Molecular Docking Study of Natural Compounds from Indonesian Medicinal plants as AKT and KRAS G12D Inhibitors Candidates

 

Aisyah, Marselina Irasonia Tan, Azzania Fibriani*

School of Life Sciences and Technology, Bandung Institute of Technology, Indonesia.

*Corresponding Author E-mail: afibriani@sith.itb.ac.id

 

ABSTRACT:

Resistance to anti-EGFR treatment in colorectal cancer may arise due to the loss of PTEN function or the presence of KRAS G12D mutation. These genetic events can lead to persistent activation of the PI3K-AKT or RAS-MAPK signaling pathways, respectively. Overcoming anti-EGFR resistance can be achieved by inhibiting these signaling pathways using AKT or KRAS G12D inhibitors. The exploration of plant-derived compounds with anticancer activity offers a promising avenue for discovering potential AKT or KRAS G12D inhibitors. Therefore, this study aimed to identify natural compounds from Indonesian medicinal plants that could be developed as AKT or KRAS G12D inhibitors using a molecular docking approach. The in-silico screening of natural compounds involved the utilization of oral drug parameters. Subsequently, the filtered natural compounds were docked into the binding sites of respective proteins. The analysis involved evaluating the AutoDock Vina scoring function and examining the ligand interactions with residues within the binding site to assess the potential of the natural compounds. The findings revealed that among the 1311 natural compounds from 320 Indonesian medicinal plant species, 274 compounds met the oral drug parameters and predicted to pose anticancer activities based on QSAR analysis. Notably, morindone and porphyrin demonstrated the highest potential for development as AKT inhibitors, while phaseollin exhibited the most potential as a KRAS G12D inhibitor.

 

KEYWORDS: Colorectal cancer, AKT, KRAS G12D, Molecular docking, Natural compound, Indonesian plant.

 

 


INTRODUCTION:

Colorectal cancer is the third most incidence cancer in the world1. The overexpression of epidermal growth factor receptor (EGFR) has been identified as a prominent factor in colorectal cancer, significantly correlating with poor prognosis. To combat this, anti-EGFR drugs like cetuximab and panitumumab are commonly employed in colorectal cancer treatment. Unfortunately, the effectiveness of anti-EGFR treatment is hampered by the prevalent issue of anti-EGFR resistance among colorectal cancer patients2. One of the main causes of this resistance is attributed to the loss of PTEN function or the presence of the KRAS G12D mutation3,4.

 

PTEN loss of function leads to hyperactivation of AKT, thereby resulting in constitutive activation of the PI3K-AKT signaling pathway5. On the other hand, the KRAS G12D mutation impairs GTP hydrolysis in KRAS, leading to the persistent activation of the RAS-MAPK signaling pathway6. Consequently, targeting AKT or KRAS G12D through the use of inhibitors represents a viable strategy to treat colorectal cancer patients with anti-EGFR resistance, primarily driven by PTEN loss or KRAS G12D mutation3.

 

Efforts have been made to develop AKT and KRAS G12D inhibitors. Some AKT inhibitor candidates showed limited efficacy in clinical trials7–9, and selectivity remains a crucial concern in the development of AKT inhibitor. Similarly, the development of KRAS G12D inhibitors still faces challenge regarding its selective binding and their ability to target the KRAS G12D in its active, which these aspects are essential for ensuring a more favorable safety profile and inhibitory effects10,11. In light of that, there is a need to discover new potential compounds with potent and selective inhibitory activities against AKT and KRAS G12D. Exploration of natural compounds is one among many strategies to identify prospective lead molecules for further development into potent inhibitors. This strategy has led to the discovery of several anticancer drugs such as Paclitaxel and Vinblastine which are developed from natural compounds of taxols and velbans,                respectively 12. This report suggests that the exploration of natural compounds derived from plants holds promise for discovering potential lead molecules in developing drugs for colorectal cancer treatment13. In the pursuit of discovering new potential compounds, molecular docking method can be performed to efficiently facilitate the identification of these compounds14,15. Molecular docking is a computational method thats can predict suitable pair of small molecule and protein based on their binding strength16. According to that explanation, this study was conducted to identify potential natural compounds from Indonesian medicinal plants that possess inhibitory effects against AKT or KRAS G12D using a molecular docking approach.

 

MATERIALS AND METHODS:

Ligands Preparation:

The natural compounds were filtered using SWISSADME webserver (www.swissadme.ch/index.php)17. The filtration was done utilizing oral drug parameters, including molecular weight (200-500 g/mol), solubility (moderate-high), high GI absorption, and Lipinski`s rule of five. Molecules that comply with Lipinski’s rule of five are probably potentials to be developed as oral drugs due to their favorable bioavailability18. Ligands that met the oral drug parameters were then subjected to QSAR analysis to predict their feasibility as anticancer agents. This analysis was conducted using the antineoplastic indicator on Way2drug web tools (http://way2drug.com/passonline)19. Ligands that exceeded a Pa threshold of > 0.3 were expected to pose moderate bioactivities20. The 2D structure of natural compounds that passed the selection was retrieved from PubChem (https://pubchem.ncbi.nlm.nih.gov/). The 3D conformation of the natural compounds was drawn on MarvinSketch by applying the dreiding force field as minimization energy. The file format was converted to pdbqt using AutoDock Tools.

 

Protein Structures Preparation:

The 3D protein structures of AKT1 (PDB ID: 6CCY), KRAS G12C (PDB ID: 6OIM), KRAS G12D (PDB ID: 6GJ8), and KRAS WT (PDB ID: 6MBT) were retrieved from Protein Data Bank (PDB) (www.rcsb.org). Protein structures from PDB may have some errors, therefore it necessitates to revise their structures and add hydrogen atoms before performing simulation docking21,22. The refinement procedure to improve protein structure quality was done using PDB_REDO webserver (https://pdb-redo.eu/)23. The water molecules and ligands co-crystallography in the protein structure were deleted using BIOVIA Discovery Studio Visualizer 2021. Polar hydrogen atoms were added to structure and the file format was converted to pdbqt using AutoDock Tools 1.5.6.

 

Binding Site Determination:

The identification of the binding site was assisted by POCASA webserver24. The binding sites for docking in AKT1 and KRAS G12C structures were in the same location of the ligand co-crystallography binding sites. Therefore, the docking process validation was needed before other ligands docking25. The docking process validation was carried out by re-docking the ligand within the determined binding site. The parameter of docking process validation is RMSD value. RMSD value < 2 Å indicates the procedure for docking is valid and can be used for other ligands docking26. The RMSD value was calculated using BIOVIA Discovery Studio Visualizer 2021.

 

Molecular Docking Analyses and Visualizations:

The prepared ligands and protein structures were docked using AutoDock Vina27. Docking simulations were conducted with grid box center settings for AKT1 (x = -11.5266, y = 16.7462, z = -32.2008); KRAS G12C (x = 3.710, y = -7.746, z = -2.207); KRAS G12D (x = 5.748, y = 15.454, z = -6.94); and KRAS WT (x = 1.243, y = -21.094, z = 51.391). The analysis of ligand interactions within binding site was done using 2D diagram generated by LigPlot+2.228. Ligplot+ is able to visualize hydrogen, hydrophobic, and sometimes covalent           bonds 29. The 3D visualization of ligand binding pose in the protein binding site was done using Chimera 1.1530.

 

RESULTS AND DISCUSSIONS:

Filtration of Natural Compounds:

Choosing appropriate molecular descriptors is important for correctly predicting the drug-likeness of new compounds, thus making it a paramount consideration in drug design31. In this study, natural compounds were filtered by considering several oral drug parameters, such as molecular weight (200-500g/mol), solubility (moderate-high), high GI absorption, and Lipinski`s rule of five. Using these parameters in the filtration step can assist us to find the feasible potential compounds to be developed as oral drug17. In this study, we collected 240 plant species from the book of Atlas Tumbuhan Obat Indonesia (Volumes 1-6) and 80 plant species from 13 journal articles32–44. The filtration results disclosed that 274 out of 1311 natural compounds from 320 Indonesian medicinal plant species met the criteria of oral drug and predicted to pose anticancer activity based on QSAR analysis, therefore they were used as ligands in molecular docking simulation. Additionally, the analysis highlighted the three predominant compound classes within the 274 selected compounds, namely flavonoid, terpenoid, and alkaloid (Table 1).


 

Table 1: Class of the filtrated compounds

Class of compound

Number of compounds

Flavonoid:

2'-hydroxychalcone, chalcone, chromene, dimethoxyflavon, dimethoxyflavone, flavan, flavanol, flavanone, flavon, flavone, flavonone, homo-isoflavan, homo-isoflavanon, isoflavonoid, methoxy flavonoid, o-methylated flavonoid, o-methylated flavonol, o-methylated isoflavone, o-methylated isoflavonoid, tetramethoxyflavone.

91

Terpenoid: abdane diterpenoid, diterpene trilactone, diterpenoid furanolactone, diterpenoid lactone, germacrane sesquiterpenoid, limonoid, menthane monoterpenoid, monoterpenoid, sesquiterpenoid, sesquiterpenoid lactone, terpene lactone, triterpenoid.

65

Alkaloid: aporphine alkaloids, furoquinoline alkaloids, indole alkaloids, isoquinoline alkaloids, pyridine alkaloids, pyrrolizidine alkaloids, pyrrolizidine-type alkaloids, quinoline alkaloids, rhoeadine alkaloids.

33

Quinone Anthraquinone, dihydroxyanthraquinone, hydroxyanthraquinone, naphthaquinone, naphthoquinone

19

Quassinoid

13

Phenol

8

Coumarin: 7-hydroxycoumarin, furanocoumarin

7

Xanthone

4

Lactone

3

Phenylpropanoid

4

Others

27

 


Validation of Molecular Docking Process:

The RMSD value of docking process validation in AKT1 and KRAS G12C were 1.88 Å and 0.44 Å, respectively. RMSD <2Å indicates the docking procedures on both proteins are valid and the grid box setting can be used for other ligands docking26. The lower the RMSD value means the closer similarity of ligand conformation produced by docking simulation to the ligand conformation from the crystallography experiment45. Superimposition of re-docked ligand conformation and ligand co-crystallography conformation is given in Figure 1.

 

Figure 1: Superimposition of re-docked ligand (red) and ligand co-crystallography (cyan) conformations (A. AKT1 (6CCY); B. KRAS G12C (6OIM))

 

AKT1 Docking Study:

In our study, the ATP binding pocket of AKT1 was used as the docking site for molecular docking simulation. The active form of AKT1 is found to be more numerous in cancer cells than normal cells as the effect of the constitutive activation of PI3K-AKT signaling pathway in cancer cells. Inhibitor that can target the ATP binding pocket of AKT1 is expected to have selective inhibition effect on the active form of AKT1, thus result in better safety because it will have lower toxic effect on normal cells7.

 

ATP binding pocket is located around G-loop, β-3, αC, hinge region, catalytic loop, and DFG motif of AKT146. There are 34 residues forming the ATP-binding pocket of AKT1 which are L156, G157, K158, G159, T160, F161, G162, K163, V164 (G-loop), A177, K179, L181 (β-3), E191, E198, L202, T211 (αC), M227, E228, Y229, A230, N231, G232, G233, E234 (hinge region), R273, D274, K276, E278, N279, M281 (catalytic loop), T291, D292, F293, G294 (DFG motif)46,47. Among these residues, F161, K179, E191, T211, E228, A230, E234, E278, M281, and D292 are known to have frequent interactions with various AKT ATP-competitive inhibitors in ATP-binding pocket of AKT147,48. Therefore, these residues can be considered as important residues for ligand binding in the ATP-binding pocket of AKT149. These important residues will be written in bold in Table 3. A ligand that forms more interactions with the important residues may have more potent inhibitory effect on the target protein50. Additionally, it is reported that ATP-binding pocket of AKT1 shares high homology with ATP-binding pocket of PKA. The sequence similarity between both ATP-binding pockets is 81%46. Both ATP-binding pockets are only distinguished by three residues particularly T211, A230, and M281 in AKT1 that are substituted into V211, V230, and L281 in PKA51. Therefore, ligand interactions with T211, A230, and M281 in ATP-binding pocket of AKT1 is important to enhance ligand`s selectivity on targeting the ATP-binding pocket of AKT152.

 

Two AKT ATP-competitive inhibitors (AZD5363 and GDC0068) that have entered clinical trials phase I and II for monotherapy were used as docking controls53. The result of docking simulation revealed 12 natural compounds with best affinity score ranging from -9 kcal/mol up to -9.8 kcal/mol (Table 2). These affinity scores were lower than AZD5363 (-7.9kcal/mol) and GDC0068 (-8.1kcal/mol) (Table 2). This result suggested that the natural compounds in Table 2 had stronger binding to AKT1 compared to AZD5363 and GDC0068.


 

Table 2: Analysis results of docking affinity score in ATP-binding pocket of AKT1, ligands total interaction with 34 residues forming ATP-binding pocket of AKT1, and ligands interaction with 10 important residues in ATP-binding pocket of AKT1

Plant Species

Compounds

PubChem ID

Class of compound

Affinity (kcal/mol)

Total Interaction with residues in ATP-Binding Pocket

Total Interaction with Important Residues

-

AZD5363 (Control)

25227436

-

-7.9

35.29%

40%

-

GDC0068 (Control)

134692697

-

-8.1

38.24%

40%

Tribulus terrestris

Hecogenin

91453

Triterpenoid

-9.8

26.47%

60%

Caesalpinia pulcherrima

Neocaesalpin P

102286692

Diterpenoid

-9.7

23.53%

50%

Tribulus terrestris

Ruscogenin

441893

Triterpenoid

-9.5

26.47%

60%

Swieteniae macrophylla

7-deacetoxy-7-oxogedunin

71300386

Limonoid

-9.4

23.53%

40%

Erythrina variegata

Erythrina fusca

Phaseollin

91572

Isoflavonoid

-9.2

35.29%

50%

Nelumbo nucifera

Roemerine

119204

Aporphine alkaloid

-9.2

29.41%

60%

Tinospora crispa

Columbin

188289

Diterpenoid

-9.1

26.47%

40%

Morinda citrifolia

Morindone

442756

Anthraquinone

-9.1

35.29%

60%

Aloe vera

Rhein

10168

Anthraquinone

-9.1

29.41%

40%

Nelumbo nucifera

Anonaine

160597

Aporphine alkaloid

-9

26.47%

30%

Alstonia scholaris

Porphyrin

66868

Tetrapyrrole

-9

35.29%

70%

Catharanthus roseus

Tetrahydroalstonine

72340

Indole alkaloid

-9

29.41%

50%

 


The analysis of ligand total interaction with 34 residues forming ATP-binding pocket of AKT1 exhibited that AZD5363 and GDC0068 could form interactions with 12 out of 34 residues (35.29%) and 13 out of 34 residues (38.24%) respectively (Table 2).  The total interaction of almost all natural compounds with 34 residues forming ATP-binding pocket of AKT1 were lower than controls, except phaseollin, morindone, and porphyrin. These natural compounds exhibited a comparable total interaction profile to AZD5363 (Table 2). On the other hand, it was found that majority of natural compounds in Table 2 including phaseollin, morindone, and porphyrin formed more interactions with 10 important residues in ATP-binding pocket of AKT1 (F161, K179, E191, T211, E228, A230, E234, E278, M281, and D292) compared to AZD5363 and GDC0068. This result suggested that phaseollin, morindone, and porphyrin were potential AKT inhibitor candidates among the natural compounds. On further analysis regarding selectivity of phaseollin, morindone, and porphyrin on targeting the ATP-binding pocket of AKT1 by analyzing their interactions with three key residues responsible for ligand binding selectivity (T211, A230, and M281), it showed that morindone and porphyrin interacted with T211, A230, and M281 whilst AZD5362 only interacted with M281 (Table 3). This result implied that morindone and porphyrin might have potential to selectively target the ATP-binding pocket of AKT1 due to their interactions with T211, A230, and M281 in ATP-binding pocket of AKT1.


 

Table 3: Binding interaction profile of phaseollin, morindone, porphyrin, and AZD5363 in ATP-binding pocket of AKT1

Compounds

Hydrogen Bonds

Hydrophobic Bonds

AZD5363

(Control)

E234

L181, G159, G162, K163, K179, E198, M227, D292, V164, M281, T291

Phaseollin

F161, G162

K158, G157, V164, E234, A177, M281, M227, T211, K179, G159

Morindone

E228, A230, T211, D292, K179, E198

Y229, L156, V164, F438, A177, M227, M281

Porphyrin

-

A230, T211, A177, E228, M281, M227, L156, K179, E234, F438, V164, D292, G157

 


Through the 3D visualization, it can be seen that the binding poses of morindone and pophyrin in ATP-binding pocket of AKT1 are located deeper than AZD5363 (Figure 2-4). This deeper binding pose may play a role in causing morindone and porphyrin to form interactions with T211, A230, and M281. Based on this study results, morindone and porphyrin were found to be the most potential natural compounds to be developed as AKT inhibitors.

 

Figure 2: 3D and 2D visualizations of AZD5363 in ATP-binding pocket of AKT1

 

 

Figure 3: 3D and 2D visualizations of Morindone in ATP-binding pocket of AKT1

 

Figure 4: 3D and 2D visualizations of Porphyrin in ATP-binding pocket of AKT1

 

KRAS Docking Study:

To identify potential natural compounds with selective inhibition effects on KRAS G12D, we performed docking experiments using filtered natural compounds against KRAS G12D, KRAS G12C, and KRAS WT. The 3D protein structure of KRAS G12D in its active state was utilized since it has been reported that cancer cells with KRAS G12D mutation predominantly express KRAS in its active state. Consequently, targeting KRAS G12D in its active state is considered more favorable for achieving optimum inhibition of cancer cells with KRAS G12D mutation 54. However, due to the unavailability of 3D protein structures of KRAS G12C and KRAS WT in their active states in the PDB database, we had to dock the filtered ligands onto KRAS G12C and KRAS WT in their inactive states. In order to ensure fairness in the prediction process for both states, we employed a strategy that targeted a binding site accessible in both the active and inactive states of KRAS. Our selection process considered multiple factors, including affinity score, ligand interactions with important residues within the defined binding site, and ligand interactions with specific mutation residues, to identify natural compounds with potential selective inhibition effects on KRAS G12D.

 

The docking site for ligand binding on KRAS was determined to be the allosteric site between the central β-sheet and α2 helix (Switch-II) as well as the α3 helix 55. Within this allosteric site, two possible binding sites for ligands exist: the Switch-II pocket (SII-P) and the Switch-II groove (SII-G)55,56. The availability of SII-P is dependent on the conformation of the Switch-II region in KRAS. SII-P is accessible only when Switch-II adopts an open conformation, which occurs when KRAS is in its inactive state. In contrast, when KRAS is in its active state, the conformation of Switch-II changes to a closed state, covering the SII-P binding site and rendering it unavailable55,56. On the other hand, SII-G remains accessible regardless of the conformation of Switch-II, making it available in both the active and inactive states of KRAS55. Targeting the
SII-G of KRAS in its active state, Zhang et al. (2020) and Gentile et al. (2017) successfully identified potential compounds that exhibited good inhibition effects on KRAS in its active state. Several important residues play a crucial role in ligand binding within SII-P and SII-G of KRAS, including V9, C12/D12, K16, T58, G60, Q61, E62, E63, R68, D69, M72, D92, H95, Y96, Q99, and R102
54,57,58, as written in bold in Table 5.

 

During this study, there were no reports of clinical trials of KRAS G12D inhibitors. Therefore, an KRAS G12C inhibitor named AMG-510 was used as docking control. AMG-510 is a KRAS G12C covalent inhibitor which has entered clinical trial phase I/II for monotherapy 59. The inhibition effect of AMG-510 only works on KRAS G12C but not on KRAS WT or other KRAS mutants 60. This specific inhibition effect is caused by the covalent interaction between AMG-510 with the mutation point of C12 in the KRAS G12C56. Specific inhibition of AMG-510 toward KRAS G12C suggests that this compound will bind stronger on KRAS G12C than other KRAS variants. Hence, the affinity score of AMG-510 toward KRAS G12C, KRAS G12D, and KRAS WT can be used as cut-off score to select natural compounds with inhibitory effects on KRAS G12D. Furthermore, as the interaction with mutation point will result in specific inhibition effect, we decided only natural compounds forming interaction with D12 in the KRAS G12D but not with C12 in the KRAS G12C and G12 in the KRAS WT that could be considered as potential compounds having selective inhibition effect on KRAS G12D.


 

Table 4: Analysis results of ligands` docking affinity scores in SII-G of KRAS proteins and ligands total interaction with important residues in SII-G of KRAS variants

Plant Species

Compounds

PubChem ID

Class of compound

Affinity (kcal/mol)

Total interaction with important residues

KRAS

G12C

KRAS G12D

KRAS WT

KRAS

G12C

KRAS G12D

KRAS WT

-

AMG-510 (control)

-

-

-8.3

-6.2

-6.2

68.75%

31.25%

37.5%

Erythrina variegata

Erythrina fusca

Phaseollin

91572

Isoflavonoid

-8.7

-7.5

-6.2

37.5%

56.25%

56.25%

Plumbago zeylanica

Chitranone

633072

Naphthaquinone

-8.6

-7.2

-6.4

50%

43.75%

37.5%

Rauvolfia serpentina

Ajmalicine

441975

Indole alkaloid

-8

-7.1

-6.3

43.75%

31.25%

37.5%

Erythrina fusca

Cristacarpin

126540

Isoflavonoid

-8.3

-6.9

-6.7

56.25%

50%

62.5%

 


The docking simulation between AMG-510 with KRAS proteins gave a result that affinity score of AMG-510 toward KRAS G12C was lower (-8.3 kcal/mol) than its affinity score toward KRAS G12D and KRAS WT (-6.2 kcal/mol) (Table 4). This result implied that AMG-510 bound stronger to KRAS G12C than other KRAS variants. This stronger binding may also a factor that cause the inhibition effect of AMG-510 only works on KRAS G12C but not on other KRAS variants60. Based on this result, we considered that a natural compound which had affinity score lower than -6.2kcal/mol was a potential compound having inhibition effect on KRAS. When we docked the filtered natural compounds to all KRAS variants, the docking result exhibited phaseollin as natural compound having the most excellent affinity score toward KRAS G12C (-8.7kcal/mol) and KRAS G12D (-7.5kcal/mol) (Table 4). This result suggested that phaseollin might give inhibitory effect on KRAS G12D because its affinity score toward KRAS G12D was lower than AMG-510. Additionaly, affinity score of phaseollin toward KRAS WT was identic with affinity score of AMG-510 (-6.2kcal/mol) (Table 4). This result indicated that the inhibition effect of phaseollin might only work on KRAS mutants but not on KRAS WT. Furthermore, phaseollin was also found to have highest total interaction with important residues in SII-G of KRAS G12D and KRAS WT. Although phaseollin had equal total interaction with important residues in SII-G of KRAS G12D and KRAS WT (Table 4), phaseollin presented different interaction profile in SII-G of KRAS G12D and KRAS WT. The difference can be obviously seen from its hydrogen interaction with D12 in the KRAS G12D, which was not found with G12 in the KRAS WT (Table 5). The ability of phaseollin to interact with D12 in the KRAS G12D may affect its stronger binding on KRAS G12D54, this hypothesis is supported by its lower affinity score on KRAS G12D than KRAS WT (Table 4). Interestingly, phaseollin was also found to only interact with D12 in the KRAS G12D but not with C12 in the KRAS G12C and G12 in the KRAS WT (Table 4). This interaction profile was similar to AMG-510 that only formed interaction with C12 in the KRAS G12C but not with D12 in the KRAS G12D and G12 in the KRAS WT (Table 5). Specific interaction on mutation point in the KRAS mutant may result in the specific inhibition effect on KRAS mutant 60.


 

Figure 5: 3D and 2D visualizations of AMG-510 in SII-G of KRAS G12D in active state

 

Figure 6: 3D and 2D visualizations of phaseollin in SII-G of KRAS G12D in active state

 

Table 5: Interaction profile between ligands and residues in SII-P/SII-G of KRAS variants

Compounds

Covalent Bonds

Hydrogen Bonds

Hydrophobic Bonds

 

KRAS G12C

KRAS G12D

KRAS WT

KRAS G12C

KRAS G12D

KRAS WT

KRAS G12C

KRAS G12D

KRAS WT

AMG-510 (control)

C12

-

-

K16

 

Q61, K88, N86

P34, A59, G60, Q61, M72, R68, V103, E62, E63, Q99, H95, Y96, G10

A66, Q99, R68, E62, R102, H95

G60, Q99, Y96, D92, H95, A11

 

Phaseollin

-

-

-

-

D12, G60, K88

T58, Y71

 

E63, M72, V103, I100, Q99, Y96, H95, D92

E62, Y96, D92, R68, Q99, R102, H95, A11

R68, G60, Q99, H95, Q61, K88, D92, Y96, V9

 

Chitranone

-

-

-

-

E62, D12, Y96

G10, D92

G60, R68, T58, M72, E63, Q99, H95, Y96

Q61, G60, K88, D92, A11, H95, G10

A11, G12, Y96, R68, Q61, G60

 

Ajmalicine

-

-

-

C12

N86

-

G60, A59, K16, A11, M72, Q99, Y96, Q61

D12, A11, Y96, H95, D92, K88, E62

Y96, D92, H95, G12, P34, A59, Q61, G60, Y71

 

Cristacarpin

-

-

-

G60

D12, K88, D92, G60

Y71, T58, Y96

C12, G10, V9, T58, M72, I100, Q99, H95, Y96, Q61

H95, R68, Q99, Y96, E62, A11

D92, H95, Q99, Q61, R68, G10, V9, G60, A11

 

 


The 3D visualization of the binding poses of AMG-510 and phaseollin provided insights into why phaseollin exhibited a better affinity score and better interactions with important residues in SII-G of KRAS G12D in active state compared to AMG-510. It can be seen that AMG-510 does not adopt a proper binding pose in SII-G of KRAS G12D in active state (Figure 5), while phaseollin demonstrates a proper binding pose (Figure 6). The proper binding pose of phaseollin in SII-G of KRAS G12D in active state may contribute to its stronger binding affinity. Additionally, phaseollin formed interactions with G60, D69, D92, Y96, Q99, and D12 SII-G of KRAS G12D in active state, similar to the interaction profile of KD2, a selective KRAS G12D peptide inhibitor. This result suggests that phaseollin is potential to be developed as selective KRAS G12D inhibitor.

 

CONCLUSION:

In the docking study of AKT1, this study found morindone and porphyrin as potential compounds to be developed as AKT inhibitors. In the docking study of KRAS, this study found phaseollin was potential to be developed as KRAS G12D inhibitor. As this study relied solely on molecular docking simulations for predictions, a molecular dynamics analysis could be an avenue for forthcoming study to validate the findings.

 

CONFLICT OF INTEREST:

The authors have no conflicts of interest.

 

ACKNOWLEDGMENTS:

The first author sincerely would thank to LPDP (Indonesia Endowment Fund for Education), Ministry of finance, Republic Indonesia for providing financial support for this study. The authors are thankful to Riyanti Weni Syavitri from Biotechnology Department, School of Life Sciences and Technology, Bandung Institute of Technology, Indonesia for her contribution to provide us several data related to Indonesian medicinal plant species and the natural compounds they contain.

 

REFERENCES:

1.      WHO. Source: Globocan 2020. Globocan 2020. 2020. available on https://gco.iarc.fr/today/data/factsheets/populations/900-world-fact-sheets.pdf.

2.      Porru M. Pompili L. Caruso C. Biroccio A. Leonetti C. Targeting KRAS in metastatic colorectal cancer: Current strategies and emerging opportunities. J Exp Clin Cancer Res. 2018; 37(1). doi:10.1186/s13046-018-0719-1

3.      Zhao B. Wang L. Qiu H. et al Mechanisms of resistance to anti-EGFR therapy in colorectal cancer. Oncotarget. 2017; 8(3): 3980-4000. doi:10.18632/oncotarget.14012

4.      Li QH. Wang YZ. Tu J. et al Anti-EGFR therapy in metastatic colorectal cancer: mechanisms and potential regimens of drug resistance. Gastroenterol Rep. 2020; 8(3): 179-191. doi:10.1093/gastro/goaa026

5.      Waniczek D. Śnietura M. Lorenc Z. Nowakowska-Zajdel E. Muc-Wierzgoń M. Assessment of PI3K/AKT/PTEN signaling pathway activity in colorectal cancer using quantum dot-conjugated antibodies. Oncol Lett. 2018; 15(1): 1236-1240. doi:10.3892/ol.2017.7392

6.      Chen CC. Er TK. Liu YY. et al Computational analysis of KRAS mutations: implications for different effects on the KRAS p.G12D and p.G13D Mutations. PLoS One. 2013; 8(2): 6-13. doi:10.1371/journal.pone.0055793

7.      Saura C. Roda D. Roselló S. et al A first-in-human phase I study of the ATP-competitive AKT inhibitor Ipatasertib demonstrates Robust and safe targeting of AKT in patients with solid tumors. Cancer Discov. 2017; 7(1): 102-113. doi:10.1158/2159-8290.CD-16-0512

8.      Crabb SJ. Griffiths G. Marwood E. et al Pan-AKT inhibitor capivasertib with docetaxel and prednisolone in metastatic castration-resistant prostate cancer: A randomized, placebo-controlled phase II trial (procaid). J Clin Oncol. 2021; 39(3): 190-201. doi:10.1200/JCO.20.01576

9.      Bang YJ. Kang YK. Ng M. et al A phase II, randomised study of mFOLFOX6 with or without the Akt inhibitor ipatasertib in patients with locally advanced or metastatic gastric or gastroesophageal junction cancer. Eur J Cancer. 2019; 108: 17-24. doi:10.1016/j.ejca.2018.11.017

10.   Wang Y. Zhang H. Li J. Niu MM. Zhou Y. Qu Y. Discovery of potent and noncovalent KRASG12D inhibitors: Structure-based virtual screening and biological evaluation. Front Pharmacol. 2022; 13(December): 1-11. doi:10.3389/fphar.2022.1094887

11.   Mao Z. Xiao H. Shen P. et al KRAS(G12D) can be targeted by potent inhibitors via formation of salt bridge. Cell Discov. 2022; 8(1). doi:10.1038/s41421-021-00368-w

12.   Shaik BB. Katari NK. Jonnalagadda SB. Role of natural products in developing novel anticancer agents: a perspective. Chem Biodivers. 2022; 19(11). doi:10.1002/cbdv.202200535

13.   Ragunathan A. Ravi L. Krishna K. Cytotoxic potential of 4-hydroxypentan-2-oneextracted from Jacaranda mimosifolia on colorectal cancer cells. Research J. Pharm. and Tech. 2018; 11(6): 2251-2257. doi:10.5958/0974-360X.2018.00417.1

14.   Purushothaman B. Suganthi N. Jothi A. Shanmugam K. Molecular Docking Studies of potential anticancer agents from Ocimum basilicum L. against human colorectal cancer regulating genes: an insilico approach. Research J. Pharm. and Tech. 2019; 12(7): 3423-3427. doi:10.5958/0974-360X.2019.00579.1

15.   Drapak I, Suleiman M, Protopopov M, Yeromina H, Sych I, Ieromina Z, Sych I PL. The use of the docking studies with the purpose of searching potential antihypertensive drugs. Research J. Pharm. and Tech. 2019; 12(10): 4889-4894. doi:10.5958/0974-360X.2019.00846.1

16.   Kodical DD. James JP. K D. Kumar P. Cyriac C. K.V G. ADMET, Molecular docking studies and binding energy calculations of pyrimidine-2-thiol derivatives as Cox inhibitors. Research. J. Pharm. and Tech. 2020; 13(9): 4200-4206. doi:10.5958/0974-360X.2020.00742.8

17.   Daina A. Michielin O. Zoete V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017; 7. doi:10.1038/srep42717

18.   Susmi MS. Kumar RS. Sreelakshmi V. Menon SV. et al A computational approach for identification of phytochemicals for targeting and optimizing the inhibitors of heat shock proteins. Research J. Pharm. and Tech. 2015; 8(9): 1199-1204. doi:10.5958/0974-360X.2015.00219.X

19.   Qais FA. Alomar SY. Imran MA. Hashmi MA. In-silico analysis of phytocompounds of Olea europaea as potential anti-cancer agents to target pkm2 protein. Molecules. 2022; 27(18). doi:10.3390/molecules27185793

20.   Renantha RR. Liga AR. Tanugroho CB. Denovian LX. Budiyanto SLAZ. Parikesit AA. Flavonoids as potential inhibitors of dengue virus 2 (DENV2) envelope protein. J. Pharm. Pharmacogn Res. 2022; 10(4): 660-675. doi:10.56499/jppres22.1375_10.4.660

21.   Pradeep P. Rengaswamy D. In silico molecular docking studies of quercetin compound against anti-inflammatory and anticancer proteins. Research J. Pharm. and Tech. 2019; 12(11): 5305-5309. doi:10.5958/0974-360X.2019.00919.3

22.   Shanmugapriya E. Velayutham R. Muthukumar VA. Molecular docking studies on naturally occurring selected flavones against protease enzyme of Dengue virus. Research J. Pharm. and Tech. 2016; 9(7): 929-932. doi:10.5958/0974-360X.2016.00178.5

23.   Joosten RP. Long F. Murshudov GN. Perrakis A. The PDB-REDO server for macromolecular structure model optimization. IUCrJ. 2014; 1: 213-220. doi:10.1107/S2052252514009324

24.   Yu J. Zhou Y. Tanaka I. Yao M. Roll: A new algorithm for the detection of protein pockets and cavities with a rolling probe sphere. Bioinformatics. 2009; 26(1): 46-52. doi:10.1093/bioinformatics/btp599

25.   Tallei TE. Tumilaar SG. Niode NJ. et al Potential of plant bioactive compounds as sars-cov-2 main protease (mpro) and spike (s) glycoprotein inhibitors: a molecular docking study. Scientifica (Cairo). 2020; 2020. doi:10.1155/2020/6307457

26.   Cosconati S. Forli S. Perryman AL. Harris R. Goodsell DS. Olson AJ. Virtual screening with autodock: theory and practice. Expert Opin Drug Discov. 2010; 5(6): 597-607. doi:10.1517/17460441.2010.484460

27.   Trott O. Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010; 31(2): 455-461. doi:10.1002/jcc.21334

28.   Laskowski RA. Swindells MB. LigPlot+: Multiple ligand-protein interaction diagrams for drug discovery. J Chem Inf Model. 2011; 51(10): 2778-2786. doi:10.1021/ci200227u

29.   LigPlot+ Operating Manual. 2021. available on https://www.ebi.ac.uk/thornton-srv/software/LigPlus/manual2/manual.html.

30.   Pettersen EF. Goddard TD. Huang CC. et al UCSF Chimera - A visualization system for exploratory research and analysis. J Comput Chem. 2004; 25(13): 1605-1612. doi:10.1002/jcc.20084

31.   Chinchole P. Wankhede SB. Comparative in silico drug likeness and in vitro study of some Schiff’s bases as potent COX-II Inhibitors. Research J Pharm and Tech. 2019; 12(10): 4973-4980. doi:10.5958/0974-360X.2019.00862.X

32.   Abbas J. Anticancer Evaluation of Plants from Indonesian Tropical Rain Forests. Indones J Cancer Chemoprevention. Published online 2011.

33.   Anggreini N. Saputri RD. Tjahjandarie TS. Tanjung M. Aktivitas antikanker senyawa pterokarpan. Pharm Med J. 2018; 1(1).

34.   Suryowati T. Damanik R. Bintang M. Handharyani E. Identifikasi komponen kimia dan aktivitas antioksidan dalam tanaman torbangun (Coleus amboinicus Lour). J Gizi Pangan. 2015; 10(3): 217-224. doi:10.25182/jgp.2015.10.3.

35.   Sutiono DR. Mario M. Reinald G. Liani O. Susanto S. Native indonesian herbs : challenges in the future for anti-cancer drugs. Cdk. 2017; 44(11): 822-826.

36.   Tanjung M. Tjahjandarie TS. Saputri RD. Kurnia BD. Rachman MF. Syah YM. Calotetrapterins A-C, three new pyranoxanthones and their cytotoxicity from the stem bark of Calophyllum tetrapterum Miq. Nat Prod Res. 2021; 35(3): 407-412. doi:10.1080/14786419.2019.1634714

37.   Asep S. Hening H. Gema SP. Gigih S. Widya MC. Sahidin. Anticancer activity of jatrophone an isolated compound from Jatropha gossypifolia plant against hepatocellular cancer cell Hep G2 1886. Biomed Pharmacol J. 2017; 10(2): 667-673. doi:10.13005/bpj/1154

38.   Bahtiar A. Vichitphan K. Han J. Leguminous plants in the Indonesian archipelago: traditional uses and secondary metabolites. Nat Prod Commun. 2017; 12(3): 461-472. doi:10.1177/1934578x1701200338

39.   Darwati I. Nurcahyanti A. Trisilawati O. Nurhayati H. Bermawie N. Wink M. Anticancer potential of kebar grass (Biophytum petersianum), an Indonesian traditional medicine. IOP Conf Ser Earth Environ Sci. 2019; 292(1). doi:10.1088/1755-1315/292/1/012063

40.   Jayani NIE. Krisnawan AH. Oktaviyanti ND. Kartini. Standardization of Phyllanthus niruri and Sonchus arvensis as components of scientific jamu. Maj Obat Tradis. 2020; 25(1): 7. doi:10.22146/mot.45955

41.   Meiyanto E. Larasati YA. The chemopreventive activity of indonesia medicinal plants targeting on hallmarks of cancer. Adv Pharm Bull. 2019; 9(2): 219-230. doi:10.15171/jcvtr.2015.24

42.   Minggarwati TS. Uji aktivitas antikanker dan identifikasi senyawa aktif dari fraksi umbi bawang sabrang (Eleutherine palmifolia (L.) Merr.) Terhadap sel kanker serviks HeLa. Universitas Islam Negeri Maulana Malik Ibrahim; 2017.

43.   Pratiwi RA. Nurlaeni Y. Screening of plant collection of cibodas botanic gardens, indonesia with anticancer properties. Biodiversitas. 2020; 21(11): 5186-5229. doi:10.13057/biodiv/d211125

44.   Restasari A. Kusrini D. Fachriyah E. Isolasi dan identifikasi fraksi teraktif dari ekstrak kloroform daun ketapang (Terminalia catappa Linn). Jurnal FMIPA Kimia UNDIP. 2009.

45.   Nauli T. Penetuan sisi aktif selulase Aspergillus niger dengan docking ligan. JKTI. 2014; 16(2): 94-100.

46.   Huck BR. Mochalkin I. Recent progress towards clinically relevant ATP-competitive Akt inhibitors. Bioorganic Med Chem Lett. 2017; 27(13): 2838-2848. doi:10.1016/j.bmcl.2017.04.090

47.   Mahajan P. Wadhwa B. Barik MR. Malik F. Nargotra A. Combining ligand‑ and structure‑based in silico methods for the identification of natural product‑based inhibitors of Akt1. Mol Divers. 2020; 24: 45-60.

48.   Bencsik JR. Xiao D. Blake JF. et al Discovery of dihydrothieno- and dihydrofuropyrimidines as potent pan Akt inhibitors. Bioorganic Med Chem Lett. 2010; 20(23): 7037-7041. doi:10.1016/j.bmcl.2010.09.112

49.   Kozakov D. Hall DR. Jehle S. et al correction for “ligand deconstruction: why some fragment binding positions are conserved and others are not,.” Proc Natl Acad Sci U S A. 2015; 112(28): E3749. doi:10.1073/pnas.1511466112

50.   Zhan W. Li D. Che J. et al Integrating docking scores, interaction profiles and molecular descriptors to improve the accuracy of molecular docking: Toward the discovery of novel Akt1 inhibitors. Eur J Med Chem. 2014; 75(March 2014): 11-20. doi:10.1016/j.ejmech.2014.01.019

51.   Urbán L. Patel VF. Vaz RJ, eds. Antitargets and Drug Safety. John Wiley & Sons, Weinheim. 2015.

52.   Chen SF. Cao Y. Chen JJ. Chen JZ. Binding selectivity studies of PKBα using molecular dynamics simulation and free energy calculations. J Mol Model. 2013; 19: 5097-5112.

53.   Lazaro G. Kostaras E. Vivanco I. Inhibitors in aktion: ATP-competitive vs allosteric. Biochem Soc Trans. 2020; 48(3): 933-943. doi:10.1042/BST20190777

54.   Zhang Z. Gao R. Hu Q. et al GTP-state-selective cyclic peptide ligands of K-Ras(G12D) block its interaction with Raf. ACS Cent Sci. 2020; 6(10): 1753-1761. doi:10.1021/acscentsci.0c00514

55.   Gentile DR. Rathinaswamy MK. Jenkins ML. et al Ras binder induces a modified switch-ii pocket in GTP and GDP states. Cell Chem Biol. 2017; 24(12): 1455-1466.e14. doi:10.1016/j.chembiol.2017.08.025

56.   Ostrem JM. Peters U. Sos ML. Wells JA. Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013; 503(7477): 548-551. doi:10.1038/nature12796

57.   Sogabe S. Kamada Y. Miwa M. et al Crystal structure of a human K-Ras G12D mutant in complex with GDP and the cyclic inhibitory peptide KRPEP-2D. ACS Med Chem Lett. 2017; 8(7): 732-736. doi:10.1021/acsmedchemlett.7b00128

58.   Buhrman G. O’Connor C. Zerbe B. et al Analysis of binding site hot spots on the surface of Ras GTPase. J Mol Biol. 2011; 413(4): 773-789. doi:10.1016/j.jmb.2011.09.011

59.   Pantsar T. The current understanding of KRAS protein structure and dynamics. Comput Struct Biotechnol J. 2020; 18: 189-198. doi:10.1016/j.csbj.2019.12.004

60.   Canon J. Rex K. Saiki AY. et al The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019; 575(7781): 217-223. doi:10.1038/s41586-019-1694-1

 

 

 

 

Received on 04.07.2023            Modified on 22.01.2024

Accepted on 26.04.2024           © RJPT All right reserved

Research J. Pharm. and Tech 2024; 17(8):3777-3785.

DOI: 10.52711/0974-360X.2024.00587