2.1. Isolation and Characterization of Eurycomalactone and Eurycomanone
Eurycomanone (
1), was isolated from
E. longifolia as a white powder. Its molecular formula C
20H
24O
9 was established by positive-ion HREIMS [M+H]
+409.3851,(calcd C
20H
24O
9 for
m/z 408.403. Comparing its
1H and
13C-NMR spectra (
Table 1) with those of (2), we found that chemical shift of methyl protons for 4 and 8-CH
3 was increased to δ
H 1.81 and 2.00 respectively. The presence of
ortho protons in C-14 and C-15 were displayed δ
H 3.25 and δ
H 5.25 respectively. 20 signals were displayed in its
13C-NMR spectrum including two carbonyl carbons at δ
C 197.89 (C-2) and δ
C 174.35 and four methylene carbons at δ
C 162.98 (C-3), 126.48 (C-4), 119.80 (C-13) and 108.71 (C-13’).
Eurycomalactone (2), was a white powder and its molecular formula was determined as C19H24O6 by HREIMS at m/z 349.1647 [M+H]+, 1H and 13C-NMR spectra. The 1H-NMR spectrum of (1), showed signals at δH 6.13 (1H, s) corresponding to vinyl proton at C-3. Four signals resolved at δH 1.64, 1.96, 1.27 and 1.18 (3H, s) indicated methyl protons in compound (1). The 13C NMR peak assignments showed four methyl carbons resonated at δC 23.64, 21.97, 12.19 and 32.33. Meanwhile signals at δ 176.31, 197.41 and 205.56 attributed to carbonyl carbon (C-2, C-3 and C-15).
The known constituent (
1) and (
2) were identified by comparison of its spectral data
1H and
13C NMR and MS with those reported in the literature [
13,
39,
40]. The structure of eurycomanone (
1) and eurycomalactone (
2) are shown in
Figure 1.
2.2. Percentage of Cells Viability and IC50 values
The in vitro anti-cancer effects of eurycomanone and eurycomalactone were evaluated against ovarian (A2780), cervical (HeLa), colorectal (HT29) cancer cells, as well as normal cardiomyocyte (H9C2) and liver (WRL-68) cell lines. Cisplatin and methotrexate were also evaluated on its in vitro anti-cancer effects as for comparison studies. Both drugs had been reported to treat various type of cancers including ovarian, cervical and colorectal cancers
[41,42,43]. Both eurycomanone and eurycomalactone gave significant in vitro anti-cancer effects, with IC
50 values ranging from 1.22 ± 0.11 µM to 4.58 ± 0.090 µM (for eurycomanone) and 1.60 ± 0.12 µM to 2.46 ± 0.081 µM (for eurycomalactone) as illustrated in
Figure 2 and tabulated in
Table 2. Cisplatin exerted comparable in vitro anti-cancer activities with the range of IC
50 between 1.38 ± 0.037 µM to 1.77 ± 0.018 µM, whereas, methotrexate exerted better in vitro anti-cancer activities with the range of IC
50 between 0.016 ± 0.00050 µM to 0.094 ± 0.0043 µM respectively (
Figure 2,
Table 2). Eurycomanone, eurycomalactone and methotrexate showed less cytotoxic effects in cardiomyocyte H9c2 normal cell line as opposed to liver WRL-68 normal cell line (
Table 2). Clinically, cisplatin was reported to have several side effects including cardiotoxicity, nephrotoxicity and neurotoxicity
[5]. Whereas, methotrexate was also reported to have adverse effects (e.g. hepatotoxicity, nephrotoxicity, gastrointestinal toxicity and death due to infections and hemorrhage) when given to cancer patients
[44].
From our current studies, the
in vitro anti-cancer analysis had shown that eurycomanone and eurycomalactone were in the potent range since the IC
50 values were less than 50 µM [
45]. To the best of our knowledge, this is the first report that revealed eurycomalactone had in vitro anti-cancer effects against ovarian cancer cells. Other studies had also reported that eurycomanone and eurycomalactone gave in vitro anti-cancer effects against other cancer cell lines. Eurycomanone was reported to have anti-cancer effects in breast [
46,
47], colon, fibrosarcoma, lung and melanoma cancer cell lines [
46] with IC
50 values ranging from 0.49-35 µM. Whereas, eurycomalactone was reported to have anti-cancer effects in murine lymphocytic leukemia (P388) and epidermoid (KB) [
48], lung cancer (A-549), breast cancer (MCF-7) [
47] and colon (26-L5), melanoma (B16-BL6), lung (LLC and A549) cancer cell lines [
49] with
IC
50 values ranging from 0.57-23.25 µM. However, the informations on the mechanisms of action of eurycomanone and eurycomalactone in killing the cancer cells are still lacking.
2.4. Molecular Docking Analysis
Molecular docking is a widely used computational technique in modern drug design for predicting drug-receptor interactions and identifying potential inhibitors. In this study, we conducted a comparative analysis of the free binding energy and binding interactions of eurycomanone and eurycomalactone with the co-crystallized ligands in the active binding sites of TNF-α (PDB ID: 2AZ5) and DHFR (PDB ID: 5HQY) (refer to
Table 5,
Figure S1, and
Figure 5). Our aim is to gain insights into the potential mechanisms of action of eurycomanone and eurycomalactone against TNF-α and DHFR. To validate the docking process, we first re-docked the co-crystallized (original) ligands as controls into the active sites of DHFR and TNF-α. The resulting score energies were -7.93 kcal/mol and -8.19 kcal/mol, respectively, with small root mean square deviations (RMSDs) of 0.98 Å for TNF-α and 0.62 Å for DHFR (
Table 5). An RMSD value of ≤ 2.0 Å is typically considered acceptable in the literature [
51,
52,
53,
54,
55]. Based on these results, we applied the same docking parameters to dock eurycomanone and eurycomalactone.
Figure S1(a, b) provides valuable insights into the interactions between co-crystalized ligands and the active binding sites of TNF-α and DHFR. In
Figure S1a, the original ligand complexed into the active binding site of TNF-α (PDB ID: 2AZ5) is observed to form a single hydrogen bond with the tyrosine residue TYR151 at a distance of 2.45 Å. In addition, the ligand forms pi-sigma and pi-alkyl interactions with TYR119 and TYR59, which are known to be important in protein-ligand recognition and may contribute to the inhibitory activity of the compound. These findings are consistent with those reported in [
36]. In
Figure S1b, the original co-crystalized ligand in PDB ID: 2AZ5 adopts a bent conformation and forms four hydrogen bonds (H-bonds) with ILE7 (3.09 Å), GLU30 (2.59 Å), ASN64 (3.44 Å), and VAL115 (3.15 Å), with relatively weak to moderate interactions. The inhibitor also establishes hydrophobic contacts with several active site residues of DHFR, including ALA9, LEU22, PHE31, PHE34, and ILE60. These interactions likely contribute to the inhibitory activity of the co-crystalized ligands. A comprehensive understanding of these interactions can shed light on the mechanism of action of TNF-α and DHFR inhibitors and facilitate the development of more effective drugs targeting these enzymes.
Table 5 presents the free binding energies of eurycomanone and eurycomalactone, compared to the co-crystallized ligand, within the active binding site of TNF-α (PDB ID: 2AZ5). Remarkably, the calculated binding free energies for eurycomanone and eurycomalactone are -8.83 kcal/mol and -7.51 kcal/mol, respectively, which are substantially similar to that of the co-crystallized ligand.
Figure 5(a,b) show detailed 2D and 3D molecular interaction analyses of eurycomanone and eurycomalactone with the active site of TNF-α. Eurycomanone is found to form three hydrogen bonds, with two of them formed between the hydroxyl group and carbonyl group of the 6-hydroxy-3-methylcyclohex-2-en-1-one ring with GLY121(A) at a distance of 2.24 Å and TYR151(B) at a distance of 1.92 Å. The third hydrogen bond is formed between the hydroxyl group of the 2-methylenecyclohex-3-en-1-ol ring and LEU120(A) at a distance of 2.02 Å. Eurycomanone also engages in pi-alkyl interactions with TYR59(A), TYR119(A), and TYR119(B) (See
Figure 5a). In the same pattern, eurycomalactone interacts with the same residues through pi-alkyl interactions with TYR59(B), TYR119(B), and TYR151(B), and its carbonyl group of the 6-hydroxy-3-methylcyclohex-2-en-1-one ring forms a single hydrogen bond interaction with TYR119(A) at a distance of 2.01 Å. These detailed molecular interaction patterns and the Ki values suggest that both eurycomanone and eurycomalactone may serve as promising hit candidates against TNF-alpha, with their strong binding affinity likely translating to a potent inhibitory effect on TNF-α activity.
- ➢
In the active binding site of DHFR (PDB ID: 5HQY), both eurycomanone and eurycomalactone exhibit free binding energies similar to the co-crystallized ligand. The calculated binding free energies for eurycomanone and eurycomalactone, which are -8.83 kcal/mol and -7.51 kcal/mol, respectively, suggest a promising potential for these compounds to bind to DHFR within its active binding site (
Table 5).
- ➢
Figure 5(c,d) illustrate the 2D and 3D molecular interactions of eurycomanone and eurycomalactone with DHFR. Eurycomanone is observed to establish two intermolecular hydrogen bonds with crucial residues in the active binding site of the enzyme (
Figure 5c). The first hydrogen bond is formed between the carbonyl group of the 3-hydroxytetrahydro-2H-pyran-2-one ring and the guanidine group of ARG70 at a distance of 2.56 Å, while the second hydrogen bond is formed between the hydroxyl group of the 3-hydroxytetrahydro-2H-pyran-2-one at a distance of 1.67 Å. In the case of eurycomalactone, the 6-hydroxy-4-methylcyclohex-2-en-1-one also forms two intermolecular hydrogen bonds with TRP24 at a distance of 2.47 Å and with GLU30 at a distance of 1.91 Å (
Figure 5d). Along with these hydrogen bonds, both eurycomanone and eurycomalactone are stabilized in the active site of DHFR through hydrophobic interactions with several residues, including ILE7, ALA9, LEU22, TRP24, PHE31, PHE34, and ILE60 for eurycomanone, and VAL8, ILE16, LEU22, PHE34, and VAL115 for eurycomalactone. These interactions bear substantial resemblance to those observed with the original co-crystallized ligand, indicating that eurycomanone and eurycomalactone may inhibit DHFR through similar mechanisms or contribute to the inhibitory activity of the compounds.
From these molecular docking analyses, eurycomanone and eurycomalactone had the potential to inhibit both TNF-α and DHFR. TNF-α and DHFR also had been reported to be involved in inhibiting cell proliferations via apoptosis by others, making them among potential targets for the treatment of cancer and other diseases [
22,
23,
27,
28]. Some anti-cancer drugs had been developed and targeted these TNF-α and DHFR as part of its mechanism in killing the cancer cells. For example, a few anti-cancer drugs that already approved by the FDA and others undergoing clinical trials include methotrexate [
56,
57], pemetrexed [
58] and pyrimethamine [
59]. Whereas, TNF-α is a cytokine involved in inflammation and targeted for the treatment of autoimmune, inflammatory disorders [
20,
28] and cancer [
60]. A few anti-cancer drugs that targeted TNF- α include doxorubicin [
61], melphalan [
60] and pembrolizumab [
62]. To the best of our knowledge, this is the first finding that eurycomanone and eurycomalactone were found to target TNF-α and DHFR via this molecular docking analysis. Nevertheless, further investigations shall be conducted to validate the inhibitory effects of eurycomanone and eurycomalactone against TNF-α and DHFR.
2.5. Lipinski’s Rule and ADMET of Eurycomanone and Eurycomalactone
It is known that Lipinski's Rule of Five (RoF) is utilised to evaluate the potential of a drug to be orally bioavailable [
63]. The assessment relies on molecular properties, including molecular weight, number of hydrogen bond donors and acceptors, and lipophilicity [
63]. It has become essential to predict the pharmacokinetic properties of a lead compound to assess its druggable potential before entering the drug development phase [
64,
65]. In this study, we assessed eurycomanone, eurycomalactone and methotrexate (control) using the ADMETlab 2.0 web service tool to evaluate RoF and their pharmacokinetic properties [
66]. The results of the predicted values for eurycomanone, eurycomalactone, and methotrexate are presented in
Table 6 and
Table 7.
According to RoF, a compound is more likely to be orally active if it has no more than one violation of the following criteria: Log P is less than 5; molecular weight is less than 500 Da; hydrogen bond donor is less than 5; and hydrogen bond acceptor is less than 10 [
63]. In
Table 6, the predicted values of these properties for three compounds, eurycomanone, eurycomalactone, and methotrexate are presented. Eurycomanone has a molecular weight of 408.14 g/mol, 9 hydrogen bond acceptors, 4 hydrogen bond donors, and a partition coefficient (logP) of 0.215. Eurycomalactone has a molecular weight of 348.16 g/mol, 6 hydrogen bond acceptors, 1 hydrogen bond donor, and a logP of 0.655. Both compounds satisfy the Lipinski's Rule of Five, as their molecular weights are less than 500, their logP values are within the acceptable range of 0-3, and their hydrogen bond acceptor and donor counts are less than 10 and 5, respectively.
On the other hand, methotrexate has a molecular weight of 454.17 g/mol, 13 hydrogen bond acceptors, 7 hydrogen bond donors, and a logP of -2.747. Methotrexate violates the Lipinski's Rule of Five since it has more than 10 hydrogen bond acceptors and more than 5 hydrogen bond donors. Therefore, methotrexate might have poor oral bioavailability. The Lipinski's Rule of Five predicted that eurycomanone and eurycomalactone are orally bioavailable, while methotrexate might have poor oral bioavailability due to its molecular properties. However, it should be acknowledged that this rule alone is not sufficient to guarantee the efficacy or safety of a drug, as it overlooks other factors that may affect the pharmacokinetic and pharmacodynamic profiles of a compound.
To address these limitations, it is necessary to complement the Rule of Five with more comprehensive ADMET evaluations that evaluate the absorption, distribution, metabolism, excretion, and toxicity of drug candidates. In this study, we utilized the ADMETlab 2.0 web service tool to evaluate the pharmacokinetic properties of the compounds under investigation, including eurycomanone, eurycomalactone, and methotrexate (See
Table 7), which were also analyzed using Lipinski's Rule of Five.
By combining the results of the Lipinski rule with the ADMET evaluation, we can obtain a more accurate and reliable prediction of the drug-likeness, pharmacokinetics and safety of the compounds, which is crucial for guiding the drug discovery and development process. Therefore, the combination of these two methods is highly recommended for identifying promising drug candidates with optimal pharmacokinetic properties, and this approach has the potential to improve the efficiency and success rate of the drug development process.
Caco-2 permeability is an important parameter when determining oral absorption and permeability in the early stages of drug development, and the ideal value is greater than −5.15 cm/s [
54,
67]. Eurycomanone and methotrexate showed low permeability, while eurycomalactone displayed higher permeability compared to the rest. An in vivo test on bioavailability studies had reported that eurycomanone is poorly bioavailable when given orally [
68] which supported this in silico finding. Whereas methotrexate is reported to be consumed either orally or intravenously [
69,
70]. To the best of our knowledge, there are no in vitro or in vivo studies being reported on pharmacokinetic properties on eurycomalactone. Eurycomanone, eurycomalactone and methotrexate, all showed high levels of human intestinal absorption satisfaction (HIA% with a value of 30%), which is a crucial parameter associated with human intestinal absorption [
54,
71], affecting the way compounds pass through biological membranes under the influence of physicochemical properties. In the body, plasma protein binding plays a significant role in the dynamics of compounds [
66,
72]. This phenomenon is well-known as plasma protein binding percent (PPB%). The predicted data suggest a low protein binding potential for eurycomanone and eurycomalactone compared to the reference value of 90%. Drugs with low protein binding may have a high therapeutic index [
66]. Another well-known parameter is the Blood-Brain Barrier (BBB), which facilitates the selective transfer of drug molecules between the blood and the brain parenchyma [
54,
66,
73]. As predicted, eurycomanone and eurycomalactone were not able to cross the BBB as compared to methotrexate, which adds to the safety profile of these compounds.
A drug's fate can be determined by its metabolism, which is characterized by the enzymatic modification or degradation of its molecules according to its therapeutic response [
74]. Cytochrome P450 (CYP) enzymes are essential for metabolism of drugs. Among 57 functional CYPs, the isoforms belonging to CYP1, CYP2 and CYP3 are responsible for the metabolism of 80% of clinical drugs [
75].
Table 7 shows eurycomanone, eurycomalactone, and methotrexate showed no inhibition on CYP metabolism on other drug. An in vitro assay on eurycomanone that were tested on these cytochromes also had shown that there was no inhibition [
76], thus indicating that eurycomanone may not interact with other drugs. The information on half-life and clearance of a drug candidate has important implications for preparing dosing regimen clinically. Half-life is defined as the time required for the concentration of a drug (typically in blood or plasma) to reduce to half of its initial value when the concentrations of the drug are in simple exponential (log−linear) decline [
77]. Short half-time periods and low clearance rates were predicted from our study for eurycomanone, eurycomalactone, and methotrexate. As a leading cause of drug withdrawals from the market, drug-induced toxicity will remain a key concern for the development of novel molecules [
52,
54,
78]. Eurycomanone and eurycomalactone showed low risk in exerting hepatotoxicity and carcinogenicity effects. Whereas, eurycomalactone had lower risk in exerting mutagenicity effect as compared to eurycomanone. Overall, eurycomanone and eurycomalactone showed good druglikeness and ADMET properties but further in vitro and in vivo studies need to be conducted to validate these predictions.