Open Access Article
Xiu Xiu Peng,
Kai Rui Feng and
Yu Jie Ren
*
School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China. E-mail: clab@sit.edu.cn
First published on 15th December 2017
The forced expression of phosphoinositide 3-kinase δ (PI3Kδ) in B cells was found to be oncogenic, rendering PI3Kδ an attractive drug target for chronic lymphocytic leukaemia. This study aimed to systemically explore the interaction mechanism of novel quinazolinone scaffold-based derivatives as PI3Kδ inhibitors using 3D-QSAR, molecular docking, pharmacophore model and molecular dynamics (MD) simulations. The 3D-QSAR models CoMFA, CoMSIA and Topomer CoMFA were established to discover critical structural factors affecting PI3Kδ inhibitory activity. The models showed suitable reliabilities (q2 0.741, 0.712 and 0.711) and predictive abilities (rpred2 0.851, 0.738 and 0.828, respectively). Contour maps indicated that the bioactivity of PI3Kδ inhibitor was affected most by electrostatic and hydrophobic fields. The Surflex-Dock and pharmacophore model result showed that enhancing the H-bond interaction of the key substituents around the 2- and 4-positions of pyrimidine with Glu826, Val828 and Asp911, as well as the electrostatic interactions of substituents around the 3-position of benzene with Ser831, Asp832 and Asn836, significantly affected the improvement in the activity and stability of the inhibitor. Based on these results, 10 novel PI3Kδ inhibitors with higher predicted activity and binding affinity were designed by introducing the heterocycles pyrrolopyridine or purine. 10 ns MD simulations further study the stable docking conformation of designed compounds, which showed strong hydrogen bond interactions with key residues Ser831 and Asp832 in a propeller-like fashion. These results provided strong guidance for the discovery and optimization of novel potent PI3Kδ selective inhibitors.
In recent years, researchers found that the B cell receptor (BCR) signalling pathway is crucial for the evolution and progression of CLL. PI3Kδ regulates B cell maturation and function by relaying signals from the BCR to downstream cytokines and chemokines. PI3Kδ is frequently over-expressed and is hyperactive in lymphoma cells; thus, PI3Kδ has been an appealing target for B cell leukaemia disease.8 Idelalisib as the first oral selective PI3Kδ inhibitor is approved by the FDA for the treatment of CLL, which greatly promoted the development of the PI3Kδ inhibitor. However, idelalisib is only suitable for patients with relapsed CLL and has severe hepatotoxicity.9 Although several PI3Kδ inhibitors, such as AMG-319,10 GS-9820,11 TGR-1202,12 IPI-145 (ref. 13) and GSK-2269557 (ref. 14) (Fig. 1), have already entered clinical trials in haematological malignancies or inflammatory diseases, these inhibitors have some disadvantages. For example, AMG-319, GS-9820 and TGR-1202 have relatively low activity compared with idelalisib and can cause inevitable side effects of diarrhea and neutropenia. GSK-2269557 has a higher activity and the selectivity of PI3Kδ over other isoforms is more than 1000; however, it causes adverse reactions of headaches and arthralgia. Therefore, finding novel potent PI3Kδ selective inhibitors with strong applicability and fewer side effects is very significant.15 Gilead Sciences further studied quinazolinone analogues, and Patel et al. claimed a series of new quinazolinone derivatives bearing a 2,4,6-triaminopyrimidine motif.16 These compounds showed better PI3Kδ inhibitory potency and improved selectivity over other PI3K isoforms compared with idelalisib. Moreover, the compounds have enhanced metabolic stability by mitigating aldehyde oxidase-mediated metabolism and shown excellent pharmacokinetic properties suitable for drug development. The pharmacokinetic and activity test in vivo of several compounds have been done in rat models and showed good results; hence, the study of such quinazolinone derivatives can promote the development of novel PI3Kδ inhibitors. However, its structure–activity relationship and the interaction mechanism have not been systematically studied by theoretical molecular modeling method. Therefore, the structure and activity of novel quinazolinone PI3Kδ inhibitors should be systematically studied by computer-aided drug design method.
The QSAR, as a mathematical technology linking chemical structure and biological activity in a quantitative manner, have been widely applied to assist the design of new drug candidates in the pharmaceutical sciences.17 Recently, the approach has speed up the lead optimization process by studying three dimensional features of chemicals.18 3D-QSAR analysis play important role in the development of targeted inhibitors for the treatment of cancer, HIV and cardio-cerebrovascular diseases.19–21 This technology, which mainly includes CoMFA,22 CoMSIA23 and Topomer CoMFA,24 can provide beneficial information on structural modifications for designing the lead compounds with desired inhibitory activity and be used to predict the activity of newly designed inhibitors. In this study, a series of quinazolinone scaffold-based derivatives as novel PI3Kδ selective inhibitors were selected to construct 3D-QSAR models, which revealed the relationship between structural characteristics and inhibitory activity. Molecular docking and pharmacophore model were used to analyze the influence of pharmacophore groups on the activity of the compounds and investigate the binding patterns of ligands with PI3Kδ receptor proteins. Molecular dynamics simulations were applied to analyse the dynamics behavior of the ligand, and obtain more detailed ligand–protein interaction information.25–28 The stepwise description of the molecular modeling was shown in Fig. S1 (see ESI†). The above systematic research is important for obtaining novel candidate compounds as PI3Kδ inhibitors and for avoiding the blindness of drug design and ensuring more efficient drug synthesis.
IC50). It was acceptable that the range of pIC50 values between the most active and the least active molecule is 4.23 log units, and the distribution of pIC50 values for the complete set was shown in Fig. S2.† All the experimental data are randomly partitioned into two subsets: a training set (30 molecules) for QSAR model generation, and a test set consisting of additional 7 molecules for model evaluation. The compounds of test set were selected in accordance with the distribution of bioactivity data and structural diversity, which is a vital factor to determine the reliability of the 3D-QSAR model. The CoMFA, CoMSIA and Topomer CoMFA models were constructed based on the same training set and test set. Furthermore, the quinazolinone derivatives as novel PI3Kδ selective inhibitors have the same structure skeleton with idelalisib, so the co-crystal structure of PI3Kδ protein with idelalisib was downloaded from the Protein Data Bank (PDB ID: 4XE0) for the molecular docking study.29
| No. | Structure | Actual pIC50 | Predicted pIC50 | CoMFA | CoMSIA | |||
|---|---|---|---|---|---|---|---|---|
| R1 | R2 | R3 | CoMFA | CoMSIA | Residual | Residual | ||
| a Test set molecules. | ||||||||
| 01 | ![]() |
H | 5-Cl | 7.004 | 7.002 | 6.851 | −0.002 | −0.154 |
| 02 | ![]() |
H | 5-Cl | 5.770 | 5.780 | 5.828 | 0.010 | 0.058 |
| 03 | ![]() |
H | 5-Cl | 8.097 | 8.247 | 8.087 | 0.150 | −0.010 |
| 04 | ![]() |
H | 5-Cl | 9.398 | 9.145 | 9.343 | −0.253 | −0.055 |
| 05 | ![]() |
H | 5-Cl | 7.523 | 7.517 | 7.522 | −0.007 | −0.001 |
| 06a | ![]() |
H | 5-Cl | 8.602 | 8.926 | 8.555 | 0.324 | −0.047 |
| 07 | –CONH2 | H | 5-Cl | 7.959 | 7.846 | 7.921 | −0.113 | −0.038 |
| 08 | –CF3 | H | 5-Cl | 8.745 | 8.674 | 8.726 | −0.071 | −0.019 |
| 09 | –CH3 | H | 5-Cl | 7.854 | 7.873 | 7.916 | 0.019 | 0.062 |
| 10a | –OCH3 | H | 5-Cl | 7.409 | 7.395 | 7.257 | −0.014 | −0.152 |
| 11 | ![]() |
H | 5-Cl | 9.523 | 9.438 | 9.547 | −0.085 | 0.024 |
| 12 | ![]() |
H | 5-Cl | 9.301 | 9.345 | 9.304 | 0.044 | 0.003 |
| 13 | ![]() |
H | 5-Cl | 9.398 | 9.457 | 9.458 | 0.059 | 0.060 |
| 14 | ![]() |
H | 5-Cl | 8.921 | 8.895 | 8.930 | −0.026 | 0.009 |
| 15 | ![]() |
H | 5-Cl | 9.523 | 9.536 | 9.526 | 0.013 | 0.003 |
| 16 | ![]() |
H | 5-Cl | 10.00 | 9.817 | 9.652 | −0.183 | −0.348 |
| 17 | ![]() |
H | 5-Cl | 8.367 | 8.472 | 8.408 | 0.105 | 0.041 |
| 18 | ![]() |
H | 5-Cl | 9.046 | 9.127 | 9.062 | 0.081 | 0.016 |
| 19 | ![]() |
H | 5-Cl | 9.000 | 9.111 | 9.049 | 0.111 | 0.049 |
| 20a | ![]() |
H | 5-Cl | 9.886 | 9.364 | 9.041 | −0.522 | −0.845 |
| 21 | –CN | 4-F | 5-Cl | 8.699 | 8.936 | 8.836 | 0.237 | 0.137 |
| 22a | –CN | 3-F | 5-Cl | 9.824 | 9.452 | 9.309 | −0.372 | −0.515 |
| 23 | –CN | 3-Cl | 5-Cl | 9.347 | 9.323 | 9.475 | −0.024 | 0.128 |
| 24 | –CN | 3-CN | 5-Cl | 8.721 | 8.703 | 8.765 | −0.018 | 0.044 |
| 25 | –CN | 3-OCH3 | 5-Cl | 9.046 | 9.026 | 9.005 | −0.020 | −0.041 |
| 26 | –CN | 3-CF3 | 5-Cl | 8.444 | 8.319 | 8.407 | −0.125 | −0.037 |
| 27 | –CN | 3-CH2CHF2 | 5-Cl | 8.796 | 8.761 | 8.799 | −0.035 | 0.003 |
| 28 | –CN | 3-CHF2 | 5-Cl | 9.268 | 9.485 | 9.363 | 0.217 | 0.095 |
| 29a | –CN | 3,5-2CN | 5-Cl | 6.824 | 6.158 | 7.620 | −0.666 | 0.796 |
| 30 | –CN | 3-Cl | 5-Cl | 8.824 | 8.747 | 8.830 | −0.077 | 0.006 |
| 5-CHF2 | ||||||||
| 31a | –CN | 3,5-2F | 5-Cl | 9.328 | 8.984 | 8.784 | −0.345 | −0.544 |
| 32 | –CN | 3,5-2F | 5-Cl | 8.538 | 8.493 | 8.490 | −0.045 | −0.048 |
| 6-F | ||||||||
| 33a | –CN | 3,5-2F | H | 7.824 | 8.323 | 8.452 | 0.499 | 0.628 |
| 34 | –CN | 3,5-2F | 5-F | 8.658 | 8.665 | 8.613 | 0.007 | −0.045 |
| 35 | –CN | 3,5-2F | 6-F | 8.319 | 8.352 | 8.353 | 0.033 | 0.034 |
| 36 | –CN | 3,5-2F | 8-F | 8.456 | 8.469 | 8.479 | 0.013 | 0.023 |
| 37 | –CN | 3,5-2F | 8-CN | 8.229 | 8.212 | 8.230 | −0.017 | 0.001 |
000 step iterations were accomplished or when the energy gradient convergence criterion of 0.005 kcal (mol−1 Å−1) was achieved.30 The minimized structures were utilized as the initial conformation for constructing 3D-QSAR models and molecular docking. Since the predictive capability and reliability of the built models are directly dependent on superimposition of molecules, the selection of template molecule is one of the important steps. Herein, CoMFA, CoMSIA and Topomer CoMFA models were constructed by ligand-based superimposition. All the compounds were superimposed to the compound 16 with highest inhibitory activity. In order to describe the structure of the molecule clearly, as shown in Fig. 2A, the whole structure was divided into region A, region B and region C. The common structure that used to align all compounds was shown in red, and the alignment results were shown in Fig. 2B.
![]() | ||
| Fig. 2 Structure of template compound 16, the common substructure (red) used in alignment (A), and the alignment of training set (B). | ||
Topomer CoMFA automates the creation of models, and its alignment is extremely rapid and objective. Identifying the R-groups for the molecules in training set was the crucial step for establishing Topomer CoMFA model.32 In this study, the template molecule 16 was split into two fragments by cutting one single bond as shown in Fig. 2A, and all the other structures were divided into R1 and R2 fragments in the same way. The 3D-QSAR models were graphically represented by field contour maps, which were able to identify functional groups and physicochemical properties affecting biological activity and binding affinity.
![]() | (1) |
Moreover, Golbraikh and Tropsha put forward another four criteria to evaluate the robustness and the predictive abilities of the established models.34 The 3D-QSAR models can be regarded as acceptable if they satisfy the following conditions:
| R2 > 0.6; 0.85 < k < 1.15; Rm2 >0.5; [(R2 − R02)/R2] < 0.1. |
The R value was calculated by the following formula:
![]() | (2) |
The slope k value of the regression line through the origin must close to 1, and the k value was calculated by the following formula:
![]() | (3) |
Rm2 value was the n-CV correlation coefficient obtained from the PLS process and defined as following equation, which was provided by Roy et al.:35
![]() | (4) |
R02 value was calculated as following equation:
![]() | (5) |
MAE value was calculated as following equation:36
![]() | (6) |
In the above equations, Ypred(test) and Ytest represented the predicted and experimental pIC50 value for each compound in the test set. Ȳtraining and Ȳtest were the average pIC50 values for each compound in the training set and test set, respectively.
In addition, the analysis of prediction errors plays a significant role in understanding systematic errors of QSAR models and determining the model acceptability. To check the predictions error of 3D-QSAR models, a series of parameters such as the absolute of average error (AE), average absolute error (AAE), number of positive and negative errors (NPE and NNE), mean positive error (MPE) and mean negative error (MNE) were calculated.37 Additionally, the Applicability Domain of CoMSIA model, which represents the chemical space from which a model is derived and where a prediction is considered to be reliable, was defined by the leverage approach.
O) that represent potential hydrogen bonds and favorable hydrophobic interactions with protein atoms were used to construct the protomol. Each probe serves as a potential alignment point for atoms in a ligand, and is scored to represent that probe's affinity for the protein. Probes with high affinity are merged into a pocket, the protomol.38 In this study, the ligand-based docking mode was adopted to generate the protomol. In docking process, all compounds are regarded to be the flexible whereas PI3Kδ protein is considered to be rigid. Subsequently, all the compounds were docked into the active pocket with other defaulted parameters.39 To predict the binding affinity of the ligand to the target protein, the default scoring function was used. Finally, 10 conformations were obtained for each compound, and the conformation with highest docking score and similar orientation to the original ligand was chosen for binding mode and dynamics simulation study.
| Parameters | CoMFA | CoMSIA | Topomer CoMFA | Constraints |
|---|---|---|---|---|
| q2 | 0.741 | 0.712 | 0.711 | >0.5 |
| ONC | 8 | 9 | 6 | — |
| r2 | 0.985 | 0.989 | 0.96 | >0.9 |
| SEE | 0.122 | 0.105 | 0.19 | ≪1 |
| F | 169.6 | 204 | 116 | ≫100 |
| rpred2 | 0.851 | 0.738 | 0.828 | >0.5 |
| MAEtraining | 0.073 | 0.053 | 0.120 | |
| MAEtest | 0.345 | 0.447 | 0.409 | |
| 0.1 × training set range | 0.423 | |||
| 0.15 × training set range | 0.635 | |||
| K | 1.017 | 1.015 | 1.006 | 0.85 < K < 1.15 |
| R02 | 0.983 | 0.968 | 0.996 | — |
| R2 | 0.872 | 0.833 | 0.869 | — |
| (R2 − R02)/R2 | −0.126 | −0.161 | −0.146 | <0.1 |
| Rm2 | 0.583 | 0.528 | 0.559 | >0.5 |
| Field contribution (%) | ||||
| Steric | 35.1 | 9.2 | ||
| Electrostatic | 64.9 | 33.8 | ||
| Hydrophobic | 20.6 | |||
| H-donor | 18.9 | |||
| H-acceptor | 17.5 | |||
These models were validated by a test set of 7 compounds, which displayed satisfactory rpred2 values of 0.851, 0.738 and 0.828 for CoMFA, CoMSIA and Topomer CoMFA models, respectively. Other external validation parameters of the models were also shown in Table 2. The CoMFA model was validated by the test set with a n-CV coefficient R2 value of 0.872, a Rm2 value of 0.583, a k value of 1.017, a R02 value of 0.983, and (R2 − R02/R2) value of −0.126. Similarly in CoMSIA model, the R2, R02, Rm2, k value and (R2 − R02/R2) value were found to be 0.833, 0.968, 0.528, 1.015 and −0.161, respectively. Simultaneously, the Topomer CoMFA analysis indicated R2 value of 0.869, R02 value of 0.996, the Rm2 value of 0.559, high k value of 1.006, and (R2 − R02/R2) value of −0.146. The results showed that the MAEtraining (0.073 for CoMFA, 0.053 for CoMSIA, 0.120 for Topomer CoMFA) and MAEtest (0.345 for CoMFA, 0.409 for Topomer CoMFA) values were both less than the 0.1 × training set range (0.423), indicating the good predictive ability of our model. In CoMSIA model, the MAEtest value of 0.447 was more than the 0.10 × training set range, but less than 0.15 × training set range indicating the moderate predictive ability of the model. All the external validation parameters of 3D-QSAR models were similar and in an acceptable limit. The plots showing the experimental and predicted pIC50 values for the total set in the CoMFA, CoMSIA and Topomer CoMFA approaches were represented in Fig. S4.† Most of points were uniformly distributed along the trend line, implying the predicted pIC50 values were in good agreement with the experimental pIC50 values. Moreover, the correlations of the experimental and predicted values showed a satisfactory predictive capability throughout the whole set of data.
The presence of bias in prediction errors from QSAR model can be easily identified from a residual plot. As shown in Fig. 4, residual plots in the test set of 3D-QSAR models indicated that the residuals were randomly distributed around zero and did not show any pattern. In addition, the parameters of prediction errors were shown in Table 3. The NPE/NNE or NNE/NPE was less than 5, the ABS(MPE/MNE) or ABS(MNE/MPE) was less than 2, and the AAE-ABS(AE) value was more than 0.5 × AAE value. The results further proved that the 3D-QSAR models have a reliable predicted ability.
| Parameters | |AE| | AAE | MPE | |MNE| | NPE | NNE | NPE/NNE or NNE/NPE | ABS (MPE/MNE) or ABS (MNE/MPE) | AAE − ABS(AE) | 0.5 × AAE |
|---|---|---|---|---|---|---|---|---|---|---|
| CoMFA | 0.156 | 0.392 | 0.412 | 0.384 | 2 | 5 | 2.5 | 1.073 | 0.235 | 0.196 |
| CoMSIA | 0.097 | 0.504 | 0.712 | 0.421 | 2 | 5 | 2.5 | 1.692 | 0.407 | 0.252 |
| Topomer CoMFA | 0.027 | 0.436 | 0.358 | 0.539 | 4 | 3 | 1.333 | 1.508 | 0.409 | 0.218 |
| hi = xi(XXT)−1xTi (i = 1,…, n) | (7) |
![]() | ||
| Fig. 7 Steric (A, B) and electrostatic(C, D) contour maps of Topomer CoMFA model around R1 and R2 fragments. | ||
In the CoMFA electrostatic contour map, as displayed in Fig. 5B, two small red contours around the cyano group of pyrimidine ring indicated that an increase in the electron density will be favourable to enhance the activity. Compound 4 and compound 3 with cyano group as substituent showed better activity than corresponding compound 1 and compound 2. Moreover, this fact was also observed between molecules 8, 7, and 9 where increasing the electron density (–CF3 > –CONH2 > –CH3) would result in increase of inhibitory activities (8.67 > 8.00 > 7.85), respectively. A large blue contour around the 4-position of region A revealed the importance of positive atomic groups. It can be explained by comparing molecule 20 (having methyl group) and 19 (having chlorine group), the group with positive charge at this position would lead to the increase of pIC50 values. Simultaneously, one medium blue cubes near 3-position of region B indicated that the positively charged groups were preferable. As can be seen from Table 1, using the positively charged substituents (–OCH3 > –CH2CHF2 > –CN > –CF3) would lead to increase of pIC50 values in the molecules (25 > 27 > 24 > 26).
The steric and electrostatic contour maps of CoMSIA and Topomer CoMFA were described in Fig. 6 and 7. Not surprisingly, the contour maps showed good agreement with that of CoMFA model, so they were not discussed. The hydrophobic contour plot was constructed through CoMSIA model, presenting in Fig. 8A. The yellow and gray color represented favorable and unfavorable hydrophobic areas, respectively. A medium yellow cube surrounding the cyano group of pyrimidine ring indicated that hydrophobic property in this region might increase the activity. Compound 7 showed less bioactivity than compounds 4 and 8, which may be due to the fact that –CONH2 was replaced by relative hydrophobic substituent –CN and –CF3. Compounds 11, 12, 13, 14 and 15 showed better inhibitory activity, because 5-position of pyrimidine contained hydrophobic substituent alkynyl. A large grey contour surrounding the 2-position of pyrimidine ring suggested that hydrophilic group was favored at this place. The compound 17 showed lower bioactivity than compound 4 due to the introduction of chlorine increased the hydrophobic property of pyrimidine. Another gray contours around the 3-position of benzene ring was found to be hydrophobic unfavorable. And when this criterion was applied to compounds 16, 23, 26 and 28, the activity was found in the order of 16 > 23 > 28 > 26.
The results of statistical analysis revealed that H-bond donor field had significant contribution on the PI3Kδ inhibitory activity. As shown in Fig. 8B, the cyan and purple contours display the favorable and unfavorable H-bond donor region respectively. Two small cyan contours near 4-position of region A suggested that hydrogen bond donor atoms were favorable in these regions. It can be demonstrated by the fact that compound 1, 4 and 16 where using –NH2 substituent at this position showed satisfactory activity. In addition, a large purple contour was around N atom of pyrimidine ring, indicating that hydrogen bond acceptor atom was conducive to the improvement of bioactivity. The nitrogen atom in this position may be beneficial to the generation of hydrogen bond between the compound and PI3Kδ protein. The H-bond acceptor contour plot of CoMSIA model containing the most active compound 16 was illustrated in Fig. 8C, and the magenta and red contours show favorable and unfavorable hydrogen bond acceptor region. A large red tetrahedron surrounding the 4-position of pyrimidine ring indicated that hydrogen bond donor group was advantaged to inhibitory activity, which was consistent with the H-bond donor contour map of CoMSIA as shown in Fig. 8B. Moreover, a magenta contour was around 5-position of pyrimidine ring (region A). For example, compound 4 and 3 which using –CN substituent at this position displayed higher activity than compound 1 and 2. Based on the information derived from CoMFA, Topomer CoMFA and CoMSIA can be utilized strategically in the design of novel quinazolinone derivatives with better PI3Kδ inhibitory activity.
N⋯H–Val828). Overall, the Surflex-Dock program can successfully reproduce the original docking conformation of PI3Kδ protein.
Subsequently, all the 37 compounds were docked into PI3Kδ ligand-binding pocket to explore the binding mode of compounds in this study. As shown in Fig. S8,† most of the quinazolinone derivatives bind in the active pocket in the similar propeller-like conformation with the original ligand. Moreover, H-bond interactions were found between the quinazolinone derivatives and several key residues, such as Glu826, Val828, Thr833, Asn836, Met752 and Asp911. In addition, the binding conformation and key interactions of the most active molecule 16 with receptor was shown in Fig. 9A. It formed four hydrogen bonds with the residues Glu826, Val828 and Asp911. The existing interaction, including hydrogen bond, electrostatic, van der Waals and hydrophobic interaction was shown in Fig. 9B. The 2-NH2 group of pyrimidine formed one hydrogen bond interaction with the amino acid Val828 at distance of 2.16 Å, and nitrogen atom at 3-position of the pyrimidine was hydrogen bonded to residue Val828 (2.09 Å). Additionally, the 5-CN of pyrimidine ring formed a hydrogen bond with Asp911 at distance of 2.80 Å, and the 4-NH2 group was hydrogen bonded to residue Glu826 (1.92 Å). It was consistent with the H-bond donor contour map of CoMSIA as shown in Fig. 8B, where 2, 4-position of the region A was found favorable as hydrogen bond donor groups. And the quinazolinone ring formed hydrophobic interactions with amino acids Met752, Trp760 and Ile777, indicated that the hydrophobic group at quinazolinone was favorable for the binding affinity. Hydrophilic amino acids Asp832, Thr833 and Asn836 were found near 3-position of the region B, indicated that the hydrophilic group at the region was favorable. As shown in Fig. 5B, negatively charged amino acids like Glu826 and Asp911 were found surrounding the 2, 4-position of the region A and validated the CoMFA electrostatic contour map where the blue cube over region A indicated electropositive groups were favorable. Moreover, the other residues, such as Phe751, Thr833, Asn836, and Met900, could also stabilize ligand through electrostatic interaction (Fig. 9B).
![]() | ||
| Fig. 9 Docking result of compound 16 (A) in the protein active site, ligand (green) and 2D interaction map of compound 16 (B) with 4XE0. | ||
Meanwhile, according to the docking results of all the compounds, the compound 06 and 16 with 2-NH2 at region A formed additional H-bond interactions with Glu826, showed better inhibitory activity and higher docking score compared with compound 03 and 04, which have not H-bond donor substituents in the same position. Meanwhile, the compound 02 with 4-NH2 at region A formed additional H-bond interactions with Val828, showed higher binding affinity compared with compound 01, which indicated H-bond donor substituents in this region was favorable. It can also be proved by the fact that compound 04 and 16 displayed higher activity than compound 03 and 07. Furthermore, the docking result of the least active molecule 02 indicated that only one hydrogen bond interaction was formed with the residue Val828 at distance of 2.07 Å. These results suggested that the higher active molecules have stronger H-bond interactions with Glu826 and Val828 than less active molecules. It can be found that H-bond interactions with Glu826 and Val828 were critical to maintain the activity and stability of the PI3Kδ inhibitor. The docking analysis of compound 22 and 31 with higher activity indicated that the electrostatic interactions with residues Ser831, Asp832 and Thr833 were improved by introducing F substituent to benzene ring. Furthermore, the docking analysis showed that most studied ligands fit into a hydrophobic pocket formed by amino acids Met752, Trp760 and Ile777, and formed electrostatic interactions with Ser831, Asp832, Thr833 and Asn836. It can be concluded that enhancing hydrogen-bond interaction with Glu826, Val828 and Asp911, and hydrophobic interaction with Met752, Trp760, Ile777, Thr833 and Met900, were very significant to improve the activity and stability of the inhibitor. Moreover, the binding interaction patterns were complementary to those of CoMFA, Topomer CoMFA and CoMSIA contour maps.
To further validate the binding mode and 3D-QSAR models, the molecular computer aided design procedure (MOLCAD) was employed. MOLCAD calculate and display the cavity depth (CD), electrostatic potential (EP), lipophilic potential (LP), and hydrogen bond site (HB) of the binding pocket. Fig. S7† depicted the MOLCAD potential surfaces structure of the binding pocket of the compound 16. As shown in Fig. S7,† the substitute at the 5-position of pyrimidine was oriented to red surface, thus hydrogen bond acceptor group would be favorable; 2-NH2 and 4-NH2 at region A were oriented to blue area, which indicated that the surface of this region was functioned as H-bond acceptors. The observations from this hydrogen bonding sites were matched with the docking results and CoMSIA H-bond donor contour map. Fig. S7† indicated that the substituent at 5-position of compound 16 were closed to the hydrophobic region of the active pocket, and it also indicated that more hydrophobic group could improve the inhibitory activity. Furthermore, the major color of the quinazolinone skeleton is brown, which indicated that hydrophobic interaction in this region is favorable. Meanwhile, the docking surface of all compounds, as shown in Fig. S8,† indicated that most compounds docked to the same binding pocket in a propeller-like conformation with compound 16. These results indicated that the MOLCAD-generated surface maps of binding site were in good consistency with the 3D-QSAR contour maps and docking poses in terms of electrostatic, hydrophobic and hydrogen-bonding potential.
![]() | ||
| Fig. 10 The best pharmacophore model aligned to five PI3Kδ inhibitors (hydrophobic features in cyan and H-bond acceptor atoms in green). | ||
In order to prioritize the designed compounds, docking analysis was performed to validate the binding modes of all designed compounds. They were showing similar binding pattern as that of known potent inhibitors with important hydrogen bonds interactions, most notably with the hinge residues Glu826, Val828 and Asp911. Two newly designed molecules which have a higher activity predicted by 3D-QSAR model, D04 and D06 were selected for an in-depth analysis. As shown in Fig. 12, compound D06 and D04 were both docked in the binding site via six hydrogen bonds in similar propeller-like conformation with original ligand. Five residues (Glu826, Val828, Ser831, Asp832 and Asp911) were suggested to be crucial because they can form hydrogen bonds with the designed molecules. The H-bond distances of D04 with the key residues were observed to be 1.89 Å (N–H⋯O
C–Glu826), 2.04 Å (N⋯H–N–Val828), 2.30 Å (N–H⋯O
C–Val828), 2.85 Å (CN⋯H–O–Asp911) respectively. Additionally, the amino of pyridine as hydrogen bond donor group formed two strong hydrogen bond interactions with the amino acid Ser831 and Asp832 at distance of 2.20 Å and 2.24 Å, respectively. The pyrimidine ring of D04 formed hydrophobic interactions with Ile777, Val828, Met 900 and Ile910 (Fig. S9†). In addition, the quinazolinone ring sandwiched between Met752, Pro758, Trp760 and Ile777. The van der Waals interactions were observed between compound D04 and the residues, Leu759, Trp760, His830, Phe908 and Ile910, and the relative position of quinazolinone ring changed in contrast to that of compound 16. As shown in Fig. S9,† the quinazolinone ring of D04 formed strong van der Waals interaction with the residues, Pro758, Leu759 and Trp760 at a relative short distance. In addition, van der Waals interactions between pyrimidine ring and key residues Phe908 and Ile910 were observed in the binding pocket, which further improved the stability of ligand in active site. Compound D06 anchored in the binding site via six hydrogen bonds as shown in Fig. 12B. H-bond interactions of D06 were almost identical with D04, observed to be 1.92 Å (N–H⋯O
C–Glu826), 2.03 Å (N⋯H–N–Val828), 2.19 Å (N–H⋯O
C–Val828), 1.88 Å (N–H⋯O
C–Ser831), 2.70 Å (N–H⋯O–H–Asp832) and 2.87 Å (CN⋯H–O–Asp911), respectively. Moreover, the hydrophobic and electrostatic interactions of the designed molecules were similar with the molecule 16 as shown in Fig. S9.† Moreover, the designed compounds all adopted the desired propeller-like docking conformation which was requisite to improve PI3Kδ-selective inhibitory activity. Compared to the known potent inhibitors, the number of hydrogen bonds and the key residues in the binding pocket and the binding affinities of the designed molecules obviously increased. Hence, these designed compounds were anticipated to be candidate compounds for the relevant researchers' experimental synthesis.
![]() | ||
| Fig. 13 Root mean square deviations and total energy of protein–ligand complexes during 10 ns simulation time. | ||
Footnote |
| † Electronic supplementary information (ESI) available. See DOI: 10.1039/c7ra10870b |
| This journal is © The Royal Society of Chemistry 2017 |