Next Article in Journal
Recovery of Residual Lead from Automotive Battery Recycling Slag Using Deep Eutectic Solvents
Previous Article in Journal
A Continuous-Wave EPR Investigation into the Photochemical Transformations of the Chromium(I) Carbonyl Complex [Cr(CO)4bis(diphenylphosphino)]+ and Reactivity with 1-hexene
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis, and Anti-Hepatocellular Carcinoma Evaluation of Sesquiterpene Lactone Epimers Trilobolide-6-O-isobutyrate Analogs

1
Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
2
Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
3
Public Research Center, Hainan Medical University, Haikou 571199, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2024, 29(2), 393; https://doi.org/10.3390/molecules29020393
Submission received: 14 December 2023 / Revised: 4 January 2024 / Accepted: 5 January 2024 / Published: 12 January 2024
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Hepatocellular carcinoma (HCC), one of the most common malignant cancers with a low 5-year survival rate, is the third leading cause of cancer-related deaths worldwide. The finding of novel agents and strategies for the treatment of HCC is an urgent need. Sesquiterpene lactones (SLs) have attracted extensive attention because of their potent antitumor activity. In this study, a new series of SL derivatives (318) were synthesized using epimers 1 and 2 as parent molecules, isolated from Sphagneticola trilobata, and evaluated for their anti-HCC activity. Furthermore, the structures of 4, 6, and 14 were confirmed by X-ray single-crystal diffraction analyses. The cytotoxic activities of 318 on two HCC cell lines, including HepG2 and Huh7, were evaluated using the CCK-8 assay. Among them, compound 10 exhibited the best activity against the HepG2 and Huh7 cell lines. Further studies showed that 10 induced cell apoptosis, arrested the cell cycle at the S phase, and induced the inhibition of cell proliferation and migration in HepG2 and Huh7. In addition, absorption, distribution, metabolism, and excretion (ADME) properties prediction showed that 10 may possess the properties to be a drug candidate. Thus, 10 may be a promising lead compound for the treatment of HCC.

Graphical Abstract

1. Introduction

Cancer is a persistent unmet global challenge claiming millions of lives yearly and taking a tremendous toll on healthcare services and world economies [1]. According to the 2020 WHO report, liver cancer has the sixth highest incidence rate (4.7%) and the third highest mortality rate (8.3%) among the 36 types of cancer, making it one of the deadliest forms of cancer [2]. The most common primary liver cancer, hepatocellular carcinoma (HCC), is one of the most prevalent and lethal tumors [3]. As our understanding of HCC biology forges ahead, a number of first- and second-line chemotherapeutic drugs, as well as new targeted agents, immune checkpoint inhibitors (ICIs), and their combination therapies, are among the options available to patients [4]. Despite progress in this field, the effectiveness of most of them is severely limited by drug resistance, as well as invasion and metastasis of cancers [5,6]. The development of new effective anti-HCC drugs is therefore essential.
Natural sesquiterpene lactones (SLs) are a family of secondary metabolites derived almost exclusively from Asteraceae/Compositae plants [7]. These natural products have been a hot topic in the field of tumor drug research because of their complexity, diversity, novelty of structures, and uniqueness of function [8]. In recent years, SL-derived drugs such as derivatives of Artemisinin (ART) and Parthenolide (PTL) (Figure 1), have shown promise for the potential treatment of various types of cancer [9,10,11,12,13,14]. ART and its derivatives such as dihydroartemisinin (DHA) and artesunate could promote cancer cell apoptosis, induce cell cycle arrest and autophagy, and inhibit cancer cell invasion as well as migration, and they represent promising candidates for cancer therapy, supported by some clinical phase I/II trials [15,16,17,18]. PTL showed promising anti-cancer properties. Nevertheless, PTL has some disadvantages, such as poor oral bioavailability and poor water solubility [19,20]. DMAPT, a derivative of PTL, effectively increased the water solubility and oral bioavailability, and has advanced into a phase I clinical trial for the treatment of acute myeloid leukemia [21]. And the other PTL derivative, ACT001, has been clinically tested in Australia and China for the treatment of glioblastoma [22].
SLs’ cytotoxic activity has been described as mainly dependent on the α-methylene-γ-lactone group, which is prone to react with suitable nucleophiles [23,24,25]. Trilobolide-6-O-isobutyrate A and B (1 and 2), a pair of C-5 epimers of SLs with high oxygen content, were isolated from the flowers of S. trilobata, they also have an α-methylene-γ-lactone group. Subsequent investigations indicated that 2 could strongly inhibit the proliferation of HCC through inhibition of the IL-6/STAT3 signaling pathway [26]. And a new series of SL derivatives were designed and synthesized using 1 and 2 as the parent molecules. Unfortunately, all of the compounds did not show significant in vitro antiproliferative activity against the tested human cancer cell lines [27].
Based on our ongoing pursuit of the investigation of 1 and 2 to further enhance their antitumor profile, a series of 1 and 2 derivatives (318) were further designed and synthesized in this work. All the compounds were evaluated for their potential inhibition of HCC cell lines. We further investigated the effects of the most active compound 10 on HepG2 and Huh7 cells, and computational absorption, distribution, metabolism, and excretion (ADME) estimation studies were performed.

2. Results and Discussion

2.1. Synthesis

Compounds 1 and 2 were isolated from the EtOH solutions of the dried flowers of S. trilobata by repeated column chromatography (CC) and these two compounds were further purified by recrystallization from EtOH following previously described methods [28]. In order to investigate whether the presence of ester groups and terminal double bonds in 1 and 2 has a positive effect on anti-hepatocellular carcinoma activities, we designed synthetic routes primarily considering the hydrolysis reaction. The acetyl groups at the C-1 and C-9 positions and the isobutyryl group at the C-6 position were removed, and two acetyl groups and one isobutyryl substituent on 1 and 2 were removed by selecting different acid and base systems to obtain hydrolysates with different degrees of ester removal. We used NaBH4 as a reductant to reduce the exocyclic double bond of C-11 (13) to the methyl group. Scheme 1 and Scheme 2 represent the synthetic pathways of all the compounds, respectively.
First, 1 was dissolved in THF and hydrolyzed by HCl solution. The crude product was further purified by silica gel CC to obtain pure products 3 and 4 [29]. Then, 1 was dissolved in THF and H2O, the hydrolysis condition was changed to alkaline KOH solution, and the reaction was at 0 °C for 2 h to prepare the pure product 5. Subsequently, the appropriate 1 and NaBH4 solids were dissolved in the CH3CN and stirred at 60 °C for 26.5 h to afford pure products 6 and 7 by HPLC [30]. In order to obtain different degrees of removal of the ester groups at the C-1, C-6, and C-9 positions and expose the hydroxyl groups, we switched to using KOH to provide alkaline conditions and MeOH as the solvent. The reaction mixture was stirred at room temperature for 6 h. Then, crude products were further purified by silica gel CC to afford pure products 8 and 9. In the hydrolysis of 2, we also optimized different acid-base systems under the conditions of HCl or H2SO4 solution, and prepared the products 10 and 12. The hydrolysis derivatives 1316 of 2 were also prepared according to the synthesis method of compounds 69. Finally, 2 was dissolved in DMSO and reacted in the presence of DBU to obtain products 17 and 18 [31].
The structures of these derivatives were firmly established by combination with the physical and chemical properties, and 1D/2D NMR and HRMS methods. The Michael addition at the α-methylene-γ-lactone of SLs with amines has been found to be highly stereo-specific and yielded exclusively a single stereoisomer with the (R*)- configuration at the newly formed C-11 chiral carbon. This was attributed to the stereo-exclusive protonation of the enolate formed during the Michael addition [32]. In this research, firstly, the absolute configuration of the parent molecules 1 and 2 was determined by X-ray crystal analysis [26]. Secondly, in our previous work, the 11R stereochemistry of the SLs derivatives was confirmed by the NOE effect between H-11 and H-6 [27]. Finally, the X-ray crystal structures of 4, 6, and 14 (Figure S55) further confirmed the R configuration at C-11.

2.2. Biological Evaluation

2.2.1. Cell Viability Assay and Structure–Activity Relationships (SARs)

The anti-HCC activity of 1, 2, and their derivatives was evaluated by the CCK-8 assay against human HCC cell lines HepG2 and Huh7, and compared with adriamycin as the working standard. As shown in Table 1, some derivatives showed comparatively significant IC50 values compared to 1 and 2. On the basis of the above activity data, a preliminary SARs was deduced. In the hydrolysates of 1 and 2, the cytotoxicity of compounds 3 and 10 was comparable or better than that of the parent molecules. It was suggested that the double bonds at the C-4, 14 or C-3, 4 positions were more favorable to enhancing the activity of HCC. Notably, the double bond between C-11 and C-13 was reduced to the methyl group in all the compounds, which were less potent than the parental compounds, further confirming SLs’ cytotoxic activity as being mainly dependent on the α-methylene-γ-lactone group [23,24,25]. Compound 10 demonstrated the best antiproliferative activity against HepG2 and Huh7 cells with IC50 values of 9.73 and 18.86 µM, respectively. Therefore, 10 was selected for subsequent antitumor experiments on HepG2 and Huh7 cell lines in vitro.

2.2.2. Anti-Proliferative Effect of Compound 10 on Huh7 and HepG2 Cells

The effects of different concentrations of compound 10 on two types of human HCC cell lines (Huh7 and HepG2) were detected by CCK-8 assay. The results indicated that 10 could dose-dependently decrease the viability of the two HCC cell lines (Figure 2A,B). In addition, 10 could also inhibit the viability of Huh7 cells in time- and dose-dependent manners (Figure 2C,D). The live/dead cell staining assay (Calcein-AM/PI double staining kit) was also employed to evaluate the in vitro cytotoxicity of 10. In the control, a bright green fluorescence (calcein AM for live cells probe) and a few red dots with fluorescence (PI for dead cells probe) were found in Huh7 and HepG2 cells (Figure 3). A decreased number of live cells for both Huh7 and HepG2 was obviously found in 10, whereas that of dead cells increased, compared with the control. Thus, the fluorescence imaging results were in agreement with those of the cytotoxicity assay.
The colony-forming assay is one of the effective methods to measure cell proliferation. When a single cell lasts for more than six generations in vitro, the descendants of the cell population are called clones. At this time, each clone contains more than 50 cells, and the size is between 0.3 and 1.0 mm. By counting the clone formation rate, the proliferation potential of a single cell can be quantitatively analyzed, and the proliferation ability and independent survival ability of the cell can be understood. Consequently, this work performed a clone-forming assay to investigate whether compound 10 inhibited proliferation. Huh7 and HepG2 cells were first incubated using 10 for a 24 h period, respectively, and followed by 14 d culture to observe a visible colony. According to Figure 4, Huh7 and HepG2 cells had significantly suppressed clone-forming capacity after 10 exposure, which was consistent with the CCK-8 assays.

2.2.3. Compound 10 Promoted Cell Apoptosis of Huh7 and HepG2 Cells

In order to investigate the proapoptotic effect of compound 10, Huh7 and HepG2 cells were stained with Annexin V-FITC and PI probe, and subjected to flow cytometry analysis. Both cells were incubated with different concentrations of 10 for 24 h. In comparison to the control group, all treatment groups of 10 significantly induced apoptosis of Huh7 and HepG2 cells (Figure 5). The percentages of total apoptotic cells increased significantly from 6.3% (control, Huh7 cells) and 6.51% (control, HepG2 cells) to 67.8% (40 μM, Huh7 cells) and 24% (10 μM, HepG2 cells), respectively. Compound 10 induced approximately 30% late apoptosis of Huh7 cells at concentrations of 40 μM, and late apoptosis was more obvious. The results established that compound 10 could significantly induce Huh7 and HepG2 cell apoptosis.

2.2.4. Compound 10 Arrested the Cell Cycle in the S Phase in Huh7 and HepG2 Cells

The cell cycle plays an important role in the operation and development of cell life due to its regulation of the division and duplication of DNA. Therefore, blocking the cell cycle is considered an effective way to eliminate tumor cells. To demonstrate the antiproliferative effect of compound 10 on the cell cycle process, flow cytometry was used to monitor PI staining treatment with 10 for 24 h; this led to an increased percentage of cells in the S phase from 39.76% and 33.22% of the untreated control to 46.27% and 36.91%, respectively (Figure 6A,B and Figure 6C,D). This result demonstrated that 10 arrested Huh7 and HepG2 cell cycle progression in the S phase.

2.2.5. Effect of Compound 10 on Cell Migration

Migration is one of the essential links in the process of tumor cell metastasis. For tumor cells to separate from the mother tumor, cross the blood vessel wall, and invade the surrounding normal tissue, they need some exercise ability. Tumor cells that are highly metastatic usually have high motility. The cell scratch method and transwell migration are simple methods to determine the migration, movement, and repair ability of cells. Thus, the effects of 10 on cell migration were further investigated. As shown in Figure 7A,B, 10 significantly inhibited the rate of wound healing in Huh7 cells after treatment at the indicated concentrations for 48 h. The number of cells that went through the membrane in transwell assays also decreased in a dose-dependent manner (Figure 7C,D). The above results suggested that 10 could effectively suppress the migration of Huh7 cells in a dose-dependent manner.

2.2.6. ADME Prediction

Many drug molecule candidates remain in phase studies without a drug molecule due to poor ADME properties. Performing theoretical ADME calculations for designed and newly synthesized compounds may aid the progression of these compounds in advanced in vitro and in vivo studies [33,34]. Therefore, some properties of the designed compounds, such as physicochemical, lipophilicity, water-solubility, pharmacokinetics, drug-likeness, and medicinal chemistry properties, were calculated using SwissADME online tools, and some of the most active properties of 10 are given in Table 2. The drug-likeness assessment of 10 was performed by predicting Lipinski’s rule of five, which includes molecular weights (MW < 500), lipophilicity (log Po/w < 5), number of hydrogen bond acceptors (HBA ≤ 10), and number of hydrogen bond donors (HBD ≤ 5) to determine the “drug-likeness” of 10. Its solubility in water is fine and favorable. Its gastrointestinal absorption was calculated as high and not able to pass through the blood–brain barrier. The findings of ADME showed that 10 has very good drug-like properties.

3. Materials and Methods

3.1. General Experimental Procedures

The 1D and 2D NMR experiments were performed on a Bruker AV-400 (Bruker Corporation, Zurich, Switzerland) instrument with tetramethylsilane as the internal standard. HR-ESI-MS spectra were acquired on a Bruker Daltonics Apex-Ultra 7.0 T (Bruker Corporation, Billerica, MA, USA) and a Q-TOF Ultima Global GAA076 LC mass spectrometer. Single-crystal data were measured with an Agilent Gemini Ultra X-ray single-crystal diffractometer (Cu Kα radiation). Preparative HPLC was conducted using an Agilent 1260 prep-HPLC system with a Waters C18 analytical HPLC column (4.6 × 250 mm, 5 μm) and a semipreparative column (9.4 × 250 mm, 7 μm). Sephadex LH-20 (Pharmacia Co. Ltd., Sandwich, UK) and silica gel (200–300 and 300–400 mesh, Qingdao Marine Chemical Inc., Qingdao, China) were used for column chromatography (CC). All solvents were purchased from Xilong Chemical Reagent Factory (Guangzhou, China). The flowers of Sphagneticola trilobata were collected from Haikou County, Hainan Province, China, in August 2018, and identified by Professor Qiong-Xin Zhong, School of Life Science, Hainan Normal University.

3.2. The Separation of SLs 1 and 2

The isolation process of 1 and 2 is shown in the supporting information.

3.3. General Procedure for the Synthesis of 1 Hydrolysis Derivatives 34

In a 25 mL round-bottomed flask, 712 mg (1.157 mmol) of 1, 2 mL of THF, and 2 mL of dilute HCl (2 mol/L) were added sequentially. The reaction was heated under reflux at 45 °C for 26 h. After the reaction was complete (CHCl3:acetone = 6:1), the reaction was detected by TLC. The pH was adjusted to 6–7 with potassium carbonate solution, and products 3 and 4 were separated by CC (CHCl3:acetone = 8:1).
(3aS,4R,4aS,8S,8aS,9R,9aS)-4-(isobutyryloxy)-8a-methyl-3,5-dimethylene-2-oxododecahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 3). Yield: 4 mg (6%), purity 93%. C23H30O8, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.18 (1H, d, J = 3.6 Hz, H-13), 5.49 (1H, d, J = 3.2 Hz, H-13), 5.42 (1H, dd, J = 8.0, 11.6 Hz, H-6), 5.30 (1H, d, J = 2.4 Hz, H-9), 5.23 (1H, dd, J = 4.4, 12.0 Hz, H-1), 4.96 (1H, t, J = 2.0 Hz, H-14), 4.85 (1H, dd, J = 2.4, 9.6 Hz, H-8), 4.76 (1H, t, J = 2.0 Hz, H-14), 3.43 (1H, m, H-7), 2.55 (1H, m, H-2″), 2.38 and 2.20 (each 1H, m, H-3), 2.16 (1H, d, J = 11.9 Hz, H-5), 1.95 (3H, s, H-2‴), 1.93 (3H, s, H-2′), 1.87 and 1.44 (each 1H, m, H-2), 1.21 (3H, s, H-15), 1.16 (6H, d, J = 7.0 Hz, H-3″, 4″); 13C NMR (100 MHz, CDCl3) δ: 175.8 (C-1″), 170.5 (C-1′), 169.3 (C-12), 169.1 (C-1‴), 141.1 (C-4), 136.8 (C-11), 120.2 (C-13), 117.4 (C-14), 73.4 (C-8), 73.0 (C-9), 70.6 (C-6), 70.4 (C-1), 55.5 (C-5), 44.6 (C-7), 40.7 (C-10), 34.1 (C-2″), 30.1 (C-3), 29.7 (C-15), 26.6 (C-2), 21.1 (C-2′), 20.5 (C-2‴), 19.5 (C-3″), 19.1 (C-4″). HRESIMS m/z 457.1822 [M+Na]+ (calcd. for C23H30O8Na, 457.1823).
(3aS,4R,4aS,8S,8aS,9R,9aS)-4-hydroxy-5,8a-dimethyl-3-methylene-2-oxo-2,3,3a,4,4a,7,8,8a,9,9a-decahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 4). Yield: 9 mg (16%), purity 97%. C19H24O7, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.20 (1H, d, J = 2.4 Hz, H-13), 6.00 (1H, d, J = 2.4 Hz, H-13), 5.51 (1H, m, H-3), 5.28 (1H, d, J = 2.8 Hz, H-9), 5.08 (1H, dd, J = 6.0, 9.6 Hz, H-1), 4.85 (1H, dd, J = 2.8, 9.6 Hz, H-8), 4.19 (1H, m, H-6), 3.25 (1H, m, H-7), 2.48, 1.97 (2H, m, H-2), 2.11 (1H, d, J = 4.4 Hz, OH), 1.95 (3H, s, H-2′), 1.91 (3H, s, H-14), 1.86 (3H, s, H-2″), 1.83 (1H, s, H-5), 1.13 (3H, s, H-15); 13C NMR (100 MHz, CDCl3) δ: 171.1 (C-1′), 170.1 (C-12), 169.1 (C-1″), 137.5 (C-11), 131.1 (C-4), 122.4 (C-3), 121.1 (C-13), 75.1 (C-6), 73.9 (C-8), 73.7 (C-9), 68.1 (C-1), 54.1 (C-5), 47.2 (C-7), 39.0 (C-10), 28.0 (C-2), 25.7 (C-14), 21.3 (C-2′), 20.6 (C-2″), 18.8 (C-15). HRESIMS m/z 387.1407 [M+Na]+ (calcd. for C19H24O7+Na, 387.1414).

3.4. General Procedure for the Synthesis of 1 Hydrolysis Derivative 5

Add 125.0 mg (0.277 mmol) 1 to a 50 mL round bottom flask and dissolve in 6 mL THF and 10 mL H2O. Add 4 mL KOH solution (0.5 mol/L) slowly at 0 °C, stirring for 2 h. After the reaction was completely detected by TLC (Petroleum ether:acetone = 1.5:1), the mixture was adjusted to pH 2–3 with 10% dilute HCl, and product 5 was purified by CC (petroleum ether:acetone = 3:1).
(3aS,4S,4aS,5S,8S,8aS,9R,9aS)-5,8,9-trihydroxy-5,8a-dimethyl-3-methylene-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 5). Yield: 73.0mg (71%), purity 92%. C19H28O7, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.25 (1H, d, J = 3.2 Hz, H-13), 5.68 (1H, d, J = 2.8 Hz, H-13), 5.65 (1H, t, J = 4.8 Hz, H-6), 4.90 (1H, dd, J = 4.0, 10.0 Hz, H-8), 4.03 (1H, dd, J = 2.8, 6.8 Hz, H-1), 3.96 (1H, d, J = 3.6 Hz, H-9), 3.27 (1H, m, H-7), 2.52 (1H, m, H-2′), 1.97 and 1.62 (each 1H, m, H-2), 1.80 (1H, d, J = 4.4 Hz, H-5), 1.77 and 1.52 (each 1H, m, H-3), 1.17 (3H, s, H-15), 1.16 (3H, s, H-14), 1.15 (6H, d, J = 7.0 Hz, H-3′, 4′); 13C NMR (100 MHz, CDCl3) δ: 176.4 (C-1′), 171.0 (C-12), 135.4 (C-11), 123.2 (C-13), 75.7 (C-8), 71.9 (C-6), 70.0 (C-10), 69.7 (2C, C-1, 9), 51.5 (C-5), 42.6 (C-7), 42.5 (C-10), 34.5 (C-3), 34.4 (C-2′), 29.4 (C-2), 25.7 (C-14), 20.7 (C-15), 18.9 (C-3′), 18.8 (C-4′). HRESIMS m/z 391.1723 [M+Na]+ (calcd. for C19H28O7+Na, 391.1727).

3.5. General Procedure for the Synthesis of 1 Hydrolysis Derivatives 69

An amount of 297 mg (0.657 mmol) of compound 1 and 88 mg of NaBH4 were dissolved in 50 mL of CH3CN and stirred at 60 °C for 26.5 h. After the reaction was complete (CHCl3: acetone = 6: 1) as detected by TLC, the pH of the mixture was adjusted to 6–7 with 10% dilute HCl, extracted with EtOAc for three times, dried with anhydrous sodium sulfate, concentrated under reduced pressure, and prepared by HPLC to obtain compounds 6 and 7. Subsequently, 23.3 mg (0.051 mmol) of 6 and 22.6 mg KOH were dissolved in 5 mL of MeOH, stirred at room temperature for 6 h, and the reaction was detected by TLC (CHCl3:acetone = 3:1). The products 8 and 9 were purified by CC (CHCl3:acetone = 6:1).
(3R,3aS,4S,4aS,5S,8S,8aS,9R,9aS)-5-hydroxy-4-(isobutyryloxy)-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 6). Yield: 86 mg (29%), purity 98%. C23H34O9, white amorphous powder. 1H-NMR (400 MHz, Methanol-d4) δ: 5.83 (1H, dd, J = 9.2, 12.0 Hz, H-6), 5.52 (1H, dd, J = 8.8, 8.8 Hz, H-1), 5.16 (1H, d, J = 2.4 Hz, H-9), 4.99 (1H, dd, J = 2.4, 10.0 Hz, H-8), 2.79 (1H, m, H-11), 2.57 (1H, m, 2″), 2.51 (1H, m, H-7), 2.23, 1.42 (2H, m, H-2), 1.98 (3H, s, H-2′),1.92 (3H, s, H-2‴), 1.96, 1.67 (2H, m, H-3), 175 (1H, d, J = 11.6 Hz, H-5), 1.38 (3H, s, H-15), 1.31 (3H, s, H-14), 1.20 (6H, d, J = 6.8 Hz, H-3″, 4″), 1.16 (3H, d, J = 7.6 Hz, H-13); 13C-NMR (100 MHz, Methanol-d4) δ: 181.5 (C-12), 178.1 (C-1″), 172.4 (C-1′), 170.9 (C-1‴), 75.9 (C-8), 75.2 (C-9), 74.3 (C-6), 70.7 (C-4), 70.2 (C-1), 54 (C-5), 49.3 (C-7), 41.5 (C-10), 41.2 (C-11), 36 (C-3), 35.6 (C-2″), 32.9 (C-15), 23.2 (C-2), 22 (C-14), 21.3 (C-2′), 21 (C-2‴), 19.7 (C-3″), 18.8 (C-4″), 17.9 (C-13). HRESIMS m/z 477.2097 [M+Na]+ (calcd. for C23H34O9+Na, 477.2095).
(3R,3aS,4S,4aS,5S,8S,8aR,9R,9aS)-8-acetoxy-5,9-dihydroxy-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 7). Yield: 80 mg (30%), purity 97%. C21H32O8, white amorphous powder. 1H NMR (400 MHz, DMSO-d6) δ: 5.61 (1H, dd, J = 8.8, 6.8 Hz, H-6), 5.26 (1H, dd, J = 7.2, 14.4 Hz, H-1), 4.79 (1H, dd, J = 2.8, 10.0 Hz, H-8), 3.53 (1H, d, J = 2.8 Hz, H-9), 2.75 (1H, m, H-11), 2.47 (1H, m, H-2″), 2.29 (1H, m, H-7), 2.18, 1.31 (2H, m, H-2), 1.95 (3H, s, H-2′), 1.79, 1.51 (2H, m, H-3), 1.55 (1H, d, J = 8.8 Hz, H-5), 1.24 (3H, s, H-15), 1.11 (6H, d, J = 7.2 Hz, H-3″, 4″), 1.08 (3H, s, H-14), 1.07 (3H, s, H-13); 13C NMR (100 MHz, DMSO-d6) δ: 179.6 (C-12), 174.9 (C-1″), 169.6 (C-1′), 75 (C-8), 71.7 (C-6), 71.6 (C-9), 70.8 (C-1), 68.6 (C-4), 51.9 (C-5), 47.3 (C-7), 40.1 (C-10), 39.1 (C-11), 34.5 (C-3), 33.5 (C-2″), 31.5 (C-15), 22.7 (C-2), 21.3 (C-14), 21.1 (C-2′), 19 (C-3″), 18.3 (C-4″), 16.5 (C-13). HRESIMS m/z 435.1987 [M+Na]+ (calcd. for C21H32O8+Na, 435.1989).
(3R,3aS,4S,4aS,5S,8S,8aS,9R,9aS)-4,5-dihydroxy-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 8). Yield: 3.5 mg (18%), purity 96%. C19H28O8, white amorphous powder. 1H-NMR (400 MHz, Methanol-d4) δ: 5.48 (1H, dd, J = 8.4, 9.2 Hz, H-1), 5.13(1H, d, J = 2.8 Hz, H-9), 4.94 (1H, dd, J = 2.8, 10.4 Hz, H-8), 4.34 (1H, dd, J = 9.6, 11.6 Hz, H-6), 2.61 (1H, m, H-11), 2.37 (1H, m, H-7), 2.22, 1.33 (2H, m, H-2), 1.93 (3H, s, H-2′), 1.90 (3H, s, H-2″), 1.71,1.30 (2H, m, H-3), 1.45 (3H, s, H-14), 1.44 (1H, d, J = 11.2 Hz, H-5), 1.36 (3H, d, J = 7.6 Hz, H-13), 1.27 (3H, s, H-15); 13C NMR (100 MHz, Methanol-d4) δ: 182.2 (C-12), 172.5 (C-1″), 171 (C-1′), 76.2 (C-8), 75.4 (C-9), 73.5 (C-1), 72.1 (C-6), 70.4 (C-4), 56.4 (C-5), 50.4 (C-7), 42.8 (C-10), 41.5 (C-11), 35.6 (C-2), 33.5 (C-15), 23.3 (C-2), 22.2 (C-14), 21.3 (C-2′), 21 (C-2″), 18.2 (C-13). HRESIMS m/z 407.1669 [M+Na]+ (calcd. for C19H28O8+Na, 407.1676).
(3R,3aS,4S,4aS,5S,8S,8aS,9R,9aS)-5,8,9-trihydroxy-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 9). Yield: 6.0 mg (31%), purity 98%. C19H30O7, white amorphous powder. 1H NMR (400 MHz, Methanol-d4) δ: 5.62 (1H, dd, J = 2.8, 3.2 Hz, H-6), 4.89 (1H, dd, J = 2.8, 10.0 Hz, H-8), 3.84 (1H, dd, J = 4.0, 5.2 Hz, H-1), 3.70 (1H, d, J = 3.6 Hz, H-9), 3.03 (1H, m, H-11), 2.54 (1H, m, H-2′), 2.38 (1H, m, H-7), 2.27, 1.63 (2H, m, H-2), 1.92, 1.50 (2H, m, H-3), 1.79 (1H, d, J = 3.2 Hz, H-5), 1.28 (3H, s, H-14), 1.24 (3H, s, H-13), 1.16 (3H, s, H-15), 1.15 (6H, d, J = 6.8 Hz, H-3′, 4′); 13C NMR (100 MHz, Methanol-d4) δ: 182.1 (C-12), 177.1 (C-1′), 76.7 (C-8), 72.9 (C-9), 72.6 (C-4), 71.9 (C-6), 70.3 (C-1), 52.0 (C-5), 48.6 (C-7), 43.3 (C-10), 39.2 (C-11), 35.4 (C-2′), 35.1 (C-3), 30.0 (C-15), 27.8 (C-2), 22.3 (C-14), 19.2 (C-3′), 19.1 (C-4′), 16.1 (C-13). HRESIMS m/z 393.1875 [M+Na]+ (calcd. for C19H30O7+Na, 393.1883).

3.6. General Procedure for the Synthesis of 2 Hydrolysis Derivatives 1012

Referring to the synthesis method of 3 and 4, compounds 10 and 11 were obtained by CC purification (CHCl3:acetone = 3:1). For further optimization of acid conditions, we replaced the previous acid with a dilute H2SO4 solution (8 mol/L) and refluxed the mixture at 45 °C for 10 h. After the reaction was completed (CHCl3:acetone = 6:1), the products 10, 11, and 12 were separated by CC (CHCl3:acetone = 8:1).
(3aS,4R,4aR,8S,8aS,9R,9aS)-9-acetoxy-8-hydroxy-5,8a-dimethyl-3-methylene-2-oxo-2,3,3a,4,4a,7,8,8a,9,9a-decahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 10). Yield: 11.2 mg (6%), purity 93%. C21H28O7, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.32 (1H, d, J = 4 Hz, H-13), 5.77 (1H, d, J = 3.2 Hz, H-13), 5.53 (1H, dd, J = 1.6, 4.0 Hz, H-6), 5.41 (1H, br s, H-3), 5.31 (1H, d, J = 4.4 Hz, H-9), 5.03 (1H, dd, J = 4.4, 8.0 Hz, H-8), 3.43 (1H, dd, J = 6.0, 10.4 Hz, H-1), 3.31 (1H, m, H-7), 2.56 (1H, m, H-2′), 2.48 (1H, H-5), 2.24, 2.07 (2H, m, H-2), 2.03 (3H, s, H-2″), 1.60 (3H, d, J = 1.2 Hz, H-14), 1.17 (6H, d, J = 7.2 Hz, H-3′, 4′), 1.11 (3H, s, H-15); 13C NMR (100 MHz, CDCl3) δ: 176.6 (C-1′), 172.3 (C-1″), 169.6 (C-12), 134.9 (C-11), 130.2 (C-4), 122.4 (C-3), 119.8 (C-13), 72.8 (C-8), 72.2 (C-9), 69.7 (C-6), 68.4 (C-1), 43.5 (C-7), 41.4 (C-10), 37.9 (C-5), 34.4 (C-2′), 29.7 (C-2), 20.7 (C-2″), 20.4 (C-14), 19.0 (C-3′), 18.8 (C-4′), 11.0 (C-15). HRESIMS m/z 415.1725 [M+Na]+ (calcd. for C21H28O7+Na, 415.1727).
(3aS,4S,4aR,5S,8S,8aS,9R,9aS)-9-acetoxy-5,8-dihydroxy-5,8a-dimethyl-3-methylene-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 11). Yield: 82.2 mg (48%), purity 95%. C21H30O8, white amorphous powder. 1H NMR (400 MHz, Methanol-d4) δ: 6.18 (1H, d, J = 4.0 Hz, H-13), 5.90 (1H, dd, J = 1.6, 3.2 Hz, H-6), 5.72 (1H, d, J = 4.0 Hz, H-13), 5.34 (1H, d, J = 4.4 Hz, H-9), 5.08 (1H, dd, J = 4.4, 8.4 Hz, H-8), 3.39 (1H, dd, J = 6.4, 8.4 Hz, H-1), 3.31 (1H, m, H-7), 2.61 (1H, m, H-2′), 1.94 (3H, s, H-2″), 1.86 (1H, d, J = 3.2 Hz, H-5), 1.68, 1.50 (2H, m, H-3), 1.64 (2H, m, H-2), 1.34 (3H, s, H-14), 1.30 (3H, s, H-15), 1.21 (6H, d, J = 7.0 Hz, H-3′, 4′); 13C NMR (100 MHz, Methanol-d4) δ: 177.5 (C-1′), 171.8 (C-12), 170.9 (C-1″), 136.8 (C-11), 119.0 (C-13), 74.5 (C-8), 73.6 (C-9), 72.2 (C-4), 72.1 (C-1), 70.0 (C-6), 45.8 (C-7), 44.5 (C-5), 43.7 (C-3), 43.1 (C-10), 35.7 (C-2′), 28.6 (C-14), 26.3 (C-2), 20.5 (C-2″), 19.3 (C-3′), 18.8 (C-4′), 13.8 (C-15). HRESIMS m/z 428.2272 [M+NH4]+ (calcd. for C21H30O8+NH4, 428.2278).
(3aS,4S,4aR,5S,8S,8aS,9R,9aS)-5,8,9-trihydroxy-5,8a-dimethyl-3-methylene-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 12). Yield: 63 mg (43%), purity 94%. C19H28O7, white amorphous powder. 1H NMR (400 MHz, Methanol-d4) δ: 6.56 (1H, d, J = 1.2 Hz, H-13), 5.82 (1H, d, J = 1.2 Hz, H-13), 5.41 (1H, dd, J = 2.4, 3.6 Hz, H-6), 4.51 (1H, d, J = 2.4 Hz, H-9), 4.25 (1H, dd, J = 2.4, 4 Hz, H-8), 3.79 (1H, dd, J = 6.4, 9.6 Hz, H-1), 3.16 (1H, m, H-7), 2.62 (1H, m, H-2′), 1.72, 1.60 (2H, m, H-2), 1.67, 1.44 (2H, m, H-2), 1.35 (1H, d, J = 2.8 Hz, H-5), 1.29 (3H, s, H-14), 1.26 (3H, s, H-15), 1.22 (6H, d, J = 7.2 Hz, H-3′, 4′); 13C NMR (100 MHz, Methanol-d4) δ: 177.3 (C-1′), 166.5 (C-12), 135.2 (C-11), 132.8 (C-13), 86.1 (C-9), 76.1 (C-6), 71.8 (C-1), 71.6 (C-4), 62.2 (C-8), 47.2 (C-7), 45.3 (C-5), 44.6 (C-10), 42.8 (C-3), 35.7 (C-2′), 28.8 (C-2), 25 (C-14), 19.5 (C-3′), 19 (C-4′), 15.2 (C-15). HRESIMS m/z 386.2166 [M+NH4]+ (calcd. for C19H28O7+NH4, 386.2173).

3.7. General Procedure for the Synthesis of 2 Hydrolysis Derivatives 1316

According to the synthesis of 6 and 7, compounds 13 and 14 were prepared by HPLC. According to the synthesis of 8 and 9, compounds 15 and 16 were purified by CC (CHCl3:acetone = 3:1).
(3R,3aS,4S,4aR,5S,8S,8aS,9R,9aS)-5-hydroxy-4-(isobutyryloxy)-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 13). Yield: 131.6 mg (43%), purity 95%. C23H34O9, white amorphous powder. 1H NMR (400 MHz, DMSO-d6) δ: 5.50 (1H, dd, J = 1.2, 2.8 Hz, H-6), 5.05 (1H, d, J = 4.4 Hz, H-9), 4.94 (1H, dd, J = 4.4, 7.6 Hz, H-8), 4.55 (1H, dd, J = 6.4, 9.2 Hz, H-1), 2.56, 1.58 (2H, m, H-2), 2.53 (1H, m, H-2″), 2.54 (1H, m, H-11), 2.34 (1H, m, H-7), 1.93 (1H, d, J = 2.8 Hz, H-5), 1.92 (3H, s, H-2′), 1.90 (3H, s, H-2‴), 1.61, 1.50 (2H, m, H-3), 1.31 (3H, s, H-15), 1.19 (3H, s, H-14), 1.16 (3H, d, J = 6.8 Hz, H-13), 1.11 (6H, d, J = 7.2 Hz, H-3″, 4″); 13C NMR (100 MHz, DMSO-d6) δ: 178.4 (C-12), 174.9 (C-1″), 169.8 (C-1′), 169.7 (C-1‴), 73.0 (C-1), 71.5 (C-8), 70.8 (C-9), 69.6 (C-4), 67.6 (C-6), 47.2 (C-7), 42.3 (C-5), 41.6 (C-3), 41.0 (C-10), 35.7 (C-2″), 33.7 (C-11), 25.5 (C-14), 23.9 (C-2), 20.8 (C-2′), 20.2 (C-2‴), 18.6 (C-3″), 18.2 (C-4″), 14.2 (C-13), 13.9 (C-15). HRESIMS m/z 477.2099 [M+Na]+ (calcd. for C23H34O9+Na, 477.2095).
(3R,3aS,4S,4aR,5S,8S,8aS,9R,9aS)-5,8,9-trihydroxy-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 14). Yield: 3 mg (1%), purity 98%. C19H30O7, white amorphous powder. 1H NMR (400 MHz, Methanol-d4) δ: 5.57 (1H, dd, J = 1.2, 2.8 Hz, H-6), 4.69 (1H, dd, J = 4.0, 7.6 Hz, H-8), 4.02 (1H, dd, J = 4.8, 9.6 Hz, H-1), 3.85 (1H, d, J = 3.6 Hz, H-9), 2.56 (1H, m, H-2′), 2.47 (1H, m, H-7), 2.28 (1H, m, H-11), 2.02 (1H, d, J = 2.8 Hz, H-5), 1.70, 1.54 (2H, m, H-3), 1.68, 1.64 (2H, m, H-2), 1.31 (3H, s, H-15), 1.23 (3H, d, J = 6.4 Hz, H-13), 1.20 (3H, s, H-14), 1.20 (6H, d, J = 6.8 Hz, H-3′, 4′); 13C NMR (100 MHz, Methanol-d4) δ: 182.7 (C-12), 177.5 (C-1′), 76.2 (C-8), 72.3 (C-1), 71.8 (C-9), 71.7 (C-4), 70.4 (C-6), 50.2 (C-5), 44.5 (C-10), 43.5 (C-3), 42.7 (C-7), 38.8 (C-11), 35.7 (C-2′), 28.6 (C-2), 26.2 (C-14), 18.4 (C-3′), 18.8 (C-4′), 15 (C-13), 13.9 (C-15). HRESIMS m/z 369.1920 [M-H]- (calcd. for C19H29O7, 369.1907).
(3R,3aS,4S,4aR,5S,8S,8aS,9R,9aS)-4,5-dihydroxy-3,5,8a-trimethyl-2-oxododecahydronaphtho[2,3-b]furan-8,9-diyl diacetate (compound 15). Yield: 17.2 mg (41%), purity 95%. C19H28O8, white amorphous powder. 1H NMR (400 MHz, DMSO-d6) δ: 5.19 (1H, d, J = 4.4 Hz, H-9), 4.97 (1H, dd, J = 4.4, 8.8 Hz, H-8), 4.67 (1H, dd, J = 5.6, 10.0 Hz, H-1), 4.57 (1H, dd, J = 1.6, 2.8 Hz, H-6), 2.54 (1H, m, H-11), 2.48 (1H, m, H-7), 1.98 (3H, s, H-2′), 1.97 (3H, s, H-2″), 1.79, 1.60 (1H, m, H-3), 1.75, 1.74 (1H, m, H-2), 1.69 (1H, d, J = 2.8 Hz, H-5), 1.58 (3H, s, H-15), 1.44 (3H, s, H-14), 1.25 (3H, d, J = 6.4 Hz, H-13); 13C NMR (100MHz, DMSO-d6) δ: 179.2 (C-12), 169.9 (C-1″), 168.7 (C-1′), 73.4 (C-1), 72.1 (C-8), 71.6 (C-9), 70.4 (C-4), 64.1 (C-6), 51.9 (C-7), 48.6 (C-5), 43.1 (C-10), 41.7 (C-3), 40.9(C-11), 35.8(C-14), 25.6 (C-2), 20.8 (C-2″), 20.3 (C-2′), 14.9 (C-15), 14.3 (C-13). HRESIMS m/z 402.2115 [M+NH4]+ (calcd. for C19H28O8+NH4, 402.2122).
(3R,3aS,4S,4aR,5S,8S,8aS,9R,9aS)-4,5,8,9-tetrahydroxy-3,5,8a-trimethyldecahydronaphtho[2,3-b]furan-2(3H)-one (compound 16). Yield: 3.6 mg (11%), purity 94%. C15H24O6, white amorphous powder. 1H NMR (400 MHz, Methanol-d4) δ: 4.75 (1H, dd, J = 8.0, 4.0 Hz, H-8), 4.46 (1H, dd, J = 2.8, 1.6 Hz, H-6), 3.98 (1H, dd, J = 9.6, 7.2 Hz, H-1), 3.81 (1H, d, J = 4 Hz, H-9), 2.68 (1H, m, H-11), 2.35 (1H, m, H-7), 1.70 (1H, d, J = 2.8 Hz), 1.65, 1.52 (4H, m, H-2, 3), 1.52 (3H, s, H-15), 1.19 (3H, s, H-14), 1.17 (3H, d, J = 7.2 Hz, H-13); 13C NMR (100 MHz, Methanol-d4) δ: 183.7 (C-12), 76.9 (C-1), 73.4 (C-8), 72.5 (C-9), 72.1 (C-4), 66.8 (C-6), 54.8 (C-7), 44.4 (C-5), 43.5 (C-10), 38.8 (C-3), 28.8 (C-2), 26.1 (C-14), 15.3 (C-15), 14.1 (C-13). HRESIMS m/z 301.1645 [M+H]+ (calcd. for C15H24O6+H, 301.1646).

3.8. General Procedure for the Synthesis of 2 Hydrolysis Derivatives 1718

The appropriate amounts of 2 (0.177 mmol) and DBU (100 ul) were dissolved in DMSO (2 mL) for the reaction. The reaction mixture was stirred at room temperature for 79 h. The pH of the mixture was adjusted to 6–7 with 10% dilute hydrochloric acid and the products 17 and 18 were purified by HPLC.
(4R,4aR,5S,8S,8aS)-8-acetoxy-5-hydroxy-3,5,8a-trimethyl-2-oxo-2,4,4a,5,6,7,8,8a-octahydronaphtho[2,3-b]furan-4-yl isobutyrate (compound 17). Yield: 15 mg (21%), purity 95%. C21H28O7, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.64 (1H, d, J = 2.8 Hz, H-6), 5.72 (1H, s, H-9), 4.61 (1H, m, H-1), 2.56 (1H, m, H-2″), 2.13 (3H, s, H-2′), 2.00, 1.66 (2H, m, H-2), 1.95 (3H, s, H-13), 1.86 (1H, d, J = 2.8 Hz, H-5), 1.80, 1.64 (2H, m, H-3), 1.42 (3H, s, H-15), 1.35 (3H, s, H-14), 1.17 (6H, d, J = 6.8 Hz, H-3″, 4″); 13C NMR (100 MHz, CDCl3) δ: 176.7 (C-1″), 170.7 (C-12), 170.6 (C-1′), 147.5 (C-8), 144.0 (C-7), 125.1 (C-11), 115.4 (C-9), 77.3 (C-1), 70.6 (C-4), 61.8 (C-6), 53.6 (C-5), 41.3 (C-3), 40.8 (C-10), 34.6 (C-2′′), 24.9 (C-14), 24.8 (C-2), 21.3 (C-2′), 19.1 (C-3″), 18.7 (C-4″), 18.0 (C-15), 9.4 (C-13). HRESIMS m/z 393.1902 [M+H]+ (calcd. for C21H29O7, 393.1907).
(4R,4aR,5S,8S,8aS)-4,5-dihydroxy-3,5,8a-trimethyl-2-oxo-2,4,4a,5,6,7,8,8a-octahydronaphtho[2,3-b]furan-8-yl acetate (compound 18). Yield: 3.1 mg (6%), purity 92%. C17H22O6, white amorphous powder. 1H NMR (400 MHz, CDCl3) δ: 6.71 (1H, d, J = 2.9 Hz, H-6), 6.12 (1H, s, -OH), 5.73 (1H, s, H-9), 5.68 (1H, t, J = 1.5 Hz, H-1), 4.64 (1H, s), 2.13 (3H, s, H-13), 2.00 (3H, s, H-1′), 2.03, 1.82 (4H, m, H-2, 3), 1.90 (1H, d, J = 2.9 Hz, H-5), 1.45 (3H, s, H-15), 1.35 (3H, s, H-14); 13C NMR (100 MHz, CDCl3) δ: 170.4 (C-1′), 166.9 (C-12), 143.5 (C-8), 127.4 (C-7), 125.6 (C-11), 115.0 (C-9), 77.2 (C-1), 70.6 (C-4), 62.0 (C-6), 53.6 (C-5), 41.3 (C-3), 40.6 (C-10), 38.9, 24.7 (C-14), 21.2(C-2′), 18.5 (C-2), 18.0 (C-15), 9.4 (C-13). HRESIMS m/z 321.1344 [M-H] (calcd. for C17H21O6, 321.1333).

3.9. X-ray Crystallographic Analysis

X-ray data were collected on an Agilent Technologies Gemini A Ultra system diffractometer with Cu-Kα radiation (λ = 1.5418 Å). The structure was solved by direct methods (SHELXS-97) and refined using full-matrix least-squares difference Fourier techniques. Crystallographic data (excluding structure factors) for 4, 6, and 14 have been deposited with the Cambridge Crystallographic Data Centre: CCDC reference numbers 2304356, 2304357, and 2304358, respectively. These data can be obtained free of charge from the Cambridge Crystallographic Data Centre via http://www.ccdc.cam.ac.uk/data_request/cif (accessed on 30 October 2023).
X-ray crystal data for 4: C19H24O7, fw = 728.76, 293 (2) K, a = 9.8333(2) Å, b = 16.4144(2) Å, c = 12.0115(3) Å, α = 90°, β = 103.202(2)°, γ = 90°, V = 1887.51(7) Å3, space group P21, Z = 2, μ (MoKα) = 0.816 mm−1, F(000) = 776.0, ρcalc = 1.282 g/cm3; 22,829 reflections measured, 6707 were unique (Rint = 0.0399); 2Θ range for data collection: 7.56 to 133.94°. Flack 0.08(12). The final R1 = 0.0352, wR2 = 0.0785 (I Page: 16 ≥ 2σ (I)).
X-ray crystal data for 6: C23H34O9, fw = 454.50, 293 (2) K, orthorhombic, a = 10.5090 (2) Å, b = 12.2547 (2) Å, c = 18.1235 (3) Å, α = 90°, β = 90°, γ = 90°, V = 2,334.03 (7) Å3, space group P212121, Z = 4, μ (MoKα) = 0.827 mm−1, F(000) = 976.0, ρcalc = 1.293 g/cm3; 14,042 reflections measured, 4127 were unique (Rint = 0.0421); 2Θ range for data collection: 8.72 to 133.8°. Flack −0.15(18). The final R1 = 0.0358, wR2 = 0.1048 (I Page: 16 ≥ 2σ (I)).
X-ray crystal data for 14: C19H30O7, fw = 370.20, 293 (2) K, monoclinic, a = 8.6601 (6) Å, b = 13.3921 (8) Å, c = 9.3091 (7) Å, α = 90°, β = 112.750 (9)°, γ = 90°, V = 995.65 (13) Å3, space group P21, Z = 2, μ (MoKα) = 0.756 mm−1, F(000) = 378.0, ρcalc = 1.179 g/cm3; 9881 reflections measured, 3473 were unique (Rint = 0.0674); 2Θ range for data collection: 10.304 to 135.168°. Flack −0.2(3). The final R1 = 0.0921, wR2 = 0.2422 (I Page: 16 ≥ 2σ (I)).

3.10. Biological Evaluation

3.10.1. Cell Lines and Cell Culture

Huh7 and HepG2 cells were obtained from the Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences. Both cancer cells were grown in MEM medium, containing 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin, and incubated at 37 °C in a humidified atmosphere containing 5% CO2.

3.10.2. Cell Cytotoxicity Assay

Compound-induced cytotoxicity was analyzed in Huh7 and HepG2 cells using CCK-8 assays. Generally, the cells were cultured in MEM supplemented with 10% FBS in 5% CO2 at 37 °C. Cells were seeded in 96-well plates at a density of 8000 cells/well. The media were replaced on the next day. Afterward, the cultures were incubated with compound 10 at 37 °C for 24 h, a CCK-8 assay was performed to examine cell viability, and the absorbance was measured at 570 nm.

3.10.3. Calcein-AM/PI Costaining

Huh7 and HepG2 cells were seeded in 6-well cell culture plates. The cells were then exposed to various doses of 10 for 24 h. The cells were rinsed three times with PBS, trypsinized, and centrifuged, and the defined amount of them was resuspended in 100 µL 1× assay buffer, in which live and dead cells were then costained by a mixture of Calcein-AM (2 mM) and PI (1.5 mM) solution for 20 min. Afterward, fluorescence microscopic images of cells were taken using an inverted fluorescent microscope (OLYMPUS IX-51, OLYMPUS, Tokyo, Japan).

3.10.4. Colony Formation Assay

Huh7 and HepG2 cells were seeded in 6-well cell culture plates (1000 cells per well). After a 24 h incubation period, different concentrations of 10 were added to the cell. The cells were cultured for 14 d in the incubator, with the media being changed every 3 d. The cells were then fixed for 30 min with 4% paraformaldehyde and stained for 30 min with 0.1% crystal violet. After washing, the plates were photographed and counted using ImageJ2×.2.1.5.0 software. The clone formation rate relative to the solvent control group was calculated.

3.10.5. Cell Apoptosis Analysis

Huh7 and HepG2 cells were seeded in 6-well cell culture plates. The cells were then grown for 24 h with varied doses of 10, and subsequent processes were carried out based on the explanatory memorandum of the Annexin V-FITC/PI Apoptosis Detection Kit (Yeasen, Cat.#: 40302ES60, Shanghai, China). Briefly, cells were digested by trypsin, centrifuged at 4 °C with 1500 rpm for 5 min, washed with pre-cooling PBS twice, and resuspended in 100 μL 1 × binding buffer before staining with 5 μL Annexin-V-FITC and 10 μL PI staining solution for 10–15 min incubation in the darkness. After diluting with 400 μL of 1 × binding buffer, the samples were examined by flow cytometry (Sysmex-Partec CyFlowTM Cube 6). Data were analyzed using FlowJo V1 (Flexera Software, Chicago, IL, USA).

3.10.6. Cell Cycle Arrest Analysis

Huh7 and HepG2 cells were seeded in 6-well cell culture plates. The cells were then exposed to various doses of 10 for 24 h. The cells were harvested, washed three times with PBS, and then fixed for at least 12 h in cold 70% ethanol at 4 °C. After that, all of the cells were washed three times in PBS to remove any remaining ethanol. The cells were then resuspended in the solution stained with propidium iodide and incubated at room temperature for 30 min in the dark. A NovoCyte Flow Cytometer (Agilent, Santa Clara, CA, USA) was used to analyze the samples.

3.10.7. Wound Healing Assay

Cells (1 × 106/mL) were seeded in 6-well plates and cultured until each well was covered. The cell monolayer was scraped vertically with a 200 μL sterile pipette tip and rinsed 3 times with sterile PBS. After that, 5% FBS medium was added with the compounds to be tested and incubated for 48 h. Images were taken at 0 and 48 h for each scratch by microscope (OLYMPUS IX-51). ImageJ software was used to compare the edge-by-edge measurements at 0 h and 24 h.

3.10.8. Cell Migration Assays

Cell migration assays were performed in the chamber of a 24-well Transwell plate. For migration experiments, starved cells were suspended in a medium without serum with a density of 1 × 105/mL. 200 μL of prepared cell suspension; various concentrations of compounds were added to the upper chamber of the chamber and 600 μL of medium (20% FBS) was added to the outer chamber of the chamber. The plates were incubated in an incubator at 37 °C for 48 h. Then, the cells in the upper chamber were swabbed, and the rest of the cells in the chamber were fixed with 4% paraformaldehyde for 30 min, stained with 0.1% crystal violet, and photographed (OLYMPUS IX-51) for observation.

3.10.9. ADME Prediction

Computational ADME analysis was performed using SwissADME (http://www.swissadme.ch/, accessed on 14 September 2023) to estimate the physiochemical, lipophilicity, water-solubility, pharmacokinetics, drug-likeness, and medicinal chemistry properties of the compound.

3.10.10. Statistical Analysis

All statistical analyses were performed using GraphPad-Prism software 9. All data are expressed as the mean ± SD for three independent tests. The statistical significance of the data between groups was acquired by either Student’s test or one-way ANOVA multiple comparisons.

4. Conclusions

In this study, a new series of SL derivatives (318) were synthesized using epimers 1 and 2 as the parent molecules, isolated from Sphagneticola trilobata, and evaluated for their anti-HCC activity. Furthermore, the structures of 4, 6, and 14 were confirmed by X-ray single-crystal diffraction analyses. The cytotoxic activities of 318 on two HCC cell lines, including HepG2 and Huh7, were evaluated using the CCK-8 assay. Among them, compound 10 exhibited the best activity against HepG2 and Huh7 cell lines with IC50 values of 9.73 and 18.86 µM, respectively. It was suggested that the double bonds at the C-4, 14 or C-3, 4 positions were more favorable to enhancing the activity of HCC. Notably, the double bond between C-11 and C-13 was reduced to the methyl group in all the compounds, which were less potent than the parental compounds, further confirming that SLs’ cytotoxic activity is mainly dependent on the α-methylene-γ-lactone group. Further studies showed that 10 induced cell apoptosis, arrested the cell cycle in the S phase, and induced the inhibition of cell proliferation and migration in HepG2 and Huh7. ADME properties prediction showed that 10 may possess the properties to be a drug candidate. Thus, 10 may be a promising lead compound for the treatment of HCC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules29020393/s1, The preparation process of 1 and 2; 1H NMR, 13C NMR, and HRESIMS of compounds 318 (Figures S1–S55) (PDF).

Author Contributions

X.Z. performed the experiments for the design, synthesis, and anti-hepatocellular carcinoma evaluation of the manuscript; X.Z. and G.Y. contributed to the cytotoxicity assays; Y.Q. and X.Y. contributed to part of the design and synthesis; G.C. contributed to part of the structure determination; Y.H. reviewed and edited the manuscript; W.C. supervised the research work and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (22167013), the Key Project of Research and Development of Hainan Province (ZDYF2021SHFZ221), the Hainan Provincial Natural Science Foundation of China (823MS043 and 221RC541), and the Specific Research Fund of the Innovation Platform for Academicians of Hainan Province (YSPTZX202030).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in Supplementary Materials.

Conflicts of Interest

The authors declare no competing financial interests.

References

  1. Mao, J.J.; Pillai, G.G.; Andrade, C.J.; Ligibel, J.A.; Basu, P.; Cohen, L.; Khan, I.A.; Mustian, K.M.; Puthiyedath, R.; Dhiman, K.S. Integrative oncology: Addressing the global challenges of cancer prevention and treatment. CA Cancer J. Clin. 2022, 72, 144–164. [Google Scholar] [CrossRef]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; 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, 209–249. [Google Scholar] [CrossRef]
  3. Forner, A.; Gilabert, M.; Bruix, J.; Raoul, J.-L. Treatment of intermediate-stage hepatocellular carcinoma. Hepatobil. Surg. Nutr. 2014, 11, 525–535. [Google Scholar] [CrossRef]
  4. Sperandio, R.C.; Pestana, R.C.; Miyamura, B.V.; Kaseb, A.O. Hepatocellular carcinoma immunotherapy. Annu. Rev. Med. 2022, 73, 267–278. [Google Scholar] [CrossRef]
  5. Ling, Y.; Gao, W.J.; Ling, C.C.; Liu, J.; Meng, C.; Qian, J.Q.; Liu, S.Q.; Gan, H.L.; Wu, H.M.; Tao, J.H.; et al. β-carboline and N-hydroxycinnamamide hybrids as anticancer agents for drug-resistant hepatocellular carcinoma. Eur. J. Med. Chem. 2019, 168, 515–526. [Google Scholar] [CrossRef] [PubMed]
  6. Ruwizhi, N.; Aderibigbe, B.A. Cinnamic acid derivatives and their biological efficacy. Int. J. Mol. Sci. 2020, 21, 5712. [Google Scholar] [CrossRef] [PubMed]
  7. Li, C.; Jones, A.X.; Lei, X.G. Synthesis and mode of action of oligomeric sesquiterpene lactones. Nat. Prod. Rep. 2016, 33, 602–611. [Google Scholar] [CrossRef] [PubMed]
  8. Ren, Y.L.; Yu, J.H.; Kinghorn, D.A. Development of anticancer agents from plant-derived sesquiterpene lactones. Curr. Med. Chem. 2016, 23, 2397–2420. [Google Scholar] [CrossRef]
  9. Yang, Z.J.; Ge, W.Z.; Li, Q.Y.; Lu, Y.X.; Gong, J.M.; Kuang, B.J.; Xi, X.N.; Wu, H.T.; Zhang, Q.; Chen, Y. Syntheses and biological evaluation of costunolide, parthenolide, and their fluorinated analogues. J. Med. Chem. 2015, 58, 7007–7020. [Google Scholar] [CrossRef]
  10. Chen, H.; Wu, G.Z.; Gao, S.; Guo, R.H.; Zhao, Z.; Yuan, H.; Liu, S.X.; Wu, J.; Lu, X.L.; Yuan, X.; et al. Discovery of potent small-molecule inhibitors of ubiquitin-conjugating enzyme UbcH5c from α-santonin derivatives. J. Med. Chem. 2017, 60, 6828–6852. [Google Scholar] [CrossRef]
  11. Janganati, V.; Ponder, J.; Jordan, C.T.; Borrelli, M.J.; NPenthala, A.R.; Crooks, P.A. Dimers of melampomagnolide B exhibit potent anticancer activity against hematological and solid tumor cells. J. Med. Chem. 2015, 58, 8896–8906. [Google Scholar] [CrossRef] [PubMed]
  12. Zhangabylov, N.S.; Dederer, L.Y.; Gorbacheva, L.B.; Vasil’eva, S.V.; Terekhov, V.S.; Adekenov, S.M. Sesquiterpene lactone arglabin influences DNA synthesis in P388 leukemia cells in vivo. Pharm. Chem. J. 2004, 38, 651–653. [Google Scholar] [CrossRef]
  13. Ghantous, A.; Gali-Muhtasib, H.; Vuorela, H.; Saliba, N.A.; Darwiche, N. What made sesquiterpene lactones reach cancer clinical trials? Drug Discovery Today 2010, 15, 668–678. [Google Scholar] [CrossRef]
  14. Li, J.; Li, S.S.; Guo, J.S.; Li, Q.Y.; Long, J.; Ma, C.; Ding, Y.H.; Yan, C.L.; Li, L.W.; Wu, Z.G.; et al. Natural product micheliolide (MCL) irreversibly activates pyruvate kinase M2 and suppresses leukemia. J. Med. Chem. 2018, 61, 4155–4164. [Google Scholar] [CrossRef]
  15. Wong, Y.K.; Xu, C.; Kalesh, K.A.; He, Y.; Lin, Q.; Wong, W.S.F.; Shen, H.M.; Wang, J. Artemisinin as an anticancer drug: Recent advances in target profiling and mechanisms of action. Med. Res. Rev. 2017, 37, 1492–1517. [Google Scholar] [CrossRef]
  16. Dai, Y.F.; Zhou, W.W.; Meng, J.; Du, X.L.; Sui, Y.P.; Dai, L.; Wang, P.Q.; Huo, H.R.; Sui, F. The pharmacological activities and mechanisms of artemisinin and its derivatives: A systematic review. Med. Chem. Res. 2017, 26, 867–880. [Google Scholar] [CrossRef]
  17. Li, Y.; Zhou, X.; Liu, J.; Yuan, X.; He, Q. Therapeutic potentials and mechanisms of artemisinin and its derivatives for tumorigenesis and metastasis. Anti-Cancer Agents Med. Chem. 2020, 20, 520–535. [Google Scholar] [CrossRef] [PubMed]
  18. Augustin, Y.; Staines, H.M.; Krishna, S. Artemisinins as a novel anti-cancer therapy: Targeting a global cancer pandemic through drug repurposing. Pharm. Ther. 2020, 216, 107706. [Google Scholar] [CrossRef]
  19. Lesiak, K.; Koprowska, K.; Zalesna, I.; Nejc, D.; Düchler, M.; Czyz, M. Parthenolide, a sesquiterpene lactone from the medical herb feverfew, shows anticancer activity against human melanoma cells in vitro. Melanoma Res. 2010, 20, 21–34. [Google Scholar] [CrossRef] [PubMed]
  20. Nasim, S.; Crooks, P.A. Antileukemic activity of aminoparthenolide analogs. Bioorg. Med. Chem. Lett. 2008, 18, 3870–3873. [Google Scholar] [CrossRef]
  21. Guzman, M.L.; Rossi, R.M.; Neelakantan, S.; Li, X.J.; Corbett, C.A.; Hassane, D.C.; Becker, M.W.; Bennett, J.M.; Sullivan, E.; Lachowicz, J.L.; et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 2007, 110, 4427–4435. [Google Scholar] [CrossRef]
  22. Zhang, Q.; Lu, Y.X.; Ding, Y.H.; Zhai, J.D.; Ji, Q.; Ma, W.W.; Yang, M.; Fan, H.G.; Long, J.; Tong, Z.H.; et al. Guaianolide sesquiterpene lactones, a source to discover agents that selectively inhibit acute myelogenous leukemia stem and progenitor cells. J. Med. Chem. 2012, 55, 8757–8769. [Google Scholar] [CrossRef] [PubMed]
  23. Rozalski, M.; Krajewska, U.; Panczyk, M.; Mirowski, M.; Rozalska, B.; Wasek, T.; Janecki, T. Synthesis and biological evaluation of 4-methylideneisoxazolidin-5-ones—A new class of highly cytotoxic α-methylidene-γ-lactones. Eur. J. Med. Chem. 2007, 42, 248–255. [Google Scholar] [CrossRef]
  24. Polo, L.M.; Castro, C.M.; Cruzado, M.C.; Collino, C.J.G.; Cuello-Carrión, F.D.; Ciocca, D.R.; Giordano, O.S.; Ferrari, M.; López, L.A. 11,13-dihydro-dehydroleucodine, a derivative of dehydroleucodine with an inactivated alkylating function conserves the anti-proliferative activity in G2 but does not cause cytotoxicity. Eur. J. Pharmacol. 2007, 556, 19–26. [Google Scholar] [CrossRef]
  25. Kupchan, S.M.; Eakin, M.A.; Thomas, A.M. Tumor inhibitors. 69. Structure-cytotoxicity relations among the sesquiterpene lactones. J. Med. Chem. 1971, 14, 1147–1152. [Google Scholar] [CrossRef] [PubMed]
  26. Zhou, X.Q.; Mao, X.M.; Fan, R.; Li, S.Y.; Shang, J.; Zhang, T.; Li, R.H.; Li, H.Q.; Hui, Y.; Chen, W.H.; et al. Trilobolide-6-O-isobutyrate suppresses hepatocellular carcinoma tumorigenesis through inhibition of IL-6/STAT3 signaling pathway. Phytother. Res. 2021, 35, 5741–5753. [Google Scholar] [CrossRef] [PubMed]
  27. Zhou, X.Q.; Yi, J.L.; Chen, G.Y.; Liu, Z.X.; Hui, Y.; Chen, W.H. Structure modification and anti-tumor activity evaluation on sesquiterpene lactones TBA and TBB from Sphagneticola trilobata. Nat. Prod. Res. 2023, 37, 3766–3771. [Google Scholar]
  28. Hui, Y.; Cao, J.; Lin, J.; Yang, J.N.; Liu, Y.J.; Han, C.R.; Song, X.P.; Dai, C.Y.; Zhang, X.P.; Chen, W.H.; et al. Eudesmanolides and other constituents from the flowers of Wedelia trilobata. Chem. Biodivers. 2018, 15, 1700411. [Google Scholar] [CrossRef]
  29. Wu, M.Y.; Han, Z.B.; Ni, H.Z.; Wang, N.Z.; Ding, K.L.; Lu, Y.X. Phosphine-catalyzed divergent domino processes between γ-substituted allenoates and carbonyl-activated alkenes. Chem. Sci. 2022, 13, 3161–3168. [Google Scholar] [CrossRef]
  30. Guo, T.L.; Mo, K.L.; Zhang, N.N.; Xiao, L.P.C.; Liu, W.L.; Wen, L.L. Embedded homogeneous ultra-fine Pd nanoparticles within MOF ultra-thin nanosheets for heterogeneous catalysis. Dalton Trans. 2021, 50, 1774–1779. [Google Scholar] [CrossRef]
  31. Wu, J.; Zhang, J.Z.; Soto-Acosta, R.; Mao, L.L.; Lian, J.H.; Chen, K.; Pillon, G.; Zhang, G.Q.; Geraghty, R.J.; Zheng, S.P. One-pot synthesis of 1-hydroxyacridones from para-quinols and ortho-methoxycarbonylaryl isocyanates. J. Org. Chem. 2020, 85, 4515–4524. [Google Scholar] [CrossRef]
  32. Wang, X.J.; Zhang, X.M.; Zheng, B.B.; Hu, N.; Xie, W.D.; Row, K. Synthesis of 13-amino telekin derivatives and their cytotoxic activity. Nat. Prod. Res. 2015, 29, 756–763. [Google Scholar] [CrossRef] [PubMed]
  33. Kardile, R.A.; Sarkate, A.P.; Lokwani, D.K.; Tiwari, S.V.; Azad, R.; Thopate, S.R. Design, synthesis, and biological evaluation of novel quinoline derivatives as small molecule mutant EGFR inhibitors targeting resistance in NSCLC: In vitro screening and ADME predictions. Eur. J. Med. Chem. 2023, 245, 114889. [Google Scholar] [CrossRef] [PubMed]
  34. Zou, J.P.; Zhang, Z.; Lv, J.Y.; Zhang, X.Q.; Zhang, Z.Y.; Han, S.T.; Liu, Y.W.; Liu, W.W.; Ji, J.; Shi, D.H. Design, synthesis and anti-cancer evaluation of genistein-1,3,5-triazine derivatives. Tetrahedron 2023, 134, 133293. [Google Scholar] [CrossRef]
Figure 1. Structures of ART, DHA, Artesunate, PTL, DMAPT, and ACT001 1 and 2.
Figure 1. Structures of ART, DHA, Artesunate, PTL, DMAPT, and ACT001 1 and 2.
Molecules 29 00393 g001
Scheme 1. Synthesis route to obtain 1 derivatives. Reagents and conditions: (a) THF, HCl (2M, aq), 45 °C, 26 h; (b) KOH (aq), THF:H2O = 1:1, 0 °C, 2 h; (c) NaBH4, CH3CN, rt, 1 h; (d) KOH, MeOH, rt, 6 h.
Scheme 1. Synthesis route to obtain 1 derivatives. Reagents and conditions: (a) THF, HCl (2M, aq), 45 °C, 26 h; (b) KOH (aq), THF:H2O = 1:1, 0 °C, 2 h; (c) NaBH4, CH3CN, rt, 1 h; (d) KOH, MeOH, rt, 6 h.
Molecules 29 00393 sch001
Scheme 2. Synthesis route to obtain 2 derivatives. Reagents and conditions: (a) THF, HCl (2M, aq), 45 °C 9 h; (b) THF, H2SO4 (8M, aq), 45 °C, 10 h; (c) NaBH4, CH3CN, rt, 1 h; (d) KOH, MeOH, rt, 6 h; (e) DMSO, DBU, rt, 79 h.
Scheme 2. Synthesis route to obtain 2 derivatives. Reagents and conditions: (a) THF, HCl (2M, aq), 45 °C 9 h; (b) THF, H2SO4 (8M, aq), 45 °C, 10 h; (c) NaBH4, CH3CN, rt, 1 h; (d) KOH, MeOH, rt, 6 h; (e) DMSO, DBU, rt, 79 h.
Molecules 29 00393 sch002
Figure 2. Cell viability of Huh7 and HepG2 cells. (A,B) Three cells were treated with indicated concentrations of compound 10 (3.125, 6.25, 12.5, 25, and 50 μM) for 24 h. (C,D) Cell viability of Huh7 cells treated with 10 at different concentrations for 12, 24, 26, 48, 60, and 72 h, respectively.
Figure 2. Cell viability of Huh7 and HepG2 cells. (A,B) Three cells were treated with indicated concentrations of compound 10 (3.125, 6.25, 12.5, 25, and 50 μM) for 24 h. (C,D) Cell viability of Huh7 cells treated with 10 at different concentrations for 12, 24, 26, 48, 60, and 72 h, respectively.
Molecules 29 00393 g002
Figure 3. Live/dead cell staining of Huh7 and HepG2 cells treated with compound 10. Scale bar: 100 μm.
Figure 3. Live/dead cell staining of Huh7 and HepG2 cells treated with compound 10. Scale bar: 100 μm.
Molecules 29 00393 g003
Figure 4. Compound 10 suppressed the colony formations of Huh7 and HepG2 cells. (A,B) Huh7 and HepG2 cells were treated with 10 for 14 d, and dyed with 0.1% crystal violet. The relative colony numbers are represented as a histogram.
Figure 4. Compound 10 suppressed the colony formations of Huh7 and HepG2 cells. (A,B) Huh7 and HepG2 cells were treated with 10 for 14 d, and dyed with 0.1% crystal violet. The relative colony numbers are represented as a histogram.
Molecules 29 00393 g004
Figure 5. Compound 10 induced apoptosis of Huh7 and HepG2 cells. (A,C) Huh7 and HepG2 cells were treated with 10 for 24 h. The apoptosis was analyzed by flow cytometry. (B,D) The apoptotic rates are represented as a histogram.
Figure 5. Compound 10 induced apoptosis of Huh7 and HepG2 cells. (A,C) Huh7 and HepG2 cells were treated with 10 for 24 h. The apoptosis was analyzed by flow cytometry. (B,D) The apoptotic rates are represented as a histogram.
Molecules 29 00393 g005
Figure 6. Compound 10 induced S phase arrest in Huh7 and HepG2 cells. (A,C) Huh7 and HepG2 cells were incubated with 10 for 24 h, respectively. The distribution of the cell cycle was analyzed by flow cytometry. (B,D) The percentage of cell cycle distribution is represented as a histogram.
Figure 6. Compound 10 induced S phase arrest in Huh7 and HepG2 cells. (A,C) Huh7 and HepG2 cells were incubated with 10 for 24 h, respectively. The distribution of the cell cycle was analyzed by flow cytometry. (B,D) The percentage of cell cycle distribution is represented as a histogram.
Molecules 29 00393 g006
Figure 7. Experimental results of the migration of cells incubated in compound 10. (A) Wound healing assay results of Huh7 cells incubated in compounds for 0 h and 48 h (scale bar = 100 μm, magnification 4×). (B) Statistical analysis of the scratch experiment and the healing rate (%) = (scratch area at 0 h -scratch area at 24 h)/scratch area at 0 h ×100%. (C) Transwell migration result of Huh7 cells treated with different concentrations of compound 10 for 48 h (scale bar = 100 μm, magnification 20×). (D) Statistical results of Transwell migration tests (n = 3).
Figure 7. Experimental results of the migration of cells incubated in compound 10. (A) Wound healing assay results of Huh7 cells incubated in compounds for 0 h and 48 h (scale bar = 100 μm, magnification 4×). (B) Statistical analysis of the scratch experiment and the healing rate (%) = (scratch area at 0 h -scratch area at 24 h)/scratch area at 0 h ×100%. (C) Transwell migration result of Huh7 cells treated with different concentrations of compound 10 for 48 h (scale bar = 100 μm, magnification 20×). (D) Statistical results of Transwell migration tests (n = 3).
Molecules 29 00393 g007
Table 1. Cytotoxic activity of compounds 118 in HepG2 and Huh7 cells.
Table 1. Cytotoxic activity of compounds 118 in HepG2 and Huh7 cells.
CompoundIC50 (μM)
HepG2Huh7CompoundHepG2Huh7
127.77 ± 0.9228.15 ± 0.871147.98 ± 2.5356.69 ± 1.09
222.12 ± 1.1314.04 ± 2.3712>70>70
314.42 ± 1.6924.17 ± 0.5713>70>70
439.43 ± 2.8444.07 ± 3.3014>70>70
515.00 ± 1.0749.69 ± 2.4615>70>70
6>70>7016>70>70
7>70>7017>70>70
8>70>7018>70>70
9>70>70adriamycin26.80 ± 1.3323.87 ± 1.62
109.73 ± 1.7218.86 ± 0.88
IC50: concentration of the compound that is required for 50% inhibition of cell growth.
Table 2. Physiochemical, lipophilicity, water-solubility, pharmacokinetics, drug-likeness, and medicinal chemistry properties of compound 10.
Table 2. Physiochemical, lipophilicity, water-solubility, pharmacokinetics, drug-likeness, and medicinal chemistry properties of compound 10.
Physicochemical PropertiesWater Solubility
FormulaC21H28O7Log S (ESOL)−3.35
Molecular weight392.44 g/molSolubility1.76 × 10−1 mg/mL; 4.49 × 10−4 mol/L
Num. heavy atoms28ClassSoluble
Num. arom. heavy atoms0Log S (Ali)−3.95
Fraction Csp30.67Solubility4.43 × 10−2 mg/mL; 1.13 × 10−4 mol/L
Num. rotatable bonds5ClassSoluble
Num. H-bond acceptors7Log S (SILICOS-IT)−2.30
Num. H-bond donors1Solubility1.99 × 100 mg/mL; 5.06 × 10−3 mol/L
Molar Refractivity100.53ClassSoluble
TPSA99.13 Å2Pharmacokinetics
LipophilicityGI absorptionHigh
Log Po/w (iLOGP)2.71BBB permeantNo
Log Po/w (XLOGP3)2.23P-gp substrateNo
Log Po/w (WLOGP)1.93CYP1A2 inhibitorNo
Log Po/w (MLOGP)2.04CYP2C19 inhibitorNo
Log Po/w (SILICOS-IT)2.12CYP2C9 inhibitorNo
Consensus Log Po/w2.20CYP2D6 inhibitorNo
DruglikenessCYP3A4 inhibitorNo
LipinskiYes; 0 violationLog Kp (skin permeation)−7.11 cm/s
GhoseYesMedicinal Chemistry
VeberYesPAINS0 alert
EganYesBrenk3 alerts: isolated_alkene, michael_acceptor_1, more_than_2_esters
MueggeYesLead-likenessNo; 1 violation: MW > 350
Bioavailability Score0.55Synthetic accessibility5.32
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhou, X.; Yi, G.; Qian, Y.; Yang, X.; Chen, G.; Hui, Y.; Chen, W. Design, Synthesis, and Anti-Hepatocellular Carcinoma Evaluation of Sesquiterpene Lactone Epimers Trilobolide-6-O-isobutyrate Analogs. Molecules 2024, 29, 393. https://doi.org/10.3390/molecules29020393

AMA Style

Zhou X, Yi G, Qian Y, Yang X, Chen G, Hui Y, Chen W. Design, Synthesis, and Anti-Hepatocellular Carcinoma Evaluation of Sesquiterpene Lactone Epimers Trilobolide-6-O-isobutyrate Analogs. Molecules. 2024; 29(2):393. https://doi.org/10.3390/molecules29020393

Chicago/Turabian Style

Zhou, Xiuqiao, Guohui Yi, Yiming Qian, Xiaorong Yang, Guangying Chen, Yang Hui, and Wenhao Chen. 2024. "Design, Synthesis, and Anti-Hepatocellular Carcinoma Evaluation of Sesquiterpene Lactone Epimers Trilobolide-6-O-isobutyrate Analogs" Molecules 29, no. 2: 393. https://doi.org/10.3390/molecules29020393

Article Metrics

Back to TopTop