Training set phase database:
At first, a phase database was created for the training set compounds (36 inhibitors with known IC50 and pChEMbL values). Stereoisomers were produced by restricting or retaining some chirality’s while allowing other chiral centers to change. Up to one low-energy conformation was created for rings with five to six members, and a maximum of four low-energy stereoisomers were preserved. Eighty-six compounds were ultimately obtained in all. Additionally, 4300 conformations in total were received, with an average of 50 conformations per molecule.
Description of the pharmacophore:
Using 36 recognized inhibitors of the BACE-1 enzyme, the common pharmacophore hypotheses were produced using the PHASE module of the Schrodinger Suite. Owing to the qualitative nature of the pharmacophore modeling process a characteristic framework was utilized which further helped in the development of model(s). In order to differentiate actives from in-actives a threshold pChEMbL value of 7.2 was used. By applying the threshold value all the compounds in training set with pChEMbL value of ≥ 7.2 were deemed active whereas compounds having pChEMbL value of less than 7.2 were deemed in-active. The pharmacophore hypotheses obtained are shown in Table. 1, while the corresponding IC50 values for each compound in the training set are shown in Fig. 4.
Out of 36 inhibitors, 27 inhibitors were deemed active according to the set criteria of the threshold value. The inactive compounds were 3, 5, 11, 12, 18, 19, 20, 21 and 27 having the IC50 values 100nM, 90nM, 120nM, 250nM, 150nM, 90nM, 130nM, 90nM and 630nM respectively. All the structures of training set molecules are shown in Fig. 6.
Table 1
shows the pharmacophore hypotheses identified by using PHASE module and their corresponding perfromance measuring metrices
Hypothesis
|
PhaseHypoScore
|
Survival Score
|
BEDROC Score
|
Site Score
|
Vector Score
|
AHRRR
|
1.328
|
6.253
|
0.953
|
0.807
|
0.961
|
HHRRR
|
1.237
|
6.275
|
0.861
|
0.760
|
0.923
|
AHHRRR
|
1.265
|
6.739
|
0.861
|
0.679
|
0.957
|
Based on the parameters mentioned in Table.1 for each of the obtained hypothesis, the hypothesis AHRRR was chosen as the best hypothesis among the bunch. Although the survival rate of the chosen hypothesis is less than the other, the PhaseHypoScore (1.328) is the highest for the chosen hypothesis.
PhaseHypoScore provides a cumulative indication on the performance of the model [53]. The intra pharmacophoric feature distances and angles are shown in Table.2 and Table.3 respectively.
Table 2
Inter-feature site distance measurements of pharmacophore model AHRRR
Site 1
|
Site 2
|
Distance in A°
|
Site 1
|
Site 2
|
Distance in A°
|
A3
|
H7
|
2.99
|
H7
|
R12
|
7.68
|
H7
|
R11
|
5.85
|
H7
|
R10
|
5.85
|
A3
|
R11
|
4.09
|
A3
|
R12
|
7.28
|
A3
|
R10
|
4.09
|
R10
|
R12
|
8.67
|
R11
|
R12
|
4.27
|
R10
|
R11
|
4.71
|
Table 3
represents the inter-feature angle measurements of pharmacophore model AHRRR
Site 1
|
Reference
|
Site 2
|
Angle (°)
|
Site 1
|
Reference
|
Site 2
|
Angle (°)
|
H7
|
R12
|
A3
|
22.8
|
R12
|
H7
|
A3
|
71.0
|
R12
|
A3
|
H7
|
86.2
|
R11
|
A3
|
R10
|
70.3
|
A3
|
R10
|
R11
|
54.9
|
A3
|
R11
|
R10
|
54.9
|
A3
|
R11
|
R12
|
121.0
|
R12
|
A3
|
R11
|
30.2
|
A3
|
R12
|
R11
|
28.8
|
R12
|
R11
|
R10
|
149.6
|
R10
|
R12
|
R11
|
15.9
|
R11
|
R10
|
R12
|
14.4
|
H7
|
A3
|
R11
|
110.4
|
R11
|
H7
|
A3
|
41.0
|
A3
|
R11
|
H7
|
28.6
|
R10
|
A3
|
H7
|
110.3
|
R10
|
H7
|
A3
|
41.0
|
A3
|
R10
|
H7
|
28.6
|
A total of 5 features were identified in the chosen common pharmacophore hypothesis. The hypothesis contained one hydrogen bond acceptor, one hydrophobic, and three aromatic ring characteristics. After alignment of all the active ligands, it was revealed that all the active ligands possess all the 5 pharmacophoric features present in the hypothesis. The aligned active ligands with the hypothesis are shown in Fig. 5.
Pharmacophore modeling Validation:
A validation procedure is essential before implementing one or more pharmacophore models for use in real-world applications. Pharmacophore validation can be carried out by utilizing a variety of techniques, including the goodness of hit list (GH), the construction of receiver operating characteristic (ROC) curves, Fischer's method, or other statistical analysis, which depends on screening a test set and decoy set (if necessary) to assess the model's capacity to distinguish between active and inactive molecules and provide an estimation of its caliber [54]. Four factors primarily describe a model's quality: sensitivity (the ability to identify active compounds), specificity (the ability to rule out inactive molecules), yield of actives (the proportion of true positives to hits), and enrichment factor (which links yield of actives to the screening dataset's composition) [55]. Currently, a variety of metrics, including Area Under Receiver Operating Characteristic (AU-ROC), Relative Information Efficiency (RIE), Binary Ensemble Averaged ROC (BEDROC), and others, are utilized to assess how well ranking techniques function in virtual screening investigations[56–59]. The chemical structures of all the compounds of the test set are shown in Fig. 7.
A metric term BEDROC is used to assess the effectiveness of a virtual screening technique. The approach has a perfect score of 1.0 for each of the three evaluated alpha values, which demonstrates its strong enrichment power. ROC is a common statistic for assessing how well a binary classification system is working [60]. With an area under the curve (AUC) of 1.0, the outcome demonstrates that the approach has perfect performance. RIE gauges how well the method separates active molecules from decoys. A RIE of 17.70 indicates that the strategy is highly effective. The average number of outperforming decoys is zero, indicating that none of the active compounds outperformed by any of the decoys. All 12 active compounds are in the top N% of decoys for all tested percentages (1%, 2%, 5%, 10%, and 20%), according to the findings of the count and percentage of actives in top N% of decoys tests. All 12 active compounds are present in the top N% of results for all tested percentages (1%, 2%, 5%, 10%, and 20%), according to the count and percentage of actives in the top N% of results. The approach has a high enrichment power, as evidenced by the Enrichment Factors (EF) with respect to N% sample size and with respect to N% actives recovered. The approach can detect 80 times more active chemical compounds than would be predicted by chance, for example, the EF value for 1% sample size is 80.17. The percentage of the active set that is recovered by the approach is known as FOD (Fraction of Overall Discovery). The outcome demonstrates that the approach did not overlook any of the active chemicals.
Database Screening:
In order to identify the possible lead compounds against BACE-1 that could provide the treatment option for Alzheimer’s disease, two databases were used for screening purpose. One of the databases was DrugCentral which contained FDA-approved drugs (4088) and the other one was Enamine diversity set database (50,240 compounds) which contains a large group of recently synthesized compounds which have not been tested yet. Both of the databases were first converted to Phase databases in order to produce the possible conformers of the compounds. The procedure for the generation of Phase database was the same as mentioned earlier. The molecules were matched with at least 4 out of 5 pharmacophoric features of the pharmacophore hypothesis (AHRRR). Only top 3 hits from each database were chosen for further analysis. The top hits were chosen on the basis of PhaseScreenScore or FitnessScore that are mentioned in Table 4. Anileridine, Umifenovir and Doxapram were top 3 hits obtained as a result of screening of DrugCentral database and have been assigned codes A1, A2 and A3 respectively. Moreover, from enamine diversity set three molecules namely N-cyclopropyl-2-(4-(2-methylthiazol-4-yl)-1H-1,2,3-triazol-1-yl)-2-phenylacetamide, N-cyclopropyl-2-(4-(6-methylpyridin-2-yl)-1H-1,2,3-triazol-1-yl)-2-phenylacetamide and 5-(2-ethoxyphenyl)-3-(pyridin-4-yl)-1,2,4-oxadiazole and were assigned the codes B1, B2 and B3 respectively.
Anileridine was found to have the highest PhaseScreenScore of 2.077 and 5-(2-ethoxyphenyl)-3-(pyridin-4-yl)-1,2,4-oxadiazole having the lowest PhaseScreenScore of 1.951. These molecules were further subjected to further analysis to determine the inhibitory potential against human BACE-1 enzyme. The chemical structures of the Hit molecules are shown in Fig. 8 and the superimposed images of the Hit molecules with the pharmacophore hypothesis are shown in Fig. 9.
Table 4
represents top 3 hits from DrugCentral and Enamine diversity set along with their PhaseScreenScores for each hit molecule.
Database
|
Top 3 HITs
|
HIT Code
|
PhaseScreenScore
|
DrugCentral
|
Anileridine
|
A1
|
2.077
|
Umifenovir
|
A2
|
2.042
|
Doxapram
|
A3
|
2.004
|
Enamine Diversity Set
|
N-cyclopropyl-2-(4-(2-methylthiazol-4-yl)-1H-1,2,3-triazol-1-yl)-2-phenylacetamide
|
B1
|
1.983
|
N-cyclopropyl-2-(4-(6-methylpyridin-2-yl)-1H-1,2,3-triazol-1-yl)-2-phenylacetamide
|
B2
|
1.981
|
5-(2-ethoxyphenyl)-3-(pyridin-4-yl)-1,2,4-oxadiazole
|
B3
|
1.951
|
Molecular Docking using GLIDE:
The docking of both reference BACE-1 inhibitors and screened Hit molecules was done against human BACE-1 and the X-ray crystal structure of human BACE-1 (PDB ID: 7MYI) was downloaded from Protein data bank having a resolution of 1.25 A°[24]. The reference inhibitors 37, 44, 43, 47, 40, 50, 39, 51, 46 and 42 (taken from test set) were docked in the active site of the protein. Moreover, the co-crystallized ligand (compound 6) with 7MYI was also re-docked and evaluated for interactions. The important residues of the active site of the proteins have already been mentioned earlier and out of all those residues the most important ones are Asp32 and Asp228 owing to the fact that they are involved in the normal catalytic process of the enzyme. The extra precision docking score and glide docking energies for all 10 reference inhibitors and screened hit molecules are all mentioned in Table 5.
Table 5
Docking and MM-GBSA results of reference inhibitors and Screened Hits against BACE-1
Docking and MM-GBSA results of reference inhibitors of BACE-1
|
Ligands
|
XP Glide Score (Kcal/mol)
|
Glide Energy (Kcal/mol)
|
MM-GBSA dG Bind (Kcal/mol)
|
Compound 6 (Co-Crystallized ligand of 7MYI Re-docked)
|
-11.009
|
-85.105
|
-77.04
|
37
|
-9.077
|
-73.049
|
-65.03
|
44
|
-9.041
|
-63.094
|
-66.61
|
43
|
-9.038
|
-73.369
|
-60.42
|
47
|
-8.702
|
-68.335
|
-70.47
|
40
|
-8.553
|
-64.308
|
-67.60
|
50
|
-8.459
|
-65.987
|
-64.66
|
39
|
-8.371
|
-62.092
|
-63.65
|
51
|
-8.215
|
-65.734
|
-47.17
|
46
|
-6.071
|
-63.093
|
-17.71
|
42
|
-5.691
|
-75.213
|
-29.70
|
Docking and MM-GBSA results of screened Hits against BACE-1
|
Anileridine (A1)
|
-6.914
|
-54.441
|
-49.14
|
Umifenovir (A2)
|
-6.144
|
-71.394
|
-39.14
|
Doxapram (A3)
|
-6.877
|
-49.927
|
-28.77
|
B1
|
-5.170
|
-62.102
|
-33.08
|
B2
|
-5.856
|
-60.838
|
-29.77
|
B3
|
-5.468
|
-42.641
|
-41.84
|
Interaction analysis of screened Hit compounds
The ligand interaction diagram (LID) of Schrodinger Suite was used to analyze the interaction pattern of reference inhibitors as well as screened hit compounds. The type of interaction, interacting atoms of protein-ligand complex and interaction distances for reference inhibitors and screened hits are shown in Tables 6 and 7 respectively. All of the reference inhibitors showed interaction with the residues of the catalytic dyad i.e. Asp32 and Asp228 either by a hydrogen bond or a salt bridge. Figure 10 shows the interaction of compound 6 (co-crystallized ligand of 7MYI when re-docked).
Moreover, there were Pi-Pi stacking and Pi-Cation interaction present in most of the reference inhibitors which also help in providing strength to the ligand-protein complex and ultimately provide stability.
Table 6
shows Interaction analysis of referenced BACE-1 inhibitors against BACE-1
Sr. No
|
Ligands
|
Type of interaction
|
Interacting atoms of protein-ligand complex
|
Distances Å
|
1.
|
Compound 6
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.17
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
1.83
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
1.83
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.85
|
Pi-cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.27
|
Hydrogen bond
|
Trp76 …. Lig (N of pyrimidine ring via water bridge)
|
1.85 with H2O molecule + 1.93 with ligand
|
7.
|
37
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.29
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
1.81
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.07
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.96
|
Pi-Cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.41
|
12.
|
44
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.02
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
1.95
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.96
|
15.
|
43
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.14
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.21
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.06
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.99
|
Pi-Cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.58
|
20.
|
47
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.09
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.07
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.97
|
Pi-Cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.59
|
24.
|
40
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.01
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.04
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
1.95
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.96
|
28.
|
50
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.00
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.04
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
1.95
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.96
|
32.
|
39
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.10
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.11
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
1.97
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.96
|
Pi-Cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.58
|
37.
|
51
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
1.89
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.00
|
Hydrogen Bond
|
Asp32 …. Lig (N+H of imidazole ring)
|
2.07
|
Salt Bridge
|
Asp32 …. Lig (N+H of imidazole ring)
|
3.01
|
Pi-Cation
|
Tyr71 …. Lig (N+H of imidazole ring)
|
6.49
|
42.
|
46
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.50
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.36
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
2.79
|
Hydrogen Bond
|
Trp76 …. Lig (C)
|
1.91
|
46.
|
42
|
Hydrogen Bond
|
Asp228 …. Lig (N of NH2)
|
2.28
|
Hydrogen Bond
|
Asp32 …. Lig (N of NH2)
|
1.92
|
However, in case of screened hit compound only Anileridine (A1) showed interaction with one of the residues of the catalytic dyad i.e. with Asp32 only. However, there were other amino acid residues which are involved in the binding interaction and the most prominent residues were Trp76 and Tyr71. The ligand atoms showed pi-pi stacking and pi-cation interaction and with a comparatively shorter distance when compared with reference inhibitors which could indicate a stronger binding with these residues. The most prominent docking score, docking energy score and ligand protein interactions were observed in Anileridine while B2 showed the least amount of residue and ligand interaction with only Trp76 involved in making one hydrogen bond with the ligand molecule. The ligand interaction diagrams for all the screened hits are shown in Fig. 11. The involvement of Tyr71 and Trp76 in binding causes the conformation of BACE-1 enzyme to close which further contribute towards a stronger inhibition potential of the compounds[27].
The binding docking score and glide energy of B1 was the least prominent among the screened hits. The reference ligand 42 also showed the least significant score of -5.691kcal.mol and Anileridine showed a docking score of -6.914kcal.mol which is better when compared with ligand 42 and 46 both of which have shown inhibitory activity towards BACE-1 enzyme in in-vitro assays and possess a known IC50 values.
Table 7
shows Interaction analysis of screened Hit molecules against BACE-1
Sr. No
|
Ligands
|
Type of interaction
|
Interacting atoms if ligand-protein complex
|
Distances Å
|
1.
|
Anileridine (A1)
|
Salt Bridge
|
Asp32 …. Lig (N of piperidine ring)
|
4.82
|
Pi-cation
|
Tyr71 …. Lig (N of piperidine ring)
|
4.87
|
Hydrogen bond
|
Phe108 …. Lig (N of NH2)
|
1.78
|
Hydrogen bond
|
Lys107 …. Lig (N of NH2)
|
2.09
|
5.
|
Umifenovir (A2)
|
Hydrogen bond
|
Trp76 …. Lig (O)
|
2.34
|
Hydrogen bond
|
Gly34 …. Lig (O)
|
2.16
|
Pi-Pi Stacking
|
Tyr71 ….Lig (Pyrole ring)
|
4.23
|
Hydrogen bond
|
Tyr198 …. Lig (O)
|
2.10
|
9.
|
Doxapram (A3)
|
Hydrogen Bond
|
Gly230 …. Lig (N of piperidine ring)
|
1.90
|
Pi-Pi Stacking
|
Tyr71 …. Lig (benzene ring)
|
4.48
|
11.
|
B1
|
Hydrogen Bond
|
Trp76 …. Lig (triazole ring)
|
2.69
|
12.
|
B2
|
Hydrogen bond
|
Trp76 …. Lig (N of triazole ring)
|
2.41
|
Pi-Pi Stacking
|
Tyr71 …. Lig (triazole ring)
|
3.96
|
14.
|
B3
|
Hydrogen bond
|
Trp76 …. Lig (O)
|
1.97
|
Pi-Pi Stacking
|
Tyr71 …. Lig (benzene ring)
|
4.35
|
Molecular Mechanics - General Born Surface Area (MM-GBSA) Analysis:
The MM-GBSA values for referenced inhibitors and the screened hits are shown in Table 5. The range of MM-GBSA for referenced inhibitors was between − 17.71kcal/mol and − 70.47kcal/mol. Out of all the screened hits anileridine (A1) had the binding free energy of -49.14kcal/mol which was considered the best among the screened hits. Obtained results suggest the formation of a stable ligand-protein complex between anileridine and BACE-1. Hereby, in view of this argument we selected anileridine for further analysis and was subjected to molecular dynamic simulation and in-silico ADMET profiling.
Molecular Dynamic Simulations analysis:
MD simulation is used to analyzed and assess the stability and dynamics profile of the docked protein-ligand complex. For a simulation time of 50ns, the docked complex of anileridine-BACE-1 was subjected to MD simulation. According to Fig. 12, The fact that the Root mean square deviations (RMSD) followed somewhat comparable tracks during the latter part of the first half (15-30ns) and early second half (30-45ns) of the simulation indicates that the entire system was well equilibrated. In contrast, the complex form's trajectory completely converged with the Apo form (no ligand) between 35ns and 40ns. Moreover, the trajectory of the complex for also converged to some extent with the Apo form 15ns to 25ns during the simulation. The RMSD for the complex and Apo forms, respectively, ranged from 0.25 to 1.0Å and 1.0 to 1.75Å during the first half of the simulation. The RMSD maintained substantially below 0.1 throughout the second half of the simulation. While the RMSD of the complex fluctuated between 0.25 and 2.0Å, that of the Apo form fluctuated between 1.75 and 2.0Å. From 35ns-40ns the complex converged with the Apo form completely and from 15ns-25ns the complex converged with the Apo form slightly to some extent. Altogether, these results showed a stable binding interaction between anileridine and BACE-1 protein upon formation of substantial interactions with key amino acids of the binding pocket.
The ligand-protein interactions were monitored throughout the time of the simulation. Protein ligand interactions were categorized into 4 major types namely: hydrogen bonds, ionic interactions, water bridges and hydrophobic interactions and are shown in Figs. 13 and 14. Throughout the trajectory, the stacked bar charts (Fig. 11) were normalized, for example a value of 0.8 is an indication that for 80% of the simulation time the specific interactions were maintained. In accordance with the stacked bar charts, the interaction fraction of ligand with Asp32 is approximately 0.8 and accompanied by 3 types of interactions. For an interaction fraction of 0.15 there were hydrogen bonds afterwards from 0.15–0.78 there were ionic interactions and finally for a very brief period of time there were water bridges. The interaction fraction of ligand with Tyr71 was well above 1.2 whereas the interaction fraction with Asp228 was around 0.2 with only ionic interaction and water bridges. The interaction fraction with Phe108 was found to be 0.7 with mostly hydrophobic interaction. Finally, the interaction fraction with Gly74 was approximately 0.6 with only hydrogen bonding as the interaction force. Other amino acids in the stacked bar charts showed minor interactions for a very short period of time.
For ligand binding to occur, hydrogen bonds are crucial. Because they have such a significant impact on drug specificity, metabolism, and adsorption, hydrogen-bonding qualities should be taken into account while developing novel drugs. Four further subtypes of hydrogen bonds can be distinguished between a protein and its ligand: backbone acceptor, backbone donor, side-chain acceptor, and side-chain donor [53].
Figure 15 shows the total time of simulation on x-axis and amino acid interaction with the ligand on y-axis. The figure shown the time of interaction of each amino acid involved throughout the time of simulation (50ns). According to this, Asp32 shows the one of the strongest interaction with the ligand. Asp32 showed continuous interaction with only minor break throughout the simulation making the interaction stable. On the other hand, Asp228, another important amino acid of the catalytic dyad as mentioned before showed interaction but for only about 12-13ns which was not shown during molecular docking. Other important and strong, continuous interactions were shown by Tyr71, Phe108 and Gly74. Gly74 started showing interaction in the late part of the first half of simulation but the interaction remained strong afterwards till the end of simulation with only minor breaks. Tyr71 showed interaction even stronger and with less breaking point than Asp32 and as mentioned before interaction of ligand with Tyr71 caused the conformation of protein to close making the inhibition even stronger. Similarly, Phe108 showed significant strong interaction with minor breaks throughout the simulation time. Moreover, Arg235 did not show any interaction in molecular docking but in MD simulations since the environment is dynamic and it also has water molecules Arg235 forms a water bridge and then a contact with the ligand atom which is also evident in Fig. 14 as well.
Figure 16 shows the torsional profile of Anileridine obtained via MD simulations. There are a total of 8 rotatable bonds in anileridine and are color coded with different colors. There are two types of plots in this figure, the bar plot and the radial plot and explain the probability density of the torsion and conformation of torsion respectively throughout the course of simulation (0-50ns). The bar and dial plots of the rotatable bond between benzene ring and piperidine ring (colored in blue) show that this bond is rotating on both x-axis and y-axis with a significant degree of freedom (almost a complete 180° rotation in both positive and negative x-axis. However, the bar and dial plots of a bond between amine and benzene ring (colored in dark orange) show that the rotation of bond is predominantly towards 90° of the positive x-axis.
Moreover, the rotatable bond between nitrogen of piperidine and -CH2 (colored in purple) shows a rotation of 90° towards negative x-axis. Similarly, the rotatable bond between the carbonyl carbon and oxygen atom (colored in pink) shows a specific rotation of 180° in both positive and negative x-axis. On a similar note, other rotatable bonds are also color coded and are their torsional profile is shown in both dial (radial) and bar plots.
ADMET Profile: Anileridine
Predictive Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) studies are the largest area of interest in drug discovery and development. Utilizing enormous databases of ADMET data associated with structures, the objective is to create computer models that link structural changes with changes in response. These models can be used to create and predict molecules with superior properties. Additionally, these databases enable users to estimate human ADMET features by extrapolating human in vitro and animal in vivo ADMET results [61]. In order to aid researchers in creating efficient dosage forms for currently existing molecules, these databases or software also offer helpful information on those molecules. For this investigation, numerous parameters concerning Anileridine (PubChem ID: 8944) were predicted using the cloud version of ADMET Predictor (TM) version 10.4.0.5, 64-bit edition.
1. Predictive Physicochemical (Absorption and Distribution) Profile:
Physicochemical properties of drug molecules play an important role in transforming a new molecular entity into a suitable dosage form. It is necessary for a drug molecule to exert its optimal activity to have good solubility, permeability, dissolution rate and pre-systemic metabolism [62]. Since then, while extensive evaluations have investigated the relationship with metabolic stability and toxicity, physicochemical properties of a molecule that are consistent with the likelihood of good oral absorption have been further developed. Thorough analyses of the physicochemical aspects of drug-target interactions have supported these conclusions. The distinctive characteristic of best-in-class pharmaceuticals, according to thermodynamic profiling, is a reliance on enthalpy, which depends on lipophilicity, to drive binding energetics [63].
The predictive ADMET profile was calculated at 7.4 pH value. The values for MLogP and Volume of distribution (Vd) indicate that the drug is hydrophobic in nature and the blood brain barrier (BBB) filter value is 99% which is an indication of drug’s ability to cross BBB. In case of Alzheimer’s disease it is imperative for the drug molecule to cross the BBB and Anileridine possesses the ability to a significant level of confidence. MLogP is one of the simplest methods developed by Moriguchi for estimating the LogP value of the drug molecules by taking structural features of the drug into consideration [64]. The BCS class of anileridine is shown in Fig. 17.
The detailed physicochemical profile of Anileridine is given in Table 8.
Table 8
shows the important physicochemical parameters and their predicted values by the model.
Sr.#
|
Parameter
|
Predictive value
|
Model used and explanation
|
1.
|
MlogP
|
2.96
|
Moriguchi estimation of log P [64].
|
2.
|
Permeation Cornea
|
180.523
|
Permeability through the rabbit cornea (cm/s x 10^7).
|
3.
|
S + logP
|
3.467
|
Simulations Plus model of log P.
|
4.
|
S + Acidic_pKa
|
None
|
Macroscopic predictions of the pKa values appear to be governed by the predominant acidic groups
|
5.
|
S + Basic_pKa
|
8.47; 4.25
|
Macroscopic predictions of the pKa values appear to be governed by the predominant basic groups
|
6.
|
Solution Factor
|
224.578
|
Universal salt solubility factor based on S + Sw model.
|
7.
|
Vd
|
3.477
|
Volume of distribution (L/kg) in human at steady state.
|
8.
|
Water Solubility (S + Sw)
|
0.15
|
Water solubility in mg/mL of the given compound predicted by the model on the basis of chemical features
|
9.
|
Diffusion coefficient
|
0.646
|
Water diffusion coefficient of nonelectrolytes at infinite dilution by Hayduk-Laudie (cm2/s x 105).
|
10.
|
S + MDCK-LE permeability assay
|
High (99%)
|
MDCK permeability classification model (low or high) built on Varma et al. data for ECCS.
|
11.
|
S + logD
|
2.367
|
log D, at 7.4 pH, based on S + logP.
|
12.
|
BBB Filter
|
Yes (99%), High
|
Determines whether a substance will be able to cross the blood–brain barrier.
|
13.
|
Permeation Skin
|
5.056
|
Permeability through human skin (cm/s x 10^7).
|
14.
|
Simulated metabolic clearance (S + CL_Metabolism)
|
Yes (99%)
|
Predicts whether clearance mechanism is metabolic.
|
15.
|
Simulated Renal Clearance (S + CL_Renal)
|
No (99%)
|
Predicts whether clearance mechanism is renal.
|
16.
|
Simulated human jejunal permeability (S + Peff)
|
2.161
|
Effective human jejunal permeability (cm/s x 10^4).
|
17.
|
hum_fup%
|
13.642
|
Percent UNBOUND to blood plasma proteins in human.
|
18.
|
ECCS_Class
|
Class 2
|
Major clearing mechanisms of drug and drug like molecules that are described in the ECCS class. based on the article of Varma et al. 1A = metabolism, 1B = hepatic uptake, 2 = metabolism, 3A = renal, 3B = either renal or hepatic uptake, and 4 = renal [65]
|
The model showed that the drug has a significant clearance via metabolic pathway and almost no renal clearance in unchanged form. Moreover, the drug has an acceptable jejunal permeability profile along with very good cornea permeation and skin permeation profile. According to the model the drug belongs to Class 2 of Extended Clearance Classification System (ECCS) which indicates that the major route of clearance of Anileridine is via metabolic route.
1.1. Solubility profile in relation to pH:
Several factors such as drug solubility, permeability, first pass metabolism and dissolution rate etc. may influence the ultimate oral absorption of the drug molecules and out of these parameters usually the poor solubility of the drug is contributed to poor oral absorption [66]. The model predicted the solubility of the drug and is shown in Fig. 18. It depicts that the drug has a good solubility profile at acidic pH and has a maximum peak level at pH value of 4.5. However, by increasing the pH value towards neutral conditions the solubility of the drug decreases many folds. Moreover, at basic pH the solubility is almost near to zero.
1.2. pKa Microstates:
The pka macro-state values display the drug's maximal concentration at a given pKa value. The concentration of hydrogen ions (pH) at which 50% of the drug resides in its ionized hydrophilic form is known as the drug's pKa (i.e., the pH at which it is in balance with its unionized lipophilic form). All local anesthetics are known to have a basic moiety in their structures and are considered to be weak bases. At physiological pH, the pKa value decreases as the lipophilicity rises [67]. The value of pKa represents the acidity and basicity of a balanced aqueous solution. For the drug to absorb, the compounds inside must be electrically neutral; otherwise, the medicinal product will not be permeable enough.
According to the model the drug possesses 0 acidic atom(s) and 2 basic atom(s): 4(-NH2)3(> N-). The model was built by only micro species contributing more than 1.0% are displayed. Moreover, Aliphatic -OH and amides groups were ignored as well as carbon protonation. The drug is 100% at a pKa values of 8.47 and 4.25 either in protonated form or in neutral form. The relationship of pKa with the microstates and macro-states of the drug is shown in Fig. 19.
1.3. Log D profile:
While conducting studies to determine the LogD profile of drug molecules it is necessary to monitor and control the pH and ionic strength especially while working with ionizable compounds [68]. The predictive LogD profile of anileridine in relation to pH suggest that at low pH the drug has almost zero diffusion rate however, as the pH starts to increase the drug will start to diffuse and by the time pH reaches 10, the diffusion rate of the drug will become optimal. The data gives the indication that by controlling and manipulating the pH the higher degree diffusion of anileridine can be achieved. The relationship of increasing pH on diffusion coefficient of anileridine is shown in Fig. 20.
2. Predictive metabolic profile:
Drug metabolism investigations are crucial for a number of reasons, including the discovery of novel chemical entities based on the identification of active metabolites, limiting potential safety risks brought on by the production of reactive or toxic metabolites, ensuring potential adequate coverage of human metabolites in animals, assisting in the prediction of human doses, and comparing preclinical metabolism in animals with that of humans [69].
From CYP450 class of metabolic enzymes only CYP2C8, CYP3A4 and CYP2D6 were found to be involved in the metabolism of anileridine. The sites of metabolism of each enzyme on the drug are shown in Fig. 21.
CYP3A4 was found to be the most prominent of the bunch to be involved in metabolism of the drug molecule. The structures of predictive metabolites and percentage of each metabolite formation is shown in Fig. 20A. Besides CYP450, UDP-glucuronosyltransferase (UGT) is also involved to some extent in the metabolism of the drug molecule. UGT1A1, UGT1A4, UGT2B7 and UGT1A3 were mostly metabolizing the drug. The chemical structures of the predictive metabolites are shown in Fig. 20B. The model predicted two major metabolites, one having sugar attached to the free amino group and the second one having sugar attached to the nitrogen of the piperidine ring. According to the predicted model UGT1A1, UGT1A4, UGT2B7 were mostly producing the metabolite 1 (M1) while UGT1A3 was predominantly making the metabolite 2 (M2). The numerical values for different parameters of enzyme metabolism are given in Table 22.
Table 9
shows important parameters of enzyme metabolism and their predicted numerical values
Sr. #
|
Parameter
|
Predictive value
|
Model used and explanation
|
1.
|
CYP3A4_Clint
|
28.799
|
According to the model, it shows the predicted CYP3A4 mediated oxidation (uL/min/mg HLM protein) which also represents the pooled atom level intrinsic clearance for the said protein.
|
2.
|
CYP3A4_HLM_Clint
|
91.149
|
According to the model, it shows the predicted CYP3A4 mediated oxidation (uL/min/mg HLM protein) in human hepatic microsomes and also represents the pooled atom level intrinsic clearance for the said protein
|
3.
|
CYP3A4_HLM_Km
|
22.581
|
Km represents the Michaelis-Menten constant for the oxidation reaction in human hepatic microsomes in unbound form and is mediated by CYP3A4 at atom level. The unit of measure a µM.
|
4.
|
CYP3A4_HLM_Vmax
|
2.058
|
Vmax is the Michaelis-Menten constant (nmol/min/mg HLM Protein) corresponding to the oxidation reaction in human hepatic microsomes. The reaction is mediated by CYP3A4.
|
5.
|
Hepatic clearance (HEP_hCLint)
|
46.234
|
Intrinsic clearance in uL/min/million cells for metabolism in human hepatocytes (unbound form).
|
6.
|
HEP_mCLint
|
22.448
|
Intrinsic clearance in uL/min/million cells for metabolism in mouse hepatocytes (unbound form).
|
7.
|
HEP_rCLint
|
486.67
|
Intrinsic clearance in uL/min/million cells for metabolism in rat hepatocytes (unbound form).
|
8.
|
CYP2D6_Vmax
|
1.736
|
Vmax is the Michaelis-Menten constant (nmol/min/mg HLM Protein) corresponding to the oxidation reaction in human hepatic microsomes. The reaction is mediated by CYP2D6.
|
Predictive transport profile:
Nowadays, it is generally accepted that drug transporters play a significant role in determining how well drugs are absorbed, excreted, and, in many circumstances, how extensively they penetrate target organs. Additionally, there is a growing understanding of how changed drug transporter function, whether brought on by genetic polymorphisms, drug-drug interactions, or environmental elements like dietary components, may cause unanticipated toxicity [70].
According to the predicted model, anileridine was found to be a substrate of p-glycoprotein (63%) as well as an inhibitor of both types Organic Cation Transporter (OCT) 1 and 2. OCT 1 and OCT 2 are transport proteins that are involved in the transport of cations across the membranes of liver and kidney respectively hence if a drug is to inhibit these transport proteins then ultimately their metabolism and renal excretion would be compromised and the drug might cause toxicity. In these instances, if the drug is a known inhibitor of any of these two transport proteins then it is suggested to decrease the dose of the drug to counter the toxicity.
3. Predictive toxicity profile:
Instead of just determining how safe a medicinal entity is, the aim of toxicity testing is to uncover any potential dangerous effects that it may have. Finding out how drug compounds behave in lab animals and whether they have any potentially harmful effects on humans directly are the main objectives of toxicity testing. Additionally, they involve administering huge doses to lab animals in order to determine any potential dangers to humans who are exposed to much lower amounts [71]. Additionally, there are currently a number of in silico tools that can estimate the prospective toxicity potential of drug molecules as well as the expected toxicity profiles of previously discovered drugs. These tools may ultimately aid researchers in developing effective dosage forms and delivery systems for the already existing drugs in order to reduce their toxic effects. The important parameters are mentioned in Table 10.
Table 10
represents the important toxicity parameters of anileridine and their predicted values
Sr. #
|
Parameter
|
Predictive value
|
Model used and Explanation
|
1.
|
Bio-concentration factor
|
10.375
|
In steady state, the bio-concentration factor (also known as the concentration ratio [Cfish/Cwater]) is a partition coefficient between fish tissues and environmental water.
|
2.
|
Chromosomal Aberration
|
Nontoxic (88%)
|
Determines if the substance will cause the mutagenic chromosomal abnormalities.
|
3.
|
hERG Filter
|
Yes (75%)
|
Determines if the substance will block the hERG potassium channel and has the potential for cardiotoxicity
|
4.
|
hERG_pIC50
|
5.503
|
Human pIC_50 (mol/L) expression of affinity to the hERG potassium channel.
|
5.
|
Reproductive toxicity
|
Nontoxic (96%)
|
Qualitative assessment of developmental and reproductive toxicity.
|
6.
|
Serum ALT
|
Normal (81%)
|
Determines whether or not a molecular entity will result in a rise in SGPT enzyme levels.
|
7.
|
Serum ALP
|
Normal (92%)
|
Determines whether or not the molecular entity will raise the levels of the enzyme alkaline phosphatase.
|
8.
|
Serum AST
|
Elevated (76%)
|
Determines whether or not the molecular entity will raise the levels of the Human SGOT enzyme.
|
9.
|
Serum GGT
|
Normal (80%)
|
Determines if a molecular entity will raise the levels of the GGT enzyme.
|
10.
|
Serum LDH
|
Normal (94%)
|
Determines if a molecular entity will result in an increase in LDH enzyme levels.
|
11.
|
Rat_TD50
|
10.927
|
TD50 (mg/kg/day in oral dose) for rat carcinogenicity over a typical lifetime.
|
12.
|
PLipidosis
|
Toxic (82%)
|
Qualitative estimation of causing phospholipidosis.
|
13.
|
Estro_Filter
|
Toxic (79%)
|
Predicts whether or not the compound possesses estrogen receptor toxicity in rat.
|
14.
|
Andro_Filter
|
Toxic (68%)
|
Predicts whether or not the compound possesses androgen receptor toxicity in rat.
|
The predictive toxicity profile revealed that the drug is cardio-toxic as the hERG filter value is 75% and hERG pIC50 is 5.503 which is significantly high. Moreover, the drug shows toxicity towards bot androgen and estrogen receptors in rat. However, the drug is not hepatotoxic as it only mildly elevates serum AST levels. The drug is non-mutagenic and non-toxic to chromosomal proteins (histone) as well as shows no reproductive toxicity.
4. Simulated Pharmacokinetic profile:
For a therapeutic molecule to be properly transformed into a formulation that would ultimately deliver the best efficacy with the fewest adverse effects, pharmacokinetic characteristics are crucial. Today, a variety of in-silico methods are utilized to calculate pharmacokinetic parameters including volume of distribution, AUC, Cmax, and Cmin [72].
Using the cloud version of ADMET Predictor (TM) version 10.4.0.5, 64-bit, a Cp vs. Time curve was plotted. The modelled pharmacokinetic characteristics were predicted using an initial dose of 10mg/kg administered intravenously and per oral. The simulated PK and biopharmaceutical profile is shown in Figure 23.
The bioavailability of the given drug was virtually 100% in the first scenario since it was administered intravenously. 7.59ng/ml and 44.96ng/ml, respectively, were the Cmin and Cmax predicted values. Additionally, AUCinf was 601.85ng-h/ml and the area under the curve (AUC) was roughly 498.58ng-h/ml. According to the Cp vs. time curve, the estimated half-life (t1/2) was 9.28 hours, and the apparent volume of distribution (Vd) was 222.41 L. Anileridine has a clearance (Cl) value of 16.61 L/h and a Clp value of 16.61 L/h.
Oral IR tablets, on the other hand, had Cmin and Cmax predicted values of 6.61ng/ml and 29.58ng/ml, respectively. Additionally, the area under the curve (AUC) was around 392.16ng-h/ml, and the AUCinf value was 482.14ng-h/ml. The apparent volume of distribution (Vd) was 222.41 L, and the estimated half-life (t1/2) from the Cp vs. time curve was 9.28 hours. Anileridine's clearance (Cl) and Clp values are both 16.61 L/h. The Tmax for anileridine when taken orally was reported to be 2.57 hours.