Next Article in Journal
Comparative Bone-Protective Effects of Tocotrienol Isomers from Palm and Annatto in Dexamethasone-Induced Osteoporotic Male Rats
Previous Article in Journal
Interplay Between KLF4, STAT, IRF, and NF-κB in VSMC and Macrophage Plasticity During Vascular Inflammation and Atherosclerosis
Previous Article in Special Issue
Multistage Molecular Simulations, Design, Synthesis, and Anticonvulsant Evaluation of 2-(Isoindolin-2-yl) Esters of Aromatic Amino Acids Targeting GABAA Receptors via π-π Stacking
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Using a Novel Consensus-Based Chemoinformatics Approach to Predict ADMET Properties and Druglikeness of Tyrosine Kinase Inhibitors

by
Evangelos Mavridis
and
Dimitra Hadjipavlou-Litina
*
Laboratory of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(20), 10207; https://doi.org/10.3390/ijms262010207
Submission received: 22 September 2025 / Revised: 14 October 2025 / Accepted: 15 October 2025 / Published: 20 October 2025
(This article belongs to the Special Issue Computational Studies in Drug Design and Discovery)

Abstract

The urgent need to reduce the cost of new drug discovery has led us to create a new, more selective screening method using free chemoinformatics tools to restrict the high failure rates of lead compounds (>90%) during the development process because of the lack of clinical efficacy (40–50%), unmanageable toxicity (30%), and poor drug-like properties (10–15%). Our efforts focused on new molecular entities (NMEs) with reported activity as tyrosine kinase inhibitors (small molecules) as a class of great potential. The criteria for the new method are acceptable Druglikeness, desirable ADME (absorption, distribution, metabolism, and excretion), and low toxicity. After a bibliographic review, we first selected the 29 most promising compounds, always according to the literature, then collected the in silico calculated data from different platforms, and finally processed them together to conclude at 14 compounds meeting the aforementioned criteria. The novelty of the present screening method is that for the evaluation of the compounds for Druglikeness, and ADMET properties (absorption, distribution, metabolism, excretion, and toxicity), the data of the different platforms were used as a whole, rather than the results of each platform individually. Additionally, we validated our new consensus-based method by comparing the final in silico results with the experimental values of FDA (Food and Drug Administration)-approved tyrosine kinase drugs. Using inferential statistics of 39 FDA-approved tyrosine kinase drugs obtained after applying our method, we delineated the intervals of the desired values of the physicochemical properties of future active compounds. Finally, molecular docking studies enhance the credibility of the applied method as an identification tool of Druglikeness.

Graphical Abstract

1. Introduction

Kinases are a ubiquitous group of enzymes that catalyze the phosphoryl transfer reaction from a phosphate donor to a receptor substrate [1]. There are 518 kinases encoded in the human genome that phosphorylate up to one-third of the proteome. Therefore, kinases have been intensively investigated as potential drug targets over the past 30 years. Virtually every signal transduction process occurs via a phosphor-transfer cascade, indicating that kinases provide multiple nodes for therapeutic intervention in many aberrantly regulated biological processes [2].
A rough classification of major kinases is based on the substrate that they phosphorylate. By adding phosphate groups to substrate proteins, protein kinases are key regulators of cell function, localization, and overall function of many proteins serving to orchestrate the activity of almost all cellular processes.
Protein kinases can be further classified according to their substrate residues as tyrosine kinases, serine/threonine kinases, histidine kinases, and cysteine kinases; more specifically, tyrosine kinases can be classified into receptor and non-receptor protein kinases. Receptor tyrosine kinases (RTKs) are membrane-spanning cell-surface proteins that play critical roles in the transduction of extracellular signals into the cytoplasm. Nonreceptor tyrosine kinases (NRTKs-cytoplasmatic), on the other hand, relay intracellular signals [3]. RTKs and NRTKs transfer a phosphoryl group from a nucleoside triphosphate donor to the hydroxyl group of tyrosine residues on protein substrates, triggering the activation of downstream signaling cascades.
Abnormal activation of tyrosine kinases due to mutations, translocations, or amplifications is implicated in the tumorigenesis, progression, invasion, and metastasis of malignancies. Tyrosine kinase inhibitors (TKIs) are designed to inhibit corresponding kinases [4].
The primary goal of drug discovery and development is to find a molecule with optimal pharmacodynamics, desirable pharmacokinetics, low toxicity, and low synthetic complexity. The pharmaceutical industry faces difficulties achieving this goal, as demonstrated by the high failure rates of lead compounds (>90%) during the development process [5].
Analyses of clinical trial data from 2010 to 2017 show four possible reasons attributed to 90% of clinical failures in drug development: lack of clinical efficacy (40–50%), unmanageable toxicity (30%), poor drug-like properties (10–15%), and lack of commercial needs and poor strategic planning (10%) [6]. Therefore, it is obvious that with the help of silico studies, Druglikeness and ADMET properties are improved to minimize poor pharmacokinetics, adverse toxicity, and, in general, low pharmaco-similarity (overall~45%). Analysis of the observed distribution of some key physicochemical properties of approved drugs, including molecular weight, hydrophobicity, and polarity, reveals that they preferentially occupy a relatively narrow range of possible values. Compounds that fall within this range are described as “druglike.” Note that this definition holds in the absence of any obvious structural similarity to an approved drug [7].
Following a thorough review of the last decade’s existing literature, we identified newly synthesized small molecules, developed as inhibitors of tyrosine kinases. These compounds either lacked or had minimal in silico studies on their Druglikeness and ADMET properties. In this study, we introduced a new comprehensive approach to assess Druglikeness and ADMET properties, utilizing a combination of data from various computational platforms. We subsequently validated the reliability of this innovative method by comparing its results with experimental data of FDA-approved tyrosine kinase inhibitor drugs, where standalone computational platforms had previously fallen short. Consequently, we evaluated and classified our examined compounds in terms of their acceptable pharmacosimilarity, desirable pharmacokinetics, and low toxicity. The compounds with the highest evaluation scores underwent additional molecular docking analyses using new protocols, to explore the binding patterns to their biological targets as referenced in the literature (Figure 1).
Finally, by leveraging the extensive data gathered through our novel approach for known drug inhibitors, and utilizing suitable statistical methods, we established confidence intervals for essential physicochemical characteristics. This will serve as a crucial resource in future ligand-based virtual screening aiming to discover new potential inhibitors of tyrosine kinases.

2. Results and Discussion

Several tools are available and useful to predict in silico Druglikeness and ADMET parameters. In this study, the collected data (last accessed on 24 January 2025) are derived from ten software and web servers (Table 1). The platforms listed are widely recognized and frequently utilized, with many of them cited more than 2000 times on Google Scholar, employing the most recent algorithms. The Toxicity Estimation Software Tool 5.1.2 (T.E.S.T) was created by the United States Environmental Protection Agency (EPA).
Based on the above information, the compounds were identified as follows:
  • In terms of complying with known rules of Druglikeness and Medicinal Chemistry such as Lipinski [20], Ghose/CMC-like [21], Veber [22], Egan [5], Muegge [23], MMDR-like [24], Leadlikeness [25], GSK [26], PAINS [27], and Brenk [28];
  • In terms of QED parameter (Quantitative Estimate of Druglikeness) [7];
  • In terms of pharmacokinetic parameters (Bioavailability, Distribution, and Excretion);
  • In terms of toxicity (carcinogenic potential and organ toxicity).
Finally, the above results were quantified, and the compounds were classified to distinguish those that presented the optimal profile (acceptable Druglikeness, desirable pharmacokinetics, low toxicity, and low synthetic complexity).

2.1. The Studied Compounds

Through extended bibliographic research conducted from 2013 to 2023, we identified and selected the most active TKI compounds from each study based on their in vitro inhibition results as IC50 values ranged from less than 1 nM to 770 nM, with one notable exception at 3200 nM.
As illustrated in Table 2, only 9 out of 29 compounds were subjected to in silico analysis regarding their Druglikeness and ADMET properties, utilizing several platforms including Discovery studio 4.0, QikProp (Schrodinger LLC), SwissADME web tool, and PreADMET 2.0, confirming that our consensus-based method will be beneficial for the initial assessment of the substances.

2.2. Calculation/Estimation of Molecular and ADMET Descriptors

For each compound, the average of their molecular descriptors was calculated (Table 3) and tested according to the Druglikeness and Medicinal Chemistry rules (Supplementary Materials, Tables S1 and S2).
The parameters that are directly influenced by the structural and biological variability among the compounds were calculated, since they are essential for evaluating the various Druglikeness rules. We observe that the individual platforms’ data regarding the MW parameter led to comparable results with minor differences, while they produce identical values for the nRbs, nHDr, and MR parameters. A slight yet significant variation is noticed for the nHAc parameter, with the Molsoft platform uniquely calculating TPSA in a different manner. Considerable discrepancies are observed in the estimation of the true value of the Log Po/w parameter, since it is plays a significant role in most Druglikeness models. This variation is correlated to the diverse prediction methods employed across platforms (e.g., ClogP, ALogP, XLogP, MLogP), utilizing different fragment-based or machine learning techniques, leading to varying results. Finally, it is evident that not every platform computes all parameters. Thus, we decided that it would be more suitable to average scores in order to encompass all results.
Accordingly, a qualitative assessment of the ADMET descriptors was performed (Table 4 and Table 5), followed by an overall ADMET evaluation and compounds classification (Supplementary Materials, Tables S3–S5). To enable a comparison among the ADMET descriptors taken from different platforms, we converted all measurements into qualitative estimates according to the explanatory theory underlying each platform. The final assessment is based on the majority principle. In the event of a tie, results from the AdmetLab 3.0, Deep-PK, and admetSAR 3.0 platforms will be considered since they have updated their algorithms more recently.
The classification of P-gp (P-glycoprotein) as a parameter describing Bioavailability or Distribution is still a subject of debate and since, the majority of the platforms listed it under Bioavailability, we followed this convention as well. Furthermore, we did not assess BBB (blood-brain barrier) penetration. Although we gathered the data, we were not focused on the compounds’ biological target specifications at this point.
Considering the toxicological characteristics, we selected the parameters for which it was possible to find a greater amount of experimental data related to approved drugs in order to validate our approach.

2.3. Method Validation

To confirm the reliability of the established screening method, we adhered to the protocols used for known tyrosine kinase inhibitor drugs [52], and the compiled findings are presented below (Table 6, Table 7, Table 8, Table 9 and Table 10). Overall, the FDA-approved drugs are assessed, focusing on compounds that attain a score exceeding 50% of the maximum, which seems to be safe.
The last acceptable compound is Mobocertinib, with a score of 3.674. It is evident that some FDA-approved drugs do not meet fundamental Druglikeness criteria, highlighting that establishing our threshold at 50% of the peak value leads us toward more secure outcomes.
In order to meet the criteria for Medicinal Chemistry (the last acceptable drug is Vandetanib), it is essential to exclude undesirable functionalities (such as chemical groups that are recognized as toxic, unstable, or causing false-positive results in biochemical tests), to possess lead-like characteristics, and to maintain a level of simplicity.
For the final determination of Bioavailability, the individual contributions of Caco-2 Permeability, Human Intestinal Absorption, MDCK Permeability, and Pgp-substrate/inhibitor were considered. P-glycoprotein (P-gp), a drug efflux pump, affects the bioavailability of therapeutic drugs and plays a potentially important role in clinical drug–drug interactions. The classification of candidate drugs as substrates or inhibitors of carrier proteins is of crucial importance in drug development. However, regarding the bioavailability of each compound by itself, the best combination is neither an inhibitor nor a substrate, while the worst is a substrate and not an inhibitor. The outcome is under investigation in any intermediate situation. Compounds above Tucatinib were considered to have a high bioavailability.
The low binding of the compound to plasma proteins is an advantageous characteristic allowing quicker results and effectiveness, since only the unbound form of the drug is active pharmacologically. Enhanced tissue penetration is achieved, along with a reduced risk of interactions with other drugs that are highly protein-bound, which may displace it and elevate free drug levels, leading to increased toxicity. However, when clearance is compromised (in cases of kidney or liver disease), the risk of toxicity is enhanced.
For cytotoxic drugs (chemotherapy agents), in terms of the clearance parameter, a high excretion rate is generally preferable to limit toxicity. It should be noted that the ideal balance is affected by various factors, so we cannot determine an optimal value for total clearance.
Upadacitinib is the last compound approved from our method, with a scoring index of 3. Herein, it is important to clarify that our current study does not aim to link scaffolds to score outcomes or suggest structural changes. We focus solely on presenting a new consensus-based screening approach for assessing Druglikeness and ADMET properties, highlighting the most promising compounds.
The in silico derived results of the molecular descriptors were then compared with the experimental data using a simple linear regression model that estimates the relationship between an independent (experimental) and a dependent (predicted) variable using a straight line. Root Mean Squared Error (RMSE) and Mean Absolute Error (MAE) are metrics used to evaluate a regression model. These metrics tell us how accurate our predictions are and what the amount of deviation from experimental values is. Errors are the differences between the predicted and the experimental values of a variable. There is a third metric—R-Squared score, usually used for regression models. This measures the amount of variation that can be explained by our model, i.e., the percentage of correct predictions returned by our model.
R M S E = 1 N i = 1 N y i y i ^ 2
M A E = y i y i ^ N
R 2 = 1 i = 1 N ( y i y i ) ^ 2 i = 1 N ( y i y ¯ ) 2
The simplest possible “model” is to always predict the mean of yi ( y ¯ ) for all inputs. —The baseline represents the worst acceptable performance. Beating it confirms your model adds value. The RMSE of this baseline is:
R M S E b a s e l i n e =   1 N i = 1 N y i y ¯ 2
where N is the number of samples, y i ^ and yi are the predicted and experimental values of the ith sample in the dataset; y ¯ is the mean value of all the experimental values.
Furthermore, the descriptive values were assessed against the experimental findings, leading to models that were evaluated based on their sensitivity (SE), specificity (SP), and accuracy (ACC).
S E = T P T P + F N
S P = T N T N + F P
A C C = T P + T N T P + T N + F P + F N
where N is the number of samples, and TP, FP, TN, and FN represent true positive, false positive, true negative, and false negative, respectively.

2.3.1. Validation Results of Consensus-Based Model

Initially, experimental data for FDA-approved drugs were collected [53,54,55,56,57] to study the correlation degree with the corresponding in silico data. Data from DrugBank [56] were also utilized (Table 11) in cases where experimental information was lacking. The regression analysis results are presented in Table 12.
In each case (MW, TPSA, and MR), Pearson correlation coefficient (r) values of 1.000, 0.995, and 0.950, were determined, respectively. These figures indicate a robust correlation with the values calculated from DrugBank. Additionally, the error variation (mean values derived from RMSE and MAE) is approximately 0.2 units for MW, 2.9 units for TPSA, and 6.4 units for MR, which are relatively small when compared to the means of the samples, also highlighting a strong relationship between the calculated values from DrugBank and the consensus-predicted figures. Comparing RMSE to RMSEbaseline, we found that the performance of our models shows improvements of 99.68%, 85.80%, and 66.93%, respectively, over the baseline, indicating a reduction in errors.
In the case of the LogPo/w parameter (Table 13), however, we observed that r < 0.700 (R = 0.645); thus, we decided to use an alternative approach. The measurement averaging from Molinspiration–Molsoft platforms showed the best coefficient, r = 0.750, compared to any other case.
The error variation is about 1.00 units for the initial case and 0.89 units for the second, indicating a slight enhancement, though it remains relatively high when compared to the sample means. Nevertheless, our final model offers a statistically significant improvement over the baseline, achieving a 32.6% reduction in errors, unlike the 20.2% reduction given in the first model. As stated previously, estimating LogPo/w is a challenging task for various reasons; primarily, LogPo/w is influenced by pH (particularly for ionizable compounds), and most predictive models still assume that the compounds are neutral. In addition, conformational flexibility and other factors contribute to increased variability in experimental LogPo/w measurements. Considering all these factors, our consensus model is currently accepted until a more effective solution is developed.
We did not consider the R2 parameter at all, because it indicates the percentage of correct predictions returned by the regression equation; however, this is not relevant to us since our primary interest is related to measuring the evaluation parameters between the predicted values ( y i ^ ) and the “real” values (yi).
We confirmed the validity of MW, LogP, TPSA, and MR, which are affected by biological variability. Furthermore, we gathered information on nHA, nHD, nRing, and nRigidB, which are 2D descriptors; however, these parameters did not show any considerable variation across platforms. Thus, it seemed less important to be compared to “real” data.
Moving to ADMET descriptors’ validation of reliability (Table 14), we have drawn the following graphs to test the ability of our method in order to detect experimental values (Figure 2).
Applying Equations (5)–(7) to each parameter, we found that the bioavailability model presents sensitivity (SE) = 0.714, specificity (SP) = 0.600, and accuracy (ACC) = 0.674, and the PPB model exhibits SE = 0.700, SP = 0.909, and ACC = 0.738, confirming both as suitable predictive models. However, the Clearance parameter is significantly underestimating true positive results with a SE = 0.0714, although it presents acceptable SP = 0.600 and ACC = 0.674. Regarding Toxicity, all individual parameters qualify as reliable the models, with the Ames test showing SE = 1.00, SP = 0.604, = ACC of 0.611; Carcinogenicity displaying SE = 0.625, SP = 0.682, and ACC = 0.667; hERG Blockers presenting SE = 0.833, SP = 0.714, and ACC = 0.769; and Hepatotoxicity yielding SE = 0.500, SP = 0.625, and ACC = 0.550. Considering that any value above 0.500 is deemed satisfactory for the reliability of our consensus model, only the Clearance parameter is excluded and thus cannot be incorporated into our screening method. Clearance, which pertains to excretion, is a vital pharmacokinetic parameter for assessing the behavior of drugs within the body; however, it is not essential for defining the pharmacosimilarity of a promising compound. Consequently, the success of the Bioavailability, Distribution, and Toxicity models mitigates the setback of the Excretion deficiency.

2.3.2. Validation Results of Individual Platforms

The same approach was primarily utilized for four distinct platforms, namely AdmetLab 3.0, pkSCM, Deep-PK, and admetSAR 3.0, as they compute most of the requested parameters (Table 1). In the physicochemical predictions, all platforms performed well with the exception of Log Po/w, which was evaluated through all platforms; each one failed to be compared to our consensus-based model, with Pearson correlation coefficients ranging from 0.314 (AdmetLab) to 0.734 (Molsoft) (Supplementary Materials, Figure S1). Regarding ADMET descriptors, our model outperformed each individual platform in terms of Bioavailability, Ames test, and Carcinogenicity, achieving similar results to Deep-PK in Hepatotoxicity. AdmetLab demonstrated marginally better performance in Distribution (SE = 0.880, SP = 0.818, ACC = 0.869) and hERG Blockers (SE = 1.000, SP = 0.857, ACC = 0.923) but did not exceed the threshold of 0.500 for any other parameter. No individual platform or consensus estimation produced satisfactory results in plasma clearance. This could be related to inadequate training of the algorithms used in the platforms. Thus, we decided to not consider our screening method on excretion parameters (Supplementary Materials, Figure S2).

2.4. Evaluation of Druglikeness, Medicinal Chemistry, and ADMET Properties

In Table 15 are the compounds that were excluded from the validated screening method following the defined criteria: Druglikeness, Medicinal Chemistry, Bioavailability (ADME), Distribution (ADME), and Overall Toxicity (Toxicity). As mentioned before, for each individual parameter, the acceptance threshold was set at >50% of the maximum score, whereas for the Distribution, only compounds with low plasma protein binding were selected.
A total of 24 compounds exceeded the Druglikeness threshold of 3.501, while 16 were rated above the acceptable medicinal chemistry score of 2. Regarding Bioavailability, 8 out of 29 examined compounds did not achieve a value greater than 2.51. In terms of Distribution, only compounds with low PPB were evaluated, and when it comes to Overall Toxicity, nine compounds achieved the minimum acceptable score of 2.01 (Supplementary Materials, Tables S1–S5).
After the excluded compounds were presented, they were arranged based on the number of hits in the table. Dark green was assigned for 5/5 hits, light green for 4/5, yellow for 3/5, red for 2/5, and white for 1/5.
Compounds that demonstrate greater than three out of five hits (≥60%) were considered appropriate (Table 16).
It is evident that Distribution is the most frequently violated property. Only two compounds among our compounds’ pool were identified to have low PPB. However, as illustrated in Table 9, a significant portion of FDA-approved drugs are actually found experimentally to exhibit high PPB; therefore, low PPB does not exclude a compound from being classified as Druglike. Additionally, Overall Toxicity is listed as the second most frequently violated property, attributed to the mechanism of action of TKIs, since they inhibit cell growth and division.
Following the application of our approach to FDA-approved drugs, 39 of the 63 compounds met the necessary criteria and were subsequently utilized in the ensuing statistical analyses.

2.5. Molecular Docking Studies

The compounds that were highlighted during the screening phase proceeded to molecular docking studies, initially focusing solely on their primary biological targets, already reported in the literature [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. Consequently, the studied biological targets include VEGFR-2, RET, MET, EGFR-HER-1, HER-2, and BTK. It is important to mention that TKI.4 was identified as a strong and highly selective small molecule inhibitor of c-MET, known as Tepotinib, which has been authorized for the treatment of advanced lung cancer in patients with specific genetic mutations. This also indicates the reliability of the computational methods and techniques used in the study. The remaining compounds that retained their initial biological focus consist of TKI.2a, TKI.2b, TKI.19 (biological target identified: VEGFR-2), TKI.6 (biological target identified: HER-2), and TKI.21b (biological target identified: EGFR). Molecular docking studies were performed and the binding efficiency of our compounds was evaluated by (i) measuring their binding affinity (kcal/mol), (ii) calculating the possibility that the pose displays a minimal Root Mean Square Deviation (RMSD) to the binding pose (CNN pose score), (iii) assessing their affinity to the biological target as determined by the CNN (CNN affinity), and, naturally, (iv) examining the interactions present in docking complexes.
The chosen X-ray crystal structure of VEGFR-2 (PDB ID: 4ASE—Tivozanib) [58] displays a ‘DFG-out’ (inactive) conformation, indicating that the kinase is primarily inactive, as the DFG (Asp-Phe-Gly) residues are oriented in a manner preventing ATP binding and obstructing the substrate binding site. The active site of VEGFR-2 is divided into four key regions: the hydrophobic regions (HYD-I and HYD-II), a hinge region, and the DFG motif region. The HYD-I region serves as the active site where ATP and type I inhibitors bind selectively, while the HYD-II region, known as the ‘Phe pocket’ or ‘allosteric site,’ is where most type II inhibitors bind specifically. Consequently, Tivozanib, as a type II inhibitor, interacts specifically with the ‘DFG-out’ conformation, thus cannot bind to the ATP binding pocket and instead binds to the receptor’s adjacent hydrophobic site. Among type I (first-generation) and type II (second-generation) inhibitors, type II inhibitors associated with the ‘DFG-out’ conformation have shown advantages regarding selectivity and off-target activity (side effects) [59]. Regarding the docking results, all three compounds displayed binding affinities and CNN pose scores ranging from −10.33 to −11.94 kcal/mol and 0.855 to 0.869, respectively, which are comparable to the co-crystallized ligand, Tivozanib, with a binding affinity of −10.87 kcal/mol and a CNN pose score of 0.925. For CNN affinity, TKI.2a (8.068) and TKI.2b (8.030) reported values that are similar to Tivozanib (8.124), except for TKI.19 (7.055). The ligand Tivozanib, when redocked in its co-crystal form, established two hydrogen bonds: one with the hinge region at Cys919 and the other with Asp1046 in the DFG domain. Hydrophobic interactions were noted in the HYD-I region with Leu840 and Phe918, as well as in the more selective HYD-II region involving Ile888, Leu889, Val899, and the gatekeeper residue Val916. Additionally, all three studied compounds exhibited H-bond interactions with Cys919; particularly, TKI.2a and TKI.2b formed two hydrogen bonds, critical for the molecule’s inhibitory activity. Regarding the DFG motif region, TKI.2a and TKI.2b showed hydrogen bonding with the amino acid Asp1046 through the urea moiety, similar to Tivozanib, while TKI.19 interacted with Glu885, another constituent of the DFG domain, through its carboxamide group. This variation may be a reason for TKI.19’s poorer performance in the CNN affinity score. Common hydrophobic interactions were observed in the HYD-I region among all compounds (Leu840, Val848), while slight variations were present in the HYD-II region between TKI.2a, TKI.2b (Leu889, Val899, Phe1047), and TKI.19 (Leu889, Val898, Ile1044). Furthermore, TKI.19 exhibited a π-π interaction with Phe1047 (Figure 3).
The HER kinase family, also known as the human epidermal growth factor receptor (HER) or epidermal growth factor receptor (EGFR), comprises four members: EGFR (HER1 or ErbB-1), HER2 (ErbB-2 or neu), HER3 (ErbB-3), and HER4 (ErbB-4). These are multidomain proteins that include an extracellular domain for ligand binding, a single transmembrane domain, and an intracellular domain with tyrosine kinase activity. In normal tissues, ERBB signaling begins when ligands bind to the extracellular domains of EGFR, HER3, or HER4, leading to either homo- or heterodimerization [60].
Unlike the other members, HER2 is not activated by ligands; instead, it serves as the preferred dimerization partner for the other ERBB family members. The selected X-ray crystal structure of HER2 (PDB ID: 7PCD—covalent inhibitor) [60] exhibits the characteristic bilobed folding of kinases. The two lobes are linked by a flexible hinge region and divided by a deep cleft that contains the ATP binding site [61]. Docking studies of TKI.6 revealed that a significant hydrogen bond formed at the hinge region with Met801, similarly to the co-crystalized inhibitor. Furthermore, while the integrated ligand showed a covalent bond with cysteine at position 805, our compound was permanently linked to Asp863 in the DFG motif domain. Comparable binding affinities were observed between the covalent inhibitor (binding affinity: −9.66 kcal/mol, CNN affinity: 7.734) and TKI.6 (binding affinity: −8.96 kcal/mol, CNN affinity: 7.684), emphasizing that the formation of a covalent bond between the inhibitor and the protein enhances binding affinity and potency. The variance in CNN pose scores between the co-crystallized inhibitor (0.814) and TKI.6 (0.920) could be attributed to the differing binding residues in the ATP binding site, indicating that the compound we studied may have a higher likelihood of adopting a favorable pose. Additionally, hydrophobic interactions with the glycine-rich nucleotide phosphate-binding loop (Leu726, Val734) contribute to the stability of the complex. Finally, although the serine located at position 783 is considered an important selectivity-determining amino acid between HER2 and EGFR activity, no interactions were observed (Figure 4).
As our molecular docking research focused on another member of the HER kinase family, namely EGFR, we utilized the surrogate crystal structure of the wild-type EGFR complexed with Mobocertinib (PDB ID: 7T4I) [62]. This research highlights important residues, including Met793 found in the hinge area, as well as specific regions like the selectivity pocket where Thr790 functions as the “gatekeeper” for ATP binding, Lys745 serves as the catalytic lysine, and Thr854 is situated in the DFG triad, along with two separate hydrophobic regions. The hydrophobic region I consists of amino acids such as Phe723, Leu747, Ile759, Met766, Leu777, and Leu788, while hydrophobic region II, located near Thr790 and comprising Leu718, Gly719, Val726, and Leu844, plays a crucial role in the binding of compounds to EGFR. Lastly, Cys797, positioned at the edge of the active site cleft and being the most solvent-exposed cysteine in the EGFR kinase domain, is responsible for forming covalent bonds with irreversible TKIs [36,63]. Both the co-crystalized ligand and TKI.21b formed hydrogen bonds with the hinge region (Met793) and the DFG motif (Thr854). However, Mobocertinib demonstrated better positioning in the active site compared to TKI.21b due to an additional hydrogen bond with Met793, another hydrogen bond with the “gatekeeper” Thr790, and a covalent bond with Cys797, which accounts for the disparity in their CNN pose scores of 0.970 and 0.860, respectively. Conversely, TKI.21b established hydrogen bonds with catalytic residue Lys745 and Asp855, along with significant hydrophobic interactions involving Leu718, Phe723, Val726, and Leu844, leading to comparable binding affinities with Mobocertinib (TKI.21b binding affinity: −8.25 kcal/mol, CNN affinity: 7.493; Mobocertinib binding affinity: −7.66 kcal/mol, CNN affinity: 8.106) (Figure 5).

Molecular Docking Validation Results

Common methods for assessing the accuracy of docking protocols include self-docking, cross-docking, and ligand enrichment. Self-docking is a highly employed technique for the preliminary evaluation of a docking program’s accuracy. As a validation technique, it aims to reproduce the original orientation of the co-crystallized ligand, whereas cross-docking assesses how well a particular receptor positions chemically diverse groups of ligands while maintaining acceptable RMSD values [64]. In the process of enriching the database, decoys are introduced among a group of active inhibitors, and the docking software is evaluated for its effectiveness in ranking the active substances. The decoy molecules mimic the active compounds by sharing similar physical characteristics; however, they must have no binding affinity for the receptor.
Docking setup was first validated by re-docking of the co-crystallized ligand in the vicinity of the binding site of the enzyme, followed by calculating the Root Mean Square Deviation (RMSD) between the final configuration and the initial coordinates. RMSD values below 2.0 Å signify consistent results, values between 2.0 Å and 3.0 Å indicate a shift from the reference position while keeping the desired orientation, and RMSD values exceeding 3.0 Å are entirely inaccurate [65]. Regarding our molecular docking investigations, we achieved reliable RMSD values for VEGFR-2 of 1.144 Å, for HER2 of 1.121 Å, and for EGFR of 1.430 Å, all remaining below 1.5 Å.
To further assess the docking protocols, cross-docking procedures were implemented. In the cross-docking analysis, each known FDA-approved ligand of the specified biological targets was docked into the receptors mentioned (PDB IDs: 4ASE, 7PCD, and 7T4I). According to Table 17, a significant majority (11 out of 18) of the known drugs achieved measurements below 2.5 Å, with 4 of them falling between 2.5 and 3 Å, confirming the reliability of our docking protocols. The only exceptions were Axitinib, Pazopanib, and Sunitinib in their molecular studies on VEGFR-2, which exhibited measurements greater than 3 Å. Notably, these three compounds share a common characteristic of demonstrating low affinity, CNN pose score, and CNN affinity values collectively.
The enrichment factor (EF) serves as a measure of the docking program’s reliability. The objective was to evaluate the ability of the receptor to differentiate between inactive substances and known active compounds by determining enrichment values. The enrichment factor was computed using the formula below [66]:
E F X % = a c t i v e s x % d a t a s e t x % a c t i v e s t o t a l d a t a s e t t o t a l
where activesx% refers to the active compounds present in the selected dataset (datasetx%), while datasettotal encompasses all compounds within that dataset, and activestotal indicates the number of active molecules included among the decoys. We defined x% as 10%, which means we aimed to determine how many active compounds exist within the top 10% of our ranked dataset. An enrichment factor (EF) exceeding 1 demonstrates that the approach is more efficient than random selection, with higher values indicating improved performance. For instance, an EF of 5 in the top 10% of the dataset implies that there are five times more active compounds present in that top 10% of the evaluated set than one would anticipate by random chance. Herein, we employed a HER2 protein (PDB ID: 3PP0) as the receptor, utilizing a dataset of 332 compounds that included 30 active compounds among 302 inactive ones. As a result, when ranking compounds based on their CNN affinity values, the enrichment factor at 10% (EF(10%)) was found to be 4.360, identifying 13 active compounds in the top 33 structures (which represents 10%) (Supplementary Materials, Table S6), while ranking based on the CNN pose score yielded an EF(10%) of 6.036, highlighting 18 actives within the top 33 structures (Supplementary Materials, Table S7). Moreover, the Receiver Operating Characteristic (ROC) Curve and the Area Under the Curve (AUC) provide valuable insights into the model’s capacity to differentiate between active and inactive compounds across various threshold settings, with the AUC serving as a summary of the model’s overall performance, as illustrated in Figure 6. It is evident that the ranking based on the CNN pose score exhibited a higher ROC-AUC compared to the CNN affinity ranking, with values of 0.930 and 0.845, respectively, indicating that utilizing the CNN pose score for ranking is a more dependable approach.
Reviewing all the docking validation parameters presented, it is confirmed that the used docking software and protocols reliably produce consistent and trustworthy results.

2.6. Statistical Results

A confidence interval refers to the probability that a population parameter will be found between a set of values for a certain proportion of times. Statisticians use confidence intervals to measure the uncertainty in an estimate of a population parameter based on a sample. Therefore, by calculating the confidence limits (a = 0.05) of the molecular descriptors for the distinguished FDA-approved drugs, we estimated the range within the true population mean (the true mean of all active tyrosine kinase inhibitors), is likely to lie.
Data obtained from the independent T-tests that we performed for each molecular descriptor of the two samples (studied compounds and FDA-approved drugs) (Table 18) (Supplementary Materials, Table S8) confirmed that mean values of the molecular descriptors of the studied compounds fall within the confidence limits of the corresponding molecular descriptors of FDA-approved drugs, with the exception of the mean value of the LogPo/w parameter.
Alongside analyzing the average values of the two datasets, we conducted a Kolmogorov–Smirnov (KS) test to evaluate the distributions of the datasets (Table 19) (Supplementary Materials, Figure S3).
Therefore, the above data suggests that we cannot reject the null hypothesis, e.g., that the molecular descriptors of the two different samples come from the same population (p > 0.05), except for the LogPo/w parameter (p < 0.05). One possible reason for the deviation in the LogPo/w value could be that FDA-approved drugs have undergone lead optimization to achieve a balanced LogPo/w for ADME, while our compounds may still be in an earlier development stage where LogPo/w has not been refined.
These findings are important because, from different samples, we can use inferential statistics to draw some conclusions about the molecular descriptors of the general population, and we will be able to more accurately approximate the molecules that are worth investigating as tyrosine kinase inhibitors.

3. Materials and Methods

In this study, we evaluated the Druglikeness and ADMET characteristics of various TKIs sourced from the literature [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51], aiming to identify the most promising and effective tyrosine kinase inhibitors with acceptable pharmacosimilarity, favorable pharmacokinetics, and minimal toxicity. To address this, we created a new consensus-based screening approach that leverages 10 well-regarded free chemoinformatics platforms, specifically Molinspiration [8], Molsoft [9], SwissADME web tool [10], Mcule [11], AdmetLab 3.0 [12,13], pkSCM [14], Deep-PK [15], admetSAR 3.0 [16,17], PreADMET 2.0 [18], and T.E.S.T 5.1.2 [19]. The forecasted data was compiled in Tables S1–S5 (Supplementary Materials).
Additionally, we conducted a comparative analysis using data from 63 FDA-approved TKIs. Bibliographic references were gathered from R. R. Shah et al., 2013 [53], J. Dulsat et al., 2023 [54], M. Viganò et al., 2023 [55], C. Knox et al., 2024 [56], and the U.S. Department of Health and Human Services Food and Drug Administration [57], and are displayed in Table 11 and Table 14. Nevertheless, for numerous compounds, we could not locate experimental data for the in silico predicted ADMET characteristics, such as skin permeability, skin sensitization, and ecological toxicity, leading us to exclude these properties from our screening method.
In molecular docking studies, the X-ray crystal structures were retrieved from the Protein Data Bank on the Research Collaboration for Structural Bioinformatics (RCSB) website www.rcsb.org (last accessed on 2 July 2025). The preparation of proteins was performed using OpenMM 8 [67]—energy minimizations were executed with the AMBER 14 or Charm36 force fields—while GypSUm-DL [68] was used to generate and minimize ligand 3D coordinates. Docking was conducted using GNINA 1.0 [69], a molecular docking tool that incorporates convolutional neural networks (CNNs) for scoring and optimizing ligands. The input files for docking were visualized using PyMOL 3.0.4 [70] and Schrödinger Maestro 14.5.131 [71]. For the validation of protocols related to molecular docking studies (including re-docking and cross-docking), we utilized suitable Python 3.2.2 scripts to identify matching atoms between the docked ligands and the co-crystallized ligands and finally compute the RMSD values. In terms of verifying the docking software, the ultimate goal involved using active structures and decoys sourced from a publicly accessible repository (InformaticsMatters, 2021) [72]. Additionally, ROC-AUC curves were created using appropriate Python scripts.
The in silico data of the FDA-approved drugs obtained from the studies were statistically analyzed using IBM SPSS Statistics (Version 29) [73]. Thus, for each physicochemical property, the confidence intervals for a 95% confidence level (i.e., corresponding significance level of 0.05 or 5%) were calculated. Finally, the two samples (39 FDA-approved drugs and 29 studied compounds) were subjected to a T-test. This analysis compares the average values of two datasets, in relation to a non-parametric Kolmogorov–Smirnov (KS) Test, which investigates if two datasets are taken from the same distribution, in order to determine if they came from the same population. The comparative statistical data along with the data tables can be found in Table 18, Table 19 and Table S6.

4. Conclusions

The novelty of the present screening method is that for the evaluation of the compounds for Druglikeness and ADMET properties, the data from the different platforms were used as a whole, rather than the results of each platform individually. After all, according to Stephen Hawking, ‘Science is beautiful when it makes simple explanations of phenomena or connections between different observations. Examples include the double helix in biology and the fundamental equations of physics.’ The reliability validation of the new method showed that it could approximate experimental values, in contrast to individual platforms validation results, unlike the validation results derived from the individual platforms.
Out of the 29 compounds listed in Table 2 from the literature, 14 compounds shown in Table 16 satisfied more than 60% of the criteria we established: Druglikeness, Medicinal Chemistry rules, Bioavailability, Distribution, and Overall Toxicity. Notably, TKI.4, TKI.16, TKI.19, TKI.21b, AIK.1, and DDK.8 exhibited the most favorable profiles. In our initial molecular docking studies, we discovered that TKI.4 is the approved drug Tepotinib, which enhances the credibility of our consensus-based method as a tool for identifying drug-like substances. Among all the other compounds, only TKI.2a, TKI.2b, TKI.19 (VEGFR-2), TKI.6 (HER2), and TKI.21b (EGFR) successfully confirmed their original biological target designation, as indicated by our newly validated molecular docking protocols that employ deep learning models for evaluation and ranking.
In addition, using inferential statistics, we demonstrated that we are 95% confident that the mean values of the molecular descriptors, for the set of active small molecules potentially acting as tyrosine kinase inhibitors, are within the confidence limits calculated from the FDA-approved drugs that picked/selected out from our screening method. These limits were defined as follows: MW [416.81, 461.47], TPSA [83.48, 95.83], MR [115.14, 128.65], LogPo/w [2.62, 3.49], nRB [5, 7], nHA [7, 8], nHD [2, 2], nRings [4, 5], nRigidB [23, 26], and nAtoms [52, 58].
At this point in our investigation, our main objective was to present a new screening method rather than to analyze the results for their possible biological significance. This will be addressed in our future work, utilizing suitable chemoinformatics tools.
Noting that the Druglikeness definition holds in the absence of any obvious structural similarity to an approved drug [7], we will aim in the future to uncover structural similarities among the selected compounds we studied and FDA-approved tyrosine kinase inhibitors by conducting molecular similarity studies. This will serve both as a manifestation of Druglikeness and as a tool to discover additional biological targets [74]. Furthermore, to assess the biological significance of proposed or newly identified targets, we will perform molecular docking studies and pharmacophore modeling, with the ultimate objective of suggesting structural modifications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms262010207/s1.

Author Contributions

Conceptualization, E.M. and D.H.-L.; methodology, E.M. and D.H.-L.; software, E.M.; validation, E.M.; formal analysis, E.M.; investigation, E.M.; resources, E.M.; data curation, E.M.; writing—original draft preparation, E.M.; writing—review and editing, D.H.-L.; visualization, E.M.; supervision, D.H.-L.; project administration, D.H.-L. 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

The data are available by the authors and through literature.

Acknowledgments

The authors are grateful to Pontiki and Patsilinakos for their helpful suggestions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cheek, S.; Zhang, H.; Grishin, N.V. Sequence and structure classification of kinases. J. Mol. Biol. 2002, 320, 855–881. [Google Scholar] [CrossRef]
  2. Ferguson, F.M.; Gray, N.S. Kinase inhibitors: The road ahead. Nat. Rev. Drug Discov. 2018, 17, 353–377. [Google Scholar] [CrossRef] [PubMed]
  3. Arora, A.; Scholar, E.M. Role of tyrosine kinase inhibitors in cancer therapy. J. Pharmacol. Exp. Ther. 2005, 315, 971–979. [Google Scholar] [CrossRef] [PubMed]
  4. Huang, L.; Jiang, S.; Shi, Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). J. Hematol. Oncol. 2020, 13, 1–23. [Google Scholar] [CrossRef] [PubMed]
  5. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of drug absorption using multivariate statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef]
  6. Sun, D.; Gao, W.; Hu, H.; Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sin. B 2022, 12, 3049–3062. [Google Scholar] [CrossRef]
  7. Bickerton, G.R.; Paolini, G.V.; Besnard, J.; Muresan, S.; Hopkins, A.L. Quantifying the chemical beauty of drugs. Nat. Chem. 2012, 4, 90–98. [Google Scholar] [CrossRef]
  8. Available online: https://www.molinspiration.com/ (accessed on 24 January 2025).
  9. Abagyan, R.; Totrov, M.; Kuznetsov, D. ICM—A New Method for Protein Modeling and Design: Applications to Docking and Structure Prediction from the Distorted Native Conformation. J. Comput. Chem. 1994, 15, 488–506. [Google Scholar] [CrossRef]
  10. 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. [Google Scholar] [CrossRef]
  11. Kiss, R.; Sandor, M.; Szalai, F.A. http://Mcule.com: A public web service for drug discovery. J. Cheminform. 2012, 4, P17. [Google Scholar] [CrossRef]
  12. Dong, J.; Wang, N.-N.; Yao, Z.-J.; Zhang, L.; Cheng, Y.; Ouyang, D.; Lu, A.-P.; Cao, D.-S. ADMETlab: A platform for systematic ADMET evaluation based on a comprehensively collected ADMET database. J. Cheminform. 2018, 10, 29. [Google Scholar] [CrossRef]
  13. Xiong, G.; Wu, Z.; Yi, J.; Fu, L.; Yang, Z.; Hsieh, C.; Yin, M.; Zeng, X.; Wu, C.; Lu, A.; et al. ADMETlab 2.0: An integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res. 2021, 49, W5–W14. [Google Scholar] [CrossRef] [PubMed]
  14. Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J. Med. Chem. 2015, 58, 4066–4072. [Google Scholar] [CrossRef] [PubMed]
  15. Available online: https://biosig.lab.uq.edu.au/deeppk/ (accessed on 24 January 2025).
  16. Yang, H.; Lou, C.; Sun, L.; Li, J.; Cai, Y.; Wang, Z.; Li, W.; Liu, G.; Tang, Y. admetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics 2019, 35, 1067–1069. [Google Scholar] [CrossRef]
  17. Cheng, F.; Li, W.; Zhou, Y.; Shen, J.; Wu, Z.; Liu, G.; Lee, P.W.; Tang, Y. AdmetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. J. Chem. Inf. Model. 2012, 52, 3099–3105, Erratum in J. Chem. Inf. Model. 2019, 59, 4959. [Google Scholar] [CrossRef] [PubMed]
  18. Lee, S.K.; Chang, G.S.; Lee, I.H.; Chung, J.E.; Sung, K.Y.; No, K.T. The PreADME: PC-based program for batch prediction of ADME properties. EuroQSAR 2004, 9, 5–10. [Google Scholar]
  19. Martin, T.M. Toxicity Estimation Software Tool; Environmental Protection Agency: Washington, DC, USA, 2020.
  20. Lipinski, C.A.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  21. Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef]
  22. Veber, D.F.; Johnson, S.R.; Cheng, H.-Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
  23. Muegge, I.; Heald, S.L.; Brittelli, D. Simple selection criteria for drug-like chemical matter. J. Med. Chem. 2001, 44, 1841–1846. [Google Scholar] [CrossRef]
  24. Oprea, T.I. Property distribution of drug-related chemical databases. J. Comput.-Aided Mol. Des. 2000, 14, 251–264. [Google Scholar] [CrossRef]
  25. Teague, S.J.; Davis, A.M.; Leeson, P.D.; Oprea, T. The design of leadlike combinatorial libraries. Angew. Chem.—Int. Ed. 1999, 38, 3743–3748. [Google Scholar] [CrossRef]
  26. Gleeson, M.P. Generation of a set of simple, interpretable ADMET rules of thumb. J. Med. Chem. 2008, 51, 817–834. [Google Scholar] [CrossRef]
  27. Baell, J.B.; Nissink, J.W.M. Seven Year Itch: Pan-Assay Interference Compounds (PAINS) in 2017—Utility and Limitations. ACS Chem. Biol. 2018, 13, 36–44. [Google Scholar] [CrossRef]
  28. Brenk, R.; Schipani, A.; James, D.; Krasowski, A.; Gilbert, I.H.; Frearson, J.; Wyatt, P.G. Lessons learnt from assembling screening libraries for drug discovery for neglected diseases. ChemMedChem 2008, 3, 435–444. [Google Scholar] [CrossRef] [PubMed]
  29. Newton, R.; Bowler, K.A.; Burns, E.M.; Chapman, P.J.; Fairweather, E.E.; Fritzl, S.J.; Goldberg, K.M.; Hamilton, N.M.; Holt, S.V.; Hopkins, G.V.; et al. The discovery of 2-substituted phenol quinazolines as potent RET kinase inhibitors with improved KDR selectivity. Eur. J. Med. Chem. 2016, 112, 20–32. [Google Scholar] [CrossRef] [PubMed]
  30. Adel, M.; Serya, R.A.; Lasheen, D.S.; Abouzid, K.A. Identification of new pyrrolo [2,3-d]pyrimidines as potent VEGFR-2 tyrosine kinase inhibitors: Design, synthesis, biological evaluation and molecular modeling. Bioorg. Chem. 2018, 81, 612–629. [Google Scholar] [CrossRef] [PubMed]
  31. El-Metwally, S.A.; Abou-El-Regal, M.M.; Eissa, I.H.; Mehany, A.B.; Mahdy, H.A.; Elkady, H.; Elwan, A.; Elkaeed, E.B. Discovery of thieno [2,3-d]pyrimidine-based derivatives as potent VEGFR-2 kinase inhibitors and anti-cancer agents. Bioorg. Chem. 2021, 112, 104947. [Google Scholar] [CrossRef]
  32. Dorsch, D.; Schadt, O.; Stieber, F.; Meyring, M.; Grädler, U.; Bladt, F.; Friese-Hamim, M.; Knühl, C.; Pehl, U.; Blaukat, A. Identification and optimization of pyridazinones as potent and selective c-Met kinase inhibitors. Bioorg. Med. Chem. Lett. 2015, 25, 1597–1602. [Google Scholar] [CrossRef]
  33. Mohamady, S.; Galal, M.; Eldehna, W.M.; Gutierrez, D.C.; Ibrahim, H.S.; Elmazar, M.M.; Ali, H.I. Dual Targeting of VEGFR2 and C-Met Kinases via the Design and Synthesis of Substituted 3-(Triazolo-thiadiazin-3-yl)indolin-2-one Derivatives as Angiogenesis Inhibitors. ACS Omega 2020, 5, 18872–18886. [Google Scholar] [CrossRef]
  34. Elmetwally, S.A.; Saied, K.F.; Eissa, I.H.; Elkaeed, E.B. Design, synthesis and anticancer evaluation of thieno [2,3-d]pyrimidine derivatives as dual EGFR/HER2 inhibitors and apoptosis inducers. Bioorg. Chem. 2019, 88, 102944. [Google Scholar] [CrossRef] [PubMed]
  35. Li, X.; Zuo, Y.; Tang, G.; Wang, Y.; Zhou, Y.; Wang, X.; Guo, T.; Xia, M.; Ding, N.; Pan, Z. Discovery of a series of 2,5-diaminopyrimidine covalent irreversible inhibitors of Bruton’s tyrosine kinase with in vivo antitumor activity. J. Med. Chem. 2014, 57, 5112–5128. [Google Scholar] [CrossRef] [PubMed]
  36. Sherbiny, F.F.; Bayoumi, A.H.; El-Morsy, A.M.; Sobhy, M.; Hagras, M. Design, Synthesis, biological Evaluation, and molecular docking studies of novel Pyrazolo [3,4-d]Pyrimidine derivative scaffolds as potent EGFR inhibitors and cell apoptosis inducers. Bioorg. Chem. 2021, 116, 105325. [Google Scholar] [CrossRef]
  37. Zheng, N.; Pan, J.; Hao, Q.; Li, Y.; Zhou, W. Design, synthesis and biological evaluation of novel 3-substituted pyrazolopyrimidine derivatives as potent Bruton’s tyrosine kinase (BTK) inhibitors. Bioorg. Med. Chem. 2018, 26, 2165–2172. [Google Scholar] [CrossRef]
  38. Mahalapbutr, P.; Leechaisit, R.; Thongnum, A.; Todsaporn, D.; Prachayasittikul, V.; Rungrotmongkol, T.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V.; Pingaew, R. Discovery of Anilino-1,4-naphthoquinones as Potent EGFR Tyrosine Kinase Inhibitors: Synthesis, Biological Evaluation, and Comprehensive Molecular Modeling. ACS Omega 2022, 7, 17881–17893. [Google Scholar] [CrossRef]
  39. Ciaffaglione, V.; Consoli, V.; Intagliata, S.; Marrazzo, A.; Romeo, G.; Pittalà, V.; Greish, K.; Vanella, L.; Floresta, G.; Rescifina, A.; et al. Novel Tyrosine Kinase Inhibitors to Target Chronic Myeloid Leukemia. Molecules 2022, 27, 3220. [Google Scholar] [CrossRef]
  40. Lamie, P.F.; El-Kalaawy, A.M.; Latif, N.S.A.; Rashed, L.A.; Philoppes, J.N. Pyrazolo [3,4-d]pyrimidine-based dual EGFR T790M/HER2 inhibitors: Design, synthesis, structure–activity relationship and biological activity as potential antitumor and anticonvulsant agents. Eur. J. Med. Chem. 2021, 214, 113222. [Google Scholar] [CrossRef]
  41. Farghaly, A.M.; AboulWafa, O.M.; Baghdadi, H.H.; El Razik, H.A.A.; Sedra, S.M.; Shamaa, M.M. New thieno [3,2-d]pyrimidine-based derivatives: Design, synthesis and biological evaluation as antiproliferative agents, EGFR and ARO inhibitors inducing apoptosis in breast cancer cells. Bioorg. Chem. 2021, 115, 105208. [Google Scholar] [CrossRef]
  42. El-Adl, K.; Ibrahim, M.; Khedr, F.; Abulkhair, H.S.; Eissa, I.H. N-Substituted-4-phenylphthalazin-1-amine-derived VEGFR-2 inhibitors: Design, synthesis, molecular docking, and anticancer evaluation studies. Arch. Pharm. 2021, 354, e2000219. [Google Scholar] [CrossRef]
  43. El-Helby, A.A.; Ayyad, R.R.A.; Sakr, H.; El-Adl, K.; Ali, M.M.; Khedr, F. Design, Synthesis, Molecular Docking, and Anticancer Activity of Phthalazine Derivatives as VEGFR-2 Inhibitors. Arch. Pharm. 2017, 350, 1700240. [Google Scholar] [CrossRef] [PubMed]
  44. Alanazi, M.M.; Mahdy, H.A.; Alsaif, N.A.; Obaidullah, A.J.; Alkahtani, H.M.; Al-Mehizia, A.A.; Alsubaie, S.M.; Dahab, M.A.; Eissa, I.H. New bis([1,2,4]triazolo)[4,3-a:3′,4′-c]quinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers: Design, synthesis, in silico studies, and anticancer evaluation. Bioorg. Chem. 2021, 112, 104949. [Google Scholar] [CrossRef]
  45. Saleh, N.M.; El-Gaby, M.S.; El-Adl, K.; El-Sattar, N.E.A. Design, green synthesis, molecular docking and anticancer evaluations of diazepam bearing sulfonamide moieties as VEGFR-2 inhibitors. Bioorg. Chem. 2020, 104, 104350. [Google Scholar] [CrossRef]
  46. Ahmed, E.Y.; Latif, N.A.A.; El-Mansy, M.F.; Elserwy, W.S.; Abdelhafez, O.M. VEGFR-2 inhibiting effect and molecular modeling of newly synthesized coumarin derivatives as anti-breast cancer agents. Bioorg. Med. Chem. 2020, 28, 115328. [Google Scholar] [CrossRef] [PubMed]
  47. Abdel-Mohsen, H.T.; El-Meguid, E.A.A.; El Kerdawy, A.M.; Mahmoud, A.E.E.; Ali, M.M. Design, synthesis, and molecular docking of novel 2-arylbenzothiazole multiangiokinase inhibitors targeting breast cancer. Arch. Pharm. 2020, 353, e1900340. [Google Scholar] [CrossRef]
  48. AboulWafa, O.M.; Daabees, H.M.; Badawi, W.A. 2-Anilinopyrimidine derivatives: Design, synthesis, in vitro anti-proliferative activity, EGFR and ARO inhibitory activity, cell cycle analysis and molecular docking study. Bioorg. Chem. 2020, 99, 103798. [Google Scholar] [CrossRef] [PubMed]
  49. Ma, B.; Bohnert, T.; Otipoby, K.L.; Tien, E.; Arefayene, M.; Bai, J.; Bajrami, B.; Bame, E.; Chan, T.R.; Humora, M.; et al. Discovery of BIIB068: A Selective, Potent, Reversible Bruton’s Tyrosine Kinase Inhibitor as an Orally Efficacious Agent for Autoimmune Diseases. J. Med. Chem. 2020, 63, 12526–12541. [Google Scholar] [CrossRef]
  50. Zhavoronkov, A.; Ivanenkov, Y.A.; Aliper, A.; Veselov, M.S.; Aladinskiy, V.A.; Aladinskaya, A.V.; Terentiev, V.A.; Polykovskiy, D.A.; Kuznetsov, M.D.; Asadulaev, A.; et al. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat. Biotechnol. 2019, 37, 1038–1040. [Google Scholar] [CrossRef] [PubMed]
  51. Osborne, J.; Birchall, K.; Tsagris, D.J.; Lewis, S.J.; Smiljanic-Hurley, E.; Taylor, D.L.; Levy, A.; Alessi, D.R.; McIver, E.G. Discovery of potent and selective 5-azaindazole inhibitors of leucine-rich repeat kinase 2 (LRRK2)—Part 1. Bioorg. Med. Chem. Lett. 2019, 29, 668–673. [Google Scholar] [CrossRef]
  52. Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2024 update. Pharmacol. Res. 2024, 200, 107059. [Google Scholar] [CrossRef]
  53. Shah, R.R.; Morganroth, J.; Shah, D.R. Cardiovascular safety of tyrosine kinase inhibitors: With a special focus on cardiac repolarisation (QT Interval). Drug Saf. 2013, 36, 295–316. [Google Scholar] [CrossRef]
  54. Dulsat, J.; López-Nieto, B.; Estrada-Tejedor, R.; Borrell, J.I. Evaluation of Free Online ADMET Tools for Academic or Small Biotech Environments. Molecules 2023, 28, 776. [Google Scholar] [CrossRef]
  55. Viganò, M.; La Milia, M.; Grassini, M.V.; Pugliese, N.; De Giorgio, M.; Fagiuoli, S. Hepatotoxicity of Small Molecule Protein Kinase Inhibitors for Cancer. Cancers 2023, 15, 1766. [Google Scholar] [CrossRef]
  56. Knox, C.; Wilson, M.; Klinger, C.M.; Franklin, M.; Oler, E.; Wilson, A.; Pon, A.; Cox, J.; Chin, N.E.; Strawbridge, S.A.; et al. DrugBank 6.0: The DrugBank Knowledgebase for 2024. Nucleic Acids Res. 2023, 52, D1265–D1275. [Google Scholar] [CrossRef] [PubMed]
  57. U.S. Department of Health and Human Services Food and Drug Administration. Available online: https://open.fda.gov/fdalabels/active_ingredient/ (accessed on 15 September 2024).
  58. Al-Sanea, M.M.; Chilingaryan, G.; Abelyan, N.; Sargsyan, A.; Hovhannisyan, S.; Gasparyan, H.; Gevorgyan, S.; Albogami, S.; Ghoneim, M.M.; Farag, A.K.; et al. Identification of novel potential vegfr-2 inhibitors using a combination of computational methods for drug discovery. Life 2021, 11, 1070. [Google Scholar] [CrossRef] [PubMed]
  59. Modi, S.J.; Kulkarni, V.M. Exploration of structural requirements for the inhibition of VEGFR-2 tyrosine kinase: Binding site analysis of type II, ‘DFG-out’ inhibitors. J. Biomol. Struct. Dyn. 2022, 40, 5712–5727. [Google Scholar] [CrossRef]
  60. Wilding, B.; Scharn, D.; Böse, D.; Baum, A.; Santoro, V.; Chetta, P.; Schnitzer, R.; Botesteanu, D.A.; Reiser, C.; Kornigg, S.; et al. Discovery of potent and selective HER2 inhibitors with efficacy against HER2 exon 20 insertion-driven tumors, which preserve wild-type EGFR signaling. Nat. Cancer 2022, 3, 821–836. [Google Scholar] [CrossRef]
  61. Aertgeerts, K.; Skene, R.; Yano, J.; Sang, B.-C.; Zou, H.; Snell, G.; Jennings, A.; Iwamoto, K.; Habuka, N.; Hirokawa, A.; et al. Structural analysis of the mechanism of inhibition and allosteric activation of the kinase domain of HER2 protein. J. Biol. Chem. 2011, 286, 18756–18765. [Google Scholar] [CrossRef]
  62. Huang, W.-S.; Li, F.; Gong, Y.; Zhang, Y.; Youngsaye, W.; Xu, Y.; Zhu, X.; Greenfield, M.T.; Kohlmann, A.; Taslimi, P.M.; et al. Discovery of mobocertinib, a potent, oral inhibitor of EGFR exon 20 insertion mutations in non–small cell lung cancer. Bioorg. Med. Chem. Lett. 2023, 80, 129084. [Google Scholar] [CrossRef]
  63. Wang, J.; Lam, D.; Yang, J.; Hu, L. Discovery of mobocertinib, a new irreversible tyrosine kinase inhibitor indicated for the treatment of non-small-cell lung cancer harboring EGFR exon 20 insertion mutations. Med. Chem. Res. 2022, 31, 1647–1662. [Google Scholar] [CrossRef]
  64. Mateev, E.; Valkova, I.; Angelov, B.; Georgieva, M. Validation Through Re-Docking, Cross-Docking and Ligand Enrichment in Various Well-Resoluted Mao-B Receptors. Artic. Int. J. Pharm. Sci. Res. 2021, 13, 1000–1007. [Google Scholar] [CrossRef]
  65. Ramírez, D.; Caballero, J. Is It Reliable to Take the Molecular Docking Top Scoring Position as the Best Solution without Considering Available Structural Data? Molecules 2018, 23, 1038. [Google Scholar] [CrossRef]
  66. Maltarollo, V.G. (Ed.) Computer-Aided and Machine Learning-Driven Drug Design. From Theory to Applications; Computer-Aided Drug Discovery and Design 3; Springer: Cham, Switzerland, 2024. [Google Scholar]
  67. Eastman, P.; Galvelis, R.; Peláez, R.P.; Abreu, C.R.A.; Farr, S.E.; Gallicchio, E.; Gorenko, A.; Henry, M.M.; Hu, F.; Huang, J.; et al. OpenMM 8: Molecular Dynamics Simulation with Machine Learning Potentials. J. Phys. Chem. B 2024, 128, 109–116. [Google Scholar] [CrossRef] [PubMed]
  68. Ropp, P.J.; Spiegel, J.O.; Walker, J.L.; Green, H.; Morales, G.A.; Milliken, K.A.; Ringe, J.J.; Durrant, J.D. Gypsum-DL: An open-source program for preparing small-molecule libraries for structure-based virtual screening. J. Cheminform. 2019, 11, 34. [Google Scholar] [CrossRef] [PubMed]
  69. McNutt, A.T.; Francoeur, P.; Aggarwal, R.; Masuda, T.; Meli, R.; Ragoza, M.; Sunseri, J.; Koes, D.R. GNINA 1.0: Molecular docking with deep learning. J. Cheminform. 2021, 13, 43. [Google Scholar] [CrossRef] [PubMed]
  70. The PyMOL Molecular Graphics System, Version 3.0.4; Schrödinger, LLC: New York, NY, USA, 2025.
  71. Schrödinger Release 2025-3: Maestro, Version 14.5.131; Schrödinger, LLC: New York, NY, USA, 2025.
  72. Available online: https://github.com/InformaticsMatters/docking-validation/tree/7ca594d166fcf6f59fd9501ef1e39efa71c4cb60/datasets/DEKOIS_2.0 (accessed on 14 September 2025).
  73. IBM Corp. IBM SPSS Statistics for Windows, Version 29.0.2.0; IBM Corp.: Armonk, NY, USA, 2023.
  74. Martin, Y.C.; Kofron, J.L.; Traphagen, L.M. Do structurally similar molecules have similar biological activity? J. Med. Chem. 2002, 45, 4350–4358. [Google Scholar] [CrossRef]
Figure 1. Flowchart describing the procedure of our research.
Figure 1. Flowchart describing the procedure of our research.
Ijms 26 10207 g001
Figure 2. Bar graphical representation of: (a) Bioavailability; (b) Plasma Protein Binding; (c) Clearance; (d) Ames test; (e) Carcinogenicity; (f) hERG; (g) Hepatotxicity assessments, showing Experimental Total = N, Identified high/positive = TP, Identified low/negative = TN, experimental low/negative − identified low/negative = FP, experimental high/positive − identified high/positive = FN.
Figure 2. Bar graphical representation of: (a) Bioavailability; (b) Plasma Protein Binding; (c) Clearance; (d) Ames test; (e) Carcinogenicity; (f) hERG; (g) Hepatotxicity assessments, showing Experimental Total = N, Identified high/positive = TP, Identified low/negative = TN, experimental low/negative − identified low/negative = FP, experimental high/positive − identified high/positive = FN.
Ijms 26 10207 g002
Figure 3. Preferred docking pose (3D) and ligand interaction diagram (2D) of (a) Tivozanib; (b) TKI.2a; (c) TKI.2b; (d) TKI.19, with VEGFR-2 (PDB ID: 4ASE). Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Cyan dashes/Green line: π-π interactions.
Figure 3. Preferred docking pose (3D) and ligand interaction diagram (2D) of (a) Tivozanib; (b) TKI.2a; (c) TKI.2b; (d) TKI.19, with VEGFR-2 (PDB ID: 4ASE). Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Cyan dashes/Green line: π-π interactions.
Ijms 26 10207 g003aIjms 26 10207 g003b
Figure 4. Preferred docking pose (3D) and ligand interaction diagram (2D) of (a) covalent inhibitor; (b) TKI.6, showing key interactions at the active site of 7PCD. Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Orange arrows: Halogen bonds; Black lines: Covalent bonds.
Figure 4. Preferred docking pose (3D) and ligand interaction diagram (2D) of (a) covalent inhibitor; (b) TKI.6, showing key interactions at the active site of 7PCD. Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Orange arrows: Halogen bonds; Black lines: Covalent bonds.
Ijms 26 10207 g004
Figure 5. Preferred docking pose (3D) and ligand interaction diagram (2D) of EGRF with (a) Mobocertinib; (b) TKI.21b, showing key interactions at the active site of 7T4I. Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Black lines: Covalent bonds.
Figure 5. Preferred docking pose (3D) and ligand interaction diagram (2D) of EGRF with (a) Mobocertinib; (b) TKI.21b, showing key interactions at the active site of 7T4I. Green core: Ligands; Magenta core: Key amino acids; Yellow dashes/Magenta arrows: Hydrogen bond interactions; Black lines: Covalent bonds.
Ijms 26 10207 g005
Figure 6. Receiver operating characteristic (ROC) curve and area under the ROC curve (AUC-ROC) ranked by (a) CNN affinity; (b) CNN pose score.
Figure 6. Receiver operating characteristic (ROC) curve and area under the ROC curve (AUC-ROC) ranked by (a) CNN affinity; (b) CNN pose score.
Ijms 26 10207 g006
Table 1. Free web servers (Molinspiration, Molsoft, SwissADME, Mcule, AdmetLab, pkSCM, Deep-PK, admetSAR, PreADMET) and software (T.E.S.T) used to predict physicochemical properties, Bioavailability, Distribution, Excretion, and Toxicity.
Table 1. Free web servers (Molinspiration, Molsoft, SwissADME, Mcule, AdmetLab, pkSCM, Deep-PK, admetSAR, PreADMET) and software (T.E.S.T) used to predict physicochemical properties, Bioavailability, Distribution, Excretion, and Toxicity.
DescriptorsSoftware/Webservers
Molinspiration [8]Molsoft [9]SwissADME [10]Mcule [11]AdmetLab 3.0 [12,13]pkSCM [14]Deep-PK [15]admetSAR 3.0 [16,17]PreADMET [18]T.E.S.T [19]
Physicochemical PropertiesMolecular weight
TPSA
Molar Refractivity
Log Po/w
Num. rotatable bonds
Num. H-bond acceptors
Num. H-bond donors
Num. Rings
Num. Rigid bonds
Num. atoms
BioavailabilityCaco-2 Permeability
Human Intestinal Absorption
MDCK Permeability
Pgp-substrate
Pgp-inhibitor
DistributionPlasma Protein Binding (PPB)
ExcretionTotal Clearance
ToxicityMutagenicity (Ames test)
Carcinogencity (rat)
hERG Blockers
Hepatotoxicity
✓ Prediction data were provided; ✕ prediction data were not provided.
Table 2. Studied compounds gathered according to their in vitro inhibition outcomes (IC50).
Table 2. Studied compounds gathered according to their in vitro inhibition outcomes (IC50).
A/ACompoundStructureReported Biological TargetIn Vitro Enzyme Inhibition Assay
IC50 (nM)
In Silico StudiesYear of PublicationReference
1TKI.1Ijms 26 10207 i001Ret44-2016[29]
2TKI.2aIjms 26 10207 i002VEGFR-211.9Docking2018[30]
3TKI.2bIjms 26 10207 i003VEGFR-213.6Docking2018
4TKI.3Ijms 26 10207 i004VEGFR-2230ADMET
/Docking
2021[31]
5TKI.4Ijms 26 10207 i005c-Met<1Docking2015[32]
6TKI.5Ijms 26 10207 i006Dual VEGFR-2/C-Met435/
654
Docking2020[33]
7TKI.6Ijms 26 10207 i007dual EGFR/HER-2278/
415
Docking2019[34]
8TKI.7aIjms 26 10207 i008BTK5Docking2013[35]
9TKI.7bIjms 26 10207 i009BTK4.4Docking2013
10TKI.8Ijms 26 10207 i010EGFR97ADMET
/Docking
2021[36]
11TKI.9Ijms 26 10207 i011BTK7.95Docking2018[37]
12TKI.10Ijms 26 10207 i012EGFR3.96ADMET
/Docking
2022[38]
13TKI.11Ijms 26 10207 i013BCR-ABL37Docking2022[39]
14TKI.13aIjms 26 10207 i014dual EGFR/HER-2420ADMET2014[40]
15TKI.13bIjms 26 10207 i015dual EGFR/HER-2220ADMET2014
16TKI.14aIjms 26 10207 i016EGFR147Docking2021[41]
17TKI.14bIjms 26 10207 i017EGFR185Docking2021
18TKI.15Ijms 26 10207 i018VEGFR-2140Docking2020[42]
19TKI.16Ijms 26 10207 i019VEGFR-2110Docking2017[43]
20TKI.17Ijms 26 10207 i020VEGFR-23200ADMET
/Docking
2021[44]
21TKI.18Ijms 26 10207 i021VEGFR-2100Docking2020[45]
22TKI.19Ijms 26 10207 i022VEGFR-2360ADMET
/Docking
2020[46]
23TKI.20aIjms 26 10207 i023VEGFR-2/FGFR-1/PDGFR-β190ADMET
/Docking
2020[47]
24TKI.20bIjms 26 10207 i024VEGFR-2/FGFR-1/PDGFR-β170ADMET
/Docking
2020
25TKI.21aIjms 26 10207 i025EGFR373Docking2020[48]
26TKI.21bIjms 26 10207 i026EGFR369Docking2020
27AIK.1Ijms 26 10207 i027BTK1Docking2020[49]
28AIK.3Ijms 26 10207 i028DDR113Docking2019[50]
29DDK.8Ijms 26 10207 i029LRRK2770Docking2019[51]
Table 3. An illustration of individual and mean value calculation of the molecular descriptors for TKI.1.
Table 3. An illustration of individual and mean value calculation of the molecular descriptors for TKI.1.
ΤΚΙ.1MolinspirationMolsoftSwissADMEMculeAdmetLabpkSCMDeep-PKadmetSARMEAN
Molecular weight (MW—
gr/mole)
329.33329.12329.33329.32329.12329.33329.33329.33329.28
Total Polar Surface Area (TPSA—Å2)76.5159.7376.5076.5076.50nd *76.5076.5074.11
Molar Refractivity (MR)ndnd89.0189.01ndndndnd89.01
Log Po/w3.732.613.113.622.543.542.412.673.03
Num. rotatable bonds (nRbs)4nd4444444
Num. Hbond acceptors (nHAc)656766666
Num. Hbond donors (nHDr)222222222
Num. Rings (nRing)ndndnd33ndndnd3
* nd = not determined by specific platform.
Table 4. An illustration of ADME descriptors and the authors’ comprehensive evaluation for TKI.18.
Table 4. An illustration of ADME descriptors and the authors’ comprehensive evaluation for TKI.18.
ΤΚΙ.18PreADMETSwissADMEAdmetLab 3.0pkSCMDeep-PKadmetSAR 3.0Authors’ Assessment of Overall
Evidence
Bioavailability
Caco-2 Permeability (LogPapp)moderatend *highmoderatemoderatemoderatemoderate
Human Intestinal Absorptionhighlowhighhighhighhighhigh
MDCK Permeability (Papp)lowndmoderatendhighlowlow
Pgp-substratendNondYesNoNoNo
Pgp-inhibitorYesndndYesNoYesYes
Distribution
Plasma Protein Binding (PPB)highndhighndhighhighhigh
Excretion
Total Clearancendndlowmoderatelowlowlow
* nd= not determined by specific platform.
Table 5. An illustration of toxicity and authors’ comprehensive evaluation for TKI.20a.
Table 5. An illustration of toxicity and authors’ comprehensive evaluation for TKI.20a.
ΤΚΙ.20aPreADMETT.E.S.TAdmetLab 3.0pkSCMDeep-PKadmetSAR 3.0Authors’ Assessment of Overall
Evidence
Carcinogenic potential
Mutagenicity (Ames test)positive positive positive negativepositive positive positive
Carcinogenicity (rat)negativend *positive ndnegativenegativenegative
Organ toxicity
hERG Blockersactive ndinactiveinactiveinactiveinactiveinactive
Hepatotoxicityndndpositive positive positive positive positive
* nd = not determined by specific platform.
Table 6. Druglikeness evaluation according to Lipinski, CMC-like, Veber, Egan, Muegge, MMDR-like, and QED rules of FDA-approved tyrosine kinase inhibitors.
Table 6. Druglikeness evaluation according to Lipinski, CMC-like, Veber, Egan, Muegge, MMDR-like, and QED rules of FDA-approved tyrosine kinase inhibitors.
IDDruglikeness
Lipinski RuleGhose/CMC-like RuleVeber RuleEgan Rule Muegge Rule MMDR-like RulesQEDScore 1
Filgotinibpasspasspasspasspasspass0.6716.671
Sunitinibpasspasspasspasspasspass0.6266.626
Fruquintinibpasspasspasspasspasspass0.556.550
Lenvatinibpasspasspasspasspasspass0.5496.549
Vandetanibpasspasspasspasspasspass0.5426.542
Axitinibpasspasspasspasspasspass0.5246.524
Gefitinibpasspasspasspasspasspass0.5186.518
Asciminibpasspasspasspasspasspass0.4986.498
Capivasertibpasspasspasspasspasspass0.4776.477
Pexidartinibpasspasspasspasspasspass0.4666.466
Pirtobrutinibpasspasspasspasspasspass0.4486.448
Erlotinibpasspasspasspasspasspass0.4186.418
Tivozanibpasspasspasspasspasspass0.3886.388
Momelotinibpasspasspasspasspasspass0.5986.598
Tofacitinibpasspasspasspasspassmid-structure0.9285.928
Ritlecitinibpasspasspasspasspassmid-structure0.8455.845
Abrocitinibpasspasspasspasspassmid-structure0.8355.835
Ruxolitinibpasspasspasspasspassmid-structure0.85.800
Upadacitinibpasspasspasspasspassmid-structure0.7335.733
Baricitinibpasspasspasspasspassmid-structure0.7175.717
Larotrectinibpasspasspasspasspassmid-structure0.675.670
Repotrectinibpasspasspasspasspassmid-structure0.6485.648
Lorlatinibpasspasspasspasspassmid-structure0.6155.615
Pemigatinibpassfailpasspasspasspass0.5725.572
Crizotinibpasspasspasspasspassmid-structure0.5335.533
Zanubrutinibpassfailpasspasspasspass0.5245.524
Futibatinibpasspasspasspasspassmid-structure0.5085.508
Deucravacitinibpasspasspassfailpasspass0.4965.496
Pazopanibpasspasspasspasspassmid-structure0.4925.492
Capmatinibpasspasspasspasspassmid-structure0.4895.489
Ibrutinibpassfailpasspasspasspass0.4675.467
Dasatinibpassfailpasspasspasspass0.4665.466
Dacomitinibpassfailpasspasspasspass0.4655.465
Afatinibpassfailpasspasspasspass0.4575.457
Erdafitinibpassfailpasspasspasspass0.4135.413
Regorafenibpassfailpasspasspasspass0.4075.407
Tepotinibpassfailpasspasspasspass0.3855.385
Tucatinibpassfailpasspasspasspass0.3585.358
Gilteritinib1 violationfailpasspasspasspass0.4284.928
Bosutinib1 violationfailpasspasspasspass0.3794.879
Selpercatinib1 violationfailpasspasspasspass0.374.870
Brigatinib1 violationfailpasspasspasspass0.3524.852
Nintedanib1 violationfailpasspasspasspass0.354.850
Cabozantinib1 violationfailpasspasspasspass0.3084.808
Pralsetinib1 violationfailpasspasspasspass0.3074.807
Pacritinibpassfailpasspasspassmid-structure0.5384.538
Acalabrutinibpassfailpasspasspassmid-structure0.4474.447
Avapritinibpassfailpasspasspassmid-structure0.3944.394
Osimertinibpassfailfailpasspasspass0.3114.311
Ponatinib1 violationfailpasspasspassmid-structure0.3943.894
Neratinib1 violationfailfailpasspasspass0.2183.718
Mobocertinib1 violationfailfailpasspasspass0.1743.674
Infigratinibfailfailpasspassfailpass0.3813.381
Ripretinibfailfailpasspassfailpass0.3233.323
Entrectinibfailfailpasspassfailpass0.2943.294
Ceritinibfailfailpasspassfailpass0.2793.279
Nilotinibfailfailpasspassfailpass0.2663.266
Alectinib1 violationfailpasspassfailmid-structure0.5823.082
Midostaurin1 violationfailpasspassfailmid-structure0.2872.787
Fedratinibfailfailfailpassfailpass0.3462.346
Quizatinibfailfailpassfailfailpass0.2572.257
Lapatinibfailfailfailpassfailpass0.1792.179
Fostamatinibfailfailfailfailfailpass0.2561.256
1 Scoring index: Pass (green) = 1, fail (red) = 0, 1 violation (yellow) = 0.5, mid-structure (blue) = 0, minimum acceptable score = 3.501.
Table 7. Criteria for the accomplishment of Medicinal Chemistry (Leadlikeness, GSK, PAINS, Brenk) rules in FDA-approved tyrosine kinase inhibitors.
Table 7. Criteria for the accomplishment of Medicinal Chemistry (Leadlikeness, GSK, PAINS, Brenk) rules in FDA-approved tyrosine kinase inhibitors.
IDMedicinal Chemistry
Leadlikeness GSK RulePAINS
(SwissADME)
Brenk
(SwissADME)
Score 1
Abrocitinibpasspasspasspass4
Ruxolitinibpasspasspasspass4
Tofacitinibpasspasspasspass4
Baricitinibfailpasspasspass3
Fruquintinibfailpasspasspass3
Repotrectinibfailpasspasspass3
Ritlecitinibpasspasspassfail3
Upadacitinibfailpasspasspass3
Alectinibfailfailpasspass2
Avapritinibfailfailpasspass2
Axitinibfailfailpasspass2
Bosutinibfailfailpasspass2
Capivasertibfailfailpasspass2
Capmatinibfailfailpasspass2
Ceritinibfailfailpasspass2
Dasatinibfailfailpasspass2
Deucravacitinibfailfailpasspass2
Entrectinibfailfailpasspass2
Erdafitinibfailfailpasspass2
Erlotinibfailpasspassfail2
Fedratinibfailfailpasspass2
Filgotinibfailfailpasspass2
Gefitinibfailfailpasspass2
Lapatinibfailfailpasspass2
Larotrectinibfailfailpasspass2
Lenvatinibfailfailpasspass2
Lorlatinibfailfailpasspass2
Midostaurinfailfailpasspass2
Nilotinibfailfailpasspass2
Pazopanibfailfailpasspass2
Pemigatinibfailfailpasspass2
Pexidartinibfailfailpasspass2
Pirtobrutinibfailfailpasspass2
Pralsetinibfailfailpasspass2
Quizatinibfailfailpasspass2
Regorafenibfailfailpasspass2
Ripretinibfailfailpasspass2
Selpercatinibfailfailpasspass2
Sunitinibfailpasspassfail2
Tepotinibfailfailpasspass2
Tivozanibfailfailpasspass2
Tucatinibfailfailpasspass2
Vandetanibfailfailpasspass2
Acalabrutinibfailfailpassfail1
Afatinibfailfailpassfail1
Asciminibfailfailpassfail1
Brigatinibfailfailpassfail1
Cabozantinibfailfailpassfail1
Crizotinibfailfailpassfail1
Dacomitinibfailfailpassfail1
Fostamatinibfailfailpassfail1
Futibatinibfailfailpassfail1
Gilteritinibfailfailfailpass1
Ibrutinibfailfailpassfail1
Infigratinibfailfailfailpass1
Mobocertinibfailfailpassfail1
Neratinibfailfailpassfail1
Nintedanibfailfailpassfail1
Osimertinibfailfailpassfail1
Pacritinibfailfailpassfail1
Ponatinibfailfailpassfail1
Zanubrutinibfailfailpassfail1
Momelotinibfailfailfailfail0
1 Scoring index: Pass (green) = 1, fail (red) = 0, minimum acceptable score = 2.
Table 8. Bioavailability evaluation (Caco-2 Permeability, HIA, MDCK prermeability, Pgp-substrate/inhibitor) of FDA-approved tyrosine kinase inhibitors.
Table 8. Bioavailability evaluation (Caco-2 Permeability, HIA, MDCK prermeability, Pgp-substrate/inhibitor) of FDA-approved tyrosine kinase inhibitors.
IDBioavailability
Caco-2
Permeability
Human Intestinal Absorption (HIA)MDCK PermeabilityPgp-SubstratePgp-
Inhibitor
Scoring 1Authors’
Assessment of Overall
Evidence
Lorlatinibhighhighhighnono5high
Ruxolitinibhighhighhighnono5high
Baricitinibhighhighmoderatenono4.5high
Ritlecitinibhighhighmoderatenono4.5high
Abrocitinibmoderatehighmoderatenono4high
Capmatinibhighhighhighyesyes4high
Deucravacitinibmoderatehighmoderatenono4high
Filgotinibmoderatehighmoderatenono4high
Fruquintinibhighhighhighnoyes4high
Futibatinibhighhighhighnoyes4high
Gefitinibhighhighhighyesyes4high
Pemigatinibhighhighhighyesyes4high
Pexidartinibhighhighhighyesyes4high
Tofacitinibmoderatehighmoderatenono4high
Vandetanibhighhighhighyesyes4high
Cabozantinibmoderatehighhighyesyes3.5high
Erlotinibhighhighmoderatenoyes3.5high
Fostamatinibmoderatehighlownono3.5high
Infigratinibmoderatehighhighyesyes3.5high
Larotrectinibhighhighmoderateyesyes3.5high
Momelotinibhighhighmoderateyesyes3.5high
Pirtobrutinibmoderatehighhighyesyes3.5high
Ripretinibmoderatehighhighnoyes3.5high
Tepotinibmoderatehighhighyesyes3.5high
Tivozanibmoderatehighhighnoyes3.5high
Acalabrutinibhighhighlowyesyes3high
Axitinibhighhighlowyesyes3high
Brigatinibmoderatehighmoderateyesyes3high
Ceritinibmoderatehighmoderateyesyes3high
Crizotinibhighhighlowyesyes3high
Dacomitinibhighhighlowyesyes3high
Fedratinibmoderatehighmoderateyesyes3high
Nilotinibmoderatehighmoderateyesyes3high
Pacritinibhighhighlowyesyes3high
Ponatinibmoderatehighmoderateyesyes3high
Pralsetinibmoderatehighmoderateyesyes3high
Quizatinibmoderatehighmoderateyesyes3high
Sunitinibmoderatehighmoderateyesyes3high
Tucatinibhighhighlowyesyes3high
Afatinibmoderatehighlowyesyes2.5low
Alectinibmoderatehighlowyesyes2.5low
Asciminibmoderatehighlownoyes2.5low
Avapritinibmoderatehighlowyesyes2.5low
Bosutinibmoderatehighlowyesyes2.5low
Dasatinibmoderatehighlowyesyes2.5low
Entrectinibmoderatehighlowyesyes2.5low
Erdafitinibmoderatehighlowyesyes2.5low
Gilteritinibmoderatehighlowyesyes2.5low
Ibrutinibmoderatehighlownoyes2.5low
Lapatinibmoderatehighlowyesyes2.5low
Midostaurinmoderatehighlowyesyes2.5low
Mobocertinibmoderatehighlowyesyes2.5low
Neratinibmoderatehighlowyesyes2.5low
Nintedanibmoderatehighlowyesyes2.5low
Osimertinibmoderatehighlowyesyes2.5low
Pazopanibmoderatehighlownoyes2.5low
Regorafenibmoderatehighlownoyes2.5low
Selpercatinibmoderatehighlowyesyes2.5low
Upadacitinibmoderatehighhighyesno2.5low
Zanubrutinibmoderatehighlowyesyes2.5low
Capivasertibmoderatehighmoderateyesno2low
Lenvatinibmoderatehighmoderateyesno2low
Repotrectinibhighhighlowyesno2low
1 Scoring index: high (green) = 1, moderate (yellow) = 0.5, low (red) = 0, No/No (green/red) = 2, No/Yes (green/green) = 1, Yes/Yes (red/green) = 1, Yes/No (red/red) = 0, minimum acceptable score = 2.51.
Table 9. Distribution (PPB) and Excretion (Total Clearance) evaluation of FDA-approved tyrosine kinase inhibitors.
Table 9. Distribution (PPB) and Excretion (Total Clearance) evaluation of FDA-approved tyrosine kinase inhibitors.
IDDistributionIDExcretion
Plasma Protein Binding
(PPB) 1
Total Clearance
AbrocitiniblowAbrocitinibhigh
AvapritiniblowAcalabrutiniblow
BaricitiniblowAfatiniblow
BrigatiniblowAlectinibhigh
CapivasertiblowAsciminiblow
CrizotiniblowAvapritinibhigh
DeucravacitiniblowAxitiniblow
ErdafitiniblowBaricitinibhigh
FilgotiniblowBosutiniblow
FruquintiniblowBrigatiniblow
FutibatiniblowCabozantiniblow
GefitiniblowCapmatiniblow
GilteritiniblowCapivasertiblow
LarotrectiniblowCeritiniblow
MobocertiniblowCrizotiniblow
NintedaniblowDacomitinibhigh
PacritiniblowDasatiniblow
PemigatiniblowDeucravacitiniblow
RepotrectiniblowEntrectiniblow
RitlecitiniblowErdafitiniblow
RuxolitiniblowErlotiniblow
SunitiniblowFedratiniblow
TofacitiniblowFilgotiniblow
UpadacitiniblowFostamatiniblow
VandetaniblowFruquintiniblow
ZanubrutiniblowFutibatiniblow
AcalabrutinibhighGefitiniblow
AfatinibhighGilteritinibhigh
AlectinibhighIbrutiniblow
AsciminibhighInfigratiniblow
AxitinibhighLapatiniblow
BosutinibhighLarotrectiniblow
CabozantinibhighLenvatiniblow
CapmatinibhighLorlatiniblow
CeritinibhighMidostaurinlow
DacomitinibhighMobocertiniblow
DasatinibhighMomelotiniblow
EntrectinibhighNeratiniblow
ErlotinibhighNilotiniblow
FedratinibhighNintedaniblow
FostamatinibhighOsimertiniblow
IbrutinibhighPacritiniblow
InfigratinibhighPazopaniblow
LapatinibhighPemigatinibhigh
LenvatinibhighPexidartinibhigh
LorlatinibhighPirtobrutiniblow
MidostaurinhighPonatiniblow
MomelotinibhighPralsetiniblow
NeratinibhighQuizatiniblow
NilotinibhighRegorafeniblow
OsimertinibhighRepotrectiniblow
PazopanibhighRipretiniblow
PexidartinibhighRitlecitiniblow
PirtobrutinibhighRuxolitinibhigh
PonatinibhighSelpercatiniblow
PralsetinibhighSunitinibhigh
QuizatinibhighTepotinibhigh
RegorafenibhighTivozaniblow
RipretinibhighTofacitinibhigh
SelpercatinibhighTucatiniblow
TepotinibhighUpadacitiniblow
TivozanibhighVandetanibhigh
TucatinibhighZanubrutiniblow
1 Color index: high = red, low = green, acceptable color = green.
Table 10. Toxicity evaluation (carcinogenic potential and organ toxicity) of FDA-approved tyrosine kinase inhibitors.
Table 10. Toxicity evaluation (carcinogenic potential and organ toxicity) of FDA-approved tyrosine kinase inhibitors.
IDToxicity
Carcinogenic PotentialOrgan ToxicityScore 1
Ames TestCarcinogencity (Rat)hERG BlockersHepatotoxicity
Baricitinibnegativenegativeinactivenegative4
Brigatinibnegativenegativeinactivenegative4
Dasatinibnegativenegativeinactivenegative4
Tofacitinibnegativenegativeinactivenegative4
Abrocitinibnegativenegativeinactivepositive 3
Bosutinibnegativenegativeactive negative3
Capivasertibnegativenegativeactive negative3
Dacomitinibnegativenegativeactive negative3
Deucravacitinibnegativenegativeinactivepositive 3
Entrectinibnegativenegativeactive negative3
Filgotinibnegativepositive inactivenegative3
Fostamatinibnegativenegativeinactivepositive 3
Neratinibnegativenegativeactive negative3
Nilotinibnegativenegativeactive negative3
Pemigatinibnegativepositive inactivenegative3
Ponatinibnegativenegativeactive negative3
Regorafenibnegativenegativeinactivepositive 3
Ruxolitinibnegativepositive inactivenegative3
Selpercatinibpositive negativeinactivenegative3
Tepotinibnegativenegativeactive negative3
Upadacitinibnegativepositive inactivenegative3
Acalabrutinibpositive negativeactive negative2
Alectinibnegativepositive active negative2
Avapritinibnegativepositive active negative2
Axitinibpositive negativeinactivepositive 2
Ceritinibnegativenegativeactive positive 2
Crizotinibpositive negativeactive negative2
Erdafitinibnegativepositive active negative2
Fedratinibnegativenegativeactive positive 2
Futibatinibpositive negativeactive negative2
Gefitinibpositive negativeactive negative2
Infigratinibnegativenegativeactive positive 2
Lapatinibpositive negativeinactivepositive 2
Lorlatinibpositive positive inactivenegative2
Midostaurinnegativepositive active negative2
Nintedanibnegativenegativeactive positive 2
Pazopanibnegativepositive inactivepositive 2
Pexidartinibnegativenegativeactive positive 2
Quizatinibnegativepositive active negative2
Vandetanibnegativepositive active negative2
Zanubrutinibpositive negativeactive negative2
Afatinibpositive negativeactive positive 1
Asciminibpositive positive active negative1
Capmatinibpositive positive active negative1
Erlotinibpositive positive active negative1
Fruquintinibpositive positive inactivepositive 1
Gilteritinibpositive positive active negative1
Larotrectinibpositive positive inactivepositive 1
Lenvatinibpositive positive active negative1
Mobocertinibnegativepositive active positive 1
Momelotinibpositive positive inactivepositive 1
Pacritinibnegativepositive active positive 1
Pirtobrutinibpositive positive active negative1
Pralsetinibnegativepositive active positive 1
Ripretinibpositive positive inactivepositive 1
Ritlecitinibpositive positive inactivepositive 1
Tivozanibnegativepositive active positive 1
Tucatinibnegativepositive active positive 1
Cabozantinibnegativepositive active positive 1
Ibrutinibpositive positive active positive 0
Osimertinibpositive positive active positive 0
Repotrectinibpositive positive active positive 0
Sunitinibpositive positive active positive 0
1 Scoring index: Positive (red) = 0, negative (green) = 1, active (red) = 0, inactive (green) = 1, minimum acceptable score = 2.01.
Table 11. DrugBank calculated, experimental, and average predicted MW, TPSA, MR, and LogPo/w values.
Table 11. DrugBank calculated, experimental, and average predicted MW, TPSA, MR, and LogPo/w values.
DrugPhysicochemical Properties
Molecular Weight (g/mol)TPSA (Å2)Molar RefractivityLog Po/w
Calculated DrugBank (yi)Predicted ( y i ^ )Calculated DrugBank (yi)Predicted ( y i ^ )Calculated DrugBank (yi)Predicted ( y i ^ )Experi-mental (yi)Predicted ( y i ^ )Molinspi-Ration/Molsoft ( y i ^ )
Abrocitinib323.42323.3590.9891.3586.0086.59*1.56-
Acalabrutinib465.52465.43118.51114.06135.72 136.510.493.042.50
Afatinib485.94485.7588.61 86.09131.38 129.90*3.95-
Alectinib482.62482.5472.36 69.80155.11 149.50*5.00-
Asciminib449.84449.66103.37 100.27104.81 113.26*3.20-
Avapritinib498.57498.48106.29 102.86164.55 144.37*2.58-
Axitinib386.47386.3970.67 76.06115.14 112.82*4.04-
Baricitinib371.42371.35120.56 119.33105.55 98.51*0.63-
Bosutinib530.45530.0882.88 80.03142.12 150.653.344.594.55
Brigatinib584.10583.8985.86 86.30164.77 176.585.174.084.53
Cabozantinib501.51501.4298.78 95.78136.12 136.59*4.49-
Capivasertib428.92428.73120.16 114.08116.97 118.97*1.67-
Capmatinib412.43412.3585.07 81.96125.27 113.93*2.94-
Ceritinib558.14557.91105.24 104.90153.86 158.71*5.62-
Crizotinib450.34450.0477.99 75.79128.43 120.721.834.273.43
Dacomitinib469.94469.7579.38 77.06129.91 132.733.924.964.90
Dasatinib488.01487.80106.51 111.29133.08 138.631.803.393.50
Deucravacitinib425.47424.64135.95 132.40138.38 113.15*1.82-
Entrectinib560.65560.5485.52 83.34161.24 163.44*4.88-
Erdafitinib446.56446.4777.33 74.91139.32 131.48*4.00-
Erlotinib393.44393.3774.73 72.77107.79 111.402.702.932.50
Fedratinib524.68524.58108.48 108.45147.88 151.66*4.97-
Filgotinib425.51425.4296.67 96.15126.12 118.09*1.98-
Fostamatinib580.46580.39186.72 183.88137.1 141.80*2.30-
Fruquintinib393.40393.3395.71 92.80106.25 106.77*2.92-
Futibatinib418.46418.39108.39 104.89122.82 119.66*2.22-
Gefitinib446.90446.7268.74 66.93117.51 121.663.204.063.82
Gilteritinib552.72552.63121.11 117.42159.84 168.434.352.673.04
Ibrutinib440.51440.4399.16 95.66138.07 131.013.973.853.49
Infigratinib560.48560.1695.09 92.08152.71 159.27*4.85-
Lapatinib581.06580.83106.35 105.71152.42 153.885.405.675.64
Larotrectinib428.44428.3886 82.90122.96 117.01*2.57-
Lenvatinib426.86426.67115.57 111.99112.21 112.863.303.243.14
Lorlatinib406.42406.35110.06 106.65121.17 111.44*2.15-
Midostaurin570.65571.7977.73 74.48162.61 169.205.894.744.66
Mobocertinib585.71585.61113.85 110.30171.52 171.32*4.52-
Momelotinib414.47414.39103.17 100.20118.46 120.29*2.61-
Neratinib557.05556.84112.4 108.35157.29 157.05*4.80-
Nilotinib529.52529.4497.62 94.10152.85 141.085.015.335.14
Nintedanib539.62539.5494.22 92.56159.1 167.003.003.173.5
Osimertinib499.62499.5387.55 84.72150.32 150.43*3.92-
Pacritinib472.59472.5068.74 67.92139.43 143.91*4.29-
Pazopanib437.52437.43119.03 118.07132.18 121.50*3.29-
Pemigatinib487.51487.4383.16 80.71125.32 136.22*2.99-
Pexidartinib417.82417.6466.49 63.64105.89 104.94*4.31-
Pirtobrutinib479.44479.36125.26 121.68127.89 115.16*3.22-
Ponatinib532.56532.4865.77 63.49152.63 150.10*4.45-
Pralsetinib533.61533.52135.53 131.05146.12 143.26*3.52-
Quizatinib560.67560.57106.16 110.87168.24 160.37*5.70-
Regorafenib482.82482.6392.35 89.51114.73 112.44*5.06-
Repotrectinib355.37355.3080.55 78.61106.42 100.58*2.28-
Ripretinib510.37510.0486.36 85.05133.78 133.195.634.955.29
Ritlecitinib285.35285.3073.91 71.5282.84 86.07*1.52-
Ruxolitinib306.37306.3283.18 80.3898.01 87.66*2.41-
Selpercatinib525.61525.52112.04 107.85158.75 152.98*3.35-
Sunitinib398.47398.4177.23 75.97116.27 116.31*3.00-
Tepotinib492.58492.5394.71 93.92154.74 145.45*3.95-
Tivozanib454.86454.68107.74 104.42120.85 120.004.314.604.28
Tofacitinib312.37312.3288.91 85.6687.8 91.201.811.030.65
Tucatinib480.53480.45110.85 106.83148.37 141.663.624.524.46
Upadacitinib380.38380.3278.32 74.9793.03 96.54*2.50-
Vandetanib475.35475.0559.51 57.76118.63 123.265.004.634.67
Zanubrutinib471.56471.48102.4899.64146.25141.35*3.49-
Average ( y ¯ )467.46-96.66-132.43-3.69--
* Experimental data for these values were not found; - predicted data for these values were not calculated.
Table 12. Regression coefficients table for MW, TPSA, and MR.
Table 12. Regression coefficients table for MW, TPSA, and MR.
Molecular Weight
ModelCoefficientsEvaluation Metrics95.0% Confidence Interval for B
BrR2RMSEMAERMSEbaselineLower BoundUpper Bound
1(Constant)0.159-----−0.1720.489
MW predicted1.0001.0001.0000.2300.15272.4690.9991.001
Dependent Variable: MWdrugbank
y = 1.000 (±0.001) × x + 0.159 (±0.331)
TPSA
1(Constant)1.057-----−1.3333.447
TPSA predicted1.0120.9950.9912.9952.71221.0900.9881.037
Dependent Variable: TPSAdrugbank
y = 1.012 (±0.024) × x + 1.057 (±2.190)
Molar Refractivity
1(Constant)12.326-----2.05822.593
MR predicted0.9170.9500.9027.2495.52121.9190.8400.994
Dependent Variable: MRdrugbank
y = 0.917 (±0.077) × x + 12.326 (±10.268)
Table 13. Regression coefficients table for LogPo/w.
Table 13. Regression coefficients table for LogPo/w.
Log Po/w
ModelCoefficientsEvaluation Metrics95.0% Confidence Interval for B
BrR2RMSEMAERMSEbaselineLower BoundUpper Bound
1(Constant)2.213-----1.0983.327
Experimental0.4810.6450.4171.1420.8921.4300.1990.763
Dependent Variable: Log Po/w Predicted
y = 0.481 (±0.282) × x + 2.213 (±1.115)
Log Po/w (Molinspiration–Molsoft)
1(Constant)1.658-----0.6152.702
Experimental0.6030.7500.5620.9700.7861.4300.3390.867
Dependent Variable: Molinspiration–Molsoft
y = 0.603 (±0.264) × x + 1.658 (±1.043)
Table 14. Experimental and predicted Bioavailability, PPB, Clearance and Toxicity (Ames test, Carcinogenicity, hERG Blockers and Hepatotoxicity) data.
Table 14. Experimental and predicted Bioavailability, PPB, Clearance and Toxicity (Ames test, Carcinogenicity, hERG Blockers and Hepatotoxicity) data.
DrugPharmacokinetic PropertiesToxicity
Bioavail-Ability 1Plasma Protein Binding (PPB) 1Clearance (L/h) 1Ames Test 1Carcinogenicity 1hERG Blockers (Human Ether-à-go-go-Related Gene) 1Hepatotoxicity 1
ExpPredExpPredExpPredExpPredExpPredExpPredExpPred
Abrocitinibhighhighlowlow*highnegativenegativenegativenegative*negativenegativepositive
Acalabrutiniblowhighhighhighhighlow*positive negativenegative*positivepositivenegative
Afatinibhighlowhighhighhighlow*positive *negative*positivepositivepositive
Alectiniblowlowhighhighhighhighnegativenegative*positive *positivepositivenegative
Asciminib*lowhighhighlowlownegativepositive *positive *positivenegativenegative
Avapritinibhighlowhighlowlowhighnegativenegative*positive *positivenegativenegative
Axitinibhighhighhighhighlowlowpositivepositive *negativenegativenegativepositivepositive
Baricitinibhighhighlowlowlowhighnegativenegativenegativenegative*negativenegativenegative
Bosutiniblowlowhighhighhighlownegativenegativenegativenegativenegativepositivepositivenegative
Brigatiniblowhighlowlowlowlownegativenegative*negative*negativepositivenegative
Cabozantinibhighhighhighhighlowlownegativenegativenegativepositive *positivepositivepositive
Capivasertiblowlowlowlowlowlow*negativenegativenegative*positivenegativenegative
Capmatinibhighhighhighhighlowlow*positive negativepositive *positivepositivenegative
Ceritinib*highhighhighlowlownegativenegative*negative*positivepositivepositive
Crizotiniblowhighhighlowhighlownegativepositive *negativepositivepositivepositivenegative
Dacomitinibhighhighhighhighlowhighnegativenegative*negative*positivenegativenegative
Dasatinibhighlowhighhighhighlownegativenegativepositivenegativenegativenegativepositivenegative
Deucravacitinibhighhighlowlowlowlownegativenegativenegativenegative*negativenegativepositive
Entrectinib*lowhighhighlowlownegativenegative*negative*positivepositivenegative
Erdafitinibhighlowhighlowlowlownegativenegative*positive *positivenegativenegative
Erlotinibhighhighhighhighlowlownegativepositive positivepositive negativepositivepositivenegative
Fedratinib*highhighhighlowlownegativenegativenegativenegative*positivepositivepositive
Filgotinib*high*low*low*negative*positive *negative*negative
Fostamatinibhighhighhighhigh*lownegativenegativenegativenegative*negativepositivepositive
Fruquintinib*highhighlowlowlownegativepositive *positive *negativepositivepositive
Futibatinib*highhighlowlowlownegativepositive *negative*positivenegativenegative
Gefitinibhighhighlowlow*lownegativepositive positivenegativepositivepositivepositivenegative
Gilteritinib*lowhighlowlowhighnegativepositive *positive *positivenegativenegative
Ibrutiniblowlowhighhighhighlownegativepositive negativepositive *positivepositivepositive
Infigratinib*highhighhighlowlownegativenegative*negative*positivenegativepositive
Lapatinib*lowhighhigh*lownegativepositive negativenegativepositivenegativepositivepositive
Larotrectiniblowhighlowlowhighlownegativepositive *positive *negativepositivepositive
Lenvatinibhighlowhighhigh*lownegativepositive *positive *positivepositivenegative
Lorlatinibhighhighlowhighlowlownegativepositive *positive *negativepositivenegative
Midostaurin*lowhighhigh*lownegativenegative*positive *positivenegativenegative
Mobocertiniblowlowhighlowhighlownegativenegative*positive *positivenegativepositive
Momelotinib*highhighhighhighlownegativepositive negativepositive *negativepositivepositive
Neratinibhighlowhighhighhighlownegativenegativenegativenegative*positivepositivenegative
Nilotiniblowhighhighhighlowlownegativenegativenegativenegativepositivepositivepositivenegative
Nintedaniblowlowhighlowhighlownegativenegativenegativenegative*positivepositivepositive
Osimertiniblowlowhighhighlowlownegativepositive positivepositive *positive*positive
Pacritinib*highhighlowlowlownegativenegativenegativepositive *positivenegativepositive
Pazopaniblowlowhighhigh*lownegativenegativepositivepositive negativenegativepositivepositive
Pemigatiniblowhighhighlowlowhighnegativenegative*positive *negativenegativenegative
Pexidartinib*highhighhighlowhighnegativenegativenegativenegative*positivenegativepositive
Pirtobrutinibhighhighhighhighlowlownegativepositive *positive *positivepositivenegative
Ponatinib*highhighhigh*lownegativenegativepositivenegative*positivenegativenegative
Pralsetinib*highhighhighlowlownegativenegative*positive *positivepositivepositive
Quizatinib*high*high*low*negative*positive *positive*negative
Regorafenibhighlowhighhigh*low*negative*negativenegativenegativepositivepositive
Repotrectiniblowlowhighlowlowlownegativepositive *positive *positivepositivepositive
Ripretinibhighhighhighhighlowlownegativepositive *positive *negativenegativepositive
Ritlecitinibhighhighlowlow*lownegativepositive positivepositive *negativenegativepositive
Ruxolitinibhighhighhighlowlowhighnegativenegativenegativepositive negativenegativenegativenegative
Selpercatinibhighlowhighhighlowlownegativepositive negativenegative*negativepositivenegative
Sunitinibhighhighhighlowlowhighnegativepositive positivepositive positivepositivepositivepositive
Tepotinibhighhighhighhighlowhigh*negative*negative*positivepositivenegative
Tivozanibhighhighhighhighlowlow*negative*positive *positivenegativepositive
Tofacitinibhighhighlowlow*highnegativenegativenegativenegative*negativenegativenegative
Tucatinib*highhighhighhighlownegativenegative*positive *positivepositivepositive
Upadacitinib*lowlowlow*lownegativenegativenegativepositive *negativenegativenegative
Vandetanibhighhighhighlow*highnegativenegative*positive positivepositivenegativenegative
Zanubrutinib*lowhighlowhighlownegativepositive *negative*positivepositivenegative
* Experimental data for these values were not found. 1 In this table, colors (red/green) are applied to enhance comparison.
Table 15. Compounds eliminated by the screening technique for each criterion, arranged by the total number of hits.
Table 15. Compounds eliminated by the screening technique for each criterion, arranged by the total number of hits.
Top 24—DruglikenessTop 16—Med. ChemistryTop 21—BioavailabilityTop 2—DistributionTop 9—Overall Toxicity
1AIK.1TKI.16TKI.16TKI.16TKI.19
2TKI.19DDK8DDK.8AIK.1DDK8
3DDK8TKI.19TKI.19 TKI.4
4TKI.21bTKI.21bTKI.21b TKI.21b
5TKI.16AIK.1TKI.4 TKI.8
6TKI.4TKI.4AIK.1 TKI.20b
7TKI.20bTKI.14bTKI.14a TKI.18
8TKI.1TKI.8TKI.14b TKI.13b
9TKI.14bTKI.6TKI.6 TKI.13a
10TKI.14aTKI.2aTKI.1
11TKI.2bTKI.2bTKI.20b
12TKI.2aTKI.14aTKI.2a
13TKI.8TKI.1TKI.2b
14TKI.6TKI.17TKI.10
15TKI.20aTKI.11TKI.5
16TKI.10TKI.15TKI.21a
17TKI.5 TKI.9
18TKI.9 TKI.20a
19TKI.21a AIK.3
20TKI.17 TKI.3
21TKI.18 TKI.7a
22TKI.13a
23TKI.13b
24TKI.11
Table 16. Compounds identified through our novel screening approach, demonstrating the percentage of criteria met, points of violation, and reported biological targets.
Table 16. Compounds identified through our novel screening approach, demonstrating the percentage of criteria met, points of violation, and reported biological targets.
D-ADMET Screening
A/ACompoundStructureCriteria Meeting (≥60%)Violation PointsReported Biological Target
1TKI.1Ijms 26 10207 i03060Distribution, Overall ToxicityRet
2TKI.2aIjms 26 10207 i03160Distribution, Overall ToxicityVEGFR-2
3TKI.2bIjms 26 10207 i03260Distribution, Overall ToxicityVEGFR-2
4TKI.4Ijms 26 10207 i03380Distributionc-Met
5TKI.6Ijms 26 10207 i03460Distribution, Overall Toxicitydual EGFR/HER2
6TKI.8Ijms 26 10207 i03560Distribution, BioavailabilityEGFR
7TKI.14aIjms 26 10207 i03660Distribution, Overall ToxicityEGFR
8TKI.14bIjms 26 10207 i03760Distribution, Overall Toxicity
9TKI.16Ijms 26 10207 i03880Overall ToxicityVEGFR-2
10TKI.19Ijms 26 10207 i03980DistributionVEGFR-2
11TKI.20bIjms 26 10207 i04060Medicinal Chemsistry,
Distribution
VEGFR-2/FGFR-1/PDGFR-β
12TKI.21bIjms 26 10207 i04180DistributionEGFR
13AIK.1Ijms 26 10207 i04280Overall ToxicityBTK
14DDK.8Ijms 26 10207 i04380DistributionLRRK2
Table 17. Results from cross-docking demonstrating affinity, CNN pose score, CNN affinity, and RMSD values for known drugs pertaining to each biological target.
Table 17. Results from cross-docking demonstrating affinity, CNN pose score, CNN affinity, and RMSD values for known drugs pertaining to each biological target.
TargetDrugAffinity (kcal/mol)CNN Pose ScoreCNN AffinityCross-Docking RMSD (Å)
VEGFR-2 (PDB ID:4ASE)Axitinib−8.530.8427.6345.500
Cabozatinib−11.860.9137.7251.059
Fruquitinib−8.740.9067.6721.499
Lenvatinib−11.030.9578.0492.776
Pazopanib−8.690.8567.4073.730
Regorafenib−11.240.8907.8331.688
Sorafenib−11.250.8827.5882.536
Sunitinib−7.350.7287.3125.250
Vandetanib−10.420.8148.0621.514
HER2 (PDB ID:7PCD)Afatinib−7.610.9257.3812.906
Capivasertib−9.710.8987.452.537
Lapatinib−9.980.8587.6092.022
Neratinib−7.510.7807.8752.432
Tucatinib−10.640.7507.6341.494
EGFR (PDB ID:7T4I)Afatinib−8.350.9007.8522.169
Dacomitinib−8.600.9328.1252.186
Gefitinib−7.930.9837.9861.862
Osimertinib−7.120.9327.9481.562
Table 18. T-test for equality of means (independent samples test).
Table 18. T-test for equality of means (independent samples test).
Independent Samples Test
Molecular DescriptorstdfSignificanceMean
Difference
Std. Error
Difference
95% Confidence Interval of the Difference
Two-Sided (p)LowerUpper
Molecular weight−0.01047.2450.992−0.2221.457−43.38442.937
TPSA0.96250.8040.3415.315.520−5.77416.392
MR0.34151.2480.7352.045.988−9.98014.060
LogPo/w3.89064.305<0.011.180.3040.5751.789
nRB−0.18951.2220.85101−1.3861.147
nHA−0.83152.7760.41000−1.1140.462
nHD0.52054.2160.60500−0.3920.666
nRings0.86060.4750.39300−0.2540.638
nRigidB1.42054.8020.16121−0.7594.442
nAtoms−1.09453.4160.279−33−7.9692.344
Equal variances not assumed.
Table 19. T-test for equality of distributions (independent-samples Kolmogorov–Smirnov test).
Table 19. T-test for equality of distributions (independent-samples Kolmogorov–Smirnov test).
Independent-Samples Kolmogorov–Smirnov Test
Molecular DescriptorsMost Extreme DifferencesSignificance
Absolute (D)PositiveNegativeTwo-Sided (p)
Molecular weight0.2010.181−0.2010.514
TPSA0.2280.228−0.1030.352
MR0.1470.147−0.0950.866
LogPo/w0.4060.4060.0000.008
nRB0.1650.078−0.1650.753
nHA0.1660.010−0.1660.748
nHD0.0710.071−0.0431.000
nRings0.1150.115−0.0260.981
nRigidB0.2100.210−0.0430.453
nAtoms0.1560.061−0.1560.815
Total N = 29 + 39= 68.
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

Mavridis, E.; Hadjipavlou-Litina, D. Using a Novel Consensus-Based Chemoinformatics Approach to Predict ADMET Properties and Druglikeness of Tyrosine Kinase Inhibitors. Int. J. Mol. Sci. 2025, 26, 10207. https://doi.org/10.3390/ijms262010207

AMA Style

Mavridis E, Hadjipavlou-Litina D. Using a Novel Consensus-Based Chemoinformatics Approach to Predict ADMET Properties and Druglikeness of Tyrosine Kinase Inhibitors. International Journal of Molecular Sciences. 2025; 26(20):10207. https://doi.org/10.3390/ijms262010207

Chicago/Turabian Style

Mavridis, Evangelos, and Dimitra Hadjipavlou-Litina. 2025. "Using a Novel Consensus-Based Chemoinformatics Approach to Predict ADMET Properties and Druglikeness of Tyrosine Kinase Inhibitors" International Journal of Molecular Sciences 26, no. 20: 10207. https://doi.org/10.3390/ijms262010207

APA Style

Mavridis, E., & Hadjipavlou-Litina, D. (2025). Using a Novel Consensus-Based Chemoinformatics Approach to Predict ADMET Properties and Druglikeness of Tyrosine Kinase Inhibitors. International Journal of Molecular Sciences, 26(20), 10207. https://doi.org/10.3390/ijms262010207

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