Skip to main content

In silico insights into the design of novel NR2B-selective NMDA receptor antagonists: QSAR modeling, ADME-toxicity predictions, molecular docking, and molecular dynamics investigations

Abstract

Based on a structural family of thirty-two NR2B-selective N-Methyl-D-Aspartate receptor (NMDAR) antagonists, two phenylpiperazine derivatives labeled C37 and C39 were conceived thanks to molecular modeling techniques, as novel NMDAR inhibitors exhibiting the highest analgesic activities (of pIC50 order) against neuropathic pain, with excellent ADME-toxicity profiles, and good levels of molecular stability towards the targeted protein of NMDA receptor. Initially, the quantitative structure-activity relationships (QSARs) models were developed using multiple linear regression (MLR), partial least square regression (PLSR), multiple non-linear regression (MNLR), and artificial neural network (ANN) techniques, revealing that analgesic activity was strongly correlated with dipole moment, octanol/water partition coefficient, Oxygen mass percentage, electronegativity, and energy of the lowest unoccupied molecular orbital, whose the correlation coefficients of generated models were: 0.860, 0.758, 0.885 and 0.977, respectively. The predictive capacity of each model was evaluated by an external validation with correlation coefficients of 0.703, 0.851, 0.778, and 0.981 respectively, followed by a cross-validation technique with the leave-one-out procedure (CVLOO) with Q2cv of 0.785, more than Y-randomization test, and applicability domain (AD), in addition to Fisher’s and Student’s statistical tests. Thereafter, ten novel molecules were designed based on MLR QSAR model, then predicted with their ADME-Toxicity profiles and subsequently examined for their similarity to the drug candidates. Finally, two of the most active compounds (C37 and C39) were chosen for molecular docking and molecular dynamics (MD) investigations during 100 ns of MD simulation time in complex with the targeted protein of NMDA receptor (5EWJ.pdb).

Peer Review reports

Introduction

N-methyl-D-aspartate (NMDA) receptors are part of ionotropic receptors physiologically activated by glutamate and glycine, which play an essential role in learning, memory, and synaptic plasticity in the central nervous system (CNS). They are mainly known for their permeability to divalent cations of calcium (Ca2+) and monovalent cations of potassium (K+) and sodium (Na+) in membrane cells, due to their structural nature as a tetrameric combination of two GluN1subunits linked to glycine and two GluN2 subunits fixed to glutamate sites, in which the distribution of each NMDA subunit is closely associated with neuronal plasticity and synaptic deficiencies [1]. Recently, it was discovered as a potential therapeutic target for neurological and mental diseases, including memory impairments, chronic pain, schizophrenia, Parkinson’s, and Alzheimer’s disorders [2,3,4,5]. In this respect, the discovery of powerful NMDA receptor antagonists is an absolute necessity for treating this class of neurodegenerative disorders, which requires a selective activation of each blocked receptor which occurs more frequently for older cells than younger ones.

Nowadays, the computer-assisted drug design (CADD) approach based on in silico techniques has taken on enormous importance in the pharmaceutical sector, aimed at discovering new drug candidates [6,7,8]. It has been widely reported in the literature to save the time and costs of experimentation before any pre-clinical and clinical tests. However, in vivo and in vitro tests remain strongly recommended for each new compound designed based on in silico studies [9,10,11,12]. Many scientific articles were focused on NMDA receptors as a major neurotransmitter in the human brain, closely involved in the excitotoxicity process responsible for the pathophysiology of various diseases, to concept the new competitive and non-competitive NMDA receptor antagonists, which could be recommended as an effective treatment for several neurological impairments such as Alzheimer’s, cerebral vascular accidents, and chronic pain [13,14,15,16].

The present work aims to concept novel NR2B-selective NMDAR antagonists using in-silico techniques based on computer molecular modeling, including QSAR modeling, ADME-Tox predictions, molecular docking, and molecular dynamics investigations.

In the first stage, QSAR technique was carried out to a structural class of thirty-two selective inhibitors of NMDA receptor, acting as analgesic agents against neuropathic pain [5, 17], in which four QSAR models were generated using MLR, PLSR, MNLR, and ANN techniques to predict the linear and nonlinear relationship between various molecular descriptors of NR2B-selective NMDAR antagonists and their analgesic activities of pIC50 order [18, 19]. The developed models were applied on a training set of twenty-six molecules and then validated on a test set of six molecules [20]. Thereafter, ten novel molecules (C33 to C42) were designed based on the most active compound (C22) and predicted in humans by their absorption, distribution, metabolism, excretion, and toxicity (ADMET), then examined for their similarity to the drug candidates [21]. In the second stage, two novel-designed molecules labeled C37 and C39, were chosen for molecular docking to study their inhibition mechanisms towards NMDA receptor, encoded in protein data bank by 5EWJ.pdb [22]. In the final stage, C37 and C39 molecules, which were predicted to have greater analgesic activity than the most active compound C22, were chosen for the molecular dynamics (MD) technique to test the thermodynamic stability of each (ligand-protein) complex [23, 24], which was equally compared to the original compound (C22) over 100 nanoseconds of MD simulation time after being complexed to the same protein target.

Materials and methods

Experimental database

A structural family of 32 NMDA receptor antagonists, which were successfully examined using experimental in vivo and in vitro assays to block the binding of [3 H]-ifenprodil to rat brain membranes as authored by Kosuke Anan, et al. [17], was chosen as an appropriate database to concept new active molecules with the highest inhibitory activities using molecular modeling techniques, where the analgesic activities against neuropathic pain of the original molecules (C1 to C32) were expressed on a decimal logarithm scale (pIC50 = - log10IC50), as presented in Table 1.

Table 1 Studied molecules and their observed activities pIC50

Molecular descriptors calculation

To establish a reliable QSAR model, we have calculated a variety of molecular descriptors as classified in Table 2, where the thermodynamic and physicochemical descriptors were calculated by MM2 method using ChemBio3D software [25], the constitutional descriptors were calculated by ACD/chemsketch software [26], at the same time the geometry of the studied compounds was optimized using the density functional theory (DFT) with the B3LYP functional [27], combined with the 6-31G + G(d, p) basis, thus the quantum descriptors were calculated with the help of Gaussian 09 package software [28].

Table 2 List of different calculated descriptors

Statistical methods

To generate the mathematical models that exhibit the quantitative relationships between the biological activity and the calculated molecular descriptors, different statistical methods were used such as Multiple Linear Regression (MLR), Partial Least Squares Regression (PLSR), Multiple Non-Linear Regression (MNLR) and Artificial Neural Network (ANN).

As a first step, the number of molecular descriptors was minimized using the Principal Component Analysis (PCA) by XLSTAT 2014 software, as a very important step to make the studied information less redundant, because it serves to reduce the size of the original variables that are correlated with each other into some synthetic variables that are independent from one another, called the principal components or factorial axes [29,30,31].

As a second step, the linear variation of these independent variables (uncorrelated descriptors) with the dependent variable (analgesic activity), was tested using MLR method with stepwise selection. In this case, where there are more predictors than observations and there is strong collinearity between these predictors, the PLS method is the best and it would be interesting to be able to study more precisely the quality of the model by minimizing the difference between the observed and calculated values [32]. Finally, the molecular descriptors used were chosen as input parameters in the multiple nonlinear regression (MNLR) and artificial neural network (ANN). All four models obtained were generated to predict the effects of these NMDA antagonists on the rat brain. Each tested model was treated according to the following statistical criteria: R, R2, and R2adj that must tend towards 1, which mean respectively: correlation coefficient, determination coefficient, and adjusted coefficient. Mean square error (MSE) should be minimal (tend towards 0), the Fisher value (F) must be inferior to the critical value of Fisher, and the probability value (p-value), it’s better to be under 5% for a 95% confidence level. To make these four QSAR models applicable, we have used the external validation technique on six new compounds constituting the test set. Also, we have performed the cross-validation method with the “leave-one-out” procedure, in order to examine the reliability of the developed models. So, the validity of the performance estimate will be obtained by performing this conventional procedure, which is realized by removing a single example from the training base, applied each time on the N-1 compounds [33]. In addition to cross-validation method, two other techniques are very important: the first one is “Y-randomization” technique; this sort of approaches is particularly appropriate when we are not sure of our model of the data generation process [34]. To do this, we were based on our full set of 32 compounds, after that we have searched randomly a new training test of 26 molecules to validate and ensure the security of the statistical tests [35].

Drug likness and in-silico ADMET predictions

To examine the drug likness of novel chemical compound, the physicochemical feautures must satisfy at least two of Lipinski’s five rules, with an acceptable ADMET profile, and good oral biovailability by the human body, without any adverse toxic effects on the human body. For this purpose, SwissADMET, pKCSM, and CLC Drug Discovery servers were carefully used to test the physicochemical and pharmacokinetic profiles [36, 37] of nine chemical compounds designed based on QSAR results.

Molecular docking modeling

Molecular docking is often used in computational chemistry to accelerate drug discovery at early stages [9]. For this project, this molecular modeling technique is based on the cell key phenomenon, where the best position of the ligand or drug candidate (the agent) is the key that can open the cell (or protein) to have finally a more stable complex by energetic order [38].

The crystal structure of amino-terminal domains of the NMDA receptor subunit GluN1 and GluN2B in complex with ifenprodil, coded in protein data bank by 5EWJ.pdb (https://www.rcsb.org/structure/5EWJ) was chosen as the targeted protein of NMDA receptor, in which can complex the inhibitors studied [39]. The responsible protein was discovered using X-ray diffraction method, in machine simulation with a resolution equal to 2.77 Å [40]. Then, it was prepared by removing all the water molecules bound, the sodium atom, and all the suspended ligands were removed, while adding the polar hydrogens using the Discovery Studio software, for the reason that the cavity method works best [41]. All this is to indicate the map coordinates of the co-crystallized ligand. After the preparation of the protein and quoting its active site, we have launched the docking calculation in AutoDock 4.2 program. In this way we have docked in the protein receptor three molecules already optimized by the DFT theory (output file): the first one is noted C22, having the greatest analgesic activity against neuropathic disease of pIC50 = 7.9, and the second ones are the new predicted molecules (C37 and C39) to compare their mechanisms of inhibition towards the most active compound. With the help of algorithm AUTOGRID We were able to centralize the grid boxes on (86.63 Å, -7.51 Å, -64.01 Å) by putting the sizes: 110, 110, and 110 in their three-dimensional structure, and running 10 genetic algorithms, with a total of two million five hundred thousand evals. At the end, we have got the strongest complex out of fifty conformations obtained [42], and we have visualized the 2D and 3D interactions of the protein-ligand by discovery studio 2020 [43].

Molecular dynamics simulation

To validate the thermodynamic stability of ligands C37, C39, and standard drug C22 with the NMDA receptor for their analgesic effect were shortlisted for performing MD simulation based upon their docking score, physicochemical analysis and observed chemical interactions with the target receptors. All the above said MD simulations were executed for a time period of 100 ns by using Desmond module of Schrodinger’s Maestro software [44]. Addition of explicit solvent molecules followed by their neutralization by adding the respective ions. The steepest-descent algorithm was used to relax the system and eliminate any steric clashes or poor contacts within atoms in order to minimize the system’s energy. Using a short series having low temperature with constant pressure (NPT) simulations, the system was brought to equilibrium. Positional constraints are applied to the system in addition to a progressive increase in temperature [45,46,47]. This makes it more likely that the system will be in a stable, balanced state prior to the simulation, in order to get the appropriate outcomes, the simulation is performed for 100 ns while taking into account the system’s energies, atom positions, and RMSD values. This aids in comprehending the system’s dynamic behavior and provides long-term intuitions on the complex’s structure and functional stability [24, 48,49,50].

Results and discussions

Statistical database

To establish the QSAR mathematical models, we have selected a set of 32 compounds from recently published work of K.Anan, et al., including NR2B-selective antagonists that Inhibit binding of [3 H]-ifenprodil to rat brain membranes [17]. Therefore, the database will be represented as a matrix of 32 rows (active molecules) and 14 columns (molecular descriptors) as illustrated in Table S1. Then, we worked randomly to divide this complete set into two subsets: the first one (training set) includes 26 compounds that are used to build the model and the second one (test set) includes 6 compounds to validate this established model.

Principal component analysis

To reduce the size of the basic data, we have applied the principal component analysis method, calculating a number of linear combinations of the original variables such a way to summarize the data with minimal loss of information, focusing on the correlation matrix given in Table S2 [51].

According to the correlation matrix, the variables that have a correlation coefficient absolutely higher than 90% are strongly correlated. So, we have found 9 following correlations:

First, the density is strongly and negatively correlated with % C and % H of -94.5% and − 96.1% respectively, also a strong positive correlation of 94.7% between % C and % H, this means that the three are correlated with each other several times. Second, it was observed that: the potential energy (P), kinetic energy (K) and the total energy (T), are totally correlated because the correlation coefficient is equal to 1 in absolute value. Third, E Homo, E gap and hardness are strongly correlated with each other, such as: r(E homo, E gap)= -90.2%, r(E homo, ƞ)= -90.4% and r(ƞ, E gap) = 99.8%. Finally, we have dropped these nine descriptors because every three variables share the same information, as they are largely correlated with each other. Thus, we have limited our study to five major uncorrelated descriptors pictured in Fig. 1.

Fig. 1
figure 1

The projection of the uncorrelated variables on the plane of the first two principal components t1 and t2

These descriptors are projected on the first two principal components t1 and t2. Since, 46.074% of the studied variables are explained by the first principal component, 24.901% of the variables contribute to the construction of the second and only 11.441% of the variables contribute to the construction of the third principal component. Therefore, we can propose that only the first two principal components are sufficient to obtain a good explanation of the data, because the variability explained by the first two principal components is: 70.97% of the total variability, as illustrated in Fig. 2.

Fig. 2
figure 2

The explanatory power of the principal components

Multiple linear regression

The Multiple Linear Regression is a statistical technique for estimating the relationship between more than two variables which have cause-effect relations [52]. For that, we have applied this technique on the training test including twenty-six observations (N = 26) and we have found the best following QSAR model as illustrated in the following equation:

$$\begin{aligned}\text{pIC}_{50}&=-5.031+42.028\ast\text{E\;lumo}+65.706\ast{\chi}-0.228\ast{\mu}\\&\;+1.308\ast \text{Log\;P}+0.244\ast\%\text{O} \end{aligned}$$
(1)

This established model shows that biological activity, is a quantitative variable significantly influenced by the following five descriptors: E lumo, χ, µ, Log P and % O, because the probability corresponding to the slope of each variable is less than 5% as noted in Table 3.

Table 3 Significance test of the slopes

To know, all the variables have a positive weight on the biological activity except the dipole moment, as shown in Fig. 3, because each increase in activity is accompanied by an increase of 42.028 kcal/mol in the energy of the lowest unoccupied molecular orbital, 65.706 in the electronegativity of the compound, 1.308 log P (Hight lipophilicity), and 0.244% of oxygen, but results in a decrease of 0.228 in the dipole moment. For a 95% confidence interval, the null hypothesis (H0) posed by the Fisher statistical test is rejected, because the calculated Fisher value (F = 24.664) is much higher than the critical value: [F (26, 5) = 2.59, p < 0.0001], as simplified by the one ANOVA test (Table 4). So, the variance is homogeneous between the dependent variable (pIC50) and the five explicative variables. In addition, the determination, correlation, and adjustment coefficients of R = 0.928, R²=0.860 and R²adjusted = 0.826, show a strong relationship between descriptors and answers. So, the first QSAR model obtained by MLR has good predictive competence, with a minimal standard error (RMSE = 0.272).

Fig. 3
figure 3

Degree of influence of the descriptors on the activity

Table 4 Analysis of variance (ANOVA 1)

Partial least squares regression (PLSR)

To model a linear relationship between a set of predictors and a response variable, so that the variance is minimal, we have applied the partial least squares regression technique, in a way that solves linear regression problems and to have finally a good quality of adjustment, and a good predictive power [53, 54]. The results of this regression technique yield the second QSAR model given in the following Eq. 2:

$$\begin{aligned}\text{pIC}_{50}&=-3.696-20.573\ast\text{E\;lumo}+37.531\ast\text{X}+2.353\text{E-02}\ast{\mu}\\&\;+1.229\ast \text{Log\;P}+0.121\ast\%\text{O}. \end{aligned}$$
(2)

This model generated by the PLSR technique, is also applied on the same training set of twenty-six observations (N = 26) and given by the following statistical criteria: the standard error (RMSE = 0.314) is minimal, the determination and correlation coefficients which are respectively: R = 0.870 and =0.758, show that there is a good quality of adjustment and a good predictive power of the model made by the regression on the partial least squares.

Multiple non-linear regression (MNLR)

Non-linear regression analysis is a type of regression analysis in which the data are modeled by a non-linear combination of several independent variables [55]. To realize this technique, we have used preprogrammed function of type:

$$\:Y=a0+{\sum\:}_{i=1}^{n}\left(ai*Xi+bi*{Xi}^{2}\right)$$
(3)

As: Y: the predicted biological activity (pIC50), Xi: the explicative variable, a0: the constant of the QSAR model, a.i.: the slope of each descriptor to one degree and bi: the slope of each descriptor of two degrees. This function presents the non-linear combination of the biological activity (pIC50) as a function of the five independent variables found previously by the linear model QSAR, based on the same training set of twenty-six observations (N = 26). So, the final results produce the third QSAR model as indicated in the following Eq. 4:

$$\begin{aligned}\text{pIC}_{50}&=23.360+64.956\ast\text{E\;lumo}-354.817\ast\text{X}-0.360\ast{\mu}\\&+3.680\ast\log\;\text{P}-0.166\ast\%\;\text{O}+297.460\ast\;\text{E\;lumo}^{2}\\&+1599.438\ast\text{X}^{2}+2.258\text{E-2}\ast\mu^{2}\\&-0.550\ast\log\;\text{P}^{2}+1.461\text{E-2}\ast\%\text{O}^{2}\end{aligned}$$
(4)

This mathematical model is defined by a good coefficient of determination (R = 0.941) and a good correlation coefficient (R2 = 0.885), in addition the model Root Mean Square Error is minimal (RMSE = 0.285), this means that the last QSAR model generated by the MNLR technique has a good predictive competence and a strong non-linear relationship between descriptors and answer.

Artificial neural network (ANN)

One of the most important steps in QSAR studies is to develop a non-linear relationship between activity and molecular descriptors, using the ANN method [56], which differs from the function of biological neurons in a number of parameters such as structures, layers, computational style, processing speed, connections, strength, storage and transmission of information [57]. In the present study, we have used three-layer neural networks, the input layer contains 5 neurons representing the selected descriptors, the hidden layer contains 3 neurons (H1, H2 and H3), and the output layer represents the observed activity values (pIC50). A parameter ρ serves a major role in the determination of the best ANN architecture, is used to identify the number of hidden neurons [58], which must be in the interval: 1 < ρ < 3 [59]. It’s given by the following Eq. 5:

$${\rho}=\frac{\:\text{T}\text{h}\text{e}\:\text{n}\text{u}\text{m}\text{b}\text{e}\text{r}\:\text{o}\text{f}\:\text{w}\text{e}\text{i}\text{g}\text{h}\text{t}}{\text{T}\text{h}\text{e}\:\text{n}\text{u}\text{m}\text{b}\text{e}\text{r}\:\text{o}\text{f}\:\text{c}\text{o}\text{n}\text{n}\text{e}\text{c}\text{t}\text{i}\text{o}\text{n}\text{s}\:\text{i}\text{n}\:\text{t}\text{h}\text{e}\:\text{N}\text{N}}$$
(5)

So, the architecture of the Artificial Neural Networks used in this work is [5–3–1], as shown in Fig. 4.

Fig. 4
figure 4

Schematic diagram of three-layer artificial neural network

The obtained results of the artificial neural network, using the JMP version 8.0 software, show a very good correlation of 97.7% between the observed and predicted values, with a minimal coded residual standard error equal to 0.189 and an external validation correlation coefficient of 98.1%, operating on 16 turns and 2.734 of over-adjustment penalty, with 75 maximum iterations, and 0.00001 of convergence criteria. This indicates that the descriptors selected by multiple linear regression, are relevant, and the model has a high statistical quality.

Validation techniques

External validation

To evaluate the accuracy of four predictive models and ensure their generalizability, it is essential to validate them externally before they are applied in clinical practice [60]. For this purpose, we have tested the six new molecules constituting the test set on a training test basis and we have arrived at the results presented in Table 5.

Table 5 The results of external validation by the MLR, PLSR and MNLR methods

Based on the set test (N = 6), the first regression established by MLR method and illustrated in Fig. 5, results an external validation correlation coefficient (R2ext = 0.703) between the observed and predicted activities, with an adjustment coefficient (R2adjusted = 0.629) and a minimal root mean square error (RMSE = 0.460). The second regression shown in Fig. 6, gives an external validation correlation coefficient (R2ext = 0.851) between the observed and predicted activities, with a good adjustment coefficient (R2adjusted = 0.814) and a minimal root mean square error (RMSE = 0.326). The third regression as presented in Fig. 7, gives an external validation correlation coefficient (R2ext = 0.778) between the observed and predicted activities, with a good adjustment coefficient (R2adjusted = 0.722) and a minimal root mean square error (RMSE = 0.398) and the last regression, results an external validation correlation coefficient of 98.1%. According to ‘Alexander Golbraikh and Alexander Tropsha’ theory, an external validation correlation coefficient greater than 0.6 shows that the established QSAR model is externally validated. Consequently, we can say that the four QSAR models obtained respectively by MLR, PLSR, and MNLR techniques are externally validated because their correlation coefficients are largely higher than 0.6, so the experimental activity can be precisely predicted using one of four established QSAR models including all five of the following descriptors: E lumo, χ, µ, Log P and % [61].

Fig. 5
figure 5

Correlation between the observed and predicted activities using MLR technique

Fig. 6
figure 6

Correlation between the observed and predicted activities using PLSR technique

Fig. 7
figure 7

Correlation between the observed and predicted activities using MNLR technique

Cross-validation

To measure the effectiveness of the QSAR models and examine their reliability, we have applied the cross-validation technique with the leave-one-out procedure, removing each observation at each iteration, so that each observation is tested exactly a once [62, 63]. This technique is applied searching each time a new model of twenty-five compounds (N-1 = 25) and predicting the biological activity of the deleted sample, as noted in Table 6, such that quadratic coefficient q2 or (r2cv) given by Eq. 6 is greater than 0.5, for that the models will be internally validated [64].

$$\:{r}^{2}cv=1-\:\frac{{\sum\:}_{1}^{26}\:\left(Ypred-Yobs\right){\:}^{2}}{{\sum\:}_{1}^{26}\:\left(Yobs-Ymean\right){\:}^{2}}=0.785$$
(6)

As:

Y pred: the LOO predicted response value, Y obs: the observed response value and Y mean: is the average of the observed response values.

A high value of r2cv = 0.785 (greater than 0.5) indicates that the candidate QSAR model is reliable, robust, and has better internal predictivity. Despite this, ‘Alexander Golbraikh and Alexander Tropsha’s study confirms that the cross-validation technique is essential but not sufficient, because the internal predictive power of the cross-validation procedure tends to be overestimated and the high value of the quadratic coefficient may result from a hazard correlation. For this reason, a Y-randomization test is necessary [61].

Table 6 Observed and predicted pIC50 values of the training set from the QSAR models

Validation using the Y-randomization test

To control the robustness of the QSAR model, we have used the Y-randomization test (or permutation test), based on repetitive randomizations of the answer data (Y). Thus, a new model is derived on a new training test of 26 compounds. But to improve the precision of the probability level, a few hundred runs of rerandomized data are usually necessary [65, 66]. The results of the validation using Y-randomization test displayed in Table S3, give the following statistical regression data of random model’s parameters:

$$\begin{aligned}&\text{Average\;r}: 0.421739, \text{Average\;r}^{2}=0.192447, \\&\text{Average\;Q}^{2}_{\text{cv}}=-0.42364,\; \text{and}\;\mathbf{cR}^{2}\mathbf{p}=\textbf{0.766375.}\end{aligned}$$

Although the re-ordered data give much lower R2 than the original data, and randomization constant (cR2p) is superior than 0.5, so we can be sure that the previous MLR QSAR model is correct, robust, and is not due to random chance.

Applicability domain

The main objective of the applicability domain (AD) for a predictive classification QSAR model is to define the area of chemical space where the model makes predictions with a given reliability [67]. For this goal, AD was carried out for the best QSAR model established by the MLR technique as given in Eq. 1, based on the leverage analysis illustrated by William diagram (Fig. 8) [68], which is performed with the assistance of SPSS software. An observation with hi > h* significantly affects the performance of the regression and can be classified as an outlier not associated to a reliable prediction. To know, the warning leverage (h*) is defined as h*=3*K/n, and K = p + 1, Where n: is the number of the training set, and p: is the number of predictor descriptors [69].

The results show that the leverage values of all chemical compounds in the training and test sets, more than ten newly designed compounds were inferior to the warning leverage h* = 0.69 (p = 5, K = 6, n = 26) without exception. So, the QSAR MLR model was predicted correctly due to the absence of outliers. Therefore, all compounds are tested in the AD, which mentions that their predicted activities are reliable [70].

Fig. 8
figure 8

William’s diagram of MLR model established by Eq: (1)

Drug similarity prediction of C22 compound, and novel molecules based on Lipinski, Ghose, Muegge, Veber and Egan rules

To arrive at a drug-like molecule, at least two of the following Lipinski’s rules must be verified: Molecular weight (≤ 500 g/mol) - Log P (High lipophilicity < 5) - Hydrogen bond donors (< 5) - Hydrogen bond acceptors (≤ 10) − 40 ≤ Molar refractive index ≤ 130. The number of rotatable bonds must be less than 10 [71, 72]. In addition, Lipinski’s violations must not exceed 1, otherwise the molecule may have bioavailability problems and a high probability of not being a drug [73]. The molecular descriptors of new designed molecules (C33 to C42) as presented in the Table 7, were calculated using Density Functional Theory (DFT) technique, and then tested with their synthetic accessibility and their drug similarity rules, specifically Lipinski, Ghose, Egan, Veber and Muegge, based on the most active compound, scored C22.

Table 7 Structural formula of the new studied compounds

The results obtained in Table 8, demonstrate that all ten compounds respect all Lipinski rules, which indicates the absence of oral bioavailability problems. All compounds are easy to synthesize, as their synthetic accessibility values are all about 3. In addition, all designed compounds meet the drug similarity rules except C36, C37 and C40 that do not verify Ghose and Egan drug similarity rules, as presented in Table 9 [74].

Table 8 Lipinski rules of the newly designed compounds
Table 9 Drug likeness prediction of the selected compounds and C22 compound based on Ghose, Muegge, Veber, and Egan rules, and their synthetic accessibility

ADMET in silico pharmacokinetics

To identify the new candidate drug with high success level and reduced experimental study duration, new compounds were designed based on the most potent inhibitor scored (C22), as presented in Table 7. This in-silico study predicts the pharmacokinetic features of Adsorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) [75] for ten compounds satisfying all conditions mentioned in Lipinski’s rule, as illustrated in Table 8. According to the results presented in Table 10, all the proposed compounds have a good absorption in the human intestine (IAH more than 70%) [72]. Except for C36, C37, C38 and C39, all the new compounds have a good distribution, as their human distribution volumes are estimated to be higher than − 0.44 Log L/kg, their BBB permeability superior than − 1 Log BB, and their CNS permeability included between − 2 and − 3 Log PS, thus do not penetrate the central nervous system [76]. C36 and C37 compounds, are potential inhibitors of cytochrome CYP450 (2C19), C33 to C39 molecules are possible inhibitors of cytochrome CYP450 (3A4), and C33-34 and C36-39 molecules could be inhibitors of cytochrome CYP450 (2C9). These last inhibitors are the only ones that have a low total clearance value, which means a great success of drug elimination by the organism. Any of these molecules having the AMES toxicity, this last property of ADMET has a major role in drug discovery [73]. Finally, we have succeeded in finding two new molecules, with highest analgesic activities (more than most active compound C22); verifying Lipinski, Muegge, Veber and Egan rules, and all in silico ADMET properties without exception, the first one noted C37 and the second one noted C39, they are predicted as powerful inhibitors of both cytochromes CYP450 (2C9) and CYP450 (3A4), with analgesic activities of pIC50 = 8.526 and pIC50 = 7.991, respectively.

Table 10 ADMET properties prediction of newly engineered compounds

Additionally, the bioavailability test confirm that all these novel compounds were predicted with an excellent oral bioavailability by the human body, because all tested molecules are part of the pink area of bioavailability radars presented in Fig. 9, as a desired part selected on the basis of the physicochemical parameters of flexibility, saturation, solubility, lipophilicity, polarity and size.

Fig. 9
figure 9

Bioavailability radars for ten designed molecules (C33 to C42)

Molecular docking

The results of molecular docking simulations shown in Fig. 10, confirm that both novel designed molecules (C37 and C39) were efffectively docked to the active sites of NMDA receptor with lowest possible binding energies in kcal/mol (-8.22 and − 8.14, respectively.), producing common intermolecular interactions, like those detected towards Gln110, Tyr109 amino acids residues (AARs), which were the same chemical bounds detected with the most active compound labeled C22, which was equally docked to the same targeted receptor with a binding energy closer than the previous ones (-7.84 kcal/mol). All other types of intermolecular interactions resulted in each (ligand-5EWJ.pdb protein) complex are presented in Table 11, in which we have equally noticed that both designed molecules (C37 and C39) in addition to the most active ligand (C22) were docked no further than protein target active sites, including Ser132 AAR in A chain and Gln110 AAR in B chain (Fig. 11), which affirm again that the studied ligands were actually docked to the active sites of the protein target, so the processes of molecular docking are successfully validated.

Fig. 10
figure 10

2D and 3D docking positions illustrating the resulted interactions for C37, C39, and C22 phenylpiperazine derivatives, in complex with NMDA receptor coded as 5EWJ.pdb, with binding energies of -8.22, -8.14, and − 7.84 kcal/mol, respectively

Table 11 Types of intermolecular interactions produced in three studied complexes
Fig. 11
figure 11

Active sites in A and B chains of NMDA receptor coded by 5EWJ.pdb code

MD simulation

Drug-receptor complex has to be sufficiently stable over a nano-scaler time range to execute the therapeutic response. As a result, the macromolecular complex underwent a 100 ns MD simulation by using Schrodinger’s Desmond software version 2022.4. The NMDA receptor’s dimeric structure is having of 710 amino acids consisting of 5502 heavy atoms out of overall 10,994 atoms. Structural alterations and RMSD analysis of the macromolecular backbone was executed during the 100 ns simulation to evaluate their thermodynamic stability. The complexed ligand C37 comprising of thirty-two heavy atoms of fifty-eight atoms in total with the presence of seven rotatable bonds. The RMSD value of the NMDA receptor’s backbone was found to fluctuate between 1.5 and 4.0 Å, whereas the bound ligand C37 exhibited some fluctuations within the macromolecular cavity with RMSD value in range of 3.0-5.5 Å followed by a sharp conformational change within the target cavity.

The atoms in a protein or ligand structure might deviate from their initial location, and this can be measured by using their RMSF value. It is an important parameter for determining the flexibility and dynamic behavior of the macromolecular complex. Protein RMSF is important because it may be used to predict protein dynamics and evaluate stability by providing information about the relative flexibility of various regions. MD based evaluation of human NMDA receptor complexed with C37 has concluded that the RMSF for Cα backbone was found to be within 0.6–3.2 Å with couple of exceptions, while for ligand C37 higher RMSF value lies withing the range of 2–4 Å within the target cavity.

The development of hydrophobic contacts, ionic interactions, and hydrogen bonds during an MD simulation are responsible for the thermodynamic permanence of a receptor-ligand complex and it is evaluated by the continuous monitoring of their strengths throughout the simulation for all the three macromolecular complexes. Throughout the simulation ligand C37 was found to be interacting with the NMDA receptor via formation of hydrophobic bonds with the amino acids Tyr109_A, Ile127_A, Tyr128_A, Ile133_A, His134_A, Met134_B, and Pro177_B, whereas amino acid Tyr109_A, Ser132_A, Leu135_A, Lys137_B, and Asp138_B via hydrogen bonding, while residues Ser108_A, Tyr109_A, Ser132_A, Ser136_A, Gln110_B, Asp113_B, Ala135_B, Asp136_B and Asp138_B are found to be interacting via water bridges.

The complexed ligand M7 comprising of thirty-two heavy atoms of forty-nine atoms in total with the presence of seven rotatable bonds. The RMSD value of the receptor’s backbone was found to fluctuate between 1.5 and 3.5 Å, whereas the bound ligand C39 exhibited stable conformation throughout the simulation within the macromolecular cavity with RMSD value in range of 6.0–9.0 Å within the target cavity.

MD based evaluation of NMDA receptor complexed with ligand C39 has concluded that the RMSF for Cα backbone was found to be within 0.8–2.4 Å with couple of exceptions, while for ligand C39 is having RMSF value within the range of 2.0–3.0 Å. indicating its high stability within the target cavity. Throughout the simulation ligand C39 was found to be interacting with NMDA receptor via formation of hydrophobic bonds with the amino acids Tyr109_A, Phe113_A, Ile127_A, Ile133_A, His134_A, and Ala107_B, whereas residues Thr105_A, Gly112_A, Ser129_A, Asp130_A, Gln105_B, Gln110_B, and Asp136_B, Val43, Ala47, Val71, Ile78, Ile90, Val120, and Val167 are found to be interacting via hydrogen bonding, while amino acid Thr105_A, Ser108_A, Arg115_A, Asp130-A, Ser132_A, Ser136_A, Thr76_B, Glu106_B, Asp113_B, Asp136_B and Asp138_BAsn46, Asp49, Glu50, Asp73, Arg76, and Gly77 are found to be interacting via water bridges.

The complexed standard drug C22 comprising of twenty-nine heavy atoms of forty-eight atoms in total with the presence of five rotatable bonds. The RMSD value of the NMDA receptor’s backbone was found to fluctuate between 1.5 and 2.7 Å, whereas the bound ligand C22 exhibited stable conformation throughout the simulation within the macromolecular cavity with RMSD value in range of 4.0-6.4 Å within the target cavity. Figure 12 demonstrates the revealed RMSD for macromolecular complexes of NMDA receptor complexed with ligand (a) C37, (b) C39, and standard drug (c) C22.

Fig. 12
figure 12

RMSD for Cα chain backbone and complexed ligand for the macromolecular complex of NMDA receptor with ligands C37, C39, and standard drug C22 respectively, detected while executing 100ns MD simulation

MD based evaluation of human NMDA receptor complexed with ligand C22 has concluded that the RMSF for Cα backbone was found to be within 0.6–2.4 Å with couple of exceptions, while for ligand C22 is having RMSF value within the range of 2.0–3.0 Å. indicating its high stability within the target cavity. RMSF for macromolecular complexes of human NMDA receptor complexed with ligand (a) C37, (b) C39, and standard drug (c) C22 was depicted in Fig. 13.

Fig. 13
figure 13

Root mean square fluctuation: Observed RMSF for the macromolecular complex of C37, C39, and standard drug C22 respectively, with human NMDA receptor detected while executing 100ns MD simulation

Throughout the simulation ligand C22 was found to be interacting with human NMDA receptor via formation of hydrophobic bonds with the amino acids Ile133_A, His171_A, Met134_B, Phe176_B and Pro177_B, whereas residues Gln110_B, Asp136_B, and Asp138_B are found to be interacting via hydrogen bonding, while amino acid Ile127_A, Ser132_A, His134_A, Glu106_B, Gln110_B, Met132_B, Ile133_B, Asp136_B, and Asp138_B are found to be interacting via water bridges. Figure 14 illustrates the interacting residues of human NMDA receptor with complexed ligand C37, C39, and standard drug C22.

Fig. 14
figure 14

Protein-ligand contacts: Protein-ligand interactions identified between human NMDA receptor complexed with ligands C37, C39, and standard drug C22. The interactions were visualized using different colored bars, with green representing hydrogen bonds, blue representing water bridges, and purple representing hydrophobic interactions

Conclusions

To concept new analgesic drugs for the treatment of neuropathic pain, four QSAR models were developed and successfully validated using different assessment methods, in which the analgesic activity was significantly affected by the physicochemical, constitutional, and quantum descriptors, in particular: dipole moment, octanol/water partition coefficient, Oxygen mass percentage, electronegativity, and energy of the lowest unoccupied molecular orbital. These results are partially qualified by molecular docking study, as the intermolecular interactions produced by the most active compound (C22) towards the active sites of protein target coded 5EWJ, confirm the production of the chemical bonds with highly electronegativity atoms, such as fluorine and chlorine in meta and para positions, respectively. This is in good agreement with the results developed by the QSAR model, as a noticeable increase in electronegativity, in the presence of an enriched mass percentage of the oxygen element renders the ligand a highly potent analgesic. These results could provide important structural information needed to optimize new good drug candidates for the treatment of neuropathic disease. Among these new drugs, two non-toxic compounds noted C37 and C39, respectively, were predicted to satisfy Lipinski, Muegge, Veber and Egan rules, with excellent ADMET profiles, and very good level of molecular stability towards NMDA receptor. For these reasons, C37 and C39 are strongly recommended to treat neuropathic pain. However, they must be subjected to experimental in vivo and in vitro investigations to examine their safety and efficacy as anti-chronic pain analgesic.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Yamamoto H, Hagino Y, Kasai S, Ikeda K. Specific roles of NMDA receptor subunits in Mental disorders. CMM. 2015;15:193–205. https://doi.org/10.2174/1566524015666150330142807.

    Article  CAS  Google Scholar 

  2. Santora VJ, Almos TA, Barido R, Basinger J, Bellows CL, Bookser BC, Breitenbucher JG, Broadbent NJ, Cabebe C, Chai C-K, Chen M, Chow S, Chung DM, Crickard L, Danks AM, Freestone GC, Gitnick D, Gupta V, Hoffmaster C, Hudson AR, Kaplan AP, Kennedy MR, Lee D, Limberis J, Ly K, Mak CC, Masatsugu B, Morse AC, Na J, Neul D, Nikpur J, Peters M, Petroski RE, Renick J, Sebring K, Sevidal S, Tabatabaei A, Wen J, Yan Y, Yoder ZW. Zook, Design and Synthesis of Novel and selective Glycine Transporter-1 (GlyT1) inhibitors with Memory Enhancing properties. J Med Chem. 2018;61:6018–33. https://doi.org/10.1021/acs.jmedchem.8b00372.

    Article  CAS  PubMed  Google Scholar 

  3. Hudson AR, Santora VJ, Petroski RE, Almos TA, Anderson G, Barido R, Basinger J, Bellows CL, Bookser BC, Broadbent NJ, Cabebe C, Chai C-K, Chen M, Chow S, Chung DM, Heger L, Danks AM, Freestone GC, Gitnick D, Gupta V, Hoffmaster C, Kaplan AP, Kennedy MR, Lee D, Limberis J, Ly K, Mak CC, Masatsugu B, Morse AC, Na J, Neul D, Nikpur J, Renick J, Sebring K, Sevidal S, Tabatabaei A, Wen J, Xia S, Yan Y, Yoder ZW, Zook D, Peters M, Breitenbucher JG. Azetidine-based selective glycine transporter-1 (GlyT1) inhibitors with memory enhancing properties. Bioorg Med Chem Lett. 2020;30:127214. https://doi.org/10.1016/j.bmcl.2020.127214.

    Article  CAS  PubMed  Google Scholar 

  4. Liu W, Jiang X, Zu Y, Yang Y, Liu Y, Sun X, Xu Z, Ding H, Zhao Q. A comprehensive description of GluN2B-selective N-methyl-D-aspartate (NMDA) receptor antagonists. Eur J Med Chem. 2020;200:112447. https://doi.org/10.1016/j.ejmech.2020.112447.

    Article  CAS  PubMed  Google Scholar 

  5. Xu Q, Hu M, Li J, Ma X, Chu Z, Zhu Q, Zhang Y, Zhu P, Huang Y, He G. Discovery of novel brain-penetrant GluN2B NMDAR antagonists via pharmacophore-merging strategy as anti-stroke therapeutic agents. Eur J Med Chem. 2022;227:113876. https://doi.org/10.1016/j.ejmech.2021.113876.

    Article  CAS  PubMed  Google Scholar 

  6. Ed-Dahmani I, El fadili M, Kandsi F, Conte R, El Atki Y, Kara M, Assouguem A, Touijer H, Lfitat A, Nouioura G, Slighoua M, Ullah R, Al-Tamimi JH, Taleb M, Abdellaoui A. Phytochemical, antioxidant activity, and toxicity of wild medicinal plant of Melitotus albus extracts, in vitro and in silico approaches, ACS Omega (2024) acsomega. https://doi.org/10.1021/acsomega.3c08314.

  7. Nouioura G, El fadili M, El Barnossi A, Loukili EH, Laaroussi H, Bouhrim M, Giesy JP, Aboul-Soud MAM, Al-Sheikh YA, Lyoussi B, Derwich Ehoussine. Comprehensive analysis of different solvent extracts of Ferula Communis L. fruit reveals phenolic compounds and their biological properties via in vitro and in silico assays. Sci Rep. 2024;14:8325. https://doi.org/10.1038/s41598-024-59087-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bouzammit R, Belchkar S, El fadili M, Kanzouai Y, Mujwar S, Alanazi MM, Chalkha M, Nakkabi A, Bakhouch M, Gal E, Gaina LI. Al Houari, New triazole-isoxazole hybrids as antibacterial agents: design, synthesis, characterization, in Vitro, and in Silico studies. Molecules. 2024;29:2510. https://doi.org/10.3390/molecules29112510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kandsi F, Lafdil FZ, El Hachlafi N, Jeddi M, Bouslamti M, Fadili ME, Seddoqi S, Gseyra N. Dysphania ambrosioides (L.) Mosyakin and Clemants: bridging traditional knowledge, photochemistry, preclinical investigations, and toxicological validation for health benefits. Naunyn-Schmiedeberg’s Arch Pharmacol. 2023. https://doi.org/10.1007/s00210-023-02658-4.

    Article  Google Scholar 

  10. Benkhaira N, El Hachlafi N, El fadili M, Jeddi M, Abdnim R, Bnouham M, Ibnsouda Koraichi S, Fikri-Benbrahim K. Unveiling the phytochemical profile, in vitro bioactivities evaluation, in silico molecular docking and ADMET study of essential oil from Clinopodium nepeta grown in Middle Atlas of Morocco. Biocatal Agric Biotechnol. 2023;102923. https://doi.org/10.1016/j.bcab.2023.102923.

  11. Assaggaf H, Hachlafi NE, El fadili M, Elbouzidi A, Ouassou H, Jeddi M, Alnasser SM, Qasem A, Attar A, AL-Farga A, Alghamdi OA, Mehana EE, Mrabti HN. Profiling, in vitro antidiabetic efficacy of Origanum Compactum Benth. Essential oil and in silico molecular docking of its major bioactive compounds. Catalysts. 2023;13:1429. https://doi.org/10.3390/catal13111429.

    Article  CAS  Google Scholar 

  12. Jeddi M, Hachlafi NE, El Fadili M, Benkhaira N, Al-Mijalli SH, Kandsi F, Abdallah EM, Ouaritini ZB, Bouyahya A, Lee L-H, Zengin G, Mrabti HN, Fikri-Benbrahim K. Antimicrobial, antioxidant, α-amylase and α-glucosidase inhibitory activities of a chemically characterized essential oil from Lavandula angustifolia Mill.,: in vitro and in silico investigations. Biochem Syst Ecol. 2023;111:104731. https://doi.org/10.1016/j.bse.2023.104731.

    Article  CAS  Google Scholar 

  13. Chtita S, Larif M, Ghamali M, Bouachrine M, Lakhlifi T. Quantitative structure–activity relationship studies of dibenzo[ a, d ]cycloalkenimine derivatives for non-competitive antagonists of N -methyl- d -aspartate based on density functional theory with electronic and topological descriptors. J Taibah Univ Sci. 2015;9:143–54. https://doi.org/10.1016/j.jtusci.2014.10.006.

    Article  Google Scholar 

  14. El fadili M, Er-rajy M, Ali Eltayb W, Kara M, Imtara H, Zarougui S, Al-Hoshani N, Hamadi A, Elhallaoui M. An in-silico investigation based on molecular simulations of novel and potential brain-penetrant GluN2B NMDA receptor antagonists as anti-stroke therapeutic agents. J Biomol Struct Dynamics. 2023;1–15. https://doi.org/10.1080/07391102.2023.2232024.

  15. El fadili M, Er-rajy M, Imtara H, Kara M, Zarougui S, Altwaijry N, Al kamaly OM, Al Sfouk A, Elhallaoui M. 3D-QSAR, ADME-Tox in silico prediction and molecular docking studies for modeling the analgesic activity against neuropathic pain of novel NR2B-selective NMDA receptor antagonists. Processes. 2022;10:1462. https://doi.org/10.3390/pr10081462.

    Article  CAS  Google Scholar 

  16. Chintha N, Jupudi S, Palathoti N, Bharathi J, Justin JA. In-silico docking and molecular dynamic introspective study of multiple targets of AChE with Rivastigmine and NMDA receptors with Riluzole for Alzheimer’s disease. J Biomolecular Structure Dynamics. 2023:1–12. https://doi.org/10.1080/07391102.2023.2167119.

  17. Anan K, Masui M, Tazawa A, Tomida M, Haga Y, Kume M, Yamamoto S, Shinohara S, Tsuji H, Shimada S, Yagi S, Hasebe N, Kai H. Discovery of NR2B-selective antagonists via scaffold hopping and pharmacokinetic profile optimization. Bioorg Med Chem Lett. 2019;29:1143–7. https://doi.org/10.1016/j.bmcl.2019.02.017.

    Article  CAS  PubMed  Google Scholar 

  18. El fadili M, Er-rajy M, Imtara H, Noman OM, Mothana RA, Abdullah S, Zerougui S, Elhallaoui M. QSAR, ADME-Tox, molecular docking and molecular dynamics simulations of novel selective glycine transporter type 1 inhibitors with memory enhancing properties. Heliyon. 2023;e13706. https://doi.org/10.1016/j.heliyon.2023.e13706.

  19. El fadili M, Er-rajy M, Ali Eltayb W, Kara M, Assouguem A, Saleh A, Al Kamaly O, Zarougui S, Elhallaoui M. In-silico screening based on molecular simulations of 3,4-disubstituted pyrrolidine sulfonamides as selective and competitive GlyT1 inhibitors. Arab J Chem. 2023;16:105105. https://doi.org/10.1016/j.arabjc.2023.105105.

    Article  CAS  Google Scholar 

  20. El fadili M, Er-Rajy M, Kara M, Assouguem A, Belhassan A, Alotaibi A, Mrabti NN, Fidan H, Ullah R, Ercisli S, Zarougui S, Elhallaoui M. QSAR, ADMET in Silico pharmacokinetics, molecular docking and molecular dynamics studies of novel bicyclo (Aryl Methyl) benzamides as potent GlyT1 inhibitors for the treatment of Schizophrenia. Pharmaceuticals. 2022;15:670. https://doi.org/10.3390/ph15060670.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Er-rajy M, El Fadili M, Hadni H, Mrabti NN, Zarougui S, Elhallaoui M. 2D-QSAR modeling, drug-likeness studies, ADMET prediction, and molecular docking for anti-lung cancer activity of 3-substituted-5-(phenylamino) indolone derivatives. Struct Chem. 2022;33:973–86. https://doi.org/10.1007/s11224-022-01913-3.

    Article  CAS  Google Scholar 

  22. El fadili M, Er-rajy M, Abdalla M, Abuelizz HA, Zarougui S, Alkhulaifi FM, Alahmady NF, Shami A, Elhalaoui M. In-silico investigations of novel tacrine derivatives potency against Alzheimer’s disease. Sci Afr. 2023;e02048. https://doi.org/10.1016/j.sciaf.2023.e02048.

  23. Er-rajy M, El fadili M, Mujwar S, Zarougui S, Elhallaoui M. Design of novel anti-cancer drugs targeting TRKs inhibitors based 3D QSAR, molecular docking and molecular dynamics simulation. J Biomol Struct Dynamics. 2023;0:1–14. https://doi.org/10.1080/07391102.2023.2170471.

    Article  CAS  Google Scholar 

  24. Er-rajy M, Fadili ME, Mujwar S, Lenda FZ, Zarougui S, Elhallaoui M. QSAR, molecular docking, and molecular dynamics simulation–based design of novel anti-cancer drugs targeting thioredoxin reductase enzyme. Struct Chem. 2023. https://doi.org/10.1007/s11224-022-02111-x.

    Article  Google Scholar 

  25. Milne GWA. Software Review of ChemBioDraw 12.0. ACS Publications. 2010. https://doi.org/10.1021/ci100385n.

    Article  Google Scholar 

  26. Österberg T, Norinder U. Prediction of drug transport processes using simple parameters and PLS statistics the use of ACD/logP and ACD/ChemSketch descriptors. Eur J Pharm Sci. 2001;12:327–37. https://doi.org/10.1016/S0928-0987(00)00189-5.

    Article  PubMed  Google Scholar 

  27. Parr RG, Weitao Y. Density-functional theory of atoms and molecules. New York: Oxford University Press; 1995. https://doi.org/10.1093/oso/9780195092769.001.0001.

    Book  Google Scholar 

  28. Frisch M, Clemente F, Revision G, Frisch A, Trucks GW, Schlegel HB, Scuseria GE, Robb MA, Cheeseman JR, Scalmani G, Barone V, Mennucci B, Petersson GA, Nakatsuji H, Caricato M, Li X, Hratchian HP. AF Izmaylov, J. Bloino, G. Zhe (2009).

  29. Lorenz G. Principal component analysis in Technology. CIRP Ann. 1989;38:107–9. https://doi.org/10.1016/S0007-8506(07)62662-6.

    Article  Google Scholar 

  30. Groth D, Hartmann S, Klie S, Selbig J, Analysis PC. Comput Toxicol. 2013;527–47. https://doi.org/10.1007/978-1-62703-059-5_22.

  31. Maćkiewicz A, Ratajczak W. Principal components analysis (PCA). Comput Geosci. 1993;19:303–42. https://doi.org/10.1016/0098-3004(93)90090-R.

    Article  Google Scholar 

  32. mem_VancolenS.pdf -. Dipl\u02c6ome Postgrade en Statistique La r\u00b4egression PLS 22 juin 2004 R\u00b4ealis\u00b4e par S\u00b4 everine Vancolen Supervis\u00b4e par le | Course Hero, (n.d.). https://www.coursehero.com/file/68552469/mem-VancolenSpdf/ (accessed November 21, 2021).

  33. Monari G. Sélection de modèles non linéaires par ‘leave-one-out’: étude théorique et application des réseaux de neurones au procédé de soudage par points, (n.d.) 114.

  34. Randomization Test - an. overview | ScienceDirect Topics, (n.d.). https://www.sciencedirect.com/topics/mathematics/randomization-test (accessed September 19, 2021).

  35. Onghena P. Randomization and the randomization test: two sides of the same Coin, in: Berger VW (Ed.), Randomization, Masking, and allocation concealment, 1st ed., Chapman and Hall/CRC, Boca Raton: Taylor & Francis,., a CRC title, part of the Taylor & Francis imprint, a member of the Taylor & Francis Group, the academic division of T&F Informa plc, 2018 2017: 185–208. https://doi.org/10.1201/9781315305110-13.

    Chapter  Google Scholar 

  36. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:42717. https://doi.org/10.1038/srep42717.

    Article  PubMed  PubMed Central  Google Scholar 

  37. pkCSM. (n.d.). http://biosig.unimelb.edu.au/pkcsm/run_example? (accessed November 15, 2021).

  38. Serrano A, Imbernón B, Pérez-Sánchez H, Cecilia JM, Bueno-Crespo A, Abellán JL. QN-Docking: an innovative molecular docking methodology based on Q-Networks. Appl Soft Comput. 2020;96:106678. https://doi.org/10.1016/j.asoc.2020.106678.

    Article  Google Scholar 

  39. Berger ML, Maciejewska D, Vanden Eynde JJ, Mottamal M, Żabiński J, Kaźmierczak P, Rezler M, Jarak I, Piantanida I, Karminski-Zamola G, Mayence A, Rebernik P, Kumar A, Ismail MA, Boykin DW, Huang TL. Pentamidine analogs as inhibitors of [3H]MK-801 and [3H]ifenprodil binding to rat brain NMDA receptors. Bioorg Med Chem. 2015;23:4489–500. https://doi.org/10.1016/j.bmc.2015.06.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kouranov A, Xie L, de la Cruz J, Chen L, Westbrook J, Bourne PE, Berman HM. The RCSB PDB information portal for structural genomics. Nucleic Acids Res. 2006;34:D302–5. https://doi.org/10.1093/nar/gkj120.

    Article  CAS  PubMed  Google Scholar 

  41. Signals Lead Discovery - Chemical analytics - PerkinElmer informatics. PerkinElmer (n.d.). https://perkinelmerinformatics.com/products/research/signals-lead-discovery/ (accessed September 19, 2021).

  42. Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, Olson AJ. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J Comput Chem. 2009;30:2785–91. https://doi.org/10.1002/jcc.21256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. BIOVIA Discovery Studio - BIOVIA - Dassault Systèmes®. (n.d.). https://www.3ds.com/products-services/biovia/products/molecular-modeling-simulation/biovia-discovery-studio/ (accessed November 17, 2021).

  44. Li Q, Zhang H, Guan S, Du J, Zhang Y, Wang S. Molecular dynamics simulation of the inhibition mechanism of factor XIa by Milvexian-like macrocyclic inhibitors. Comput Theor Chem. 2023;1225:114131. https://doi.org/10.1016/j.comptc.2023.114131.

    Article  CAS  Google Scholar 

  45. Shah K, Mujwar S. Delineation of a novel non-steroidal anti-inflammatory drugs derivative using molecular docking and pharmacological assessment. IJPS. 2022;84. https://doi.org/10.36468/pharmaceutical-sciences.959.

  46. Mujwar S, Tripathi A. Repurposing benzbromarone as antifolate to develop novel antifungal therapy for Candida albicans. J Mol Model. 2022;28:193. https://doi.org/10.1007/s00894-022-05185-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Behl T, Kumar K, Brisc C, Rus M, Nistor-Cseppento DC, Bustea C, Aron RAC, Pantis C, Zengin G, Sehgal A, Kaur R, Kumar A, Arora S, Setia D, Chandel D, Bungau S. Exploring the multifocal role of phytochemicals as immunomodulators. Biomed Pharmacother. 2021;133:110959. https://doi.org/10.1016/j.biopha.2020.110959.

    Article  CAS  PubMed  Google Scholar 

  48. Er-rajy M, El fadili M, Mujwar S, Zarougui S, Elhallaoui M. Design of novel anti-cancer drugs targeting TRKs inhibitors based 3D QSAR, molecular docking and molecular dynamics simulation. J Biomol Struct Dynamics. 2023;41:11657–70. https://doi.org/10.1080/07391102.2023.2170471.

    Article  CAS  Google Scholar 

  49. Gupta SM, Behera A, Jain NK, Kumar D, Tripathi A, Tripathi SM, Mujwar S, Patra J, Negi A. Indene-derived hydrazides targeting acetylcholinesterase enzyme in Alzheimer’s: design, synthesis, and Biological evaluation. Pharmaceutics. 2022;15:94. https://doi.org/10.3390/pharmaceutics15010094.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shinu P, Sharma M, Gupta GL, Mujwar S, Kandeel M, Kumar M, Nair AB, Goyal M, Singh P, Attimarad M, Venugopala KN, Nagaraja S, Telsang M, Aldhubiab BE, Morsy MA. Computational design, synthesis, and pharmacological evaluation of Naproxen-Guaiacol Chimera for Gastro-sparing anti-inflammatory response by selective COX2 inhibition. Molecules. 2022;27:6905. https://doi.org/10.3390/molecules27206905.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Nemati M, Oveisi MR, Abdollahi H, Sabzevari O. Differentiation of bovine and porcine gelatins using principal component analysis. J Pharm Biomed Anal. 2004;34:485–92. https://doi.org/10.1016/S0731-7085(03)00574-0.

    Article  CAS  PubMed  Google Scholar 

  52. Uyanık GK, Güler N, Study on multiple linear regression analysis. Proc Social Behav Sci. 2013;106:234–40. https://doi.org/10.1016/j.sbspro.2013.12.027.

    Article  Google Scholar 

  53. Rosipal R, Krämer N. Overview and recent advances in partial least squares. In: Saunders C, Grobelnik M, Gunn S, Shawe-Taylor J, editors. Subspace, Latent structure and feature selection. Berlin Heidelberg, Berlin, Heidelberg: Springer; 2006. pp. 34–51. https://doi.org/10.1007/11752790_2.

    Chapter  Google Scholar 

  54. Polat E, Gunay S. A New Robust partial least squares regression method based on multivariate MM-estimators. Int J Math Statistics™. 2017;18:82–99. http://www.ceser.in/ceserp/index.php/ijms/article/view/5002. accessed November 20, 2021.

    Google Scholar 

  55. Bagchi J, Si T. Nonlinear regression analysis using multi-verse optimizer, ArXiv:2005.10642 [Cs, Stat] (2020). http://arxiv.org/abs/2005.10642 (accessed September 18, 2021).

  56. Mozafari Z, Arab Chamjangali M, Arashi M. Combination of least absolute shrinkage and selection operator with bayesian regularization artificial neural network (LASSO-BR-ANN) for QSAR studies using functional group and molecular docking mixed descriptors. Chemometr Intell Lab Syst. 2020;200:103998. https://doi.org/10.1016/j.chemolab.2020.103998.

    Article  CAS  Google Scholar 

  57. Eluyode OS, Akomolafe DT. Comparative study of biological and artificial neural networks, (2013) 12.

  58. Mbarki S, Hallaoui ME, Dguigui K. 3D-Qsar for α-glucosidase inhibitory activity of N-(phenoxyalkyl) phthalimide derivatives, vol. 7; 2012.

  59. Hadni H, Elhallaoui M. Molecular docking and QSAR studies for modeling the antimalarial activity of hybrids 4-anilinoquinoline-triazines derivatives with the wild-type and mutant receptor pf-DHFR. Heliyon. 2019;5:e02357. https://doi.org/10.1016/j.heliyon.2019.e02357.

    Article  PubMed  PubMed Central  Google Scholar 

  60. de Rezende KB, L.A. da Cunha AJ, Amim Junior J, Bornia RG. External validation of the fetal medicine foundation algorithm for the prediction of preeclampsia in a Brazilian population. Pregnancy Hypertens. 2019;17:64–8. https://doi.org/10.1016/j.preghy.2019.05.006.

    Article  PubMed  Google Scholar 

  61. Golbraikh A, Tropsha A. Beware of q2! J Mol Graph Model. 2002;20:269–76. https://doi.org/10.1016/S1093-3263(01)00123-1.

    Article  CAS  PubMed  Google Scholar 

  62. Rafało M. Cross validation methods: analysis based on diagnostics of thyroid cancer metastasis. ICT Express. 2021;S2405959521000552. https://doi.org/10.1016/j.icte.2021.05.001.

  63. Monari G. Sélection de modèles non linéaires par leave-one-out: étude théorique et application des réseaux de neurones au procédé de soudage par points, phdthesis, Université Pierre et Marie Curie - Paris VI, 1999. https://pastel.archives-ouvertes.fr/pastel-00000676 (accessed November 20, 2021).

  64. Chtita S, Mohammed B, Lakhlifi T. Modélisation De molécules organiques hétérocycliques biologiquement actives par des méthodes QSAR/QSPR. Recherche De Nouveaux médicaments. (n d) 188.

  65. Wold S, Eriksson L, Clementi S. Statistical validation of QSAR results. In: van de Waterbeemd H, editor. Methods and principles in Medicinal Chemistry. 1st ed. Wiley; 1995. pp. 309–38. https://doi.org/10.1002/9783527615452.ch5.

  66. van der Voet H. Comparing the predictive accuracy of models using a simple randomization test. Chemometr Intell Lab Syst. 1994;25:313–23. https://doi.org/10.1016/0169-7439(94)85050-X.

    Article  Google Scholar 

  67. Klingspohn W, Mathea M, ter Laak A, Heinrich N, Baumann K. Efficiency of different measures for defining the applicability domain of classification models. J Cheminform. 2017;9:44. https://doi.org/10.1186/s13321-017-0230-2.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Chtita S, Belhassan A, Bakhouch M, Taourati AI, Aouidate A, Belaidi S, Moutaabbid M, Belaaouad S, Bouachrine M, Lakhlifi T. QSAR study of unsymmetrical aromatic disulfides as potent avian SARS-CoV main protease inhibitors using quantum chemical descriptors and statistical methods. Chemometr Intell Lab Syst. 2021;210:104266. https://doi.org/10.1016/j.chemolab.2021.104266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Pourbasheer E, Riahi S, Ganjali MR, Norouzi P. Quantitative structure–activity relationship (QSAR) study of interleukin-1 receptor associated kinase 4 (IRAK-4) inhibitor activity by the genetic algorithm and multiple linear regression (GA-MLR) method. J Enzyme Inhib Med Chem. 2010;25:844–53. https://doi.org/10.3109/14756361003757893.

    Article  CAS  PubMed  Google Scholar 

  70. Hadni H, Elhallaoui M. 3D-QSAR, docking and ADMET properties of aurone analogues as antimalarial agents. Heliyon. 2020;6:e03580. https://doi.org/10.1016/j.heliyon.2020.e03580.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Lipinski CA. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discovery Today Technol. 2004;1:337–41. https://doi.org/10.1016/j.ddtec.2004.11.007.

    Article  CAS  Google Scholar 

  72. Egan WJ, Merz Kenneth M, Baldwin JJ. Prediction of drug absorption using Multivariate statistics. J Med Chem. 2000;43:3867–77. https://doi.org/10.1021/jm000292e.

    Article  CAS  PubMed  Google Scholar 

  73. Belhassan A, Zaki H, Chtita S, Alaqarbeh M, Alsakhen N, Benlyas M, Lakhlifi T, Bouachrine M. Camphor, Artemisinin and Sumac phytochemicals as inhibitors against COVID-19: computational approach. Comput Biol Med. 2021;136:104758. https://doi.org/10.1016/j.compbiomed.2021.104758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. El Mchichi L, El Aissouq A, Kasmi R, Belhassan A, El-Mernissi R, Ouammou A, Lakhlifi T, Bouachrine M. In silico design of novel pyrazole derivatives containing thiourea skeleton as anti-cancer agents using: 3D QSAR, drug-likeness studies, ADMET prediction and molecular docking. Mater Today Proc. 2021;45:7661–74. https://doi.org/10.1016/j.matpr.2021.03.152.

    Article  CAS  Google Scholar 

  75. Tian S, Wang J, Li Y, Li D, Xu L, Hou T. The application of in silico drug-likeness predictions in pharmaceutical research. Adv Drug Deliv Rev. 2015;86:2–10. https://doi.org/10.1016/j.addr.2015.01.009.

    Article  CAS  PubMed  Google Scholar 

  76. Clark DE. In silico prediction of blood–brain barrier permeation. Drug Discovery Today. 2003;8:927–33. https://doi.org/10.1016/S1359-6446(03)02827-7.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors extend their appreciation to the Researchers Supporting Project, King Saud University, Riyadh, Saudi Arabia for funding this work through grant no. RSPD2024R566.

Funding

This research was funded by the Researchers Supporting Project (No. RSPD2024R566), King Saud University, Riyadh, Saudi Arabia.

Author information

Authors and Affiliations

Authors

Contributions

Author Contributions: M.EF: wrote the main manuscript text, M.ER and S.M: software, A.A and R.B, H.A: reviewed the manuscript, M.K and S.E: analysis and validation, M.E; Supervised and reviewed the experiments.

Corresponding author

Correspondence to Mohamed El fadili.

Ethics declarations

Competing interests

The authors declare no competing interests.

Institutional review board statement

Not applicable.

Conflict of interest

The authors declare that they are no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

El fadili, M., Er-rajy, M., Mujwar, S. et al. In silico insights into the design of novel NR2B-selective NMDA receptor antagonists: QSAR modeling, ADME-toxicity predictions, molecular docking, and molecular dynamics investigations. BMC Chemistry 18, 142 (2024). https://doi.org/10.1186/s13065-024-01248-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13065-024-01248-6

Keywords