Synthesis, and Evaluation of Novel N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino) acetamide derivatives by In silico investigations as an anticancer scaffold

 

Dipanjan Karati1*, Shankar Thapa2, Mahalakshmi Suresha Biradar3

1Department of Pharmaceutical Technology, School of Pharmacy,

Techno India University, Kolkata, West Bengal, 700091, India.

2Department of Pharmaceutical Technology, Department of Pharmaceutical Technology,

Universal College of Medical Science, Siddharthnagar, 32900, Nepal.

3Department of Pharmaceutical Technology, Al-Ameen College of Pharmacy, Bengaluru, 560027, India.

*Corresponding Author E-mail: karatibabai@gmail.com, tshankar551@gmail.com, mahalakshmisb12@gmail.com

 

ABSTRACT:

This study demonstrates the scope for N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino)acetamide to develop as promising anticancer agents. Several N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino)acetamide derivatives were synthesized by reflux method and assessed their anticancer activity by in silico research. The yield of the compounds was moderate to high. The functional groups were measured by the IR ranges at 3300cm-1 (NH), 1656 cm-1 (CO) and 1180 (C=S) wavelength. The synthesized compounds have high binding energy against 7A2A protein as an EGFR inhibitor. Compound C2 and C4 showed best affinity of -7.7 and -7.2kcal/mol, correspondingly. The molecules have been tested for their toxicity. The result shows that all the compounds have affinity towards the EGFR protein. The toxicity prediction suggests that all the synthesized substances are relatively safe, having a low likelihood of causing harm at the given doses.

 

KEYWORDS: Pyrimidine, Acetamide derivatives, Molecular docking, Anticancer, EGFR.

 

 


1.       INTRODUCTION:

Cancer remains a global health crisis, with over 19 million new cases and nearly 10 million deaths annually.1 Among these, cancers involving dysregulated signaling pathways like the Epidermal Growth Factor Receptor (EGFR) pathway account for a significant portion.2 EGFR, a transmembrane receptor tyrosine kinase, is crucial for cell proliferation and survival, and its abnormal activation is linked to various cancers, including lung, colorectal, and head and neck cancers.3-6

 

While targeted EGFR therapies have advanced, drug resistance and side effects remain challenges. Small molecule inhibitors, such as Gefitinib and Erlotinib, block the ATP-binding site of EGFR, showing promising therapeutic effects.7-8 Dihydropyrimidin derivatives, in particular, have gained attention for their ability to inhibit EGFR signaling and tumor growth due to their structural versatility.9-11 Designing novel scaffolds to enhance efficacy and reduce side effects requires understanding the molecular interactions of EGFR inhibitors.12-14 Exploiting dihydropyrimidin scaffolds has led to compounds with varying potency, offering insights into structure-activity relationships and optimization strategies.15

 

2.       MATERIALS AND METHODS:

2.1 General Information:

All the chemicals employed in the experiments were of the analytical chemical grade. Loba Chemical supplied extra pure methanol, benzaldehyde, chloro acetyl chloride, malononitrile, substituted aniline derivatives, and thiourea.

 

2.2 Chemistry:

2.2.1 General procedure for the synthesis of compound A: A mixture of 2 mmol/L of benzaldehyde, 2 mmol/L of malononitrile, 3 mmol/L of thiourea, and 0.8 mmol/L of NH4Cl was heated in an oil bath with stirring for 4 hours at 1100 C in a 100 mL round-bottom flask. TLC was used to track the reaction's progress. The solid product was obtained by cooling the reaction liquid to room temperature and then pouring it over crushed ice after the reaction had finished.

 

2.2.2 Synthesis of Compound B: In order to create compound B, one mole of compound A was added to 100millilitres of toluene solvent in a 250 milliliter, clean, round-bottomed flask. The mixture was well mixed and heated using reflux. Next, one mole of dissolved chloroacetyl chloride was added gradually. The entire reaction material was then refluxed for two hours. Following this process, the precipitate was filtered, dried, and then recrystallized using pure ethyl alcohol.

 

2.2.3 Synthesis of Compound C1-C5: In the presence of K2CO3 (1 mols), an equimolar amount of compound B (1mols) and substituted aniline (1mols) in chloroform solvent were heated under reflux (figure 1). The residue was agitated with water after the excess solvent was extracted using a vacuum and it was subsequently filtered and cleaned with 2% NaHCO3 and water. After that, 100% ethyl alcohol was used to recrystallize the crude product.


 

Figure 1: Synthetic scheme of compounds C1-C5

 


2.2.4 Compound C1: Yield 78%. IR (KBr, cm−1): 3414 (NH), 1667 (C = O), 2219 (CN), 1180 (C=S), 700 (C-Cl), 1390 (NO2). 1H NMR (DMSO-d6, 400 MHz) d (ppm): in table 1. MASS: 440.05 (C19H13ClN6O3S).

 

Table 1: NMR data[DMSO-d6, 400 MHz) d (ppm)] of Compound 1

Sr. No.

Type of H

Number of H

δ Value (ppm)

1.

H of NH

1

13.03

2.

H of NH

1

6.28

3.

H of NH

1

9.21

4.

Aromatic H

5

7.56-7.98

5.

H of CH2

2

3.77

6.

H of 1-benzene

3

6.98-7.58

 

2.2.5 Compound C2: Yield 80%. IR (KBr, cm−1): 3414 (NH), 1667 (C = O), 2219 (CN), 1180 (C=S), 700 (C-Cl), 1390 (NO2). 1H NMR has been depicted in table 2. MASS: 440.05 (C19H13ClN6O3S).

 

 

 

Table 2: NMR data of Compound 2

Sr. No.

Type of H

Number of H

δ Value(ppm)

1.

H of NH

1

13.03

2.

H of NH

1

5.74

3.

H of NH

1

9.21

4.

Aromatic H

5

7.56-7.98

5.

H of CH2

2

3.77

6.

H of 1-benzene

3

6.87-8.13

 

2.2.6 Compound C3: Yield 75%. IR (KBr, cm−1): 3414 (NH), 1667 (C = O), 2219 (CN), 1180 (C=S), 680 (C-Br). 1H NMR has been depicted in table 3. MASS: 439.01 (C19H14BrN5OS).

 

Table 3: NMR data of Compound 3

Sr. No.

Type of H

Number of H

δ Value(ppm)

1.

H of NH

1

13.03

2.

H of NH

1

6.28

3.

H of NH

1

9.21

4.

Aromatic H

5

7.56-7.98

5.

H of CH2

2

3.77

6.

H of 1-benzene

4

6.59-7.13

2.2.7 Compound C4: Yield 85%. IR (KBr, cm−1): 3414 (NH), 1667 (C = O), 2219 (CN), 1180 (C=S), 1390 (NO2). 1H NMR has been depicted in table 4. MASS: 406.08 (C19H14N6O3S).

 

Table 4: NMR data of Compound 4

Sr. No.

Type of H

Number of H

δ Value(ppm)

1.

H of NH

1

13.03

2.

H of NH

1

8.74

3.

H of NH

1

9.21

4.

Aromatic H

5

7.56-7.98

5.

H of CH2

2

3.77

6.

H of 1-benzene

4

7.08-8.20

 

2.2.8 Compound C5: Yield 83%. IR (KBr, cm−1): 3414 (NH), 1667 (C = O), 2219 (CN), 1180 (C=S), 1390 (NO2). 1H NMR has been depicted in table 5. MASS: 361.10 (C19H15N5OS).

 

Table 5: NMR data of Compound 5

Sr. No.

Type of H

Number of H

δ Value(ppm)

1.

H of NH

1

13.03

2.

H of NH

1

6.28

3.

H of NH

1

9.21

4.

Aromatic H

5

7.56-7.98

5.

H of CH2

2

3.77

6.

H of 1-benzene

5

6.67-7.08

 

2.3 Pharmacokinetic properties and drug likeliness studies:

The absorption, distribution, metabolism, and excretion (ADME) prediction and drug likeliness study were conducted using the Swiss ADME web server (http://www.swissadme.ch/index.php).16-17 Finally, results were analyzed to prioritize compounds with promising drug-like characteristics for further investigation.

 

2.4 Molecular docking:

2.4.1      Protein and ligand preparation:

The procedure for conducting molecular docking begins with the preparation of both protein and ligand structures. The protein structure of EGFR (PDB ID: 7A2A)18 was obtained from the Protein Data Bank, while ligand structures, specifically N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino) acetamide derivatives, are generated using a Chem Draw software 20.1.1.125.19 Open Babel 2.4.1 software was used to convert all drug structures from. cdx files to. sdf MDL MOL format in a single file.20 The ligands were prepared by minimizing its energy for the purpose of docking.21-22

 

2.4.2 Binding pocket and grid box generation:

Subsequently, using visualization software BIOVIA discovery studio 2021, the binding pocket of the protein around the co-crystal ligand was identified, and a grid box (x-axis = -16.304, y-axis = 10.562, and z-axis = 1.274) was defined around this region to guide the docking simulations. The dimensions of the grid box encompassed the binding pocket adequately while allowing ample space (center = 10) for ligand movement during docking (Figure 2).23-26

 

2.4.3 Docking process and result visualization:

AutoDock Vina 1.5.727-29 was then executed with the provided configuration files for each ligand, initiating docking calculations that explore various ligand orientations and conformations within the defined binding site of the protein. Upon completion, AutoDock Vina 1.5.7 generated output files (.pdbqt) containing predicted binding poses and corresponding binding affinities for each ligand. Finally, the docking results were analyzed and visualized using BIOVIA discovery studio 2021 software.30

 

2.4.4 Docking result validation:

Docking results were validated in PyMOL using the redocking principle. Co-crystal native ligand was removed from the protein, redocked into the protein structure, and poses compared. Binding affinities were assessed, and interactions visually inspected. Iterative refinements were made for accuracy, ensuring agreement with experimental data and known binding patterns, facilitating reliable predictions of ligand-protein interactions for further analysis and interpretation.31,32

 

2.5 Biological activity prediction:

Biological activity prediction was carried out using the PASS (Prediction of Activity Spectra for Substances) web server (https://way2drug.com/passonline/ predict.php).33-34

 

2.6 Toxicity prediction:

Toxicity prediction was performed using the ProTox-II web server (https://comptox.charite.de/protox3/) [35-36]. Initially, the chemical structures of the compounds were submitted to the server.

 

3.    RESULTS AND DISCUSSION:

3.1 Chemistry:

Using the IR ranges at 3300 cm-1 (NH), 1656 cm-1 (CO), and 1180 cm-1 (C=S) wavelength, the functional groups were determined. Compound C2 and compound C4 were found to be having least binding energy of -7.7 and -7.2 kcal/mol correspondingly. The synthetic compound exhibits a binding energy of -6.6 to -7.8 kcal/mol when compared to the standard medication. The amino acids Met793, Val726, Leu (718, 799, 844), Arg841, and Ala749 make up the co-crystal7G9, which is located close to the active site of the enzyme and could accommodate all five compounds (C1–C5) snugly. After closely examining the docked complexes, this was found. The potential amino acids Met793, Arg841, Asp800, and Cys797 are also involved in interaction. Compounds C2, C4, and their binding affinity of -7.7 and -7.6 kcal/mol, correspondingly, demonstrate their significant affinity for the EGFR active site. Through hydrogen bonds at 2.05 Å, the carbonyl group (C=O) of the molecule C2 interacts with Met793.

 

3.2      Pharmacokinetic properties and drug likeliness studies:

Table 6 presents various physicochemical and pharmacokinetic properties of five compounds (C1, C2, C3, C4, and C5), which are crucial for understanding their behavior in biological systems. The Log Po/w values represent the partition coefficient between octanol and water, which indicates the lipophilicity of the compounds. The LogPo/w values range from 1.28 to 2.21, suggesting that the compounds exhibit varying degrees of lipophilicity, with C3 being the most lipophilic and C4 being the least.37

 

The "HBA" (Hydrogen Bond Acceptors) and "HBD" (Hydrogen Bond Donors) represent the number of hydrogen bond acceptor and donor groups present in each compound, respectively. These properties are crucial for understanding a compound's ability to interact with other molecules through hydrogen bonding, which can impact its binding affinity and biological activity. The "RB" (Rotatable Bonds) indicates the number of rotatable bonds in each compound. Rotatable bonds are important for assessing the flexibility of a molecule, which can influence its conformational changes and interactions with biological targets. The "TPSA" (Topological Polar Surface Area) provides information about the surface area of a compound that is polar or capable of forming hydrogen bonds. This property is relevant for predicting a compound's membrane permeability and oral bioavailability. The "Lipinski’s Rule Violation" indicates whether each compound violates Lipinski's Rule of Five, which is a widely used guideline in drug discovery for predicting a compound's likelihood of being orally bioavailable.38 Overall, none of the compounds in the table violate Lipinski's Rule.


 

Table 6: Pharmacokinetic properties of compounds

Compound code

LogPo/w

LogS

Log Kp

GI abs.

BS

BBB

Pg-S

CYP1A2 inh.

C1

1.66

Poorly soluble

-6.95

High

0.55

No

No

No

C2

1.66

Poorly soluble

-6.95

High

0.55

No

No

No

C3

2.21

Poorly soluble

-6.77

High

0.55

No

No

Yes

C4

1.28

Poorly soluble

-6.79

High

0.55

No

No

No

C5

1.66

Poorly soluble

-6.79

High

0.55

No

No

Yes

LogPo/w (Consensus) = Lipophilicity, LogS = Water solubility, Log Kp= Skin permeability, BS= Bioavailability score, GI abs. = Gastrointestinal Absorption, Pg-S = Pg substrate, CYP1A2 inh. = Cytochrome P1A2 inhibitor.

 

Table 7: Drug likeliness studies of compounds

Compound code

Ml.Wt.

HBA

HBD

RB

TPSA

SA

Lipinski’s rule Violation

C1

424.80

6

3

7

132.70

3.15

No

C2

424.80

6

3

7

132.70

3.13

No

C3

424.25

4

3

6

86.88

2.95

No

C4

390.35

6

3

7

132.70

3.09

No

C5

345.35

4

3

6

86.88

2.91

No

Ml.Wt.= Molecular Weight, HBA= Hydrogen Bond Acceptor, HBD = Hydrogen Bond Donor, RB = Rotatable Bond, TPSA = Topological Polar Surface Area, SA = Synthetic Accessibility.

 


3.3  Molecular docking:

The choice of EGFR as the docking target was made because the crystal structure was available and it complexed with Spebrutinib (7G9). The resemblance in structure between the co-crystal ligand Spebrutinib (7G9) (Figure 3) and our proposed N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino) acetamide derivatives has sparked our interest in selecting the protein. Compound C2 and compound C4 had the lowest binding energies of -7.7 and -7.2 kcal/mol, respectively. The synthesized molecule exhibits a binding energy ranging from -6.6 to -7.8 kcal/mol, as stated in table 8. All the compounds (C1-C5) can tightly fit into the active site of the enzyme near the co-crystal 7G9, composed of the amino acids Met793, Val726, Leu (718, 799, 844), Arg841, and Ala749. This finding was made by meticulous analysis of the docked complexes. Furthermore, the amino acid residues Met793, Arg841, Asp800, and Cys797 play a crucial role in the interaction. The compounds C2 and C4 exhibit a significant affinity for the active site of EGFR, as indicated by their respective binding energies of -7.7 and -7.6 kcal/mol. The molecule C2 forms hydrogen bonds with Met793 by the interaction of its carbonyl group (C=O) at a distance of 2.05 Å, as depicted in figure 4. In the interaction Met793 donates a hydrogen to the compound C2. Moreover, NO2 group accepts hydrogen donated by Cys797 at a distance of 3.49 Å to form conventional hydrogen bonding. A similar type of interaction was seen in compound C4, where the carbonyl group (C=O) establishes a hydrogen connection with Met793 but at a distance of 1.90 Å (figure 5). Based on the comparable binding affinities and interactions of the potential compounds C2 and C4, the synthesized compounds may effectively suppress EGFR. Compound C5 exhibited a greater binding energy (lower affinity) of -6.6 kcal/mol (figure 6).

 


 

Table 8: Interaction results of docking studies for compounds (C1 – C5) and respective cocrystal.

Interaction

Binding Energy (kcal/mol)

Interacting amino acids

Nature of Bond

Bond length(Å)

C1-7A2A

-6.8

Arg841, Asp800

Hydrogen bond

2.32, 3.11

C2-7A2A

-7.7

Met793, Cys797

Hydrogen bond

2.05, 3.49

C3-7A2A

-6.7

Met793

Hydrogen bond

2.03

C4-7A2A

-7.6

Met793

Hydrogen bond

1.90

C5-7A2A

-6.6

Cys793

Hydrogen bond

2.04

C6-7A2A (Osimertinib)

-6.8

Gly719

Hydrogen bond

2.75

C7-7A2A (Co-crystal ligand, Spebrutinib 7G9)

-7.5

Met793

Hydrogen bond

2.06

 

 


Figure 2: Binding site identification from BIOVIA discovery studio.

 

Figure 3: Co-crystal ligand Spebrutinib(7G9)

 

Figure 4: 2D and 3D molecular interaction of compound C2 with EGFR (PDBID = 7A2A) protein

Figure 5: 2D and 3D molecular interaction of compound C4 with EGFR (PDBID = 7A2A) protein

 

Figure 6: 2D and 3D molecular interaction of C5 with EGFR (PDBID =7A2A) protein

 

3.4 Biological activity prediction:

All five designed compounds screened for anticancer activity. All the compounds (except C4) showed the significant activity against bone cancer. Compounds C3, C4, and C5 showed the activity against pancreatic cancer. Interestingly, compound C4 reports the Pa value of 0,158 against bladder cancer which is greater value than Pi (0,112). Compound C3 is active as epidermal growth factor antagonist having Pa value (0.049) greater than Pi (0.031). Detail activity values are expressed in table 9.

 

Table 9: Predicted biological activity of compounds

Compound code

Anticancer activity

Remarks

Pa

Pi

C1

0,231

0,064

Antineoplastic

(bone cancer)

C2

0,224

 

0,206

0,083

 

0,176

Antineoplastic

(bone cancer)

Antineoplastic

(pancreatic cancer)

C3

0,049

 

0,266

 

0,219

0,031

 

0,018

 

0,152

Epidermal growth factor antagonist

Antineoplastic

(bone cancer)

Antineoplastic

(pancreatic cancer)

C4

0,158

 

0,221

0,112

 

0,148

Antineoplastic

(bladder cancer)

Antineoplastic

(pancreatic cancer)

C5

0,247

0,036

Antineoplastic

(bone cancer)

 

3.5 Toxicity prediction:

The table 10 includes statistics on the toxicity prediction of five N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino) acetamide derivatives (C1, C2, C3, C4, and C5). Each compound is classified as "Non-toxic" for organ toxicity, signifying that, these compounds are not anticipated to induce harm to the specified organ (kidney, liver, and heart) [39-42]. The LD50 values range from 560 mg/kg to 1500 mg/kg, showing different levels of acute toxicity. Regardless of these differences, all substances are categorized as "Class IV" toxicity, indicating a generally low level of acute toxicity.

 

Table 10: Toxicity prediction of synthesized compounds

Compound code

Organ Toxicity

LD50 (mg/kg)

Toxicity class

C1

Non-toxic

1500

Class IV

C2

Non-toxic

560

Class IV

C3

Non-toxic

560

Class IV

C4

Non-toxic

759

Class IV

C5

Non-toxic

759

Class IV

Organ toxicity includes; nephrotoxicity, hepatotoxicity, and cardiotoxicity.

 

4.    CONCLUSION:

Here, we have synthesized five novel N-(5-cyano-6-phenyl-2-thioxo-2,3-dihydropyrimidin-4-yl)-2-(phenylamino)acetamide congeners with anticancer predictions in acceptable chemical yields. According to the in silico investigative study, these synthesized scaffolds displayed an affinity for the anticancer protein EGFR (7A2A). It will lead scientists in the future to perform in vivo cancer studies of these congeners as a potent anticancer moiety. Lipinski's Rule is not broken by any of the compounds, indicating that they could have promising drug-like qualities and be developed further as chemical probes or pharmaceutical agents. According to the estimated toxicity levels, this analysis indicates that these compounds are generally safe and unlikely to be harmful at the recommended dosages.

 

5. AVAILABILITY OF DATA AND MATERIALS:

The data supporting the findings of the article is available within the article.

 

6. REFERENCES:

1.      WHO, “Cancer,” WHO, 2023. https://www.who.int/health-topics/cancer#tab=tab_1 (accessed Apr. 07, 2024).

2.      Liu Q, Yu S, Zhao W, Qin S, Chu Q, Wu K. EGFR-TKIs resistance via EGFR-independent signaling pathways. Molecular Cancer. 2018; 17: 1-9. doi: 10.1186/s12943-018-0793-1.

3.      Bou Antoun N, Chioni AM. Dysregulated signalling pathways driving anticancer drug resistance. International Journal of Molecular Sciences. 2023; 24(15): 12222. doi: 10.3390/ ijms241512222.

4.      Wee P, Wang Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers. 2017; 9(5): 52, doi: 10.3390/cancers9050052.

5.      Zubair T, Bandyopadhyay D. Small molecule EGFR inhibitors as anti-cancer agents: discovery, mechanisms of action, and opportunities. International Journal of Molecular Sciences. 2023 Jan 31; 24(3): 2651, doi: 10.3390/ijms24032651.

6.      Jain MS, Barhate SD, Shimpi RD. Mobocertinib is an Oral kinase inhibitor targeted against EGFR and used in the treatment of Non-small cell Lung cancer: A Review. Asian Journal of Pharmaceutical Research. 2022; 12(2): 179-82.

7.      Dhivya S, Gangopadhyay S, Kumar SP, Sarkar G. Serum creatinine and eGFR are affected in female hypothyroid patients with poor Thyroid control. Asian Journal of Pharmacy and Technology. 2020; 10(4): 241-4.

8.      Cascone T, Morelli MP, Ciardiello F. Small molecule epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in non-small cell lung cancer. Annals of Oncology. 2006; 17: ii46-8, doi: 10.1093/annonc/mdj921.

9.      Naishima NL, Faizan S, Raju RM, Sruthi AS, Veena NG, Sharma GK, Vasanth KS, Shivaraju VK, Ramu R, Kumar BP. Design, synthesis, analysis, evaluation of cytotoxicity against MCF-7 breast cancer cells, 3D QSAR studies and EGFR, HER2 inhibition studies on Novel Biginelli 1, 4-dihydropyrimidines. Journal of Molecular Structure. 2023; 1277: 134848, doi: 10.1016/ j.molstruc.2022.134848.

10.   Elzahabi HS, Nossier ES, Alasfoury RA, El-Manawaty M, Sayed SM, Elkaeed EB, Metwaly AM, Hagras M, Eissa IH. Design, synthesis, and anti-cancer evaluation of new pyrido [2, 3-d] pyrimidin-4 (3H)-one derivatives as potential EGFRWT and EGFRT790M inhibitors and apoptosis inducers. Journal of Enzyme Inhibition and Medicinal Chemistry. 2022; 37(1): 1053-76. doi: 10.1080/14756366.2022.2062752.

11.   Xie Z, Wu K, Wang Y, Pan Y, Chen B, Cheng D, Pan S, Guo T, Du X, Fang L, Wang X. Discovery of 4, 6-pyrimidinediamine derivatives as novel dual EGFR/FGFR inhibitors aimed EGFR/FGFR1-positive NSCLC. European Journal of Medicinal Chemistry. 2020; 187: 111943, doi: 10.1016/ j.ejmech.2019.111943.

12.   Yadav TT, Moin Shaikh G, Kumar MS, Chintamaneni M, Yc M. A Review on Fused Pyrimidine Systems as EGFR Inhibitors and Their Structure–Activity Relationship. Frontiers in Chemistry. 2022; 10: 861288, doi: 10.3389/fchem.2022.861288.

13.   Wang YT, Yang PC, Zhang JY, Sun JF. Synthetic Routes and Clinical Application of Representative Small-Molecule EGFR Inhibitors for Cancer Therapy. Molecules. 2024; 29(7): 1448. doi: 10.3390/molecules29071448.

14.   M. K. Mahapatra MK. Karuppasamy M. Fundamental considerations in drug design, no. January. 2022.

15.   Lin X, Li X, Lin X. A review on applications of computational methods in drug screening and design. Molecules. 2020 Mar 18; 25(6): 1375. doi: 10.3390/molecules25061375.

16.   Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports. 2017 Mar 3; 7(1): 42717, doi: 10.1038/srep42717.

17.   Morak-Młodawska B, Jeleń M, Martula E, Korlacki R. Study of lipophilicity and ADME properties of 1, 9-diazaphenothiazines with anticancer action. International Journal of Molecular Sciences. 2023 Apr 9; 24(8): 6970, doi: 10.3390/ijms24086970.

18.   Niggenaber J, Heyden L, Grabe T, Müller MP, Lategahn J, Rauh D. Complex crystal structures of EGFR with third-generation kinase inhibitors and simultaneously bound allosteric ligands. ACS Medicinal Chemistry Letters. 2020 Nov 5; 11(12): 2484-90, doi: 10.1021/acsmedchemlett.0c00472.

19.   Mendelsohn LD. ChemDraw 8 ultra, windows and macintosh versions. Journal of chemical information and computer sciences. 2004 Nov 22; 44(6): 2225-6, doi: 10.1021/ci040123t.

20.   O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: An open chemical toolbox. Journal of cheminformatics. 2011 Dec; 3: 1-4.

21.   Madhavi Sastry G, Adzhigirey M, Day T, Annabhimoju R, Sherman W. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments. Journal of computer-aided molecular design. 2013 Mar; 27: 221-34, doi: 10.1007/s10822-013-9644-8.

22.   Sriharsha SN, Jainab NH, Biradar MS, Thapa S, Venkatesh ES, Bidye DP, Pujar G, Dixit S. Novel β-L-1, 3-thiazolidine pyrimidine nucleoside analogues: Design, synthesis, molecular docking, and anti-HIV activity. Journal of Molecular Structure. 2023 Dec 15; 1294: 136304. doi: 10.1016/j.molstruc.2023.136304.

23.   Gazgalis D, Zaka M, Abbasi BH, Logothetis DE, Mezei M, Cui M. Protein binding pocket optimization for virtual high-throughput screening (vHTS) drug discovery. ACS omega. 2020 Jun 10; 5(24): 14297-307, doi: 10.1021/acsomega.0c00522.

24.   Gote S, Thapa S, Dubey S, Nargund SL, Biradar MS. Computational investigation of quinazoline derivatives as Keap1 inhibitors for Alzheimer's disease. Informatics in Medicine Unlocked. 2023 Jan 1; 41: 101334, doi: 10.1016/ j.imu.2023.101334.

25.   Otuokere IE, Amaku FJ, Alisa CO. In silico geometry optimization, excited-state properties of (2E)-N-Hydroxy-3-[3-(Phenylsulfamoyl) Phenyl] prop-2-enamide (Belinostat) and its molecular docking studies with Ebola Virus glycoprotein. Asian Journal of Pharmaceutical Research. 2015; 5(3): 131-7.

26.   Patadiya N, Vaghela V. Design, in-silico ADME Study and molecular docking study of novel quinoline-4-on derivatives as Factor Xa Inhibitor as Potential anti-coagulating agents. Asian Journal of Pharmaceutical Research. 2022; 12(3): 207-11.

27.   Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of computational chemistry. 2010 Jan 30; 31(2): 455-61, doi: 10.1002/jcc.21334.

28.   Pawar S, Kulkarni C, Gadade P, Pujari S, Kakade S, Rohane SH, Redasani VK. Molecular Docking using different Tools. Asian Journal of Pharmaceutical Research. 2023 Dec 1; 13(4).

29.   Shinde SA, Mhetar MJ, Parit AG, Thombre AR. In-silico investigation and ADMET prediction of potential antifungal phytochemicals against lanosterol 14-alpha demethylase inhibitors. Asian Journal of Pharmaceutical Research. 2024; 14(1): 33-8.

30.   Thapa S, Nargund SL, Biradar MS. Molecular design and in-silico analysis of trisubstituted benzimidazole derivatives as ftsz inhibitor. Journal of Chemistry. 2023 Mar 1; 2023: 1-9, doi: 10.1155/2023/9307613.

31.   Seeliger D, de Groot BL. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. Journal of computer-aided molecular design. 2010 May; 24(5): 417-22 doi: 10.1007/s10822-010-9352-6.

32.   Pant L, Thapa S, Dahal B, Khadka R, Biradar MS. In Silico and In Vitro Studies of Antibacterial Activity of Cow Urine Distillate (CUD). Evidence-Based Complementary and Alternative Medicine. 2024 Jan 8;2024, doi: https://doi.org/10.1155/2024/ 1904763.

32.

33.   Lagunin A, Stepanchikova A, Filimonov D, Poroikov V. PASS: prediction of activity spectra for biologically active substances. Bioinformatics. 2000 Aug; 16(8): 747-8, doi: 10.1093/ bioinformatics/16.8.747.

34.   Filimonov DA, Lagunin AA, Gloriozova TA, Rudik AV, Druzhilovskii DS, Pogodin PV, Poroikov VV. Prediction of the biological activity spectra of organic compounds using the PASS online web resource. Chemistry of Heterocyclic Compounds. 2014 Jun; 50: 444-57, doi: 10.1007/s10593-014-1496-1.

35.   Drwal MN, Banerjee P, Dunkel M, Wettig MR, Preissner R. ProTox: a web server for the in silico prediction of rodent oral toxicity. Nucleic acids research. 2014 Jul 1; 42(W1): W53-8, doi: 10.1093/nar/gku401.

36.   Thapa S, Biradar MS, Nargund SL, Ahmad I, Patel H, Lamsal A. Synthesis, molecular docking, molecular dynamic simulation studies, and anti-tubercular activity evaluation of substituted benzimidazole derivatives, doi: 10.1155/2024/9986613.

37.   Waters LJ, Quah XL. Predicting skin permeability using HuskinDB. Scientific data. 2022 Sep 23; 9(1): 584, doi: 10.1038/s41597-022-01698-4.

38.   Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced drug delivery reviews. 1997 Jan 15;23(1-3):3-25, doi: 10.1016/j.addr.2012.09.019.

39.   Morris-Schaffer K, McCoy MJ. A review of the LD50 and its current role in hazard communication. ACS Chemical Health & Safety. 2020 Dec 21; 28(1): 25-33, doi: 10.1021/acs.chas.0c00096.

40.   Tripathi J, Mahto MK, Bhaskar M, Shahbazi S. Molecular docking and toxicity analysis of novel atorvastatin structural analogues with HMG-CoA reductase. Asian Journal of Research In Chemistry. 2012; 5(3): 386-9.

41.   Sravani M, Duganath N, Gade DR, Reddy CH. Insilico analysis and docking of imatinib derivatives targeting BCR-ABL oncoprotein for chronic myeloid leukemia. Asian Journal of Research in Chemistry. 2012; 5(1): 153-8.

42.   Govindammal M, Prasath M, Sathya B, Selvapandiyan M. Investigation on the Binding Interaction Between Clomipramine and Doxepin with LeuT by Molecular Docking Analysis. Asian Journal of Research in Chemistry. 2017; 10(4): 486-90.

43.   Chauhan R, D'Souza HL, Shabnam RS, Abraham J. Phytochemical and cytotoxicity analysis of seeds and leaves of Adenanthera pavonina. Research Journal of Pharmacy and Technology. 2015; 8(2): 198-203.

 

 

 

 

Received on 15.05.2024      Revised on 19.08.2024

Accepted on 12.09.2024      Published on 22.10.2024

Available online from October 31, 2024

Asian J. Research Chem.2024; 17(5):278-284.

DOI: 10.52711/0974-4150.2024.00048

©AandV Publications All Right Reserved

 

This work is licensed under a Creative Commons Attribution-Non Commercial-Share Alike 4.0 International License. Creative Commons License.

Description: Description: Creative Commons License