Next Article in Journal
Uprolides N, O and P from the Panamanian Octocoral Eunicea succinea
Next Article in Special Issue
Pyridine and p-Nitrophenyl Oxime Esters with Possible Photochemotherapeutic Activity: Synthesis, DNA Photocleavage and DNA Binding Studies
Previous Article in Journal
Aza-Michael Mono-addition Using Acidic Alumina under Solventless Conditions
Previous Article in Special Issue
Discovery of Novel Allopurinol Derivatives with Anticancer Activity and Attenuated Xanthine Oxidase Inhibition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel 5-Substituted 2-(Aylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides: Synthesis, Molecular Structure, Anticancer Activity, Apoptosis-Inducing Activity and Metabolic Stability

by
Beata Żołnowska
1,*,
Jarosław Sławiński
1,*,
Aneta Pogorzelska
1,
Krzysztof Szafrański
1,
Anna Kawiak
2,3,
Grzegorz Stasiłojć
4,
Mariusz Belka
5,
Szymon Ulenberg
5,
Tomasz Bączek
5 and
Jarosław Chojnacki
6
1
Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, Gdańsk 80-416, Poland
2
Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, ul. Abrahama 58, Gdańsk 80-307, Poland
3
Laboratory of Human Physiology, Medical University of Gdańsk, ul. Tuwima 15, Gdańsk 80-210, Poland
4
Laboratory of Cell Biology, Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, ul. Dębinki 1, Gdańsk 80-211, Poland
5
Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, Gdańsk 80-416, Poland
6
Department of Inorganic Chemistry, Gdańsk University of Technology, Narutowicza 11/12, Gdańsk 80-233, Poland
*
Authors to whom correspondence should be addressed.
Molecules 2016, 21(6), 808; https://doi.org/10.3390/molecules21060808
Submission received: 13 May 2016 / Revised: 8 June 2016 / Accepted: 17 June 2016 / Published: 22 June 2016

Abstract

:
A series of novel 5-substituted 2-(arylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl) benzenesulfonamide derivatives 2760 have been synthesized by the reaction of aminoguanidines with an appropriate phenylglyoxal hydrate in glacial acetic acid. A majority of the compounds showed cytotoxic activity toward the human cancer cell lines HCT-116, HeLa and MCF-7, with IC50 values below 100 μM. It was found that for the analogues 3638 the naphthyl moiety contributed significantly to the anticancer activity. Cytometric analysis of translocation of phosphatidylserine as well as mitochondrial membrane potential and cell cycle revealed that the most active compounds 37 (HCT-116 and HeLa) and 46 (MCF-7) inhibited the proliferation of cells by increasing the number of apoptotic cells. Apoptotic-like, dose dependent changes in morphology of cell lines were also noticed after treatment with 37 and 46. Moreover, triazines 37 and 46 induced caspase activity in the HCT-116, HeLa and MCF-7 cell lines. Selected compounds were tested for metabolic stability in the presence of pooled human liver microsomes and NADPH, both R2 and Ar = 4-CF3-C6H4 moiety in 2-(R2-methylthio)-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides simultaneously increased metabolic stability. The results pointed to 37 as a hit compound with a good cytotoxicity against HCT-116 (IC50 = 36 μM), HeLa (IC50 = 34 μM) cell lines, apoptosis-inducing activity and moderate metabolic stability.

Graphical Abstract

1. Introduction

For decades chemotherapeutics have played the most important role in the fight against cancer. Unfortunately, the serious toxicities of conventional cytotoxic medicines have impelled and are still impelling researchers to focus on the development of new potent and selective anticancer drugs.
Heterocyclic scaffolds, especially nitrogen-containing heterocyclic compounds, play an important role in the design of novel drugs because of their utility for various biological receptors with a high degree of binding affinity. Among the heterocycles, the triazines, with their wide biological profile, occupy a prominent position [1]. The 1,2,4-triazine ring as one of the most ubiquitous heterocycles in Nature, and it has been reported to possess a broad spectrum of biological properties, including anticonvulsant [2], neuroprotective [3], sedative [4], anxiolytic [5], benzodiazepine receptor inhibitory activity [6], antiparkinson [7], antidepressant [8], anti-inflammatory [9], antimicrobial [10], antiparasitic [11] activities. Some 1,2,4-triazines have become well-known drugs, such as the antiviral azaribine [12], the anticonvulsant lamotrigine [13], the anti-inflammatory and analgesic apazone [14], the antibacterial ceftriaxone [15], the insecticide pymetrozine [16] or vardenafil, useful in the treatment of male erectile dysfunction [17]. In addition, there are also many reports indicating significant anticancer properties for the 1,2,4-triazine fragment. For example, tirapazamine (I, Figure 1) is currently in various clinical trial phases for the treatment of human non-small cell, cervical, ovarian, head and neck cancers. Tirapazamine works by inducing DNA damage in poorly oxygenated tumor cells [18]. The 6-azauridine II, known as an inhibitor of de novo pyrimidine biosynthesis, is an effective agent for remission induction of acute myelocytic leukemia in children [19]. Close analogs of 6-azauridine, S-alkyl derivatives of 1,2,4-triazinone III, show cytotoxic activities against human breast cancer (MCF-7), colon carcinoma (HCT-116) and hepatocellular carcinoma (Hep-G2) cell lines [20]. Some 1,2,4-triazine analogs IV have distinct antiptoliferative activities against non-small cell lung cancer (NCI-H460), breast cancer (MCF-7), CNS cancer (SF-268), with a low cytotoxicity [21].
Our systematic studies on arylsulfonamides has resulted in promising anticancer agents [22,23,24,25,26,27,28,29], and among them we have reported that some trisubstituted 1,2,4-triazines V (Figure 1), exhibited anticancer activity against colon, CNS, melanoma, ovarian, breast, renal, leukemia cell lines [29]. Searching for innovative low-molecular chemotherapeutics, a structure-based molecular hybridization strategy has been applied as a common approach to develop multi-targeted compounds [30]. Thus, we designed and synthesized a series of new 4-chloro-2-mercapto-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides VI (Figure 1), bearing a disubstituted 1,2,4-triazine and a 2-mercaptobenzenesulfonamide moiety in the molecular skeleton, (Figure 1) as potential anticancer agents. Various substituent groups were introduced to the triazine ring as well as the benzenesulfonamide fragment in positions 2 and 5 for structure-activity relationship (SAR) analysis. The prepared compounds were evaluated for their antiproliferative activities against three human cancer cell lines, namely, HCT-116 (colon cancer), HeLa (cervical cancer) and MCF-7 (breast cancer). For the most active compounds apoptosis- inducing activities on HCT-116, HeLa and MCF-7 cell lines were further investigated.
In vitro tests for metabolic stability, which is one of the important parameters characterizing pharmacokinetic properties of a molecule, were performed on selected compounds. The experiments were run in the presence of human liver microsomes and NADPH.

2. Results and Discussion

2.1. Chemistry

The starting substrates, 6-chloro-3-methylthio-7-R1-1,1-dioxo-1,4,2-benzodithiazines 15 [31,32], 3-amino-6-chloro-7-R1-1,1-dioxo-1,4,2-benzodithiazines 610 [33,34], N-(benzenesulfonyl)cyanamide potassium salts 1118 [22,25,33,35] as well as aminoguanidines 1920 and 2325 [29,33,35,36] were prepared according to the respective known methods. Novel substrates 2122 and 26 were synthesized analogously by the reaction of hydrazine monohydrochloride with the potassium salts 1314 as was shown in Scheme 1. Finally, 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides 2760 were readily obtained by treatment of aminoguanidines 1926 with the appropriate phenylglyoxal hydrate in glacial acetic acid at reflux for 24–45 h as outlined in Scheme 2.
The structures of the final compounds 2760 were confirmed by IR, 1H-NMR and 13C-NMR spectroscopy. 1H-NMR spectra showed the absence of characteristic of aminoguanidine NH signals at 4 and 6 ppm, confirming the success of the condensation reactions between the amine and carbonyl groups of the reagents leading to the heterocyclic ring formation. In addition, the presence in the 1H-NMR spectra of a singlet at about 9 ppm corresponding to the H-6 proton in the 1,2,4-triazine fragment was observed. Moreover, X-ray analysis was done to confirm the proposed structures using the representative compound 59.
Compound 59 crystallizes in the triclinic system in the space group P 1 ¯ . The asymmetric unit contains two sulfonamide molecules and four solvating dimethylformamide (DMF) molecules. The unit cell of the crystal contains two asymmetric parts (Z = 2). The two independent sulfonamide molecules have the same topological atom connectivity (constitution) but cannot be superimposed on each another (they are almost related by a mirror plane passing through SO2 group and bisecting the S–N–C and S–C–C planes). The solvent DMF molecules are not only filling the voids in the structure, but also participate in hydrogen bonding—two of carbonyl groups are acceptors of hydrogen from N-H of 1,2,4-triazine residue (O14-N4, O16-N9, see Table 1). The other two DMF molecules use their CO groups as acceptors for amidic NH groups (O13-N6, O15-N1, Table 1). Other intermolecular interactions responsible for crystal packing are π-π stacking operating between R1 (N3, C22, N4, N5, C23, C24) and R5 (C25–C30) with the distance between ring geometry centers (centroids) of 3.8252(3)Å and R6 (N8, C54, N9, N10, C55, C56) and R10 (C57–C52) 3.8952(3)Å. All other rings are separated by more than 4.4Å and interactions among them were neglected.
The sulfonamide group seems to be deprotonated in the solid state, as there is a C–H bond from a DMF molecule directed to the sulfonamidic N (Figure 2) which would not be beneficial without assumed ionization of the –SO2NH– fragment. Additionally, protonation of the nitrogen atom in position 2 of the 1,2,4-triazine is required by electrical neutrality and by formation of hydrogen bond to the carbonyl group from the neighbor DMF molecule. Thus, two charge-assisted hydrogen bonds are created: C–H···N(−)–S and (+)N–H···O between the sulfonamide and the solvating DMF.
Bond lengths within the triazine ring are not helpful to attribute double bonds in the substructure. The 1,2,4-triazine residue is flat and almost coplanar with 3,4-dimethoxyphenyl which may be a consequence of beneficial π-π interactions with the neighbor molecule in crystal. The second solvent DMF molecule forms hydrogen bond with the amide N–H proton donor in the other branch of the main molecule. The other functional groups are not unusual and their geometry will therefore not be further analyzed.

2.2. Biological Evaluations

2.2.1. Cytotoxic Activity

Compounds 2760 were evaluated in vitro for their effects on the viability of three human cancer cell lines: HCT-116 (colon cancer), HeLa (cervical cancer) and MCF-7 (breast cancer). The concentration required for 50% inhibition of cell viability IC50 was calculated and compared with the reference drug cisplatin, the results were shown in Table 2. To describe of cytotoxic potency, the following scale was applied: IC50 < 25 μM—very strong, 25 ≤ IC50 ≤ 50 μM—strong, 50 < IC50 < 75 μM—moderate, 75 ≤ IC50 ≤ 100 μM—weak, IC50 >100 μM—inactive compounds.
The most active compounds 37 and 38 belonged to the 1-naphthyl series (R2 = 1-naphthyl) and showed outstanding average cytotoxic activity (47 μM and 50 μM, respectively) against all tested cell lines, HCT-116, HeLa and MCF-7. Moreover, compounds 3436 and 46 moderately inhibited all cell lines viability, exhibiting average IC50 values below 71 μM. Among tested analogues only six compounds (17.6%) were completely inactive with average IC50 > 100 μM.
As shown in Table 2, the HCT-116 cell line presented the relatively highest susceptibility and was affected by eleven compounds (3338, 46, 47, 52, 56 and 57) in the range of IC50 values 36–75 μM. Meanwhile, the HeLa cell line was susceptible toward six compounds (3438 and 53) with IC50; 34–74 μM, and the MCF-7 by six compounds (30, 34, 37, 38, 46 and 52) with IC50 values ranging between 59 and 70 μM.
We found that, among 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzene-sulfonamides with R2 = 1-naphthyl, presence of Ar = 4-CF3-C6H4 group provided strong cytotoxicity of 37 toward the HeLa (IC50 = 34 μM) as well as HCT-116 (IC50 = 36 μM) cells and replacement of 1-naphthyl substituent in this compound by the smaller aromatic group, i.e., 34 (R2 = 4-CF3-C6H4), 40 (R2 = 3,5-dimethylisoxazol-4-yl) caused decrease in activity to IC50; 51–85 μM or the loss of activity when R2 = Ph (compd 28, see Table 2). It should be noted, that replacement of Ar = 4-CF3-C6H4 group (37) by Ar = 4-MeO-C6H4 (38) or 3-F-C6H4 (36) did not contribute to improving the anticancer activity. The presence of Ar = 4-CF3-C6H4 also influenced the strongest cytotoxic activity of 46 (R1 = 4-Cl-C6H4NHCO, R2 = Ph, IC50 = 59 μM) toward the MCF-7 cell line. Nevertheless, the exchange of R1 = 4-Cl-C6H4NHCO in compd 46 to small methyl group strongly inactivate the compound 28 (R1 = Me, R2 = Ph) but introduction of 4-Me-C6H4NHCO as the R1 substituent in compd 52 decreased its cytotoxic activity to IC50 = 69 μM.

2.2.2. Investigation of Apoptotic Activity

The ability to induce apoptosis in cancer cells is a desired feature of a potential chemotherapeutic agent. Thus, the apoptosis-inducing activity was studied through biochemical markers, such as: DNA fragmentation, loss of mitochondrial membrane potential (Δψm), phosphatidylserine translocation and caspase activation. Apoptotic-like changes in morphology of tested cell lines were also evaluated. The experiments were performed with the most active compounds (37 and 46) exhibited the highest activity in MTT tests.

Cell Morphology

To evaluate the changes in morphology of the treated cells, they were incubated with increasing concentrations of 37 (HCT-116, HeLa) or 46 (MCF-7) for 24 h and observed using light microscope. The most characteristic morphological changes (shrinkage of the cells, detachment from the surface) occurred in the HCT-116 and HeLa cells treated with 37 (Figure 3).

Cell Cycle Analysis

One of the most common mechanisms in anticancer drug treatment is changes in cell cycle, which can be measured by DNA content [37]. Cell cycle distribution in HCT-116, HeLa and MCF-7 cells was examined to determine cycle arrest and/or presence of sub-G1 after treatment with compounds 37 and 46. Cells were coincubated with trazines and a reference drug (cisplatin) for 24 h, respectively. Cell populations data are expressed as the mean ± SD of at least three independent experiments.
Sub-G1 cell population increased in dose dependent manner (HCT-116 from 3.6% ± 1.66% (control) to 48.6% ± 13.5%; HeLa from 5.7% ± 3.4% (control) to 21% ± 3.4%) after treatment with compound 37 for 24 h. Figure 4 shows one representative experiment. Similar results were obtained for MCF-7 cells coincubated with compound 46 for 24 h (from 5.0% ± 3.8% (control) to 20% ± 5.6%) (Figure 4).
Coincubation of HCT-116, HeLa and MCF-7 with appropriate triazines (37 and 46) caused appearance of sub-G1 stage and high concentrations led to fragmentation of DNA, resulting in a sub-G0/G1 peak of cell cycle. A significant accumulation of the HCT-116, HeLa and MCF-7 population in the sub-G1 phase indicated that the cells had undergone programmed cell death (apoptosis) for all of the analyzed compounds. Triazine coincubation did not cause significant differences in the cell cycle phases of the analyzed cell lines.

Mitochondrial Membrane Potential (Δψm) Analysis

Loss of the mitochondrial membrane potential (Δψm) is one of the earliest indicators of an apoptosis [38]. It can be detected using fluorescent dye JC-1 (5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide) exited with blue laser. JC-1 spontaneously forms dimer complexes (J aggregates) with red fluorescent or remain as a monomers with green fluorescence in cells with high and low Δψm, respectively.
As shown in Figure 5 HCT-116, HeLa and MCF-7 cells in the control group, high red fluorescence (J aggregates) were observed. However, exposure of cells even to low and moderate concentrations of compounds 37 and 46 remarkably decreased Δψm which is the earliest indicators of an apoptotic cell death.

Translocation of Phosphatidylserine to Outer Leaflet of Cell Membrane

The quantification of cell death was evaluated by measuring the exposure of phosphatidylserine on the outer leaflet of plasma membrane. Four subpopulations were identified according to their fluorescence: PI-low/FITC-low (live cells), PI-high/FITC-low (necrotic cells), PI-low/FITC-high (early apoptotic cells), PI-high/FITC-high (late apoptotic cells).
Appearance of increased population of PI-low/FITC-high (early apoptotic cells), PI-high/FITC-high (late apoptotic cells) gates was noticed for HCT-116 and HeLa cells coincubated with compound 37 (Figure 6). The increased levels of apoptotic cells were concentration dependent. For MCF-7 cells treated with compound 46 the results were not conclusive, as the differences between treated cells and control cells were statistically significant only for low 25 μM of 46 (Figure 6).

Caspase Activation

Caspase activation plays a central role in the execution of apoptosis and is required for the occurrence of its biochemical and morphological hallmarks, such as DNA fragmentation, formation of apoptotic bodies and chromatin condensation. The ability of the examined compounds to induce caspase activity was determined with the use of a fluorescent labeled caspase inhibitor, a carboxyfluorescein (FAM) derivative of valylalanylaspartic acid (VAD) fluoromethyl ketone (FMK).
FAM-VAD-FMK contains a target sequence recognized by active caspases (caspases 1 through 9). Binding to this sequence inhibits the enzymatic activity of caspases and allows for the determination of their activity through the direct measurement of fluorescent intensity of the bound inhibitor. The results of the research showed that the tested compounds 37 and 46 induced caspase activity in the examined cells lines, as shown by an increase in FAM-VAD-FMK fluorescence in the cell population with activated caspases (Figure 7). The strongest influence on caspase activation was noticed for the HeLa cell line, where that cell population increased by 31% in the presence of 37 at a concentration of 100 μM.

2.2.3. Metabolic Stability

Metabolism as one of the drugs’ pharmacokinetic characteristics can be evaluated during the early preclinical stage to assessment of degree of drug candidate conversion into a set of metabolites. Metabolic stability can be assessed by incubation of a potent drug in a presence of liver microsomes and NADPH to give an insight to metabolic properties [39,40,41].
For further evaluation of metabolic stability the compounds with outstanding activities and varied structural features including R1, R2 and Ar substituents (30, 31, 3438, 46, 47, 52) were selected. Human liver microsomes were chosen as a model enzymatic system and in vitro metabolic half-life was assessed (Table 3).
Based on the enzymatic test results some structure-metabolic stability relationships can be noted. The most stable compound 34 bears a 4-CF3-C6H4 moiety in both the R1 and Ar positions. Replacement of Ar = 4-CF3-C6H4 (34) by 4-MeO-C6H4 (compound 35) resulted in decrease of the t1/2 value from >60 to 42.4 min, while an analogous change between 46 and 47 caused a significant decrease of t1/2 from >60 to 17.5 min. On the other hand, taking into account the results for 30 (Ar = 3-MeO-C6H4) and 31 (Ar = 3,4-diMeO-C6H3), it was found that an additional methoxy (MeO) group in the Ar substituent slightly decreased the metabolic stability. These facts suggest that the methoxy substituent is a major soft spot in the described series of compounds and moreover, its undesirable influence can be diminished by incorporation of CF3 instead of a MeO substituent.
Comparison of t1/2 for 34 (R1 = 4-CF3-C6H4) and 37 (R1 = 1-naphthyl) indicated that 1-naphthyl (R1) also seems to decrease metabolic stability. We assumed that the undesirable influence of the 1-naphthyl (R1) and 4-MeO-C6H4 (Ar) substituents accelerated in the case of compound 38, which was characterized as the least stable derivative. Further synthesis in this group of compounds should avoid incorporation of the two abovementioned substituents. Although a 4-CF3-C6H4 (R1) substituent is preferable for metabolic stability in comparison with a 1-naphthyl moiety, the high biological response observed for 37 predisposes it to be a lead compound, as it shows a good balance between stability and cytotoxic properties.
In order to explain the differences between the metabolic stability of distinctive compounds (3435, 3738) we applied a tool accessible on-line for accurate prediction of xenobiotic metabolism sites, called XenoSite Cytochrome P450 Prediction Models [42]. Among the available models, one is able to predict which atoms on a molecule are likely to be oxidized by human liver microsomes. In silico results showed that the least stable derivative 38 has three more sites vulnerable for metabolic biotransformation than 34 (see Figure 8). Moreover, a slight decrease of metabolic stability of 35 and 37 compared to 34 may resulted from presence of additional sites of oxidation on methoxy group and methylene linker (35, Figure 8) as well as on methylene linker and naphthalene ring (37, Figure 8).

3. Materials and Methods

3.1. General Information

The melting points were uncorrected and measured using Boethius PHMK apparatus (Veb Analytic, Dresden, Germany). IR spectra were measured on Thermo Mattson Satellite FTIR spectrometer (Thermo Mattson, Madison, WI, USA) in KBr pellets; an absorption range was 400–4000 cm−1. 1H-NMR and 13C-NMR spectra were recorded on a Varian Gemini 200 apparatus (Varian, Palo Alto, CA, USA) or a Varian Unity Plus 500 apparatus. Chemical shifts are expressed at δ values relative to Me4Si (TMS) as an internal standard. The apparent resonance multiplicity is described as: s (singlet), d (doublet), dd (doublet of doublets), t (triplet), m (multiplet), and br (broad) signal. Elemental analyses were performed on 2400 Series II CHN Elemental Analyzer (PerkinElmer, Shelton, CT, USA) and the results were within ±0.4% of the theoretical values. Thin-layer chromatography (TLC) was performed on Kieselgel 60 F254 plates (Merck, Darmstadt, Germany) and visualized by UV. The commercially unavailable substrates were obtained according to the following methods described previously: 6-chloro-3-methylthio-7-R1-1,1-dioxo-1,4,2-benzo-dithiazines 15 [31,32], 3-amino-6-chloro-7-R1-1,1-dioxo-1,4,2-benzodithiazines 610 [33,34], N-(2-alkylthio-4-chloro-5-R1-benzenesulfonyl)cyanamide potassium salts 1118 [22,25,33,35], 1-amino-2-(4-chloro-5-R1-2-alkylthiobenzenesulfonyl)guanidines 1920 and 2325 [25,29,33,35,36]. The NMR spectra of newly synthesized compounds 2122, 2660 are given in the Supplementary Materials.

3.2. Synthesis

3.2.1. Procedures for the Preparation of Aminoguanidines 2122 and 26

A mixture of the appropriate cyanamide potassium salt 13, 14 or 18 (7 mmol) and hydrazine monohydrochloride (0.48 g, 7 mmol) in anhydrous toluene (14 mL) was refluxed with stirring for 3–9 h. After cooling to room temperature, the reaction mixture was left in the freezer overnight. The precipitate was collected by filtration, dried and suspended in water (9 mL). The solid was separated and crystallized from ethanol (2122) or p-dioxane (26). In this manner, the following aminoguanidines were obtained.
1-Amino-2-{4-chloro-5-methyl-2-[(naphthalen-1-ylmethyl)thio]benzenesulfonyl}guanidine (21). Starting from N-{4-chloro-5-methyl-2-[(naphthalen-1-ylmethyl)thio]benzenesulfonyl}cyanamide monopotassium salt 13 (3.09 g), the title compound 21 was obtained (2.55 g, 80%) mp 217–219 °C; IR (KBr)νmax 3453, 3348, 3217 (NH), 2925, 2853 (C-H), 1385, 1133 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.31 (s, 3H, CH3), 4.44 (s, 2H, C-NH2), 4.74 (s, 2H, SCH2), 6.93 (s, 2H, NNH2), 7.46 (t, 1H, arom), 7.51 (s, 1H, H-3, arom), 7.57–7.59 (m, 3H, arom), 7.84 (s, 1H, H-6, arom), 7.87 (d, J = 8.3 Hz, 1H, arom), 7.95 (d, J = 7.8 Hz, 1H, arom), 8.23 (d, J = 8.3 Hz, 1H, arom), 8.38 (s, 1H, NH) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 19.7, 35.2, 124.9, 126.1, 126.6, 126.9, 128.6, 128.8, 129.2, 131.1, 132.1, 132.3, 132.8, 134.1, 136.6, 137.3, 140.4, 155.9, 157.6 ppm; anal. C 52.20, H 4.30, N 12.61% calcd. for C19H19ClN4O2S2, C 52.47, H 4.40, N 12.88%.
1-Amino-2-{4-chloro-5-methyl-2-[(3,5-dimethylisoxazol-4-ylmethyl)thio]benzenesulfonyl}guanidine (22). Starting from N-{4-chloro-5-methyl-2-[(3,5-dimethylisoxazol-4-ylmethyl)thio]benzenesulfonyl}cyanamide monopotassium salt 14 (2.87 g), the title compound 22 was obtained (2.34 g, 84%) mp 204–206 °C; IR (KBr)νmax 3479, 3345, 3341 (NH); 2926, 2851 (C–H); 1268, 1131 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.21 (s, 3H, CH3), 2.30–2.31 (m, 6H, 2×CH3), 4.08 (s, 2H, SCH2), 4.49 (s, 2H, CNH2), 6.97 (s, 2H, NNH2), 7.43 (s, 1H, H-3, arom), 7.82 (s,1H, H-6, arom), 8.39 (s, 1H, NH) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 10.4, 11.3, 19.7, 25.3, 109.7, 129.7, 131.0, 133.0, 135.1, 136.6, 141.8, 159.4, 160.3, 167.3 ppm; anal. C 41.88, H 4.56, N 17.62% calcd. for C14H18ClN5O3S2, C 41.63, H 4.49, N 17.34%.
1-Amino-2-(2-benzylthio-4-chloro-5-methoxyphenylcarbamoylbenzenesulfonyl)guanidine (26). Starting from N-(2-benzylthio-4-chloro-5-methoxyphenylcarbamoylbenzenesulfonyl)cyanamide monopotassium salt 18 (3.68 g), the title compound 26 was obtained (2.52 g, 70%) mp 238–240 °C; IR (KBr) νmax 3312, 3212 (NH), 2921, 2851 (C–H), 1678 (C=O), 1541, 1510 (C=C, C=N), 1312, 1134 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 3.75 (s, 3H, OCH3), 4.41 (s, 2H, SCH2), 4.54 (s, 2H, NH2), 6.92–7.03 (m, 4H, arom), 7.29–7.41 (m, 3H, arom), 7.48–7.64 (m, 5H, arom), 7.97 (m, 1H, arom), 8.52 (s, 1H, NH), 10.43 (s, 1H, NHCO) ppm; anal. C 50.56, H 4.15, N 13.15 calcd. for C22H22ClN5O4S2, C 50.81, H 4.26, N 13.47%.

3.2.2. Procedures for the Preparation of 4-Chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides 2760

A mixture of the appropriate aminoguanidine 1926 (1 mmol) and phenylglyoxal hydrate (1 mmol) in glacial acetic acid (2.5 mL) was refluxed with stirring for 24–45 h. The solid of the compound was obtained either after stirring overnight at room temperature, or after additional stirring on an ice bath for 12 h (compds. 3031, 4244, 47). The formed precipitate was collected by filtration, washed with glacial acetic acid (2 × 1 mL) and dried. The final products 2760 were purified as described below.
2-Benzylthio-4-chloro-N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]-5-methylbenzenesulfonamide (27). Starting from 19 (0.385 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 33 h, the crude 27 (0.431 g) was obtained. The crude 27 was crystallized twice from EtOH and the title compound 27 (0.290 g, 58%) was isolated mp 216–218 °C; IR (KBr) νmax 3430, 3196 (NH), 3074, 2921 (C–H), 1585, 1556 (C=C, C=N), 1302, 1159 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.29 (s, 3H, CH3), 4.31 (s, 2H, SCH2), 7.09–7.10 (m, 3H, arom), 7.26–7.37 (m, 2H, arom), 7.50–7.67 (m, 4H, arom), 7.82–7.86 (m, 1H, arom), 8.13 (s, 1H, H-6, arom), 9.31 (s, 1H, triazine), 14.25 (brs, 1 H, SO2NH) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 19.1, 36.2, 114.8, 115.3, 121.1, 125.1, 127.3, 128.3, 128.4, 129.1, 131.6, 131.7, 132.5, 132.7, 134.9, 135.1, 135.4, 136.5, 137.6, 160.1, 165.0 ppm; anal. C 55.32, H 3.36, N 11.36% calcd. for C23H18ClFN4O2S2, C 55.14, H 3.29, N 11.18%.
2-Benzylthio-4-chloro-5-methyl-N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]benzenesulfonamide (28). Starting from 19 (0.385 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 33 h, the title compound 28 (0.298 g, 72%) was obtained without further purification mp 287–289 °C; IR (KBr) νmax 3457, 3182 (NH), 3089, 2929 (C–H), 1585, 1560 (C=C, C=N), 1320, 1123 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.37 (s, 3H, CH3), 4.32 (s, 2H, SCH2), 7.10–7.13 (m, 3H, arom), 7.28–7.30 (m, 2H, arom), 7.57 (s, 1H, H-3, arom), 7.92–7.96 (m, 2H, H-6, arom), 8.15–8.17 (m, 3H, arom), 9.42 (s, 1H, triazine), 14.10 (brs, 1 H, SO2NH) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 19.1, 36.2, 121.3, 126.1, 126.2, 126.3, 127.3, 128.2, 128.5, 129.2, 129.4, 129.5, 132.3, 133.2, 135.7, 136.4, 136.7, 137.7, 137.8, 137.9 ppm; anal. C 52.45, H 3.41, N 10.28% calcd. for C23H18ClFN4O2S2, C 52.32, H 3.29, N 10.17%.
2-Benzylthio-4-chloro-N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]-5-methylbenzenesulfonamide (29). Starting from 19 (0.385 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 31 h, the crude 29 (0.381 g) was obtained. The crude 29 was crystallized from MeCN and the title compound 29 (0.324 g, 63%) was isolated mp 232–234 °C; IR (KBr) νmax 3188 (NH), 2918, 2831 (C–H), 1606, 1575 (C=C, C=N), 1533 (NHdef.), 1298, 1180 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.40 (s, 3H, CH3), 3.88 (s, 3H, OCH3), 4.27 (s, 2H, SCH2), 7.05–7.17 (m, 4H, arom), 7.24–7.31 (m, 3H, arom), 7.48 (s, 1H, H-3, arom), 7.95 (d, 2H, J = 8.93 Hz, arom), 8.12 (s, 1H, H-6), 9.08 (s, 1H, triazine) ppm; anal. C 56.02, H 3.96, N 10.91% calcd. for C24H21ClN4O3S2, C 56.19, H 4.13, N 10.92%.
2-Benzylthio-4-chloro-N-[5-(3-methoxyphenyl)-1,2,4-triazin-3-yl]5-methylbenzenesulfonamide (30). Starting from 19 (0.385 g) and 3-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the title compound 30 (0.400 g, 78%) was obtained without further purification mp 198–200 °C; IR (KBr) νmax 3199 (NH), 2934 (C–H), 1631, 1582 (C=C, C=N), 1561 (NHdef.), 1283, 1132 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.28 (s, 3H, CH3), 3.81 (s, 3H, OCH3), 4.29 (s, 2H, SCH2), 7.10–7.13 (m, 3H, arom), 7.26–7.32 (m, 3H, arom), 7.42–7.52 (m, 4H, H-3, arom.), 8.08 (s, 1H, H-6, arom), 9.27 (s, 1H, triazine) ppm; anal. C 56.13, H 4.01, N 10.96% calcd. for C24H21ClN4O3S2, C 56.19, H 4.13, N 10.92%.
2-Benzylthio-4-chloro-N-[5-(3,4-dimethoxyphenyl)-1,2,4-triazin-3-yl]-5-methylbenzenesulfonamide (31). Starting from 19 (0.385 g) and 3,4-dimethoxyphenylglyoxal hydrate (0.212 g) after stirring for 32 h, the title compound 31 (0.443 g, 82%) was obtained without further purification mp 198–200 °C; IR (KBr) νmax 3447, 3125 (NH), 3000, 2936 (C–H), 1615, 1554, 1519 (C=C, C=N), 1316, 1133 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.35 (s, 3H, CH3), 3.80 (s, 3H, OCH3), 3.88 (s, 3H, OCH3), 4.28 (s, 2H, SCH2), 7.08–7.19 (m, 4H, arom), 7.28–7.32 (m, 2H, arom), 7.47 (s, 1H, H-3, arom), 7.56 (d, J = 1.91 Hz, 1H, arom), 7.67 (dd, J1 = 8.50 Hz, J2 = 2.00 Hz, 1H, arom), 8.08 (s, 1H, H-6, arom), 9.20 (s, 1H, triazine) ppm; anal. C 55.08, H 4.13, N 10.11% calcd. for C25H23ClN4O4S2, C 55.29, H 4.27, N 10.32%.
2-Benzylthio-4-chloro-5-methyl-N-[5-(3,4,5-trimethoxyphenyl)-1,2,4-triazin-3-yl]benzenesulfonamide (32). Starting from 19 (0.385 g) and 3,4,5-trimethoxyphenylglyoxal hydrate (0.242 g) after stirring for 33 h, the title compound 32 (0.525 g, 92%) was obtained without further purification mp 163–164 °C; IR (KBr) νmax 3436 (NH), 3073, 2936 (C–H), 1594, 1550 (C=N, C=C), 1332, 1129 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.28 (s, 3H, CH3), 3.79 (s, 3H, OCH3), 3.85 (s, 6H, 2×OCH3), 4.32 (s, 2H, SCH2), 7.17–7.19 (m, 3H, arom), 7.31–7.35 (m, 4H, arom), 7.52 (s, 1H, H-3, arom), 8.04 (s, 1H, H-6, arom), 9.34 (brs, 1H, triazine), 14.25 (brs, 1H, SO2NH) ppm; 13C-NMR (50 MHz, DMSO-d6) δ: 19.1, 36.3, 56.7, 60.7, 107.1, 127.4, 127.7, 127.9, 128.5, 129.2, 132.0, 132.1, 135.4, 136.4, 137.3, 146.2, 153.6 ppm; anal. C 54.82, H 4.56, N 10.07% calcd. for C26H25ClN4O5S2, C 54.49, H 4.39, N 9.78%.
4-Chloro-N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]-5-methyl-2-[4-(trifluoromethyl)benzylthio]benzenesulfonamide (33). Starting from 20 (0.451 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 33 h, the crude 33 (0.446 g) was obtained. The crude 33 was purified by heating under reflux with EtOH and the title compound 33 (0.330 g, 58%) was isolated mp 212–214 °C; IR (KBr) νmax 3436, 3186 (NH), 3092, 2923, 2851 (C–H), 1589, 1554 (C=C, C=N), 1326, 1126 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.35 (s, 3H, CH3), 4.42 (s, 2H, SCH2), 7.39 (d, J = 8.30 Hz, 2H, arom), 7.43–7.53 (m, 4H, arom), 7.54–7.65 (m, 2H, arom), 7.76 (d, J = 7.80 Hz, 1H, arom), 8.08 (s, 1H, H-6, arom), 9.28 (brs, 1H, triazine) ppm; anal. C 50.71, H 3.11, N 9.89% calcd. for C24H17ClF4N4O2S2, C 50.66, H 3.01, N 9.85%.
4-Chloro-5-methyl-2-[4-(trifluoromethyl)benzylthio]-N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]-benzenesulfonamide (34). Starting from 20 (0.451 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 33 h, the crude 34 (0.447 g) was obtained. The crude 34 was purified by heating under reflux with EtOH and the title compound 34 (0.331 g, 53%) was isolated mp 256–259 °C; IR (KBr) νmax 3444 (NH), 3078, 2925 (C–H), 1581, 1556 (C=C, C=N), 1325, 1131 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.33 (s, 3H, CH3), 4.42 (s, 2H, SCH2), 7.40 (d, 2H, J = 7.80 Hz, arom), 7.49 (d, 2H, J = 8.30 Hz, arom), 7.60 (s, 1H, H-3, arom), 7.89 (d, 2H, J = 8.30 Hz, arom), 8.08 (d, J = 8.30 Hz, 2H, arom), 8.13 (s, 1H, H-6, arom), 9.39 (brs, 1H, triazine) ppm; anal. C 48.66, H 3.00, N 9.12% calcd. for C25H17ClF6N4O2S2, C 48.51, H 2.77, N 9.05%.
4-Chloro-N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]-5-methyl-2-[4-(trifluoromethyl)benzylthio]benzene-sulfonamide (35). Starting from 20 (0.451 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 29 h, the crude 35 (0.462 g) was obtained. The crude 35 was purified by heating under reflux with EtOH and the title compound 35 (0.387 g, 66%) was isolated mp 255–258 °C; IR (KBr) νmax 3435 (NH), 3084, 2931 (C–H), 1579, 1536 (C=C, C=N), 1299, 1125 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.37 (s, 3H, CH3), 3.86 (s, 3H, OCH3), 4.39 (s, 2H, SCH2), 7.04 (d, 2H, J = 8.8 Hz, arom), 7.44 (d, 2H, arom), 7.48-7.58 (m, 3H, arom), 7.89 (d, 2H, J = 7.8 Hz, arom), 8.10 (s, 1H, H-6, arom), 9.04 (brs, 1H, triazine), 14.51 (brs, 1H, SO2NH) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 19.7, 35.8, 56.5, 88.9, 115.4, 123.6, 124.9, 125.6, 128.1, 128.3, 130.3, 132.9, 134.8, 142.4 ppm; anal. C 51.75, H 3.49, N 9.68% calcd. for C25H20ClF3N4O3S2, C 51.68, H 3.47, N 9.64%.
4-Chloro-N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]-5-methyl-2-[(naphthalene-1-yl)methylthio]benzene-sulfonamide (36). Starting from 21 (0.435 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 43 h, the crude 36 (0.417 g) was obtained. The crude 36 was purified by heating under reflux with EtOH and the title compound 36 (0.277 g, 50%) was isolated mp 234–237 °C; IR (KBr) νmax 3432 (NH), 3052, 2922 (C–H), 1552, 1524 (C=C, C=N), 1442, 1168 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.40 (s, 3H, CH3), 4.75 (s, 2H, SCH2), 7.26 (t, 1H, arom), 7.42–7.57 (m, 5H, arom), 7.58–7.64 (m, 1H, arom), 7.68 (s, 1H, H-3, arom), 7.74 (d, J = 8.30 Hz, 1H, arom), 7.81–7.82 (m, 1H, arom), 7.86–7.89 (m, 1H, arom), 8.08–8.12 (m, 2H, arom), 9.20 (brs, 1H, triazine) ppm; anal. C 58.53, H 3.45, N 9.95% calcd. for C27H20ClFN4O2S2, C 58.85, H 3.66, N 10.17%.
4-Chloro-5-methyl-2-[(naphthalene-1-yl)methylthio]-N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]-benzenesulfonamide (37). Starting from 21 (0.435 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 30 h, the crude 37 (0.526 g) was obtained. The crude 37 was purified by heating under reflux with EtOH and the title compound 37 (0.417 g, 69%) was isolated: mp 247–251 °C; IR (KBr) νmax 3438 (NH), 3080, 2927 (C–H), 1580, 1554 (C=C, C=N), 1323, 1162 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.38 (s, 3H, CH3), 4.76 (s, 2H, SCH2), 7.28 (t, 1H, arom), 7.39–7.57 (m, 3H, arom), 7.67 (s, 1H, H-3, arom), 7.76 (d, J = 7.8 Hz,1H, arom), 7.82–8.02 (m, 3H, arom), 8.02–8.28 (m, 4H, arom), 9.30 (brs, 1H, triazine) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 19.6, 34.9, 124.8, 125.9, 126.6, 126.9, 128.6, 128.8, 129.1, 129.1, 130.0, 131.9, 132.2, 132.7, 133.5, 133.9, 137.0 ppm; anal. C 55.63, H 3.20, N 8.99% calcd. for C28H20ClF3N4O2S2, C 55.95, H 3.35, N 9.32%.
4-Chloro-N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]-5-methyl-2-[(naphthalene-1-yl)methylthio]benzene-sulfonamide (38). Starting from 21 (0.435 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 40 h, the crude 38 (0.461 g) was obtained. The crude 38 was purified by heating under reflux with EtOH and the title compound 38 (0.389 g, 75%) was isolated mp 262–265 °C; IR (KBr) νmax 3450 (NH), 3060, 2925 (C–H), 1607, 1541 (C=C, C=N), 1264, 1176 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.42 (s, 3H, CH3), 3.88 (s, 3H, OCH3), 4.73 (s, 2H, SCH2), 7.06 (d, J = 8.8 Hz, 2H, arom), 7.30 (t, 1H, arom), 7.44–7.56 (m, 3H, arom), 7.60 (s, 1H, H-3, arom), 7.78 (d, J = 8.3 Hz, 1H, arom), 7.84–8.02 (m, 3H, arom), 8.09 (d, J = 7.8 Hz, 1H, arom), 8.13 (s, 1H, H-6 arom), 9.00 (brs, 1H, triazine), 14.39 (brs, 1H, SO2NH) ppm; 13C-NMR (125 MHz, DMSO-d6): δ 18.2, 37.1, 55.2, 110.9, 113.7, 115.4, 115.8, 117.7, 122.0, 122.7, 124.9, 125.9, 126.6, 128.4, 128.7, 130.3, 131.5, 132.7, 133.1, 133.4, 136.3, 136.5, 137.8, 142.7, 150.9, 168.8 ppm; anal. C 59.56, H 4.09, N 9.89% calcd. for C28H23ClN4O3S2, C 59.72, H 4.12, N 9.95%.
4-Chloro-2-[(3,5-dimethylizoxazol-4-yl)methylthio]-N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]-5-methyl-benzenesulfonamide (39). Starting from 22 (0.404 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 25 h, the crude 39 (0.380 g) was obtained. The crude 39 was purified by heating under reflux with EtOH and the title compound 39 (0.178 g, 33%) was isolated mp 208–211 °C; IR (KBr) νmax 3435 (NH), 3071, 2963, 2925, 2854, 2737 (C–H), 1551, 1514 (C=C, C=N), 1326, 1156 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.06 (s, 3H, CH3), 2.11 (s, 3H, CH3), 2.41 (s, 3H, CH3), 4.10 (s, 2H, SCH2), 7.34–7.46 (s, 1H, arom), 7.48–7.66 (m, 2H, arom), 7.72 (s, 1H, H-3, arom), 7.77 (d, J = 7.8 Hz, 1H, arom), 8.09 (s, 1H, H-6, arom), 9.24 (bs, 1H, triazine) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 10.3, 11.2, 19.6, 25.6, 110.3, 115.3, 115.5, 125.6, 130.7, 131.9, 132.0, 133.9, 135.3, 135.3, 135.4, 159.9, 161.9, 163.9, 166.7 ppm; anal. C 51.05, H 3.75, N 13.70% calcd. for C22H19ClFN5O3S2, C 50.81, H 3.68, N 13.47%.
4-Chloro-2-[(3,5-dimethylizoxazol-4-yl)methylthio]-5-methyl-N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]benzenesulfonamide (40). Starting from 22 (0.404 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 45 h, the crude 40 (0.516 g) was obtained. The crude 40 was purified by heating under reflux with EtOH and the title compound 40 (0.250 g, 43%) was isolated mp 218–221 °C; IR (KBr) νmax 3434 (NH), 3072, 2754 (C-H), 1550, 1519 (C=C,C=N), 1325, 1156 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.08 (s, 3H, CH3), 2.14 (s, 3H, CH3), 2.39 (s, 3H, CH3), 4.11 (s, 2H, SCH2), 7.67 (s, 1H, H-6, arom), 7.90 (d, J = 8.3 Hz, 2H, arom), 8.12 (d, 2H, arom), 8.15 (s, 1H, H-6, arom), 9.36 (brs, 1H, triazine) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 10.3, 11.2, 19.6, 25.6, 110.0, 121.1, 123.3, 125.4, 126.5, 126.6, 127.6, 129.9, 130.4, 133.7, 135.2, 137.0, 160.0, 166.9 ppm; anal. C 51.15, H 3.55, N 10.10% calcd. for C23H19ClF3N5O3S2 , C 50.84, H 3.20, N 9.88%.
4-Chloro-2-[(3,5-dimethylizoxazol-4-yl)methylthio]-N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]-5-methyl-benzenesulfonamide (41). Starting from 22 (0.404 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 24 h, the crude 41 (0.435 g) was obtained. The crude 41 was purified by heating under reflux with EtOH and the title compound 41 (0.364 g, 68%) was isolated mp 224–227 °C; IR (KBr) νmax 3418, 3218 (NH), 3071, 2925, 2847, (C–H), 1577, 1541 (C=C, C=N), 1267, 1157 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 2.08 (s, 3H, CH3), 2.13 (s, 3H, CH3), 2.43 (s, 3H, CH3), 3.87 (s, 3H, OCH3), 4.08 (s, 2H, SCH2), 7.03 (d, J = 8.8 Hz, 2H, arom), 7.62 (s, 1H, H-3, arom), 7.86 (d, J = 8.30 Hz, 2H, arom), 8.10 (s, 1H, H-6, arom), 9.03 (brs, 1H, triazine), 14.40 (brs, 1H, SO2NH) ppm; 13C-NMR (125 MHz, DMSO-d6) δ 10.3, 11.2, 19.7, 25.5, 56.5, 110.2, 115.4, 125.0, 130.1, 133.5, 160.0, 166.8 ppm; anal. C 52.03, H 4.20, N 13.39% calcd. for C23H22ClN5O4S2, C 51.92, H 4.17, N 13.16%.
4-Benzylthio-2-chloro-5-{N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-phenylbenzamide (42). Starting from 23 (0.451 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 31 h, the crude 42 (0.486 g) was obtained. The crude 42 was crystallized from MeCN and the title compound 42 (0.383 g, 67%) was isolated mp 283–286 °C; IR (KBr) νmax 3409, 3323 (NH), 2923, 2843 (C–H), 1664 (C=O), 1604, 1576 (C=C, C=N), 1535 (NHdef.), 1315, 1174 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.77 (s, 3H, OCH3), 4.38 (s, 2H, SCH2), 7.04 (d, J = 9.09 Hz, 2H, arom), 7.14–7.22 (m, 4H, arom), 7.32–7.42 (m, 4H, arom), 7.63 (s, 1H, H-3, arom), 7.73 (d, J = 7.67 Hz, 2H, arom), 7.94 (d, J = 8.92 Hz, 2H, arom), 8.26 (s, 1H, H-6, arom), 9.09 (s, 1H, triazine), 10.63 (s, 1H, NHCO), 14.50 (s, 1H, SO2NH) ppm; anal. C 58.03, H 2.99, N 11.18% calcd. for C30H24ClN5O4S2, C 58.29, H 3.19, N 11.33%.
4-Benzylthio-2-chloro-5-{N-[5-(3-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-phenylbenzamide (43). Starting from 23 (0.451 g) and 3-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the crude 43 (0.453 g) was obtained. The crude 43 was purified by heating under reflux with EtOH and the title compound 43 (0.399 g, 71%) was isolated mp 270–272 °C; IR (KBr) νmax 3436, 3326 (NH), 2935 (C–H), 1681 (C=O), 1603, 1554 (C=N, C=C), 1314, 1137 (SO2) cm−1; 1H-NMR (500 MHz, DMSO-d6) δ 3.73 (s, 3H, OCH3), 4.39 (s, 2H, SCH2), 7.11–7.20 (m, 5H, arom), 7.34–7.42 (m, 6H, arom), 7.63–7.68 (m, 4H, H-3, arom), 8.21 (s, 1H, H-6, arom), 9.23 (s, 1H, triazine), 10.54 (s, 1H, NHCO) ppm; anal. C 58.49, H 3.30, N 11.42% calcd. for C30H24ClN5O4S2, C 58.29, H 3.19, N 11.33%.
4-Benzylthio-2-chloro-5-{N-[5-(3,4-dimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-phenylbenzamide (44). Starting from 23 (0.451 g) and 3,4-dimethoxyphenylglyoxal hydrate (0.212 g) after stirring for 32 h, the crude 44 (0.500 g) was obtained. The crude 44 was crystallized from EtOH and the title compound 44 (0.407 g, 68%) was isolated mp 252–253 °C; IR (KBr) νmax 3419, 3328 (NH), 2933 (C–H), 1651 (C=O), 1600, 1540 (C=N, C=C), 1318, 1133 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.72 (s, 3H, OCH3), 3.78 (s, 3H, OCH3), 4.40 (s, 2H, SCH2), 6.99–7.24 (m, 6H, arom), 7.32–7,39 (m, 3H, arom), 7.61–7.71 (m, 5H, arom), 8.25 (s, 1H, arom), 9.17 (s, 1H, triazine), 10.57 (s, 1H, NHCO) ppm; anal. C 57.50, H 4.03, N 10.81% calcd. for C31H26ClN5O5S2, C 57.45, H 4.04, N 10.81%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]sulfamoyl}benzamide (45). Starting from 24 (0.524 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 33 h, the title compound 45 (0.470 g, 73%) was obtained without further purification mp 280–282 °C; IR (KBr) νmax 3429, 3276 (NH), 3065, 2923 (C–H), 1657 (C=O), 1558, 1527 (C=N, C=C), 1310, 1166 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 4.42 (s, 2H, SCH2), 7.17–7.36 (m, 3H, arom), 7.43–7.63 (m, 6H, arom), 7.66–7.75 (m, 4H, arom), 7.93 (d, J = 9.61 Hz, 1H, arom), 8.25 (s, 1H, H-6, arom), 9.28 (s, 1H, triazine), 10.72 (s, 1H, CONH), 14.45 (brs, 1H, SO2NH) ppm; anal. C 54.58, H 3.22, N 11.01% calcd. for C29H20Cl2FN5O3S2, C 54.36, H 3.15, N 10.93%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (46). Starting from 24 (0.524 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 33 h, the crude 46 (0.561 g) was obtained. The crude 46 was purified by heating under reflux with MeCN and the title compound 46 (0.553 g, 80%) was isolated mp 273–275 °C; IR (KBr) νmax 3290 (NH), 3062, 2922 (C–H), 1658 (C=O), 1560, 1529 (C=C, C=N), 1325, 1165 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 4.43 (s, 2H, SCH2), 7.16–7.28 (m, 3H, arom), 7.34–7.46 (m, 6H, H-3, arom), 7.73–7.77 (m, 3H, arom), 7.85–7.89 (m, 2H, arom), 8.09–8.13 (m, 2H, arom), 8.30 (s, 1H, H-6, arom), 9.36 (s, 1H, triazine), 10.80 (s, 1H, NHCO) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 35.9, 121.4, 126.3, 126.4, 126.6, 127.5, 128.0, 128.3, 128.6, 129.0, 129.2, 129.3, 129.6, 130.9, 132.2, 132.8, 133.4, 134.3, 136.2, 136.4, 137.9, 140.8, 155.1, 163.9 ppm; anal. C 52.36, H 3.15, N 10.23% calcd. for C30H20Cl2F3N5O3S2, C 52.18, H 2.92, N 10.14%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}benzamide (47). Starting from 24 (0.524 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 30 h, the crude 47 (0.502 g) was obtained. The crude 47 was purified by heating under reflux with MeCN and the title compound 47 (0.363 g, 56%) was isolated mp 282–284 °C; IR (KBr) νmax 3411 (NH), 2925, 2843 (C–H), 1663 (C=O), 1604, 1577 (C=N, C=C), 1537 (NHdef.), 1314, 1174 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.79 (s, 3H, OCH3), 4.39 (s, 2H, SCH2), 7.03 (d, J = 8.97 Hz, 2H, arom), 7.17–7.23 (m, 3H, arom), 7.32–7.37 (m, 2H, arom), 7.44 (d, J = 8.88 Hz, 2H, arom), 7.64 (s, 1H, H-3, arom), 7.75 (d, J = 8.92 Hz, 2H, arom), 7.94 (d, 2H, arom), 8.27 (s, 1H, H-6, arom), 9.09 (s, 1H, triazine), 10.74 (s, 1H, NHCO) ppm; anal. C 55.01, H 3.46, N 10.58% calcd. for C30H23Cl2N5O4S2, C 55.22, H 3.55, N 10.87%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(3-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (48). Starting from 24 (0.524 g) and 3-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the crude 48 (0.358 g) was obtained. The crude 48 was purified by heating under reflux with MeCN and the title compound 48 (0.356 g, 56%) was isolated mp 256–258 °C; IR (KBr) νmax 3318 (NH), 3088, 2921 (C–H), 1684 (C=O), 1597, 1553 (C=C, C=N), 1311, 1162 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.76 (s, 3H, OCH3), 4.42 (s, 2H, SCH2), 7.20–7.38 (m, 4H, arom), 7.43–7.46 (m, 6H, arom), 7.67–7.75 (m, 4H, arom), 8.26 (s, 1H, H-6, arom), 9.27 (s, 1H, triazine), 10.70 (s, 1H, NHCO) ppm; anal. C 55.36, H 3.60, N 10.80% calcd. for C30H23Cl2N5O4S2, C 55.22, H 3.55, N 10.87%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(3,4-dimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (49). Starting from 24 (0.524 g) and 3,4-dimethoxyphenylglyoxal hydrate (0.212 g) after stirring for 33 h, the crude 49 (0.570 g) was obtained. The crude 49 was crystallized from dry p-dioxane and the title compound 49 (0.264 g, 39%) was isolated: mp: 257–259 °C; IR (KBr) νmax 3435, 3327 (NH), 2921 (C–H), 1656 (C=O), 1600, 1544 (C=C, C=N), 1318, 1132 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.76 (s, 3H, OCH3), 3.78 (s, 3H, OCH3), 4.41 (s, 2H, SCH2), 6.99–7.04 (m, 1H, arom), 7.23–7.26 (m, 3H, arom), 7.39–7.45 (m, 4H, arom), 7.63–7.72 (m, 3H, arom), 8.27 (s, 1H, H-6, arom), 9.19 (s, 1H, triazine), 10.69 (s, 1H, CONH) ppm; anal. C 54.60, H 3.75, N 10.27% calcd. for C31H25Cl2N5O5S2, C 54.55, H 3.69, N 10.26%.
4-Benzylthio-2-chloro-N-(4-chlorophenyl)-5-{N-[5-(3,4,5-trimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (50). Starting from 24 (0.524 g) and 3,4,5-trimethoxyphenylglyoxal hydrate (0.242 g) after stirring for 33 h, the crude 50 (0.385 g) was obtained. The crude 50 was crystallized from dry p-dioxane and the title compound 50 (0.182 g, 47%) was isolated mp 260–262 °C; IR (KBr) νmax 3301, 3102 (NH), 2925, 2853 (C–H), 1682 (C=O), 1596, 1552 (C=C, C=N), 1333, 1131 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.69 (s, 3H, OCH3), 3.79 (s, 6H, 2×OCH3), 4.43 (s, 2H, SCH2), 7.24–7.46 (m, 8H, arom), 7.64–7.73 (m, 4H, arom), 8.22 (s, 1H, H-6, arom), 9.31 (s, 1H, triazine), 10.66 (s, 1H, CONH), 14.5 (brs, 1H, SO2NH) ppm; anal. C 53.58, H 3.56, N 9.49% calcd. for C32H27Cl2N5O6S2, C 53.93, H 3.82, N 9.83%.
4-Benzylthio-2-chloro-5-{N-[5-(3-fluorophenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-(4-methylphenyl)benzamide (51). Starting from 25 (0.504 g) and 3-fluorophenylglyoxal hydrate (0.170 g) after stirring for 33 h, the crude 51 (0.470 g) was obtained. The crude 51 was purified by heating under reflux with EtOH and the title compound 51 (0.322 g, 52%) was isolated mp 266–268 °C; IR (KBr) νmax 3427, 3278 (NH), 3064, 2921 (C–H), 1654, 1528 (C=C, C=N), 1316, 1165 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.30 (s, 3H, CH3), 4.41 (s, 2H, SCH2), 7.17–7.20 (m, 4H, arom), 7.33–7.35 (m, 2H, H-3, arom), 7.45–7.68 (m, 7H, arom), 7.95 (d, J = 9.9 Hz, 1H, arom), 8.23 (s, 1H, H-6, arom), 9.28 (s, 1H, triazine), 10.49 (s, 1H, NHCO), 14.5 (brs, 1H, SO2NH) ppm; anal. C 58.22, H 3.80, N 11.31% calcd. for C30H23ClFN5O3S2, C 58.09, H 3.74, N 11.29%.
4-Benzylthio-2-chloro-N-(4-methylphenyl)-5-{N-[5-(4-trifluoromethylphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (52). Starting from 25 (0.504 g) and 4-trifluoromethylphenylglyoxal hydrate (0.220 g) after stirring for 33 h, the crude 52 (0.560 g) was obtained. The crude 52 was purified by heating under reflux with EtOH and the title compound 52 (0.370 g, 55%) was isolated mp 277–279 °C; IR (KBr) νmax 3343, 3347 (NH), 3091, 2920 (C–H), 1668 (C=O), 1558, 1517 (C=C, C=N), 1316, 1120 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.31 (s, 3H, CH3), 4.43 (s, 2H, SCH2), 7.16–7.21 (m, 5H, arom), 7.34–7.36 (m, 2H, arom), 7.62 (d, J = 8.0 Hz, 2H, arom), 7.72 (s, 1H, H-3, arom), 7.89 (d, J = 8.2 Hz, 2H, arom), 8.11 (d, 2H, arom), 8.28 (s, 1H, H-6, arom), 9.36 (s, 1H, triazine), 10.61 (s, 1H, NHCO), 14.13 (brs, 1H, SO2NH) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 20.8, 35.9, 119.8, 126.3, 126.4, 127.5, 128.3, 128.5, 128.6, 129.2, 129.5, 129.6, 130.9, 132.6, 133.4, 134.4, 136.2, 136.4, 136.5, 136.9, 140.5, 155.0, 163.6 ppm; anal. C 55.28, H 3.10, N 10.09% calcd. for C31H23ClF3N5O3S2, C 55.56, H 3.46, N 10.45%.
4-Benzylthio-2-chloro-5-{N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-(4-methylphenyl)-benzamide (53). Starting from 25 (0.504 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the title compound 53 (0.490 g, 77%) was obtained without further purification mp 255–258 °C; IR (KBr) νmax 3315 (NH), 3087, 2921, 2851 (C–H), 1675 (C=O), 1577, 1540 (C=C, C=N), 1315, 1162 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.31 (s, 3H, CH3), 3.78 (s, 3H, OCH3), 4.40 (s, 2H, SCH2), 7.05 (d, J = 8.3 Hz, 2H, arom), 7.18–7.21 (m, 5H, arom), 7.34–7.36 (m, 2H, arom), 7.62–7.65 (m, 3H, H-3, arom), 7.95 (d, J = 8.3 Hz, 2H, arom), 8.27 (s, 1H, H-6, arom), 9.10 (s, 1H, triazine), 10.57 (s, 1H, NHCO), 14.5 (brs, 1H, SO2NH ) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 20.8, 35.9, 55.7, 115.2, 119.9, 124.5, 127.5, 127.9, 128.6, 129.1, 129.5, 130.5, 131.7, 132.4, 133.3, 133.7, 136.3, 133.7, 136.6, 140.2, 154.2, 163.7, 164.6 ppm; anal. C 58.72, H 4.03, N 10.98% calcd. for C31H26ClN5O4S2, C 58.90, H 4.15, N 11.08%.
4-Benzylthio-2-chloro-5-{N-[5-(3-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-(4-methylphenyl)benzamide (54). Starting from 25 (0.504 g) and 3-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the title compound 54 (0.480 g, 76%) was obtained without further purification mp 237–239 °C; IR (KBr) νmax 3328 (NH), 3029, 2918 (C–H), 1678 (C=O), 1554, 1519 (C=C, C=N), 1314, 1162 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.31 (s, 3H, CH3), 3.77 (s, 3H, OCH3), 4.42 (s, 2H, SCH2), 7.19–7.21 (m, 6H, arom), 7.36–7.46 (m, 4H, arom), 7.58–7.69 (m, 4H, H-3, arom), 8.24 (s, 1H, H-6), 9.28 (s, 1H, triazine), 10.50 (s, 1H, NHCO) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 20.8, 35.9, 55.7, 114.2, 119.9, 120.1, 121.3, 127.5, 127.9, 128.6, 129.2, 129.4, 130.5, 130.9, 132.5, 133.2, 133.7, 136.2, 136.5, 140.3, 154.2, 159.9, 163.5 ppm; anal. C 59.06, H 4.25, N 11.12% calcd. for C31H26ClN5O4S2, C 58.90, H 4.15, N 11.08%.
4-Benzylthio-2-chloro-5-{N-[5-(3,4-dimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-(4-methylphenyl)-benzamide (55). Starting from 25 (0.504 g) and 3,4-dimethoxyphenylglyoxal hydrate (0.212 g) after stirring for 33 h, the crude 55 (0.570 g) was obtained. The crude 55 was purified by heating under reflux with EtOH and the title compound 55 (0.480 g, 72%) was isolated mp 250–252 °C; IR (KBr) νmax 3329, 3093 (NH), 2921, 2852 (C–H), 1640 (C=O), 154, 1518 (C=N, C=C), 1318, 1132 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.30 (s, 3H, CH3), 3.74 (s, 3H, OCH3), 3.79 (s, 3H, OCH3), 4.41 (s, 2H, SCH2), 7.00–7.04 (m, 1H, arom), 7.15–7.25 (m, 5H, arom), 7.37–7.42 (m, 2H, arom), 7.56–7.68 (m, 5H, arom), 8.25 (s, 1H, H-6, arom), 9.19 (s, 1H, triazine), 10.49 (s, 1H, NHCO) ppm; anal. C 56.03, H 4.19, N 10.36% calcd. for C32H28ClN5O5S2, C 56.23, H 4.26, N 10.58%.
4-Benzylthio-2-chloro-N-(4-methylphenyl)-5-{N-[5-(3,4,5-trimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (56). Starting from 24 (0.504 g) and 3,4,5-trimethoxyphenylglyoxal hydrate (0.242 g) after stirring for 33 h, the crude 56 (0.516 g) was obtained. The crude 56 was purified by heating under reflux with EtOH and the title compound 56 (0.464 g, 67%) was isolated mp 259–261 °C; IR (KBr) νmax 3435, 3308 (NH), 3033, 2924 (C–H), 1640 (C=O), 1558, 1522 (C=C, C=N), 1342, 1129 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 2.29 (s, 3H, CH3), 3.71 (s, 3H, OCH3), 3.80 (s, 6H, 2×OCH3), 4.43 (s, 2H, SCH2), 7.14–7.26 (m, 5H, arom), 7.40–7.63 (m, 7H, H-3, arom), 8.20 (s, 1H, H-6, arom), 9.32 (s, 1H, triazine), 10.45 (s, 1H, NHCO), 14.20 (brs, 1H, SO2NH) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 20.8, 35.9, 56.2, 60.5, 106.6, 119.8, 127.4, 127.6, 127.7, 128.7, 129.3, 129.4, 130.0, 132.3, 133.2, 133.9, 136.2, 136.4, 140.4, 143.5, 153.5, 154.5, 163.4 ppm; anal. C 56.89, H 4.02, N 9.95% calcd. for C33H30ClN5O6S2, C 57.26, H 4.37, N 10.12%.
4-Benzylthio-2-chloro-N-(4-methoxyphenyl)-5-{N-[5-(4-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}benzamide (57). Starting from 26 (0.520 g) and 4-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the title compound 57 (0.571 g, 88%) was obtained without further purification mp 270–272 °C; IR (KBr) νmax 3331 (NH), 3092, 2932, 2835 (CH2, CH3), 1676 (C=O), 1608, 1543 (C=N, C=C), 1313, 1162 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.77 (s, 3H, OCH3), 3.79 (s, 3H, OCH3), 4.39 (s, 2H, SCH2), 6.96 (d, J = 9.0 Hz, 2H, arom), 7.05 (d, J = 9.00 Hz, 2H, arom), 7.17–7.20 (m, 3H, arom), 7.33–7.35 (m, 2H, arom), 7.63–7.68 (m, 3H, arom), 7.95 (d, J = 8.8 Hz, 2H, arom), 8.26 (s, 1H, H-6, arom), 9.10 (s, 1H, triazine), 10.49 (s, 1H, NHCO) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 35.8, 55.5, 55.9, 114.3, 115.3, 121.4, 124.5, 127.5, 127.9, 128.6, 129.1, 130.5, 131.7, 132.2, 132.5, 133.8, 136.3, 140.1, 156.0, 163.5, 164.6 ppm; anal. C 57.50, H 4.09, N 10.85% calcd. for C31H26ClN5O5S2, C 57.44, H 4.04, N 10.81%.
4-Benzylthio-2-chloro-N-(4-methoxyphenyl)-5-{N-[5-(3-methoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}benzamide (58). Starting from 26 (0.520 g) and 3-methoxyphenylglyoxal hydrate (0.182 g) after stirring for 33 h, the crude 58 (0.550 g) was obtained. The crude 58 was purified by heating under reflux with EtOH and the title compound 58 (0.470 g, 72%) was isolated mp 242–244 °C; IR (KBr) νmax 3333 (NH), 3083, 2922 (CH2, CH3), 2360 (OCH3), 1679 (C=O), 1600, 1552, 1510 (C=N, C=C), 1307, 1162 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.77 (s, 6H, 2×OCH3), 4.41 (m, 2H, SCH2), 6.96 (d, J = 8.8 Hz, 2H, arom), 7.19–7.21 (m, 4H, arom), 7.35–7.45 (m, 4H, arom), 7.59–7.68 (m, 4H, arom), 8.23 (s, 1H, arom), 9.27 (s, 1H, triazine), 10.43 (s, 1H, NHCO) ppm; 13C-NMR (50 MHz, DMSO-d6) δ 35.9, 55.5, 55.7, 114.2, 120.0, 121.3, 121.4, 127.5, 128.0, 128.6, 129.2, 130.5, 130.9, 132.2, 132.5, 133.7, 136.2, 140.2, 156.0, 160.0, 163.2 ppm; anal. C 57.32, H 3.88, N 10.70% calcd. for C31H26ClN5O5S2, C 57.44, H 4.04, N 10.81%.
4-Benzylthio-2-chloro-5-{N-[5-(3,4-dimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-N-(4-methoxyphenyl)-benzamide (59). Starting from 26 (0.520 g) and 3,4-dimethoxyphenylglyoxal hydrate (0.212 g) after stirring for 33 h, the title compound 59 (0.625 g, 92%) was obtained without further purification mp 269–271 °C; IR (KBr) νmax 3393, 3262 (NH), 2924, 2851 (C–H), 1679 (C=O), 1543, 1514 (C=C, C=N), 1312, 1126 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.76-3.78 (m, 9H, 3×OCH3), 4.41 (s, 2H, SCH2), 6.92–7.06 (m, 3H, arom), 7.22–7.25 (m, 3H, H-3, arom), 7.38–7.49 (m, 2H, arom), 7.59–7.67 (m, 5H, arom), 8.26 (s, 1H, H-6, arom), 9.19 (s, 1H, triazine), 10.43 (s, 1H, NHCO) ppm; 13C-NMR (50 MHz. DMSO-d6) δ 35.9, 55.5, 55.9, 111.5, 112.3, 114.2, 121.3, 124.5, 127.6, 127.7, 128.7, 129.2, 132.2, 132.3, 133.8, 136.1, 136.2, 140.4, 149.3, 154.5, 156.0, 163.3 ppm; anal. C 56.52, H 4.05, N 10.22% calcd. for C32H28ClN5O6S2, C 56.68, H 4.17, N 10.33%.
4-Benzylthio-2-chloro-N-(4-methoxyphenyl)-5-{N-[5-(3,4,5-trimethoxyphenyl)-1,2,4-triazin-3-yl]sulfamoyl}-benzamide (60). Starting from 26 (0.520 g) and 3,4,5-trimethoxyphenylglyoxal hydrate (0.242 g) after stirring for 33 h, the crude 60 (0.610 g) was obtained. The crude 60 was purified by heating under reflux with EtOH and the title compound 60 (0.519 g, 73%) was isolated mp 263–265 °C; IR (KBr) νmax 3309 (NH), 2924, 2834 (C–H), 1639 (C=O), 1558, 1513 (C=N, C=C), 1334, 1132 (SO2) cm−1; 1H-NMR (200 MHz, DMSO-d6) δ 3.71-3.79 (m, 12H, 4×OCH3), 4.42 (s, 2H, SCH2), 6.92 (d, J = 8.9 Hz, 2H, arom), 7.23–7.26 (m, 3H, arom), 7.40–7.46 (m, 4H, arom), 7.53–7.62 (m, 3H, arom), 8.19 (s, 1H, H-6, arom), 9.32 (s, 1H, triazine), 10.38 (s, 1H, NHCO) ppm; anal. C 55.84, H 3.88, N 9.55% calcd. for C33H30ClN5O7S2, C 55.97, H 4.27, N 9.89%.

3.3. X-ray Structure Determination

Crystals of 59 were grown from DMF solution. All specimens obtained were in form of very thin needles, having poor diffraction power. The copper radiation (almost no diffraction was observed with the Mo lamp) and long exposure times (4 min) were applied. The obtained signal to noise ratio was still smaller than usually, so final Rint and R1 indices are above the regular standards for small molecules. Nevertheless, structure solution is with no doubts generally correct. Diffraction intensity data were collected on an IPDS 2T dual-beam diffractometer (STOE & Cie GmbH, Darmstadt, Germany) at 120.(2) K with Cu-Kα radiation of a microfocus X-ray source (50 kV, 0.6 mA, λ = 154.186 pm, GeniX 3D Cu High Flux, Xenocs, Sassenage, France), The crystal was thermostated in nitrogen stream at 120 K using CryoStream-800 device (Oxford CryoSystem, Oxford, UK) during the entire experiment. Data collection and data reduction were controlled by X-Area 1.75 program [43]. An absorption correction was performed on the integrated reflections by a combination of frame scaling, reflection scaling and a spherical absorption correction. Outliers have been rejected according to Blessing’s method [44].
The structure was solved using direct methods with SHELXS-13 program and refined by SHELXL-2013 [45] program run under control of WinGx [46]. All C-H type hydrogen atoms were attached at their geometrically expected positions and refined as riding on heavier atoms with the usual constraints. The N-H hydrogen atoms were found in the differential Fourier electron density map and were refined without constraints.
Crystallographic data for the analysis have been deposited with the Cambridge Crystallographic data Centre, CCDC reference number is 1471508. Copies of this information may be obtained free of charge from CCDC, 12 Union Road, Cambridge, CB21EZ, UK (Fax: +44-1223-336033; E-mail: [email protected] or www: http://www.ccdc.cam.ac.uk).

3.4. Cell Culture and Cell Viability Assay

All chemicals, if not stated otherwise, were obtained from Sigma-Aldrich (St. Louis, MO, USA). The MCF-7 cell line was purchased from Cell Lines Services (Eppelheim, Germany), the HeLa and HCT-116 cell lines were obtained from the Department of Microbiology, Tumor and Cell Biology, Karolinska Institute (Stockholm, Sweden). Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum, 2 mM glutamine, 100 units/mL penicillin, and 100 μg/mL streptomycin. Cultures were maintained in a humidified atmosphere with 5% CO2 at 37 °C in an incubator (HeraCell, Heraeus, Langenselbold, Germany).
Cell viability was determined using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazoliumbromide) assay. Stock solutions of the studied compounds were prepared in 100% DMSO. Working solutions were prepared by diluting the stock solutions with DMEM medium, the final concentration of DMSO did not exceed 0.5% in the treated samples. Cells were seeded in 96-well plates at a density of 5 × 103 cells/well and treated for 72 h with the examined compounds in the concentration range 1–100 μM (1, 10, 25, 50 and 100 μM). Following treatment, MTT (0.5 mg/mL) was added to the medium and cells were further incubated for 2 h at 37 °C. Cells were lysed with DMSO and the absorbance of the formazan solution was measured at 550 nm with a plate reader (1420 multilabel counter, Victor, Jügesheim, Germany). The optical density of the formazan solution was measured at 550 nm with a plate reader (Victor 1420 multilabel counter). The experiment was performed in triplicate. Values are expressed as the mean ± SD of at least three independent experiments.

3.4.1. Cell Morphology

The HCT-116, HeLa and MCF-7cells were seeded on 24-well plates (5 × 104/per well) in 300 μL of medium and incubated with 37 (HCT-116, HeLa) or 46 (MCF-7) for 24 h. Cells incubated with increasing concentrations of compounds (25–100 μM) were observed after 24 h using light microscope Axiovert 200 (Zeiss, Oberkochen, Germany).

3.4.2. Cell Cycle Analysis

Cells were coincubated with compounds and a reference drug (cisplatin) for 24 h, respectively. Following treatment, cells were fixed in cold 70% ethanol for 30 min. Fixed cells were stained for 15 min in buffer containing 500 μg/mL RNAse A (EURx, Gdansk, Poland) and 2.5 μg/mL of DAPI (Sigma-Aldrich, Munich, Germany). The fluorescence of DAPI was measured at 440 ± 40 nm with a flow cytometer (LSR II, BD Biosciences, Mountain View, CA, USA). Each experiment was performed at least in triplicate. Data were analyzed off-line using the BD FACSDiva (BD Biosciences).

3.4.3. Mitochondrial Membrane Potential (Δψm) Analysis

The HCT-116, HeLa nad MCF-7 cells were coincubated with trazines and a reference drug (cisplatin) for 24 h, respectively. Half an hour before the end of 24 h-treatment 2.5 μg/mL of JC-1 (5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide) was added to the cell medium. As a negative control 100 μM CCCP (carbonyl cyanide 3-chlorophenylhydrazone) was used 15 min before JC-1 treatment. Following treatment cells were twice washed with PBS and analyzed with flow cytometer (LSR II, BD Biosciences).
Green and red fluorescence were observed using FL-1 (530 ± 30 nm) and FL-2 (585 ± 42 nm) channels, respectively. Percentage of cell population in red or green fluorescence gate indicated the level of Δψm. Decreasing red fluorescence cell population indicated lower Δψm.

3.4.4. Translocation of Phosphatidylserine to Outer Leaflet of Cell Membrane

Analysis was performed using FITC-conjugated annexin-V (Annexin V-FITC Apoptosis Kit I, BD Biosciences) according to the manufacturer’s instructions (BD Biosciences) as described previously [47]. HCT-116, HeLa and MCF-7 cells were coincubated for 24 h with traizines 37 and 46, respectively. Following treatment cells were washed in PBS, centrifuged and then resuspended in binding buffer. Afterwards, the cells were incubated for 15 min at 37 °C with FITC-conjugated annexin-V and propidium iodide. The samples were then analyzed using a LSR II flow cytometer (BD Biosciences) using 525 ± 20 nm (Annexin V-FITC) and 570 ± 26 nm (PI). The subpopulations were identified according to their fluorescence: PI-low/FITC-low (live cells), PI-high/FITC-low (necrotic cells), PI-low/FITC-high (early apoptotic cells), PI-high/FITC-high (late apoptotic cells).

3.4.5. Caspase Activity Determination

Caspase activity was determined with the FLICA Apoptosis Detection Kit (Immunochemistry Technologies, Bloomington, MN, USA) according to the manufacturer’s instructions. Cells were treated with the examined compounds for the indicated periods of time after which cells were collected and suspended in a buffer containing the caspase inhibitor—a carboxyfluorescein-labeled fluoromethyl ketone peptide. Cells were subsequently incubated for 1 h 37 °C under 5% CO2 cells and next were washed with washing buffer. The fluorescence intensity of fluorescein was determined with flow cytometry (BD FACSCalibur) and caspase activity was determined as the amount of fluorescence emitted from caspase inhibitors bound to the caspases.

3.4.6. Statistical Analysis

Statistical differences between control and triazine coincubated cells were determined using the t-student test, after checking the normal distribution of data. The analyses were performed using 6 to 15 replicates run in at least three independent experiments.

3.5. Metabolic Stability

Stock solutions of studied compounds were prepared at concentration of 10 mM in DMSO. Working solutions were prepared by dilution of stock with reaction buffer or acetonitrile, final concentration of organic solvent did not exceed 1%. Incubation mixture contained 10 μM of a studied derivative, 1 mM of NADPH (Sigma-Aldrich) and 0.5 mg/mL of human liver microsomes (HLM, Sigma-Aldrich) in potassium phosphate buffer (0.1 M, pH 7.4). Incubation was carried out in thermostat at 37 °C and started by addition of studied compound. 50 μL samples were taken after 5, 15, 30, 45 and 60 min. Enzymatic reaction was terminated by the addition of the equal volume of ice-cold acetonitrile containing 10 μM of alprazolam (Sigma-Aldrich) serving as internal standard (IS). Control incubations were performed without NADPH to assess chemical instability. After collection, samples were immediately centrifuged (10 min, 10,000 rpm) and resulted supernatant was directly analyzed or kept in −80 °C until LC-MS analysis. Natural logarithm of a compound over IS peak area ratio was plotted versus incubation time. Metabolic half-time (t1/2) was calculated from the slope of the linear regression, as reported in [48].
LC-MS analysis was performed on an Agilent 1260 system coupled to SingleQuad 6120 mass spectrometer (Agilent Technologies, Santa Clara, CA, USA). Ascentis Express ES-CN (2.1 mm × 100 mm, 2.7 μm, Supelco Analytical, Bellefonte, PA, USA) was used in reversed-phase mode with gradient elution starting with 50% of phase A (10 mM ammonium formate in water) and 50% of phase B (10 mM ammonium formate in acetonitrile-water mixture, 95:5 v/v). The amount of phase B was linearly increased to 100% in 5 min. Total analysis time was 10 min at 25 °C, flow rate was 0.4 mL/min and the injection volume was 20 μL. The mass spectrometer was equipped with electrospray ion source and operated in positive ionization. Mass analyzer was set individually to each compound to detect [M + H]+ pseudomolecular ions. MSD parameters of the ESI source were as follows: nebulizer pressure 50 psig (N2), drying gas 13 mL/min (N2), drying gas temperature 300 °C, capillary voltage 3.5 kV, fragmentor voltage 100 V.

4. Conclusions

We have synthesized a series of novel 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamide derivatives using aminoguanidines and appropriate phenylglyoxal hydrates in glacial acetic acid. The molecular structures of novel compounds were confirmed by NMR, IR, and also by single crystal X-ray structure analysis for representative compound 59. All N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides were tested for their in vitro cytotoxic activity against the HCT-116, HeLa and MCF-7 cell lines. The most active compounds 37 and 38 belonged to the 1-naphthyl series (R2 = 1-naphthyl) and showed outstanding average cytotoxic activity against all tested cell lines with special regard to the HCT-116 (IC50: 36–38 μM) and HeLa (IC50: 34–42 μM) cell lines. The MCF-7 cell line occurred less sensitive to 1,2,4-triazine derivatives, the strongest cytotoxic potency showed 46. The ability to induce apoptosis in cancer cells was studied through biochemical markers, such as: DNA fragmentation, loss of mitochondrial membrane potential, phosphatidylserine translocation and caspase activation. Apoptotic-like changes in morphology of HCT-116, HeLa, MCF-7 cell lines, significant accumulation of the HCT-116, HeLa and MCF-7 population in the sub-G1 phase and DNA fragmentation, loss of Δψm, appearance of increased population of early and late apoptotic cells, induction of caspase activity indicated apoptosis of the cells in presence of 37 and 46. In vitro tests for metabolic stability in the presence of human liver microsomes showed that both R2 and Ar substituents (4-CF3-C6H4) substantially increased the metabolic stability of 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides. The results pointed to 37 as a hit compound with a good biological response against HCT-116 (IC50 = 36 μM) and HeLa (IC50 = 34 μM) cell lines with apoptosis-inducing activity, and satisfactory metabolic stability.

Supplementary Materials

Supplementary materials can be accessed at: https://www.mdpi.com/1420-3049/21/6/808/s1.

Acknowledgments

The project was financed by National Science Centre based on the decision number DEC-2013/09/B/NZ7/00048.

Author Contributions

B.Ż. and J.S. created the concept, and designed the study. A.P. and K.S. performed chemical research. B.Ż. and J.S. analyzed both chemical and biological data and wrote the manuscript together. A.K. and G.S. tested the biological activity and made cytometric analysis of the compounds and interpreted the results. J.C. performed crystallographic analysis. M.B, S.U. and T.B. carried out all experiments concerning the metabolic stability. All Authors read and approved the final version of the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Singla, P.; Luxami, V.; Paul, K. Triazine as a promising scaffold for its versatile biological behavior. Eur. J. Med. Chem. 2015, 102, 39–57. [Google Scholar] [CrossRef] [PubMed]
  2. Costanzo, A.; Guerrini, G.; Ciciani, G.; Bruni, F.; Selleri, S.; Costa, B.; Martini, C.; Lucacchini, A.; Aiello, P.M.; Ipponi, A. Benzodiazepine receptor ligands. 4. Synthesis and pharmacological evaluation of 3-heteroaryl-8-chloropyrazolo[5,1-c][1,2,4]benzotriazine 5-oxides. J. Med. Chem. 1999, 42, 2218–2226. [Google Scholar] [CrossRef] [PubMed]
  3. Irannejad, H.; Amini, M.; Khodagholi, F.; Ansari, N.; Tusi, S.K.; Sharifzadeh, M.; Shafiee, A. Synthesis and in vitro evaluation of novel 1,2,4-triazine derivatives as neuroprotective agents. Bioorg. Med. Chem. 2010, 18, 4224–4230. [Google Scholar] [CrossRef] [PubMed]
  4. Bennett, G.B.; Babington, R.G.; Deacon, M.A.; Eden, P.L.; Kerestan, S.P.; Leslie, G.H.; Ryan, E.A.; Mason, R.B.; Minor, H.E. A potent, new, sedative-hypnotic agent: 5,7-dihydro-5,5,7,7-tetramethyl-3-(3-nitrophenyl)furo[3,4-e]-as-triazine 4-oxide. J. Med. Chem. 1981, 24, 490–496. [Google Scholar] [CrossRef] [PubMed]
  5. Guerrini, G.; Ciciani, G.; Cambi, G.; Bruni, F.; Selleri, S.; Melani, F.; Montali, M.; Martini, C.; Ghelardini, C.; Norcini, M.; et al. Novel 3-aroylpyrazolo[5,1-c][1,2,4]benzotriazine 5-oxides 8-substituted, ligands at GABAA/benzodiazepine receptor complex: Synthesis, pharmacological and molecular modeling studies. Bioorg. Med. Chem. 2008, 16, 4471–4489. [Google Scholar] [CrossRef] [PubMed]
  6. Szárics, E.; Riedl, Z.; Nyikos, L.; Hajós, G.; Kardos, J. Interaction of novel condensed triazine derivatives with central and peripheral type benzodiazepine receptors: Synthesis, in vitro pharmacology and modelling. Eur. J. Med. Chem. 2006, 41, 445–456. [Google Scholar] [CrossRef] [PubMed]
  7. Congreve, M.; Andrews, S.P.; Doré, A.S.; Hollenstein, K.; Hurrell, E.; Langmead, C.J.; Mason, J.S.; Ng, I.W.; Tehan, B.; Zhukov, A.; et al. Discovery of 1,2,4-triazine derivatives as adenosine A(2A) antagonists using structure based drug design. J. Med. Chem. 2012, 55, 1898–1903. [Google Scholar] [CrossRef] [PubMed]
  8. Trepanier, D.L.; Shriver, K.L.; Eble, J.N. Aryl-substituted triazines with antidepressant activity. J. Med. Chem. 1969, 12, 257–260. [Google Scholar] [CrossRef] [PubMed]
  9. Liu, C.; Lin, J.; Wrobleski, S.T.; Lin, S.; Hynes, J.; Wu, H.; Dyckman, A.J.; Li, T.; Wityak, J.; Gillooly, K.M.; et al. Discovery of 4-(5-(cyclopropylcarbamoyl)-2-methylphenylamino)-5-methyl-N-propylpyrrolo[1,2-f][1,2,4]triazine-6-carboxamide (BMS-582949), a clinical p38α MAP kinase inhibitor for the treatment of inflammatory diseases. J. Med. Chem. 2010, 53, 6629–6639. [Google Scholar] [CrossRef] [PubMed]
  10. Sztanke, K.; Pasternak, K.; Rajtar, B.; Sztanke, M.; Majek, M.; Polz-Dacewicz, M. Identification of antibacterial and antiviral activities of novel fused 1,2,4-triazine esters. Bioorg. Med. Chem. 2007, 15, 5480–5486. [Google Scholar] [CrossRef] [PubMed]
  11. Kusch, P.; Deininger, S.; Specht, S.; Maniako, R.; Haubrich, S.; Pommerening, T.; Lin, P.K.; Hoerauf, A.; Kaiser, A. In vitro and in vivo antimalarial activity assays of seeds from Balanites aegyptiaca: Compounds of the extract show growth inhibition and activity against Plasmodial Aminopeptidase. J. Parasitol. Res. 2011, 2011. [Google Scholar] [CrossRef] [PubMed]
  12. Morrey, J.D.; Smee, D.F.; Sidwell, R.W.; Tseng, C. Identification of active antiviral compounds against a New York isolate of West Nile virus. Antivir. Res. 2002, 55, 107–116. [Google Scholar] [CrossRef]
  13. Centorino, M.B.; Catalano, G.; Catalano, M.C. Lamotrigine induced whole body tics: A case report and literature review. Curr. Drug Saf. 2016, 11, 189–191. [Google Scholar] [CrossRef] [PubMed]
  14. Grennan, D.M.; McLeod, M.; Watkins, C.; Dick, W.C. Clinical assessment of azapropazone in rheumatoid arthritis. Curr. Med. Res. Opin. 1976, 4, 44–49. [Google Scholar] [CrossRef] [PubMed]
  15. Patil, U.N.; Jambulingappa, K.L. A combination strategy of Ceftriaxone, Sulbactam and disodium edetate for the treatment of multi-drug resistant (MDR) septicaemia: A retrospective, observational study in Indian Tertiary Care Hospital. J. Clin. Diagn. Res. 2015, 9, FC29–FC32. [Google Scholar] [CrossRef] [PubMed]
  16. Ucherek, M.M.; Wroblewska, J.; Modzelewska-Banachiewicz, B.; Gospodarek, E. Biological activity of methyl 2-[5-oxo-3,4-di-(2-pirydyl)-1,4,5,6-tetrahydro-1,2,4-triazine-6-ylidene]acetate. Acta Polon. Pharm. Drug Res. 2008, 65, 789–791. [Google Scholar]
  17. Fahmy, U.A. Nanoethosomal transdermal delivery of vardenafil for treatment of erectile dysfunction: Optimization, characterization, and in vivo evaluation. Drug Des. Dev. Ther. 2015, 9, 6129–6137. [Google Scholar] [CrossRef] [PubMed]
  18. Hay, M.P.; Pruijn, F.B.; Gamage, S.A.; Liyanage, H.D.; Kovacs, M.S.; Patterson, A.V.; Wilson, W.R.; Brown, J.M.; Denny, W.A. DNA-targeted 1,2,4-benzotriazine 1,4-dioxide: Potent analogues of the hypoxia selective cytotoxin tirapazamine. J. Med. Chem. 2004, 47, 475–488. [Google Scholar] [CrossRef] [PubMed]
  19. Walters, T.R.; Aur, R.J.A.; Hernandez, K.; Vietti, T.; Pinkel, D. 6-Azauridine in combination chemotherapy of childhood acute myelocytic leukemia. Cancer 1972, 29, 1057–1060. [Google Scholar] [CrossRef]
  20. Saad, H.A.; Moustafa, A.H. Synthesis and anticancer activity of some new S-glycosyl and S-alkyl 1,2,4-triazinone derivatives. Molecules 2011, 16, 5682–5700. [Google Scholar] [CrossRef] [PubMed]
  21. Mohareb, M.R.; Mohamed, A.A. The Reaction of cyanoacetylhydrazine with ω-bromo(4-methyl)acetophenone: Synthesis of heterocyclic derivatives with antitumor activity. Molecules 2010, 15, 3602–3617. [Google Scholar] [CrossRef] [PubMed]
  22. Żołnowska, B.; Sławiński, J.; Belka, M.; Bączek, T.; Kawiak, A.; Chojnacki, J.; Pogorzelska, A.; Szafrański, K. Synthesis, molecular structure, metabolic stability and QSAR studies of a novel series of anticancer N-acylbenzenesulfonamides. Molecules 2015, 20, 19101–19129. [Google Scholar] [CrossRef] [PubMed]
  23. Żołnowska, B.; Sławiński, J.; Pogorzelska, A.; Chojnacki, J.; Vullo, D.; Supuran, C.T. Carbonic anhydrase inhibitors. Synthesis, and molecular structure of novel series N-substituted N’-(2-arylmethylthio-4-chloro-5-methylbenzenesulfonyl)guanidines and their inhibition of human cytosolic isozymes I and II and the transmembrane tumor-associated isozymes IX and XII. Eur. J. Med. Chem. 2014, 71, 135–147. [Google Scholar]
  24. Sławiński, J.; Brzozowski, Z.; Żołnowska, B.; Szafrański, K.; Pogorzelska, A.; Vullo, D.; Supuran, C.T. Synthesis of a new series of N4-substituted 4-(2-aminoethyl)benzenesulfonamides and their inhibitory effect on human carbonic anhydrase cytosolic isozymes I and II and transmembrane tumor-associated isozymes IX and XII. Eur. J. Med. Chem. 2014, 84, 59–67. [Google Scholar] [CrossRef] [PubMed]
  25. Sławiński, J.; Żołnowska, B.; Orlewska, C.; Chojnacki, J. Synthesis and molecular structure of novel 2-(alkylthio)-4-chloro-N-(4,5-dihydro-5-oxo-1H-1,2,4-triazol-3-yl)-5-methylbenzenesulfonamides with potential anticancer activity. Mon. Chem. 2012, 143, 1705–1718. [Google Scholar] [CrossRef] [PubMed]
  26. Brożewicz, K.; Sławiński, J. Synthesis and in vitro activity of novel 2-(benzylthio)-4-chloro-5-(1,3,4-oxadiazol-2-yl)benzenesulfonamide derivatives. Mon. Chem. 2012, 143, 975–984. [Google Scholar] [CrossRef] [PubMed]
  27. Brożewicz, K.; Sławiński, J. 1-(2-Mercaptobenzenesulfonyl)-3-hydroxyguanidines—Novel potent antiproliferatives, synthesis and in vitro biological activity. Eur. J. Med. Chem. 2012, 55, 384–394. [Google Scholar] [CrossRef] [PubMed]
  28. Sławiński, J.; Brożewicz, K.; Fruziński, A.; Główka, M.L. Synthesis and antitumor activity of novel N′-(2-benzylthiobenzenesulfonyl)-1H-pyrazole-1-amidine derivatives. Heterocycles 2011, 83, 1093–1109. [Google Scholar] [CrossRef]
  29. Sławiński, J.; Gdaniec, M. Synthesis, molecular structure and in vitro antitumor activity of new 4-chloro-2-mercaptobenzenesulfonamide derivatives. Eur. J. Med. Chem. 2005, 40, 377–389. [Google Scholar] [CrossRef] [PubMed]
  30. Nepali, K.; Sharma, S.; Sharma, M.; Bedi, P.M.S.; Dhar, K.L. Rational approaches, design strategies, structure activity relationship and mechanistic insights for anticancer hybrids. Eur. J. Med. Chem. 2014, 77, 422–487. [Google Scholar] [CrossRef] [PubMed]
  31. Brzozowski, Z.; Sławiński, J. 1,1-Dioxo-1,4,2-benzodithiazine derivatives. I. Synthesis of various 7-carboxy-3-mercapto-1,1-dioxo-1,4,2-benzodithiazine. Acta Polon. Pharm. 1984, 41, 5–13. [Google Scholar]
  32. Brzozowski, Z.; Sławiński, J. 1,1-Dioxo-1,4,2-benzodithiazine derivatives. II Synthesis of some 3-mercapto-1,1-dioxo-1,4,2-benzodithiazine derivatives. Acta Polon. Pharm. 1984, 41, 133–139. [Google Scholar]
  33. Sławiński, J. Syntheses and Some Reactions of 3-Amino-6-choro-7-methyl-1,1-dioxo-1,4,2-benzodithiazine. Pol. J. Chem. 2001, 75, 1309–1316. [Google Scholar]
  34. Brzozowski, Z.; Sławiński, J. Syntheses and some reactions of 3-amino-6-chloro-7-methyl-1,1-dioxo-1,4,2-benzodithiazine. Pol. J. Chem. 2007, 81, 1419–1426. [Google Scholar]
  35. Sławiński, J.; Żołnowska, B.; Pirska, D.; Kędzia, A.; Kwapisz, E. Synthesis of 2-(4-chloro-2-mercaptobenzenesulfonyl)-3,5-dihydroxy-1,1-dioxo-2H-1,2,4,6-thiatriazine derivatives with potential anti-HIV and anticancer activities. J. Enzyme Inhib. Med. Chem. 2013, 28, 41–51. [Google Scholar] [CrossRef] [PubMed]
  36. Sławiński, J.; Pogorzelska, A.; Żołnowska, B.; Kędzia, A.; Ziółkowska-Klinkosz, M.; Kwapisz, E. Synthesis and anti-yeast evaluation of novel 2-alkylthio-4-chloro-5-methyl-N-[imino-(1-oxo-(1H)-phthalazin-2-yl)methyl]benzenesulfonamide derivatives. Molecules 2014, 19, 13704–13723. [Google Scholar] [CrossRef] [PubMed]
  37. Singh, M.; Modi, A.; Narayan, G.; Singh, S.K. Benzothiazole derivatives bearing amide moiety: Potential cytotoxic and apoptosis-inducing agents against cervical cancer. Anticancer Drugs 2016, 27, 519–532. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Y.; Perchellet, E.M.; Ward, M.M.; Lou, K.; Hua, D.H.; Perchellet, J.P. Rapid collapse of mitochondrial transmembrane potential in HL-60 cells and isolated mitochondria treated with anti-tumor 1,4-anthracenediones. Anticancer Drugs 2005, 16, 953–967. [Google Scholar] [CrossRef] [PubMed]
  39. Kassel, D.B. Applications of high-throughput ADME in drug discovery. Curr. Opin. Chem. Biol. 2004, 8, 339–345. [Google Scholar] [CrossRef] [PubMed]
  40. Korfmacher, W.A. Advances in the integration of drug metabolism into the lead optimization paradigm. Mini Rev. Med. Chem. 2009, 9, 703–716. [Google Scholar] [CrossRef] [PubMed]
  41. Baranczewski, P.; Stanczak, A.; Sundberg, K.; Svensson, R.; Wallin, A.; Jansson, J.; Garberg, P.; Postlind, H. Introduction to in vitro estimation of metabolic stability and drug interactions of new chemical entities in drug discovery and development. Pharmacol. Rep. 2006, 58, 453–472. [Google Scholar] [PubMed]
  42. Zaretzki, J.; Matlock, M.; Swamidass, S.J. XenoSite: Accurately predicting CYP-mediated sites of metabolism with neural networks. J. Chem. Inf. Model. 2013, 53, 3373–3383. [Google Scholar] [CrossRef] [PubMed]
  43. STOE & Cie GmbH. X-Area 1.75, Software Package for Collecting Single-Crystal Data on STOE Area-Detector Diffractometers, for Image Processing, Scaling Reflection Intensities and for Outlier Rejection; STOE & Cie: Darmstadt, Germany, 2015. [Google Scholar]
  44. Blessing, R.H. Routines employed in the SORTAV program for identifying and downweighting mismeasured outliers in merging multiply measured data sets are described. J. Appl. Cryst. 1997, 30, 421–426. [Google Scholar] [CrossRef]
  45. Sheldrick, G.M. A short history of SHELX. Acta Crystallogr. 2008, A64, 112–122. [Google Scholar] [CrossRef] [PubMed]
  46. Farrugia, L.J. WinGX and ORTEP for Windows: An update. J. Appl. Cryst. 2012, 45, 849–854. [Google Scholar] [CrossRef]
  47. Stasiłojć, G.; Pinto, S.; Wyszkowska, R.; Wejda, M.; Słomińska, E.M.; Filipska, M.; Koszałka, P.; Swierczyński, J.; O’Connor, J.E.; Bigda, J.J. U937 variant cells as a model of apoptosis without cell disintegration. Cell. Mol. Biol. Lett. 2013, 18, 249–262. [Google Scholar] [CrossRef] [PubMed]
  48. Obach, R.S. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab. Dispos. 1999, 27, 1350–1359. [Google Scholar] [PubMed]
  • Sample Availability: Samples of the compounds 2122, 2660 are available from the authors.
Figure 1. Structures of known anticancer 1,2,4-triazines (IV) and novel 2-(arylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamide derivatives (VI).
Figure 1. Structures of known anticancer 1,2,4-triazines (IV) and novel 2-(arylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamide derivatives (VI).
Molecules 21 00808 g001
Scheme 1. Synthesis of 1-amino-2-[4-chloro-5-R1-2-(R2-methylthio)benzenesulfonyl]guanidine (1926). Reagents and Conditions: (a) 25% NH3 aq/EtOH, r.t., 144 h; (b) Excess K2CO3, THF, reflux, 24 h; (c) R1CH2Cl, ethanol or water, r.t. or 0 °C, 1–4 h; (d) NH2-NH2 × HCl, dry toluene, reflux 3–9 h.
Scheme 1. Synthesis of 1-amino-2-[4-chloro-5-R1-2-(R2-methylthio)benzenesulfonyl]guanidine (1926). Reagents and Conditions: (a) 25% NH3 aq/EtOH, r.t., 144 h; (b) Excess K2CO3, THF, reflux, 24 h; (c) R1CH2Cl, ethanol or water, r.t. or 0 °C, 1–4 h; (d) NH2-NH2 × HCl, dry toluene, reflux 3–9 h.
Molecules 21 00808 sch001
Scheme 2. Synthesis of 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides 2760.
Scheme 2. Synthesis of 4-chloro-2-(R2-methylthio)-5-R1-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides 2760.
Molecules 21 00808 sch002
Figure 2. View of half of the independent unit (one sulfonamide and two DMF molecules) showing the general arrangement of atoms in 59 and N–H···O type hydrogen bonds.
Figure 2. View of half of the independent unit (one sulfonamide and two DMF molecules) showing the general arrangement of atoms in 59 and N–H···O type hydrogen bonds.
Molecules 21 00808 g002
Figure 3. Morphology of cell lines: HCT-116 (a); HeLa (b) treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 (c) treated with 46 (0–100 μM) and cisplatin (100 μM). Light microscopy photographs, with 10× objective, of cells treated for 24 h.
Figure 3. Morphology of cell lines: HCT-116 (a); HeLa (b) treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 (c) treated with 46 (0–100 μM) and cisplatin (100 μM). Light microscopy photographs, with 10× objective, of cells treated for 24 h.
Molecules 21 00808 g003aMolecules 21 00808 g003b
Figure 4. HCT-116, HeLa and MCF-7 cell cycle 24 h induction with 1,2,4-triazine derivatives (representative results). HCT-116 (a); HeLa (b) treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 (c) treated with 46 (0–100 μM) and cisplatin (100 μM).
Figure 4. HCT-116, HeLa and MCF-7 cell cycle 24 h induction with 1,2,4-triazine derivatives (representative results). HCT-116 (a); HeLa (b) treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 (c) treated with 46 (0–100 μM) and cisplatin (100 μM).
Molecules 21 00808 g004
Figure 5. Mitochondrial membrane potential (Δψm). Cytometric analysis of Δψm with 2.5 μg/mL JC-1 in: HCT-116 (a), HeLa (b) treated with 37 (0–100 μM) 24 h, carbonyl cyanide m-chlorophenylhydrazone (CCCP) (100 μM) for 15 min or cisplatin (100 μM) 24 h; MCF-7 (c) treated with 46 (0–100 μM) 24 h, CCCP (100 μM) for 15 min or cisplatin (100 μM) 24 h. Results represent the mean of number of cells with high Δψm (high JC-1 fluorescence) ± SD (n = 8). Statistically significant differences between treated and control cells are indicated (* p < 0.05; *** p < 0.005).
Figure 5. Mitochondrial membrane potential (Δψm). Cytometric analysis of Δψm with 2.5 μg/mL JC-1 in: HCT-116 (a), HeLa (b) treated with 37 (0–100 μM) 24 h, carbonyl cyanide m-chlorophenylhydrazone (CCCP) (100 μM) for 15 min or cisplatin (100 μM) 24 h; MCF-7 (c) treated with 46 (0–100 μM) 24 h, CCCP (100 μM) for 15 min or cisplatin (100 μM) 24 h. Results represent the mean of number of cells with high Δψm (high JC-1 fluorescence) ± SD (n = 8). Statistically significant differences between treated and control cells are indicated (* p < 0.05; *** p < 0.005).
Molecules 21 00808 g005
Figure 6. Features of dying process of HCT-116 (a); HeLa (b) and MCF-7 (c) cell lines (representative results). Dotblots show cells stained with Annexin V-FITC Apoptosis Kit. HCT-116, HeLa cells treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 cells treated with 46 (0–100 μM) and cisplatin (100 μM) for 24 h. Results show the mean of number of cells in each quadrant Q1, Q2, Q3, Q4 (necrotic cells, late apoptotic cells, alive cells, apoptotic cells, respectively).
Figure 6. Features of dying process of HCT-116 (a); HeLa (b) and MCF-7 (c) cell lines (representative results). Dotblots show cells stained with Annexin V-FITC Apoptosis Kit. HCT-116, HeLa cells treated with 37 (0–100 μM) and cisplatin (100 μM); MCF-7 cells treated with 46 (0–100 μM) and cisplatin (100 μM) for 24 h. Results show the mean of number of cells in each quadrant Q1, Q2, Q3, Q4 (necrotic cells, late apoptotic cells, alive cells, apoptotic cells, respectively).
Molecules 21 00808 g006
Figure 7. Induction of caspase activity in HCT-116, HeLa and MCF-7 cells by 1,2,4-triazine derivatives. HCT-116 (a), HeLa (b) and MCF-7 (c) cells were treated with the indicated concentrations of compounds 37 and 46 for 24 h (a), 72 h (b) and 72 h (c), respectively. Enzyme activity was determined by flow cytometry with the use of a caspase inhibitor, FAM-VAD-FMK. Values represent mean ± SD of three independent experiments.
Figure 7. Induction of caspase activity in HCT-116, HeLa and MCF-7 cells by 1,2,4-triazine derivatives. HCT-116 (a), HeLa (b) and MCF-7 (c) cells were treated with the indicated concentrations of compounds 37 and 46 for 24 h (a), 72 h (b) and 72 h (c), respectively. Enzyme activity was determined by flow cytometry with the use of a caspase inhibitor, FAM-VAD-FMK. Values represent mean ± SD of three independent experiments.
Molecules 21 00808 g007
Figure 8. Sites of metabolism predicted for 34, 35, 37 and 38 by XenoSite software [42]. Yellow color indicates more vulnerability to biotransformation than blue. Some significant differences are additionally pointed out by red arrows.
Figure 8. Sites of metabolism predicted for 34, 35, 37 and 38 by XenoSite software [42]. Yellow color indicates more vulnerability to biotransformation than blue. Some significant differences are additionally pointed out by red arrows.
Molecules 21 00808 g008
Table 1. Crystal data and structure refinement of 59.
Table 1. Crystal data and structure refinement of 59.
Empirical FormulaC38H42ClN7O8S2
Formula weight824.35
Temperature120(2) K
Wavelength1.54186 Å
Crystal systemTriclinic
Space group P 1 ¯
Unit cell dimensionsa = 7.8336(6) Å, α = 84.84(3)°
b = 22.4284(8) Å, β = 86.95(4)°
c = 22.2739(9) Å, γ = 85.13(5)°
Volume3879.4(5) Å3
Z4
Density (calculated)1.411 Mg/m3
Absorption coefficient2.399 mm−1
F(000)1728
Crystal size0.03 × 0.03 × 0.25 mm3
Theta range for data collection1.984 to 64.981°
Index ranges−8 ≤ h ≤ 8, −23 ≤ k ≤ 26, −25 ≤ l ≤ 23
Reflections collected19645
Independent reflections11676 [R(int) = 0.1757]
Completeness to θ = 67.686°83.1%
Refinement methodFull-matrix least-squares on F2
Data/restraints/parameters11,676/0/1014
Goodness-of-fit on F21.253
Final R indices [I > 2σ(I)]R1 = 0.1476, wR2 = 0.3359
R indices (all data)R1 = 0.2313, wR2 = 0.4260
Extinction coefficient0.0114(11)
Table 2. Cytotoxicity of compounds 2760 toward human cancer cell lines a.
Table 2. Cytotoxicity of compounds 2760 toward human cancer cell lines a.
CompoundIC50 (μM)
HCT-116HeLaMCF-7
2775 ± 184 ± 180 ± 2
28200 ± 4100 ± 6120 ± 5
29720 ± 36190 ± 7110 ± 2
3085 ± 290 ± 467 ± 1
31140 ± 7150 ± 677 ± 2
3288 ± 387 ± 487 ± 4
3367 ± 176 ± 388 ± 1
3451 ± 173 ± 168 ± 3
3568 ± 369 ± 178 ± 2
3649 ± 255 ± 296 ± 4
3736 ± 134 ± 270 ± 3
3838 ± 242 ± 169 ± 1
3983 ± 293 ± 195 ± 3
4077 ± 185 ± 298 ± 3
4197 ± 285 ± 199 ± 1
4277 ± 282 ± 183 ± 2
4392 ± 3130 ± 1087 ± 2
44160 ± 6160 ± 890 ± 5
4597 ± 282 ± 190 ± 1
4664 ± 180 ± 259 ± 2
4775 ± 579 ± 178 ± 3
4880 ± 380 ± 287 ± 3
49110 ± 2120 ± 681 ± 5
50115 ± 3130 ± 8120 ± 4
51190 ± 13*95 ± 5
5270 ± 2*69 ± 1
5376 ± 274 ± 288 ± 2
5484 ± 582 ± 196 ± 5
55150 ± 3105 ± 4150 ± 6
5675 ± 280 ± 289 ± 3
5773 ± 180 ± 190 ± 1
5898 ± 288 ± 692 ± 6
59140 ± 5150 ± 2100 ± 5
60230 ± 18230 ± 5270 ± 13
Cisplatin3.8 ± 0.22.2 ± 0.23 ± 0.1
a Analysis was performed using the MTT assay after 72 h of incubation. Values are expressed as the mean ± SD of at least three independent experiments; * Viability of cell lines at 100 μM of tested compounds were approximately 100%.
Table 3. Results of metabolic stability study.
Table 3. Results of metabolic stability study.
CompoundIn Vitro Metabolic Half-Life t1/2 (min)
3024.9
3117.6
34>60 (154)
3542.4
3613.9
3732.8
38<5
46>60 (78.1)
4717.5
5226.5

Share and Cite

MDPI and ACS Style

Żołnowska, B.; Sławiński, J.; Pogorzelska, A.; Szafrański, K.; Kawiak, A.; Stasiłojć, G.; Belka, M.; Ulenberg, S.; Bączek, T.; Chojnacki, J. Novel 5-Substituted 2-(Aylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides: Synthesis, Molecular Structure, Anticancer Activity, Apoptosis-Inducing Activity and Metabolic Stability. Molecules 2016, 21, 808. https://doi.org/10.3390/molecules21060808

AMA Style

Żołnowska B, Sławiński J, Pogorzelska A, Szafrański K, Kawiak A, Stasiłojć G, Belka M, Ulenberg S, Bączek T, Chojnacki J. Novel 5-Substituted 2-(Aylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides: Synthesis, Molecular Structure, Anticancer Activity, Apoptosis-Inducing Activity and Metabolic Stability. Molecules. 2016; 21(6):808. https://doi.org/10.3390/molecules21060808

Chicago/Turabian Style

Żołnowska, Beata, Jarosław Sławiński, Aneta Pogorzelska, Krzysztof Szafrański, Anna Kawiak, Grzegorz Stasiłojć, Mariusz Belka, Szymon Ulenberg, Tomasz Bączek, and Jarosław Chojnacki. 2016. "Novel 5-Substituted 2-(Aylmethylthio)-4-chloro-N-(5-aryl-1,2,4-triazin-3-yl)benzenesulfonamides: Synthesis, Molecular Structure, Anticancer Activity, Apoptosis-Inducing Activity and Metabolic Stability" Molecules 21, no. 6: 808. https://doi.org/10.3390/molecules21060808

Article Metrics

Back to TopTop