Skip to main content

Novel phenoxy-((phenylethynyl) selanyl) propan-2-ol derivatives as potential anticancer agents

Abstract

Selenocompounds protect against damage to healthy cells and induce the death of tumor cells by apoptosis; for this reason, they are attractive compounds for cancer research. In the present study, two series of novel phenoxy-((phenylethynyl) selanyl) propan-2-ol derivatives were synthesized, and their anti-proliferation activities were evaluated. Of the 23 compounds synthesized, most showed potent anti-proliferative activity against human cancer cell lines. Specifically, compounds 3h, 3g, and 3h-2, which had a 2- or 4-position halogen substituent on 1-((phenylethynyl)selanyl)-3-phenoxypropan-2-ol, exhibited the best anti-proliferative activity against tumor cells. Flow cytometry demonstrated that 3h, 3g, and 3h-2 induced G2/M phase arrest and apoptosis in A549 cells. Cellular studies demonstrated that the induction of apoptosis by 3h correlated with changes in the expression of cell cycle-related proteins and apoptosis-related proteins. Xenograft tumor experiments in nude mice revealed that compound 3h has antitumor effects in vivo and no evident toxic effects in nude mice. In addition, compound 3h alleviated cisplatin-induced liver and kidney damage. These findings uncover the applicability of compound 3h as a novel lead compound for cancer treatment.

Peer Review reports

Introduction

There were almost 19.3 million new cancer cases worldwide in 2020 and more than 10.0 million deaths from cancer. By 2040, those figures will almost double [1]. The limitations of current cancer treatments include the lack of selectivity for healthy versus cancer cells, the development of multidrug resistance, and toxicities caused by most current cancer therapies. It is urgent to identify potential therapeutic targets and effective drugs [2, 3].

Apoptosis is a defense mechanism against the formation and progression of cancer, involving the activation, expression, and regulation of several genes [4]. Most current anticancer drugs work through the apoptotic signaling pathway to trigger cell death [5]. The selective regulation of tumor cell apoptosis has become the focus of antitumor drug research [6,7,8].

Selenium (Se) is a trace element with multiple roles in human health [9,10,11]. Selenoproteins mediate a series of biological effects, including antioxidant defense (protect normal cells from oxidative damage), anti-inflammatory actions, maintenance of thyroid function, and immune response regulation [12]. Epidemiologic evidence indicates an inverse association between Se status and cancer risk [13]. Some organoselenium compounds inhibit tumor cell growth in many xenograft models of cancer [14,15,16] and have synergistic effects when combined with chemotherapy drugs [17,18,19,20]. The effectiveness of selenium compounds as chemopreventive agents in vivo is associated with their ability to induce apoptosis, inhibit tumor cell migration and invasion, and regulate the cell cycle [21].

We have designed and synthesized 23 novel phenoxy-((phenylethynyl) selanyl) propan-2-ol derivatives. The proliferation inhibition, cell migration destruction, apoptosis, and cell cycle arrest in cancer cells were examined in relation to these compounds. Subsequently, the xenograft tumor in vivo experiment and protection of normal cells against cisplatin-induced damage were performed on the most potent derivative 3h.

Methods

Reagents and antibodies

Fetal bovine serum (PA500) was purchased from Newzerum (Christchurch, New Zealand). DMEM- and RPMI 1640 medium Phosphate-buffered saline (PBS) and penicillin/streptomycin were purchased from Gibco. The DeadEnd™ Fluorometric TUNEL System TB235 kit (G3250) was from Promega (Southampton, UK), and the Annexin V-FITC/PI Apoptosis Detection Kit (A211-01, Vazyme) and MTT cell proliferation and cytotoxicity assay kit (G020-1-2, Nanjing Jiancheng) were also used. Paraformaldehyde was purchased from Macklin (Shanghai, China). Dimethyl sulfoxide (DMSO) was purchased from Solarbio (Beijing). DAPI was purchased from SouthernBiotech (Birmingham, USA). The following antibodies were used: Cyclin D1 (Abmart, T55404), Bcl2 (Abmart, T40056), Bax (Santa, SC-493), cleaved Caspase 3 (Affinity Biosciences, AF7022) and β-actin (TransGen Biotech, HC201-01).

Synthesis procedures of 1-phenoxy-3-((phenylethynyl) selanyl) propan-2-ol derivatives

The synthesis procedure of 1-phenoxy-3-((phenylethynyl) selanyl) propan-2-ol derivatives was a copper-catalyzed three-component cascade, as previously reported [22]. Briefly, propionic acids (0.2 mmol), epoxides (0.6 mmol), Se (0.6 mmol), CuCl2 (0.02 mmol), TBAI (0.4 mmol), 1, 10-phen (0.02 mmol), and Cs2CO3 (0.6 mmol) in H2O (2 mL) were placed in a 25-mL glass. The reaction mixture was stirred at 50 ℃ for 24 h. After it was cooled, 10 mL of ethyl acetate was added, and the mixture was filtered through diatomaceous earth. The organic phase was dried over Na2SO4 and concentrated under reduced pressure. The residue was then purified by chromatography to provide the final product.

Characterizations of products in detail are as follows:

1-phenoxy-3-(phenylethynylselanyl) propan-2-ol (3a)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (60.4 mg, 91% yield). 1 H NMR (500 MHz, CDCl3): δ 7.36–7.34 (m, 2 H), 7.27–7.23 (m, 5 H), 6.97–6.90 (m, 3 H), 4.37 (brs, 1 H), 4.16–4.10 (m, 2 H), 3.17 (dd, J = 12.50, 5.40 Hz, 1 H), 3.07 (dd, J = 12.50, 6.90 Hz, 1 H), 2.84–2.83 (m, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.4, 131.6, 129.6, 128.4, 128.3, 123.2, 121.4, 114.7, 99.6, 70.3, 69.6, 69.5, 32.8.

1-phenoxy-3-(p-tolylethynylselanyl)propan-2-ol (3b)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (65.7 mg, 95% yield), Mp = 49–50 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.29–7.25 (m, 4 H), 7.08 (d, J = 7.90 Hz, 2 H), 6.96 (t, J = 7.40 Hz, 1 H), 6.92 (d, J = 8.20 Hz, 2 H), 4.39–4.37 (m, 1 H), 4.18–4.12 (m, 2 H), 3.17 (dd, J = 12.40, 5.25 Hz, 1 H), 3.07 (dd, J = 12.40, 5.25 Hz, 1 H), 2.77–2.76 (m, 1 H), 2.33 (s, 3 H); 13 C NMR (125 MHz, CDCl3): δ 158.3, 138.6, 131.6, 129.6, 129.1, 121.3, 120.1, 114.6, 99.8, 70.3, 69.6, 68.3, 32.7, 21.5; HRMS (ESI): calcd for C18H18O2Se [M + H]+ 347.0544, found 347.0540.

1-(mesitylethynylselanyl)-3-phenoxypropan-2-ol (3c)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (58.3 mg, 78% yield). 1 H NMR (500 MHz, CDCl3): δ 7.27–7.23 (m, 2 H), 6.95 (t, J = 7.30 Hz, 1 H), 6.90 (d, J = 8.15 Hz, 1 H), 6.83 (s, 2 H), 4.41–4.38 (m, 1 H), 4.17–4.11 (m, 2 H), 3.19 (dd, J = 12.50, 5.40 Hz, 1 H), 3.06 (dd, J = 12.50, 5.40 Hz, 1 H), 2.76–2.75 (m, 1 H), 2.36 (s, 6 H), 2.26 (s, 3 H); 13 C NMR (125 MHz, CDCl3): δ 158.4, 140.3, 137.8, 129.5, 127.6, 121.3, 120.1, 114.6, 97.7, 75.7, 70.4, 69.6, 33.3, 21.2, 20.9; HRMS (ESI): calcd for C20H22O2Se [M + H]+ 375.0858, found 375.0853.

1-((4-tert-butylphenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3d)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (72.9 mg, 94% yield). 1 H NMR (500 MHz, CDCl3): δ 7.31–7.27 (m, 6 H), 6.98–6.91 (m, 3 H), 4.39 (dt, J = 16.50, 5.30 Hz, 1 H), 4.19–4.13 (m, 2 H), 3.18 (dd, J = 12.50, 5.40 Hz, 1 H), 3.08 (dd, J = 12.50, 5.40 Hz, 1 H), 1.30 (s, 9 H); 13 C NMR (125 MHz, CDCl3): δ 158.3, 151.8, 131.4, 129.5, 125.3, 131.3, 120.1, 114.6, 99.7, 70.3, 69.6, 68.3, 34.8, 32.8, 31.1; HRMS (ESI): calcd for C21H24O2Se [M + H]+ 389.1014, found 389.1007.

1-(biphenyl-4-ylethynylselanyl)-3-phenoxypropan-2-ol (3e)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (59.6 mg, 73% yield), Mp = 78–79 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.59–7.56 (m, 2 H), 7.51–7.50 (m, 2 H), 7.44–7.41 (m, 4 H), 7.36–7.33 (m, 1 H), 7.28–7.24 (m, 2 H), 6.98–6.92 (m, 3 H), 4.39 (brs, 1 H), 4.19–4.14 (m, 2 H), 3.21 (dd, J = 12.50, 5.40 Hz, 1 H), 3.10 (dd, J = 12.50, 5.40 Hz, 1 H), 2.77 (brs, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.3, 141.1, 140.3, 132.9, 132.0, 129.6, 128.9, 127.7, 127.1, 127.0, 127.0, 126.9, 122.0, 121.4, 114.6, 99.5, 70.3, 70.2, 69.6, 32.9; HRMS (ESI): calcd for C23H20O2Se [M + H]+ 409.0701, found 409.0732.

1-((4-methoxyphenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3f)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (65.2 mg, 90% yield), Mp = 59–60 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.33–7.27 (m, 4 H), 6.97 (t, J = 7.3 Hz, 1 H), 6.93–6.91 (m, 2 H), 6.82–6.80 (m, 2 H), 4.39 (brs, 1 H), 4.19–4.13 (m, 2 H), 3.80 (s, 3 H), 3.17 (dd, J = 12.50, 5.40 Hz, 1 H), 3.07 (dd, J = 12.50, 5.40 Hz, 1 H), 2.74 (brs, 1 H); 13 C NMR (125 MHz, CDCl3): δ 159.8, 158.3, 133.4, 129.5, 121.3, 115.3, 114.6, 113.9, 99.5, 70.3, 69.6, 67.3, 55.3, 32.8; HRMS (ESI): calcd for C18H18O3Se [M + H]+ 363.0494.

1-((4-fluorophenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3g)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (43.4 mg, 62% yield). 1 H NMR (500 MHz, CDCl3): δ 7.32–7.24 (m, 4 H), 6.97–6.89 (m, 5 H), 4.37–4.34 (m, 1 H), 4.15–4.10 (m, 2 H), 3.17 (dd, J = 12.50, 5.50 Hz, 1 H), 3.07 (dd, J = 12.51, 5.50 Hz, 1 H), 2.85 (d, J = 5.40 Hz, 1 H); 13 C NMR (125 MHz, CDCl3): δ 162.5 (d, JF = 249.9 Hz), 158.4 (d, JF = 19.7 Hz), 133.6 (d, JF = 8.3 Hz), 129.6, 121.4, 121.3, 119.3 (d, JF = 3.2 Hz), 115.6, 115.5, 114.7, 98.4, 70.3, 69.5, 69.4, 32.8; 19 F NMR (470 MHz, CDCl3): δ -110.37 (s, 1 F); HRMS (ESI): calcd for C17H15FO2Se [M + H]+ 351.0294, found 351.0283.

1-((2-chlorophenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3h)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (60.0 mg, 82% yield). 1 H NMR (500 MHz, CDCl3): δ 7.34–7.32 (m, 2 H), 7.27–7.22 (m, 2 H), 7.18–7.12 (m, 2 H), 6.94–6.88 (m, 3 H), 4.41–4.40 (m, 1 H), 4.12–4.11 (m, 2 H), 3.20 (dd, J = 12.50, 5.40 Hz, 1 H), 3.09 (dd, J = 12.50, 5.40 Hz, 1 H), 2.94–2.93 (d, J = 3.90 Hz, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.4, 135.6, 133.0, 129.6, 129.2, 129.1, 126.5, 123.2, 121.4, 121.3, 114.7, 96.5, 76.1, 70.4, 69.5, 33.2; HRMS (ESI): calcd for C17H15ClO2Se [M + H]+ 366.9998, found 366.9999.

1-((3-chlorophenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3i)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (62.2 mg, 85% yield). 1 H NMR (500 MHz, CDCl3): δ 7.30–7.29 (m, 1 H), 7.27–7.22 (m, 3 H), 7.19–7.14 (m, 2 H), 6.97–6.94 (m, 1 H), 6.90–6.88 (m, 2 H), 4.36–4.33 (m, 1 H), 4.15–4.09 (m, 2 H), 3.18 (dd, J = 12.45, 5.50 Hz, 1 H), 3.07 (dd, J = 12.45, 5.50 Hz, 1 H), 2.91–2.90 (m, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.3, 134.1, 131.3, 129.6, 129.5, 128.6, 124.8, 121.5, 114.7, 114.6, 98.2, 71.8, 70.2, 69.5, 32.9; HRMS (ESI): calcd for C17H15ClO2Se [M + H]+ 366.9998, found 366.9999.

1-((4-bromophenyl)ethynylselanyl)-3-phenoxypropan-2-ol (3j)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (65.6 mg, 80% yield), Mp = 74–75 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.38–7.36 (m, 2 H), 7.27–7.23 (m, 2 H), 7.16–7.14 (m, 2 H), 6.97–6.93 (m, 1 H), 6.90–6.88 (m, 2 H), 4.36–4.33 (m, 1 H), 4.15–4.09 (m, 2 H), 3.18 (dd, J = 12.43, 5.50 Hz, 1 H), 3.07 (dd, J = 12.43, 5.50 Hz, 1 H), 2.88–2.83 (m, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.3, 132.9, 131.5, 129.6, 129.5, 122.5, 122.1, 121.4, 121.3, 114.6, 98.5, 71.2, 70.2, 69.5, 32.8; HRMS (ESI): calcd for C17H15BrO2Se [M + H]+ 410.9493, found 410.9504.

4-((2-hydroxy-3-phenoxypropylselanyl)ethynyl)benzonitrile (3k)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (46.4 mg, 65% yield), Mp = 71–72 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.55 (d, J = 8.1 Hz, 2 H), 7.35 (d, J = 8.1 Hz, 2 H), 7.30 (t, J = 7.8 Hz, 2 H), 7.00 (t, J = 7.4 Hz, 1 H), 6.94 (d, J = 8.1 Hz, 2 H), 4.42–4.37 (m, 1 H), 4.18 (d, J = 4.9 Hz, 2 H), 3.28 (dd, J = 12.5, 5.6 Hz, 1 H), 3.17 (dd, J = 12.5, 6.6 Hz, 1 H), 2.80 (brs, 1 H); 13 C NMR (125 MHz, CDCl3): δ 158.2, 131.9, 131.6, 129.6, 127.9, 131.5, 118.4, 114.6, 111.2, 98.2, 76.3, 69.9, 69.4, 33.1; HRMS (ESI): calcd for C18H15NO2Se [M + H]+ 358.0340, found 358.0325.

1-phenoxy-3-(thiophen-3-ylethynylselanyl)propan-2-ol (3L)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (54.7 mg, 81% yield). 1 H NMR (500 MHz, CDCl3): δ 7.33–7.27 (m, 3 H), 7.21 (dd, J = 3.6, 1.2 Hz, 1 H), 7.02–6.95 (m, 4 H), 4.42–4.38 (m, 1 H), 4.21–4.14 (m, 2 H), 3.21 (dd, J = 12.5, 5.4 Hz, 1 H), 3.11 (dd, J = 12.4, 6.9 Hz, 1 H), 2.79 (brs, 1 H);13 C NMR (125 MHz, CDCl3): δ158.4, 129.9, 129.6, 129.5, 125.2, 122.3, 121.4, 121.3, 114.7, 94.5, 70.3, 69.5, 69.1, 68.8, 32.8; HRMS (ESI): calcd for C15H14O2SSe [M + H]+ 338.9952, found 338.9954.

1-(phenylethynylselanyl)-3-(p-tolyloxy)propan-2-ol (4a)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (63.6 mg, 92% yield). 1 H NMR (500 MHz, CDCl3): δ 7.36–7.34 (m, 2 H), 7.30–7.24 (m, 3 H), 7.05 (d, J = 8.40 Hz, 2 H), 6.81 (d, J = 8.40 Hz, 2 H), 4.37–4.35 (m, 1 H), 4.15–4.09 (m, 2 H), 3.18 (dd, J = 12.50, 5.40 Hz, 1 H), 3.08 (dd, J = 12.5, 5.40 Hz, 1 H), 2.76 (brs, 1 H), 2.27 (s, 3 H); 13 C NMR (125 MHz, CDCl3): δ 156.2, 131.6, 130.6, 130.0, 128.3, 128.2, 123.2, 114.5, 99.6, 70.5, 69.6, 69.5, 32.8, 20.4; HRMS (ESI): calcd for C18H18O2Se [M + H]+ 347.0545, found 347.0541.

1-(4-bromophenoxy)-3-(phenylethynylselanyl)propan-2-ol (4b)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (61.5 mg, 75% yield). 1 H NMR (500 MHz, CDCl3): δ 7.35–7.25 (m, 7 H), 6.81–6.77 (m, 2 H), 4.38–4.36 (s, 1 H), 4.15–4.09 (m, 2 H), 3.17 (dd, J = 12.50, 5.40 Hz, 1 H), 3.07 (dd, J = 12.50, 5.40 Hz, 1 H), 2.74 (brs, 1 H); 13 C NMR (125 MHz, CDCl3): δ 157.4, 132.4, 131.6, 128.4, 128.3, 123.0, 116.4, 113.5, 99.7, 70.5, 69.4, 69.2, 32.7.

1-(phenylethynylselanyl)-3-(4-(trifluoromethoxy)phenoxy)propan-2-ol (4c)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (64.1 mg, 77% yield), Mp = 48–49 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.35–7.34 (m, 5 H), 7.12–7.11 (m, 2 H), 6.91–6.89 (m, 2 H), 4.39–4.38 (m, 1 H), 4.18–4.12 (m, 2 H), 3.18 (dd, J = 12.50, 5.40 Hz, 1 H), 3.08 (dd, J = 12.50, 5.40 Hz, 1 H), 2.74–2.73 (m, 1 H); 13 C NMR (125 MHz, CDCl3): δ 156.8, 143.2, 131.5, 128.4, 128.3, 123.6, 123.0, 122.4, 121.5, 119.5, 117.5, 115.4, 99.7, 70.7, 69.4, 69.2, 32.7; 19 F NMR (470 MHz, CDCl3): δ -58.4 (s, 3 F). HRMS (ESI): calcd for C18H15F3O3Se [M + H]+ 417.0211, found 417.0216.

1-(4-nitrophenoxy)-3-(phenylethynylselanyl)propan-2-ol (4d)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (54.3 mg, 72% yield), Mp = 71–72 ℃. 1 H NMR (500 MHz, CDCl3): δ 8.16–8.12 (m, 2 H), 7.33–7.24 (m, 5 H), 6.99–6.96 (m, 2 H), 4.45–4.42 (m, 1 H), 4.30–4.25 (m, 2 H), 3.20 (dd, J = 12.50, 5.40 Hz, 4 H), 3.09 (dd, J = 12.50, 5.40 Hz, 1 H), 2.73–2.72 (m, 1 H); 13 C NMR (125 MHz, CDCl3): δ 163.2, 141.9, 131.6, 128.6, 128.3, 125.9, 122.8, 114.6, 99.8, 70.8, 69.3, 68.9, 32.5.

1-(3-(diethylamino)phenoxy)-3-(phenylethynylselanyl)propan-2-ol (4e)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (66.9 mg, 83% yield). 1 H NMR (500 MHz, CDCl3): δ 7.38–7.36 (m, 2 H), 7.28–7.25 (m, 3 H), 7.10–7.07 (m, 1 H), 6.33–6.31 (m, 1 H), 6.24–6.21 (m, 2 H), 4.37–4.36 (m, 1 H), 4.17–4.11(m, 2 H), 3.30 (dd, J = 14.0, 7.0 Hz, 4 H), 3.19 (dd, J = 12.50, 5.40 Hz, 1 H), 3.09 (dd, J = 12.50, 5.40 Hz, 1 H), 2.74–2.73 (m, 1 H), 1.14 (s, 6 H); 13 C NMR (125 MHz, CDCl3): δ 159.7, 149.3, 131.6, 130.0, 128.3, 123.2, 105.7, 100.8, 99.6, 98.9, 70.1, 69.6, 69.5, 44.4, 32.9, 12.6; HRMS (ESI): calcd for C21H25NO2Se [M + H]+ 404.1123, found 404.1129.

1-(naphthalen-2-yloxy)-3-(phenylethynylselanyl)propan-2-ol (4f)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford light yellow solid (68.7 mg, 90% yield), Mp = 63–64 ℃. 1 H NMR (500 MHz, CDCl3): δ 7.76–7.71 (m, 2 H), 7.65 (d, J = 8.18 Hz, 1 H), 7.43–7.40 (m, 1 H), 7.35–7.32 (m, 3 H), 7.28–7.20 (m, 3 H), 7.17–7.15 (m, 2 H), 4.48–4.43 (m, 1 H), 4.31–4.25 (m, 2 H), 3.23 (dd, J = 12.50, 5.40 Hz, 4 H), 3.13 (dd, J = 12.50, 5.40 Hz, 1 H), 2.81 (brs, 1 H); 13 C NMR (125 MHz, CDCl3): δ 156.3, 134.4, 131.6, 129.6, 129.3, 128.3, 128.2, 127.6, 126.8, 126.5, 123.9, 123.1, 118.6, 107.1, 99.7, 70.4, 69.6, 69.4, 32.8; HRMS (ESI): calcd for C21H18O2Se [M + H]+ 383.0544, found 383.0537.

1-(benzo[d][1,3]dioxol-5-yloxy)-3-(phenylethynylselanyl)propan-2-ol (4g)

Following the general procedure, using (petroleum ether: EtOAc = 7 : 1) as the eluant to afford yellow oil liquid (58.6 mg, 78% yield). 1 H NMR (500 MHz, CDCl3): δ 7.37–7.35 (m, 2 H), 7.29–7.27 (m, 3 H), 6.67 (d, J = 8.45 Hz, 1 H), 6.50 (d, J = 2.45 Hz, 1 H), 6.34 (dd, J = 8.45 Hz, 2.45 Hz, 1 H), 5.90 (s, 2 H), 4.36–4.33 (m, 1 H), 4.11–4.05 (m, 2 H), 3.17 (dd, J = 12.50, 5.40 Hz, 4 H), 3.07 (dd, J = 12.50, 5.40 Hz, 1 H), 2.72 (d, J = 5.15 Hz, 1 H); 13 C NMR (125 MHz, CDCl3): δ 153.8, 148.3, 142.1, 131.6, 128.3, 128.2, 123.1, 107.9, 105.9, 101.2, 99.6, 98.3, 71.3, 69.5, 69.4, 32.8; HRMS (ESI): calcd for C18H16O4Se [M + H]+ 377.0286, found 377.0280.

1-(5-isopropyl-2-methylphenoxy)-3-(phenylethynylselanyl)propan-2-ol (4h)

Following the general procedure, using (petroleum ether: EtOAc = 7: 1) as the eluant to afford yellow oil liquid (60.5 mg, 78% yield). 1 H NMR (500 MHz, CDCl3): δ 7.34–7.32 (m, 2 H), 7.28–7.24 (m, 3 H), 7.04 (d, J = 7.60 Hz, 1 H), 6.76–6.74 (m, 1 H), 6.70 (s, 1 H), 4.42–4.39 (m, 1 H), 4.18 (d, J = 4.95 Hz, 2 H), 3.23 (dd, J = 14.0, 7.0 Hz, 4 H), 3.11 (dd, J = 12.4, 6.3 Hz, 1 H), 2.84–2.78 (m, 1 H), 2.19 (s, 3 H), 1.20 (d, J = 6.90 Hz, 6 H); 13 C NMR (125 MHz, CDCl3): δ 156.3, 148.1, 131.5, 130.6, 128.3, 124.0, 123.2, 118.8, 109.8, 99.5, 70.3, 69.8, 69.5, 34.1, 33.0, 24.1, 15.8; HRMS (ESI): calcd for C21H24O2Se [M + H]+ 389.1014, found 389.1010.

1-(((2-fluorophenyl)ethynyl)selanyl)-3-phenoxypropan-2-ol (3 h-1)

Following the general procedure, using (petroleum ether: EtOAc = 9:1) as the eluent afforded a yellow liquid (53.9 mg, 77% yield). 1 H NMR (400 MHz, CDCl3): δ 7.38 (td, J = 7.6, 1.9 Hz, 1 H), 7.31 (dd, J = 10.7, 5.2 Hz, 3 H), 7.10 (td, J = 8.7, 8.0, 2.9 Hz, 2 H), 6.99 (dd, J = 18.0, 7.8 Hz, 3 H), 4.45 (q, J = 5.5 Hz, 1 H), 4.24–4.17 (m, 2 H), 3.26 (dd, J = 12.5, 5.4 Hz, 1 H), 3.16 (dd, J = 12.5, 6.9 Hz, 1 H), 2.97 (t, J = 6.6 Hz, 1 H); 13 C NMR (100 MHz, CDCl3): δ 162.91 (d, J = 251.3 Hz), 158.39, 133.43, 130.08 (d, J = 7.9 Hz), 129.66, 124.04 (d, J = 3.7 Hz), 121.41, 115.56 (d, J = 20.8 Hz), 114.68, 111.90 (d, J = 15.7 Hz), 92.90, 75.62, 70.36, 69.56, 33.04; 19 F NMR (375 MHz, CDCl3): -109.96; HRMS (ESI): calcd for C17H16FO2Se [M + H]+ 351.0299, found 351.0301.

1-(((2-bromophenyl)ethynyl)selanyl)-3-phenoxypropan-2-ol (3h-2)

Following the general procedure, using (petroleum ether: EtOAc = 9:1) as the eluant afforded a yellow liquid (64.8 mg, 79% yield). 1 H NMR (400 MHz, CDCl3): δ 7.60 (dd, J = 8.0, 1.3 Hz, 1 H), 7.42 (dd, J = 7.7, 1.7 Hz, 1 H), 7.35–7.30 (m, 2 H), 7.26 (td, J = 7.6, 1.3 Hz, 1 H), 7.17 (td, J = 7.7, 1.7 Hz, 1 H), 7.03–6.97 (m, 3 H), 4.51 (q, J = 5.4 Hz, 1 H), 4.24–4.17 (m, 2 H), 3.30 (dd, J = 12.4, 5.3 Hz, 1 H), 3.17 (dd, J = 12.4, 7.0 Hz, 1 H), 3.03 (t, J = 5.6 Hz, 1 H); 13 C NMR (100 MHz, CDCl3): δ 158.39, 133.15, 132.44, 129.69, 129.39, 127.16, 125.37, 125.24, 121.43, 114.72, 98.34, 75.69, 70.41, 69.53, 33.17; HRMS (ESI): calcd for C17H16BrO2Se [M + H]+ 410.9499, found 410.9502.

1-(((2-methoxyphenyl)ethynyl)selanyl)-3-phenoxypropan-2-ol (3h-3)

Following the general procedure, using (petroleum ether: EtOAc = 9:1) as the eluent afforded a yellow liquid (65.9 mg, 91% yield). 1 H NMR (400 MHz, CDCl3): δ 7.38 (dd, J = 7.6, 1.8 Hz, 1 H), 7.34–7.30 (m, 3 H), 7.00 (dd, J = 17.5, 7.9 Hz, 3 H), 6.95–6.88 (m, 2 H), 4.51 (q, J = 5.5 Hz, 1 H), 4.24–4.17 (m, 2 H), 3.88 (d, J = 1.3 Hz, 3 H), 3.27–3.11 (m, 3 H); 13 C NMR (100 MHz, CDCl3): δ 160.24, 158.49, 133.31, 129.88, 129.64, 121.32, 120.54, 114.68, 112.45, 110.62, 96.17, 73.59, 70.54, 69.86, 55.80, 32.89; HRMS (ESI): calcd for C18H19O3Se [M + H]+ 363.0499, found 363.0500.

Cell culture

The human lung adenocarcinoma cell line A549, the human hepatoblastoma cell line HepG2, and the human colon carcinoma cell line RKO were purchased from the Shanghai Cell Bank of the Chinese Academy of Sciences (Shanghai, China). These cells were cultured in RPMI 1640 or DMEM (4.5 g/l glucose) supplemented with 10% fetal bovine serum in a humidified incubator with 5% CO2 at 37 °C.

Cell viability assay

Compounds were dissolved in DMSO and diluted with the culture medium. For the MTT assay, the cells were grown in 96-well plates (5 × 103 cells/well) for 24 h and treated with various concentrations of synthetic compounds for 48 h. The cells were incubated with the MTT assay kit for another 4 h. The dark blue crystals (formazan) were dissolved in DMSO. The absorbance was measured using a multifunction microplate reader (Molecular Devices, Flex Station 3) at 570 nm. The cytotoxic effects of each compound were expressed as IC50 values. All experiments were performed in triplicate in three independent experiments.

Flow cytometric analysis

A549 cells were incubated in six-well plates (3 × 105 cells/well) for 24 h. The cells were treated in the presence or absence of compounds 3h, 3g, and 3h-2 at 1 µmol for 12 h. The cells were then incubated with an Annexin V-FITC/PI Apoptosis Detection Kit according to the manufacturer’s instructions. Almost 10,000 events were collected for each sample and were analyzed by flow cytometry (Beckman Coulter, Epics XL). The results were calculated using EXPO32 ADC analysis software.

Western blot analysis

The harvested cells were lysed with cold RIPA lysis buffer (Solarbio, Beijing, China) with a protease inhibitor cocktail (Thermo Fisher Scientific, MA) to obtain total protein. Protein concentrations were determined using the modified Coomassie bright blue method. An aliquot of protein was separated using different sodium dodecyl sulfate‒polyacrylamide gel electrophoresis and transferred onto 0.45-µm polyvinylidene difluoride membranes (Solarbio, Beijing, China). Blots were cut prior to hybridization with antibodies during blotting. Membranes were blocked with 5% skim milk at room temperature for 2 h and incubated overnight at 4 °C with primary antibodies against Cyclin D1 (1:1000), Bax (1:1000), Bcl2 (1:1000), cleaved caspase 3 (1:1000), or β-actin (1:1000). Protein bands were visualized using a chemiluminescence reagent (TransGen Biotech, DW101-01) with horseradish peroxidase-conjugated secondary antibodies and quantified using ImageJ software (version 1.44p, NIH).

TUNEL assay

A549 cells were fixed with 4% paraformaldehyde and stained using a TUNEL System TB235 kit. Stained cells were visualized using a Nikon confocal microscope.

Cell migration assay

A549 cells were seeded in 24-well plates (1 × 104 per well) and grown to confluence before serum starvation for 12 h. The monolayers were scratched with a 10-µl pipet or needle tip. We removed the culture medium and washed the cells with PBS three times to eliminate floating cells. The medium for cell culture was replaced with serum-free RPMI 1640 medium and incubated with compounds 3h, 3g, and 3h-2 (1 µmol). The changes in the wound area were measured 24 or 48 h later. The cell images were detected using a light microscope (Nikon, Japan).

RNA isolation and real-time PCR

Total RNA was extracted from A549 cells incubated for 24 h with or without the addition of compounds of 1 µmol 3h using Trizol reagent. We used a NanoDrop™ One device (Thermo Scientific, ND-ONE-W) to measure RNA concentrations. cDNA was reverse-transcribed using a SuperScript™ II Reverse Transcriptase kit (TransGen Biotech, AT341). qRT‒PCR was performed using PerfectStart™ Green qPCR SuperMix (TransGen Biotech, AQ601) on a CFX96 Touch Real-Time PCR Detection System (Bio-Rad, Inc., Hercules, USA). The primers are listed in Supplementary Table S1. The relative amounts of the mRNAs were expressed as 2−ΔΔCT.

In vivo xenograft Tumor model study

Five-week-old male BALB/c nude mice (18–20 g, n = 10) were purchased from Charles River Laboratories (Beijing, China). The Animal Care and Use Committee of Wenzhou Medical University, China, approved all animal procedures. The nude mice were maintained in an animal chamber free of pathogens and fed with sterilized water and chow. The xenograft model was established by subcutaneous injection of A549 cells in the logarithmic growth phase into the right axilla. When the tumor volume reached approximately 100 mm3, the xenograft tumor-bearing nude mice were randomly divided into two groups (five in each setting): vehicle-treated and 3h-treated. The mice in the 3h-treated group were intraperitoneally injected with compound 3h (2.5 mg/kg, once a day), and the mice in the vehicle-treated group were intraperitoneally injected with the same volume of PBS. The body weight and tumor volume were measured every other day during the experiment. The tumor volume was calculated as follows: volume (mm3) = 0.5 × length (mm) × width (mm)2. After two weeks, all mice were euthanized with pentobarbital sodium (100 mg/kg, i.p.), and the tumors were peeled off, photographed, harvested, and weighed.

Protection of Selenocompounds against cisplatin damage to normal cells

Prior to treatment, renal podocytes (MPC) and liver (AML-12) cells were cultured in serum-free media for 10–12 h and divided into: (i) Normal group; (ii) Cisplatin group (20 µmol cisplatin), and (iii) Compound 3h intervention groups (20 µmol cisplatin-treated with 0.01, 0.1, 1, 10 µmol compounds 3h). MTT and TUNEL experiments were carried out.

Statistical analysis

All statistical analyses were performed using GraphPad Prism 9.0. Data are presented as the mean ± SEM. Each set of experiments was repeated independently at least three times. Student’s-t test was performed to determine the significance of differences between pairs. For the comparison of more than two groups, one-way ANOVA was used. A value of P < 0.05 was considered statistically significant.

Results and discussion

Chemistry

The synthesis route of 3a-3L is shown in Fig. 1. Phenylpropionic acid with different substituted functional groups (1), Se powder, and glycidylphenyl ether epoxide (2a) were used as substrates. CuCl2 was used as the catalyst, 1, 10-phen was the efficient ligand, Cs2CO3 was the base, and TBAI was the phase transfer catalyst in H2O at 50 °C to obtain compounds 3a-3L in the yields of 62%~94%. For compounds 3b-3k, they are alkyl (3b-3d), phenyl (3e), methoxy (3f), halogen (3g-3j), and cyano (3k) substituted 1-((phenylethynyl)selanyl)-3-phenoxypropan-2-ol, respectively.

Fig. 1
figure 1

General synthesis of selenium-containing compounds 3a‒3L

The synthesis route of 4a‒4h is shown in Fig. 2. Phenylpropiolic acid (1a), Se powder, and various glycidyl phenyl ether epoxides (2) were used as reagents. The synthesis conditions were consistent with 3a-3L. Glycidyl phenyl ether epoxides (2) were synthesized by reacting various substituted phenols with epichlorohydrin in the presence of phase transfer catalysts and sodium hydroxide. Different substituents of glycidyl phenyl ether epoxides were used to investigate the effects of changes in these functional groups. In the method section, all the synthesized chemical structures were characterized by ESI-MS, 1 H NMR, and 13 C NMR.

Fig. 2
figure 2

General synthesis of selenium-containing compounds 4a‒4h

Structure-activity study

Se-organic compounds reduce the viability of various cancer cell lines [23]. Inspired by these studies, we designed and synthesized two series of structurally related selenium-containing molecules, and their antitumor activity was tested in A549 cells (non-small cell lung carcinoma) [24], HepG2 cells (liver hepatocellular) [25] and RKO cells (human colorectal carcinoma) [26]. A structure-activity relationship analysis based on three cancer cell lines showed that the anti-proliferative activity of 3a‒3L was significantly better than that of 4a‒4h. For compounds 3a–3L, the functional groups alkyl (3b‒3d), methoxyl (3f), halogen (3g–3j), phenyl (3e), and cyano (3k) were substituted for aryl propionic acid 1. We found that arylpropiolic acids bearing electron-deficient substituents (3g–3L, 3i–3k) showed better activity than those bearing electron-donating substituents (3a, 3b, 3d, and 3f). When the phenyl ring carried a methyl group, the anti-proliferation activity of compounds 3a‒3c against cancer cells increased with the number of substituents; compound 3c showed the most potent anti-proliferation activity. Compound 3d, a tert-butyl substitution, showed no inhibitory activity, possibly due to its rigid structure. Compounds 3e‒3k contain electron-withdrawing groups. Compounds 3h and 3g showed the most potent tumor inhibition activity; their structures are ortho- and meso-substituted phenyls with a chlorine atom, respectively. Thiophene was substituted for phenyl in compound 3L, which reduced its antitumor activity (Table 1).

Table 1 Anti-proliferative activity of compounds 3a‒3L against A549, HepG2, and RKO cell lines

The anti-proliferative activities of skeleton structure compounds 4a‒4h were also analyzed. None of these compounds had very high antitumor proliferation activity, except for 4c and 4h (Table 2).

Table 2 Anti-proliferative activity of compounds 4a‒4h against A549, HepG2, and RKO cell lines

Because compound 3h showed an excellent inhibitory effect in all cancer cell lines, we synthesized compounds 3h-1, 3h-2, and 3h-3 to determine whether other electron-absorbing or electron donor groups linked to phenyl would generate a better tumor-inhibiting compound. We found that these derivatives exhibited good antitumor activity, and compound 3h-2 was the best (Table 3). These data suggest that 3h, 3g, and 3h-2 have the potential for further investigation. Nonetheless, the antitumor activity of Se compounds synthesized from 3h was not superior to that of 3h, suggesting that some core structures with tumor inhibition can be introduced in subsequent structural optimization to enhance antitumor activity [27, 28].

Table 3 Anti-proliferative activity of compounds 3h-1‒3h-3 against A549, HepG2, and RKO cell lines

Effect of compounds 3h, 3g, and 3h-2 on A549 cell migration

Since compounds 3h, 3g, and 3h-2 showed suitable anti-proliferative activities against cancer cells, we measured the ability of 3h, 3g, and 3h-2 to inhibit cell migration. Typical images were taken at the beginning of the experiment (0h) and after treatment with compounds or vehicle for 24 or 48 h. Compounds 3h, 3g, and 3h-2 inhibited the migration ability of A549 cells in a time-dependent manner (Fig. 3A). The cell migration rate was decreased by 60.2% after treatment with 1 µmol of 3h for 48 h. The same result was observed after treatment with 3g and 3h-2, with the cell migration rate decreasing by 46.4% and 39.6%, respectively (Fig. 3B>). These results suggest that 3h, 3g, and 3h-2 inhibit the migration and invasion of A549 cells and may be chemotherapy agents for metastatic cancer.

Fig. 3
figure 3

Effect of 3h, 3g, and 3h-2 on the migration of A549 cells. (A, B) A549 cells treated with compound 3h, 3g, and 3h-2 (1 µmol) in free serum DMEM for 48 h. (A) Representative images of A549 cells at 0 h, 24 h, and 48 h post scratch (n = 5). Scale bars = 50 μm. (B) The quantitative ratio of the cell migration area to the scratch area at 0 h. All data are expressed as mean ± SEM. ***p < 0.001, **p < 0.01 compared to 0 h

Effect of compounds 3h, 3g, and 3h-2 on A549 cell apoptosis using Annexin V-FITC/PI

We conducted several experiments in vitro to determine whether the tumor inhibition effect was mediated by apoptosis. The ability of compounds 3h, 3g, and 3h-2 to induce apoptosis was evaluated by measuring apoptosis using flow cytometry in human cancer cells. Treating A549 cells with 3h, 3g, and 3h-2 at 1 µmol for 12h led to early-stage cell apoptosis at 25.49%, 10.83%, and 7.68%, respectively. There was only 4.39% apoptosis in untreated cells (Fig. 4A and B). This finding suggests that the inhibition of cell proliferation mediated by 3h, 3g, and 3h-2 is related to increased apoptosis, especially for 3h.

Fig. 4
figure 4

Apoptosis analysis of A549 lung tumor cells using flow cytometry. (A) Dot plot representation of annexin-V-FITC fluorescence (X-axis) vs. PI fluorescence (Y-axis) of apoptotic A549 (annexin-V positive) cells treated with 3h, 3g, and 3h-2 (1 µmol) for 12h (n = 3). (B) Early apoptotic cell percentages were obtained by EXPO32 ADC analysis software. All data are expressed as mean ± SEM. ***p < 0.001, **p < 0.01 compared to the control group

Effect of compounds 3h, 3g, and 3h-2 on cancer and normal cells

TUNEL staining showed that A549 cells treated with 3h, 3g, and 3h-2 exhibited more significant numbers of apoptotic cells with fragmented nuclei than the control (Fig. 5A). The order of TUNEL-positive cells was as follows: 3h > 3g > 3h-2 > Control (Fig. 5B). These results suggest that compounds 3h, 3g, 3h-2, and (especially) 3h inhibit cell proliferation and promote apoptosis in A549 cells.

To investigate whether these compounds are active against normal cells, we conducted a toxicity experiment on podocyte cells (MPC) and hepatocyte cells (AML-12), which are two normal cell types involved in drug metabolism. The MTT results indicated that the three representative compounds exhibited almost no toxic side effects on normal liver cells and renal tubular cells even at a concentration of 10 µM, far exceeding the IC50 value of the compounds for cancer cell lines (Supplementary Fig. S3).

Fig. 5
figure 5

TUNEL staining of compounds 3h, 3g, and 3h-2 in A549 cells. (A) Representative images of TUNEL staining of A549 cells treated with compounds 3h, 3g, and 3h-2 at 1 µmol concentration for 24 h (n = 4). Scale bars = 100 μm. (B) Quantification of TUNEL-positive cells in panel A. All data are expressed as mean ± SEM. ***p < 0.001, **p < 0.01 compared to the control group; n.s., not significant

Mechanistic study of compound 3h on A549 cells at the protein level

Cyclin D1 promotes cell proliferation by promoting the cell cycle transition from G1 to S phase via binding and activating CDK4, a Cyclin-dependent kinase specific to the G1 phase. The abundance of Cyclin D1 was less than that of the control after treatment with compound 3h at 1 µmol (Fig. 6A and B).

The effect of compound 3h on apoptosis was also tested. After treatment with compound 3h, the pro-apoptotic proteins cleaved Caspase 3 and Bax were dose-dependently upregulated, while the anti-apoptotic protein Bcl2 was downregulated in A549 cells (Fig. 6A C-E).

Fig. 6
figure 6

Cell signaling in A549 cells treated with 3h at different concentrations (0.1, 1, and 10 µmol) for 24h. (A) Western blotting analysis of the expression of Cyclin D1, Bcl2, Bax, and cleaved Caspase 3 (n = 3). β-actin was used as a loading control. (B-E) Statistical analysis of the relative protein intensity of (B) Cyclin D1, (C) cleaved Caspase 3, (D) Bax, and (E) Bcl2 in A549 cells. Full-length gels are presented in Supplementary Fig. S1. All data are expressed as mean ± SEM. *p < 0.05, **p < 0.01 compared to the control group; n.s., not significant

Proliferation and apoptosis-related mRNA levels

We then investigated the correlation of 3h, which induced cell cycle arrest, with alterations in the expression of proteins that regulate cell division. CDK1, CDK2, and Cyclin B1 induce eukaryotic cells to enter mitosis. We measured the relative mRNA levels and found that CDK1, CDK2, and Cyclin B1 mRNA were decreased after treatment with compound 3h (Fig. 7A-C). Real-time PCR quantification revealed that Cyclin D1 levels were markedly higher in the control group than in the 3h-treated group (Fig. 7D). The mRNA levels of the cell proliferation-related proteins Ki67 and PCNA showed the same trend (Fig. 7E and F).

p21 is a member of the cyclin-dependent kinase inhibitor family. It is associated with tumor inhibition and inhibits CDK activity. qRT‒PCR quantification revealed that the expression of p21 was markedly higher in the 3h-treated group than in the control group (Fig. 7G). These results suggest that compound 3h induces cell cycle arrest and reduces cell proliferation in A549 cells. In addition, Bax/Bcl2 mRNA levels were significantly increased after treatment with compound 3h at 1 µmol (Fig. 7H).

Fig. 7
figure 7

Relative mRNA levels of (A) CDK1, (B) CDK2, (C) Cyclin B1, (D) Cyclin D1, (E) Ki67, (F) PCNA, (G) p21, and (H) Bax/Bcl2 genes in the A549 cells that were treated with 3h at 1 µmol concentrations or PBS control for 24 h. Relative mRNA levels were normalized to that of β-actin. The data were from six independent measurements. All data are expressed as mean ± SEM. ***p < 0.001, *p < 0.05 compared to the control group

Antitumor activity of compound 3h in a xenograft tumor model

A xenograft model was established by subcutaneous injection of A549 cells at the logarithmic growth phase into the right axilla of mice. When the tumor size reached approximately 100 mm3, the mice were randomly divided into two groups (PBS-treated and 3h-treated groups), with five mice per group. The mice were intraperitoneally injected with compound 3h at 2.5 mg/kg daily during the observation period. After treatment for 14 days, the tumor was excised and weighed. The mean tumor volume in the PBS-treated group was 1.94 times that of the 3h-treated group (Fig. 8A and B). Treatment with 3h significantly reduced tumor size and weight (Fig. 8C). In addition, we also evaluated the impact of 3h on the growth and apoptosis of mouse tumors using Western blotting. Compared with the control group, the expression of Cyclin D1 in total tumor lysates from mice after 3h treatment was significantly decreased, while the pro-apoptotic proteins c-Caspase3 and Bax were upregulated and the anti-apoptotic protein Bcl2 was downregulated (Fig. 8D and E). Although compound 3h exerts strong antitumor activity, the specific mechanisms and targets of this series of compounds still need to be further elucidated.

Fig. 8
figure 8

In vivo antitumor effect of compound 3h. (A-E) A549 cells were injected into the flanks of nude mice. When the tumor volume reached approximately 100 mm3, the nude mice were sorted into two groups (n = 5) and one group was intraperitoneally injected with 3h (2.5mg/kg, once a day) for 14 days. (A) Images of sacrificed mice and excised tumors in each group. (B) The development of tumor size during the treatment period was recorded. (C) At the end of the experiment, tumors were resected and weighed. (D) Western blotting analysis of the expression of Cyclin D1, c-Caspase3, Bax, and Bcl2 in total tumor lysates from vehicle- and 3h-treated mice (n = 3). β-actin was used as a loading control. (E) Expression levels were quantitated using ImageJ software. Full-length gels are presented in Supplementary Fig. S2. All data are expressed as mean ± SEM. ***p < 0.001, **p < 0.01, *p < 0.05 compared to the control group

Protection of compound 3h against cisplatin-induced damage to normal cells

Nephrotoxicity and hepatotoxicity are significant dose-limiting side effects in cisplatin-based chemotherapy [29, 30]. Se compounds protect normal cells from damage. Considering the organoselenium compounds have synergistic effects when combined with chemotherapeutic drugs, we explored the protective activity of 3h against cisplatin-induced MPC and AML-12 cells. As shown in Fig. 9A and C, the viability of MPC and AML-12 cells was reduced after treatment with cisplatin, which was significantly increased in a dose-dependent manner after 3h treatment. TUNEL results showed that the number of positive cells significantly decreased after 3h treatment compared with the cisplatin-induced group (Fig. 9B and D). These findings suggest that compound 3h has a protective effect on normal cells, and a synergistic effect can be achieved when combined with cisplatin.

Fig. 9
figure 9

MTT assay and TUNEL staining in MPC and AML-12 cells. (A) MPC cells were cultured with 20 µmol cisplatin or 20 µmol cisplatin combined with 0.01, 0.1, 1, and 10 µmol 3h for 12 h, and cell viability was determined by MTT test. (B) Representative images and quantification of TUNEL staining of MPC cells treated with 20 µmol cisplatin or 20 µmol cisplatin combined with 1 µmol 3h for 12 h. Scale bars = 100 μm. (C) AML-12 cells were cultured with 20 µmol cisplatin or 20 µmol cisplatin combined with 0.01, 0.1, 1, 10 µmol 3h for 12 h, and cell viability was determined by MTT assay. (D) Representative images and quantification of TUNEL staining of AML-12 cells treated with 20 µmol cisplatin or 20 µmol cisplatin and 1 µmol 3h for 24h. Scale bars = 100 μm. n = 4 per group. All data are expressed as mean ± SEM. ***p < 0.001, **p < 0.01 compared to the Cis group

Conclusion

Selenocompounds promote cell death via apoptosis and protect cells against oxidative stress-induced death. The anticancer effect of Se might be mediated by the prevention of damage to healthy cells and apoptosis of tumor cells [31]. In the present study, two series of novel phenoxy-((phenylethynyl) selanyl) propan-2-ol derivatives were synthesized, and their anti-proliferation activities were evaluated. These compounds exhibited intense anti-proliferation activity against three human cancer cells, with IC50 values in the submicromolar concentration range. Compounds 3h, 3g, and 3h-2 exhibited the best activity against cancer cells. Further flow cytometry analysis showed that 3h, 3g, and 3h-2 induced G2/M phase arrest and apoptosis of A549 cells. Cellular studies showed that apoptosis induction by 3h was associated with expression changes in several cell cycle-related proteins (e.g., Cyclin B1, Cyclin D1, CDK1, and CDK2) and apoptosis-related proteins (e.g., Bcl-2, Bax, and cleaved-Caspase 3). After treatment with compound 3h, the level of cleaved caspase 3 and the ratio of Bax to Bcl-2 protein level significantly increased. The xenograft tumor experiment in nude mice revealed that compound 3h has antitumor activity in vivo without evident toxicity. Compound 3h protected against cisplatin-induced injury to normal cells. These in vitro and in vivo findings suggest that the 1-phenoxy-3-((phenylethynyl) selanyl) propan-2-ol derivative 3h is a promising lead compound for developing anticancer drugs.

Data Availability

All data generated or analyzed during this study are included in this published article.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Ehudin MA, Golla U, Trivedi D, Potlakayala SD, Rudrabhatla SV, Desai D, Dovat S, Claxton D, Sharma A. Therapeutic benefits of Selenium in Hematological malignancies. Int J Mol Sci. 2022;23(14):7972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L. Current challenges in Cancer Treatment. Clin Ther. 2016;38(7):1551–66.

    Article  PubMed  Google Scholar 

  4. Cui H, Hu Z, Yang K, Huang J, Wu Y, Chen Q, Wei R, Wang P, Wang H, Li H, Chen Y, Lu T, Yao Y, Zhu Y. Design and synthesis of highly TRAIL expression HDAC inhibitors based on ONC201 to promote apoptosis of Colorectal cancer. Eur J Med Chem. 2022;238:114484.

    Article  CAS  PubMed  Google Scholar 

  5. Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D’Orazi G. Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging. 2016;8(4):603–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. D’Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582–92.

    Article  PubMed  Google Scholar 

  7. Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: Disease message and therapeutic target potentials. Biosci Rep. 2019;39(1):BSR20180992.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17(7):395–417.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Brown KM, Arthur JR. Selenium, selenoproteins and human health: a review. Public Health Nutr. 2001;4(2B):59359–9.

    Article  Google Scholar 

  10. Mistry HD, Broughton Pipkin F, Redman CW, Poston L. Selenium in reproductive health. Am J Obstet Gynecol. 2012;206(1):21–30.

    Article  CAS  PubMed  Google Scholar 

  11. Moghadaszadeh B, Beggs AH. Selenoproteins and their impact on human health through diverse physiological pathways. Physiol (Bethesda). 2006;21:307–15.

    CAS  Google Scholar 

  12. Ibrahim SAZ, Kerkadi A, Agouni A. Selenium and health: an update on the Situation in the Middle East and North Africa. Nutrients. 2019;11(7):1457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zeng H, Cheng WH, Johnson LK. Methylselenol, a selenium metabolite, modulates p53 pathway and inhibits the growth of colon Cancer xenografts in Balb/c mice. J Nutr Biochem. 2013;24(5):776–80.

    Article  CAS  PubMed  Google Scholar 

  14. Tagaram HR, Desai D, Li G, Liu D, Rountree CB, Gowda K, Berg A, Amin S, Staveley-O’Carroll KF, Kimchi ET. A selenium containing inhibitor for the treatment of Hepatocellular Cancer. Pharmaceuticals (Basel). 2016;9(2):18.

    Article  PubMed  Google Scholar 

  15. Karelia DN, Sk UH, Singh P, Gowda ASP, Pandey MK, Ramisetti SR, Amin S, Sharma AK. Design, synthesis, and identification of a novel napthalamide-isoselenocyanate compound NISC-6 as a dual Topoisomerase-IIα and akt pathway inhibitor, and evaluation of its anti-melanoma activity. Eur J Med Chem. 2017;135:282–95.

    Article  CAS  PubMed  Google Scholar 

  16. Gao X, Li X, Mu J, Ho CT, Su J, Zhang Y, Lin X, Chen Z, Li B, Xie Y. Preparation, physicochemical characterization, and anti-proliferation of selenium nanoparticles stabilized by Polyporus Umbellatus polysaccharide. Int J Biol Macromol. 2020;152:605–15.

    Article  CAS  PubMed  Google Scholar 

  17. Bhattacharya A. Methylselenocysteine: a promising antiangiogenic agent for overcoming drug delivery barriers in solid malignancies for therapeutic synergy with anticancer Drugs. Expert Opin Drug Deliv. 2011;8(6):749–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bosland MC, Schlicht MJ, Deng Y, Lü J. Effect of Dietary Methylseleninic Acid and Se-Methylselenocysteine on Carcinogen-Induced, Androgen-promoted prostate carcinogenesis in rats. Nutr Cancer. 2022;74(10):3761–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Qi Y, Fu X, Xiong Z, Zhang H, Hill SM, Rowan BG, Dong Y. Methylseleninic acid enhances paclitaxel efficacy for the treatment of triple-negative Breast cancer. PLoS ONE. 2012;7(2):e31539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Martins IL, Miranda JP, Oliveira NG, Fernandes AS, Gonçalves S, Antunes AM. Synthesis and biological activity of 6-selenocaffeine: potential modulator of chemotherapeutic Drugs in Breast cancer cells. Molecules. 2013;18(5):5251–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zeng H, Combs GF Jr. Selenium as an anticancer nutrient: roles in cell proliferation and Tumor cell invasion. J Nutr Biochem. 2008;19(1):1–7.

    Article  PubMed  Google Scholar 

  22. Yao Y, Wang C, Ma Y, Zhang J, Sun D, Chen L, Huang L, Wu G. Copper-catalyzed decarboxylative alkylselenation of Propiolic acids with Se Powder and Epoxides. Adv Synth Catal. 2021;363(7):1930–4.

    Article  CAS  Google Scholar 

  23. Ramos-Inza S, Encío I, Raza A, Sharma AK, Sanmartín C, Plano D. Design, synthesis and anticancer evaluation of novel Se-NSAID hybrid molecules: identification of a Se-indomethacin analog as a potential therapeutic for Breast cancer. Eur J Med Chem. 2022;244:114839.

    Article  CAS  PubMed  Google Scholar 

  24. Lu X, Yang L, Chen J, Zhou J, Tang X, Zhu Y, Qiu H, Shen J. The action and mechanism of myrislignan on A549 cells in vitro and in vivo. J Nat Med. 2017;71(1):76–85.

    Article  CAS  PubMed  Google Scholar 

  25. Zanka K, Kawaguchi Y, Okada Y, Nagaoka S. Epigallocatechin Gallate induces upregulation of LDL receptor via the 67 kDa laminin receptor-independent pathway in HepG2 cells. Mol Nutr Food Res. 2020;64(7):e1901036.

    Article  PubMed  Google Scholar 

  26. Oehme I, Bösser S, Zörnig M. Agonists of an ecdysone-inducible mammalian expression system inhibit Fas ligand- and TRAIL-induced apoptosis in the human colon carcinoma cell line RKO. Cell Death Differ. 2006;13(2):189–201.

    Article  CAS  PubMed  Google Scholar 

  27. An B, Wang B, Hu J, Xu S, Huang L, Li X, Chan ASC. Synthesis and biological evaluation of selenium-containing 4-Anilinoquinazoline derivatives as Novel Antimitotic agents. J Med Chem. 2018;61(6):2571–88.

    Article  CAS  PubMed  Google Scholar 

  28. Pang Y, An B, Lou L, Zhang J, Yan J, Huang L, Li X, Yin S. Design, synthesis, and Biological evaluation of Novel Selenium-Containing isocombretastatins and Phenstatins as Antitumor agents. J Med Chem. 2017;60(17):7300–14.

    Article  CAS  PubMed  Google Scholar 

  29. Negishi K, Noiri E, Maeda R, Portilla D, Sugaya T, Fujita T. Renal L-type fatty acid-binding protein mediates the bezafibrate reduction of cisplatin-induced acute kidney injury. Kidney Int. 2008;73(12):1374–84.

    Article  CAS  PubMed  Google Scholar 

  30. Hwang DB, Won DH, Shin YS, Kim SY, Kang BC, Lim KM, Che JH, Nam KT, Yun JW. Ccrn4l as a pre-dose marker for prediction of cisplatin-induced hepatotoxicity susceptibility. Free Radic Biol Med. 2020;148:128–39.

    Article  CAS  PubMed  Google Scholar 

  31. Bartolini D, Sancineto L, Fabro de Bem A, Tew KD, Santi C, Radi R, Toquato P, Galli F. Selenocompounds in cancer therapy: an overview. Adv Cancer Res. 2017;136:259–302.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Natural Science Foundation of Zhejiang Province (LTY21H030001, LQ22H170001), the Science and Technology Project of Wenzhou (Y2020086, Y20210215), the Fundamental Research Funds for Wenzhou Medical University (KYYW202009, KYYW20211), and the Students’ scientific and technological innovation activity plan (new talent plan)of Zhejiang province (2023R413076).

Author information

Authors and Affiliations

Authors

Contributions

JS, JZ, and WX: Conception, Methodology. BP, QW, JL, HZ, and RZ: Data curation, Visualization, Investigation. WX and YD: Validation. WX: Writing- Original draft. JS and JZ: Writing- Reviewing and Editing. GZ: Writing - Revision after review. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Guanghui Zhu, Jie Zhou or Jian Sun.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Ethics approval and consent to participate

All animal procedures were approved by the Institutional Animal Care and Use Committee of Wenzhou Medical University. We confirmed that all methods were carried out in accordance with the relevant guidelines and regulations of the Institutional Animal Care and Use Committee of Wenzhou Medical University, China. The study is reported in accordance with ARRIVE guidelines.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

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

Xu, W., Du, Y., Pan, B. et al. Novel phenoxy-((phenylethynyl) selanyl) propan-2-ol derivatives as potential anticancer agents. BMC Chemistry 17, 172 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s13065-023-01076-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13065-023-01076-0

Keywords