In silico Exploration of Quinazolinone-incorporated-chalcones as EGFR inhibitors (T790M mutated) to Combat Lung Cancer
Praveen Kumar Arora1*, Sushil Kumar2, Sandeep Kumar Bansal3 and Tarun Virmani4
1Pt. L.R. College of Pharmacy, Faridabad, Haryana, India
2School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh, India.
3Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, Uttar Pradesh, India.
4Department of Pharmaceutical Sciences, Amity University, Greater Noida, Uttar Pradesh, India.
Corresponding Author E-mail:procarora@gmail.com
DOI : http://dx.doi.org/10.13005/ojc/400521
Article Received on : 16 Aug 2024
Article Accepted on : 28 Sep 2024
Article Published : 31 Oct 2024
Reviewed by: Dr. Parasuraman S
Second Review by: Dr. Naresh Batham
Final Approval by: Dr. Ioana Stanciu
The current research concentrates on the insilico exploration of quinazolinone-incorporated chalcones (42 ligands) as anti-lung-cancer agents by evaluating their ability to inhibit mutated EGFR (T790M mutation) by docking studies employing autodock 4. The observed free binding energies of the ligands were -45.44 KJ/mol to -34.64 KJ/mol and the observed inhibition constants range was 11.04 nM to 853.47 nM. In the docking studies, when compared with the reference EGFR TKIs (erlotinib, afatinib, and naquotinib), all the docked 42 ligands were found to have higher potency and the compound C19 was found as the most potent ligand (binding energy = -45.44 KJ/mol and inhibition constant = 11.04 nM). As per the Osiris property explorer prediction, ligand C6 was with the highest drug score (0.42) followed by ligand C9(0.35).
KEYWORDS:Anticancer agents; Cytotoxicity; Chalcones; Docking; EGFR inhibitors; Non-small cell lung cancer (NSCLC)
Download this article as:Copy the following to cite this article: Arora P. K, Kumar S, Bansal S. K, Virmani T. Insilico exploration of Quinazolinone-incorporated-chalcones as EGFR inhibitors (T790M mutated) to combat Lung Cancer. Orient J Chem 2024;40(5). |
Copy the following to cite this URL: Arora P. K, Kumar S, Bansal S. K, Virmani T. Insilico exploration of Quinazolinone-incorporated-chalcones as EGFR inhibitors (T790M mutated) to combat Lung Cancer. Orient J Chem 2024;40(5). Available from: https://bit.ly/3UKAe5d |
Introduction
Globally, lung cancer is the most prevalent cancer to be diagnosed and the primary cause of cancer-related mortality1,2. The substantial therapeutic potential of the quinazolinone/quinazoline scaffold was discovered through a review of the literature. Recently, certain new trimethoxy quinazolines have been identified by Altamimi et al. as promiscuous EGFR inhibitors3.
Chalcones serve as an important pharmacophoric site in a variety of anticancer compounds, such as butein, isoliquiritigenin, and other natural and synthetic anticancer compounds4. The chalcone structural motif (1,3-diaryl-prop-2-ene-1-one) exhibits a variety of biological activities. Molecular hybridization, a method of drug designing, involves joining two distinct bioactive scaffolds to create a single hybrid molecule. The design of new anticancer drugs have been shown to benefit greatly from the molecular hybridization approach, for example,para-aminoquinazoline-chalcone conjugates5, quinoline incorporated chalcones6,benzimidazole incorporated chalcones7, chalcones having thiazole heterocyclic ring8, chalcones having thiazole ring9, 2-arylquinazolin-4-one naphthylchalcone hybrids10, 2-(4-methoxyphenyl)quinazolin-4-one- chalcone derivatives11 etc., All favour employing the chalcone structural motif in the search for novel anticancer medicines.
Patients with lung cancer typically have a poor prognosis and a low survival rate, and this is largely due to EGFR over-expression and EGFR mutation. The harsh side effects and poor prognosis connected with traditional chemotherapeutic therapies have been the impetus for the creation of EGFR-TKIs12-14.First, second, and third-generation EGFR inhibitors are presently employed for treating NSCLC 15. When treating NSCLC associated with the T790M EGFR mutation, first-generation (e.g. erlotinib) and second-generation (e.g. afatinib) EGFR inhibitors produce unsatisfactory results15.In clinical studies, it has been revealed that T790M mutation is also induced by first or second-generation EGFR TKIs15. At present, EGFR with T790M mutation can only be targeted satisfactorily by the third-generation EGFR TKIs (e.g. naquotinib, osimertinib or lazertinib ) but in the course of treatment, these drugs also suffered from EGFR resistance15.The present study concentrates on the insilico investigations of the chalcone ligands to assess their toxicity risks, drug score, drug-likeness, physiochemical parameters, pharmacokinetic parameters, and their ability to inhibit mutated EGFR, taking erlotinib, afatinib and naquotinib as reference drugs for the comparison.
Materials and Methods
Osiris property explorer was used to predict toxicity risks, solubility, cLogP, TPSA, drug-likeness and drug-score. The Swiss ADME online server was used to predict the pharmacokinetic parameters. MarvinSketch18.23 was utilized for drawing chemical structures of ligands and for optimization of their energy. The T790M mutation carrying epidermal growth factor receptor (EGFR) (Protein data bank ID: 5Y9T) was obtained from the Protein data bank (https://www.rcsb.org). AutoDock 4.0 MGL tools were used to investigate molecular docking. Using the discovery studio visualizer, the ligand-EGFR (5Y9T) complex was observed.
Docking run (target 5Y9T)
Preparation of target protein (5Y9T) and the preparation of ligands were done as per the reported procedures10. Validated docking experiments using autodock 4 were done as per the reported procedure11.
Results and Discussion
Osiris Property explorer Toxicity Predictions
The OSIRIS property explorer predicts the results using color code. The red and orange colors indicate that ligand is associated with high risk and medium risk of undesired effects (mutagenicity, tumorigenicity, irritant, and reproductive effect) respectively. Whereas a green color rules out such undesired effects (Table 1).
Table 1: Osiris property explorer toxicity predictions |
Figure 1: Structural sites responsible for the toxicities (Osiris predictions) |
The compound C5 is predicted as tumorigenic (high risk) by Osiris property explorer owing to the presence of ortho-dimethylphenyl site at the chalcone site. The compound C7 is predicted as mutagenic (high risk) by Osiris property explorer owing to the site 3-(furan-2-yl)prop-2-enoyl. As shown in Fig.1., the compound C12 is predicted as irritating (high risk) as well as it possesses deleterious reproductive effects (moderate risk) owing to the site 3-(4-methoxyphenyl)prop-2-enoyl. The compounds (C15- C28) possessing a 2-furyl substituent on the quinazolinone were predicted as irritants (high risk). The compound C21 besides irritant (high risk) is also predicted as mutagenic (high risk) owing to the site (2E)-3-(furan-2-yl)prop-2-enoyl. The compound 35 is also mutagenic (high risk) because of this site. The compound C33 besides being irritant (high risk) is also predicted as mutagenic (high risk) owing to the site 4-methylphenyl and is also tumorigenic (medium risk) because of its 3,4,5-trimethylphenyl site. The compounds C12, C26, and C40 besides irritant (high risk) are also predicted to have a reproductive effect (medium risk) owing to the site (2E)-3-(4-methoxyphenyl)prop-2-enoyl.
Osiris property explorer drug score predictions
The drug score value combines all other predictions into one total. The drug score value takes into account several parameters viz. cLogP, logS, molweight, drug-likeness, and probable toxicity (mutagenicity, tumorigenicity, irritating effects, reproductive effects). The Osiris property explorer predicted a higher drug score for erlotinib (0.38) than afatinib (0.24), this prediction complies with the conclusion of the comparative clinical trial phase-3 studies between erlotinib and afatinib that concluded that the incidence of stomatitis and diarrhea, was more with afatinib when compared with erlotinib16 . In the following table (Table 2), the ligands are arranged in descending order of their respective drug scores.
Table 2: Osiris property explorer drug score prediction
Compound Code |
cLogP |
Solubility |
MW |
TPSA |
Drug likeness |
Drug score |
C6 |
4.6 |
-6.12 |
429 |
62.63 |
5.49 |
0.42 |
C9 |
5.25 |
-6.62 |
444 |
69.97 |
5.08 |
0.35 |
C10 |
5.18 |
-6.63 |
474 |
79.2 |
5.59 |
0.33 |
C34 |
5.2 |
-6.85 |
463 |
62.63 |
5.39 |
0.33 |
C11 |
5.11 |
-6.65 |
504 |
88.43 |
6.18 |
0.32 |
C1 |
5.6 |
-6.91 |
428 |
49.74 |
1.84 |
0.3 |
C13 |
5.46 |
-6.95 |
488 |
68.2 |
6.95 |
0.3 |
C20 |
3.79 |
-5.8 |
419 |
75.77 |
4.79 |
0.3 |
C2 |
5.94 |
-7.26 |
442 |
49.74 |
3.5 |
0.29 |
C14 |
5.39 |
-6.97 |
518 |
77.43 |
7.92 |
0.28 |
C37 |
5.86 |
-7.35 |
478 |
69.97 |
4.96 |
0.27 |
C3 |
6.36 |
-7.41 |
456 |
49.74 |
4.68 |
0.26 |
C8 |
6.21 |
-7.66 |
462 |
49.74 |
5.6 |
0.26 |
C38 |
5.79 |
-7.37 |
508 |
79.2 |
5.47 |
0.26 |
C4 |
6.29 |
-7.6 |
456 |
49.74 |
2.27 |
0.25 |
C23 |
4.44 |
-6.3 |
434 |
83.11 |
4.43 |
0.25 |
C29 |
6.21 |
-7.65 |
462 |
49.74 |
1.78 |
0.25 |
C39 |
5.72 |
-7.39 |
538 |
88.43 |
6.07 |
0.25 |
C24 |
4.37 |
-6.32 |
464 |
92.34 |
5 |
0.24 |
C30 |
6.55 |
-7.99 |
476 |
49.74 |
3.38 |
0.24 |
C41 |
6.07 |
-7.68 |
522 |
68.2 |
6.84 |
0.24 |
C7 |
4.79 |
-6.59 |
418 |
62.88 |
5.08 |
0.23 |
C25 |
4.3 |
-6.33 |
494 |
101.5 |
5.56 |
0.23 |
C42 |
6 |
-7.7 |
552 |
77.43 |
7.83 |
0.23 |
C27 |
4.65 |
-6.63 |
478 |
81.34 |
6.38 |
0.22 |
C31 |
6.97 |
-8.15 |
490 |
49.74 |
4.57 |
0.22 |
C36 |
6.81 |
-8.38 |
496 |
49.74 |
4.75 |
0.22 |
C15 |
4.79 |
-6.59 |
418 |
62.88 |
1.28 |
0.21 |
C28 |
4.58 |
-6.65 |
508 |
90.57 |
7.29 |
0.21 |
C32 |
6.89 |
-8.34 |
490 |
49.74 |
2.16 |
0.21 |
C16 |
5.13 |
-6.94 |
432 |
62.88 |
2.85 |
0.2 |
C17 |
5.55 |
-7.1 |
446 |
62.88 |
4.05 |
0.19 |
C22 |
5.39 |
-7.33 |
452 |
62.88 |
4.92 |
0.19 |
C35 |
5.39 |
-7.33 |
452 |
62.88 |
4.95 |
0.19 |
C26 |
4.72 |
-6.61 |
448 |
72.11 |
4.49 |
0.18 |
C18 |
5.48 |
-7.28 |
446 |
62.88 |
1.61 |
0.17 |
C21 |
3.98 |
-6.28 |
408 |
76.02 |
4.21 |
0.17 |
C5 |
6.63 |
-7.94 |
470 |
49.74 |
4.98 |
0.15 |
C12 |
5.53 |
-6.93 |
458 |
58.97 |
5.12 |
0.15 |
C40 |
6.14 |
-7.67 |
492 |
58.97 |
4.99 |
0.12 |
C19 |
5.82 |
-7.63 |
460 |
62.88 |
4.32 |
0.1 |
C33 |
7.24 |
-8.68 |
504 |
49.74 |
3.61 |
0.06 |
Erlotinib Reference |
3.07 |
-3.53 |
393 |
74.73 |
-6.73 |
0.38 |
Afatinib Reference |
3.64 |
-5.48 |
485 |
88.61 |
-4.11 |
0.24 |
Naquotinib Reference |
2.54 |
-3.51 |
563 |
120.1 |
0.72 |
0.11 |
Osiris property explorer predicts that the compound C6 has the maximum drug score (0.42) among all the investigated compounds in the compound library and it is the only compound that has a drug score greater than the reference erlotinib (0.38; first generation EGFR inhibitor). In the library, eighteen compounds (C1- C4, C6, C8- C11, C13, C14, C20, C23, C29, C34, C37- C39) have greater drug score than the reference afatinib (0.24; second generation EGFR inhibitor). Except for C19 and C33, the other ligands have a greater drug score than naquotinib (0.11; third generation EGFR inhibitor).
Swiss ADME predictions for absorption, P-gp substrate, CYP enzymes inhibition
P-glycoprotein is a membrane transporter pump that is one of the main energy-dependent efflux mechanisms. P-glycoprotein effluxes many anticancer drugs out of the cells which can substantially reduce or demolish the activity and is one of the important reasons for drug resistance17. None of the ligands was found to be a P-glycoprotein substrate (Table 3), hence, the virtual investigations predict that none of the lignads would be effluxed by P-glycoprotein, from the tumor cells.
CYP superfamily enzymes are mainly expressed in the liver. Cytochrome P450 (CYP) enzymes play a primary role in metabolic phase-1 oxidative reactions that render the xenobiotics (drugs) hydrophilic by introducing polar handles and make them eligible for phase-2 metabolic reactions which subsequently make the drug pharmacodynamically neutral ( i.e. termination of biological activity) and facilitates its renal clearance. CYP1A2 is involved in the metabolism of about 10% of clinically used drugs, except the ligand C21 none of the ligands is its inhibitors 18. It implies that except the ligand C21 (Table 3), no other ligand in the library slows down the metabolism of the drugs primarily metabolized by CYP1A2. In the present study, all the ligands (C1-C42) are found to be the inhibitors of the CYP2C19 enzyme (Table 3). Proguanil, rabeprazole, omeprazole, lansoprazole, pantoprazole, amitriptyline, clomipramine, amitriptyline, diazepam, S-mephenytoin, phenobarbitone, cyclophosphamide, clopidogrel, nelfinavir, and warfarin are among the drugs that primarily undergo oxidative metabolism by CYP2C19 enzyme19. Hence, all the ligands would slow down the metabolism of these drugs and slow down their renal clearance. CYP2C9 is one of the most important CYP superfamily enzymes, it has been estimated to contribute to the oxidative phase-1 metabolism of approximately 15% of all therapeutic agents that are subjected to biotransformation by CYP enzymes. The ligands C2-C5, C8, C19, C29-C33, C36, and C37 being non-inhibitors of CYP2C9 (Table 3) would not slow down the metabolism and renal excretion of those therapeutic agents (e.g. warfarin, ibuprofen, etc) primarily metabolized by CYP2C9 enzyme 20. In the metabolism of a large number of clinically important drugs, the CYP2D6 enzyme act as a catalyst (~20% of commonly used therapeutic agents) including neuroleptics, antidepressants, antiarrhythmics, opioids, and lipophilic β-adrenoceptor blockers. In the present study, none of the ligands is found to be the inhibitor of CYP2D6 (Table 3) which implies that the ligands would not slow down the metabolism of the drugs primarily metabolized by CYP2D6 21. CYP3A4 enzyme having unusually low substrate specificity is involved in the metabolism of about 60% of currently known therapeutics. The ligands C12-C14, C20, C21, C26-C28, C41 and C42 being inhibitors of CYP3A4 would slow down the metabolism of the varieties of the drugs which are metabolized by the CYP3A4 enzyme22.
Table 3: Swiss ADME predictions for absorption, P-gp substrate, CYP enzymes inhibition
Compound Code |
Gastrointestinal absorption |
Blood –brain-barrier permeant |
Pgp substrate |
Cyp1A2 inhibitor |
Cyp2C19 inhibitor |
Cyp2C9 inhibitor |
Cyp2D6 inhibitor |
Cyp3A4 inhibitor |
Bioavail ability Score |
Alert for PAINS |
C1 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C2 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C3 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C4 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C5 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C6 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C7 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C8 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C9 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C10 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C11 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C12 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C13 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C14 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C15 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C16 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C17 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C18 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C19 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C20 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C21 |
High |
No |
No |
Yes |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C22 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C23 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C24 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C25 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C26 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C27 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C28 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C29 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C30 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C31 |
Low |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C32 |
Low |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C33 |
Low |
No |
No |
No |
Yes |
No |
No |
No |
0.17 |
Zero |
C34 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C35 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C36 |
Low |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C37 |
High |
No |
No |
No |
Yes |
No |
No |
No |
0.55 |
Zero |
C38 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C39 |
Low |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C40 |
High |
No |
No |
No |
Yes |
Yes |
No |
No |
0.55 |
Zero |
C41 |
High |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
C42 |
Low |
No |
No |
No |
Yes |
Yes |
No |
Yes |
0.55 |
Zero |
Erlotinib Reference |
High |
Yes |
No |
Yes |
Yes |
Yes |
Yes |
Yes |
0.55 |
Zero |
Afatinib Reference |
High |
No |
Yes |
No |
Yes |
Yes |
Yes |
Yes |
0.55 |
Zero |
Naquotinib Reference |
High |
No |
Yes |
No |
No |
No |
No |
Yes |
0.17 |
One |
Docking, Docked pose and Binding with target protein:
The respective .dlg files included the ligand’s inhibition constant (KI) and free energy of binding (BE). The complex of the target protein and the ligand’s best-fit pose was saved in the .pdb file . The Discovery studio software was used to see the binding posture of the ligand and its interactions with target protein.
In Molecular docking studies of 42 ligands, It was observed that all the ligands occupied the reported binding site of the target molecular protein5Y9T23 (Fig.2.). All the designed ligands exhibited good affinity for the molecular target protein 5Y9T as the free binding energies were observed in the range of -45.44 KJ/mol to -34.64 KJ/mol, the inhibition constants of the ligands were observed in the range of 11.04 nM to 853.47 nM (Table 4).
Table 4: Docking data
Compound |
Ar |
Docking Rank |
Constant of Inhibition (IC) (in nM) |
Binding Energy (in KJ/mol) |
Hydrogen bonds |
Hydrogen bonding |
Other interacting amino acids |
No. of amino acids interacting |
C1 |
Phenyl |
22 |
127.35 |
-39.37 |
1 |
MET793(2.28) |
LEU718, |
10 |
C2 |
4-Tolyl |
13 |
49.16 |
-41.71 |
1 |
MET793(1.80) |
ASP855, |
10 |
C3 |
4-Ethylphenyl |
11 |
40.19 |
-42.22 |
1 |
MET793(1.81) |
MET790, |
10 |
C4 |
3,4-Dimethylphenyl |
6 |
21.64 |
-43.76 |
1 |
MET793(1.76) |
PRO794, |
11 |
C5 |
3,4,5-Trimethylphenyl |
3 |
14.56 |
-44.73 |
1 |
MET793(1.68) |
ALA743, |
10 |
C6 |
Pyridin-3-yl |
36 |
346.06 |
-36.86 |
1 |
MET793(2.44) |
LEU718, |
8 |
C7 |
Furan-2-yl |
38 |
471.17 |
-36.11 |
1 |
MET793(2.38) |
ALA743, |
8 |
C8 |
4-Chlorophenyl |
16 |
78.74 |
-40.54 |
1 |
MET793(2.33) |
LEU844, |
7 |
C9 |
4-Hydroxyphenyl |
28 |
254.35 |
-37.66 |
1 |
MET793(2.07) |
LEU844, |
9 |
C10 |
4-Hydroxy-3-methoxyphenyl |
15 |
69.25 |
-40.88 |
3 |
LYS716(2.10), |
LEU718, |
10 |
C11 |
4-Hydroxy-3,5-dimethoxyphenyl |
14 |
60.47 |
-41.21 |
3 |
LYS716(2.24), |
LEU718, |
10 |
C12 |
4-Methoxyphenyl |
19 |
116.31 |
-39.58 |
1 |
MET793(2.17) |
LEU844, |
9 |
C13 |
3,4-Dimethoxyphenyl |
18 |
89.47 |
-40.25 |
3 |
LYS716(4.91), |
LEU718, |
9 |
C14 |
3,4,5-Trimethoxyphenyl |
17 |
84.59 |
-40.38 |
3 |
LYS716(2.01), |
LEU718, |
11 |
C15 |
Phenyl |
32 |
286.74 |
-37.36 |
1 |
MET793(1.89) |
PRO794, |
8 |
C16 |
4-Tolyl |
8 |
31.66 |
-42.8 |
2 |
MET793(1.84), |
PRO794, |
13 |
C17 |
4-Ethylphenyl |
7 |
28.28 |
-43.1 |
2 |
MET793(1.80), |
PRO794, |
10 |
C18 |
3,4-Dimethylphenyl |
4 |
14.5 |
-44.73 |
2 |
MET793(1.73), |
MET790, |
11 |
C19 |
3,4,5-Trimethylphenyl |
1 |
11.04 |
-45.44 |
2 |
MET793(1.87), |
PRO794, |
12 |
C20 |
Pyridin-3-yl |
33 |
293.59 |
-37.28 |
1 |
LYS745(1.83) |
LEU844, |
9 |
C21 |
Furan-2-yl |
42 |
853.47 |
-34.64 |
1 |
MET793(1.97) |
LYS728, |
10 |
C22 |
4-Chlorophenyl |
26 |
230.48 |
-37.87 |
2 |
MET793(1.99), |
SER720, |
14 |
C23 |
4-Hydroxyphenyl |
41 |
589.14 |
-35.56 |
1 |
MET793(2.34) |
ALA743, |
9 |
C24 |
4-Hydroxy-3-methoxyphenyl |
40 |
561.85 |
-35.69 |
1 |
MET793(2.52) |
LEU718, |
9 |
C25 |
4-Hydroxy-3,5-dimethoxyphenyl |
25 |
173.71 |
-38.58 |
3 |
LYS728(2.64), |
LEU718, |
10 |
C26 |
4-Methoxyphenyl |
34 |
306.24 |
-37.2 |
1 |
MET793(2.11) |
ALA743, |
10 |
C27 |
3,4-Dimethoxyphenyl |
30 |
265.5 |
-37.53 |
3 |
GLY796(2.70), |
LEU718, |
9 |
C28 |
3,4,5-Trimethoxyphenyl |
21 |
118.48 |
-39.54 |
2 |
LYS716(2.42), |
LEU844, |
10 |
C29 |
Phenyl |
27 |
233.16 |
-37.87 |
1 |
MET793(3.00) |
VAL726, |
10 |
C30 |
4-Tolyl |
10 |
39.61 |
-42.26 |
1 |
MET793(1.97) |
LEU792, |
14 |
C31 |
4-Ethylphenyl |
9 |
35.56 |
-42.51 |
1 |
MET793(2.00) |
LEU792, |
12 |
C32 |
3,4-Dimethylphenyl |
5 |
15.89 |
-44.52 |
1 |
MET793(1.95) |
LEU718, |
13 |
C33 |
3,4,5-Trimethylphenyl |
2 |
13.04 |
-45.02 |
1 |
MET793(1.68) |
ALA743, |
10 |
C34 |
Pyridin-3-yl |
35 |
322.13 |
-37.07 |
1 |
MET793(2.71) |
LEU844, |
9 |
C35 |
Furan-2-yl |
39 |
479.18 |
-36.07 |
1 |
ARG841(5.47) |
ASP837, |
7 |
C36 |
4-Chlorophenyl |
29 |
261.09 |
-37.57 |
1 |
MET793(2.68) |
LEU844, |
9 |
C37 |
4-Hydroxyphenyl |
24 |
128.44 |
-39.33 |
1 |
MET793(2.14) |
LEU718, |
10 |
C38 |
4-Hydroxy-3-methoxyphenyl |
37 |
356.01 |
-36.82 |
1 |
LYS716(2.77) |
LEU844, |
8 |
C39 |
4-Hydroxy-3,5-dimethoxyphenyl |
20 |
116.49 |
-39.58 |
2 |
MET793(2.36), |
LEU718, |
11 |
C40 |
4-Methoxyphenyl |
31 |
273.28 |
-37.45 |
1 |
MET793(2.65) |
GLY719, |
8 |
C41 |
3,4-Dimethoxyphenyl |
12 |
43.45 |
-42.01 |
1 |
MET793(1.86) |
ALA743, |
12 |
C42 |
3,4,5-Trimethoxyphenyl |
23 |
125.89 |
-39.37 |
3 |
LYS716(2.36), |
LEU718, ASP855, |
9 |
Reference Erlotinib |
2.37 micro-molar |
-32.13 |
3 |
LYS860 |
GLU758, |
7 |
||
Reference Afatinib |
1.30 micro-molar |
-33.60 |
2 |
MET793(1.87), |
ALA743, LEU792, |
9 |
||
Reference Naquotinib |
1.55 micro-molar |
-33.18 |
3 |
ASP855(3.01), |
LEU844, |
10 |
Figure 2: Ligands binding with EGFR (5Y9T) (a) C19 (b) C33 (c) C6 (d) C9 |
Figure 3: Ligands interactions with EGFR (5Y9T) (a) C19 (b) C33 (c) C6 (d) C9 |
C19 and C33 interactions (most potent ligands)
Insilico docking studies revealed that the compound C19 has the highest affinity (binding energy = -45.44 KJ/mol and inhibition constant = 11.04 nM) and the compound C33 was observed as 2nd most potent ligand (binding energy = -45.02 KJ/mol and inhibition constant = 13.04 nM) for the molecular target protein 5Y9T. The ligand-target interaction studies (Fig.3.), revealed that in both the highly potent ligands (compoundC19 & compound C33)the carbonyl oxygen of the chalcone site acts as an H-bond acceptor with the MET793 amino acid while in compound C19 the oxygen of furan ring also acts as H-bond acceptor with LYS745 amino acid. The quinazolinone ring system makes Pi-Pi and Pi-sigma bindings with PHE723 and VAL726 respectively. The aromatic ring (furan in C19 and phenyl in C33) makes Pi-anion interaction with the carboxylate site of ASP855. The phenyl ring at N-3 of quinazolinone makes Pi-sigma and Pi-alkyl bonding with VAL726 and ALA743, respectively. The “3,4,5-trimethylphenyl” substituent on the beta-unsaturated carbon (w.r.t. carbonyl group of chalcone) makes Pi-lone pair binding with PRO794, Pi-sigma binding with LEU718 and alkyl/Pi-alkyl type of bindings with LYS728 and LEU792.
C6 (highest drug score) & C9 (2nd highest drug score) interactions
Insilico docking studies revealed that the compound C6 has the binding energy = -45.44 KJ/mol and inhibition constant = 11.04 nM) while the compound C9 has the binding energy = -45.02 KJ/mol and inhibition constant = 13.04 nM, for the molecular target protein 5Y9T. The ligand-target interaction studies (Fig.3.) revealed that in both the high drug score ligands (compound C6 & compound C9)the carbonyl oxygen of the chalcone site acts as an H-bond acceptor with the MET793 amino acid. The quinazolinone ring system makes Pi-sigma interaction with VAL726. 2-Phenyl substituent of quinazolinone makes Pi-anion interaction with the carboxylate site of ASP855. The phenyl ring at N-3 of quinazolinone makes a Pi-alkyl bond with LEU844. Aromatic substituents (pyridinyl in C6 and 4-hydroxyphenyl in C9) at the beta-unsaturated carbon (w.r.t. carbonyl group of chalcone) make Pi-lone pair interaction with PRO794.
The review of the literature showed that the requirement to treat NSCLC with T790M mutant EGFR remains a significant unmet need. Madhavi et al., has reported certain “quinazoline integrated chalcones derivatives” as strong cytotoxic agents against cancer cell lines (colorectal, breast, melanoma, and lung cancer), with an IC50 range of 0.10 to 0.19 mM5. The molecular docking studies and invitro cytotoxic studies of quinazolinone-naphthyl chalcones and 2-methoxyquinazolinone chalcones have enlightened that quinazolinone-chalcone conjugates are potent anticancer agents and can be the promiscuous chemical entities to target T790M mutated EFGR10,11. Le et al., discovered that the methyl substituted quinazolinone derivatives were potent inhibitors of wild-type EGFR, with IC50 (mM) values ranging from 0.01-0.5424. Zhang et al. observed that several analogs of 2-phenoxymethylquinazolinone were potent EGFR (wild-type) inhibitors, with IC50 values ranging from 0.047 to 2.71 (mM)25. The third-generation EGFR TKIs, such as naquotinib, osimertinib or lazertinib, raised hope for NSCLC patients with T790M mutation; however, these medications are associated with substantial side effects, and during treatment, patients develop EGFR resistance15,26. Thus, there is still a great unmet need for the treatment of NSCLC with the T790M mutant EGFR26. The present work involves the insilico exploration of low molecular weight chemical ligands for their drug score, toxicity, pharmacokinetic parameters, and their ability to inhibit mutated EGFR. As the explored ligands were not found to be the P-glycoprotein substrates (Table 3), none of the ligands would be effluxed by P-glycoprotein, from the tumor cells. The insilico exploration revealed that all the tested ligands are potent, have good target affinity, and have a good bioavailability Score. Moreover, the tested ligands have good synthesis feasibility10,11. The present research can be taken forward for the synthesis of ligands with high drug scores (e.g. C6 & C9) and good target affinity (e.g. C19 & C33) and their invitro/invivo evaluation for EGFR (T790M mutated) inhibition without wasting time, material, and money. Hence, the present insilico exploration gives momentum to the discovery of the drugs that can target mutated EGFR, to defeat resistance hindrances.
Acknowledgement
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Conflict of Interest
The author declare that we have no conflict of interest.
Funding Sources
There is no funding Sources
References
- Li, C.; Lei, S.; Ding, L.; Xu, Y.; Wu, X.; Wang, H.; Zhang, Z.; Gao, T.; Zhang, Y.; Li, L. Chin Med J (Engl), 2023 ,136(13),1583-1590, doi: 10.1097/CM9.0000000000002529.
CrossRef - Rebecca, L.S.; Kimberly, D.M.; Hannah, E.F.; Ahmedin, J. CA Cancer J Clin, 2022, 72, 7–33.
- Altamimi, A.S.; El-Azab, A.S.; Abdelhamid, S.G.; Alamri, M.A.; Bayoumi, A.H.; Alqahtani, S.M.; Alabbas, A.B.; Altharawi, A.I.; Alossaimi, M.A.; Mohamed, M.A. Molecules, 2021,26(10),2992, doi: 10.3390/molecules26102992.
CrossRef - Ouyang, Y. Li. J.; Chen, X. Fu. X.; Sun, S.; Wu, Q. Biomolecules, 2021, 11(6), 894, doi: 10.3390/biom11060894.
CrossRef - Madhavi, S.; Sreenivasulu, R.; Yazala, J.P.; Raju, R.R. Saudi Pharm J, 2017, 25,275-279.
CrossRef - Abbas, S.H.; Abd El-Hafeez, A.A.; Shoman, M.E.; Montano, M.M.; Hassan, H.A. Bioorg Chem, 2019, 82,360-377.
CrossRef - Zhou, W.; Zhang, W.; Peng, Y.; Jiang, Z.H.; Zhang, L.; Du, Z. Molecules, 2020, 25, 3180-3197.
CrossRef - Kasetti, A.B.; Singhvi, I.; Nagasuri, R.; Bhandare, R.R.; Shaik, A.B. Molecules. 2021, 26, 2847-2862.
CrossRef - Alam, M.J.; Alam, O.; Perwez, A.; Rizvi, M.A.; Naim, M.J.; Naidu, V.G.M.; Imran, M.; Ghoneim, M.M.; Alshehri, S.; Shakeel, F. Pharmaceuticals (Basel),2022,15, 280-299.
CrossRef - Arora, P. K.; Kumar, S.; Bansal, S. K.; Sharma, P. C. Orient J Chem, 2023,39(2),231-245.
CrossRef - Arora, P. K.; Kumar, S.; Bansal, S. K.; Sharma, P. C. Rasayan J. Chem, 2023, 16(3), 1104-1115.
CrossRef - Bao, S.M.; Hu, Q.H.; Yang, W.T.; Wang, Y.; Tong, Y.P.; Bao, W.D. Anticancer Agents Med Chem,2019,19, 984-991.
CrossRef - Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Signal Transduct Target Ther; 2020, 5, 22-34.
CrossRef - Peng, W.; Yao, C.; Pan, Q.; Zhang, Z.; Ye, J.; Shen, B.; Zhou, G.; Fang, Y. Front Oncol, 2023,13,1120278, doi: 10.3389/fonc.2023.1120278.
CrossRef - Boch, T.; Köhler, J.; Janning, M.; Loges, S. Cancer Biol Med, 2022 ,19(11),1543–64, doi: 10.20892/j.issn.2095-3941.2022.0540.
CrossRef - Soria, J.C.; Felip, E.; Cobo, M.; Lu, S.; Syrigos, K.; Lee, K.H.; Göker, E.; Georgoulias, V.; Li, W.; Isla, D.; Guclu, S.Z.; Morabito, A.;Min, Y.J.; Ardizzoni, A.; Gadgeel, S.M.; Wang, B.; Chand, V.K.; Goss, G.D. Lancet Oncol, 2015, 16,897-907.
CrossRef - Heming, C. P.; Muriithi ,W.; Wanjiku, M. L.; Niemeyer, F. P.; Moura-Neto, V.; Aran, V. Heliyon, 2022 ,8(10),e11171. doi: 10.1016/j.heliyon.2022.e11171.
CrossRef - Esteves, F.; Almeida, C.M.M.; Silva, S.; Saldanha, I.; Urban, P.; Rueff, J.; Pompon, D.; Truan, G.; Kranendonk, M. Biomolecules, 2023 ,13(7),1083, doi: 10.3390/biom13071083.
CrossRef - Botton, M.R.; Whirl-Carrillo, M.; Del, T. A.L.; Sangkuhl, K.; Cavallari, L.H.; Agúndez, J.A.G.; Duconge, J.; Lee, M.T.M.; Woodahl, E.L.; Claudio-Campos, K.; Daly, A.K.; Klein, T.E,; Pratt, V.M.; Scott, S.A.; Gaedigk, A. Clin Pharmacol Ther, 2021 ,109(2),352-366, doi: 10.1002/cpt.1973.
CrossRef - Sangkuhl, K.; Claudio-Campos, K.; Cavallari, L.H.; Agundez, J.A.G.; Whirl-Carrillo, M.; Duconge, J.; Del Tredici, A.L.; Wadelius, M; Rodrigues, B. M.; Woodahl, E.L.; Scott, S.A.; Klein, T.E.; Pratt, V.M.; Daly, A.K.; Gaedigk, A. Clin Pharmacol Ther, 2021, 110(3),662-676, doi: 10.1002/cpt.2333.
CrossRef - Taylor, C.; Crosby, I.; Yip, V.; Maguire, P.; Pirmohamed, M.; Turner, R.M. Genes (Basel), 2020,11(11),1295, doi: 10.3390/genes11111295.
CrossRef - Klyushova, L.S.; Perepechaeva, M.L.; Grishanova, A.Y. Biomedicines, 2022 ,10(11),2686, doi: 10.3390/biomedicines10112686.
CrossRef - Hirano, T.; Yasuda, H.; Hamamoto, J.; Nukaga, S.; Masuzawa, K.; Kawada, I.; Naoki, K.; Niimi, T.; Mimasu, S.; Sakagami, H.; Soejima, K.; Betsuyaku, T. Mol Cancer Ther,2018, 17, 740-750.
CrossRef - Le, Y.; Gan, Y.; Fu, Y.; Liu, J.; Li,W.; Zou, X.; Zhou, Z.; Wang, Z.; Ouyang, G.; Yan, L. J Enzyme Inhib Med Chem, 2020, 35, 555-564, doi:10.1080/14756366.2020.1715389.
CrossRef - Zhang,Y.; Wang, Q.; Li, L.; Le, Y.; Liu, L.; Yang, J.; Li, Y.; Bao, G.; Yan, L. J Enzyme Inhib Med Chem, 2021, 36,1205-1216.
CrossRef - Agema, B.C.; Veerman; G.D.M.; Steendam, C.M.J.; Lanser, D.A.C.; Preijers, T.; Van der Leest, C.; Koch, B.C.P.; Dingemans, A.C.; Mathijssen, R.H.J.; Koolen, S.L.W. Ther Adv Med Oncol,2022, 14,1-10.
CrossRef
This work is licensed under a Creative Commons Attribution 4.0 International License.