Skip to main content
Saudi Journal of Biological Sciences logoLink to Saudi Journal of Biological Sciences
. 2022 Feb 4;29(5):3313–3325. doi: 10.1016/j.sjbs.2022.01.058

Development of antidiabetic drugs from benzamide derivatives as glucokinase activator: A computational approach

Amena Ali 1
PMCID: PMC9280248  PMID: 35844378

Abstract

Hyperglycemia is a condition known for the impairment of insulin secretion and is responsible for diabetes mellitus. Various small molecule inhibitors have been discovered as glucokinase activators. Recent studies on benzamide derivatives showed their importance in the treatment of diabetes as glucokinase activator. The present manuscript showed a computation study on benzamide derivatives to help in the production of potent glucokinase activators. In the present study, pharmacophore development, 3D-QSAR, and docking studies were performed on benzamide derivatives to find out the important features required for the development of a potential glucokinase activator. The generated pharmacophore hypothesis ADRR_1 consisted of essential features required for the activity. The resultant statistical data showed high significant values with R2 > 0.99; 0.98 for the training set and Q2 > 0.52; 0.71 for test set based on atom-based and field-based models, respectively. The potent compound 15b of the series showed a good docking score via binding with different amino acid residues such as (NH…ARG63), (SO2…ARG250, THR65), and π-π staking with (phenyl……TYR214). The virtual screening study used 3563 compounds from ZINC database and screened hit compound ZINC08974524, binds with similar amino acids as shown by compound 15b and crystal ligand with docking scores SP (-11.17 kcal/mol) and XP (-8.43 kcal/mol). Compounds were further evaluated by ADME and MMGBSA parameters. Ligands and ZINC hits showed no violation of Lipinski rules. All the screened compounds showed good synthetic accessibility. The present study may be used by researchers for the development of novel benzamide derivatives as glucokinase activator.

Keywords: Glucokinase activator, Benzamide, 3D-QSAR, Virtual screening, Pharmacophore analysis, Docking

1. Introduction

Glucokinase is a hexokinase isozyme, consist of 465 amino acids (molecular weight = 50 kD) present in pancreatic β-cells and liver (postprandial state). Glucokinase catalyzes a reaction that involve the transfer of phosphate from ATP to glucose and the generation of glucose 6-phosphate which is the first step in the direction of synthesis of glycogen and glycolysis (Matschinsky, 1996, Antoine et al., 2009). This reaction is also representing the first rate-limiting step in glucose metabolism. Glucokinase activator (GK-A) worked through two different mechanisms known as lowering the blood glucose level in the liver and increasing insulin secretion in pancreatic β-cells. Therefore, it becomes an interesting target in the present scenario to treat diabetes. Various GK-As have been synthesized, some are under clinical studies and showed promising results to lower blood glucose levels in healthy people and type-2 diabetes mellitus (T2-DM) patients. Glucokinase activator is responsible for several side effects such as hypoglycemia and testicular toxicity (American Diabetes Association, 2009, Waring et al., 2011). To eliminate these side effects, frequent dose regimens and dose titration are preferred. There are two different approaches described to treat hypoglycemic state, one is related to the designing of partial activators (Pfefferkorn et al. 2011) and the second approach is to restrict liver-selective glucose activators (Bebernitz et al., 2009, Massa et al., 2011, Pfefferkorn et al., 2012, Park, 2012, Park et al., 2013).

In the direction to treat diabetes, various benzamide derivatives have been developed as GK-As (Matschinsky and Porte, 2010, Mao et al., 2012). Park, et al. identified a novel phenylethyl benzamide GK-A (Park, 2012, Park et al., 2013). Furthermore, Park et al. synthesized a series of pyrazole benzamide derivatives as GK-A having 3-methylpyridine and 4-phenoxymethyl sulfone groups for the treatment of T2-DM. Grewal et al. synthesized a series of benzamide 3,5-disubstituted analogues and evaluated them for GK activation activity. These analogues showed considerable antihyperglycemic activity in the animal models (Grewal et al. 2019). In a study, Charaya et al. synthesized thiazole-2-yl benzamide derivatives from benzoic acid and evaluated them for GK activation activity (Charaya et al. 2018). A benzamide derivative PF-04937319 (1) is under phase-1 clinical trial for the treatment of diabetes.

graphic file with name fx1.jpg

On the basis of previously synthesized compounds (Park, 2012, Park et al., 2013), structure-based drug design was performed with pharmacophore development, 3D QSAR, and docking simulations for the determination of potent GK-A by using the PHASE module (Schrodinger). Pharmacophore development determines the important features required for the activity and can be used for 3D QSAR and virtual screening studies. Field and atom-based 3D-QSAR models were developed for the determination of statistical data results of correlation between molecules and their properties. A virtual screening study on the ZINC database generated the potent compounds as GK-A. Docking study revealed the important interactions with amino acids required for activity. On the other hand, the MMGBSA (Molecular Mechanics Generalized Born Surface Area) method was used to predict the binding free energy of the docked molecules. The current findings of the study may be utilized as a guiding tool for the development of novel and effective GK-A.

2. Methodologies

2.1. Software

The 3D-QSAR, pharmacophore modeling and docking were performed by Schrodinger module (Park et al., 2014, Park et al., 2015).

2.2. Dataset

A dataset of 43 benzamide derivatives was taken for the development of pharmacophore, 3D-QSAR, virtual screening, and docking studies (Table 1). The IC50 values taken from biological activities were converted into pIC50. Data set were divided into active for ‘higher activity’ and inactive for ‘lower activity’ compounds and the remaining were counted in the category of intermediate. The alignment on common scaffold of 43 benzamide derivatives is given in Fig. 1A. The PDB ID-3A0I was used for the docking studies. The crystal ligand of this protein used for comparison of docking outcomes are presented in Fig. 1B (Andreoli et al. 2014).

Table 1.

In vitro IC50 and PIC50 values for GK activity of pyrazole benzamide derivatives (Park, 2012, Park et al., 2013).

Inline graphic
15a-18k
Inline graphic
19a-19e
Inline graphic
13a1-22e1
C R1 R2 AP-A (IC50, µM) AP-A (pIC50) C R1 R2 AP-A (IC50, µM) AP-A (pIC50)
16a H H 0.095 7.02 16b1 graphic file with name fx5.gif 0.301 6.52
16b H F 0.238 6.62 16c1 graphic file with name fx6.gif 0.025 7.60
15a graphic file with name fx7.gif H 0.103 6.99 18a1 graphic file with name fx8.gif 0.021 7.68
15b graphic file with name fx9.gif H 0.005 8.30 18b1 graphic file with name fx10.gif 0.012 7.92
18a graphic file with name fx11.gif H 50 4.30 18c1 graphic file with name fx12.gif 0.03 7.52
18b graphic file with name fx13.gif H 0.212 6.67 18d1 graphic file with name fx14.gif 0.034 7.47
18c graphic file with name fx15.gif H 0.688 6.16 19 graphic file with name fx16.gif 0.027 7.57
18d graphic file with name fx17.gif H 2.43 5.61 21 graphic file with name fx18.gif 0.039 7.41
18e graphic file with name fx19.gif H 7.46 5.13 20a1 graphic file with name fx20.gif 0.037 7.43
18f graphic file with name fx21.gif H 50 4.30 20b1 graphic file with name fx22.gif 0.163 6.79
18 g graphic file with name fx23.gif H 0.008 8.10 20c1 graphic file with name fx24.gif 0.006 8.22
18 h graphic file with name fx25.gif H 0.033 7.48 20d1 graphic file with name fx26.gif 0.006 8.22
18i graphic file with name fx27.gif H 0.463 6.33 20e1 graphic file with name fx28.gif 0.006 8.22
18j graphic file with name fx29.gif H 0.106 6.97 20f1 graphic file with name fx30.gif 0.02 7.70
18 k graphic file with name fx31.gif H 50 4.30 20 g1 graphic file with name fx32.gif 0.039 7.41
19a CH3 0.807 6.09 20 h1 graphic file with name fx33.gif 0.009 8.05
19b graphic file with name fx34.gif 50 4.30 22a1 graphic file with name fx35.gif 0.053 7.28
19d graphic file with name fx36.gif 0.441 6.36 22b1 graphic file with name fx37.gif 0.046 7.34
19e graphic file with name fx38.gif 0.315 6.50 22c1 graphic file with name fx39.gif 0.006 8.22
13a1 graphic file with name fx40.gif 1.87 5.73 22d1 graphic file with name fx41.gif 0.007 8.15
13b1 graphic file with name fx42.gif 0.006 8.22 22e1 graphic file with name fx43.gif 0.016 7.80
16a1 graphic file with name fx44.gif 7.7 5.11

C: Compound; AP-A: Antiproliferative activity.

Fig. 1.

Fig. 1

(1A) Common scaffold alignment of 43 benzamide derivatives; (1B) Binding interactions of crystal ligand with protein 3AOI.

2.3. Preparation of ligands

The molecular structure of benzamide derivatives was converted from 2D to 3D by using LigPrep of Schrodinger software. The OPLS_2005 force field was taken for the preparation of ligands. Further, the prepared molecules were taken for 3D-QSAR and docking simulations (Schrodinger, 2017, Ali and Ali, 2021).

2.4. Pharmacophore development

The pharmacophore model was developed by using PHASE module, where all 43 ligands were aligned on common scaffold and generated conformers using the macromodel search method (Ali and Ali 2021). PHASE module consists six different pharmacophore features including hydrogen bond acceptor (HBA), hydrogen bond donor (HBD), aromatic ring, hydrophobic group, positive ionizable, and negative ionizable groups (Rajeswari et al. 2014). The maximum number of sites was fixed to 5 which further responsible for the generation of the topmost 20 different hypotheses. For the generation of these hypotheses, ligands were categorized into active, inactive, and intermediate.

The active ligand was set as for those which had pIC50 value of more than 8 whereas inactive ligands considered for those had pIC50 value less than 6.6, except these all-other ligands were set as intermediates. The final data consisted of 9 actives and 14 inactive ligands. The hypothesis was generated by using 9 actives and 1 Å box size and 2 Å site distance. The all-generated hypotheses were ranked based on different scores depicted in Table 2 (Crisan et al., 2019, Sakkiah et al., 2014). The hypothesis ADRR_1 (Fig. 2) showed the top scoring features with one HBA, one HBD, and two aromatic rings. The hypothesis determines the essential features required for the binding with receptor for a particular activity. A total of 20 pharmacophore hypotheses were developed which were ranks according to the score hypothesis (Table 2).

Table 2.

The generated pharmacophore hypotheses.

N. Hypothesis Phasehypo-S Survival-S Inactive-S Site-S Vector-S Volume-S Selectivity-S
1 DHHRR_1 1.25 5.061 1.433 0.689 0.927 0.537 2.063
2 ADRR_1 1.18 5.032 1.234 0.982 0.922 0.409 1.955
3 AHHRR_1 1.17 4.964 1.283 0.725 0.957 0.502 1.936
4 AAHHR_1 1.16 4.829 1.329 0.772 0.957 0.521 1.733
5 ADHRR_1 1.15 4.813 1.698 0.775 0.898 0.411 1.775
6 ADHHR_1 1.14 4.805 1.237 0.696 0.926 0.544 1.793
7 ADHHR_2 1.13 4.798 1.269 0.701 0.983 0.465 1.804
8 AAHHR_2 1.11 4.777 1.333 0.607 0.937 0.525 1.864
9 ADHHR_3 1.10 4.764 1.464 0.671 0.961 0.491 1.797
10 AAHRR_1 1.09 4.719 1.298 0.854 0.724 0.481 1.883
11 AADHR_1 1.09 4.713 1.778 0.985 0.951 0.480 1.451
12 HHRR_1 1.09 4.572 1.391 0.742 0.923 0.554 1.509
13 AHHR_1 1.08 4.531 1.129 0.873 0.905 0.545 1.363
14 DHRR_1 1.07 4.524 1.645 0.824 0.846 0.410 1.489
15 AHHR_2 1.07 4.508 1.616 0.740 0.900 0.537 1.485
16 DHHR_1 1.06 4.469 1.513 0.668 0.944 0.548 1.465
17 ADHR_1 1.06 4.459 1.548 0.996 0.928 0.467 1.222
18 AAHR_1 1.06 4.426 1.766 0.995 0.937 0.465 1.183
19 AHRR_1 1.00 4.394 1.683 0.762 0.886 0.409 1.383
20 AHRR_2 0.96 4.389 1.386 0.923 0.695 0.498 1.495

S: Scores

Fig. 2.

Fig. 2

Pharmacophore model (ADRR_1). Model represents various features by arrow like acceptor (A; pink-colour), donor (D: grey colour), hydrophobic and aromatic ring (R: brown colour) features.

2.5. 3D-QSAR

3D-QSAR models were developed by two techniques known as field-based and atom-based QSAR. The developed models were showed the essential parameters required for activity by correlating the structure features with biological activity. All 43 benzamide derivatives were separated into a training set with 75% and test set with 25% compounds using 4 PLS factors (Kar et al. 2010). Best models were generated by QSAR models (atom and field-based) described in Table 3. Statistics for atom-type fraction and field type fraction were summarized in Table 4, Table 5. In the atom-based model, 35 molecules were selected for the training, and 8 molecules for the test set, whereas in field-based 31 and 12 molecules were taken for the training and the test set, respectively (Table 6). The contour maps generated by atom-based and field-based models are presented in Fig. 3, Fig. 4, respectively. These maps are represented as hydrophobic, steric, donor, acceptor, and electrostatic fields (Peng et al. 2017).

Table 3.

Statistical data generated by atom based (A.B.) and field based (F.B.) models.

P.F. R2
R2 CV
Q2
SD
RMSE
Stability
P-r
F
A.B. F.B. A.B. F.B. A.B. F.B. A.B. F.B. A.B. F.B. A.B. F.B. A.B. F.B. A.B. F.B.
1 0.80 0.63 0.59 0.40 0.41 0.72 0.59 0.55 1.06 0.65 0.89 0.91 0.67 0.88 81.90 41.30
2 0.95 0.86 0.57 0.54 0.45 0.61 0.57 0.56 1.02 0.77 0.63 0.77 0.75 0.82 195.20 71.20
3 0.98 0.94 0.59 0.61 0.50 0.69 0.59 0.31 0.97 0.69 0.63 0.72 0.80 0.87 251.20 124.50
4 0.99 0.98 0.62 0.64 0.52 0.71 0.62 0.19 0.96 0.66 0.65 0.70 0.82 0.86 299.80 271.90

P.F.: PLS factor; A.B.: Atom based; F.B.: Field based

Table 4.

Statistical fraction data procured from atom based model.

Factors Hydrogen donor Hydrophobic group Positive group Electron withdrawing group Other
1 0.013 0.675 0.247 0.065 0.013
2 0.012 0.700 0.243 0.044 0.012
3 0.012 0.703 0.241 0.045 0.012

Table 5.

Statistical fraction data procured from field based model.

Factors Steric Electrostatic Hydrophobic Hydrogen Acceptor Hydrogen Donor
1 0.361 0.101 0.242 0.226 0.07
2 0.411 0.098 0.262 0.151 0.077
3 0.403 0.099 0.276 0.138 0.085

Table 6.

The IC50 value (Actual vs predicted) generated by atom-based and field-based 3D-QSAR model using PLS factor 4.

N. Ligand QSAR set (A.B.) Observed activity Predicted activity (A.B) QSAR set
(F.B)
Predicted activity (F.B)
1 15b Training 8.301 6.70 Training 8.49
2 13b1 Training 8.222 6.91 Training 8.18
3 20c1 Training 8.222 7.50 Training 8.32
4 20d1 Training 8.222 8.16 Training 8.17
5 20E_1 Test 8.222 8.16 Test 7.88
6 22c1 Training 8.222 7.29 Test 7.26
7 22d1 Training 8.155 8.12 Training 8.16
8 18g Test 8.097 7.92 Training 8.01
9 20h1 Test 8.046 7.17 Test 7.47
10 18b1 Training 7.921 7.76 Training 8.05
11 22E_1 Training 7.796 7.85 Training 7.60
12 20f1 Training 7.699 7.50 Training 7.88
13 18a1 Training 7.678 7.69 Training 7.73
14 16c1 Training 7.602 7.29 Training 7.29
15 19 Training 7.569 7.60 Test 7.76
16 18c1 Training 7.523 7.66 Test 7.43
17 18h Training 7.481 7.25 Training 7.32
18 18d1 Test 7.468 7.35 Training 7.62
19 20a1 Training 7.432 7.52 Training 7.53
20 20g1 Training 7.409 7.42 Test 7.16
21 21 Training 7.409 7.43 Training 7.23
22 22b1 Training 7.337 7.30 Training 7.16
23 22a1 Training 7.276 7.28 Training 7.41
24 16a Training 7.02 6.89 Training 6.92
25 15a Test 6.987 6.87 Training 6.69
26 18j Training 6.975 6.34 Test 6.62
27 20b1 Training 6.788 6.64 Training 6.99
28 18b Training 6.674 6.76 Test 6.24
29 16b Training 6.62 6.38 Training 6.23
30 16b1 Training 6.521 6.35 Training 6.18
31 19e Test 6.502 6.60 Training 6.48
32 19d Training 6.356 6.27 Training 6.31
33 18i Training 6.334 6.32 Training 6.33
34 18c Test 6.162 6.25 Test 5.16
35 19a Training 6.093 6.29 Training 5.99
36 13a1 Training 5.728 6.01 Training 5.86
37 18d Training 5.614 5.57 Test 6.09
38 18e Training 5.127 5.03 Test 4.65
39 16a1 Training 5.114 6.72 Training 5.23
40 18a Training 4.301 4.18 Test 5.70
41 18f Training 4.301 5.47 Training 4.45
42 18k Test 4.301 5.69 Training 4.37
43 19b Training 4.301 5.23 Training 4.44

A.B.: Atom based; F.B.: Field based

Fig. 3.

Fig. 3

3D contour maps, atom-based: (3A) EWG; (3B) HBD group; (3C) Hydrophobic group; (3D) Positive ionic group.

Fig. 4.

Fig. 4

Different field contour maps: (4A) Electrostatic (blue {favored}, red {disfavored}); (4B) HBA (red {favored}, magenta {disfavored}); (4C) HBD (purple {favored}, cyan {disfavored}); (4D) Steric (green {favored}, yellow {disfavored}).

2.6. Target protein prediction

The potent compounds of the series were selected and submitted to the Swiss-Target-Prediction tool. The said tool predicted various protein targets, in which GK-A was the most appropriate target for the selected molecules.

2.7. Docking analysis

The PDB ID-3A0I consists a three-dimensional structure of GK-A and was downloaded through protein data bank (Tagami et al. 2010). The docking study was performed to determine the binding scores between ligand and receptor by using the Glide module. The SP and XP methodologies were used for all 43 ligands with PDB ID-3A0I (Table 7). The MMGBSA based rescoring technique was used for the prediction of binding free energy calculation between ligands and receptor molecule (Table 8). However, DFT studies can give more accurate assessment of binding/docking (Van Mourik et al., 2014).

Table 7.

Docking scores generated through the Glide module.

N. Ligand D.S. (XP)
kcal/mol
D.S. (SP)
kcal/mol
N. Ligand DS (XP)
kcal/mol
DS (SP)
kcal/mol
1 18d −8.854 −12.715 23 19d −10.014 −9.62
2 19b −10.686 −12.412 24 13a1 −11.856 −9.56
3 19e −11.07 −11.731 25 13b1 −10.242 −9.519
4 19a −10.768 −11.467 26 18b −10.21 −9.396
5 16b −9.207 −11.361 27 18a −9.761 −9.059
6 16a −9.261 −11.296 28 19 −8.507 −9.03
7 15a −9.351 −11.284 29 18b1 −8.284 −9.001
8 18k −9.258 −11.149 30 18d1 −9.406 −8.769
9 15b −9.047 −11.088 31 20d1 −9.615 −8.707
10 18h −8.742 −10.866 32 22b1 −8.48 −8.701
11 18c −8.522 −10.622 33 20h1 −11.053 −8.694
12 18e −7.234 −10.615 34 16b1 −8.827 −8.653
13 20E_1 −8.191 −10.613 35 20c1 −8.548 −8.653
14 18f −9.513 −10.536 36 20g1 −8.464 −8.57
15 18g −11.172 −10.443 37 22a1 −7.801 −8.548
16 18j −7.131 −10.427 38 16a1 −12.379 −8.513
17 18i −9.313 −10.184 39 22E_1 −9.187 −8.274
18 18a1 −10.037 −9.95 40 22c1 −7.757 −7.804
19 20a1 −8.082 −9.897 41 20b1 −5.211 −7.388
20 18c1 −11.886 −9.767 42 20f1 −10.515 −6.959
21 16c1 −10.364 −9.703 43 22d1 −7.929 −6.476
22 21 −8.751 −9.679

DS: Docking score; Extra precision: XP; SP: Standard precision.

Table 8.

The scores calculated by different docking methodologies used in the present study with their rescoring values calculated by MMGBSA method.

C PDB ID: 3AOI
D.S. (XP)
kcal/mol
D.S. (SP)
kcal/mol
D.S. (HTVS)
kcal/mol
dG bind
kcal/mol
18g −11.172 −10.443 −10.65 −81.31
13b1 −10.242 −9.519 −7.85 −74.62
19e −11.07 −11.73 −6.38 −83.57
19a −10.77 −11.47 −9.26 −80.57
16b −9.21 −11.36 −10.78 −84.33
16a −9.26 −11.30 −11.47 −81.96
15b −9.05 −11.09 −9.24 −89.31
ARRY-403 −10.259 −10.101 −9.26 −72.62
RO-5305552 −6.707 −9.074 −7.75 −39.15
Piragliatin −8.345 −8.144 −7.24 −39.35
Glucokinase activator 1 −3.36 −5.782 −8.37 −0.96
ZINC08974524 −8.43 −11.17 −10.26 −19.55
ZINC00656909 −8.715 −10.887 −8.62 −56.96
ZINC31812808 −8.40 −8.223 −9.37 −76.4
ZINC14791611 −8.294 −10.879 −8.28 −29.58
ZINC05204145 −7.984 −7.323 −8.32 −53.31
ZINC01114130 −7.854 −10.21 −9.65 −37.62
ZINC09712705 −7.764 −8.962 −9.78 −47.96
ZINC14462664 −7.65 −10.997 −7.47 −75.4
ZINC02474054 −6.186 −4.208 −6.25 −8158

D.S.: Docking score; XP: Extra precision; SP: Standard precision; HTVS: High-throughput virtual screening.

2.8. ADME prediction studies

The top scored compounds were analyzed by different ADME properties such as drug-likeness, solubility, and pharmacokinetic studies. The QikProp software was used to calculate ADME properties (Table 9). Further, the Swiss-ADME tool was used to evaluate additional parameters like cytochrome profile of the drug with permeation through different other barriers.

Table 9.

ADME predictions of top scored compounds.

C QP log Po/w1 QPP- Caco2 QP log B.B.3 QPP-MDCK4 QP log Khsa5 MetR6 PHOA7
18g 3.524 62.806 −1.768 46.094 0.214 6 66.803
13b1 3.606 2683.955 −0.698 1438.153 −0.05 3 100
19e 3.411 290.144 −2.14 130.171 −0.097 4 78.033
19a 3.746 323.686 −1.674 146.509 0.346 2 93.806
16b 3.784 251.098 −1.595 332.05 0.266 3 92.054
16a 3.544 300.192 −1.505 262.463 0.187 3 92.039
15b 3.472 76.153 −1.903 56.786 −0.114 7 67.996
ARRY-403 2.664 170.959 −1.879 200.448 −0.156 6 82.508
GK-A 1 4.515 22.249 −2.361 47.821 0.115 3 64.54
Piragliatin 3.048 435.091 −1.217 374.987 0.038 7 92.016
RO-5305552 2.775 199.82 −2.015 174.056 −0.177 4 84.373
ZINC08974524 4.32 2282.231 −0.392 2329.878 0.362 4 100

Predicted [1: Octanol/water partition coefficient; 2: Caco-2 cell permeability (nm/s); 3: Brain/blood partition coefficient; 4: Apparent MDCK cell permeability (nm/s); 5: Human serum albumin binding]; 6: Number of metabolic reactions; 7: Percent human oral absorption.

3. Results

3.1. Analysis of pharmacophore modeling

The generated pharmacophore hypotheses with different scores are presented in Table 2. The hypothesis ADRR_1 was chosen as the best hypothesis with phase hypo-score = 1.18, survival score = 5.03, and site score = 0.98. The field-based and atom-based QSAR studies showed reliable statistical parameters with different evaluation factors. The results showed internal validation parameters such as R2 values 0.99, 0.98; R2CV values 0.62, 0.64; Q2 values 0.52, 0.71; SD values 0.62, 0.19; RMSE values 0.96, 0.66 and F values 299, 271 for atom-based and field-based models, respectively (Table 3). The scatter plots for both models are shown in Fig. 5.

Fig. 5.

Fig. 5

Correlation between test (5 A & C) and training (5B & D) set compounds by using atom and field-based 3D QSAR models.

3.2. 3D-QSAR

The contour maps showed the correlation between different bioactivities by various substituents on the core moiety (Fig. 3). The correlation between actual and predicted activities of test and training-set compounds for atom-based QSAR is depicted in Fig. 5. The contour maps generated by field-based model include HBA, HBD, steric, electrostatic, and hydrophobic fields (Fig. 4). Different substituent groups on the potent compound (16a) described by various colour responsible for increase or decrease in activity. The correlation between actual and predicted activities of test and training-set compounds for field-based QSAR is depicted in Fig. 5.

3.3. Docking and virtual screening studies

The PDB ID-3A0I was taken for docking purposes to evaluate binding interactions of potent ligands and ZINC compounds. The docking scores were compared with the observed activity. Compounds, 18g, 13b1, 19e, 19a, 16b, 16a, and 15b showed binding interactions with important amino acids required for GK-A. The amino acids bind with both compounds 15b and 18g were ARG250 and THR65, ARG63 as shown in Fig. 6. Compound 16a also showed good binding interactions described in Fig. 7. The docking score of the potent compounds of the series compared with standard drugs such as glucokinase activator 1, piragliatin, ARRY-403, and RO-5305552 are described in Table 8. The binding interactions of other potent compounds 19e and 19 are displayed in Fig. 8.

Fig. 6.

Fig. 6

Ligand interaction diagram for higher scoring compound 15b (6A) and 18 g (6B) in docking study.

Fig. 7.

Fig. 7

(7A) 2D and (7B) 3D docking interactions represented by compound 16a.

Fig. 8.

Fig. 8

Docking interactions represented by compound 19e (8A) and 19 (8B).

3.4. MMGBSA-based rescoring

The rescoring of docked structure of ligand and protein was performed by the MMGBSA-based method (molecular mechanics energies combined with the Poisson–Boltzmann or generalized Born and surface area continuum solvation). The screened ZINC hit compound ZINC08974524 (complex of ZINC08974524: 3A0I) showed docking score of −8.428 kcal/mol. Other ZINC hits such as ZINC00656909, ZINC31812808, ZINC14791611, ZINC05204145, ZINC01114130, ZINC09712705 and ZINC14462664 showed XP docking scores −8.715, −8.40, −8.294, −7.984, −7.854, −7.764 and −7.65 kcal/mol, respectively. All ZINC compounds showed negative value of dG binding energies as compared with standards (Table 8).

3.5. ADME properties calculation

ADME properties were calculated by the QikProp module, comprise of one another SwissADME tool. These parameters were within the acceptable range for ligands and ZINC hits (Table 9). The compounds of the series showed drug-likeness properties with no violation of the Lipinski rule. The bioavailability score of compounds showed the value of 0.55.

4. Discussion

The final hypothesis consisted of one HBD, one HBA, and two aromatic ring structures. The hypothesis (ADRR_1) showed alignment with other molecules of the series and displayed good correlation between structure and bioactivity. The features of the hypothesis were further taken for screening of ZINC compounds from the ZINCPHARMER (http://zincpharmer.csb.pitt.edu/) online tool. The atom based QSAR maps indicated the influence on bioactivity by the addition of substituents on the nucleus. The blue contour maps showed an increase in activity, whereas red maps showed decrease in activity. The compound 16a showed alignment with pharmacophore hypothesis ADRR_1 with different colours on their substituents. The electron-withdrawing group (EWG) substitution on benzamide derivatives exhibited increase in the activity, whereas EWG substitution on phenyl ring showed decrease in activity, as represented by red maps (Fig. 3). The HBD group addition showed no changes in activity. The hydrophobic group addition connected to the amide group displayed increase in activity, whereas the introduction of such group at phenyl ring decreased the potency and showed mixed activity throughout the ring. However, the addition of positive and negative ionic groups showed decrease in the activity.

In field based QSAR contour map electrostatic group contains the blue colour at the amide group linked heterocyclic compounds that showed the introduction of electron positive group at the site responsible for increase in the activity. The phenyl ring connected to heterocyclic ring with electron positive group may decrease or increase the activity. The HBA group introduction at phenyl ring connected group may increase the activity. The addition of HBD group at the amide group may increase the activity. The group contains steric field with green color in phenyl ring substituted group may be responsible for increase in activity. The potent compound 15b of the series showed good docking score using interactions with amino acid residues (NH…ARG63), (SO2…ARG250, THR65), and π-π staking with (phenyl……TYR214). Compound 16a also showed good binding interactions with amino acid residues such as TRY214, ARG250, THR65, and ARG63 (docking score −11.296 kcal/mol), important for GK-A activity (Fig. 7). The docking scores and the amino acid residues of the potent compounds of the series compared with standard drugs such as glucokinase activator 1 (−3.36 kcal/mol), piragliatin (−8.345 kcal/mol), ARRY-403 (−10.259 kcal/mol), and RO-5305552 (−6.707 kcal/mol) (Table 8). Furthermore, the other potent compounds 19e and 19 from both the series showed good binding interactions with THR65, ARG63, ARG250, and TYR214 essential amino acids that are important for a GK-A drug (Fig. 8).

The ZINC library was downloaded through the Swiss screening database. Total 3563 compounds were downloaded by ZINC database and further screened by different docking methodologies using Glide module. After applying Lipinski rule, the compounds were filtered through HTVS docking process. The top 50% of the compounds from this process were further taken for SP, and the top 20% compounds were finally taken for XP. Top hit, namely ZINC08974524 (Fig. 9) showed best docking score in SP (−11.17 kcal/mol), XP (−8.43 kcal/mol), and HTVS (−10.26 kcal/mol). The binding interactions of all the ZINC compounds were similar to crystal ligand interactions. The RMSD value was used as a parameter to check the binding pattern of different compounds from crystal ligand. These binding interactions of active compounds and ZINC derivatives displayed similar interaction as shown by crystal ligand of PDB ID-3A0I. The Zinc hit compounds may be used for the in vivo evaluation as GK-A. Furthermore, ZINC screening data suggested that the binding interactions were similar to the compounds taking for 3D QSAR study. So, these compounds may be designed further for the synthesis of potent compounds as GK-A against diabetes.

Fig. 9.

Fig. 9

SP (9A) and XP (9B) ligand interaction diagram for higher scoring compound (ZINC08974524) screened through ZINC database.

In ADME analysis, compounds showed solubility range from low to high values. The GI absorption was low for 16a and high for 19a compounds. The SwissTargetPrediction results exhibited that compounds have high target specificity for GK-A. All compounds showed good synthetic accessibility within the range of 2–4. These compounds may be useful for the generation of novel compounds as GK-A. The compounds screened through ZINC database showed good ADME properties (Table 9). However, cytochrome profiling for ZINC hits shown inhibitory activities against CYP3A4, CYP2C19, and CYP2C9. All the compounds of the series and ZINC hits showed noncarcinogenic activities.

5. SAR optimization

The pharmacophore model, 3D QSAR, virtual screening, and Zinc hit compounds may be used as a basis for the production of novel compounds as GK-A depicted in Fig. 10. The present SAR optimized by the 3D QSAR study revealed that the substitutions on benzamide scaffold with different characteristic features for the development of novel GK-A. The core moiety in the place of thiazole ring can be replaced by some electropositive atoms and hydrophobic groups such as long carbon chain and phenyl ring responsible for increase in activity. The oxygen atom of the benzamide group is more prominent for activity, or if it is replaced by less electronegative groups such as sulphur or nitrogen, then the activity may be diminished. The pyridine ring connected to the benzamide scaffold can be replaced by electropositive groups that results an increase in activity. These features can be used for the further development and synthesis of novel derivatives as GK-A.

Fig. 10.

Fig. 10

Substitutions on benzamide scaffold with different characteristic features for the development of future compounds as GK-A.

6. Conclusion

The present manuscript revealed a computation study on benzamide derivatives processed in a sequence to produce potent GK-As, where ligands and ZINC hits showed no violation of the Lipinski rules. All screened compounds displayed good synthetic accessibility. Based on 3D-QSAR, pharmacophore development, SwissTargetPrediction, virtual screening, and molecular docking studies, we may design novel molecules with good ADME properties and low toxicity. ADRR_1 is determined as the best pharmacophore in the study. The 3D QSAR study showed the best statistical data by atom-based and field-based models, consecutively. The binding interactions of compounds showed important amino acids required for activity. Fig. 10 showed the importance of different substituents on core moiety which may help for the development of novel compounds as GK-A. Furthermore, the present study may be helpful for researchers as a guiding tool for the development of novel benzamide derivatives as GK-A.

Declaration of Competing Interest

The author declare that there is no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgment

Dr Amena Ali is thankful for Schrödinger team for training and license. This research received no grant.

Footnotes

Peer review under responsibility of King Saud University.

References

  1. Ali A., Ali A. Integrated computational approaches on pyrazoline derivatives as B-Raf kinase inhibitors for the development of novel anticancer agents. J. Mol. Struct. 2021;1230:129861. doi: 10.1016/j.molstruc.2020.129861. [DOI] [Google Scholar]
  2. American Diabetes Association, 2009. Diagnosis and classification of diabetes mellitus. Diabetes Care. 32, S62-S67. [DOI] [PMC free article] [PubMed]
  3. Andreoli M., Persico M., Kumar A., Orteca N., Kumar V., Pepe A., Mahalingam S., Alegria A.E., Petrella L., Sevciunaite L., Camperchioli A., Mariani M., Di Dato A., Novellino E., Scambia G., Malhotra S.V., Ferlini C., Fattorusso C. Identification of the first inhibitor of the GBP1:PIM1 interaction. Implications for the development of a new class of anticancer agents against paclitaxel resistant cancer cells. J. Med. Chem. 2014;57(19):7916–7932. doi: 10.1021/jm5009902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Antoine M., Boutin J.A., Ferry G. Binding kinetics of glucose and allosteric activators to human glucokinase reveal multiple conformational states. Biochemistry. 2009;48:5466–5482. doi: 10.1021/bi900374c. [DOI] [PubMed] [Google Scholar]
  5. Bebernitz G.R., Beaulieu V., Dale B.A., Deacon R., Duttaroy A., Gao J., Grondine M.S., Gupta R.C., Kakmak M., Kavana M., Kirman L.C., Liang J., Maniara W.M., Munshi S., Nadkarni S.S., Schuster H.F., Stams T., Denny I.S., Taslimi P.M., Vash B., Caplan S.L. Investigation of functionally liver selective glucokinase activators for the treatment of type 2 diabetes. J. Med. Chem. 2009;52:6142–6152. doi: 10.1021/jm900839k. [DOI] [PubMed] [Google Scholar]
  6. Charaya N., Pandita D., Grewal A.S., Lather V. Design, synthesis and biological evaluation of novel thiazol-2-yl benzamide derivatives as glucokinase activators. Comput. Biol. Chem. 2018;73:221–229. doi: 10.1016/j.compbiolchem.2018.02.018. [DOI] [PubMed] [Google Scholar]
  7. Crisan L., Borota A., Bora A., Pacureanu L. Diarylthiazole and diarylimidazole selective COX-1 inhibitor analysis through pharmacophore modeling, virtual screening, and DFT-based approaches. Struc. Chem. 2019;30:2311–2326. [Google Scholar]
  8. Grewal A.S., Kharb R., Prasad D.N., Dua J.S., Lather V. Design, synthesis and evaluation of novel 3,5-disubstituted benzamide derivatives as allosteric glucokinase activators. BMC Chem. 2019;13:1–14. doi: 10.1186/s13065-019-0532-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Kar S., Harding A.P., Roy K., Popelier P.L.A. QSAR with quantum topological molecular similarity indices: toxicity of aromatic aldehydes to Tetrahymena pyriformis. SAR QSAR Environ. Res. 2010;21:149–168. doi: 10.1080/10629360903568697. [DOI] [PubMed] [Google Scholar]
  10. Mao W., Ning M., Liu Z., Zhu Q., Leng Y., Zhang A. Design, synthesis, and pharmacological evaluation of benzamide derivatives as glucokinase activators. Bioorg. Med. Chem. 2012;20:2982–2991. doi: 10.1016/j.bmc.2012.03.008. [DOI] [PubMed] [Google Scholar]
  11. Massa M.L., Gagliardino J.J., Francini F. Liver glucokinase: An overview on the regulatory mechanisms of its activity. IUBMB Life. 2011;63:1–6. doi: 10.1002/iub.411. [DOI] [PubMed] [Google Scholar]
  12. Matschinsky F.M., Porte D. Glucokinase activators (GKAs) promise a new pharmacotherapy for diabetics. F1000 Med. Rep. 2010;2:43. doi: 10.3410/M2-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Matschinsky F.M. A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes. 1996;45:223–241. doi: 10.2337/diab.45.2.223. [DOI] [PubMed] [Google Scholar]
  14. Park K. Identification of YH-GKA, a novel benzamide glucokinase activator as therapeutic candidate for type 2 diabetes mellitus. Arch. Pharm. Res. 2012;35:2029–2033. doi: 10.1007/s12272-012-1201-9. [DOI] [PubMed] [Google Scholar]
  15. Park K., Lee B.M., Hyun K.H., Han T., Lee D.H., Choi H.H. Design and synthesis of acetylenyl benzamide derivatives as novel glucokinase activators for the treatment of T2DM. ACS Med. Chem. Lett. 2015;6:296–301. doi: 10.1021/ml5004712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Park K., Lee B.M., Hyun K.H., Lee D.H., Choi H.H., Kim H., Chong W., Kim K.B., Nam S.Y. Discovery of 3-(4-methanesulfonylphenoxy)-N-[1-(2-methoxy-ethoxymethyl)-1H-pyrazol-3-yl]-5-(3-methylpyridin-2-yl)-benzamide as a novel glucokinase activator (GKA) for the treatment of type 2 diabetes mellitus. Bioorg. Med. Chem. 2014;22(7):2280–2293. doi: 10.1016/j.bmc.2014.02.009. [DOI] [PubMed] [Google Scholar]
  17. Park K., Lee B.M., Kim Y.H., Han T., Yi W., Lee D.H., Choi H.H., Chong W., Lee C.H. Discovery of a novel phenylethyl benzamide glucokinase activator for the treatment of type 2 diabetes mellitus. Bioorg. Med. Chem. Lett. 2013;23:537–542. doi: 10.1016/j.bmcl.2012.11.018. [DOI] [PubMed] [Google Scholar]
  18. Peng X.X., Feng K.R., Ren Y.J. Molecular modeling studies of quinazolinone derivatives as novel PI3Kd selective inhibitors. RSC Adv. 2017;7:56344–56358. [Google Scholar]
  19. Pfefferkorn J.A., Guzman-Perez A., Oates P.J., Litchfield J., Aspnes G., Basak A., Benbow J., Berliner M.A., Bian J., Choi C., Freeman-Cook K., Corbett J.W., Didiuk M., Dunetz J.R., Filipski K.J., Hungerford W.M., Jones C.S., Karki K., Ling A., Li J.-C., Patel L., Perreault C., Risley H., Saenz J., Song W., Tu M., Aiello R., Atkinson K., Barucci N., Beebe D., Bourassa P., Bourbounais F., Brodeur A.M., Burbey R., Chen J., D'Aquila T., Derksen D.R., Haddish-Berhane N., Huang C., Landro J., Lee Lapworth A., MacDougall M., Perregaux D., Pettersen J., Robertson A., Tan B., Treadway J.L., Liu S., Qiu X., Knafels J., Ammirati M., Song X.i., DaSilva-Jardine P., Liras S., Sweet L., Rolph T.P. Designing glucokinase activators with reduced hypoglycemia risk: discovery of N, N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as a clinical candidate for the treatment of type 2 diabetes mellitus. Med. Chem. Commun. 2011;2(9):828. doi: 10.1039/c1md00116g. [DOI] [Google Scholar]
  20. Pfefferkorn J.A., Guzman-Perez A., Litchfield J., Aiello R., Treadway J.L., Pattersen J., et al. Discovery of (S)-6-(3cyclopentyl-2-(4-(trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic acid as a hepatoselective glucokinase activator clinical candidate for treating type 2 diabetes mellitus. J. Med. Chem. 2012;55:1318–1333. doi: 10.1021/jm2014887. [DOI] [PubMed] [Google Scholar]
  21. Rajeswari M., Santhi N., Bhuvaneswari V. Pharmacophore and Virtual Screening of JAK3 inhibitors. Bioinfo. 2014;10:157–163. doi: 10.6026/97320630010157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Sakkiah S., Senese S., Yang Q., Lee K.W., Torres J.Z. Dynamic and multi-pharmacophore modeling for designing polo-box domain inhibitors. PloS One. 2014;9 doi: 10.1371/journal.pone.0101405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Schrodinger. 2017. LigPrep, Schrodinger, LLC, NY.
  24. Tagami S., Sekine S., Kumarevel T., Hino N., Murayama Y., Kamegamori S., Yamamoto M., Sakamoto K., Yokoyama S. Crystal structure of bacterial RNA polymerase bound with a transcription inhibitor protein. Nature. 2010;468:978–982. doi: 10.1038/nature09573. [DOI] [PubMed] [Google Scholar]
  25. Van Mourik T., Bühl M., Gaigeot M.P. Density functional theory across chemistry, physics and biology. Philosophical Trans. Ser. A, Math. Phys. Eng. Sci. 2014;372(2011):20120488. doi: 10.1098/rsta.2012.0488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Waring M.J., Brogan I.J., Coghlan M., Johnstone C., Jones H.B., Leighton B., McKerrecher D., Pikea K.G., Robba G.R. Overcoming retinoic acid receptor-α based testicular toxicity in the optimisation of glucokinase activators. Med. Chem. Commun. 2011;2:771–774. [Google Scholar]

Articles from Saudi Journal of Biological Sciences are provided here courtesy of Elsevier

RESOURCES