Abstract
The reaction of 3,4-dichlorophenyl-1,3,4-oxadiazole-2( 3H )-thione with piperidine derivatives via Mannich reaction was used to generate eleven novel compounds in moderate to good yields. Synthesized molecules were characterized according to their structure with 1H NMR, 13C NMR and FT-IR spectral foundations, which were compatible with literature informations. Antimicrobial activity and cytotoxicity studies were done by disc diffusion and NCI-60 sulphordamine B assay methods. The antimicrobial test results revealed that synthesized compounds have better activity against gram-positive species than gram-negative ones. A total analysis of the antibacterial, antifungal, and antiyeast activity revealed that newly synthesized compounds were really active against Bacillus cereus , Bacillus ehimensis, and Bacillus thuringiensis species . For cytotoxicity, among three different cancer cell lines (HCT116, MCF7, HUH7) compounds 5c, 5d, 5e, 5f, 5g, 5i, 5j and 5k were seemed especially effective on HUH7 cancer cell line via moderate to good activity. More significantly, against liver carcinoma cell line (HUH7) most of the compounds of the series ( 5c-5g and 5i-5j ) have better IC50 values (IC50= 18.78 µM) than 5-Florouracil (5-FU) and also compound 5d possessed 10.1 µM value, which represents good druggable cytotoxic activity. Further, the molecules were also screened for in silico chemoinformatic and toxicity data to gather the predicted bioavailibity and safety measurements.
Keywords: 1,3,4-Oxadiazole; piperidine; antibacterial activity; cytotoxicity
1. Introduction
Antimicrobial drugs are known as a group of pharmaceuticals that preserve various defensive effects against bacteria, fungi, virus, and parasites. It is well recognized that many antimicrobial agents are necessary to treat life-threatening infections, but improved bacterial resistance against these medications may cause worser consequences for human health. Antimicrobial resistance occurs naturally over time, when responsible strains of a microorganism exchange among microorganisms usually through genetic material. This proceeding problem of currently marketed antimicrobials have speed up the birth of new variants of mechanisms about resistance and fairly fast rise in the number of microorganisms which spread among all over the world [1–4].
To prevent the risk of increase in antimicrobial resistance, different therapy strategies should be applied according to the type of microorganisms (drug-resistant or not) and disease severity. Since 2005, World Health Organization (WHO) has developed a criteria to order antimicrobials according to their relative importance in human medicine and the study has been used by clinicians to preserve the usage quality of currently available drugs [5]. This commercially available medicines which are used for infection and cancer treatments have a common point: the emergence of resistance against multiple drugs [6,7]. Other related problems are insufficient selectivity and undesirable side effects consequences of the patients [6,8]. Hence, there is a massive requirement for the improvement of new antimicrobial and anticancer therapies, with higher selectivity, besides serving fewer side effects than current ones. So, the main goal is resistance prevention, good potential of action, and last but not least, diminished side effects [9–12]. As well as developing antimicrobial treatment strategies, new studies on antimicrobials have been maintained as cancer theraphy due to strong evidences about the relationship between microbial diseases and cancer. This data supports the combined usage of antimicrobial and anticancer medications in clinical area. In addition, studies on direct antiproliferative activity of certain antimicrobials and prophylactic effect of antimicrobials against post-chemotheraphy infections due to immunosupression are being discussed with their possible mechanisms about cancer treatment [13]. Today, it is reported in the literature that antiproliferative activity of clinically used antimicrobials is related to the topoisomerase enzyme inhibition [14,15], degradation of tumorigenic proteins [16,17], destabilization [18], antiangiogenic effects and apoptosis [19–24].
1,3,4-Oxadiazole is generally used entity for pharmacophore development and has been investigated because of its good metabolic profile and hydrogen-bonding capacity within the receptor site. Presence of azole group (N=C-O) also elevates lipophilicity feature of compound, which provides advantage for its transportation through cell membrane to reach the target site and show various biological activities [25]. These co-operative properties provide great benefits to obtain desired antimicrobial and anticancer activity within various proven in vitro and in vivo models. In 2013, Du and coworkers dealed with modeling 1,3,4-oxadiazole ring to obtain both of anticancer and antimicrobial effect by targeting thymidylate synthase (TS), which is an important enzyme for DNA synthesis. Newly synthesized compounds were identified as potent inhibitors against two kinds TS proteins with IC50 values of 0.47–1.4 µM [26].
Nowadays, dual antimicrobial-anticancer activity of 1,3,4-oxadiazole core structure has been an important concern. Ahsan et al. dealed with disubstituted derivatives of 1,3,4-oxadiazole with antimicrobial-anticancer activity capacity ,and synthesized analogs showed moderate to severe potency for this binary physiological topics [27]. In another research, Savariz and coworkers studied with 3,5-disubstituted-1,3,4-oxadiazole pharmacophore group, which were derived with different functional moieties via Mannich reaction. Results showed that both anticancer and antimicrobial activity of synthesized series have intermediate to excellent effect, and especially one of the compounds, which has a heterocyclic ring from third position of core structure, improved antitumor activity, which was found to be 4.5 timefold compared to the precursor molecule [28]. Under the shadow of previous experiences, Selvaraj and coworkers synthesized fifteen 1,3,4-oxadiazole derivatives, and they were found to have moderate to severe inhibitor effect against different microbial and cancer cell lines in 2017 [29].
Based on the statements above, to develop new, potent antimicrobial and anticancer agents, we aimed to synthesize a new series of 3,5-disubstituted-1,3,4-oxadiazole-2( 3H )-thione derivatives carrying different piperidine side chains. The compounds were evaluated for their antimicrobial and cytotoxicity profile to investigate the effect of molecular variations on activity against different bacteria, fungi, and yeasts. Further, compounds were tested in against various cancer cell lines for their cytotoxic activity.
2. Experimental
2.1. Materials and measurements
Melting points of compounds were checked by Mettler Toledo FP62 capillary melting point apparatus (Mettler-Toledo, Greifensee, Switzerland) and are uncorrected. Infrared spectral data were obtained by the use of Perkin-Elmer Spectrum One series FT-IR apparatus (Version 5.0.1) (Perkin Elmer, Norwalk, CT, USA), with potassium bromide pellets and the frequencies were presented in cm–1. The 1H-NMR spectra were checked via Varian Mercury-400 FT-NMR spectrometer (Varian, Palo Alto, CA, USA) using tetramethylsilane as the internal reference, with dimethyl Sulfoxide (DMSO-d6), as solvent, the chemical shifts were reported in parts per million (ppm), and coupling constants (J) were given in hertz (Hz). Elemental analyses were done by LECO 932 CHNS instrument (Leco-932, St. Joseph, MI, USA) and were within ± 0.4% of the theoretical values.
2.2. Chemistry
2.2.1. General procedure for the synthesis of 5-(3,4-dichlorophenyl)-1,3,4-oxadiazole-2(3H)-thione (4)
Solution of aroyl hydrazine (3.13 mmol) and carbon disulfide (6.27 mmol) in absolute ethanol (15 mL) were mixed in cold media (0 °C) and after the addition of potassium hydroxide (3.13 mmol) in one portion, the mixture was refluxed for 8 h. After the reaction was over, solvent was evaporated and the residue was acidified with 2M hydrochloric acid and extracted with ethyl acetate (2 × 20 mL). The organic layers were washed with water and dried with anhydrous sodium sulphate. Filtration and concentration in vacuo gave a solid, which was recrystallized from ethanol to give the compound [30].
2.2.2. General procedure for the synthesis of 5-(3,4-dichlorophenyl)-3-[(substitutedpiperidine)methyl]-1,3,4-oxadiazole-2(3H)-thione derivatives (5a-5k)
A mixture of 5-(3,4-dichlorophenyl)-1,3,4-oxadiazole-2(3 H )-thione (0.71 g, 3 mmol), an appropriate N-substituted amine (3 mmol) and 37% formaldehyde solution (1 mL) in ethanol (15 mL), was refluxed 3–5 h. The crude products were either precipitated or it was necessary to add water in case of not precipitated. The crude products were filtered, washed with water, dried, and crystallized from ethanol or ethanol/water.
2.2.2.1. 5-(3,4-Dichlorophenyl)-3-[(4-phenylpiperidin-1-yl)methyl]-1,3,4-oxadiazole-2(3H)-thione (5a)
White powder, yield 87%, Mp 182.0°C; FT-IR (KBr) νmax : 3075-3024 (Aromatic C-H), 1611 (C=N), 1435 (C=C), 1318 (C=S), 1238 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.07 (1H, s, phenyl H2), 7.79 (1H, d, J =10 Hz, phenyl H5), 7.61 (1H, bd, J = 8.8 Hz, phenyl H6), 7.34-7.19 (5H, m, phenyl H2’+H3’+H4’+H5’+H6’), 5.11 (2H, s, -C H 2-), 3.16 (2H, bd, J =11.6 Hz, piperidine H2), 2.65 (2H, t, J =10.8 Hz, piperidine H6), 1.76 (2H, bd, J =11.2 Hz, piperidine H3), 1.65 (1H, m, piperidine H4), 1.66–1.62 (2H, m, piperidine H5); C13-NMR (100 MHz, DMSO) δ 179.9 ( C =S), 159.8 ( C =N), 151.5 ( C 1 ’ , phenyl), 134.9 ( C 4 , phenyl), 132.4 ( C 3 , phenyl), 131.8 ( C 5 , phenyl), 128.7 ( C 3 ’+C 5 ’ , phenyl), 126.7 ( C 4 ’ , phenyl), 126.2 ( C 2 , phenyl), 126.1 ( C 6 , phenyl), 125.1 ( C 2 ’+C 6 ’ , phenyl), 122.1 ( C 1 , phenyl), 73.5 (N- C H2-N), 69.0 ( C 3 , piperidine), 46.2 ( C 1 +C 5 , piperidine), 37.7 ( C 2 +C 4 , piperidine); Anal. Calcd. for C20H19Cl2N3OS: C, 57.15; H, 4.56; N, 10.00; S, 7.63. Found: C, 57.32; H, 4.56; N, 10.14; S, 7.66.
2.2.2.2. 5-(3,4-Dichlorophenyl)-3-[(4-hydroxy-4-phenylpiperidin-1-yl)methyl]-1,3,4-oxadiazole-2(3H)-thione (5b)
White powder, yield 69.23%, Mp 182.1°C; FT-IR (KBr) νmax : 3456 (O-H), 3077 (Aromatic C-H), 1435 (C=N), 1417 (C=C), 1317 (C=S), 1235 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.06 (1H, s, phenyl H2), 7.86 (2H, d, J = 1.2 Hz, phenyl H5+H6), 7.46 (2H, d, J = 8 Hz, phenyl H2’+H6’), 7.36–7.18 (3H, m, phenyl H3’+H4’+H5’), 5.10 (2H, s, N-C H 2-N), 4.79 (1H, bs, O H ), 2.94 (4H, t, J = 9.6 Hz, piperidine H2+H6), 1.93-1.91 (2H, m, piperidine H5), 1.59 (2H, d, J = 12 Hz, piperidine H3,); C13-NMR (100 MHz, DMSO) δ 177.7 ( C =S), 156.6 ( C =N), 149.7 (phenyl C 1 ’ ), 134.9 (phenyl C 4 ), 132.4 (phenyl C 3 ), 131.8 (phenyl C 5 ), 127.7 (phenyl C 3 ’+C 5 ’ ), 127.6 (phenyl C 4 ’ ), 126.1 (phenyl C 6 ), 126.1 (phenyl C 2 ), 124.6 (phenyl C 2 ’+C 6 ’ ), 122.8 (phenyl C 1 ), 73.5 (piperidine C 3 ), 69.0 (N- C H2-N), 46.2 (piperidine C 1 +C 5 ), 37.7 (piperidine C 2 +C 4 ); Anal. Calcd. for C20H19Cl2N3O2S: C, 55.05; H, 4.39; N, 9.63; S, 7.35. Found: C, 54.85; H, 4.27; N, 9.72; S, 7.42.
2.2.2.3. 5-(3,4-Dichlorophenyl)-3-[(4-acetyl-4-phenylpiperidin-1-yl)methyl]-1,3,4-oxadiazole-2(3H)-thione (5c)
White powder, yield 52.48%, Mp 149.8°C; FT-IR (KBr) νmax : 2924-2831 (Aliphatic C-H), 1698 (C=O), 1606 (C=N), 1422 (C=C), 1330 (C=S), 1244 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.01 (1H, d, J = 1.6 Hz, phenyl H2), 7.86–7.82 (2H, m, phenyl H5+H6), 7.38–7.31 (5H, m, aromatic H2’+H3’+H4’+H5’+H6’), 5.01 (2H, s, N-C H 2-N), 2.92 (2H, d, J = 12.4 Hz, piperidine H2), 2.63 (2H, t, J =10.8 Hz, piperidine H6), 2.43 (2H, d, J = 11.2 Hz, piperidine H5), 1.97–1.89 (2H, m, piperidine H3), 1.84 (3H, s, CO-C H 3); C13-NMR (100 MHz, DMSO) δ 208.6 ( C =O), 177.7 ( C =S), 156.8 ( C =N), 141.2 (phenyl C 1 ’ ), 134.8 (phenyl C 4 ), 132.4 (phenyl C 3 ), 131.8 (phenyl C 5 ), 128.7 (phenyl C 3 ’ +C 5 ’ ), 127.6 (phenyl C 4 ’ ), 127.0 (phenyl C 6 ), 126.2 (phenyl C 2 +C 6 ), 126.1 (phenyl C 2 ), 122.9 (phenyl C 1 ), 70.5 (N- C H2-N), 53.5 (piperidine C 3 ), 47.4 (piperidine C 1 +C 5 ), 32.1 (piperidine C 2 +C 4 ), 25.4 (CO C H3); Anal. Calcd. for C22H21Cl2N3O2S: C, 57.15; H, 4.58; N, 9.09; S, 6.93. Found: C, 57.35; H, 4.40; N, 9.03; S, 6.55.
2.2.2.4. 5-(3,4-Dichlorophenyl)-3-[(4-cyano-4-phenylpiperidin-1-yl)methyl]-1,3,4-oxadiazole-2(3H)-thione (5d)
White crystals Yield 47.68%, , Mp 168.7°C; FT-IR (KBr) νmax : 3091 (Aromatic C-H), 2931 (Aliphatic C-H), 2238 (CºN), 1607 (C=N), 1432–1415 (C=C), 1322 (C=S), 1249 (C-O-C) cm–1; 1H NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.10 (1H, s, phenyl H2), 7.89 (2H, d, J = 0.8 Hz, phenyl H5+H6), 7.53 (2H, d, J = 8.0 Hz, phenyl H2’+H6’), 7.44 (2H, t, J = 7.6 Hz, phenyl H3’+H5’), 7.37 (1H, t, J = 7.6 Hz, phenyl H4’), 5.13 (2H, s, N-C H 2-N), 3.25 (2H, d, J = 12.4 Hz, piperidine H2), 2.87 (2H, t, J = 10.8 Hz, piperidine H5), 2.16 (2H, d, J = 12.4 Hz, piperidine H6), 2.03 (2H, t, J = 12.8 Hz, piperidine H3); C13-NMR (100 MHz, DMSO) δ 177.5 ( C =S), 156.8 ( C =N), 139.9 (phenyl C 1 ’), 135.1 (phenyl C 4 ), 132.4 (phenyl C 3 ), 131.8 (phenyl C 5 ), 129.0 (phenyl C 3 ’ +C 5 ’ ), 128.0 (phenyl C 4 ’ ), 127.7 (phenyl C 6 ), 126.2 (phenyl C 2 ), 125.5 (phenyl C 2 ’ +C 6 ’ ), 122.6 (phenyl C 1 ), 121.6 ( C ≡N), 70.1 (N- C H2-N), 47.5 (piperidine C 1 +C 5 ), 41.3 (piperidine C 3 ), 35.3 (piperidine C 2 + C 4 ); Anal. Calcd. for C21H18Cl2N4OS: C, 56.63; H, 4.07; N, 12.58; S, 7.20. Found: C, 56.54; H, 4.20; N, 12.57; S, 7.24.
2.2.2.5. 3-[(4-Benzylpiperidin-1-yl)methyl]-5-(3,4-dichlorophenyl)-1,3,4-oxadiazole-2(3H)-thione (5e)
White crystals, Yield 56.92%, Mp 94.8°C; FT-IR (KBr) νmax : 3024 (Aromatic C-H), 2914 (Aliphatic C-H), 1617 (C=N), 1436-1418 (C=C), 1318 (C=S), 1233 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ =8.04 (1H, s, phenyl H2), 7.84 (2H, d, J = 1.6 Hz, phenyl H5+H6), 7.25 (2H, t, J = 7.2 Hz, phenyl H2’+H6’), 7.14–7.11 (3H, m, phenyl H3’, H4’, H5’), 5.03 (2H, s, N-C H 2-N), 3.01 (2H, d, J = 11.6 Hz, piperidine H2), 2.45 (2H, d, J = 6.8 Hz, N-C H 2-Phenyl), 2.43 (2H, d, J = 11.6 Hz, piperidine H6), 1.54 (2H, d, J = 12 Hz, piperidine H3), 1.44–1.40 (1H, m, piperidine H4), 1.17 (2H, q, J = 10.6 Hz, piperidine H5); C13-NMR (100 MHz, DMSO) δ 177.8 ( C =S), 156.8 ( C =N), 140.1 (phenyl C 1 ’), 134.5 (phenyl C 4 ), 132.4 (phenyl C 3 ), 131.7 (phenyl C 5 ), 128.8 (phenyl C 3 ’ +C 5 ’), 128.0 (phenyl C 2 ’ +C 6 ’), 127.5 (phenyl C 2 ), 126.0 (phenyl C 4 ’), 125.7 (phenyl C 6 ), 123.2 (phenyl C 1 ), 71.0 (N- C H2-N), 50.0 (piperidine C 1 +C 5 ), 42.1 (N- C H2-Phenyl), 36.5 (piperidine C 3 ), 31.5 (piperidine C 2 +C 4 ); Anal. Calcd. for C21H21Cl2N3OS: C, 58.07; H, 4.87; N, 9.67; S, 7.38. Found: C, 58.24; H, 4.91; N, 9.85; S, 7.55.
2.2.2.6. 5-(3,4-Dichlorophenyl)-3-{[4-(morpholin-4-yl)piperidin-1-yl]methyl}-1,3,4-oxadiazole-2(3H)-thione (5f)
White powder, yield 59.55%, Mp 156.8°C; FT-IR (KBr) νmax : 2938 (Aromatic C-H), 1610 (C=N), 1448 (C=C), 1326 (C=S), 1246 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.04 (1H, s, phenyl H2), 7.84 (2H, d, J = 2 Hz, phenyl H5+H6), 5.02 (2H, s, N-C H 2-N), 3.58 (5H, bs, morpholine C H 2O, piperidine H4), 3.33 (5H, bs, morpholine NC H 2, piperidine H2), 3.09 (2H, d, J = 11.2 Hz, piperidine H6), 1.79 (2H, d, J = 11.2 Hz, piperidine H3), 1.42 (2H, bs, piperidine H5); C13-NMR (100 MHz, DMSO) δ 178.3 ( C =S), 157.2 ( C =N), 132.1 (phenyl C 4 ), 131.6 (phenyl C 3 ), 127.2 (phenyl C 5 ), 125.8 (phenyl C 6 ), 123.5 (phenyl C 2 ), 122.0 (phenyl C 1 ), 70.4 (N- C H2-N), 65.7 (morpholine C 2 +C 3 ), 63.0 (piperidine C 3 ), 60.8 (morpholine C 1 +C 4 ), 49.0 (piperidine C 1 +C 5 ), 27.1 (piperidine C 2 +C 4 ); Anal. Calcd. for C18H22Cl2N4O2S: C, 50.35: H, 5.16: N, 13.05; S, 7.47. Found: C, 49.61; H, 4.81; N, 12.92; S, 7.75.
2.2.2.7. 1-{[5-(3,4-Dichlorophenyl)-2-thioxo-1,3,4-oxadiazol-3(2H)-yl]methyl}piperidine-4-carboxylic acid (5g)
White powder, yield 49.55%, Mp 181.3°C; FT-IR (KBr) νmax : 3424 (O-H), 3042 (Aromatic C-H) 2941 (Aliphatic C-H), 1725 (C=O), 1554 (C=N), 1441–1419 (C=C), 1320 (C=S), 1239 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.01 (1H, s, phenyl H2), 7.82 (2H, s, phenyl H5+H6), 5.04 (2H, s, N-C H 2 - N), 3.00 (4H, t, J = 11.4 Hz, piperidine H2+H6), 2.00 (1H, d, J = 12.0 Hz, piperidine H4,), 1.82 (2H, d, J = 12.0 Hz, piperidine H3), 1.60–1.51 (2H, m, piperidine H5); C13-NMR (100 MHz, DMSO) δ 179.1 ( C =O), 175.7 ( C =S), 157.7 ( C =N), 133.4 (phenyl C 4 ), 132.1 (phenyl C 3 ), 131.6 (phenyl C 2 +C 6 ), 126.9 (phenyl C 5 ), 125.5 (phenyl C 1 ), 70.6 (N- C H2-N), 49.9 (piperidine C 1 +C 5 ), 42.5 (piperidine C 3 ), 27.6 (piperidine C 2 + C 4 ); Anal. Calcd. for C15H15Cl2N3O3S: C, 46.40; H, 3.89; N, 10.82; S, 8.26. Found: C, 47.42; H, 4.49; N, 10.88; S, 7.75.
2.2.2.8. 1-{[5-(3,4-Dichlorophenyl)-2-thioxo-1,3,4-oxadiazol-3(2H)-yl]methyl}piperi-dine-3-carboxylic acid (5h)
White powder, yield 70.83%, Mp 181.7°C; FT-IR (KBr) νmax : 3421 (O-H), 2934 (Aromatic C-H), 1710 (C=O), 1609 (C=N), 1437–1414 (C=C), 1328 (C=S), 1235 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 12.25 (1H, bs, COO H ), 8.05 (1H, t, J = 0.8 Hz, phenyl H2), 7.87–7.85 (2H, m, phenyl H5+H6), 5.06 (2H, s, N-C H 2 -N), 3.34 (1H, bs, piperidine H3), 3.13 (1H, dd, J = 11.2 Hz, J’ = 3.6 Hz piperidine H2), 2.91 (1H, dd, J = 11.2 Hz, J’ = 3.6 Hz, piperidine H2’), 2.66 (2H, t, J = 10.0 Hz, piperidine H6), 1.78–1.63 (2H, m, piperidine H4), 1.47-1.31 (2H, m, piperidine H5); C13-NMR (100 MHz, DMSO) δ 177.4 ( C =O), 174.6 ( C =S), 156.6 ( C =N), 134.9 (phenyl C 4 ), 132.3 (phenyl C 3 ), 131.7 (phenyl C 5 ), 127.7 (phenyl C 6 ), 126.2 (phenyl C 2 ), 122.8 (phenyl C 1 ), 70.8 (N- C H2-N), 52.1 (piperidine C 1 ), 50.0 (piperidine C 5 ), 41.0 (piperidine C 2 ), 25.7 (piperidine C 3 ), 23.9 (piperidine C 4 ); Anal. Calcd. for C15H15Cl2N3O3S: C, 46.40; H, 3.89; N, 10.82; S, 8.26. Found: C, 46.15; H, 3.88; N, 10.86; S, 8.53.
2.2.2.9. Ethyl 1-{[5-(3,4-dichlorophenyl)-2-thioxo-1,3,4-oxadiazol-3(2H)-yl]methyl}piperidine-4-carboxylate (5i)
White crystals, yield 85.13%, Mp 163.8°C; IR (KBr) νmax : 2950 (Aromatic C-H), 1720 (C=O), 1611 (C=N), 1443–1421 (C=C), 1324 (C=S), 1241 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.06 (1H, s, phenyl H2), 7.86 (2H, s, phenyl H5+H6), 5.04 (2H, s, N-C H 2-N), 4.03 (2H, q, J = 6.8 Hz, COO-C H 2-CH3), 3.02 (2H, d, J = 12 Hz, piperidine H2), 2.56 (2H, d, J = 10.8 Hz, piperidine H6), 2.24-2.21 (1H, m, piperidine H4), 1.81 (2H, d, J = 10.4 Hz, piperidine H3), 1.57–1.52 (2H, m, piperidine H5), 1.15 (3H, t, J = 6.8 Hz, COO-CH2-C H 3); C13-NMR (100 MHz, DMSO) δ 177.5 ( C =O), 174.0 ( C =S), 156.6 ( C =N), 134.9 (phenyl C 4 ), 132.3 (phenyl C 3 ), 131.7 (phenyl C 5 ), 127.7 (phenyl C 6 ), 126.2 (phenyl C 2 ), 122.8 (phenyl C 1 ), 70.8 (N- C H2-N), 59.7 (O- C H2CH3), 49.1 (piperidine C 1 + C 3 + C 5 ), 28.7 (piperidine C 2 + C 4 ), 13.9 (O-CH2 C H3); Anal. Calcd. for C17H19Cl2N3O3S: C, 49.04; H, 4.60; N, 10.09; S, 7.70. Found: C, 48.64; H, 4.53; N, 10.00; S, 8.72.
2.2.2.10. Ethyl 1-{[5-(3,4-dichlorophenyl)-2-thioxo-1,3,4-oxadiazol-3(2H)-yl]methyl}piperidine-3-carboxylate (5j)
White crystals, yield 75.51%, Mp 132.9°C; FT-IR (KBr) νmax : 2934 (Aromatic C-H), 1727 (C=O), 1607 (C=N), 1414 (C=C), 1335 (C=S), 1180 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.06 (1H, d, J = 1.6 Hz, phenyl H2), 7.87 (2H, t, J = 1.6 Hz, phenyl H5+H6), 5.05 (2H, s, N-C H 2-N), 4.06 (2H, q, J = 7.2 Hz, COO-C H 2CH3), 3.12 (1H, dd, J = 11.2 Hz, J’ = 3.2 Hz, piperidine H2), 2.92-2.87 (1H, m, piperidine H2’), 2.72 (1H, t, J = 11.2 Hz, piperidine H6), 2.59–2.52 (2H, m, piperidine-H6’+H4), 1.76–1.63 (2H, m, piperidine H3), 1.47–1.35 (2H, m, piperidine H5), 1.17 (3H, t, J = 7.2 Hz, COO-CH2C H 3); C13-NMR (100 MHz, DMSO) δ 177.5 ( C =O), 172.8 ( C =S), 156.7 ( C =N), 134.9 (phenyl C 4 ), 132.3 (phenyl C 3 ), 131.7 (phenyl C 5 ), 127.6 (phenyl C 6 ), 126.1 (phenyl C 2 ), 122.8 (phenyl C 1 ), 70.7 (N- C H2-N), 59.7 (O- C H2CH3), 51.9 ( C 1 , piperidine), 49.9 ( C 5 , piperidine), 40.9 ( C 2 , piperidine), 25.5 ( C 3 , piperidine), 23.7 ( C 4 , piperidine), 13.9 (O-CH2 C H3); Anal. Calcd. for C17H19Cl2N3O3S: C, 49.04; H, 4.60; N, 10.09; S, 7.70. Found: C, 48.64; H, 3.94; N, 9.99; S, 6.80.
2.2.2.11. Ethyl 1-{[5-(3,4-dichlorophenyl)-2-thioxo-1,3,4-oxadiazol-3(2H)-yl]methyl}piperidine-2-carboxylate (5k)
White crystals, yield 32.05%, Mp 113.6°C; FT-IR (KBr) νmax 2938 (Aromatic C-H), 1730 (C=O), 1448-1414 (C=C), 1321 (C=S), 1187 (C-O-C) cm–1; 1H-NMR (DMSO- d 6 , 400 MHz) ppm: δ = 8.04 (1H, d, J = 2.0 Hz, phenyl H2), 7.89 (1H, d, J = 8.0 Hz, phenyl H6), 7.84 (1H, dd, J = 8.8 Hz, J’ = 2.0 Hz phenyl H5), 5.13 (2H, s, N-C H 2-N), 4.09–4.02 (2H, m, COO-C H 2-CH3), 3.69 (1H, t, J = 6.0 Hz, piperidine H2), 3.24–3.21 (2H, m, piperidine H6), 1.77–1.67 (2H, m, piperidine H3), 1.51-1.48 (2H, m, piperidine H5), 1.40–1.30 (2H, m, piperidine H4), 1.18 (3H, t, J = 6.8 Hz, COO-CH2-C H 3 ); C13-NMR (100 MHz, DMSO) δ 177.1 ( C =O), 172.5 ( C =S), 156.3 ( C =N), 134.9 (phenyl C 4 ), 132.3 (phenyl C 3 ), 131.8 (phenyl C 5 ), 127.5 (phenyl C 6 ), 126.0 (phenyl C 2 ), 122.7 (phenyl C 1 ), 68.9 (N- C H2-N), 59.9 (O- C H2CH3), 48.2 (piperidine C 1 + C 5 ), 29.0 (piperidine C 2 ), 24.8 (piperidine C 4 ), 20.9 (piperidine C 3 ), 13.9 (O-CH2 C H3); Anal. Calcd. for C17H19Cl2N3O3S: C, 49.04; H, 4.60; N, 10.09; S, 7.70. Found: C, 49.13; H, 4.72; N, 10.20; S, 7.74.”
2.3. Biological assays
2.3.1. Antimicrobial activity
2.3.1.1. Disc diffusion method
Dimethylsulfoxide (DMSO) was used to dissolve and prepare the synthesized compounds with a concentration of 10 mg mL–1. The lyophilized compounds sterilized by filtration via 0.45 mm millipore filters. Disc diffusion method was performed by using 100 mL of suspension containing 108 colony forming units (CFU) mL–1 of bacteria, 106 CFU mL–1 of yeast and 104 spore mL–1 of fungi spread on nutrient agar (NA), sabour dextrose agar (SDA), and potato dextrose agar (PDA) medium, in sequence. A total of 15 mL of each synthesized compounds (300 mg/disc) at the concentration of 10 mg mL–1 were impregnated to the discs (6 mm in diameter). DMSO impregnated discs were used for negative controls. The compounds and negative controls were located in the inoculated agar. In order to determine the sensitivity of one strain/isolate standard, ofloxacin and nystatin were used as positive references for bacterial and fungus-yeast strains, respectively. The incubation at 37°C of inoculated plates took 24 h for bacterial strains, 48 h for yeast and 72 h for fungi isolates. The incubation of plant related microorganisms were held at 27°C, differently [31].
2.3.2. Cytotoxic activity
The human cancer cell lines were grown in Dulbecco’s Modified Eagle’s Medium (DMEM), with 10% fetal bovine serum (FBS) and 1% penicillin. They were incubated in 37 °C incubators containing 5% CO2 and 95% air. Cancer cells (range of 2000 cells/well to 5000 cells/well) were inoculated into 96-well plates in 200 μL of media and incubated in 37 °C incubators containing 5% CO2 and 95% air. After a 24 h incubation period, one plate for each cell line was fixed with 100 μL of 10% ice-cold trichloroacetic acid (TCA). This plate represents the behavior of the cells just prior to compound treatment and is accepted as the time-zero plate. The compounds to be tested were solubilized in dimethyl sulfoxide (DMSO) to a final concentration of 40 mM and stored at +4°C. While treating the cells with the compounds, the corresponding volume of the compound was applied to the cell to achieve the desired drug concentration and diluted through serial dilution (40, 20, 10, 5, 2.5 µM). After drug treatment, the cells were incubated in 37 °C incubators containing 5% CO2 and 95% air for 72 h. Following the termination of the incubation period after drug treatment, the cells were fixed with 100 μL 10% ice-cold TCA and incubated in the dark at +4°C for 1 h. Then, the TCA was washed away with ddH2O five times and the plates were left to air dry. In the final step, the plates were stained with 100 μL of 0.4% SRB (cat.86183-5 g from Sigma) solution in 1% acetic acid solution. Following staining, the plates were incubated in dark for 10 min at room temperature. The unbound dye was washed away using 1% acetic acid and the plates were left to air dry. To measure the absorbance results, the bound stain was then solubilized using 200 μL of 10 mM Tris-Base. Camptothecin was the positive control and 5-Fluorouracil (5-FU) was standard drug for the cytotoxic effect. The OD values were obtained at 515nm [32].
2.4. In silico chemo-informatic and toxicity measurements
For determination of drug-like physicochemical, pharmacokinetics, and toxicity parameters, a combination of various online screening tools were used, which included MedChem Designer 5.5 (MedChem Designer, version 5.5.0.112019), Chem&BioDraw 12.0 (ChemDraw version 12.0.2.10762019), SwissAdme (Swiss Institute of Bioinformatics2013_http://www.swissadme.ch/). Toxicity prediction of these newly synthesized compound 5a-5k series were retrieved from Lazar software (Version 1.4.2) which is a web-based application as https://lazar.in-silico.ch [33].
3. Results and discussion
3.1. Chemistry
5-(3,4-Dichlorophenyl)-3-[(substitutedpiperidine)methyl]-1,3,4-oxadiazole-2( 3H )-thione derivatives ( 5a-5k ) were prepared via Mannich reaction. According to chemical procedure of piperidine, derivatives were reacted with (3,4-dichlorophenyl)-1,3,4-oxadiazole-2( 3H )-thione ( 4 ) group in alcoholic media (Figure). Compounds 4 , 5a-5k were characterized by FT-IR, 1H NMR, 13C NMR spectroscopy, and purity of compounds were checked with elemental analysis. All results of spectral and elemental analysis were found compatible with literature data [34–37]. Compounds 4, 5a-5k were tested for their antimicrobial and cytotoxic properties.
Figure.
Synthesis of (3-4-dichlorophenyl)-1,3,4-oxadiazole-2(3H)-thione derivatives.
The FT-IR spectrum of compounds displayed a strong band in range of 3080–2900 cm-1 which assigned to aromatic carbon-hydrogen sp2 hybridizations in common for all compounds. Imine (C=N) and thione (C=S) groups in 1,3,4-oxadiazole-2( 3H )-thione structure, generated two characteristic signals approximately at 1610 and 1330 cm–1. Compounds 5c, 5g-5k had an extra sharp signal around 1740–1680 cm–1, which corresponded to the carbonyl group, and compound 5d showed a spesific band at 2238 cm–1, which was claimed as nitrile group.
1H NMR spectra of compounds 4, 5a-5k demonstrated hydrogen signals of aromatic structures in the range of 8.00–7.00 ppm. Two proton integrationed and singlet coupled signal in the range of 5.13–5.01 ppm values was a strong evidence for methylene bridge protons between 1,3,4-oxadiazole and piperidine moieties which obtained via Mannich reaction procedure. Variable but compatible integrated signals between 3.60–1.30 ppm confirmed different piperidine protons for each compound. Compound 5c have an acetyl group and preserved a singlet signal in 1.84 ppm for alpha protons. Signals for compounds 5i-5k , which have different positioned ethyl ester groups on piperidine moiety emerged at 4.06–4.02 for methylene protons (-COOC H 2-) and 1.18–1.15 ppm values for methyl protons (-COOCH2C H 3). Integration and multiplicity of signals for all compounds were compatible with literature data.
13C NMR spectra of compounds 4, 5a-5k preserved two characteristic signals related to thione ( C =S) and imine ( C =N) groups at 179 and 158 ppm values originated from 1,3,4-oxadiazole-2( 3H )-thione structure. Carbon signal of ketone carbonyl in compound 5b , carboxy and ester carbonyl in compounds 5g-5k were appeared at different ppm values due to different chemical environments of carbonyl functional groups. While ketone carbonyl carbon of 5c showed signal at 208 ppm, carboxy and ester carbonyl carbons of compound 5g-5k indicated their carbonyl carbon signals in the range of 179–174 ppm. Due to shielded-deshielded properties of carbon atoms in magnetic field of 13C NMR, signal of ketone carbonyl in 5c occured in downfield region while carboxylic acid ( 5g, 5h ) and ester ( 5i-5k ) carbonyl carbons shifted through upfield part of the scale. In this direction, moderately shielded aromatic structures in compounds 4, 5a-k gave their spesific signals in the range of 138–122 ppm. 13C NMR signals of remaining carbons associated with methylene and piperidine groups were observed at 70–65 and 69–32 ppm.
3.2 Biological evaluation
3.2.1. Antimicrobial activity
Antimicrobial activity was tested by measuring the zone of inhibition against test organisms with disc diffusion assay method, and results were summarized in Table 1 and Table 2 with positive control ofloxacin. Eleven compounds were screened for their antibacterial activity against three gram-negative ( E. coli, P. aeruginosa, P. vulgaris ) and sixteen gram-positive bacterial strains. ( Staphylococcus spp, Micrococcus spp, Bacillus spp ). They were also evaluated for their antifungal potential against six fungal strains ( Aspergillus spp, F. oxysporium, B. cinerea, Penicillium, Candida spp ) and antiyeast activity against three yeast strains ( K. marxianus, P. membranaefaciens, S. occidentalis ). Ofloxacin and nystatin were used as positive controls. Antimicrobial data of compounds and reference drugs were given in Tables 1 and Table 2.
Table 1.
Antibacterial activities of newly synthesized compounds.
| Test microorganisms | 4 | 5a | 5b | 5c | 5d | 5e | 5f | 5g | 5h | 5i | 5j | 5k | Ofloxacin | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Escherichia coli | - | - | - | - | - | - | - | - | - | - | - | - | 22 | Zone of inhibition in mm |
| Pseudomonas aeruginosa | - | - | - | - | - | - | - | - | 8 | - | - | - | 22 | |
| Pseudomonas vulgaris | 14 | 14 | 12 | 12 | 10 | 14 | 10 | 10 | 12 | 12 | 14 | 14 | 23 | |
| Staphylococcus aureus | 10 | 10 | 12 | 10 | 12 | 14 | 14 | 12 | 12 | 12 | 14 | 12 | 22 | |
| Staphylococcus cohnii | 12 | 10 | 20 | 14 | 14 | 11 | 14 | 14 | 14 | 12 | 14 | 12 | 24 | |
| Micrococcus lylae | 14 | 12 | 10 | 10 | 10 | 10 | 10 | 11 | 12 | 11 | 11 | 9 | 13 | |
| Micrococcus luteus | 14 | 9 | 12 | 9 | 9 | 10 | 9 | 12 | 12 | 9 | 10 | 9 | 13 | |
| Bacillus megaterium | 14 | 12 | 15 | 14 | 11 | 12 | 16 | 14 | 15 | 16 | 12 | 12 | 26 | |
| Bacillus lentimorbus | 24 | 24 | 22 | 20 | 18 | 16 | 14 | 18 | 20 | 16 | 18 | 16 | 26 | |
| Bacillus subtilis | 15 | 15 | 12 | 12 | 12 | 17 | 15 | 14 | 17 | 16 | 15 | 15 | 21 | |
| Bacillus licheniformis | 22 | 13 | 20 | 15 | 14 | 13 | 14 | 17 | 14 | 13 | 14 | 13 | 25 | |
| Bacillus pumilus | 20 | 10 | 15 | 14 | 14 | 15 | 18 | 20 | 19 | 13 | 16 | 12 | 20 | |
| Bacillus mycoides | 20 | 12 | 23 | 20 | 15 | 17 | 20 | 20 | 22 | 15 | 19 | 19 | 25 | |
| Bacillus cereus | 14 | 12 | 16 | 16 | 14 | 14 | 15 | 15 | 16 | 14 | 14 | 13 | 15 | |
| Bacillus ehimensis | 30 | 20 | 32 | 24 | 18 | 20 | 30 | 26 | 28 | 24 | 28 | 22 | 21 | |
| Bacillus thuringiensis | 14 | 13 | 18 | 15 | 14 | 12 | 13 | 15 | 16 | 12 | 12 | 11 | 14 | |
| Bacillus sphaericus | 9 | - | 11 | 8 | 8 | 10 | 10 | - | 10 | 8 | 10 | 10 | 17 | |
| Bacillus marinus | 24 | 15 | 22 | 12 | 14 | 20 | 20 | 22 | 22 | 20 | 24 | 20 | 30 | |
| Bacillus laevolacticus | 26 | 12 | 21 | 14 | 15 | 17 | 20 | 17 | 20 | 13 | 19 | 17 | 30 | |
Table 2.
Antifungal and antiyeast activities of newly synthesized compounds.
| Test microorganisms | 4 | 5a | 5b | 5c | 5d | 5e | 5f | 5g | 5h | 5i | 5j | 5k | Nystatin | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Aspergillus spp | 9 | - | - | - | - | - | - | - | - | - | - | - | 12 | Zone of inhibition in mm |
| Fusarium oxysporium | 12 | 8 | 10 | 9 | 8 | 9 | 9 | 10 | 13 | 8 | 12 | 9 | 14 | |
| Botrytis cinerea | 17 | 12 | 15 | 10 | 9 | 10 | 10 | 11 | 12 | 10 | 10 | 12 | 25 | |
| Penicillium spp | 19 | - | 12 | - | - | 1 | - | 14 | 16 | - | 13 | 11 | 14 | |
| Candida albicans | 14 | 10 | 16 | 14 | 12 | 14 | 16 | 14 | 16 | 14 | 12 | 12 | 20 | |
| Candida parapsilosis | 14 | 8 | 14 | 14 | 12 | 12 | 11 | 12 | 14 | 11 | 13 | 12 | 20 | |
| Kluyveromyces marxianus | 12 | 11 | 14 | 12 | 11 | 11 | 12 | 14 | 16 | 14 | 15 | 15 | 20 | |
| Pichia membranaefaciens | 15 | 12 | 18 | 12 | 12 | 16 | 16 | 18 | 18 | 13 | 15 | 15 | 18 | |
| Schwanniomyces occidentalis | 17 | 10 | 20 | 14 | 10 | 14 | 16 | 13 | 20 | 12 | 14 | 14 | 20 | |
In vitro disc diffusion test was carried out to evaluate newly synthesized compounds ( 4, 5a-k ) for their antibacterial activities towards pathogenic gram-positive and gram-negative bacteria, and ofloxacin was used as positive control under the same conditions. As shown in Table 1, compound series displayed serious inhibition of growth (mm) in certain bacterial strains. Especially compounds 5b, 5c, 5f-5k showed considerable antibacterial activity against gram-positive Bacillus spp . when compared to ofloxacin, while inhibition of growth values (mm) of other compounds in the series were either equal or lower than reference molecule. In vitro antibacterial screening results also revealed that, for all bacterial strains, lower inhibition values than reference material meaned as an indicator of antibacterial inability especially for compounds 5a, 5d and 5e (Table 1). None of compounds didn’t show any antibacterial activity against E.coli and P. Aeruginosa but compounds 5b, 5f, 5g, 5h and 5j showed statistically significant antibacterial activity against B. ehimensis when they compared with ofloxacin.
Chemical nature of substitution pattern related to piperidine derivatives of 5-(3,4-dichlorophenyl)-1,3,4-oxadiazole-2( 3H )-thione compounds was important point to establish biological activity-functional group relationship. Therefore, newly synthesized molecules were designed to emphasize this correlation. As a comparable result, compounds 5a, 5b, 5c and 5e , which have a common phenyl group, showed different biological responses for B. ehimensis strains. Other substituents and phenyl groups that were located on the fourth position piperidine moiety had significant functionality differences for activity. Based on above part, strong electron-donating hydroxyl containing compound 5b , weak electron-withdrawing acetyl containing 5c , strong electron-withdrawing nitrile containing 5d , and only phenyl substituted piperidine containing compound 5a were good examples to elucidate this phenomenon. According to structure-activity relationship (SAR) studies in literature , electron-donating groups provide an elevation of inhibitor level against spesific bacterial strains [38]. The results obtained in parallel with this information was compound 5b , which had high level of antimicrobial property specifically against B. ehimensis due to its electron-donating hydroxyl moiety. In addition of these four compounds ( 5a-d ), morpholine containing compound 5f was also found more active than reference drug on B. ehimensis strain. Heteroatoms in morpholine and their bonding capasities with active site of bacteria were serious indicators for biologic activity of 5f , which was an open point for further studies on heterocyclic structure substituted piperidine rings. Besides, substituent variation and their effects on biologic acitiy, structural design was modulated also to generate position effect on activity. So, on piperidine moiety, differently located carboxylic acid containing compounds 5g, 5h and differently positioned ethyl ester containing compounds 5i , 5j and 5k were added to series. Results clearly claimed that position differences of one substituent on piperidine did not cause a significant difference for their antimicrobial response (Table 1).
In vitro antimicrobial activity of compounds 4, 5a-k were further assessed in terms of antifungal and antiyeast activities relative to nystatin according to disc diffusion assay method . The results were presented in Table 2 and a review of data revealed that all compounds were possessed moderate activities against Candida spp and no inhibition against Aspergillus spp except compound 4 . The best and comparable results were obtained against Penicillium sp. and F. oxysporium for compounds 4, 5g, and 5h . According to antiyeast activity profile, compounds showed weak to moderate activity against K. Marxianus wherease compounds 5b, 5g and 5h were equipotent against P. membranaefaciens and S. occidentalis when they were compared with nystatin. Also compound 5b , which has strong electron-donating hydroxyl group showed the best activity against all fungal and yeast strains according to other phenyl containing compounds 5a, 5c, and 5d . This unclear activity profile and lipophilicity relationship might be seen as described in previous study [39] . Due to the consistent results generated from antibacterial, antifungal and antiyeast activities, synthesized compounds were further analysed for their cytotoxic activities on certain cancer cell lines.
3.2.2. Cytotoxicity study
All synthesized target compounds 5a-5k were screened for their cytotoxic activity against three cancer cell lines: colon (HCT116), breast (MCF7), and liver (HUH7) with sulphorhodamine B (SRB) assay in triplicate aplication where 5-flourouracil (5-FU) was used as positive control. The IC50 values obtained for these compounds were shown in Table 3.
Table 3.
IC50 value for tested compounds 5a-5k against cancer cell lines.
| 5a | 5b | 5c | 5d | 5e | 5f | 5g | 5h | 5i | 5j | 5k | 5-FU | ||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| HCT116* | NI | NI | NI | 18.2 | 33.3 | 47.9 | 44.1 | 80.4 | NI | NI | 13.9 | 30.7 | IC50 (µM) |
| MCF7* | NI | 40.9 | NI | NI | 26.8 | NI | 29.0 | 37.4 | NI | 45.0 | 25.2 | 3.5 | |
| HUH7* | NI | 27.5 | 11.8 | 10.1 | 18.3 | 16.4 | 14.0 | 34.6 | 17.8 | 15.2 | 11.9 | 18.78 | |
*All the experiments were conducted in triplicate (1 < R2 < 0.8). NI: no inhibition.
The results of cytotoxicity studies revealed that activities of the compounds were not impressive against colon and breast cancer cells, all of the compounds showed cell viability with IC50 values ranging from 13.9–80.4 µM concentrations. It was noteworthy that the cytotoxic effects were more pronounced against liver carcinoma cell line, HUH7. Most of the compounds of the series ( 5c-5g and 5i-5j ) have better IC50 values than 5-FLU (IC50 = 18.78 µM) and also compound 5d possessed 10.1 µM value, which represents good druggable cytotoxic activity.
Our results indicate that the addition of phenyl and carbonyl group as substituents enhances the antimicrobial activity of the prepared compounds. It indicates that the structural differences is an important factor for the activity. It is notable, also, that phenyl and carbonyl substitutions in compounds 5c, 5d, 5f, 5g, 5i-5k lead to not only to an increase in the antimicrobial activity on certain bacterial and fungal strains, but also to a significant level of anticancer activity against liver carcinoma cell line (HUH7). According to the preliminary antimicrobial activity, compound 5b showed the best inhibitor activity against Bacillus spp strains, and compound 5d was evaluated to have strongest cytotoxic activity against liver carcinoma cells (HUH7). A total analysis of the antibacterial, antifungal, and antiyeast activity revealed that newly synthesized compounds were really active against Bacillus cereus , Bacillus ehimensis and Bacillus thuringiensis species .
3.3. In silico chemo-informatic and toxicity measurements
The chemo-informatic features of newly synthesized molecules were evaluated by computational tools. According to in silico data, compounds 5a-5k showed acceptable consequences for Lipinski’s rule of five (RO5) analysis; molecular weight (MW) (<500 dalton), hydrogen bond acceptor (HBA) (<10), hydrogen bond donor (HBD) (<5) and logP (<5) values [40]. In the situation of one deviation, corresponds to poor absorption of compounds. However, there are plenty of examples are available for RO5 violation amongst the existing drugs [41,42]. Furthermore, the polar surface area (PSA) of a molecule is defined as the surface sum over all polar atoms, primarily oxygen and nitrogen with their attached hydrogen atoms. The PSA value of a molecule reflects the ability to permeate cells, which is used for drug’s optimization. Previous researches showed the standard value of PSA as <89 A2 [43] in which this measure is supported by our newly synthesized compound 5a-5k serie. On the other hand, number of rotatable bond is a measurement for molecular flexibility and is significant in determining oral bioavailability of the compounds (rule of three-number of rotatable bonds ≤ 3), which explains oral usage of compound 5a-5k might be decrease bioavailibility (Table 4) [44].
Table 4.
Chemo-informatic data of compound 5a-5k.
![]() | ||||||||
|---|---|---|---|---|---|---|---|---|
| Compound | R1 | R2 | MW (Da) | HBA | HBD | RB | LogP | PSA |
| 5a | 4-phenyl | - | 420.36 | 4 | 0 | 4 | 6.30 | 28.07 |
| 5b | 4-phenyl | 4-hydroxy | 436.35 | 5 | 1 | 4 | 5.10 | 48.30 |
| 5c | 4-phenyl | 4-acetyl | 462.39 | 5 | 0 | 5 | 5.69 | 45.14 |
| 5d | 4-phenyl | 4-cyano | 445.36 | 5 | 0 | 4 | 6.32 | 51.86 |
| 5e | 4-benzyl | - | 434.38 | 4 | 0 | 5 | 6.72 | 28.07 |
| 5f | 4-morpholine | - | 429.39 | 6 | 0 | 4 | 3.60 | 40.54 |
| 5g | 4-carboxylic acid | - | 388.27 | 6 | 1 | 4 | 3.95 | 65.37 |
| 5h | 3-carboxylic acid | - | 388.27 | 6 | 1 | 4 | 4.09 | 65.37 |
| 5i | 4-(ethyloxycarboxyl) | - | 416.32 | 6 | 0 | 6 | 4.55 | 54.37 |
| 5j | 3-(ethyloxycarboxyl) | - | 416.32 | 6 | 0 | 6 | 4.69 | 54.37 |
| 5k | 2-(ethyloxycarboxyl) | - | 416.32 | 6 | 0 | 6 | 4.81 | 54.37 |
*HBA (Hydrogen Bind Acceptor) and HBD (hydrogen bind donor) values of compound 5a-5k were calculated by MedChem Designer 5.5.
*MW (molecular weight), logP and PSA (polar surface area) values of compound 5a-5k were calculated by Chem & Bio Draw 12.0.
*RB (rotatable bond) value of compound 5a-5k were calculated by SwissAdme.
Toxicity prediction is a tool is an useful method in the drug discovery due to many of the newly synthesized potential candidates had failed in clinical trial evaluation because of some pharmacokinetics and toxicity problems. In silico predictions are improved to overcome such scenario in this detailed process. Meanwhile, toxicity predictions were clearly revelead that all our novel compounds are seemed to have no predictable central nervous system side effects, carcinogenicity, and mutagenicity (Table 5).
Table 5.
In silico predicted toxicity measurements of compound 5a-5k.
![]() | |||||
|---|---|---|---|---|---|
| Compound | R1 | R2 | BBBP | CG | MG |
| 5a | 4-phenyl | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5b | 4-phenyl | 4-hydroxy | Penetrating | Non-carcinogenic | Non-mutagenic |
| 5c | 4-phenyl | 4-acetyl | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5d | 4-phenyl | 4-cyano | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5e | 4-benzyl | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5f | 4-morpholine | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5g | 4-carboxylic acid | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5h | 3-carboxylic acid | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5i | 4-(ethyloxycarboxyl) | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5j | 3-(ethyloxycarboxyl) | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
| 5k | 2-(ethyloxycarboxyl) | - | Non-penetrating | Non-carcinogenic | Non-mutagenic |
* BBBP: Blood brain barrier penetration; CG: Carcinogenicity; MG: Mutagenicity.
4. Conclusion
In summary, we report the efficient synthesis, characterization, antimicrobial and cytotoxic activity evaluation of new compound series which contain different substituted piperidine bearing 1,3,4-oxadiazole-2( 3H )-thione structures. According to biological consequences, phenyl and carbonyl group that substituted to piperidine ring seemed to have supportive property on antimicrobial activity of the novel compounds. Some compounds like 5 c, 5d, 5f, 5g, 5i-5k that contain phenyl and carbonyl group revealed not only antimicrobial effect on certain bacterial and fungal strains but also significant level of anticancer activity against liver carcinoma cell line (HUH7). Especially compound 5b showed the best inhibitor activity against Bacillus spp whereas compound 5d represent valuable effect against liver carcinoma cells (HUH7). Besides, evaluating biological properties, compounds were predicted for the chemo-informatic and possible toxicity features within some software programmes. Synthesized molecules were calculated as to qualified the criteria to be a drug as per Lipinski Rule of Five and in silico predicted toxicology results were seemed as all of them have no mutagenic or carcinogenic profiles.
Supplementary Information
1H and 13C NMR spectra of all compounds, 5a-5k, are given as supplementary information at www.ias.ac.in/chemsci.
Acknowledgments
Authors gratefully acknowledge Prof. Dr. Rengül Çetin-Atalay and İrem Durmaz for the cytotoxicity assay.
References
- Penesyan A Gillings M Paulsen IT Antibiotic discovery: combatting bacterial resistance in cells and in biofilm communities. Molecules . 2015;20:5286–5298. doi: 10.3390/molecules20045286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tetz G Tetz V. In vitro antimicrobial activity of a novel compound, Mul-1867, against clinically important bacteria. Antimicrobial Resistance and Infection Control . 2015;4:45–45. doi: 10.1186/s13756-015-0088-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies J Davies D Origins and evolution of antibiotic resistance. Microbiology and Moleculer Biology Reviews . 2010;74:417–433. doi: 10.1128/MMBR.00016-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pitta E Tsolaki E Geronikaki A Petrović J Glamočlija J -Thiazolidinone derivatives as potent antimicrobial agents: microwave- assisted synthesis, biological evaluation and docking studies. Medicinal Chemistry Communications . 2015;6:319–319. [Google Scholar]
- Critically important antimicrobials for human medicine, 6th revision 2019.
- Baguley BC Multiple drug resistance mechanisms in Cancer. Molecular Biotechnology . 2010;46:308–316. doi: 10.1007/s12033-010-9321-2. [DOI] [PubMed] [Google Scholar]
- Theuretzbacher U Accelerating resistance, inadequate antibacterial drug pipelines and international responses. International Journal of Antimicrobial Agents . 2012;39:295–299. doi: 10.1016/j.ijantimicag.2011.12.006. [DOI] [PubMed] [Google Scholar]
- Mandell LA Ball P Tillotson G Antimicrobial safety and tolerability: differences and dilemmas. Clinical Infectious Diseases . 2001;32:72–79. doi: 10.1086/319379. [DOI] [PubMed] [Google Scholar]
- Lincke CR Bliek AM Schuurhuis GJ Velde-Koerts T Smit JJ Multidrug resistance phenotype of human BRO melanoma cells transfected with a wild-type human mdr1 complementary DNA. Cancer Research . 1990;50:1779–1785. [PubMed] [Google Scholar]
- Arias CA Murray BE Antibiotic-resistant bugs in the 21st Century — a clinical super-challenge. The New England Journal of Medicine . 2019;360:439–443. doi: 10.1056/NEJMp0804651. [DOI] [PubMed] [Google Scholar]
- Kakde D Jain D Shrivastava V Kakde R Patil AT Cancer therapeutics-opportunities, challenges and advances in drug delivery. Journal of Applied Pharmaceutical Science . 2011;1:1–10. [Google Scholar]
- Felício MR Silva ON Gonçalves S Santos NC Franco OL Peptides with dual antimicrobial and anticancer activities. A Review. Frontiers in Chemistry . 2017;5 doi: 10.3389/fchem.2017.00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alibek K Bekmurzayeva A Mussabekova A Sultankulov B Using antimicrobial adjuvant theraphy in cancer treatment: a review. Infectious Agents and Cancer . 2012;7:33–33. doi: 10.1186/1750-9378-7-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otova B Hrdy J Votruba I Holy A In vivo modulation of angiogenic gene expression by acyclic nucleoside phosphonates pmedap and pmeg. Anticancer Research . 2009;29:1295–1302. [PubMed] [Google Scholar]
- Mondello C Scovassi AI Telomeres, telomerase, and apoptosis. Biochemistry and cell biology-biochimie et biologie cellulaire. 2004;82:498–507. doi: 10.1139/o04-048. [DOI] [PubMed] [Google Scholar]
- Fumo G Akin C Metcalfe DD -demethoxygeldanamycin (17-aag) is effective in down-regulating mutated, constitutively activated kit protein in human mast cells. Blood . 2004;103:1078–1084. doi: 10.1182/blood-2003-07-2477. [DOI] [PubMed] [Google Scholar]
- Mahalingam D Swords R Carew JS Nawrocki ST Bhalla K Targeting hsp90 for cancer therapy. British Journal of Cancer . 2009;100:1523–9. doi: 10.1038/sj.bjc.6605066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattson DM Ahmad IM Dayal D Parsons AD Aykin-Burns N Cisplatin combined with zidovudine enhances cytotoxicity and oxidative stress in human head and neck cancer cells via a thiol-dependent mechanism. Free Radical Biology and Medicine . 2009;46:232–7. doi: 10.1016/j.freeradbiomed.2008.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding WQ Liu BL Vaught JL Yamauchi H Lind SE Anticancer activity of the antibiotic clioquinol. Cancer Research . 2005;65:3389–3389. doi: 10.1158/0008-5472.CAN-04-3577. [DOI] [PubMed] [Google Scholar]
- Hye JJ Yonghyo K Hyang BL Ho JK Antiangiogenic Activity of the Lipophilic Antimicrobial Peptides from an Endophytic Bacterial Strain Isolated from Red Pepper Leaf. Molecules and Cells . 2004;38:273–8. doi: 10.14348/molcells.2015.2320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curiel TJ Tregs and rethinking cancer immunotherapy. Journal of Clinical Investigation . 2007;117:1167–74. doi: 10.1172/JCI31202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assouline S Culjkovic B Cocolakis E Rousseau C Beslu N Molecular targeting of the oncogene eif4e in acute myeloid leukemia (aml): A proof-of-principle clinical trial with ribavirin. Blood . 2009;114:257–60. doi: 10.1182/blood-2009-02-205153. [DOI] [PubMed] [Google Scholar]
- Kentsis A Topisirovic I Culjkovic B Shao L Borden KLB Ribavirin suppresses eif4e-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mrna cap. Proceedings of the National Academy of Sciences (Proceedings of the National Academy of Sciences of the United States of America) . 2004;101:18105–10. doi: 10.1073/pnas.0406927102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halasi M Zhao H Dahari H Bhat UG Gonzalez EB Thiazole antibiotics against breast cancer. Cell Cycle . 2010;9:1214–17. doi: 10.4161/cc.9.6.10955. [DOI] [PubMed] [Google Scholar]
- Boström J Hogner A Llinas A Wellner E Plowright AT Oxadiazoles in Medicinal Chemistry. Journal of Medicinal Chemistry . 2012;55:1817–30. doi: 10.1021/jm2013248. [DOI] [PubMed] [Google Scholar]
- Du Q Li D Pi Y Li J Sun J -oxadizole thioether derivatives targeting thymidylate synthase as dual anticancer/antimicrobial agents. Novel . 2013;1:2286–97. doi: 10.1016/j.bmc.2013.02.008. [DOI] [PubMed] [Google Scholar]
- Ahsan MJ Sharma J Bhatia S Goyal PK Shank-hala K et al. Synthesis of 2,5-disubstituted-1 . 2014;11:413–413. [Google Scholar]
- Savariz FC Formagio ASN Barbosa VA Foglio MA Carvalho JE Synthesis, antitumor and antimicrobial activity of novel 1-substitutedphenyl-3-[3-alkylamino(methyl)-2-thioxo- Journal of Brazilian Chemical Society . 2010;21:4–4. [Google Scholar]
- Selvaraj K Kulanthai K Sadhasivam G Synthesis, characterization and biological evaluation of novel 2,5-disubstituted- Saudi Pharmaceutical Journal . 2017;25:4–4. doi: 10.1016/j.jsps.2016.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koksal M Bilge SS Bozkurt A Sahin S Isik S Synthesis, characterization and anti-inflammatory activity of new 5-(3,4-dichlorophenyl)-2-(aroylmethyl)thio-1, Arzneimittel-Forschung . 2008;58:4–oxadiazoles. doi: 10.1055/s-0031-1296550. [DOI] [PubMed] [Google Scholar]
- Manual of Clinical Microbiology ASM. pp. 1482–1482.
- Skehan P Storeng R Scudiero D Monks A McMahon J New colorimetric cytotoxicity assay for anticancer-drug screening. Journal of National Cancer Institute . 1990;82:1107–1112. doi: 10.1093/jnci/82.13.1107. [DOI] [PubMed] [Google Scholar]
- Glück J Buhrke T Frenzel F Braeuning A Lampen A In silico genotoxicity and carcinogenicity prediction for food-relevant secondary plant metabolites. Food and Chemical Toxicology . 2018;116:298–306. doi: 10.1016/j.fct.2018.04.024. [DOI] [PubMed] [Google Scholar]
- Sowjanya C RamaBharathi V Kalpana Devi G Rajitha G Synthesis and evaluation of some novel 3-[5-phenyl- Journal of Chemical and Pharmaceutical Research . 2011;4:4–4. [Google Scholar]
- Romano E Soria NAJ Rudyk R Silvia A. -thiol by using DFT calculations. Theoretical study of the infrared spectrum of 5-phenyl-1 . 2012;38:4–4. [Google Scholar]
- Parikh K Joshi D Synthesis and evaluation of 2-(5-(aryl. Journal of Chemical Sciences . 2014;1:4–4. [Google Scholar]
- Sankhe NM Durgashivaprasad E Kutty NG Rao JV Narayanan K -oxadiazole derivatives induce apoptosis in HepG2 cells through p53 mediated intrinsic pathway. Novel . 2019;2:5–5. [Google Scholar]
- Chikhalia KH Vashi DB Patel MJ Synthesis of a novel class of some 1,3,4-oxadiazole derivatives as antimicrobial agents. Journal of Enzyme Inhibition and Medicinal Chemistry . 2009;24:617–22. doi: 10.1080/14756360802318936. [DOI] [PubMed] [Google Scholar]
- Modh RP Shah D Chikhalia KH -oxadiazole derivatives: Design, synthesis, antibacterial and antifungal studies. Indian Journal of Chemistry . 2013;1:2–2. [Google Scholar]
- Jadhav BP Yadav AR Gore MG Concept of drug likeness in pharmaceutical research. International. Journal of Pharma and Bio Sciences . 2015;6:142–54. [Google Scholar]
- Bakht MA Yar MS Abdel-Hamid SG Al-Qasoumi SI Samad A Molecular properties prediction, synthesis and antimicrobial activity of some newer oxadiazole derivatives. European Journal of Medicinal Chemistry . 2010;45:5862–69. doi: 10.1016/j.ejmech.2010.07.069. [DOI] [PubMed] [Google Scholar]
- Tian S Wang J Li Y Li D Xu L The application of in silico drug-likeness predictions in pharmaceutical research. Advanced Drug Delivery Reviews . 2015;86:2–10. doi: 10.1016/j.addr.2015.01.009. [DOI] [PubMed] [Google Scholar]
- Ghose AK Herbertz T Hudkins RL Dorsey BD Mallamo JP Knowledge-Based, Central Nervous System (CNS) Lead Selection and Lead Optimization for CNS Drug Discovery. ACS Chemical Neuroscience . 2013;3:50–68. doi: 10.1021/cn200100h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veber DF Johnson SR Cheng HY Smith BR Ward KW Molecular properties that influence the oral bioavailability of drug candidates. Journal of Medicinal Chemistry . 2002;45:2615–2615. doi: 10.1021/jm020017n. [DOI] [PubMed] [Google Scholar]



