Arabian Journal of Chemistry (2016) xxx, xxx–xxx

King Saud University Arabian Journal of Chemistry

www.ksu.edu.sa www.sciencedirect.com

ORIGINAL ARTICLE Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors

Garima Verma a, Mohemmed Faraz Khan a, Wasim Akhtar a, Mohammad Mumtaz Alam a, Mymoona Akhter a, Ozair Alam a, Syed Misbahul Hasan b, Mohammad Shaquiquzzaman a,* a and Lab, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard, New Delhi 110062, India b College of Pharmacy, Al Jouf University, PO Box-2994, Sakaka, Al Jouf, Saudi Arabia

Received 20 June 2016; accepted 17 September 2016

KEYWORDS Abstract Pharmacophore modeling, molecular docking and in silico ADME prediction have been 3D-QSAR; performed for quinazoline based EGFR inhibitors. This study has been carried out to determine Molecular docking; the binding mode and drug likeliness nature of compounds. A five point model (AAARR.7) was gen- Quinazoline; erated using 64 compounds. The generated model was found to be statistically significant as it had a Epidermal growth factor high correlation coefficient (R2 = 0.9433), cross validation coefficient (Q2 = 0.8493) and F value of receptor 97.10 at 6 component PLS factor. The results of external validation were also indicative of high pre- dictive power (R2 = 0.86). The generated model also passed Tropsha’s test for predictive ability and Y-randomization test. The Domain of Applicability (APD) of the model was also successfully defined to ascertain that a given prediction can be considered reliable. In order to evaluate the effectiveness of the docking protocol, co-crystallized was extracted from the ligand binding domain of the pro- tein and was re-docked into the same position. The conformer obtained on re-docking and the co- crystallized ligand were superimposed and the root mean square deviation between the two was found to be 1.005 A˚. Outcomes of this study provide an insight for designing novel EGFR inhibitors. Ó 2016 The Authors. Production and hosting by Elsevier B.V. on behalf of King Saud University. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Abbreviations: R2, regression coefficient; Q2, cross validation correlation coefficient; Gscore, Glide score; RMSD, root mean square deviation; A, hydrogen bond acceptor; R, aromatic ring; H, hydrophobic; PLS, partial least square; SD, standard deviation; EGFR, epidermal growth factor receptor; APD, domain of applicability * Corresponding author. E-mail address: [email protected] (M. Shaquiquzzaman). Peer review under responsibility of King Saud University.

Production and hosting by Elsevier http://dx.doi.org/10.1016/j.arabjc.2016.09.019 1878-5352 Ó 2016 The Authors. Production and hosting by Elsevier B.V. on behalf of King Saud University. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 2 G. Verma et al.

1. Introduction design process (Cichero et al., 2014, 2016, 2011a, 2011b; Cichero and Fossa, 2012). With these facts, we herein report the pharmacophore Cancer is one of the leading causes of death in both developing and modeling, 3D-QSAR, molecular docking and ADME prediction of developed countries (Han et al., 2016). The total number of new cancer quinazoline analogs to provide an insight into the key structural fea- cases is projected to increase from 14 million in 2012 to 24 million in tures required for designing EGFR targeting agents. 2035 (Liu et al., 2016). Despite the availability of a broad range com- pounds, cancer therapy still has many limitations. Side effects and 2. Materials and methods resistance to the current agents are the major ones (Pape et al., 2016). These factors necessitate the need for development of novel 2.1. Data set and safer anticancer agents. The Epidermal Growth Factor Receptor (EGFR), a member of ErbB family which signals through a tyrosine kinase (TK) is involved A set comprising of 64 compounds bearing quinazoline moiety in regulation of growth, migration and apoptosis of tumor cells (Yan were selected from the available literatures (Qin et al., 2016a, et al., 2015). It has been described in the nucleus of primary tumors 2016b; Qin et al., 2015). The selected compounds for the data (Rodrigues et al., 2016). Hence, it is considered as one of the important set shared the same assay procedure with significant variations targets for the development of novel anticancer agents. in their structures and profiles. Inhibitory potencies of EGFR inhibitors, such as gefitinib and erlotinib have made signif- the compounds included in data set, reported as IC50 values icant progress in the treatment of patients with cancer (Johnson et al., varied from 2.0 to 16,500 nM and were converted into molar 2005; Ciardiello and Tortora, 2008). Gefitinib and erlotinib inhibit the values. These were then converted into pIC values using tyrosine kinase activity by competitively binding to the adenosine 50 triphosphate (ATP) binding pocket of the intracellular EGFR TK the formula given below: domain, thereby blocking downstream signal transduction pathways ¼ ½ pIC50 log10 IC50 in cancer cells. However, the success of such EGFR tyrosine kinase inhibitors is limited by the up-regulation of bypass signaling pathways The 3D structures of ligands were generated using the builder and acquired point mutations (Oxnard et al., 2011). After treatment of panel in Maestro and subsequently optimized using LigPrep 10–16 months with these inhibitors, most of the cancer patients acquire module (v3.1, Schro¨ dinger 2016-1). Partial atomic charges an additional gate keeper mutation (T790M) (Kobayashi et al., 2005). were ascribed and possible ionization states were generated The secondary T790M mutation decreases sensitivity to gefitinib or at a pH of 7.0. The OPLS_2005 force field was used for opti- erlotinib and increases the binding affinity for ATP (Ou, 2012). In mization for production of low energy conformer of the ligand order to overcome the T790M mutation related resistance, several irre- (Shivakumar et al., 2010). The energy minimization was per- versible EGFR second-generation covalent inhibitors, which form a formed for each ligand till it reached a root mean square devi- covalent bond with Cys797 within the EGFR active site, have shown ˚ preclinical activity against EGFR with T790M mutation (Xia et al., ation (RMSD) cutoff of 0.01 A. The resulting structures were 2014). Afatinib was the first approved irreversible EGFR inhibitor then taken for performing modeling studies. In the current by the US FDA for the treatment of late-stage cancer patients with study, we used phase (v4.0) shape screening for flexible align- actively mutated EGFR. Other examples include canertinib and ment of the selected EGFR inhibitors. dacomitinib (Dungo and Keating, 2013). Structural information provided by the co-crystal structures of gefi- 2.2. Pharmacophore 3D-QSAR modeling tinib or erlotinib (Yun et al., 2007; Stamos et al., 2002) with EGFR has played a major role in rational design of potent EGFR inhibitors. Analysis of binding mode of gefitinib suggests various key interactions Phase (v4.6, Schro¨ dinger 2016-1) was used to generate phar- with the ATP binding pocket of EGFR, including the critical hydrogen macophore and 3D-QSAR models for EGFR inhibitors bond of the N1 of the quinazoline moiety with the hinge residue Met (Dixon et al., 2006). The prepared ligands were imported for 793 of active site, the deep hydrophobic pocket filled up by the 3- developing pharmacophore model panel of Phase with their chloro-4-fluoro aniline substituent and the solvent region occupation respective biological activity values. The ligands were assigned of 6-morpholinopropoxy group. A variety of polar functional groups as actives with a threshold of pIC50 > 7.7 and inactives with a are generally accommodated with a little affection on the efficacy by threshold of pIC50 < 6.0. Remaining compounds were consid- the solvent region (Yin et al., 2014). A good example of this strategy ered as moderately active. Phase (v4.0) has been proven to be is the success of icotinib, which mimics erlotinib with a crown ether an important tool for flexible ligand superposition (Sastry fused quinazoline (Liu et al., 2015; Guan et al., 2014). et al., 2011; Miller et al., 1999). In the current study, we used The EGFR kinase domain adopts a bilobate-fold and characteristic phase (v4.0) shape screening for flexible alignment of the NH2 terminal lobe (N-lobe) is composed of mainly b-strands and a- helix. COOH lobe (C-lobe) is mostly a-helical. These two lobes are sep- selected EGFR inhibitors. The most active compound 3 was arated by a cleft, similar to the one possessed by ATP. ATP analogs taken as the template and pharmacophore type volume scoring and ATP competitive inhibitors are known to bind in this cleft. Cat- was kept as default. The default settings were used, with a alytic machinery of N-lobe includes glycine-rich nucleotide phosphate maximum of 10 conformations per rotatable bonds and with binding loop (Gly 695 – Gly 700). C-lobe comprises of DFG motif this a maximum of 100 conformers were generated. Conforma- (Asp 831 – Gly 833), presumptive catalytic loop (Asp 813), the cat- tions of these compounds were varied and at most one align- alytic loop (Arg 812 – Asn 818) and the A-loop (Asp 831 – Val852). ment for every ligand was retained. However, Erlotinib, an approved EGFR inhibitor is found in the cleft The selection of training and test sets is the most important between the N- and C-terminal lobes (Stamos et al., 2002). and also the most difficult step, which often consumes much Emergence of resistance and side effects of the currently used agents further push the scientists across the globe to engage themselves time in building QSAR models. Random division is a widely in the development of novel EGFR targeting agents (Tang et al., 2013; used approach in establishing the QSAR model robustness Costa et al., 2008; Zheng et al., 2014). Nowadays, computational stud- (Wang et al., 2015). Therefore, for assigning training and test ies based on ligand- and structure-based approaches, are considered set for 64 compounds, random selection was made using the effective tools in medicinal chemistry, useful to accelerate the drug Automated Random Selection option present in Phase (v4.0)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 3 software. In this method, ten trials (70, 75, 80, 85 and 90%) is the result of a substantial extrapolation of the model and were run. The trials were based on structural diversity and a may not be reliable. wide range of activity – the range of biological activity of the APD can also be defined using similarity measurements test-set was similar to that of the training set. Thus, based on the Euclidean distances among all training and test the test set chosen was a true representative of the training set, compounds. The distance of a test compound to its nearest as per recommendations of Golbraikh and Tropsha, 2003. neighbor in the training set is compared to a predefined thresh- Resultantly, 20 compounds were included in the test set and old (APD), and the prediction is considered unreliable when remaining 44 compounds were included in training set. Phar- the distance is higher than that. APD was calculated based macophore sites for these compounds included default set of on the following formula: chemical features of Phase: three hydrogen bond acceptors ¼h iþ r (A) and two aromatic rings (R). Pharmacophore matching tol- APD d Z ˚ erance was set as 1 A. A total of 19 variants were generated by Calculation of hdi and r was performed as follows: first, the keeping 5 and 4 as the maximum and minimum number of average of Euclidean distances between all pairs of training sites respectively. It was specified that at least 9 of the actives compounds was calculated. Next, the set of distances that were must match. Finally, it resulted in 12 possible combinations of lower than the average was formulated. hdi and r were finally features which could give rise to common pharmacophores. calculated as the average and standard deviation of all dis- tances are included in this set. Z is an empirical cutoff value 2.3. Model validation and for this work, it was chosen as 0.5 (Zhang et al., 2006). Enalos Domain – similarity node that executes the aforemen- The QSAR model AAARR.7 with 6 component PLS factor tioned procedure was included in our workflow and used to was characterized as the best model. Pharmacophoric model assess domain of applicability of the proposed model. For was validated by its accuracy in prediction of the activity of the calculation of the domain of applicability, we have used training set ligands. The predicted EGFR inhibitory activity Enalos Domain KNIME nodes. of training set ligands elicited a correlation (R2) of 0.85 with observed EGFR inhibitory activity. The efficacy of model 2.5. Y-randomization test AAHR.11 was also examined with external validation (Table 1: Supplementary Material and Fig. 4). Graph of actual value vs. Y-Randomization technique ensures the robustness of a predicted value and residual value vs. predicted value was QSAR model (Tropsha et al., 2003; Shen et al., 2004). The plotted. dependent variable vector is randomly shuffled and a new The predictive power of the produced 3D-QSAR model QSAR model is developed, using the given modeling algo- was also assessed by using Enalos Model Acceptability Criteria rithm. The procedure is repeated several times and the new KNIME node (Melagraki and Afantitis, 2013), which includes QSAR models are expected to have low R2 and Q2 values. If all proposed tests by Tropsha (Zhang et al., 2006). Criteria fol- the opposite happens then an acceptable QSAR model cannot lowed in developing activity/property predictors, especially for be obtained for the specific modeling method and data. continuous QSAR, are as follows: (i) correlation coefficient R between the predicted and observed activities; (ii) coefficients 2 2.6. Molecular docking of determination (predicted versus observed activities R0 and 02 observed versus predicted activities R 0 for regressions through the origin); and (iii) slopes k and k0 of regression lines through The catalytic domain of EGFR enzyme in complex with erlo- the origin. The criterions for the acceptability of the QSAR tinib (PDB Code: 1M17 resolution 2.6 A˚) was obtained from model are represented in left column of Fig. 5. protein data bank (Stamos et al., 2002) and prepared using the protein preparation wizard (Sastry et al., 2013), available 2.4. Domain of applicability in Schro¨ dinger suite 2016-1. Crystallographic water molecules i.e. without 3H bonds were deleted and hydrogen bonds corre- sponding to pH 7 were added considering the appropriate - In order to screen a QSAR model for new compounds, its ization states for both acidic and basic amino acid residues. domain of application (APD)(Tropsha et al., 2003; Shen OPLS_2005 force field was used for energy minimization of et al., 2004) must be defined and predictions for only those the crystal structure (Shivakumar et al., 2010). Active site compounds that fall into this domain may be considered reli- was defined with a radius of 14 A˚around the ligand present able. Extent of Extrapolation (Tropsha et al., 2003) and Eucli- in crystal structure. Grid box was generated at a centroid of dean Distances are two simple approaches to define the APD. active site. For docking, low energy conformations of all the Extent of Extrapolation is based on the calculation of the compounds were docked into the catalytic domain of EGFR leverage (hi) for each chemical, where the QSAR model is used protein (PDB Code: 1M17) using Grid based Ligand Docking to predict its activity. with Energetics (Glide v7.0, Schro¨ dinger 2016-1) (Lipinski 1 et al., 2001) in extra precision mode without applying any con- h ¼ x ðXTXÞ xT i i i straints. The best docked structure was identified using Glide

In the above equation, xi is the row vector containing the k score function, Glide energy and Glide Emodel energy model parameters of the query compound and X is the n k (Table 2: Supplementary Material). The lowest energy docked matrix containing the k model parameters for each one of compound 3 was selected for further studies. A map of the n training compounds. A leverage value greater than hydrophobic and hydrophilic fields for inhibitor 3 was also 3k/n is considered large. It means that the predicted response generated (Fig. 8).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 4 G. Verma et al.

2.7. Lipinski’s rule for drug likeliness and in silico ADME and survival actives (Dixon et al., 2006), as shown in Table 1. prediction Based on the sites, a maximum of five features were allowed for developing the hypotheses. 3D-QSAR model was gener- We further predicted the drug-like behavior of the compounds ated using Partial Least Square (PLS) regression statistics ˚ through the analysis of pharmacokinetic profile of the com- and keeping the grid spacing of 1 A. The number of PLS fac- pounds by using QikProp module (v4.3, Schro¨ dinger 2016-1). tors included in model development is 6 because an incremen- The compounds prepared by LigPrep module (v3.1, Schro¨ din- tal increase in the statistical significance and predictivity was observed till 6 PLS factor (Table 2). ger 2016-1) were utilized for the calculation of pharmacoki- 2 2 netic parameters by QikProp v4.3. The program QikProp The best fitted model AAARR.7 (R = 0.9433, Q = 0.8493 v4.3, utilizes the method of Jorgensen19 to compute pharma- and F = 97.10) consists of three hydrogen bond acceptors and cokinetic properties and descriptors. Physically significant two aromatic ring features with highest survival score (6.202). descriptors and pharmaceutically relevant properties of all The hypothesis AAARR.7 is shown in Fig. 1. The distance the test compounds such as molecular weight, log p, H-bond and angles between different sites of the model AAARR.7 are donors, and H-bond acceptors according to the Lipinski’s rule presented in Tables 3 and 4 respectively. This is also apparent of five were analyzed. Lipinski’s rule of five is a rule of thumb from the comparison from the deduction of survival inactive to evaluate drug likeness, or determine whether a chemical from survival active. It deducts inactive features from the compound with a certain pharmacological or biological activ- hypothesis and was decisively highest for the hypothesis ity has properties that would make it a likely orally active drug AAARR.7. Among the pharmacophore features, intersite in humans. The rule describes molecular properties important distances and angles between site points were found to be the for drug in the human body, including its principal attribute and the point of difference between active absorption, distribution, metabolism and (ADME) and inactive is due to the interstitial site distances as evident (Cheng et al., 2014). by the pharmacophore hypothesis AAARR.7alignment over active (pIC50 > 7.7) and inactive compounds (pIC50 < 6.0) in Fig. 2a and b respectively. Using the model, AAARR.7, fitness 3. Results and discussion score of all the ligands was also evaluated (Table 5). 3.1. Pharmacophore and 3D-QSAR models 3.2. Model validation, domain of applicability and Y- randomization test Phase (v4.0) (Dixon et al., 2006) module of Schro¨ dinger 2016-1 was used for the development of pharmacophore model. Atom based 3D-QSAR unveiled the effect of substituents on activity. The predictability and validity of the common pharmacophore The generated hypotheses were scored and ranked in accor- model, AAARR.7 (test set), based on active compounds were 2 dance with their vector, volume, site scores, survival scores judged by cross validation coefficient (Q = 0.84) (Table 2).

Table 1 Score of different parameters of the hypotheses. S. No. Hypothesis Survival score Survival inactive Site Vector Matches Activity Inactive 1 AAARR.7 6.202 4.215 1.00 1.000 9 8.036 1.988 2 AAARR.25 5.816 4.310 0.68 0.859 9 8.036 1.507 3 AAARR.94 5.755 4.249 0.66 0.838 9 8.036 1.507 4 AAARR.23 5.444 3.929 0.50 0.797 9 7.963 1.514 5 AAARR.24 5.398 3.656 0.41 0.826 9 8.699 1.743 6 AAARR.19 5.388 3.664 0.47 0.797 9 7.963 1.723 7 AAARR.93 5.388 3.664 0.47 0.797 9 7.963 1.723 8 AAARR.95 5.354 3.356 0.44 0.779 9 8.174 1.998 9 AAARR.96 5.354 3.356 0.44 0.779 9 8.174 1.998 A: Acceptor; R: Aromatic ring.

Table 2 PLS statistical parameters of the model AAARR.7. PLS SD R2 FP Stability RMSE Q2 Pearson-R 1 0.6002 0.5030 40.50 1.459e007 0.9624 0.6358 0.5582 0.7619 2 0.5052 0.6568 37.30 8.758e010 0.9717 0.5333 0.6891 0.8370 3 0.3623 0.8280 61.00 1.373e014 0.8017 0.3744 0.8468 0.9320 4 0.3046 0.8816 68.90 1.237e016 0.7706 0.3791 0.8429 0.9260 5 0.2531 0.9205 83.30 9.158e019 0.7523 0.3804 0.8418 0.9222 6 0.2167 0.9433 97.10 2.468e020 0.7013 0.3713 0.8493 0.9244 SD: Standard deviation of regression;R2: Regression coefficient; F: Ratio of the model variance to the observed activity variance (variance ratio); P: Significance level of variance ratio; Q2: Cross validated correlation coefficient for the test set; RMSE: the RMS error in the test set predictions.

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 5

Figure 1 Hypothesis, AAARR.7. Pink spheres with arrow, hydrogen bond acceptor (A); orange open circle, aromatic ring (R).

Table 3 Angles between the pharmacophoric sites of AAARR.7. Table 4 Intersite distances between the pharmacophoric sites of AAARR.7. Entry Site 1 Site 2 Distance Entry Site 1 Site 2 Site 3 Angle AAARR.7 A3 A4 2.821 AAARR.7 A3 A5 4.800 AAARR.7 A4 A3 A5 28.5 AAARR.7 A3 R12 3.747 AAARR.7 A4 A3 R12 99.7 AAARR.7 A3 R13 2.798 AAARR.7 A4 A3 R13 59.5 AAARR.7 A4 A5 2.682 AAARR.7 A5 A3 R12 71.3 AAARR.7 A4 R12 5.056 AAARR.7 A5 A3 R13 31.0 AAARR.7 A4 R13 2.788 AAARR.7 R12 A3 R13 40.3 AAARR.7 A5 R12 5.055 AAARR.7 A3 A4 A5 121.4 AAARR.7 A5 R13 2.803 AAARR.7 A3 A4 R12 46.9 AAARR.7 R12 R13 2.422 AAARR.7 A3 A4 R13 59.9 AAARR.7 A5 A4 R12 74.6 AAARR.7 A5 A4 R13 61.6 AAARR.7 R12 A4 R13 13.0 Regression coefficient of the training set was 0.94, which AAARR.7 A3 A5 A4 30.1 exhibited relevance of the model. Stability of the generated AAARR.7 A3 A5 R12 44.6 model ranges from 0.7013 to 0.9624 on the maximum scale AAARR.7 A3 A5 R13 31.0 of 1. F value was found to be 97.10. Additionally, P value of AAARR.7 A4 A5 R12 74.7 AAARR.7 A4 A5 R13 61.1 2.468e020 and Pearson r of 0.9244 indicated greater degree AAARR.7 R12 A5 R13 13.6 of confidence on the model. Standard deviation (SD) value AAARR.7 A3 R12 A4 33.4 of 0.2167 and root mean square error (RMSE) of 0.3713 indi- AAARR.7 A3 R12 A5 64.1 cate the stability of the generated model for prediction of AAARR.7 A3 R12 R13 48.3 unknown compounds in the test. Scatter plots for experimental AAARR.7 A4 R12 A5 30.8 and predicted activities of ligands elicited significant linear cor- AAARR.7 A4 R12 R13 15.0 relation and moderate difference between the experimental and AAARR.7 A5 R12 R13 15.8 predicted values as shown in Fig. 3a and b. In order to evalu- AAARR.7 A3 R13 A4 60.7 ate the efficacy of the generated model, it was further validated AAARR.7 A3 R13 A5 118.0 using an external test set (Golbraikh and Tropsha, 2002a). The AAARR.7 A3 R13 R12 91.4 AAARR.7 A4 R13 A5 57.3 calculated pIC values of the compounds included in pre- 50 AAARR.7 A4 R13 R12 152.1 dicted and external test set are given in Table 5 and Table 1 AAARR.7 A5 R13 R12 150.6 (Supplementary Material) respectively. A plot of experimental vs. predicted pIC50 of external test set is shown in Fig. 4a and a

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 6 G. Verma et al.

Figure 2 (a) Mapping of the active compounds onto the pharmacophore. (b) Mapping of inactive compounds onto the pharmacphore.

plot of residual vs. predicted value is shown in Fig. 4b. These able and could be used in the design of new EGFR inhibitors two plots are vital for the predictive ability of QSAR. Scatter within this structural motif of molecules. residual plots were used to identify the outliers from the QSAR It is important that the limitations of the model are also model (Golbraikh and Tropsha, 2002b). Fig. 4b elicits that described via the APD. This gives an important indication as there is no outlier in the study. Thereby, the model is consid- the user can freely and creatively design novel molecules but ered as stable and as expected. It was able to sort the experi- will be warned for the reliability of the prediction when the mental pIC50 values for compounds in the external test set. structural characteristics cannot be tolerated by the model. Predictive correlation coefficient R2 value of 0.86 was observed After model validation, the APD of our model was also for the external test set for the developed QSAR model. An R2 defined to ascertain that a given prediction can be considered value of more than 0.5 between the predicted and experimental reliable. Enalos Domains – Similarity (Euclidean Distances) values renders the model to be good and able to predict the and Leverage (Extent of Extrapolation) nodes that execute inhibitory activity of compounds not included in the model the aforementioned procedure are included in our workflow development process (Golbraikh and Tropsha, 2002a, 2002b). and were used to assess APD of the proposed model. In Eucli- Furthermore, the Enalos Model Acceptability Criteria dean Distances method, the APD limit value was defined equal KNIME node has also been applied to the data. The model to 0.633, whereas in Extent of Extrapolation method, the APD passed Tropsha’s recommended tests for predictive ability limit was defined as 0.15. These limits were defined on the basis and the results are depicted in Fig. 5. These results suggest that of equations provided in Methods section. In both types of this alignment can effectively take into consideration the APD prediction, all compounds in the test set had values in ligand-receptor interactions, and the QSAR model is thus reli- the range of the APD. The predictions for all compounds that

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 7

Table 5 Calculated pIC50 for compounds in predicted set. Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness F Cl

N NH 1t 7.64 7.82 0.18 2.56 N O

O O

NNH 2 7.96 8.02 0.06 2.56 N O

O O F Cl

NNH 3t 8.69 8.26 0.40 3.00 N O

O O

N NH

4t N O 8.03 7.81 0.22 2.58

O O

O

N NH 5 N O 8.35 8.22 0.13 2.81

O O

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 8 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness

Cl

N NH 6 N O 8.17 8.32 0.15 2.59

O O

O- N+ O

N NH 7 8.01 8.10 0.09 2.98 N O

O O

F Cl

N NH

N 8t O 7.88 7.12 0.76 2.56

O O

O

N NH 9 N O 7.67 7.79 0.12 2.94

O O

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 9

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness F

N NH

N 10 O 6.69 6.90 0.21 2.57

O O

O

O

N NH N O 11 6.79 6.70 0.09 2.94 O O

O

F O- N+ O

N NH

12 N O 7.35 6.86 0.49 2.52

O O

O

O- N+ O

N NH

13t N O 7.12 6.84 0.28 2.74

O O

O

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 10 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness Cl O- N+ O

N NH

14t N O 7.44 6.82 0.62 2.53

O O

O

F N

N NH

15 N O 6.41 6.52 0.11 2.77

O O

O

F Cl

N NH

N O 16 7.61 7.46 0.15 2.83

O O

O

N NH N O 17 7.56 7.45 0.11 2.90 O O

O

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 11

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness F O- N+ O

N NH 18 7.08 7.30 0.22 2.53 N O

O O

O- N+ O

N NH 19t 6.73 7.32 0.59 2.94 N O

O O

Cl O- N+ O

N NH 20t 6.98 7.31 0.33 2.53 N O

O O

F N

N NH 21 6.50 6.88 0.38 2.78 N O

O O

N

N NH t 22 N O 6.38 7.12 0.74 2.96

O O

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 12 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness F Cl

N NH 23 7.53 7.51 0.02 2.57 N O

O O

N NH

24 N O 7.62 7.70 0.08 2.56

O O

F O- N+ O

N NH

25 N O 6.63 6.57 0.06 2.87

O O

O

-O N+ O

N NH

26 N O 6.44 6.52 0.08 2.88

O O

O

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 13

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness F N

N NH

27 N O 6.45 6.25 0.20 2.89

O O

O

F Cl

N NH t 28 N O 6.60 6.85 0.25 2.86

O O O

N NH

29 N O 6.32 6.66 0.34 2.76

O O O F Cl

N NH

t 30 N O 7.68 7.55 0.13 2.83

O O O

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 14 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness

N NH

31 N O 7.77 7.53 0.24 2.89

O O O F Cl

N NH

32 N O 7.23 7.05 0.18 2.55

O O

O

N NH N O 33t 7.23 7.24 0.01 2.54 O O

O

F Cl

N NH

N O

34t 7.58 7.49 0.09 2.66 O O

N

O

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 15

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness Br

N NH

N O

35 O 7.73 7.40 0.33 2.68 O

N

O Cl

N NH

N O

36 O 7.65 7.39 0.26 2.68 O

N

O F

N NH N O

37 O 7.07 7.44 0.37 2.68 O

N

O F F F

N NH N O

38 7.01 7.21 0.20 2.78 O O

N

O

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 16 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness O N

39 6.61 6.60 0.01 1.36 N O N F N NO H Cl O N

40 NH 7.20 7.20 0.00 1.39 N O N

N O Br O N

41t NH 7.04 7.19 0.15 1.39 N O N

N O O N

42 NH 6.72 6.84 0.12 1.37 N O N

N O F F F O N

43 NH 6.08 6.23 0.15 1.35 N O N

N O

O N

44 NH 6.85 6.57 0.28 1.36 N O N

N O Cl O F N

45 NH 7.27 6.94 0.33 1.36 N O N N O

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 17

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness O F N

46 NH 6.36 6.48 0.12 1.39 N O N

N O O Cl N

47t NH 6.45 6.39 0.06 1.37 N O N N O O Br N

48 NH 6.08 6.33 0.25 1.37 N O N

NO O N

49t NH 6.22 6.31 0.09 1.37 N O N NO O N

50 NH 6.28 6.17 0.11 1.38 N O N NO Cl O N

51 NH 6.78 6.62 0.16 1.36 N O N NO F NH+

52t NH 6.33 6.51 0.18 1.37 O N N NO

(continued on next page)

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 18 G. Verma et al.

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness Cl NH+

53 NH 6.93 6.77 0.16 1.37 N O N

N O Br NH+

54 NH 7.00 6.83 0.17 1.37 N O N

N O

NH+

55 NH 6.341 6.61 0.27 1.37 N O N

N O

NH+

56t NH 6.67 6.82 0.15 1.36 N O N N O Cl F NH +

57 NH 6.86 6.98 0.12 1.37 N O N NO F

O 58 O 4.98 4.89 0.09 1.97 N O N O

O N

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 19

Table 5 (continued) Ligand name Chemical structure Experimental activity Predicted activity Residual activity Fitness

O

t O 59 5.15 4.94 0.21 1.98 N O N O

O N

F

60 O 5.06 5.07 0.01 2.01 N O N O

O N

O 61 4.78 5.09 0.31 2.02 N O N O

O N

F F F

62 5.05 5.03 0.02 1.97 O N O N O

O N

O

63 O 4.99 5.05 0.06 2.00 N O N O

O N F

O 64t N O N O 5.00 5.58 0.58 1.098

O N

t test.

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 20 G. Verma et al.

Figure 4 (a) Plot of actual value vs. predicted value of external training set (y = 1.20x +(2.08), R2 = 0.86). (b) Plot of residual value vs predicted value.

Figure 3 Scatter plot of the observed versus phase-predicted activity for (a) training set (y = 0.94x + 0.39, R2 = 0.94) and (b) test set compounds with best fit line (y = 0.79x + 1.46, R2 = 0.85).

fell inside the domain of applicability of the model can be con- sidered reliable. Figure 5 Screenshot of results obtained by Enalos Model The model was further validated by applying Y- Acceptability Criteria KNIME node. randomization. Several random shuffles of the Y vector were performed and the low R2 and Q2 values that were obtained show that the good results in the original model use not due for developing the new QSAR model, including the selection of to a chance correlation or structural dependency of the train- the most appropriate descriptors. The results of the Y- ing set. It should be noted that for each random permutation randomization test are presented in Table 3 (Supplementary of the Y vector, the complete training procedure was followed Material).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 21

3.3. 3D-QSAR contour map analysis arise on the application of QSAR model to the most active and least active ligands. Contour plot analysis was performed to identify the effect of The hydrogen-bond donor nature for the most active com- spatial arrangement of structural features such as electrostatic, pound 3 (Fig. 6a) and the least active compound 61 (Fig. 6b) ionic, hydrophobic, H-bond donor and H-bond acceptor when compared showed their most favorable region with blue regions on antitubulin activity. Their individual positive con- color and unfavorable regions with red color. Hydrogen-bond tribution is shown in blue cubes and negative contribution is donor mapping revealed that favorable regions lied near the indicated by red cubes. Fig. 6a–e shows the comparison of amine group of aniline present at fourth position of quinazo- most significant favorable and unfavorable interactions, which line (Fig. 6a), indicating their importance for activity as com-

Figure 6 QSAR model visualized in the context of favorable and unfavorable hydrogen bond donor effects in (a) compound 3 (8.69) and (b) compound 61 (4.78). QSAR model visualized in the context of favorable and unfavorable hydrophobic interactions in (c) compound 3 (8.69) and (d) compound 61 (4.78). QSAR model visualized in the context of favorable and unfavorable electron withdrawing groups in (e) compound 3 (8.69) and (f) compound 61 (4.78).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 22 G. Verma et al. pared to the least active compound 61, devoid of amine group at the meta position of aniline attached to quinazoline. There at the same steric position (Fig. 6b). Therefore, the presence of is a big blue region at the aforementioned position indicating aniline with hydrogen donor amine group is vital for the preference of electron withdrawing group substitution over EGFR inhibitory activity. This assumption is further sup- it. This observation is consistent with trend of activity that ported by the less activity of non-aniline containing com- when the meta position of aniline attached to quinazoline is pounds 58–64, when compared to all other aniline containing substituted with electron-withdrawing group such as nitro or compounds 1–57. The presence of blue cubes near the oxygen cyano in compounds 12–15, the activity decreased dramati- (at A3 pharmacophoric feature) of dioxane ring attached to cally. Similarly, among compounds 18–29 substituted with sec- the fifth and sixth carbon position of quinazoline indicates ondary carbon chain at seventh position of quinazoline, its importance for activity as compared to the least active com- compounds 18–22 and 25–27, with the same electron with- pound 61, devoid of the ring in its backbone structure. drawing (nitro or cyano) substitution at meta position of ani- The 3-morpholinopropyl derivatives (39–42 and 44–45) line exhibited 10 times less potency against EGFR than the showed higher potency values than the related 3-(piperidin-1- most active compound 3. yl)propyl analogs (52–57). This indicates that the increased hydrophilicity of 6-position substituent of quinazoline may 3.4. Molecular docking be beneficial for enhancing the inhibitory activity against EGFR. The scores of docking studies are shown in Table 2 (Supple- The comparison between the electron withdrawing group mentary Material). From 63 listed total-scores, pIC of most effects of the most and least active compounds can be seen 50

Figure 7 (a) Binding mode of compound 3 in the catalytic pocket of 1M17. (b) 2D-ligand interaction diagram of 3 in the catalytic pocket of 1M17. (c) Hydrophobic interactions of 3 in the catalytic pocket of 1M17. (d) Overlay of docked pose (magenta) of Erlotinib with its crystal structure conformation (cyan).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 23 compounds keep in touch with the Glide score. Docking study (Supplementary Material). The optimum value of rotatable revealed that interactions were dominated by the hydrophobic- bonds (0–15) and polar surface area (7–200 A˚) holds a great ity and aromaticity due to the presence of quinazoline moiety. importance on the oral of the drug molecules. The interactions were dominated in the region of Met 769, Leu The active test quinazoline derivatives demonstrated results 694, Phe 699, Pro 770, Phe 771 and Cys 773 amino acid residue of the descriptors to be in the prescribed range thus owing content due to pronounced existence of active site in the region good bioavailability. Intestinal absorption or permeation is (Fig. 7a and b). In general, quinazoline ring exhibited also one of the important factors to be studied in concern with hydrophobic interactions with Ile 765, Ala 719, Met 769, the absorption of the drug , which was further con- Leu 694, and Leu 768 the key residues necessary for binding firmed by predicted Caco-2 cell permeability (QPPCaco), used of inhibitors (Fig. 7c). 1,4-Dioxane ring, attached to quinazo- as model for gut–blood barrier. Caco-2 cell permeability pre- line ring showed hydrophobic interactions with Phe 699, Val diction of the test compounds indicates excellent results pre- 702, Cys 773, and Phe 771. Phenyl ring substituted with fluoro and chloro groups, attached to quinazoline ring via NH link- age exhibited hydrophobic interactions with Phe 832, Leu 764, Leu 753, Met 742, Ile 720 and Ile 765. It also had polar interactions with Thr 830 and Thr 766 residues. A hydrogen bond of 2.14 A˚was also seen in case of quinazoline ring with Met 769, as shown in Fig. 7c with pink color. This is also evi- dent by analyzing the generated map of hydrophobic and hydrophilic fields for inhibitor 3 (Fig. 8), quinazoline moiety, a part of 1,4-dioxane, and phenyl ring are buried in the hydrophobic pocket (orange color) and some parts of 1,4- dioxane and phenyl ring are in hydrophilic pocket (cyan color). Our Glide XP-docking result also revealed solvent exposure in the region of 1,4-dioxane and ethoxy substitution plays an important role in stabilization of inhibitor at the active site. In addition, the accuracy of the docking procedure is deter- mined by examining how closely the lowest energy poses (bind- ing conformation) predicted by the object scoring function, and Glide score (GScore) almost resembles an experimental binding mode as determined by X-ray crystallography. The root mean square deviation (RMSD) between the predicted conformation and the observed X-ray crystallographic confor- mation of colchicines (PDB Code: 1M17) equal to 1.0057 A˚ Figure 8 Map of hydrophobic and hydrophilic fields for (Fig. 7d), a value that suggests the reliability of Glide XP dock- inhibitor 3 into the catalytic protein of EGFR (1M17). ing mode in reproducing the experimentally observed binding mode of EGFR inhibitors and the parameter set for Glide XP docking are reasonable to reproduce the X-ray structure. Further, similar orientation was observed between the super- position of conformation 3 best XP-docking pose and 3D- QSAR pose (RMSD: 1.005 A˚)(Fig. 9).

3.5. Lipinski’s rule for drug likeliness and in silico ADME prediction

Different pharmacokinetic parameters of the compounds taken for the study were calculated using ADME predictions by QikProp v4.3. The compounds were assessed for their basic parameters of Lipinski’s rule of 5 and other pharmacokinetic parameters. Table 4 (Supplementary Material) shows the results obtained from QikProp with their permissible range. In general, an orally active compound should not have more than 2 violations of the Lipinski rule. The active test com- pounds in present study were not found to violate the rule more than the maximum permissible limits and thus proving their drug likeness properties. The optimum values of the descriptors, polar surface area and rotatable bonds also have great influence on the oral bioavailability of the drug molecules. The important parame- Figure 9 Superimposition of conformations of inhibitor 3: best ters with their permissible ranges are delineated in Table 4 docking pose and pose of the AAARR.7 model (RMSD: 1.005 A˚).

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 24 G. Verma et al. dicting good intestinal absorption. Further, the test results for first mouse trace amine-associated receptor 5 (TAAR5) antago- QPlogkhsa descriptor of QikProp indicating the predicted val- nists. Med. Chem. Comm. 7, 353–364. ues of human serum albumin binding indicated that test mole- Cichero, E., Fossa, P., 2012. Docking-based 3D-QSAR analyses of cules were found to fall in the permissible range (1.5 to 1.5). pyrazole derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors. J. Mol. Model. 18, 1573–1582. Also, the QikProp descriptor for blood/brain partition coeffi- Cichero, E., Liqresti, A., Allara, M., Lazzati, Z., D’Ursi, P., cient QPlogBB showed reliable prediction for all the test com- Marabotti, A., Milanesi, L., Spallarossa, A., Ranise, A., Fossa, pounds and reference drugs. The cell permeability of the blood P., 2011b. Homology modeling in tandem with 3D-QSAR analyses: brain barrier mimic MDCK cells (QPPMDCK) also displayed a computational approach to depict the binding site of the reliable results falling in the prescribed range. The aqueous sol- human CB2 receptor. Eur. J. Med. Chem. 46, 4489–4505. ubility parameter (QPlog S) of the test entities was assessed Costa, D.B., Nguyen, K.S.H., Cho, B.C., Sequist, L.V., Jackman, D. and the compounds were also found to be in the permissible M., Riely, G.J., Yeap, B.Y., Halmos, B., Kim, J.H., Janne, P.A., range (<0.5). Huberman, M.S., Pao, W., Tenen, D.G., Kobayashi, S., 2008. Small effects of erlotinib in EGFR mutated non-small cell lung cancers with resistance to gefitinib. Clin. Cancer Res. 14, 7060– 4. Conclusion 7067. Dixon, S.I., Smondyrev, A.M., Knoll, E.H., Rao, S.N., Shaw, D.E., Combined computational approach was applied to give an insight into Friesner, R.A., 2006. PHASE: a new engine for pharmacophore the structural basis and inhibition mechanism for a series of quinazo- perception, 3D-QSAR model development, and 3D database line derivatives as EGFR inhibitors. 3D-QSAR modeling was per- screening: 1. Methodology and preliminary results. J. Comput. formed to provide a structural framework for understanding the Aided Mol. Des. 20, 647–671. structure activity relationship of these compounds. The atom based Dungo, R.T., Keating, G.M., 2013. Afatinib: first global approval. 3D-QSAR generated model AAARR.7 exhibited good correlation Drugs 73, 1503–15145. and predictive power and satisfactory agreement between experiment Golbraikh, A., Tropsha, A., 2002a. Bewqre of q2I. Mol. Gr. Model. and theory. Molecular docking studies were performed to produce pos- 20, 269–276. sible binding poses for these compounds to EGFR. The low value of Golbraikh, A., Tropsha, A., 2002b. Predictive QSAR modeling based RMSD between the initial complex structure and the energy minimized on diversity sampling of experimental datasets for the training and final average complex structure suggests that the derived docked com- test set selection. J. Comput. Aided Mol. Des. 5, 231–243. plex is close to equilibrium. Further, ADME predictions were per- Golbraikh, A., Tropsha, A., 2003. QSAR modeling using chirality formed for these compounds. descriptors derived from molecular topology. J. Chem. Inform. Comput. Sci. 43, 144–154. Conflict of interest Guan, Y., Zhao, H., Meng, J., Yan, X., Jiao, S., 2014. Dramatic response to high-dose icotinib in a lung adenocarcinoma patient The authors declare no competing financial interest. after erlotinib failure. Lung Cancer 83, 305–307. Han, X., Luo, J., Wu, F., Hou, X.Y., Yan, G., Zhou, M., Zhang, M., Pu, C., Li, R., 2016. Synthesis and biological evaluation of novel Acknowledgments 2,3-dihydrochromeno [3,4-d]imidazol-4(1H)-one derivatives as potent anticancer cell proliferation and migration agents. Eur. J. The authors are thankful to Jamia Hamdard, New Delhi. Med. Chem. 114, 232–243. Johnson, J.R., Cohen, M., Sridhara, R., Chen, Y.F., Williams, G.M., Appendix A. Supplementary material Gobburu, J., Booth, J., Benson, B., Leighton, K., Hsieh, J., Chidambaram, N., Zimmerman, P., Pazdur, R., 2005. Approval summary for erlotinib for treatment of patients with locally Supplementary data associated with this article can be found, advanced or metastatic non-small cell lung cancer after failure of in the online version, at http://dx.doi.org/10.1016/j.arabjc. at least one prior chemotherapy regimen. Clin. Cancer Res. 11, 2016.09.019. 6414–6421. Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A., Kocher, O., References Meyerson, M., Johnson, B.E., Eck, M.J., Tenen, D.G., Halmos, B., 2005. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792. Cheng, W., Yuan, Y., Qiu, N., Peng, P., Sheng, R., Hu, Y., 2014. Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 2001. Identification of novel 4-anilinoquinazline derivatives as potent Experimental and computational approaches to estimate solubility EGFR inhibitors both under normoxia and hypoxia. Bioorg. Med. and permeability in and development settings. Adv. Chem. 22, 6796–6805. Drug Deliv. Rev. 46, 3–26. Ciardiello, F., Tortora, G., 2008. EGFR antagonists in cancer Liu, C.J., Yu, S.L., Liu, Y.P., Dai, X.J., Wu, Y., Li, R.J., Tao, J.C., treatment. N. Engl. J. Med. 358, 1160–1174. 2016. Synthesis, cytotoxic activity evaluation and HQSAR study of Cichero, E., Buffa, L., Fossa, P., 2011a. 3,4,5-Trisubstituted-1,2,4-4H- novel isosteviol derivatives as potential anticancer agents. Eur. J. triazoles as WT and Y188L mutant HIV-1 non-nucleoside reverse Med. Chem. 115, 26–40. transcriptase inhibitors: docking-based CoMFA and CoMSIA Liu, D., Zhang, L., Wu, Y., Jiang, J., Tan, F., Wang, Y., Liu, Y., Hu, analyses. J. Mol. Model. 17, 1537–1550. P., 2015. Icotinib might be effective for the treatment of lep- Cichero, E., Espinoza, S., Franchini, S., Guarenito, S., Brasili, L., tomeningeal carcinomatosis in non-small cell lung cancer with Gainetdinov, R.R., Fossa, P., 2014. Further insights into the sensitive EGFR mutations. Lung Cancer 89, 268–273. of the human trace amine-associated receptors: Melagraki, G., Afantitis, A., 2013. Enalos KNIME nodes: exploring discovery of novel ligands for TAAR1 by a corrosion inhibition of steel in acidic medium. Chem. Intel. Lab. approach. Chem. Biol. Drug Des. 84, 712–720. Syst. 123, 9–14. Cichero, E., Espinoza, S., Tonelli, M., Franchini, S., Gerasimov, A.S., Miller, M.D., Sheridan, S.K., Kearsley, S.K., 1999. SQ: a program for Sorbi, S., Gainetdinov, R.R., Brasili, L., Fossa, P., 2016. A rapidly producing pharmacophorically relevant molecular super- homology modelling-driven study leading to the discovery of the positions. J. Med. Chem. 42, 1505–1514.

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019 Quinazoline based EGFR inhibitors 25

Ou, S.H., 2012. Second-generation irreversible epidermal growth using molecular dynamics free energy perturbation and the OPLS factor receptor (EGFR) tyrosine kinase inhibitors (TKIs): a better force field. J. Chem. Theory Comput. 6, 1509–1519. mousetrap? A review of the clinical evidence. Crit. Rev. Oncol. Stamos, J., Sliwkowski, M.X., Eigenbrot, C., 2002. Structure of the Hematol. 83, 407–421. epidermal growth factor receptor kinase domain alone and in Oxnard, G.R., Arcila, M.E., Chmielecki, J., Ladanyi, M., Miller, V.A., complex with a 4-anilinoquinazoline inhibitor. J. Biol. Chem. 277, Pao, W., 2011. New strategies in overcoming acquired resistance to 46265–46272. epidermal growth factor receptor tyrosine kinase inhibitors in lung Tang, J., Salama, R., Gadgeel, S.M., Sarkar, F.H., Ahmad, A., 2013. cancer. Clin. Cancer Res. 17, 5530–5537. Erlotinib resistance in lung cancer: current progress and future Pape, V.F.S., Toth, S., Furedi, A., Szebenyi, K., Lovrics, A., Szabo, P., perspectives. Front. Pharmacol. 4, 1–9. Wiese, M., Szakacs, G., 2016. Design, synthesis and biological Tropsha, A., Gramatica, P., Gombar, V.K., 2003. The importance of evaluation of thiosemicarbazones, hydrazinobenzothiazoles and being earnest: validation is the absolute essential for successful arylhydrazones as anticancer agents with a potential to overcome application and interpretation of QSPR models. QSAR Comb. Sci. multidrug resistance. Eur. J. Med. Chem. 117, 335–354. 22, 69–77. Qin, X., Han, X., Hu, L., Li, Z., Geng, Z., Wang, Z., Zeng, C., Xiao, Wang, J., Li, Y., Yang, Y., Zhang, J., Du, J., Zhang, S., Yang, L., X., 2015. Design, synthesis and biological evaluation of Quinox- 2015. Profiling the interaction mechanism of indole based deriva- alin-2(1H)-one derivatives as EGFR tyrosine kinase inhibitors. tives targeting the HIV-1 gp120 receptor. RSC Adv. 5, 78278– Anticancer Agents Med. Chem. 15, 267–273. 78298. Qin, X., Li, Z., Yang, L., Liu, P., Hu, L., Zeng, C., Pan, Z., 2016a. Xia, G., Chen, C., Zhang, J., Shao, J., Zhang, Y., Huang, W., Zhang, Discovery of new [1,4]dioxino[2,3-f]quinazoline-based inhibitors of L., Qi, W., Sun, X., Li, B., Xiang, Z., Ma, C., Xu, J., Deng, H., Li, EGFR including the T790M/L858R mutant. Bioorg. Med. Chem. Y., Li, P., Miao, H., Han, J., Liu, Y., Shen, J., Yu, Y., 2014. A http://dx.doi.org/10.1016/j.bmc.2016.01.003. chemical tuned strategy to develop novel irreversible EGFR-TK Qin, X., Lv, Y., Liu, P., Li, Z., Hu, L., Zeng, C., Yang, L., 2016b. inhibitors with improved safety and pharmacokinetic profiles. J. Novel morpholin-3-one fused quinazoline derivatives as tyrosine Med. Chem. 57, 9889–9900. kinase inhibitors. Bioorg. Med. Chem. Lett. 26, 1571–1575. Yan, Y., Xiao, Z.Y., Song, Y., Kang, Z.T., Wang, P., Sun, X.L., 2015. Rodrigues, M.A., Gamba, C.O., Faria, J.A.Q.A., Ferreira, E., Goes, 99mTc-HYNIC-MPG: a novel SPECT probe for targeting mutated A.M., Gomes, D.A., Cassali, G.D., 2016. Inner nucleus membrane EGFR. Bioorg. Med. Chem. Lett. 25, 1647–1652. localization of epidermal growth factor receptor (EGFR) in Yin, K.-H., Hsieh, Y.-H., Sulake, R.S., Wang, S.-P., Chao, J.-I., Chen, spontaneous canine model of invasive micropapillary carcinoma C.P., 2014. Optimization of gefitinib analogues with potent of the mammary gland. Pathol. Res. Pract. 212, 340–344. anticancer activity. Bioorg. Med. Chem. Lett. 2014 (24), 5247–5250. Sastry, G.M., Adzhigirey, M., Day, T., Annabhimoju, R., Sherman, Yun, C.H., Boggon, T.J., Li, Y., Woo, M.S., Greulich, H., Meyerson, W., 2013. Protein and ligand preparation: parameters, protocols M., Eck, M.J., 2007. Structures of lung cancer-derived EGFR and influence on virtual screening enrichments. J. Comput. Aided mutants and inhibitor complexes: mechanism of activation and Mol. Des. 27, 221–234. insights into differential inhibitor sensitivity. Cancer Cell 11, 217– Sastry, G.M., Dixon, S.L., Sherman, W., 2011. Rapid shape-based 227. ligand alignment and virtual screening method based on atom/ Zhang, S., Golbraikh, A., Oloff, S., Kohn, H., Tropsha, A., 2006. A feature-pair similarities and volume overlap scoring. J. Chem. Inf. novel automated lazy learning QSAR (ALL-QSAR) approach: Model. 51, 2455–2466. method development, applications, and virtual screening of chem- Shen, M., Beguin, C., Golbraikh, A., Stables, J., Kohn, H., Tropsha, ical databases using validated ALL-QSAR models. J. Chem. Inf. A., 2004. Application of predictive QSAR models to database Model. 46, 1984–1995. mining: identification and experimental validation of novel anti- Zheng, H., Wang, Q., Shi, H., Zhang, H., Hu, F., Li, B., 2014. convulsant compounds. J. Med. Chem. 47, 2356–2364. Favorable response to icotinib in a lung cancer patient with a Shivakumar, D., Williams, J., Wu, Y., Damm, W., Shelley, J., special mutation at exon 19 of eoidermal growth factor receptor. Sherman, W., 2010. Prediction of absolute solvation free energies Thorac. Cancer 5, 358–361.

Please cite this article in press as: Verma, G. et al., Pharmacophore modeling, 3D-QSAR, docking and ADME prediction of quinazoline based EGFR inhibitors. Arabian Journal of Chemistry (2016), http://dx.doi.org/10.1016/j.arabjc.2016.09.019