Next Article in Journal
A Green Bioactive By-Product Almond Skin Functional Extract for Developing Nutraceutical Formulations with Potential Antimetabolic Activity
Next Article in Special Issue
Design, Synthesis and Evaluation of Antioxidant and NSAID Derivatives with Antioxidant, Anti-Inflammatory and Plasma Lipid Lowering Effects
Previous Article in Journal
Systematic Analysis of 2′-O-Alkyl Modified Analogs for Enzymatic Synthesis and Their Oligonucleotide Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Biological Evaluation of 2-Substituted Quinazolin-4(3H)-Ones with Antiproliferative Activities

1
Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
2
Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium
3
Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
4
Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438 Frankfurt, Germany
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(23), 7912; https://doi.org/10.3390/molecules28237912
Submission received: 12 November 2023 / Revised: 26 November 2023 / Accepted: 29 November 2023 / Published: 2 December 2023
(This article belongs to the Special Issue Design, Synthesis and Biological Evaluation of Heterocyclic Compounds)

Abstract

:
Sixteen new 2-substituted quinazolines were synthesized using a straightforward methodology starting from 2-methoxybezoic acid or 3-methoxy-2-naphthoic acid. The anti-proliferative activity of the target compounds was evaluated against nine cancer cell lines. Additionally, all the compounds were screened for their potency and selectivity against a panel of 109 kinases and four bromodomains, using Differential Scanning Fluorimetry (DSF). Compound 17 bearing a 2-methoxyphenyl substitution along with a basic side chain displayed a remarkable profile against the majority of the tested cell lines.

1. Introduction

Cancer is a complex disease that arises from the accumulation of genetic mutations and aberrant cellular signaling. The uncontrolled proliferation and metastasis of cancer cells are the hallmarks of this disease. Among the many different classes of chemotherapeutic agents used against malignant diseases, quinazoline derivatives have been extensively studied as anti-cancer compounds [1,2,3]. Furthermore, this scaffold has been used as a lead compound for the synthesis of various analogues [4,5,6,7].
Characterized by their quinazoline ring structure, these compounds have emerged as potent modulators of key cellular signaling pathways, offering promise for the development of selective and targeted interventions against cancer [8,9,10]. By inhibiting tyrosine kinases, enzymes that play a central role in cell growth and differentiation, quinazolinone-based inhibitors hold the potential to disrupt the aberrant signaling cascades that drive tumorigenesis. Their ability to target specific kinases associated with different cancer types highlights their importance in the pursuit of personalized therapeutic strategies [3,11,12].
Notable examples for approved and clinically used quinazoline-based drugs are Gefitinib [13] and the muti targeted kinase inhibitor Vandetanib [14], which have significantly impacted the treatment of non-small-cell lung cancer (NSCLC) by targeting, among others, the epidermal growth factor receptor (EGFR) (Figure 1). Following extensive Structure–Activity Relationship (SAR) studies in this class of compounds, it has been proposed that the substitution with one or two polar side chains and a substituted aniline plays an important role in their activity and selectivity as well as for optimizing the ADME (Absorption, Distribution, Metabolism, and Excretion) profile. Furthermore, Idelalisib, a PI3Kδ inhibitor, is approved for the treatment of chronic lymphocytic leukemia [15,16].
Beyond their role as kinase inhibitors, quinazolinones possess a rich array of capabilities that extend their utility in oncology. They have the potential to induce apoptosis in cancer cells, a process that triggers programmed cell death and is vital for slowing tumor growth [17,18]. Compounds I and II (Figure 1) are examples of known quinazoline analogs that inhibit tubulin polymerization. Recent studies show that 2-aryl-substituted quinazolines show moderate antiproliferative potency against various cell lines. Many other quinazoline analogs are under investigation for their antiproliferative activity [1,2,19,20,21].
In an effort to explore the optimal structural requirements for 2-aryl-substituted quinazolines, we designed a number of new analogs and present herein their synthesis and biological evaluation. In the new derivatives, a substituted phenyl or naphthyl ring is incorporated at position 2 of the quinazolinone moiety. In addition, a basic side chain was positioned at C8 aiming to identify the optimal structural requirements for biological activity. Furthermore, we modulated the aminoalkyl side chain to identify the optimal moiety for interaction with the hinge region of kinases, resulting in the preparation of various analogs with different aminoalkyl chains.

2. Results and Discussion

2.1. Chemistry

For the synthesis of the target derivatives 17–37, we used as starting material 2-methoxybezoic acid (1) or 3-methoxy-2-naphthoic acid (2), which were converted to the corresponding anhydrides by treatment with ethyl chloroformate (Scheme 1). Subsequent reaction with 2-amino-3-nitrobenzoic acid in the presence of Na2CO3 provided acids 3 and 4, respectively. The next step concerns the synthesis of the benzoxazinone analogs 5 and 6 upon heating in acetic acid anhydride, followed by reaction with ammonia to yield the intermediate amides 7 and 8 [22,23]. Ring closure of the amides 7 and 8, upon treatment with 5% aq. NaOH solution under reflux, provided the nitro quinazolinones 9 and 10 [24]. The nitro group was then easily reduced by hydrogenation to afford the amino derivatives 11 and 12, which were converted to the corresponding amides 13–16 by treatment with chloroacetyl chloride or 3-chloropropionyl chloride.
Reaction of these amides with the suitable amines resulted in the target amino-substituted compounds 17–20 (Scheme 2) [25]. The methoxy compounds 17 and 18 were then efficiently demethoxylated upon treatment with BBr3 to provide the phenol analogs 21 and 22, respectively. For the preparation of hydroxy analogs 25 and 26, we followed a slightly different approach. Specifically, chlorides 13 and 14 were first treated with potassium acetate in DMF to provide the corresponding acetates 23 and 24 that were saponified to provide the corresponding quinazolinones 25 and 26, respectively. Interestingly, the reaction of chloride 13 with potassium acetate in methanol gave compound 27 upon intramolecular cyclisation.
For the synthesis of the amides 32–35 and 36–37, we followed a similar strategy. Therefore, chlorides 15 and 16 were converted into the corresponding amines 32–35 by treatment with suitable secondary amines (Scheme 3).
Finally, the amides 32 and 33 underwent Lewis acid-mediated deprotection to provide the target amines 36 and 37, respectively. Cis-diols 30 and 31 were prepared by catalytic syn-hydroxylation of the acrylamides 28 and 29, with osmium tetroxide and N-methylmorpholine-N-oxide as the oxidizing agent [26].

2.2. Biological Assays

2.2.1. Growth Inhibiting Activity

All compounds were evaluated for their anti-proliferative activities against nine human cancer cell lines: LN-229 (glioblastoma), Capan-1 (pancreatic adenocarcinoma), Hap-1 (chronic myeloid leukemia), HCT-116 (colorectal carcinoma), NCI-H460 (lung carcinoma), DND-41 (acute lymphoblastic leukemia), HL-60 (acute myeloid leukemia), K-562 (chronic myeloid leukemia), and Z-138 (non-Hodgkin lymphoma). Staurosporine and Docetaxel were used as positive controls to validate the assay. As negative controls, the untreated cell lines were used, which allowed us to measure the baseline response in the absence of compound treatment. The results of the MTT dye reduction assay, expressed as 50% inhibitory concentrations (IC50) in µM, are depicted in Table 1.
Most of the new compounds exhibited no cytotoxic activity, with only five of them (specifically 17, 21, 25, 32, and 34) demonstrating moderate inhibitory effects on cell growth in the low micromolar potency region. It is apparent that all five of these compounds are phenyl-substituted, indicating that the presence of the naphthyl group is unfavorable for the cytotoxic activity within this class of compounds. The results obtained suggest that the demethylation of the methoxy derivative 17, resulting in the hydroxy analog 21, significantly reduces antiproliferative activity in the tested cell lines. Similarly, the data imply that dimethylamino substitution enhances activity when compared to cyclopropylamino substitution. Conversely, increasing the side chain length from two (compounds 17–20) to three carbons (compounds 32–35) diminished antiproliferative activity.
The phenyl-substituted analog 17 was the most potent inhibitor within the cell lines tested, indicating that the methoxy phenyl substitution, along with a dimethylaminoacetamido side chain, clearly enhances cytotoxic activity. Upon direct comparison of their activity against all tested cell lines, it was evident that most of the compounds were more cytotoxic against the chronic myeloid leukemia cell line Hap-1.

2.2.2. Kinases and Bromodomain Inhibition

All the synthesized compounds were screened against a panel of 113 proteins (109 kinases and 4 bromodomains), using Differential Scanning Fluorimetry (DSF) in order to assess their activity against these potential target molecules (Tables S1–S8, supporting information). The screening data highlighted 17 promising kinase targets with significant temperature shifts (Table 2). Compounds 17, 21, and 25 exhibited interesting profiles by binding to five kinases, with ΔTm values comparable to the positive controls, Staurosporine, Silmitasertib, GW779439X, and GSK626616. Notably, compound 17 demonstrated the highest potency in the assay, stabilizing most of the highlighted kinases, with ΔTm values half of those observed with the positive control.
DSF data measured on compound 17 demonstrated significant temperature stabilization of BMPK2, GSK3B, and MEK5 with ΔTm of 9 °C, 5.1 °C, and, 5.3 °C, respectively. ΔTm values were slightly lower for non-methoxylated compound 21, except for CK2A2 and PIM3 with ΔTm values at 3.9 °C and 6.5 °C, compared to 1.8 °C and 3.2 °C of compound 17. Bulkier substituents in the side chain (compound 19) resulted in a loss of potency to around half of that seen in compound 17, except for DAPK3, MST3, and DYRK1A with ΔTm at 4.1 °C, 3 °C, and 5 °C, respectively. Compounds 32 and 34, where the side chain was extended, demonstrated a loss of their potency, except for GSG2 with ΔTm values around 3 °C. Furthermore, this increase in the side chain’s length, combined with a phenolic substituent on the quinazoline ring (compound 36), led to a complete loss of potency across the tested kinases.
Naphthyl-substituted compounds either bearing a 2-methoxy or a 2-hydroxy substitution were not potent against all targets, except for compounds 18 and 20 which retained their ΔTm values against GSK3b at around 4 °C, comparable to compound 17. The addition of a polar side chain favored the stabilization of MEK5 and DYRK2, with compounds 25 and 30 showing interesting ΔTm values at 6.5 and 6.1 °C for MEK5 and 3.8 °C and 3.7 °C for DYRK2, respectively. Furthermore, these compounds exhibited a selectivity profile favoring MEK5 over the other tested MEKs, namely MEK1 and MAP2K7 (see Supplementary Material). This provides valuable insights for further investigation.

3. Materials and Methods

3.1. General Information

All commercially available reagents and solvents were purchased from Alfa Aesar (Ward Hill, MA, USA) and used without any further purification. Melting points were determined on Büchi apparatus and were uncorrected. One-dimensional (1H NMR, 13C NMR) and two-dimensional (COSY, NOESY, HMBC, HSQC-DEPT135) spectra were carried out on a Bruker Avance III-600 MHz spectrometer (Karlsruhe, Germany). Chemical shifts (δ) are expressed in ppm while coupling constants (J) are in Hz. The multiplicity of vertices is expressed as s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet of doublets), and m (multiple). 1H-NMR and 13C-NMR are available online in the supplementary material. Flash chromatography was performed on Merck silica gel (40–63 μm) with the indicated solvent system using gradients of increasing polarity in most cases (Merck KGaA—Darmstadt, Germany). The reactions were monitored by analytical thin-layer chromatography (Merck pre-coated silica gel 60 F254 TLC plates, 0.25 mm layer thickness). Mass spectra were recorded on a UPLC Triple TOF-MS (UPLC: Acquity of Waters (Milford, MA 01757, USA), SCIEX Triple TOF-MS 5600+ (Framingham, MA 01701, USA)).

3.2. Synthesis of Compounds 3–37

2-(2-Methoxybenzamido)-3-nitrobenzoic acid (3). To a solution of 2-methoxybenzoic acid (1) (150 mg, 1.08 mmol) in ACN (20 mL) at 0 °C, Et3N (275 μL, 1.97 mmol) and ClCO2Et (0.1 mL, 1.08 mmol) were added under argon. The resulting mixture was stirred for 30 min at room temperature; after which, Na2CO3 (195 mg, 2.96 mmol) and 2-amino-3-nitrobenzoic acid (200 mg, 1.08 mmol) were added, and the mixture was heated at 50 °C for 24 h. The reaction mixture was then vacuum evaporated, diluted with water, and acidified with 9% aq. HCl solution (pH ≈ 3). The precipitate was filtered and air-dried to give crude 3, which was purified by column chromatography (silica gel) using a mixture of CH2Cl2/CH3OH 100/2–100/16 as the eluent to afford 180 mg (57%) of the title compound as a yellow solid. Mp.: 134–136 °C (EtOAc). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 8.24 (dd, J = 7.8, 1.6 Hz, 1H, H-6), 8.11 (dd, J = 8.1, 1.6 Hz, 1H, H-4), 7.96 (dd, J = 7.8, 1.9 Hz, 1H, H-6′), 7.61 (td, J = 8.4, 1.8 Hz, 1H, H-4′), 7.45 (t, J = 8.0 Hz, 1H, H-5), 7.26 (dd, J = 8.4, 1.0 Hz, 1H, H-3′), 7.11 (dd, J = 8.1, 1.0 Hz, 1H, H-5′), 4.05 (s, 3H, OCH3).13C NMR (151 MHz, DMSO-d6) δ (ppm) 167.3 (NHCO), 163.1 (COOH), 157.8 (C-2′), 144.8 (C-3), 134.8 (C-6), 134.4 (C-4′), 131.7 (C-6′), 130.8 (C-2), 128.1 (C-4), 126.5, (C-1), 124.4 (C-5), 120.9, (C-5′), 120.1, (C-1′), 112.5 (C-3′), 56.1 (OCH3).
2-(3-Methoxy-2-naphthamido)-3-nitrobenzoic acid (4). This compound was synthesized by an analogous procedure as described for the preparation of compound 3, using 3-methoxy-2-naphthoic acid (2). Yield: 55%. Mp.: 145–147 °C (EtOAc). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 12.03 (brs, 1H, D2O exch., NH), 8.62 (s, 1H, H-1′), 8.26 (dd, J = 7.8, 1.6 Hz, 1H, H-6), 8.19 (dd, J = 8.2, 1.6 Hz, 1H, H-4), 8.05 (d, J = 8.2 Hz, 1H, H-8′), 7.92 (d, J = 8.2 Hz, 1H, H-5′), 7.64–7.58 (m, 2H, H-4′, H-6′), 7.52 (t, J = 8.0 Hz, 1H, H-5), 7.45 (dd, J = 8.1, 6.8, 1.2 Hz, 1H, H-7′), 4.17 (s, 3H, OCH3). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 167.1 (NHCO), 163.0 (COOH), 154.4 (C-3′), 144.9 (C-3), 136.0 (C-4a’), 134.9 (C-6), 133.6 (C-1′), 130.6 (C-2), 129.1 (C-8′), 128.9 (C-6′), 128.6 (C-4), 127.5 (C-8a’), 126.4 (C-5′), 125.5 (C-1), 124.9 (C-5), 124.7 (C-7′), 121.3 (C-2′), 107.3 (C-4′), 56.1 (OCH3).
2-(2-Methoxyphenyl)-8-nitroquinazolin-4(3H)-one (9). A suspension of compound 3 (1 g, 3.16 mmol) in (CH3CO)2O (6.83 mL, 72.28 mmol) was refluxed for 2 h. The volatiles were then vacuum evaporated, and the residue was treated with NH3 (0.5 M solution in THF, 15 mL). After completion of the reaction, the solvent was vacuum evaporated, and the residue was dissolved in 5% aq. NaOH solution (10 mL) and refluxed for 10 min. After cooling, the reaction mixture was diluted with water and acidified with 9% aq. HCl solution (pH ≈ 3). The precipitate was filtered, washed with water, and air-dried to give compound 9 (750 mg, 79.8%), practically pure, which was used for the next step without any further purification. Mp.: 168–170 °C (EtOAc). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.37 (dd, J = 8.0, 1.5 Hz, 1H, H-5), 8.30 (dd, J = 7.8, 1.5 Hz, 1H, H-7), 7.66 (m, 2H, H-6, H-6′), 7.57 (td, J = 8.8, 1.8 Hz, 1H, H-4′), 7.22 (d, J = 8.4 Hz, 1H, H-3′), 7.11 (t, J = 7.5 Hz, 1H, H-5′), 3.88 (s, 3H, OCH3).13C NMR (151 MHz, DMSO-d6) δ (ppm) 159.9 (CO), 157.4 (C-2′), 154.8 (C-2), 146.8 (C-8), 140.8 (C-8a), 133.1 (C-4′), 130.7 (C-6′), 129.7 (C-5), 128.1, (C-7), 126.2 (C-6), 122.5, (C-4a), 121.8, (C-1′), 120.7 (C-5′), 112.2 (C-3′), 56.0 (OCH3).
2-(3-Methoxynaphthalen-2-yl)-8-nitroquinazolin-4(3H)-one (10). This compound was synthesized by an analogous procedure as described for the preparation of compound 9. Yield: 80%. Mp.: 186–188 °C (EtOAc-n-Pentane). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.41 (d, J = 8.0 Hz, 1H, H-5), 8.33 (d, J = 7.8 Hz, 1H, H-7), 8.16 (s, 1H, H-1′), 7.98 (d, J = 6.25 Hz, 1H, H-8′), 7.91 (d, J = 6.24 Hz, 1H, H-5′), 7.70 (t, J = 7.9 Hz, 1H, H-6), 7.58 (t, J = 7.9 Hz, 1H, H-6′), 7.54 (s, 1H, H-4′), 7.43 (td, J = 8.0, 1.0 Hz, 1H, H-7′), 3.95 (s, 3H, OCH3). 13C NMR (101 MHz, DMSO-d6) δ (ppm) 160.0 (CO), 154.9 (C-3′), 154.4 (C-2), 146.8 (C-8), 140.7 (C-8a), 135.4 (C-4a’), 131.0 (C-1), 129.7 (C-5), 128.5 (C-6′), 128.2 (C-8′), 128.1 (C-7), 127.4 (C-8a’), 126.7 (C-5′), 126.4 (C-6), 124.6 (C-7′), 124.2 (C-2′), 122.6 (C-4a), 106.7 (C-4′), 56.0 (OCH3).
2-Chloro-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (13). A solution of 9 (200 mg, 0.67 mmol) in abs. EtOH (10 mL) was hydrogenated in the presence of Pd/C (15 mg), under pressure (50 psi), at room temperature, for 4 h. After completion of the reaction, the mixture was filtered through a Celite pad, and the filtrate was evaporated to dryness to provide the amino derivative 11. Without further purification, the oily residue was dissolved in THF (10 mL) and CH2Cl2 (5 mL). To this solution, Na2CO3 (210 mg, 2.01 mmol) and chloroacetyl chloride (59 μL, 0.74 mmol) were added. The resulting suspension was stirred for 15 min at room temperature. The volatiles were then vacuum evaporated, and the residue was diluted with water and acidified with 9% aq. HCl solution (pH ≈ 3). The precipitate was filtered, washed with water, and air dried to afford the title compound 13 (161 mg, 70%), practically pure, which was used for the next step without any further purification. Mp.: 132–134 °C (MeOH). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 12.20 (brs, 1H, D2O exch., NH), 10.20 (brs, 1H, D2O exch., NHCO), 8.62 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 7.90 (dd, J = 7.6, 1.8 Hz, 1H, H-6′), 7.86 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.57 (td, J = 8.8, 1.8 Hz, 1H, H-4′), 7.52 (t, J = 8.0 Hz, 1H, H-6), 7.23 (d, J = 8.3 Hz, 1H, H-3′), 7.14 (t, J = 7.5 Hz, 1H, H-5′), 4.54 (s, 2H, CH2), 3.90 (s, 3H, OCH3).13C NMR (151 MHz, DMSO-d6) δ (ppm) 164.9 (NHCO), 160.9 (CO), 157.4 (C-2′), 152.0 (C-2), 138.9 (C-8a), 133.2 (C-4a), 132.8 (C-4′), 130.9 (C-6′), 126.7 (C-6), 122.4 (C-7), 121.9 (C-5′), 120.9 (C-8), 120.7 (C-5), 120.2 (C-1′), 112.1 (C-3′), 56.0 (OCH3), 43.6 (CH2).
2-Chloro-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (14). This compound was synthesized by an analogous procedure as described for the preparation of compound 13. Yield: 77%. Mp.: 156–158 °C (THF-n-Pentane). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 12.45 (brs, 1H, D2O exch., NH), 10.24 (brs, 1H, D2O exch., NHCO), 8.67 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 8.41 (s, 1H, H-1′), 7.93 (d, J = 8.1 Hz, 1H, H-8′), 7.89 (d, J = 8.2 Hz, 1H, H-5′), 7.59 (dd, J = 8.2, 1.3 Hz, 1H,H-6′), 7.57–7.52 (m, 2H, H-6, H-4′), 7.45 (dd, J = 8.0, 1.2 Hz, 1H, H-7′), 4.55 (s, 2H, CH2), 3.98 (s, 3H, OCH3). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 164.8 (NHCO), 160.9 (CO), 154.6 (C-3′), 151.8 (C-2), 138.8 (C-8a), 135.3 (C-4a’), 133.3 (C-4a), 131.3 (C-1′), 128.4 (C-6′), 128.0 (C-8′), 127.6 (C-8a’), 126.7 (C-6), 126.6 (C-5′, C-7′), 124.5 (C-2′), 122.4 (C-7), 121.0 (C-8), 120.3 (C-2′), 106.6 (C-4′), 56.9 (OCH3), 43.6 (CH2).
3-Chloro-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (15). This compound was synthesized by an analogous procedure as described for the preparation of compound 13. Yield: 67%. Mp.: 153–155 °C (MeOH). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 12.18 (brs, 1H, D2O exch., NH), 9.76 (brs, 1H, D2O exch., NHCO), 8.63 (dd, J = 8.3, 1.6 Hz, 1H, H-7), 7.96 (dd, J = 7.6, 1.8 Hz, 1H, H-6′), 7.83 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.57 (td, J = 8.5, 1.8 Hz, 1H, H-4′), 7.49 (t, J = 8.0 Hz, 1H, H-6), 7.22 (dd, J = 8.6, 1.0 Hz, 1H, H-3′), 7.13 (td, J = 7.5, 1.0 Hz, 1H, H-5′), 3.94–3.85 (m, 5H, OCH3, COCH2CH2), 3.06 (t, J = 6.3 Hz, 2H, COCH2CH2).13C NMR (101 MHz, DMSO-d6) δ (ppm) 168.5 (NHCO), 160.9 (CO), 157.4 (C-2′), 151.6 (C-2), 138.9 (C-8a), 134.0 (C-4a), 132.6 (C-4′), 131.2 (C-6′), 126.5 (C-6), 123.4 (C-7), 122.1 (C-5′), 120.9 (C-8), 120.6 (C-5), 119.9 (C-1′), 111.9 (C-3′), 55.9 (OCH3), 40.7 (COCH2CH2), 30.7 (COCH2CH2).
3-Chloro-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (16). This compound was synthesized by an analogous procedure as described for the preparation of compound 13. Yield: 81%. Mp.: 167–169 °C (THF-n-Pentane). 1H NMR (600 MHz CDCl3) δ (ppm) 10.96 (brs, 1H, D2O exch., NH), 9.58 (s brs, 1H, D2O exch., NHCO), 8.95 (s, 1H, H-1′), 8.87 (dd, J = 7.9, 1.4 Hz, H-7), 8.00 (dd, J = 8.0, 1.4 Hz, H-5), 7.96 (d, J = 8.1 Hz, H-8′), 7.81 (d, J = 8.2 Hz, 1H, H-5′), 7.59 (dd, J = 8.1, 1.2 Hz, 1H, H-6′), 7.51–7.44 (m, 2H, H-6, H-7′), 7.35 (s, 1H, H-4′), 4.16 (s, 3H, OCH3), 4.01 (t, J = 6.3 Hz, 2H, COCH2CH2), 3.06 (t, J = 6.3 Hz, 2H, COCH2CH2). 13C NMR (151 MHz, CDCl3) δ (ppm) 168.0 (NHCO), 161.4 (CO), 154.8 (C-3′), 150.3 (C-2), 138.6 (C-8a), 136.0 (C-4a’), 133.9 (C-4a), 133.1 (C-1′), 129.2 (C-6′), 129.1 (C-8′), 128.5 (C-8a’), 127.3 (C-6), 126.7 (C-5′), 125.4 (C-7′), 122.9 (C-7), 120.9 (C-5), 120.7 (C-8), 120.6 (C-2′), 107.5 (C-4′), 56.4 (OCH3), 41.3 (COCH2CH2), 40.2 (COCH2CH2).
2-(Dimethylamino)-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (17). To a solution of chloride 13 (80 mg, 0.23 mmol) in anh. THF (10 mL), a 5.6 M ethanolic solution of dimethylamine (4.66 mmol) was added dropwise and the resulting mixture was heated at 100 °C, in an autoclave apparatus, for 65 h. After cooling, the solvent was vacuum evaporated and the oily residue was purified by column chromatography (silica gel, CH2Cl2/MeOH 95/5) to afford 17 (59 mg, 73.2%). Mp.: >230 °C (EtOAc-n-Pentane). IR (Nujol) ν max/cm−1: 1671.98 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 11.19 (brs, 1H, D2O exch., NH), 11.10 (brs, 1H, D2O exch., NHCO), 8.78 (dd, J = 7.9, 1.4 Hz, 1H, H-7), 8.62 (dd, J = 8.0, 1.8 Hz, 1H, H-6′), 7.92 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.53 (td, J = 8.4, 1.8 Hz, 1H, H-4′), 7.41 (t, J = 8.0 Hz, 1H, H-6), 7.14 (td, J = 8.1, 1.0 Hz, 1H, H-5′), 7.09 (dd, J = 8.4, 1.0 Hz, 1H, H-3′), 4.08 (s, 3H, OCH3), 3.22 (s, 2H, CH2), 2.52 (s, 6H, (CH3)2). 13C NMR (151 MHz, CDCl3) δ (ppm) 169.1 (NHCO), 161.7 (CO), 158.4 (C-2′), 149.7 (C-2), 138.9 (C-8a), 134.0 (C-4a), 133.6 (C-4′), 131.2 (C-6′), 127.0 (C-6), 122.3 (C-7), 121.8 (C-5′), 120.9 (C-8), 120.2 (C-5), 119.5 (C-1′), 112.3 (C-3′), 64.1 (CH2), 56.4 (OCH3), 46.3 (CH3)2).
2-(Dimethylamino)-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (18). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 57%. Mp.: >230 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1668.23 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 11.19 (brs, 2H, D2O exch., NH, NHCO), 9.16 (s, 1H, H-1′), 8.89 (dd, J = 7.9, 1.4 Hz, 1H, H-7), 7.98 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.89 (d, J = 8.1 Hz, 1H, H-8′), 7.82 (d, J = 8.2 Hz, 1H, H-5′), 7.59 (td, J = 8.1, 1.2 Hz, 1H, H-6′), 7.48 (m, 2H, H-6, H-7′), 7.37 (s, 1H, H-4′), 4.19 (s, 3H, OCH3), 3.27 (s, 2H, CH2), 2.61 (s, 6H, (CH3)2).13C NMR (151 MHz, CDCl3) δ (ppm) 169.2 (NHCO), 161.7 (CO), 155.3 (C-3′), 149.8 (C-2), 139.0 (C-8a), 136.1 (C-4a’), 134.2 (C-4a), 133.0 (C-1′), 129.0 (C-6′), 128.8 (C-8′), 128.8 (C-8a’), 127.3 (C-6), 126.9 (C-5′), 125.5 (C-7′), 122.6 (C-7), 121.1 (C-8), 120.7 (C-2′), 120.4 (C-5), 107.6 (C-4′), 64.4 (CH2), 56.5 (OCH3), 46.6 ((CH3)2).
2-(Cyclopropylamino)-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (19): This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 63%. Mp.: >230 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1669.11 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 11.07 (brs, 1H, D2O exch., NH), 10.91 (brs, 1H, D2O exch., NHCO), 8.88 (dd, J = 7.8, 1.3 Hz, 1H, H-7), 8.57 (dd, J = 8.0, 1.8 Hz, 1H, H-6′), 7.93 (dd, J = 7.8, 1.3 Hz, 1H, H-5), 7.55 (dd, J = 7.9, 1.8 Hz, 1H, H-4′), 7.41 (t, J = 7.9 Hz, 1H, H-6), 7.19 (t, J = 7.6 Hz, 1H, H-5′), 7.10 (d, J = 8.5 Hz, 1H, H-3′), 4.08 (s, 3H, OCH3), 3.64 (s, 2H, CH2), 2.37 (m, 1H, CH-cyclopropyl), 0.63–0.52 (m, 4H, CH2-cyclopropyl). 13C NMR (151 MHz, CDCl3) δ (ppm) 170.6 (NHCO), 161.7 (CO), 158.3 (C-2′), 149.8 (C-2), 138.8 (C-8a), 134.1 (C-4a), 133.7 (C-4′), 131.4 (C-6′), 127.1 (C-6), 122.3 (C-7), 121.8 (C-5′), 120.9 (C-8), 120.3 (C-5), 119.5 (C-1′), 112.3 (C-3′), 56.4 (OCH3), 54.1 (CH2), 31.7 (CH-cyclopropyl), 6.8 (CH2-cyclopropyl); HRMS (ESI+) m/z 365.1617 (calcd for C20H21N4O3+, 365.1608).
2-(Cyclopropylamino)-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (20). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 88%. Mp.: >230 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1667.75 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 10.99 (brs, 1H, D2O exch., NH), 10.95 (brs, 1H, D2O exch., NHCO), 9.01 (s, 1H, H-1′), 8.93 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 7.99–7.94 (m, 2H, H-5, H-8′), 7.81 (d, J = 8.2 Hz, 1H, H-5′), 7.58 (dd, J = 8.1, 1.2 Hz, 1H, H-6′), 7.50–7.42 (m, 2H, H-6, H-7′), 7.32 (s, 1H, H-4′), 4.14 (s, 3H, OCH3), 3.66 (s, 2H, CH2), 2.35 (m, 1H, CH-cyclopropyl), 0.63–0.57 (m, 2H, CH2-cyclopropyl), 0.63–0.57 (m, 2H, CH2-cyclopropyl). 13C NMR (151 MHz, CDCl3) δ (ppm) 170.8 (NHCO), 161.6 (CO), 155.0 (C-3′), 149.9 (C-2), 138.9 (C-8a), 136.0 (C-4a’), 134.2 (C-4a), 132.9 (C-1′), 129.1 (C-6′), 128.9 (C-8′), 128.6 (C-8a’), 127.3 (C-6), 126.8 (C-5′), 125.2 (C-7′), 122.5 (C-7), 121.1 (C-8), 120.9 (C-2′), 120.3 (C-5), 107.5 (C-4′), 56.4 (OCH3), 54.2 (CH2), 32.1 (CH-cyclopropyl), 7.0 (CH2-cyclopropyl); HRMS (ESI+) m/z 415.1770 (calcd for C24H23N4O3+, 415.1765).
2-(Dimethylamino)-N-(2-(2-hydroxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (21). BBr3 (0.52 mL, 0.51 mmol, 10% solution in CH2Cl2) was added dropwise to a solution of 17 (30 mg, 0.085 mmol) in anh. CH2Cl2 (5 mL), at −40 °C, under argon, and the reaction mixture was stirred at this temperature for 10 min and at 0 °C for 24 h. Afterwards, MeOH (10 mL) and a saturated NaHCO3 solution were added (0.5 mL) and stirring was continued for 10 min. Most of the organic solvents were vacuum evaporated. The residue was then extracted with EtOAc, the organic phase was washed with a saturated NaHCO3 solution, water, and brine, dried (anh. Na2SO4), and concentrated to dryness. The crude product was purified by column chromatography (silica gel), using a mixture of CH2Cl2/CH3OH 100/2 to 100/8 as the eluent, to afford 13 mg (45%) of the title compound. Mp.: >230 °C (EtOAc). IR (Nujol) ν max/cm−1: 1668.51 (CO). 1H NMR (600 MHz, CDCl3-MeOD) δ (ppm) 8.80 (dd, J = 7.9, 1.4 Hz, 1H, H-7), 8.45 (dd, J = 8.3, 1.7 Hz, 1H, H-6′), 8.01 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.55–7.50 (m, 2H, H-6, H-4′), 7.13–7.11 (m, 2H, H-3′, H-5′), 3.31 (s, 2H, CH2), 2.62 (s, 6H, (CH3)2). 13C NMR (151 MHz, CDCl3-MeOD) δ (ppm) δ 170.7 (NHCO), 163.0 (CO), 158.4 (C-2′), 151.9 (C-2), 139.3 (C-8a), 134.2 (C-4′), 133.5 (C-4a), 129.6 (H-6′), 127.2 (C-6), 123.5 (C-7), 121.0 (C-5′, C-8), 120.6 (C-5), 118.0 (C-3′), 116.4 (C-1′), 64.1 (CH2), 46.3 (CH3)2); HRMS (ESI+) m/z 339.1457 (calcd for C18H19N4O3+, 339.1452).
2-(Dimethylamino)-N-(2-(3-hydroxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (22). This compound was synthesized by an analogous procedure as described for the preparation of compound 21. Yield: 40%. Mp.: >230 °C (EtOAc). IR (Nujol) ν max/cm−1: 1670.07 (CO). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 10.84 (brs, 2H, D2O exch., NHCO, NH), 8.81 (s, 1H, H-1′), 8.61 (d, J = 8.4 Hz, 1H, H-7), 7.85–7.88 (m, 2H, H-5′, H-8′), 7.79 (d, J = 8.0 Hz, 1H, H-5), 7.55–7.47 (m, 2H, H-6′, H-7′), 7.43–7.35 (m, 2H, H-6, H-4′), 3.30 (m, 2H, CH2), 2.55 (s, 6H, (CH3)2). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 167.8 (NHCO), 161.1 (CO), 154.0 (C-3′), 152.1 (C-2), 138.7 (C-8a), 135.8 (C-4a’), 133.2 (C-4a), 130.7 (C-1′), 128.4 (C-6′, C-8a’), 128.2 (C-8′), 127.0 (C-6), 126.5 (C-5′), 125.9 (C-7′), 124.0 (C-7), 121.1 (C-8), 120.9 (C-2′), 120.0 (C-5), 119.9 (C-2′), 110.9 (C-4′), 62.4 (CH2), 45.4 ((CH3)2); HRMS (ESI+) m/z 389.1615 (calcd for C22H21N4O3+, 389.1608).
2-Hydroxy-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (25). To a solution of 13 (100 mg, 0.29 mmol) in anh. DMF (20 mL), under argon, was added CH3COOK (57 mg, 0.58 mmol) and the mixture was heated at 50 °C for 2 h. After completion of the reaction, the mixture was poured into water and acidified with 9% aq. HCl solution (pH ≈ 3). The precipitate was filtered, washed with water, and air dried to afford crude intermediate 23 (65 mg, 0.18 mmol), which was purified by column chromatography (silica gel) using a mixture of CH2Cl2/EtOAc 100/10 to CH2Cl2/EtOAc 25/10 as the eluent. This compound was then dissolved in MeOH (10 mL) and treated with 30% aq. NaOH solution at room temperature for 2 h. The resulting precipitate was filtered and purified by column chromatography (silica gel), using a mixture of CH2Cl2/CH3OH 100/4 to CH2Cl2/CH3OH 100/12 as the eluent, to afford 30 mg (52.6%) of the title compound 25. Mp.: 113–115 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1668.11 (CO). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 10.53 (brs, 1H, D2O exch., NHCO), 8.64 (dd, J = 7.8, 1.4 Hz, 1H, H-7), 7.74 (dd, J = 8.0, 1.5 Hz, 1H, H-6′), 7.68 (dd, J = 7.6, 1.8 Hz, 1H, H-5), 7.45 (dd, J = 8.9, 1.8 Hz, 1H, H-4′), 7.31 (t, J = 7.9 Hz, 1H, H-6), 7.14 (d, J = 8.3 Hz, 1H, H-3′), 7.05 (td, J = 7.4, 1.0 Hz, 1H, H-5′), 4.03 (s, 2H, CH2), 3.83 (s, 3H, OCH3). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 174.3 (NHCO), 170.6 (CO), 157.3 (C-2′, C-2), 140.0 (C-8a), 132.7 (C-4a), 132.8 (C-4′), 130.5 (C-6′), 127.8 (C-6), 124.3 (C-8), 120.7 (C-5′), 120.2 (C-7), 119.5 (C-5), 119.0 (C-1′), 112.2 (C-3′), 61. 9 (CH2), 55.9 (OCH3); HRMS (ESI+) m/z 326.1141 (calcd for C17H16N3O4+, 326.1135).
2-Hydroxy-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acetamide (26). This compound was synthesized by an analogous procedure as described for the preparation of compound 25. Yield: 58%. Mp.: 198–200 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1669.74 (CO). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 10.53 (brs, 1H, D2O exch., NHCO), 8.80 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 8.32 (s, 1H, H-1′), 7.96 (d, J = 8.1 Hz, 1H, H-8′), 7.93 (d, J = 8.2 Hz, 1H, H-5′), 7.81 (dd, J = 8.2, 1.42Hz, 1H, H-5), 7.59 (dd, J = 8.2, 1.2 Hz, 1H, H-6′), 7.55 (s, 1H, H-4′), 7.52 (t, J = 8.0 Hz, 1H, H-6), 7.45 (td, J = 8.0, 6.8, 1.2 Hz, 1H, H-7′), 4.06 (s, 2H, CH2), 3.98 (s, 3H, OCH3). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 171.0 (NHCO), 164.4 (CO), 155.7 (C-3′, C-2), 140.6 (C-8a), 135.0 (C-4a’), 133.1 (C-4a), 130.6 (C-1′), 128.5 (C-8′), 128.2 (C-8a’), 127.6 (C-6′, C-6), 126.9 (C-5′), 124.5 (C-7′), 121.4 (C-8), 120.1 (C-5, C-7), 119.4 (C-2′), 106.7 (C-4′), 62.4 (CH2). 56.2 (OCH3); HRMS (ESI+) m/z 376.1287 (calcd for C21H18N3O4+, 376.1292).
5-(2-Methoxyphenyl)-3-hydro-1H,7H-pyrazino[3,2,1-ij]quinazoline-2,7-dione (27). A suspension of 13 (40 mg, 0.11 mmol) and CH3COOK (23 mg, 0.23 mmol) in dry MeOH (20 mL) was stirred under argon, at room temperature, for 18 h. The solvent was vacuum evaporated, the residue was dissolved in CH2Cl2, washed with water, dried (anh. Na2SO4), and evaporated to dryness. Flash chromatography on silica gel using a mixture of cyclohexane/EtOAc 1/1 as the eluent provided the title compound 27 (15 mg, 45.5%). Mp.: 202–204 °C (THF-n-Pentane). 1H NMR (600 MHz, CDCl3) δ (ppm) 7.92 (dd, J = 8.0, 1.3 Hz, 1H, H-8), 7.49 (td, J = 8.4, 1.8 Hz, 1H, H-4′), 7.43–7.34 (m, 2H, H-9, H-6′), 7.19 (dd, J = 7.8, 1.3 Hz, 1H, H-10), 7.09 (td, J = 7.6, 0.9 Hz, 1H, H-5′), 7.00 (d, J = 8.4 Hz, 1H, H-3′), 4.74 (d, J = 18.0 Hz, 1H, CHH), 4.39 (d, J = 17.9 Hz, 1H, CHH), 3.82 (s, 3H, OCH3). 13C NMR (151 MHz, CDCl3) δ (ppm) 168.4 (CO-7), 162.4 (CO-2), 159.9 (C-5), 155.7 (C-2′), 132.7 (C-4′), 129.9 (C-6′), 127.2 (C-10b), 127.1 (C-9), 122.4 (C-8), 122.3 (C-10a), 121.8 (C-5′), 119.9 (C-7a), 118.7 (C-10), 111.5 (C-3′), 55.9 (OCH3), 49.5 (CH2); HRMS (ESI+) m/z 308.1037 (calcd for C17H14N3O3+, 308.1030).
N-(2-(2-Methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)acrylamide (28). A solution of 15 (120 mg, 0.34 mmol) and Et3N (0.47 mL, 3.34 mmol) in anh. THF (20 mL) was stirred for 18 h at 70 °C, under argon. The solvent was vacuum evaporated, the residue was dissolved in CH2Cl2, washed with water, dried (anh. Na2SO4), and evaporated to dryness. Flash chromatography on silica gel using a mixture of cyclohexane/EtOAc 1/1 as the eluent provided 90 mg (83.3%) of the title compound 28. Mp.: 190–192 °C (THF-n-Pentane). 1H NMR (400 MHz, CDCl3) δ (ppm) 10.98 (brs, 1H, D2O exch., NH), 9.46 (brs, 1H, D2O exch., NHCO), 8.95 (dd, J = 8.0, 1.3 Hz, 1H, H-7), 8.43 (dd, J = 7.9, 1.8 Hz, 1H, H-6′), 7.98 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.57 (dd, J = 8.6, 1.8 Hz, 1H, H-4′), 7.48 (t, J = 8.0 Hz, 1H, H-6), 7.21 (t, J = 7.6 Hz, 1H, H-5′), 7.12 (d, J = 8.4 Hz, 1H, H-3′), 6.78 (dd, J = 17.0, 1.2 Hz, 1H, COCH=CHH), 6.29 (dd, J = 16.9, 1.8 Hz, 1H, COCH=CHH), 5.86 (dd, J = 9.4, 2.0 Hz, 1H, COCH=CHH), 4.09 (s, 3H, OCH3). 13C NMR (101 MHz, CDCl3) δ (ppm) 163.6 (NHCO), 161.5 (CO), 158.1 (C-2′), 150.1 (C-2), 138.5 (C-8a), 134.1 (C-4a), 133.8 (C-4′), 131.8 (COCH=CH2), 131.1 (C-6′), 127.8 (COCH=CH2), 127.2 (C-6), 122.8 (C-7), 122.0 (C-5′), 120.8 (C-8), 120.51(C-5), 119.5 (C-1′), 112.2 (C-3′), 56.4 (OCH3); HRMS (ESI+) m/z 322.1192 (calcd for C18H16N3O3+, 322.1186).
N-(2-(3-Methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)acrylamide (29). This compound was synthesized by an analogous procedure as described for the preparation of compound 28. Yield: 40.7%. Mp.: 210–212 °C (THF-n-Pentane). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 12.41 (brs, 1H, D2O exch., NH), 9.86 (brs, 1H, D2O exch., NHCO), 8.74 (dd, J = 7.9, 1.4 Hz, 1H, H-7), 8.47 (s, 1H, H-1′), 8.00 (d, J = 8.2 Hz, 1H, H-8′), 7.92 (d, J = 8.2 Hz, 3H, H-5′), 7.88 (dd, J = 7.9, 1.4 Hz, 1H, H-5), 7.58 (td, J = 8.2, 6.8, 1.3 Hz, 1H, H-6′), 7.56–7.51 (m, 2H, H-6, H-4′), 7.45 (dd, J = 8.2, 1.2 Hz, 1H, H-7′), 6.78 (dd, J = 16.9, 10.2 Hz, 1H, COCH=CHH), 6.31 (dd, J = 16.9, 1.7 Hz, 1H, COCH=CHH), 5.78 (dd, J = 10.3, 1.8 Hz, 1H, COCH=CHH), 3.97 (s, 3H, OCH3). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 163.4 (NHCO), 160.9 (CO), 154.6 (C-3′), 151.7 (C-2), 139.1 (C-8a), 135.2 (C-4a’), 134.0 (C-4a), 132.2 (COCH=CH2), 131.6 (C-1′), 128.4 (C-8′), 127.9 (C-6′), 127.6 (C-8a’), 127.3 (COCH=CH2), 126.6 (C-6), 126.5 (C-5′), 124.3 (C-7′), 124.2 (C-8), 123.5(C-7), 121.0 (C-2′), 120.0 (C-5), 106.4 (C-4′), 55.9 (OCH3); HRMS (ESI+) m/z 372.1338 (calcd for C22H18N3O3+, 372.1343).
2,3-Dihydroxy-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (30). To a solution of osmium tetroxide (2.5% in isopropanol) (0.23 mL, 0.0036 mmol) and N-methylmorpholine N-oxide (14.78 mg, 0.126 mmol) in THF (10 mL), compound 28 (30 mg, 0.09 mmol) was added. The reaction mixture was stirred at rt for 3 days. A saturated NaHSO3 solution (0.5 mL) was then added, the mixture was stirred at rt for 60 min, and the volatiles were vacuum evaporated. The residue was then extracted with EtOAc, and the organic phase was washed with a saturated NaHCO3 solution, water, and brine, dried (anh. Na2SO4), and concentrated to dryness. Flash chromatography on silica gel using a mixture of CH2Cl2/CH3OH 100/0–100/5 as the eluent provided 13 mg (40%) of the title compound 30. Mp.: 210–212 °C (EtOH). IR (Nujol) ν max/cm−1: 1669.52 (CO). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 12.19 (brs, 1H, D2O exch., NH), 10.61 (brs, 1H, D2O exch., NHCO), 8.80 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 7.82 (dd, J = 8.0, 1.8 Hz, 1H, H-6′), 7.80 (dd, J = 7.8, 1.3 Hz, 1H, H-5), 7.57 (dd, J = 8.9, 1.8 Hz, 1H, H-4′), 7.49 (t, J = 8.0 Hz, 1H, H-6), 7.23 (d, J = 8.4 Hz, 1H, H-3′), 7.12 (td, J = 7.5, 1.0 Hz, 1H, H-5′), 6.26 (d, J = 5.1 Hz, 1H, CHOH), 4.84 (t, J = 5.3 Hz, 1H, CH2OH), 4.12 (td, J = 5.1, 3.2 Hz, 1H, CH), 3.90 (s, 3H, OCH3), 3.73–3.63 (m, 2H, CH2). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 171.2 (NHCO), 160.9 (CO), 157.4 (C-2′), 151.7 (C-2), 138.3 (C-8a), 133.5 (C-4a), 132.6 (C-4′), 130.5 (C-6′), 126.7 (C-6), 122.1 (C-7), 120. (C-8a, C-5′), 120.60 (C-5), 119.2 (C-1′), 112.1 (C-3′), 73.6 (CH), 63.6 (CH2), 55.9 (OCH3); HRMS (ESI+) m/z 356.1248 (calcd for C18H18N3O5+, 356.1241).
2,3-Dihydroxy-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (31). This compound was synthesized by an analogous procedure as described for the preparation of compound 30. Yield: 41%. Mp.: 216–218 °C (EtOH). IR (Nujol) ν max/cm−1: 1661.24 (CO). 1H NMR (600 MHz, DMSO-d6) δ (ppm) 10.66 (brs, 1H, D2O exch., NHCO), 8.81 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 8.37 (s, 1H, H-1′), 7.97 (d, J = 8.1 Hz, 1H, H-8′), 7.93 (d, J = 8.2 Hz, 3H, H-5′), 7.83 (dd, J = 8.1, 1.4 Hz, 1H, H-5), 7.59 (dd, J = 8.2, 1.3 Hz, 1H, H-6′), 7.56 (s, 1H, H-4′), 7.52 (t, J = 8.0 Hz, 1H, H-6), 7.46 (dd, J = 7.4, 1.0 Hz, 1H, H-7′), 3.99 (s, 3H, OCH3), 6.27 (d, J = 5.1 Hz, 1H,CHOH), 4.85 (t, J = 5.9 Hz, 1H, CH2OH), 4.13 (td, J = 5.1, 3.2 Hz, 1H, CH), 3.90 (s, 3H, OCH3), 3.71–3.65 (m, 2H, CH2). 13C NMR (151 MHz, DMSO-d6) δ (ppm) 171.2 (NHCO), 161.0 (CO), 154.7 (C-3′), 151.6 (C-2), 138.3 (C-8a), 135.3 (C-4a’), 133.7 (C-4a), 131.0 (C-1′), 128.4 (C-8′), 128.0 (C-6′), 127.6 (C-8a’), 126.9 (C-6), 126.6 (C-5′), 124.5 (C-7′), 124.1 (C-8), 120.9 (C-7), 120.9 (C-5), 119.3 (C-2′), 106.7 (C-4′), 73.5 (CH), 63.6 (CH2), 56.0 (OCH3); HRMS (ESI+) m/z 406.1405 (calcd for C22H20N3O5+, 406.1397).
3-(Dimethylamino)-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (32). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 57%. Mp.: 208–210 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1669.28 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 10.92 (brs, 1H, D2O exch., NHCO), 8.93 (dd, J = 8.0, 1.5 Hz, 1H, H-7), 8.56 (dd, J = 7.9, 1.8 Hz, 1H, H-6′), 7.97 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.55 (dd, J = 8.8, 1.8 Hz, 1H, H-4′), 7.44 (t, J = 8.0 Hz, 1H, H-6), 7.17 (d, J = 7.6 Hz, 1H, H-5′), 7.11 (d, J = 8.4 Hz, 1H, H-5′), 4.08 (s, 3H, OCH3), 2.70 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.67 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.37 (s, 6H, (CH3)2). 13C NMR (101 MHz, CDCl3) δ (ppm) 171.4 (NHCO), 161.6 (CO), 157.9 (C-2′), 149.6 (C-2), 138.8 (C-8a), 135.1 (C-4a), 133.4 (C-4′), 131.5 (C-6′), 126.9 (C-6), 123.5 (C-7), 121.4 (C-5′), 120.8 (C-8), 120.0 (C-5), 119.8 (C-1′), 112.0 (C-3′), 56.2 (COCH2CH2), 55.6 (OCH3), 45.3 ((CH3)2), 35.4 (COCH2CH2); HRMS (ESI+) m/z 367.1770 (calcd for C20H23N4O3+, 367.1765).
3-(Dimethylamino)-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (33). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 60%. Mp.: >230 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1665.49 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 11.00 (brs, 1H, D2O exch., NH), 10.68 (brs, 1H, D2Oexch., NHCO), 8.93 (dd, J = 8.1, 1.5 Hz, 1H, H-7), 8.77 (s, 1H, H-1′), 7.97 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.91 (d, J = 8.2 Hz, 1H, H-8′), 7.81 (d, J = 8.2 Hz, 1H, H-5′), 7.57 (dd, J = 8.1, 1.2 Hz, 1H, H-6′), 7.49–7.42 (m, 2H, H-6, H-7′), 7.33 (s, 1H, H-4′), 4.12 (s, 3H, OCH3), 2.76–2.67 (m, 4H, COCH2CH2), 2.30 (s, 6H, (CH3)2). 13C NMR (151 MHz, CDCl3) δ (ppm) 171.3 (NHCO), 161.6 (CO), 154.6 (C-3′), 150.0 (C-2′), 139.1 (C-8a), 135.9 (C-4a’), 135.3 (C-4a), 132.9 (C-1′), 128.7 (C-6′, C-8′), 128.4 (C-8a’), 127.3 (C-6), 126.9 (C-5′), 125.3 (C-7′), 123.7 (C-7), 121.9 (C-8), 121.1 (C-2′), 120.3 (C-5), 107.3 (C-4′), 56.3 (OCH3), 55.5 (COCH2CH2). 45.2 ((CH3)2), 35.3 (COCH2CH2); HRMS (ESI+) m/z 417.1929 (calcd for C24H25N4O3+, 417.1921).
3-(Cyclopropylamino)-N-(2-(2-methoxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (34). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 61%. Mp.: 187–189 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1666.56 (CO). 1H NMR (400 MHz, CDCl3) δ (ppm) 9.98 (brs, 1H, D2O exch., NHCO), 8.80 (dd, J = 8.0, 1.4 Hz, 1H, H-7), 8.38 (dd, J = 7.9, 1.8 Hz, 1H, H-6′), 7.87 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.48 (dd, J = 8.4, 1.8 Hz, 1H, H-4′), 7.36 (t, J = 8.0 Hz, 1H, H-6), 7.11 (d, J = 7.6 Hz, 1H, H-5′), 7.02 (dd, J = 8.5, 1.0 Hz, 1H, H-3′), 4.00 (s, 3H, OCH3), 3.14 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.71 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.17 (t, J = 6.8, Hz, 1H, CH-cyclopropyl), 0.40–0.33 (m, 4H, CH2-cyclopropyl). 13C NMR (101 MHz, CDCl3) δ (ppm) 171.1 (NHCO), 161.5 (CO), 157.9 (C-2′), 149.8 (C-2), 138.5 (C-8a), 134.4 (C-4a), 133.5 (C-4′), 131.3 (C-6′), 126.9 (C-6), 123.0 (C-7), 121.7 (C-5′), 120.7 (C-8), 120.1 (C-5), 119. (C-1′), 112.0 (C-3′), 56.2 (OCH3), 45.5 (COCH2CH2), 37.6 (COCH2CH2), 30.4 (CH-cyclopropyl), 6.3 (CH2-cyclopropyl); HRMS (ESI+) m/z 379.1759 (calcd for C21H23N4O3+, 379.1765).
3-(Cyclopropylamino)-N-(2-(3-methoxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (35). This compound was synthesized by an analogous procedure as described for the preparation of compound 17. Yield: 82%. Mp.: >230 °C (THF-n-Pentane). IR (Nujol) ν max/cm−1: 1664.59 (CO). 1H NMR (600 MHz, CDCl3) δ (ppm) 10.10 (brs, 1H, D2O exch., NHCO), 8.88 (m, 2H, H-7, H-1′), 7.96 (dd, J = 8.0, 1.4 Hz, H-5), 7.92 (d, J = 8.1 Hz, H-8′), 7.80 (d, J = 8.2 Hz, 1H, H-5′), 7.57 (td, J = 8.1, 1.2 Hz, 1H, H-6′), 7.51–7.44 (m, 2H, H-6, H-7′), 7.32 (s, 1H, H-4′), 4.13 (s, 3H, OCH3), 3.21 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.77 (t, J = 6.1 Hz, 2H, COCH2CH2), 2.20 (s, 1H, CH-cyclopropyl), 0.43–0.34 (m, 4H, CH2-cyclopropyl). 13C NMR (151 MHz, CDCl3) δ (ppm) 171.1 (NHCO), 161.5 (CO), 154.9 (C-3′), 150.0 (C-2′), 138.8 (C-8a), 136.0(C-4a’), 134.6 (C-4a), 132.9 (C-1′), 129.0 (C-6′), 128.9 (C-8′), 128.5 (C-8a’), 127.3 (C-6), 126.8 (C-5′), 125.3 (C-7′), 123.3 (C-7), 121.0 (C-8), 120.3 C-5, C-2′), 107.4 (C-4′), 56.4 (OCH3), 45.7 (COCH2CH2), 37.9 (COCH2CH2) 30.6 (CH-cyclopropyl), 6.3 (CH2-cyclopropyl); HRMS (ESI+) m/z 429.1915 (calcd for C25H25N4O3+, 429.1912).
3-(Dimethylamino)-N-(2-(2-hydroxyphenyl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (36). This compound was synthesized by an analogous procedure as described for the preparation of compound 21. Yield: 41%. Mp.: >230 °C (EtOAc). IR (Nujol) ν max/cm−1: 1665.17 (CO). 1H NMR (600 MHz, CDCl3-MeOD) δ (ppm) 8.10 (dd, J = 7.8, 1.4 Hz, 1H, H-7), 7.99 (dd, J = 8.1, 1.6 Hz, 1H, H-6′), 7.94 (dd, J = 8.0, 1.4 Hz, 1H, H-5), 7.40–7.44 (m, 2H, H-6, H-4′), 7.06 (dd, J = 8.3, 1.1 Hz, 1H, H-3′), 6.94 (d, J = 7.6 Hz, 1H, H-5′), 3.63 (t, J = 6.6 Hz, 2H, COCH2CH2)), 3.21 (t, J = 6.5 Hz, 2H, COCH2CH2), 3.04 (s, 6H, (CH3)2). 13C NMR (151 MHz, CDCl3-MeOD) δ (ppm) 170.2 (NHCO), 161.5 (CO), 162.3 (C-2′), 153.5 (C-2), 139.8 (C-8a), 135.2 (C-4′), 132.0 (C-4a), 130.0 (C-6′), 129.1 (C-5′), 127.8 (C-6), 124.3 (C-7), 121.6 (C-8a), 120.9 (C-5), 118.3 (C-3′), 114.3 (C-1′), 54.6 (COCH2CH2), 43.8 ((CH3)2), 31.1 (COCH2CH2); HRMS (ESI+) m/z 353.1617 (calcd for C19H19N4O3+, 353.1608).
3-(Dimethylamino)-N-(2-(3-hydroxynaphthalen-2-yl)-4-oxo-3,4-dihydroquinazolin-8-yl)propanamide (37). This compound was synthesized by an analogous procedure as described for the preparation of compound 21. Yield: 42%. Mp.: 214–216 °C (EtOAc). IR (Nujol) ν max/cm−1: 1664.10 (CO). 1H NMR (600 MHz, CDCl3-MeOD) δ (ppm) 8.72 (s, 1H, H-1′), 8.31 (d, J = 7.9 Hz, 1H, H-7), 7.94 (d, J = 7.9 Hz, 1H, H-8′), 7.89 (d, J = 8.2 Hz, 1H, H-5′), 7.68 (d, J = 8.3 Hz, 1H, H-5), 7.49 (dd, J = 8.2, 1.2 Hz, 1H, H-6′), 7.38–7.31 (m, 2H, H-6, H-7′), 7.31 (s, 1H, H-4′), 3.34 (m, 2H, COCH2CH2), 3.08 (t, J = 6.5 Hz, 2H, COCH2CH2), 2.83 (s, 6H, (CH3)2). 13C NMR (151 MHz, CDCl3-MeOD) δ (ppm) 170.2 (NHCO), 162.7 (CO), 156.8 (C-3′), 152.3 (C-2), 138.7 (C-8a), 135.7 (C-4a’), 133.2 (C-4a), 130.4 (C-1′), 128.9 (C-6′), 128.5 (C-8′), 127.6 (C-8a’), 126.6 (C-6, C-5′), 125.8 (C-7′), 122.3 (C-7), 122.1 (C-8), 122.1 (C-2′), 121.0 (C-5), 111.5 (C-4′), 54.4 (COCH2CH2). 43.7 ((CH3)2), 32.2 (COCH2CH2); HRMS (ESI+) m/z 403.1772 (calcd for C23H23N4O3+, 403.1765).

3.3. Biological Assays and Experiments

Cell Viability MTS Assays

The human cancer cell lines Capan-1, HCT-116, NCI-H460, LN-229, HL-60, K-562, and Z-138 were acquired from the American Type Culture Collection (ATCC, Manassas, VA, USA), while the DND-41 cell line was purchased from the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ Leibniz-Institut, Braunschweig, Germany) and the Hap-1 cell line which was ordered from Horizon Discovery (Horizon Discovery Group, UK). All cell lines were cultured as recommended by the suppliers. Culture media were purchased from Gibco (Gibco Life Technologies, Merelbeke, Belgium) and supplemented with 10% fetal bovine serum (HyClone, Cytiva, MA, USA).
Adherent cell lines were seeded at a density between 500 and 1500 cells per well in 384-well plates (Greiner). After overnight incubation, cells were treated with seven different concentrations of the test compounds, ranging from 100 to 0.006 µM. Docetaxel and Staurosporine were included as positive controls to validate the assay conditions. Untreated cell lines (i.e., without compound treatment) were used as negative controls.
Suspension cell lines were seeded at densities ranging from 2500 to 5000 cells per well in 384-well culture plates containing the test compounds at the same concentration points. Cells were incubated for 72 h with compounds and were then analyzed using the CellTiter 96® AQueous One Solution Cell Proliferation Assay (MTS) reagent (Promega) according to the manufacturer’s instructions. Absorbance of the samples was measured at 490 nm using a SpectraMax Plus 384 (Molecular Devices), and OD values were used to calculate the 50% inhibitory concentration (IC50). Compounds were tested in two independent experiments [27].
Differential scanning fluorimetry (DSF). Thermal melting experiments were measured using an Mx3005p Real Time PCR machine (Stratagene). Proteins were buffered in 10 mM HEPES pH 7.5 and 500 mM NaCl and run in a 96-well plate at a final concentration of 2 μM in 20 μL volume. Compounds were added at a final concentration of 10 μM from stock solutions in DMSO. SYPRO Orange was used as a fluorescence probe at a dilution of 1:1000. Excitation and emission filters for the SYPRO Orange dye were set to 465 nm and 590 nm, respectively. The temperature was raised with a step of 3 °C per minute from 25 °C to 96 °C and fluorescence readings were taken at each interval. Data collection was made in triplicates. For the data analysis, the baselines of the denatured and native states were approximated by a linear fit. The observed temperature shifts, ΔTm, were recorded as the difference between the transition midpoints of sample and reference wells containing the protein without the ligand in the same plate and determined by non-linear least squares fit [28].

4. Conclusions

A set of 16 newly synthesized 2-aryl-substituted quinazolinones were assessed for their anti-proliferative effects against nine different tumor cell lines. Most of the compounds exhibited moderate to good anti-proliferative activity, with the most promising outcomes observed in the case of phenyl-substituted analogs. All the naphthyl-substituted compounds were found to be inactive against the tested cell lines. In contrast, only the 2-methoxyphenyl-substituted compounds demonstrated interesting activity. Notably, compound 17 exhibited the highest potency, displaying significant efficacy against the Capan-1, HCT-116, Hap-1, NCI-H460, Z-138, and DND-41 cell lines. Considering its potential clinical relevance and therapeutic utility, compound 17 could be further optimized. While the precise mechanism underlying its tumor-suppressing function remains unclear, our results suggest that in the case of the 2-phenyl analog, the side chain significantly enhances its anti-cancer activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28237912/s1, DSF, 1H-NMR and 13C-NMR are available online.

Author Contributions

All authors contributed to the writing and gave approval to the final version of the manuscript. M.K., D.K., A.D.K. and I.K.K. performed the chemical synthesis experiments, analyzed the results, and wrote the manuscript. L.P., D.S. and S.D.J. designed the biological experiments, performed cell viability assays, analyzed the results, and wrote the manuscript. A.K. and S.K. designed the biological experiments, analyzed the results, and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

S.K. and A.K. are grateful for support from the Structural Genomics Consortium (SGC), a registered charity (no: 1097737) that receives funds from Bayer AG, Boehringer Ingelheim, Bristol Myers Squibb, Genentech, Genome Canada through Ontario Genomics Institute, EU/EFPIA/OICR/McGill/KTH/Diamond Innovative Medicines Initiative 2 Joint Undertaking [EUbOPEN grant 875510], Janssen, Merck KGaA, Pfizer, and Takeda, and by the German Cancer Research Center DKTK and the Frankfurt Cancer Institute (FCI).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Data are contained within the article and supplementary materials.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Wdowiak, P.; Matysiak, J.; Kuszta, P.; Czarnek, K.; Niezabitowska, E.; Baj, T. Quinazoline Derivatives as Potential Therapeutic Agents in Urinary Bladder Cancer Therapy. Front. Chem. 2021, 9, 765552. [Google Scholar] [CrossRef] [PubMed]
  2. Shagufta; Ahmad, I. An Insight into the Therapeutic Potential of Quinazoline Derivatives as Anticancer Agents. Med. Chem. Commun. 2017, 8, 871–885. [Google Scholar] [CrossRef] [PubMed]
  3. Zayed, M.F. Medicinal Chemistry of Quinazolines as Anticancer Agents Targeting Tyrosine Kinases. Sci. Pharm. 2023, 91, 18. [Google Scholar] [CrossRef]
  4. Murthy Boddapati, S.N.; Babu Bollikolla, H.; Geetha Bhavani, K.; Singh Saini, H.; Ramesh, N.; Babu Jonnalagadda, S. Advances in Synthesis and Biological Activities of Quinazoline Scaffold Analogues: A Review. Arab. J. Chem. 2023, 16, 105190. [Google Scholar] [CrossRef]
  5. Auti, P.S.; George, G.; Paul, A.T. Recent Advances in the Pharmacological Diversification of Quinazoline/Quinazolinone Hybrids. RSC Adv. 2020, 10, 41353–41392. [Google Scholar] [CrossRef] [PubMed]
  6. Alsibaee, A.M.; Al-Yousef, H.M.; Al-Salem, H.S. Quinazolinones, the Winning Horse in Drug Discovery. Molecules 2023, 28, 978. [Google Scholar] [CrossRef] [PubMed]
  7. Asif, M. Chemical Characteristics, Synthetic Methods, and Biological Potential of Quinazoline and Quinazolinone Derivatives. Int. J. Med. Chem. 2014, 2014, e395637. [Google Scholar] [CrossRef]
  8. Khodair, A.I.; Alsafi, M.A.; Nafie, M.S. Synthesis, Molecular Modeling and Anti-Cancer Evaluation of a Series of Quinazoline Derivatives. Carbohydr. Res. 2019, 486, 107832. [Google Scholar] [CrossRef]
  9. Ahmed, M.F.; Magdy, N. Design and Synthesis of 4-Substituted Quinazolines as Potent EGFR Inhibitors with Anti-Breast Cancer Activity. Anti-Cancer Agents Med. Chem. 2017, 17, 832–838. [Google Scholar] [CrossRef]
  10. Dwivedi, A.R.; Kumar, V.; Prashar, V.; Verma, A.; Kumar, N.; Parkash, J.; Kumar, V. Morpholine Substituted Quinazoline Derivatives as Anticancer Agents against MCF-7, A549 and SHSY-5Y Cancer Cell Lines and Mechanistic Studies. RSC Med. Chem. 2022, 13, 599–609. [Google Scholar] [CrossRef]
  11. Li, Y.; Xiao, J.; Zhang, Q.; Yu, W.; Liu, M.; Guo, Y.; He, J.; Liu, Y. The Association between Anti-Tumor Potency and Structure-Activity of Protein-Kinases Inhibitors Based on Quinazoline Molecular Skeleton. Bioorg. Med. Chem. 2019, 27, 568–577. [Google Scholar] [CrossRef]
  12. Jin, H.; Dan, H.-G.; Rao, G.-W. Research Progress in Quinazoline Derivatives as Multi-Target Tyrosine Kinase Inhibitors. Heterocycl. Commun. 2018, 24, 1–10. [Google Scholar] [CrossRef]
  13. Kumar, P.; Mangla, B.; Javed, S.; Ahsan, W.; Musyuni, P.; Ahsan, A.; Aggarwal, G. Gefitinib: An Updated Review of Its Role in the Cancer Management, Its Nanotechnological Interventions, Recent Patents and Clinical Trials. Recent Pat. Anti-Cancer Drug Discov. 2023, 18, 448–469. [Google Scholar] [CrossRef] [PubMed]
  14. Al-Ghusn, A.I.; Bakheit, A.H.; Attwa, M.W.; AlRabiah, H. Chapter Four—Vandetanib. In Profiles of Drug Substances, Excipients and Related Methodology; Al-Majed, A.A., Ed.; Academic Press: Cambridge, MA, USA, 2023; Volume 48, pp. 109–134. [Google Scholar]
  15. Gerson, S.L.; Caimi, P.F.; William, B.M.; Creger, R.J. Chapter 57—Pharmacology and Molecular Mechanisms of Antineoplastic Agents for Hematologic Malignancies. In Hematology, 7th ed.; Hoffman, R., Benz, E.J., Silberstein, L.E., Heslop, H.E., Weitz, J.I., Anastasi, J., Salama, M.E., Abutalib, S.A., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 849–912. ISBN 978-0-323-35762-3. [Google Scholar]
  16. Awan, F.T.; Kharfan-Dabaja, M.A. Chapter 12—Hematopoietic Cell Transplantation for Chronic Lymphocytic Leukemia. In Hematopoietic Cell Transplantation for Malignant Conditions; Bashir, Q., Hamadani, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 185–190. ISBN 978-0-323-56802-9. [Google Scholar]
  17. Zahran, S.S.; Ragab, F.A.; El-Gazzar, M.G.; Soliman, A.M.; Mahmoud, W.R.; Ghorab, M.M. Antiproliferative, Antiangiogenic and Apoptotic Effect of New Hybrids of Quinazoline-4(3H)-Ones and Sulfachloropyridazine. Eur. J. Med. Chem. 2023, 245, 114912. [Google Scholar] [CrossRef] [PubMed]
  18. Alsantali, R.I. Design, Synthesis, and Anticancer Activity of New Quinazoline Derivatives Containing Acetylhydrazide Moiety as EGFR Inhibitors and Apoptosis Inducers. Russ. J. Bioorg. Chem. 2023, 49, 645–654. [Google Scholar] [CrossRef]
  19. Alagarsamy, V.; Chitra, K.; Saravanan, G.; Solomon, V.R.; Sulthana, M.T.; Narendhar, B. An Overview of Quinazolines: Pharmacological Significance and Recent Developments. Eur. J. Med. Chem. 2018, 151, 628–685. [Google Scholar] [CrossRef]
  20. Khadka, D.B.; Tran, G.H.; Shin, S.; Nguyen, H.T.M.; Cao, H.T.; Zhao, C.; Jin, Y.; Van, H.T.M.; Chau, M.V.; Kwon, Y.; et al. Substituted 2-Arylquinazolinones: Design, Synthesis, and Evaluation of Cytotoxicity and Inhibition of Topoisomerases. Eur. J. Med. Chem. 2015, 103, 69–79. [Google Scholar] [CrossRef]
  21. Amrhein, J.A.; Beyett, T.S.; Feng, W.W.; Krämer, A.; Weckesser, J.; Schaeffner, I.K.; Rana, J.K.; Jänne, P.A.; Eck, M.J.; Knapp, S.; et al. Macrocyclization of Quinazoline-Based EGFR Inhibitors Leads to Exclusive Mutant Selectivity for EGFR L858R and Del19. J. Med. Chem. 2022, 65, 15679–15697. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, P.-F.; Jensen, A.A.; Bunch, L. From Methaqualone and Beyond: Structure–Activity Relationship of 6-, 7-, and 8-Substituted 2,3-Diphenyl-Quinazolin-4(3H)-Ones and in Silico Prediction of Putative Binding Modes of Quinazolin-4(3H)-Ones as Positive Allosteric Modulators of GABAA Receptors. ACS Chem. Neurosci. 2020, 11, 4362–4375. [Google Scholar] [CrossRef]
  23. Ye, Z.; Gao, Y.; Bakshi, R.K.; Chen, M.-H.; Rohrer, S.P.; Feighner, S.D.; Pong, S.-S.; Howard, A.D.; Blake, A.; Birzin, E.T.; et al. Modeling Directed Design and Biological Evaluation of Quinazolinones as Non-Peptidic Growth Hormone Secretagogues. Bioorg. Med. Chem. Lett. 2000, 10, 5–8. [Google Scholar] [CrossRef]
  24. Nathubhai, A.; Haikarainen, T.; Hayward, P.C.; Muñoz-Descalzo, S.; Thompson, A.S.; Lloyd, M.D.; Lehtiö, L.; Threadgill, M.D. Structure-Activity Relationships of 2-Arylquinazolin-4-Ones as Highly Selective and Potent Inhibitors of the Tankyrases. Eur. J. Med. Chem. 2016, 118, 316–327. [Google Scholar] [CrossRef]
  25. Georgakopoulos, A.; Kalampaliki, A.D.; Gioti, K.; Hamdoun, S.; Giannopoulou, A.F.; Efferth, T.; Stravopodis, D.J.; Tenta, R.; Marakos, P.; Pouli, N.; et al. Synthesis of Novel Xanthone and Acridone Carboxamides with Potent Antiproliferative Activities. Arab. J. Chem. 2020, 13, 7953–7969. [Google Scholar] [CrossRef]
  26. Panteleon, V.; Kostakis, I.K.; Marakos, P.; Pouli, N.; Andreadou, I. Synthesis of Some New Spiropyranoquinolines and Evaluation of Their Free Radical Scavenging Activity. Chem. Pharm. Bull. 2009, 57, 446–452. [Google Scholar] [CrossRef] [PubMed]
  27. Van de Walle, T.; Theppawong, A.; Grootaert, C.; De Jonghe, S.; Persoons, L.; Daelemans, D.; Van Hecke, K.; Van Camp, J.; D’hooghe, M. Synthesis and Cytotoxic Evaluation of Monocarbonyl Curcuminoids and Their Pyrazoline Derivatives. Monatsh. Chem. 2019, 150, 2045–2051. [Google Scholar] [CrossRef]
  28. Niesen, F.H.; Berglund, H.; Vedadi, M. The Use of Differential Scanning Fluorimetry to Detect Ligand Interactions That Promote Protein Stability. Nat. Protoc. 2007, 2, 2212–2221. [Google Scholar] [CrossRef]
Figure 1. Structures of Gefitinib, Vandetanib, Idelalisib, and other known quinazoline kinase inhibitors (IIII).
Figure 1. Structures of Gefitinib, Vandetanib, Idelalisib, and other known quinazoline kinase inhibitors (IIII).
Molecules 28 07912 g001
Scheme 1. Reagents and conditions: (a) ACN, Et3N, ClCO2Et, 30 min, 0 °C/2-amino-3-nitrobenzoic acid, Na2CO3, 50 °C, 24 h; (b) (CH3CO)2O, reflux, 2 h; (c) NH3, THF, 12 h; (d) 5% aq. NaOH sol., reflux, 10 min; (e) EtOH abs., H2, Pd/C, 50 psi, 4 h; (f) 13–14: ClCOCH2Cl, Na2CO3, THF, rt, 15 min; 15–16: ClCOCH2CH2Cl, Na2CO3, THF, rt, 15 min.
Scheme 1. Reagents and conditions: (a) ACN, Et3N, ClCO2Et, 30 min, 0 °C/2-amino-3-nitrobenzoic acid, Na2CO3, 50 °C, 24 h; (b) (CH3CO)2O, reflux, 2 h; (c) NH3, THF, 12 h; (d) 5% aq. NaOH sol., reflux, 10 min; (e) EtOH abs., H2, Pd/C, 50 psi, 4 h; (f) 13–14: ClCOCH2Cl, Na2CO3, THF, rt, 15 min; 15–16: ClCOCH2CH2Cl, Na2CO3, THF, rt, 15 min.
Molecules 28 07912 sch001
Scheme 2. Reagents and conditions: (a) suitable amine, THF anh., autoclave, 100 °C, 65 h; (b) BBr3, CH2Cl2, −40 °C, 10 min, then 0 °C, 24 h; (c) DMF, CH3COOK, 50 °C, 2 h; (d) MeOH, 30% aq. NaOH sol., rt, 2 h; (e) MeOH, CH3COOK, rt, 18 h.
Scheme 2. Reagents and conditions: (a) suitable amine, THF anh., autoclave, 100 °C, 65 h; (b) BBr3, CH2Cl2, −40 °C, 10 min, then 0 °C, 24 h; (c) DMF, CH3COOK, 50 °C, 2 h; (d) MeOH, 30% aq. NaOH sol., rt, 2 h; (e) MeOH, CH3COOK, rt, 18 h.
Molecules 28 07912 sch002
Scheme 3. Reagents and conditions: (a) Et3N, THF anh., 70 °C, 18 h; (b) OsO4, N-methylmorpholine-N-oxide, THF anh., rt, 60 h; (c) suitable amine, THF anh., autoclave, 100 °C, 65 h; (d) BBr3, CH2Cl2, −40 °C, 10 min, then 0 °C, 24 h.
Scheme 3. Reagents and conditions: (a) Et3N, THF anh., 70 °C, 18 h; (b) OsO4, N-methylmorpholine-N-oxide, THF anh., rt, 60 h; (c) suitable amine, THF anh., autoclave, 100 °C, 65 h; (d) BBr3, CH2Cl2, −40 °C, 10 min, then 0 °C, 24 h.
Molecules 28 07912 sch003
Table 1. Accumulative results of the anti-proliferative activities for all the synthesized compounds.
Table 1. Accumulative results of the anti-proliferative activities for all the synthesized compounds.
CompIC50 (µM)
LN-229Capan-1Hap-1HCT-116NCI-H460DND-41HL-60K-562Z-138
GlioblastomaPancreatic AdenocarcinomaChronic Myeloid LeukemiaColorectal CarcinomaLung CarcinomaAcute Lymphoblastic LeukemiaAcute Myeloid LeukemiaChronic Myeloid LeukemiaNon-Hodgkin Lymphoma
1742.61.82.32.21.45.510.612.72.6
18>100>100>100>10057.1>10098.5>100>100
19>100>100>100>100>100>100>100>100>100
20>100>100>100>100>100>100>100>100>100
2121.845.534.473.1>10051.253.852.437.7
22>100>10051.3>100>100>100>100>100>100
2532.937.039.29.549.851.848.942.241.9
26>100>10051.3>100>100>100>100>100>100
30>100>10051.3>100>100>100>100>100>100
31>100>10051.3>100>100>100>100>100>100
3246.735.348.242.027.637.655.045.215.3
33>10078.770.149.1>100>100>100>10051.7
3466.49.911.313.132.510.89.654.011.8
35>100>10051.3>100>100>100>100>100>100
3673.6>100>10040.8>10095.3>100>10060.1
3767.4>10073.863.7>100>100>100>100>100
Docetaxel (nM)2.84.92.02.32.82.910.519.42.0
Staurosporine (nM)44.744.437.463.754.559.758.350.548.8
Table 2. DSF results on 17 most selected kinases, measured as ΔTm (°C).
Table 2. DSF results on 17 most selected kinases, measured as ΔTm (°C).
AAK1CAMKK2CK2A2CLK1DYRK2HIPK2GAKDAPK3DYRK1AGSG2MST3GSK3BMAP3K5PIM3BMP2KBMPR2MEK5
175.25.21.84.62.31.14.72.04.02.60.65.12.73.29.03.25.3
214.34.33.93.92.50.83.92.13.21.50.74.71.76.57.02.62.8
192.22.21.03.11.41.22.44.15.00.83.02.42.03.03.31.85.2
342.62.61.63.21.60.72.01.42.73.10.42.40.32.54.51.43.3
200.70.70.92.11.10.51.00.92.2−0.40.34.10.31.01.31.51.5
351.41.40.72.41.40.61.41.22.40.40.22.60.23.52.50.92.2
182.12.11.33.41.50.62.01.32.3−0.1−0.13.90.71.24.82.12.8
330.90.90.92.11.10.61.32.02.00.30.42.10.32.91.80.41.7
303.93.91.84.63.72.02.82.12.83.10.83.05.13.07.02.86.1
321.71.71.42.51.10.51.51.62.03.00.01.50.31.92.80.62.2
310.70.71.22.51.00.50.60.51.40.30.10.80.81.21.30.82.5
253.33.31.34.23.82.32.81.75.32.90.23.71.92.75.02.86.5
330.10.10.90.00.30.6−0.20.4−0.40.50.30.90.02.70.00.21.5
360.20.22.00.30.40.30.30.21.20.8−0.10.5−0.22.80.80.30.6
Staurosporine13.023.82.811.97.04.68.916.312.88.99.111.216.919.618.32.513.4
Silmitasertib 15.7
GW779439X 11.6
GSK626616 6.6
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Karelou, M.; Kampasis, D.; Kalampaliki, A.D.; Persoons, L.; Krämer, A.; Schols, D.; Knapp, S.; De Jonghe, S.; Kostakis, I.K. Synthesis and Biological Evaluation of 2-Substituted Quinazolin-4(3H)-Ones with Antiproliferative Activities. Molecules 2023, 28, 7912. https://doi.org/10.3390/molecules28237912

AMA Style

Karelou M, Kampasis D, Kalampaliki AD, Persoons L, Krämer A, Schols D, Knapp S, De Jonghe S, Kostakis IK. Synthesis and Biological Evaluation of 2-Substituted Quinazolin-4(3H)-Ones with Antiproliferative Activities. Molecules. 2023; 28(23):7912. https://doi.org/10.3390/molecules28237912

Chicago/Turabian Style

Karelou, Maria, Dionysis Kampasis, Amalia D. Kalampaliki, Leentje Persoons, Andreas Krämer, Dominique Schols, Stefan Knapp, Steven De Jonghe, and Ioannis K. Kostakis. 2023. "Synthesis and Biological Evaluation of 2-Substituted Quinazolin-4(3H)-Ones with Antiproliferative Activities" Molecules 28, no. 23: 7912. https://doi.org/10.3390/molecules28237912

Article Metrics

Back to TopTop