Next Article in Journal
Reduced Element for Adaptive Finite Element Analysis of First-Order IVP with Built-in Error Estimator in Maximum Norm
Previous Article in Journal
A Comprehensive Approach for an Interpretable Diabetic Macular Edema Grading System Based on ConvUNext
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparative Analysis of Structural Analogs of Dipyridothiazines with m-Xylene and a Lutidine Moiety—In Silico, In Vitro, and Docking Studies

by
Emilia Martula
1,
Beata Morak-Młodawska
2,*,
Małgorzata Jeleń
2,
Paulina Strzyga-Łach
3,
Marta Struga
3,
Katarzyna Żurawska
4,
Anna Kasprzycka
4,5 and
Weronika Bagrowska
6
1
Doctoral School, The Medical University of Silesia, 40-055 Katowice, Poland
2
Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
3
Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
4
Biotechnology Centre, The Silesian University of Technology, Krzywoustego Street 8, 44-100 Gliwice, Poland
5
Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, The Silesian University of Technology, Krzywoustego Street 4, 44-100 Gliwice, Poland
6
Tunneling Group, Biotechnology Centre, The Silesian University of Technology, Krzywoustego Street 8, 44-100 Gliwice, Poland
*
Author to whom correspondence should be addressed.
Appl. Sci. 2024, 14(16), 7263; https://doi.org/10.3390/app14167263
Submission received: 28 July 2024 / Revised: 13 August 2024 / Accepted: 14 August 2024 / Published: 18 August 2024

Abstract

:
Dimers of dipyridothiazines with an m-xylene moiety are presented in terms of a comparative analysis with anticancer active structural analogs containing a lutidine system. The synthesis of new isomeric dimers was described, the structure of which was confirmed by 1H, 13C and 2D NMR, and HR MS spectroscopic methods. The preliminary prediction of the pharmacological profile using the Way2Drug server indicated the anticancer potential of the tested derivatives. In vitro biological activity tests were performed on a normal skin cell line (HaCaT) and five cancer cell lines, including human primary colon cancer (SW480), human metastatic colon cancer (SW620), human breast adenocarcinoma (MDA-MB-231), human lung carcinoma (A-549), and human glioblastoma (LN-229), which indicated low cytotoxic activity. In order to explain the surprisingly low activity, a comparative structural analysis of the tested analogs compared to the dimers with the lutidine system was performed using quantum mechanics and molecular docking in relation to histone deacetylase. Molecular docking indicated the different binding sites of the analyzed dimers, which explained the differences in the activity.

1. Introduction

Phenothiazines are completely synthetic heterocyclic systems that have no precursor or equivalent in the world of living nature [1,2]. Although they were discovered more than 140 years ago, to this day, the molecules are still very popular in the world of medical, pharmaceutical, as well as chemical research [3,4,5].
In the 1950s, phenothiazines containing dialkylaminoalkyl substituents in their structure triggered revolutionary changes in psychiatry by showing phenomenal antipsychotic activity to treat schizophrenia or manic states. To this day, these drugs are still used in medicine [1]. Scientific research in the area of phenothiazines is constantly being carried out, revealing many further interesting pharmacological activities, including anticancer [6,7,8,9,10], antimicrobial [11,12], antifungal [13], antiviral [14,15], antioxidant [16], or the reversal of multidrug resistance activities [17]. This topic is so rich that its results are published in numerous review papers each year around the world [18,19,20,21,22]. In 2006, English researcher Mitchell named the phenothiazine “parent molecule”, which fully reflects the application nature of this chemical compound [22].
The structure of phenothiazines is also modified at various levels, which leads to the creation of new, previously undescribed derivatives with interesting biological activities [23]. Such a group of modified phenothiazines includes dipyridothiazines having two pyridine rings in their structure instead of benzene rings [21]. Dipyridothiazines have a valuable anticancer effect against numerous cancers, such as melanoma, glioma, lung, colon, breast, and ovarian cancer, consisting of the induction of apoptosis through its mitochondrial activation and the possibility of partial DNA intercalation [21,24]. Dipyridothiazines also showed significant immunosuppressive and anti-inflammatory activity by inhibiting TNF alpha and interleukin 6 levels, as well as low cytotoxicity towards normal cells [21].
Dimeric derivatives of dipyridothiazines, in which two dipyridothiazine units were connected by selected organic linkers, such as o-xylene, p-xylene, and 2,6-dimethylpyridine (lutidine), have recently shown highly promising anticancer activities in relation to colon (SW480) and breast (MCF 7) cancer lines. Additionally, these compounds were characterized by their relatively low cytotoxicity in relation to normal myoblast (L6) cell lines [25]. Among dimeric dipyridothiazine derivatives, the highest anticancer potential was demonstrated by compounds that contained a linker in their structure, which was 2,6-dimethylpyridine, i.e., lutidine. Additionally, pyridine and its substituted derivatives are known as six-membered heterocycles with broad pharmacological activity profiles [26,27].
The idea of this work was to synthesize new dimeric derivatives of dipyridothiazines (1,6-, 1,8-, 2,7-, and 3,6-diazaphenothiazines 14) with an m-xylene linker (69) as structural analogs of the early described dimers with the lutidine moiety (6a9a) [25] (Scheme 1, Figure 1) in order to comparatively analyze their biological activity and determine the structure–activity relationship in the area of in vitro anticancer research.
The structure of the new compounds was clearly confirmed by the NMR, 2D NMR spectroscopy, and mass spectrometry HR MS. In silico analyses of probable molecular targets were performed using the Way2Drug server. The compounds were tested for anticancer activity in relation to the following cancer cell lines: human primary colon cancer (SW480), human metastatic colon cancer (SW620), human breast adenocarcinoma (MDA-MB-231), human lung carcinoma (A-549), and human glioblastoma (LN-229), and the human immortal keratinocyte cell line from adult human skin (HaCaT). To elucidate the structure–activity relationship, quantum mechanical calculations and a molecular docking study were performed against the selected histone deacetylase target.

2. Materials and Methods

2.1. Chemistry

The NMR spectra were recorded on Bruker Advance spectrometers (1H at 600 MHz, 13C at 150 MHz) in CDCl3 and DMSOd6. Two-dimensional COSY, ROESY, HSQC, and HMBC spectra of compound 8 were recorded on a Bruker Advance spectrometer at 600 MHz. All NMR analyses were performed at room temperature using the standard TopSpin 3.7.0 software. The HR MS spectra (ESI—Electro Spray Ionisation) were run on a Brucker Impact II. The samples were prepared as solutions in acetonitrile. Melting points were determined in open capillary tubes on a Boetius melting point apparatus. TLC was performed on silica gel 60 F254 (Merck 1.05735) with CHCl3-EtOH (10:1 v/v) and on Al2O3 60 F254 neutral (type E) (Merck 1.05581) with CHCl3-EtOH (10:1 v/v) as eluents.
The parent starting dipyridothiazines 14 (Scheme 1) and lutidine derivatives 6a9a were prepared according to reference [25].

General Procedure for Synthesis of Compounds (69)

To a solution of the selected diazaphenothiazine (14) (0.201 mg, 1 mmol) in dry DMF (10 mL), NaH (0.028 g, 1.2 mmol, 60% NaH in mineral oil was washed out with hexane) was added. The reaction mixture was stirred at room temperature for 1 h and then the linker α,α′-dichloro-m-xylene (0.5 mmol) was added, and the stirring was continued for 24 h. The progress of the reaction was monitored using TLC. The mixture was poured into water (15 mL), extracted with CHCl3 (3 × 10 mL), and dried using Na2SO4. The obtained product was purified by column chromatography (aluminum oxide, CHCl3-Ethanol 10:1) to give the following:
1,3-bis((10H-dipyrido [2,3-b:2′,3′-e][1,4]thiazin-10-yl)methyl)benzene (6)
(214 mg, 85%), mp = 268–270 °C
1H NMR (DMSOd6) δ: 5.16 (s, 4H, 2 CH2), 6.69 (dd, J = 8.4 Hz, J = 1.2 Hz, 2H), 6.78 (dd, J = 7.2 Hz, J = 4.8 Hz, 2H), 6.84 (dd, J = 8.4 Hz, J = 4.8 Hz, 2H), 6.93 (s, 1H, Hd-m-xylen), 7.18 (d, J = 7.2 Hz, 2H, Hb-m-xylen), 7.31 (t, J = 7.2 Hz, 1H, Hc-m-xylen), 7.37 (dd, J = 7.2 Hz, J = 1.2 Hz, 2H), 7.68 (dd, J = 4.8 Hz, J = 1.2 Hz, 2H), and 7.82 (dd, J = 4.8 Hz, J = 1.2 Hz, 2H). 13C NMR: 47.58, 115.14, 119.10, 121.95, 122.56, 124.59, 125.45, 129.28, 135.11, 137.16, 137.92, 142.85, 143.17, 145.41, and 151.89. HR MS (EI) m/z for [C28H21N6S2 + H], calc. 505.1264; found: 505.1283 (100%).
1,3-bis((10H-dipyrido [3,2-b:3′,4′-e][1,4]thiazin-10-yl)methyl)benzene (7)
(209 mg, 83%), mp = 262–264 °C
1H NMR (CDCl3) δ: 5.42 (s, 4H, 2 CH2), 6.90 (m, 2H), 7.17 (d, J = 6.0 Hz, 2H), 7.18 (d, J = 7.8 Hz, 2 Hb-m-xylen), 7.21 (m, 2H), 7.27 (m, 1H, Hc-m-xylen), 7.70 (s, 1H, Hd-m-xylen), 8.05 (m, 2H), 8.14 (d, J = 6.0 Hz, 2H), and 8.19 (s, 2H). 13C NMR: 48.02, 111.95, 120.20, 121.59, 125.52, 126.67, 128.23, 130.03, 134.95, 135.05, 140.91, 143.84, 147.14, 151.13, and 152.90. HR MS (EI) m/z for [C28H21N6S2 + H], calc. 505.1264; found: 505.1283 (100%).
1,3-bis((10H-dipyrido [3,4-b:3′,4′-e][1,4]thiazin-10-yl)methyl)benzene (8)
(216 mg, 86%), mp = 256–257 °C
1H NMR (CDCl3) δ: 5.06 (s, 4H, 2 CH2), 6.35 (d, J = 5.7 Hz, 2H, 2 H9), 6.94 (s, 1H, Hd-m-xylen), 6.97 (d, J = 4.8 Hz, 2H, 2 H4), 7.25 (d, J = 7.2 Hz, 2H, 2 Hb-m-xylen), 7.45 (t, J = 7.2 Hz, 1H, Hc-m-xylen), 7.70 (s, 2H, 2 H1), 7.96 (d, J = 5.7 Hz, 2H, 2 H8), 8.01 (s, 2H, 2 H6), and 8.10 (d, J = 4.8 Hz, 2H, 2 H3). 13C NMR: 51.08 CCH2, 109.41 C9, 117.19 C9a, 121.56 Cd, 123.48 C4, 126. 06 Cb, 130.79 Cc, 133.35 C10a, 134.46 Ca, 135.56 C1, 137.22 C4a, 143.44 C6, 145.70 C3, 147.14 C8, and 151.47 C5a. HR MS (EI) m/z for [C28H21N6S2 + H], calc. 505.1264; found: 505.1229 (100%).
1,3-bis((10H-dipyrido [2,3-b:4′,3′-e][1,4]thiazin-5-yl)methyl)benzene (9)
(199 mg, 79%), mp = 288–290 °C
1H NMR (DMSOd6) δ: 5.04 (s, 4H, 2 CH2), 6.44 (d, J = 5.4 Hz, 2H), 6.81 (m, 2H), 6.88 (m, 2H), 6.97 (s, 1H, Hd-m-xylen), 7.27 (d, J = 7.2 Hz, 2H, Hb-m-xylen), 7.44 (t, J = 7.2 Hz, 1H, Hc-m-xylen), 7.90 (d, J = 4.2 Hz, 2H), 7.95 (d, J = 5.4 Hz, 2H), and 8.02 (s, 2H). 13C NMR (CDCl3): 50.10, 110.06, 117.65, 121.69, 128.61, 129.01, 131.28, 134.04, 136.33, 137.99, 145.02, 146.85, 149.14, and 149.63. HR MS (EI) m/z for [C28H21N6S2 + H], calc. 505.1264; found: 505.1299 (100%).

2.2. In Silico Target Prediction

The prediction of the biological targets and cytotoxicity towards the cancer cell lines was performed using the Way2Drug server [28,29]. This server analyzes the biological activity of an organic drug based on molecular recognition against its database.

2.3. Biological Evaluation

2.3.1. Cell Line and Culture

Human primary colon cancer (SW480) and metastatic colon cancer (SW620), human breast adenocarcinoma (MDA-MB-231), human lung carcinoma (A-549), human glioblastoma (LN-229), and human immortal keratinocyte (HaCaT) cell lines were obtained from the American Type Culture Collection (ATCC) in Rockville, MD, USA. The following cell lines were cultured in the recommended medium according to ATCC protocols: SW480 and SW620 in Minimum Essential Medium (MEM), and MDA-MB-231, A-549, LN-229, and HaCaT cells in Dulbecco’s Modified Eagle Medium (DMEM). The culture medium was supplemented with 10% fetal bovine serum (FBS), penicillin (100 U/mL), and streptomycin (100 μg/mL). Cells were maintained in a humidified incubator at 37 °C with 5% CO2. Passaging was performed when the cells reached 80–90% confluence using 0.25% trypsin (Gibco Life Technologies, Gibco, Grand, Island, NY, USA) treatment before being utilized for the experiments.

2.3.2. MTT Cell Viability Assay

Cell viability was determined through the enzymatic conversion of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide salt (MTT) to insoluble formazan crystals by the mitochondrial dehydrogenases present in the viable cells [30]. Cells were seeded into 96-well plates at a density of 1 × 104 cells per well and allowed to adhere for 24 h at 37 °C in a CO2 incubator. Following the 24 h incubation period, the culture medium was replaced with fresh medium, and the cells were exposed to varying concentrations of the compounds (ranging from 10 to 100 μM). Then, they were further incubated for 72 h at 37 °C in a CO2 incubator. Untreated cells served as controls. Subsequently, MTT solution (0.5 mg/mL in serum-free medium) was added, and the samples were incubated for 4 h at 37 °C in a CO2 incubator. Following this, the medium was removed, and formazan crystals were solubilized by adding an isopropanol-DMSO mixture (1:1). The absorbance of the dissolved crystals was measured at a wavelength of 570 nm using a UVM 340 reader (ASYS Hitech GmbH, Eugendorf, Austria). The IC50 values were calculated using GraphPad Prism 8 software (GraphPad Software).

2.4. The Statistical Analysis

The statistical analysis was performed using GraphPad Prism 9 [31] software (Graph Pad Software, San Diego, CA, USA). The results were reported as mean ± SD from a minimum of three independent experiments. Statistical significance of the differences between the values was assessed using an analysis of variance with Dunnett’s multiple comparison post hoc test, and significance was defined as p < 0.05.

2.5. Quantum Mechanical Calculations

Electron density and bond length calculations were performed using the standard B3LYP functional set and the 6-31+G(d,p) basis set, using the Gaussian16 program [32], and visualized using the widely used Avogadro program [33].

2.6. Molecular Docking Analysis

  • Protein preparation
The HDAC4 histone deacetylase structure (PDB ID: 2VQJ) was used for docking. The HDAC4 structure was crystallized with the inhibitor, which was manually removed from the structure. The protonation of the enzyme was performed with an H++ server at a pH of 7.5 [34,35].
  • Ligand preparation
The structures of two ligands, 8 and 8a, were created using MarvinSchetch 19.22.0 software. File conversion, including Gasteiger charge assignment, was performed using the OpenBabel 2.3.2 tool [36].
  • Molecular Docking
The first docking was carried out using the DiffDock software Version 1.1.2 [37]. Blind docking on default settings was used. Docking was also performed using AutoDock Vina software Version 4.2.6 [38,39] implemented on the TAMARIND BIO server. The docking site was defined as the inhibitor binding pocket in the crystallographic structure. This pocket was also detected during the blind docking using DiffDock software Version 1.1.2, during the analysis of the selected ligands.

3. Results and Discussion

3.1. Chemistry Part

The preparation of new dipyridothiazine dimers was carried out using previously synthesized, selected dipyridothiazines (10H-1,6-, 1,8-, 2,7-, and 3,6-diazaphenothiazines 14), for which the promising anticancer activities were determined [25]. In the next step, selected dipyridothiazines were subjected to alkylation reactions with α,α′-dichloro-m-xylene. The reactions were carried out in anhydrous DMF in the presence of the strong base NaH at room temperature, which led to the formation of the final dimers 69 (Scheme 1). The obtained dimers were the structural analogs of the dimers with the lutidine moiety, which do not have the nitrogen atom characteristic of the lutidine system. The progress of the reaction was monitored using thin-layer chromatography (TLC). The crude solid reaction products were purified using column chromatography. All of the new compounds were obtained in good yields (79–86%).

3.2. Structural Identification Using Spectroscopic Methods

The identification of the structure of new organic compounds is a fundamental problem both in organic chemistry and in the drug design process [40,41]. Due to the fact that chemical reactions may be accompanied by rearrangements or parallel or subsequent reactions, proving the structure of the obtained product is essential. For this purpose, advanced NMR spectroscopic techniques and high-resolution HR MS mass spectrometry were used. The identification of the molecule’s structure was carried out on the basis of the following 1H, 13C NMR spectra, and 2D NMR correlations: COSY (COrrelation SpectroscopY), ROESY (Rotating frame nuclear Overhauser Effect SpectroscopY), HSQC (Heteronuclear Single Quantum Correlation), HMBC (Heteronuclear Multiple Bond Correlation), and HR MS mass spectrometry. The full structural analysis performed for dimer 8 allowed for the assignment of individual hydrogen and carbon atoms in the molecules. The NMR spectra confirmed the correct number of both protons and carbon atoms in the new compounds. The molecular mass and purity of the compounds were confirmed by high-resolution mass spectrometry (HR MS). All of the spectra of compounds 69 are included in Supplementary Materials File S1.

3.3. In Silico Target Prediction

The new dimers 69 were analyzed in silico using the Way2Drug server, which includes the PASS (Prediction of Activity Spectra for Substances) application, which allows for the prediction of the potential biological activities of the molecules and the probability of cytotoxicity toward cancer cell lines [28,29]. The obtained results are summarized in Table 1 and Table 2.
The results show that the new dimers 69 have a high probability of biological action aimed at the anticancer potential related to the mechanism of histone deacetylase stimulation, the inhibition of glycosylphosphatidylinositol phospholipase D, which may directly lead to the activation of the mitochondrial apoptosis pathway. It is worth noting that the anticancer potentials of dipyridothiazines 14 were also experimentally confirmed in our earlier studies, which was the beginning of the search for new derivatives in the group of dipyridothiazines with even better activities [21,24]. Moreover, previous in silico analyses performed for dimers with the lutidine, o-xylene, and p-xylene systems also indicated a high potential for anticancer activity, which was confirmed [25].
Furthermore, the PASS program indicated the likelihood of dimer activity in neurodegenerative diseases, antipsychotic and antiallergic effects, and the inhibition of cytochrome 450.
More importantly, in addition to the probable mechanisms of biological action, the PASS program indicated, for the new dimeric derivatives, the likelihood of cytotoxic effects on breast, kidney, colon, pancreatic, and ovarian cancer cell lines, melanoma, and non-small-cell lung cancer. It is worth pointing out that previous studies on dipyridothiazines 14 confirmed this promising anticancer potential [21], which was an inspiration to conduct further cytotoxicity tests of the new dimers 69.

3.4. Biological Part—Analysis of Cytotoxicity towards Cancer and Normal Cells

Taking into account the results of the in silico simulations and the previously promising anticancer activities and low cytotoxicity of the obtained dipirydothiazines 14 [21] and dimers with a lutidine linker 6a9a [25], the new derivatives 69 were tested for their anticancer activity. The cytotoxicity and antiproliferative activity of new dimers were determined in vitro in relation to the following five cancer cell lines: human primary colon cancer (SW480), human metastatic colon cancer (SW620), human breast adenocarcinoma (MDA-MB-231), human lung carcinoma (A-549), and human glioblastoma (LN-229), and to the normal line human immortal keratinocyte cell line from adult human skin (HaCaT) using the MTT assay [30]. The study used doxorubicin and cisplatin as reference drugs. The data are expressed as the mean SD, IC50 (µM)—the half-maximal growth inhibitory concentration after culturing the cells for 72 h with the individual compound—and the SI (Selectivity Index), which was calculated using the following formula: SI = IC50 for normal cell line/IC50 cancer cell line. The results are summarized in Table 3, and show that compounds 69 were not toxic to the normal keratinocyte HaCaT cell line in the range of the tested concentration. The analysis of the anticancer activity showed that the tested derivatives were mostly inactive (IC50 > 100 µM). Dimer 6 had poor activity in relation to SW480, MDA-MB-231, and A-549 cell lines (IC50 = 81–92 µM). Dimer 7, which contains two 1,8-dipyridothiazine units in its structure, was the most active in the group of tested compounds in relation to the MDA-MB-231 cell line (IC50 = 67.3 µM). The results of the biological assay were very surprising when analyzed in relation to the previously obtained results of the anticancer activity of structural analogs 6a9a, as shown in Figure 1 [25].
Lutidine derivatives 6a9a showed activity in the range of IC50 = 0.1–11 µM towards MCF 7 and IC50 = 4.5–25.9 µM towards the SW480 cell lines [25]. This led us to consider the reason for the deactivation of the obtained dimers 69. To explain this phenomenon, quantum chemical calculations and preliminary molecular docking analyses were used.

3.5. Quantum Mechanical Calculations and Molecular Docking Study

In order to understand and explain the differences in the activity and inactivity of the dimers with an m-xylene and lutidine linker, measurements of the electron density of the selected dimer 8 containing 2,7-diazaphenothiazine in its structure and its analog with the lutidine 8a system were performed using the Gaussian program [32]. Dimer 8 was selected for quantum mechanical studies and molecular docking because it was the only one that showed inactivity against all of the tested cancer cell lines (IC50 > 100 µM). A visualization of the obtained results is presented in Figure 2 and Figure 3 using the Avogadro program [33]. These measurements show that the presence of the lutidine system in dimer 8a causes a differentiation of the electron charges on the thiazine nitrogen atom in 1,4-thiazine rings directly connected to the CH2 moiety (Figure 3). Additionally, the lutidine system contains a basic nitrogen atom with a lone electron pair, which causes the entire molecule to be enriched with electrons. This situation was not observed in dimer 8 with the m-xylene linker (Figure 2).
Additionally, bond length measurements were made, with particular attention paid to the connecting linkers. In the dimer 8a containing lutidine, there are CH2-Clutidine bonds with a length of 1.516 Å and a characteristic Clutidine-Nlutidine bond with a length of 1.362 Å. These bonds are essentially shorter than the analogous bonds found in dimer 8 (CH2-Cm-xylene with a length of 1.521 Å; Cm-xylene-Cm-xylene bond with a length of 1.405 Å) (Supplementary Materials File S1). The structural analysis carried out using quantum mechanical calculations to some extent explains the differences in the structure of isomeric dimers 8 and 8a, which had a direct impact on the affinity for protein molecular targets.
In the next step, in order to more deeply explain the weak activity of the dimers with the m-xylene system, preliminary molecular docking studies of dimer 8 and its lutidine derivative 8a were performed in relation to the histone deacetylase (HDAC4), indicated as a molecular target by the PASS program. The selected histone deacetylase (HDAC4) and the dimers 8 and 8a were prepared for docking according to the literature data (see Section 2.6) [34,35,36]. Two types of docking were performed using two kinds of software. The first one was performed using the DiffDock software Version 1.1.2 [37] with the default settings. The second docking was also performed using the AutoDock Vina software Version 4.2.6 [39], implemented on the TAMARIND BIO server, where the docking site was defined as the inhibitor-binding pocket in the crystallographic structure. This pocket was also detected during blind docking using DiffDock software when analyzing the selected ligands. The docking results are summarized in Table 4, which presents the confidence values of the DiffDock software, as well as the binding affinity calculated by the AutoDock software. The results indicate that both dimers 8 and 8a dock with a similar strength.
In a further stage of the docking study, an interaction analysis was performed for the best position from each complex using the Protein Ligand Interaction Profiler (PLIP) server [39] (Table 5).
The analysis showed that the tested dimers 8 and 8a bind in different parts of the pocket, depending on the software used. Both docking programs indicated that dimer 8 had fewer binding sites than dimer 8a.
In the complexes obtained using the DiffDock server, the ligands bind near the zinc atom, Phe168 and His198 amino acids, while the complexes obtained using the AutoDock software in the left part of the pocket also interact with Phe168 (Figure 4).
The obtained results were also compared with the position of the trifluoromethyl ketone inhibitor in the structure of the tested protein. This inhibitor binds to the pocket into which test compounds 8 and 8a were docked using DiffDock software (Figure 5).
Analyzing the interactions of both dimers in this binding pocket, it can be noticed that dimer 8 is located at a considerable distance from the zinc atom and interacts only with Phe168 and His198 (Figure 6).
Meanwhile, the 8a dimer, having a lutidine system in its structure, is located closer to the zinc atom compared to its analog 8, and has four interactions with the following amino acids: Phe168, Phe277, His153, and His198 (Figure 7).
The performed quantum mechanical studies and preliminary molecular docking in relation to histone deacetylase explain the weak anticancer activity of the tested compounds compared to previously obtained active dimers with a lutidine linker.

4. Conclusions

The synthesis of dipyridothiazine dimers with an m-xylene moiety is presented in the light of a comparative analysis with their anticancer active analogs containing a lutidine linker in their structure. The structure of the new compounds was clearly confirmed by the NMR and HRMS spectroscopic techniques. The compounds were subjected to initial in silico screening using the Way2Drug server to determine the molecular targets and the likelihood of cytotoxicity against various types of cancer cells. In the next stage, cytotoxicity tests were performed against normal cell lines (HaCaT) and five cancer cell lines (SW480, SW620, MDA-MB-231, A-549, and LN-229), which showed weak anticancer activity and low cytotoxicity compared to the previously described structural analogs with the lutidine system. Quantum mechanical calculations of the electron density and bond length were performed to elucidate the structural differences that directly affected the conformation of the molecule, as well as interactions with the molecular targets. In addition, preliminary molecular docking studies were performed on histone deacetylase, showing different binding sites to the selected molecular target. Quantum mechanical calculations and molecular docking explained the anticancer activity research results to some extent.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/app14167263/s1, File S1: Supporting materials on spectral analysis of all compounds and the results of quantum mechanical calculations are included.

Author Contributions

Conceptualization, B.M.-M. and E.M.; methodology, E.M., P.S.-Ł., M.S., K.Ż., A.K. and W.B.; software, E.M., B.M.-M., M.J., P.S.-Ł., M.S., K.Ż., A.K. and W.B.; validation, E.M.; formal analysis, E.M., B.M.-M., M.J., M.J., P.S.-Ł., M.S., K.Ż., A.K. and W.B.; investigation, E.M. and B.M.-M.; resources, E.M., B.M.-M., M.J., P.S.-Ł., M.S., K.Ż., A.K. and W.B.; data curation, E.M. and B.M.-M.; writing—original draft preparation, E.M. and B.M.-M.; writing—review and editing, B.M.-M., E.M. and M.J.; visualization, B.M.-M.; supervision, E.M. and B.M.-M.; project administration, B.M.-M.; funding acquisition, E.M. and B.M.-M. All authors have read and agreed to the published version of the manuscript.

Funding

Emilia Martula was supported by a research subsidy from the Medical University of Silesia in Katowice (grant BNW-2-015/N/3/F, BNW-2-051/K/4/F); A.P.C. was funded by the Metropolis of Upper Silesia and Zagłebie Basin as part of the implementation of the project entitled “Support for scientific activity of doctoral students and employees at the doctoral level of the Silesian Medical University in Katowice”, which was implemented within the framework of the Metropolitan Science Support Fund program for 2022–2024 (grant agreement nos. RW/27/2024 and PCTT/621/2024).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. López-Muñoz, F.; Alamo, C.; Cuenca, E.; Shen, W.W.; Clervoy, P.; Rubio, G. History of the Discovery and Clinical Introduction of Chlorpromazine. Ann. Clin. Psychiatry 2005, 17, 113–135. [Google Scholar] [CrossRef]
  2. Jaszczyszyn, A.; Gąsiorowski, K.; Świątek, P.; Malinka, W.; Cieślik-Boczula, K.; Petrus, J.; Czarnik-Matusewicz, B. Chemical structure of phenothiazines and their biological activity. Pharmacol. Rep. 2012, 64, 16–23. [Google Scholar] [CrossRef]
  3. Otręba, M.; Stojko, J.; Rzepecka-Stojko, A. Phenothiazine derivatives and their impact on the necroptosis and necrosis processes. A review. Toxicology 2023, 492, 153528. [Google Scholar] [CrossRef] [PubMed]
  4. Kumar, A.; Vigato, C.; Boschi, D.; Lolli, M.L.; Kumar, D. Phenothiazines as anti-cancer agents: SAR overview and synthetic strategies. Eur. J. Med. Chem. 2023, 254, 115337. [Google Scholar] [CrossRef] [PubMed]
  5. Swoboda, D.; Nycz, J.E.; Karaush-Karmazin, N.; Minaev, B.; Książek, M.; Kusz, J.; Podsiadły, R. Synthesis and Spectroscopic Characterization of Selected Phenothiazines and Phenazines Rationalized Based on DFT Calculation. Molecules 2022, 27, 7519. [Google Scholar] [CrossRef]
  6. Duarte, D.; Vale, N. Antipsychotic Drug Fluphenazine against Human Cancer Cells. Biomolecules 2022, 12, 1360. [Google Scholar] [CrossRef]
  7. Otręba, M.; Stojko, J.; Rzepecka-Stojko, A. The role of phenothiazine derivatives in autophagy regulation: A systematic review. J. Appl. Toxicol. 2023, 43, 474–489. [Google Scholar] [CrossRef]
  8. Lopes, R.M.; Souza, A.C.S.; Otręba, M.; Rzepecka-Stojko, A.; Tersariol, I.L.S.; Rodrigues, T. Targeting autophagy by antipsychotic phenothiazines: Potential drug repurposing for cancer therapy. Biochem. Pharmacol. 2024, 222, 116075. [Google Scholar] [CrossRef]
  9. Xu, F.; Xi, H.; Liao, M.; Zhang, Y.; Ma, H.; Wu, M.; Xue, Q.; Sun, H.; Zhang, Y.; Xia, Y. Repurposed antipsychotic chlorpromazine inhibits colorectal cancer and pulmonary metastasis by inducing G2/M cell cycle arrest, apoptosis, and autophagy. Cancer Chemother. Pharmacol. 2022, 89, 331–346. [Google Scholar] [CrossRef]
  10. Li, A.; Chen, X.; Jing, Z.; Chen, J. Trifluoperazine induces cellular apoptosis by inhibiting autophagy and targeting NUPR1 in multiple myeloma. FEBS Open Bio. 2020, 10, 2097–2106. [Google Scholar] [CrossRef]
  11. Posso, M.C.; Domingues, F.C.; Ferreira, S.; Silvestre, S. Development of Phenothiazine Hybrids with Potential Medicinal Interest: A Review. Molecules 2022, 27, 276. [Google Scholar] [CrossRef]
  12. Rineh, A.; Dolla, N.K.; Ball, A.R.; Magana, M.; Bremner, J.B.; Hamblin, M.R.; Tegos, G.P.; Kelso, M.J. Attaching the NorA Efflux Pump Inhibitor INF55 to Methylene Blue Enhances Antimicrobial Photodynamic Inactivation of Methicillin-Resistant Staphylococcus aureus in Vitro and in Vivo. ACS Infect. Dis. 2017, 3, 756–766. [Google Scholar] [CrossRef]
  13. Brilhante, R.S.N.; Gotay, W.J.P.; Pereira, V.S.; de Oliveira, J.S.; Pereira-Neto, W.A.; Castelo-Branco, D.d.S.C.M.; de Aguiar Cordeiro, R.; Sidrim, J.J.C.; Rocha, M.F.G. Antifungal Activity of Promethazine and Chlorpromazine Against Planktonic Cells and Biofilms of Cryptococcus neoformans/Cryptococcus gattii Complex Species. Med. Mycol. 2020, 58, 906–912. [Google Scholar] [CrossRef]
  14. Piccini, L.E.; Castilla, V.; Damonte, E.B. Inhibition of dengue virus infection by trifluoperazine. Arch. Virol. 2022, 167, 2203–2212. [Google Scholar] [CrossRef]
  15. Simanjuntak, Y.; Liang, J.J.; Lee, Y.L.; Lin, Y.L. Repurposing of prochlorperazine for use against dengue virus infection. J. Infect. Dis. 2015, 211, 394–404. [Google Scholar] [CrossRef]
  16. Egbujor, M.C.; Tucci, P.; Buttari, B.; Nwobodo, D.C.; Marini, P.; Saso, L. Phenothiazines: Nrf2 activation and antioxidant effects. J. Biochem. Mol. Toxicol. 2024, 38, e23661. [Google Scholar] [CrossRef] [PubMed]
  17. Takács, D.; Csonka, Á.; Horváth, Á.; Windt, T.; Gajdács, M.; Riedl, Z.; Hajós, G.; Amaral, L.; Molnár, J.; Spengler, G. Reversal of ABCB1-related Multidrug Resistance of Colonic Adenocarcinoma Cells by Phenothiazines. Anticancer Res. 2015, 35, 3245–3251. [Google Scholar]
  18. González-González, A.; Vazquez-Jimenez, L.K.; Paz-González, A.D.; Bolognesi, M.L.; Rivera, G. Recent Advances in the Medicinal Chemistry of Phenothiazines, New Anticancer and Antiprotozoal Agents. Curr. Med. Chem. 2021, 28, 7910–7936. [Google Scholar] [CrossRef]
  19. Ohlow, M.J.; Moosmann, B. Phenothiazine: The seven lives of pharmacology’s first lead structure. Drug Discov. Today 2011, 16, 119–131. [Google Scholar] [CrossRef] [PubMed]
  20. Pluta, K.; Morak-Młodawska, B.; Jeleń, M. Synthesis and properties of diaza-, triaza- and tetraazaphenothiazines. J. Heterocycl. Chem. 2009, 46, 355–391. [Google Scholar] [CrossRef]
  21. Morak-Młodawska, B.; Jeleń, M.; Pluta, K. Phenothiazines Modified with the Pyridine Ring as Promising Anticancer Agents. Life 2021, 11, 206. [Google Scholar] [CrossRef]
  22. Mitchell, S.C. Phenothiazine: The parent molecule. Curr. Drug Targets 2006, 7, 1181–1189. [Google Scholar] [CrossRef]
  23. Marraffa, J.M. Phenothiazines. Encycl. Toxicol. (Fourth Ed.) 2024, 7, 539–542. [Google Scholar] [CrossRef]
  24. Zhang, J.; Ming, C.; Zhang, W.; Okechukwu, P.N.; Morak-Młodawska, B.; Pluta, K.; Jeleń, M.; Akim, A.M.; Ang, K.P.; Ooi, K.K. 10H-3,6-Diazaphenothiazines induce G2/M phase cell cycle arrest, caspase-dependent apoptosis and inhibits cell invasion of A2780 ovarian carcinoma cells through regulation on NF-κB and [BIRC6-XIAP] complexes. Drug Des. Dev. Ther. 2017, 11, 3045–3063. [Google Scholar] [CrossRef] [PubMed]
  25. Martula, E.; Morak-Młodawska, B.; Jeleń, M.; Okechukwu, N.P.; Balachandran, A.; Tehirunavukarasu, P.; Anamalay, K.; Ulaganathan, V. Synthesis and structural characterization of novel dimers of dipyridothiazine as promising antiproliferative agents. Molecules 2023, 28, 7662. [Google Scholar] [CrossRef]
  26. Li, J.J. Heterocyclic Chemistry in Drug Discovery; Wiley: Hoboken, NJ, USA, 2013; ISBN 978-1118148907. [Google Scholar]
  27. Singh, D.; Shewale, D.J.; Sengupta, A.; Soppina, V.; Kanvah, S. Lutidine deriva-tives for live-cell imaging of the mitochondria and endoplasmic reticulum. Org. Biomol. Chem. 2022, 20, 7047–7055. [Google Scholar] [CrossRef] [PubMed]
  28. Way2Drug Understanding Chemical-Biological Interactions, Predictive Services. Available online: www.way2drug.com/passonline/index.php (accessed on 18 December 2023).
  29. Druzhilovskiy, D.S.; Rudik, A.V.; Filimonov, D.A.; Gloriozova, T.A.; Lagunin, A.A.; Dmitriev, A.V.; Pogodin, P.V.; Dubovskaya, V.I.; Ivanov, S.M.; Tarasova, O.A.; et al. Computational platform Way2Drug: From the prediction of biological activity to drug repurposing. Russ. Chem. Bull. 2017, 66, 1832–1841. [Google Scholar] [CrossRef]
  30. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  31. GraphPad Prism. Available online: https://www.graphpad.com/updates/prism-900-release-notes (accessed on 15 February 2024).
  32. Gaussian 16; Revision C.01; Gaussian, Inc.: Wallingford, CT, USA, 2019.
  33. Avogadro: An Open-Source Molecular Builder and Visualization Tool. Version 1.2.0. Available online: http://avogadro.cc/ (accessed on 12 March 2024).
  34. Zhang, X.; Green, T.J.; Tsao, J.; Qiu, S.; Luo, M. Role of Intermolecular Interactions of Vesicular Stomatitis Virus Nucleoprotein in RNA Encapsidation. J. Virol. 2008, 82, 674–682. [Google Scholar] [CrossRef]
  35. Gordon, J.C.; Myers, J.B.; Folta, T.; Shoja, V.; Heath, L.S.; Onufriev, A. H++: A server for estimating pKas and adding missing hydrogens to macromolecules. Nucleic Acids Res. 2005, 33, W368–W371. [Google Scholar] [CrossRef]
  36. O’Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open Babel: An open chemical toolbox. J. Cheminform. 2011, 3, 33. [Google Scholar] [CrossRef] [PubMed]
  37. Corso, G.; Deng, A.; Fry, B.; Polizzi, N.; Barzilay, R.; Jaakkola, T. Deep Confident Steps to New Pockets: Strategies for Docking Generalization. Published as a conference paper at ICLR. arXiv 2024, arXiv:2402.18396. [Google Scholar] [CrossRef]
  38. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [PubMed]
  39. Salentin, S.; Schreiber, S.; Haupt, V.J.; Adasme, M.F.; Schroeder, M. PLIP: Fully automated protein–ligand interaction profiler. Nucleic Acids Res. 2015, 43, W443–W447. [Google Scholar] [CrossRef]
  40. Webster, F.X.; Kiemle, D.J.; Silverstein, R.M.; Bryce, D.L. Spectrometric Identification of Organic Compounds, 8th ed.; John Wiley & Sons Inc.: Hoboken, NJ, USA, 2014; ISBN 978-0-470-61637-6. [Google Scholar]
  41. Hermann, C.K.F.; Morrill, T.C.; Shriner, R.L.; Fuson, R.C. Systematic Identification of Organic Compounds, 9th ed.; Wiley: Hoboken, NJ, USA, 2023; ISBN 9781119799665. [Google Scholar]
Scheme 1. Route of the synthesis of isomeric dipyridothiazine dimers 69.
Scheme 1. Route of the synthesis of isomeric dipyridothiazine dimers 69.
Applsci 14 07263 sch001
Figure 1. Structure of the investigated dimers with the m-xylene linker (69) and lutidine moiety (6a9a).
Figure 1. Structure of the investigated dimers with the m-xylene linker (69) and lutidine moiety (6a9a).
Applsci 14 07263 g001
Figure 2. Visualization of the electron density of compound 8 (electron density was calculated in the Gaussian program, visualized in the Avogadro program). Electron density is represented by a color gradient, with red representing a higher electron density and dark blue representing a lower electron density.
Figure 2. Visualization of the electron density of compound 8 (electron density was calculated in the Gaussian program, visualized in the Avogadro program). Electron density is represented by a color gradient, with red representing a higher electron density and dark blue representing a lower electron density.
Applsci 14 07263 g002
Figure 3. Visualization of the electron density of the structural analog 8a with a lutidine system as a linker (electron density was calculated in the Gaussian program, visualized in the Avogadro program). Electron density is represented by a color gradient, with red representing a higher electron density and dark blue representing a lower electron density.
Figure 3. Visualization of the electron density of the structural analog 8a with a lutidine system as a linker (electron density was calculated in the Gaussian program, visualized in the Avogadro program). Electron density is represented by a color gradient, with red representing a higher electron density and dark blue representing a lower electron density.
Applsci 14 07263 g003
Figure 4. Picture of the pockets occupied by docked compounds 8 and 8a. The light-pink color indicates the pocket where compounds are docked with DiffDock and the light-blue color indicates the pocket where compounds are docked with AutoDock Vina. The orange sticks indicate the following amino acids: Phe168 and His198.
Figure 4. Picture of the pockets occupied by docked compounds 8 and 8a. The light-pink color indicates the pocket where compounds are docked with DiffDock and the light-blue color indicates the pocket where compounds are docked with AutoDock Vina. The orange sticks indicate the following amino acids: Phe168 and His198.
Applsci 14 07263 g004
Figure 5. Position of the trifluoromethylketone inhibitor in the ligand-binding pocket obtained using DiffDock docking. The dark-blue sphere indicates the zinc atom. Orange sticks indicate the following amino acids: Phe168 and His198.
Figure 5. Position of the trifluoromethylketone inhibitor in the ligand-binding pocket obtained using DiffDock docking. The dark-blue sphere indicates the zinc atom. Orange sticks indicate the following amino acids: Phe168 and His198.
Applsci 14 07263 g005
Figure 6. Position of dimer 8 in the binding pocket obtained using DiffDock docking and the interaction with amino acids, which are indicated by black dashed lines. The dark-blue sphere indicates the zinc atom. The orange sticks indicate the following amino acids: Phe168 and His198.
Figure 6. Position of dimer 8 in the binding pocket obtained using DiffDock docking and the interaction with amino acids, which are indicated by black dashed lines. The dark-blue sphere indicates the zinc atom. The orange sticks indicate the following amino acids: Phe168 and His198.
Applsci 14 07263 g006
Figure 7. Position of dimer 8a in the binding pocket obtained using DiffDock docking and the interaction with amino acids, which are indicated by black dashed lines. The dark-blue sphere indicates the zinc atom. The orange sticks indicate the following amino acids: Phe168, Phe277, His153, and His198.
Figure 7. Position of dimer 8a in the binding pocket obtained using DiffDock docking and the interaction with amino acids, which are indicated by black dashed lines. The dark-blue sphere indicates the zinc atom. The orange sticks indicate the following amino acids: Phe168, Phe277, His153, and His198.
Applsci 14 07263 g007
Table 1. Probability (%) of activity spectra of dimers (69) using the PASS program.
Table 1. Probability (%) of activity spectra of dimers (69) using the PASS program.
No.Probability of Activity Spectrum
6(28%)
Histone deacetylase stimulant
(77%) Glycosylphosphatidylinositol phospholipase D inhibitor(47%) Transcription factor inhibitor(33%) Alzheimer’s disease treatment(46%) Antiallergic
7(29%)
Mitochondrial processing peptidase inhibitor
(83%) Glycosylphosphatidylinositol phospholipase D inhibitor(18%) Antipsychotic(30%) Cytochrome P450 inhibitor(10%) MAP3K8 inhibitor
8(75%)
Neurodegenerative diseases treatment
(84%) Glycosylphosphatidylinositol phospholipase D inhibitor(65%)
Histone deacetylase stimulant
(42%) Alzheimer’s disease treatment(41%) Cytochrome P450 inhibitor
9(76%) Histone deacetylase stimulant(66%) Antiallergic(60%) Neurodegenerative diseases treatment(58%) Alzheimer’s disease treatment(59%) Antiasthmatic
Table 2. The probable cytotoxic effect (%) on various cancer cell lines of dimers (69).
Table 2. The probable cytotoxic effect (%) on various cancer cell lines of dimers (69).
No.Probability of Cytotoxicity towards Cancer Cell Lines
6Breast cancer MDA-MB-468 (62%)Melanoma
UACC-257 (51%)
Non-small-cell lung carcinoma NCI-H322M (49%)Colon carcinoma HCT-116 (42%)Pancreatic carcinoma YAPC (43%)
7Breast cancer MDA-MB-468 (59%)Melanoma
UACC-257 (55%)
Renal carcinoma 786 (45%)Colon carcinoma HCT-116 (39%)Pancreatic carcinoma YAPC (44%)
8Breast cancer MDA-MB-468 (39%)Melanoma
UACC-257 (54%)
Renal carcinoma 786-0 (49%)Colon carcinoma HCT-116 (55%)Ovarian adenocarcinoma OVCAR-4 (43%)
9Breast cancer MDA-MB-468 (44%)Melanoma
UACC-257 (49%)
Renal carcinoma 786-0 (44%)Colon carcinoma HCT-116 (54%)Ovarian adenocarcinoma OVCAR-4 (37%)
Table 3. Cytotoxic activity (IC50, µM) of the studied compounds estimated by the MTT assay.
Table 3. Cytotoxic activity (IC50, µM) of the studied compounds estimated by the MTT assay.
No.Cancer CellsNormal Cells
SW480SW620MDA-MB-231A-549LN-229HaCaT
IC50SIIC50SIIC50SIIC50SIIC50SIIC50
692.6 ± 5.311.08>100183.2 ± 3.381.2081.5 ± 5.711.22>1001>100
7>1000.93>1000.9367.3 ± 4.131.38>1000.93>1000.9392.7 ± 5.00
8>1001>1001>1001>1001>1001>100
9>1001>100196.5 ± 1.371.03>1001>1001>100
Doxorubicin0.7 ± 0.10.40.3 ± 0.11.01.6 ± 0.230.190.2 ± 0.091.51.1 ± 0.120.270.3 ± 0.1
Cisplatin10.4 ± 0.90.66.7 ± 1.10.97.8 ± 0.980.813.2 ± 1.245.082.6 ± 0.152.426.3 ± 0.7
Table 4. Docking results obtained from the DiffDock and AutoDock Vina tools.
Table 4. Docking results obtained from the DiffDock and AutoDock Vina tools.
PoseDiffDock ConfidenceAutodock Vina Binding Affinity [kcal/mol]DiffDock ConfidenceAutoDock Vina
Binding Affinity [kcal/mol]
88a
1−1.26−8.88−1.38−9.03
2−1.34−8.77−1.42−9.01
3−1.46−8.74−1.51−8.98
4−1.61−8.65−1.59−8.93
5−1.91−8.61−1.60−8.91
6−2.06−8.61−1.73−8.85
7−2.18−8.58−1.93−8.53
8−2.38−8.50−1.98−8.61
9−3.25−8.48−2.08−8.54
10−3.67-−4.15-
Average−2.11−8.65−1.94−8.82
Deviation0.810.130.810.20
Table 5. Detected protein–ligand interactions for the analyzed complexes.
Table 5. Detected protein–ligand interactions for the analyzed complexes.
DiffDockAutoDock VinaDiffDockAutoDock
Vina
88a
Phe168, His198Lys20, His21, Arg37, Ser123, Phe168His158, Phe168, His198, Phe227Arg37, Pro155, Asn119, Phe168, His198, His332
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

Martula, E.; Morak-Młodawska, B.; Jeleń, M.; Strzyga-Łach, P.; Struga, M.; Żurawska, K.; Kasprzycka, A.; Bagrowska, W. Comparative Analysis of Structural Analogs of Dipyridothiazines with m-Xylene and a Lutidine Moiety—In Silico, In Vitro, and Docking Studies. Appl. Sci. 2024, 14, 7263. https://doi.org/10.3390/app14167263

AMA Style

Martula E, Morak-Młodawska B, Jeleń M, Strzyga-Łach P, Struga M, Żurawska K, Kasprzycka A, Bagrowska W. Comparative Analysis of Structural Analogs of Dipyridothiazines with m-Xylene and a Lutidine Moiety—In Silico, In Vitro, and Docking Studies. Applied Sciences. 2024; 14(16):7263. https://doi.org/10.3390/app14167263

Chicago/Turabian Style

Martula, Emilia, Beata Morak-Młodawska, Małgorzata Jeleń, Paulina Strzyga-Łach, Marta Struga, Katarzyna Żurawska, Anna Kasprzycka, and Weronika Bagrowska. 2024. "Comparative Analysis of Structural Analogs of Dipyridothiazines with m-Xylene and a Lutidine Moiety—In Silico, In Vitro, and Docking Studies" Applied Sciences 14, no. 16: 7263. https://doi.org/10.3390/app14167263

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