1. Introduction
Phenothiazines (PTHs) are heterocyclic compounds based on sulfur and nitrogen. Due to their high biological activity, the phenothiazine ring system is one of the leading structures in the pharmaceutical industry particularly in the development of new drugs [
1,
2,
3]. A comprehensive review of the literature shows that phenothiazine derivatives have wide application in both psychiatric states (e.g., antipsychotic, neuroleptic) and non-psychiatric states, such as antiviral or anti-proliferative effects [
1,
2,
3]. Chlorpromazine belongs to the aliphatic group of phenothiazines. This compound was introduced to psychiatric practice in 1952 and has influenced advances in the treatment of mental illness. Over the past few decades, much attention has been paid to the synthesis of phenothiazine derivatives and their study for a variety of pharmacological activities, including antibiotics, painkillers, sedatives, and antivirals [
1,
2,
3]. In recent years, in vitro studies have confirmed also its usefulness as a promising antitumor agent showing activity against various cancers, such as glioma [
4]. Other studies have suggested that work has pointed to the use of this drug to combat psychotic episodes in patients diagnosed with COVID-19, alone or in combination with hydroxychloroquine [
5,
6,
7,
8]. The compounds containing chlorpromazine have also been shown to have potentially anticancer properties against the cancer cells of colon, breast, lung, brain, as well as leukemia and lymphoma [
5,
6,
7,
8].
These observations highlight the need to develop new phenothiazine derivatives, including chlorpromazine analogs, as effective drugs against these diseases. For a new molecule, including phenothiazine derivatives, to become an effective drug, it must not only exhibit biological activity, but also be safe for the potential patient and have a favorable ADME (absorption, distribution, metabolism, and excretion) profile which determines the pharmacokinetic properties of individual bioactive compounds as drug candidates. The ADME parameters need to be predicted early, during the first stages of drug development. This can help to reduce the number of compounds with undesirable ADME properties studied in later phases of drug development, i.e., in clinical trials. Among the various physicochemical properties, lipophilicity is one of those that can affect the ADME profile of active molecules. Lipophilicity is a key parameter in drug design, and thus in the pharmaceutical industry, because it determines the permeability of the compound through the membrane in the biological system and is helpful in optimizing the structure of new drug molecules [
9,
10,
11,
12,
13]. Lipophilicity of organic compounds is expressed as log P, the base-10 logarithm of the partition coefficient (P), which is defined as the ratio of the compound’s concentration in two immiscible solvents: a non-polar organic phase and a polar aqueous phase at equilibrium [
9]. Current methods of determining the lipophilicity parameter include, in addition to the traditional flask shaking technique, various chromatographic systems for thin-layer chromatography and high-performance liquid chromatography (HPLC—high-performance liquid chromatography) [
9,
14,
15]. In this method, lipophilicity chromatographic parameters (R
M0 and logk
w) are obtained from the retention factor R
M or k and extrapolated to the zero content of the organic modifier in the applied mobile phase. Due to its ease of use and the possibility to analyze several compounds simultaneously on the same plate, TLC (thin-layer chromatography) is a method often used in the study of the lipophilicity of different groups of newly synthesized compounds as potential drug candidates. Numerous applications of thin-layer chromatography in the evaluation of the lipophilicity of pharmaceuticals including potential candidates for new drugs are reported in the literature [
16,
17,
18,
19,
20,
21,
22]. In addition to experimental methods, a wide range of computational tools for predicting ADME parameters, including lipophilicity, have been developed in recent decades, such as SwissADME, pkCSM, Molinspiration, and others [
23,
24,
25,
26,
27,
28,
29]. Existing calculation methods for log P are classified into four groups:
- -
atom-based methods;
- -
fragment-based methods;
- -
topology-based methods;
- -
structural-property-based methods.
It is known that the logP computational algorithms from different families have their respective advantages and disadvantages. Some calculation methods are more or less suited for specific heterogeneous compounds. The poor predictive power of software packages might be explained by insufficient coverage of the chemical surface by measured compounds. The calculation algorithms are as good as the data that they are based on. Generally, the additional corrections introduced to them allows one to achieve better prediction accuracy for the determination of the theoretical parameter of lipophilicity. Correction factors are introduced to rectify the calculated logP when some special substructures occur in the molecule. As it was well described by Mannhold et coworkers [
30], there are structural and interaction factors. Structural factors concern the chain bonds, ring bonds, and branch bonds. Interaction factors consider aliphatic proximity, electronic effects through π-bonds as well as special ortho effects [
30]. The first type of algorithms, i.e., atom-based methods and atom additive methods such as AlogP, are suitable for small molecules without complex structures. These methods cut molecules down to single atoms and do not apply correction rules. An advantage of atom-based methods is that ambiguities are avoided; a shortcoming is the failure to deal with long-range interaction [
30]. Therefore, to overcome these shortcomings, the adjusted atom-based or hybrid methods are implemented like XlogP or Silicos-IT logP, respectively. Currently, a new version of the XlogP3 algorithm adopts an optimized classification scheme of 87 atom types as well as two correction factors accounting for internal H-bonds and amino acids. Fragment-based methods divide molecules into fragments and apply correction factors to account for intramolecular interactions (e.g., ClogP) for log P calculation are similarly based on summing up of the hydrophobicity contribution of each fragment in a molecule, i.e., fragment constant (hydrophobicity contribution of each fragment) which are determined by the experimental value of log P. The above-mentioned Clog P is the most frequently used log P calculation tool. Recent versions include the basic fragmental values which were derived from the measured log P data of simple-molecule complex hydrocarbons, whose measured values were not the sum of the fragment values; the differences were defined in terms of correction factors. Thanks to the additional correction factors taking into account additional interactions, such as hydrophilicity shield effect and hydrogen bonding, these methods are better predictors for large molecules. Next, the third family is topology- or graph-based methods (e.g., Mlog P) which use topological descriptors generated by means of 2D structures. Their main advantage is speed. The last group is structural-property-based methods. These methods use a physical–chemical perspective and 3D structure to calculate log P values. Among the computational methods, DFT-based implicit solvent models provide a physically grounded approach by estimating solvation free energies in water and 1-octanol to compute log P. Owing to systematic error cancelation between solvents, they offer reasonable accuracy (typically with mean absolute errors of around 0.6 log units) and are computationally more efficient than explicit solvent simulations. Although generally outperformed by empirical fragment-based methods for well-represented neutral compounds, DFT-based models are advantageous when dealing with novel or structurally diverse molecules outside standard training sets [
31].
Prediction of lipophilicity parameters and other ADME descriptors in silico allows for early reduction of new drugs with undesirable ADME properties. In several papers, experimental values of lipophilicity parameters were compared with theoretical values of the partition coefficient obtained using software and web servers to predict ADME parameters [
17,
20,
21,
22,
29]. Theoretical values of the lipophilicity descriptors of the studied compounds may be helpful. However, predictions will not be enough without the experimental evaluation of drug candidates during the drug discovery and development process.
Thus, our work aimed to determine the lipophilic parameters of previously synthesized chlorpromazine derivatives (
1-
17)—
Figure 1—both experimentally using thin-layer reverse phase chromatography (RP-TLC) and computationally, employing various software tools. In addition, other key ADMET parameters that describe the pharmacokinetic behavior of drugs were determined. Chlorpromazine (
18) (known for years as a drug) was also used in the studies as a reference compound to compare the tested parameters of the new substances. The results of our previous work confirmed the antibacterial and pro-apoptotic activity of these novel compounds in relation to the following cancer cell lines: A549, MiaPaCa-2, and HCT-116 [
32].
2. Results and Discussion
This study presents an analysis of the lipophilic properties and other ADME parameters obtained in silico for quinoline analogues of chlorpromazine that were previously synthesized and exhibit diverse anticancer and antimicrobial activity. The new derivatives were obtained via multi-step reactions based on 6
H-8-chloroquinobenzothiazine [
32]. This tetracyclic phenothiazine was obtained via the reaction of 2-amino-4-chlorobenzenethiol and 3-bromo-2-chloroquinoline. The tested 8-chloroquinobenzothiazines exhibited promising activity against A549 cells without affecting HaCaT cells. Compounds
2,
9,
15, and
17 (
Figure 1) showed the most promising cytotoxicity against A549 cells and a higher selectivity index (SI = 7.6–10.7) than the reference compound, doxorubicin (SI = 0.14–0.15). Compounds
2 and
16 showed the highest selectivity index (143), with IC
50 values of 1.6 µM and 0.7 µM, respectively, against HCT-116 cells, while showing no cytotoxic effects on HaCaT cells. Compound
5, on the other hand, showed high cytotoxicity against HCT-116 cells (7.7 µM) (
Table S1). Studies on the mechanisms of cytotoxic action of the discussed substances confirmed their proapoptotic activity, especially in terms of inducing late apoptosis or necrosis in cancer cell lines A549, MiaPaCa-2, and HCT-116. The activity of new chlorpromazine analogues against standard Gram-positive bacteria (various strains of
S. aureus and
S. epidermidis) and Gram-negative bacteria (
E. coli and
P. aeruginosa) was also tested. 8-Chloroquinobenzothiazines
1,
2,
3,
9, and
15 showed moderate antibacterial activity, mainly against standard strains of staphylococci. The most significant activity against standard strains was observed for compound
15 (MIC = 2–8 µg/mL) [
32].
Studies to determine and analyze the lipophilicity parameters of the new substances began by determining the lipophilicity parameters (log P
calc) using selected computational programs. The values obtained computationally are presented in
Table 1.
Analyzing the obtained results, one can observe large differences in the values of the lipophilicity parameters calculated with different programs, i.e., log Pcalc. Differences in the values obtained for individual substances reach even more than three units. For example, for 8-chloroquinobenzothiazine 17, the iLOGP program provided a value of 3.67, while the WLOGP and Clog P programs yielded 7.26 and 7.81, respectively. In the case of substance 14, the MLOGP program produced a value of 3.04, and the Clog P program yielded 6.76. For most of the tested substances, the lowest values for log P were obtained using the iLOGP program. The exceptions were 8-chloroquinobenzothiazines 14 and 16 for which the lowest values of the log Pcalcd parameter were calculated using the MLOGP program. The highest values of this parameter reaching up to 7.90 for compound no 12 were obtained from the Clog P program. The calculated log P values were not sufficiently precise in terms of possible contributions from conformation, folding, hydration, ion pair formation, and intra- and intermolecular hydrogen bond formation. The low predictive power of the programs can be attributed to the nonplanarity of the four-ring quinobenzothiazine system as well as the boat conformation of the central thiazine ring (such conformation was determined via X-ray analysis of the selected compounds and may additionally result from an unpredictable conformation of long substituents at the thiazine nitrogen atom. These results show that, although computational techniques are a cheap, fast way to estimate lipophilicity parameters of new drug candidates, especially in early stages of research, experimental methods still cannot be omitted.
In connection with this, in the next stage of the study, the parameters of lipophilicity of the tested compounds were determined experimentally. The experimental lipophilicity of substances
1–
18 was tested using the RP-TLC method. This technique was used to determine the retardation coefficient R
f for each compound in eight mobile phases consisting of acetone and Tris buffer in different volume ratios (50–85% acetone). Next, the R
f values were converted to the chromatographic parameter R
M, and then the obtained values were extrapolated to 0% acetone content in the mobile phase obtaining the lipophilicity parameter R
M0 (
Table S2). In addition to this, due to a relationship observed between R
M0 and the slope of these linear plots (b), the lipophilicity parameter C
0 was also determined (
Table S2).
The parameter R
M0 can be correlated to an experimental log P lipophilicity parameter of chemical molecules (log P
TLC) by applying a calibration curve. The calibration curve was obtained by determining the R
f value for standard substances with a known log P
lit value using the RP-TLC method under conditions analogous to those of the tested substances. After calculating the R
M and R
M0 values, the dependence of the log P
lit parameter on the experimentally obtained R
M0 values for standards was determined (
Table 2).
A relationship characterized by a high correlation coefficient was obtained. Using the standard curve equation:
log P
TLC parameters for the tested compounds
1–
18 were calculated. The log P
TLC values of the tested compounds obtained in this way are presented in
Table 3. The experimentally determined log P
TLC values for the tested quinoline analogues of chlorpromazine ranged from 3.70 to 6.22. The highest lipophilicity was determined for derivative
17 with a
p-toluenesulfonamidobutyl substituent at the thiazine nitrogen atom and the lowest for derivative
7 with an acetylaminopropyl substituent. The values of the log P
TLC parameter for the tested substances
1–
5, containing dialkylaminoalkyl substituents, were similar and were in the range of 3.77 to 4.20. Replacing the pyrrolidine ring (compound
3) with a piperazine ring (compound
4) did not affect this parameter’s value. A greater change can be observed by changing the substituent to 1-methyl-2-piperidine (compound
5). Among derivatives
6–
11 containing differently substituted propyl fragments and for derivatives
12–
17 with four-carbon linkers, the highest values of the log P
TLC parameter were obtained for derivatives
11 and
17 containing a
p-toluenesulfonamide fragment. In addition, compounds with the butylene chain (
6–
11) were more lipophilic than compounds with the propylene linker (
12–
17). In our previous studies on the lipophilicity of a large group of 6,9-disubstituted quinobenzothiazines (with H, Cl, and SCH
3 at position 9 of the quinobenzothiazine ring) we found the same pattern [
38]. The lipophilicity of derivative
2 with a dimethylaminopropyl substituent was slightly higher than the lipophilicity of chlorpromazine
18 used as a reference substance. The introduction of the quinoline system in place of one of the benzene rings of the thiazine system increased the value of the lipophilicity parameter by nearly 0.5.
Figure 2 shows a comparison of all theoretical lipophilicity parameters (log P
calc) and experimental (log P
TLC) of the examined 8-chloroquinobenzothiazines
1–
17 and chlorpromazine
18.
As can be seen in the graph shown in
Figure 2, the closest log P values (overlapping curves) were obtained using computational algorithms such as XLOGP3, WLOGP, MLOGP, and SILCOS-IT, as well as the RP-TLC method as log P
TLC for compounds
1–
7. This fact indicates that the four different algorithms used to calculate log P are suitable for the rapid estimation of the lipophilicity of the first group of tested compounds designated as
1–
7. The analysis of
Figure 3 confirms the previous observations regarding the different power of prediction of computational algorithms used to calculate the theoretical value of the partition coefficient of the studied compounds in relation to the RP-TLC method. Therefore, in order to compare both, in the further stage of the analysis, the experimental log P
TLC values of all tested compounds were compared with the calculated values (log P
calc) using a statistical tool, namely cluster analysis. Interpretation of dendrogram of all lipophilicity parameters of examined compounds
1–
17 (8-chloroquinobenzothiazines) and chloropromazine
18 presented in
Figure 3 shows that the biggest similarities in the case of all compounds indicate the theoretical log P values obtained by means of SILICOS-IT and Mlog P algorithms. These two partition coefficients form one cluster with the smallest Euclidean distance. The observed similarity of experimental values of the partition coefficient as log P
TLC with Xlog P3 values confirms that this model of calculation based on the atom-based method including corrective factors can be a good tool to obtain reliable results of lipophilicity parameters for all the compounds studied.
Figure 4 shows the dendrogram of similarity of the tested compounds (8-chloroquinobenzothiazines)
1–
17 and chloropromazine
18 based on both lipophilicity parameters, i.e., theoretical (Clog P, XLOGP3, WLOGP, MLOGP, SILICOS-IT, iLOGP) and chromatographic one (log P
TLC).
Analysis of the dendrogram shown in
Figure 4 indicates that cluster analysis allowed one to group all tested compounds
1–
18 into several smaller and then one large cluster taking into account the lipophilicity properties as the key parameter in describing the ADME profile of the studied compounds. The highest similarity given lipophilic properties indicates compound
4 and
1 (the smallest Euclidean distance) with diethylamine and piperidine substituents, respectively, and derivatives
2 and
3 with dimethylamine and pyrrolidine substituents. Substance
5, also containing a dialkylaminoalkyl substituent, shows less similarity to compounds
1–
4. This may be due to the different position of the nitrogen atom in the piperidine ring than in substance
4. A common cluster, although isolated from the rest, is formed by substances
11 and
17 containing a
p-toluenesulfonic fragment in the substituent at the thiazine nitrogen atom, and substances
7 and
13 with acetylaminoalkyl substituents. On the other hand, the smallest similarity to other compounds (the greatest distance) is observed at the dendrogram in the case of compound
10.
Next, to illustrate the relationship between the theoretical partition coefficients and the chromatographic (i.e., experimental) lipophilicity parameter (log P
TLC), a heat map was generated (
Figure 5).
As can be seen in
Figure 5, the Clog P parameter was found to be the closest to the experimental value. The MLOGP parameters were found to deviate the most from the log P
TLC value. A significant similarity can also be observed in the values obtained with the XLOGP program. These log P values were calculated by means of atom-based and fragment contribution methods [
27]. Correlating experimentally determined and calculated log P values remains crucial for understanding and predicting lipophilicity. As demonstrated in recent systematic studies of a series of fluorinated compounds, computational methods—from fragment-based models to quantum mechanical approaches—can reflect general trends in lipophilicity, although discrepancies often arise for specific structural motifs. These differences underscore the importance of careful model selection and validation against experimental data when investigating the effects of subtle chemical modifications on log P [
39].
In order for a newly synthesized substance to be considered as a potential drug candidate, it must exhibit specific pharmacokinetic, pharmacological, and toxicological properties. To assess the suitability of a given substance as a drug candidate, it is necessary first to assess its similarity to the drug and determine its properties in terms of absorption, distribution, metabolism, and excretion (ADME). A preliminary assessment of the similarity of a molecule to the drug is performed early in the research process to accelerate the discovery and development of new drugs. There are several methods for assessing the drug similarity of a tested substance. These include the rules proposed by Lipinski, Ghose, Veber, Egan, and Muegge. Each of these rules aims to determine whether a chemical compound with a specific pharmacological or biological activity has properties that make it an active orally administered drug. The criteria on which these rules are based refer to the properties of the substance that are relevant to pharmacokinetic processes (absorption, distribution, metabolism, and excretion). In order to confirm the bioavailability of the tested compounds
1–
18, their compliance with the rules of Lipinski, Ghose, Veber, and Egan was checked using the SwissADME platform. The bioavailability parameters that allow us to determine the drug-likeness of the tested substances are presented in
Table 4.
The molar mass of the tested compounds
1–
17 is in the range of 369.91 (compound
2) to 510.07 (compound
17), the value of the lipophilicity parameter MLOGP is in the range of 3.04 (for substance
14) to 5.27 (for substance
5), the number of n-HBA hydrogen bond acceptors for all substances does not exceed 10, and for each of the tested compounds
1–
17, the number of hydrogen bond donors is below 5. Lipinski’s rule assumes that a substance with good bioavailability should have a molecular weight < 500, no more than five hydrogen bond donors, no more than ten hydrogen bond acceptors, and a lipophilicity parameter value MLOGP < 5. A substance is considered to meet this rule if it exhibits three of these parameters. The results of fitting the tested substances to the Lipinski rule are presented in
Table 5. Of the substances tested, only compound
17 does not comply with this rule. The table also shows which parameters fall outside the range given for the Lipinski rule. Because the Lipinski Rule of Five (Ro5) remains a common benchmark for assessing oral drug similarity, we decided to address it in this study, although its limitations are increasingly recognized. As the recent literature has shown, Ro5 should be viewed as a flexible guideline rather than a strict rule, especially given its limited applicability to certain classes of compounds [
40]. More advanced metrics, such as ligand lipophilic efficiency (LLE), molecular flexibility, and three-dimensionality, now offer a more nuanced understanding of drug similarity in modern lead optimization. According to Ghose’s rule, a drug-like substance should have a molecular weight in the range of 160–480, a molar refractive index in the range of 40–130, a WLOGP of 0.4 to 5.6, and an atom count of 20 to 70. As shown in
Table 5, six of the seventeen substances tested do not meet these criteria (
5,
6,
11,
12,
14, and
17). However, all the 8-chloroquinobenzothiazines tested meet Veber’s rule, which assumes that a substance that is a good drug candidate should have at most 10 rotatable bonds and a TPSA of at most 140 Å. Egan’s rule based on the parameters TPSA and WLOGP (WLOG ≤ 5.88, TPSA ≤ 131.6 Å
2) is not fulfilled only by substances
11 and
17 with
p-toluenesulfonamidoalkyl substituents. However Muegge’s rule is fulfilled only by three of the tested substances, 8-chloroquinobenzothiazines
7,
10, and
16. This rule is based on the highest number of parameters, and the tested substances do not meet it due to the value of the lipophilicity parameter XLOGP3 being higher than 5 [
41]. In terms of similarity to chlorpromazine used as a reference, which can be assessed based on the rules of Lipinski, Ghose, Veber, Egan, and Muegge, the tested derivatives containing dialkylaminoalkyl substituents
1–
4 and derivatives containing three-carbon chains in the substituent at the thiazine nitrogen atom
7–
10 may exhibit similar properties (
Table 5).
In order for a newly obtained chemical molecule to be considered as a potential drug, it must demonstrate a favorable ADME profile, good bioavailability, and no side effects. Predictive methods are inexpensive and convenient for the initial assessment of these parameters at the first stage of the evaluation of new substances in terms of their suitability as potential therapeutic substances [
42].
To initially assess the ADME profile of the synthesized compounds, pkCSM was used, a program that is often used in the initial evaluation of new drug candidates [
43]. Using this program, the parameters responsible for absorption (water solubility, Caco-2 permeability, intestinal absorption, skin permeability), distribution (VDss, unbound fraction, BBB permeability, and CNS permeability), excretion, and toxicity (total clearance, max. tolerated dose, oral rat acute toxicity, oral rat chronic toxicity, T. Pyriformis toxicity, and minnow toxicity) were calculated for the discussed substances.
The parameter concerning water solubility obtained from the pkCSM program is given as logS (S—solubility expressed in mol/L). All tested 8-chloroquinobenzothiazines
1–
17 are characterized by poor solubility in water, which is due to their chemical structure (four six-membered condensed rings). Parameter values range from −6.26 (compound
12) to −4.92 (compound
3). The value of this parameter for chlorpromazine
18 used as a reference is −4.89 (
Table 6). Due to the functional and morphological similarity of Caco-2 cells to the human intestinal epithelium, the study of the permeability of compounds through the monolayer of Caco-2 cells is the most commonly used in vitro method to determine absorption of orally administered drugs [
44,
45].
The Caco-2 permeability calculated using the pkCSM algorithms is given as the logarithm of the apparent permeability coefficient (log Papp). Compounds for which the calculated log Papp at 10
−6 cm/s is greater than 0.9 are considered to have high Caco-2 permeability. Based on the results obtained for all tested 8-chloroquinobenzothiazines
1–
17 (
Table 6), it can be predicted that they will show high Caco-2 cell permeability (values ranged from 0.97 to 1.18). The calculated intestinal absorption values are given in percentages and indicate what percentage of the substance will be absorbed in the human intestine. The values obtained for the tested substances
1–
17 showed that all the compounds have a very high probability of high intestinal absorption; for all the compounds the value of this parameter was greater than 90 and close to the value obtained for chlorpromazine
18. Also, skin permeability is considered as an essential parameter to be considered when delivering active substances. It is also a parameter that makes it possible to determine the risk of using a given substance [
46,
47].
This parameter obtained from the pkCSM program is expressed as logKp and a compound is considered to have relatively low skin permeability if its logKp > −2.5. For all the tested substances, the obtained value of this parameter is in the range of −2.74 to −2.61 and indicates poor permeability (
Table 6).
A key parameter influencing the bioavailability of a biologically active substance is its solubility in water. Substances that are poorly soluble in body fluids are difficult to dissolve, which reduces their bioavailability. In this case, various strategies can be used to increase solubility and improve absorption (formulation modifications, the use of specific carriers) [
48].
The obtained values of the solubility parameter for all the new 17 compounds indicate their poor solubility, which results from their chemical structure. The obtained values of the parameters range from −6.30 for quinobenzothiazine
15 to −5.12 for quinobenzothiazine
4. For chlorpromazine, this parameter was −4.89 (
Table 6). The absorption parameters of the tested quinobenzothiazines were also evaluated for potential interactions with
p-glycoproteins. Almost all of them (except substances
8 and
10) can be substrates for
p-glycoprotein, and all of them can be inhibitors of
p-glycoprotein I and
p-glycoprotein II (
Table S3).
Using the pkCSM program, the unbound fraction (Fu) parameter was also calculated (
Table 7). It is also a pharmacokinetic parameter that affects the effectiveness of the drug and any side effects that may occur (glomerular filtration in the kidneys, total clearance hepatic metabolism). Determining this parameter is of great importance because the part of the drug that has not been bound to the molecular target may lead to interactions with other proteins, enzymes, and receptors [
49].
For the tested 6-substituted 8-chloroquinobenzothiazines 1–17, the value of this parameter is in the range of 0.02 for compound 9 to 0.16 for compound 17. For chlorpromazine used in the study as a reference substance, a value of 0.08 was obtained. The results indicate a low content of the unbound fraction in plasma. The obtained results indicate that the largest amount of unbound substance in plasma may remain in the case of compounds 11 and 17 containing p-toluenesulfonamide substituents.
For medicinal substances, it is also important to determine the degree to which they cross the blood–brain barrier. This parameter is given as logBB and is defined as the logarithmic ratio of the drug concentration in the brain to its concentration in the plasma. In the pkCSM calculation model, a substance can cross the blood–brain barrier if logBB is greater 0.3. If the obtained value of this parameter is less than −1, the substance is distributed to the brain to a small extent [
50,
51]. The calculated permeability through the blood–brain barrier for the tested substances
1–
17 ranges from −0.36 (for compound
16) to 0.58 (for compound
1). Values greater than 0.3 were obtained for substances
1–
5 having dialkylaminoalkyl substituents at the thiazine nitrogen atom, similarly to chlorpromazine
18 used as a reference (logBB = 0.89). According to the predictions obtained, the remaining substances will have poor permeability of the blood–brain barrier.
Permeability to the central nervous system is given as log PS. The value of this parameter obtained using the pkCSM program ranges from −2.16 for derivative
16 to −1.42 for derivative
1, so 8-chloroquinobenzothiazines
1–
9 and
11–
14 can penetrate the central nervous system. Substances with log PS > −2 are considered to penetrate the central nervous system, while substances with log PS < −3 do not (
Table 7,
Figure S1).
An important parameter influencing the metabolism of xenobiotics (phase I of metabolism) is their susceptibility to catalysis by cytochrome P450 (CYP) enzymes. If a substance is a substrate for this enzyme, it is converted into metabolites by binding to the active site of the enzyme; however, inhibitors may be their substrates or non-substrates. It is believed that the CYP3A, CYP1A2, CYP2C9, CYP2C19, and CYP2D6 isoforms of this enzyme, occurring primarily in the liver and intestinal wall, have a large share in drug metabolism. Predictions of the interactions of the tested quinobenzothiazines with these enzymes obtained from the pkCSM program gave varied results. All quinobenzothiazines can be CYP3A4 substrates, while only three (compounds
1,
3, and
4) are substrates for the CYP2D6 isoform). However, all the tested compounds may be CYP3A4 inhibitors and almost all (except compounds
11 and
17) may be CYP1A2 inhibitors (
Table S4). As is known, chlorpromazine is extensively metabolized in the liver and kidneys, and its metabolism is mainly due to cytochrome P450 isoenzymes: CYP2D6, CYP1A2, and CYP3A4.
One of the most important pharmacokinetic parameters to consider when designing drug candidates is the clearance rate. Clearance is divided into three general categories: metabolic conversion, renal excretion, and hepatobiliary excretion. The mechanism determining the clearance rate is determined by the physicochemical properties of the substance, with lipophilic molecules tending to be metabolized and hydrophilic, and polar molecules tending to be passively or actively excreted. This is one of the main parameters describing the elimination of a substance from the body, which means the volume of plasma from which substances are removed per unit of time. Using the pkCSM program, the total clearance (CLtot) can be calculated, which determines the efficiency of drug elimination from the entire body without indicating specific elimination mechanisms. This parameter is useful, among other things, for determining the dosing rate [
52,
53]. Among the 8-chloroquinobenzothiazines studied, CLtot values were diverse and ranged from 0.03 (for compound
17) to 0.77 mL/min/kg (for compound
3). The highest values, similar to the total clearance values for the chlorpromazine
18, were obtained for derivatives with dialkylaminoalkyl substituents.
For the tested 8-chloroquinobenzothiazines, the maximum tolerated dose, acute oral toxicity in rats, T. Pyriformis toxicity, and toxicity to fish were also calculated. The obtained results are summarized in
Table 8. The maximum recommended tolerated dose (MRTD) allows for the preliminary determination of the toxic dose of a given substance for humans. The obtained results of the MRTD parameter values are given as log (mg/kg/day). Obtaining this parameter facilitates the determination of the maximum recommended initial dose. According to the model used in the pkCSM program, if the calculated value is less than or equal to 0.48 log (mg/kg/day), it is considered low, and if it is higher, it is considered high. For most of the tested derivatives (
1–
5,
7–
10,
13–
16), the obtained results indicate a low maximum recommended dose. To determine in silico toxicity in the pkCSM program, a model based on T. Pyriformis, a protozoan bacterium, is used, the toxicity of which is considered a toxic endpoint. The value of this parameter is designated as plGC
50 (negative logarithm of the concentration required to inhibit 50% of growth in log µg/L). The tested compound can be assessed as toxic if the plGC
50 value is greater than −0.5 log µg/L. On the other hand, the toxicity parameter for roach is given as LC
50, i.e., the concentration of the substance necessary to cause death of 50% of the flathead roach. According to this model, a substance for which the LC
50 value is less than 0.5 mM (logLC
50 < −0.3) is considered highly toxic. Hepatotoxicity and no skin sensitization are predicted for all the tested substances (similar to chlorpromazine
18) (
Table S5).
Comparing the log PTLC data obtained with the results of cytotoxicity tests against various cell lines (A549, HCT-116, MiaPaCa-2), it was observed that compounds with moderate lipophilicity (log PTLC in the range of 4.6–5.2) showed the highest antitumor activity while maintaining high selectivity against normal cells (HaCaT). For example, compound 2 (log PTLC = 5.05) showed strong cytotoxicity against A549 (IC50 = 8.2 µM) and HCT-116 (IC50 = 1.6 µM) cells, achieving high selectivity indices of 7.6 and 39, respectively. A similar profile was shown by compound 9 (log PTLC = 4.66), which achieved the highest SI for the A549 line (SI = 10.7). Particularly outstanding was compound 16 (log PTLC = 4.90), which selectively and strongly inhibited the growth of HCT-116 cells (IC50 = 0.7 µM, SI = 143), while showing a lack of toxicity to non-cancer cells. In contrast, excessive lipophilicity, as in the case of compound 17 (log PTLC = 6.22), although correlated with good activity against A549 (IC50 = 9.45 µM), no longer led to equally favorable SI values against other lines. Compound 15 (log PTLC = 4.82), despite its low IC50 value (6.98 µM), had very low selectivity (SI = 0.1), indicating its potential non-selective toxicity.