Next Article in Journal
Acetylcholinesterase Inhibitors among Zingiber officinale Terpenes—Extraction Conditions and Thin Layer Chromatography-Based Bioautography Studies
Next Article in Special Issue
Organosilicon Compounds, SILA-409 and SILA-421, as Doxorubicin Resistance-Reversing Agents in Human Colon Cancer Cells
Previous Article in Journal
Use of Lead Isotopic Ratios as Geographical Tracer for Lambrusco PDO Wines
Previous Article in Special Issue
Synthesis, Characterization and Biological Evaluation of Novel Dihydropyranoindoles Improving the Anticancer Effects of HDAC Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Docking Studies and Biological Activity of New Benzimidazole- Triazolothiadiazine Derivatives as Aromatase Inhibitor

by
Ulviye Acar Çevik
1,2,
Betül Kaya Çavuşoğlu
3,
Begüm Nurpelin Sağlık
1,2,
Derya Osmaniye
1,2,
Serkan Levent
1,2,
Sinem Ilgın
4,
Yusuf Özkay
1,2,* and
Zafer Asım Kaplancıklı
1
1
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
2
Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
3
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bülent Ecevit University, Zonguldak 67100, Turkey
4
Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Turkey
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(7), 1642; https://doi.org/10.3390/molecules25071642
Submission received: 3 March 2020 / Revised: 31 March 2020 / Accepted: 1 April 2020 / Published: 2 April 2020
(This article belongs to the Special Issue Anticancer Agents: Design, Synthesis and Evaluation)

Abstract

:
In the last step of estrogen biosynthesis, aromatase enzyme catalyzes the conversion of androgens to estrogens. Aromatase inhibition is an important way to control estrogen-related diseases and estrogen levels. In this study, sixteen of benzimidazole-triazolothiadiazine derivatives have been synthesized and studied as potent aromatase inhibitors. First, these compounds were tested for their anti-cancer properties against human breast cancer cell line (MCF-7). The most active compounds 5c, 5e, 5k, and 5m on MCF-7 cell line were subject to further in vitro aromatase enzyme inhibition assays to determine the possible mechanisms of action underlying their activity. Compound 5e showed slight less potent aromatase inhibitory activity than that of letrozole with IC50 = 0.032 ± 0.042 µM, compared to IC50 = 0.024 ± 0.001 µM for letrozole. Furthermore, compound 5e and reference drug letrozole were docked into human placental aromatase enzyme to predict their possible binding modes with the enzyme. Finally, ADME parameters (absorption, distribution, metabolism, and excretion) of synthesized compounds (5a5p) were calculated by QikProp 4.8 software.

1. Introduction

Aromatase is a member of the cytochrome P450 enzyme that catalyzes the biosynthesis of estrogens from androgens [1,2]. Estrogen levels have been shown to be higher in breast cancer cells. Inhibition of aromatase is one of the effective current therapeutic strategies for controlling estrogen-dependent breast cancer. Therefore, one of the commonly used classes of drugs for the management of estrogen-dependent cancer is aromatase inhibitors (AIs) [1,3,4].
Aromatase inhibitors that have been used clinically can be categorized as first-, second-, and third generations based on their evolution time or steroidal or steroidal or non-steroidal aromatase inhibitors (NSAIs) based on their structural similarity with steroids [5,6].
Aminoglutethimide is the prototype nonsteroidal inhibitor of aromatase. Problems with the side effects and selectivity of aminoglutethimide led to the development of the second-generation of nonsteroidal aromatase inhibitor (Fadrazole bearing imidazole structure) [7]. However, this compound still has some nonselective inhibitory activity with respect to progesterone, corticosterone, and aldosterone biosynthesis. Competitive nonsteroidal inhibitors can also be constructed with a triazole ring, which is found in the third generation of aromatase inhibitors [8,9,10]. Most of the nonsteroidal aromatase inhibitors of therapeutic importance act covalently bind to the substrate-binding site of aromatase by coordinate the heme iron [11,12,13]. Especially, the heterocyclic nitrogen atom of triazole and imidazole plays an important role by coordinating with the heme iron of the aromatase enzyme [12,13]. Some studies have shown that imidazole and triazole derivatives have promising aromatase inhibition [14,15,16,17].
Recent clinical studies have shown that aromatase inhibitors, especially the third-generation, are more effective than fulvestrant and tamoxifen because of their lower side effects and higher clinical efficacy. Nevertheless, the development of acquired resistance after prolonged AIs therapy, undesirable side effects (bone loss, cardiac events, increased rash, insomnia, headaches, and arthralgia) limits their use in clinical practice. Thus, the search for new potent molecules that impair cancer growth, strongly inhibit aromatase enzyme, and present fewer side effects is of major importance [18,19,20,21].
In the previous study, 3-[4-(5-methyl-1H-benzo[d]imidazol-2-yl)phenyl]-6- (substituted phenyl) -7H-[1,2,4] triazolo [3,4-b] [1,3,4]thiadiazines derivatives have been synthesized and promising compounds have been obtained that need further development as a new class of aromatase inhibitors [22]. In this study, new benzimidazole-triazolothiadiazine derivatives were synthesized and structure of these compounds was characterized by spectroscopic data. Their antiproliferative activities against MCF-7 were evaluated. To identify the possible modes of action, aromatase inhibition experiments were performed for the most active compounds against the MCF-7 cell line. Finally, in silico prediction of pharmacokinetic profiles (ADME) were calculated for physicochemical properties of these drug candidates.

2. Results and Discussion

2.1. Chemistry

Synthesis of the target compounds was accomplished according to the steps illustrated in Scheme 1. Substituents of the synthesized compounds (5a5p) were shown in Table 1. The starting intermediated compound (1a,1b) was prepared by the reaction of 1,2-phenylenediamine and sodium metabisulfite with 4-formylbenzoic acid methyl ester as described in a previous study [23]. The ester derivative (1a,1b) reacted with hydrazine hydrate producing compounds 2a, 2b in a microwave synthesis reactor. Then, the compounds-(5-substitüe-1H-benzimidazol-2-yl)benzoic acid hydrazide derivatives (2a,2b) were treated with carbon disulfide in NaOH solution affording, after acidic treatment, compounds 3a, 3b. The reaction of compound 3a, 3b with hydrazine hydrate in the presence of ethanol produced compounds 4a, 4b. Then cyclization of compounds 4a, 4b with appropriate phenacyl bromide in the presence of anhydrous ethanol provided the desired final products (5a5p). The characterization of these new derivatives was done by their spectroscopic (1H NMR, 13C NMR and Mass) data. 1H NMR and 13C NMR spectra of the synthesized compounds were observed at the expected region. Mass spectra (HRMS) of all the synthesized compounds showed that the molecular ion [M + H]+ peak is in agreement with their molecular formula (see Supplementary Materials).

2.2. Cytotoxicity Assay

The newly synthesized compounds were evaluated for their in vitro anticancer potential against MCF-7 breast cancer cell lines by MTT assay [24] using cisplatin as a reference standard. The 50% inhibitory concentration (IC50) values were determined for these compounds. In addition, the cytotoxic activities of compounds 5c, 5e, 5k, and 5m were assessed against healthy NIH3T3 [25] cells, in order to express the selectivity toward carcinogenic cells. The IC50 values of test compounds were determined as the mean IC50 of 4 independent experiments. Results are presented in Table 2, as IC50 values are in μM and revealed that some of the tested compounds were remarkably more cytotoxic than the cisplatin against MCF-7 cell lines. Concerning MCF-7 cell line, the most potent compounds were the 4-cyano derivatives 5e and 5m with IC50 = 0.016 ± 0.001 and 0.018 ± 0.001 μM, respectively compared to IC50 = 0.020 ± 0.009 μM for the reference drug cisplatin. Furthermore, the most promising activity was observed for the compounds 5c and 5k bearing 4-fluorophenyl derivatives with IC50= 0.119 ± 0.005 and 0.110 ± 0.005 μM, respectively compared to IC50 = 0.020 ± 0.009 μM for the reference drug cisplatin.
Benzimidazole-triazolothiadiazine derivatives led to a promising increase in the anti-proliferative activity. The synthesized compounds can be divided into two groups as nonsubstituted benzimidazole and 5-chlorobenzimidazole. Test compounds showed variable activities against MCF-7. In particular, compounds 5e and 5m displayed the highest cytotoxic activity against MCF-7 cell line. The most active compounds 5e and 5m carried a 4-cyanophenyl substituent. The chlorine substituent in the fifth position (5m) of the benzimidazole ring did not significantly affect the activity. Compounds 5c and 5k carrying fluorine substituents in the fourth position of the phenyl ring showed lower activity than compounds 5e and 5m bearing cyano group in the fourth position of the phenyl ring.

2.3. Aromatase Inhibition Assay

The in vitro anti-aromatase activity of the most active compounds 5c, 5e, 5k, and 5m was valued using commercial fluorometric assay kit (Aromatase-CYP19A Inhibitor Screening kit, Bio Vision) with letrozole as the reference drug. Results are presented in Table 3. The IC50 values of compounds were in the sub-micromolar range (2.276 ± 0.106–0.03 2 ± 0.001 µM). The best value was shown by compound 5e with IC50 value (0.032 ± 0.001 µM).

2.4. Molecular Docking

After the most active derivative was selected as compound 5e according to in vitro aromatase enzyme inhibition assay and letrozole, molecular docking studies were performed to explain its binding modes with human aromatase enzyme active site. For this purpose, the crystal structure of human aromatase enzyme (PDB ID: 3EQM) [26] was retrieved from the Protein Data Bank server (www.pdb.org). The 2D and 3D docking poses of compound 5e are presented in Figure 1 and Figure 2. The docking poses of letrozole are presented in Figure 3 and Figure 4.
According to docking poses, it is understood that compound 5e displays compatible settlement with the enzyme active region. The benzimidazole ring in the structure forms two π–π interactions with Arg115 and Phe134. Also, it can be seen from 2D docking pose, this compound is in interaction with Hem molecules and Cys437 via its nitrogen atoms of triazolothiadiazine ring by salt bridge. There is another π–π interaction between 4-cyanophenyl ring and HEM molecule. The nitrogen atom of cyano group at C-4 position of phenyl ring interacts with hydroxyl of Ser314 doing hydrogen bonding. It is thought that this interaction is important for compound 5e in terms of explaining its inhibitory activity. It is seen that the presence of an electron withdrawing group such as cyano at this position is a positive contribution to the activity.
As seen in the docking poses, letrozole is settled down in the enzyme active site properly. For letrozole, it is seen that benzonitrile ring creates π–π interaction with HEM molecule. Another π–π interaction is observed between 1,2,4-triazole ring and Arg115. Also, this 1,2,4-triazole ring forms two hydrogen bonds via its second and fourth nitrogen atoms with amino groups of Ala438 and Arg115, respectively.

2.5. Theoretical Determination of ADME Properties

QikProp allows to provide acceptable ranges for comparing the predicted properties of compounds with those of 95% of known drugs and estimate drug-likeness properties. The drug-likeness of a compound was assessed according to Jorgensen’s rule of three [27], which regards PCaco (>22 nm/s), logS (>−5.7), primary metabolites (PM) (<7), and Lipinski’s rule of five [28], which considers number of hydrogen bond acceptors (≤10) and donors (≤5), molecular weight (<500 Da), and octanol/water partition coefficient (≤5).
Table 4. presents the predicted ADME properties of all compounds. According to Lipinski’s rule of five and Jorgensen’s rule of three, all compounds (5a5p) are in accordance with the rule by causing no more than one violation. Consequently, according to predictions of ADME properties, it can be suggested that the active compounds may have a good pharmacokinetic profile.

3. Materials and Methods

3.1. Chemistry

Whole chemicals employed in the synthetic procedure were purchased from Sigma-Aldrich Chemicals (Sigma-Aldrich Corp., St. Louis, MO, USA) or Merck Chemicals (Merck KGaA, Darmstadt, Germany). Melting points of the obtained compounds were determined by MP90 digital melting point apparatus (Mettler Toledo, OH, USA) and were uncorrected. Microwave syntheses were realized by using a Monowave 300 high-performance microwave reactor (Anton-Paar, Austria). 1H NMR and 13C NMR spectra of the synthesized compounds were registered by a Bruker 300 MHz and 75 MHz digital FT-NMR spectrometer (Bruker Bioscience, Billerica, MA, USA) in DMSO-d6, respectively. Splitting patterns were designated as follows: s: singlet; d: doublet; t: triplet; m: multiplet in the NMR spectra. Coupling constants (J) were reported as Hertz. M+1 peaks were determined by Shimadzu LC/MS ITTOF system (Shimadzu, Tokyo, Japan). All reactions were monitored by thin-layer chromatography (TLC) using Silica Gel 60 F254 TLC plates (Merck KGaA, Darmstadt, Germany).

3.1.1. Synthesis of 4-(5-substitüe-1H-benz[d]imidazol-2-yl)benzoic Acid Methyl Ester (1a,1b)

Yields: 82–78%. 4-(5-substitüe-1H-benz[d]imidazol-2-yl)benzoic acid hydrazide derivatives (2a,2b) (yields: 76–72%) and 5-[4-(5(6)-substituted-1H-benz[d]imidazol-2-yl)phenyl]-1,3,4-oxadiazole-2-thiol derivatives (3a,3b) (yields: 78–7 %) were prepared following the reported procedures [23].

3.1.2. 4-Amino-5-(4-(5-substitüe-1H-benzo[d]imidazol-2-yl)phenyl)-4H-1,2,4-triazole-3-thiol (4a,4b)

Compound 3a or 3b (0.02 mol) was dissolved in ethanol. Hydrazine hydrate (5 mL) was added to the mixture. The reaction mixture was heated at 240 °C and 10 bar for 10 min under microwave synthesis reactor (Anton-Paar Monowave 300). After the reaction ended, the product was washed with water, dried, and crystallized using ethanol (96%) [22]. Yields: 68–64%.

3.1.3. 3-(4-(5-substitüe-1H-benzo[d]imidazol-2-yl)phenyl)-6-(substitüephenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4]thiadiazine (5a5p)

Compound 4a or 4b (0.003 mol) and appropriate phenacyl bromide (0.003 mol) in ethanol was heated under reflux for 8 h. After the reaction ended, the product was filtered and dried [22].
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-phenyl-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5a): Yields: 77%. Mp 230.7–232.9 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.52 (2H, s, -CH2-), 7.52-7.55 (2H, m, aromatic CH), 7.59–7.67 (3H, m, Aromatic CH), 7.83–7.86 (2H, m, aromatic CH), 8.06–8.09 (2H, m, aromatic CH), 8.34 (2H, d, J=8.67 Hz, 1,4-disubstitutedbenzene), 8.43 (2H, d, J=8.64 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.31, 114.89, 125.84, 126.55, 128.13, 128.50, 129.13, 129.65, 132.64, 133.78, 134.17, 143.96, 149.21, 151.22, 156.82, 163.07. HRMS (m/z): [M + H]+ calcd for C23H16N6S: 409.1224; found: 409.1230.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-chlorophenyl)-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5b): Yields: 72%. Mp 190.9–192.6 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.50 (2H, s, -CH2-), 7.56–7.59 (2H, m, aromatic CH), 7.65–7.70 (3H, m, aromatic CH), 7.86–7.89 (2H, m, aromatic CH), 8.06–8.09 (2H, m, aromatic CH), 8.31 (2H, d, J=8.61 Hz, 1,4-disubstitutedbenzene), 8.57 (2H, d, J=8.61 Hz, 1,4- Disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ=23.27, 114.67, 125.49, 126.35, 128.87, 129.11, 129.32, 129.45, 129.73, 129.92, 130.11, 130.71, 132.62, 133.01, 137.50, 144.01, 148.52, 151.17, 156.87. HRMS (m/z): [M + H]+ calcd for C23H15N6SCl: 443.0843; found: 443.0840.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-fluorophenyl)-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5c): Yields: 69%. Mp 274.2–277.1 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.50 (2H, s, -CH2-), 7.12 (1H, s, aromatic CH), 7.29 (1H, s, aromatic CH), 7.46–7.52 (4H, m, aromatic CH), 7.81–7.84 (2H, m, aromatic CH), 8.33 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.51 (2H, d, J=8.76 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.27, 114.32, 114.90, 116.76 (d, J = 21.98 Hz), 126.40, 127.68, 127.96, 128.43, 128.56, 128.69, 128.87, 128.95, 130.32 (d, J = 2.77 Hz), 130.78 (d, J = 8.43 Hz), 134.60, 143.66, 151.30, 155.71, 166.36 (d, J = 249.53 Hz). HRMS (m/z): [M + H]+ calcd for C23H15N6FS: 427.1137; found: 427.1136.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-methylphenyl)-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5d): Yields: 70%. Mp 207.5–209.1 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 2.41 (3H, s, CH3), 4.49 (2H, s, -CH2-), 7.41 (2H, d, J = 8.13 Hz, 1,4-disubstitutedbenzene), 7.52–7.55 (2H, m, benzimidazole CH), 7.83–7.86 (2H, m, benzimidazole CH), 7.97 (2H, d, J = 8.25 Hz, 1,4-disubstitutedbenzene), 8.33 (2H, d, J = 8.58 Hz, 1,4-disubstitutedbenzene), 8.43 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ=21.55, 23.18, 114.86, 125.84, 126.42, 128.08, 128.24, 128.47, 129.02, 129.22, 129.71, 130.20, 130.90, 134.11, 142.96, 144.00, 147.35, 149.13, 151.07, 156.69. HRMS (m/z): [M + H]+ calcd for C24H18N6S: 423.1381; found: 423.1386.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-cyanophenyl)-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5e): Yields: 68%. Mp 259.3–263.1 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.53 (2H, s, -CH2-), 7.56–7.58 (2H, m, benzimidazole CH), 7.85–7.88 (2H, m, benzimidazole CH), 8.06 (2H, d, J = 8.37 Hz, 1,4-disubstitutedbenzene), 8.21 (2H, d, J = 8.46 Hz, 1,4-disubstitutedbenzene), 8.30 (2H, d, J = 8.49 Hz, 1,4-disubstitutedbenzene), 8.42 (2H, d, J = 8.49 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.35, 114.58, 114.71, 118.68, 125.51, 126.39, 127.20, 128.48, 128.74, 128.87, 129.19, 129.43, 129.84, 133.03, 133.44, 137.97, 143.97, 148.72, 151.25, 155.45. HRMS (m/z): [M + H]+ calcd for C24H15N7S: 434.1186; found: 434.1182.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-bromophenyl)-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5f): Yields: 74%. Mp 284.8–286.2 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.50 (2H, s, -CH2-), 7.53–7.56 (3H, m, aromatic CH), 7.81–7.85 (3H, m, aromatic CH), 7.99–8.02 (2H, m, aromatic CH), 8.32 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.42 (2H, d, J = 8.70 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.20, 114.91, 125.04, 125.96, 126.50, 127.03, 128.56, 129.19, 130.09, 130.90, 132.37, 132.67, 133.00, 142.94, 143.96, 147.34, 149.17, 151.22, 155.95. HRMS (m/z): [M + H]+ calcd for C23H15N6SBr: 487.0311; found: 487.0335.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(2,4-difluorohenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5g): Yields: 72%. Mp 208.5–210.7 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.42 (2H, s, -CH2-), 7.46–7.49 (4H, m, aromatic CH), 7.79–7.82 (3H, m, aromatic CH), 7.29–7.35 (2H, m, aromatic CH), 8.45–8.51 (2H, m, aromatic CH). 13C-NMR (75 MHz, DMSO-d6): δ = 25.37, 113.01 (dd, J1 = 3.08 Hz, J2 = 21.45 Hz), 106.08 (d, J = 26.01 Hz), 114.05, 114.45, 119.72 (dd, J1 = 3.02 Hz, J2 = 11.51 Hz), 126.56, 127.17, 128.25, 128.75, 129.15, 132.37, 132.67 (dd, J1 = 3.12 Hz, J2 = 10.36 Hz), 134.23, 135.55, 143.74, 148.22, 151.06, 153.70, 164.66 (d, J = 251.18 Hz), 164.83 (d, J = 250.91 Hz). HRMS (m/z): [M + H]+ calcd for C23H14N6SF2: 445.1053; found: 445.1041.
3-(4-(1H-benzo[d]imidazol-2-yl)phenyl)-6-(2,4-dichlorophenyl)-7H-[1,2,4]triazole[3,4-b] [1,3,4] thiadiazine (5h): Yields: 77%. Mp 190.7–193.4 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.36 (2H, s, -CH2-), 7.34–7.37 (3H, m, aromatic CH), 7.65 (1H, dd, J1 = 2.04 Hz, J2 = 8.34 Hz, aromatic CH), 7.70–7.73 (2H, m, aromatic CH), 7.78–7.81 (1H, m, aromatic CH), 8.20 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.40 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 26.23, 115.05, 115.32, 124.22, 124.72, 126.99, 127.78, 128.08, 128.29, 128.57, 128.96, 129.59, 130.38, 132.93, 133.05, 133.74, 136.86, 143.65, 149.92, 151.62, 156.53. HRMS (m/z): [M + H]+ calcd for C23H14N6SCl2: 477.0457; found: 477.0450.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-phenyl-7H-[1,2,4]triazole[3,4-b][1,3,4] thiadiazine (5i): Yields: 80%. Mp: 247.3–249.6 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.50 (2H, s, -CH2-), 7.49 (1H, dd, J1 = 1.89 Hz, J2=8.67 Hz, aromatic CH), 7.59–7.62 (3H, m, aromatic CH), 7.78–7.84 (2H, m, aromatic CH), 8.03–8.06 (2H, m, aromatic CH), 8.27 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.37 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.32, 114.60, 116.29, 125.95, 126.40, 128.11, 128.47, 128.97, 129.20, 129.63, 129.94, 132.62, 133.27, 133.71, 135.39, 143.97, 150.42, 151.04, 156.76. HRMS (m/z): [M + H]+ calcd for C23H15N6SCl: 443.0826; found: 443.0840.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-chlorophenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5j): Yields: 70%. Mp: 226.0–228.9 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.46 (2H, s, -CH2-), 7.33–7.37 (1H, m, benzimidazole CH), 7.63 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene), 7.68–7.73 (2H, m, benzimidazole CH), 8.04 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene), 8.19 (2H, d, J = 8.46 Hz, 1,4-disubstitutedbenzene), 8.39 (2H, d, J = 8.43 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.13, 115.14, 116.51, 124.18, 127.79, 128.20, 128.33, 128.83, 128.98, 129.19, 129.68, 129.87, 130.51, 131.57, 132.60, 137.42, 143.58, 151.40, 155.59. HRMS (m/z): [M + H]+ calcd for C23H14N6SCl2: 477.0427; found: 477.0450.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-fluorophenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5k): Yields: 71%. Mp: 249.1–250.9 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.48 (2H, s, -CH2-), 7.38 (1H, dd, J1 = 1.98 Hz, J2 = 8.58 Hz, aromatic CH), 7.46–7.48 (2H, m, aromatic CH), 7.71–7.74 (1H, m, aromatic CH), 7.77 (1H, s, aromatic CH), 8.12–8.14 (2H, m, aromatic CH), 8.25 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.40 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.45, 114.95, 116.45, 116.77 (d, J = 22.09 Hz), 124.83, 128.07, 128.21, 128.39, 128.84, 128.89, 130.30 (d, J = 2.99 Hz), 130.85 (d, J = 8.93 Hz), 135.26, 137.58, 143.69, 150.99, 151.27, 155.73, 164.82 (d, J = 249.42 Hz). HRMS (m/z): [M + H]+ calcd for C23H14N6FSCl: 461.0741; found: 461.0746.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-methylphenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5l): Yields: 65%. Mp: 250.7–252.3 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.46 (2H, s, -CH2-), 7.40 (2H, d, J = 8.07 Hz, 1,4-disubstitutedbenzene), 7.45 (1H, dd, J1 = 1.95 Hz, J2 = 8.67 Hz, benzimidazole CH), 7.76–7.79 (1H, m, benzimidazole CH), 7.82–7.83 (1H, m, benzimidazole CH), 7.95 (2H, d, J = 8.31 Hz, 1,4-disubstitutedbenzene), 8.28 (2H, d, J = 8.67 Hz, 1,4-disubstitutedbenzene), 8.37 (2H, d, J = 8.67 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 21.54, 23.15, 114.88, 116.44, 125.26, 127.74, 128.08, 128.21, 128.95, 129.24, 129.72, 130.21, 130.90, 134.62, 136.89, 142.95, 143.90, 150.99, 151.15, 156.66. HRMS (m/z): [M + H]+ calcd for C24H17N6SCl: 457.0994; found: 457.0997.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-cyanophenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5m): Yields: 62%. Mp: 254.0–255.7 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.53 (2H, s, -CH2-), 7.50 (1H, dd, J1 = 1.95 Hz, J2 = 8.70 Hz, benzimidazole CH), 7.85–7.86 (1H, m, aromatic CH), 8.06–8.08 (3H, m, aromatic CH), 8.21 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene), 8.26 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.38 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 23.30, 114.57, 114.71, 118.70, 125.83, 126.85, 128.48, 128.86, 129.11, 129.37, 129.81, 133.46, 133.58, 135.73, 137.98, 143.88, 150.56, 151.33, 155.41. HRMS (m/z): [M + H]+ calcd for C24H14N7SCl: 468.0782; found: 468.0793.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(4-bromophenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5n): Yields: 76%. Mp: 243.4–247.3 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.48 (2H, s, -CH2-), 7.43 (1H, dd, J1 = 1.98 Hz, J2 = 8.67 Hz, benzimidazole CH), 7.75 (1H, s, aromatic CH), 7.80–7.82 (3H, m, aromatic CH), 7.99 (2H, d, J = 8.70 Hz, 1,4-disubstitutedbenzene), 8.26 (2H, d, J = 8.58 Hz, 1,4-disubstitutedbenzene), 8.37 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ=23.14, 115.01, 116.52, 124.92, 126.45, 128.10, 129.05, 130.07, 131.76, 132.00, 132.15, 132.67, 133.00, 141.45, 143.77, 148.89, 149.69, 151.39, 155.91. HRMS (m/z): [M + H]+ calcd for C23H14N6SClBr: 520.9930; found: 520.9945.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(2,4-difluorohenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5o): Yields: 66%. Mp: 182.2–185.0 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.39 (2H, s, -CH2-), 7.28–7.34 (1H, m, aromatic CH), 7.42–7.48 (1H, m, aromatic CH), 7.48–7.52 (2H, m, aromatic CH), 7.77–7.83 (2H, m, aromatic CH), 7.93–8.01 (1H, m, aromatic CH), 8.25 (2H, d, J = 8.64 Hz, 1,4-disubstitutedbenzene), 8.44 (2H, d, J = 8.58 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 25.31, 105.76 (d, J = 26.01 Hz), 106.28, 113.19 (dd, J1 = 2.63 Hz, J2 = 20.86 Hz), 114.74, 116.39, 119.76 (dd, J1 = 3.73 Hz, J2 = 7.83 Hz), 125.45, 127.49, 128.32, 128.94, 129.14, 129.40, 132.71 (dd, J1 = 3.21 Hz, J2 = 9.80 Hz), 134.22, 143.80, 151.60, 151.26, 153.78, 161.28 (d, J = 252.62 Hz), 164.46 (d, J = 256.22 Hz). HRMS (m/z): [M + H]+ calcd for C23H13N6F2SCl: 479.0632; found: 479.0652.
3-(4-(5-chloro-1H-benzo[d]imidazol-2-yl)phenyl)-6-(2,4-dichlorophenyl)-7H-[1,2,4]triazole [3,4-b][1,3,4] thiadiazine (5p): Yields: 68%. Mp: 114.4–116.5 °C. 1H-NMR (300 MHz, DMSO-d6): δ = 4.35 (2H, s, -CH2-), 7.25 (1H, dd, J1 = 1.74 Hz, J2 = 8.52 Hz, aromatic CH), 7.65 (2H, dd, J1 = 2.07 Hz, J2 = 8.34 Hz, aromatic CH), 7.88 (2H, d, J = 8.34 Hz, aromatic CH), 7.90 (1H, m, aromatic CH), 8.16 (2H, d, J = 8.61 Hz, 1,4-disubstitutedbenzene), 8.32 (2H, d, J = 8.58 Hz, 1,4-disubstitutedbenzene). 13C-NMR (75 MHz, DMSO-d6): δ = 26.33, 106.76, 125.55, 127.29, 127.35, 128.58, 128.90, 129.64, 130.37, 131.63, 132.35, 132.96, 133.06, 133.44, 133.77, 136.56, 136.84, 143.51, 149.25, 151.77, 156.48. HRMS (m/z): [M + H]+ calcd for C23H13N6SCl3: 511.0037; found: 511.0061.

3.2. Cytotoxicity Assay

The anticancer activity of compounds 5a5p were screened according to the MTT assays. The MTT assays were performed as previously described [24,25]. Cisplatin was used as the reference drug for the MCF7 cell line in the MTT assays.

3.3. Aromatase Inhibition Assay

This method was carried out according to the kit procedure (Bio Vision, Aromatase (CYP19A) Inhibitor Screening Kit (Fluorometric)). The compounds were dissolved in dimethyl sulfoxide (DMSO) and added to the assay in at least seven concentrations ranging from 10−3–10−9 M. The recombinant human aromatase stock was prepared by reconstituting with 1 mL of aromatase assay buffer. The contents were mixed thoroughly by vortexing to obtain a homogeneous solution and the solution was transferred to a 15-mL conical tube. The volume was brought to 2450 µL with the aromatase assay buffer and 50 µl of NADPH production system (100X) was added for a final total volume of 2.5 mL. Letrozole was used as a positive inhibition control. For solvent control, a small aliquot of aromatase assay buffer containing the organic solvent was used to dissolve the test compounds that were prepared. Reaction wells containing test compounds and the corresponding no inhibitor controls (which may also serve as a solvent control), as well as a background control (containing no fluorogenic Aromatase Substrate) were prepared. The plate was incubated for at least 10 min at 37 °C to allow test ligands to interact with the aromatase. After incubation, 30 µl of the aromatase substrate/NADP+ mixture was added to each well. Immediately (within 1 min), the fluorescence at Ex/Em = 488/527 nm was measured.

3.4. Molecular Docking

A structure-based in silico procedure was applied to discover the binding modes of compound 5e to human aromatase enzyme active site. The crystal structures of human aromatase (PDB ID: 3EQM) [26] was retrieved from the Protein Data Bank server (www.pdb.org).
The structures of ligands were built using the Schrödinger Maestro [29] interface and then were submitted to the Protein Preparation Wizard protocol of the Schrödinger Suite 2016 Update 2 [30]. The ligands were prepared by the LigPrep 3.8 [31] to assign the protonation states at pH 7.4 ± 1.0 and the atom types, correctly. Bond orders were assigned, and hydrogen atoms were added to the structures. The grid generation was formed using Glide 7.1 [32]. Flexible docking runs were performed with single precision docking mode (SP).

3.5. Theoretical Determination of ADME Properties

Physicochemical parameters of obtained compounds (5a5p) were evaluated by using QikProp 4.8 [33].

4. Conclusions

Inhibition of aromatase has proved to be an effective approach for the treatment of hormone-dependent breast cancer in the postmenopausal women. Imidazole and triazole groups are important rings for the development new potent aromatase inhibitors with high affinity for the enzyme. A series of benzimidazole-triazolothiadiazine derivatives have been synthesized with different substituents at benzimidazole and phenyl rings. The newly synthesized compounds were tested for their anti-cancer properties against human breast cancer cell line (MCF-7). The synthesized compounds were then tested in in vitro aromatase assay and two compounds (5e and 5m) exhibited activity similar to letrozole.

Supplementary Materials

The following are available online.

Author Contributions

Z.A.K., Y.Ö., conceived and designed the experiments; D.O., U.A.Ç. performed the synthesis; S.L. performed analysis studies; B.N.S., U.A.Ç., and S.I. performed activity tests; D.O., U.A.Ç., B.N.S., B.K.Ç. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

This study was financially supported by Anadolu University Scientific Projects Fund, Project No: 1905S032.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chamduang, C.; Pingaew, R.; Prachayasittikul, V.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Novel triazole-tetrahydroisoquinoline hybrids as human aromatase inhibitors. Bioorg. Chem. 2019, 93, 103327. [Google Scholar] [CrossRef] [PubMed]
  2. Doiron, J.; Richard, R.; Touré, M.M.; Picot, N.; Richard, R.; Čuperlović-Culf, M.; Robichaud, G.A.; Touaibia, M. Synthesis and structure–activity relationship of 1-and 2-substituted-1, 2, 3-triazole letrozole-based analogues as aromatase inhibitors. Eur. J. Med. Chem. 2011, 46, 4010–4024. [Google Scholar] [CrossRef] [PubMed]
  3. Henneberta, O.; Montes, M.; Favre-Reguillon, A.; Chermetted, H.; Ferroudc, C.; Mortin, R. Epimerase activity of the human 11β-hydroxysteroid dehydrogenase type 1 on 7-hydroxylated C19-steroids. J. Steroid Biochem. Mol. Biol. 2009, 114, 57–63. [Google Scholar] [CrossRef] [PubMed]
  4. Leechaisit, R.; Pingaew, R.; Prachayasittikul, V.; Worachartcheewan, A.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Synthesis, molecular docking, and QSAR study of bis-sulfonamide derivatives as potential aromatase inhibitors. Bioorg. Med. Chem. 2019, 27, 115040. [Google Scholar] [CrossRef]
  5. Brueggemeier, R.W.; Hackett, J.C.; Diaz-Cruz, E.S. Aromatase inhibitors in the treatment of breast cancer. Endocr. Rev. 2005, 26, 331–345. [Google Scholar] [CrossRef] [Green Version]
  6. Cepa, M.M.; da Silva, E.J.T.; Correia-da-Silva, G.; Roleira, F.M.; Teixeira, N.A. Synthesis and biochemical studies of 17-substituted androst-3-enes and 3,4-epoxyandrostanes as aromatase inhibitors. Steroids 2008, 73, 1409–1415. [Google Scholar] [CrossRef] [Green Version]
  7. Bonfield, K.; Amato, E.; Bankemper, T.; Agard, H.; Steller, J.; Keeler, J.M.; Roy, D.; McCallum, A.; Paula, S.; Ma, L. Development of a new class of aromatase inhibitors: Design, synthesis and inhibitory activity of 3-phenylchroman-4-one (isoflavanone) derivatives. Bioorg. Med. Chem. 2012, 20, 2603–2613. [Google Scholar] [CrossRef] [Green Version]
  8. Rampogu, S.; Baek, A.; Bavi, R.S.; Son, M.; Cao, G.P.; Kumar, R.; Park, C.; Zeb, A.; Rana, R.M.; Park, S.J.; et al. Identification of Novel Scaffolds with Dual Role as Antiepileptic and Anti-Breast Cancer. IEEE/ACM Trans. Comput. Biol. Bioinform. 2018, 16, 1663–1674. [Google Scholar] [CrossRef]
  9. Trunet, P.F.; Vreeland, F.; Royce, C.; Chaudri, H.A.; Cooper, J.; Bhatnagar, A.S. Clinical use of aromatase inhibitors in the treatment of advanced breast cancer. J. Steroid. Biochem. Mol. Biol. 1997, 61, 241–245. [Google Scholar] [CrossRef]
  10. Steele, R.E.; Mellor, L.B.; Sawyer, W.K.; Wasvary, J.M.; Browne, L.J. In vitro and in vivo studies demonstrating potent and selective estrogen inhibition with the nonsteroidal aromatase inhibitor CGS 16949A. Steroids 1987, 50, 147161. [Google Scholar] [CrossRef]
  11. Kato, S.; Endoh, H.; Masuhiro, Y.; Kitamoto, T.; Uchiyama, S.; Sasaki, H.; Masushige, S.; Gotoh, Y.; Nishida, E.; Kawashima, H.; et al. Activation of the estrogen receptor through phosphorylation by mitogen-activated protein kinase. Science 1995, 270, 1491–1494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sable, P.M.; Potey, L.C. Synthesis and antiproliferative activity of imidazole and triazole derivatives of flavonoids. Pharm. Chem. J. 2018, 52, 438–443. [Google Scholar] [CrossRef]
  13. Gilardi, G.; Di Nardo, G. Heme iron centers in cytochrome P450: Structure and catalytic activity. Rend. Lincei 2017, 28, 159–167. [Google Scholar] [CrossRef] [Green Version]
  14. Asadi, P.; Khodarahmi, G.; Farrokhpour, H.; Hassanzadeh, F.; Saghaei, L. Quantum mechanical/molecular mechanical and docking study of the novel analogues based on hybridization of common pharmacophores as potential anti-breast cancer agents. Res. Pharm. Sci. 2017, 12, 233. [Google Scholar]
  15. Mojaddami, A.; Sakhteman, A.; Fereidoonnezhad, M.; Faghih, Z.; Najdian, A.; Khabnadideh, S.; Rezaei, Z. Binding mode of triazole derivatives as aromatase inhibitors based on docking, protein ligand interaction fingerprinting, and molecular dynamics simulation studies. Res. Pharm. Sci. 2017, 12, 21. [Google Scholar] [CrossRef] [Green Version]
  16. Song, Z.; Liu, Y.; Dai, Z.; Liu, W.; Zhao, K.; Zhang, T.; Dai, Y. Synthesis and aromatase inhibitory evaluation of 4-N-nitrophenyl substituted amino-4H-1,2,4-triazole derivatives. Bioorg. Med. Chem. 2016, 24, 4723–4730. [Google Scholar] [CrossRef]
  17. Adhikari, N.; Amin, S.A.; Jha, T.; Gayen, S. Integrating regression and classification-based QSARs with molecular docking analyses to explore the structure-antiaromatase activity relationships of letrozole-based analogs. Can. J. Chem. 2017, 95, 1285–1295. [Google Scholar] [CrossRef]
  18. Prachayasittikul, V.; Pingaew, R.; Worachartcheewan, A.; Sitthimonchai, S.; Nantasenamat, C.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Aromatase inhibitory activity of 1, 4-naphthoquinone derivatives and QSAR study. EXCLI J. 2017, 16, 714. [Google Scholar]
  19. Augusto, T.V.; Amaral, C.; Varela, C.L.; Bernardo, F.; da Silva, E.T.; Roleira, F.F.; Costa, S.; Teixeria, N.; Correia-da-Silva, G. Effects of new C6-substituted steroidal aromatase inhibitors in hormone-sensitive breast cancer cells: Cell death mechanisms and modulation of estrogen and androgen receptors. J. Steroid. Biochem. Mol. Biol. 2019, 195, 105486. [Google Scholar] [CrossRef]
  20. Shoombuatong, W.; Schaduangrat, N.; Nantasenamat, C. Towards understanding aromatase inhibitory activity via QSAR modeling. EXCLI J. 2018, 17, 688. [Google Scholar]
  21. Prior, A.M.; Yu, X.; Park, E.J.; Kondratyuk, T.P.; Lin, Y.; Pezzuto, J.M.; Sun, D. Structure-activity relationships and docking studies of synthetic 2-arylindole derivatives determined with aromatase and quinone reductase 1. Bioorg. Med. Chem. Lett. 2017, 27, 5393–5399. [Google Scholar] [CrossRef] [PubMed]
  22. Acar Çevik, U.; Sağlık, B.N.; Osmaniye, D.; Levent, S.; Kaya Çavuşoğlu, B.; Karaduman, A.B.; Ozkay, Y.; Kaplancıklı, Z.A. Synthesis and docking study of benzimidazole–triazolothiadiazine hybrids as aromatase inhibitors. Arch. Pharm. 2020, e2000008. [Google Scholar] [CrossRef] [PubMed]
  23. Çevik, U.A.; Osmaniye, D.; Çavuşoğlu, B.K.; Sağlik, B.N.; Levent, S.; Ilgin, S.; Ozkay, Y.; Kaplancikli, Z.A. Synthesis of novel benzimidazole–oxadiazole derivatives as potent anticancer activity. Med. Chem. Res. 2019, 28, 2252–2261. [Google Scholar] [CrossRef]
  24. Evren, A.E.; Yurttaş, L.; Eksellı, B.; Akalın-Cıftcı, G. Novel tri-substituted thiazoles bearing piperazine ring: Synthesis and evaluation of their anticancer activity. Lett. Drug Des. Discov. 2019, 16, 547–555. [Google Scholar] [CrossRef]
  25. Çevik, U.A.; Sağlık, B.N.; Ardıç, C.M.; Özkay, Y.; Atlı, Ö. Synthesis and evaluation of new benzimidazole derivatives with hydrazone moiety as anticancer agents. Turk. J. Biochem. 2018, 43, 151–158. [Google Scholar] [CrossRef]
  26. Ghosh, D.; Griswold, J.; Erman, M.; Pangborn, W. Structural basis for androgen specificity and oestrogen synthesis in human aromatase. Nature 2009, 457, 219. [Google Scholar] [CrossRef] [Green Version]
  27. Jorgensen, W.L.; Duffy, E.M. Prediction of drug solubility from structure. Adv. Drug Deliv. Rev. 2002, 54, 355–366. [Google Scholar] [CrossRef]
  28. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  29. Maestro, version 10.6; Schrödinger, LLC: New York, NY, USA, 2016.
  30. The Schrödinger Suite 2016 Update 2; Schrödinger, LLC: New York, NY, USA, 2016.
  31. LigPrep, version 3.8; Schrödinger, LLC: New York, NY, USA, 2016.
  32. Glide, version 7.1; Schrödinger, LLC: New York, NY, USA, 2016.
  33. QikProp, version 4.8; Schrödinger, LLC: New York, NY, USA, 2016.
Sample Availability: Samples of the compounds 5a5p are available from the authors.
Scheme 1. The reaction sequence for the synthesis of the compounds (5a5p).
Scheme 1. The reaction sequence for the synthesis of the compounds (5a5p).
Molecules 25 01642 sch001
Figure 1. Two-dimensional interaction mode of compound 5e in the enzyme active site (Human aromatase PDB Code: 3EQM).
Figure 1. Two-dimensional interaction mode of compound 5e in the enzyme active site (Human aromatase PDB Code: 3EQM).
Molecules 25 01642 g001
Figure 2. The interacting mode of compound 5e in the active region of human aromatase. The inhibitor, colored with maroon, and the important residues, colored with purple, in the active site of the enzyme are presented by tube model.
Figure 2. The interacting mode of compound 5e in the active region of human aromatase. The inhibitor, colored with maroon, and the important residues, colored with purple, in the active site of the enzyme are presented by tube model.
Molecules 25 01642 g002
Figure 3. Two-dimensional interaction mode of letrozole in the enzyme active site (Human aromatase PDB Code: 3EQM).
Figure 3. Two-dimensional interaction mode of letrozole in the enzyme active site (Human aromatase PDB Code: 3EQM).
Molecules 25 01642 g003
Figure 4. The interacting mode of letrozole in the active region of human aromatase. The inhibitor, colored with orange, and the important residues, colored with turquoise, in the active site of the enzyme are presented by tube model.
Figure 4. The interacting mode of letrozole in the active region of human aromatase. The inhibitor, colored with orange, and the important residues, colored with turquoise, in the active site of the enzyme are presented by tube model.
Molecules 25 01642 g004
Table 1. The synthesized compounds (5a5p).
Table 1. The synthesized compounds (5a5p).
Comp.RR1R2
5a-H-H-H
5b-H-Cl-H
5c-H-F-H
5d-H-CH3-H
5e-H-CN-H
5f-H-Br-H
5g-H-F-F
5h-H-Cl-Cl
-Cl-H-H
5j-Cl-Cl-H
5k-Cl-F-H
5l-Cl-CH3-H
5m-Cl-CN-H
5n-Cl-Br-H
5o-Cl-F-F
5p-Cl-Cl-Cl
Table 2. IC50 (µM) values of compounds (5a5p) (n = 4).
Table 2. IC50 (µM) values of compounds (5a5p) (n = 4).
Comp.MCF-7NIH3T3
5a0.142 ± 0.007-
5b0.0414 ± 0.001-
5c0.119 ± 0.00581.8 ± 2.4
5d0.194 ± 0.006-
5e0.016 ± 0.001≥1000
5f0.318 ± 0.019-
5g0.245 ± 0.004-
5h0.342 ± 0.017-
0.205 ± 0.005-
5j0.258 ± 0.011-
5k0.110 ± 0.00577.6 ± 2.1
5l0.352 ± 0.019-
5m0.018 ± 0.001≥1000
5n0.264 ± 0.011-
5o0.373 ± 0.011-
5p0.147 ± 0.001-
Cisplatin0.020 ± 0.009≥1000
Table 3. IC50 (µM) values of compounds 5c, 5e, 5k, 5m, and letrozole.
Table 3. IC50 (µM) values of compounds 5c, 5e, 5k, 5m, and letrozole.
Comp.Aromatase Inhibition
5c1.716 ± 0.042
5e0.032 ± 0.001
5k2.276 ± 0.106
5m1.562 ± 0.064
Letrozole0.024 ± 0.001
Table 4. Calculated ADME parameters.
Table 4. Calculated ADME parameters.
Comp.MWRBDMMVDHBAHBPSAlogPlogSPCacologBBPMDCKPMCNS%HOAVRFVRT
5a408.48013.5391230.5814.566.815.226−7.5771145.907−0.3611066.2822010011
5b442.925011.5361274.69114.566.8135.722−8.3231145.871−0.2072630.5682010011
5c426.47011.2941246.68914.566.8155.463−7.9441145.883−0.2551928.0382010011
5d422.506014.1751289.51714.566.815.533−8.1451145.907−0.3881066.2823010011
5e433.48917.9681297.2781692.6064.468−8.534236.976−1.255194.1192−295.60701
5f487.376011.9131283.59614.566.8135.799−8.4391145.858−0.1982828.3582010011
5g444.461012.5981251.33714.565.5955.544−8.021167.271−0.22431.4012010011
5h477.37012.7091287.74314.564.65.897−8.4221199.257−0.0993927.1412010011
5i442.925012.6091274.52814.566.8065.721−8.3211145.207−0.2082625.132010011
5j477.37010.6971318.63914.566.8096.217−9.0681145.171−0.0546476.3182010011
5k460.915010.4551290.63714.566.8115.957−8.6881145.183−0.1014746.7262010011
5l456.951013.2291333.46514.566.8066.028−8.8911145.207−0.2352625.133010011
5m467.93417.4091341.2261692.6024.96−9.274236.832−1.117477.9112−210001
5n521.821011.0461327.54414.566.8096.294−9.1841145.158−0.0446963.2672092.63121
5o478.906011.991295.26614.565.5916.038−8.7641166.535−0.0465985.5892010011
5p511.815012.0561331.67614.564.5976.391−9.1661198.4870.0559667.8772193.55321
MW: Molecular weight RB: Number of rotatable bonds (recommended value: 0-15) DM: Computed dipole moment (recommended value: 1-12.5) MV: Total solvent-accessible volume (recommended value: 500-2000) DHB: Estimated number of hydrogen bond donors (recommended value: 0-6) AHB: Estimated number of hydrogen bond acceptors (recommended value: 2-20) PSA: Van der Waals surface area of polar nitrogen and oxygen atoms and carbonyl carbon atoms (recommended value: 7-200) logP: Predicted octanol/water partition coefficient (recommended value: -2-6.5) logS: Predicted aqueous solubility (recommended value: -6.5-0.5) PCaco: Predicted apparent Caco-2 cell permeability (recommended value: <25 poor, >500 great) logBB: Predicted brain/blood partition coefficient (recommended value: -3-1.2) PMDCK: Predicted apparent MDCK cell permeability (recommended value: <25 poor, >500 great) PM: Number of likely metabolic reactions (recommended value: 1-8) CNS: Predicted central nervous system activity (recommended value: –2 (inactive), +2 (active)) %HOA: Predicted human oral absorption percent (recommended value: >80% is high, <25% is poor) VRF: Number of violations of Lipinski’s rule of five. The rules are: MW < 500, logP < 5, DHB ≤ 5, AHB≤ 10, Positive PSA value. VRT: Number of violations of Jorgensen’s rule of three. The three rules are: logS > -5.7, PCaco > 22 nm/s, PM < 7.

Share and Cite

MDPI and ACS Style

Acar Çevik, U.; Kaya Çavuşoğlu, B.; Sağlık, B.N.; Osmaniye, D.; Levent, S.; Ilgın, S.; Özkay, Y.; Kaplancıklı, Z.A. Synthesis, Docking Studies and Biological Activity of New Benzimidazole- Triazolothiadiazine Derivatives as Aromatase Inhibitor. Molecules 2020, 25, 1642. https://doi.org/10.3390/molecules25071642

AMA Style

Acar Çevik U, Kaya Çavuşoğlu B, Sağlık BN, Osmaniye D, Levent S, Ilgın S, Özkay Y, Kaplancıklı ZA. Synthesis, Docking Studies and Biological Activity of New Benzimidazole- Triazolothiadiazine Derivatives as Aromatase Inhibitor. Molecules. 2020; 25(7):1642. https://doi.org/10.3390/molecules25071642

Chicago/Turabian Style

Acar Çevik, Ulviye, Betül Kaya Çavuşoğlu, Begüm Nurpelin Sağlık, Derya Osmaniye, Serkan Levent, Sinem Ilgın, Yusuf Özkay, and Zafer Asım Kaplancıklı. 2020. "Synthesis, Docking Studies and Biological Activity of New Benzimidazole- Triazolothiadiazine Derivatives as Aromatase Inhibitor" Molecules 25, no. 7: 1642. https://doi.org/10.3390/molecules25071642

Article Metrics

Back to TopTop