Next Article in Journal
Generative Language Models for Personality-Based Utterances in Novels: A Character Clustering Approach
Previous Article in Journal
Seismic Assessment and Strengthening of a Load-Bearing Masonry Structure Considering SSI Effects
Previous Article in Special Issue
MultiScaleFusion-Net and ResRNN-Net: Proposed Deep Learning Architectures for Accurate and Interpretable Pregnancy Risk Prediction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Combining QSAR and Molecular Docking for the Methodological Design of Novel Radiotracers Targeting Parkinson’s Disease

by
Juan A. Castillo-Garit
1,*,†,
Mar Soria-Merino
2,†,
Karel Mena-Ulecia
3,4,
Mónica Romero-Otero
2,
Virginia Pérez-Doñate
5,
Francisco Torrens
6 and
Facundo Pérez-Giménez
7
1
Instituto Universitario de Investigación y Desarrollo Tecnológico (IDT), Universidad Tecnológica Metropolitana, Ignacio Valdivieso 2409, San Joaquín, Santiago 8940577, Chile
2
Radiopharmacy Unit, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
3
Departamento de Ciencias Biológicas y Químicas, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco 4810399, Chile
4
Núcleo de Investigación en Bioproductos y Materiales Avanzados (BioMa), Vicerectoría de Investigación y Postgrado, Universidad Católica de Temuco, Temuco 4810399, Chile
5
Microbiology Service, Hospital Universitario de la Ribera, 46600 Valencia, Spain
6
Institut Universitari de Ciència Molecular, Universitat de Valencia, 46010 Valencia, Spain
7
Research Unit of Radiopharmacy and Stability Drugs, Universitat de Valencia, 46010 Valencia, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2025, 15(15), 8134; https://doi.org/10.3390/app15158134
Submission received: 24 March 2025 / Revised: 17 July 2025 / Accepted: 18 July 2025 / Published: 22 July 2025
(This article belongs to the Special Issue Application of Artificial Intelligence in Biomedical Informatics)

Abstract

Parkinson’s disease (PD) is a neurodegenerative disorder marked by the progressive loss of dopaminergic neurons in the nigrostriatal pathway. The dopamine active transporter (DAT), a key protein involved in dopamine reuptake, serves as a selective biomarker for dopaminergic terminals in the striatum. DAT binding has been extensively studied using in vivo imaging techniques such as Single-Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET). To support the design of new radiotracers targeting DAT, we employ Quantitative Structure–Activity Relationship (QSAR) analysis on a structurally diverse dataset composed of 57 compounds with known affinity constants for DAT. The best-performing QSAR model includes four molecular descriptors and demonstrates robust statistical performance: R2 = 0.7554, Q2LOO = 0.6800, and external R2 = 0.7090. These values indicate strong predictive capability and model stability. The predicted compounds are evaluated using a docking methodology to check the correct coupling and interactions with the DAT. The proposed approach—combining QSAR modeling and docking—offers a valuable strategy for screening and optimizing potential PET/SPECT radiotracers, ultimately aiding in the neuroimaging and early diagnosis of Parkinson’s disease.

1. Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s disease, affecting approximately 1% of the global population over the age of 60 [1,2,3]. It is caused by the degeneration of dopaminergic neurons located in the substantia nigra pars compacta [4]. This neuronal loss leads to a decrease in dopamine release in the striatum, which results in the hallmark symptoms of PD, which are bradykinesia, rigidity, postural instability, tremors, and a significant decline in patients’ quality of life [5].
The diagnosis of PD still relies on clinical criteria and remains challenging [6], particularly in its early stages when symptoms may resemble those of other disorders [7] such as multisystem atrophy, progressive supranuclear palsy, essential tremor, and vascular or drug Parkinsonism [5,6,8]. Therefore, the use of tools that allow for an accurate and early diagnosis of PD is crucial, given their confirmed impact on both prognosis and treatment outcomes [9]. One such approach involves evaluating the integrity of the nigrostriatal dopaminergic pathway using dopamine active transporters (DATs) [10]. The DATs are a group of transmembrane proteins associated with chloride channels, located at the presynaptic terminals of dopaminergic neurons [11]. They are responsible for the reuptake of dopamine after its release into the synaptic cleft [11,12]. In patients with PD, because of the degeneration of these neurons, the density of DATs is significantly reduced [13].
Non-invasive molecular neuroimaging techniques, such as SPECT (Single-Photon Emission Computed Tomography) and PET (Positron Emission Tomography), enable in vivo visualization of various tissues and organs using radiopharmaceuticals [12,14]. Several PET and SPECT radiotracers have been developed to target DATs [15]. Among them are radio-iodinated cocaine analogues, such as 123I-Ioflupane (123I-N-ω-fluoropropyl-2β-carbomethoxy-3β-(4-iodophenyl)nortropane) and 123I-β-CIT (123I-2β-carbomethoxy-3β-(4-iodophenyl)tropane) [16]. These SPECT radiopharmaceuticals are widely used to assess the integrity of the nigrostriatal dopaminergic pathway and have demonstrated good diagnostic sensitivity and specificity for suspected PD cases [15]. However, both tracers require pre-treatment with a thyroid-blocking agent to protect the thyroid gland from radioactive iodine uptake [17]. Additionally, imaging with 123I-β-CIT must be conducted 24 h post-injection, which can be inconvenient for outpatient settings [18]. There are also PET radiotracers with affinity for DAT, such as 11C-N-(3-iodoprop-2(E)-enyl)-2β-carbomethoxy-3β-(4′-methylphenyl)nortropane (11C-PE2I) [16]. In spite of its high specificity, the short half-life of 11C (approximately 20 min) limits its clinical utility, as it requires on-site cyclotron production [19]. These limitations underscore the need for the development of new DAT-targeting radiopharmaceuticals that offer improved pharmacokinetic properties and logistical advantages, enabling better assessment of the nigrostriatal pathway in the early diagnosis of PD.
The estimated cost and time required to bring a new drug to market are approximately USD 2.8 billion and 7 to 12 years [20]. Moreover, of the roughly 40,000 compounds tested in animals, only five typically proceed to human trials, and just one in five of those that reach clinical studies ultimately receives approval [21]. This reflects a significant expenditure of financial, human, and time resources. In response, virtual (computational) screening has emerged as a promising alternative to the traditional “trial-and-error” approach in identifying new chemical entities with biological activity. As a result, the pharmaceutical industry has shown growing interest in computational methodologies [22]. Among these approaches, QSAR (Quantitative Structure–Activity Relationship) modeling stands out as a valuable in silico technique connecting chemical structure with pharmacological activity. QSAR offers clear advantages in terms of cost- and time-efficiency, especially during the early stages of drug discovery, by enabling the prioritization of the most promising compounds for further evaluation [23,24,25,26].
In recent years, numerous computational studies have been undertaken to identify novel compounds with potential radiopharmaceutical applications (references). Notably, some of these investigations have targeted specific biological systems, such as the dopamine (D2) receptor and the adenosine A2A receptor, the latter with an emphasis on antagonist identification. A significant number of studies have also concentrated on modeling interactions with the dopamine transporter. However, many of these efforts have relied on relatively limited compound databases or congeneric series, which restricts the generalizability and predictive power of their findings.
In recent years, a growing number of computational studies have aimed to identify novel compounds with potential radiopharmaceutical applications [27,28,29,30]. These efforts have usually focused on specific molecular targets, including the dopamine (D2) receptor [27] and the adenosine A2A receptor (particularly in the context of antagonist discovery) [28]. In parallel, several studies have focused on modeling interactions with the dopamine transporter [29,30]. In spite of their contributions, many of these studies are based on relatively small datasets or congeneric compound series, which limits the extrapolation, robustness, and external validation of the resulting models. Given this context, the main aim of the present work is to propose a well-validated computational model based on a structurally diverse dataset, capable of identifying the key structural features required for DAT affinity, and to integrate it with molecular docking studies. This combined approach aims to facilitate the prediction of novel radiopharmaceutical candidates with potential utility in the diagnosis of Parkinson’s disease.

2. Materials and Methods

2.1. Dataset

The database used in the QSAR study is composed of 57 compounds that were taken from the previously published literature [31,32]. These compounds cover different pharmacological groups, are structurally diverse, and belong to the Binding Database. All of them have affinity reports for the DAT, evaluated by determining the value of the dissociation equilibrium constant (Kd) at DAT binding, using this parameter to define the affinity of the molecules to be bound to the transporter. The Kd value was determined by measuring the activity of the 3H-labeled radioligand bound to the DAT using an adjustment of the method proposed by Pristupa et al. [33]. The Kd values ranged from 70 to 52,000 nM (Table 1); because of the large affinity span of the compounds, we decided to apply a transformation and use Log Kd as the dependent variable. All standard parametric assumptions (including linearity, normality of residuals, homoscedasticity, and absence of multicollinearity) were evaluated using appropriate statistical diagnostics. The complete set of assumption checks, including plots and test results, is available in the Supplementary Materials.

2.2. Calculation of Molecular Descriptors

To calculate the molecular descriptors (MDs) used in this study, we employed Dragon software version 7.0.10 [34], developed by the Milano Chemometrics and QSAR Research Group. Dragon is one of the most widely recognized tools for molecular descriptor calculation. The study focused exclusively on 0D to 2D descriptor families, including constitutional indices, topological indices, functional group counts, molecular properties, and 2D-autocorrelations, among others. A total of 3839 descriptors were initially generated. After that, the variables with constant or near-constant values were removed and, consequently, not used in further analysis. In addition, variables with a standard deviation (SD) lower than 5% and a pair correlation over 85% were also discarded from subsequent analysis.

2.3. Multiple Linear Regression Model

MLR (multiple linear regression) is the process that obtains a linear relationship between DAT affinity and molecular descriptors using the OLS (ordinary least squares) method [35]. We used the QSARINS (QSAR-Insubria) software (version 2.2.4 year 2019) [36] developed by the QSAR Research Unit, University of Insubria, Italy. It allows the creation of MLR models using the GA (genetic algorithm) method for variable selection [35,37]. This software permits dividing the database into training and prediction sets by either modeled endpoint responses or structure; in our case, we used the first option. Figure 1 shows the structures of the molecules included in the training (learning) set, and Figure 2 shows the structures of the compounds used in the prediction set. The best model was selected based on several criteria such as a high value of R2 (good fitting) and Q2LOO (robustness), a low value for R2–Q2LOO (stability) and KXX (low correlation of descriptors), and the greatest R2ext (predictability), among others, so that it has a slight difference between fitting, cross-validation, and external validation parameters.

2.4. Validation of the QSAR-MLR Model

The obtained model must be carefully evaluated and rigorously validated to ensure its reliability. QSARINS provides several tools to assess whether the model complies with the standards established by the Organization for Economic Cooperation and Development (OECD) for the development, validation, acceptance, and use of QSAR models. Adhering to these guidelines enhances confidence in the predictive reliability of the model. According to OECD principles, a valid QSAR-MLR model must satisfy the following criteria [38,39]:
1
A defined endpoint;
2
An unambiguous algorithm;
3
A defined domain of applicability;
4
Appropriate measures of goodness-of-fit, robustness, and predictability;
5
A mechanistic interpretation, if possible.
To comply with the 1st principle, the dissociation equilibrium constant (Kd) between the molecules and the DAT was used as the endpoint. For compliance with the 2nd principle, molecular descriptors were calculated using Dragon software, while the QSARINS platform was employed to develop the MLR model as we explained in the previous section. This algorithm allows us to correlate the chemical structure descriptors and their binding affinity for DAT.
The definition of the applicability domain (AD) of the model is considered a critical aspect in chemometric studies and is the 3rd principle. Only predictions for compounds that fall within the AD may be considered reliable, but not the extrapolations of the models, even if they show robustness and significance. In order to define the AD of the model, the Williams graph/plot was used, so that reliable predictions of the model have leverage values lower than the critical leverage, ranging within ±3 standard deviations [40]. Compounds falling outside these ranges were considered outliers. In addition, the Insubria graph (generated by QSARINS) was utilized to assess further the reliability of predictions. This graphical tool is particularly useful for visualizing the position of molecules with unknown experimental responses and comparing them with those in the prediction set, helping to evaluate the confidence in their predicted values.
Following the 4th principle, the goodness of the fit was evaluated using the coefficient of determination R2 and its modified form R2adj; the latter accounts for the number of descriptors in the model and penalizes the inclusion of non-informative variables. To evaluate the robustness of the model, several internal validation strategies were employed. First, the Q2_LOO metric was used, applying Leave-One-Out (LOO) cross-validation, where each compound in the dataset is sequentially excluded, and the model is recalculated to predict the excluded compound. Additionally, QSARINS provides a more rigorous validation approach, Leave-Many-Out (LMO) cross-validation, which was also applied. In this method, between 5% and 30% of the dataset was randomly excluded in each iteration, allowing us to examine the model’s stability and predictive behavior under more challenging conditions. To ensure that the model is not the result of a casual correlation, a randomization test (Y-scrambling) was performed [41]. In this procedure, the response values (Kd) are randomly shuffled, breaking any real association with the molecular descriptors. A valid model is expected to show a significant drop in performance under these conditions, confirming that the original model’s predictive power is not because of chance.
To further confirm the predictive capability of the model, an external validation was carried out. The external set included several widely used reference radiopharmaceuticals, which were excluded from model training and used solely to assess predictability. The affinity values of these compounds (see Figure 2) were predicted using the final MLR model equation, providing an independent evaluation of the model’s generalization ability.

2.5. Virtual Screening and Docking Experiment

With the aim of proving the possibilities of our approach to identifying new compounds with potential affinity for DAT, we performed a virtual screening of several compounds that have different modes of action in the central nervous system (CNS). The structure of these compounds is shown in Figure 3. We performed virtual screening with eleven compounds to evaluate the performance of our model with respect to those compounds and identified new compounds with possible affinity for DAT. The compounds were selected based on the following criteria: (1) chemical diversity (representing different chemical scaffolds to explore diverse interactions within the binding site of the target); (2) drug-likeness and physicochemical properties (the compounds comply with Lipinski’s Rule of Five and exhibit acceptable ADME profiles); (3) commercial availability (compounds readily purchasable from chemical suppliers for potential future in vitro validation). After that, docking experiments were performed to evaluate the affinity of the predicted compound with the receptor. The optimized structure of each molecule was used for docking experiments using Autodock Vina software v1.2.3 [42]. The dopamine transporter was the protein used in this study, which was downloaded from the Protein Data Bank with code 8Y2D, resolved at 2.80 Å [43]. The dopamine was prepared in vitro by the addition of hydrogen atoms at the physiological pH of 7.4 and by the elimination of the water molecules around the protein. Ligands were prepared to account for rotatable bonds using Autodock Tool v1.5.7. This same program was used for the treatment of the dopamine transporter protein before the docking experiments, setting the grid size to 40 × 40 × 40 Å3 located at the center of mass of L-dopamine in the crystal structure of the dopamine transporter.
The coordinates of the grid box were x = 112.241 Å, y = 105.521 Å, and z = 106.058 Å, with a grid spacing of 0.375 Å, covering a volume of 30 × 30 × 30 Å3 and located in the center of mass of L-dopamine in the crystal structure of the dopamine transporter to completely cover the binding site and adjacent regions, allowing flexibility in the exploration of poses [44,45], which is consistent with similar studies in G-protein-coupled receptors (GPCRs) [46]. The mode number was 50, and the energy rank was set to 1 kcal/mol. The best docking poses were selected following two criteria: the relative total binding energy score and the positional root-mean-square deviation (RMSD) [47,48], comparing each docking ligand result with the position of L-dopamine in the crystallographic structure of the dopamine transporter from the Protein Data Bank [49,50,51]. Each RMSD value was calculated using the Pymol program v2.5.0 [52,53].

2.6. Molecular Dynamic Simulation Protocol

The starting point for preparing the complexes for molecular dynamics simulations was to obtain the best poses derived from docking experiments. These complexes were those formed by the compounds Diethylpropion, Procaine, and Benztropine (see Figure 3) with the dopamine transporter, whose PDB ID is 8Y2D, resolved at 2.80 Å [43]. The protein was prepared by adding hydrogen atoms at physiological pH (pH = 7.4) using the UCSF Chimera software version 1.14 [54,55,56]. The force field used was CHARMM36 for proteins [57], and parameters for organic molecules were obtained from the SwissParam web server [58].
The ligand–protein complexes were placed in a lipid bilayer formed by the POPC lipid model, which was constructed using the CHARMM-GUI server [59,60,61]. This lipid bilayer contains 55,160 lipid molecules. These complexes, along with the membrane, were placed into a cubic water box of 15.0 × 15.0 × 15.0 Å3 centered on the center of mass of each ligand using the TIP3P water model [62,63,64]. All the complexes were submitted to 2000 steps for energy minimization using the conjugate gradient methodology [65,66] at a temperature of 298.15 K using the weak coupling algorithm [67,68,69]. The Van der Waals cutoff was fixed to 12 Å, under the NPT ensemble (constant number of particles, pressure, and temperature). The complexes studied were submitted to a 1.0 fs time step under the velocity Verlet algorithm [70,71] and 2.0 ns of equilibration and 100 ns of molecular dynamics simulation using the NAMD 2.13 software package [72,73,74,75].

3. Results

3.1. Development of the QSAR-MLR Model

Large numbers of models were obtained while exploring the best combinations of molecular descriptors that show a high correlation with the response variable (LogKd). Therefore, an analysis of the model’s parameters was performed considering the principle of parsimony. Based on this analysis, a QSAR-MLR model was developed to evaluate the binding affinity for DAT (so that it can describe the maximum information with the least number of descriptors). The equation and the statistical parameters of the best model are as follows:
Log Kd = 3.538 (±0.204) − 0.403 (±0.204) × O-058 + 0.162 (±0.039) × CATS2D_03_AL
− 0.133 (±0.039) * CATS2D_08_LL − 0.537 (±0.291) * B08[C-N]
R2 = 0.755R2adj = 0.729R2 − R2adj = 0.026LOF = 0.195
Kxx = 0.236ΔK = 0.076RMSEtr = 0.358MAEtr = 0.276
RSStr = 5.377CCCtr = 0.861s = 0.381F = 28.566
where R2 is the coefficient of determination, R2adj is adjusted R2, s is the standard error of estimate, F is the variance ratio, and LOF is Friedman lack of fit. The parameter Kxx is the correlation among descriptors, and ΔK is the difference of the correlations between the descriptors (Kx) and the descriptors plus the responses (Kx). Moreover, RMSEtr is the root-mean-square error of fit (for the training set), RSStr is the residual sum of squares in the fit, and CCCtr is the concordance correlation coefficient calculated over the training set [76]. Table 2 shows the results of the training set, and Figure 4 shows the scatter plot of predicted vs. observed values.

3.2. Applicability Domain of the Model

The leverage (h) and standardized residual approach, described in the literature as a Williams plot (Figure 5A), was used for the analysis of the applicability domain. The study shows the graph for the training and prediction sets. In addition, the Insubria plot allows easy visual inspection of the diagonal values of the leverage values (HAT) against the predicted responses for each compound. This second approach was introduced in the QSARINS software (Version 2.2.4 year 2019) and is shown in Figure 5B.

3.3. Validation of the Developed Model

The results of the internal validation experiments demonstrated both fitness and stability of the model. The most important techniques for internal validation are LOO, LMO, and Y-scrambling. The values achieved in these experiments showed a Q2LOO value of 0.680, an RMSEcv value of 0.409, and MAEcv = 0.316. Figure 6A shows the values predicted by LOO vs. the experimental LogKd values. The model performance for internal validation using LMO was Q2LMO = 0.648; Figure 6B shows the correlation between descriptors and DAT binding affinity (Q2LMO vs. KXY values).
The results of the Y-scrambling procedure are shown in Figure 7: the values of R2 and Q2 of every iteration (yellow and red circles, respectively) and their averages were R2Y-scr = 0.098 and Q2Y-scr = −0.174, as well as the respective values of these parameters for the model (light and dark blue, for R2 and Q2, respectively).
The predictability of the model was tested using the set of external predictions of 12 compounds. Its parameters were R2ext = 0.709, RMSEext = 0.339, MAEext = 0.302, PRESSext = 1.381, Q2-F1 = 0.298, Q2-F2 = 0.250, Q2-F3 = 0.780, CCCext = 0.772, r2m_aver = 0.424, and r2m_ delta = 0.331, where R2ext is the external determination coefficient; RMSEext is the root-mean-square error in the external prediction; MAEext is the mean absolute error in the external prediction; PRESSext is the predictive residual sum of squares (external validation); Q2-F1, Q2-F2, and Q2-F3 are the explained variances in the external prediction; CCCext is the concordance correlation coefficient; and r2m-aver and r2m-delta are the Roy criteria, average and delta, respectively [76]. The predictions of compounds in the external set are shown in Figure 4, Figure 5 and Figure 6 (blue circles) and Table 3.

3.4. Comparison with Other Approaches

In this study, our MLR model is compared with other approaches from the literature in the task of predicting DAT inhibitors. The results obtained by using the MLR-based model are shown in Table 4, where the models obtained with other approaches are also shown.
A comparative analysis of the presented models reveals key differences in performance, dataset composition, and validation rigor. As shown in Table 4, the previously developed studies to identify novel compounds with potential radiopharmaceutical applications were all based on congeneric series, which limits their applicability to chemical structures that differ from those used during model training. In contrast, our model was developed using a dataset with greater structural diversity, thereby enhancing its ability to predict compounds outside the original chemical space.
We would like to highlight that the model targeting the adenosine A2A receptor inhibitors [28] achieved the highest internal coefficient of determination (R2 = 0.901) among all the models; however, it also exhibited the lowest external R2 value (0.540), indicating limited predictive performance on the test set. Similarly, the PLS model for dopamine D2 receptor [27] showed acceptable internal performance (R2 = 0.731; Q2LOO = 0.623) as well as external performance (R2ext = 0.742), but also relied on a small, structurally homogeneous dataset.
Among the models targeting the dopamine transporter (DAT), the PLS model reported by de Oliveira et al. [30] achieved the highest external predictivity (R2ext = 0.997), along with strong internal validation metrics (Q2LOO = 0.761; Q2LMO = 0.746). However, such a pronounced discrepancy between internal fit values and external predictability is uncommon in computational modeling and is likely attributable to the specific selection of the external test set. The models reported by Mavel et al. [29] were built using the largest dataset (69 compounds), which is advantageous in terms of statistical power; however, the absence of appropriate cross-validation and external validation metrics weakens confidence in their robustness and predictive reliability. By contrast, the model proposed in the present work stands out because of its use of a structurally diverse dataset and its comprehensive validation strategy. In spite of a slightly lower adjusted R2 (0.755), it demonstrated consistent performance across internal (Q2LOO = 0.680; Q2LMO = 0.648; RMSEcv = 0.409) and external metrics (R2ext = 0.709), suggesting a better balance between robustness, generalizability, and practical utility for identifying novel radiopharmaceutical candidates targeting DAT.

3.5. Virtual Screening and Docking Experiment

The selected compounds were evaluated in the model to predict their LogKd value, and the docking experiment was used to determine the binding mode of the ligand to the dopamine transporter pocket. The predicted LogKd values, obtained with the computational model, and the binding affinities achieved in the docking experiment are shown in Table 5.
Figure 8 shows that the docked structures fit acceptably with the available L-dopamine X-ray crystal structures; all inhibitors were properly oriented at the active center of the dopamine transporter (Figure 8). To validate the docking experiments, we redocked dopamine with its receptor using the same methodology as with the other compounds. The RMSD obtained from the redocking of dopamine (reference ligand) was 0.85 Å, which is within the acceptable range (<2.0 Å) according to the reviewed literature [42]. This result confirms that the docking parameters we used in this work accurately reproduce the crystallographic pose found in the Protein Data Bank (PDB: 8Y2D) [43]. The best poses were conserved and were those that had a ∆G ≤ −6.0 kcal/mol, which is the common threshold for strong protein–ligand interactions [77].

4. Discussion

The development of new computational models demands a suitable goodness-of-fit, robustness, applicability domain, and predictability. The model developed in this study showed an R2 value of 0.755, indicating a suitable fit for modeling DAT binding affinity. This means that the model explains more than 75% of the experimental variance. There was no overfitting in the model, thus representing a good fit with a minimum number of descriptors, because of the low value of the LOF parameter (0.195) and an R2adj = 0.729; the LOF is used to penalize the addition of descriptors in the model equation and should be as low as possible, while the R2adj value should be as similar as possible to the R2 value, which the developed model complied with. The correlation between the descriptors of the model was low since the KXX value (0.236) was small, indicating that there was no redundant information in the selected descriptors. In addition, the correlation between the descriptors and the response variable was appropriate, according to the ΔK parameter (0.076), with a small error in the calculations of both training and prediction estimates (RMSEtr = 0.358; MAEtr = 0.276; s = 0.381). Figure 4 shows the values predicted by the model equation against the experimental LogKd values for both training and prediction series. As can be seen, most of the points are close to the line, and three compounds with atypical behavior in comparison to the rest of the molecules in the database were detected as outliers and not used to develop the model. Table 2 shows the predicted values for the compounds in the training set and the predictions in the LOO experiments as well as the residuals with respect to experimental values and leverage values (which were used to establish the applicability domain). The predictions are quite similar to the experimental values for most compounds. The molecular descriptors included in the model can be seen in Table 6 with the general interpretation of the molecular feature that describes each descriptor and the descriptor block to which they belong in the software.
A critical aspect in QSAR studies is the definition of the AD of the models. Next, we used two approaches to establish the chemical space described by our model. First, we used a Williams graph (Figure 5A): the AD is defined as the area at the left of a leverage threshold h* = 0.3571 and within ±3 standard deviations. As shown in the figure, most compounds are within the applicability domain of the model. There is only one compound (49, belonging to the training set) with a leverage value (h = 0.2327) greater than the critical leverage (h*), although it shows the SD value within the limits; therefore, the prediction should be considered with caution. In addition, we used another approach available in QSARINS software, the Insubria graph (Figure 5B), which plots the leverage values against the predicted responses for each compound. The results agree with the previous approach, identifying the same compound outside the AD with a leverage value greater than the cutoff value, but in this approach two compounds in the prediction set (36 and 56) present LogKd values that were slightly overestimated; see Table 3 for details.
To evaluate the robustness and stability of the model, several internal validation strategies were performed. First, an LOO cross-validation technique, which iteratively excludes a compound of the dataset: The variance explained in the prediction by LOO was Q2LOO = 0.680, with a small error in the predictions (RMSEcv = 0.409 and MAEcv = 0.316); these results are shown in Figure 6A. Second, an internal validation by LMO was performed, which was developed by leaving out 30% of the dataset to study the behavior of our model. The value obtained for this experiment (Q2LMO = 0.648) was like the value previously achieved in LOO, although it is important to notice that this technique gives better results with larger databases. The Q2LMO values (red circles) were similar to each other and distributed around the Q2 of the model (blue circle), corroborating its good fit and stability (see Figure 6B). In the Y-scrambling experiment, the absence of change in correlation for the model was checked. The expected behavior is that both values of R2Y-scr and Q2Y-scr (for each iteration) should differ appreciably from the R2 and Q2 values of the model. Both values of R2 and Q2 for every iteration are shown in Figure 6 (yellow and red circles, respectively), and their averages were R2Y-scr = 0.098 and Q2Y-scr = −0.174, quite far from the respective values of these parameters for the model (light and dark blue for R2 and Q2, respectively), confirming its validity.
To evaluate the model’s ability to predict new compounds, an external validation was also performed. The procedure was carried out by applying the model equation developed with the training set to the set of external predictions. Based on the results with the external set, we can say that the model has a good predictive power (R2ext = 0.709), with lower value of 0.339 for RMSEext. As can be seen in Figure 5A, all compounds in the prediction set are within the applicability domain of the model. Table 3 shows the predicted values for the compounds in the prediction set and the residuals with respect to experimental values and the leverage values. The predicted values are quite similar to the experimental values, demonstrating the predictive power of the developed model.
The validated model was then used to predict the affinity for DAT of new molecules of interest. For this, we selected a set of eleven compounds with different actions over the CNS; among the activities of these compounds, we can find psychostimulant, centrally acting sympathomimetic agent, anorectic psychostimulant, centrally acting anticholinergic, a radiopharmaceutical used to diagnose Parkinsonism, nasal decongestant for systemic use, and psychedelic. This feature ensures that they can reach the DAT and makes the evaluation of these compounds in virtual screening more interesting. This group of compounds was evaluated with the computational model and assessed by docking to estimate the binding affinities for the DAT.
Among the compounds selected for virtual screening, 123I-Ioflupane (one of the most widely used DAT radiotracers) was chosen, which achieved a good prediction for the LogKd value of 3.088; taking this as a point of reference, we highlighted the compounds with affinity for DAT potentially better than 123I-Ioflupane. We found that Modafinil and Armodafinil seem to be the better compounds showing the lowest LogKd value of 2.386, followed by Benztropine with 2.449 and Procaine with a value of 2.713, slightly lower than the 3.088 shown by 123I-Ioflupane. The results obtained with the QSAR model fully agree with those obtained in the docking experiments, which are described below.
The results of the docking experiments are shown in Table 5. As can be seen, all the ligands present binding energies (ΔGbinding) between −5.278 and −7.303 kcal/mol, with the most negative energy corresponding to the complex formed between the ligand Ioflupane (009) and the dopamine transporter, with a binding free energy of −7.303 kcal/mol. The stability of this complex is based on the two H-bond interactions with Asp68 and Ser72, in addition to the π-π stacking interaction with Tyr355 (Sandwich Type) (Figure 9C).
The second binding energy corresponded to the complex formed between Modafinil (001) and the dopamine receptor, with a binding free energy of −6.669 kcal/mol. The stability of this complex is based on two π-π stacking interactions shared with Phe332 (T-Shaped and Sandwich Type) (Figure 9A). The third and fourth most negative binding free energies were found between the complexes formed between Procaine (003) (−6.452 kcal/mol) and Armodafinil (010) (−6.436 kcal/mol). The stability of the Procaine–dopamine transporter complex is based on one H-bond interaction with Asp79, and two π-π stacking interactions with Phe320 (Sandwich Type) and Phe326 (T-Shaped Type) (Figure 9B). The stability of the Armodafinil–dopamine transporter complex is based on two hydrogen bond (H-Bond) interactions shared with Leu459 (Figure 9D), in addition to the π-π stacking interaction with Phe462 (T-Shaped Type) (Figure 9D). The least stable complex of all those subjected to docking experiments was the one formed between the ligand Phenmetrazine (004) and the dopamine transporter, as well as the least stable complex, corresponding to Phentermine (002) and the dopamine transporter. These two ligands were oriented away from the active center of the protein with loss of non-covalent interactions (Figure 10).
Below, we present a detailed comparison of the results of our docking experiments (ΔGbinding and RMSD) with recently published data from computational and experimental studies. This comparison serves to validate the consistency and reliability of our findings. For example, a recent study developed robust QSAR models on approximately 61 DAT inhibitors, including drugs similar to ours, and obtained ΔGbinding values very similar to ours. Phenmetrazine (L-004) in this study behaved very similarly to ours, with ΔGbinding ≈ −6.8 kJ/mol [78].
To support the validity of our data, we compared the docking results with recent experimental affinities in the scientific literature. Modafinil has been estimated to have a Ki ≈ 2600 nM (≈2.6 µM) per human DAT, which is in agreement with our finding of ΔGbinding ≈ −5.99 kJ/mol, reflecting a low affinity for the dopamine receptor, a result that is in agreement with our study. Furthermore, an analogous relationship between experimental affinity in this study and the binding energies observed in our set of molecules (with several ligands in the range −7.0 to −7.5 kJ/mol with RMSD < 2 Å) has been found, corroborating the predictive power of our docking experiments [79].
Molecular dynamics simulations provide us with information about the behavior of ligand–protein complexes over time at the atomic level [70,80,81,82]. In our case, they also allow us to determine the stability of the systems and whether the interactions found in the docking experiments are maintained over time. To this end, we analyzed several parameters obtained from the trajectory, which will be discussed below. One of the important parameters when analyzing the stability of ligand–protein complexes over time is the RMSD [47,83,84]. The behavior of this variable over time for all the complexes studied is shown in Figure 11. As can be seen in Figure 2, all complexes subjected to molecular dynamics simulations maintained low RMSD values (<2 Å), indicating that these complexes remained stable throughout the simulation period. This result is consistent with that obtained from the docking experiments. The lowest RMSD values were found in the complex formed by Benztropine and the dopamine receptor, meaning that it was the most stable complex over time. This complex had the most negative energy obtained in the docking experiments, coinciding with the results obtained from the docking experiments and the molecular dynamics experiments; therefore, we can expect that this molecule could be a good candidate as a radiotracer for Parkinson’s disease diagnosis.
One of the parameters that could explain the stability of compounds over time is hydrogen bond interactions (Figure 12). As can be seen in Figure 12, the number of hydrogen bond interactions during the trajectory was low. The highest number of hydrogen bond interactions was found in the complex formed by Procaine and the dopamine transporter. Given the structure of this molecule (Figure 3), there is only one possibility of hydrogen bond interaction as the hydrogen acceptor: the carbonyl oxygen of this ligand, which interacts strongly and stably with Asp79 of the dopamine transporter with an occupancy of 88.81% (Table 7). This interaction could give the complex strong stability during the simulation time.
The second complex with stable hydrogen bond interactions over time was found in the complex formed by diethylpropion with the dopamine transporter. This complex maintained strong interactions with Asp421 with an occupancy of 60.98% (Table 7), which is considered a stable interaction over time. This behavior could explain the low RMSD values found during the 100 ns trajectory.
In the case of the complex formed by Benztropine and the dopamine transporter, no stable hydrogen bond interactions were found over time. Given the structure of this molecule (Figure 11), it has little chance of interacting by hydrogen bond with the amino acids of the dopamine transporter, because it only has one hydrogen acceptor group and it is an oxygen of the ether group within Benztropine. The stability of this complex found in the RMSD parameter could be because of another type of interaction such as π-π staking, Van del Waals interactions, or solvation type.
Another parameter usually used to analyze ligand–protein complexes is the radius of gyration. This variable provides information about the degree of compaction of a complex during the simulation time. As can be seen in Figure 12, the two most compact complexes were those formed by Procaine and Benztropine, with radii of gyration values below 4 Å. This result is consistent with the RMSD results and with the results obtained from the docking experiments, so we can conclude that these two compounds could be good markers for Parkinson’s disease.

5. Conclusions

In the present study, a QSAR-MLR model was developed by using molecular descriptors calculated with the Dragon software, which adequately predicts the binding affinity for DAT. The model developed with the QSARINS software fulfilled all regulatory principles established by the OECD. The robustness of the model was tested through several internal validation techniques (LOO, LMO, and Y-scrambling), and its predictability was tested using a set of external predictions (external validation). The developed model was used to perform a virtual screening of compounds with different actions on the CNS. The model predicts four compounds with LogKd values lower than the 123I-Ioflupane value; these compounds should present good binding affinities for DAT. The docking experiments corroborate the results of the model prediction and give information about the main interactions of these compounds with the active site. The combination of both strategies strengthens the probabilities of identifying new compounds with potential affinity for DAT. In conclusion, the proposed computational tool is efficient in the identification of novel DAT radiotracers.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/app15158134/s1.

Author Contributions

Conceptualization, J.A.C.-G., M.S.-M., and F.P.-G.; methodology, M.S.-M. and K.M.-U.; software, K.M.-U.; validation, M.R.-O. and V.P.-D.; formal analysis, V.P.-D.; investigation, J.A.C.-G. and K.M.-U.; resources, M.R.-O.; data curation, M.S.-M. and V.P.-D.; writing—original draft preparation, J.A.C.-G. and M.S.-M.; writing—review and editing, J.A.C.-G., F.P.-G., and F.T.; visualization, F.P.-G.; supervision, F.T. and F.P.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data required to reproduce the results of this paper can be found directly in the main text (Table 1 and Figure 1, Figure 2 and Figure 3).

Acknowledgments

The authors are grateful to Paola Gramatica and her group of the University of Insubria for kindly providing the QSARINS software for this study. Castillo-Garit, J.A. thanks the program ‘Estades Temporals per a Investigators Convidats’ for a fellowship to work at Valencia University in 2018. Torrens, F. thanks the Universitat de Valencia for the Special Research Actions Funding 2024. Powered@NLHPC: This research was partially supported by the supercomputing infrastructure of the NLHPC (ECM-02) of the Universidad de Chile.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
PDParkinson’s disease
DATDopamine active transporter
QSARQuantitative Structure–Activity Relationship
KdDissociation equilibrium constant
MDsMolecular descriptors
SDStandard deviation
SPECTSingle-Photon Emission Computed Tomography
PETPositron Emission Tomography
MLRMultiple linear regression
GAGenetic algorithm
OECDOrganization for Economic Cooperation and Development
ADApplicability domain
CVCross-validation
LOOLeaving One Out
LMOLeaving Many Out
CNSCentral nervous system
RMSDRoot-mean-square deviation
HATLeverage values
PDBProtein data bank

References

  1. Hafeez, M.; Eoff, E.; Wei, J.; Azhar, G. Missed Insights for Earlier Management of Parkinson’s Disease and the Value of Dopamine Transporter (DAT) Scans. Geriatrics 2024, 9, 126. [Google Scholar] [CrossRef] [PubMed]
  2. Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef] [PubMed]
  3. Kouli, A.; Torsney, K.M.; Kuan, W.-L. Parkinson’s Disease: Etiology, Neuropathology, and Pathogenesis. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Exon Publications: Brisbane City, QLD, Australia, 2018; pp. 3–26. [Google Scholar]
  4. Balasubramanian, S.; Mehmood, K.T.; Al-Baldawi, S.; Zúñiga Salazar, G.; Zúñiga, D. Behind the Mask: Parkinson’s Disease and Depression. Cureus 2024, 16, e52663. [Google Scholar] [CrossRef] [PubMed]
  5. Postuma, R.B.; Berg, D.; Stern, M.; Poewe, W.; Olanow, C.W.; Oertel, W.; Obeso, J.; Marek, K.; Litvan, I.; Lang, A.E. MDS clinical diagnostic criteria for Parkinson’s disease. Mov. Disord. 2015, 30, 1591–1601. [Google Scholar] [CrossRef] [PubMed]
  6. Rizzo, G.; Copetti, M.; Arcuti, S.; Martino, D.; Fontana, A.; Logroscino, G. Accuracy of clinical diagnosis of Parkinson disease: A systematic review and meta-analysis. Neurology 2016, 86, 566–576. [Google Scholar] [CrossRef] [PubMed]
  7. Xiromerisiou, G.; Boura, I.; Barmpounaki, E.; Georgoulias, P.; Dardiotis, E.; Spanaki, C.; Valotassiou, V. The Utilization and Impact of Dopamine Transporter Imaging in Diagnosing Movement Disorders at a Tertiary Care Hospital in Greece. Biomedicines 2025, 13, 970. [Google Scholar] [CrossRef] [PubMed]
  8. Höglinger, G.U.; Respondek, G.; Stamelou, M.; Kurz, C.; Josephs, K.A.; Lang, A.E.; Mollenhauer, B.; Müller, U.; Nilsson, C.; Whitwell, J.L. Clinical diagnosis of progressive supranuclear palsy: The movement disorder society criteria. Mov. Disord. 2017, 32, 853–864. [Google Scholar] [CrossRef] [PubMed]
  9. O’sullivan, S.S.; Williams, D.R.; Gallagher, D.A.; Massey, L.A.; Silveira-Moriyama, L.; Lees, A.J. Nonmotor symptoms as presenting complaints in Parkinson’s disease: A clinicopathological study. Mov. Disord. 2008, 23, 101–106. [Google Scholar] [CrossRef] [PubMed]
  10. Thobois, S.; Prange, S.; Scheiber, C.; Broussolle, E. What a neurologist should know about PET and SPECT functional imaging for parkinsonism: A practical perspective. Park. Relat. Disord. 2018, 59, 93–100. [Google Scholar] [CrossRef] [PubMed]
  11. Nepal, B.; Das, S.; Reith, M.E.; Kortagere, S. Overview of the structure and function of the dopamine transporter and its protein interactions. Front. Physiol. 2023, 14, 1150355. [Google Scholar] [CrossRef] [PubMed]
  12. Zuo, J.; Kang, J.; Hong, J.; Li, J.; Fang, Y.; Liu, C.; Xie, M.; Chen, Z. Synthesis and Preliminary Evaluation of an In Vivo Stable 131I-Labeled Deuterated Tropane for Mapping Dopamine Transporter. J. Label. Compd. Radiopharm. 2025, 68, e4147. [Google Scholar] [CrossRef] [PubMed]
  13. Palermo, G.; Ceravolo, R. Molecular Imaging of the Dopamine Transporter. Cells 2019, 8, 872. [Google Scholar] [CrossRef] [PubMed]
  14. Kerstens, V.S.; Fazio, P.; Sundgren, M.; Halldin, C.; Svenningsson, P.; Varrone, A. [18F]FE-PE2I DAT correlates with Parkinson’s disease duration, stage, and rigidity/bradykinesia scores: A PET radioligand validation study. EJNMMI Res. 2023, 13, 29. [Google Scholar] [CrossRef] [PubMed]
  15. de Natale, E.R.; Niccolini, F.; Wilson, H.; Politis, M. Chapter Five—Molecular Imaging of the Dopaminergic System in Idiopathic Parkinson’s Disease. In International Review of Neurobiology; Politis, M., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 141, pp. 131–172. [Google Scholar]
  16. Darcourt, J.; Booij, J.; Tatsch, K.; Varrone, A.; Vander Borght, T.; Kapucu, Ö.L.; Någren, K.; Nobili, F.; Walker, Z.; Van Laere, K. EANM procedure guidelines for brain neurotransmission SPECT using 123 I-labelled dopamine transporter ligands, version 2. Eur. J. Nucl. Med. Mol. Imaging. 2010, 37, 443–450. [Google Scholar] [CrossRef] [PubMed]
  17. Agencia Europea de, M. Ficha Técnica de DATSCAN (Ioflupano [123I]) Solución Inyectable. 2022. Available online: https://www.sen.es/pdf/2025/GuiaParkinson2024.pdf (accessed on 24 March 2025).
  18. Booij, J.; Tissingh, G.; Boer, G.; Speelman, J.; Stoof, J.; Janssen, A.; Wolters, E.C.; Van Royen, E. [123I] FP-CIT SPECT shows a pronounced decline of striatal dopamine transporter labelling in early and advanced Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 1997, 62, 133–140. [Google Scholar] [CrossRef] [PubMed]
  19. Weng, C.-C.; Huang, S.-L.; Chen, Z.-A.; Lin, K.-J.; Hsiao, T.; Yen, T.-C.; Kung, M.-P.; Wey, S.-P.; Hsu, C.-H. [18F] FP-(+)-DTBZ PET study in a lactacystin-treated rat model of Parkinson disease. Ann. Nucl. Med. 2017, 31, 506–513. [Google Scholar] [CrossRef] [PubMed]
  20. DiMasi, J.A.; Grabowski, H.G.; Hansen, R.W. Innovation in the pharmaceutical industry: New estimates of R&D costs. J. Health Econ. 2016, 47, 20–33. [Google Scholar] [CrossRef] [PubMed]
  21. Galvez, J.; Galvez-Llompart, M.; Garcia-Domenech, R. Introduction to molecular topology: Basic concepts and application to drug design. Curr. Comput.-Aided Drug Des. 2012, 8, 196–223. [Google Scholar] [CrossRef] [PubMed]
  22. Marrero-Ponce, Y.; Martínez-Albelo, E.R.; Casañola-Martín, G.M.; Castillo-Garit, J.A.; Echevería-Díaz, Y.; Zaldivar, V.R.; Tygat, J.; Borges, J.E.R.; García-Domenech, R.; Torrens, F.; et al. Bond-based linear indices of the non-stochastic and stochastic edge-adjacency matrix. 1. Theory and modeling of ChemPhys properties of organic molecules. Mol. Div. 2010, 14, 731–753. [Google Scholar] [CrossRef] [PubMed]
  23. Castillo-Garit, J.A.; del Toro-Cortés, O.; Vega, M.C.; Rolón, M.; Rojas de Arias, A.; Casañola-Martin, G.M.; Escario, J.A.; Gómez-Barrio, A.; Marrero-Ponce, Y.; Torrens, F.; et al. Bond-based bilinear indices for computational discovery of novel trypanosomicidal drug-like compounds through virtual screening. Eur. J. Med. Chem. 2015, 96, 238–244. [Google Scholar] [CrossRef] [PubMed]
  24. Brito-Sánchez, Y.; Castillo-Garit, J.A.; Le-Thi-Thu, H.; González-Madariaga, Y.; Torrens, F.; Marrero-Ponce, Y.; Rodríguez-Borges, J.E. Comparative study to predict toxic modes of action of phenols from molecular structures. SAR QSAR Environ. Res. 2013, 24, 235–251. [Google Scholar] [CrossRef] [PubMed]
  25. Canizares-Carmenate, Y.; Mena-Ulecia, K.; Perera-Sardina, Y.; Torrens, F.; Castillo-Garit, J.A. An approach to identify new antihypertensive agents using Thermolysin as model: In silico study based on QSARINS and docking. Arab. J. Chem. 2019, 12, 4861–4877. [Google Scholar] [CrossRef]
  26. Castillo-Garit, J.A.; Marrero-Ponce, Y.; Barigye, S.J.; Medina-Marrero, R.; Bernal, M.G.; de la Vega, J.M.G.; Torrens, F.; Arán, V.J.; Pérez-Giménez, F.; García-Domenech, R.; et al. In silico Antibacterial Activity Modeling Based on the TOMOCOMD-CARDD Approach. J. Braz. Chem. Soc. 2015, 26, 1218–1226. [Google Scholar]
  27. De, P.; Roy, K. QSAR modeling of PET imaging agents for the diagnosis of Parkinson’s disease targeting dopamine receptor. Theor. Chem. Acc. 2020, 139, 176. [Google Scholar] [CrossRef]
  28. De, P.; Roy, J.; Bhattacharyya, D.; Roy, K. Chemometric modeling of PET imaging agents for diagnosis of Parkinson’s disease: A QSAR approach. Struct. Chem. 2020, 31, 1969–1981. [Google Scholar] [CrossRef]
  29. Mavel, S.; Mincheva, Z.; Méheux, N.; Carcenac, Y.; Guilloteau, D.; Abarbri, M.; Emond, P. QSAR study and synthesis of new phenyltropanes as ligands of the dopamine transporter (DAT). Bioorg. Med. Chem. 2012, 20, 1388–1395. [Google Scholar] [CrossRef] [PubMed]
  30. de Oliveira, L.H.D.; Cruz, J.N.; dos Santos, C.B.R.; de Melo, E.B. Multivariate QSAR, similarity search and ADMET studies based in a set of methylamine derivatives described as dopamine transporter inhibitors. Mol. Div. 2024, 28, 2931–2946. [Google Scholar] [CrossRef] [PubMed]
  31. Carlier, P.R.; Lo, M.M.; Lo, P.C.; Richelson, E.; Tatsumi, M.; Reynolds, I.J.; Sharma, T.A. Synthesis of a potent wide-spectrum serotonin-, norepinephrine-, dopamine-reuptake inhibitor (SNDRI) and a species-selective dopamine-reuptake inhibitor based on the gamma-amino alcohol functional group. Bioorg. Med. Chem. Lett. 1998, 8, 487–492. [Google Scholar] [CrossRef] [PubMed]
  32. Tatsumi, M.; Groshan, K.; Blakely, R.D.; Richelson, E. Pharmacological profile of antidepressants and related compounds at human monoamine transporters. Eur. J. Pharmacol. 1997, 340, 249–258. [Google Scholar] [CrossRef] [PubMed]
  33. Pristupa, Z.B.; Wilson, J.M.; Hoffman, B.J.; Kish, S.J.; Niznik, H.B. Pharmacological heterogeneity of the cloned and native human dopamine transporter: Disassociation of [3H]WIN 35,428 and [3H]GBR 12,935 binding. Mol. Pharmacol. 1994, 45, 125–135. [Google Scholar] [CrossRef] [PubMed]
  34. Dragon, version 7.0.10; Software for Molecular Descriptor Calculation; Kode srl: Cagliari, Italy, 2017. Available online: https://chm.kode-solutions.net (accessed on 24 March 2025).
  35. Gramatica, P.; Chirico, N.; Papa, E.; Cassani, S.; Kovarich, S. QSARINS: A new software for the development, analysis and validation of QSAR MLR models. J. Comput. Chem. 2013, 34, 2121–2132. [Google Scholar] [CrossRef]
  36. Gramatica, P.; Chirico, N.; Papa, E.; Cassani, S.; Kovarich, S. QSARINS, Software for QSAR MLR Model Development and Validation; QSAR Research Unit in Environmental Chemistry and Ecotoxicology, University of Insubria: Varese, Italy, 2013; (Version 2.2.4 year 2019).
  37. Gramatica, P.; Cassani, S.; Chirico, N. QSARINS-chem: Insubria datasets and new QSAR/QSPR models for environmental pollutants in QSARINS. J. Comput. Chem. 2014, 35, 1036–1044. [Google Scholar] [CrossRef] [PubMed]
  38. Worth, A.P.; Bassan, A.; De Bruijn, J.; Gallego-Saliner, A.; Netzeva, T.; Patlewicz, G.; Pavan, M.; Tsakovska, I.; Eisenreich, S. The role of the European Chemicals Bureau in promoting the regulatory use of (Q)SAR methods. SAR QSAR Environ. Res. 2007, 18, 111–125. [Google Scholar] [CrossRef] [PubMed]
  39. Canizares-Carmenate, Y.; Hernandez-Morfa, M.; Torrens, F.; Castellano, G.; Castillo Garit, J.A. Larvicidal activity prediction against <i>Aedes aegypti</i> mosquito using computational tools. J. Vector Borne Dis. 2017, 54, 164–171. [Google Scholar] [PubMed]
  40. Atkinson, A.C. Plots, Transformations, and Regression: An Introduction to Graphical Methods of Diagnostic Regression Analysis; Clarendon Press: Oxford, UK, 1985. [Google Scholar]
  41. Rücker, C.; Rücker, G.; Meringer, M. y-Randomization and Its Variants in QSPR/QSAR. J. Chem. Inf. Model. 2007, 47, 2345–2357. [Google Scholar] [CrossRef] [PubMed]
  42. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Y.; Wang, X.; Meng, Y.; Hu, T.; Zhao, J.; Li, R.; Bai, Q.; Yuan, P.; Han, J.; Hao, K.; et al. Dopamine reuptake and inhibitory mechanisms in human dopamine transporter. Nature 2024, 632, 686–694. [Google Scholar] [CrossRef] [PubMed]
  44. Halgren, T.A.; Murphy, R.B.; Friesner, R.A.; Beard, H.S.; Frye, L.L.; Pollard, W.T.; Banks, J.L. Glide:  A New Approach for Rapid, Accurate Docking and Scoring. 2. Enrichment Factors in Database Screening. J. Med. Chem. 2004, 47, 1750–1759. [Google Scholar] [CrossRef] [PubMed]
  45. Friesner, R.A.; Banks, J.L.; Murphy, R.B.; Halgren, T.A.; Klicic, J.J.; Mainz, D.T.; Repasky, M.P.; Knoll, E.H.; Shelley, M.; Perry, J.K.; et al. Glide:  A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy. J. Med. Chem. 2004, 47, 1739–1749. [Google Scholar] [CrossRef] [PubMed]
  46. Manglik, A.; Kruse, A.C.; Kobilka, T.S.; Thian, F.S.; Mathiesen, J.M.; Sunahara, R.K.; Pardo, L.; Weis, W.I.; Kobilka, B.K.; Granier, S. Crystal structure of the µ-opioid receptor bound to a morphinan antagonist. Nature 2012, 485, 321–326. [Google Scholar] [CrossRef] [PubMed]
  47. Bell, E.W.; Zhang, Y. DockRMSD: An open-source tool for atom mapping and RMSD calculation of symmetric molecules through graph isomorphism. J. Cheminform. 2019, 11, 40. [Google Scholar] [CrossRef] [PubMed]
  48. Velázquez-Libera, J.L.; Durán-Verdugo, F.; Valdés-Jiménez, A.; Núñez-Vivanco, G.; Caballero, J. LigRMSD: A web server for automatic structure matching and RMSD calculations among identical and similar compounds in protein-ligand docking. Bioinformatics 2020, 36, 2912–2914. [Google Scholar] [CrossRef] [PubMed]
  49. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef] [PubMed]
  50. Berman, H.M.; Battistuz, T.; Bhat, T.N.; Bluhm, W.F.; Bourne, P.E.; Burkhardt, K.; Feng, Z.; Gilliland, G.L.; Iype, L.; Jain, S.; et al. The Protein Data Bank. Acta Crystallogr. D 2002, 58, 899–907. [Google Scholar] [CrossRef] [PubMed]
  51. Berman, H.; Henrick, K.; Nakamura, H. Announcing the worldwide Protein Data Bank. Nat. Struct. Mol. Biol. 2003, 10, 980. [Google Scholar] [CrossRef] [PubMed]
  52. DeLano, W.L. PyMOL: An open-source molecular graphics tool. CCP4 Newsl. Protein Crystallogr. 2002, 40, 82–92. [Google Scholar]
  53. Yuan, S.; Chan, H.C.S.; Hu, Z. Using PyMOL as a platform for computational drug design. WIREs Comput. Mol. Sci. 2017, 7, e1298. [Google Scholar] [CrossRef]
  54. Meng, E.C.; Goddard, T.D.; Pettersen, E.F.; Couch, G.S.; Pearson, Z.J.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Tools for structure building and analysis. Protein Sci. 2023, 32, e4792. [Google Scholar] [CrossRef] [PubMed]
  55. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Meng, E.C.; Couch, G.S.; Croll, T.I.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Structure visualization for researchers, educators, and developers. Protein Sci. 2021, 30, 70–82. [Google Scholar] [CrossRef] [PubMed]
  56. Goddard, T.D.; Huang, C.C.; Meng, E.C.; Pettersen, E.F.; Couch, G.S.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Meeting modern challenges in visualization and analysis. Protein Sci. 2018, 27, 14–25. [Google Scholar] [CrossRef] [PubMed]
  57. Huang, J.; Rauscher, S.; Nawrocki, G.; Ran, T.; Feig, M.; de Groot, B.L.; Grubmüller, H.; MacKerell, A.D. CHARMM36m: An improved force field for folded and intrinsically disordered proteins. Nat. Methods 2017, 14, 71–73. [Google Scholar] [CrossRef] [PubMed]
  58. Zoete, V.; Cuendet, M.A.; Grosdidier, A.; Michielin, O. SwissParam: A fast force field generation tool for small organic molecules. J. Comput. Chem. 2011, 32, 2359–2368. [Google Scholar] [CrossRef] [PubMed]
  59. Jo, S.; Kim, T.; Iyer, V.G.; Im, W. CHARMM-GUI: A web-based graphical user interface for CHARMM. J. Comput. Chem. 2008, 29, 1859–1865. [Google Scholar] [CrossRef] [PubMed]
  60. Brooks, B.R.; Brooks Iii, C.L.; Mackerell Jr, A.D.; Nilsson, L.; Petrella, R.J.; Roux, B.; Won, Y.; Archontis, G.; Bartels, C.; Boresch, S.; et al. CHARMM: The biomolecular simulation program. J. Comput. Chem. 2009, 30, 1545–1614. [Google Scholar] [CrossRef] [PubMed]
  61. Lee, J.; Cheng, X.; Swails, J.M.; Yeom, M.S.; Eastman, P.K.; Lemkul, J.A.; Wei, S.; Buckner, J.; Jeong, J.C.; Qi, Y.; et al. CHARMM-GUI Input Generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM Simulations Using the CHARMM36 Additive Force Field. J. Chem. Theory Comput. 2016, 12, 405–413. [Google Scholar] [CrossRef] [PubMed]
  62. Boonstra, S.; Onck, P.R.; van der Giessen, E. CHARMM TIP3P Water Model Suppresses Peptide Folding by Solvating the Unfolded State. J. Phys. Chem. B 2016, 120, 3692–3698. [Google Scholar] [CrossRef] [PubMed]
  63. Piskulich, Z.A.; Cui, Q. Hydrogen bonding blues: Vibrational spectroscopy of the TIP3P water model. J. Chem. Phys. 2025, 162, 014104. [Google Scholar] [CrossRef] [PubMed]
  64. Rosas Jiménez, J.G.; Fábián, B.; Hummer, G. Faster Sampling in Molecular Dynamics Simulations with TIP3P-F Water. J. Chem. Theory Comput. 2024, 20, 11068–11081. [Google Scholar] [CrossRef] [PubMed]
  65. Mrad, H.; Fakhari, S.M. Optimization of unconstrained problems using a developed algorithm of spectral conjugate gradient method calculation. Math. Comput. Simul. 2024, 215, 282–290. [Google Scholar] [CrossRef]
  66. Farahmand-Tabar, S.; Abdollahi, F.; Fatemi, M. Robust Conjugate Gradient Methods for Non-smooth Convex Optimization and Image Processing Problems. In Handbook of Formal Optimization; Kulkarni, A.J., Gandomi, A.H., Eds.; Springer Nature: Singapore, 2023; pp. 1–25. [Google Scholar]
  67. Ghimenti, F.; Berthier, L.; Szamel, G.; van Wijland, F. Irreversible Boltzmann samplers in dense liquids: Weak-coupling approximation and mode-coupling theory. Phys. Rev. E 2024, 110, 034604. [Google Scholar] [CrossRef] [PubMed]
  68. Hünenberger, P.H. Thermostat Algorithms for Molecular Dynamics Simulations. In Advanced Computer Simulation: Approaches for Soft Matter Sciences I; Holm, C., Kremer, K., Eds.; Springer: Berlin/Heidelberg, Germany, 2005; pp. 105–149. [Google Scholar]
  69. Yong, X.; Zhang, L.T. Thermostats and thermostat strategies for molecular dynamics simulations of nanofluidics. J. Chem. Phys. 2013, 138, 084503. [Google Scholar] [CrossRef] [PubMed]
  70. Spreiter, Q.; Walter, M. Classical Molecular Dynamics Simulation with the Velocity Verlet Algorithm at Strong External Magnetic Fields. J. Comput. Phys. 1999, 152, 102–119. [Google Scholar] [CrossRef]
  71. Omelyan, I.P.; Mryglod, I.M.; Folk, R. Optimized Verlet-like algorithms for molecular dynamics simulations. Phys. Rev. E 2002, 65, 056706. [Google Scholar] [CrossRef] [PubMed]
  72. Phillips, J.C.; Hardy, D.J.; Maia, J.D.C.; Stone, J.E.; Ribeiro, J.V.; Bernardi, R.C.; Buch, R.; Fiorin, G.; Hénin, J.; Jiang, W.; et al. Scalable molecular dynamics on CPU and GPU architectures with NAMD. J. Chem. Phys. 2020, 153, 044130. [Google Scholar] [CrossRef] [PubMed]
  73. Acun, B.; Hardy, D.J.; Kale, L.V.; Li, K.; Phillips, J.C.; Stone, J.E. Scalable molecular dynamics with NAMD on the Summit system. IBM J. Res. Dev. 2018, 62, 4:1–4:9. [Google Scholar] [CrossRef] [PubMed]
  74. Phillips, J.C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R.D.; Kalé, L.; Schulten, K. Scalable molecular dynamics with NAMD. J. Comput. Chem. 2005, 26, 1781–1802. [Google Scholar] [CrossRef] [PubMed]
  75. Theoretical and Computational Biophysics Group. NAMD 3.0 User’s Guide; University of Illinois at Urbana-Champaign: Champaign, IL, USA, 2023. [Google Scholar]
  76. Castillo-Garit, J.A.; Abad, C.; Casañola-Martin, G.M.; Jones Barigye, S.; Torrens, F.; Torreblanca, A. Prediction of Aquatic Toxicity of Benzene Derivatives to Tetrahymena pyriformis According to OECD Principles. Curr. Pharm. Des. 2016, 22, 5085–5094. [Google Scholar] [CrossRef] [PubMed]
  77. Ciancetta, A.; Cuzzolin, A.; Moro, S. Alternative Quality Assessment Strategy to Compare Performances of GPCR-Ligand Docking Protocols: The Human Adenosine A2A Receptor as a Case Study. J. Chem. Inf. Model. 2014, 54, 2243–2254. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, K.H.; Won, S.J.; Oyinloye, P.; Shi, L. Unlocking the Potential of High-Quality Dopamine Transporter Pharmacological Data: Advancing Robust Machine Learning-Based QSAR Modeling. bioRxiv 2024. [Google Scholar] [CrossRef]
  79. Aggarwal, S.; Mortensen, O.V. Discovery and Development of Monoamine Transporter Ligands. Adv. Neurobiol. 2023, 30, 101–129. [Google Scholar] [CrossRef] [PubMed]
  80. Badar, M.S.; Shamsi, S.; Ahmed, J.; Alam, M.A. Molecular Dynamics Simulations: Concept, Methods, and Applications. In Transdisciplinarity; Rezaei, N., Ed.; Springer International Publishing: Cham, Switzerland, 2022; pp. 131–151. [Google Scholar]
  81. Karplus, M.; McCammon, J.A. Molecular dynamics simulations of biomolecules. Nat. Struct. Mol. Biol. 2002, 9, 646–652. [Google Scholar] [CrossRef] [PubMed]
  82. Kurt, B.; Jürgen, H.; Walter, K.; Wolfgang, P.; Fathollah, V. Molecular dynamics simulations. J. Phys. Condens. Matter. 2004, 16, S429–S453. [Google Scholar] [CrossRef]
  83. Sargsyan, K.; Grauffel, C.; Lim, C. How Molecular Size Impacts RMSD Applications in Molecular Dynamics Simulations. J. Chem. Theory Comput. 2017, 13, 1518–1524. [Google Scholar] [CrossRef] [PubMed]
  84. Brüschweiler, R. Efficient RMSD measures for the comparison of two molecular ensembles. Proteins Struct. Funct. Bioinf. 2003, 50, 26–34. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular structures of compounds used in the training set.
Figure 1. Molecular structures of compounds used in the training set.
Applsci 15 08134 g001
Figure 2. Molecular structures of compounds used in the prediction set.
Figure 2. Molecular structures of compounds used in the prediction set.
Applsci 15 08134 g002
Figure 3. Molecular structure of the set of compounds used in the virtual screening and docking experiments.
Figure 3. Molecular structure of the set of compounds used in the virtual screening and docking experiments.
Applsci 15 08134 g003
Figure 4. Scatter plot of experimental LogKd values vs. those predicted by the model equation.
Figure 4. Scatter plot of experimental LogKd values vs. those predicted by the model equation.
Applsci 15 08134 g004
Figure 5. Analysis of the applicability domain: (A) Williams plot—leverage values vs. standardized residuals; (B) Insubria graph—leverage values vs. predicted data.
Figure 5. Analysis of the applicability domain: (A) Williams plot—leverage values vs. standardized residuals; (B) Insubria graph—leverage values vs. predicted data.
Applsci 15 08134 g005
Figure 6. Internal validation of the model: (A) Scatter plot of experimental LogKd vs. that predicted by LOO experiment; (B) plot of Q2 values of LMO validations compared to the Q2 value of the original model.
Figure 6. Internal validation of the model: (A) Scatter plot of experimental LogKd vs. that predicted by LOO experiment; (B) plot of Q2 values of LMO validations compared to the Q2 value of the original model.
Applsci 15 08134 g006
Figure 7. Scatter plot of the Y-scrambled model compared to the original model.
Figure 7. Scatter plot of the Y-scrambled model compared to the original model.
Applsci 15 08134 g007
Figure 8. Alignment of all docked ligands in complex with the dopamine transporter: (A) represents the re-docked result of L-dopamine (in yellow), obtained from Protein Data Bank X-ray crystallography structure (PDB id: 8Y2D), and the L-dopamine re-docked poses (in magenta). (B) represents the best docking poses of other dopamine transporter inhibitors.
Figure 8. Alignment of all docked ligands in complex with the dopamine transporter: (A) represents the re-docked result of L-dopamine (in yellow), obtained from Protein Data Bank X-ray crystallography structure (PDB id: 8Y2D), and the L-dopamine re-docked poses (in magenta). (B) represents the best docking poses of other dopamine transporter inhibitors.
Applsci 15 08134 g008
Figure 9. Representation of the interactions of the ligands in the active center of the dopamine transporter: (A) Modafinil (001), (B) Procaine (003), (C) Ioflupane (009), and (D) Armodafinil (010).
Figure 9. Representation of the interactions of the ligands in the active center of the dopamine transporter: (A) Modafinil (001), (B) Procaine (003), (C) Ioflupane (009), and (D) Armodafinil (010).
Applsci 15 08134 g009
Figure 10. Graphical representation of the ligands with the lowest binding energy calculated in the docking experiments at the dopamine transporter pocket.
Figure 10. Graphical representation of the ligands with the lowest binding energy calculated in the docking experiments at the dopamine transporter pocket.
Applsci 15 08134 g010
Figure 11. Behavior of the RMSD parameter during the 100 ns of simulation time.
Figure 11. Behavior of the RMSD parameter during the 100 ns of simulation time.
Applsci 15 08134 g011
Figure 12. Radius of gyration of the alpha carbon of the protein in complex with the studied molecules during 100 ns of simulation time.
Figure 12. Radius of gyration of the alpha carbon of the protein in complex with the studied molecules during 100 ns of simulation time.
Applsci 15 08134 g012
Table 1. Reported affinity for DAT expressed as dissociation equilibrium constant (Kd) obtained from the Binding Database.
Table 1. Reported affinity for DAT expressed as dissociation equilibrium constant (Kd) obtained from the Binding Database.
NoLigandKd (nM)Log KdNoLigandKd (nM)Log Kd
1BDBM8207193003.96830BDBM5001424720503.312
2BDBM500694473602.55631BDBM5002809192003.964
3BDBM5006944635003.54432BDBM5001085985003.929
4BDBM5006945242,0004.62333BDBM8144865303.815
5BDBM500694493402.53134BDBM8243718,0004.255
6BDBM50069453701.84535BDBM5010197310003.000
7BDBM5006945021,4004.33036BDBM3100594003.973
8BDBM5006944512,0004.07937BDBM501806614202.623
9BDBM5036656710803.03338BDBM11277711403.057
10BDBM5006945172603.86139BDBM8521843403.637
11BDBM3013036003.55640BDBM5010541784003.924
12BDBM5006944227,0004.43141BDBM5017606221003.322
13BDBM5006944324,0004.38042BDBM5002278410803.033
14BDBM500694443402.53143BDBM5011385151003.708
15BDBM5006945443103.63444BDBM5007344474003.869
16BDBM500694481402.14645BDBM5024041037803.577
17BDBM224164902.69046DB0065513,5004.130
18BDBM5002071232503.51247BDBM1886081003.908
19BDBM2287043103.63448DB0066120,0004.301
20BDBM500483925202.71649BDBM5001771241,0004.613
21BDBM8522039403.59550BDBM5002272329003.462
22BDBM2587028,1004.44951BDBM862072202.342
23BDBM7797021903.34052BDBM5035949928003.447
24BDBM3522931903.50453BDBM8146610603.025
25BDBM8521918,3004.26254BDBM5001767422003.342
26BDBM500280661292.11155DB0098824003.380
27BDBM8254853103.72556BDBM3523416,2004.210
28BDBM5007952712,1004.08357BDBM500219522072.316
29BDBM8243852,0004.716
Table 2. Experimental and predicted values (expressed as log Kd) of DAT affinity for the compounds in the training set.
Table 2. Experimental and predicted values (expressed as log Kd) of DAT affinity for the compounds in the training set.
CompoundExp. a EndpointPredictedResidualPred. LOO bPred. LOO Res.Leverage Value c
13.9683.9740.0063.9750.0070.066
22.5563.0660.5103.1020.5460.065
44.623-outlier-
52.5312.8780.3472.9090.3780.080
61.8452.1830.3382.2720.4270.208
74.3304.3490.0194.3510.0210.122
84.0793.653−0.4263.594−0.4850.122
93.0333.1450.1123.1510.1180.049
103.8614.1870.3264.2270.3660.110
124.431-outlier-
134.3803.491−0.8893.372−1.0080.118
153.6343.545−0.0893.542−0.0930.041
162.1462.021−0.1251.985−0.1610.221
172.6903.0120.3223.0330.3430.062
183.5123.164−0.3493.130−0.3820.087
202.7162.8210.1052.8430.1270.171
213.5953.326−0.2693.305−0.2900.072
224.4494.298−0.1514.283−0.1660.094
233.3403.6790.3393.6960.3560.048
254.2624.136−0.1264.126−0.1360.077
262.1112.6010.4902.6540.5430.097
273.7253.164−0.5623.110−0.6150.087
284.0833.974−0.1093.966−0.1170.066
294.716-outlier-
303.3122.907−0.4052.861−0.4510.103
313.9643.9740.0103.9750.0110.066
323.9293.9740.0453.9770.0480.066
344.2554.058−0.1984.034−0.2210.106
353.0003.0010.0013.0020.0020.106
372.6233.1160.4933.1390.5160.043
383.0573.001−0.0562.995−0.0620.106
403.9243.538−0.3863.488−0.4360.115
423.0333.1450.1123.1510.1180.049
433.7083.538−0.1703.516−0.1920.115
443.8693.276−0.5933.225−0.6450.079
453.5774.2980.7214.3730.7960.094
464.1304.108−0.0224.105−0.0260.129
473.9083.9530.0453.9580.0500.093
484.3013.909−0.3923.764−0.5380.270
494.6134.313−0.3004.114−0.4990.400
512.3422.310−0.0322.299−0.0430.260
523.4473.5380.0913.5500.1030.115
533.0253.5710.5463.7540.7290.250
553.3804.1870.8074.2860.9060.110
572.3162.177−0.1392.127−0.1890.266
a Experimental values; b LOO: Leave-One-Out; c the critical Leverage value is h* = 0.3571.
Table 3. Experimental and predicted values (expressed as LogKd) of DAT affinity for the compounds in the prediction set.
Table 3. Experimental and predicted values (expressed as LogKd) of DAT affinity for the compounds in the prediction set.
CompoundExp. a EndpointPredictedResidualLeverage Value b
33.5443.7070.1630.045
113.5563.116−0.4400.043
142.5312.421−0.1100.118
193.6344.0320.3980.070
243.5043.9740.4700.066
333.8153.650−0.1650.059
363.9734.5110.5380.139
393.6373.164−0.4730.087
413.3223.001−0.3210.106
503.4623.5380.0760.115
543.3423.5170.1750.047
564.2104.5110.3010.139
a Experimental value; b the critical leverage value is h* = 0.3571.
Table 4. Comparison between computational models developed to identify novel compounds with potential radiopharmaceutical applications.
Table 4. Comparison between computational models developed to identify novel compounds with potential radiopharmaceutical applications.
TargetTechniqueStructuresNo Var.N (Train)N (Test)R2R2adjQ2LOOQ2LMORMSEcvR2extRef.
D2PLSCongeneric52770.7310.6960.623nrnr0.742[27]
A2AICP-MLRCongeneric525100.9010.8750.850nrnr0.540[28]
DATPLSCongeneric226100.842nr0.7610.7460.5460.997[30]
DATMLRCongeneric1069np0.8140.692nrnrnrnr[29]
DATPLSCongeneric1069np0.8070.732nrnrnrnr[29]
DATGFACongeneric1069np0.7180.67nrnrnrnr[29]
DATMLRDiverse445120.7550.7290.6800.6480.4090.709TW
PLS: partial least squares; nr: not reported; ICP-MLR: Intelligent Consensus Predictor Multiple Linear Regression; MLR: Multiple Linear Regression; GFA: Genetic Function Approximation; np: not performed; TW: this work.
Table 5. Predicted values of LogKd, calculated binding free energies (kcal/mol), RMSD (Å), and number of H-bond interactions of the first-rank Autodock Vina poses for the complexes studied.
Table 5. Predicted values of LogKd, calculated binding free energies (kcal/mol), RMSD (Å), and number of H-bond interactions of the first-rank Autodock Vina poses for the complexes studied.
MoleculesPredicted LogKdΔGbinding (kcal/mol)RMSD (Å) (Rank)N° H-Bond
Modafinil (001)2.386−6.6692.187 (5)0
Phentermine (002)3.538−5.2782.449 (2)1
Procaine (003)2.713−6.4522.227 (1)1
Phenmetrazine (004)4.024−5.3363.063 (2)0
Diethylpropion (005)3.517−5.6102.814 (3)1
Benztropine (006)2.449−6.4171.568 (2)2
Fencamfamine (007)3.405−5.9933.070 (1)1
Sibutramine (008)4.053−6.0122.795 (2)1
Ioflupane (009)3.088−7.3031.127 (1)2
Armodafinil (010)2.386−6.4362.838 (2)2
Pseudoephedrine (011)4.024−5.7422.332 (2)1
In bold are highlighted the best results.
Table 6. Molecular descriptors selected in the obtained model.
Table 6. Molecular descriptors selected in the obtained model.
DescriptorStructural Feature DescribedDescriptor Block
O-058Hydrogen bonding capacity(Atom-centered fragments) = O
CATS2D_03_ALLipophilicityCATS2D Acceptor-Lipophilic at lag 03
CATS2D_08_LLLipophilicityCATS2D Lipophilic-Lipophilic at lag 08
B08[C-N]Presence/absence of C-N at topological distance 82D Atom pairs
Table 7. H-bond occupancies analysis during 100 ns of simulation time.
Table 7. H-bond occupancies analysis during 100 ns of simulation time.
ComplexesH-Bond InteractionOccupancy (%)
Diethylpropion-8Y2DASP421-Side60.98%
PHE320-Main15.76%
Procaine-8Y2DASP79-Side88.81%
VAL78-Main15.16%
Benztropine-8Y2DLEU459-Main0.08%
ASP231-Side0.04%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Castillo-Garit, J.A.; Soria-Merino, M.; Mena-Ulecia, K.; Romero-Otero, M.; Pérez-Doñate, V.; Torrens, F.; Pérez-Giménez, F. Combining QSAR and Molecular Docking for the Methodological Design of Novel Radiotracers Targeting Parkinson’s Disease. Appl. Sci. 2025, 15, 8134. https://doi.org/10.3390/app15158134

AMA Style

Castillo-Garit JA, Soria-Merino M, Mena-Ulecia K, Romero-Otero M, Pérez-Doñate V, Torrens F, Pérez-Giménez F. Combining QSAR and Molecular Docking for the Methodological Design of Novel Radiotracers Targeting Parkinson’s Disease. Applied Sciences. 2025; 15(15):8134. https://doi.org/10.3390/app15158134

Chicago/Turabian Style

Castillo-Garit, Juan A., Mar Soria-Merino, Karel Mena-Ulecia, Mónica Romero-Otero, Virginia Pérez-Doñate, Francisco Torrens, and Facundo Pérez-Giménez. 2025. "Combining QSAR and Molecular Docking for the Methodological Design of Novel Radiotracers Targeting Parkinson’s Disease" Applied Sciences 15, no. 15: 8134. https://doi.org/10.3390/app15158134

APA Style

Castillo-Garit, J. A., Soria-Merino, M., Mena-Ulecia, K., Romero-Otero, M., Pérez-Doñate, V., Torrens, F., & Pérez-Giménez, F. (2025). Combining QSAR and Molecular Docking for the Methodological Design of Novel Radiotracers Targeting Parkinson’s Disease. Applied Sciences, 15(15), 8134. https://doi.org/10.3390/app15158134

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop