Corresponding author: Ammar A. Razzak Mahmood ( kubbaammar1963@gmail.com ) Academic editor: Plamen Peikov
© 2021 Ammar I. Al-Bayati, Ammar A. Razzak Mahmood, Zainab A. Al-Mazaydeh, Majdoleen S. Rammaha, Rheda I. Al-bayati, Fatima Alsoubani, Lubna H. Tahtamouni.
This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Citation:
Al-Bayati AI, Mahmood AAR, Al-Mazaydeh ZA, Rammaha MS, Al-bayati RI, Alsoubani F, Tahtamouni LH (2021) Synthesis, docking study, and in vitro anticancer evaluation of new flufenamic acid derivatives. Pharmacia 68(2): 449-461. https://doi.org/10.3897/pharmacia.68.e66788
|
Novel compounds (6–10) were synthesized and confirmed by spectroscopic analysis, including AT-IR, 1HNMR and CHNS. Their cytotoxic effect was evaluated by MTT assay against two cancer cell lines and two normal cell types. Compound 7 exhibited anticancer activity against MCF-7 breast cancer cell line (GI50 = 63.9 µg/ml, 148 µM), without any effect against A549 lung cancer cells, or the normal cells. Compound 7 caused cytotoxicity in MCF-7 breast cancer cells by apoptotic cell death, as suggested by fragmented nuclei after DAPI staining and agarose gel electrophoresis. In addition, treating MCF-7 cells with compound 7 resulted in an increase in the level of caspase 9 mRNA level, and its activation. Moreover, compound 7-treated MCF-7 cells showed enhanced cytochrome c release from the mitochondria to the cytosol, signifying an induction of the intrinsic apoptotic pathway. Finally, compound 7 exhibited epidermal growth factor receptor (EGFR) kinase inhibitory activity at (EC50 = 0.13 µM), which was matched by molecular docking studies that showed compound 7 might be an important EGFR kinase inhibitor.
Apoptosis intrinsic pathway, EGFR binding site, Phenylisothiocyanate, Thiadiazole
Cancer, uncontrolled cell growth, is a molecularly heterogeneous disease and a leading cause of death worldwide. According to the American Cancer Society, there is a substantial increase in cancer rates, particularly in African regions (
Heterocyclic compounds are of particular interest to medicinal chemists, because of their remarkable chemical and versatile biological profiles. Despite considerable advances in research progress into heterocyclic ring systems, attempts are always being made to find new heterocyclic compounds with versatile bioactivity (
Diverse mechanisms of action are being attributed to the anticancer effects of 1,3,4-thiadiazole moiety, such as inhibiting DNA, RNA, and protein synthesis, inhibiting carbonic anhydrase (Supuran and Scozzafava, 2000), phosphodiesterase-7 (PDE7), histone deacetylase, or acting as an adenosine A3 receptor antagonist. Some thiadiazole derivatives used as HIV protease inhibitors were investigated as potential anti-cancer drugs. These agents are capable of preventing the activation of growth factor receptors and downstream Akt signaling, resulting in endoplasmic reticulum stress, autophagy and cell death (
The epidermal growth factor receptor (EGFR) is expressed in 60% of triple negative breast cancers (TNBCs) and triggers disease progression. Efforts to try to inhibit EGFR in unselected TNBC patients had a marginal impact on health outcomes (
EGFR-dependent tumors that are initially susceptible to EGFR TKIs (tyrosine kinase inhibitors) may develop threonine mutation of 790 (Mishra et al. 2008). Substitution of this residue in EGFR with bulky methionine may cause resistance by steric interference with TKI binding, including gefitinib and erlotinib. New compounds and drug combinations offer an opportunity to delay or overcome resistance mechanism to EGFR TKIs (
Flufenamic acid was purchased from Sigma-Aldrich. The infrared spectra were recorded using Shimadzu Specac GS 10800-R IR Affnity-1 Spectrometer (v = cm-1). CHNS microanalysis was carried out using a Euro EA3000 elemental analyzer.
1HNMR spectra of the synthesized compounds were measured on AVANCE-III 400MHz Nanobay FT-NMR spectrometer, using tetramethylsilane (TMS) as an internal standard, the chemical shift was displayed once as (δ, ppm), and DMSO-d6 was utilized as a solvent.
Flufenamic ethyl ester was prepared in accordance with the method developed by Curtius and Geoble (1888). In a 500 mL round bottom flask with ground glass joint, a mixture of flufenamic acid (0.01 mol, 1.39 g) and 100 mL abs. EtOH was added, and the flask was fitted with a rubber cork carrying an inlet tube as well as a calcium chloride guard tube. Hydrochloric acid dried by bubbling through conc. H2SO4 was passed into the mixture till enough gas was dissolved. The flask was fitted with a reflux condenser and the mixture was refluxed. The flufenamic acid completely went into solution within about 30 min. After a total refluxing of 6 h, the flask was allowed to cool, the solution was neutralized with 10% Na2CO3 and extracted by four portions of diethyl ether. The ether was evaporated by rotary evaporator and the pure crystals were obtained by recrystallization from 60% EtOH. A colorless flufenamic acid ethyl ester, yield (89%), m.p = 144–146 °C, Rf. = 0.9, AT-IR (v = cm-1): 3313 str of sec. (NH2); 3000–3100 (Ar-CH) str, 2981 str of methyl (CH), 2930 str of methelyne (CH), 1685 str of (C = O) conjugated ester carbonyl, 1523,1454, str of Ar (C = C), 1165 (C-O) str of ester 794, 748, 698 str of ortho & meta substitution.
1H NMR (500 MHz, DMSO-d6, δ ppm): 9.36 (s, 1 H, NH) 7.93 (br d, J=7.81 Hz, 1 H, Ar-H) 7.51–7.56 (m, 5 H Ar-H) 7.32 (br t, J=8.29 Hz, 1 H, Ar-H) , 6.95 (t, J=7.62 Hz, 1 H, Ar-H) ,4.31 (q, 2H, CH2 aliph), 1.31 (t, 3H, CH3 aliph).
Flufenamic ethyl ester, (0.037 mol, 5.6 g) and hydrazine hydrate 99.5% (an excess amount of 0.185 mol, 9.0 mL) were added to 40 mL of EtOH into 250 mL round bottom flask and the mixture was first refluxed at 80 °C for 12 h after which, it was set to be stirred overnight at room temperature (RT). It was noticed that the colorless solution changed into pale pink with the time. At the end, half of the solvent was removed under reduced pressure, and the residue was poured into ice. The precipitate was filtered and washed with ice-cold EtOH to afford a product, afterwards, it was recrystallized from 70% EtOH to yield compound (Hydrazide), yield (80%), m.p = 149–151 °C, Rf. = 0.74, AT-IR (v = cm-1): 3317, 3305 doublets, str of prim (NH2), 3305, 3197 str of sec (NH), 1616 bend of (NH), 1523, 1496, 1465 str (Ar C = C) skeleton, 1581 bend of (NH) (amide II band), 1284 str of (C-N), 790, 752, 694 str of ortho &meta substitutions.
1H NMR (500 MHz, DMSO-d6, δ ppm): 9.85 (br s, 1 H, sec.NH-amine), 9.52 (s, 1 H, NH-amide), 7.61 (br d, J = 7.81 Hz, 1 H, Ar-H), 7.48 (br t, J = 7.81 Hz, 1 H, Ar-H), 7.37–7.42 (m, 4 H, Ar-H), 7.23 (br d, J = 7.32 Hz, 1 H, Ar-H), 6.95 (t, J = 7.32 Hz, 1 H, Ar-H), 4.54 (br s, 2 H, NH2).
To a solution of compound (hydrazide) (0.00364 mol, 0.5 g) in 25 mL of EtOH, the following were added separately [(1): p-chloro phenylisothiocyanate (0.00364 mol, 0.78 g), (2): p-fluoro phenylisothiocyanate, (0.00364 mol, 0.558g), (3): p-bromophenylthiocyanate, (0.00364 mol, 0.558g), (4) phenylisothiocyanate, (0.00364 mol, 0.657 g), and (5): p-methylphenylisothiocyanate (0.00364 mol, 0.544g)]. The reaction mixture was stirred at 40–50 ⁰C for 4 h, and then it was kept stirring overnight at RT. Half of the solvent was removed under reduced pressure, and the residue was poured into ice. The precipitate was filtered and washed with ice-cold EtOH, to give a product and recrystallized from 70% EtOH to yield the corresponding final compounds, (Scheme 1).
Off-white crystals, yield (94%), m.p= (190–194 °C) , Rf = 0.89, AT-IR (v = cm-1): 3363 str of sec (NH), 3271 str (NH) of thioamide, 3159 str (NH) of amide, 2962 str of (Ar-CH), 1651 str (C = O) of amide (amide I band), 1581 bend (NH), 1519, 1491, 1446, 1423 str of (Ar C = C) skeleton, 1257 str (C-N), 1211 str (C = S), 829 str p-Cl substitution, 794, 740, 698 str ortho &meta substitutions.
1H NMR (400 MHz, DMSO-d6,δ = ppm):10.62 (br s, 1 H, sec.NH-amide), 9.80 (s, 1 H, sec,NH-amine), 9.60 (br s, 1 H, sec.NH-thioamide), 7.88 (br s, 1 H, Ar-H), 7.31–7.53 (m, 9 H Ar-H and sec.NH), 7.21 (br d, J=7.46 Hz, 1 H, Ar-H), 6.99 (br t, J = 7.40 Hz, 1 H, Ar-H).
CHNS analysis: Calcd. for (C21H16ClF3N4OS) C, 54.25; H, 3.47; N, 12.05; S, 6.90. Observed: C, 54.35; H, 3.69; N, 12.28; S, 6.72.
White crystals, yield (87.5%), m.p = (104–208 ⁰C) , Rf = 0.73, AT-IR (v = cm-1): 3363 str (NH) of sec. amide, 3228 str (NH) of thioamide, 3174 str (NH) of amide, 1654 str (C = O) of amide (amide I band), 1604 bend (NH) overlapped with Ar-str of (C = C) skeleton, 1581 bend (NH) of (amide II band), 1504 str of (Ar C = C) skeleton, 1253 str (C-N), 1211 str (C = S), 833 p-F substitution, 783, 744, 694 str ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ ppm) 10.62 (br s, 1 H, sec.NH-amide), 9.73 (br s, 1 H, sec.NH), 7.89 (br s, 1 H, Ar-H), 7.41–7.53 (m, 7 H, Ar-H, & NH), 7.26 (br d, J = 7.46 Hz, 1 H, Ar- H), 7.17 (br t, J=8.68 Hz, 2 H, Ar-H), 6.99 (t, J = 7.40 Hz, 1 H, Ar-H).
CHNS analysis: Calcd. for (C21H16F4N4OS) C, 56.25; H, 3.60; N, 12.49; S, 7.15. Observed: C, 56.41; H, 3.62; N, 12.36; S, 6.96.
White powder, yield (95%), m.p = (180–185 ⁰C), Rf = 0.89, FTIR (v = cm-1): 3356 str of sec. amide (NH), 3275 str of thioamide (NH), 3155 str of amide (NH), 1651 str of(C = O) amide (amide I band), 1581 str of (C=N) overlapped with Ar- str of (C = C) of skeleton, 1519 bend of (amide II band) (NH), 1257str (C-N), 1215 str of (C = S), 825 p-Br substitution, 794, 740, 698 str ortho &meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ ppm): 10.62 (br s, 1 H, secNH-amide), 9.81 (br s, 1 H,sec. NH), 9.61 (br s, 1 H, NH-thioamide), 7.88 (br s, 1 H, Ar-H), 7.39–7.53 (m, 10 H, Ar-H & NH), 7.25 (br d, J = 7.21 Hz, 1 H, Ar-H), 7.00 (br t, J = 7.15 Hz, 1 H, Ar-H).
CHNS analysis: Calcd. for (C21H16BrF3N4OS) C, 49.52; H, 3.17; N, 11.00; S, 6.30. Observed: C, 49.70; H, 3.01; N, 11.03; S, 6.34.
Off-white powder, yield (90%), m.p = (190–195 ⁰C), Rf = 0.63, AT-IR (v =cm-1): 3371 str of sec. amide (NH), 3228 str of thioamide (NH), 3163 str of amide (NH), 1651 str of (C = O), amide (amide I band), 1597 str of (C=N) overlapped with Ar-str vibrations of (C=C) skeleton, 1527 bend of (amide II band) (NH), 1257str (C-N), 1215 str (C = S), 786, 740, 692 str ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ ppm): 10.62 (br s, 1 H, sec.NH-amide), 9.82 (br s, 1 H, sec.NH), 9.59 (br s, 1 H, NH-thioamide), 7.80 (br s, 1 H, Ar-H), 7.46–7.66 (m, 6 H, Ar-H & NH), 7.31–7.41 (m, 4 H, Ar-H), 7.27 (br d, J = 7.46 Hz, 1 H, Ar-H) ,7.17 (m, 1 H, Ar-H) 7.00 (br t, J = 7.34 Hz, 1 H, Ar-H).
CHNS analysis: Calcd. for (C21H17F3N4OS) C, 58.60; H, 3.98; N, 13.02; S, 7.45. Observed: C, 58.80; H, 4.20; N, 13.23; S, 7.23.
White powder, yield (92%), m.p = (189–192 ⁰C) , Rf = 0.64, ATIR (v = cm-1): 3360 str of sec. amide (NH), 3267 str of thioamide (NH), 3228 str of amide (NH), 3140 str of Ar-(CH), 1651 str of(C = O) amide (amide I band), 1581 str of (C=N) overlapped with aromatic -stretching of (C = C) skeleton, 1519 bend of (amide II band) (NH), 1330 (CH) sym in-plane bend of (–CH3), 1257 str (C-N), 1215 str (C = S), 817 p-CH3-substitution, 794, 740, 663 ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ ppm) 10.59 (br s, 1 H, sec.NH-amide), 9.75 (br s, 1 H, sec.NH), 9.62 (br s, 1 H,NH-thioamide), 7.87 (br s, 1 H, Ar-H), 7.42–7.53 (m, 6 H, Ar-H & NH), 7.21–7.40 (m, 3 H, Ar-H), 7.14 (br d, J = 7.83 Hz, 2 H, Ar-H), 6.99 (br t, J = 7.34 Hz, 1 H, Ar-H) , 2.29 (s, 3 H, CH3-aliph).
CHNS analysis: Calcd. for (C22H19F3N4OS) C, 59.45; H, 4.31; N, 12.61; S, 7.21. Observed: C, 59.61; H, 4.31; N, 12.67; S, 7.15.
Hydrazinecarbothioamide compounds (1-5) (0.001 mol) were added separately to 5 mL conc.H2SO4 at 0 °C, and stirred for 3 h at RT. It was noticed that a clear yellow solution appeared. The reaction mixture was neutralized with 2N NaOH and filtered, afterwards, it was washed with a plenty amount of H2O. The precipitate was recrystallized with 70% EtOH to afford the tiled compounds (6–10).
Pale brown powder, yield (42%), m.p = (75–77 ⁰C), Rf = 0.52, ATIR (v =cm-1): 3259,3194 str of sec. amine (NH), 3043str of Ar (CH), 1616,1600,1581,1562 bend of (NH), and str (C = N), 1492, 1516 str of Ar-(C = C), 1219 str (C-N), 624 str (C-S-C), 821 str p-Cl-substitution, 794, 729, 694 ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ = ppm): 10.57 (s, 1 H, sec. NH-TDZ), 9.12 (s, 1 H, NH), 7.98 (dd, J = 7.82, 1.10 Hz, 1 H, Ar-H), 7.69–7.72 (m, 2 H, Ar-H), 7.38–7.43 (m, 6 H, Ar-H), 7.18–7.24 (m, 3 H, Ar-H).
CHNS analysis: Calcd. for (C21H14ClF3N4S) C, 56.44; H, 3.16; N, 12.54; S, 7.18. Observed: C, 56.66; H, 3.32; N, 12.71; S, 7.26.
Pale brown powder, yield (30%), m.p (83–86 ⁰C), Rf = 0.30, ATIR (v = cm-1): 3259,3217 str of sec. amine (NH), 2997 str of Ar(CH), 1624, 1581 bend of (NH) and str (C = N), 1485,1423 str of Ar-(C = C), 1226 str (C-N), 613 str (C-S-C), 821 p-F- substitution, 750, 729, 698 ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ = ppm) 10.46 (s, 1 H, sec.NH TDZ), 9.13 (s, 1 H, NH), 7.97 (d, J = 7.46 Hz, 1 H, Ar-H), 7.68–7.70 (m, 2 H, Ar-H), 7.46–7.52 (m, 3 H, Ar-H), 7.17–7.24 (m, 6 H, Ar-H).
CHNS analysis: Calcd. for (C21H14F4N4S) C, 58.60; H, 3.28; N, 13.02; S, 7.45. Observed: C, 58.81; H, 3.48; N, 13.24; S, 7.63.
Brown powder, yield (35%), m.p = (88–92 ⁰C), Rf = 0.9, FTIR (v = cm-1): 3278,3190 str of sec. amine (NH), 3035 str of Ar-(C-H), 1612,1597,1562 bend of (NH), and str (C = N), 1492,1527,1423 str of Ar-(C = C), 1219 str (C-N), 613 str (C-S-C), 817 p-Br substitution, 794, 744,732,698 ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6,δ = ppm,): 10.58 (s, 1 H, NH-TDZ), 9.11 (s, 1 H, sec. NH), 7.98 (d, J = 7.83 Hz, 1 H, Ar-H), 7.65 (d, J = 8.93 Hz, 2 H, Ar-H), 7.43–7.53 (m, 5 H, Ar-H), 7.19–7.23 (m, 4H, Ar-H). CHNS analysis: Calcd. for (C21H14BrF3N4S) C, 51.34; H, 2.87; N, 11.40; S, 6.53. Observed: C, 51.23; H, 3.10; N, 11.61; S, 6.47.
Pale brown powder, yield (50%), m.p = (90–93 ⁰C), Rf = 0.74, ATIR (v = cm-1): 3263,3190 str of sec. amine (NH), 3055,3028 str of Ar-(CH), 1597,1566 bend of (NH) and str (C = N), 1496,1539,1445,1419 str of Ar-(C = C), 1226 str (C-N), 632 str (C-S-C), 790,752,729,664 ortho & meta str substitutions.
1H NMR (400 MHz, DMSO-d6, δ = ppm): 10.44 (s, 1 H, sec.NH), 9.15 (s, 1 H, NH-TDZ), 7.97 (d, J = 7.57 Hz, 1 H, Ar-H), 7.66 (d, J = 7.82 Hz, 2 H, Ar-H), 7.35–7.46 (m, 5 H, Ar-H), 7.19–7.25 (m, 4 H, Ar-H), 7.01 (t, J = 7.34 Hz, 1 H, Ar-H).
CHNS analysis: Calcd. for (C21H15F3N4S) C, 61.16; H, 3.67; N, 13.58; S, 7.77. Observed: C, 58.25; H, 4.05; N, 12.92; S, 8.71.
Brown powder, yield (46%), m.p (92–96 ⁰C), Rf = 0.89, FTIR (v = cm-1): 3248,3186 str of sec. amine (NH), 3116,3032 str of Ar-(CH), 1616,1597,1570,1535 bend of (NH) and str (C = N), 1535,1516,1469,1450,1423 str of Ar-(C = C), 1219 str (C-N), 617 str (C-S-C), 817 p-methyl substitution, 790,748,729,694 ortho & meta substitutions.
1H NMR (400 MHz, DMSO-d6, δ = ppm) 10.34 (s, 1 H, NH-TDZ), 9.17 (s, 1 H,sec.NH), 7.94 (d, J = 7.54 Hz, 1 H, Ar-H), 7.54 (d, J = 8.31 Hz, 2 H, Ar-H), 7.38–7.50 (m, 3 H, Ar-H), 7.12–7.28 (m, 6 H, Ar-H), 2.26 (s, 3H, aliph. CH3).
CHNS analysis: Calcd. for (C22H17F3N4S) C, 61.96; H, 4.02; N, 13.14; S, 7.52. Observed: C, 30.48; H, 2.22; N, 6.58; S, 7.14.
Human A549 lung cancer cell line was cultured in Ham’s F-12K (Kaighn’s) medium (Gibco, USA) supplemented with 10% fetal bovine serum (FBS; Capricorn Scientific, Germany), human MCF-7 breast cancer cells were cultured in RPMI-1640 medium (Euroclone, Italy) supplemented with 10% FBS, The mammary non-tumorigenic MCF10A cell line was grown in DMEM/F12 (Gibco, USA) supplemented with 5% horse serum (Invitrogen, USA), EGF (20 ng/mL, Sigma), hydrocortisone (0.5 mg/mL, Sigma), cholera toxin (100 ng/mL, Sigma) and insulin (10 μg/mL, Sigma) and white blood cells (WBC) were cultured in RPMI-1640, supplemented with 5% FBS. Trypsin-EDTA (Lonza, Switzerland) was used throughout for subcultures. Cell growth was attained at 37 °C in 5% carbon dioxide and 95% air.
The cytotoxicity of the parent compounds (1–5) and the various 1,3,4-thiadiazole derivatives (6–10) against different cell lines (A549, MCF-7 and MCF10A), and WBC was evaluated using an MTT assay (tetrazolium salt reduction) assay (
Apoptosis was identified by microscopic analysis of apoptotic nuclei upon DAPI staining (
The quantity of caspase 4, caspase 8, caspase 9 mRNA was assessed by qRT-PCR (
Control and GI50-treated cells were washed 3 times with cold PBS, and then lysed with protein lysis buffer [10% sodium dodecyl sulfate (SDS), 1 M Tris buffer pH 7.5, 1 M sodium florid (NaF), 1M dithiothreitol (DTT), 0.1 M ethylene glycol tetraacetic acid (EGTA), and D.W on ice. The cell lysate was collected and boiled for 5 min and sonicated. Aliquots of lysates were diluted in 4x SDS-PAGE sample buffer (0.5 M Tris-HCl pH 6.8, 2% SDS, 20% glycerol, 20% 2-mercaptoethanol and 0.16% bromophenol blue), and proteins were resolved by electrophoresis on 10% or 12.5% SDS-polyacrylamide gels. Proteins were transferred onto nitrocellulose membranes and were blocked using 2% (w/v) BSA in Tris-buffered saline (TBS), and subjected overnight at 4°C to the primary antibodies: rabbit polyclonal anti-caspase 4 (1:1000; Invitrogen, USA), mouse monoclonal anti-caspase 8 (1:1000; Invitrogen, USA), mouse monoclonal anti-caspase 9 (1:1000; Invitrogen, USA), mouse monoclonal anti-GAPDH (1:6000; CHEMICON, USA), rabbit polyclonal anti-tubulin (1:3000; abcam, USA), diluted in 1% BSA in TBS containing 0.05% Tween 20. After washing and incubation with appropriate secondary antibodies conjugated to IRDye 680 or 800 nm fluorescent dyes, the membranes were washed, and the bands were analyzed on FluorChem R system (Oxford, UK). Signals were validated using AlphaView software (ProteinSimple, USA) (
5x107 control and GI50-treated cells were recovered by centrifugation at 2,900 RPM for 5 min, washed with ice cold PBS, and centrifuged at 2,900 RPM for 5 min, and re-suspended with 1X cytosolic extraction buffer mix containing dithiothreitol (DTT), and protease inhibitors. The cells were incubated on ice for 10 min, homogenized and centrifuged at 3,100 RPM for 10 min. The supernatant was centrifuged again at 12,000 RMP for 30 min to obtain the cytosolic fraction, while the pellet was re-suspended in mitochondrial extraction buffer to collect the mitochondrial fraction (Cytochrome c Release Apoptosis Assay Kit, Abcam, USA).
The in vitro inhibitory activity of the synthesized compound(s) against EGFR was accomplished using EGFR(T790M/L858R) Kinase Assay Kit (BPS Bioscience, USA). Briefly, EGFR and its substrate were incubated with the synthesized compounds in enzymatic buffer for 40 min at 30 °C to launch the enzymatic reaction. The reaction was terminated by addition of detection reagent (Kinase-Glo Max reagent, Promega), followed by incubation at RT for 15 min. The remaining activity of EGFR kinase was observed by measuring chemiluminescence using a Biotek Synergy HT Multi-Mode Microplate Reader (VT, USA). The concentration-percent remaining EGFR kinase activity curve was also used to calculate the concentration that caused 50% kinase activity inhibition (the effective concentration that inhibits 50% of EGFR kinase activity; EC50). All samples and controls were verified in duplicate.
All experiments were performed in duplicates or triplicates. The results were presented as average ± SEM. Statistical analysis was carried out using GraphPad Prism version 5.0. (GraphPad Software, San Diego, CA, USA). The Student›s t-test was also used to test for significant differences in means. The p value <0.05 was considered to be significant (
In terms of choosing molecular targets, the newly synthesized compounds (6–10) may function on, and try comparing them with other ligands, and to determine the pharmacophoric functionality that may enable binding to the critical amino acid(s) at the target site, the target site was chosen by the protein data bank (https://www.rcsb.org/). Target compounds are tested in practice against many active sites, and the outcomes will then determine the proper protein for molecular docking. Upon selecting a particular protein, some operations were carried out, and provide insight into the molecular binding modes of the tested compounds within the pocket of the epidermal growth factor receptor tyrosine kinase (ATP binding site of EGFR kinase), using the MOE 2015 software. The binding sites were generated from co-crystallized ligand in crystal protein (PDB codes: 1ywn) (
The cytotoxicity of the five parent compounds (1–5) and the five different thiadiazole derivatives (6–10) was investigated against two human cancer cell lines (A549 and MCF-7), and two normal cell types (MCF10A and WBC) by the in vitro cytotoxicity MTT assay. The corresponding GI50 (50% Growth Inhibition) values for each compound against cancer cells are shown in Table
Compound | A549 | MCF-7 |
1 | No effectb (20%)* | No effectb (16%)* |
2 | No effectb (17%)* | No effectb (40%)* |
3 | No effectb (0%)* | No effectb (27%)* |
4 | No effectb (16%)* | No effectb (28%)* |
5 | No effectb (0%)* | No effectb (8%)* |
6 | No effectb (15%)* | 75.2 µg/mL (169.1µM)a |
7 | No effectb (11%)* | 63.9 µg/mL (148 µM)a |
8 | No effectb (11%)* | No effectb (25%)* |
9 | No effectb (4%)* | No effectb (14%)* |
10 | No effectb (0%)* | 24.7 µg/mL (57.92 µM)a |
The results presented in Fig.
Compound 7 induces apoptosis in MCF-7 breast cancer cells. A) Fluorescence images of MCF-7 breast cancer cells showing fragmented nuclei after DAPI staining. Scale bar: 10 µm, B) Detection of DNA fragmentation by agarose gel electrophoresis. Cells were treated and genomic DNA was extracted and electrophoresed on 1.5% agarose gels. M: DNA molecular weight marker; C: vehicle-treated control cells; 5%: 5% DMSO-treated cells; 7: Compound 7-treated cells. Three experiments were performed with similar results.
In order to investigate the role of caspase activation downstream of compound 7 induction of apoptosis, the mRNA levels of caspase 4, caspase 8 and caspase 9 were tested by qRT-PCR (Fig.
Compound-7 induced apoptosis is dependent on caspase 9 activation and cytochrome c release. A) qRT-PCR analysis of caspase 4, 8 and 9 mRNA in compound 7-treated cells (GI50) as compared to vehicle-treated control cells [set as 1 arbitrary unit (a.u.)]. Values were normalized to β-actin. Bars = mean ± SEM. of three independent experiments performed in triplicates. * P < 0.05 compared to vehicle-treated control cells, B) Representative Western blots showing cleavage “activation” of procaspase 9 to the active forms p35/p37, C) Representative Western blot showing the release of cytochrome c from the mitochondria (M) into the cytosol (C) of treated MCF-7 breast cancer cells (GI50). Equal protein loading was controlled by staining membranes with Ponceau S (a representative section of the stained membrane is shown). The experiment was repeated three times.
Compound 7 was tested for its EGFR kinase inhibitory activity using a commercial EGFR Kinase Assay Kit. 5% DMSO was used as a negative control (Fig.
Molecular docking process was carried out using CDOCKER protocol. CDOCKER is a grid-based molecular docking methodology that employs CHARMM-based molecular dynamics (MD) system to dock the ligands into a binding pocket. The receptor was held rigid while the ligands were allowed to be flexible during the refining process. The docking score (CDOCKER interaction energy) of the best fitting poses with the active site, the ATP binding site of EGFR kinase was recorded (Table
Show (∆G) kcal/mol of tested candidates against (EGFR) target site PDB ID: 1ywn.
Comp. NO. | Interactions | Score (∆G) kcal/mol |
RMSD Value/ ⁰A | |
---|---|---|---|---|
H.B | pi | |||
1 | 3 | 0 | -6.84 | 1.97 |
2 | 4 | 0 | -6.82 | 1.95 |
3 | 3 | 0 | -6.40 | 1.74 |
4 | 3 | 0 | -6.68 | 1.59 |
5 | 1 | 0 | -6.99 | 1.48 |
6 | 2 | 1 | -8.12 | 1.27 |
7 | 2 | 1 | -7.79 | 1.22 |
8 | 2 | 1 | -8.12 | 1.39 |
9 | 2 | 1 | -7.70 | 1.29 |
10 | 1 | 1 | -7.31 | 1.56 |
Erlotinib | 0 | 3 | -6.36 | 1.68 |
The binding mode of compound 6 exhibited an energy binding of -8.12 kcal/mol. The 1,3,4-thiadiazole ring formed a hydrogen bonding with Lys721 at a distance of 2.12 °A, as well as, the p-chloro phenyl group formed one hydrogen bonding with Met742, at a distance of 3.17 °A. The hydrophobic phenyl moiety formed one pi-interaction with Val702 (Fig.
Meanwhile, tri-fluro phenyl group formed a new pi- interaction with Asp831, and hydrophobic phenyl moiety formed one pi-interaction with Val702 (Fig.
Absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction was carried out with the ADMET descriptor module of the small molecules protocol of the pre ADMET online software.
The partition coefficient (Clogp) values of the target compounds are greater than > 5, and they still follow the Lipiniski rule of five, that states the number of hydrogen bond donors (HBD), and hydrogen bond acceptors (HBA) are within the Lipinski’s limit range (Beg et al. 2020). Whereas the Clogs for these compounds ranges from -7.48 to -6.45, this implies poor solubility of the synthesized derivatives in water. As a general principle, if the bioactivity score is large, there is a high possibility that the compound under investigation will be active. Therefore, a compound having bioactivity score of more than 0.00 is most likely to have a significant biological activity, while values between -0.50 to 0.00 are expected to be moderately active, and if the score is less than -0.50 it is presumed to be inactive (Ezekiel A et al. 2017). As for the log Kp for the skin permeability, which reflects the transport of the molecules through the epidermis, the more negative the log Kp, the less the skin permeates the molecule (
The extent of absorption of GIT molecules appears to be low, yet, these derivatives possess low penetration to the blood brain barrier (BBB), and would have no effect on CNS activity. Finally, the data generated in (Table
Properties | Comp1 | Comp 2 | Comp3 | Comp 4 | Comp 5 | Comp 6 | Comp 7 | Comp 8 | Comp 9 | Comp 10 |
M.wt | 464.89 | 448.44 | 509.34 | 430.45 | 444.47 | 446.88 | 430.42 | 491.33 | 412.43 | 426.46 |
c log p < 5 | 5.36 | 5.09 | 5.55 | 4.88 | 5.03 | 6.15 | 5.88 | 6.24 | 5.60 | 5.9 |
c log s | -7.04 | -6.61 | -7.36 | -6.45 | -6.75 | -7.17 | -6.73 | -7.48 | -6.58 | -6.87 |
nHBA ≤10 | 4 | 5 | 4 | 4 | 4 | 5 | 6 | 5 | 5 | 5 |
nHBD ≤5 | 4 | 4 | 4 | 4 | 4 | 2 | 2 | 2 | 2 | 2 |
nrtb | 9 | 9 | 9 | 9 | 9 | 6 | 6 | 6 | 6 | 6 |
n. violation | 1 | 1 | 2 | 1 | 1 | 1 | 1 | 1 | 1 | 1 |
B.A score | 0.55 | 0.55 | 0.17 | 0.55 | 0.55 | 0.55 | 0.55 | 0.55 | 0.55 | 0.55 |
(log Kp) cm/s | -4.08 | -4.35 | -4.30 | -4.31 | -4.14/ | -4.08 | -4.36 | -4.31 | -4.32 | -4.15 |
G.I. absorption | low | Low | low | low | low | low | low | low | low | low |
Mutagenic | None | None | None | None | None | None | None | None | None | None |
Tumorigenic | None | None | None | None | None | None | None | None | None | None |
Irritant | None | None | None | None | None | None | None | None | None | None |
LL score | 3 | 3 | 3 | 3 | 3 | 2 | 2 | 2 | 2 | 2 |
TPSA≤ 140 ⁰A | 97.28 | 97.28 | 97.28 | 97.28 | 97.28 | 78.06 | 78.08 | 78.08 | 78.08 | 78.08 |
BBB | NO | NO | NO | NO | NO | NO | NO | NO | NO | NO |
Novel thiadiazole compounds (6–10) were synthesized from the parent flufenamic acid, and confirmed by spectroscopic analysis, including AT-IR, 1HNMR and CHNS. Their cytotoxic activity against two human cancer cell lines (A549 and MCF-7), and the non-tumorigenic MCF10A and WBC was evaluated by the MTT assay. Three of the five derivatives (compounds 6, 7 and 10) showed significant cell growth inhibition against MCF-7 cell line. Compound 7 exhibited the lowest and more consistent anticancer activity against MCF-7 cell line (GI50 = 63.9 µg/mL), with no effect on A549 cell line or normal cells.
Compound 7 caused cytotoxicity against MCF-7 breast cancer cells by induction of apoptosis, as indicated by fragmented nuclei after DAPI staining, and agarose gel electrophoresis. Moreover, compound 7 led to an increase in caspase 9 mRNA level and its activation. Also, compound 7-treated MCF-7 cells showed enhanced cytochrome c release from the mitochondria to the cytosol, indicating an induction of the intrinsic apoptotic pathway. Finally, compound 7 showed EGFR kinase inhibitory activity (EC50 = 0.13 µM), which was matched by the molecular docking studies, that showed compound 7 may be considered as plausible inhibitor of EGFR kinase. This study could serve as a basis for optimizing other derivatives for better interaction with EGFR kinase enzyme.
We thank the Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad for their support, and work facilities in the laboratory.