1. Introduction
Oxidative stress can cause cell injury and death, which may be related to numerous diseases and conditions, such as liver damage, aging, cancer, stroke, Alzheimer’s disease, and Parkinson’s disease [
1,
2]. The well-known capability of flavonoids to scavenge reactive oxygen species (ROS) is frequently cited as the key property underlying the prevention of and/or reduction in oxidative stress-related chronic diseases and age-related disorders, such as cardiovascular diseases, carcinogenesis, and neurodegeneration. However, many studies have suggested that the therapeutic activity of these compounds involves other properties their ability to directly bind to target peptides [
3], inducing the inhibition of key enzymes, the modulation of cell receptors or transcription factors, as well as the perturbation of protein (or peptide) aggregates, who are known to regulate many cell functions.
Flavonoids have a broad spectrum of biological activities and administering high dosages could trigger side effects. One strategy to improve the potency and selectivity of flavonoids is to take advantage of the dimeric nature of biflavonoids, thereby facilitating simultaneous interactions through the binding of multiple sites of a biological target [
4,
5,
6]. Similar to flavonoid dimers, flavonolignan dimers or simply bi-flavonolignans are also an emerging class of dimeric compounds that unlike bi-flavonoids, which are very widespread in nature, consist of synthetic dimers of few flavonolignans isolated from the milk thistle
Silybum marianum [L. Gaertn. (Asteraceae)] [
7]. In this frame, recently we reported the synthesis of new silibinin dimers in which the two monomer units are linked through a phosphodiester bridge, between two aliphatic OH functions (Phosphate-Linked Silybin dimers,
Figure 1) [
8].
Silibinin is a diastereoisomeric mixture of two flavonolignans, namely, silybin A (SilA) and silybin B (SilB) (
1a and
1b,
Figure 1), in a ratio of approximately 1:1, extracted from milk thistle seeds [
9,
10]. Silibinin has been used as a traditional drug to treat a range of liver disorders, including hepatitis and cirrhosis. The manifold inhibitory effects of silibinin against various cancer cells include growth inhibition, anti-inflammation, cell cycle regulation, apoptosis induction, chemo-sensitization, inhibition of angiogenesis, reversal of multi-drug resistance, and inhibition of invasion and metastasis [
11,
12]. Many in vitro and in vivo reports on the activity of silibinin, clearly neglect the structure-activity relationship of the pair of diastereoisomers, using silibinin, the natural mixture of the two flavonolignans SilA and SilB, for all experiments. These studies have mostly disregarded this aspect because of the difficulty separating, on a preparative scale, two diastereoisomers (
1a and
1b). In the 2021 a very in-depth study by Kren et al. [
13] on the central role of stereochemistry in the pharmacological properties of silybin, highlights how it is necessary to continue studying these flavonolignans, together with the other silymarin flavonolignans, never neglecting their optical purity.
As a part of our continuing research effort on the synthesis of newly modified silibinin [
14,
15,
16], in 2017, starting from silibinin (
1ab), dimers 3-3, 3-9′′′ and 9′′-9′′ phosphodiester were obtained (
Figure 1) [
8]. Dimers, obtained as mixture of diastereoisomers, were very soluble in water and stable in both human serum and alkaline phosphatase. Despite silibinin (
1ab) and silybins (
1a and
1b) [
17,
18] not having strong antioxidant activity, dimers 9′′-9′′ showed a strong radical-scavenging ability. In particular, the ability to scavenge
1O
2 in H
2O was tested, and a higher reactivity towards HO
● (about two times) was estimated for the 3-9′′ and 9′′-9′′ dimers with respect to silibinin. Starting from these results, it seemed interesting to investigate the structure–activity relationships of dimers 9′′-9′′, obtained from diastereoisomerically pure silybin monomers (SilA
1a and SilB
1b,
Figure 1).
Herein, we report an improvement of 9′′-9′′ PLSd dimers synthesis and a systematic study on the ability to scavenge HO● radicals as well as their antiproliferative effect on many human tumor cell lines of different histological origins or metastatic potential. Human dermal fibroblasts (HDFs) were used as healthy cells to evaluate the selectivity of action of the examined metabolites towards tumor cells. Furthermore, apoptosis induction was investigated in leukemia cells treated with the examined compounds.
3. Materials and Methods
3.1. General Methods and Materials
All chemicals were purchased from Sigma–Aldrich (Milano, Italy). HPLC–grade MeCN and MeOH were purchased from Carlo Erba Reagents and Sigma-Aldrich, respectively. Reactions were monitored by TLC (F254 precoated silica gel plates, Merck) and column chromatography (Merck Kieselgel 60, 70–230 mesh, Milano, Italy). HPLC analysis of dimers 6aa, 6bb and 6ab, was performed with a Shimadzu LC–8A PLC system (Shimadzu Analytical and Measuring Instruments, Milano, Italy) equipped with a Shimadzu SCL–10A VP System control and a Shimadzu SPD–10A VP UV–Vis detector. Mass spectrometric analyses were performed on AB SCIEX TOF/TOF 5800 in positive or negative mode and Waters Micromass ZQ Instrument (Waters, Milano, Italy) equipped with an electrospray source in positive mode. The NMR spectra were recorded at 25 °C on an NMR spectrometer Bruker DRX, Bruker Advance (Bruker Italia Srl, Milano, Italy) and INOVA-500 NMR instrument (Varian, Milan, Italy), referenced in ppm to residual solvent signals (CDCl3, at δH 7.27, δC 77.0; CD3OD, at δH 3.31, δC 49.0 and DMSO-d6, δH 2.50, δC 39.5. 31P NMR spectra were recorded using D3PO4 (85 wt. % in D2O, 98 atoms %D) as an external standard, referenced to residual solvent signals (δP 0.0 ppm). Data for 1H NMR are reported as follows: chemical shift (ppm), multiplicity (s = singlet, br = broad, d = doublet, t = triplet, and m = multiplet), coupling constant (Hz), integration, and assignment. The 1H signals were assigned by using 1H/1H COSY, 1H/13C HSQC, and 1H/13C HMBC. NMR data were processed using Bruker Topspin 3.6.1 software. The proton-detected heteronuclear correlations were measured using a gradient heteronuclear single-quantum coherence (HSQC) experiment, optimized for 1JHC = 155 Hz, and a gradient heteronuclear multiple bond coherence (HMBC) experiment, optimized for nJHC = 8 Hz.
Silybin A and silybin B were obtained by HPLC purification of silibinin purchased from Sigma–Aldrich (S0417) as reported by us [
21]. The experimental procedures to the synthesis of building blocks
2 and
4, are described in detail only for the stereoisomers of silybin A: the same reaction conditions (temperature, stoichiometric ratios, time of reaction) were used for silybin B.
3.2. Synthesis of 3,5,7,4′′-O-tetra-isobutyryl-silybin 2a (or 2b)
Silybin A (1a, 730 mg, 1.51 mmol), previously co-evaporated several times with anhydrous THF and dissolved in anhydrous pyridine (4 mL), was reacted with DMTCl (666 mg, 1.96 mmol). The reaction mixture, left at room temperature for 2 h under stirring, was then diluted with MeOH and concentrated under reduced pressure. The crude was then diluted with DCM, transferred into a separatory funnel, washed once with a saturated NaHCO3 aqueous solution, and then once with H2O. The organic phase, dried over anhydrous Na2SO4, was filtered, and then concentrated under reduced pressure. The crude was next purified on a silica gel column, eluting with DCM/MeOH (98:2, v/v) in the presence of 1% of pyridine, affording pure 9′′-O-(4,4′-dimethoxytriphenylmethyl)-silybin A as a pale amorphous solid (1.14 g, 1.44 mmol) in a 96% yield.
In total, 1.14 g (1.44 mmol) of product dissolved in anhydrous DCM (20 mL), adding Et3N (841 μL, 6.05 mmol) and pyridine (1.2 mL, 14.4 mmol), was reacted with isobutyryl chloride (638 μL, 6.05 mmol). The mixture was left under stirring at 0 °C for 15 min and then diluted with MeOH and concentrated under reduced pressure. The crude was then diluted with DCM, transferred into a separatory funnel, washed one time with a saturated NaHCO3 aqueous solution, and then once with H2O. The organic phase, dried over anhydrous Na2SO4, was filtered, and then concentrated under reduced pressure. The crude was next purified on a silica gel column, eluting with n-hexane/EtOAc (7:3, v/v) in the presence of 1% of pyridine furnishing pure 3,5,7,4′′-O-tetra-isobutyryl-9′′-O-(4,4′-dimethoxytriphenylmethyl)-silybin A as pale amorphous solid (1.06 g, 1.0 mmol) in a 70% yield.
In a solution, 0.1 M of product (1.06 g, 1.0 mmol) in MeOH/DCM (6:1 v/v) was added 1% (p/v) of I2 (100 mg). The solution was left under stirring at room temperature for 1 h and then added Na2S2O3 and concentrated under reduced pressure. The crude was then diluted with DCM, transferred into a separatory funnel, washed one time with a saturated NaHCO3 aqueous solution, and then once with H2O. The organic phase, dried over anhydrous Na2SO4, was filtered, and then concentrated under reduced pressure. The crude was next purified on a silica gel column, eluting with n-hexane/EtOAc (6:4, v/v), leading pure 3,5,7,4′′-O-tetra-isobutyryl-silybin A (2a) as pale amorphous solid (686 mg, 0.9 mmol) in a 90% yield.
2a 690 mg (60% starting from 1a). Rf = 0.5 (n-hexane/EtOAc 6:4 v/v silica gel). 1H-NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz): 7.13–6.96 (complex signals, 6H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.74 (d, J = 2.2, 1H, H-6); 6.55 (d, J = 2.2, 1H, H-8); 5.66 (d, J = 12.2, 1H, H-3); 5.35 (d, J = 12.2, 1H, H-2); 5.02 (d, J = 8.0, 1H, H-7′′); 4.02–3.97 (m, 1H, H8′′); 3.87–3.82 (overlapped signals, 4H, OCH3 and H-9′′a); 3.57 (dd, J = 12.5, 3.6, 1H, H-9′′b); 2.97–2.71 (m, 3H, CH of isobutyryl (ibu) groups in 5, 7 and 4′′); 2.59–2.49 (m, 1H, CH of ibu in 3); 1.37–1.22 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.12–0.97 (m, 6H, CH3 of ibu group in 3). 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm): 185.0; 175.3; 175.0 (2C); 174.1; 162.4; 156.5; 151.7; 151.6; 144.1; 143.6; 140.5; 134.5; 128.6; 123.0; 120.8; 119.8; 117.0, 116.5, 111.1 (2C); 110.7; 108.7; 81.1; 78.3; 76.0; 72.9; 61.4; 56.1; 34.2; 34.0; 33.9; 33.6; 19.0; 18.8; 18.7; 18.5. MS (MALDI-TOF, positive ions): m/z calculated for C41H46O14 = 762.289; found: 763.996 [M + H]+, 785.229 [M + Na]+, 801.652 [M + K]+.
2b 644 mg (55% starting from 1b). Rf = 0.5 (n-hexane/EtOAc 6:4 v/v silica gel). 1H NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz): 7.11–6.96 (complex signals, 6H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.75 (d, J = 2.2, 1H, H-6); 6.55 (d, J = 2.2, 1H, H-8); 5.67 (d, J = 12.1, 1H, H-3); 5.36 (d, J = 12.2, 1H, H-2); 5.04 (d, J = 8.0, 1H, H-7′′); 4.02–3.97 (m, 1H, H8′′); 3.86–3.80 (overlapped signals, 4H, OCH3 and H-9′′a); 3.56 (dd, J = 12.4, 3.6, 1H, H-9′′b); 2.96–2.73 (m, 3H, CH of ibu groups in 5, 7 and 4′′); 2.60–2.50 (m, 1H, CH of ibu group in 3); 1.35–1.26 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.02–0.92 (m, 6H, CH3 of ibu group in 3). 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm): 185.0; 175.1; 175.0 (2C); 174.1; 162.4; 156.5; 151.7; 151.6; 144.1; 143.6; 140.6; 134.5; 128.5; 123.1; 120.9; 119.7; 117.1, 116.4, 111.3; 111.1; 110.8; 108.7; 81.0; 78.3; 75.9; 72.8; 61.4; 56.0; 34.2; 34.0; 33.9; 33.6; 19.0; 18.8; 18.7; 18.5. MS (MALDI-TOF, positive ions): m/z calculated for C41H46O14 = 762.289; found: 763.425 [M + H]+, 785.784 [M + Na]+, 801.358 [M + K]+.
3.3. Synthesis of Phosphoramidite Silybin 4a (or 4b)
To 3,5,7,4′′-tetra-O-iBu-silybin A (2a, 0.26 mmol; 200 mg) dissolved in anhydrous DCM (4.5 mL), DIEA (181 μL, 1.05 mmol), and 2-cyanoethyl-N,N-diisopropylamino-chlorophosphoramidite 3 (73 μL, 0.31 mmol) were mixed. After 20 min the solution was concentrated and silica gel chromatography of the residue (eluent n-hexane/EtOAc 6:4, v/v, with 3% v/v of Et3N), afforded desired compound 4a in 86% yield.
4a 219 mg (0.22 mmol, 86%). Rf = 0.8 (n-hexane/EtOAc 6:4, v/v silica gel). 1H NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz, mixture of diastereoisomers): 7.11–6.94 (complex signals, 6H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.74 (d, J = 2.2, 1H, H-6); 6.55 (d, J = 2.2, 1H, H-8); 5.66 (d, J = 12.2, 1H, H-3); 5.65 (d, J = 12.2, 1H, H-3); 5.35 (d, J = 12.2, 1H, H-2); 5.34 (d, J = 12.2, 1H, H-2); 5.02 (d, J = 7.8, 1H, H-7′′); 4.14–4.06 (m, 1H, H-8′′); 3.97–3.49 (m, 9H, OCH3, 2H-9′′, OCH2CH2CN, N[CH(CH3)2]2; 2.97–2.48 (m, 6H, CH of ibu groups, OCH2CH2CN); 1.36–1.26 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.20–1.07 (m, 15H, CH3 of ibu groups in 3, N[CH(CH3)2]2). 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm, mixture of diastereoisomers): 185.0 (2C); 175.2 (2C); 175.0 (2C); 174.1; 162.5; 156.5; 151.7; 151.5 (2C); 144.2; 143.5; 143.4; 140.5(2C); 134.6; 128.3; 128.2; 123.0; 122.9; 120.9; 120.8; 119.9; 119.8; 117.6; 117.5; 117.1; 116.9; 116.5; 116.4; 111.4; 111.2; 111.1; 110.7, 108.7; 81.2; 81.1; 76.2; 75.8; 72.9 (2C); 62.5; 62.2; 62.0; 58.7; 58.6; 58.5; 58.3; 56.0 (2C); 43.3; 43.2; 34.2; 34.0; 33.9; 33.6; 24.6; 24.5; 20.4; 20.3 (3C); 19.0; 18.8; 18.7; 18.5. 31P NMR (CDCl3, 161.98 MHz, 25 °C, δ ppm): 150.3; 149.9. MS (ESI-TOF, positive ions): m/z calculated for C50H63N2O15P = 962.397; found: 964.229 [M + H]+, 986.033 [M + Na]+, 1002.553 [M + K]+.
4b 202 mg (0.21 mmol, 80%). Rf = 0.8 (n-hexane/EtOAc 6:4, v/v, silica gel). 1H NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz, mixture of diastereoisomers): 7.11–6.94 (complex signals, 6H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.75 (d, J = 2.2, 1H, H-6); 6.55 (d, J = 2.2, 1H, H-8); 5.67 (d, J = 12.1, 1H, H-3); 5.35 (d, J = 12.1, 1H, H-2); 5.34 (d, J = 12.1, 1H, H-2); 5.02 (d, J = 7.8, 1H, H-7′′); 4.14–4.05 (m, 1H, H-8′′); 3.97–3.50 (m, 9H, OCH3, 2H-9′′, OCH2CH2CN, N[CH(CH3)2]2; 2.96–2.49 (m, 6H, CH of ibu groups, OCH2CH2CN); 1.36–1.26 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.20–1.07 (m, 15H, CH3 of ibu groups in 3, N[CH(CH3)2]2) ppm. 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm, mixture of diastereoisomers): 185.0; 175.1 (2C); 175.0; 174.9; 174.1; 162.4; 156.5; 151.7; 144.3; 144.2; 143.5 (2C); 140.5 (2C); 134.7; 128.2; 128.1; 123.0; 122.9; 121.0; 119.9; 119.8; 117.6; 117.5; 117.1; 116.5; 116.3; 111.1; 110.8, 108.7; 81.1; 81.0; 76.1; 75.8; 72.8; 62.7; 62.5; 62.2; 62.1; 58.7; 58.5 (2C); 58.3; 56.0 (2C); 43.3; 43.2; 34.2; 34.0; 33.9; 33.6; 24.6 (2C); 24.5 (2C); 20.3 (3C); 19.0; 18.8; 18.7 (3C); 18.5. 31P NMR (CDCl3, 161.98 MHz, 25 °C, δ ppm): 150.2; 149.8. MS (ESI-TOF, positive ions): m/z calculated for C50H63N2O15P = 962.397; found: 964.215 [M + H]+, 986,212 [M + Na]+, 1002.322 [M+K]+.
3.4. General Procedure for the Synthesis of Phosphotriester dimers 5aa, 5bb and 5ab
In total, 219 mg (0.22 mmol) of phosphoramidites 4a and the building block 2a 155 mg (0.20 mmol) previously dried and kept under reduced pressure, were reacted with a 0.25 M 4,5-dicyanoimidazole solution in anhydrous MeCN (1.5 mL, 0.37 mmol). To obtain the 5bb dimer, the phosphoramidite 4b and the derivative 2b were coupled under the same conditions as previously reported. To obtain the 5ab dimer, the best yields were obtained by coupling the phosphoramidite 4a and the derivative 2b. The reaction was left under stirring at r.t. and monitored by TLC with an eluent system n-hexane/EtOAc (6:4, v/v). After 30 min, the reaction was over, and then a 5.5 M tert-Butyl hydroperoxide (TBHP) solution in decane (150 μL) was added and left stirring at r.t. After 30 min the reaction mixture was, concentrated under reduced pressure, and purified by flash chromatography, eluting with n-hexane/EtOAc (7:3, v/v), to afford pure 5 (5aa, 5bb and 5ab) yellow-brown amorphous powder in 83%, 80% and 77% yields, respectively.
5aa 272 mg (0.17 mmol, 83%). Rf = 0.5 (n-hexane/EtOAc 1:1, v/v, silica gel). 1H-NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz): 7.11–6.91 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.76 (s, 2H, H-6); 6.55 (s, 2H, H-8); 5.66 (d, J = 12.1, 2H, H-3); 5.36 (d, J = 12.1, 2H, H-2); 4.93 (t, J = 8.0, 2H, H-7′′); 4.32–3.95 (m, 8H, H8′′, 2H9′′, OCH2CH2CN); 3.86–3.80 (overlapped signals, 6H, OCH3); 2.95–2.49 (m, 10H, CH of ibu groups, OCH2CH2CN); 1.39–1.25 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.14–1.08 (m, 6H, CH3 of ibu groups in 3); 1.01–0.96 (m, 6H, CH3 of ibu groups in 3). 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm): 184.9; 175.1; 175.0; 174.1; 162.4; 156.5; 151.7; 143.6; 143.4; 143.3; 140.8; 133.7; 128.9; 123.3; 123.2; 121.3; 121.1; 119.8 (2C); 117.2; 117.1; 116.6; 116.5; 116.4; 111.5, 111.4; 111.1; 110.7, 108.7; 80.9; 76.0; 75.9; 75.7; 72.8; 72.7; 66.5; 62.3; 56.0; 34.2; 34.0; 33.9; 33.6; 19.0; 18.8; 18.7 (C2); 18.5. 31P NMR (161 MHz, CDCl3, 25 °C, δ ppm): -2.4. MS (MALDI-TOF, positive ions): m/z calculated for C85H94NO30P = 1639.560; found: 1641.838 [M+H]+, 1663.652 [M + Na]+, 1679.154 [M + K]+.
5bb 262 mg (0.16 mmol, 80%). Rf = 0.5 (n-hexane/EtOAc 1:1, v/v, silica gel). 1H NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz): 7.11–6.91 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.76 (s, 2H, H-6); 6.55 (s, 2H, H-8); 5.66 (d, J = 12.1, 2H, H-3); 5.36 (d, J = 12.1, 2H, H-2); 4.93 (t, J = 8.0, 2H, H-7′′); 4.32–3.95 (m, 8H, H8′′, 2H9′′, OCH2CH2CN); 3.86–3.80 (overlapped signals, 6H, OCH3); 2.95–2.49 (m, 10H, CH of ibu groups, OCH2CH2CN); 1.36–1.24 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.13–1.09 (m, 6H, CH3 of ibu groups in 3); 1.02–0.98 (m, 6H, CH3 of ibu groups in 3) ppm. 13C-NMR (125 MHz, CDCl3, 25 °C, δ ppm, J Hz): 184.9; 175.1; 175.0 (2C); 174.1; 162.4; 156.5; 151.7 (2C); 143.6; 143.4; 143.3; 140.8; 133.7; 128.9; 123.3 (2C); 121.2; 121.1; 119.8 (2C); 117.2; 117.1; 116.6 (2C); 116.4; 111.5, 111.4; 111.1; 110.7, 108.7; 80.9; 75.9; 75.7; 72.8; 66.5 (2C); 62.3; 62.2; 56.0; 34.2; 34.0; 33.9; 33.6; 19.0; 18.8; 18.7; 18.5. 31P NMR (161 MHz, CDCl3, 25 °C, δ ppm): -2.4. MS (MALDI-TOF, positive ions): m/z calculated for C85H94NO30P = 1639.560; found: 1641.554 [M + H]+, 1663,159 [M + Na]+, 1679.555 [M + K]+.
5ab 262 mg (0.15 mmol, 77%). Rf = 0.5 (n-hexane/EtOAc 1:1, v/v, silica gel). 1H NMR (400 MHz, CDCl3, 25 °C, δ ppm, J Hz): 7.12–6.88 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′); 6.76–6.73 (m, 2H, H-6); 6.56–6.54 (m, 2H, H-8); 5.69–5.61 (m, 2H, H-3); 5.38–5.32 (m, 2H, H-2); 4.99–5.90 (m, 2H, H-7′′); 4.35–3.97 (m, 8H, H8′′, 2H9′′, OCH2CH2CN); 3.88–3.79 (overlapped signals, 6H, OCH3); 2.96–2.48 (m, 10H, CH of ibu groups, OCH2CH2CN); 1.36–1.24 (m, 18H, CH3 of ibu groups in 5, 7 and 4′′); 1.13–1.08 (m, 6H, CH3 of ibu groups in 3); 1.02–0.96 (m, 6H, CH3 of ibu groups in 3) ppm. 13C NMR (100 MHz, CDCl3, 25 °C, δ ppm): 184.9; 175.1; 175.0 (2C); 174.1; 162.4; 156.5; 151.7 (2C); 143.6; 143.4; 143.3; 140.8; 133.7; 128.9; 123.3 (2C); 121.2; 121.1; 119.8 (2C); 117.2; 117.1; 116.6 (2C); 116.4; 111.5, 111.4; 111.1; 110.7, 108.7; 80.9; 76.1; 76.0; 75.9; 75.7; 72.8; 66.5 (2C); 62.3; 62.2; 56.0; 34.2; 34.0; 33.9; 33.6; 19.0; 18.8; 18.7 (2C); 18.5. 31P NMR (161 MHz, CDCl3, 25 °C, δ ppm): -2.3; -2.6. MS (MALDI-TOF, positive ions): m/z calculated for C85H94NO30P = 1639.560; found: 1641.497 [M + H]+, 1663.132 [M + Na]+, 1679.115 [M + K]+.
3.5. General Procedure for the Synthesis of Dimers 6aa, 6bb and 6ab
In total, 200 mg (0.12 mmol) of dimer
5aa were treated with 7mL of a mixture conc. aq NH
3/MeOH (1:1,
v/
v) for 5 h at 50 °C, leading to full removal of the
ibu and 2-cyanoethyl (CE) groups. The mixture was dried under reduced pressure and suspended in a buffer solution and then purified on RP-HPLC carried out on Phenomenex Kromasil
® C18 column (10 μm particle size, 10.0 mm × 250 mm i.d.) using a linear gradient of MeCN in in 0.1 M Ammonium Acetate in H
2O (pH 7.0) (pH 7.0) from 5% to 95% over 20 min at a flow rate of 6 mL/min with detection at 288, 260 nm. Compound thus obtained was converted into the corresponding sodium salts by cation exchange on a DOWEX (Na
+ form) resin to obtain homogeneous samples
6aa in 77% yield. RP-HPLC analysis was carried out on Luna C18 (2) (5 μm particle size, 150 mm × 4.6 mm i.d.) using a linear gradient of MeCN in in 0.1 M ammonium acetate in H
2O (pH 7.0) from 5% to 95% over 20 min at a flow rate of 0.8 mL/min with detection at 288 nm. The purity of the
6aa product was 99.6% (see
SM).
6aa 97 mg (0.09 mmol, 77%). t
R = 13.3 min (99.6% purity, see
Figure S1 in SM).
1H NMR (400 MHz, DMSO-d6 + 5% D
2O, 25 °C, δ ppm,
J Hz); 7.11–6.74 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′) 5.85 (s, 4H, H-6 and H-8); 5.05 (d,
J = 11.0, 2H, H-2); 4.82 (d,
J = 7.6, 2H, H-7′′); 4.57 (d,
J = 11.0, 1H, H-3); 4.35–4.27 (m, 2H, H8′′); 3.80–3.65 (overlapped signals, 8H, OCH
3, H-9′′a); 3.58– 3.47 (m, 2H, H-9′′b) ppm.
13C NMR (100 MHz, DMSO-d6 + 5% D
2O, 25 °C, δ ppm): 197.8; 168.2; 163.7; 162.8; 148.0; 147.5; 143.8; 143.6; 130.5; 127.6; 121.8; 120.8; 116.8; 116.7; 115.7; 112.1, 100.6; 96.7; 95.7; 82.9; 77.1; 76.2; 71.8; 63.6; 56.0.
31P NMR (161 MHz, DMSO-d6 + 5% D
2O, 25 °C, δ ppm): -1.6. MS (MALDI-TOF, negative ions):
m/
z calculated for C
50H
43O
22P = 1026.198; found: 1025.355 [M − H]
-.
6bb 103 mg (0.10 mmol, 82%). t
R = 13.4 min (98.7% purity, see
Figure S2 in SM).
1H NMR (400 MHz, DMSO-d6, 25 °C, δ ppm,
J Hz): 11.90 (s, 2H, OH-5); 11.08 (s, 2H, OH-7); 9.19 (s, 2H, OH-4′′); 7.10–6.73 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′) 5.91 (s, 4H, H-6 and H-8); 5.85–5.79 (m, 2H, OH-3); 5.05 (d,
J = 11.1, 2H, H-2); 4.83 (d,
J = 7.8, 2H, H-7′′); 4.60 (d,
J = 11.1, 1H, H-3); 4.58 (d,
J = 11.1, 1H, H-3); 4.34–4.26 (m, 2H, H8′′); 3.79–3.65 (overlapped signals, 8H, OCH
3, H-9′′a); 3.58– 3.48 (m, 2H, H-9′′b) ppm.
13C NMR (100 MHz, DMSO-d6, 25 °C, δ ppm,
J Hz): 198.2; 167.4; 163.7; 162.9; 148.0; 147.5; 143.8; 143.5; 130.5; 127.6; 121.6; 120.8; 117.0; 116.8; 115.7; 112.1, 100.8; 96.5; 95.5; 83.0; 77.1; 76.2; 71.9; 63.6; 56.0.
31P NMR (161 MHz, DMSO-d6, 25 °C, δ ppm): -1.7. MS (MALDI-TOF, negative ions):
m/
z calculated for C
50H
43O
22P = 1026.198; found: 1025.196 [M − H]
−.
6ab 100 mg (0.10 mmol, 80%). t
R = 13.5 min (99.2% purity, see
Figure S3 in SM).
1H NMR (400 MHz, MeOD-d4, 25 °C, δ ppm,
J Hz): 7.15–6.79 (complex signals, 12H, H-2′, H-5′, H-6′, H-2′′, H-5′′, H-6′′) 5.92 (s, 4H, H-6 and H-8); 5.02–4.94 (m, 4H, H-2 and H7′′); 4.53 (d, 11.5, 1H, H-3); 4.26–4.20 (m, 2H, H8′′); 3.91–3.80 (overlapped signals, 8H, OCH
3, H-9′′a); 3.79– 3.75 (m, 2H, H-9′′b) ppm.
13C NMR (100 MHz, MeOD-d4, 25 °C, δ ppm): 196.9; 167.3; 163.9; 162.9; 147.7; 146.9; 143.9; 143.8; 143.6; 130.0; 127.8; 120.6; 120.3; 116.5; 116.3; 116.1; 114.9; 110.9, 100.4; 96.9; 94.9; 83.3 (2C); 77.0; 76.1; 76.0; 72.2; 64.0; 55.1.
31P NMR (161 MHz, MeOD-
d4, 25 °C, δ ppm,
J Hz): -0.2. MS (MALDI-TOF, negative ions):
m/
z calculated for C
50H
43O
22P = 1026.198; found: 1025.144 [M − H]
−.
3.6. Hydroxyl Radical (∙OH) Generation and Reactivity Estimation
The reactivity constant between new silybin dimers and hydroxyl radical has been determined using Laser flash Photolysis system. The spectroscopic equipment and method have been described elsewhere, and only an introduction is given below [
8].
The formation of di-thiocyanate radical anion (SCN
2●−) through reactivity of photogenerated hydroxyl radicals (HO
●) with thiocyanate (SCN
- in the presence of hydrogen peroxide. The second-order rate constants between HO
● and dimers were determined following reactions (R1–R4 in the
Section 2.2) and using the following equation:
where Abs
0 and Abs are the absorption of
at 475 nm in absence and presence of dimers;
and
are the second-order rate constants of HO
● with thiocyanate and dimers at different concentrations. The plot of
vs. the concentration of dimers (
6aa,
6bb or
6ab) can be fitted with a liner correlation and slope used to determine the value of
. The results presented here were the mean of three replicates. Means were compared by one way analysis of variance (ANOVA) and significant differences were assessed by post hoc tests of least significant. Differences with a
p value of < 0.05 were considered significance.
3.7. Culture Conditions
Human T lymphoblastoid (Jurkat) and human metastatic melanoma (WM266) cell lines were grown in RPMI medium supplemented with heat inactivated 10% fetal bovine serum (FBS), 2.5 mM glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin (Euroclone). Human cervix adenocarcinoma cell line (HeLa), human lung carcinoma (A549), human pancreatic cancer (PANC), human glioblastoma (U87) normal human fibroblasts (HDF) were grown in DMEM supplemented with 10% fetal bovine serum (FBS), 1% glutamine, 100 U/mL penicillin and 100 μg/mL streptomycin (Euroclone, Milano, Italy). Cells were maintained in humidified air containing 5% CO
2, at 37 °C [
30].
3.8. Antiproliferative Activity
Cells were plated at density of 10,000 cells/well for Jurkat, 2000/well for WM266 and HDF, 1200 cells/well for HeLa, and 1000 for PANC, U87 and A375 in 96-well microplates (Thermofisher, Waltham, MA, USA). After 24 h incubation, cells were treated with increasing concentrations of synthetized compounds previously solubilized in DMSO at 50 mM concentration. Cell proliferation was determined by using (2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt (CCK-8, Sigma Aldrich) for Jurkat cells [
31], and the 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide assay (MTT, Sigma Aldrich, Sigma Aldrich, St. Louis, MO, USA)) for HeLa, WM266, PANC, U87 and HDF cells [
32], after 48 h treatment. Plates were then analyzed by using a microplate reader (Enspire, Perkin Elmer Italia Spa, Milano, Italy) at 450 (CCK-8) or 570 nm (MTT).
The results are presented as the percentage of proliferating cells respect to the control (vehicle treated cells) and are expressed as means ± SE of, at least, three independent experiments performed in triplicate. The statistical analysis was performed using Student’s t-test, unpaired, two-sided, p < 0.05 was considered significant. The IC50 values were calculated by GraphPad Prism software.
3.9. Apoptosis Assay
The apoptosis analysis was performed on Jurkat cells seeded at 2.5 × 10
5 cells/mL in a 6-well plate. The cells were incubated in the absence or presence of 200 μM concentration of examined compounds at 37 °C and apoptosis induction was analysed after 48 h by double staining with annexin V/FITC and propidium iodide (PI) (eBioscience, Affimetrix Santa Clara, CA, USA) [
33]. The cells undergoing apoptosis were quantified using a flow cytometer equipped with a 488 nm argon laser (Becton Dickinson, Franklin Lakes, NJ, USA) by Cell Quest software. All FACS analyses were performed at least 2 times.