Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Investigating chronic myeloid leukemia therapeutics: AI-optimized design and synthesis of 4-methylthiazole-2-amine derivatives with translocation control activity

Zaheer Ahmada, Labaina Shakoora, Sajid Mahmoodb, Aneeqa Batoolc, Nusrat Shafiq*c, Aniqa Moveedc, Syeda Aaliya Shehzadid, Mohamed Mohanye, Lei Zhangf, Salim S. Al-Rejaieg and Humaira Razzaqa
aDepartment of Chemistry, University of Wah, The Quaid Avenue, Wah Cantt 47040, Pakistan. E-mail: dr.zaheer.ahmad@uow.edu.pk
bDepartment of Chemistry, University of Education Lahore, Vehari Campus, Vehari, Pakistan
cSynthetic and Natural Product Drug Discovery Lab., Department of Chemistry, Government College Women University, Faisalabad-3800, Pakistan. E-mail: dr.nusratshafiq@gcwuf.edu.pk; gqumarin@gmail.com
dSulaiman Bin Abdullah Aba Al-Khail-Centre for Interdisciplinary Research in Basic Sciences (SA-CIRBS), International Islamic University, Islamabad-44000, Pakistan
eDepartment of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
fMOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, P. R. China
gDepartment of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia

Received 23rd September 2025 , Accepted 20th November 2025

First published on 5th December 2025


Abstract

A series of 4-methylthiazole-2-amine derivatives (3a–3f, 6g–j and 7) were prepared via a Hantzsch-type multicomponent reaction and characterized by FT-IR, 1H-NMR and 13C-NMR spectroscopy techniques. The cytotoxic effects of the synthesized products on the chronic myeloid leukemia K562 and U937 cell lines were tested using the MTT assay. Compounds 3a, 3b, 3c, 6g, 6h and 6i exhibited high cytotoxic potential with IC50 values ranging from 1.5 to 5 0 µM, which showed a dose-dependent inhibition. Stable ligand–receptor interaction was observed by molecular docking studies against the chosen CML-associated proteins (2GQG, 5MO4, 2AZ5 and 5MAR) with the highest docking scores obtained for 6h and 6i (−8.37 and −8.97 kcal mol−1, respectively). Strong binding affinities (ΔG = −53.36 kcal mol−1) were confirmed by MM-GBSA calculations. Further, density functional theory (by using B3LYP/6-311G basic function set) was used to gain information on the electronic configurations, HOMO–LUMO gaps and charge distribution, which favored stability of the molecules and reactivity. The combined experimental and computational findings indicated that compounds 6h and 6i are potential scaffolds that can be used to develop new thiazole-based anticancer agents against chronic myeloid leukemia.


1 Introduction

Many natural products contain five-membered heterocycles having nitrogen and sulphur heteroatoms at positions 1 and 3. For example, in vitamin B1 (1), the thiazolium ring acts as an electron sink, and its coenzyme structure is essential for the decarboxylation of α-keto acids (2). It is used as an intermediate in the preparation of drugs and dyes. The aromatic nature of the thiazole is due to the delocalization of the lone pair of electrons on the sulfur atom.1 The most demanding and investigated class of aromatic five-membered heterocycles is thiazoles. Hantzsch and Weber were the first scientists who illustrated the structure of thiazole in 1887 (1) (Fig. 1).
image file: d5ra07232h-f1.tif
Fig. 1 Basic core structure of thiazole.

Thiazoles are essential due to their countless biological and pharmaceutical properties.2 Many thiazole drugs are antioxidant, anti-inflammatory, anti-Alzheimer, antidiabetic, antimicrobial, anti-HIV, anticancer, antihypertensive and anticonvulsant.3 Thiazoles are also present in many clinically utilized drugs, such as abafungin (antifungal drug), ritonavir (antiretroviral drug) and sulfathiazole (antimicrobial drug).4 Nizatidine is a drug used as antagonist and drug of neonicotinoids class act as (insects) behavior product.5 Nitazoxanide is a broad-spectrum medication play a role in traeting the diarrhea (as antiparasitic agent)6 and in many other work such as anti-inflammatory agents. In the development of novel therapeutic agents and drug, design the most recent rumor of thiazole core structure have been put forwarded many applications (Fig. 2).


image file: d5ra07232h-f2.tif
Fig. 2 Examples of thiazole-bearing drugs.

The properties of thiazoles are comparable to those of oxazoles, and the nitrogen atom with an unshared pair of electrons is vital in nature. Thiazoles holds a noticeable position within the drug innovation process among the distinctive aromatic heterocycles.7 This ring structure is found in a few marketed drugs. Therefore, thiazoles are often included in the core structure for the amalgamation of chemical libraries.8 Consequently, the thiazole core has been much considered within the field of medicinal and organic chemistry.

Since decades, huge populations are affected by cancer. Next to coronary disease, cancer is the second most dangerous disease. In accordance with the world health organization (WHO) report, the number of latest cases expected to increase by 70% more than two decades. Central and South America, Africa and Asia account for 60% of the world's total latest yearly cancer cases and 70% of the world's cancer deaths. By adoptation of suitable treatment for cancer control, struggle against cancer is taken as worldwide confront in this distressing condition. According to the stages and types of cancers, various treatment options such as surgery, chemotherapy, radiation therapy, targeted therapy and precision medicine, hormone therapy, immunotherapy, and stem cell transplant are available. Among all these treatment options, chemotherapy is broadly acknowledged and chosen due to its different administration routes such as oral, intraperitoneal, intravenous, intra-arterial, intrathecal and topical modes.9

Now-a-days, most of the studies are focused on discovering the accurate target and developing potential chemotherapeutic agents for the control, treatment and eradication of this lethal disease. The medicines currently available on the market generally have nitrogen-containing heterocyclic compounds.10 Furthermore, it is confirmed that the flexible thiazole core has significant anti-cancer effects, and is found in numerous novel chemotherapeutic agents.11 There are numerous examples of medicines that contain the thiazole core as a fundamental structure. These drugs have revealed significant anti-cancer activities (Fig. 3).


image file: d5ra07232h-f3.tif
Fig. 3 Antitumor drugs containing a thiazole ring.

2 Materials and methods

2.1. General procedure for the synthesis of thiazole derivatives

Different ketones having alpha hydrogen were used for the preparation of 2,4-disubtituted thiazole derivatives. The Hantzsch mechanism was followed for the preparation of thiazole derivatives. Three- and four-component single-step reactions occurred (Scheme 1).
image file: d5ra07232h-s1.tif
Scheme 1 General reaction mechanism for the synthesis of thiazole derivatives followed by the Hantzch synthesis.
2.1.1. General procedure for the synthesis of thiazole derivatives (3a–f). For the preparation of thiazole derivatives, a mixture of substituted ketones (0.2 mole), amides (0.2 mole), thiourea (0.3 mole) and bromine (0.2 mole) was added to a round-bottom flask and refluxed for 2 h at 110 °C. The reaction mixture was cooled to room temperature (25 °C) and then 100 mL water was added. Then, the solution was filtered off and washed with ethyl acetate to remove unreacted parts especially bromine or iodine. Excess of ethyl acetate was removed and the filtrate was poured into boiling water. Filter off the hot solution immediately. Then, it was set aside and left for half an hour. The pH of the solution was maintained at 8–9 by making it alkaline with a ammonium hydroxide solution. The products obtained were filtered. The solid product was washed sequentially with water (2 × 150 mL). Aqueous ethanol (1[thin space (1/6-em)]:[thin space (1/6-em)]1) was used to crystallize the product (Scheme 2).
image file: d5ra07232h-s2.tif
Scheme 2 General reaction for the preparation of thiazole derivatives (3a–f).
2.1.2. General procedure for the synthesis of compounds 6g–j. A mixture of amino thiazole derivatives (0.02 mole) and chloroacetyl chloride (0.5 g) in 20 mL DCM was taken in a 200 mL round-bottom flask. Sodium bicarbonate (NaHCO3) was added to the mixture and refluxed for several hours with constant stirring. After completion, the reaction mixture was poured into ice-cold water (200 mL). Then the precipitate was filtered-off and crystalized with ethanol (Scheme 3).
image file: d5ra07232h-s3.tif
Scheme 3 General reaction of compounds 7d–e.
2.1.3. General procedure for the synthesis of azothiazole (7). For the synthesis of azothiazole (7) (Scheme 4), an aqueous solution of NaNO2 and acidic solution of aniline were prepared. Then, 0.01 mole of NaNO2 in 12.5 mL water and 0.01 mole of C6H5NH2 in 7.5 mL of hydrochloric acid were prepared. Then, a NaNO2 solution was added to an aniline solution drop-wise maintaining at 5 °C, and a diazonium salt was formed. A diazonium salt was added to cool the mixture of 2-amino-4-phenyl thiazole (0.01 mole) and 7.5 g of CH3COONa in ethanol. The reaction mixture was continuously stirred at a temperature of 5 °C for 2 h. Water was added to the mixture, and the solution was filtered. A solid mass was washed with water and recrystallized with aqueous ethanol (9[thin space (1/6-em)]:[thin space (1/6-em)]1).
image file: d5ra07232h-s4.tif
Scheme 4 Reaction for the preparation of azothiazole (7).

2.2. Characterization of the targeted synthesized compounds (3a–f, 6g–j and 7)

2.2.1. 2-Amino-4-methylthiazole (3a). Physical appearance: white crystals; yield: 74.5%; RF value: 0.75; FTIR (cm−1): 3370.24 (–NHstretch), 1590.53 (C[double bond, length as m-dash]N), 1083.20 (C–Nstretch), 2962.08 (CHstretch), 1406.71 (C[double bond, length as m-dash]Cring), 730.04 (thiazole nucleus); 1HNMR (DMSO, 300 MHz): δ 7.06 (s, 2H, 2-NH2), 6.48 (s, 1H, H-5), 2.28 (s, 3H, 4-CH3); 13CNMR (DMSO, 125 MHz): δ 169.1 (C-2), 147.8 (C-4), 101.3 (C-5), 16.8 (4-CH3); molecular formula: C4H6N2S; molecular weight: 114.17 g mol−1.
2.2.2. 2-Amino-4-phenyl thiazole (3b). Physical appearance: yellow crystals; yield: 80%; RF value: 0.69; FT-IR (cm−1): 3249.51 (NHstretch); 1596.71 (C[double bond, length as m-dash]N); 1021.65 (C–Nstretch); 1530.66 (C[double bond, length as m-dash]Caromatic); 1440.74 (C[double bond, length as m-dash]Cring); 710.95 (thiazole nucleus); 1HNMR (DMSO, 300 MHz) δ 6.998 (s, –NH2), 7.78 (dd, J = 2.00 Hz, 2H, H-7, 11), 7.38 (d, 2H, J = 2.04 Hz, H-8, 10), 7.27 (dd, 1H, J = 1.02 Hz, H-9), 7.074 (s, 1H, H-5); 13CNMR (DMSO, 125 MHz): δ 168.6 (C-2), 150.29 (C-4), 125.98 (C-9), 127.63 (C-7, 11), 128.92 (C-8, 10), 135.38 (C-6), 101.94 (C-5); molecular formula: C9H8N2S; molecular weight: 176.24 g mol−1.
2.2.3. Bis-(2-aminothiazole-4-yl)-methanone (3c). Physical appearance: brown crystals; yield: 83%; RF value: 0.89; FT-IR value: 3318.45 (NHstretch), 1655.26 (C[double bond, length as m-dash]N), 1129.41 (C–Nstretch), 1706.78 (C[double bond, length as m-dash]Ostretch), St of thiazole nucleus, 720.08; C[double bond, length as m-dash]C (ring), 1430.75; 1HNMR (DMSO, 300 MHz): δ 6.99 (s, 4H, 2,2′-NH2), 8.38 (s, 2H, H-5,5′); 13CNMR (DMSO, 125 MHz): δ 194.9 (C-6), 168.9 (C-2,2′), 144.9 (C-4,4′), 113.5 (C-5,5′); molecular formula: C7H6N4S2O; molecular weight: 226.00 g mol−1.
2.2.4. N4-Isopropyl-5-methylene-4,5-dihydrothiazole-2-4-diamine (3d). Physical appearance: green crystals; yield: 76.5%; RF value: 0.67; FTIR (cm−1): 3277.59 (NHstretch), 1620.42 (C[double bond, length as m-dash]N), 1153.88 (C–Nstretch), 1253.89 (CHaromatic), 1650.82 (C[double bond, length as m-dash]Cring), 1549.06_(NHbend) (CHstretch), 2876.22 (CH3), 2932.69 (CHstretch) 727.51 (thiazole nucleus); 1HNMR (DMSO, 300 MHz): δ 7.06 (s, 2H, –NH2), 6.48 (s, 1H, H-5), 2.28 (s, 3H, 4-CH3); 13CNMR (DMSO, 125 MHz): δ 169.1 (C-2), 147.8 (C-4), 101.3 (C-5), 16.8 (4-CH3); molecular formula: C7H13N3S; molecular weight: 171.26 g mol−1.
2.2.5. 5,6-Dihydro-4H-cyclopenta[d]thiazole-2-amine (3e). Physical Appearance: white powder; yield: 79%; RF value: 0.61; FTIR (cm−1): 3376.24 (NHstretch), 1608.08 (C[double bond, length as m-dash]N), 1081.78 (C–Nstretch), 3147.59 (CHaromatic), 1441.36 (C[double bond, length as m-dash]Cring), 727.51 (thiazole nucleus); 1HNMR (DMSO, 300 MHz): δ 7.06 (s, 2H, –NH2), 6.48 (s, 1H, H-5), 2.28 (s, 3H, 4-CH3); 13CNMR (DMSO, 125 MHz): δ 169.1 (C-2), 147.8 (C-4), 101.3 (C-5), 16.8 (4-CH3); molecular formula: C6H8N2S; molecular weight: 140.21 g mol−1.
2.2.6. 5-Methylene-4-5-dihydrothiazole-2-4-diamine (3f). Physical Appearance: white crystals; yield: 83.6%; RF value: 0.61; FTIR (cm−1): 3346.20 (NHstretch), 1602.63 (C[double bond, length as m-dash]N), 1081.30 (C–Nstretch), 3118.98 (CHaromatic), 1665.89 (C[double bond, length as m-dash]Cring), 727.51 (thiazole nucleus); 1HNMR (DMSO, 300 MHz): δ 7.06 (s, 2H, –NH2), 6.48 (s, 1H, H-5), 2.28 (s, 3H, 4-CH3); 13CNMR (DMSO, 125 MHz): δ 169.1 (C-2), 147.8 (C-4), 101.3 (C-5), 16.8 (4-CH3); molecular formula: C4H7N3S; molecular weight: 129.18 g mol−1.
2.2.7. 2-Chloro-N-(4-phenylthiazole-2-yl)acetamide (6g). Physical appearance: brownish crystals; yield: 80%; RF value: 0.70; FT-IR value: 3249.36 (N–Hstretch), 1566.86 (C[double bond, length as m-dash]N), 1137.87 (C–N), 1263.03 (C–Naromatic), 1654.79 (C[double bond, length as m-dash]Ostretch), 1566.86 (CHaromatic), 669.81 (Clstretch), 718.66 (thiazolo nucleus); 1HNMR (DMSO, 300 MHz): δ 12.68 (s, 2H, NH2-2), 7.92 (dd, 2H J = 1 Hz, H-7,11), 7.45 (dd, 2H, J = 0.94 Hz, H-8,10), 7.34 (dd, 1H, J = 0.58 Hz, H-9), 7.67 (s, 1H, H-5), 4.43 (s, 2H, CH2-12); 13CNMR (DMSO, 125 MHz): δ 165.6 (C[double bond, length as m-dash]O), 157.92 (C-2), 149.53 (C-4), 134.58 (C-6), 129.22 (C-8,10), 128.35 (C-7,11), 126.15 (C-9), 109.03 (C-5), 42.79 (–CH2); molecular formula: C11H9ClN2OS; molecular weight: 252.72 g mol−1.
2.2.8. 2-Chloro-N-(5,6-dihydro-4H cyclopenta[d]thiazol-2-yl)acetamide (6h). Physical appearance: yellow crystals; yield: 78%; RF value: 0.78; FT-IR (cm−1): 3319.15 (NHstretch), 1584.04 (NHbending), 1640.60 (C[double bond, length as m-dash]Nstretch), 1228.02 (C–Naromatic); 1117.57 (CNstretch), 1667.22 (C[double bond, length as m-dash]Ostretch), 2979.28 (CHaromatic), 672.37 (Clstretch); 1HNMR (DMSO, 300 MHz): δ 12.5 (2H, s, 2-NH2), 2.87 (2H, t, 6-CH2), 2.54 (2H, m, 7-CH2), 2.52 (2H, m, 8-CH2); 13CNMR (DMSO, 125 MHz): δ 164.2 (C[double bond, length as m-dash]O), 148.2 (C-4), 118.9 (C-5), 28.2 (C-6), 26.0 (C-7), 30.9 (C-8); molecular formula: C8H9ClN2OS; molecular weight: 216.69 g mol−1.
2.2.9. N,N′-(4,4′-Carbonylbis(thiazole-4,2-diyl))bis(2-chloroacetamide) (6i). Physical appearance: grey powder; yield: 73%; RF value: 0.57; FTIR (cm−1): 3383.72 (NHstretch), 1574.54 (NHbending), 1630.14 (C[double bond, length as m-dash]Nstretch), 1224.23 (C–Naromatic), 1118.70 (C–Nstretch), 1654.82 (C[double bond, length as m-dash]Ostretch), 670.82 (Clstretch); 1HNMR (DMSO, 300 MHz): δ 12.4 (2H, s, 2,2′-NH), 4.87 (4H, t, 8,8′-CH2), 8.98 (2H, s, 5,5′-CH2); 13CNMR (DMSO, 125 MHz): δ 192.9 (C-9), 164.5 (C-7,7′), 144.2 (C-4,4′), 113.9 (C-5,5′), 42.7 (C-8,8′), 26.0 (C-7), 30.9 (C-8); molecular formula: C11H10ClN4O3S2; molecular weight: 379.23 g mol−1.
2.2.10. 2-Chloro-N-(4-methylthiazole-2-yl)acetamide (6j). Physical appearance: white powder; yield: 82%; RF value: 0.59; FTIR (cm−1): 3264.01 (N–Hstretch), 1582.5 (C[double bond, length as m-dash]N), 1072.49 (C–Nstretch), 1287.11 (C–Naromatic), 1683.96 (C[double bond, length as m-dash]Ostretch), 2919.77 (CHstretch), 670.34 (Clstretch), 722.57 (thiazole nucleus); 1HNMR (DMSO, 300 MHz): δ 12.68 (s, 1H, 6-NH-), 7.67 (s, 1H, H-5), 4.43 (s, 2H, CH2-8); 13CNMR (DMSO, 125 MHz): δ 165.6 (C[double bond, length as m-dash]O), 162.92 (C-2), 144.7 (C-4), 104.3 (C-5), 42.79 (8-CH2) molecular formula: C6H7ClN2OS; molecular weight: 190.65 g mol−1.
2.2.11. 4-Phenyl-5-(phenyldiazenyl)thiazole-2-amine (7). Physical appearance: grey powder; yield: 69%; RF value: 0.66 FT-IR value: 3246.76 (NHstretch), 1597.30 (C[double bond, length as m-dash]N), 1038 (C–Nstretch), 1282.04 (C–Naromatic), 1517.64 (N[double bond, length as m-dash]N), 1483.26 (CHaromatic), 1441.32 (C[double bond, length as m-dash]Caromatic), 772.17 (thiazolo nucleus); 1HNMR (DMSO, 300 MHz): δ 8.61 (2H, s, 2-NH2), 8.20 (2H, dd, J = 8, 1.0 Hz, H-7,11), 7.38 (2H, dd, J = 1.0, 2.5 Hz, H-8,10), 7.45 (1H, ddd, J = 1.5 Hz, 2.5 Hz, H-9), 7.26 (2H, dd, J = 8.5, 1.0 Hz, H-7′, 11′), 7.34 (2H, dd, J = 8.0 Hz, H-8′,10′), 7.10 (1H, dd, J = 8.0 Hz, H-9′); 13CNMR (DMSO, 125 MHz): δ 1770.2 (C-2), 154.4 (C-4), 138.0 (C-5), 133.1 C-6), 126.1 (C-7,11), 128.9 (C-8,10), 128.6 (C-9), 128.6 (C-6′), 127.9 (C-7′,11′), 128.6 (C-8′, 10′), 126.2 (C-9′) molecular formula: C15H12N4S; molecular weight: 280.35 g mol−1.

2.3. Anti-tumor activity of the synthesized compounds

Different thiazole derivatives were prepared and screened by the MTT assay.12 For this purpose, two chronic myeloid leukemia model cell lines, namely, K562 and U937, were selected as translocation controls. The targeted cell lines were purchased from the German Collection of Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany) through the local vendor (Musaji Adam & Sons, Lahore). RPMI1640 media supplemented with 10% fetal bovine serum (FBS; Gibco/Invitrogen, Karlsruhe, Germany), 1% penicillin/streptomycin (P/S) and 1% L-glutamine were used to culture the cell lines. This assay was performed using a previously reported method.12 For this purpose, the compounds at concentrations of 0.4 µM, 0.6 µM, 0.8 µM, and 1.0 µM were used. The cell viability was calculated, IC50 was determined using GraphPad Prism and further statistical analyses of results were done by applying two-way ANOVA for the proper presentation of results.

2.4. Molecular docking study

Molecular docking of the synthesized compounds from thiazole and azothiazol was performed using Maestro Schrodinger 13.4. The protein structures are generally refined for their topologies, incomplete and terminal amide groups, bond ordering, formal charges and missing hydrogen atoms.13 Water molecules larger than 5 Å were eliminated. The most stable ionization states that were produced in the protein structure were selected. The orientations of the remaining water molecules were adjusted.14 Lastly, the protein structure was minimized by reorienting side-chain hydroxyl groups and steric conflicts using the OPLS2005 force field.15
2.4.1. Ligand preparation. A dataset of thiazole derivatives were prepared by the synthesis of thiazole and azothiazol. Thiazole derivative structures were identified by the characterization of these compounds. The structures were drawn by using ChemDraw (ChemDraw 19.1), then names, and smiles of these thiazole derivatives were generated (as shown in Table S1 (SI)). For molecular docking, thiazole derivatives are saved in the SDF format along with the reference drug. The Maestro Schrodinger suite 2022 software was used for ligand preparation through the LigPrep interface.16 By follwoing the Schrodinger software's specifications, all ligands were selected from the workspace, eliminate the tautomer, neutralize and create the ligand.17
2.4.2. Receptor grid generation. For receptor grid generation, the ligands remained in the crystal structure of the prepared protein. For checking the accuracy of docking studies, the lowest energy pose of the co-crystallized ligands was determined by the scoring function, which closely reflects the experimental binding mode determined by X-ray crystallography.17 The co-crystallized ligand was separated from the protein's binding site using the Glide docking approach. The receptor grid and the ligands were used to perform the glide docking of the synthesized compounds.18

We used the Maestro Schrodinger version 13.5 software, which use the step wise preparation by following the sequence of protein preparation, receptor grid generation, ligand preparation and docking. Protein amino acid interaction with receptor (ligand) was enclosed by active site grid.19 Through molecular docking study, these ligands were converted into 3D structures and the best docking score was recorded.

2.5. MM-GBSA calculation

Ligand–protein complexes were subjected to MM-GBSA built in Maestro Schrodinger 13.5 version to calculate the binding energy. Using this software, proteins were prepared by the removal of water and minimization of H-bonds, and ligands were also prepared by the LigPrep tool using the OPLS2005 force field.20 Ligand–protein complexes were optimized using Prime with the OPLS 2005 force field. Different factors of energies were calculated for the MM-GBSA study such as binding free energy, coulomb binding free energy, covalent binding free energy, hydrogen bond binding free energy and lipophilic binding free energy.21

2.6. Optimization of geometry parameters and spectral data by DFT studies

Density functional theory (DFT) calculations were performed to check the optimization geometry of the compounds using the Gaussian 09W and Gauss View 6.0.16 software. The B3LYP 6-311G basis set was used to calculate the energy gap between the LUMO and HOMO orbitals and the stability and reactivity of the compounds.22
2.6.1. Computational details of the protocol. Theoretical analyses of compounds 6h (2-chloro-N-(5,6-dihydro-4H cyclopenta[d]thiazol-2-yl)acetamide) and 6i (N,N′-(4,4′-carbonylbis(thiazole-4,2-diyl))bis(2-chloroacetamide)) were performed using the Gaussian and Gauss View program.23 Geometry optimization and frequency calculation were also conducted by the density functional theory (DFT) calculation of quantum mechanics with the B3LYP parameter and the 6-311G basis set as correlation functions.24 The B3LYP model gives the stable IR and NMR DFT calculations with good experimental calculation of IR and NMR according to studies. Transition energies of the lowest unoccupied molecular orbital (LUMO) and the highest occupied molecular orbital (HOMO), energy gap, FMO, MEP, dipole moments and other properties were calculated using this model.25 The vibrational data of IR obtained from the Gauss View was computed to Origin program to generate the IR spectra. Proton and carbon NMR spectra were generated using the GIAO (gauge independent atomic orbital) approach and compared with the experimental calculations. Using tetramethylsilane (TMS HF/6-31G(d)) GIAO as the internal reference, the optimized parameters obtained from the B3LYP/6 311G method were used to determine the proton and carbon isotropic shielding.26 The natural bond orbital (NBO) was obtained using the second order of perturbation theory from the B3LYP 6-311G basis set. Further studies of natural population analysis (NPA) and Mulliken's population analysis (MPA) were calculated from the second order of perturbation theory NBOs.27
2.6.2. Hirshfeld population analysis. To check the charge distribution at the atomic level and understand the non-covalent nature of compounds 6h and 6i, the Hirshfeld surface population analysis and CM5 atomic charge calculation were performed on the basis of the DFT results. All the optimization of geometry for targeted compounds 6h and 6i was done at the B3LYP 6-311G level using the Gaussian 09W software. The components of dipole moment (Dx, Dy, and Dz), Hirshfeld atomic charges (Q-H) and CM5 charges (Q-CM5) were determined from the converged wave-function through the population analysis tools, and further process proceeded with Multiwfn.28 This study gave an idea about partial atomic charges and charge separation directions in the molecule that are fundamental to explaining the electrostatic interactions, intra-molecular polarization and ability to bind with biological targets.29 Red color area indicates the atoms with positive dipole contributions and blue to violet color area indicates the negative dipole contributions, consistent with the electron-withdrawing or electron-donating characteristics.30 The gradient in the plot, observed as red (e-poor) to blue (e-rich) direction, indicates significant polarization in the molecular structure, which could have a significant impact on the orientation and strength of the non-covalent interactions in the biological context.
2.6.3. Non-covalent bond interactions (NCI) study. Non-covalent interactions (NCI) for targeted compounds 6h and 6i were analyzed by the reduced density gradient (RDG) analysis due to the density of electrons, which was calculated by the density functional theory (DFT) calculation. The optimized geometry of compounds 6h and 6i was determined using the Gaussian software at the B3LYP 6-311G level. The respective wave function (wfn) file was created and used for RDG analysis. The mathematical operations were performed by applying the Multiwfn program that calculates the value of RDG and also determines the area of weak interactions within the molecular system.31 The visualization of the RDG isosurfaces was performed using the Visual Molecular Dynamics (VMD) software. This tool set can offer this kind of in-depth analysis of several non-covalent forces, such as van der Waals forces, hydrogen bonding and steric effects, by studying the distribution of electron density and gradient.32
2.6.4. Electron localization function (ELF) and localized orbital locator (LOL). The electronic structure of the 6h and 6i compounds was determined in the framework of density functional theory (DFT), providing insights into the character of electron localization and bonding interaction of these compounds as well as their electron localization function (ELF) and localized orbital locator (LOL). These gauges allow precious information about covalent bonding, lone pairs and delocalized electrons and ELF gives an indication of the likelihood of occurrence of electron pairs and the LOL looks at the distribution of kinetic energy density.33 A set of ELF and LOL calculations were performed using the Multiwfn program, allowing a quantitative and visual analysis of the features of the localization of the wavefunctions and their convergence with the DFT results.34 The resulting 3D isosurfaces and contour plots were visualized in VMD (Visual Molecular Dynamics), which made the bonding environments of the 6h and 6i structures clear to interpret.

3 Results and discussion

3.1. Chemistry

The synthesized compounds were characterized for their structure by spectroscopic analysis. The FT-IR spectral data of all compounds displayed characteristic peaks of the thiazole nucleus at 772.17 cm−1 to 672.37 cm−1 with other functional group peaks like –NH and –C[double bond, length as m-dash]C– at their specific values compared to the literature. The 1H-NMR spectral data of compound 3a displayed a singlet at δ 7.06 ppm for the amino group with two more singlets at 6.48 ppm and 2.28 ppm for the ring proton and methyl moiety present at the 5-position of the ring, respectively. The 13C-NMR spectra were also found in accordance with the 1H-NMR data displaying a specific carbon signal at δ 169.1 ppm for quarterly carbon attached to an amino group shifting downfield. A signal was observed at δ 147.8 ppm corresponding to the carbon attached to a methyl group whose signal at δ 16.8 ppm and a signal at δ 101.3 ppm correspond to the methane proton.35 The spectral data of 3b and 3c were also found to be similar to 3a except the presence of a phenyl ring in place of methyl in compound 3b and the presence of a carbonyl group with the double signals of the thaizole skeleton, confirming the dimer of compound 3c. Basic spectral signals of compounds 3d, 3e and 3f were found to be similar to 3a except the additional signals of substituents found in them. Similarly, the 1H-NMR spectra of compound 7 displayed the additional signals of a biphenyl skeleton in addition to the thaizole signals as in the case of 3b. The 13C-NMR spectral data of compound 7 also support the 1H-NMR data, confirming the structure of 7. All spectral analyses of 6g were found to be the same as that for compound 3b with additional signals of a –OCH2-moiety both in proton and in carbon NMR data. Similarly, the spectral data of 6h were observed to be similar to 6g in the presence of the propene ring and the absence of signals for the phenyl group. Spectral data (1H- and 13C-NMR) of 6i were found to be in accordance with those of 3c with additional signals for the –COCH2 moiety. Similarly, spectral analysis of 6j displayed the signals similar to that of compound 3a with additional signals for the –COCH2 moiety. All the spectral analysis of targeted compounds elucidated their structures.

3.2. Anti-tumor activity of the synthesized compounds

Targeted compounds were screened for their anti-tumor potential against chronic myeloid leukemia models K562 and U937, pro-monocytic model cell lines, by the MTT assay. The compounds 3a, 3b, 3c and 6g showed excellent inhibition against the mentioned cancer cell lines. The results are shown in Fig. 4–8 as graphical representation for both cell lines. The absorbance values obtained by the MTT assay were used to measure the cell toxicity and cell viability by measuring the activity of cellular enzymes. The cells were incubated with the MTT reagent, which was converted by cell's mitochondrial enzymes into a colored formazan product. The amount of formazan product was proportional to the number of viable cells present. To calculate the cell viability, the absorbance of the treated cultures was compared with the absorbance of untreated cultures. The percentage cytotoxicity was calculated using the following equation:
% Cytotoxicity = 100% − % cell viability

Overall, the compounds showed a dose–response manner of activity; however, the compounds 3d, 3e, 3f, 6j and 7 did not show any significant toxicity against both cell lines at all concentrations, while the compound 6h exhibited significant toxicity against both cell lines, with 82% toxicity against U937 and 84% against K562 at 10 µM concentration. Among the compounds 3b and 3c, the compound 3c showed outstanding activity against U937 with 57% toxicity at 10 µM concentration. In summary, compound 6g, 6i, 6h, 3b and 3c have potential to be promising anti-tumor compounds; however, the in vivo study could support the findings of the in vitro study.

3.2.1. Antitumor activity of targeted compounds against the cell line K562. The graph (Fig. 4) showed the dose–response curves of a number of compounds tested on the K562 cancer cell line with molar absorbance (%) versus the levels of the compound (µM). The concentration in which 50 percent inhibition is observed, the IC50 value, is labeled next to each compound. The highest antitumor activity was observed to be exhibited by compound 3a with the lowest value of IC50 of 3.523, followed by the next powerful compound 3c (4.664), 6g (4.938), 6h (5.004) and 6i (5.083) (Fig. 4). These compounds (3c, 6g, 6h and 6i) show the good inhibitory concentration (IC50) as all the compounds are under 50 value against K562 cell line. The compounds were evaluated at multiple concentrations: 0.4 µM, 0.6 µM, 0.8 µM, and 1.0 µM, as shown in Fig. 4 and 5 supported by the descriptive data reported. These data indicate that 3a has a great potential against the K562 cell line, and it is the most effective analog compared with the others.36 The well-defined sigmoidal curves support the standard dose–response effect, which enhances the validity of IC50 measurements of this screening test.
image file: d5ra07232h-f4.tif
Fig. 4 Graphical representation of the K562 cell line inhibitory concentration of the synthesized drugs.

image file: d5ra07232h-f5.tif
Fig. 5 Graphical representation of two-way ANOVA of all the synthesized drugs against the K562 cell line.

3.2.1.1. Two-way ANOVA application on anti-tumor results against the K562 cell line. The ANOVA values (Table 1 and Fig. 5) of the antitumor activity of the compounds (3a, 3c, 6g, 6h and 6i) against the K562 cell line have indicated extreme variability of all factors. The row factor (compounds) obtained an F-value of 31[thin space (1/6-em)]988 (*P* < 0.0001), which meant that there was a lot of variation between the effects of the compounds. The interaction term and the column factor (which would be the likely concentrations) also displayed significant F-values of 19[thin space (1/6-em)]297 and 4[thin space (1/6-em)]089, respectively, with *P* < 0.0001. This is supported by the fact that the residual variance (MS = 0.5) is very low, which implies that the results are accurate and statistically significant.
Table 1 Two-way ANOVA results of all the synthesized drugs against the K562 cell line
ANOVA table SS DF MS F (DFn, DFd) P Value
Interaction 30[thin space (1/6-em)]666 15 2044 F (15, 24) = 4089 P < 0.0001
Row factor 47[thin space (1/6-em)]982 3 15[thin space (1/6-em)]994 F (3, 24) = 31[thin space (1/6-em)]988 P < 0.0001
Column factor 48[thin space (1/6-em)]244 5 9649 F (5, 24) = 19[thin space (1/6-em)]297 P < 0.0001
Residual 12.00 24 0.5000    



3.2.1.1.1 Tukey's test. Tukey's post hoc indicated that there is a statistically significant difference between Row 1 and every other row, with the greatest mean difference occurring between Row 1 and Row 3 (78.67, q = 385.4). The mean differences as observed by the comparison of Row 2, Row 3 and Row 4 are smaller, and the least significant of them is the comparison of Row 2 to Row 4 (difference in means = −5.33, q = 26.13). Compared to Row 1, the strong statistical significance is also illustrated by high q values (>300). In general, the variability between Row 1 and the others is very high, and the differences between the second, third and fourth rows are relatively not high (Table S2: SI).
3.2.1.1.2 Bonferroni's test. The multiple comparison test developed by Bonferroni proves that the differences between Row 1 and the others are highly significant, with the greatest average difference being between Row 1 and Row 3 (78.67, t = 272.5). The results of the comparison (Row 2 vs. 3; t = 23.67, Row 2 vs. 4; t = 18.48 and Row 3 vs. 4; t = 42.15) are also statistically different having lower t values. The standard error is very small (0.2887) and the t values are very large; hence, there is a likelihood that all the pairwise comparisons are significant even after Bonferroni's correction. These findings lead to the conclusion that Row 1 is significantly more different than the other rows, whereas the other rows are different to a smaller degree (Table S3: SI).
3.2.2. Antitumor activity data against cell line U937. The anticancer properties of the chosen compositions against the U937 cell line and IC50 were determined from the plot of molar absorbance (percent) against concentration (µM). The most promising compound in tests of cytotoxicity activity was 6h with an IC50 value of 1.515, followed by 6i (2.453), 3c (3.540 2), 3b (5.289 2), and 6g (5.706 2) (Fig. 6). The weakest activity was observed with the compound 3a whose IC50 is 150.8 µM. These compounds also show good inhibitory concentration (IC50) against the U937 cell line, and the compounds were evaluated at multiple concentrations: 0.5 µM, 0.6 µM, 0.7 µM, 0.8 µM, 0.9 µM and 1.0 µM, as shown in Fig. 6 by the descriptive data. These findings indicate the fact that 6h and 6i were the best candidates that can be further examined in terms of anticancer activity against U937 cells.37
image file: d5ra07232h-f6.tif
Fig. 6 Graphical representation of the U937 cell line inhibitory concentration of the synthesized drugs.

3.2.2.1. Two-way ANOVA application on anti-tumor results against the U937 cell line. The two-way ANOVA was applied on the results for compounds tested to reveal the effect on the U937 cell line showed that the differences are significantly high across the factors (Table 2 and Fig. 7). The row factor (compounds) exhibits an F-value of 43[thin space (1/6-em)]429 (P < 0.0001), which indicates that there is a large variation in the antitumor activity among the compounds. The column factor (warmer-probable concentrations) also reveals a huge influence with an F-value of 15[thin space (1/6-em)]201 (P < 0.0001). The interaction effect (compound × concentration) is also significant, F = 12[thin space (1/6-em)]051, and shows different patterns of response to treatments. The reliability of the model is established by the low residual variance.
Table 2 Two-way ANOVA of all the synthesized drugs against the U937 cell line
ANOVA table SS DF MS F (DFn, DFd) P Value
Interaction 120[thin space (1/6-em)]506 15 8034 F (15, 24) = 12[thin space (1/6-em)]051 P < 0.0001
Row factor 86[thin space (1/6-em)]859 3 28[thin space (1/6-em)]953 F (3, 24) = 43[thin space (1/6-em)]429 P < 0.0001
Column factor 50[thin space (1/6-em)]671 5 10[thin space (1/6-em)]134 F (5, 24) = 15[thin space (1/6-em)]201 P < 0.0001
Residual 16.00 24 0.6667    



image file: d5ra07232h-f7.tif
Fig. 7 Graphical representation of the U937 cell line inhibitory concentration of the synthesized drugs.

3.2.2.1.1 Turkey's test. All the pairs of compounds give a significant difference, as observed in Tukey's test of the U937 cell line. The greatest contrast is that between Row 2 and Row 4 (mean diff. = 96.67, q = 410.1), Row 3 and Row 4 (q = 376.2) and Row 1 and Row 2 (q = 336.6). Even the smallest difference, Row 2 vs. Row 3 (q = 33.94), is statistically significant (because of the low standard error). These findings support the fact that all the compounds behave differently but in a significant manner in their antitumor effects in the U937 cell line, and Row 4 behaves differently, as represented in Table S4 in the SI.
3.2.2.1.2 Bonferroni's test. Bonferroni's post-hoc test of the U937 cell line affirms as highly true the existence of differences between compound pairs. The largest difference was calculated between Rows 2 and 4 (t = 290.0) and 3 and 4 (t = 266.0), which means that a high variation was observed in terms of antitumor effects (Table S5, SI). The differences between Row 1 and Row 2 (t = 238.0) and Row 1 and Row 3 (t = 214.0)38 were also quite significant. The smallest contrast, Row 2 vs. Row 3 (t = 24.0) is still significant, even after Bonferroni's correction because the standard error is very small. The findings demonstrate clear and strong activity variances among the compounds that are tested (Table S5: SI).
3.2.3. Structure–activity relationship of the targeted compounds. The structure–activity relationship of targeted compounds was found to be mainly structured/influenced at the 4-position at the ring of thiazole as a site of substitution (Fig. 8), which is predominant in the anticancer activity. Adding small electron-donating alkyl groups (compound 3a and 6g) greatly increased the cytotoxicity, but phenyl-aromatic usage (3b, 6h) increased π–π stacking interactions with key residues, which reduced activity moderate to high. Electron-withdrawing acyl (–COCH3) groups of compounds 3e and 6j significantly lowered the activity. At position-2, unsubstituted hydrogen preferred the most favorable binding orientation, whereas a flexible linker like –CH2–CH2– in compound 6i increased the flexibility at the binding pocket, leading to intensive H-bonding and halogen interactions. Amino substituents in compound 3f that were bulky reduced the activity because of steric hindrance. Altogether, 3a, 6g, 6h and 6i proved to be the most effective ones, and the evidence of their power was based on the in vitro cytotoxicity and docking evidence.
image file: d5ra07232h-f8.tif
Fig. 8 Structure–activity relationship of thaizole derivatives.

3.3. Docking analysis

On the basis of electrostatic interaction between the protein and the ligand, molecular interaction also predicts the binding conformation that can be quantified. All interactions were observed to be docking scores of ligands. The docking score is shown as a negative energy value (kcal mol−1). A lower energy value shows strong interaction between the protein and ligands.39 Docking studies give the best binding conformation ligands and interaction of residues and ligands at enzymes active sites. 5MO4 and 2GQG proteins were selected for the chronic myeloid leukemia model K562 cell line and 2AZ5 and 5MAR proteins for the chronic myeloid leukemia model U937 cell line. Docking result showed that the good binding energy of ligands than the standard drug (5-fluorouracil) was observed, as 6 synthesized compounds out of 12 compounds showed strong interaction with 2GQG protein, 6 compounds with 5MO4 protein, 4 compounds with 2AZ5 protein and 5 compounds with 5MAR protein shown in Table 3.
Table 3 Docking scores with the corresponding interactions
S. no. PDB ID Compounds Docking score Type of interaction Amino acid residue Distance
1 2GQG 5-Fluorouracil −7.064 H-bond MET318 1.91
H-bond MET318 1.86
3a −6.727 H-bond MET318 2.38
H-bond MET318 1.83
3b −6.245 H-bond LEU248 2.42
3c −6.472 H-bond GLU286 2.42
3d −6.147 H-bond GLU286 1.89
3e −7.688 H-bond MET318 2.21
H-bond MET318 1.88
3f −7.606 H-bond MET318 2.20
H-bond MET318 2.21
H-bond GLU316 1.87
6g −6.700 H-bond THR315 2.18
6h −8.370 H-bond MET318 2.28
H-bond MET318 1.87
6i −8.973 H-bond MET318 2.09
H-bond THR315 2.28
Halogen bond TYR253 3.19
Halogen bond GLY251 3.09
6j −7.284 H-bond MET318 2.40
H-bond MET318 1.86
7 −7.184 H-bond GLU316 1.78
2 5MO4 5-Fluorouracil −6.329 H-bond MET337 2.10
Pi–Pi stacking TYR272 5.02
3a −5.590
3b −7.447 Pi–Pi stacking TYR272 4.81
3c −5.870 H-bond ASP400 2.32
3d −6.741 H-bond GLU305 2.63
3e −6.791 H-bond MET337 2.22
Pi–Pi stacking TYR272 5.14
3f −6.212 H-bond ALA288 2.18
H-bond ILE332 2.12
6g −8.441 H-bond ASP400 2.32
Pi–Pi stacking TYR272 5.03
6h −7.292 H-bond ASP400 1.88
Pi-cation LYS290 6.55
6i −7.384 H-bond ASP400 2.40
H-bond GLU305 2.24
6j −6.154 H-bond ASP400 2.55
7 −6.044
3 2AZ5 5-Fluorouracil −5.598 H-bond TYR B:151 1.97
H-bond LEU B:120 2.74
3a −4.960 H-bond SER B:60 2.06
Pi–Pi stacking TYR B:119 4.98
3b −5.923 H-bond LEU A:120 1.85
Pi–Pi stacking TYR B:119 5.35
3c −5.054 H-bond LEU A:120 2.27
H-bond LEU A:120 2.16
H-bond SER B:60 2.47
Pi–Pi stacking TYR B:119 5.12
3d −5.335 H-bond SER B:60 2.22
3e −5.494 H-bond GLY A:121 1.99
3f −5.613 H-bond GLY A:121 1.84
H-bond TYR B:151 2.34
6g −5.339 H-bond LEU A:120 3.47
Pi–Pi stacking TYR A:59 4.18
6h −5.838
6i −5.118 Halogen bond TYR B:119 3.42
H-bond GLY A:121 2.34
6j −5.030 H-bond GLY A:121 2.19
7 −6.289 H-bond SER A:60 2.19
Pi–Pi stacking TYR B:59 5.09
Pi–Pi stacking TYR B:119 5.49
4 5MAR 5-Fluorouracil −6.676 H-bond LYS 287 2.19
H-bond ASN286 1.85
H-bond ASP95 1.68
3a −5.130 H-bond ALA85 2.73
H-bond GLY261 2.12
3b −4.653 H-bond SER263 2.65
3c −6.292 H-bond GLU288 1.83
H-bond ARG97 2.01
H-bond PHE96 1.89
3d −5.071 H-bond ASP95 1.95
H-bond GLU288 2.07
3e −5.246 H-bond ALA85 2.37
3f −5.528 H-bond GLU288 1.68
H-bond ASN286 2.09
6g −5.398 Halogen bond ARG97 2.82
Halogen bond TYR104 2.42
6h −5.977 H-bond SER263 2.56
H-bond ALA85 2.28
6i −5.535 H-bond ARG A:97 2.60
H-bond SER A:263 2.04
H-bond HIS A:187 2.31
Halogen bond ARG A:97 3.48
Halogen bond TYR A:104 2.59
6j −5.534 H-bond SER263 2.21
H-bond ALA85 1.97
Halogen bond ARG97 2.64
7 −4.378


3.3.1. 2GQG protein. With protein 2GQG, out of all docked compounds (Fig. S1–S5: SI and Table 3), six compounds showed a good binding score as compared to the standard drug. Compound 3e showed 2H-bond interactions with the amino acid residue MET318 along with distances of 2.21 and 1.88 Å (Fig. 9(a and b)). Compound 3f showed 3H-bond interactions with the amino acid residues MET318, MET318 and GLU316 with distances of 2.20, 2.21 and 1.87 Å (Fig. 9(c and d)). Compound 6h showed 2H-bond interactions with the amino acid residue MET318 with distances of 2.28 and 1.87 Å (Fig. 10(a and b)). Compound 6i showed 2H-bond and 2 halogen bond interactions with the amino acid residues MET318, THR315, TYR253 and GLY251 with distances of 2.09, 2.28, 3.19 and 3.09 Å (Fig. 10(c and d)). Compound 6j showed 2H-bond interactions with the amino acid residue MET318 along with distances 2.40 and 1.86 Å (Fig. 11(a and b)). Out of the docked compounds, compound 7 show 1H-bond interaction with the amino acid residue GLU316 with a distance of 1.78 Å (Fig. 11(c and d)). In comparison with the above-mentioned compounds, 5-flourouracil standard drug shows −7.064 docking score (Fig. S6: SI).
image file: d5ra07232h-f9.tif
Fig. 9 (a) 3D-interaction of 3e in complex with the protein 2GQG; (b) 2D interaction of 3e in complex with the protein 2GQG; (c) 3D interaction of 3f in complex with the protein 2GQG; and (d) 2D interaction of 3f in complex with the protein 2GQG.

image file: d5ra07232h-f10.tif
Fig. 10 (a) 3D interaction of 6h in complex with the protein 2GQG; (b) 2D interaction of 6h in complex with the protein 2GQG; (c) 3D interaction of 6i in complex with the protein 2GQG; and (d) 2D interaction of 6i in complex with the protein 2GQG.

image file: d5ra07232h-f11.tif
Fig. 11 (a) 3D interaction of 6j in complex with the protein 2GQG; (b) 2D interaction of 6j in complex with the protein 2GQG; (c) 3D interaction of compound 7 in complex with the protein 2GQG; and (d) 2D interaction of compound 7 in complex with the protein 2GQG.
3.3.2. Docking analysis in complex with the 5MO4 protein. From protein 5MO4 docking results of all compounds (Fig. S7–S12: SI and Table 3), 6 compounds show a good binding score as compared to the standard drug. Compound 3b showed the Pi–Pi stacking interaction with the amino acid residue TYR272 with a distance of 4.81 Å (Fig. 12(a and b)). Compound 3d showed 1H-bond interaction with the amino acid residue GLU305 with a distance of 2.63 Å (Fig. 12(c and d)). Compound 3e showed 1H-bond and Pi–Pi stacking interactions with the amino acid residues MET337 and TYR272 with distances of 2.22 and 5.14 Å (Fig. 13(a and b)). Compound 6g showed 1H-bond and Pi–Pi stacking interactions with the amino acid residues ASP400 and TYR272 with distances of 2.32 and 5.03 Å (Fig. 13(c and d)). Compound 6h showed 1H-bond and Pi-cation interactions with the amino acid residues ASP400 and LYS290 with distances of 1.88 and 6.55 Å (Fig. 14(a and b)). Compound 6i showed 2H-bond interactions with the amino acid residues ASP400 and GLU305 with distances of 2.40 and 2.24 Å (Fig. 14(c and d)). The standard drug 5-flourouracil showed a docking score of −6.329 with the 5MO4 protein as compared to those mentioned above (Fig. S12, SI).
image file: d5ra07232h-f12.tif
Fig. 12 (a) 3D interaction of 3b in complex with the protein 5MO4; (b) 2D interaction of 3b in complex with the protein 5MO4; (c) 3D interaction of 3d in complex with the protein 5MO4; (d) 2D interaction of 3d in complex with the protein 5MO4.

image file: d5ra07232h-f13.tif
Fig. 13 (a) 3D interaction of 3e in complex with the protein 5MO4; (b) 2D interaction of 3e in complex with the protein 5MO4; (c) 3D interaction of 6g in complex with the protein 5MO4; and (d) 2D interaction of 6g in complex with the protein 5MO4.

image file: d5ra07232h-f14.tif
Fig. 14 (a)3D interaction of 6h in complex with the protein 5MO4; (b) 2D interaction of 6h in complex with the protein 5MO4; (c) 3D interaction of 6i in complex with the protein 5MO4; and (d) 2D interaction of 6i in complex with the protein 5MO4.
3.3.3. 2AZ5 protein. From the protein 2AZ5, out of all the compounds, 4 compounds showed a good binding score in comparison to the standard drug (Fig. S13–S18: SI and Table 3). Compound 3b showed 1H-bond and Pi–Pi stacking interactions with the amino acid residues LEU A: 120 and TYR B: 119 with distances of 1.85 and 5.35 (Fig. 15(a and b)). Compound 3f showed 2H-bond interactions with the amino acid residues GLY A: 121 and TYR B: 151 with distances of 1.84 and 2.34 (Fig. 15(c and d)). Compound 6h showed no interaction with the amino acid residue, but the docking score is higher than that of the standard drug (Fig. 16(a and b)). Compound 7 showed 1H-bond and 2Pi–Pi stacking interactions with the amino acid residues SER A:60, TYR B:59 and TYR B:119 with distances of 2.19, 5.09 and 5.49 (Fig. 16(c and d)). 5-Flourouracil standard drug showed −5.598 docking score with the 2AZ5 protein (Fig. S19: SI).
image file: d5ra07232h-f15.tif
Fig. 15 (a) 3D interaction of 3b in complex with the protein 2AZ5; (b) 2D interaction of 3b in complex with the protein 2AZ5; (c) 3D interaction of 3f in complex with the protein 2AZ5; and (d) 2D interaction of 3f in complex with the protein 2AZ5.

image file: d5ra07232h-f16.tif
Fig. 16 (a) 3D interaction of 6h in complex with the protein 2AZ5; (b) 2D interaction of 6h in complex with the protein 2AZ5; (c) 3D interaction of compound 7 in complex with the protein 2AZ5; and (d) 2D interaction of compound 7 in complex with the protein 2AZ5.
3.3.4. 5MAR protein. From the protein 5MAR, 5 compounds showed a good binding score as compared to the standard drug and other compounds (Fig. S20–S25: SI and Table 3). Compound 3c showed 3H-bond interactions with the amino acid residues GLU288, ARG97 and PHE96 with distances of 1.83, 2.01 and 1.89 (Fig. 17a and b). Compound 3f showed 2H-bond interactions with the amino acid residues GLU288 and ASN286 with distances of 1.68 and 2.09 (Fig. 17c and d). Compound 6h showed 2H-bond interactions with the amino acid residues SER263 and ALA85 with distances of 2.56 and 2.28 (Fig. 18a and b). Compound 6i showed 3H-bond and 2 halogen bond interactions with the amino acid residues ARG A:97, SER A:263, HIS A:187, ARG A:97 and TYR A:104 with distances of 2.60, 2.04, 2.31, 3.48 and 2.59 (Fig. 18c and d). Compound 6j showed 2H-bond and 1 halogen bond interactions with the amino acid residues SER263, ALA85 and ARG97 with distances of 2.21, 1.97 and 2.64 (Fig. 19a and b). In comparison to the above-mentioned compounds, the 5-flourouracil standard drug showed −6.676 docking score with the 5MAR protein (Fig. S26: SI).
image file: d5ra07232h-f17.tif
Fig. 17 (a)3D interaction of 3c in complex with the protein 5MAR; (b) 2D interaction of 3c in complex with the protein 5MAR; (c) 3D interaction of 3f in complex with the protein 5MAR; and (d) 2D interaction of 3f in complex with the protein 5MAR.

image file: d5ra07232h-f18.tif
Fig. 18 (a) 3D interaction of 6h in complex with the protein 5MAR; (b) 2D interaction of 6h in complex with the protein 5MAR; (c) 3D interaction of 6i in complex with the protein 5MAR; and (d) 2D interaction of 6i in complex with the protein 5MAR.

image file: d5ra07232h-f19.tif
Fig. 19 (a) 3D interaction of 6j in complex with the protein 5MAR and (b) 2D interaction of 6j in complex with the protein 5MAR.

3.4. MM-GBSA analysis of complexes

After molecular docking studies, protein–ligand complexes were subjected to the Prime MM-GBSA method to calculate the binding free energy.40 MMBSA was performed on chronic myeloid leukemia model K562 cell line proteins (5MO4 and 2GQG) and chronic myeloid leukemia model U937 cell line proteins (2AZ5 and 5MAR) with all ligands. On protein preparation, the 5MO4 protein was screened out among all other proteins because the highest binding free energy was observed as −53.36, respectively. The other proteins 2GQG, 2AZ5 and 5MAR showed −51.54, −36.05 and −21.92 kcal mol−1 negative binding free energies values. 5MO4 protein shows the negative binding free energy value of Coulomb, lipophilic and hydrogen bonding contributes positively while covalent binding free energy values contribute unfavorably (Table 4). Higher value of protein–ligand complex binding free energy suggested that the higher the affinities of protein–ligand docking along with their docking score validation will be higher.
Table 4 Binding free energy study of protein–ligand complexes using the Prime MM-GBSA method
S. no. Protein–ligand complex ΔG bind (kcal mol−1) ΔG bind coulomb ΔG bind covalent ΔG bind H-bond ΔG bind lipophilic ΔG bind packing
1 5MO4–6g −53.36 −11.96 3.07 −1.08 −21.11 −1.79
2 5MO4–3b −36.05 −7.2 3.33 −0.75 −17.53 −1.91
3 5MO4–6i −44.94 −16.36 2.26 −1.35 −12.13 0
4 5MO4–6h −45.11 −11.48 −0.97 −0.58 −13.53 0
5 5MO4–3e −34.27 −6.4 1.12 −0.77 −14.61 −0.84
6 5MO4–3d −36.19 −11.74 1.36 −1.61 −13.24 0
7 5MO4–5-fluorouracil −23.95 −6.27 0.15 −0.55 −4.7 −0.79
8 5MO4–3f −24.86 −14.56 1.32 −1.6 −4.28 0
9 5MO4–6j −36.41 −10.81 −0.94 −1.01 −8.84 0
10 5MO4–7 −4.96 −0.84 14.93 −0.44 −20.87 −0.8
11 5MO4–3c −25.01 −11.35 3.26 −1.46 −5.71 0
12 5MO4–3a −17.65 −5.09 0.26 −0.76 −5.11 0


3.5. Geometry and spectroscopic evaluation by density functional theory analysis

Density functional theory (DFT) was used for the detailed molecular structure study of the synthesized compounds and their structure–activity relationship by calculating their chemical descriptors and geometrical parameters.41 The B3LYP functional set was used to evaluate the closely related theoretical values in comparison with the experimental values. The 2D structure of these synthesized compounds was converted into a 3D structure using PerkinElmer ChemDraw and ChemDraw 3D. Then these compounds were run on the Gaussian09W software, and DFT calculations were done for optimizing their structures42 The DFT calculation results for ionization potential, electron affinity, energy gap, absolute softness and hardness, electronegativity, nucleophilic and electrophilic index and chemical potential values are presented in Table 5.
Table 5 Chemical descriptors of compounds 6h and 6i calculated by the B3LYP 6-311G basis set of DFT
Parameters 6h Compound 6i Compound
EHOMO −0.27911 (−7.5949 hartree) −0.27423 (−7.4622 hartree)
ELUMO −0.04535 (−1.2340 hartree) −0.07854 (−2.1372 hartree)
Ionization potential (I) 7.5949 7.4622
Electron affinity (A) 1.2340 2.1372
Energy gap (ΔE) 6.3609 5.325
Absolute hardness (η) 3.1804 2.6625
Absolute softness (σ) 0.3144 0.3755
Electronegativity (χ) 4.4144 4.7997
Chemical potential (CP) −4.4144 −4.7997
Electrophilicity index (ω) 3.0635 4.3262
Nucleophilicity index (N) 0.3264 0.2311
Additional electronic charges (ΔNmax) 1.3880 1.8027
Global softness (S) 0.1572 0.1877


3.5.1. Natural bond orbital (NBO) analysis. Interpretation of the data provided by natural bond analysis (NBO) regarding the intra-molecular and intermolecular interactions, affected the molecular stabilization. Natural bond analysis gives information about electron density distribution between Lewis and non-Lewis-type donors that are bonding and anti-bonding orbitals from the second-order perturbation theory study. NBO analysis was performed using the Gaussian 09 program, which provides an understanding of the intramolecular and intermolecular interactions with their stability.43 Stabilization energy (E2) of compound 6h for electron acceptor interaction is higher than the electron donor interaction. Eqn (1) illustrates the stabilization energy (E2) calculation according to the second-order perturbation theory:
 
image file: d5ra07232h-t1.tif(1)
where E2 is the stabilization energy, F(i, j) is the diagonal, and εj and εi are also off diagonal of the Fock matrix element in NBOs. The structural representation and numbering of 6h (2-chloro-N-(5,6-dihydro-4H cyclopenta[d]thiazol-2-ylacetamide)) and 6i (N,N′-(4,4′-carbonylbis(thiazole-4,2-diyl))bis-(2-chloroacetamide)) compounds are represented in Fig. S27 (SI).

Natural Bond Orbital (NBOs) analysis for the lead compound 6h (2-chloro-N-(5, 6-dihydro-4H cyclopenta[d]thiazol-2-yl)acetamide) mentioned in Table shows the highest transition observed as π (C2–N3) to π* (C4–C5) with 19.16 kcal mol−1 stabilization energy. Its highest energy value shows high stabilization and the strongest interaction between the electron donor and acceptor. The second highest transition is π (C4–C5) to π* (C2–N3) with a high stabilization energy of 10.6 kcal mol−1. The σ (C4–C8)-to-σ* (C7–C8) transition shows the smallest stabilization energy as 0.51 kcal mol−1. Its lowest stabilization energy shows weak interactions between the electron donor (σ) and acceptor (σ*). Other σ (C10–C12)-to-σ* (C10–C11) transitions also exhibit the lowest stabilization energy of 0.51 kcal mol−1. Similarly, LP1 (N9) to π* (C10–C11), LP1 (N9) to π* (C2–N3) and LP2 (S1) to π* (C2–N3) transitions were observed at the highest stabilization energies of 56.91 kcal mol−1, 49.68 kcal mol−1 and 31.07 kcal mol−1, while the LP2 (O11)-to-σ* (C2–N9) transition produces the lowest stabilization energy of 0.61 kcal mol−1 that shows the low interaction energy (Table 6).

Table 6 Second-order perturbation theory analysis of Fock matrix in NBO basis for the compound 6ha
S. no. Donor (i) Type Acceptor (j) Type E2 (kcal mol−1) EiEj Fi, j
a *E(2) means the energy of the hyper conjugative interaction (stabilization energy in kcal mol−1). *Energy difference E(j)E(i) between the donor and the acceptor i and j NBO orbitals. *F(i, j) is the Fock matrix element between the i and j NBO orbitals.
1 S1–C2 Σ C4–C8 σ* 1.47 1.12 0.036
2 S1–C2 Σ C5–C6 σ* 7.11 1.12 0.08
3 S1–C2 Σ N9–H20 σ* 2.91 1.05 0.05
4 S1–C5 Σ C2–N9 σ* 4.11 1.22 0.064
5 S1–C5 Σ N3–C4 σ* 0.74 1.22 0.027
6 S1–C5 Σ C4–C5 σ* 1.1 1.37 0.035
7 S1–C5 Σ C4–C8 σ* 4.25 1.19 0.064
8 S1–C5 Σ C5–C6 σ* 0.86 1.19 0.029
9 S1–C5 Σ C6–C7 σ* 0.68 1.15 0.025
10 C2–N3 Σ C2–N9 σ* 2.75 1.32 0.054
11 C2–N3 Σ N3–C4 σ* 0.93 1.32 0.031
12 C2–N3 Σ C4–C8 σ* 6.17 1.29 0.08
13 C2–N3 Σ N9–C10 σ* 3.45 1.28 0.06
14 C2–N3 Π C2–N3 π* 0.9 0.3 0.016
15 C2–N3 Π C4–C5 π* 19.16 0.36 0.078
16 C2–N9 Σ C2–N3 σ* 2.16 1.4 0.049
17 C2–N9 Σ N3–C4 σ* 2.35 1.34 0.05
18 C2–N9 Σ N9–C10 σ* 2.09 1.3 0.047
19 C2–N9 Σ N9–H20 σ* 0.6 1.24 0.024
20 C2–N9 Σ C10–O11 σ* 1.12 1.4 0.035
21 N3–C4 Σ C2–N3 σ* 0.6 1.3 0.025
22 N3–C4 Σ C2–N9 σ* 7.63 1.24 0.087
23 N3–C4 Σ C4–C5 σ* 2.18 1.39 0.049
24 N3–C4 σ C4–C8 σ* 1.32 1.21 0.036
25 N3–C4 σ C5–C6 σ* 2.43 1.21 0.048
26 C4–C5 σ S1–C5 σ* 0.88 0.99 0.026
27 C4–C5 σ C2–N9 σ* 1.51 1.19 0.038
28 C4–C5 σ N3–C4 σ* 1.91 1.19 0.043
29 C4–C5 σ C4–C8 σ* 3.08 1.16 0.053
30 C4–C5 σ C5–C6 σ* 4.24 1.16 0.063
31 C4–C5 σ C8–H18 σ* 0.53 1.21 0.023
32 C4–C5 π C2–N3 π* 10.6 0.26 0.05
33 C4–C5 π C6–H14 σ* 3.65 0.74 0.048
34 C4–C5 π C6–H14 σ* 3.09 0.65 0.041
35 C4–C5 π C8–H18 σ* 2.65 0.75 0.041
36 C4–C5 π C8–H19 σ* 2.79 0.73 0.042
37 C4–C8 σ S1–C5 σ* 5.24 0.86 0.06
38 C4–C8 σ C2–N3 σ* 2.12 1.12 0.044
39 C4–C8 σ N3–C4 σ* 1.41 1.06 0.035
40 C4–C8 σ C4–C5 σ* 3.56 1.21 0.059
41 C4–C8 σ C6–C7 σ* 0.56 0.99 0.021
42 C4–C8 σ C7–C8 σ* 0.51 0.96 0.02
43 C4–C8 σ C7–H17 σ* 0.66 1.02 0.023
44 C4–C8 σ C8–H18 σ* 1.08 1.08 0.031
45 C4–C8 σ C8–H19 σ* 0.86 1.06 0.027
46 C5–C6 σ S1–C5 σ* 0.57 0.89 0.02
47 C5–C6 σ N3–C4 σ* 4.56 1.09 0.063
48 C5–C6 σ C4–C5 σ* 4.85 1.24 0.07
49 C5–C6 σ C6–C7 σ* 0.82 1.03 0.026
50 C5–C6 σ C6–H14 σ* 1.02 1.1 0.03
51 C5–C6 σ C6–H15 σ* 0.62 1.01 0.022
52 C5–C6 σ C7–C8 σ* 0.53 0.99 0.02
53 C5–C6 σ C7–H17 σ* 1.36 1.05 0.034
54 C6–C7 σ S1–C5 σ* 9.1 0.85 0.078
55 C6–C7 σ N3–C4 σ* 0.52 1.05 0.021
56 C6–C7 σ C4–C8 σ* 0.54 1.02 0.021
57 C6–C7 σ C5–C6 σ* 1.8 1.02 0.038
58 C6–C7 σ C6–H14 σ* 0.98 1.06 0.029
59 C6–C7 σ C6–H15 σ* 0.54 0.97 0.02
60 C6–H14 σ C4–C5 σ* 1.15 1.11 0.032
61 C6–H14 σ C4–C5 π* 3.52 0.54 0.042
62 C6–H14 σ C5–C6 σ* 0.54 0.93 0.02
63 C6–H14 σ C7–C8 σ* 0.88 0.86 0.025
64 C6–H14 σ C7–H16 σ* 0.89 0.85 0.025
65 C6–H14 σ C7–H17 σ* 1.01 0.92 0.027
66 C6–H15 σ C4–C5 σ* 1.97 1.09 0.041
67 C6–H15 σ C4–C5 π* 2.93 0.52 0.038
68 C6–H15 σ C7–C8 σ* 1.5 0.84 0.032
69 C6–H15 σ C7–H16 σ* 1.22 0.83 0.029
70 C7–C8 σ S1–C5 σ* 1.09 0.81 0.027
71 C7–C8 σ N3–C4 σ* 7.08 1.02 0.076
72 C7–C8 σ C4–C8 σ* 0.93 0.99 0.027
73 C7–C8 σ C5–C6 σ* 0.54 0.99 0.021
74 C7–C8 σ C6–H15 σ* 0.66 0.94 0.022
75 C7–C8 σ C8–H18 σ* 0.76 1.04 0.025
76 C7–C8 σ C8–H19 σ* 0.67 1.01 0.023
77 C7–H16 σ C4–C8 σ* 0.52 0.88 0.019
78 C7–H16 σ C5–C6 σ* 0.58 0.88 0.02
79 C7–H16 σ C6–H14 σ* 1.29 0.92 0.031
80 C7–H16 σ C6–H15 σ* 1.52 0.83 0.032
81 C7–H16 σ C8–H18 σ* 1.15 0.93 0.029
82 C7–H16 σ C8–H19 σ* 1.37 0.9 0.031
83 C7–H17 σ C4–C8 σ* 0.87 0.9 0.025
84 C7–H17 σ C5–C6 σ* 1.16 0.91 0.029
85 C7–H17 σ C6–H14 σ* 1.2 0.95 0.03
86 C7–H17 σ C8–H18 σ* 0.89 0.95 0.026
87 C8–H18 σ C4–C5 σ* 1.52 1.1 0.037
88 C8–H18 σ C4–C5 π* 2.47 0.54 0.035
89 C8–H18 σ C6–C7 σ* 0.82 0.89 0.024
90 C8–H18 σ C7–H16 σ* 0.55 0.85 0.019
91 C8–H18 σ C7–H17 σ* 0.84 0.92 0.025
92 C8–H19 σ C4–C5 σ* 1.15 1.08 0.032
93 C8–H19 σ C4–C5 π* 5.11 0.52 0.049
94 C8–H19 σ C6–C7 σ* 1.11 0.87 0.028
95 C8–H19 σ C7–H16 σ* 1.28 0.83 0.029
96 N9–C10 σ C2–N3 σ* 1.94 1.37 0.046
97 N9–C10 σ C2–N9 σ* 2.6 1.3 0.052
98 N9–C10 σ C12–Cl13 σ* 0.84 0.9 0.025
99 N9–H20 σ S1–C2 σ* 6.65 0.79 0.066
100 N9–H20 σ C2–N3 σ* 0.94 1.15 0.03
101 N9–H20 σ C2–N9 σ* 0.66 1.09 0.024
102 N9–H20 σ C10–O11 σ* 1.65 1.16 0.039
103 N9–H20 σ C10–C12 σ* 5.37 0.91 0.063
104 C10–O11 σ C2–N9 σ* 3.04 1.48 0.06
105 C10–O11 σ N9–C10 σ* 0.94 1.44 0.033
106 C10–O11 σ C10–C12 σ* 0.66 1.3 0.027
107 C10–O11 π C10–O11 π* 1.06 0.35 0.018
108 C10–O11 π C12–H21 σ* 0.74 0.77 0.021
109 C10–O11 π C12–H22 σ* 1.58 0.8 0.032
110 C10–C12 σ N9–H20 σ* 2.07 0.99 0.04
111 C10–C12 σ C10–O11 σ* 0.51 1.15 0.022
112 C12–Cl13 σ N9–C10 σ* 2.74 1.03 0.048
113 C12–Cl13 σ C10–O11 σ* 0.59 1.14 0.023
114 C12–H21 σ C10–O11 σ* 2.56 1.07 0.047
115 C12–H21 σ C10–O11 π* 1.59 0.54 0.028
116 C12–H21 σ C12–Cl13 σ* 0.79 0.6 0.02
117 C12–H22 σ C10–O11 π* 4.2 0.55 0.046
118 C12–H22 σ C12–Cl13 σ* 0.91 0.61 0.021
119 S1 LP1 C2–N3 σ* 3.14 1.15 0.054
120 S1 LP1 C4–C5 σ* 2.41 1.23 0.049
121 S1 LP1 C5–C6 σ* 0.8 1.06 0.026
122 S1 LP1 C12–Cl13 σ* 0.75 0.68 0.02
123 S1 LP2 C2–N3 π* 31.07 0.24 0.078
124 S1 LP2 C4–C5 π* 25.32 0.29 0.078
125 N3 LP1 S1–C2 σ* 17.31 0.5 0.084
126 N3 LP1 C2–N9 σ* 3.22 0.8 0.046
127 N3 LP1 C4–C5 σ* 6.29 0.95 0.07
128 N3 LP1 C4–C8 σ* 0.79 0.77 0.023
129 N9 LP1 S1–C2 σ* 0.99 0.43 0.02
130 N9 LP1 C2–N3 π* 49.68 0.25 0.102
131 N9 LP1 C10–O11 π* 56.91 0.26 0.112
132 O11 LP1 N9–C10 σ* 1.51 1.1 0.037
133 O11 LP1 C10–C12 σ* 1.66 0.95 0.036
134 O11 LP2 C2–N9 σ* 0.61 0.7 0.019
135 O11 LP2 N9–C10 σ* 22.05 0.66 0.109
136 O11 LP2 C10–C12 σ* 22.14 0.52 0.096
137 Cl13 LP2 C12–H21 σ* 3.02 0.7 0.041
138 Cl13 LP2 C12–H22 σ* 3.55 0.73 0.046
139 Cl13 LP3 C10–C12 σ* 4.91 0.57 0.048
140 Cl13 LP3 C12–H22 σ* 1.26 0.71 0.027
141 Cl13 LP3 C12–H22 σ* 0.62 0.74 0.019
142 C2–N3 π* C4–C5 π* 41.07 0.06 0.07
143 C2–N3 π* C10–O11 π* 1.02 0.01 0.005
144 C4–C5 π* C6–H14 σ* 1.05 0.43 0.047
145 C4–C5 π* C6–H15 σ* 0.79 0.34 0.036
146 C4–C5 π* C8–H18 σ* 0.85 0.44 0.043
147 C4–C5 π* C8–H19 σ* 1.06 0.42 0.046
148 C10–O11 π* C12–Cl13 σ* 1.24 0.06 0.021
149 C10–O11 π* C12–H22 σ* 1.06 0.45 0.053


NBO analysis of the compound 6i (N,N′-(4,4′-carbonylbis(thiazole-4,2-diyl)))-bis-(2-chloroacetamide) mentioned in Table 7 shows the highest transition observed as π (C10–N11) to π* (C7–C8) with 19.53 kcal mol−1 stabilization energy. This compound has the highest energy value, which shows high stabilization and the strongest interaction between the electron donor (π) and the acceptor (π*). The second highest transition is π (C7–C8) to π* (C6–O12) with a high stabilization energy of 17.91 kcal mol−1, while σ (C16–Cl17)-to-σ* (C14–O15) transition shows the smallest stabilization energy of 0.51 kcal mol−1. Its lowest stabilization energy shows weak interactions between the electron donor and acceptor. Similarly, LP1 (N13)-to-π* (N1–C2) and LP1 (N18)-to-π* (C19–O20) transitions were observed at the highest stabilization energy of 55.92 kcal mol−1 and 53.66 kcal mol−1, while LP3 (Cl17)-to-σ* (C16–H27) transition produced the lowest stabilization energy of 0.53 kcal mol−1 that shows the low interaction energy (Table 7).

Table 7 Second-order perturbation theory analysis of Fock matrix in NBO basis for the compound 6ia
S. no. Donor (i) Type Acceptor (j) Type E2 (kcal mol−1) EiEj Fi, j
a *E(2) means the energy of the hyper-conjugative interaction (stabilization energy in kcal mol−1). *Energy difference E(j)E(i) between the donor and acceptor i and j NBO orbitals. *F(i, j) is the Fock matrix element between the i and j NBO orbitals.
1 N1–C2 σ N1–C5 σ* 0.73 1.28 0.027
2 N1–C2 σ C2–N13 σ* 2.63 1.33 0.053
3 N1–C2 σ C5–C6 σ* 3.27 1.31 0.059
4 N1–C2 σ N13–H25 σ* 1.39 1.31 0.038
5 N1–C2 π N1–C2 π* 1.31 0.31 0.019
6 N1–C2 π C4–C5 π* 15.52 0.33 0.067
7 N1–C5 σ N1–C2 σ* 0.52 1.27 0.023
8 N1–C5 σ C2–N13 σ* 8.26 1.19 0.089
9 N1–C5 σ C4–C5 σ* 1.33 1.26 0.037
10 N1–C5 σ C4–H23 σ* 2.48 1.18 0.049
11 N1–C5 σ C5–C6 σ* 0.92 1.17 0.03
12 N1–C5 σ C6–C7 σ* 1.1 1.17 0.032
13 N1–C5 σ C6–O12 π* 0.74 0.75 0.022
14 C2–S3 σ C4–H23 σ* 3.34 1.15 0.055
15 C2–S3 σ C5–C6 σ* 0.81 1.13 0.027
16 C2–S3 σ N13–C14 σ* 4.65 1.12 0.065
17 C2–S3 σ N13–H25 σ* 0.74 1.13 0.026
18 C2–N13 σ N1–C2 σ* 2.78 1.39 0.056
19 C2–N13 σ N1–C5 σ* 2.79 1.26 0.053
20 C2–N13 σ N13–C14 σ* 1.42 1.28 0.039
21 C2–N13 σ N13–H25 σ* 0.84 1.29 0.029
22 C2–N13 σ C14–O15 σ* 1.1 1.42 0.035
23 S3–C4 σ N1–C2 σ* 0.68 1.21 0.026
24 S3–C4 σ N1–C5 σ* 1.53 1.08 0.036
25 S3–C4 σ C2–N13 σ* 4.28 1.13 0.063
26 S3–C4 σ C5–C6 σ* 4.98 1.12 0.067
27 C4–C5 σ N1–C5 σ* 0.79 1.12 0.027
28 C4–C5 σ C2–N13 σ* 1.2 1.18 0.034
29 C4–C5 σ C4–H23 σ* 1.55 1.17 0.038
30 C4–C5 σ C5–C6 σ* 2.57 1.16 0.049
31 C4–C5 σ C6–O12 σ* 1.2 1.26 0.035
32 C4–C5 π N1–C2 π* 8.86 0.26 0.046
33 C4–C5 π C4–C5 π* 0.53 0.28 0.011
34 C4–C5 π C6–C7 σ* 1.19 0.71 0.026
35 C4–C5 π C6–O12 σ* 1.2 0.81 0.029
36 C4–C5 π C6–O12 π* 12.07 0.29 0.053
37 C4–H23 σ N1–C5 σ* 3.59 0.95 0.052
38 C4–H23 σ C2–S3 σ* 0.52 0.72 0.018
39 C4–H23 σ S3–C4 σ* 0.69 0.73 0.02
40 C4–H23 σ C4–C5 σ* 1.47 1.07 0.035
41 C5–C6 σ N1–C2 σ* 1.94 1.19 0.043
42 C5–C6 σ N1–C5 σ* 0.8 1.06 0.026
43 C5–C6 σ S3–C4 σ* 2.53 0.84 0.041
44 C5–C6 σ C4–C5 σ* 3.33 1.18 0.056
45 C5–C6 σ C6–C7 σ* 1.34 1.1 0.035
46 C5–C6 σ C6–O12 σ* 1.26 1.2 0.035
47 C5–C6 σ C7–C8 σ* 3 1.17 0.053
48 C6–C7 σ N1–C5 σ* 2.07 1.07 0.042
49 C6–C7 σ C4–C5 π* 0.75 0.68 0.022
50 C6–C7 σ C5–C6 σ* 1.49 1.1 0.037
51 C6–C7 σ C6–O12 σ* 1.06 1.21 0.032
52 C6–C7 σ C7–C8 σ* 3.19 1.17 0.055
53 C6–C7 σ C7–N11 σ* 1.53 1.13 0.037
54 C6–C7 σ C8–S9 σ* 2.09 0.86 0.038
55 C6–C7 σ C10–N11 σ* 3.11 1.18 0.054
56 C6–O12 σ C4–C5 σ* 1.36 1.55 0.041
57 C6–O12 σ C5–C6 σ* 1.94 1.46 0.048
58 C6–O12 σ C6–C7 σ* 1.78 1.47 0.046
59 C6–O12 σ C7–N11 σ* 1.35 1.49 0.04
60 C6–O12 π N1–C5 σ* 1.13 0.75 0.026
61 C6–O12 π C4–C5 σ* 0.72 0.87 0.022
62 C6–O12 π C4–C5 π* 4.8 0.36 0.04
63 C6–O12 π C7–C8 π* 6.82 0.34 0.047
64 C7–C8 σ C6–C6 σ* 1.51 1.16 0.038
65 C7–C8 σ C6–C7 σ* 3.33 1.16 0.056
66 C7–C8 σ C7–N11 σ* 1.43 1.19 0.037
67 C7–C8 σ C8–H24 σ* 1.93 1.2 0.043
68 C7–C8 σ C10–N18 σ* 1.39 1.12 0.035
69 C7–C8 π C6–O12 π* 17.91 0.29 0.065
70 C7–C8 π C7–C8 π* 0.76 0.28 0.013
71 C7–C8 π C10–N11 π* 12.84 0.25 0.054
72 C7–N11 σ C6–C7 σ* 1.13 1.25 0.034
73 C7–N11 σ C6–O12 σ* 1.32 1.36 0.038
74 C7–N11 σ C7–C8 σ* 1.87 1.32 0.044
75 C7–N11 σ C8–H24 σ* 1.82 1.29 0.043
76 C7–N11 σ C10–N11 σ* 1.27 1.32 0.037
77 C7–N11 σ C10–N18 σ* 8.27 1.21 0.09
78 C8–S9 σ C6–C7 σ* 4.85 1.16 0.068
79 C8–S9 σ C7–N11 σ* 1.25 1.18 0.034
80 C8–S9 σ C10–N11 σ* 0.65 1.23 0.025
81 C8–S9 σ C10–N18 σ* 4.18 1.11 0.061
82 C8–H24 σ C7–C8 σ* 1.89 1.07 0.04
83 C8–H24 σ C7–N11 σ* 3.81 1.02 0.056
84 C8–H24 σ S9–C10 σ* 0.74 0.7 0.021
85 S9–C10 σ C6–C7 σ* 0.98 1.14 0.03
86 S9–C10 σ C8–H24 σ* 3.48 1.17 0.057
87 S9–C10 σ N18–C19 σ* 4.13 1.12 0.061
88 S9–C10 σ N18–H28 σ* 0.73 1.14 0.026
89 C10–N11 σ C6–C7 σ* 4.01 1.33 0.066
90 C10–N11 σ C7–N11 σ* 1.33 1.35 0.038
91 C10–N11 σ C10–N18 σ* 1.99 1.29 0.045
92 C10–N11 σ N18–H28 σ* 1.16 1.33 0.035
93 C10–N11 π C7–C8 π* 19.53 0.33 0.076
94 C10–N11 π C10–N11 π* 0.79 0.31 0.015
95 C10–N18 σ C7–N11 σ* 2.74 1.31 0.054
96 C10–N18 σ C10–N11 σ* 2.1 1.36 0.048
97 C10–N18 σ N18–C19 σ* 1.3 1.27 0.037
98 C10–N18 σ N18–H28 σ* 0.78 1.29 0.028
99 C10–N18 σ C19–O20 σ* 1.69 1.38 0.043
100 N13–C14 σ C2–S3 σ* 2.78 0.98 0.047
101 N13–C14 σ C2–N13 σ* 2.52 1.26 0.051
102 N13–C14 σ N13–H25 σ* 0.52 1.24 0.023
103 N13–C14 σ C14–C16 σ* 0.58 1.17 0.024
104 N13–C14 σ C16–Cl17 σ* 1.33 0.95 0.032
105 N13–H25 σ N1–C2 σ* 5.17 1.19 0.07
106 N13–H25 σ C2–N13 σ* 0.54 1.12 0.022
107 N13–H25 σ C14–O15 σ* 1.3 1.23 0.036
108 N13–H25 σ C14–C16 σ* 3.49 1.03 0.054
109 C14–O15 σ C2–N13 σ* 2.74 1.48 0.057
110 C14–O15 σ N13–C14 σ* 0.54 1.45 0.025
111 C14–O15 σ C14–C16 σ* 1.63 1.39 0.043
112 C14–O15 π C14–O15 π* 0.9 0.36 0.017
113 C14–O15 π C16–H26 σ* 1.43 0.75 0.029
114 C14–O15 π C16–H27 σ* 1.53 0.72 0.03
115 C14–C16 σ N13–H25 σ* 2.12 1.08 0.043
116 C14–C16 σ C14–O15 σ* 1.29 1.21 0.035
117 C16–Cl17 σ N13–C14 σ* 3.77 1.08 0.058
118 C16–Cl17 σ C14–O15 σ* 0.51 1.22 0.022
119 C16–H26 σ C14–O15 σ* 2.24 1.06 0.044
120 C16–H26 σ C14–O15 π* 5.94 0.51 0.052
121 C16–H26 σ C16–Cl17 σ* 0.58 0.64 0.017
122 C16–H27 σ C14–O15 σ* 2.15 1.06 0.043
123 C16–H27 σ C14–O15 π* 4.64 0.51 0.046
124 C16–H27 σ C16–Cl17 σ* 0.71 0.64 0.019
125 N18–C19 σ S9–C10 σ* 2.26 0.98 0.043
126 N18–C19 σ C10–N18 σ* 2.12 1.23 0.046
127 N18–C19 σ N18–H28 σ* 0.69 1.27 0.026
128 N18–C19 σ C19–C21 σ* 0.63 1.2 0.025
129 N18–C19 σ C21–Cl22 σ* 1.48 0.97 0.034
130 N18–H28 σ C10–N11 σ* 5.02 1.19 0.069
131 N18–H28 σ C19–O20 σ* 0.95 1.21 0.03
132 N18–H28 σ C19–C21 σ* 3.52 1.05 0.055
133 C19–O20 σ C10–N18 σ* 2.56 1.41 0.054
134 C19–O20 σ N18–C19 σ* 0.92 1.43 0.033
135 C19–O20 σ C19–C21 σ* 1.48 1.38 0.041
136 C19–O20 π C19–O20 π* 0.85 0.35 0.016
137 C19–O20 π C21–H29 σ* 1.48 0.75 0.03
138 C19–O20 π C21–H30 σ* 2.2 0.7 0.035
139 C19–C21 σ N18–C19 σ* 0.55 1.09 0.022
140 C19–C21 σ N18–H28 σ* 2.28 1.11 0.045
141 C19–C21 σ C19–O20 σ* 1.22 1.2 0.034
142 C21–Cl22 σ N18–C19 σ* 3.35 1.08 0.055
143 C21–Cl22 σ C19–O20 σ* 0.61 1.19 0.024
144 C21–H29 σ C19–O20 σ* 2.43 1.04 0.045
145 C21–H29 σ C19–O20 π* 4.06 0.51 0.043
146 C21–H30 σ C19–O20 σ* 0.97 1.03 0.028
147 C21–H30 σ C19–O20 π* 5.29 0.5 0.049
148 C21–H30 σ C21–Cl22 σ* 0.68 0.63 0.019
149 N1 LP1 C2–S3 σ* 17.41 0.52 0.085
150 N1 LP1 C2–N13 σ* 2.25 0.8 0.039
151 N1 LP1 C4–C5 σ* 5.23 0.86 0.061
152 N1 LP1 C5–C6 σ* 0.91 0.78 0.024
153 N1 LP1 C16–Cl17 σ* 1.46 0.48 0.024
154 S3 LP1 N1–C2 σ* 3.63 1.17 0.058
155 S3 LP1 C4–C5 σ* 2.08 1.16 0.044
156 S3 LP2 N1–C2 π* 33.32 0.23 0.08
157 S3 LP2 C4–C5 π* 21.08 0.26 0.067
158 S9 LP1 C7–C8 σ* 2.04 1.16 0.044
159 S9 LP1 C10–N11 σ* 3.16 1.16 0.054
160 S9 LP2 C7–C8 π* 23.71 0.26 0.071
161 S9 LP2 C10–N11 π* 31.24 0.23 0.076
162 N11 LP1 C6–C7 σ* 1.87 0.79 0.035
163 N11 LP1 C7–C8 σ* 6.61 0.86 0.069
164 N11 LP1 S9–C10 σ* 17.14 0.5 0.083
165 N11 LP1 C10–N18 σ* 2.62 0.75 0.041
166 N11 LP1 C21–Cl22 σ* 1.21 0.48 0.022
167 O12 LP1 C5–C6 σ* 1.96 1.1 0.042
168 O12 LP1 C6–C7 σ* 2.58 1.11 0.048
169 O12 LP2 C5–C6 σ* 18.86 0.68 0.102
170 O12 LP2 C6–C7 σ* 17.86 0.68 0.099
171 O12 LP2 C7–N11 σ* 0.59 0.7 0.019
172 N13 LP1 N1–C2 π* 55.92 0.25 0.107
173 N13 LP1 C14–O15 π* 49.44 0.27 0.107
174 O15 LP1 N13–C14 σ* 1.03 1.07 0.03
175 O15 LP1 C14–C16 σ* 2.5 1.02 0.046
176 O15 LP2 C2–N13 σ* 1.1 0.69 0.025
177 O15 LP2 N13–C14 σ* 23.29 0.65 0.111
178 O15 LP2 C14–C16 σ* 20.73 0.59 0.1
179 Cl17 LP2 C16–H26 σ* 4.3 0.67 0.048
180 Cl17 LP2 C16–H27 σ* 4.72 0.64 0.049
181 Cl17 LP3 N13–C14 σ* 0.84 0.68 0.022
182 Cl17 LP3 C14–C16 σ* 5.73 0.63 0.054
183 Cl17 LP3 C16–H26 σ* 2.13 0.67 0.034
184 Cl17 LP3 C16–H27 σ* 0.53 0.64 0.017
185 N18 LP1 C10–N11 π* 51.61 0.24 0.102
186 N18 LP1 C19–O20 π* 53.66 0.27 0.111
187 O20 LP1 N18–C19 σ* 1.65 1.09 0.038
188 O20 LP1 C19–C21 σ* 2.26 1.04 0.044
189 O20 LP2 C10–N18 σ* 0.58 0.63 0.017
190 O20 LP2 N18–C19 σ* 23.66 0.65 0.112
191 O20 LP2 C19–C21 σ* 19.2 0.6 0.097
192 Cl22 LP2 C21–H29 σ* 4.01 0.68 0.047
193 Cl22 LP2 C21–H30 σ* 3.31 0.63 0.041
194 Cl22 LP3 N18–C19 σ* 0.64 0.69 0.019
195 Cl22 LP3 C19–C21 σ* 6.09 0.64 0.056
196 Cl22 LP3 C21–H29 σ* 0.54 0.68 0.017
197 Cl22 LP3 C21–H30 σ* 2.38 0.63 0.035
198 N1–C2 π* C4–C5 π* 69.71 0.02 0.063
199 N1–C2 π* C14–O15 π* 0.56 0.03 0.006
200 C4–C5 π* C4–C5 σ* 0.7 0.51 0.042
201 C4–C5 π* C6–C7 σ* 0.57 0.43 0.033
202 C6–O12 π* N1–C5 σ* 0.59 0.39 0.042
203 C7–C8 π* C6–O12 π* 105.05 0.02 0.077
204 C10–N11 π* C7–C8 π* 84.14 0.03 0.071
205 C10–N11 π* C19–O20 π* 0.53 0.03 0.006
206 C14–O15 π* C16–H26 σ* 0.79 0.39 0.044
207 C14–O15 π* C16–H27 σ* 1.07 0.36 0.05
208 C19–O20 π* C21–H29 σ* 0.83 0.4 0.045
209 C19–O20 π* C21–H30 σ* 1.58 0.35 0.057


3.5.2. Mulliken's population and natural population analysis. Mulliken's population analysis (MPA) and natural population analysis (NPA) of compounds 6h and 6i were determined by the B3LYP method with the 6-311G basis set for the detailed study of reactive sites of the biologically active compounds, as shown in Table S1 (SI). The Mulliken population analysis can be used to describe the phenomena of transformation of atomic charge in reaction, electronegativity equalization and electrostatic potential of system.44 In MPA and NPA analysis all hydrogen atoms show positive charges and all oxygen and nitrogen show negative charges. If hydrogen is attached with a negatively charged carbon atom, then it shows the charge transfer from hydrogen to carbon atom. Similarly, in chemical bonding, nitrogen and oxygen atoms show the same behavior.45 Our goal is to assess the reactivity of the depicted charges using the quantum chemical technique and to provide a detailed description of the electron distribution of these compounds. Charge analysis of compound 6h indicates that N9–H20–O11 is involved in strong intramolecular hydrogen bonding as compared to N3, while C2, C4 and C5 are positively charged and give the best pathway to other chemical moieties for attachment. All hydrogen atoms transform their charges to carbon atoms, and S1 and Cl13 atoms show electron-withdrawing behavior. The graphical representation of compound 6h with Mulliken's population analysis (MPA) and natural population analysis (NPA) is shown in Fig. 20.
image file: d5ra07232h-f20.tif
Fig. 20 Atomic charge analysis: (A) graphical representation of Mulliken's charges for compound 6h, (B) graphical representation of Mulliken's charges with summed hydrogen atoms for compound 6h and (C) natural population charges (NPA) for compound 6h.

The charge analysis of compound 6i indicates that N13–H25–O15 and N18–H28–O20 are involved in strong intramolecular hydrogen bonding as compared to other negatively charged nitrogen N1 and N11. C2, C5, C7 and C10 are also positively charged, providing a path for new moieties to attach. O12, O15 and O20 are the strong carbonyl acceptors and provide an ideal site for intramolecular hydrogen bonding. The graphical representation of compound 6i with Mulliken's population analysis (MPA) and natural population analysis (NPA) is shown in Fig. 21.


image file: d5ra07232h-f21.tif
Fig. 21 Atomic charge analysis: (A) graphical representation of Mulliken's charges for compound 6i, (B) graphical representation of Mulliken's charges with summed hydrogen atoms for compound 6i and (C) natural population charges (NPA) for compound 6i.
3.5.3. Frontier molecular orbital (FMO) analysis. Frontier molecular orbital (FMO) is an important parameter in DFT that is used to check the chemical strength of compounds. Quantum orbitals as the lowest unoccupied molecular orbital (LUMO) and highest occupied molecular orbitals (HOMO) give us qualitative information about HOMO that can donate its electron to LUMO for the measurement of chemically reactive descriptors.46 Energy gap can also be calculated by HOMO–LUMO and provide us information about stability and chemical reactivity of compounds. Contour and energy diagram of frontier molecular orbitals of both compounds along with energy gap is shown in Fig. 22 and 23.
image file: d5ra07232h-f22.tif
Fig. 22 Contour diagram of HOMO–LUMO of compounds 6h (A) and 6i (B) with their energy gap.

image file: d5ra07232h-f23.tif
Fig. 23 HOMO–LUMO energy diagram of compounds 6h (A) and 6i (B) with their energy gap.

The above-mentioned HOMO–LUMO energy values of compounds 6h and 6i are calculated at the B3LYP 6-311G basis set of DFT. Frontier molecular orbitals (FMOs) can also be used to calculated the values of energy gap, softness and hardness, electronegativity and chemical potential. All the calculated energy parameters with their values are shown in Table 5. First descriptive parameter EHOMO is shown in Table 5, and its high value shows that it can donate electron to acceptor molecules. A higher value of EHOMO is also favorable to the biological activity of compound. Compound 6i is a better electron donor than compound 6h due to its high value. The second descriptive parameter is ELUMO, and its low value shows that it can accept electrons from the donating molecule. A lower value of ELUMO also increases the biological activity of the compound. Compound 6i is a better electron acceptor than compound 6h due to its lower value. Third descriptive parameter is energy gap (ΔE) calculated by the HOMO–LUMO values, where the HOMO is more stable than the LUMO. Compound 6i shows a smaller value of energy gap than compound 6h. A smaller energy gap value shows that compound 6i is chemically more reactive and biologically more active.

Frontier molecular orbital (FMO) studies can investigate the structure–activity relationship and bioactivities of these synthesized compounds for drug designing. The HOMO value is higher in compound 6i, which shows that it could be a good electron donor drug. Compound 6i is more reactive and more accepting than the other compound 6h due to its lower ionization potential (I), higher electron affinity (A), lower absolute hardness (η), higher absolute softness (σ), higher electronegativity (χ), lower chemical potential (CP) and higher electrophilicity index (ω).

3.5.4. Molecular electrostatic potential (MEP). The determination of chemical mechanism and investigation of compounds can be done by calculating the MEP contour and map. Hydrogen bonding interaction and electrophilic–nucleophilic behavior of compounds can also be interpreted by MEP. MEP calculations using the Gaussian09W software give us some important indication about molecules by negative, positive and neutral electrostatic potential. The MEP scale gives us information about the relationship of physicochemical properties with the structure of compounds of drugs.47 The MEP contour and map of compounds 6h and 6i were calculated using the same B3LYP 6-311G basis set of DFT.

In MEP, the negatively charged red-yellow color region of electrostatic potential shows the electrophilic attack sites, where O atoms can react with electrophiles, while the positively charged blue region of electrostatic potential showed nucleophilic attack sites where H atoms can react with nucleophiles. The green color showed the neutral reactivity, and the benzene ring remained neutral.48 The molecular electrostatic potential (MEP) contour and map are shown in Fig. 24.


image file: d5ra07232h-f24.tif
Fig. 24 Molecular electrostatic potential (MEP) contour and map of compounds 6h and 6i.
3.5.5. IR analysis. In IR spectroscopic analysis, functional group moieties were determined by vibrational modes, which are present in a molecule. Theoretical calculations were performed for compounds 6h and 6i to gain vibrational analysis information with the 6-311G B3LYP basis set to generate FTIR spectra using an FT-Raman spectrophotometer, which is presented in Table 8. As we study these compounds, compound 6h contains 22 atoms with 60 normal vibrations modes, while compound 6i contains 30 atoms with 84 normal modes of vibrations by these calculations. Fig. 20a and b show the comparison graphs between the experimental and theoretical FTIR data of compounds 6h and 6i.
Table 8 Chemical shift (δ) values of 1H and 13C NMR of compounds 6h and 6i
Carbon δC experimental δC theoretical Proton δH experimental δH theoretical
6h Compound
10-C 164.2 158.8581 20-H 12.5 7.985457
2-C 157.5015 21-H 4.28 6.401765
4-C 148.2 153.7083 22-H 4.28 5.7118
5-C 118.9 124.8353 17-H 2.54 4.081958
12-C 47.99194 14-H 2.87 3.871611
7-C 26.0 29.11105 18-H 2.52 3.657483
8-C 30.9 21.45014 15-H 2.87 3.482611
6-C 28.2 18.3616 16-H 2.54 3.102562
      19-H 2.52 2.79344
[thin space (1/6-em)]
6i Compound
6-C 192.9 197.9515 24-H 8.98 7.796086
14-C 188.8477 23-H 8.98 7.606245
19-C 187.3156 25-H 12.4 7.030382
10-C 177.1132 28-H 12.4 6.484283
2-C 172.8862 27-H 4.87 5.484209
5-C 113.9 172.265 30-H 4.87 5.305767
7-C 164.5 166.9709 26-H 4.87 5.073771
4-C 144.2 158.5567 29-H 4.87 4.94707
8-C 42.7 148.9024      
16-C 67.20365      
21-C 65.9386      



3.5.5.1. N–H, C–N and C[double bond, length as m-dash]N vibrations. The IR vibrational peaks value range for heterocyclic compound where amines (N–H) stretching vibration are from 3200 cm−1 to 3500 cm−1.49 As the N–H, C[double bond, length as m-dash]O, and C–H vibrational modes show the maximum intensity and high polarity. In compound 6h, two polar stretching vibrations are shown for the N–H group at 3310.79 cm−1 and 3344.59 cm−1, while the experimental value peak assigned to the N–H stretching vibration appears at 3319.15 cm−1. There is a little difference between experimental and theoretical wavenumbers because it is sensitive to hydrogen bonding with the oxygen atom of carbonyl functional groups in the H–N–C–O interaction. The bending N–H vibration ranges from 1500 cm−1 to 1650 cm−1, and this one is experimentally observed at 1584.04 cm−1. We observed the C[double bond, length as m-dash]N functional group stretching vibrations at 1640.60 cm−1 experimentally that well match to range found in litrature start from 1610 cm−1 to 1660 cm−1. The thiazole ring consists of a 5-membered ring with a delocalized π-system. The C–N interaction is in plane ring bending vibration. Which is observed from 1020 cm−1 to 1360 cm−1 theoretically and experimentally, and the aromatic C–N vibration is observed at 1117.57 cm−1 and the C–N stretching vibration at 1228.02 cm−1 (Fig. 25).
image file: d5ra07232h-f25.tif
Fig. 25 Theoretical vibrational analysis of compound 6h in the IR study.

In compound 6i, one stretching vibration is shown for the N–H functional group at 3371.92 cm−1 while experimentally at 3383.72 cm−1. The experimental and theoretical values show a difference in wavenumber due to hydrogen bonding with the oxygen atom of carbonyl group in both H–N–C–O interactions. The N–H bending vibrations are observed theoretically at 1545.17 cm−1 and 1604.15 cm−1 and experimentally at 1574.54 cm−1. In compound 6i, the C[double bond, length as m-dash]N functional group experimentally shows stretching vibrations at 1630.14 cm−1, which is in the range from 1610 cm−1 to 1660 cm−1. C–N vibrations are stretched in-plane that are observed at 1132.11 cm−1 and 1229.10 cm−1 theoretically and experimentally observed at 1118.70 cm−1 and 1224.23 cm−1 (Fig. 26).


image file: d5ra07232h-f26.tif
Fig. 26 Theoretical vibrational analysis of compound 6i in the IR study.

3.5.5.2. Carbonyl group (C[double bond, length as m-dash]O) vibrations. The IR vibrational range for the carbonyl group (C[double bond, length as m-dash]O) was reported to be from 1630 cm−1 to 1850 cm−1,50 while the stretching vibrational range for the ketonic carbonyl (C[double bond, length as m-dash]O) group is from 1650 cm−1 to 1850 cm−1. In compound 6h, two modes of vibrations are at 1676.28 cm−1 and 1766.76 cm−1 with the highest potential energy distribution (PED) contribution of 72%. This ketonic carbonyl (C[double bond, length as m-dash]O) group, which is highly polarized (π-bond), shows the symmetric stretching vibrational mode at 1676.28 cm−1, as predicted by the IR data. The experimental value of this ketonic carbonyl group is shown at 1667.22 cm−1. In compound 6i, three modes of vibrations are shown at 1655.08 cm−1, 1667.57 cm−1 and 1711.73 cm−1 with the highest PED contribution. Theoretically, this ketonic carbonyl (C[double bond, length as m-dash]O), which is polar in nature, shows the stretching vibrations at 1655.08 cm−1, while the experimental value of the carbonyl functional group was observed at 1654.82 cm−1.
3.5.5.3. Aromatic ring C–H vibrations. The aromatic ring C–H vibrations are reported at 3000 cm−1 to 3100 cm−1, and the aliphatic C–H vibrations are reported at 2850 cm−1 to 2960 cm−1.50 In the IR study, theoretical and experimental calculations of C–H vibrations are observed right after the N–H vibrations. In compound 6h, the aromatic ring C–H vibrations range from 2885.84 cm−1 to 3068.29 cm−1 theoretically and C–H vibrations are observed at 2979.28 cm−1 for experimental calculations. In compound 6i, the C–H vibrational modes are theoretically observed at 2964.47 cm−1 and 3053.76 cm−1 but experimentally no C–H stretching vibration peaks were observed in 6i compound.
3.5.5.4. C–Cl vibrations. The stretching vibrational frequency for C–Cl ranges between 600 cm−1 and 800 cm−1 (Bisong et al., 2020). Chlorine is a heavy atom (in mass in comparison to hydrogen and carbon) and electron withdrawing that lowers the vibrational wavenumber due to this heavy atom of chlorine. In compound 6h, the stretching vibrations of C–Cl occur from 627.95 cm−1 to 792.11 cm−1. The C–Cl vibrational frequency mode was observed at 679.62 cm−1 theoretically and at 672.37 cm−1 experimentally. In compound 6i, the stretching vibrations of C–Cl are observed in the range from 622.31 cm−1 to 795.62 cm−1. The C–Cl functional group was observed at 661.28 cm−1 wavenumber theoretically and at 670.82 cm−1 wavenumber experimentally.
3.5.6. NMR analysis. Nuclear magnetic resonance (NMR) is a spectroscopic technique used to understand the organic compound structure. The computational study was performed using Gaussian 09W for theoretical information, which is later used for the validation of experimental results. The computational study was conducted using the GIAO method for proton (1H) and carbon (13C) NMR spectra of the synthesized compounds 6h and 6i at the B3LYP 6-311G basis set.51 The chemical shift values of 1H and 13C NMR for these compounds are presented in ppm along with the reference of TMS in Table 8.

Theoretical and experimental shift values of 13C NMR for these compounds are accordance to each other and their experimental values are observed by dissolving in DMSO solvent. For 6h compound, experimental chemical shift value range from C-6 to C-10 is 164.2 ppm–28.2 ppm. In 13C NMR analysis, 10-C in 6h compound show the highest peak in theoretical study at 158.85 ppm and in experimental show at 164.2 ppm due to the ketonic group presence. 4-C and 5-C in compound 6h show aromatic carbon shift values at 153.70 ppm and 124.83 ppm (Fig. S28 (SI)).

For the 1H NMR study, the theoretical values of compound 6h were compared with the experimental values. In the proton NMR, 20-H shows the anomalously high chemical shift value at 7.985 ppm, while the experimental value at 12.5 ppm confirms the intramolecular H-bonding because this is attached to the nitrogen atom. In experimental proton NMR analysis, protons 21-H, 22-H shows same peaks at 4.28 ppm, 14-H, 15-H shows same peaks at 2.87 ppm while 18-H, 19-H also show same peaks at 2.52 ppm. The experimental and theoretical analysis comparison for proton and carbon NMR of compound 6h in graphical representation is shown in Fig. S29 (SI).

For compound 6i, both proton and carbon NMR chemical shift values were analyzed by dissolving this compound in a DMSO solvent. In 13C NMR spectral data analysis, 6-C in 6i compound show the highly de-shielded effect with the highest peak at 192.9 ppm in experimental spectrum and in theoretical spectral data show chemical shift value at 197.95 ppm due to the carbonyl group adjacent to electron withdrawing oxygen atom (O-12). In compound 6i, 8-C adjacent to S-9 shows the lowest chemical shift value for theoretical 148.90 ppm and experimental 42.7 ppm due to the electron-donating effect from sulfur and the shielding effect. In compound 6i, 2-C and 5-C show the same aromatic carbon shift values at 172.88 ppm and 172.26 ppm (Fig. S30: SI).

In the 1H NMR analysis of 6i compound, 23-H to 30-H protons show the experimental chemical shift range from 12.4 to 4.87 ppm and the theoretical range from 7.796 to 4.947 ppm; 26-H, 27-H, 29-H and 30-H show the same peak value at 4.87 ppm; 24-H proton attaches on 8-C, which is near S-9, showing the deshielding effect due to potential hydrogen bonding, at chemical shifts of 7.80 ppm theoretically and 8.98 ppm experimentally; and 25-H and 28-H protons attach to the electronegative atom (N13, N18), showing intramolecular hydrogen bonds due to the strong deshielding effect and a high chemical shift value at 12.4 ppm. The comparison of theoretical and experiment analyses of 1H NMR and 13C NMR for compound 6i in graphical representation is shown in Fig. S31 (SI).

3.5.7. Hirshfeld surface population analysis. The charge distribution analysis of the compound 6h by Hirshfeld surface showed the details of its electronic distribution and charge of atoms. The sulfur atom (atom 1) has a moderately positive Hirshfeld charge (Q-H = 0.0526), indicating that there is a weak electron donation. The nitrogen atoms present at position 3 and 9 in compound 6h have quite negative Hirshfeld charges (Q-H = −0.223 and −0.100), which suggests that they are highly electronegative and partake of electron-rich sites (Table 9). Oxygen atom (atom 11) displays a great negative charge (Q-H = −0.280) consistent with its functionality as a major site of electron withdrawals. Carbon atoms are distributed with positive–negative Hirshfeld charge degrees, starting with slightly positive (e.g., atom 2 at Q-H = 0.101) through to about modestly negative (e.g., atoms 5 to 8 at Q-H = −0.045 to −0.052), due to the distinct binding surroundings in the molecule. These hydrogen atoms are uniformly and strongly positively charged and the maximum possible Hirshfeld value is found in atom 20 (Q-H = 0.160), probably because such an atom is attached to a highly electronegative or conjugated system. The above discussed trends are confirmed by CM5 charges (Q-CM5) as the values of electronegative atoms are highly negative (up to −0.430 at nitrogen), whereas the values of hydrogen atoms are large positive (e.g., +0.362 in atom 20). The molecule is therefore significantly polarized toward charges. The chlorine atom (atom 13) also has a slightly negative charge (Q-H = −0.106), and this takes part in electron-withdrawing properties in its immediate vicinity. In general, the Hirshfeld and CM5 studies of compound 6h promote a sense of the existence of easily recognizable regions of electron richness and electron deficiency, which is essential in terms of understanding its chemical reactivity and possible interaction (Fig. 27 and Table 9).
image file: d5ra07232h-f27.tif
Fig. 27 Graphical representation of Hirshfeld dipoles for compound 6h.
Table 9 Hirshfeld charges and dipoles on all atoms of compound 6h
S. no. Atoms Q-H S–H Dx Dy Dz Q-CM5
1 S 0.052581 0 −0.10383 0.10676 0.012915 0.032333
2 C 0.101472 0 0.072774 −0.01084 −0.00544 0.253843
3 N −0.22317 0 −0.03542 −0.23115 −0.0091 −0.39461
4 C 0.025584 0 −0.01655 0.140314 0.011738 0.085696
5 C −0.05204 0 0.027068 −0.10138 0.004714 −0.04235
6 C −0.05184 0 −0.02789 −0.01269 0.00077 −0.15253
7 C −0.04559 0 0.005154 −0.00031 0.008675 −0.15263
8 C −0.04892 0 −0.00988 0.011386 0.0008 −0.14731
9 N −0.10035 0 −0.01429 0.016501 0.004748 −0.42946
10 C 0.191464 0 −0.031 0.009398 −0.00082 0.279809
11 O −0.28036 0 −0.15658 −0.12058 0.004974 −0.31793
12 C −0.00226 0 0.045451 0.013045 0.000319 −0.09221
13 Cl −0.10608 0 −0.11253 0.020695 −0.00369 −0.11463
14 H 0.043254 0 −0.02216 −0.07298 0.1212 0.097898
15 H 0.045233 0 −0.02097 −0.10242 −0.10264 0.099879
16 H 0.040062 0 −0.0553 −0.00196 −0.13263 0.095082
17 H 0.041387 0 −0.11082 −0.01974 0.0903 0.095657
18 H 0.04457 0 −0.03512 0.055456 0.127172 0.100274
19 H 0.047426 0 −0.04472 0.102661 −0.09445 0.103257
20 H 0.160124 0 −0.01795 0.205089 −0.00488 0.362167
21 H 0.058132 0 −0.04787 −0.05699 0.128317 0.118357
22 H 0.059331 0 −0.04155 −0.06482 −0.12844 0.119429


The Hirshfeld analysis for the charge distribution of compound 6i offers the specific observation of the electronic structure: The computed Hirshfeld charges (Q-H) confirm that electronegative atoms, i.e., oxygen (e.g., atoms 12 and 15) have quite negative values (Q-H = 0.268–0.270), which means that they very actively attract electrons. The nitrogen atoms (e.g., atoms present at position-1, 11, 13 and 18) also have negative charges (Q-H 10 −0.10 to −0.20), in agreement with the electronegativity (Table 10). Contrastingly, the hydrogen atoms portray a positive charge because the values acquire high values of up to Q-H = 0.156 (atom 25), implying that they may be involved in polar bonding. The Hirshfeld charges of sulfur atoms (atoms 3 and 9) are moderately positive (∼0.078 to 0.101), and the carbon atoms exhibit different values, which are indicators of different bonding conditions at all points of the molecule. These observations are supported by the Q-CM5 charges, which give good charge estimate that takes into consideration the molecular dipoles where oxygen and nitrogen atoms have very negative values (up to −0.43) and hydrogen has highly positive values (up to 0.35). There is also a slight negative charge on the Cl atoms (atoms 17 and 22), which is indicative of certain level of localization of the electronic density. Altogether, the distribution of the charge in compound 6i indicates the presence of specific areas of electron density, which affects the chemical reactivity and could undergo biological processes or interactions with other molecules (Fig. 28 and Table 10).


image file: d5ra07232h-f28.tif
Fig. 28 Graphical representation of Hirshfeld dipoles for compound 6i.
Table 10 Hirshfeld charges and dipoles on all atoms of compound 6i
S. no. Atoms Q-H S–H Dx Dy Dz Q-CM5
1 N −0.1934 0 0.124148 −0.1293 −3.7 × 10−5 −0.35937
2 C 0.112533 0 −0.05472 −0.04975 0.000005 0.249657
3 S 0.078672 0 −0.01072 0.154275 −8 × 10−6 0.069008
4 C −0.05531 0 0.028256 −0.06034 −8 × 10−6 −0.08712
5 C 0.028273 0 −0.02864 0.067994 −1.6 × 10−5 0.092246
6 C 0.128231 0 0.008871 −0.01393 −9.4 × 10−5 0.162595
7 C 0.027198 0 −0.01117 −0.09745 −6.3 × 10−5 0.087222
8 C −0.03866 0 0.019808 0.078705 0.000055 −0.09341
9 S 0.10149 0 −0.09922 −0.07953 −6.4 × 10−5 0.108364
10 C 0.112308 0 0.067035 −0.00326 −1.6 × 10−5 0.268064
11 N −0.20562 0 −0.01548 0.170965 0.000133 −0.3839
12 O −0.26821 0 0.134528 −0.13756 0.000034 −0.29944
13 N −0.10363 0 0.002612 −0.00013 −3.7 × 10−5 −0.43416
14 C 0.204433 0 0.036435 −0.01995 0.000017 0.291084
15 O −0.2701 0 0.177188 0.081351 0.000013 −0.30814
16 C 0.006357 0 −0.03602 0.002269 −7 × 10−6 −0.08273
17 Cl −0.12025 0 0.120082 −0.05079 0.000017 −0.12857
18 N −0.10332 0 −0.00086 0.000943 0.000071 −0.43433
19 C 0.203106 0 −0.01197 0.036074 −3.4 × 10−5 0.290233
20 O −0.26493 0 −0.18201 0.057322 −3.9 × 10−5 −0.30305
21 C 0.002207 0 0.033044 −0.02387 0.000003 −0.08757
22 Cl −0.10675 0 −0.04483 0.09918 −3 × 10−6 −0.11516
23 H 0.063078 0 0.143825 −0.03779 −2.8 × 10−5 0.126208
24 H 0.084987 0 −0.09943 0.159465 0.000157 0.144925
25 H 0.155833 0 −0.03043 −0.2079 −2.3 × 10−5 0.355682
26 H 0.069127 0 0.069964 0.05358 0.131787 0.131809
27 H 0.069126 0 0.06999 0.053625 −0.13173 0.131819
28 H 0.156973 0 0.162255 0.133737 −0.00015 0.357041
29 H 0.063251 0 −0.08074 0.010005 0.131151 0.125613
30 H 0.063227 0 −0.08083 0.00993 −0.13106 0.125606


3.5.8. Non-covalent bond interactions (NCI) analysis. Fig. 29 presents the diagram detailing non-covalent interaction (NCI) and reduced density gradient (RDG) plot of the compound 6h using density functional theory (DFT). Fig. 29 (part A) depicts the molecular structure of 6h, as shown in the colored isosurfaces to indicate the presence of different non-covalent interactions. Green isosurfaces show weak van der Waals forces, blue surfaces show strong attractive forces including hydrogen bonding and red colors are abolished by steric repulsion. The RDG versus (λ2)ρ plot shown in Fig. 29 (part B) is utilized to determine the nature and the strength of these interactions. Attractive interactions (blue) are represented by the negative values of (λ2)ρ, which are close to zero values, indicated by green color (van der Waals interactions), and repulsive interactions, indicated by red color, by a positive value of (λ2)ρ. This analysis helps to have a detailed visualization of how non-covalent forces are adding to the stability and electron structure of the compound 6h.
image file: d5ra07232h-f29.tif
Fig. 29 (A) Non-covalent interaction (NCI) of compound 6h. (B) Graphical representation of the reduced density gradient (RDG) of compound 6h.

Fig. 30 shows the non-covalent interaction (NCI) and reduced density gradient (RDG) of compound 6i, which provides an understanding and visualizes weak intermolecular and intramolecular interactions using DFT-calculated electron density. In Fig. 30 (part A), the 3D isosurface of the NCI is displayed the molecular conformation that green surfaces demonstrate van der Waals weak interactions, blue surfaces denote an attractive hydrogen bond formation interaction and red surfaces exemplify steric repulsion. Fig. 30 (part B) presents the RDG vs. sign (λ2)ρ scatter plot, whose distribution of points tells something about the strength and nature of non-covalent interactions. In the graphical representation, blue peaks represent the presence of attractive forces, red peaks indicate the presence of repulsive forces and the green areas indicate the presence of weak dispersive forces. The work was analyzed by the NCI and RDG performed using the Multiwfn program and isosurfaces analyzed using VMD, providing a close insight into the non-covalent interaction surface of compound 6i.


image file: d5ra07232h-f30.tif
Fig. 30 (A) Non-covalent interaction (NCI) of compound 6i. (B) Graphical representation of the reduced density gradient (RDG) of compound 6i.
3.5.9. Electron localization function (ELF) and localized orbital locator (LOL) analysis. In Fig. 31 the analysis of the electron localization function (ELF) and localized orbital locator (LOL) of compound 6h are presented, which provide information about all electron pair localization and chemical bonding. Fig. 31 (part A) shows the ELF contour map with large values (dark areas around the carbon atoms), showing strong electron localization, which is usually attributed to bonding pairs or lone pairs. This visualization can be used to determine the areas of covalent bonding and electron density. The three-dimensional version of the LOL surface (top) and the associated 2D contour map (bottom), which increased the LOL values in both cases (darker colors), similarly indicate the localized regions of electrons, as shown in Fig. 31 (part B). The LOL analysis is an addition to the ELF since it highlights the presence of localized orbitals, especially within the bonding and non-bonding regions. Collectively, these roles use a detailed view to represent the electronic structure, which, in turn, is useful to make interpretations about the chemical bonding and reactivity of the molecular system and the distribution of lone pairs.
image file: d5ra07232h-f31.tif
Fig. 31 (A) Graphical representation of the electron localization function (ELF) for compound 6h. (B) Graphical representation of the localized orbital locator (LOL) for compound 6h.

Fig. 32 represents the electron localization function (ELF) and localized orbital locator (LOL) calculations of the compound 6i carried out using density functional theory (DFT) to investigate the electronic structure and bonding of the compound. Fig. 32 part (A) presents the ELF contour map, in which dense lines surrounding nitrogen, carbon and hydrogen atoms represent the regions of great electron localization including bonding and lone pairing regions. It is worthy to note that the high localization around the nitrogen atoms implies the existence of the lone-pair electrons, which adds to the electronic behavior of the compound 6i. A 2D contour map of the LOL is presented in Fig. 32 (part B), and a 3D surface plot of the LOL is also shown, giving an idea of how localized orbitals are spatially distributed. The colored areas, particularly red and yellow domains, signal the great localization that usually pertains to bonding between atoms in the conjugated core of compound 6i. The ELF and LOL represented in Fig. 32 explains the better bonding system and delocalization of electrons which further assists in learning more on reactivity and stability of this compound.


image file: d5ra07232h-f32.tif
Fig. 32 (A) Graphical representation of the electron localization function (ELF) for compound 6i. (B) Graphical representation of the localized orbital locator (LOL) for compound 6i.

4 Conclusion

Thiazole derivatives have been synthesized via two routes: the reaction of aminothiazole and chloroacetyl chloride leads to the formation of phenyl thiazole acetamide and the reaction of aminothiazoles and diazonium salt leads to the excellent yield of azothiazole. Two cell lines of chronic myeloid leukemia model, namely, K562 and U937 were used for biological potential evaluation against cancer. By using these cell lines, 2-amino-4-methylthiazole (3a), 2-amino-4-phenylthiazole (3b), bis(2-aminothiazole-4-yl)methanone (3c) and 2-chloro-N-(4-phenylthiazole-2-yl acetamide) (6j) compounds show poor results towards cytotoxicity. After the in vitro study, an in silico study was performed for further analyses of these thiazole derivatives. The docking studies were performed to check the pharmacologically active compounds with four proteins. Six compounds (3e, 3f, 6h, 6i, 6j and 7) were screened out from the 2GQG protein, 6 compounds (3b, 3e, 3d, 6g, 6h and 6i) from the 5MO4 protein, 4 compounds (3b, 3f, 6h and 7) from the 2AZ5 protein and 5 compounds (3c, 3f, 6h, 6i and 6j) from the 5MAR protein. The MMGBSA study showed excellent results with the 5MO4 protein. Further screening of two compounds (6h and 6i) was carried out using density functional theory, as these two compounds showed a good binding affinity score with the receptor site of all proteins. The DFT analysis provided us the detail about structural parameters and molecular geometry. After that a research created between experimental and computational studies. Overall, the lead synthesized compounds 6h and 6i proved to be effective drugs for future use due to the enhanced activity and cost-effective method.

Author contributions

Zaheer Ahmad: supervision, editing, evaluation of results; Labaina Shakorr: synthesis, biological activity, drafting; Aneeqa Batool: library preparation, molecular docking; Nusrat Shafiq: conceptualization, paper drafting, write up, data analysis, literature and funding; Syeda Aaliya Shehzadi: two-way ANOVA application on activity data; Mohamed Mohany: helped reviewing and data analysis; Lei Zhang & Salim S. Al-Rejaie: validation, proof editing; Humaira Razzaq & Sajid Mahmood: resolution and alignment of all figures.

Conflicts of interest

The authors have no conflict of interest.

Data availability

All data files are available as supplementary information (SI). Supplementary information: Tables S1, S2, S3, S4 and S5. Fig S1−S31. All 1H-NMR and 13C-NMR spectra. See DOI: https://doi.org/10.1039/d5ra07232h.

Acknowledgements

The authors acknowledge and appreciate the Ongoing Research Funding Program (ORF-2025-120), King Saud University, Riyadh, Saudi Arabia. The authors thank the Pakistan Science Foundation (Grant # CRP/PSF/TH-22) for supporting this study.

References

  1. E. Abele, R. Abele and E. Lukevics, Oximes of five-membered heterocyclic compounds with two heteroatoms 2: Reactions and biological activity, Chem. Heterocycl. Compd., 2007, 43, 945–977 CrossRef CAS.
  2. N. Siddiqui, M. F. Arshad, W. Ahsan and M. S. Alam, Thiazoles: A Valuable Insight into the Recent Advances and Biological Activities, Int. J. Pharmaceut. Sci. Drug Res., 2009, 1, 136–143 CrossRef CAS.
  3. N. Siddiqui, S. K. Arya, W. Ahsan and B. Azad, Diverse biological activities of Thiazoles: A Retrospect, Int. J. Drug Dev. Res., 2011, 3, 55–67 CAS.
  4. S. J. Kashyap, V. K. Garg, P. K. Sharma, N. Kumar, R. Dudhe and J. K. Gupta, Thiazoles: having diverse biological activities Review Article, Med. Chem. Res., 2012, 21, 2123–2132 CrossRef CAS.
  5. D. Goulson, REVIEW: An overview of the environmental risks posed by neonicotinoid insecticides, J. Appl. Ecol., 2013, 50, 977–987 CrossRef.
  6. L. M. Fox and L. D. Saravolatz, Nitazoxanide: a new thiazolide antiparasitic agent, Clin. Infect. Dis., 2005, 40, 1173–1180 CrossRef CAS PubMed.
  7. J. B. Sperry and D. L. Wright, Furans, thiophenes and related heterocycles in drug discovery, Curr. Opin. Drug Discovery Dev., 2005, 8, 723–740 CAS.
  8. R. E. Dolle and K. H. Nelson, Comprehensive Survey of Combinatorial Library Synthesis: 1998, J. Comb. Chem., 1999, 1, 235–282 CrossRef CAS PubMed.
  9. C. Zhang, G. Li, C. Fan, J. Xu, J. Cao and N. L. Shen Liu, Comparison of efficacy of different route of administration of chemotherapy on unresectable, advanced gastric cancer, World J. Surg. Oncol., 2012, 10, 1–14 CrossRef PubMed.
  10. S. Seth, A Comprehensive Review on Recent advances in Synthesis & Pharmacotherapeutic potential of Benzothiazoles, Inflamm. Anti-Allergy Agents Med. Chem., 2015, 14, 98–112 CrossRef CAS PubMed.
  11. F. Al-Omran, R. M. Mohareb and A. A. El-Khair, New route for synthesis, spectroscopy, and X-ray studies of 2-[aryl-(6′-hydroxy-4′,4′-dimethyl-2′-oxocyclohex-6′-enyl)methyl]-3-hydroxy-5,5-dimethylcyclohex-2-enone and 1,8-dioxo-octahydroxanthenes and antitumor evaluation, Med. Chem. Res., 2014, 23, 1623–1633 CrossRef CAS.
  12. U. Arshad, S. Ahmed, N. Shafiq, Z. Ahmad, A. Hassan, N. Akhtar, S. Parveen and T. Mehmood, Structure-Based Designing, Solvent Less Synthesis of 1,2,3,4-Tetrahydropyrimidine-5-carboxylate Derivatives: A Combined In Vitro and In Silico Screening Approach, Molecules, 2021, 26, 1–22 Search PubMed.
  13. P. Schneider and G. Schneider, De novo design at the edge of chaos: Miniperspective, J. Med. Chem., 2016, 59, 4077–4086 CrossRef CAS PubMed.
  14. S. A. A. Anand, C. Loganathan, K. Saravanan and S. Kabilan, Comparison of molecular docking and molecular dynamics simulations of 1, 3-thiazin-4-one with MDM2 protein, Int. Lett. Chem., Phys. Astron., 2015, 60, 161–167 Search PubMed.
  15. S. Roy, R. Sharma and A. Kumar, 17 Molecular Docking and Molecular Dynamics in Signal Analysis, Signal Analysis in Pharmacovigilance: Principles and Processes, 2024, p. 210 Search PubMed.
  16. S. Chinnasamy, G. Selvaraj, A. C. Kaushik, S. Kaliamurthi, A. S. Nangraj, C. Selvaraj, S. K. Singh, R. Thirugnanasambandam, K. Gu and D.-q. Wei, Identification of potent inhibitors against Aurora kinase A using molecular docking and molecular dynamics simulation studies, Preprints.org, preprint, 2019,  DOI:10.20944/preprints201908.0238.v1.
  17. O. O. Elekofehinti, I. R. Adetoyi, H. O. Popoola, F. O. Ayodeji, F. A. Taiwo, M. O. Akinjiyan, O. F. Koledoye, O. Iwaloye and A. E. Adegboyega, Discovery of potential epidermal growth factor receptor inhibitors from black pepper for the treatment of lung cancer: an in-silico approach, Silico Pharmacol., 2024, 12, 28 CrossRef PubMed.
  18. A. O. Fadaka, R. T. Aruleba, N. R. S. Sibuyi, A. Klein, A. M. Madiehe and M. Meyer, Inhibitory potential of repurposed drugs against the SARS-CoV-2 main protease: a computational-aided approach, J. Biomol. Struct. Dyn., 2022, 40, 3416–3427 CrossRef CAS PubMed.
  19. V. T. Sabe, G. F. Tolufashe, C. U. Ibeji, S. B. Maseko, T. Govender, G. E. Maguire, G. Lamichhane, B. Honarparvar and H. G. Kruger, Identification of potent L, D-transpeptidase 5 inhibitors for Mycobacterium tuberculosis as potential anti-TB leads: Virtual screening and molecular dynamics simulations, J. Mol. Model., 2019, 25, 1–16 CrossRef PubMed.
  20. S. V. Pattar, S. A. Adhoni, C. M. Kamanavalli and S. S. Kumbar, In silico molecular docking studies and MM/GBSA analysis of coumarin-carbonodithioate hybrid derivatives divulge the anticancer potential against breast cancer, Beni-Suef Univ. J. Basic Appl. Sci, 2020, 9, 1–10 CrossRef.
  21. N. Muddagoni, R. Bathula, M. Dasari and S. R. Potlapally, Homology modeling, virtual screening, prime-MMGBSA, AutoDock-identification of inhibitors of FGR protein, Biointerface Res. Appl. Chem., 2021, 11, 11088–11103 CAS.
  22. A. A. Braga, G. Ujaque and F. Maseras, A DFT study of the full catalytic cycle of the Suzuki− Miyaura cross-coupling on a model system, Organometallics, 2006, 25, 3647–3658 CrossRef CAS.
  23. A. Tomberg, An introduction to computational chemistry using G09W and Avogadro software, Gaussian 09w Tutorial, 2013, pp. 1–36 Search PubMed.
  24. M. Zunita, M. David and W. Natola, Prediction of CO2 with amine functionalized ionic liquids interaction using density functional theory, Case Stud. Chem. Environ. Eng., 2023, 8, 100355 CrossRef CAS.
  25. S. M. Putra, T. K. Wungu and I. Arif, Ab-Initio Calculation of Chlorophyll-b UV-Vis Absorbance Spectra using Gaussian 09 based Density Functional Theory (DFT), Int. J. Nanoelectron. Mater., 2021, 14, 11–26 Search PubMed.
  26. G. Kotan and H. Yüksek, Experimental (Ft-Ir, Nmr) And Theoretical (B3pw91, B3lyp, Hf) Analyses Of 2-((3-Ethyl-4, 5-Dihydro-1h-1, 2, 4-Triazol-5-On-4-Yl)-Azomethine)-Benzoic Acid, Caucasian J. Sci., 2019, 6, 64–75 Search PubMed.
  27. M. Sheikhi, E. Balali and H. Lari, Theoretical investigations on molecular structure, NBO, HOMO-LUMO and MEP analysis of two crystal structures of N-(2-benzoyl-phenyl) oxalyl: A DFT study, J. Phys. Theor. Chem., 2016, 13, 155–169 Search PubMed.
  28. S. C. North, K. R. Jorgensen, J. Pricetolstoy and A. K. Wilson, Population analysis and the effects of Gaussian basis set quality and quantum mechanical approach: main group through heavy element species, Front. Chem., 2023, 11, 1152500 CrossRef CAS PubMed.
  29. H. Benaissi, M. Drissi, S. Yahiaoui, Y. Megrouss, A. Chouaih and F. Hamzaoui, Hirshfeld surface analysis, topological features and nonlinear optical properties of phthalonitrile derivative: low temperature experimental charge density and quantum chemistry studies, J. Optoelectron. Biomed. Mater., 2018, 10, 73–82 Search PubMed.
  30. G. Amudha, R. Santhakumari, D. Chandrika, S. Mugeshini, N. Rajeswari and S. Sagadevan, Growth, spectroscopic and Hirshfeld surface analysis on pyridine urea single crystal, J. Mol. Struct., 2022, 1257, 132606 CrossRef CAS.
  31. N. E. Amraoui and D. Hammoutè, DNA base pairs sensors: DFT, QTAIM and NCI-RDG study, Comput. Chem., 2024, 12, 75–90 CrossRef CAS.
  32. D. A. Dhas and S. Balachandran, Structural, Spectroscopic, and CH⋯O Hydrogen Bonding Interaction on Structure (Monomer and Dimer) Vibrational Spectroscopic, Fukui, NCI, AIM, and RDG Analysis Molecular Docking and Molecular Dynamic Simulation of Biological Active Pencycuron, Polycyclic Aromat. Compd., 2023, 43, 8494–8543 CrossRef.
  33. S. Janani, H. Rajagopal, S. Muthu, S. Javed and A. Irfan, Structural, electronic properties (different solvents), chemical reactivity, ELF, LOL, spectroscopic insights, molecular docking and in vitro anticancer activity studies on methyl (4-nitro-1-imidazolyl) acetate, J. Indian Chem. Soc., 2022, 99, 100438 CrossRef CAS.
  34. F. Akman, A. Demirpolat, A. S. Kazachenko, A. S. Kazachenko, N. Issaoui and O. Al-Dossary, Molecular structure, electronic properties, reactivity (ELF, LOL, and Fukui), and NCI-RDG studies of the binary mixture of water and essential oil of Phlomis bruguieri, Molecules, 2023, 28, 2684 CrossRef CAS PubMed.
  35. D. P. Khrustalev, A. A. Suleimenova and S. D. Fazylov, Synthesis of 2-amino-4-phenylthiazole under Conditions of microwave Irradiation, Russ. J. Appl. Chem., 2008, 81, 863–900 CrossRef.
  36. J. Sun, B. R. Liu, W. J. Hu, L. X. Yu and X. P. Qian, In vitro anticancer activity of aqueous extracts and ethanol extracts of fifteen taditional Chinese medicines on human digestive tumor cell lines, Phytother Res., 2007, 21, 1102–1104 CrossRef CAS PubMed.
  37. Y. Ohsaki, A. F. Gazdar, H.-C. Chen and B. E. Johnson, Antitumor activity of magainin analogues against human lung cancer cell lines, Cancer Res., 1992, 52, 3534–3538 CAS.
  38. Y. K. Janapati and S. Junapudi, Progress in experimental models to investigate the in vivo and in vitro antidiabetic activity of drugs, Anim. Models Exp. Med., 2024, 7, 297–309 CrossRef CAS PubMed.
  39. J. J. Sahayarayan, K. S. Rajan, M. Nachiappan, D. Prabhu, R. G. R. Rao, J. Jeyakanthan, A. H. Mahmoud, O. B. Mohammed and A. M. Morgan, Identification of potential drug target in malarial disease using molecular docking analysis, Saudi J. Biol. Sci., 2020, 27, 3327–3333 CrossRef PubMed.
  40. B. K. Kumar, n. Faheem, K. V. G. C. Sekhar, R. Ojha, V. K. Prajapati, A. Pai and S. Murugesan, Pharmacophore based virtual screening, molecular docking, molecular dynamics and MM-GBSA approach for identification of prospective SARS-CoV-2 inhibitor from natural product databases, J. Biomol. Struct. Dyn., 2022, 40, 1363–1386 CrossRef CAS PubMed.
  41. P. T. Tasli, Ç. K. Atay, T. Demirturk and T. Tilki, Experimental and computational studies of newly synthesized azo dyes based materials, J. Mol. Struct., 2020, 1201, 127098 CrossRef CAS.
  42. G. Khanum, A. Ali, S. Shabbir, A. Fatima, N. Alsaiari, Y. Fatima, M. Ahmad, N. Siddiqui, S. Javed and M. Gupta, Vibrational Spectroscopy, Quantum Computational and Molecular Docking Studies on 2-[(1H-benzimidazol-1-yl)-methyl] benzoic acid, Crystals, 2022, 12, 337 CrossRef CAS.
  43. S. Rekha, S. Tamilselvan, V. Vetrivelan, J. C. Mishma, S. Kadaikunnan, G. Abbas and S. Muthu, Effect of different solvents, molecular level vibrational energies, electronic, electrostatic, donar-acceptor and pharmaceutical studies on 3-methoxy phenyl acetonitrile-anti depressant agent, J. Mol. Liquids, 2023, 386, 122308 CrossRef CAS.
  44. K. Chaitanya, C. Santhamm, K. Prasad and V. Veeraiah, Molecular structure, vibrational spectroscopic (FT-IR, FT-Raman), first order hyperpolarizability, NBO analysis, HOMO and LUMO analysis, thermodynamic properties of 3, 5-dimethylbenzophenone by ab inito HF and density functional method, J. At. Mol. Sci., 2012, 3(1), 1–22 Search PubMed.
  45. Z. Demircioğlu, C. A. Kaştaş and O. Büyükgüngör, Theoretical analysis (NBO, NPA, Mulliken Population Method) and molecular orbital studies (hardness, chemical potential, electrophilicity and Fukui function analysis) of (E)-2-((4-hydroxy-2-methylphenylimino) methyl)-3-methoxyphenol, J. Mol. Struct., 2015, 1091, 183–195 CrossRef.
  46. T. B. Issa, F. Sayari, H. Ghalla and L. Benhamada, Synthesis, crystal structure, DFT calculations and molecular docking of L-pyroglutamic acid, J. Mol. Struct., 2019, 1178, 436–449 CrossRef CAS.
  47. K. Ramirez-Balderrama, E. Orrantia-Borunda and N. Flores-Holguin, Calculation of global and local reactivity descriptors of carbodiimides, a DFT study, J. Theor. Comput. Chem., 2017, 16(3), 1750019 CrossRef.
  48. S. Parveen, A. Batool, N. Shafiq, M. Rashid, A. Sultan and G. F. Wondmie, et al., Developmental landscape of computational techniques to explore the potential phytochemicals from Punica granatum peels for their antioxidant activity in Alzheimer’s disease, Front. Mol. Biosci., 2023, 10, 1252178 CrossRef CAS PubMed.
  49. B. Chandralekha, R. Hemamalini, S. Muthu and S. Sevvanthi, Spectroscopic (FT-IR, FT-RAMAN, NMR, UV–Vis) investigations, computational analysis and molecular docking study of 5-bromo-2-hydroxy pyrimidine, J. Mol. Struct., 2020, 1218, 128494 CrossRef CAS.
  50. E. A. Bisong, H. Louis, T. O. Unimuke, J. O. Odey, E. I. Ubana and M. M. Edim, et al. Vibrational, electronic, spectroscopic properties, and NBO analysis of p-xylene, 3, 6-difluoro-p-xylene, 3, 6-dichloro-p-xylene and 3, 6-dibromo-pxylene: DFT study, Heliyon, 2020, 6(12), e05783 CrossRef CAS PubMed.
  51. G. Bella and A. Rotondo, Theoretical prediction of 13C NMR spectrum of mixed triglycerides by mean of GIAO calculations to improve vegetable oils analysis, Chem. Phys. Lipids, 2020, 232, 104973 CrossRef CAS PubMed.

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.