Skip to main content

Novel tetrahydroisoquinolines as DHFR and CDK2 inhibitors: synthesis, characterization, anticancer activity and antioxidant properties

Abstract

In this study, we synthesized new 5,6,7,8-tetrahydroisoquinolines and 6,7,8,9-tetrahydrothieno[2,3-c]isoquinolines based on 4-(N,N-dimethylamino)phenyl moiety as expected anticancer and/or antioxidant agents. The structure of all synthesized compounds were confirmed by spectral date (FT-IR, 1H NMR, 13C NMR) and elemental analysis. We evaluated the anticancer activity of these compounds toward two cell lines: A459 cell line (lung cancer cells) and MCF7 cell line (breast cancer cells). All tested compounds showed moderate to strong anti-cancer activity towards the two cell lines. Compound 7e exhibited the most potent cytotoxic activity against A549 cell line (IC50: 0.155 µM) while compound 8d showed the most potent one against MCF7 cell line (IC50: 0.170 µM) in comparison with doxorubicin. In addition, we examined the effect of compounds 7e and 8d regarding the growth of A549 and MCF7 cell lines, employing flow cytometry and Annexin V-FITC apoptotic assay. Our results showed that compound 7e caused cell cycle arrest at the G2/M phase with a 79-fold increase in apoptosis of A459 cell line. Moreover, compound 8d caused cell cycle arrest at the S phase with a 69-fold increase in apoptosis of MCF7 cell line. Furthermore, we studied the activity of these compounds as enzyme inhibitors against several enzymes. Our findings by docking and experimental studies that compound 7e is a potent CDK2 inhibitor with IC50 of 0.149 µM, compared to the Roscovitine control drug with IC50 of 0.380 µM. We also found that compound 8d is a significant DHFR inhibitor with an IC50 of 0.199 µM, compared to Methotrexate control drug with IC50 of 0.131 µM. Evaluation of the antioxidant properties of ten compounds was also studied in comparison with Vitamin C. Compounds 1, 3, 6, 7c and 8e have higher antioxidant activity than Vitamin C which mean that these compounds can used as potent antioxidant drugs.

Graphical Abstract

Peer Review reports

Introduction

Nowadays cancer is one of the most dangerous diseases in the world and it has risen to the position of the leading cause of death around the globed due to the inherent resistance of many types of cancer to conventional radiotherapy and chemotherapy [1]. So many strategies have been admitted treating cancer patients. One modality is through inhibition of cell cycle regulators enzymes of cancer cells such as inhibition of CDKs [2] and DHFR enzymes [3], epidermal growth factor (EGF) [2], Ras, and Tubulin proteins [4]. CDKs (cyclin-dependent kinases) are serine/threonine kinases enzymes that play a crucial role in regulating eukaryotic cell cycle [5], apoptosis, differentiation, and transcription. So, controlling CDKs activity has emerged as a promising therapeutic approach [5, 6]. CDK2 is one of CDK families which exist as an inactive form [5, 6], upon binding to its regulatory partners cyclin A or cyclin E. Which formed a functional heterodimeric complex to control cell cycle progression [7, 8]. Previous studies found that CDK2 is over-activated in many types of cancer [8]. Which makes CDK2 inhibitions is a desirable target for cancer treatment [9, 10]. CDK2 inhibitors could be classified as ATP-competitive and non-ATP-competitive based on their binding site [11]. Roscovitine and Flavopiridol are the most common commercial CDK2 inhibitors drugs where their structure based on heterocyclic moiety [12].

Dihydrofolate reductase enzyme (DHFR) is responsible for reduction of dihydrofolate (DHF) to tetrahydrofolate (THF). THF is essential for DNA synthesis, cell growth, and the production of raw materials for cell proliferation in both normal and cancer cells [13]. Therefor inhibitions of DHFR is an important target to prevent cell spreading [14]. Moreover DHFR enzyme required to maintain bacterial growth [15, 16]. Due to its critical role in nucleotide biosynthesis. Hence inhibitors of DHFR have been proven in as effective agents for treating bacterial infections [16]. Methotrexate is the most effective commercial drug for DHFR inhibition which contain heterocyclic atoms. In addition it has been approved to be effective in reducing cancer symptoms in children with acute lymphoblastic leukemia [14, 15].

Generally heterocyclic compounds were reported to be used as CDK2 inhibitors as reported in previous work such as pyridazines derivatives [5]. Oxindoles compounds [7], 6-Substituted 2-Arylaminopurines compounds [8], and Thiazolone compounds [11]. In addition, Recent literature showed that all new DHFR inhibitors contain heterocyclic moieties in their structure such as pyridine, quinoline and isoquinoline moieties [14, 17].

Isoquinoline ring is one of the heterocyclic compounds which reported to has various biological activities, including antimicrobial [18], anti-oxidant [19], anti-inflammatory [19, 20], antipyretic [20], antihypertensive [21], antitumor [22,23,24,25] and anti-proliferative effects [26, 27]. Many isoquinoline alkaloids, including cepharanthine, berberine, and tetrandrine, have shown anti-inflammatory effect [28]. Therefore, a huge effort has been spent in developing novel and effective isoquinoline derivatives. Furthermore, increased interest in partially hydrogenated isoquinoline derivatives is related to the presence of an isoquinoline fragment in molecules of many alkaloids, which give new biologically active compounds. Synthetic 1,2,3,4- and 5,6,7,8-tetrahydroisoquinoline derivatives were reported to exhibit antitumor [29,30,31,32], antihypertensive and neurotropic activities [33].

In view of the above observations, the current work was designed to synthesize and characterize some new (5,6,7,8-tetrahydroisoquinolin-3-yl)Thio compounds and related 6,7,8,9-tetrahyrothieno[2,3-c]isoquinolines incorporating 4-(N,N-dimethylamino) phenyl moiety to be examined as anticancer agents and antioxidant drugs. Dimethylamino moiety was chosen in this work because of its remarkable antioxidant activities [34] as they associate to the proton donors active groups in the surfaces like amino or methyl groups. These groups can interact by inter molecular reactions on the surface of DPPH to give antioxidant activities through hydrogen atom transfer reaction [35] in comparison with vitamin C drug. In addition to the tetrahydroisoquinolines anticancer [31, 32] properties in comparison with doxorubicin control and compounds 7e and 8d were the most potent compounds. Furthermore, the effect of compounds 7e and 8d on induced apoptosis and cell cycle arrest of the cancer cell lines were also included. Moreover, the enzyme inhibitory activities and molecular docking of two selective tetrahydroisoquinolines 7e and 8d were studied.

Materials and methods

Chemicals and instrumentations

Chemicals: chemicals of this work (4-(N,N-dimethylaminobenzaldhyde, Cyanothioacetamide, Piperidine, Methyl iodide, Ethyl Chloroacetate, 2-Chloroacetamide, Chloroacetonitrile or N-aryl-2-Chloroacetamides, Ethanol, Sodium acetate.3H2O, Sodium carbonate) were purchased from Sigma Aldrich Co.

Instrumentations: Melting points were determined on a Gallan-Kamp apparatus and are uncorrected. The purity of the compounds was ensured by TLC and the spectroscopic analysis.

IR spectra were recorded on a Shimadzu 470 IR-spectrophotometer (KBr; νmax in cm−1). The 1H and 13C NMR spectra were recorded on Varian A5 500 MHz spectrometer using DMSO-d6 as a solvent and tetramethylsilane (TMS) as an internal reference. Coupling constants (J values) are given in Hertz (Hz). Elemental analyses were performed on a Perkin Elmer 2400 LS Series CHN/O analyzer.

Cell lines: The in vitro human breast cancerous cell line (MCF7), lung cancerous  cell lines (A549) and normal cell lines were purchased from Serum and Vaccine formulation in Cairo-Egypt.

Molecular docking: Molecular docking studies were performed in (I Mole Lab for bioinformatics, Cairo, Egypt).

Softwares: The biological data was analyzed and plot by Graphpad prism, Cell qust, ANOVA, Origin Lab, AutoDock Vina 1.1.2, Mestrenova and Excel software.

7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylaminophenyl)-5,6,7,8-tetrahydroisoquinoline-3(2H)-thione (1)

A mixture of 2,4-diacetyl-5-hydroxy-5-methyl-3-(4-(N,N-dimethylaminophenyl) cyclohexanone (3.3 g,10 mmol), 2-cyanothioacetamide (1.0 g,10 mmol) and piperidine (0.8 mL, 10 mmol) in ethanol (30 mL) was refluxed for 2 h. The yellow crystals that formed on cooling were collected, washed with methanol, and dried in air to give compound 1. Yield: 98%; m. p: 283–284 °C. IR: 3432 (O–H), 3273 (N–H); 3142 (C–H, sp2); 2885 (C–H, sp3); 2216 (C≡N); 1709 (C=O); 1619 (C=N). 1H NMR: δ 13.78 (s, 1H, NH); 6.88 (d, 2H, J = 10 Hz, Ar–H); 6.61 (d, J = 10 Hz, 2H, Ar–H), 4.83 (s, 1H, OH); 4.27 (d, J = 10 Hz, 1H, C8H); 3.45 (d, J = 10 Hz, 2H: C5H and C7H), 3.28(s,1H, C6H) 2.87 (m, 7H: C5H and N(CH3)2); 2.09 (s, 3H, CH3, attached to C-1); 1.90 (s, 3H, COCH3); 1.24 (s, 3H, CH3) ppm. 13C NMR: δ 209.97, 182.75, 178.99, 174.94, 155.49, 155.41, 152.98, 149.21, 129.18, 129.03, 125.05, 116.90, 113.88, 113.01, 68.16, 68.07, 66.22, 56.49, 31.55, 28.11, 28.01, 19.01 ppm. Anal. Calcd. for C22H25N3O2S (395.17): C, 66.81; H, 6.37; N, 10.62%. Found: C, 66.61; H, 6.40; N, 10.78%.

Reaction of compound 1 with methyl iodide, ethyl chloroacetate, 2-chloroacetamide, chloroacetonitrile or N-aryl-2-chloroacetamides 2a–e: synthesis of compounds 3, 4, 5, 6 and 7a–e

A mixture of 1 (3.95 g, 10 mmol), methyl iodide (0.7 mL, 10 mmol), ethyl chloroacetate (1 mL, 10 mmol), 2-chloroacetamide (0.93 g, 10 mmol), chloroacetonitrile (0.8 mL, 10 mmol)or N-aryl-2-chloroacetamide 2a–e (10 mmol), and sodium acetate trihydrate (1.50 g, 11 mmol) in ethanol (100 mL) was refluxed for one hour. The reaction mixture was then allowed to stand at room temperature overnight. After that the precipitate was collected and recrystallized from ethanol as colorless crystals of title compounds 3, 4, 5, 6, and 7a–e respectively.

7-Acetyl-4-cyano-1,6-dimethyl-3-methylthio-6-hydroxy-8-(4-N,N-dimethyl-aminophenyl)-5,6,7,8-tetrahydroisoquinoline (3)

Yield: 94%; m.p.: 162–163 °C. IR: 3510 (O–H); 2967, 2909 (C–H, sp2); 2217 (C≡N); 1696 (C=O, acetyl); 1612 (C=N). 1H NMR: δ 6.83 (d, J = 5 Hz, 2H, Ar–H); 6.61 (t, J = 5 Hz, 2H, Ar–H); 4.78 (s, 1H, OH), 4.39 (d, J = 5 Hz, 1H, C8H), 3.18 (dd, J = 7,10 Hz, 3H: C7H and C5H2), 2.86 (m, 9H: SCH3 and N(CH3)2), 2.11(d, J = 7 Hz, 3H, at C-1), 2.00 (s, 3H, CH3, COCH3), 1.25(d, J = 10 Hz, 3H, CH3) ppm. 13C NMR: δ 209.69, 165.72, 161.03, 157.43, 149.23, 148.72, 130.71, 130.08, 128.60, 115.30, 112.40, 104.17, 67.58, 66.31, 43.28, 42.06, 31.12, 27.61, 24.78, 23.73, 14.54. Anal. Calcd. for C23H27N3O2S (409.18): C, 67.45; H, 6.65; N, 10.26%. Found: C, 67,42; H: 6.58, N; 10.30%.

Ethyl 2-((7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio)acetate (4)

Yield: 78%; m.p.: 159–160 °C. IR: 3506 (O–H); 2983, 2964, 2809 (C–H, sp3); 2215 (C≡N); 1740 (C=O, ester); 1695 (C=O, acetyl). 1H NMR: δ 6.81 (d, J = 10 Hz, 2H, Ar–H), 6.58 (d, J = 10 Hz, 2H, Ar–H), 4.81 (s, 1H, OH), 4.38 (d, J = 9 Hz, 1H, C8H), 4.05 (m, 4H: SCH2 and OCH2), C5H and), 3.22 (d, J = 10 Hz, 1H, C5H), 2.87 (d, J = 10 Hz, 8H: C7H, C5H and N(CH3)2), 2.11 (s, 3H, CH3, at C-1), 1.93 (s, 3H, COCH3), 1.25 (s, 3H, CH3), 1.12 (d, J = 5 Hz, 3H, CH3 of ester group) ppm. 13C NMR: δ 209.62, 168.58, 160.96, 156.15, 149.43, 148.74, 130.57, 128.63, 115.09, 112.38, 103.71, 67.59, 66.28, 60.90, 42.02, 40.00, 31.98, 31.10, 27.58, 24.49, 14.00. Anal. Calcd. for C26H31N3O4S(481.20): C, 64.84; H, 6.49; N, 8.72%. Found: C, 64.98; H, 6.44; N, 8.51%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]acetamide (5)

Yield: 85%; m.p.: 196–197 °C. IR: 3562 (O–H); 3436, 3295, 3181 (NH2); 2971, 2809 (C–H, sp3); 2219 (C≡N); 1698 (C=O, acetyl); 1667 (C=O, amide). 1H NMR: δ 7.50 (s, 1H, NH), 7.05 (s, 1H, NH), 6.82 (d, J = 10 Hz, 2H, Ar–H), 6.60 (d, J = 9 Hz, 2H, Ar–H), 4.75 (s, 1H, OH), 4.39 (d, J = 15 Hz, 1H, C8H), 3.88 (d, J = 12 Hz, 15 Hz, 2H, SCH2), C5H and), 3.26 (d, J = 10 Hz, 1H, C5H), 2.89 (m, 8H: C7H, C5H and N(CH3)2), 2.11 (s, 3H, CH3, at C-1), 1.99 (s, 3H, COCH3), 1.26 (s, 3H, CH3) ppm. 13C NMR: δ 210.02, 169.55, 161.45, 157.33, 149.80, 149.20, 131.21, 130.84, 129.14, 115.73, 112.95, 104.19, 68.07, 66.77, 43.77, 42.50, 33.82, 31.59, 28.07, 25.11.

Anal. Calcd. for C24H28N4O3S (452.19): C, 63.69; H, 6.24; N, 12.38%. Found: C, 63.37; H, 6.18; N, 12.41%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]acetonitrile (6)

Yield:90%; m.p.: 145 °C. IR: 3537 (O–H); 2966, 2924,2801 (C–H, sp3); 2246 (C≡N, non conjugated); 2217 (C≡N, conjugated); 1698 (C=O, acetyl). 1H NMR: δ 6.85 (d, J = 10 Hz, 2H, Ar–H), 6.61 (d, J = 10 Hz 2H, Ar–H), 4.79 (s, 1H, OH), 4.44 (d, J = 8 Hz, 1H, C8H), 4.32 (s, 2H, SCH2), 3.27 (d, 1H, C5H), 2.92(d, J = 8 Hz, 2H, C7H and C5H), 2.89 (d, J = 10 Hz, 6H: N(CH3)2), 2.12 (s, 3H, CH3, at C-1), 2.07 (s, 3H, COCH3), 1.27 (s, 3H, CH3) ppm. 13C NMR: δ 210.26, 162.04, 154.32, 150.40, 149.25, 132.05, 130.91, 129.19, 118.20, 115.25, 112.95, 104.66, 68.09, 66.69, 43.83, 42.55, 31.63, 27.98, 25.14, 15.74 ppm. Anal. Calcd. for C24H26N4O2S (434.18): C, 66.33; H, 6.03; N, 12.89%. Found: C, 65.72; H, 5.71; N, 13.09%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-phenylacetamide (7a)

Yield: 80%; m.p.: 209–210 °C. IR: 3459 (O–H); 3247 (N–H); 2971, 2805 (C–H, sp3); 2211 (C≡N); 1706 (C=O, acetyl); 1683 (C=O, amide). 1H NMR: δ 10.21 (s, 1H, NH), 7.52 (d, J = 10 Hz, 2H, Ar–H), 7.27 (t, J = 10 Hz, 2H, Ar–H), 7.02 (m, 1H, Ar–H), 6.80 (d, J = 10 Hz, 2H, Ar–H), 6.57 (d, J = 10 Hz, 2H, Ar–H), 4.80 (s, 1H, OH), 4.37 (d, J = 10 Hz, 1H, C8H), 4.1 (dd, J = 10 Hz, 13 Hz, 2H, SCH2), 3.23(d, J = 17 Hz, 1H, C5H), 2.87 (m, 4H: C7H and C5H), 2.83 (s, 6H, N(CH3)2), 2.10 (s, 3H, CH3, at C-1), 1.92 (s, 3H, COCH3), 1.24 (s, 3H, CH3). 13C NMR: δ 217.44, 209.63, 166.10, 160.95, 156.74, 149.35, 148.72, 138.90, 130.61, 130.48, 128,69, 128.61, 123.26, 119.04, 115.19, 112.38, 103.66, 67.57, 66.28, 43.29, 41.99, 34.68, 31.06, 27.58, 24.54. Anal. Calcd. for C30H32N4O3S (528.22): C, 68.16; H, 6.10; N, 10.60%. Found: C, 68.10; H, 6.15; N, 10.46%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-(4-tolyl)acetamide (7b)

Yield: 95%; m.p.:198–199 °C. IR: 3436 (O–H); 3251 (N–H); 3119 (C–H, sp2); 2964,2908 (C–H, sp3); 2216 (C≡N); 1706 (C=O, acetyl); 1675 (C=O, amide). 1H NMR: δ 10.11 (s, 1H, NH), 7.39 (d, J = 9 Hz, 2H, Ar–H), 7.07 (d, J = 8 Hz, 2H, Ar–H), 6.80 (d, J = 9 Hz, 2H, Ar–H), 6.57 (d, J = 9 Hz, 2H, Ar–H), 4.79 (s, 1H, OH), 4.37 (d, J = 10 Hz, 1H, C8H), 4.085 (dd, J = 4, 7 Hz, 2H, SCH2), 3.23 (d, J = 17 Hz, 1H, C5H), 2.87 (m, 2H, C7H and C5H), 2.83 (s, 6H, N(CH3)2), 2.22 (s, 3H, CH3 of 4-tolyl group), 2.10 (s, 3H, CH3, at C-1), 1.92 (s, 3H, COCH3), 1.24 (s, 3H, CH3) ppm. 13C NMR: δ 209.62, 165.83, 160.93, 156.77, 149.33, 148.70, 136.40, 132.16, 130.61,130.45, 129.06, 128.61, 119.05, 115.18, 112.37, 103.64, 67.56, 66.27, 43.28, 41.98, 34.64, 31.05, 27.57, 24.53, 20.38 ppm. Anal. Calcd. For C31H34N4O3S (542.24): C, 68.61; H, 6.31; N, 10.32%. Found: C, 68.52; H, 6.45; N, 10.11%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-,5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-(4-chlorophenyl)acetamide (7c)

Yield: 96%; m.p.: 214–215 °C. IR: 3458 (O–H); 3242 (N–H); 2966, 2804 (C–H, sp3); 2214 (C≡N); 1685 (2C=O, acetyl and amide); 1610 (C=N). 1H NMR: δ 10.36 (s, 1H, NH), 7.55 (d, J = 10 Hz, 2H, Ar–H), 7.32 (t, J = 10 Hz, 2H, Ar–H), 6.80 (d, J = 9 Hz, 2H, Ar–H), 6.57 (d, J = 8 Hz, 2H, Ar–H), 4.80 (s, 1H, OH), 4.37 (d, J = 10 Hz, 1H, C8H), 4.11 (dd, J = 12,15 Hz 2H, SCH2), 3.23 (d, J = 17 Hz, 1H, C5H), 2.89 (m, 2H, C7H and C5H), 2.84 (s, 6H, N(CH3)2), 2.10 (s, 3H, CH3, at C-1), 1.90 (s, 3H, COCH3), 1.25 (s, 3H, CH3) ppm. 13C NMR: δ 209.62, 166.32, 160.93, 156.67, 149.34, 148.71, 137.86, 130.58, 130.49,128.60, 126.81, 120.56, 115.16, 112.36, 103.66, 67.51, 66.26, 43.28, 41.99, 34.69, 31.07, 27.57, 24.50 ppm. Anal. Calcd. For C30H31ClN4O3S (562.18): C, 63.99; H, 5.55; N, 9.95%. Found: C, 64.15; H, 5.48; N, 9.84%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-(4-acetylphenyl)acetamide (7d)

Yield:93%; m.p.: 205 °C. IR: 3490 (O–H); 3244 (N–H); 3033 (C–H, sp2); 2922 (C–H, sp3); 2215 (C≡N); 1690 (3C=O, acetyl and amide); 1614 (C=N). 1H NMR: δ 10.62 (s, 1H, NH), 7.89 (d, J = 10 Hz, 2H, Ar–H), 7.67 (d, J = 10 Hz, 2H, Ar–H), 6.80 (d, J = 13 Hz, 2H, Ar–H), 6.55 (d, 2H, Ar–H), 4.80 (s, 1H, OH), 4.36 (d, J = 10 Hz, 1H, C8H), 4.15 (dd, J = 11, 13 Hz 2H, SCH2), 3.23 (d, J = 20 Hz, 1H, C5H), 2.87 (d, J = 12 Hz, 2H, C7H and C5H), 2.82 (s, 6H, N(CH3)2), 2.50 (s, 3H, COCH3 attached to phenyl group and overlapped with solvent proton), 2.10 (s, 3H, CH3, at C-1), 1.88 (s, 3H, COCH3), 1.24 (s, 3H, CH3). 13C NMR: δ 209.26, 196.36, 166.83, 160.75, 156.63, 149.38, 148.70,143.72, 131.68, 130.57, 129.45, 128.61, 118.22,115.33, 112.57, 103.72, 67.57, 66.27, 43.28, 41.97, 34.82, 31.05, 27.57, 26.34, 24.47. Anal. Calcd. for: C32H34N4O4S: (570.23): C, 67.35; H, 6.00; N, 9.82%. Found: C, 67.00; H, 5.88; N, 9.79%.

2-[(7-Acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-N,N-dimethylamino-phenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-(naphthalen-1-yl)acetamide (7e)

Yield: 88%; m.p.: 194–195 °C. IR: 3506 (O–H); 3288 (N–H); 3114 (C–H, sp2); 2968–2804 (C–H, sp3); 2217 (C≡N); 1696 (2 C=O, acetyl and amide); 1611 (C=N). 1H NMR: δ 10.20 (s, 1H, NH), 7.94 (d, J = 10 Hz, 2H, Ar–H), 7.75 (d, J = 7 Hz, 1H, Ar–H), 7.57 (d, J = 8 Hz, 1H, Ar–H), 7.46 (d, J = 10, 2H, Ar–H), 7.33 (m, 1H, Ar–H), 6.85 (d, J = 9 Hz, 2H, Ar–H), 6.59 (d, J = 8 Hz, 2H, Ar–H), 4.83 (s, 1H, OH), 4.42 (d, J = 10 Hz, 1H, C8H), 4.30 (dd, J = 15, 17 Hz, 2H, SCH2), 3.27 (d, J = 20 Hz, 1H, C5H), 2.93 (d, J = 10 Hz, 1H, C7H), 2.86 (m, 7H: C5H and N(CH3)2), 2.12 (s, 3H, CH3, at C-1), 2.04 (s, 3H, COCH3), 1.27 (s, 3H, CH3). 13C NMR: δ 202.84, 166.93, 161.10, 156.89, 149.39, 148.73, 133.61, 130.86, 128.69, 128.03, 125.48, 122.72, 121.71, 115.36, 112.24, 103.43, 67.61, 66.28, 43.33, 42.07, 34.17, 31.14, 27.60, 24.69. Anal. Calcd. for: C34H34N4O3S (578.24): C, 70.56; H, 5.92; N, 9.68%. Found: C, 70.43; H, 5.89; N, 9.85%.

7-Acetyl-1-amino-2-(N-arylcarbamoyl)-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylaminophenyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinolines 8a–d: general procedures

Method A

To a suspension of 7a–e (10 mmol) in abs. ethanol (60 mL), anhydrous sodium carbonate (0.30 g) was added. The reaction mixture was refluxed for 3 h. The yellow crystals that formed while hot were collected, washed with water, dried in air, and then crystallized from dioxane to give 8a–e.

7-Acetyl-1-amino-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylaminophenyl)-N-phenyl-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamide (8a)

Yield: 96%; m.p.: 260 °C. IR:3501, 3451 (O–H, NH2 and NH); 3123 (C–H, sp2); 2990, 2810(C–H, sp3); 1695 (C=O, acetyl); 1631 (C=O, amide). 1H NMR: δ 9.40 (s, 1H, NH), 7.69 (d, J = 8 Hz, 2H, Ar–H), 7.33 (d, J = 8 Hz, 2H, Ar–H), 7.07 (m, 3H, Ar–H), 6.78 (br s, 2H, NH2), 6.59 (d, J = 9 Hz, 2H, Ar–H), 4.70 (br s, 1H, OH), 4.48 (d, J = 10 Hz, 1H, C6H), 3.57 (d, J = 17 Hz, 1H, C9H), 3.39 (d, 1H, C7H), 2.84 (m,7H: C9H and N(CH3)2), 2.14 (s, 3H, CH3, at C-5), 2.04 (s, 3H, COCH3), 1.29 (s, 3H, CH3). 13C NMR: δ 210.27, 164.37, 158.77, 155.97, 149.45, 148.61, 141.95, 138.89, 131.73, 130.04, 128.47, 128.34, 123.38, 122.88, 121.24, 112.43, 96.88, 67.18, 66.59, 42.39, 40.05, 31.19, 28.02, 24.65. Anal. Calcd. for C30H32N4O3S (528.22): C, 68.16; H, 6.10; N, 10.60%. Found: C, 68.02; H, 6.00; N, 10.27%.

7-Acetyl-1-amino-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylaminophenyl)-N-(4-tolyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamide (8b)

Yield:93%; m.p.: 289–290 °C. IR: 3394, 3327 (O–H, NH2, NH); 2915, 2798 (C–H, sp3); 1703 (C=O, acetyl); 1614 (C=N). 1H NMR: δ 9.33 (s, 1H, NH), 7.58 (s, 2H, Ar–H), 7.15 (s, 2H, Ar–H), 7.02 (d, J = 64 Hz, 2H, Ar–H), 6.76 (s, 2H, NH2), 6.59 (d, J = 10 Hz, 2H, Ar–H), 4.66 (s, 1H, OH), 4.48 (d, J = 94 Hz, 1H, C6H), 3.57 (m, 2H, C9H and C7H), 2.85 (m, 7H, C9H and N(CH3)2), 2.28 (s, 3H, CH3 of 4-tolyl group), 2.14 (s, 3H, CH3, at C-5), 2.03 (s, 3H, COCH3), 1.28 (s, 3H, CH3). 13C NMR: δ 209.98, 209.69, 164.24, 158.91, 158.61, 158.31, 158.14, 158.00, 155.72, 149.22, 142.52, 129.06, 123.34, 121,47, 121.42, 118.74, 118.28, 118.23, 118.19, 116.44, 114.14, 111.85, 103.52, 97.26, 67.27, 66,35, 43.94, 42.69, 31.03, 27.99, 24.46, 20.49 Anal. Calcd. for C31H34N4O3S (542.24) C, 68.61; H, 6.31; N, 10.32%. Found; C, 68.57; H, 6.66; N, 10.24%.

7-Acetyl-1-amino-N-(4-chlorophenyl)-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethyl-aminophenyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamide (8c)

Yield: 83%; m.p.: 295 °C. IR: 3416, 3325 (O–H, NH2, NH); 2916 (C–H, sp3);1703 (C=O, acetyl); 1614 (C=N. 1H NMR: δ 9.67 (s, 1H, NH), 7.93 (s, 2H, NH2), 7.65 (d, J = 10 Hz, 2H, Ar–H), 7.35 (d, J = 10 Hz, 2H, Ar–H), 7.24 (d, J = 10 Hz, 2H, Ar–H), 7.06(d, J = 10 Hz, 2H, Ar–H), 4.66 (s, 1H, OH), 3.59 (d, J = 17 Hz, 1H, C6H), 3.31 (d, 1H, C9H), 3.03 (m, 7H: C7H and N(CH3)2), 2.83 (d, J = 10, 1H, C9H), 2.14 (s, 3H, CH3, at C-5), 2.02 (s, 3H, COCH3), 1.29(s, 3H, CH3). 13C NMR: δ 165.34, 161.46, 158.98, 158.69, 158.39, 158.09, 155.94, 153.55, 149.8, 147.95, 143.16,139.75, 138.01, 129.51, 128.73, 128.36, 127.23, 123.26, 122.78, 118.39, 116.64,114.33, 112.03, 96.85, 67.06, 66.14, 44.02, 42.27,42,21, 31.12, 28.00, 24.48. Anal. Calcd. for C30H31ClN4O3S (562.18): C, 63.99; H, 5.55; N, 9.95%. Found: C, 64.15; H, 5.49; N, 9.62%.

7-Acetyl-N-(4-acetylphenyl)-1-amino-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylaminophenyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamide (8d)

Yield:89%; m.p.: 301–302 °C. IR: 3424 (O–H); 3320 (N–H); 2916 (C–H, sp3); 1705 (C=O, acetyl); 1681 (C=O, amide). 1H NMR: δ 9.71 (s, 1H, NH), 7.91 (m, 6H, Ar–H), 7.17 (d, J = 10 Hz, 2H, Ar–H), 7.04 (s, 2H, NH2), 4.63 (s, 1H, OH), 3.60 (d, J = 8 Hz,1H, C6H), 3.39 (d, J = 10 Hz, 1H, C9H), 3.02 (s, 6H, N(CH3)2), 2.84 (d, J = 10 Hz, 1H, C7H), 2.53 (s, 4H: C9H and COCH3 attached to phenyl group and ovellaped with solvent protons), 2.17 (s, 3H, CH3, at C-5), 2.03 (s, 3H, COCH3), 1.30 (s, 3H, CH3). 13C NMR: δ 202.91, 196.84,164.62, 159.00, 158.78, 158.70, 158.40, 158.00, 156.21, 150.29,143.71, 142.98, 131.74, 129.63, 129.37, 129.13,128.98, 123.01, 120.02,118.96, 117.58, 116.64, 114.34, 112.03, 96.38, 67.33, 66.28, 43.46, 42.72, 42.20, 31.13, 28.00, 26.43, 24.57. Anal. Calcd. for: C32H34N4O4S: (570.23): C, 67.35; H, 6.00; N, 9.82%. Found: C, 67.51; H, 6.09; N, 9.74%.

7-Acetyl-1-amino-N-(naphthalen-1-yl)-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylminophenyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamide (8e)

Yield: 94%; m.p.: 288–290 °C. IR:3440, 3391 (O–H, NH2, NH); 3050 (C–H, sp2); 2910 (C–H, sp3); 1702 (C=O, acetyl); 1633 (C=O, amide). 1H NMR: δ 9.69 (s, 1H, NH), 7.51–7. 95 (m, 7H, Ar–H of 2-naphthyl group), 6.97 (br s, 2H, NH2), 6.78 (d, J = 15 Hz, 2H, Ar–H), 6.60 (d, J = 17 Hz, 2H, Ar–H), 4.65 (s, 1H, OH), 4.50 (d, J = 16 Hz,1H, C6H), 3.55 (d, J = 17 Hz, 1H, C9H), 3.38 (d, J = 13 Hz, 1H, C7H), 2.86 (d, J = 12 Hz, 7H: C9H and N(CH3)2), 2.14 (s, 3H, CH3, at C-5), 2.04 (s, 3H, COCH3), 1.28 (s, 3H, CH3). 13C NMR: δ 200.05, 165.03, 158.52, 156.05, 148.68, 141.88, 133.34, 131.80, 129.92, 128.47, 125.87, 123.46, 112.45, 67.23, 66.27, 42.63, 41.96, 31.4, 28.02, 24.63. Anal. Calcd for: C34H34N4O3S (578.24): C, 70.56; H, 5.92; N, 9.68%. Found: C, 70.79; H, 5.79; N, 9.42%.

Method B

A mixture of 1 (3.95 g, 10 mmol), N-aryl-2-chloroacetamide 2a–e (10 mmol) and anhydrous sodium carbonate (1.35 g) in ethanol (100 mL) was refluxed for three hours. The precipitate that formed on cooling was collected and recrystallized from dioxane as yellow crystals of 8a–e (94–98%).

Biological evaluation

In vitro cytotoxic activity

In vitro cytotoxic activity of all synthesized compounds against two human breast cell line (MCF7) and lung cell lines (A549) was evaluated according to the MTT method [23,24,25, 37, 38]. Firstly, Growth the cell line medium in 96 well tissue culture plate was injected with 105 cells/mL (100 uL/plate well) of the cell line and incubated at 37 °C for 24 h to develop a monolayer sheet then the formed growth medium was poured from 96 well microtiter plates after the confluent sheet of cells. After that preparing the isoquinoline samples stock solutions in DMSO and diluted the concentrations to started from 0.0487, 0.0975, 0.195, 0.391, 0.781, 1.562, 3.125, 6.25, 12.50, 25.00 µM. Secondly, add 0.1 mL of each concentration tetrahydroisoquinoline sample to each plate. The plates were incubated at 37 °C. Thirdly MTT solution (5 mg/mL in PBS) is prepared. Add 20 µL of MTT solution to each well plates. And shaking in 150 rpm for 5 min, to mix the MTT into the media. Then incubate at (37 °C, 5% CO2) for 1–5 h. Finally read the optical density at 560 nm and subtract background at 620 nm.

Cell cycle analysis

The cell cycle arrests of compound 7e against A549 and compound 8d against MCF7 at their IC50 values were carried out according to Abcam method (code ab139418), (www.abcam.co.jp). Thus, A549 and MCF7 cells were collected and fixed with 75% ice-cold ethanol before being stored at − 20 °C for 1 h after being treated with an IC50 dose of our compounds 7e, 8d. Then centrifuged the cells and washed twice with ice-cold PBS, and incubated for 20 min at 4 °C. A cell cycle assay was used to assess the cell cycle (Propidium Iodide Flow Cytometry Kit [ab13941]. Then perform statistical analysis for the result by the Cell quest software on the cell fractions in sub-G0/G1, S, and G2/M phases [38].

Annexin-V FITC apoptosis assay

The Annexin-V FITC apoptosis assay of compounds 7e against A549 cell line and 8d against MCF7 cell line at their IC50 values were carried out according to (BioVision) protocol (code k101-25). (www.biovision.com). Thus, cell line were treated with the IC50 concentration of the compounds for 24 h then collected by trypsin, and centrifuged then rinsed with PBS and suspended in 0.5 mL of binding buffer, then dual-stained with Annexin V-FITC (5 μL) and propidium iodide (5 μL) in the dark for 15 min at RT. These stained cells were measured using flow cytometry with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. The results were then analyzed with the Cell quest software [39,40,41].

Molecular docking

Protein preparation

The three-dimensional crystal structures of cyclin-dependent kinase 2 (CDK2, PDB ID 1AQ1) and dihydrofolate reductase (DHFR, PDB ID 1BOZ) were obtained from the Protein Data Bank (PDB). The protein structures were prepared using AutoDockTools 1.5.6. All water molecules were removed and hydrogen atoms were added. Gasteiger charges were assigned and nonpolar hydrogen were merged.

Ligand preparation

The 3D structures of ligand 1 (7e compound) and ligand 2 (8d compound) were built and energetically minimized using Avogadro 1.2.0 with the MMFF94 force field. Ligand atom types were assigned and rotatable bonds were defined using AutoDockTools. Both ligands were converted to PDBQT format required for docking calculations.

Molecular docking

Molecular docking studies were performed in (I Mole Lab for bioinformatics, Cairo, Egypt) by using AutoDock Vina 1.1.2. For each protein target, a docking grid box was generated to cover the active site based on a co-crystalized ligand. The exhaustiveness parameter was set to 8. Docking was performed with the prepared proteins and ligands to generate 9 binding poses per ligand. The best binding poses based on docking score were visually analyzed using Biovia Discovery Studio 2020 for interactions with key active site residues.

CDK2 inhibitors assay

The CDK2/cyclin A2 protein kinase assay was performed according to the bioscience protocol (code #79599) (www.bpsbioscience.com).

Firstly, prepare the master mixture (6 μL of 5 × Kinase assay buffer 1 + 1 µL of ATP (500 µM) + 5 µL of 10 × CDK substrate peptide 1 + 13 μL of distilled water).then add 25 μL of master mixture to every well of the 96-well plate. Add 20 ng of Cyclin A2 and 30 ng of different CDK2 mutant protein into the wells as indicated along with 0.155 µM of our synthesized compound 7e. Incubate at 30 °C for 45 min. After the 45-min reaction, add 50 µL of Kinase-Glo Max reagent to each well. After that cover the plate with aluminum foil and incubate the plate for 15 min at RT. Then Measure luminescence after subtracted The value of blank from all readings using the microplate reader. The relative kinase activity of Cyclin A2/wild-type CDK2 group is set as 100%. The data was analysied and plot by Graphpad prism software [42, 43].

DHFR inhibitors assay

The DHFR inhibitors assay kit was performed according to abcam (code ab283374); (www.abcam.co.jp).

Firstly, Dilute 2 μL Dihydrofolate Reductase with798 μL DHFR Assay Buffer. Then add 98 μL of diluted Dihydrofolate Reductase into desired well(s) containing the out synthesized 8d compound. Add 40 μL of diluted NADPH to each well containing the test samples. Incubate at room temperature for 10–15 min. Add 60 μL of diluted DHFR substrate to each well containing the test samples vortex briefly and keep on ice. Measure the absorbance immediately at 340 nm. Then calculate the inhibition concentration of 8d compound. The data was analyzed and plot by Graphpad prism software [44, 45].

Antioxidant activity

The antioxidant activity of ten compounds was determined using DPPH [32,33,34]. A solution 1: prepared by dissolving DPPH (0.002 g) in ethanol (50 mL etnanol). Solution 2: prepared by dissolving different weights 0.05, 0.01 g of each sample in 1 mL of DMSO then take 10 µL of each sample solution with 1 mL ethanol. Then mix 1 mL of solution 1 with 1 mL of solution 2 then vortex the resulting mixture in the dark for about 30 min. The absorbance of the mixture was measured by spectrophotometer at λmax = 517 nm against blank 1 mL absolute ethanol and compared to the ascorbic acid (Vitamin C).

Results and discussion

Synthesis

Refluxing of 2,4-diacetyl-5-hydroxy-5-methyl-3-(4-(N,N-dimethylaminophenyl) cyclohexanone with 2-cyanothioacetamide in ethanol in the presence of piperidine as a basic catalyst resulted in regioselective cyclocondensation reaction affording, 7-acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-(N,N-dimethylaminophenyl)-5,6,7,8-tetrahydroiso-quinoline-3(2H)-thione (1) in 98% yield (Scheme 1).

Scheme 1
scheme 1

Synthesis of compounds 1,3–6,7a–e and 8a–e

Compound 1 underwent S-alkylation reactions upon treatment with some halo reagents namely; methyl iodide, ethyl chloroacetate, 2-chloroacetamide, chloroacetonitrile or N-aryl-2-chloroacetamides 2a–e in refluxing ethanol containing slightly excess molar amounts of sodium acetate trihydrate to give 3-ethylthio-5,6,7,8-tetrahydroisoquinoline 3, ethyl (5,6,7,8-tetrahydroisoquinolin-3-ylthio)acetate 4, (5,6,7,8-tetrahydroisoquinolin-3-ylthio)acetamide 5, (5,6,7,8-tetrahydroisoquinolin-3-ylthio)acetonitrile 6 and 2-[(7-acetyl-4-cyano-1,6-dimethyl-6-hydroxy-8-(4-(N,N-dimethylaminophenyl)-5,6,7,8-tetrahydroisoquinolin-3-yl)thio]-N-arylacetamides 7a–e, respectively (Scheme 1).

On heating of compounds 7a–e with catalytic amounts of anhydrous sodium carbonate in abs. ethanol, they underwent intramolecular Thorpe-Ziegler cyclization affording 7-acetyl-1-amino-N-aryl-5,8-dimethyl-8-hydroxy-6-(4-N,N-dimethylamino-phenyl)-6,7,8,9-tetrahydrothieno[2,3-c]isoquinoline-2-carboxamides 8a–e in nearly quantitative yield (Scheme 1). Compounds 8a–e were also synthesized via reaction of 1 with the respective N-aryl-2-chloroacetamides 2a–e by heating in abs. ethanol in the presence of slightly excess molar amounts of anhydrous sodium carbonate (Scheme 1).

Characterization

The structures of all newly synthesized compounds were confirmed by FT-IR, 1H NMR and 13C NMR as well as elemental analyses (cf. Experimental part section and Additional file 1: Figs. S1–S45).

Anticancer activities

In vitro cytotoxicity

Our newly synthesized compounds 1, 3–6, 7a–e, and 8a–e were studied for their in vitro cytotoxic activities against two selective cell lines MCF7 and A549 (which our compounds show high activities towards them by using a way to drug predication program) by using the MTT assay method [36, 37]. In this work, doxorubicin was used as a positive control drug for comparison with the synthesized compounds under the same experimental conditions. Ten concentrations of each compound and doxorubicin ranging from 0.04875 to 25 μM were tested to reach the concentration which could cause death for 50% of the cancer cells (IC50). The cell viability and toxicity percentage are given in supplementary data (Additional file 1: Tables S1–S6), and summarized in Table 1 and Fig. 1. These results indicated that all synthesized compounds possess high cytotoxic activity against the two cell lines under investigation compared with that of doxorubicin, with IC50 values ranging from 0.117 to 3.800 μM (Table 1).

Table 1 Cytotoxicity (IC50) of compounds 1, 3–6, 7a–7e, 8a–e and doxorubicin as a standard against both MCF7, A549 cell lines
Fig. 1
figure 1

Anticancer activity of synthesized compounds compared with Doxorubicin as a standard at different concentrations from 0.048 to 25 μM. a- Compounds 1 and 3–6. b- Compounds 7a–7e. c- Compounds 8a–8e against MCF7 cell line respectively. d -Compounds 1 and 3–6. e -Compounds 7a–7e.f -Compounds 8a–8e against the A549 cell line respectively

In more details on structure–activity relationship, we noticed that: (i) the cytotoxic activity of compounds 1 and 3–6 against MCF7 cells obeys the order 6 > 3 > 5 > 1 > 4, whereas that of the same compounds obeys approximately opposite order against A549 cells as 4 > 5 > 1 > 3 > 6; (ii) 4-substituted phenylcarbamoylmethylthio derivatives 7b–d exhibited stronger cytotoxic activity than the parent unsubstituted one 7a against both MCF7 and A549 cell lines; (iii) among the arylcarbamoylmethylthioisoquinolines 7a–e and arylcarbamoyl thienoisoquinolines 8a–e, naphalen-1-yl derivative 7e exhibited the highest cytotoxic activity against A549 cell line and 4-chlorophenyl derivative 8d showed the highest activity against MCF7 cell line, respectively. Moreover the toxicity of these two compounds against normal human fetal lung fibroblast WI-38 cell line were investigated in this study which show that 7e and 8d compounds not toxic and safe for normal lung cell line with IC50 19.7 µM, 23.3 µM respectively in comparison with Doxorubicin  IC50 11.43 µM (Table 2) the test details presented in Additional file 1: Table S6a.

Table 2 Cytotoxicity (IC50) of compounds 7e and 8d and Doxorubicin against normal cell line WI-38 cell line

By calculating the selectivity index of these compounds 7e, 8d and Doxorubicin ((SI) = IC50 of compound in noncancerous cell line (WI-38)\IC50 of compound in cancer cell (A549)). They show very high selectivity index SI = 127, 45, 52 respectively. therefore these compounds belong of a selected potential anticancer drugs.Cyclization of arylcarbomyl-methylthioisoquinolines 7a and 7c into the corresponding arylcarbomylthienoiso-quinolines 8a and 8c resulted in increasing the anticancer activity towards both MCF7 and A549 cell lines; (v) cyclization of tolylcarbomylmethylthioisoquinolines 7b into the corresponding tolylcarbomylthieno[2,3-c]isoquinolines 8b decreases the anticancer activity towards MCF7 cell line and (vi) cyclization of carbomylmethylthioisoquinolines 7e into the corresponding carbomylthienoisoquinoline 8e decreases the anticancer activity towards both MCF7 and A549 cell lines (Fig. 1, and Table 1).

Cell cycle analysis in MCF7 and A549 Cells

The high cytotoxic activity of compound 7e against A549 (IC50 0.155 µM) and compound 8d against the MCF7 cell line (IC50 0.170 µM) prompted us to further investigate the growth inhibitory mechanism of the target conjugates to study the mechanism of the cell cycle by using flow cytometric analysis [46,47,48]. Both regulation of cell cycle progression and apoptosis induction have been considered significant strategies to control the proliferation of different cancer cells, accordingly, we primarily examined the growth inhibition mechanism of compounds 7e and 8d in relation to cell cycle progression and regulation in A549 and MCF7 cancer cells, respectively (Fig. 2, and Table 3).

Fig. 2
figure 2

Cell cycle analysis of A549 and MCF7 cells treated with compounds 7e and 8d

Table 3 Cell cycle analysis of A549 and MCF7 cells treated with compounds 7e and 8d

The impact on cell cycle distribution was assessed by a DNA flow cytometry analysis, through incubation of A549 cells with compound 7e at its IC50 concentration (IC50 0.155 µM) and incubation of MCF7 cells with compound 8d at its IC50 concentration (IC50 0.170 µM) for 48 h (Fig. 2). From the obtained results, it was found that: (i) A549 cells exposed to compound 7e significantly arrested at the G2/M phase of the cell cycle with an escalation in G2/M phase fraction from 11.60 (in control cells) to 23.61% (in 7e-treated A549 cells) and (ii) MCF7 cells treated with compound 8d had a significant decrease in G0-G1 and G2/M phases than control cells in contrast (iii) S phase was significantly increased in treated cells as an indication of cell cycle arrest; i.e. increased from 29.81 (control) to 37.92 (8d-treated cells). The antiproliferative mechanism of our compounds was explored from the aforementioned obtained result; compounds of type 7e compound arrested the cell cycle at G2/M phase of the cell cycle whereas compounds of type 8d compound arrested the cell cycle at S phase (Fig. 2).

Apoptosis assay in A549 and MCF7 cell lines

To further investigate whether the anti-proliferative activity for compound 7e or 8d is harmonious with the apoptosis induction [47,48,49,50] within A549 or MCF7 cells pointed out by the increased cell population in G2/M phase in 7e-treated A549 cells and S phase in 8d-treated MCF7 cells, respectively, and AnnexinV-FITC/PI dual staining analysis was used for the apoptosis assay (Fig. 3).

Fig. 3
figure 3

 Apoptosis results of compounds 7e and 8d on A549 and MCF7 cell lines respectively. a. Control A549  b. Compd. 7e \A549 and c. MCF7 control. d. Compd. 8d\MCF7

The results of the Annexin V-FITC/PI assay suggested that: (i) treatment of A549 cells with compound 7e led to early and late cellular apoptosis, which proved through the significant increase the percentage of the apoptotic cells in both the early apoptotic phase (from 0.36 to 26.85%) and the late apoptotic phase (from 0.18 to 15.61%) that indicates a high increase in total apoptosis when compared to the untreated control (Fig. 3a, b), (ii) compound 8d caused a considerable increase in early and late apoptosis of MCF7 cells than control cells; i.e. the early and late apoptotic population increased from 0.55 to 22.38% and from 0.27 to 26.96%, respectively (Fig. 3c, d), and (iii) treating A549 cells with compound 7e increases the population of necrotic cell from 1.41 (control) to 3.73% keeping the necrosis minimally contributing. Also, the population of necrotic cells increases from 1.89 (control) to 5.04% upon the subjection of the MCF7 cells to compound 8d (Fig. 4). From the above results, an overall 79-fold increase in A549 cellular apoptosis after treatment with compounds 7e and 69-fold increase in MCF7 cellular apoptosis after treatment with 8d compound In comparison to the control. We observed that our targeted substances, 7e and 8d, have the potential to function as a biological mechanism for inhibiting cell growth, thus leading to cytotoxic effects against the MCF7 and A549 cell line (Fig. 4).

Fig. 4
figure 4

Apoptosis/necrosis assessment of A549 and MCF7 cells after treatment with compounds 7e against A549 and 8d against MCF7. Different cell populations were plotted as a percentage of total events. Data are presented as mean ± SD; n = 3

Molecular docking

The docking studies revealed that compound 7e had stronger binding affinity (− 10.3 kcal/mol) to CDK2 compared to the standard STU299 (− 11.5 kcal/mol). The interactions analysis showed that 7e formed hydrogen bonds, amid pi-sulfate, alkyl, pi-alkyl, and pi-sigma interactions with key amino acid residues in the CDK2 binding site like GLU 12, VAL 18, LYS 33, and LEU 134 (Table 4, Fig. 5). In contrast, STU299 showed hydrogen bonds, C–H bonds, alkyl, pi-alkyl, and pi-sigma interactions with residues like GLY 13, GLN 131, LEU 134, VAL 18, ILE 10. The additional pi-sulfate and amid interactions of 7e with GLU 12 likely contribute to its better binding over STU299.

Table 4 ∆G and binding affinity (Kcal/mol) for CDK2 docking interaction with compound 7e in comparison its standard stu299
Fig. 5
figure 5

3D and 2D docking interaction of compound 7e with CDK2 in compered to the slandered STU299

For DHFR, compound 8d had a stronger binding affinity (− 9.5 kcal/mol) than the standard PRD400 (− 8.5 kcal/mol). The interactions analysis revealed 8d forms hydrogen bonds, C–H bonds, alkyl, and pi-sigma interactions with key residues like VAL 115, GLN 35, PHE 34 in the DHFR binding site (Table 5, Fig. 6). Meanwhile, PRD400 showed hydrogen bonds, C–H bonds, alkyl, and pi-alkyl interactions with residues such as LYS 55, ALA 9, ILE 16, SER 59, GLY 117, ILE 7, PHE 34. The extra pi-sigma interaction of 8d with PHE 34 may enhance its binding over PRD400.

Table 5 ∆G and binding affinity (Kcal/mol) for DHFR docking with 8d with its standard PRD400
Fig. 6
figure 6

3D and 2D docking interaction of compound 8d with DHFR in compered to the slandered PRD400

Overall, the docking results indicate compounds 7e and 8d bind more strongly to CDK2 and DHFR respectively compared to the standard inhibitors. The additional interactions formed by 7e and 8d with key active site residues likely contribute to their enhanced binding affinity.

Enzyme inhibitory activities

The promising anti-proliferative impact of compounds 7e and 8d, in addition to their cell cycle disruption and pro-apoptotic effects, proved a further exploration for their possible inhibitory activities against many enzymes such as RET (encodes a receptor tyrosine kinase) and CDK2 (Cyclin-dependent kinase 2) treated with compound 7e, and DHFR (Dihydrofolate reeducates), Eef2 Kinase (Eukaryotic elongation factor 2kinase)and IKB kinase (inhibitory kappa B kinase) treated with compound 8d.

Inhibitory activity of compound 7e towards CDK2

Compound 7e showed significant CDK2 cyclin A inhibitory activity in comparison with the reference; Roscovitine Table 6. Due to the nature of isoquinoline moiety [51, 52]. From the docking study the inhibition mechanism of compound 7e with interaction with CDK2 with hydrogen bonding and other bonds; they may deactivate the binding site in CDK2 and either its partners or substrates resulting in specific inhibition of CDK2. The obtained results in Table 6 and Fig. 7a and for more enzyme inhibition test details presented in supplementary data (Additional file 1: Table S7) showed that the tested compound 7e exhibited significant inhibitory action against CDK2 with IC50 value 0.149 ± 0.007 in comparison with the control; Roscovitine which showed IC50 of 0.380 ± 0.008 µM (reference of CDK2 inhibitor).

Table 6 CDK2 inhibitory activity of compound 7e
Fig. 7
figure 7

a-  CDK2 inhibitory activity of compound 7e. b- DHFR inhibitory activity of compound 8d.

DHFR inhibitory activity of compound 8d

Our results obtained indicated that compound 8d which contains tetrahydrothieno[2,3-c]isoquinoline [14, 53, 54] moiety showed high inhibitory activity towards DHFR enzyme in comparison with the reference; methotrexate show Table 7, Fig. 7b and for more enzyme inhibition test details was presented in supplementary data (Additional file 1: Table S8). Thus, compound 8d exhibited good inhibitory activity towards DHFR with IC50 value 0.199 ± 0.016 in comparison with that Methotrexate (IC50 of 0.131 ± 0.007).

Table 7 DHFR inhibitory activity of the compound 8d
Other enzyme inhibitory activity

Compounds7e and 8d exhibited moderate inhibitory activity towards other enzymes under investigation in comparison with their control for more enzyme inhibition test details show Additional file 1 (Table 8 and Additional file 1: Tables S9–S11).

Table 8 Enzyme inhibitory activity of compounds 7e and 8d

In vitro antioxidant behavior

Ten newly synthesized compounds were studied as in vitro antioxidants by measuring of their DPPH scavenging activity which is represented as a percentage % [32]. Results are represented by mean ± SD of three replicates. Table 9 showed the percentage of DPPH scavenging activity of the tested compounds in a dose-dependent relationship compared with Vitamin C (ascorbic acid) as a standard. The higher dose concentration of 0.05 μg/mL represents higher antioxidant activity. Compounds 1, 3, 6, 7c and 8e have higher result than Vitamin C itself. Compound 8e show the highest significant result which suggests that this compound can be used as excellent antioxidant drugs. The high antioxidant activity is referred to the presence of C=O, NH2, and OH groups like ascorbic acid [55, 56] which can be easily oxidized and reduced and can be used as antioxidant drugs. (Fig. 8 and Table 9).

Fig. 8
figure 8

Antioxidant activity of compounds 1, 3, 6, 7a, 7b, 7c, 7d, 7e, 8a and 8e

Table 9 DPPH Scavenging activity of 5,6,7,8-tetrahydrothieno[2,3-c]isoquinolines 1, 3, 6 and 7a–e, and 6,7,8,9-tetrahydrothieno[2,3-c]isoquinolines 8a, b

Conclusion

In this paper, We successfully synthesized and characterized of novel two series of substituted methylthiotetrahydroisoquniolines and related tetrahydrothieno[2,3-c]isoquinolines. All synthesized compounds were evaluated for their anticancer activity towards A549 and MCF7 cell lines, and showed promising results. Moreover, the cell cycle arrest and apoptosis induction of the two representative compounds was studied. Compound 7e caused cell cycle arrest of A549 cell line at G2/M phase and compound 8d arrest the cell cycle of MCF7 cell line at S phase. Compounds 7e and 8d compounds caused high increase in the early and late apoptosis and necrosis. Furthermore, compound 7e showed significant inhibition of CDK2 enzyme while compound 8d exhibited significant activity as a DHFR inhibitors. In the future we intend to synthesis new series of tetrahydrothieno[2,3-c]isoquinolines to studied their anticancer activity not only in vitro but also in vivo and examined the anticancer activity of these compounds in patient samples as potent anticancer drugs.

Availability of data and materials

All data generated or analyzed during this study are in this published article and supplementary information.

References

  1. Hawash M, Jaradat N, Eid MA, Abubaker A, Mufleh O, Hroub Q, Sobuh S. Synthesis of novel isoxazole–carboxamide derivatives as promising agents for melanoma and targeted nano-emulgel conjugate for improved cellular permeability. BMC Chem. 2022;16(1):Article No. 47.

    Article  PubMed  Google Scholar 

  2. Hawash M. Highlights on specific biological targets; cyclin-dependent kinases, epidermal growth factor receptors, ras protein, and cancer stem cells in anticancer drug development. Drug Res. 2019;69(9):471–8.

    Article  CAS  Google Scholar 

  3. Bruyere C, Meijer L. Targeting cyclin-dependent kinases in anti-neoplastic therapy. Curr Opin Cell Biol. 2013;25(6):772–9.

    Article  CAS  PubMed  Google Scholar 

  4. Hawash M. Recent advances of tubulin inhibitors targeting the colchicine binding site for cancer therapy. Biomolecules. 2022;12(12):Article No. 1843.

    Article  PubMed  Google Scholar 

  5. Sabt A, Eldehna WM, Al-Warhi T, Alotaibi OJ, Elaasser MM, Suliman H, Abdel-Aziz HA. Discovery of 3,6-dissubsttituted pyridazines as a novel class of anticancer agent targeting cyclin-dependent kinase 2: synthesis, biological evolution and in silico insights. J Enzyme Inhib Med Chem. 2020;35(1):1616–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tetsu O, Cormick MF. Proliferation of cancer cells despite CDK2 inhibition. Cancer Cell. 2003;3(3):233–45.

    Article  CAS  PubMed  Google Scholar 

  7. Tarfah A, Mahmoud FA, Hadia A, Ohoud JA, Mohammad MA, Ghada HA, Hanaa YA, Mahmoud ME, Wagdy ME, Hatem A. Novel [(N-alkyl-(3)indolylmethylene)hydrazono]oxindoles arrest cell cycle and induce cell apoptosis by inhibiting CDK2 and Bcl-2: synthesis, biological evaluation and in silico studies. J Enzyme Inhib Med Chem. 2020;35(1):1300–9.

    Article  Google Scholar 

  8. Christopher RC, Elizabeth A, Suzannah JH, Mathew PM, Benoit C, Bernard TG, Ian RH, Lisa KH, Svitlana K, Christopher JM, et al. Cyclin-dependent kinase (CDK) inhibitors: structure–activity relationships and insights into the CDK-2 selectivity of 6-substituted 2-arylaminopurines. J Med Chem. 2017;60(5):1746–67.

    Article  Google Scholar 

  9. Jiawei Z, Yichao G, HongzhiJie LY, Xin H, Liming L, Senlin X, Zhipeng F, Byung-wook K, Lina G, Lili D, et al. Inhibition of the CDK2 and cyclin a complex leads to autophagic degradation of CDK2 in cancer cells. Nat Commun. 2022;13(1):Article No. 2835.

    Article  Google Scholar 

  10. Nchez-Martínez CS, Gelbert LM, Lallena MJ, Dios DA. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs. Bioorg Med Chem Lett. 2015;25(17):3420–35.

    Article  Google Scholar 

  11. Nour EA, Eman HK, Wafaa HA, Mohamed IA, Asmaa AM, Nasser SM. Design and synthesis of new CDK2 inhibitors containing thiazolone andthiazolthione scafold with apoptotic activity. Chem Pharm Bull. 2011;69(1):106–17.

    Google Scholar 

  12. Aravinda P, Jayashree BS. Novel benzylidene benzofuranone analogues aspotential anticancer agents: design, synthesis and in vitro evaluation based on CDK2 inhibition assays. Biotech. 2022;12(10):256–78.

    Google Scholar 

  13. Maria VR, Ornella R, Mery LF, Giampaolo B, Elisa V, Daniela R, Simona C. DHFR inhibitors: reading the past for discovering novel anticancer agents. Molecules. 2019;24(6):1140–59.

    Article  Google Scholar 

  14. Kristen ML, Narendran G, Dennis LW, Amy CA. Elucidating features that drive the design of selective antifolates using crystal structures of human dihydrofolate reductase. J Biochem. 2013;52(41):15–52.

    Google Scholar 

  15. Juan H, Wenliang Q, Qi A, Tao Y, Youfu L. Dihydrofolate reductase inhibitors for use as antimicrobial agents. Eur J Med Chem. 2020;1(195):112268.

    Google Scholar 

  16. Mohamed HE, Kamal ME, Ashraf HB, Khaled E, Mohamed A, Hany EA, Ahmed AA, Hamada SA. The antimicrobial potential and pharmacokinetic profiles of novel quinoline-based scaffolds: synthesis and in silico mechanistic studies as dual DNA gyrase and DHFR inhibitors. New J Chem. 2021;45(31):13986–4004.

    Article  Google Scholar 

  17. Rana RM, Rampogu S, Abid NB, Zeb A, Parate S, Lee G, Yoon S, Kim Y, Kim D, Lee KW. In silico study identified methotrexate analog as potential inhibitor of drug resistant human dihydrofolate reductase for cancer therapeutics. Molecules. 2020;25(15):3510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Galán A, Moreno L, Párraga J, Serrano A, Sanz MJ, Cortes D, Cabedo N. Novel isoquinoline derivatives as antimicrobial agents. Bioorg Med Chem. 2013;21(11):3221–30.

    Article  PubMed  Google Scholar 

  19. Alagumuthu M, Sathiyanarayanan KI, Arumugam S. Molecular docking, design, synthesis, in vitro antioxidant and anti-inflammatory evaluations of new isoquinoline derivatives. Int J Pharm Sci. 2015;7(12):200–8.

    CAS  Google Scholar 

  20. Manikandan A, Sivakumar AA. Analgesic, anti-inflammatory and antipyretic evaluations of new isoquinoline derivatives. J Pharm Sci. 2016;8(4):339–43.

    CAS  Google Scholar 

  21. Watanuki S, Matsuura K, Tomura Y, Okada M, Okazaki T, Ohta M, Tsukamoto S. Synthesis and pharmacological evaluation of 1-isopropyl-1,2,3,4-tetrahydroisoquinoline derivatives as novel antihypertensive agents. Chem Pharm Bull. 2011;59(8):1029–37.

    Article  CAS  Google Scholar 

  22. Pingaew R, Prachayasittikul S, Ruchirawat S. Synthesis, cytotoxic and antimalarial activities of benzoyl thiosemicarbazone analogs of isoquinoline and related compounds. Molecules. 2010;15(2):988–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hassaneen HM, Wardkhan WW, Mohammed YS. Novel route to isoquinoline[2,1-g][1,6]naphthyridine, pyrazolo[5,1-a]isoquinoline and pyridazino[4′,5′: 3,4]pyrazolo[5,1-a]isoquinoline derivatives with evaluation of antitumor activities. Z Naturforsch B. 2013;68(b):895–904.

    Article  Google Scholar 

  24. Kakhki S, Shahosseini S, Zarghi A. Design, synthesis and cytotoxicity evaluation of new 2-aryl-5,6-dihydropyrrolo[2,1-a]isoquinoline derivatives as topoisomerase inhibitors. Iran J Pharm Res. 2014;13:71–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Partik Y, Ashish K, Islam A, Vishal N. Recent development of tetrahydroquinoline/isoquinoline based compounds as anticancer agents. Med Chem. 2021;21(17):1587–622.

    Google Scholar 

  26. Cushman M, Jayaraman M, Vroman JA, FuKunaga AK, Fox BM, Kohlhagen G, Strumberg D, Pommier Y. Synthesis of new indeno[1,2-c]isoquinolines:cytotoxic non-camptothecin to poisomerase 1 inhibitors. J Med Chem. 2000;43(20):3688–98.

    Article  CAS  PubMed  Google Scholar 

  27. Rashad AS, Ibrahim A, Mohmed M. Cytotoxcicity evaluation of a new set of2-aminobeno[de]isoquoinoline-1,3-diones. Int J Mol Sci. 2014;15(12):22483–91.

    Article  Google Scholar 

  28. Sarbadhikary P, George BP, Abrahamse H. Inhibitory role of berberine, an isoquinoline alkaloid, on NLRP3 inflammasome activation for the treatment of inflammatory diseases. Molecules. 2021;26(20):Article No. 6238.

    Article  PubMed  Google Scholar 

  29. Faheem KK, Chandra S, Chander S, Kunjiappan S, Murugesan S. Medicinal chemistry perspective of 1,2,3,4-tetrahydroisoquinoline analogs biological activities and SAR studies. RSC Adv. 2021;11(20):12254–87.

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  30. Gangapuram M, Eyunni S, Redda KK. Synthesis and pharmacological evolution of tetrahydroisoquinolines as anti breast cancer agents. J Cancer Sci Ther. 2014;6(5):161–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gao Y, Tu N, Liu X, Lu K, Chen S, Guo J. Progress in the total synthesis of antitumor tetrahydroisoquinoline alkaloids. Chem Biodivers. 2003;20(5):e202300172.

    Article  Google Scholar 

  32. Sayed EM, Hassanien R, Farhan N, Aly HF, Mahmoud K, Mohamed SK, Mague JT, Bakhite EA. Nitrophenyl-group-containing heterocycles. I. Synthesis, characterization, crystal Structure, anticancer Activity, and antioxidant properties of some new 5,6,7,8-tetrahydroisoquinolines bearing 3(4)-nitrophenyl group. ACS Omega. 2022;7(10):8767–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dermerson CA, Philipp AH, Humber LG, Kraml MJ, Chares MP, Tom H, Vavra I. Pyrrolo[4,3,2-de]isoquinoline with central nervous system andantihypertensive activities. J Med Chem. 1974;17(11):1140–5.

    Article  Google Scholar 

  34. Brahmayya M, Venkateswara B, Viplavaprasad U, Basaveswara Rao MV, Babua KR, Babua BK, Rajkumar K, Praveen C, Giribabu N, Vijaya M, Padmarao CV, Srinivasa NR. Synthesis of quinolines and their In vitro antioxidant activities under solvent free conditions by using the SiO2–Zn–MgO as a novel and reusable catalyst. J Appl Pharm Sci. 2012;2(10):041–4.

    CAS  Google Scholar 

  35. Sirassu N, Kumara SB, Vasudeva RN. One-pot synthesis of novel 1,2,3-triazole-pyrimido[4,5-c]isoquinoline hybrids and evaluation of their antioxidant activity. Synth Commun. 2018;74(10):1220–6.

    Google Scholar 

  36. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65:55–63.

    Article  CAS  PubMed  Google Scholar 

  37. Toshiak S, Kenji A, Masami I, Masanao N, Seiichiro S, Noriyuki S, Fumio Y, Kyoji T, Yoshie H. Synthesis and in vitro cytotoxicity of 1,2,3,4-tetrahydroiso-quinoline derivative. Eur J Med Chem. 2006;41(2):241–52.

    Article  Google Scholar 

  38. Hawash M, Kahraman DC, Ergun SG, Cetin-Atalay R. Synthesis of novel indole-isoxazole hybrids and evaluation of their cytotoxic activities on hepatocellular carcinoma cell lines. BMC Chem. 2021;15:Article No. 66.

    Article  PubMed  Google Scholar 

  39. Hawash M, Qneibi M, Jaradat N, Abualhasan M, Amer J, Amer E. The impact of filtered water-pipe smoke on healthy versus cancer cells and their neurodegenerative role on AMPA receptor. Drug Chem ToxicoI. 2022;25(5):2292–300.

    Article  Google Scholar 

  40. Mohammed FZ, Rizzk YW, El Deen IM, Mourad AAE, El Behery M. Design, synthesis, cytotoxic screening and molecular docking studies of novel hybrid thiosemicarbazone derivatives as anticancer agents. Chem Biodivers. 2021;18(12):Article No. e2100580.

    Article  PubMed  Google Scholar 

  41. Mohammed FZ, Rizzk YW, Mohey El-Deen I, Gad EM, El Behery M, Mahdy ARE. Discovery of 2-amino-4H-1,3, 4-thiadiazine-5(6H)-one derivatives and their in vitro antitumor investigation. Med Chem Drug Disc. 2022;7:Article No. e20210433.

    Google Scholar 

  42. Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 2015;14(2):130–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang J, Gan Y, Li H, Yin J, He X, Lin L, Xu S, Fang Z, Kim B, Gao L, Ding L, Zhang E, Ma X, Li J, Li L, Xu Y, Horne D, Xu R, Yu H, Gu Y, Huang W. Inhibition of the CDK2 and Cyclin A complex leads to autophagic degradation of CDK2 in cancer cells. Nat Commun. 2022;13:Article No. 2835.

    Article  PubMed  ADS  Google Scholar 

  44. Salem IM, Mostafa SM, Salama I, Elsabbah OA, Hagazy WAH, Ibrahim TS. Human dihydrofolate reductase inhibition effect of 1-Phenylpyrazolo[3,4–d]pyrimidines: synthesis, antitumor evaluation and molecular modeling study. Bioorgan Chem. 2022;129:Article No. 106207.

    Article  Google Scholar 

  45. Zhu Z, Chen C, Zhang J, Lai F, Feng J, Wu G, Xia J, Zhang W, Han Z, Zhang C, Yang Q, Wang Y, Liu B, Li T, Wu S. Exploration and biological evaluation of 1,3-diamino-7H-pyrrol[3,2-f]quinazoline derivatives as dihydrofolate reductase inhibitors. J Med Chem. 2023;66(20):13946–67.

    Article  CAS  PubMed  Google Scholar 

  46. Lili X, Guozheng H, Zhihui Z, Shasha T, Yingying W, Huanwu H, Xiaowei L, Ying L, Feize L, Huajun Z. LFZ-4-46, a tetrahydroisoquinoline derivative, induces apoptosis and cell cycle arrest via induction of DNA damage and activation of MAPKs pathway in cancer cells. Anticancer Drugs. 2021;32(8):842–54.

    Article  Google Scholar 

  47. Megda FM, Hamdi MH, Ismail AA. Cytotoxicity, molecular modeling, cellcycle arrest and apoptotic induction induced by novel tetrahdro [1,2,4]triazolo[3,4-a] isoquinoline chalcones. Eur J Med Chem. 2018;143:532–41.

    Article  Google Scholar 

  48. Tian X, Li Y, Shen Y, Li Q, Wang Q, Feng L. Apoptosis and inhibition of proliferation of cancer cells induced by cordycepin (review). Oncol Lett. 2015;10(2):595–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sun X, Liu M, Gao L, Mao Y, Zhao D, Zhuang J, Liu LAA. noveltetrahydroisoquinoline (THIQ) analogue induces mitochondria-dependent apoptosis. Eur J Med Chem. 2018;150:719–28.

    Article  CAS  PubMed  Google Scholar 

  50. Darwish MIM, Moustafa AM, Youssef AM, Mansour M, Yousef AI, El Omri A, Shawki HH, Mohamed MF, Hassaneen HM, Abdelhamid AI, Oishi H. Novel tetrahydro[1,2,4]triazolo[3,4-a]isoquinoline chalcones suppress Breast Carcinoma through cell cycle arrests and apoptosis. Molecules. 2023;28(8):3338–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Deng Y, Shipps J, Zhao L, Siddiqui MA, Popovici-Mullar J, Curran PJ, Duca JS, Hruza AW, Fischmann TO, Madison VS, et al. Modulating the interaction between CDK2 and cyclin A with A quinoline-based inhibitors. Bioorg Med Chem Lett. 2001;24(1):199–203.

    Article  Google Scholar 

  52. Yaoguang H, Wenwu L, Shuoqi H, Deping L, Chang X, Xiaowen J, Mingue L, Xin L, Chengze Z, Limeng W, et al. Discovery of novel benzofuro[3,2-b]quinolone derivative as dual CDK2/TOPO 1 inhibitors. Bioorg Chem. 2022;126:Article No. 105870.

    Article  Google Scholar 

  53. Yousry AA, Sondos MA, Sadia AH, Abeer MA, Ahmed AA, Ahmed R. One-pot strategy for thiazole tethered 7-ethoxyquinolone hybrids: synthesis and potential antimicrobial agent as dihydrofolate reductase (DHFR) inhibitors with molecular docking study. J Mol Struct. 2021;1242:130748.

    Article  Google Scholar 

  54. Wang M, Yang J, Yuan M, Xue L, Li H, Tian C, Wang X, Liu J, Zhang Z. Synthesis and antiproliferative activity of a series of novel 6-substituted pyrido[3,2-d]pyrimidines as potential nonclassical lipophilic antifolates targeting dihydrofolate reductase. Eur J Med Chem. 2017;128:88–97.

    Article  CAS  PubMed  Google Scholar 

  55. Annie SA, Arun SA, Kuppusamy R, Isaac SR. In vitro antioxidant studies on the benzyl tetrahydroisoquinoline alkaloid berberine. Biol Pharm Bull. 2006;29(9):1906–10.

    Article  Google Scholar 

  56. Ahmed KO, Saripah SS, Yuhanis MB, Mohd AN, Saadon AA, Khaliigah A, Marc L. Two new isoquinoline alkaloids from the bark of Alphonsea cylindrical king and their antioxidant activity. Phytochem Lett. 2019;29:11–40.

    Google Scholar 

Download references

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

EMS: Investigation, Methodology, Writing-original draft, Visualization, Software, Validation. EAB: Conceptualization, Formal analysis, Supervision, Investigation. RH: Investigation, Methodology. Writing-review and editing. NF: Writing-original draft, Writing-review and editing. HFA: Investigation, Methodology. Writing-review and editing. SGM: Conceptualization, Formal analysis, Supervision, Investigation, Methodology, Writing-original draft, Writing-review and editing. NAH: Conceptualization, Formal analysis, Supervision, Investigation, Methodology, Writing-original draft, Writing-review and editing.

Corresponding authors

Correspondence to Eman M. Sayed or Etify A. Bakhite.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Fig. S1

. FT-IR spectrum of Compound 1. Fig. S2. 1H NMR spectrum of Compound 1. Fig. S3. 13C NMR spectrum of compound 1. Fig. S4. FT-IR spectrum of compound 3. Fig. S5. 1H NMR spectrum of compound 3. Fig. S6. 13C NMR spectrum of compound 3. Fig. S7. FT-IR spectrum of compound 4. Fig. S8. 1H NMR spectrum of compound 4. Fig. S9. 13C NMR spectrum of compound 4. Fig. S10. FT-IR spectrum of compound 5. Fig. S11. 1H NMR spectrum of compound 5. Fig. S12. 13C NMR spectrum of compound 5. Fig. S13. FT-IR spectrum of compound 6. Fig. S14. 1H NMR spectrum of compound 6. Fig. S15. 13C NMR spectrum of compound 6. Fig. S16. FT-IR spectrum of compound 7a. Fig. S17. 1H NMR spectrum of compound 7a. Fig. S18. 13C NMR spectrum of compound 7a. Fig. S19. FT-IR spectrum of compound 7b. Fig. S20. 1H NMR spectrum of compound 7b. Fig. S21. 13C NMR spectrum of compound 7b. Fig. S22. FT-IR spectrum of compound 7c. Fig. S23. 1H NMR spectrum of compound 7c. Fig. S24. 13C NMR spectrum of compound 7c. Fig. S25. FT-IR spectrum of compound 7d. Fig. S26. 1H NMR spectrum of compound 7d. Fig. S27. 13C NMR spectrum of compound 7d. Fig. S28. FT-IR spectrum of compound 7e. Fig. S29. 1H NMR spectrum of compound 7e. Fig. S30. 13C NMR spectrum of compound 7e. Fig. S31. FT-IR spectrum of compound 8a. Fig. S32. 1H NMR spectrum of compound 8a. Fig. S33. 13C NMR spectrum of compound 8a. Fig. S34. FT-IR spectrum of compound 8b. Fig. S35. 1H NMR spectrum of compound 8b. Fig. S36. 13C NMR spectrum of compound 8b. Fig. S37. FT-IR spectrum of compound 8c. Fig. S38. 1H NMR spectrum of compound 8c. Fig. S39. 13C NMR spectrum of compound 8c. Fig. S40. FT-IR spectrum compound 8d. Fig. S41. 1H NMR spectrum compound 8d. Fig. S42. 13C NMR spectrum of compound 8d. Fig. S43. FT-IR spectrum compound 8e. Fig. S44. 1H NMR spectrum compound 8e. Fig. S45. 13C NMR spectrum of compound 8e. Table S1. Raw date of toxicity and viability of compounds 1,3–6 against MCF7. Table S2. Raw date of toxicity and viability of compounds 7a–e against MCF7. Table S3. Raw date of toxicity and viability of compounds 8a–e against MCF7. Table S4. Raw date of toxicity and viability of compounds 1,3–6 against A549. Table S5. Raw date of toxicity and viability of compounds 7a–e against A549. Table S6. Raw date of toxicity and viability of compounds 8a–e against A549. Table S7. CDK2 inhibitor detailed results. Table S8. DHFR inhibitor detailed results. Table S9. Eef2 inhibitor detailed results. Table S10. IKB inhibitor detailed results. Table S11. RET inhibitor detailed results.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sayed, E.M., Bakhite, E.A., Hassanien, R. et al. Novel tetrahydroisoquinolines as DHFR and CDK2 inhibitors: synthesis, characterization, anticancer activity and antioxidant properties. BMC Chemistry 18, 34 (2024). https://doi.org/10.1186/s13065-024-01139-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13065-024-01139-w

Keywords