Next Article in Journal
The Modification of Substrate in the Soilless Cultivation of Raspberries (Rubus Idaeus L.) as a Factor Stimulating the Biosynthesis of Selected Bioactive Compounds in Fruits
Next Article in Special Issue
Synthesis and Biological Evaluation of 3-Amino-4,4-Dimethyl Lithocholic Acid Derivatives as Novel, Selective, and Cellularly Active Allosteric SHP1 Activators
Previous Article in Journal
Ameliorative Effect of Citrus Lemon Peel Extract and Resveratrol on Premature Ovarian Failure Rat Model: Role of iNOS/Caspase-3 Pathway
Previous Article in Special Issue
Lysinated Multiwalled Carbon Nanotubes with Carbohydrate Ligands as an Effective Nanocarrier for Targeted Doxorubicin Delivery to Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Cytotoxic Evaluation, and Structure-Activity Relationship of Substituted Quinazolinones as Cyclin-Dependent Kinase 9 Inhibitors

by
Hamad M. Alkahtani
1,*,
Amer Alhaj Zen
2,
Ahmad J. Obaidullah
1,
Mohammed M. Alanazi
1,
Abdulrahman A. Almehizia
1,
Siddique Akber Ansari
1,
Fadilah Sfouq Aleanizy
3,
Fulwah Yahya Alqahtani
3,
Rana M. Aldossari
4,
Raghad Abdullah Algamdi
1,
Lamees S. Al-Rasheed
1,
Sami G. Abdel-Hamided
5,
Alaa A.-M. Abdel-Aziz
1 and
Adel S. El-Azab
1,*
1
Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
2
Chemistry & Forensics Department, Clifton Campus, Nottingham Trent University, Nottingham Ng11 8NS, UK
3
Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
4
Department of Pharmacology & Toxicology, College of Pharmacy, 11 Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
5
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(1), 120; https://doi.org/10.3390/molecules28010120
Submission received: 10 November 2022 / Revised: 4 December 2022 / Accepted: 6 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Anticancer Agents: Design, Synthesis and Evaluation III)

Abstract

:
Cyclin-dependent kinase 9 (CDK9) plays a critical role in transcriptional elongation, through which short-lived antiapoptotic proteins are overexpressed and make cancer cells resistant to apoptosis. Therefore, CDK9 inhibition depletes antiapoptotic proteins, which in turn leads to the reinstatement of apoptosis in cancer cells. Twenty-seven compounds were synthesized, and their CDK9 inhibitory and cytotoxic activities were evaluated. Compounds 7, 9, and 25 were the most potent CDK9 inhibitors, with IC50 values of 0.115, 0.131, and 0.142 μM, respectively. The binding modes of these molecules were studied via molecular docking, which shows that they occupy the adenosine triphosphate binding site of CDK9. Of these three molecules, compound 25 shows good drug-like properties, as it does not violate Lipinski’s rule of five. In addition, this molecule shows promising ligand and lipophilic efficiency values and is an ideal candidate for further optimization.

1. Introduction

Cyclin-dependent kinases (CDKs), a family of Ser/Thr PKs belonging to the CMGC [Cyclin-dependent kinases (CDKs), Mitogen-activated protein kinases (MAPKs), Glycogen synthase kinases (GSKs), and Cdc2-like kinases (CLKs)] superfamily, play an essential role in controlling and regulating the cell cycle and the transcription of genes in eukaryotes [1,2,3,4,5,6]. In humans, 20 different CDKs (1–20) have been discovered and can be divided into two main categories according to their primary function. CDK1-4 and CDK6 regulate cell cycle phases [3,4,5,6,7], whereas CDK7-9 and CDK11 promote transcription for cell growth, differentiation, and viral pathogenesis [3,4,5,6,8,9]. CDK9 is overexpressed in various solid and hematological malignancies, where CDK9 is the principal regulator that stimulates transcriptional elongation, through which short-lived antiapoptotic proteins (e.g., Mcl-1) are overexpressed, leading to cancer cells becoming resistant to apoptosis [9]. Pharmacological inhibition of CDK9 induces apoptosis in cancer cells, making it a potential drug target in oncology [10,11,12,13,14,15]. It is expected that selective CDK9 inhibitors will lead to an improvement in the toxicity profile of currently available CDK9 inhibitors [16]. Moreover, unlike other transcriptional CDKs, such as CDK7 and CDK8, CDK9 has been validated as a druggable target for treating other diseases, such as cardiac hypertrophy and HIV infection [9,17]. Several CDK9 inhibitors have been synthesized, with many entering clinical trials. The vast majority of these small molecules are potent competitive inhibitors. In addition, they are effective against hematological malignancies as well as solid tumors such as breast cancer [11,18,19,20,21]. These include, derivatives of flavonoids (flavopiridol, I), thiazoles (SNS-032, II), pyrimidines (CDKI-73, III), pyridines (AZD4573, IV), and benzimidazoles (5,6-dichlorobenzimidazole 1-beta-D-ribofuranoside (BDR), V) [11,14,15,18,19,22]. Quinazoline derivatives are known for their wide range of therapeutic activities, such as anticancer, antiviral, antihypertensive, and antidiabetic [23,24,25]. This wide range of therapeutic activities exhibited by quinazoline derivatives is mediated by inhibiting several molecular targets, such as protein kinases, carbonic anhydrases, antitumor and cyclooxygenases (COX) [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42]. In addition, they exhibit promising CDK9 inhibitory activity (such as compound VI and VII) that warrants further investigation and improvement (Figure 1) [43,44].
As a continuation of our studies on quinazoline derivatives (VI) as promising CDK9 inhibitors that exhibit cytotoxic activity [45], several quinazolin-4-ones linked to substituted anilides and 4-sulfamoylphenethyl were designed, as shown in Figure 2. In addition, the introduction of different substitutions at position 6 of the quinazolin-4-one ring (VII) was performed to improve CDK9 inhibitory activity via possible interaction with the gatekeeper region of CDK9. It was expected that such modifications would improve the antiproliferative activity of these molecules against cancer cells by increasing their affinity for the CDK9 enzyme. Molecular docking studies were carried out on this new series of compounds to explore the structural requirements for inhibitory activity toward CDK9.

2. Results and Discussion

2.1. Chemistry

2-Mercapto-3-(4-sulfamoylphenethyl)quinazolin-4-one (1) was prepared, as reported previously [46], in a yield of 95%. Compounds 216 were prepared in excellent yields (>90%) by treating compound 1 with 2-chloro-N-substituted-amide and potassium carbonate at room temperature in acetone, as reported previously in the literature, the reaction scheme of which is shown in Scheme 1 [26].
Compounds 1720 and 29 were prepared as reported previously (Figure 3) [31,47,48,49]. 2-(3-Bromobenzamido)-5-methylbenzoic acid (23) was prepared in 97% yield via the reaction of 5-methylanthranilic acid (21) with 3-bromobenzoyl chloride (22) in pyridine at room temperature (Scheme 2). Compound 23 underwent hot cyclization in acetic anhydride to give compound 24 in 95% yield.
Multiple spectroscopic techniques were used to confirm the structures of the target compounds 2328. The chemical structure of compound 23 was established by the presence of the carboxylic (COOH), amide (CONH), and methyl (CH3) peaks at 12.06, 8.51, and 2.33 ppm in 1H NMR and 170.52, 163.37 and 20.73 ppm in 13C NMR, respectively. 2-(3-Bromophenyl)-6-methyl-4H-benzo[d][1,3]oxazin-4-one (24) was confirmed by the disappearance of the carboxylic and amide peaks of compound 23 in NMR spectra. In addition to the occurrence of a new carbonyl group due to 4H-benzo[d][1,3]oxazin-4-one at 159.02 ppm in 13C NMR. Boiling compound 24 in formamide gave 2-(3-bromophenyl)-6-methylquinazolin-4(3H)-one (25) in 80% yield. Its formation was confirmed by the disappearance of the carbonyl group of benzo[d][1,3]oxazin-4-one (24) at 159.02 ppm and the appearance of a new carbonyl group peak due to the quinazoline nucleus at 165.30 ppm in 13C NMR spectrum. 2-(3-Bromophenyl)-6-methylquinazoline-4(3H)-thione (26) was prepared in a yield of 68% by boiling compound 25 with phosphorus pentasulfide (P2S5) in toluene. Its successful formation was confirmed by singlet peaks related to the thioamide (CSNH) group at 13.94 ppm in 1H NMR and 187.62 ppm in 13C NMR spectra, respectively. Furthermore, 3-amino-2-(3-bromophenyl)-6-methylquinazolin-4(3H)-one (27) was obtained in a yield of 87% by heating compound 24 with hydrazine hydrate in ethanol, while 2-(3-bromophenyl)-3-hydroxy-6-methylquinazolin-4(3H)-one (28) was obtained in a yield of 84% by heating compound 24 with hydroxylamine hydrochloride in dry pyridine. Compounds 27 and 28 were identified by the presence of new characteristic amine (NH2) and hydroxyl (OH) peaks at 5.66 and 11.73 ppm, respectively, in 1H NMR spectra, as shown in Scheme 2.

2.2. Structure-Activity Relationship (SAR) Analysis

The CDK9 inhibition activities of the target compounds were evaluated, with flavopiridol used as a reference. Compound 1 has a similar scaffold to those of several reported CDK9 inhibitors and shows promising CDK9 inhibitory activity with an IC50 value of 0.644 μM [39]. Therefore, it was selected as a hit compound and modified with different substituents at the quinazoline position 2 to improve its inhibition properties. The results show that introducing acetamide and acetanilide groups at position 2 in compounds 2 and 3 showed better inhibition than the hit compound, with IC50 values of 0.454 and 0.421 μM, respectively, as shown in Table 1. To investigate the effects that other functional groups substituted at the p-position of the acetanilide ring have on improving the potency of the compound, analogs 48 with various substituents, such as methyl, acetyl, methoxy, and ethoxy groups, were prepared and evaluated. Compound 4, with a p-methyl group, displays lower inhibitory activity than compounds (13), with an IC50 value of 0.788 μM, as shown in Table 1.
Furthermore, substituting the p-methyl group for a larger group, such as an acetyl group used to generate compound 5, was found to be detrimental as it was less potent than the previous analogs, showing an IC50 value of 0.829 μM, as shown in Table 1. However, introducing a methoxy group at the p-position gave compound 6, which led to a recovery in the inhibitory activity, exhibiting an IC50 value of 0.463 μM. Interestingly, when an ethoxy group was introduced, as in compound 7, the inhibitory activity increased by around 6-fold compared to the hit compound 1, with the compound exhibiting an IC50 value of 0.115 μM. However, replacing the p-methoxy group with 3,4,5-trimethoxy groups in the acetanilide ring led to compound 8, which demonstrated less potency than the hit compound, exhibiting an IC50 value of 0.501 μM. Interestingly, introducing a halogen (such as Br, Cl, or F) at the p-position of acetanilide led to the generation of compounds 911, which show excellent inhibition of CDK9.
Furthermore, introducing bromine at this position, as in the formation of compound 9, seemed to be more favorable to the inhibitory activity compared with the introduction of other halogens at the same position, with compound 9 showing an IC50 value of 0.131 μM. Analogously, increasing the lipophilicity of the benzylacetamide substituents in compounds 1214 could be essential for improving their CDK9 inhibitory activity compared to their corresponding acetanilide-containing counterparts, such as compounds 6 and 11. It was observed that the phenylpropanamide substituents in compounds 15 and 16, which exhibit IC50 values of 0.444 and 0.350 μM, respectively, were not conducive to improving their CDK9 inhibitory activity when compared to the corresponding phenylacetanilide substituents in compounds 3 and 10, which have IC50 values of 0.421 and 0.193 μM. Among the investigated compounds, compounds 7 and 9 showed the best CDK9 inhibition activities, with IC50 values of 0.115 and 0.131 μM, respectively.
Compounds 1720 and 2329 were synthesized to investigate their SAR further (Table 2). Compounds 1720 are 4-quinazolinone derivatives with different substituents at 2 and 6 positions. Keeping position 2 unsubstituted while introducing iodine at position 6 resulted in the formation of compound 17, which showed sub-micromolar activity against CDK9, exhibiting an IC50 value of 0.639 μM. Keeping the iodine at position 6 while introducing a thiophene ring at position 2, as in compound 18, led to a doubling of the potency of the compound, with it exhibiting an IC50 value of 0.296 μM. Introducing p-tolyl and chlorine groups at positions 2 and 6, respectively, leading to the formation of compound 19, did not lead to any significant improvement in the CDK9 inhibitory activity, with the compound showing an IC50 value of 0.282 μM. However, replacing the p-tolyl group at position 2 with a p-chlorophenyl group and the chlorine with a nitro group, which is firmly electron withdrawing, led to the formation of compound 20, which exhibited a 1-fold reduction in potency against CDK9. Five bromophenyl derivatives with a methyl group at position 6 were synthesized to further explore the compounds’ SAR. The quinazolin-4-one derivative (25), with a (3-bromophenyl) moiety at position 2, showed significant improvement in CDK9 inhibitory activity, whereas the quinazoline-4-thione analog 26 exhibited a 2-fold reduction in inhibitory activity, with IC50 values of 0.142 and 0.289 μM, respectively. Replacing 4-quinazolinone with benzo[d][1,3]oxazin-4-one, as in compound 24, led to a 3-fold reduction in CDK9 inhibitory activity compared with compound 25. Interestingly, intermediate 23, used in the synthesis of compound 24, was tested and shown to be more potent, with IC50 values of 0.210 and 0.486 μM, respectively. N-hydroxy derivative 28 was shown to be a less potent inhibitor of CDK9 when compared to compound 25, showing IC50 values of 0.210 and 0.142 μM, respectively. Compound 27, the N-amino analog of compound 25, showed an approximately 6-fold decrease in potency against CDK9. A quinazoline-2,4-dione analog, compound 29, was also tested and exhibited inhibitory activity against CDK9, with an IC50 value of 0.589 μM.
Breast cancer often occurs with dysregulation in CDK9 levels, and several studies have shown the efficacy of CDK9 inhibitors in breast cancer [20,21]. Therefore, the antiproliferative activity of compounds 120 and 23-29 was evaluated against the breast cancer cell line MCF-7 by the metabolic assay MTT. Compounds 116 showed potent cytotoxic activities with IC50 values ranging from 0.16 to 4.65 μM, with compounds 4 and 5 being the most potent cytotoxic agents, which could be due to their multitarget inhibitory activities against EGFR, HER2, and VEGFR2 as well as CDK9 [26]. However, compounds 1720 and 2329 were significantly less potent than compounds 116, with IC50 values ranging from 3.88 to 28.7 μM.

2.3. Molecular Docking

Molecular docking experiments were performed using the genetic algorithm docking program GOLD 5.2 to rationalize the observed potency of compounds 7, 9, and 25. In addition, flavopiridol was used as a reference compound to compare the binding pattern. The modeled complexes with CDK9 are shown in Figure 4.
At the binding cavity of CDK9, flavopiridol, compounds 7, 9, and 25 occupy the adenosine triphosphate (ATP) binding site. Compounds 7 and 9 display similar binding modes. These conformations, however, are different from the ones shown by flavopiridol. The benzene ring of the quinazolinone is in contact with the Phe105 residue in the hinge region. In addition, the sulphonamide moieties of compounds 7 and 9 bind differently, forming hydrogen bonds with the Thr29 and Glu107 residues in the enzyme, respectively.
Moreover, the Ethoxy group of compound 7 forms a hydrogen bond with Lys48, whereas there is no interaction between the enzyme and the anilide moiety of compound 9. This suggests that the sulphonamide moiety is essential for the activity, whereas the acetanilide groups could be necessary for the potency in the context of 2-mercapto-3-(4-sulfamoylphenethyl)quinazolin-4-one. However, compound 25 adopts a similar orientation to that of flavopiridol. Unlike compounds 7 and 9, the NH group of compound 25 forms a hydrogen bond with Glu107. Moreover, the methyl group of compound 25 is in contact with Phe103 of the gatekeeper region of the enzyme. In addition, the m-bromophenyl group in compound 25 occupies a similar position to that occupied by the o-chlorophenyl ring of flavopiridol.

2.4. Analyses of the Physicochemical Properties of the Compounds

2.4.1. Lipinski’s Rule of Five

Lipinski’s rule of five was used to evaluate the drug-like properties of compounds 120 and 2329. DataWarrior was used to estimate the molecular weight (MW), CLogP, hydrogen bond acceptors (HBAs), and hydrogen bond donors (HBD) for each molecule, and the values are presented in Table 3. The data in the table show that compounds 416 have molecular weights of >500 Da. In addition, compound 8 offers an additional violation of Lipinski’s rule, with HBAs >10. However, all the compounds satisfy Lipinski’s rule regarding lipophilicity and the number of HBDs, with CLogP and HBD values below 5.

2.4.2. Ligand Efficiency (LE)

The LE is a property that describes the potency per heavy atom of a drug [50,51,52]. The LE values of the synthesized compounds were obtained using DataWarrior according to the following equation [50,52]:
L E = R T ln I C 50 N
N represents the number of heavy atoms, i.e., non-hydrogen atoms in the drug, R is the universal gas constant, T is the absolute temperature in degrees Kelvin, and IC50 is CDK9 IC50 in mol/L.
LE is an essential metric in lead optimization, which allows the comparison of the affinity of molecules according to their size. Compounds with LE values higher than 0.3 are considered promising lead compounds. The LE values for the target compounds in this study are presented in Table 4, which shows that the LE values of the synthesized compounds are between 0.2 and 0.7. Except for compounds 316, the LE values fall into an acceptable range and are >0.3 [52,53].

2.4.3. Ligand Lipophilic Efficiency (LLE)

LLE is used to link the potency of a compound to its lipophilicity [52,53]. The challenge in drug discovery is optimizing a compound’s activity while maintaining lipophilicity at a constant value. For this reason, LLE is considered an effective strategy to control the lipophilicity of a molecule to avoid any “molecular obesity” during lead optimization. The LLE values for compounds 1–20 and 23–29 shown in Table 4 were obtained using DataWarrior according to the following equation [52]:
L L E = p I C 50   C L o g P
pIC50 is the negative log of the CDK9 IC50 and CLogP is the calculated LogP value.
An acceptable lead compound should have an LLE value of ≥ 5. Compounds 2, 17, and 27 show good LLE values, i.e., LLE > 5 [52,53]. However, the other compounds have values that are below the recommended limit.
In conclusion, compound 17 is a good candidate for lead optimization since it has the lowest non-hydrogen atoms (N), an acceptable LE value of 0.708, and an acceptable LLE value of 5.20.

3. Materials and Methods

3.1. Chemistry

Chemicals and solvents were obtained from suppliers and used directly without any purification. Agilent 6320 Ion Trap mass spectrometer was used to generate mass spectra (MS). Melting Point Apparatus Barnstead 9100 Electrothermal was used to record the final compounds’ melting points (uncorrected). IR spectra were obtained using an FT-IR Perkin-Elmer spectrometer. Bruker 700 Ultrashield NMR spectrometer was run at 700 MHz and 175 MHz to generate 1H and 13C spectra, respectively. Compounds 120 and 29 were synthesized as reported previously [22,27,40,41,42,43]. the newly synthesized compounds were re-crystalized from ethanol. IR, NMR and mass spectra of compounds 23–28 are available in the Supplementary Material of this article.
2-(3-Bromobenzamido)-5-methylbenzoic acid (23). Equimolar amounts of 2-amino-5-methylbenzoic acid (20 mmol, 3.0 g) and 3-bromobenzoyl chloride (20 mmol, 4.40 g) were stirred at room temperature in 20 mL of anhydrous pyridine for 3 h. The solvent was then removed in vacuo, and the resulting crude solid was washed with 5% HCl ice–water before being filtered and dried to give the final product in 97% yield. M.P. 200–202 °C; IR (KBr, cm−1) ν: 3233 (NH), 2600 (COOH), 1653 (C=O); 1H NMR (DMSO-d6): δ 12.06 (s, 1H), 8.51 (s, 1H), 8.50 (d, 1H, J = 8.47 Hz), 8.08 (s, 1H), 7.93 (d, 1H, J = 7.70 Hz), 7.84 (t, 2H, J = 6.79 and 7.98 Hz), 7.55 (t, 1H, J = 7.84 Hz), 7.47 (d, 1H, J = 8.47 Hz), 2.33 (s, 3H); 13C NMR (DMSO-d6): δ 170.52, 163.37, 138.74, 137.27, 135.22, 135.20, 132.94, 131.73, 131.64, 130.26, 126.49, 122.64, 120.65, 117.46, 20.73; MS [M–OH; 316 and M–OH + 2: 318; M–COOH; 288 and 290].
2-(3-Bromophenyl)-6-methyl-4H-benzo[d][1,3]oxazin-4-one (24). 2-(3-Bromobenzamido)-5-methylbenzoic acid (23) (15 mmol, 4.74 g) was boiled in acetic anhydride for 4 h, after which the reaction mixture was cooled, and the solid was collected by filtration and dried to give the final product in 95% yield. M.P. 155–157 °C; IR (KBr, cm−1) ν: 1753 (C=O); 1H NMR (DMSO-d6): δ 8.20 (s, 1H), 8.11 (d, 1H, J = 7.77 Hz), 7.92 (s, 1H), 7.83 (d, 1H, J = 7.77 Hz), 7.75 (d, 1H, J = 8.12 Hz), 7.60 (d, 1H, J = 8.12 Hz), 7.54 (t, 1H, J = 7.91 and 7.84 Hz), 2.45 (s, 3H); 13C NMR (DMSO-d6): δ 159.02, 154.70, 144.23, 139.53, 138.39, 135.57, 132.78,131.66, 130.37, 128.05, 127.35, 127.02, 122.55, 117.12, 21.22; MS [m/z: 315 and M + 2: 317].
2-(3-Bromophenyl)-6-methylquinazolin-4(3H)-one (25). 2-(3-Bromophenyl)-6-methyl-4H-benzo[d][1,3]oxazin-4-one (24) (5 mmol 1.58 g) was heated in formamide (7 mL) for 10 h, after which the reaction mixture was cooled, and the resulting solid was filtered and dried to give the final product in 80% yield. M.P. 320–322 °C; IR (KBr, cm−1) ν: 3334 (NH), 1660, (C=O); 1H NMR (DMSO-d6): δ 8.35 (s, 1H), 8.15 (d, 1H, J = 7.00 Hz), 7.95 (s, 1H), 7.78 (d, 1H, J = 7.211 Hz), 7.68 (s, 2H), 7.51 (t, 1H, J = 7.28 and 7.35 Hz), 2.46 (s, 3H); 13C NMR (DMSO-d6): δ 165.30, 162.66, 150.70, 146.91, 137.16, 136.41, 135.55, 134.33, 131.22, 130.74, 127.88, 127.13, 125.72, 122.35, 21.34; MS [m/z: 314 and M + 2: 316].
2-(3-Bromophenyl)-6-methylquinazoline-4(3H)-thione (26). 2-(3-Bromophenyl)-6-methylquinazolin-4(3H)-one (25) (3 mmol, 945 mg) was heated with phosphorus pentasulfide in dry toluene (5 mL) for 12 h, after which time the reaction mixture was cooled, and the solid obtained was filtered and dried to give the final product in 68% yield. M.P. 259–260 °C; IR (KBr, cm−1) ν: 3102 (NH), 1241 (C=S); 1H NMR (DMSO-d6): δ 13.94 (s, 1H), 8.40 (s, 1H), 8.34 (s, 1H), 8.15 (d, 1H, J = 7.84 Hz), 7.79 (d, 1H, J = 7.91 Hz), 7.74 (dd, 1H, J = 1.33 Hz), 7.70 (d, 1H, J = 8.26 Hz), 7.51 (t, 1H, J = 7.91 Hz); 2.49 (s, 3H); 13C NMR (DMSO-d6): δ 187.62, 149.86, 142.67, 138.70, 137.49, 134.78, 134.45, 131.42, 131.05, 128.94, 128.66, 127.94, 127.89, 122.13, 21.54; MS [m/z: 330 and M + 2: 332].
3-Amino-2-(3-bromophenyl)-6-methylquinazolin-4(3H)-one (27). 2-(3-Bromophenyl)-6-methyl-4H-benzo[d][1,3]oxazin-4-one (24) (5 mmol 1.58 g) was heated with absolute hydrazine hydrate (3 mL) and absolute ethanol (3 mL) for 8 h, after which time the reaction mixture was cooled. The separated solid was filtered and dried to give the final product an 87% yield. M.P. 170–172 °C; IR (KBr, cm−1) ν: 3401, 3308 (NH2), 1668 (C=O); 1H NMR (DMSO-d6): δ 8.00 (t, 1H, J = 1.68 and 1.75 Hz), 7.99 (s, 1H), 7.81 (d, 1H, J = 7.84 Hz), 7.70 (tt, 1H, J = 0.98 and 0.77 Hz), 7.67 (dd, 1H, J = 1.82 Hz), 7.64 (d, 1H, J = 8.119 Hz), 7.45 (t, 1H, J = 7.91), 5.66 (s, 2H), 2.48 (s, 3H); 13C NMR (DMSO-d6): δ 161.53, 154.06, 145.12, 137.56, 137.28, 136.21, 132.66, 132.60, 130.09, 129.16, 127.83, 125.72, 120.96, 120.47, 21.35; MS [m/z: 329 and M + 2: 331].
2-(3-Bromophenyl)-3-hydroxy-6-methylquinazolin-4(3H)-one (28). 2-(3-Bromophenyl)-6-methyl-4H-benzo[d][1,3]oxazin-4-one (24) (5 mmol 1.58 g) was heated with hydroxylamine hydrochloride (6 mmol, 417 mg) in dry pyridine (10 mL) for 15 h. The reaction mixture was cooled, and the solvent was removed in a vacuo. The solid obtained was then washed with 5% HCl ice–water and filtered, dried, and recrystallized from ethanol to give the final product an 84% yield. M.P. 235–237 °C; IR (KBr, cm−1) ν: 3301(OH), 1670 (C=O); 1H NMR (DMSO-d6): δ 11.73 (s, 1H), 9.00 (d, 2H, J = 9.24 Hz), 7.84 (d, 1H, J = 7.77 Hz), 7.76 (dd, 1H, J = 0.84 and 0.92 Hz), 7.68 (q, 2H, J = 8.19 and 8.26 Hz), 7.49 (t, 1H, J = 7.91 Hz), 2.48 (s, 3H); 13C NMR (DMSO-d6): δ 158.55, 151.48, 144.59, 137.43, 136.12, 135.47, 133.39, 132.41, 130.54, 128.97, 127.99, 125.64, 121.81, 121.36, 21.33; MS [M–OH: 313 and M–OH + 2: 315].

3.2. Metabolic Assay

The antiproliferative activity of the twenty-seven compounds was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) metabolic assay against the MCF-7 cell line, according to a previous method [26].

3.3. CDK9 Kinase Assay

In vitro luminescent CDK9 kinase assay was performed as reported previously using Kinase- Glo® MAX as a detection reagent [43]. Briefly, 5 μL of each inhibitor in concentrations ranging from 10 μM to 1 nM (10 μM, 1 μM, 0.1 μM, 0.01 μM, and 0.001 μM) and 10 μL of enzyme substrate were mixed in 20 μL of kinase assay buffer (obtained from BPS Bioscience, catalog #79334) at room temperature. Then 20 μL of 5 ng/μL CDK9/cyclin T was added to the mixture to initiate the reaction. After 45 min, 50 μL of Kinase-Glo® Max reagent was added, and the resulting mixture was incubated for 15 min at room temperature. The chemiluminescence was measured microplate reader, and IC50 values were calculated using Prism 8.0 (GraphPad Software, San Diego, CA, USA).

3.4. Molecular Docking

Molecular docking was performed according to the procedure reported previously using the X-ray crystal structure of flavopiridol in a complex with CDK9 (PDB ID: 3BLR) which was retrieved from the PDB Data Bank (URL: http://www.rcsb.org; accessed on 20 September 2022) [26].

4. Conclusions

Twenty-seven compounds were synthesized, and their CDK9 inhibitory and cytotoxic activities were evaluated. Compounds 7, 9, and 25 were the most potent CDK9 inhibitors, with IC50 values of 0.115, 0.131, and 0.142 μM, respectively. The binding modes of these molecules were studied using molecular docking, which showed that they occupy the ATP binding site of CDK9. Of these three molecules, compound 25 shows good drug-like properties since it does not violate Lipinski’s rule of five. In addition, this molecule shows promising LE and LLE values and is an ideal candidate for further optimization.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28010120/s1, 1H NMR, 13C NMR, FT-IR, and mass spectra of compounds 2328.

Author Contributions

H.M.A. and A.S.E.-A. Conceived, planned, and carried out the experiments and contributed to interpreting and validating the results and writing the manuscript. A.A.Z. and S.G.A.-H. contributed to the research’s design and implementation, the analysis and interpretation of the result, and the manuscript’s writing. M.M.A. and S.A.A. contributed to the results’ design, visualization, and investigation. R.M.A. and A.A.-M.A.-A. Contributed to the design, carried out the methodology, and wrote the draft. R.A.A. and L.S.A.-R. contributed to the formal analysis and data curation along with the original draft preparation. A.J.O. and A.A.A. contributed to the design and performance of Molecular docking. F.Y.A. and F.S.A.; contributed to resources and writing—review and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research project was supported by the Researchers Supporting Project number (RSP-2021/340), King Saud University, Riyadh, Saudi Arabia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available by request from the corresponding author.

Acknowledgments

This research project was supported by the Researchers Supporting Project number (RSP-2021/340), King Saud University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sánchez-Martínez, C.; Lallena, M.J.; Sanfeliciano, S.G.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015–2019). Bioorg. Med. Chem. Lett. 2019, 29, 126637. [Google Scholar] [CrossRef]
  2. Diab, S.; Yu, M.; Wang, S. CDK7 Inhibitors in Cancer Therapy: The Sweet Smell of Success? J. Med. Chem. 2020, 63, 7458–7474. [Google Scholar] [CrossRef] [PubMed]
  3. Cheng, W.; Yang, Z.; Wang, S.; Li, Y.; Wei, H.; Tian, X.; Kan, Q. Recent development of CDK inhibitors: An overview of CDK/inhibitor co-crystal structures. Eur. J. Med. Chem. 2019, 164, 615–639. [Google Scholar] [CrossRef]
  4. Tutone, M.; Almerico, A.M. Recent advances on CDK inhibitors: An insight by means of in silico methods. Eur. J. Med. Chem. 2017, 142, 300–315. [Google Scholar] [CrossRef] [PubMed]
  5. Asghar, U.; Witkiewicz, A.K.; Turner, N.C.; Knudsen, E.S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 2015, 14, 130–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Malumbres, M. Cyclin-dependent kinases. Genome Biol. 2014, 15, 122. [Google Scholar] [CrossRef] [Green Version]
  7. Sánchez-Martínez, C.; Gelbert, L.M.; Lallena, M.J.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs. Bioorg. Med. Chem. Lett. 2015, 25, 3420–3435. [Google Scholar] [CrossRef]
  8. Krystof, V.; Baumli, S.; Furst, R. Perspective of Cyclin-dependent kinase 9 (CDK9) as a Drug Target. Curr. Pharm. Des. 2012, 18, 2883–2890. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, S.; Fischer, P.M. Cyclin-dependent kinase 9: A key transcriptional regulator and potential drug target in oncology, virology and cardiology. Trends Pharmacol. Sci. 2008, 29, 302–313. [Google Scholar] [CrossRef]
  10. Lukasik, P.M.; Elabar, S.; Lam, F.; Shao, H.; Liu, X.R.; Abbas, A.Y.; Wang, S.D. Synthesis and biological evaluation of imidazo[4,5-b]pyridine and 4-heteroaryl-pyrimidine derivatives as anti-cancer agents. Eur. J. Med. Chem. 2012, 57, 311–322. [Google Scholar] [CrossRef]
  11. Alkahtani, H.M.; Abbas, A.Y.; Wang, S. Synthesis and biological evaluation of benzo[d]imidazole derivatives as potential anti-cancer agents. Bioorg. Med. Chem. Lett. 2012, 22, 1317–1321. [Google Scholar] [CrossRef]
  12. Shao, H.; Shi, S.H.; Foley, D.W.; Lam, F.; Abbas, A.Y.; Liu, X.R.; Huang, S.L.; Jiang, X.R.; Baharin, N.; Fischer, P.M.; et al. Synthesis, structure–activity relationship and biological evaluation of 2,4,5-trisubstituted pyrimidine CDK inhibitors as potential anti-tumour agents. Eur. J. Med. Chem. 2013, 70, 447–455. [Google Scholar] [CrossRef] [PubMed]
  13. Shao, H.; Shi, S.; Huang, S.; Hole, A.J.; Abbas, A.Y.; Baumli, S.; Liu, X.; Lam, F.; Foley, D.W.; Fischer, P.M.; et al. Substituted 4-(Thiazol-5-yl)-2-(phenylamino)pyrimidines Are Highly Active CDK9 Inhibitors: Synthesis, X-ray Crystal Structures, Structure–Activity Relationship, and Anticancer Activities. J. Med. Chem. 2013, 56, 640–659. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, R.; Wierda, W.G.; Chubb, S.; Hawtin, R.E.; Fox, J.A.; Keating, M.J.; Gandhi, V.; Plunkett, W. Mechanism of action of SNS-032, a novel cyclin-dependent kinase inhibitor, in chronic lymphocytic leukemia. Blood 2009, 113, 4637–4645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Walsby, E.; Pratt, G.; Shao, H.; Abbas, A.Y.; Fischer, P.M.; Bradshaw, T.D.; Brennan, P.; Fegan, C.; Wang, S.; Pepper, C. A novel Cdk9 inhibitor preferentially targets tumor cells and synergizes with fludarabine. Oncotarget 2014, 5, 375–385. [Google Scholar] [CrossRef] [Green Version]
  16. Wu, T.; Qin, Z.; Tian, Y.; Wang, J.; Xu, C.; Li, Z.; Bian, J. Recent Developments in the Biology and Medicinal Chemistry of CDK9 Inhibitors: An Update. J. Med. Chem. 2020, 63, 13228–13257. [Google Scholar] [CrossRef]
  17. Kryštof, V.; Chamrád, I.; Jorda, R.; Kohoutek, J. Pharmacological targeting of CDK9 in cardiac hypertrophy. Med. Res. Rev. 2010, 30, 646–666. [Google Scholar] [CrossRef]
  18. Richter, A.; Schoenwaelder, N.; Sender, S.; Junghanss, C.; Maletzki, C. Cyclin-Dependent Kinase Inhibitors in Hematological Malignancies—Current Understanding, (Pre-)Clinical Application and Promising Approaches. Cancers 2021, 13, 2497. [Google Scholar] [CrossRef]
  19. Mandal, R.; Becker, S.; Strebhardt, K. Targeting CDK9 for Anti-Cancer Therapeutics. Cancers 2021, 13, 2181. [Google Scholar] [CrossRef]
  20. Mitra, P.; Yang, R.-M.; Sutton, J.; Ramsay, R.G.; Gonda, T.J. CDK9 inhibitors selectively target estrogen receptor-positive breast cancer cells through combined inhibition of MYB and MCL-1 expression. Oncotarget 2016, 7, 9069–9083. [Google Scholar] [CrossRef]
  21. Cheng, S.-S.; Qu, Y.-Q.; Wu, J.; Yang, G.-J.; Liu, H.; Wang, W.; Huang, Q.; Chen, F.; Li, G.; Wong, C.-Y.; et al. Inhibition of the CDK9–cyclin T1 protein–protein interaction as a new approach against triple-negative breast cancer. Acta Pharm. Sin. B 2022, 12, 1390–1405. [Google Scholar] [CrossRef] [PubMed]
  22. Deep, A.; Marwaha, R.K.; Marwaha, M.G.; Jyoti, J.; Nandal, R.; Sharma, A.K. Flavopiridol as cyclin dependent kinase (CDK) inhibitor: A review. New J. Chem. 2018, 42, 18500–18507. [Google Scholar] [CrossRef]
  23. Hameed, A.; Al-Rashida, M.; Uroos, M.; Ali, S.A.; Arshia; Ishtiaq, M.; Khan, K.M. Quinazoline and quinazolinone as important medicinal scaffolds: A comparative patent review (2011–2016). Expert Opin. Ther. Pat. 2018, 28, 281–297. [Google Scholar] [CrossRef] [PubMed]
  24. 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] [PubMed]
  25. Khan, I.; Zaib, S.; Batool, S.; Abbas, N.; Ashraf, Z.; Iqbal, J.; Saeed, A. Quinazolines and quinazolinones as ubiquitous structural fragments in medicinal chemistry: An update on the development of synthetic methods and pharmacological diversification. Bioorg. Med. Chem. 2016, 24, 2361–2381. [Google Scholar] [CrossRef]
  26. Alkahtani, H.M.; Abdalla, A.N.; Obaidullah, A.J.; Alanazi, M.M.; Almehizia, A.A.; Alanazi, M.G.; Ahmed, A.Y.; Alwassil, O.I.; Darwish, H.W.; Abdel-Aziz, A.A.-M.; et al. Synthesis, cytotoxic evaluation, and molecular docking studies of novel quinazoline derivatives with benzenesulfonamide and anilide tails: Dual inhibitors of EGFR/HER2. Bioorg. Chem. 2020, 95, 103461. [Google Scholar] [CrossRef]
  27. El-Azab, A.S.; Abdel-Aziz, A.A.-M.; Bua, S.; Nocentini, A.; El-Gendy, M.A.; Mohamed, M.A.; Shawer, T.Z.; AlSaif, N.A.; Supuran, C.T. Synthesis of benzensulfonamides linked to quinazoline scaffolds as novel carbonic anhydrase inhibitors. Bioorg. Chem. 2019, 87, 78–90. [Google Scholar] [CrossRef]
  28. Abdel-Aziz, A.A.-M.; Abou-Zeid, L.A.; ElTahir, K.E.H.; Mohamed, M.A.; Abu El-Enin, M.A.; El-Azab, A.S. Design, synthesis of 2,3-disubstitued 4(3H)-quinazolinone derivatives as anti-inflammatory and analgesic agents: COX-1/2 inhibitory activities and molecular docking studies. Bioorg. Med. Chem. 2016, 24, 3818–3828. [Google Scholar] [CrossRef]
  29. Abdel-Aziz, A.A.-M.; Abou-Zeid, L.A.; ElTahir, K.E.H.; Ayyad, R.R.; El-Sayed, M.A.-A.; El-Azab, A.S. Synthesis, anti-inflammatory, analgesic, COX-1/2 inhibitory activities and molecular docking studies of substituted 2-mercapto-4(3H)-quinazolinones. Eur. J. Med. Chem. 2016, 121, 410–421. [Google Scholar] [CrossRef]
  30. Alanazi, A.M.; Abdel-Aziz, A.A.-M.; Shawer, T.Z.; Ayyad, R.R.; Al-Obaid, A.M.; Al-Agamy, M.H.M.; Maarouf, A.R.; El-Azab, A.S. Synthesis, antitumor and antimicrobial activity of some new 6-methyl-3-phenyl-4(3H)-quinazolinone analogues: In silico studies. J. Enzym. Inhib. Med. Chem. 2016, 31, 721–735. [Google Scholar] [CrossRef]
  31. El-Azab, A.S.; Abdel-Hamide, S.G.; Sayed-Ahmed, M.M.; Hassan, G.S.; El-Hadiyah, T.M.; Al-Shabanah, O.A.; Al-Deeb, O.A.; El-Subbagh, H.I. Novel 4(3H)-quinazolinone analogs: Synthesis and anticonvulsant activity. Med. Chem. Res. 2013, 22, 2815–2827. [Google Scholar] [CrossRef]
  32. El-Azab, A.S.; ElTahir, K.E.H. Synthesis and anticonvulsant evaluation of some new 2,3,8-trisubstituted-4(3H)-quinazoline derivatives. Bioorg. Med. Chem. Lett. 2012, 22, 327–333. [Google Scholar] [CrossRef] [PubMed]
  33. El-Azab, A.S.; ElTahir, K.E. Design and synthesis of novel 7-aminoquinazoline derivatives: Antitumor and anticonvulsant activities. Bioorg. Med. Chem. Lett. 2012, 22, 1879–1885. [Google Scholar] [CrossRef]
  34. El-Azab, A.S.; ElTahir, K.E.H.; Attia, S.M. Synthesis and anticonvulsant evaluation of some novel 4(3H)-quinazolinones. Monatsh. Chem. 2011, 142, 837–848. [Google Scholar] [CrossRef]
  35. El-Azab, A.S.; Al-Omar, M.A.; Abdel-Aziz, A.A.-M.; Abdel-Aziz, N.I.; El-Sayed, M.A.-A.; Aleisa, A.M.; Sayed-Ahmed, M.M.; Abdel-Hamide, S.G. Design, synthesis and biological evaluation of novel quinazoline derivatives as potential antitumor agents: Molecular docking study. Eur. J. Med. Chem. 2010, 45, 4188–4198. [Google Scholar] [CrossRef]
  36. El-Azab, A.S. Synthesis of Some New Substituted 2-Mercaptoquinazoline Analogs as Potential Antimicrobial Agents. Phosphorus Sulfur Silicon Relat. Elem. 2007, 182, 333–348. [Google Scholar] [CrossRef]
  37. Aziza, M.A.; Nassar, M.W.; Abdel Hamide, S.G.; El Hakim, A.E.; El-Azab, A.S. Synthesis and antimicrobial activities of some new 3-heteroaryl-quinazolin-4-ones. Indian J. Heterocy. Ch. 1996, 6, 25–30. [Google Scholar]
  38. Alanazi, A.M.; Al-Suwaidan, I.A.; Abdel-Aziz, A.A.-M.; Mohamed, M.A.; El Morsy, A.M.; El-Azab, A.S. Design, synthesis and biological evaluation of some novel substituted 2-mercapto-3-phenethylquinazolines as antitumor agents. Med. Chem. Res. 2013, 22, 5566–5577. [Google Scholar] [CrossRef]
  39. Al-Suwaidan, I.A.; Abdel-Aziz, A.A.-M.; Shawer, T.Z.; Ayyad, R.R.; Alanazi, A.M.; El-Morsy, A.M.; Mohamed, M.A.; Abdel-Aziz, N.I.; El-Sayed, M.A.-A.; El-Azab, A.S. Synthesis, antitumor activity and molecular docking study of some novel 3-benzyl-4(3H)quinazolinone analogues. J. Enzym. Inhib. Med. Chem. 2016, 31, 78–89. [Google Scholar] [CrossRef] [Green Version]
  40. El-Azab, A.S.; Al-Dhfyan, A.; Abdel-Aziz, A.A.-M.; Abou-Zeid, L.A.; Alkahtani, H.M.; Al-Obaid, A.M.; Al-Gendy, M.A. Synthesis, anticancer and apoptosis-inducing activities of quinazoline–isatin conjugates: Epidermal growth factor receptor-tyrosine kinase assay and molecular docking studies. J. Enzym. Inhib. Med. Chem. 2017, 32, 935–944. [Google Scholar] [CrossRef] [Green Version]
  41. El-Azab, A.S.; Abdel-Aziz, A.A.-M.; Ghabbour, H.A.; Al-Gendy, M.A. Synthesis, in vitro antitumour activity, and molecular docking study of novel 2-substituted mercapto-3-(3,4,5-trimethoxybenzyl)-4(3H)-quinazolinone analogues. J. Enzym. Inhib. Med. Chem. 2017, 32, 1229–1239. [Google Scholar] [CrossRef] [PubMed]
  42. El-Azab, A.S.; Abdel-Aziz, A.A.-M.; Ghabbour, H.A.; Bua, S.; Nocentini, A.; Alkahtani, H.M.; Alsaif, N.A.; Al-Agamy, M.H.M.; Supuran, C.T. Carbonic Anhydrase Inhibition Activities of Schiff’s Bases Based on Quinazoline-Linked Benzenesulfonamide. Molecules 2022, 27, 7703. [Google Scholar] [CrossRef] [PubMed]
  43. El-Azab, A.S.; Abdel-Aziz, A.A.-M.; AlSaif, N.A.; Alkahtani, H.M.; Alanazi, M.M.; Obaidullah, A.J.; Eskandrani, R.O.; Alharbi, A. Antitumor activity, multitarget mechanisms, and molecular docking studies of quinazoline derivatives based on a benzenesulfonamide scaffold: Cell cycle analysis. Bioorg. Chem. 2020, 104, 104345. [Google Scholar] [CrossRef] [PubMed]
  44. Sancineto, L.; Iraci, N.; Massari, S.; Attanasio, V.; Corazza, G.; Barreca, M.L.; Sabatini, S.; Manfroni, G.; Avanzi, N.R.; Cecchetti, V.; et al. Computer-Aided Design, Synthesis and Validation of 2-Phenylquinazolinone Fragments as CDK9 Inhibitors with Anti-HIV-1 Tat-Mediated Transcription Activity. ChemMedChem 2013, 8, 1941–1953. [Google Scholar] [CrossRef]
  45. Abdalla, A.N.; Abdallah, M.E.; Aslam, A.; Bader, A.; Vassallo, A.; Tommasi, N.; Malki, W.H.; Gouda, A.M.; Mukhtar, M.H.; El-Readi, M.Z.; et al. Synergistic Anti Leukemia Effect of a Novel Hsp90 and a Pan Cyclin Dependent Kinase Inhibitors. Molecules 2020, 9, 25. [Google Scholar] [CrossRef]
  46. Bozdag, M.; Alafeefy, A.M.; Carta, F.; Ceruso, M.; Al-Tamimi, A.-M.S.; Al-Kahtani, A.A.; Alasmary, F.A.S.; Supuran, C.T. Synthesis 4-[2-(2-mercapto-4-oxo-4H-quinazolin-3-yl)-ethyl]-benzenesulfonamides with subnanomolar carbonic anhydrase II and XII inhibitory properties. Bioorg. Med. Chem. 2016, 24, 4100–4107. [Google Scholar] [CrossRef]
  47. Kulkarni, S.S.; Singh, S.; Shah, J.R.; Low, W.-K.; Talele, T.T. Synthesis and SAR optimization of quinazolin-4(3H)-ones as poly(ADP-ribose)polymerase-1 inhibitors. Eur. J. Med. Chem. 2012, 50, 264–273. [Google Scholar] [CrossRef]
  48. Alanazi, A.M.; Abdel-Aziz, A.A.M.; Al-Suwaidan, I.A.; Abdel-Hamide, S.G.; Shawer, T.Z.; El-Azab, A.S. Design, synthesis and biological evaluation of some novel substituted quinazolines as antitumor agents. Eur. J. Med. Chem. 2014, 79, 446–454. [Google Scholar] [CrossRef]
  49. Cho, N.-C.; Cha, J.H.; Kim, H.; Kwak, J.; Kim, D.; Seo, S.-H.; Shin, J.-S.; Kim, T.; Park, K.D.; Lee, J.; et al. Discovery of 2-aryloxy-4-amino-quinazoline derivatives as novel protease-activated receptor 2 (PAR2) antagonists. Bioorg. Med. Chem. 2015, 23, 7717–7727. [Google Scholar] [CrossRef]
  50. Hopkins, A.L.; Groom, C.R.; Alex, A. Ligand efficiency: A useful metric for lead selection. Drug Discov. Today 2004, 9, 430–431. [Google Scholar] [CrossRef]
  51. Abad-Zapatero, C.; Metz, J.T. Ligand efficiency indices as guideposts for drug discovery. Drug Discov. Today 2005, 10, 464–469. [Google Scholar] [CrossRef] [PubMed]
  52. Roskoski, R., Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2020 update. Pharmacol. Res. 2020, 152, 104609. [Google Scholar] [CrossRef] [PubMed]
  53. Hopkins, A.L.; Keserü, G.M.; Leeson, P.D.; Rees, D.C.; Reynolds, C.H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discov. 2014, 13, 105–121. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of some known CDK9 inhibitors.
Figure 1. Structures of some known CDK9 inhibitors.
Molecules 28 00120 g001
Figure 2. Rational design of quinazoline derivatives as CDK9 inhibitors (120 and 2329).
Figure 2. Rational design of quinazoline derivatives as CDK9 inhibitors (120 and 2329).
Molecules 28 00120 g002
Scheme 1. Synthesis of compounds 216.
Scheme 1. Synthesis of compounds 216.
Molecules 28 00120 sch001
Figure 3. Structures of compounds 17-20 and 29.
Figure 3. Structures of compounds 17-20 and 29.
Molecules 28 00120 g003
Scheme 2. Synthesis routes of compounds 2328.
Scheme 2. Synthesis routes of compounds 2328.
Molecules 28 00120 sch002
Figure 4. Binding modes of (A) flavopiridol, (B) compound 7, (C) compound 9, and (D) compound 25 with CDK9.
Figure 4. Binding modes of (A) flavopiridol, (B) compound 7, (C) compound 9, and (D) compound 25 with CDK9.
Molecules 28 00120 g004
Table 1. Structures of compounds 116 and their biological activity.
Table 1. Structures of compounds 116 and their biological activity.
Molecules 28 00120 i001
CompdStructureRCDK9 IC50 (μM)aCytotoxicity IC50 (μM), MCF-71,a
1--0.644 ± 0.0070.65 ± 0.15
2AH0.454 ± 0.0062.31 ± 0.39
3APh0.421 ± 0.0060.82 ± 0.08
4A4-tolyl0.788 ± 0.0120.16 ± 0.02
5A4-acetylphenyl0.829 ± 0.0140.18 ± 0.09
6A4-methoxyphenyl0.463 ± 0.0074.65 ± 1.38
7A4-ethoxyphenyl0.115 ± 0.0021.57 ± 0.04
8A3,4,5-trimethoxyphenyl0.501 ± 0.0092.48 ± 0.32
9A4-bromophenyl0.131 ± 0.0021.72 ± 0.26
10A4-chlorophenyl0.193 ± 0.0031.04 ± 0.07
11A4-fluorophenyl0.336 ± 0.0053.86 ± 0.78
12A4-flourobenzyl0.218 ± 0.0040.69 ± 0.23
13A4-methoxybenzyl0.334 ± 0.0060.84 ± 0.15
14A3,4-dimethoxybenzyl0.229 ± 0.0040.59 ± 0.10
15BPh0.444 ± 0.0070.67 ± 0.18
16B4-chlorophenyl0.350 ± 0.0060.73 ± 0.01
Flavopiridol--0.020 ± 0.0080.04 ± 0.001
1 Antiproliferative activity by the metabolic assay MTT-48 h. Data are expressed as mean ± SD. a all of the values in the table showed statistical differences (1-way ANOVA and post hoc Tukey-Kramer multiple comparison tests, p < 0.05) compared with the untreated control group.
Table 2. Structures of compounds 1720 and 23-29 and their biological activity.
Table 2. Structures of compounds 1720 and 23-29 and their biological activity.
Molecules 28 00120 i002
CompoundXYR1R2CDK9 (μM) aCytotoxicity IC50 (μM), MCF-7 1,a
17ONHHI0.639 ± 0.0043.88 ± 0.12
18ONH2-thienylI0.296 ± 0.00321.4 ± 0.68
19ONH4-tolylCl0.282 ± 0.00312.3 ± 0.39
20ONH4-chlorophenylNO20.434 ± 0.0036.07 ± 0.19
23----0.210 ± 0.00311.5 ± 0.37
24OO3-bromophenylCH30.486 ± 0.00410.4 ± 0.33
25ONH3-bromophenylCH30.142 ± 0.00116.8 ± 0.54
26SNH3-bromophenylCH30.289 ± 0.0025.21 ± 0.17
27ON-NH23-bromophenylCH30.835 ± 0.00628.7 ± 0.92
28ON-OH3-bromophenylCH30.210 ± 0.0026.27 ± 0.2
29----0.589 ± 0.00217.4 ± 0.56
Flavopiridol----0.020 ± 0.0080.04 ± 0.001
1 Antiproliferative activity by the metabolic assay MTT-48 h. Data are expressed as mean ± SD. aAll of the values in the table showed statistical differences (1-way ANOVA and post hoc Tukey-Kramer multiple comparison tests, p < 0.05) when compared with the untreated control group.
Table 3. Summary of the physicochemical properties of compounds 1–20 and 23-29.
Table 3. Summary of the physicochemical properties of compounds 1–20 and 23-29.
CompoundMW (Da)CLogPHBAHBDNo. of Violations
13611.83610
24180.87820
34952.93820
45093.28821
55372.80921
65252.86921
75393.27921
85852.721122
95733.66821
105293.54821
115133.03821
125272.74821
135392.57921
145692.501022
155093.34821
165433.95821
172721.00310
183542.51310
192713.16310
203021.89610
233343.36420
243163.42300
253153.28310
263313.61210
273301.97410
283312.62410
291620.51420
Table 4. LE and LLE values of compounds 1–20 and 23-29.
Table 4. LE and LLE values of compounds 1–20 and 23-29.
CompoundIC50 (μM)pIC50NLELLE
10.6446.19240.3544.36
20.4546.34280.3115.47
30.4216.38340.2573.44
40.7886.10350.2392.83
50.8296.08370.2253.28
60.4636.33360.2413.47
70.1156.94370.2573.67
80.5016.30400.2163.58
90.1316.88350.2703.22
100.1936.71350.2633.18
110.3366.47350.2543.44
120.2186.66360.2543.92
130.3346.48370.2403.90
140.2296.64390.2344.14
150.4446.35350.2493.01
160.356.46360.2462.51
170.6396.19120.7085.20
180.2966.53170.5274.02
190.2826.55190.4733.39
200.4346.36210.4164.47
230.216.68200.4583.32
240.4866.31190.4562.89
250.1426.85190.4943.57
260.2896.54190.4722.93
270.8356.08200.4174.11
280.216.68200.4584.06
290.5896.23120.7125.72
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

Alkahtani, H.M.; Zen, A.A.; Obaidullah, A.J.; Alanazi, M.M.; Almehizia, A.A.; Ansari, S.A.; Aleanizy, F.S.; Alqahtani, F.Y.; Aldossari, R.M.; Algamdi, R.A.; et al. Synthesis, Cytotoxic Evaluation, and Structure-Activity Relationship of Substituted Quinazolinones as Cyclin-Dependent Kinase 9 Inhibitors. Molecules 2023, 28, 120. https://doi.org/10.3390/molecules28010120

AMA Style

Alkahtani HM, Zen AA, Obaidullah AJ, Alanazi MM, Almehizia AA, Ansari SA, Aleanizy FS, Alqahtani FY, Aldossari RM, Algamdi RA, et al. Synthesis, Cytotoxic Evaluation, and Structure-Activity Relationship of Substituted Quinazolinones as Cyclin-Dependent Kinase 9 Inhibitors. Molecules. 2023; 28(1):120. https://doi.org/10.3390/molecules28010120

Chicago/Turabian Style

Alkahtani, Hamad M., Amer Alhaj Zen, Ahmad J. Obaidullah, Mohammed M. Alanazi, Abdulrahman A. Almehizia, Siddique Akber Ansari, Fadilah Sfouq Aleanizy, Fulwah Yahya Alqahtani, Rana M. Aldossari, Raghad Abdullah Algamdi, and et al. 2023. "Synthesis, Cytotoxic Evaluation, and Structure-Activity Relationship of Substituted Quinazolinones as Cyclin-Dependent Kinase 9 Inhibitors" Molecules 28, no. 1: 120. https://doi.org/10.3390/molecules28010120

Article Metrics

Back to TopTop