Facile synthesis of some 5-(3-substituted-thiophene)-pyrimidine derivatives and their pharmacological and computational studies Chimica Techno Acta ARTICLE published by Ural Federal University 2021, vol. 8(4), № 20218401 eISSN 2411-1414; chimicatechnoacta.ru DOI: 10.15826/chimtech.2021.8.4.01 1 of 13 Facile synthesis of some 5-(3-substituted-thiophene)- pyrimidine derivatives and their pharmacological and computational studies S.H. Sukanya a, Talavara Venkatesh a* , S.J. Aditya Rao b, N. Shivakumara c, Muthipeedika Nibin Joy d a: Department of P.G. Studies and Research in Chemistry, Jnanasahyadri, Kuvempu University, Shankaraghatta 577451, Karnataka, India b: Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India c: Department of Chemistry, Ramaiah Institute of Technology, Bangalore 560054, India d: Innovation Center for Chemical and Pharmaceutical Technologies, Institute of Chemical Technology, Ural Federal University, 19 Mira St., Yekaterinburg 620002, Russia * Corresponding author: venkateshatalwar@gmail.com This article belongs to the regular issue. © 2021, The Authors. This article is published in open access form under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/). Abstract A series of 5-(3-substituted-thiophene)-pyrimidine derivatives (3a-d) were synthesized via Knoevenagel condensation reaction in aqueous ethanol using H2O2:HCl as a catalyst. Their pharmacological effects were evaluated. Analytical and spectroscopic methods confirmed the structures of the target molecules. The antibacterial activity studies revealed that compounds 3b and 3d exhibited the most effective zone of inhibition against bacterial strains E. coli and S. aureus, respectively. The in vitro cytotoxicity was carried out by MTT assay against MCF-7 cell line. The results showed the excellent selectivity for all four compounds, among which the compound 3a exhibited re- markable cytotoxicity with a minimum cell viability range of 23.68 to 44.16%. The interaction of compounds with calf thymus DNA was de- termined using UV-absorption spectroscopy. The results confirmed that all the synthesized compounds interacted strongly with CT DNA through electrostatic or groove binding. In silico ADME-toxicology studies indicated that all the molecules under investigation are non- toxic with good oral bioavailability. The drug-likeness score indicat- ed that they are suitable as drug-leads. In silico molecular docking the specified compound 3b bounds with GlcN-6-P and P38 MAPk with a minimum binding energy of –7.9 and –6.4 kcal/mol, respectively. DFT study demonstrated that the compound 3d was chemically and biologically more reactive due to less energy gap. Keywords biological studies DNA binding ADME-toxicology study SAR study molecular docking and DFT studies Received: 02.09.2021 Revised: 11.10.2021 Accepted: 12.10.2021 Available online: 15.10.2021 1. Introduction Heterocyclic compounds play a predominant role in medic- inal chemistry and synthetic organic chemistry due to their massive biological importance. The synthesis of ni- trogen and sulphur containing fused heterocyclic com- pounds with multi-structures in one molecule has attract- ed the attention of medicinal chemists and researchers due to their multifaceted pharmacological activities [1–2]. Among them, pyrimidine and thiophene have been recog- nized as key scaffolds owing to their important biological significances and interesting therapeutic properties in- cluding anti-tubercular [3], anticancer [4], anti-HIV [5], antibacterial [6], antifungal [7], antitumor [8], also used as potent EGFR inhibitor [9, 10], protein kinase inhibitors [11–14] and 5-HT7 receptors [15]. Moreover, the heterocyclic compounds increase the strength of the molecules by forming hydrogen bonds with DNA. Hence, the interactive study of heterocyclic moieties with DNA is essential for estimating their anticancer activ- ity and elucidates the viable mechanism of their action. Therefore, DNA binding is considered as an essential ex- http://chimicatechnoacta.ru/ https://doi.org/10.15826/chimtech.2021.8.4.01 http://creativecommons.org/licenses/by/4.0/ https://orcid.org/0000-0001-9637-3909 Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 2 of 13 perimental step to measure anticancer drugs activity be- cause most anticancer drugs specifically target the DNA [16]. Heterocyclic compounds appear to be most effective against various cancers. Around 60% of the medicines used for cancer treatment are based on heterocyclic moie- ties. Among the various heterocyclic moieties, nitrogen and sulphur-based compounds are effective against differ- ent types of cancers [17–19]. Cancer is a group of diseases in which the abnormal cells grow uncontrollably by disregarding the normal rules of cell division. Cancer is caused by many external (tobacco, chemi- cals, radiation and infectious organisms) as well as some in- ternal factors (inherited mutations, hormones, immune condi- tions and random mutations). There are different types of can- cer such as breast cancer, bladder cancer, kidney cancer, leu- kaemia, liver cancer, lung cancer, melanoma, pancreatic can- cer, prostate cancer, thyroid cancer etc. [20]. The breast can- cer is one of the most frequently diagnosed cancers and the leading cause of death in females worldwide, with more than 1.5 million new cases recorded every year. It is also the fifth- highest cause of death in the world [21]. The modern methods of chemotherapy suffer from the main disadvantages: side effects and drug resistance. Therefore, continued search for novel and safer anticancer drugs remains essential [22]. Pollution is one of the critical problems that the syn- thetic organic chemist faces in designing the organic reac- tions for the synthesis of pharmacologically active com- pounds. Thus, development of an environmentally-friendly chemical process that induces necessary organic transfor- mation is one of the most important goal of sustainable progression [23]. Therefore, "green chemistry" becomes a promising approach that meets the requirements of the chemical and pharmaceutical industries. The replacement of hazardous solvents with eco-friendly solvents is an ac- ceptable and valuable approach in a chemical reaction. Hence, the use of a hypochlorous acid (HOCl) as a green halogenating agent [24] is highly advantageous over other catalysts, including cost-effectiveness, efficiency and read- ily availability. Earlier, our research group has reported the synthesis of different pyrimidine derivatives and other biologically important heterocyclic compounds [25–27]. Some of the drugs containing pyrimidine nucleus available in the market are depicted below (Fig. 1). Based on the above findings, we herein report the ap- plication of H2O2:HCl as a green halogenating catalyst for the synthesis of 5-(3-substituted-thiophene)-pyrimidine derivatives of considerable pharmacological relevance. 2. Experimental 2.1. Materials and Method All chemicals and calf thymus DNA were purchased from Al- drich Chemical Company, and the reaction was performed at refluxed condition and solvents were used without further purification. Analytical TLC was performed with E. Merck silica gel GF254 glass plates. Visualization of the developed chromatogram was performed by UV light (254 and 356 nm). The melting points of the products were determined in open capillary tubes and uncorrected. The ATR-IR spectra were obtained using Bruker FTIR Alpha spectrometer. The 1H NMR and 13C NMR spectra were recorded on Bruker 400 MHz and 100 MHz in DMSO-d6 as a solvent. Mass spectra were ob- tained by Agilent 1200 series LC and Micromass Q spectrome- ter. The DNA binding studies were carried on Elico SL 159 UV-Visible spectrophotometer in 200–500 nm range equipped with 1.0 cm quartz cell at room temperature. The anticancer activity was carried out in the Department of Mi- crobiology, Maratha Mandal's NGH Institute of Dental Sci- ences and Research Centre, Belgaum, Karnataka. The compu- tational studies were carried out by Density functional theory (DFT)/B3LYP method using Gaussian 09 software using 6- 31G (d, p) basis set at gaseous phase. 2.2. General procedure for the synthesis of 5-(3- substituted-thiophene)-pyrimidine derivatives (3a-d) The synthesis of 5-(3-substituted-thiophene)-pyrimidine derivatives (3a-d) were carried out by the reaction of bar- bituric/thiobarbituric acid (1, 1mmol) with 3-substituted- thiophene-2-carboxaldehyde (2, 1mmol) in the presence of 15 mL aqueous ethanol using 6% H2O2:HCl (2:1) as a cata- lyst. The reaction mixture was refluxed at 80 °C for about 10–15 min and the progress of the reaction was monitored by TLC (Ethyl acetate and Petroleum ether). After the completion of the reaction, the reaction mixture was cooled to room temperature and poured into the 100 mL flake ice with vigorous stirring to get solid precipitated out. The crude mixture was filtered, washed and recrystal- lized with absolute ethanol to afford pure solid products. 2.2.1. 5-(Thiophen-2-ylmethylidene)pyrimidine- 2,4,6(1H,3H,5H)-trione (3a) Yield: 96%, Yellow solid; MP: 280–282 °C. IR (ATR, υ cm–1): 3373 (NH), 1654 (C=O), 1529 (C=C). 1H NMR (400 MHz, DMSO-d6, δ ppm): 7.31–7.33 (t, J= 8 Hz, 1H, Ar-H), 8.13–8.24 (m, 2H, Ar-H), 8.53 (s, 1H, CH), 11.17 (s, 1H, NH), 11.21 (s, 1H, NH). 13C NMR (100 MHz, DMSO-d6, δ ppm): 112.035, 128.808, 136.776, 142.526, 146.116, 146.291, 150.679, 163.453 and 163.950 (C=O). HRMS: m/z 221.9054 [M+H]+. Anal. Calcd for C9H6N2O3S: C 48.64, H 2.72 and N 12.61%. Found: C 48.59, H 2.68 and N 12.53%. 2.2.2. 5-[(3-Methylthiophen-2-yl)methylidene]pyrimidine- 2,4,6(1H,3H,5H)-trione (3b) Yield: 94%, Yellow solid; MP: 302–306 °C. IR (ATR, υ cm–1): 3225 (NH), 2858 (CH3), 1703 (C=O), 1542 (C=C). 1H NMR (400 MHz, DMSO-d6, δ ppm): 2.24 (s, 3H, CH3), 7.21–7.23 (d, J= 8 Hz, 1H, Ar-H), 8.16–8.18 (d, J= 8 Hz, 1H, Ar-H), 8.53 (s, 1H, CH), 11.17 (s, 1H, NH), 11.24 (s, 1H, NH). 13C NMR (100 MHz, DMSO-d6, δ ppm): 19.922 (CH3), 114.870, 135.252, 136.011, 145.166, 147.180, 154.915, 158.106, 167.728 and 168.497 (C=O). HRMS: m/z 235.9774 [M+H]+. Anal. Calcd for C10H8N2O3S: C 50.84, H 3.41 and N 11.86%. Found: C 50.79, H 3.36 and N 11.80%. Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 3 of 13 HN H NO O F 5-Fluorouracil (Anticancer) N H NO O F O HO HO Floxuridine (Anticancer) N H NH N O O Cl Cl Uramustine (Anticancer) HN N O O O F Tegafur (anticancer) HN N H SH O O 5-thiouracil (Anticancer) N N NH2 O O O H2N Trimethoprim (Antibacterial) N N S N N O N N HN S O O Pictilisib (Anticancer) N N N O F N H O S Lorediplon (Insomnia) N N N O S N O Indiplon (Sedative & hypnotics) Fig. 1 Some of the drugs containing pyrimidine nucleus available in the market 2.2.3. 5-(Thiophen-2-ylmethylidene)-2- thioxodihydropyrimidine-4,6(1H,5H)-dione (3c) Yield: 93%, Green solid; MP: 320–322 °C. IR (ATR, υ cm–1): 3106 (NH), 1760 (C=O), 1559 (C=C), 1156 (C=S). 1H NMR (400 MHz, DMSO-d6, δ ppm): 6.86–6.88 (d, J = 8 Hz, 1H, Ar-H), 8.20 (s, 1H, CH), 8.34–8.36 (d, J = 8 Hz, 2H, Ar-H), 12.20 (s, 1H, NH), 12.30 (s, 1H, NH). 13C NMR (100 MHz, DMSO-d6, δ ppm): 114.713, 116.139, 124.434, 139.254, 157.012, 160.480, 162.825, 164.168 (C=O) and 178.617 (C=S). HRMS: m/z 237.9467 [M+H]+. Anal. Calcd for C9H6N2O2S2: C 45.36, H 2.54 and N 11.76%. Found: C 45.30, H 2.49 and N 11.71%. 2.2.4. 5-[(3-Methylthiophen-2-yl)methylidene]-2- thioxodihydropyrimidine-4,6(1H,5H)-dione (3d) Yield: 95%, Green solid; MP: 310–312 °C. IR (ATR, υ cm–1): 3108 (NH), 2852 (CH3), 1704 (C=O), 1516 (C=C), 1200 (C=S). 1H NMR (400 MHz, DMSO-d6, δ ppm): 2.24 (s, 3H, CH3), 7.22–7.24 (d, J= 8 Hz, 1H, Ar-H), 8.17–8.19 (d, J = 8 Hz, 1H, Ar-H), 8.54 (s, 1H, CH), 11.18 (s, 1H, NH), 11.25 (s, 1H, NH). 13C NMR (100 MHz, DMSO-d6, δ ppm): 26.806 (CH3), 114.280, 128.031, 128.903, 129.867, 130.391, 130.892, 142.472, 163.552 (C=O) and 173.358 (C=S). HRMS: m/z 251.9608 [M+H]+. Anal. Calcd for C10H8N2O2S2: C 47.60, H 3.20 and N 11.10%. Found: C 47.55, H 3.16 and N 11.06%. 2.3. Pharmacological studies 2.3.1. Antibacterial activity Antibacterial activity of the synthesized compounds (3a-d) was carried out by the agar well diffusion method [28] using two bacterial strains (Gram-negative and Gram- positive) Escherichia coli (MTCC-1559) and Staphylococ- cus aureus (MTCC-902). DMSO was used as negative con- trol and Ciprofloxacin as standard drug. The test com- pounds were dissolved in DMSO at two different concen- trations, 20 and 40 µg/mL. 2.3.2. Cytotoxicity In vitro cytotoxicity was assessed by the MTT assay meth- od [29] against MCF-7 (Breast cancer) cell line. The cells were seeded in a 96-well flat-bottom microplate and maintained at 37 °C in 95% humidity and 5% CO2 over- night. Different concentration (200, 100, 50, 25, 12.5 and 6.25 µg/mL) of samples were treated. The cells were incu- bated for another 48 h, and the wells were washed twice with PBS. 20 µL of the MTT staining solution was added to each well, and the plate was incubated at 37 °C for 4 h. The medium with MTT was discarded, and 100 µL of Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 4 of 13 DMSO was added to each well to dissolve the formazan crystals. The absorbance was recorded at 570 nm using a microplate reader. The percentage of cell survival was calculated by using the following formula: % of cell survival = Mean O. D. of test compound Mean O. D. of Negative control  100. (1) 2.3.3. DNA binding study DNA binding study was assessed by using electronic spec- troscopy. A solution of CT DNA in 50 mM Tris-HCl/50 mM NaCl buffer solution was prepared at pH 6.9–7.01. In buff- er solution, the ratio of absorption values of CT DNA at 260 and 280 nm is 1.8–1.9, indicating that DNA was free of proteins [30]. Then a concentrated stock solution of DNA was prepared in 50 mM Tris-HCl, 50 mM NaCl in double distilled water at pH 6.9–7.01. The concentration of CT DNA was determined per nucleotide by taking the ab- sorption coefficient (6600 dm3 mol–1 cm–1) at 260 nm [31]. Stock solutions were stored at 4 °C. 2 mL of the solution was taken containing a fixed concentration of the com- pounds (3a-d) with CT DNA (0 to 350 µL of a 0.5025–6.067010–7 M stock CT DNA solution). A blank solution containing the same concentration of DNA was used as a reference. Solutions were prepared by mixing the compound and CT DNA in DMSO medium and then recording the UV absorption spectra by adding 25 to 350 µL DNA to the compound. The spectra were recorded against a blank solution containing the same concentration of DNA (4.090910–6 mol L–1). The intrinsic binding con- stant Kb was obtained by using the following equation [32]: [DNA] (∈A−∈B) = [DNA] (∈B−∈F ) + 1 Kb(∈B−∈F) , (2) where, ∈𝐴 ,∈𝐵 and ∈𝐹 corresponds to the apparent, bound and free compound extinction coefficients, respectively. A plot of [DNA] (∈A−∈F ) versus [DNA] gave a slope of 1 (∈B−∈F) and Y-intercept equal to 1 Kb(∈B−∈F) . Hence Kb is the ratio of slope to intercept. The % of hyperchromicity or hypochromicity (% H) for the CT DNA/[Ligand] was obtained from (∈A – ∈F)/ ∈F 100. 2.3.4. In silico oral bioavailability assessment and ADME- toxicology studies The oral bioavailability of the synthetic molecules (3a-d) can be predicted by considering their structural properties to screen based on the Rule of five or Lipinski rule-of-five (RO5) filter [33]. Rule of five employs the molecular prop- erties necessary to filter candidate drug's pharmacokinet- ics (PK) and pharmacodynamics (PD) [34–36]. Oral bioavailability assessment was done using Osiris Data warrior V.4.4.3 [37] based on total molecular weight, ClogP, H-acceptors, H-donors, rotatable bonds as part of RO5 filters, along with TPSA (Topological polar surface ar- ea) and drug-likeness assessment [38]. Pharmacodynamic properties like mutagenicity, tumorigenicity, reproductive effects, irritancy, Ames toxicity and hepatotoxicity were predicted using the admetSAR server. Bioactivity scores were predicted using the molinspiration server for GPCR ligand, ion channel modulator, kinase inhibitor, nuclear receptor inhibitor, protease inhibitor, enzyme inhibitor. Pharmacokinetic properties like blood-brain barrier pene- tration, human intestinal absorption, Caco-2 permeability and CYP450 2D6 substrate were predicted by submitting each molecule individually to the admetSAR server [39]. 2.3.5. In silico molecular docking studies The docking of the synthesized compounds to the binding pocket of glucosamine-6-phosphate synthase (GlcN-6-P) and P38 MAP kinase was carried out using the Autodock- Vina program [40]. The co-crystallized structure of GlcN- 6-P (PDB ID: 2VF5) and P38 MAP kinase (PDB ID: 1OUK) were retrieved from protein databank, and their substrate binding sites were identified using pdbsum server [41, 42]. A grid box of dimensions 40 x 50 x 40 Å with X, Y and Z coordinates at 32.198, 16.709 and –3.151 for GlcN-6-P and 56 x 60 x 48 Å with X, Y and Z coordinates at 44.746, 34.234 and 32.603 for P38 MAPk were created respective- ly. For the obtained molecules, all the torsions were al- lowed to rotate during docking. The grid box was set around the residues forming the active pocket. The bind- ing interactions were visualized using Biovia Discovery Studio Visualizer V.20.1 and Schrodinger-Maestro V.12.7. The in silico studies were performed on a local machine equipped with AMD Ryzen 5 six-core 3.4 GHz processor, 8 GB graphics and 16 GB RAM with Microsoft Windows 10 operating system. 2.3.6. Computational studies Computational studies of synthesized compounds (3a-d) were conducted by using the Gaussian 09 software [43] with the help of the density functional theory at Becke-3-Lee-Yang-parr (DFT)/B3LYP level with 6-31G (d,p) basis set [44]. The energy minimization pro- cess has been conducted at the same level in the gas phase to obtain a stable structure. The 3D representation of the optimized structure was presented in a molecular visuali- zation program Gauss view 5.0 and the output was pro- cessed using the Avogadro software [45]. 3. Results and discussion 3.1. Chemistry In this report, we developed a convenient and straightfor- ward method for the synthesis of 5-(3-substituted- thiophene)-pyrimidine derivatives (3a-d) via Knoevenagel condensation of barbituric/thiobarbituric acid (1) with 3-substituted-thiophene-2-carboxaldehyde (2) in aqueous ethanol using H2O2:HCl as catalyst (Scheme 1). A possible mechanism for the formation of 5-(3-substituted-thiophene)-pyrimidine derivatives is shown in Scheme 2. Firstly, hypochlorous acid (HOCl) forms by the reaction of H2O2 with HCl, and it acts as a Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 5 of 13 powerful oxidizing agent. Then the reaction is initiated by the generation of carbanion 2 from active methylene com- pound 1. The carbanion 2 attacks the carbonyl carbon of aldehyde 3 to form intermediate 4, which undergo subse- quent dehydration to desired Knoevenagel product 5. HOCl increases the abstraction of acidic proton from active methylene compound and electrophilicity of carbonyl group of aldehydes due to hydrogen bonding. Firstly, we studied the effect of catalyst on the reac- tion. In the previous reports, the same reaction was car- ried out in the presence of different catalysts such as CuO NPs, PVP-Ni NPs, Fe3O4 NPs, L-tyrosine, NH2SO3H, EAN, Bi(NO3)3.5H2O, and also in the absence of catalyst (Table 1). We studied the influence of catalyst in the pro- gress of reaction as well as on the increased product yield. The results were not very encouraging. Therefore, we con- cluded that the best result was obtained in the presence of green halogenating catalyst 6% H2O2:HCl, where a further increase in the quantity of catalyst doesn't significantly affect reaction kinetics. Consequently, to study the effect of temperature on synthesized compound 3a, we carried out the reaction at room temperature, 50 °C and 80 °C (Table 2). As a result, an increase in the reaction temperature decreased the re- action time from 60 to 20 and 20 to 10 min, respectively. Still, the yield of the product was not affected by an in- crease in temperature. The structures of the intended 5-(3-substituted- thiophene)-pyrimidine derivatives (3a-d) were confirmed by IR, 1H NMR, 13C NMR and HRMS spectral data. IR spec- trum of compound 3a showed that the absorption band in the region 3373 cm–1 is attributed to the amide stretching vibration, and the absorption band at 1654 cm–1 corre- sponds to stretching vibration of the carbonyl group (C=O). Another stretching vibrational band at 1529 cm–1 corresponds to C=C bond. The 1H NMR spectrum of com- pound 3a exhibited two singlet peaks at δ 11.21 and 11.17 ppm, which corresponds to two NH protons of pyrimidine nucleus (s, 2H, NH) and another singlet peak at δ 8.53 ppm due to CH proton (s, 1H, CH). A multiplet peak was observed in the range of δ 8.24–8.13 ppm, corre- sponds to two aromatic protons (m, 2H, Ar-H) and a tri- plet peak at 7.33–7.31 ppm due to one aromatic proton (t, J = 8 Hz, 1H, Ar–H). HN NH X O O S R H O HN N H O X O S R 1 2 3(a-d) + H2O2:HCl Reflux/ 10-15min X= O & S, R= H & CH3 Scheme 1 Synthesis of 5-(3-substituted-thiophene)-pyrimidine derivatives (3a-d) N H N H O O X H H O H Cl N H N H O O X _ S O H H O Cl R N H NH S O X O OH H R N H NH S O X O R H 2 O 2 + HCl HOCl + H2O 1 3 4 5 2 - HOCl - H2O - H2O Scheme 2 Possible mechanism of synthesized compounds (3a-d) In addition, the 13C NMR spectrum of compound 3a exhib- ited peaks at δ 163.950 and 163.453 ppm, which corre- spond to carbonyl carbons. The mass spectrum showed molecular ion peak [M+H]+ at m/z is 221. 9054 corresponds to the molecular weight of compound 3a (Supporting information: S1 to S16). The physical and analytical data of synthesized compounds (3a-d) were appended in Table 3. 3.2. Pharmacological effect 3.2.1. Antibacterial activity The synthesized 5-(3-substituted-thiophene)-pyrimidine de- rivatives (3a-d) were screened for their in vitro antibacterial activity at two different concentrations (20 and 40 µg/mL). All four compounds showed appreciable antibacterial ac- tivity with a varied zone of inhibition in the range of 3.3 to 3.8 and 7.1 to 7.8 mm against E. coli and 3.0 to 3.7 and 7.4 to 7.9 mm against S. aureus, respectively (Table 4). Table 1 Effect of catalysts on synthesized compound 3a Entry Catalyst Solvent Temperature, °C Time, min Yield, % 1 H2O2: HCl EtOH:H2O Reflux 10 96 2 CuO NPs – RT 10 93 [46] 3 PVP-Ni NPs Ethylene glycol Reflux 10 87 [47] 4 Fe3O4 NPs EtOH Reflux 30 70 [48] 5 L-tyrosine H2O RT 16 93 [49] 6 NH2SO3H – Grinding 120 96 [50] 7 – Ionic liquids RT 10 96 [51] 8 Bi(NO3)35H2O EtOH Reflux 20 95 [52] 9 – EtOH Reflux 120 89 [53] Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 6 of 13 Table 2 Effect of temperature on synthesized compounds 3a Entry Temperature, °C Time, min Yield, % 1 RT 60 96 2 50 20 96 3 80 10 96 The results revealed that compounds 3b (3.8 and 7.8 mm) and 3d (3.7 and 7.9 mm) having electron- donating group (methyl) on C-3 of the thiophene ring ex- hibited the most inhibitory effect against bacterial strains E. coli and S. aureus, respectively, compared to the stand- ard drug Ciprofloxacin. 3.2.2. Cytotoxicity All the four synthesized compounds were investigated for their in vitro cytotoxicity against MCF-7 (Breast cancer) cell line (Fig. 2). The plot details compound concentration versus the survival fraction (Fig. 3). The percentages of cell survival of the tested compounds are listed in Table 5. In vitro cytotoxicity results revealed that all four com- pounds displayed a superior selectivity against the MCF-7 cell line. Among the compounds tested, compound 3a ex- hibited promising cytotoxicity with a minimum cell sur- vival range of 23.68 to 44.16% at the concentration range of 200 to 6.25 µg/mL. Whereas 3b, 3c and 3d displayed reliable selectivity at all the concentrations with cell sur- vival ranges of 29.00 to 50.93%, 31.31 to 66.82% and 26.95 to 53.12%, respectively. 3.2.3. DNA binding study DNA binding was assessed using electronic spectroscopy. The UV-absorption spectral studies were employed to examine the binding mode of compounds to CT DNA, which involves the changes in absorbance and wavelength [55]. The mole- cules are bound to DNA with two modes (covalent or non- covalent) of binding. The covalent binding led to bathochrom- ism and hyperchromism due to breaking the DNA structure when a compound interacted with DNA covalently. While in non-covalent binding, there are "electrostatic", "groove", and "intercalative" types of interactions. Table 4 Antibacterial activity results of synthesized compounds (3a-d) Compd. Zone of inhibition in mm Escherichia coli Staphylococcus aureus Concentration in µg/mL 20 40 20 40 3a 3.5 7.8 3.0 7.5 3b 3.8 7.8 3.1 7.6 3c 3.3 7.2 3.5 7.4 3d 3.4 7.1 3.7 7.9 Ciprofloxacin 4.0 8.0 4.2 8.4 Table 3 Physical and analytical data of synthesized compounds (3a-d) Entry Comp. X R Product Yield (%) MP (oC) Observed Reported 1 3a O H HN N H O O O S 98 278–280 271 [54] 2 3b O CH3 HN N H O O O S 95 302–306 – 3 3c S H HN N H O S O S 94 320–322 – 4 3d S CH3 HN N H O S O S 96 310–312 – Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 7 of 13 Fig. 2 Images of anticancer study of the synthesized compounds (3a-d) Fig. 3 A graph of % of surviving cells of compounds (3a-d) at dif- ferent concentration against MCF-7 cell line Table 5 Percentage of cell viability against MCF-7 cell line of the synthesized compounds (3a-d) Concent- ration in µg/mL Mean cell Viability of MCF-7 3a 3b 3c 3d 6.25 44.16±0.76 50.93±0.42 66.82±0.41 53.12±0.34 12.5 38.11±0.82 48.68±0.31 58.1±0.13 46.88±0.52 25 34.09±1.2 43.72±0.52 48.6±0.23 43.93±0.42 50 32.37±0.82 40.23±0.61 40.03±0.82 40.03±0.62 100 30.06±0.62 36.80±0.20 37.38±0.61 37.85±0.34 200 23.68±0.41 29.00±0.16 31.31±0.42 26.95±0.52 Values are Mean ±SE, N=3, *P<0.01 vs. Control Decreased absorption (hypochromic shifts) and red- shift (bathochromic shift) revealed the intercalative bind- ing of compounds with DNA. Lower hypo- chromic/hyperchromic effect with no or negligible batho- chromic shift led to the electrostatic binding. Minor or no effect and, rarely, some hyperchromism show the groove binding [56–58]. The DNA binding efficiency of synthesized compounds (3a-d) was monitored by comparing their absorption spectra with and without CT DNA. The absorption spectra were car- ried out at a fixed concentration of synthesized compounds and varying with DNA concentrations (25–350 µL of 0.502510–7 to 6.067010–7 mol L–1) under the physiological condition of pH 7.01. The absorption spectra of all the syn- thesized compounds (3a-d) exhibited absorption bands at 235 to 240 nm due to π–π* transitions (Fig. 4). The Kb values of compounds (3a-d) are found to be 1.1216107, 1.4072107, 1.0634107 and 3.4872107 respectively are shown in Table 6. These Kb values confirm that all the synthesized compounds interacted strongly with CT DNA. Among the four com- pounds, compound 3d showed a prominent binding ability with CT DNA compared to other compounds. The absorption bands of the compounds were affected due to the gradual increase of CT DNA concentration resulting hyperchrom- ism/hypochromism. No/or negligible blue/red shift indicates strong interaction of the compounds with CT DNA mainly through electrostatic or groove binding [59]. The kinetics and thermodynamics of compounds-DNA interaction in terms of binding constant (Kb) and Gibbs free energy change (ΔG) were evaluated using the classical Van't Hoff's equation, ΔG= –2.303RT logKb. The negative ΔG values confirmed spon- taneous binding of compounds with CT DNA through the formation of stable complexes. 3.2.4. In silico ADME-toxicology studies The bioavailability and drug-likeness were estimated for all the synthesized compounds (3a-d) based on the mo- lecular properties. The results indicated that all the four compounds under study could pass through Lipinski's fil- ter without any violation, demonstrating a positive drug- likeness score indicating their suitability as drug-leads. In silico pharmacokinetic studies showed that all the mol- ecules under investigation could penetrate the blood-brain barrier and are readily absorbed by the human intestine while they are impermeable to Caco-2, and non-substrate Cytochromes P450 (CYP450) group of enzymes (Table 7). Table 6 DNA binding results of synthesized compounds (3a-d) Compd. λmax, nm Δλmax, nm % H Kb, M–1 ΔG, kJ/mol Free Bound 3a 240 239 1 6.187910–4 1.1216107 –40.205 3b 236 236 0 6.015710–4 1.4072107 –40.767 3c 239 239 0 1.156010–3 1.0634107 –40.073 3d 236 236 0 1.238810–3 3.4872107 –43.015 Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 8 of 13 Fig. 4 The electronic absorption spectra of compounds (3a-d) in the absence and presence of increasing amounts of CT DNA. Arrow (↓) shows the change in the absorbance with increase the DNA concentration. Inset: plot of [DNA]/( ∈A–∈F) Vs [DNA] Table 7 Bioavailability, drug likeness and in silico pharmacokinetic assessment of synthesized compounds (3a-d) Comp. Bioavailability and Drug likeness In silico Pharmacokinetics T o ta l M o le c u la r w e ig h t c L o g P H -A c c e p to r s H -D o n o r s R o ta ta b le B o n d s P o la r S u r fa c e A re a D r u g l ik e n e s s H u m a n i n te s ti n a l a b s o r p ti o n C a c o -2 p e r m e a b il it y B lo o d b r a in b a r ri e r C Y P 4 5 0 2 D 6 s u b s tr a te 3a 222.224 0.2739 5 2 1 103.51 5.2698 +0.982 –0.779 +0.982 –0.873 3b 236.251 0.6178 5 2 1 103.51 4.9763 +0.728 –0.753 +0.980 –0.889 3c 238.291 0.6344 4 2 1 118.53 4.2229 +0.979 –0.698 +0.977 –0.871 3d 252.318 0.9783 4 2 1 118.53 3.9143 +0.987 –0.673 +0.976 –0.886 In silico pharmacodynamics studies revealed that all the four molecules are non-mutagenic, non- tumorigenic, non-irritant, AMES non-toxic with high reproductive effects with possible hepatotoxicity. The bioactivity assessment indicated that the molecules do not belong to the GPCR group of ligands, do not modu- late ion channels, non-kinase inhibitors, non-nuclear receptor ligands, non-protease and non-enzyme inhibi- tors (Table 8). 3.2.5. Structure-activity relationship (SAR) studies The evaluation of the antibacterial activity of the newly synthesized compounds (3a-d) revealed that the presence of electron-donating methyl group has a significant effect on enhancing their potency. The compounds 3b and 3d have a methyl group at C-3 of the thiophene ring. This could have improved their cell permeability, which im- proved their activity profile compared to 3a and 3c. The combination of heterocyclic rings like pyrimidine and thi- ophene is presumed to be the main reason for the pro- found cytotoxicity of the newly synthesized compounds (3a-d). Furthermore, the presence of the urea group in the pyrimidine ring could have significantly contributed to the superior cytotoxicity of 3a and 3b compared to other syn- thesized molecules. Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 9 of 13 3.2.6. In silico molecular docking studies The results revealed that compound 3b bound with GlcN- 6-P and P38 MAPk with a minimum binding energy of –7.9 and –6.4 kcal/mol, respectively. 3a, 3d and 3c interacted with a binding energy of –7.6 and –6.4, –7.4 and –6.2 and –7.4 and –6.0 with GlcN-6-P and P38 MAPk targets, re- spectively. The interaction of all the molecules with GlcN- 6-P and P38 MAPk were compared with antibacterial agent Ciprofloxacin (–7.7 kcal/mol) and anticancer agent 5-fluorouracil (–4.7 kcal/mol) (Table 9, Fig.5). The compu- tational methods in drug discovery have gained enormous importance in modern drug research. They play a critical role in reducing the virtual chemical space in synthesizing, modifying, and screening chemical drugs against a specific disease target. Their effectiveness can be validated using microbial pathogens due to their simpler and clearer cellu- lar understandings. Similarly, cell death modalities can also be studied using in vitro cell culture studies, particu- larly focusing on necrosis, apoptosis, necroptosis, au- tophagic cell death, etc. [42]. In the present study, in silico molecular docking studies were performed to predict the most effective binding among the synthesized molecules to appropriate targets [60, 61]. The studied molecules showed remarkable binding interactions with the selected target proteins, supporting their remarkable antimicrobial and anticancer effects. 3.2.7. DFT studies Frontier molecular orbitals (FMOs) containing the highest occupied molecular orbital (HOMO) and lowest unoccupied molecular orbital (LUMO), as well as the energy gap (ΔE = EHOMO –ELUMO) were considered to be very effective pa- rameters in chemical quantum chemistry [62]. FMOs also delivered important information about chemical reactivity, biological activity and kinetic stability of the molecules [63]. The optimized HOMO and LUMO structures of synthesized compounds (3a-d) are shown in Fig. 6 and 7. The HOMO and LUMO of compounds were helpful in determining of various global reactivity parameters such as ionization energy (I = –EHOMO), electron affinity (A = –ELUMO), chemical hard- ness (ƞ = 1/2 (I – A)), chemical softness (σ = 1/ƞ), electroneg- ativity (χ = 1/2 (I + A)), chemical potential (µ = –χ), and elec- trophilicity index (ω = µ2/2ƞ) helps to study about donor– acceptor interaction and intramolecular charge transfer (ICT) ability of synthesized compounds [64]. The calculated HOMO-LUMO energies and global reac- tivity parameters of the synthesized compounds (3a-d) are displayed in Table 10. A smaller HOMO-LUMO energy gap (ΔE) indicates a soft molecule, while a larger gap indicates a hard molecule. Lower energy gap, less ionization poten- tial, electron affinity, chemical hardness, electronegativi- ty, electrophilicity index values and more softness values indicate that a molecule is more chemically and biological- ly active with low kinetic stability [65]. Fig. 5 Binding interaction of compounds 3a (a and b), 3b (c and d), 3c (e and f), and 3d (g and h) with GlcN-6-P along with standard drug ciprofloxacin (i and j) (a) and Binding interaction of com- pounds 3a (a and b), 3b (c and d), 3c (e and f), and 3d (g and h) with P38 MAPk along with standard drug 5-fluorouracil (I and j) (b) Table 8 In silico pharmacodynamics and bioactivity assessment of synthesized compounds (3a-d) Comp. In silico Pharmacodynamics Bioactivity score M u ta g e n ic T u m o ri g e n ic R e p r o d u c ti v e E ff e c ti v e Ir ri ta n t A e r o b ic b io d e g r a d a b il - it y A m e s t o x ic it y H e p a to to x ic it y G P C R l ig a n d Io n c h a n n e l m o d u la to r K in a s e in h ib it o r N u c le a r r e c e p to r li g a n d P r o te a s e i n h ib it o r E n z y m e i n h ib it o r 3a NONE NONE HIGH NONE –0.590 –0.706 +0.925 –1.12 –1.58 –0.86 –1.26 –1.30 –0.69 3b NONE NONE HIGH NONE –0.562 –0.694 +0.950 –1.07 –1.44 –0.90 –0.91 –1.32 –0.73 3c HIGH NONE HIGH NONE –0.859 –0.731 +0.900 –1.43 –1.77 –1.41 –1.73 –1.32 –1.00 3d HIGH NONE HIGH NONE –0.843 –0.711 +0.850 –1.35 –1.62 –1.42 –1.35 –1.34 –1.03 Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 10 of 13 DFT studies data reveals that the energy gaps (ΔE) of compounds 3a, 3b, 3c and 3d are 0.16038 eV, 0.15988 eV, 0.12799 eV and 0.12636 eV, respectively. 3a, 3b and 3c, 3d moleculs show nearly similar energy gap due to the simi- lar structure. Among them 3d molecule shows less energy gap (0.12636 eV) and more softness value (15.8277 eV); hence, it is chemically more reactive compared to the oth- er molecules. 3a molecule has more electronegative value (0.19923 eV); hence, it has more tendency to attract a bonding electron pairs compared to the other molecules. Table 9 Binding energies of synthesized compounds (3a-d) with GlcN-6-P and P38 MAPk targets Antibacterial activity Anticancer activity Compd. Binding energy in kcal/mol Compd. Binding energy in kcal/mol Ciprofloxacin –7.7 5-Fluorouracil –4.7 3a –7.6 3a –6.4 3b –7.9 3b –6.4 3c –7.4 3c –6.0 3d –7.4 3d –6.2 Conclusions We described a mild, easy and green protocol for synthesiz- ing 5-(3-substituted-thiophene)-pyrimidine derivatives (3a- d) using H2O2:HCl as a catalyst under reflux condition. This synthetic approach has a short reaction time, excellent yield, and clean reactions make this procedure a magnifi- cent alternative to the existing methods. Furthermore, this method is environmentally greener and safer. The activity results revealed that the compounds 3b and 3d exhibited more potent antibacterial activity against E. coli and S. au- reus than the standard drug Ciprofloxacin. In vitro cytotoxi- city results disclosed the outstanding selectivity on MCF-7 cell line, mainly compound 3a exhibiting the most effective cytotoxicity with a minimum cell survival range of 23.68 to 44.16%. DNA binding results indicated that all the synthe- sized compounds interacted strongly with CT DNA, and compound-DNA complexes were stabilized by electrostatic or groove binding. In silico ADME-toxicology results showed that all the four compounds are non-toxic and suitable for oral bioavailability and drug-likeness, indicating their suit- ability as drug-leads. Table 10 Chemical parameters of synthesized compounds (3a-d) Entry EHOMO (eV) ELUMO, eV ΔE EHOMO– ELUMO, eV I, eV A, eV ƞ, eV σ, eV χ, eV µ, eV ω, eV D, Debye 3a –0.27942 –0.11904 0.16038 0.27942 0.11904 0.08019 12.4703 0.19923 –0.19923 0.24749 4.6744 3b –0.27411 –0.11423 0.15988 0.27411 0.11423 0.07994 12.5093 0.19417 –0.19417 0.23581 5.2238 3c –0.25095 –0.12296 0.12799 0.25095 0.12296 0.06399 15.6274 0.18695 –0.18695 0.27310 5.4300 3d –0.24793 –0.12157 0.12636 0.24793 0.12157 0.06318 15.8277 0.18475 –0.18475 0.27012 5.9267 Fig. 6 Optimized structures of synthesized compounds (3a-d) Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 11 of 13 Fig. 7 DFT study of synthesized compounds (3a-d) In silico molecular docking results showed compound 3b bound with GlcN-6-P and P38 MAPk with a least binding energy of –7.9 and –6.4 kcal/mol, respectively. DFT study result revealed that the compound 3d was chemically and biologically more reactive with low kinetic stability due to less energy gap. In future, the obtained compounds can be used as antibiotics, anticancer agents, dyes and pigments in compounds as food industries. Acknowledgements The authors are thankful to the Chairman, Department of Chemistry, Kuvempu University, Shankaraghatta, for providing the laboratory facilities and SAIF, Mysore Uni- versity, for providing the spectral data. Supporting information Spectroscopic spectra's (IR, 1HNMR, 13CNMR and HRMS) of the synthesized compounds are provided in the supporting information file. Conflict of Interest The authors declare that there is no conflict of interests regarding the publication of this work. References 1. Singh PK, Silakari O. The current status of O‐heterocycles: A synthetic and medicinal overview. Med Chem. 2018;13:1071– 1087. doi:10.1002/cmdc.201800119 2. Pathania S, Narang RK, Rawal RK. Role of sulphur- heterocycles in medicinal chemistry: An update. Eur J Med Chem. 2019;180:486–508. doi:10.1016/j.ejmech.2019.07.043 https://doi.org/10.1002/cmdc.201800119 https://doi.org/10.1016/j.ejmech.2019.07.043 Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 12 of 13 3. Venkatesh T, Bodke YD, Manjunatha B, Kumar SR. Synthesis, antitubercular activity and molecular docking study of substi- tuted [1,3]dioxino[4,5-d] pyrimidine derivatives via facile CAN catalyzed Biginelli reaction. Nucleosides, Nucleotides and Nucleic Acids. 2021:1–13. doi:10.1080/15257770.2021.1972310 4. Sravanthi B, Kaviarasan L, Praveen TK, Sai Kiran PSS, Pa- vankumar C, Gowramma B. Synthesis and pharmacological evaluation of 1,3,4-thiadiazole bearing pyrimidine derivatives as STAT3 inhibitor for treatment of breast cancer. J Iran Chem Soc. 2020;17:2359–2370. doi:10.1007/s13738-020-01932-z 5. Kang D, Feng D, Ginex T, Zou J, Wei F, Zhao T, Huang B, Sun Y, Desta S, Clercq ED, Pannecouque C, Zhan P, Liu X. Explor- ing the hydrophobic channel of NNIBP leads to the discovery of novel piperidinesubstituted thiophene[3,2-d]pyrimidine derivatives as potent HIV-1 NNRTIs. Acta Pharm Sin B. 2020;10:878–894. doi:10.1016/j.apsb.2019.08.013 6. Prasad P, Anirudhdha GK and Patel MP. Microwave assisted one-pot synthetic route to imidazo[1,2-a]pyrimidine deriva- tives of imidazo/triazole clubbed pyrazole and their pharma- cological screening. New J Chem. 2018;42:12666–12676. doi:10.1039/C8NJ00670A 7. Zia UHK, Arif UK, Pingyu W, Yongmei C, Dandan K, Shafiul- lah K, Kamran T. In vitro pharmacological screening of three newly synthesised pyrimidine derivatives. Nat Prod Res. 2015;29:933–938. doi:10.1080/14786419.2014.964707 8. Ghada SM. New Potential Antitumor Pyrimidine Derivatives: Synthesis and Cytotoxicity Evaluation. Polycycl Aromat Compd. 2021:1–17. doi:10.1080/10406638.2021.1936086 Zhen X, Cilong C, Lingjia Z, Zhihui Z, Qian Z, Feiyi Y, Zunhua9. Y, Pengwu Z, Shan X, Wufu Z. Discovery of thiapyran- pyrimidine derivatives as potential EGFR inhibitors. Bioorg Med Chem. 2020;28:1–13. doi:10.1016/j.bmc.2020.115669 10. Zhen X, Zhihui Z, Cilong C, Qian Z, Lingjia Z, Zunhua Y, Xin Li, Liying Yu, Pengwu Z, Shan Xu, Wufu Z. Design, synthesis and antitumor activity of novel thiophene-pyrimidine deriva- tives as EGFR inhibitors overcoming T790M and L858R/T790M mutations. Eur J Med Chem. 2020;203:1–43. doi:10.1016/j.ejmech.2020.112511 11. Elshaymaa IE. Thieno[2,3-d]pyrimidine derivatives: Synthet- ic approaches and their FLT3 kinase inhibition. J Heterocyclic Chem. 2020;57:2067–2078. doi:10.1002/jhet.3934 12. Khalil NA, Ahmed EM, Zaher AF, El-Zoghbi MS, Sobh EA. Syn- thesis of certain benzothieno[3,2-d]pyrimidine derivatives as a selective SIRT2 inhibitors. Eur J Med Chem. 2020;187:1–56. doi:10.1016/j.ejmech.2019.111926 13. Shipilovskikh SA, Rubtsov AE. One-Pot Synthesis of Thieno[3,2-e]pyrrolo[1,2-a]pyrimidine Derivative Scaffold: A Valuable Source of PARP-1 Inhibitors. J Org Chem. 2019;84:15788–15796. doi:10.1021/acs.joc.9b00711 14. Lei S, Chengjuan C, Xueting H, Hao H, Manman W, Jianqiu Z, Yile Y, Jianming L, Tiantai Z, Dayong Z. Design, synthesis, and pharmacological evaluation of 4- or 6-phenylpyrimidine derivatives as novel and selective Janus kinase 3 inhibitors. Eur J Med Chem. 2020;191:1–18. doi:10.1016/j.ejmech.2020.112148 15. Giuseppe R, Loredana S, Valeria P, Marialuisa C, Sebastiano I, Emanuele A, Mario S, Alfredo C, Antonio R, Giuseppe F, Maria NM. [1]Benzothieno[3,2-d]pyrimidine derivatives as ligands for the serotonergic 5-HT7 receptor. Eur J Med Chem. 2019;183:1–16. doi:10.1016/j.ejmech.2019.111690 16. Ali I, Wani WA, Saleem K, Hsieh MF. Anticancer metallodrugs of glutamic acid sulphonamides: in silico, DNA binding, he- molysis and anticancer studies. RSC Adv. 2014;4:29629– 29641. doi:10.1039/C4RA02570A 17. Aarti A, Archana K, Dr. Amarjit KA, Dr. Nithish C, Yudhvir S. Recent advances made on anticancer drugs the therapeutic potential of the aromatic heterocyclic compounds. Int J Pharm Sci Rev Res. 2019;58:104–113. 18. Jawaid A, Ahsan AK, Zulphikar Ali, Rafi H, Shahar YM. Struc- ture-activity relationship (SAR) study and design strategies of nitrogen-containing heterocyclic moieties for their anti- cancer activities. Eur J Med Chem. 2017;125:143–189. doi:10.1016/j.ejmech.2016.09.023 19. Prabodh CS, Kushal KB, Archana S, Diksha S, Aakash D. Thia- zole-containing compounds as therapeutic targets for cancer therapy. Eur J Med Chem. 2020;188:1–47. doi:10.1016/j.ejmech.2019.112016 20. Garima M, Sumitra N, Pramod KS. Cancer: An overview. J Cancer Res. 2015;8:1–9. doi:10.5829/idosi.ajcr.2015.8.1.9336 21. Nathan MR and Schmid P. A review of fulvestrant in breast cancer. Oncol Ther. 2017;5:17–29. doi:10.1007/s40487-017-0046-2 22. Akhtar J, Khan AA, Ali Z, Haider R, Yar MS. Structure-activity relationship (SAR) study and design strategies of nitrogen containing heterocyclic moieties for their anticancer activi- ties. Eur J Med Chem. 2016;125:143–189. doi:10.1016/ j.ejmech.2016.09.023 23. Koohshari M, Dabiri M, Salehi P. Catalyst-free domino reac- tion in water/ethanol: an efficient, regio- and chemoselective one-pot multi-component synthesis of pyranopyrazole deriva- tives. RSC Adv. 2014;4:10669–10671. doi:10.1039/C3RA47639A 24. Daniel RB, De Visser SP, Shaik S, Neumann R. Electrophilic aromatic chlorination and haloperoxidation of chloride cata- lyzed by polyfluorinated alcohols: a new manifestation of template catalysis. J Am Chem Soc. 2003;125:12116–12117. doi:10.1021/ja0364524 25. Venkatesh T, Bodke YD, Nagaraj K, Kumar SR. One-pot syn- thesis of novel substituted phenyl-1,5-dihydro-2h- benzo[4,5]thiazolo[3,2-a]pyrimido[4,5-d]pyrimidine deriva- tives as potent antimicrobial agents. Med Chem. 2018;8:1–7. doi:10.4172/2161-0444.1000488 26. Venkatesh T, Bodke YD, Kenchappa R, Telkar S. Synthesis, antimicrobial and antioxidant activity of chalcone derivatives containing thiobarbitone nucleus. Med Chem. 2016;6:440– 448. doi:10.4172/2161-0444.1000383 27. Sukanya SH, Venkatesh T, Kumar SR, Bodke YD. Facile TiO2 NPs catalysed synthesis of substituted-4-Hydroxy/methoxy benzylidene derivatives as potent antioxidant and anti- tubercular agents. Chem Data Collect. 2021;33:1–16. doi:10.1016/j.cdc.2021.100713 28. Venkatesh T, Bodke YD, Joy NM, Vinoda BM, Shiralgi Y, Dhananjaya BL. Synthesis of some novel 5,7-disubstituted-2- phenyl-5H-[1,3,4]thiadiazolo[3,2-a]pyrimidine derivatives and evaluation of their biological activity. Lett Org Chem. 2016;13:661–671. doi:10.2174/1570178613666161017123123 29. Sukanya SH, Venkatesh T, Aditya Rao SJ, Joy MN. Efficient L- Proline catalyzed synthesis of some new (4-substituted- phenyl)-1,5-dihydro-2H-pyrimido[4,5- d][1,3]thiazolo[3,2a]- pyrimidine-2,4(3H)-diones bearing thiazolopyrimidine deriv- atives and evaluation of their pharmacological activities. J Mol Struct. 2021;1247:1–13. doi:10.1016/j.molstruc.2021.131324 30. Shivakumara N, Murali Krishna P. Synthesis, Spectral Char- acterization, and Evaluation of their DNA interactions. Curr Chem Lett. 2019;8:157–168. doi:10.5267/j.ccl.2019.004.004 31. Ali I, Mukhtar SD, Hsieh MF, Alothman ZA, Alwarthan A. Fac- ile synthesis of indole heterocyclic compounds based micel- larnano anticancer drugs. RSC Adv. 2018;8:37905–37914. doi:10.1039/c8ra07060a 32. Wolfe A, Shimer GH, Meehan T. Polycyclic aromatic hydro- carbons physically intercalate into duplex regions of dena- tured DNA. Biochemistry. 1987;26:6392–6396. 33. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experi- mental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 1997;23:3–25. doi:10.1016/S0169-409X(96)00423-1 34. Aditya R, Venugopal T, Jayanna N, Paramesha M, Ramesh C. Bioactive isolates of Morus species as antibacterial agents and their insilico profiling. Lett. Drug Des Discov. 2020;17:1– 9. doi:10.2174/1570180817999201104120815 https://doi.org/10.1080/15257770.2021.1972310 https://doi.org/10.1007/s13738-020-01932-z https://doi.org/10.1016/j.apsb.2019.08.013 https://doi.org/10.1039/C8NJ00670A https://doi.org/10.1080/14786419.2014.964707 https://doi.org/10.1080/10406638.2021.1936086 https://doi.org/10.1080/10406638.2021.1936086 https://doi.org/10.1016/j.bmc.2020.115669 https://doi.org/10.1016/j.ejmech.2020.112511 https://doi.org/10.1002/jhet.3934 https://doi.org/10.1016/j.ejmech.2019.111926 https://doi.org/10.1021/acs.joc.9b00711 https://doi.org/10.1016/j.ejmech.2020.112148 https://doi.org/10.1016/j.ejmech.2019.111690 https://doi.org/10.1039/C4RA02570A https://doi.org/10.1016/j.ejmech.2016.09.023 https://doi.org/10.1016/j.ejmech.2019.112016 https://doi.org/10.1007/s40487-017-0046-2 https://doi.org/10.1016/%20j.ejmech.2016.09.023 https://doi.org/10.1039/C3RA47639A https://doi.org/10.1021/ja0364524 https://doi.org/10.4172/2161-0444.1000488 https://doi.org/10.4172/2161-0444.1000383 https://doi.org/10.1016/j.cdc.2021.100713 https://doi.org/10.2174/1570178613666161017123123 https://doi.org/10.1016/j.molstruc.2021.131324 https://doi.org/10.5267/j.ccl.2019.004.004 https://doi.org/10.1039/c8ra07060a https://doi.org/10.1016/S0169-409X(96)00423-1 https://doi.org/10.2174/1570180817999201104120815 Chimica Techno Acta 2021, vol. 8(4), № 20218401 ARTICLE 13 of 13 35. Jarrahpour A, Motamedifar M, Zarei M, Youssoufi MH, Mimouni M, Chohan ZH. Petra, Osiris, and Molinspiration to- gether as a guide in drug design: Predictions and correlation structure/antibacterial activity relationships of new n- sulfonyl monocyclic β-Lactams. Phosphorus Sulfur Silicon Relat Elem. 2010;185:491–497. doi:10.1080/10426500902953953 36. Raghavendra S, Aditya Rao SJ, Kumar V, Ramesh CK. Multiple ligand simultaneous docking (MLSD): A novel approach to study the effect of inhibitors on substrate binding to PPO. Comput Biol Chem. 2015;59:81–86. doi:10.1016/j.compbiolchem.2015.09.008 37. Sander T, Freyss J, Von Korff M, Rufener C. DataWarrior: An open-source program for chemistry aware data visualization and analysis. J Chem Inf Model 2015;55:460–473. doi:10.1021/ci500588j 38. Venkatesh T, Bodke YD, Aditya Rao SJ. Facile CAN catalyzed one pot synthesis of novel indol-5,8-pyrimido[4,5- d]pyrimidine derivatives and their pharmacological study. Chem Data Collect. 2020;25:1–13. doi:10.1016/j.cdc.2019.100335 39. Cheng F, Li W, Zhou Y, Shen J, Wu Z, Liu G. AdmetSAR. A comprehensive source and free tool for assessment of chemi- cal ADMET properties. J Chem Inf Model. 2012;52:3099– 3105. doi:10.1021/ci300367a 40. Trott O, Olson JA. AutoDockVina: Improving the speed and accuracy of docking with a new scoring function, efficient op- timization and multithreading. J Comput Chem. 2010;31:455– 461. doi:10.1002/jcc.21334 41. Aditya Rao SJ, Ramesh CK, Raghavendra S, Paramesha M. Dehydroabietylamine, A diterpene from carthamus tinctori- ous L. showing antibacterial and anthelmintic effects with computational evidence. Curr Comput Aided Drug Des. 2020;16:231–237. doi:10.2174/1573409915666190301142811 42. Aditya Rao SJ, Shivayogi SM, Satyanarayana JK, Kumaran RC. Characterization of isolated compounds from Morus spp. and their biological activity as anticancer molecules. Bioimpacts 2021;11:1–11. doi:10.34172/bi.2021.09 43. Frisch MJEA, Trucks GW, Schlegel HB, Scuseria GE, Robb MA, Cheeseman JR, Nakatsuji H. Gaussian 09, Revision d. 01, Gaussian. Inc. Wallingford CT 2009;201. 44. Becke AD. A new mixing of Hartree-Fock and local density‐ functional theories. J Chem Phys. 1993;98:1372–1377. 45. Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR. Avogadro an advanced semantic chemical edi- tor, visualization, and analysis platform. Aust J Chem. 2012;4:1–33. doi:10.1186/1758-2946-4-17 46. Dighore NR, Anandgaonker PL, Gaikwad ST, Rajbhoj AS. Sol- vent free green synthesis of 5-arylidine barbituric acid deriv- atives catalyzed by copper oxide nanoparticles. Res J Chem Sci. 2014;4:93–98. 47. Khurana JM, Kanika Vij. Nickel nanoparticles catalyzed knoevenagel condensation of aromatic aldehydes with barbi- turic acids and 2-thiobarbituric acids. Catal Lett. 2010;138:104–110. doi:10.1007/s10562-010-0376-2 48. Fattahi M, Davoodnia A, Pordel M. Efficient one-pot synthesis of some new pyrimido[5,4:5,6]pyrido[2,3-d]pyrimidines catalyzed by magnetically recyclable Fe3O4 nanoparticles. Russ J Gen Chem. 2017;87:863–867. doi:10.1134/S1070363217040326 49. Thirupathi G, Venkatanarayana M, Dubey PK, BharathiKuma- ri Y. Facile and green syntheses of 5-arylidene-pyrimidine- 2,4,6-triones and 5-arylidene-2-thioxo-dihydro-pyrimidine-4, 6-diones using L-tyrosine as an efficient and eco-friendly cat- alyst in aqueous medium. Chem Sci Trans. 2013;2:441–446. doi:10.7598/cst2013.385 50. Tai Li J, Guang Dai H, Liu D, Shuang Li T. Efficient method for synthesis of the derivatives of 5‐arylidene barbituric acid catalyzed by aminosulfonic acid with grinding. Synth Com- mun. 2006;36:789–794. doi:10.1080/00397910500451324 51. Hu Y, Chen ZC, Le ZG, Zheng QG. Organic reactions in ionic liquids: ionic liquid promoted knoevenagel condensation of aromatic aldehydes with (2‐thio) barbituric acid. Synth Commun. 2004;34:4521–4529. doi:10.1081/SCC-200043210 52. Shabeer M, Barbosa LCA, Karak M, Coelho ACS. Thiobarbitu- rates as potential antifungal agents to control human infec- tions caused by Candida and Cryptococcus species. Med Chem Res. 2018;27:1043–1049. doi:10.1007/s00044-017-2126-0 53. Yan Q, Cao R, Yi W, Chen Z, Wen H, Ma L, Song H. Inhibitory effects of 5-benzylidene barbiturate derivatives on mushroom tyrosinase and their antibacterial activities. Eur J Med Chem. 2009;44:4235–4243. doi:10.1016/j.ejmech.2009.05.023 54. Egorova S, Ivanova VN, Putokhin NI. Thiophene aldehyde and its derivatives. Chem Heterocycl Comp. 1967;3:654. doi:10.1007/BF00468337 55. Ramesh G, Daravath S, Ganji N, Rambabu A, Venkateswarlu K. Facile synthesis, structural characterization, DNA binding, incision evaluation, antioxidant and antimicrobial activity studies of Cobalt(II), Nickle(II) and Copper(II) complexes of 3-amino-5-(4-fluorophenyl)isoxazole derivatives. J Mol Struct. 2019;1202:1–41. doi:10.1016/j.molstruc.2019.127338 56. Ali I, Haque A, Saleem K, Hsieh MF. Curcumin-I Knoevenagel's condensates and their Schiff's bases as anti- cancer agents: synthesis, pharmacological and simulation studies. Bioorg Med Chem. 2013;21:3808–3820. doi:10.1016/j.bmc.2013.04.018 57. Indumathy R, Kanthimathi M, Weyhermuller T, Nair BU. Co- balt complexes of terpyridine ligands: crystal structure and nuclease activity. Polyhedron 2008;27:3443–3450. doi:10.1016/j.poly.2008.08.003 58. Zhao P, Xu LC, Huang JW, Liu J, Yu HC, Zheng KC, Ji LN. Ex- perimental and DFT studies on DNA binding and photocleav- age of two cationic porphyrins: effects of the introduction of a carboxyphenyl into pyridiniumporphyrin. Spectrochim Acta A Mol Biomol Spectrosc. 2008;71:1216–1223. doi:10.1016/j.saa.2008.03.031 59. Hajian R, Ekhlasi E, Daneshvar R. Spectroscopic and electro- chemical studies on the interaction of epirubicin with fish sperm DNA. J Chem. 2012;9:1587–1598. doi:10.1155/2012/738678 60. Campillos M, Kuhn M, Gavin AC, Jensen LJ, Bork P. Drug tar- get identification using side-effect similarity. Science. 2008;321:263–266. doi:10.1126/science.1158140 61. Keiser MJ, Roth BL, Armbruster BN, Ernsberger P, Irwin JJ, Shoichet BK. Relating protein pharmacology by ligand chem- istry. Nat Biotechnol. 2007;25:197–206. doi:10.1038/nbt1284 62. Ali A, Khalid M, Rehman MF, Haq S, Ali A, Tahir MN, Ashfaq M, Rasool F, Braga AAC. Efficient synthesis, SC-XRD, and the- oretical studies of O-Benzenesulfonylated pyrimidines: Role of noncovalent interaction influence in their supramolecular network. ACS Omega. 2020;5:15115–15128. doi:10.1021/acsomega.0c00975 63. Ooretir C, Kaniskan N. Frontier orbital theory and chemical reactivity: The utility of spectroscopy and molecular orbital calculations. Recent Experimental and Computational Ad- vances in Molecular Spectroscopy. 1993:351–367. 64. Anup Pandith, Young Jun Seo. Label-free sensing platform for miRNA-146a based on chromofluorogenic pyrophosphate recognition. J Inorg Biochem. 2020;203:1–41. doi:10.1016/j.jinorgbio.2019.110867 65. George J, Prasana JC, Muthu S, Kuruvilla TK, Savanthi S, Saji RS. Spectroscopic (FT-IR, FT-Raman) and Quantum mechani- cal study on N-(2,6 dimehyl phenyl)-2-{4-[2hydroxy-3- (2methoxyphenoxyl)propyl]piperazin-1yl}acetamide. J Mol Struct. 2018;1171:268–278. doi:10.1016/j.molstruc.2018.05.106 https://doi.org/10.1080/10426500902953953 https://doi.org/10.1016/j.compbiolchem.2015.09.008 https://doi.org/10.1021/ci500588j https://doi.org/10.1016/j.cdc.2019.100335 https://doi.org/10.1021/ci300367a https://doi.org/10.1002/jcc.21334 https://doi.org/10.2174/1573409915666190301142811 https://doi.org/10.34172/bi.2021.09 https://doi.org/10.1186/1758-2946-4-17 https://doi.org/10.1007/s10562-010-0376-2 https://doi.org/10.1134/S1070363217040326 https://doi.org/10.7598/cst2013.385 https://doi.org/10.1080/00397910500451324 https://doi.org/10.1081/SCC-200043210 https://doi.org/10.1007/s00044-017-2126-0 https://doi.org/10.1016/j.ejmech.2009.05.023 https://doi.org/10.1007/BF00468337 https://doi.org/10.1016/j.molstruc.2019.127338 https://doi.org/10.1016/j.bmc.2013.04.018 https://doi.org/10.1016/j.poly.2008.08.003 https://doi.org/10.1016/j.saa.2008.03.031 https://doi.org/10.1155/2012/738678 https://doi.org/10.1126/science.1158140 https://doi.org/10.1038/nbt1284 https://doi.org/10.1021/acsomega.0c00975 https://doi.org/10.1016/j.jinorgbio.2019.110867 https://doi.org/10.1016/j.molstruc.2018.05.106