Next Article in Journal
Real-Time Monitoring of the Cytotoxic and Antimetastatic Properties of Cannabidiol in Human Oral Squamous Cell Carcinoma Cells Using Electric Cell-Substrate Impedance Sensing
Next Article in Special Issue
A Comprehensive Study of Gemfibrozil Complexation with β-Cyclodextrins in Aqueous Solution Using Different Analytical Techniques
Previous Article in Journal
Identification of Iguania Ancestral Syntenic Blocks and Putative Sex Chromosomes in the Veiled Chameleon (Chamaeleo calyptratus, Chamaeleonidae, Iguania)
Previous Article in Special Issue
Molecular Tuning of IR-786 for Improved Brown Adipose Tissue Imaging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Antiproliferative Evaluation and QSAR Analysis of Novel Halogen- and Amidino-Substituted Benzothiazoles and Benzimidazoles

by
Valentina Rep Kaulić
1,
Livio Racané
2,
Marijana Leventić
3,
Domagoj Šubarić
4,
Vesna Rastija
4,
Ljubica Glavaš-Obrovac
3,* and
Silvana Raić-Malić
1,*
1
Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 20, 10000 Zagreb, Croatia
2
Department of Applied Chemistry, Faculty of Textile Technology, University of Zagreb, Prilaz Baruna Filipovića 28, 10000 Zagreb, Croatia
3
Department of Medicinal Chemistry, Biochemistry and Laboratory Medicine, Faculty of Medicine Osijek, University Josip Juraj Strossmayer of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
4
Faculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek, Vladimira Preloga 1, 31000 Osijek, Croatia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15843; https://doi.org/10.3390/ijms232415843
Submission received: 24 November 2022 / Revised: 8 December 2022 / Accepted: 9 December 2022 / Published: 13 December 2022
(This article belongs to the Collection Feature Papers in Molecular Pharmacology)

Abstract

:
Syntheses of 6-halogen-substituted benzothiazoles were performed by condensation of 4-hydroxybenzaldehydes and 2-aminotiophenoles and subsequent O-alkylation with appropriate halides, whereas 6-amidino-substituted benzothiazoles were synthesized by condensation of 5-amidino-2-aminothiophenoles and corresponding benzaldehydes. While most of the compounds from non-substituted and halogen-substituted benzothiazole series showed marginal antiproliferative activity on tested tumor cell lines, amidino benzazoles exhibited stronger inhibitory activity. Generally, imidazolyl benzothiazoles showed pronounced and nonselective activity, with the exception of 36c which had a strong inhibitory effect on HuT78 cells (IC50 = 1.6 µM) without adverse cytotoxicity on normal BJ cells (IC50 >100 µM). Compared to benzothiazoles, benzimidazole structural analogs 45a45c and 46c containing the 1,2,3-triazole ring exhibited pronounced and selective antiproliferative activity against HuT78 cells with IC50 < 10 µM. Moreover, compounds 45c and 46c containing the methoxy group at the phenoxy unit were not toxic to normal BJ cells. Of all the tested compounds, benzimidazole 45a with the unsubstituted phenoxy central core showed the most pronounced cell growth inhibition on THP1 cells in the nanomolar range (IC50 = 0.8 µM; SI = 70). QSAR models of antiproliferative activity for benzazoles on T-cell lymphoma (HuT78) and non-tumor MDCK-1 cells elucidated the effects of the substituents at position 6 of benzazoles, demonstrating their dependence on the topological and spatial distribution of atomic mass, polarizability, and van der Waals volumes. A notable cell cycle perturbation with higher accumulation of cells in the G2/M phase, and a significant cell increase in subG0/G1 phase were found in HuT78 cells treated with 36c, 42c, 45a45c and 46c. Apoptotic morphological changes, an externalization of phosphatidylserine, and changes in the mitochondrial membrane potential of treated cells were observed as well.

Graphical Abstract

1. Introduction

Due to its high prevalence, complexity, and dangerous mortality rate, cancer has been listed as the second leading cause of death worldwide, accounting for nearly 10 million deaths in 2020, or nearly one in six deaths [1]. Since 2020, cancer chemotherapy has been severely impacted by the COVID-19 pandemic, resulting in delays in diagnosis and treatment, which may lead to an increase in advanced stage disease and ultimately increased mortality [2]. Given the limited efficacy of currently available anticancer drugs and the rapid development of resistance due to genetic mutations of accessible targets, there is a growing need to design and develop new drug candidates with higher efficacy and lower toxicity [3].
As in recent years, many drugs authorized in 2020 contain nitrogen aromatic heterocycles [4]. Nitrogen aromatic heterocycle-based compounds exhibit anticancer effects through either cell growth arrest or induction of cell differentiation and apoptosis [5]. Owing to structure similarity to the natural occurring purines, benzimidazole and benzothiazole derivatives are useful scaffolds in drug discovery of anticancer agents [6,7,8,9,10]. Some benzimidazole-based compounds such as abemaciclib, bendamustine, crenolanib, dovitinib, galeterone, glasdegib, liarozole, nocodazole, pracinostat, selumetinib, and veliparib have been approved for the treatment of various cancers [11]. Moreover, benzimidazole [11,12,13] and benzothiazole [14,15,16] hybrids exhibit dual or multiple antiproliferative activities and, therefore, have the potential to increase efficacy and overcome cancer drug resistance. The 2-arylbenzothiazole derivative CJM-126 (I) (Figure 1) was found to exhibit potent growth inhibition on human-derived breast carcinoma MCF-7 cell lines, including estrogen receptor-positive MCF-7wt cells with an IC50 value < 0.001 µM [17,18]. Another example of highly potent anticancer agent is benzothiazole analog MKT-077 (II), a water-soluble rhodocyanine dye, acting as an inhibitor of heat shock protein 70 (Hsp 70) [19]. 6-Fluorobenzothiazole (PMX 610) (III) exhibited potent and selective in vitro antitumor properties in human cancer cell lines (e.g., colon, non-small cell lung, and breast subpanels) [20,21,22], while compound 5F203 (IV) proved efficient in nanomolar range against MCF-7 cells. Its prodrug Phortress (V), with high bioavailability tested in clinical trials, showed activity against renal, breast, ovarian and colorectal solid carcinoma. Its mechanism of action involves in vivo hydrolysis to release 5F203 (IV), which is further metabolized by the P450 enzyme CYP1A1 to a highly reactive species, which attacks and breaks DNA strands, ultimately leading to cell death [9,17,23,24].
We have found that, among diverse benzimidazole amidines, imidazolyl benzimidazole with benzyl-1,2,3-triazole VI (Figure 2) exhibits potent growth-inhibitory activity against non-small cell lung cancer A549 cells, which was associated with induction of apoptosis and primary necrosis [25]. 1-(p-Chlorophenyl)-1,2,3-triazole-tagged benzimidazole VII also showed selective inhibitory effect on A549 cells, inducing p38 MAPK- and NF-κB-mediated apoptosis [26]. Moreover, in the series of benzothiazole amidines, VIII and IX, which exhibited strong antiproliferative effect on colorectal cancer SW620 and MCF-7 cell lines, respectively, also showed noncovalent interaction with 6-amidinobenzothiazole ligands, demonstrating both minor groove binding, and intercalating mode of DNA interaction [27].
In view of the biological importance of benzothiazole and benzimidazole pharmacophores, and as a part of ongoing research focused on the development of new anticancer agents [25,26,27], we have designed and synthesized a small library of 2-aryl-substituted benzothiazole and benzimidazole entities aiming to evaluate their antiproliferative activities (Figure 2). In this context, halogen and amidino benzothiazoles were linked via phenoxymethylene spacer to diverse aromatic subunits, to ensure the distribution of highly hydrophilic and hydrophobic parts of the structure. Antiproliferative effects of novel 6-halogen, 6-imidazolyl and 6-pyrimidinyl benzothiazole derivatives, and previously prepared benzimidazoles [28], were evaluated on selected human tumor cell lines. The results of quantitative structure–activity relationship (QSAR) analysis on T-cell lymphoma (HuT78) and Madin–Darby canine kidney cells (MDCK1) were compared and discussed. Imidazolyl benzothiazole 36c, and pyrimidinyl benzimidazoles 42c, 45a, 45b, 45c, and 46c with potent and selective antiproliferative activity were also evaluated in cell-cycle perturbation and mitochondrial membrane potential assays.

2. Results and Discussion

2.1. Chemistry

A library of 22 previously published pyrimidinyl benzimidazole [28] derivatives was expanded with novel 55 halogen- and amidine-substituted benzothiazole analogs, prepared by a multi-step synthetic route as shown in Scheme 1, Scheme 2, Scheme 3 and Scheme 4. Benzoyl (15a15c, 16a16c, 17a17c) and picolyl (18a18c, 19a19c, 20a20c) 6-halogen-substituted and 6-unsubstituted benzothiazoles were prepared by a four-step synthesis. The key intermediates 9a9c, 10a10c and 11a11c were prepared in moderate reaction yields (33%–78%) by condensation of corresponding 4-hydroxybenzaldehydes 8a8c and 2-aminotiophenoles 57 using sodium metabisulfite (Na2S2O5) as a mild oxidant (Scheme 1). A base-promoted O-alkylation of 2-(4-hydroxyphenyl)benzothiazoles (9a9c, 10a10c) with corresponding halides gave target 6-halogen-substituted benzothiazole derivatives with benzoyl (15a15c, 16a16c) and picolyl (18a18c, 19a19c) units, and introduced the phenoxymethylene linker in low to moderate yields (22–66%), whereas 6-unsubstituted benzothiazole analogs 17a17c and 20a20c were isolated in good to high yields (41–81%). O-Propargylated intermediates (12a12c, 13a13c, 14a14c) for the synthesis of 1,2,3-triazolyl linked 2-arylbenzothiazole derivatives (21a26a, 21b26b and 21c26c) were prepared. To evaluate the effect of triazole moieties on biological activity, 1H-1,2,3-triazole benzothiazole derivatives 21a21c, 22a22c and 23a23c were synthesized via a regioselective copper(I) catalyzed cycloaddition, with copper(I) iodide and trimethylsilylazide, while benzothiazoles 24a24c, 25a25c, and 26a26c with 1-benzyl-1,2,3-triazole unit were obtained by the one-pot click reaction with benzyl azide formed in situ using Cu(II) acetate as a catalyst (Scheme 2).
To further explore the influence of the amidino substituent in C-6 of the benzothiazole core and to improve solubility, in addition to a series of pyrimidinyl benzimidazole derivatives prepared according to the procedure previously reported by our group [28], a series of 6-imidazolyl 38a38c, 39a, 39c, 40a40c, 41a41c and 6-pirimidinyl 42a, 42b, 43c, 44a44c, 45a, 45c benzothiazoles was synthesized as shown in Scheme 3 and Scheme 4.
Firstly, benzaldehyde precursors 27a27c, 28a28c, and 29a29c were obtained through O-alkylation of 4-hydroxybenzaldehydes. Followed by a copper(I)-catalyzed Huisgen 1,3-dipolar cycloaddition reaction of benzaldehydes 27a27c, 1,2,3-triazole-substituted benzaldehydes 30a30c, 31a31c were obtained [28]. Amidino-substituted 2-aminothiophenole 32 and 33 were prepared from 6-cyanobenzothiazole by the Pinner method [29,30]. Benzothiazoles 34a34c, 35a, 35c, 36a36c and 37a37c were prepared by cyclocondensation of amidino-2-aminothiophenole 32 with benzaldehyde precursors (27a27c, 28a28c, 29a29c) in acetic acid, while the benzothiazoles 38a, 38b, 39c, 40a40c, 41a and 41c [31] were synthesized from amidino-2-aminothiophenole 33 with corresponding benzaldehydes. Finally, targeted amidino-substituted benzothiazole hydrochlorides were prepared by an acid-base reaction.

2.2. Evaluation of Antiproliferative Activity

The antiproliferative activities of novel benzothiazole derivatives (15a26a, 15b26b, 15c26c, 34a38a, 40a, 41a, 34b, 36b38b, 40b, 34c37c, and 39c41c on human tumor cell lines, including cervical adenocarcinoma (HeLa), colon adenocarcinoma (CaCo-2), T-cell lymphoma (HuT78), and non-tumor Madin–Darby canine kidney (MDCK-1) cells and human fibroblasts (BJ) are presented in Table 1 and Table 2 as well as in Tables S1 and S2 (Supplementary Materials). 5-Fluorouracil (5-FU) was used as the reference drug. In order to assess the impact of the benzimidazole moiety as benzothiazole bioisoster on the antiproliferative activity and to compare inhibitory effects of benzothiazoles and their benzimidazole analogs, activity of previously synthesized amidino benzimidazoles 42a49a, 42b45b, 47b49b, 42c46c, 48c and 49c was also evaluated (Table 1, Table 2, Tables S1 and S2).
According to the results presented in the Table 1, the majority of compounds from non-substituted and halogen-substituted benzothiazole series showed marginal antiproliferative activity on tested tumor cell lines. From benzothiazoles without 1,2,3-triazole moiety 15a20a, 15b20b, and 15c20c, benzothiazoles with benzoyl moiety showed to be less active compared to their analogs with the picolyl aromatic unit. In line, 6-chlorobenzimidazole 18a containing pyridinyl exhibited the best inhibitory activity (IC50 = 13.2 µM) on T-cell lymphoma (HuT78) cells, while benzothiazole 20b, with fluorine attached at phenyl central unit, had moderate activity on CaCo-2 cells (IC50 = 28.7 µM), which was lower to that of 5-FU. Among 4-(1,2,3-triazolylmethoxy)phenyl benzothiazoles 21a26a, 21b26b and 21c26c, compounds 21a21c, 22a22c and 23a23c with terminal 1H-1,2,3-triazole ring exhibited higher activity than those with 1-benzyl-1,2,3-triazole 24a24c, 25a25c and 26a26c. Although cytotoxic in contact with normal MDCK-1 cells, these compounds exhibited only marginal inhibitory effects on growth of normal human fibroblasts (BJ). 6-Chlorobenzothiazoles 21a and 21b and 6-fluorobenzothiazole 22b had the best activity on HuT78 cells (21a: IC50 = 6.8 µM, 21b: IC50 = 3.6 µM, 22b: IC50 = 9.1 µM). In comparison, 5-FU did not exhibit antiproliferative effect on HuT78 cells. Overall, our data suggest increased antiproliferative activity of compounds with fluorine substituent attached at aromatic central unit, while methoxy group-substituted entities demonstrated decreased inhibition efficiency in HuT78 cells. Conversely, the methoxy group in 6-unsubstituted benzothiazoles 26a26c with 1,4-disubstituted 1,2,3-triazole improved growth inhibition in both HeLa (IC50 = 23.3 µM) and HuT78 (IC50 = 23.7 µM) cell lines.
Nineteen compounds from the amidino benzothiazole and twenty two compounds from amidino benzimidazole series (Table 2) showed stronger growth inhibition than halogen- and unsubstituted benzothiazole derivatives. Imidazolyl benzothiazoles showed strong antiproliferative activity on all tested tumor cell lines. However, these compounds were also toxic on both normal cell lines, MDCK1 and BJ cells. 5-FU showed to be less cytotoxic on MDCK1 and BJ cells with values of selectivity index (SI) from 6.7 to 9.3. Interestingly, the introduction of the 1H-1,2,3-triazole moiety at phenyl in compounds 36a36c, resulted in less cytotoxicity against normal MDCK1 and BJ cells, while maintaining excellent growth-inhibitory effect on HuT78 cell lines (36a: IC50 = 4.4 µM; 36b: IC50 = 1.8 µM; 36c: IC50 = 1.6 µM) with SI of 8.8, 18.1 and 62.5, respectively, with respect to the inhibition of BJ cells (Table S2). The methoxy group at phenyl in 36c caused a selective and pronounced antiproliferative effect on HuT78 cells. Replacement of the 6-imidazolyl with the 6-pyrimidinyl group in 38a, 38b, 39c, 40a40c, 41a and 41c reduced their inhibitory activity. From pyrimidinyl benzothiazole derivatives, only 38b and 41a expressed strong albeit not selective activity on HuT78 cells. Some 6-pyrimidinyl benzothiazoles were devoid of any antitumor activities. Thus, pyrimidinyl derivatives 40a40c and 41c compared to their imidazolyl analogs 36a36c and 37c did not exhibit inhibitory effect on tested cell lines, except for 40b which showed only moderate activity against HuT78 cells (IC50 = 37.3 µM).
Among benzimidazole amidines 42a49a, 42b45b, 47b49b, 42c46c, 48c and 49c, the strongest antiproliferative activity on HuT78 cells was observed for analogs 45a (IC50 = 4.8 µM), 45b (IC50 = 5.5 µM), and 45c (IC50 = 4.1 µM) with 1-benzyl-1,2,3-triazole substituent at the phenoxy core. A similar effect was observed for compound 46c (IC50 = 4.1 µM), containing 1-ethylmorpholino-1,2,3-triazole side chain, with SI (compared to the inhibition of BJ cells) of 11.6, 14.9, 23.4 and 19.6, respectively (Table S2). Their benzothiazole analogs 37a37c were more cytotoxic to human normal fibroblasts (BJ) with SI of 2.7, 2.5 and 1.6, respectively. Compared to benzothiazoles 34a and 38a with benzoyl moiety, benzimidazole structural analog 42a exhibited significant and selective antiproliferative activity against T-cell lymphoma (IC50 = 7.0 µM on HuT78 cells, IC50 > 100 µM on MDCK1 and BJ cells). As shown in Table 2, benzimidazoles containing picolyl 43a43c and 1H-1,2,3-triazole 44a44c moiety exhibited in turn decreased inhibitory activity compared to those of benzothiazole congeners. Moreover, introduction of aliphatic moiety in benzimidazole amidines 47a49a, 47b48b and 48c49c resulted in a loss of antiproliferative activity.
Of the 77 benzazole analogs that were evaluated for their antiproliferative activity, twelve benzazoles (36a, 36c, 38b, 39c, 42a42c, 43a, 45a45c and 46c) with marked and selective activity were chosen for evaluation on additional tumor cell lines, i.e., colorectal adenocarcinoma, metastatic (SW620), human breast adenocarcinoma (MDA-MB-231), promyelocytic leukemia (HL60) and human monocytic leukemia (THP1) cell lines (Table 3).
Interestingly, amidino benzothiazole 36c with the methoxy group at the phenoxy core did not exhibit growth inhibition on evaluated cell lines showing selective antiproliferative activity only on HuT78 cells (Table 2 and Table 3). Similarly, amidino benzimidazole 46c with methoxy substituent showed notable antiproliferative activity only on HuT78 cell line (Table 2), along with moderate activity on HL60 cells (Table 3). In contrast to selective antiproliferative activity of 36c on HuT78 cells, its structural analog 36a with the unsubstituted phenoxy unit showed inhibitory activity on additional set of tumor cells, particularly on SW620 cells, with IC50 values ranging from 5.6 µM to 33.0 µM. Benzothiazoles with benzoyl 38b and picolyl 39c moiety also exhibited moderate inhibitory potency on additional tumor cell lines. Among amidino benzimidazoles, 1-benzyl-1,2,3-triazole analogs 45a and 45b showed the best antiproliferative activity on both sets of tumor cell lines. Of all tested compounds, benzimidazole 45a with the unsubstituted phenoxy unit showed the most pronounced cell growth inhibition efficiency on THP1 cells in nanomolar range (IC50 = 0.8 µM; SI = 69.5; Table S3). Its fluoro-substituted structural analog 45b had also marked activity on THP1 cell line with IC50 = 2.5µM and SI = 32.4, while methoxy-substituted analog 45c demonstrated only moderate activity (IC50 = 25.1µM; SI > 44) on HL60 cells (Table S3).
To assess whether cell cycle disturbance is a possible mechanism of action for the antiproliferative activity of the tested compounds, six analogs (36c, 42c, 45a45c, and 46c) were tested in cell cycle perturbation experiments on HuT78 cells. As shown in Figure 3a, 24 h post-treatment effects of all applied compounds (5 µM) induced disturbance of cell cycle division in treated compared to control (untreated) cells. Enrichment of the G0/G1 cell fraction was evident in cell lines treated with all tested analogs, whereas decrease in G2/M phase was noticed only following 42c, 45a, 45b, and 45c cell treatment. Untreated cells followed normal diploid distribution exhibiting regular proliferative features [32].
In our experimental conditions, after 48 h of treatment of HuT78 cells, all tested compounds expressed similar effect pattern. A significant cell cycle perturbation characterized by cell accumulation in the G2/M and subG0/G1 phase was evident in treated compared to the non-treated cells (Figure 3b). A SubG1 peak is indicative of DNA fragmentation, plausibly due to apoptosis-induced cell. A decrease in the polyploid cell number of treated compared to the untreated cells was also observed. Polyploidy is a common tumor feature, and pan-cancer analyses confirm that 28.2–37% of human cancers undergo polyploidization [33,34]. Polyploidy enables large phenotypic leaps, providing tumors with access to many different therapy-resistant phenotypes.
Since apoptosis is frequently accompanied by complex mitochondrial changes, alterations in the mitochondrial membrane potential may signal early apoptotic events or, may reflect changes in the apoptotic signaling pathways [35]. In response to multiple intracellular stress conditions, mitochondrial membranes can become permeabilized due to the pore-forming activity of proapoptotic Bcl-2 protein family members. Alternatively, mitochondria can lose their structural integrity after the mitochondrial permeability transition, a phenomenon that is initiated at the mitochondrial inner membrane [36]. In both cases, permeabilized mitochondria allow the release of proapoptotic proteins into the cellular cytoplasm.
To prove apoptosis as a mechanism of treated cells death, we used two methods for apoptosis detection, tracing signs of phosphatildylserine translocation to the extracellular membrane and accompanying changes in the mitochondrial membrane potential (∆Ψm). Results of Annexin-V flow cytometry measurements in selected cell lines, 24 h after compound exposure, showed no significant increase in the proportion of apoptotic cells in treated versus nontreated cells. However, the number of apoptotic cells increased (Figure 4a,b) following 48 h post-treatment with 36c, 42c, 45a45c and 46c. A statistically significant difference was observed in comparison to cells treated with 42c, 45a and 45b (Figure 4b).
To determine the effects of selected compounds on the function of mitochondria, ΔΨm was assessed in HuT78 cells after 48 h of treatment by chosen compounds (36c, 42c, 45a45c and 46c). Changes in the (∆Ψm) were measured using TMRE (Tetramethylrhodamine, Ethyl Ester, Perchlorate) dye. Flow cytometric analysis showed statistically significant changes in mitochondrial membrane potential. Obtained results are consistent with the results of previously published studies which showed that some benzothiazole derivatives have the potential to induce apoptosis of B and T lymphoma cells by the intrinsic pathway through disruption of mitochondrial membranes [8,37].
The given results suggest that disruption of mitochondrial membrane potential produced by tested compounds can lead to cytotoxicity and cell death by apoptosis and/or necrosis as shown in Figure 5.

2.3. QSAR Study

Cytotoxic effects of 77 benzothiazoles and benzimidazoles against normal MDCK-1 cells were ranked by activities, and 17 compounds were chosen for the test set. The best QSAR model expressed by multiple linear regression equation generated by five molecular descriptors is:
log IC50 = −13.88 − 8.18 SIC1 − 2.27 GATS4p − 2.54 BEHv6 + 14.87 BELp1 + 6.26 R7m
Ntrain = 60; Ntest = 17.
Williams plot (Figure 6), which shows the applicability domain of model (1a) detected molecule 25b as a border outlier and no molecule out of the warning leverage. Outlying behavior of compound 25b was expected since it demonstrated lowest antiproliferative activity against MDCK-1 cells among two fluoro-substituted benzothiazoles (16b, 19b, 22b) (Table 1). Removing molecule 25b from the training set, and subsequent re-analysis produced a following improved QSAR model:
log IC50 = −14.56 − 8.45 SIC1 − 2.37 GATS4p − 2.70 BEHv6 + 15.61 BELp1 + 5.93 R7m
Ntrain = 59; Ntest = 17.
Experimental and calculated logIC50 by Equation (1a,b), as well as values of included descriptors are shown in the Supplementary Materials, Table S4.
QSAR study for antiproliferative activity on T-cell lymphoma (HuT78) cells was performed on a total of 59 molecules. The set was split into 12 molecules in the test set by activity ranking method, and the remaining 47 candidates were part of the training set. Considering the number of molecules in the dataset, the number of descriptors in the model was limited to four.
Log IC50 = 5.14 − 3.38 MATS8v − 2.44 Mor30m + 1.63 Mor09p − 4.12 E2u
Ntrain = 47; Ntest = 12.
Experimental and calculated logIC50 for HuT78 cell line by Equation (2a), as well as values of included descriptors are shown in the Supplementary Materials, Table S5. Inspection of Williams plot (Figure 7) for the applicability domain of model (2a) revealed three outliers (25b, 39c, 47b), which have been removed subsequently from the original set. Molecule 25b had a lower calculated value of logIC50 than the measured value, probably because of the presence of two fluorine atoms at the positions R1 and R2 (Table 1), while molecules 39c and 47b exhibited higher activity than calculated. Performing the QSAR analysis on dataset without these outliers, with the same descriptors, resulted on the model of better quality:
log IC50 = 5.9 − 2.95 MATS8v − 2.51 Mor30m + 1.79 Mor09p − 4.98 E2u
Ntrain = 45; Ntest = 11.
Calculated log IC50 by Equation (2a,b), as well as values of included descriptors are shown in the Supplementary Materials, Table S5. The statistical results of QSAR models (1a, 1b, 2a, and 2b) are presented in Table 4. Low collinearity among descriptors in all the models was confirmed by a low value of the global correlation among descriptors (KXX) and the difference between correlation among the descriptors and response variable (KXY) and KXXK) higher than 0.05 [38]. The absence of collinearity in models 1a and 2a was also verified by the values of the correlation coefficient (R ≤ 0.7) in the correlation matrix (Table 5 and Table 6, respectively).
Statistical parameters, presented in Table 4, confirmed that all four models satisfied fitting abilities: coefficients of determination (R2tr) were greater than 0.60, and higher than adjusted coefficient of determination (R2adj). The concordance correlation coefficient of the training set (CCCtr) was also higher than 0.80 [39]. In order to assess the internal prediction power and stability of QSAR models, leave-one-out (LOO) cross validation technique was performed. The statistical significance of the models was proven by the cross-validated correlation coefficient (Q2LOO), which was higher than 0.05 for all models, but the largest was for model (2b). The differences between R2 and Q2LOO did not exceed 0.2–0.3. Additionally, the root-mean-square errors of the cross-validated method (RMSEcv) were higher than root-mean-square error of the training set (RMSEtr). An average value of squared correlation coefficients (r2m) between the observed and LOO predicted values of the compounds is a measure for internal validation. Their values were > 0.5 for all models, except for the model (1a). Similarly, the absolute differences between the observed and leave-one-out predicted values of the compounds (Δr2m) were < 0.2 for all models, except for model (1a), which indicates the low predictive ability of the model (1a), despite of its large Q2LOO (0.64) [40]. After the exclusion of the outlier, molecule 25b, the model (1b) exhibited better internal predictivity. Y-Randomization test was performed to check the robustness of the obtained QSAR models. The values of both coefficients, R2Yscr (Y-scramble correlation coefficients) and Q2Yscr (Y-scramble cross-validation coefficients) were <0.02, implying that models were not obtained by chance [38]. Predictive power of obtained QSAR models were validated by parameters of the external validation, i.e., the coefficients of determination of validation set (R2ext) were > 0.60, concordance correlation coefficient of the test set (CCCext) was ≥ 0.85 (except for model (1a)), and the root-mean-square error of the external validation set (RMSEex), and mean absolute error of the external validation set (MAEex) were close to zero. The external performance of all four models in terms of external explained variance (Q2F1, Q2F2, Q2F3), which should be > 0.60 was satisfying [41,42]. The chemical domain of applicability defined the structural, physicochemical, and response space of the obtained models. Williams plots (Figure 1 and Figure 2), except mentioned outliers, did not detect structurally influential chemicals in models, in which leverage in the original variable space (h) was not higher than warning leverage (h*) [43]. Generally, QSAR model (2b) for antiproliferative activity on HuT78 cell provided better statistical quality with better predictability in comparation to the model for MDCK-1 cells (1a).
QSAR model for cytotoxic effects against non-tumor MDCK-1 cells contained two BCUT (Burden eigenvalues) descriptors; BELp1 was the lowest negative eigenvalue num. 1 weighted by polarizability, while the BEHv6 was the positive highest eigenvalue n. 6 of Burden matrix weighted by atomic van der Waals volumes [44]. Amidino benzothiazoles and amidino benzimidazoles had higher values of BEHv6 than their halogen- and unsubstituted benzothiazole derivatives (Table S4), and thus, according to the negative values of BEHv6 coefficient in the model (1a), lower values of logIC50, meaning that they were more toxic against MDCK-1 cells. For example, 6-imidazolyl benzothiazoles had larger substituents at the position R1 than their unsubstituted benzothiazole analogs, and therefore had higher values of BEHv6 and stronger inhibition against MDCK-1 cells (17a, BEHv6 = 2.173; logIC50 = 2.00, and 34a, BEHv6 = 3.085; logIC50 = 0.45; 20a, BEHv6 = 2.719; logIC50 = 1.82, and 35a, BEHv6 = 2.934; logIC50 = 0.43; 23a, BEHv6 = 2.711; logIC50 = 1.61, and 36a, BEHv6 = 2.766; logIC50 = 1.50). Values of descriptor BEHv6 also explain the decrease in cytotoxicity against normal MDCK1 cells by replacement of 1-benzyl-1H-1,2,3-triazole moiety in 37a (BEHv6 = 3.127; logIC50 = 0.18) by substituent of lower total atomic van der Waals volumes, 1H-1,2,3-triazole in 36a (BEHv6 = 2.766; logIC50 = 1.5). Descriptor BELp1 discriminates well benzothiazoles from benzimidazoles analog, while it was not sensitive to changes of substituents at phenoxymethylene unit within the groups of ac compounds (Table S4). The presence of sulphur atom in benzothiazole, instead of a nitrogen atom in benzimidazole, determined their lower values of BELp1 descriptors (Table S4), and thus lower values of logIC50 (stronger antiproliferative effect on MDCK-1 cells), which is in accordance with the positive coefficient of these descriptors in models (1a) and (1b). Thus, benzothiazole 41a had logIC50 of 1.76 (BELp1 = 2.02), while its benzimidazole analog 45a had higher value of logIC50 (logIC50 = 2.00, BELp1 = 2.039). Similarly, benzimidazole 42a exhibited logIC50 of 2.00 (BELp1 2.039) that is higher to that of its benzothiazole analog 38a (logIC50 = 1.43, BELp1 2.02). R-GETAWAY (Geometry, Topology, and Atom-Weights AssemblY) descriptor R7m, encoded the information about the 3D distribution of atomic mass at the topological distance 7 [45,46]. Introduction of the pyrimidinyl group at 6-position of amidino benzothiazoles (34a, 34b, 35c, 37a37c) enhanced the values of the R7m descriptor compared to their 6-imidazolyl analogs (38a, 38b, 39c, 41a41c) (Table S4). Therefore, in accordance with the positive coefficient of R7m in Equation (1a,b), 6-pyrimidinyl benzothiazoles were found to be less toxic on the MDCK-1 cells compared to their 6-imidazolyl analogs. Descriptor R7m was extremely sensitive to the difference in the 3D distribution of atomic mass at the topological distance 7 between the benzothiazoles without 1,2,3-triazole moiety. Benzothiazoles with the picolyl aromatic unit (18a18c, 19a19c, 20a20c) had lower values of R7m, and therefore decreased values of logIC50 (more active against MDCK-1 cells) compared to their analogs with benzoyl moiety (15a15c, 16a16c, 17a17c). Descriptor GATS4p is a Geary 2D autocorrelations descriptor that reflects a level of independence of polarizability of one atom in the molecular structure on the polarizability of other atoms at the spatial lag 4 [46]. Highest values of these descriptors had 6-fluorobenzothiazoles (16a16c, 19a19c, 22a22c), since they possess highly polarizable sulphur atom at the topological distance 4 from strongly electrophilic fluorine atom. These compounds were found to be particularly toxic on MDCK-1 cells, which is in accordance with the negative coefficient of GATS4p in Equations (1) and (1b). Descriptor SIC1 is structural information content with the first order of symmetry neighborhood of vertices in a hydrogen-filled graph [47]. Molecules containing three adjacent nitrogen atoms in 1,2,3-triazole moiety had enhanced values of SIC1 descriptor (Table S4). This is especially expressed in benzothiazoles with terminal 1H-1,2,3-triazole ring (21a21c, 22a22c, 23a23c), which strongly inhibited MDCK-1 cells.
QSAR models (2a and 2b) for the antiproliferative activities against HuT78 cell contain two 3D-MoRSE (Molecular Representation of Structures based on Electronic diffraction) descriptors, Mor30m and Mor09p. Descriptors Mor30m and Mor09p reflect the contribution of the 3D distribution of atomic mass at a scattering parameter s = 29 Å−1, and atomic polarizability at the scattering parameter s = 8 Å−1 [48]. The negative coefficient of the descriptor Mor30m in models (2a) and (2b) showed that its higher values correspond to a higher antitumor effect (Table S5). Because of the presence of sulphur atom, amidino benzothiazoles (34a38a, 34b, 36b38b, 40b, 34c41c) had higher values of Mor30m than amidino benzimidazoles, which implies lower values of logIC50, therefore stronger antiproliferative activity on HuT78 cells. Among amidino benzimidazoles, 1,4-disubstited 1,2,3-triazoles (45a45c, 46c) showed to be the most active (Table 2). These compounds had higher values of Mor30m than their unsubstituted 1,2,3-triazole analogs (44a44c) (Table S5) that caused stronger antiproliferative activity on HuT78 cells. The descriptor Mor09p is sensitive to the position of atoms with higher polarizability. For example, compound 36c had higher negative value of Mor09p (−1.518, Table S5) and thus lower value of logIC50 (0.2) than compound 36b with fluorine at the position R2 (Mor09p = -1.488, logIC50 = 0.26). Therefore, substituents with less polarizability decreased the activity against the HuT78 cells. Descriptor MATS8v is Moran autocorrelation of lag 8/weighted by atomic van der Waals volumes [48]. This autocorrelation descriptor represents atomic van der Waals volumes at the topological distances 8. Among benzothiazoles 21a21c, 22a22c and 23a23c with terminal 1H-1,2,3-triazole ring, compounds with the methoxy group (21c, 22c and 23c) had higher values of MATS8v. The oxygen atom from the methoxy group is at the topological distances 8 from the atom at the R1 position. Since oxygen atoms had higher van der Waals volumes than hydrogen or fluorine atoms, these compounds had higher values of MATS8v than compounds 21a, 21b, 22a, 22b, 23a, 23b and lowest activity against HuT78. Descriptor E2u is 2nd component accessibility directional WHIM (Weighted Holistic Invariant Molecular descriptors) index/unweighted. E is distribution embedded along axes, and it is also directional [48]. Descriptor E2u represents a dispersion measure of the projected atoms along the second principal axis, accounting for the molecular size along this principal direction. The compounds 21a21c, 22a22c and 23a23c with terminal 1H-1,2,3-triazole ring had higher values of E2u descriptor than 1-benzyl-1,2,3-triazoles 24a24c, 25a25c and 26a26c, and therefore exhibited higher activity against HuT78.

3. Materials and Methods

3.1. General

All the solvents and chemicals were purchased from commercial suppliers Aldrich (St. Louis, MO, USA) and Acros (Geel, Belgium). For monitoring the progress of a reaction and for comparison purpose, thin-layer chromatography (TLC) was performed on precoated Merck silica gel 60F-254 plates (Merck, Kenilworth, NJ, USA) using an appropriate solvent system, and the spots were detected under ultraviolet (UV) light (254 nm). For column chromatography, 0.063–0.2 mm silica gel (Fluka, Seelze, Germany) was employed, glass column was slurry-packed under gravity. Nuclear magnetic resonance (NMR) spectroscopic data for 1H and 13C nuclei (Figures S1–S65, Supplementary Materials) were recorded at room temperature on a spectrometer Bruker Avance (Bruker, Billerca, MA, USA) 300 MHz and 600 MHz. All NMR spectra were measured in deuterated dimethyl sulfoxide, DMSO with tetramethylsilane as an internal standard. Individual resonances were assigned on the basis of their chemical shifts, signal intensities, multiplicity of resonances, and H–H coupling constants. Melting points were recorded using Kofler micro hot-stage (Reichert, Wien, Austria) and Thermovar HT1BT1 (Reichert, Wien, Austria). Elemental analyses for carbon, hydrogen, and nitrogen were performed on a Perkin–Elmer 2400 elemental analyzer. Analyses are indicated as symbols of elements, and the analytical results obtained are within 0.4% of the theoretical value.

3.2. Experimental Procedure for Preparation of Compounds

Compounds 6-chlorobenzothiazol-2-amine 3 [49], 6-fluorobenzothiazol-2-amine 4 [50], 2-amino-5-chlorobenzenethiol 5 [51], 2-amino-5-fluorobenzenethiol 6 [52], 27a [53], 27b [54], 27c [55], 28a [54], 28b [54], 28c [54], 29a [55], 29b [54], 29c [54], 30a [54], 30b [28], 30c [28], 31a [25], 31b [54], 31c [54], 32 [29], 33 [30], hydrochloride 38a [28], hydrochloride 38b [28], hydrochloride 39c [28], hydrochloride 40a [28], hydrochloride 40b [28], hydrochloride 40c [28], hydrochloride 41a [28], hydrochloride 41c [28], were synthesized in accordance with procedures given in the literature.

3.2.1. General Procedure for Preparation of 2-(4-Hydroxyphenyl)benzothiazole Derivatives 9a9c, 10a10c, and 11a11c

To a solution of the corresponding 2-aminobenzenethiole (57, 1 eq) in DMF corresponding benzaldehyde (8a8c, 1.1 eq) and Na2S2O5 (1.1 eq) were added and reaction mixture was stirred at 100 °C for 2 h. Solvent was evaporated and the residue was purified by column chromatography (CH2Cl2:CH3OH = 50:1).
6-Chloro-2-(4-hydroxyphenyl)benzothiazole 9a. Compound 9a was prepared using the above-mentioned procedure from 5 (1.60 g, 10 mmol) and 8a (1.34 g, 11 mmol) to obtain 9a as brown powder (1.66 g, 63%; m.p. 169–172 °C). 1H NMR (600 MHz, DMSO) δ 10.27 (1H, s, OH), 8.24 (1H, d, J = 2.1 Hz), 7.96 (1H, d, J = 8.7 Hz), 7.92 (2H, d, J = 8.7 Hz), 7.52 (1H, dd, J = 8.6, 2.2 Hz), 6.93 (2H, d, J = 8.7 Hz). 13C NMR (151 MHz, DMSO) δ 168.45, 160.73, 152.48, 135.64, 129.21, 129.12, 126.84, 123.64, 123.35, 121.78, 116.11.
6-Chloro-2-(3-fluoro-4-hydroxyphenyl)benzothiazole 9b. Compound 9b was prepared using the above-mentioned procedure from 5 (1.60 mg, 10 mmol) and 8b (1.54 mg, 11 mmol) to obtain 9b as brown powder (1.74 g, 62%; m.p. 183–186 °C). 1H NMR (600 MHz, DMSO) δ 10.76 (1H, s, OH), 8.26 (1H, d, J = 2.1 Hz), 7.98 (1H, d, J = 8.7 Hz), 7.85 (1H, dd, J = 11.9, 2.1 Hz), 7.74 (1H, dd, J = 8.4, 1.9 Hz), 7.54 (1H, dd, J = 8.7, 2.2 Hz), 7.13 (1H, t, J = 8.6 Hz). 13C NMR (75 MHz, DMSO) δ 167.73 (d, JCF = 2.7 Hz), 151.57 (d, JCF =241,5 Hz), 152,75, 148.87 (d, JCF = 12.1 Hz), 136.31, 130.05, 127.50, 124.97 (d, JCF = 2.8 Hz), 124.54 (d, JCF = 6.6 Hz), 124.08, 122.38, 118.84 (d, JCF = 3.3 Hz), 115.20 (d, JCF = 20.1 Hz).
6-Chloro-2-(4-hydroxy-3-methoxyphenyl)benzothiazole 9c. Compound 9c was prepared using the above-mentioned procedure from 5 (1.60 mg, 10 mmol) and 8c (1.67 mg, 11 mmol) to obtain 9c as beige powder (1.14 g, 39%; m.p. 219–222 °C). 1H NMR (300 MHz, DMSO) δ 9.90 (1H, s, OH), 8.25 (1H, d, J = 2.1 Hz), 7.99 (1H, d, J = 8.7 Hz), 7.61 (1H, d, J = 1.9 Hz), 7.59–7.47 (2H, m), 6.95 (1H, d, J = 8.2 Hz), 3.90 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 168.52, 152.40, 150.27, 148.09, 135.71, 129.26, 126.85, 123.91, 123.37, 121.76, 121.39, 115.91, 110.07, 55.68 (OCH3).
6-Fluoro-2-(4-hydroxyphenyl)benzothiazole 10a. Compound 10a was prepared using the above-mentioned procedure from 6 (1.43 g, 10 mmol) and 8a (1.34 g, 11 mmol) to obtain 10a as white powder (1.84 g, 75%; m.p. 203–205 °C). 1H NMR (300 MHz, DMSO) δ 10.24 (1H, s, OH), 8.05–7.95 (2H, m), 7.91 (2H, d, J = 8.7 Hz), 7.36 (1H, td, J = 9.1, 2.7 Hz), 6.93 (2H, d, J = 8.7 Hz). 13C NMR (75 MHz, DMSO) δ 168.05 (d, JCF = 3.2 Hz), 161.00, 159.95 (d, JCF = 242.3 Hz), 151.02, 135.80 (d, JCF = 11.6 Hz), 129.45, 124.31, 123.88 (d, JCF = 9.4 Hz), 116.57, 115.21 (d, JCF = 24.6 Hz), 109.05 (d, JCF = 27.2 Hz).
6-Fluoro-2-(3-fluoro-4-hydroxyphenyl)benzothiazole 10b. Compound 10b was prepared using the above-mentioned procedure from 6 (1.43 g, 10 mmol) and 8b (1.54 g, 11 mmol) to obtain 10b as beige powder (1.02 g, 39%; m.p. 182–185 °C). 1H NMR (300 MHz, DMSO) δ 10.74 (1H, s, OH), 8.03 (2H, dd, J = 9.0, 6.4, 3.8 Hz), 7.84 (1H, dd, J = 11.9, 2.1 Hz), 7.73 (1H, dd, J = 8.4, 1.5 Hz), 7.39 (1H, td, J = 9.1, 2.7 Hz), 7.13 (1H, t, J = 8.7 Hz). 13C NMR (75 MHz, DMSO) δ 160.11 (d, JCF = 242.8 Hz), 151.56 (d, JCF = 242.8 Hz), 150.83 (d, JCF = 1.4 Hz), 148.64 (d, JCF = 12.2 Hz), 135.93, 124.81 (d, JCF = 2.9 Hz), 124.68, 124.16 (d, JCF = 9.5 Hz), 118.83 (d, JCF = 3.3 Hz), 115.44 (d, JCF = 24.8 Hz), 115.07 (d, JCF = 20.2 Hz), 109.14 (d, JCF = 27.4 Hz).
6-Fluoro-2-(4-hydroxy-3-methoxyphenyl)benzothiazole 10c. Compound 10c was prepared using the above-mentioned procedure from 6 (1.43 g, 10 mmol) and 8c (1.67 g, 11 mmol) to obtain 10c as beige powder (0.93 g, 33%; m.p. 206–208 °C). 1H NMR (600 MHz, DMSO) δ 9.86 (1H, s, OH), 8.02 (2H, dd, J = 8.7, 3.5 Hz,), 7.61 (1H, d, J = 1.8 Hz), 7.49 (1H, dd, J = 8.2, 1.9 Hz), 7.37 (1H, td, J = 9.1, 2.6 Hz), 6.95 (1H, d, J = 8.2 Hz), 3.90 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 167.64 (d, JCF = 3.1 Hz), 159.48 (d, JCF = 242.3 Hz), 150.46, 150.05, 148.09, 135.38 (d, JCF = 11.8 Hz), 124.09, 123.41 (d, JCF = 9.5 Hz), 121.22, 115.89, 114.74 (d, JCF = 24.7 Hz), 109.95, 108.53 (d, JCF = 27.3 Hz), 55.67 (OCH3).
2-(4-Hydroxyphenyl)benzothiazole 11a. Compound 11a was prepared using the above-mentioned procedure from 7 (1.25 g, 10 mmol) and 8a (1.34 g, 11 mmol) to obtain 11a as beige powder (1.79 g, 78%; m.p. 224–227 °C). 1H NMR (300 MHz, DMSO) δ 10.25 (1H, s, OH), 8.08 (1H, d, J = 8.0 Hz), 8.00 (1H, d, J = 8.1 Hz), 7.96 (2H, d, J = 8.7 Hz), 7.51 (1H, t, J = 7.7 Hz), 7.41 (1H, t, J = 7.6 Hz), 6.97 (2H, d, J = 8.7 Hz). 13C NMR (75 MHz, DMSO) δ 167.92, 160.99, 154.20, 134.58, 129.51, 126.86, 125.34, 124.52, 122.76, 122.54, 116.55.
2-(3-Fluoro-4-hydroxyphenyl)benzothiazole 11b. Compound 11b was prepared using the above-mentioned procedure from 7 (1.25 g, 10 mmol) and 8b (1.54 g, 11 mmol) to obtain 11b as yellow powder (1.92 g, 78%; m.p. 199–201 °C). 1H NMR (300 MHz, DMSO) δ 10.72 (1H, s, OH), 8.11 (1H, d, J = 7.5 Hz), 8.02 (1H, d, J = 7.8 Hz), 7.87 (1H, dd, J = 12.0, 2.1 Hz), 7.76 (1H, dd, J = 8.4, 1.4 Hz), 7.53 (1H, t, J = 8.2 Hz), 7.44 (1H, t, J = 8.1 Hz), 7.14 (1H, t, J = 8.7 Hz). 13C NMR (75 MHz, DMSO) δ 166.68, 153.98, 151.57 (d, JCF = 242.8 Hz), 148.61 (d, JCF = 12.1 Hz), 134.76, 127.03, 125.67, 125.00, 124.84 (d, JCF = 2.9 Hz), 122.98, 122.67, 118.81 (d, JCF = 3.3 Hz), 115.12 (d, JCF = 20.1 Hz).
2-(4-Hydroxy-3-methoxyphenyl)benzothiazole 11c. Compound 11c was prepared using the above-mentioned procedure from 7 (1.25 g, 10 mmol) and 8c (1.67 g, 11 mmol) to obtain 11c as a beige powder (2.19 g, 85%; m.p. 185–187 °C). 1H NMR (300 MHz, DMSO) δ 9.86 (1H, s, OH), 8.08 (1H, d, J = 7.4 Hz), 8.01 (1H, d, J = 7.7 Hz), 7.64 (1H, d, J = 2.0 Hz), 7.59–7.47 (2H, m), 7.41 (1H, t, J = 8.1 Hz), 6.96 (1H, d, J = 8.2 Hz), 3.91 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 167.50, 153.61, 150.01, 148.06, 134.14, 126.40, 124.89, 124.30, 122.27, 122.05, 121.25, 115.87, 110.05, 55.67 (OCH3).

3.2.2. General Procedure for O-Alkylation of Propargylated Benzothiazole Derivatives 12a12c, 13a13c, 14a14c and Target Analogs 15a15c, 16a16c, 17a17c, 18a18c, 19a19c, 20a20c

To a solution of the corresponding heterocyclic base (9a9c, 10a10c, 11a11c; 1 eq) in acetonitrile, K2CO3 (3 eq) was added and stirred for 30 min. Corresponding alkyl halogenide (1.2 eq) was added and the reaction mixture was stirred for 12 h at room temperature. The solvent was evaporated and the residue was purified by column chromatography (CH2Cl2:CH3OH = 50:1).
6-Chloro-2-(4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 12a. Using the above-mentioned procedure from 9a (500 mg, 1.91 mmol) and propargyl bromide (174 µL, 2.29 mmol), compound 12a was obtained as beige powder (471.8 mg, 82%; m.p. 158–161 °C). 1H NMR (300 MHz, DMSO) δ 8.29 (1H, d, J = 2.1 Hz), 8.05 (2H, d, J = 8.9 Hz), 8.01 (1H, d, J = 8.7 Hz), 7.55 (1H, dd, J = 8.7, 2.2 Hz), 7.18 (2H, d, J = 8.9 Hz), 4.93 (2H, d, J = 2.3 Hz, OCH2), 3.65 (1H, t, J = 2.3 Hz, CH). 13C NMR (151 MHz, DMSO) δ 167.91, 159.83, 152.40, 135.83, 129.53, 128.89, 126.98, 125.80, 123.62, 121.90, 115.66, 78.74(OCH2CCH), 78.70(OCH2CCH), 55.71 (OCH2CCH).
6-Chloro-2-(3-fluoro-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 12b. Using the above-mentioned procedure from 9b (500 mg, 1.79 mmol) and propargyl bromide (163 µL, 2.15 mmol), compound 12b was obtained as beige powder (500.5 mg, 88%; m.p. 142–145 °C). 1H NMR (300 MHz, DMSO) δ 8.32 (1H, d, J = 2.2 Hz), 8.03 (1H, d, J = 8.7 Hz), 7.99–7.87 (3H, m), 7.57 (1H, dd, J = 8.7, 2.2 Hz), 7.43 (1H, t, J = 8.5 Hz), 5.03 (2H, d, J = 2.3 Hz, OCH2), 3.72 (1H, t, J = 2.3 Hz, CH). 13C NMR (151 MHz, DMSO) δ 166.73, 152.19, 151.76 (d, JCF = 246.2 Hz), 147.68 (d, JCF = 10.7 Hz), 136.02, 129.87, 127.15, 126.27 (d, JCF = 7.0 Hz), 124.24 (d, JCF = 3.0 Hz), 123.84, 122.01, 115.93, 114.54 (d, JCF = 20.2 Hz), 79.34 (OCH2CCH), 78.24 (OCH2CCH), 56.67 (OCH2CCH).
6-Chloro-2-(3-methoxy-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 12c. Using the above-mentioned procedure from 9c (500 mg, 1.71 mmol) and propargyl bromide (156 µL, 2.05 mmol), compound 12c was obtained as beige powder (497.4 mg, 88%; m.p. 153–156 °C). 1H NMR (300 MHz, DMSO) δ 8.26 (1H, d, J = 2.1 Hz), 8.01 (1H, d, J = 8.7 Hz), 7.68–7.57 (2H, m), 7.53 (1H, dd, J = 8.7, 2.2 Hz), 7.18 (1H, d, J = 8.3 Hz), 4.90 (2H, d, J = 2.3 Hz, OCH2), 3.88 (3H, s, OCH3), 3.62 (1H, t, J = 2.3 Hz, CH). 13C NMR (75 MHz, DMSO) δ 168.58, 152.82, 149.92, 136.39, 130.07, 127.49, 126.55, 124.14, 122.37, 121.18, 114.30, 110.21, 79.25(OCH2CCH), 56.55 (OCH2CCH), 56.16 (OCH3).
6-Fluoro-2-(4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 13a. Using the above-mentioned procedure from 10a (500 mg, 2.04 mmol) and propargyl bromide (186 µL, 2.45 mmol), compound 13a was obtained as beige powder (338.5 mg, 58%; m.p. 132–135 °C). 1H NMR (600 MHz, DMSO) δ 8.06–8.00 (4H, m), 7.39 (1H, td, J = 9.1, 2.6 Hz), 7.20–7.16 (2H, m), 4.93 (2H, d, J = 2.3 Hz, OCH2), 3.65 (1H, t, J = 2.3 Hz, CH). 13C NMR (151 MHz, DMSO) δ 167.00 (d, JCF = 2.5 Hz), 159.63, 159.60 (d, JCF = 242.7 Hz), 150.46, 135.51 (d, JCF = 11.9 Hz), 128.72, 125.96, 123.67 (d, JCF = 9.5 Hz), 115.61, 114.89 (d, JCF = 24.9 Hz), 108.63 (d, JCF = 27.1 Hz), 78.76 (OCH2CCH), 78.69 (OCH2CCH), 55.68 (OCH2CCH).
6-Fluoro-2-(3-fluoro-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 13b. Using the above-mentioned procedure from 10b (500 mg, 1.89 mmol) and propargyl bromide (172 µL, 2.27 mmol), compound 13b was obtained as beige powder (314.7 mg, 55%; m.p. 125–128 °C). 1H NMR (400 MHz, DMSO) δ 8.10–8.02 (2H, m), 7.93 (1H, dd, J = 11.9, 2.2 Hz), 7.91–7.86 (1H, m), 7.45–7.38 (2H, m), 5.03 (2H, d, J = 2.4 Hz, OCH2), 3.72 (1H, t, J = 2.4 Hz, CH). 13C NMR (101 MHz, DMSO) δ 166.35, 160.26 (d, JCF = 243.1 Hz), 152.27 (d, JCF = 246.1 Hz), 150.78, 147.99 (d, JCF = 10.6 Hz), 136.25 (d, JCF = 11.9 Hz), 126.93 (d, JCF = 6.9 Hz), 124.57 (d, JCF = 2.6 Hz), 124.45 (d, JCF = 9.6 Hz), 116.42, 115.64 (d, JCF = 24.8 Hz), 114.92 (d, JCF = 20.2 Hz), 109.24 (d, JCF = 27.4 Hz), 79.84 (OCH2CCH), 78.77 (OCH2CCH), 57.15 (OCH2CCH).
6-Fluoro-2-(3-methoxy-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 13c. Using the above-mentioned procedure from 10c (500 mg, 1.82 mmol) and propargyl bromide (166 µL, 2.18 mmol), compound 13c was obtained as a beige powder (223.0 mg, 39%; m.p. 145–148 °C). 1H NMR (300 MHz, DMSO) δ 8.05 (2H, dt, J = 8.5, 3.6 Hz), 7.65 (1H, d, J = 2.1 Hz), 7.60 (1H, dd, J = 8.3, 2.1 Hz), 7.39 (1H, td, J = 9.1, 2.7 Hz), 7.20 (1H, d, J = 8.4 Hz), 4.92 (2H, d, J = 2.4 Hz, OCH2), 3.91 (3H, s, OCH3), 3.64 (1H, t, J = 2.3 Hz, CH). 13C NMR (75 MHz, DMSO) δ 167.67 (d, JCF = 3.2 Hz), 160.13 (d, JCF = 242.7 Hz), 150.89, 149.93, 149.72, 136.08 (d, JCF = 11.8 Hz), 126.73, 124.20 (d, JCF = 9.5 Hz), 121.01, 115.41 (d, JCF = 24.8 Hz), 114.30, 110.09, 109.11 (d, JCF = 27.3 Hz), 79.30 (OCH2CCH), 79.23 (OCH2CCH), 56.55 (OCH2CCH), 56.15 (OCH3).
2-(4-(Prop-2-yn-1-yloxy)phenyl)benzothiazole 14a. Using the above-mentioned procedure from 11a (500 mg, 2.19 mmol) and propargyl bromide (200 µL, 2.63 mmol), compound 14a was obtained as white powder (517.1 mg, 88%; m.p. 136–140 °C). 1H NMR (600 MHz, DMSO) δ 8.12 (1H, d, J = 7.8 Hz), 8.06 (2H, d, J = 8.8 Hz), 8.02 (1H, d, J = 8.1 Hz), 7.53 (1H, t, J = 8.2 Hz), 7.44 (1H, t, J = 8.1 Hz), 7.18 (2H, d, J = 8.9 Hz), 4.93 (2H, d, J = 2.3 Hz, OCH2), 3.64 (1H, t, J = 2.3 Hz, CH). 13C NMR (151 MHz, DMSO) δ 166.87, 159.63, 153.62, 134.26, 128.78, 126.52, 126.17, 125.15, 122.51, 122.20, 115.61, 78.78 (OCH2CCH), 78.67 (OCH2CCH), 55.69 (OCH2CCH).
2-(3-Fluoro-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 14b. Using the above-mentioned procedure from 11b (500 mg, 2.04 mmol) and propargyl bromide (186 µL, 2.45 mmol), compound 14b was obtained as beige powder (439.3 mg, 76%; m.p. 134–138 °C). 1H NMR (300 MHz, DMSO) δ 8.17–8.12 (1H, m), 8.07–8.02 (1H, m), 7.99–7.88 (2H, m), 7.58–7.51 (1H, m), 7.50–7.38 (2H, m), 5.03 (2H, d, J = 2.4 Hz, OCH2), 3.71 (1H, t, J = 2.4 Hz, CH). 13C NMR (75 MHz, DMSO) δ 153.91, 152.26 (d, JCF = 245.9 Hz), 147.96 (d, JCF = 10.6 Hz), 134.96, 127.17, 127.09, 125.97, 124.60 (d, JCF = 3.3 Hz), 123.21, 122.82, 116.42 (d, JCF = 1.6 Hz), 114.97 (d, JCF = 20.2 Hz), 79.82 (OCH2CCH), 78.77 (OCH2CCH), 57.14 (OCH2CCH).
2-(3-Methoxy-4-(prop-2-yn-1-yloxy)phenyl)benzothiazole 14c. Using the above-mentioned procedure from 11c (500 mg, 1.94 mmol) and propargyl bromide (177 µL, 2.33 mmol), compound 14c was obtained as beige powder (397.1 mg, 69%; m.p. 122–125 °C). 1H NMR (600 MHz, DMSO) δ 8.11 (1H, d, J = 7.8 Hz), 8.04 (1H, d, J = 8.1 Hz), 7.68 (1H, d, J = 2.1 Hz), 7.63 (1H, dd, J = 8.3, 2.1 Hz), 7.53 (1H, t, J = 8.2 Hz), 7.44 (1H, t, J = 8.1 Hz), 7.21 (1H, d, J = 8.4 Hz), 4.91 (2H, d, J = 2.4 Hz, OCH2), 3.91 (3H, s, OCH3), 3.63 (1H, t, J = 2.3 Hz, CH). 13C NMR (151 MHz, DMSO) δ 167.03, 153.54, 149.45, 149.22, 134.33, 126.52, 126.47, 125.18, 122.52, 122.17, 120.53, 113.86, 109.74, 78.82 (OCH2CCH), 78.70 (OCH2CCH), 56.07 (OCH2CCH), 55.67 (OCH3).
6-Chloro-2-(4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 15a. Using the above-mentioned procedure from 9a (80.0 mg, 0.31 mmol) and 2-bromoacetophenone (74.0 mg, 0.37 mmol), compound 15a was obtained as a grey powder (53.2 mg, 45%; m.p. 192–195 °C). 1H NMR (300 MHz. DMSO) δ 8.28 (1H, d, J = 2.0 Hz), 8.10–7.96 (5H, m), 7.72 (1H, t, J = 7.4 Hz), 7.64–7.51 (3H, m), 7.17 (2H, d, J = 8.9 Hz), 5.75 (2H, s, OCH2). 13C NMR (151 MHz. DMSO) δ 194.03 (C=O), 167.98, 160.71, 152.43, 135.81, 134.24, 133.87, 129.47, 128.86, 128.84, 127.87, 126.96, 125.47, 123.58, 121.88, 115.50, 70.29 (OCH2). Anal.calcd. for C21H14ClNO2S (Mr = 379.86): C 66.40, H 3.72, N 3.69; found: C 66.17, H 3.71, N 3.67.
6-Chloro-2-(3-fluoro-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 15b. Using the above-mentioned procedure from 9b (80.0 mg, 0.29 mmol) and 2-bromoacetophenone (69.3 mg, 0.35 mmol), compound 15b was obtained as a white powder (43.4 mg, 38%; m.p. 192–195 °C). 1H NMR (300 MHz. DMSO) δ 8.31 (1H, d, J = 2.1 Hz), 8.08–8.00 (3H, m), 7.96 (1H, dd, J = 12.0, 2.1 Hz), 7.82 (1H, d, J = 8.6 Hz), 7.73 (1H, t, J = 7.4 Hz), 7.65–7.53 (3H, m), 7.32 (1H, t, J = 8.7 Hz), 5.87 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 193.60 (C=O), 166.83, 151.50 (d, JCF = 245.8 Hz), 152.22, 148.73, 148.67, 136.00, 134.08, 133.96, 129.80, 128.85, 127.87, 127.12, 125.79 (d, JCF = 6.8 Hz), 124.15 (d, JCF = 2.9 Hz), 123.79, 121.98, 115.63, 114.55 (d, JCF = 20.2 Hz), 70.85 (OCH2). Anal.calcd. for C21H13ClFNO2S (Mr = 397.85): C 63.40, H 3.29, N 3.52; found: C 63.18, H 3.29, N 3.49.
6-Chloro-2-(3-methoxy-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 15c. Using the above mentioned procedure from 9c (60.0 mg,  0.21 mmol) and 2-bromoacetophenone (50.2 mg, 0.25 mmol) compound 15c was obtained as grey powder (65.2 mg, 77%; m.p. 163–166 °C. 1H NMR (300 MHz, DMSO) δ 8.27 (1H, d, J = 2.0 Hz), 8.04 (3H, t, J = 7.6 Hz), 7.77–7.64 (2H, m), 7.64–7.52 (4H, m), 7.07 (1H, d, J = 8.5 Hz), 5.74 (2H, s, OCH2), 3.94 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 194.52 (C=O), 168.64, 152.85, 150.91, 149.65, 136.36, 134.74, 134.36, 130.00, 129.33, 128.38, 127.46, 126.11, 124.09, 122.35, 121.23, 113.91, 110.38, 71.07 (OCH2), 56.23(OCH3). Anal.calcd. for C22H16ClNO3S (Mr = 409.88): C 64.47, H 3.93, N 3.42; found: C 64.24, H 3.93, N 3.40.
6-Fluoro-2-(4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 16a. Using the above-mentioned procedure from 10a (90.0 mg, 0.37 mmol) and 2-bromoacetophenone (88.4 mg, 0.44 mmol), compound 16a was obtained as a yellow powder (68.7 mg, 51%; m.p. 164–167 °C). 1H NMR (600 MHz, DMSO) δ 8.08–7.97 (6H, m), 7.72 (1H, t, J = 7.4 Hz), 7.60 (2H, t, J = 7.8 Hz), 7.39 (1H, td, J = 9.1, 2.7 Hz), 7.16 (2H, d, J = 8.8 Hz), 5.74 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 194.06 (C=O), 167.08 (d, JCF = 3.0 Hz), 160.52, 159.58 (d, JCF = 242.7 Hz), 150.49, 135.49 (d, JCF = 11.8 Hz), 134.24, 133.87, 128.83, 128.70, 127.87, 125.64, 123.64 (d, JCF = 9.5 Hz), 115.46, 114.87 (d, JCF = 24.7 Hz), 108.61 (d, J CF= 27.4 Hz), 70.27 (OCH2). Anal.calcd. for C21H14FNO2S (Mr = 363.41): C 69.41, H 3.88, N 3.85; found: C 69.14, H 3.88, N 3.83.
6-Fluoro-2-(3-fluoro-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 16b. Using the above-mentioned procedure from 10b (90.0 mg, 0.34 mmol) and 2-bromoacetophenone (81.2 mg, 0.41 mmol), compound 16b was obtained as an orange powder (57.0 mg, 60%; m.p. 148–152 °C). 1H NMR (600 MHz, DMSO) δ 8.11–8.00 (H4, m), 7.94 (1H, dd, J = 12.0, 1.9 Hz), 7.80 (1H, d, J = 8.5 Hz), 7.72 (1H, t, J = 7.4 Hz), 7.60 (2H, t, J = 7.7 Hz), 7.41 (1H, td, J = 9.0, 2.6 Hz), 7.31 (1H, t, J = 8.6 Hz), 5.86 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 194.13 (C=O), 166.41, 160.22 (d, JCF = 242.9 Hz), 152.00 (d, JCF = 245.7 Hz), 150.79, 149.01 (d, JCF = 10.4 Hz), 136.21 (d, JCF = 11.8 Hz), 134.57, 134.47, 129.35, 128.37, 126.44 (d, JCF = 6.8 Hz), 124.43 (t, JCF = 6.4 Hz), 124.40 (d, JCF = 9.6 Hz), 116.11, 115.60 (d, JCF = 24.9 Hz), 114.93 (d, JCF = 20.3 Hz), 109.22 (d, JCF = 27.4 Hz), 71.32 (OCH2). Anal.calcd. for C21H13F2NO2S (Mr = 381.40): C 66.13, H 3.44, N 3.67; found: C 65.90, H 3.43, N 3.66.
6-Fluoro-2-(3-methoxy-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 16c. Using the above-mentioned procedure from 10c (90.0 mg, 0.33 mmol) and 2-bromoacetophenone (78.8 mg, 0.40 mmol), compound 16c was obtained as a beige powder (53.5 mg, 41%; m.p. 178–181 °C). 1H NMR (600 MHz, DMSO) δ 8.08–8.00 (4H, m), 7.71 (1H, t, J = 7.4 Hz), 7.66 (1H, d, J = 2.0 Hz), 7.59 (2H, t, J = 7.8 Hz), 7.53 (1H, dd, J = 8.4, 2.0 Hz), 7.39 (1H, td, J = 9.0, 2.6 Hz), 7.06 (1H, d, J = 8.5 Hz), 5.72 (2H, s, OCH2), 3.93 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 194.07 (C=O), 167.25, 159.60 (d, JCF = 242.7 Hz), 150.42, 150.24, 149.18, 135.56 (d, JCF = 11.7 Hz), 134.27, 133.84, 128.82, 127.87, 125.83, 123.67 (d, JCF = 9.4 Hz), 120.58, 114.89 (d, JCF = 24.8 Hz), 113.48, 109.86, 108.60 (d, JCF = 27.3 Hz), 70.60 (OCH2), 55.76 (OCH3). Anal.calcd. for C22H16FNO3S (Mr = 393.43): C 67.16, H 4.10, N 3.56; found: C 66.91, H 4.09, N 3.54.
2-(4-(2-Oxo-2-phenylethoxy)phenyl)benzothiazole 17a. Using the above-mentioned procedure from 11a (120 mg, 0.53 mmol) and 2-bromoacetophenone (126.6 mg, 0.64 mmol), compound 17a was obtained as a beige powder (64.3 mg, 35%; m.p. 150–155 °C). 1H NMR (300 MHz, DMSO) δ 8.13–8.07 (1H, m), 8.07–7.97 (5H, m), 7.75–7.67 (1H, m), 7.62–7.54 (2H, m), 7.54–7.47 (1H, m), 7.45–7.37 (1H, m), 7.15 (2H, d, J = 8.9 Hz), 5.73 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 194.58 (C=O), 167.45, 161.00, 154.13, 134.73, 134.38, 129.34, 129.26, 128.37, 127.00, 126.33, 125.61, 122.96, 122.68, 115.94, 70.77 (OCH2). Anal.calcd. for C21H15NO2S (Mr = 345.42): C 73.02, H 4.38, N 4.06; found: C 72.72, H 4.37, N 4.03.
2-(3-Fluoro-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 17b. Using the above-mentioned procedure from 11b (120 mg, 0.49 mmol) and 2-bromoacetophenone (117.0 mg, 0.59 mmol), compound 17b was obtained as a beige powder (103.5 mg, 58%; m.p. 174–177 °C). 1H NMR (600 MHz, DMSO) δ 8.13 (1H, d, J = 7.8 Hz), 8.06–8.02 (3H, m), 7.96 (1H, dd, J = 12.0, 2.1 Hz), 7.83–7.80 (1H, m), 7.73 (1H, t, J = 7.4 Hz), 7.60 (2H, t, J = 7.8 Hz), 7.56–7.52 (1H, m), 7.48–7.43 (1H, m), 7.32 (1H, t, J = 8.7 Hz), 5.86 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 193.64 (C=O), 165.78, 153.44, 151.51 (d, JCF = 245.6 Hz), 148.49 (d, JCF = 10.3 Hz), 134.44, 134.10, 133.96, 128.85, 127.87, 126.64, 126.17 (d, JCF = 6.8 Hz), 125.40, 124.01 (d, JCF = 2.5 Hz), 122.67, 122.28, 115.60, 114.47 (d, J = 20.2 Hz), 70.84 (OCH2). Anal.calcd. for C21H14FNO2S (Mr = 363.41): C 69.41, H 3.88, N 3.85; found: C 69.12, H 3.87, N 3.83.
2-(3-Methoxy-4-(2-oxo-2-phenylethoxy)phenyl)benzothiazole 17c. Using the above-mentioned procedure from 11c (120 mg, 0.47 mmol) and 2-bromoacetophenone (112.3 mg, 0.56 mmol), compound 17c was obtained as a beige powder (99.2 mg, 56%; m.p. 184–186 °C). 1H NMR (400 MHz, DMSO) δ 8.11 (1H, d, J = 7.5 Hz), 8.07–8.02 (3H, m), 7.75–7.68 (2H, m), 7.63–7.50 (4H, m), 7.44 (1H, t, J = 7.6 Hz), 7.07 (1H, d, J = 8.5 Hz), 5.73 (2H, s, OCH2), 3.95 (3H, OCH3). 13C NMR (101 MHz, DMSO) δ 194.58 (C=O), 167.61, 154.07, 150.71, 149.65, 134.80, 134.77, 134.37, 129.34, 128.39, 127.02, 126.51, 125.65, 122.99, 122.68, 121.11, 113.93, 110.39, 71.09 (OCH2), 56.25 (OCH3). Anal.calcd. for C22H17NO3S (Mr = 375.44): C 70.38, H 4.56, N 3.73; found: C 70.13, H 4.55, N 3.71.
6-Chloro-2-(4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 18a. Using the above-mentioned procedure from 9a (80.0 mg, 0.31 mmol) 2-(bromomethyl)pyridine hydrobromide (94.1 mg, 0.37 mmol), compound 18a was obtained as a beige powder (72.0 mg, 66%; m.p. 199–204 °C). 1H NMR (300 MHz, DMSO) δ 8.60 (1H, d, J = 4.1 Hz), 8.27 (1H, d, J = 2.1 Hz), 8.04 (2H, d, J = 8.9 Hz), 8.00 (1H, d, J = 8.7 Hz), 7.86 (1H, td, J = 7.7, 1.7 Hz), 7.54 (2H, dd, J = 8.7, 2.2 Hz), 7.37 (1H, dd, J = 7.5, 4.9 Hz), 7.22 (2H, d, J = 8.9 Hz), 5.30 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 167.96, 160.83, 156.10, 152.42, 149.15, 137.09, 135.81, 129.49, 129.01, 126.97, 125.53, 123.60, 123.11, 121.90, 121.79, 115.64, 70.48 (OCH2). Anal.calcd. for C19H13ClNO2S (Mr = 352.84): C 64.68, H 3.71, N 7.94; found: C 64.44, H 3.71, N 7.90.
6-Chloro-2-(3-fluoro-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 18b. Using the above-mentioned procedure from 9b (80.0 mg, 0.29 mmol) 2-(bromomethyl)pyridine hydrobromide (88.0 mg, 0.35 mmol), compound 18b was obtained as a white powder (43.0 mg, 40%; m.p. 211–215 °C). 1H NMR (600 MHz, DMSO) δ 8.57 (1H, ddd, J = 4.9, 1.6, 1.0 Hz), 8.22 (1H, d, J = 2.1 Hz), 7.98 (1H, d, J = 8.7 Hz), 7.89 (1H, dd, J = 12.0, 2.2 Hz), 7.86–7.79 (2H, m), 7.55–7.50 (2H, m), 7.41 (1H, t, J = 8.6 Hz), 7.34 (1H, ddd, J = 7.5, 4.8, 0.8 Hz), 5.35 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 166.77, 155.71, 151.95 (d, JCF = 246.4 Hz), 152.30, 149.19, 148.90 (d, JCF = 10.5 Hz), 137.01, 136.08, 129.92, 127.09, 126.18 (d, JCF = 6.9 Hz), 124.34 (d, JCF = 3.2 Hz), 123.80, 123.17, 121.86, 121.83, 116.18, 114.57 (d, JCF = 20.3 Hz), 71.67 (OCH2). Anal.calcd. for C19H12ClFN2OS (Mr = 370.83): C 61.54, H 3.26, N 7.55; found: C 61.31, H 3.25, N 7.52.
6-Chloro-2-(3-methoxy-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 18c. Using the above-mentioned procedure from 9c (60.0 mg, 0.21 mmol) 2-(bromomethyl)pyridine hydrobromide (63.7 mg, 0.25 mmol), compound 18c was obtained as a grey powder (64.0 mg, 81%; m.p. 173–176 °C). 1H NMR (600 MHz, DMSO) δ 8.18 (ddd, J = 4.8, 1.5, 0.8 Hz, 1H), 7.85 (d, J = 2.2 Hz, 1H), 7.60 (d, J = 8.7 Hz, 1H), 7.45 (td, J = 7.7, 1.8 Hz, 1H), 7.26 (d, J = 2.1 Hz, 1H), 7.18 (dd, J = 8.4, 2.1 Hz, 1H), 7.13 (dd, J = 8.7, 2.2 Hz, 2H), 6.95 (ddd, J = 7.4, 4.8, 0.8 Hz, 1H), 6.80 (d, J = 8.5 Hz, 1H), 4.86 (2H, s, OCH2), 3.51 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 168.13, 156.18, 152.33, 150.54, 149.38, 149.14, 137.06, 135.86, 129.54, 126.98, 125.69, 123.60, 123.10, 121.84, 121.80, 120.94, 113.52, 109.74, 70.91 (OCH2), 55.75 (OCH3). Anal.calcd. for C20H15ClN2O2S (Mr = 382.86): C 62.74, H 3.95, N 7.32; found: C 62.51, H 3.94, N 7.28.
6-Fluoro-2-(4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 19a. Using the above-mentioned procedure from 10a (70.0 mg, 0.29 mmol) 2-(bromomethyl)pyridine hydrobromide (88.0 mg, 0.35 mmol), compound 19a was obtained as a beige powder (55.9 mg, 58%; m.p. 165–170 °C). 1H NMR (300 MHz, DMSO) δ 8.60 (1H, d, J = 4.8 Hz), 8.08–7.97 (4H, m), 7.86 (1H, td, J = 7.7, 1.7 Hz), 7.55 (1H, d, J = 7.8 Hz), 7.43–7.33 (2H, m), 7.22 (2H, d, J = 8.9 Hz), 5.30 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 161.13, 160.08 (d, JCF = 242.5 Hz), 156.64, 150.98, 149.67, 137.55, 135.98 (d, JCF = 11.9 Hz), 129.34, 126.17, 124.15 (d, JCF = 9.5 Hz), 123.59, 122.26, 116.09, 115.39 (d, JCF = 24.9 Hz), 109.14 (d, JCF = 27.3 Hz), 70.97 (OCH2). Anal.calcd. for C19H13FN2OS (Mr = 336.38): C 67.84, H 3.90, N 8.33; found: C 67.57, H 3.90, N 8.29.
6-Fluoro-2-(3-fluoro-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 19b. Using the above-mentioned procedure from 10b (70.0 mg, 0.27 mmol) 2-(bromomethyl)pyridine hydrobromide (81.9 mg, 0.32 mmol), compound 19b was obtained as a yellow powder (38.3 mg, 40%; m.p. 190–195 °C). 1H NMR (300 MHz, DMSO) δ 8.61 (1H, d, J = 4.0 Hz), 8.12–8.01 (2H, m), 7.98–7.81 (3H, m), 7.57 (1H, d, J = 7.8 Hz), 7.49–7.35 (3H, m), 5.38 (2H, s OCH2). 13C NMR (151 MHz, DMSO) δ 165.91, 159.73 (d, JCF = 243.1 Hz), 155.62, 151.71 (d, JCF = 245.9 Hz), 150.29, 149.25, 148.60 (d, JCF = 10.7 Hz), 137.16, 135.71 (d, JCF = 11.7 Hz), 126.03 (d, JCF = 6.7 Hz), 124.26 (d, JCF = 3.0 Hz), 123.91 (d, JCF = 9.6 Hz), 123.27, 121.87, 115.74, 115.12 (d, JCF = 24.9 Hz), 114.37 (d, JCF = 20.0 Hz), 108.74 (d, JCF = 27.4 Hz), 71.23 (OCH2). Anal.calcd. for C19H12F2N2OS (Mr = 354.37): C 64.40, H 3.41, N 7.91; found: C 64.16, H 3.41, N 7.87.
6-Fluoro-2-(3-methoxy-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 19c. Using the above-mentioned procedure from 10c (70.0 mg, 0.25 mmol) 2-(bromomethyl)pyridine hydrobromide (75.9 mg, 0.30 mmol), compound 19c was obtained as grey powder (62.4 mg, 66%; m.p. 152–156 °C). 1H NMR (300 MHz, DMSO) δ 8.60 (1H, d, J = 4.1 Hz), 8.12–7.97 (2H, m), 7.87 (1H, td, J = 7.7, 1.6 Hz), 7.67 (1H, d, J = 1.8 Hz), 7.56 (2H, dd, J = 11.0, 4.7 Hz), 7.46–7.30 (2H, m), 7.21 (1H, d, J = 8.5 Hz), 5.28 (2H, s, OCH2), 3.93 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 167.73 (d, JCF = 3.1 Hz), 160.11 (d, JCF = 242.6 Hz), 156.72, 150.89, 150.84, 149.86, 149.64, 137.55, 136.05 (d, JCF = 12.0 Hz), 126.35, 124.17 (d, JCF = 9.5 Hz), 123.59, 122.27, 121.27, 115.40 (d, JCF = 24.7 Hz), 113.97, 110.08, 109.11 (d, JCF = 27.3 Hz), 71.38 (OCH2), 56.22 (OCH3). Anal.calcd. for C20H15FN2O2S (Mr = 366.41): C 65.56, H 4.13, N 7.65; found: C 65.31, H 4.12, N 7.61.
2-(4-(Pyridin-2-ylmethoxy)phenyl)benzothiazole 20a. Using the above-mentioned procedure from 11a (120 mg, 0.53 mmol) 2-(bromomethyl)pyridine hydrobromide (160.9 mg, 0.64 mmol), compound 20a was obtained as a beige powder (57.8 mg, 34%; m.p. 132–136 °C). 1H NMR (600 MHz, DMSO) δ 8.61 (1H, dd, J = 4.7, 1.5, 0.7 Hz), 8.11 (1H, d, J = 7.8 Hz), 8.05 (2H, d, J = 8.8 Hz), 8.01 (1H, d, J = 8.1 Hz), 7.86 (1H, td, J = 7.7, 1.7 Hz), 7.55 (1H, d, J = 7.8 Hz), 7.54–7.51 (1H, m), 7.45–7.40 (1H, m), 7.39–7.35 (1H, m), 7.22 (2H, d, J = 8.8 Hz), 5.30 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 166.91, 160.63, 156.17, 153.63, 149.17, 137.04, 134.24, 128.89, 126.50, 125.89, 125.12, 123.08, 122.48, 122.19, 121.76, 115.57, 70.49 (OCH2). Anal.calcd. for C19H14N2OS (Mr = 318.39): C 71.68, H 4.43, N 8.80; found: C 71.34, H 4.43, N 8.75.
2-(3-Fluoro-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 20b. Using the above-mentioned procedure from 11b (120 mg, 0.49 mmol) 2-(bromomethyl)pyridine hydrobromide (148.7 mg, 0.59 mmol), compound 20b was obtained as a beige powder (36.9 mg, 22%; m.p. 136–142 °C). 1H NMR (300 MHz, DMSO) δ 8.61 (1H, d, J = 4.1 Hz), 8.14 (1H, d, J = 7.9 Hz), 8.04 (1H, d, J = 7.8 Hz), 7.96 (1H, dd, J = 12.0, 2.0 Hz), 7.92–7.82 (2H, m), 7.60–7.50 (2H, m), 7.42 (3H, dd, J = 13.0, 7.3, 4.4 Hz), 5.38 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 156.14, 153.83, 152.21 (d, JCF = 245.8 Hz), 149.75, 149.07 (d, JCF = 10.5 Hz), 137.66, 134.93, 130.52, 127.16, 126.74 (d, JCF = 6.6 Hz), 125.93, 124.78 (d, JCF = 3.1 Hz), 123.77, 123.18, 122.81, 122.36, 116.22, 114.92 (d, JCF = 20.1 Hz), 71.72 (OCH2). Anal.calcd. for C19H13FN2OS (Mr = 336.38): C 67.84, H 3.90, N 8.33; found: C 67.55, H 3.89, N 8.28.
2-(3-Methoxy-4-(pyridin-2-yl)methoxy)phenyl)benzothiazole 20c. Using the above-mentioned procedure from 11c (120 mg, 0.47 mmol) 2-(bromomethyl)pyridine hydrobromide (142.7 mg, 0.56 mmol), compound 20c was obtained as a beige powder (99.4 mg, 61%; m.p. 110–115 °C). 1H NMR (400 MHz, DMSO) δ 8.61 (1H, dd, J = 4.8, 1.7, 0.9 Hz), 8.11 (1H, d, J = 7.3 Hz), 8.04 (1H, d, J = 7.7 Hz), 7.87 (1H, td, J = 7.7, 1.8 Hz), 7.70 (1H, d, J = 2.1 Hz), 7.60 (1H, dd, J = 8.4, 2.1 Hz), 7.58–7.50 (2H, m), 7.46–7.41 (1H, m), 7.38 (1H, dd, J = 7.5, 4.8, 1.1 Hz), 7.21 (1H, d, J = 8.5 Hz), 5.28 (2H, s, OCH2), 3.94 (3H, s, OCH3). 13C NMR (101 MHz, DMSO) δ 167.60, 156.76, 154.06, 150.82, 149.86, 149.65, 137.56, 134.81, 127.02, 126.57, 125.66, 123.59, 123.00, 122.68, 122.28, 121.30, 113.97, 110.18, 71.39 (OCH2), 56.23 (OCH3). Anal.calcd. for C20H16N2O2S (Mr = 348.42): C 68.95, H 4.63, N 8.04; found: C 68.68, H 4.62, N 8.00.

3.2.3. General Procedure for Preparation of Target 1H-1,2,3-Triazole-substituted Benzothiazole Analogs 21a21c, 22a22c and 23a23c

The reaction mixture of compounds 12a12c, 13a13c, 14a14c, CuI (0.1 eq) and the trimethylsilyl azide (1.5 eq) was dissolved in a mixture of DMF:MeOH  =  1:1 (2 mL). The reaction mixture was stirred at 100 °C for 12 h. The solvent was removed under reduced pressure and purified by column chromatography with CH2Cl2.
6-Chloro-2-(4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 21a. Compound 21a was prepared using the above-mentioned procedure from 12a (200 mg. 0.67 mmol) and trimethylsilyl azide (132 µL, 1.00 mmol) to obtain 21a as a beige powder (88.1 mg, 38%; m.p. 220–223 °C). 1H NMR (300 MHz, DMSO) δ 15.13 (1H, s, NH), 8.25 (1H, d, J = 2.1 Hz), 8.05–7.94 (4H, m), 7.53 (1H, dd, J = 8.7, 2.2 Hz), 7.21 (2H, d, J = 8.8 Hz), 5.29 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 167.96, 160.67, 152.41, 135.80, 129.48, 128.94, 126.96, 125.46, 123.58, 121.88, 115.55, 61.10 (OCH2). Anal.calcd. for C16H11ClN4OS (Mr = 342.80): C 56.06, H 3.23, N 16.34; found: C 55.84, H 3.23, N 16.26.
6-Chloro-2-(3-fluoro-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 21b. Compound 21b was prepared using the above-mentioned procedure from 12b (200 mg, 0.63 mmol) and trimethylsilyl azide (124 µL, 0.95 mmol) to obtain 21b as a yellow powder (35.7 mg, 15%; m.p. 205–209 °C). 1H NMR (400 MHz, DMSO) δ 15.10 (1H, s, NH), 8.31 (1H, d, J = 2.1 Hz), 8.03 (1H, d, J = 8.7 Hz), 7.93 (1H, dd, J = 11.9, 2.1 Hz), 7.91–7.86 (1H, m), 7.57 (2H, dd, J = 8.7, 2.2 Hz), 5.40 (2H, s, OCH2). 13C NMR (101 MHz, DMSO) δ 167.33, 152.71, 152.20 (d, JCF = 246.0 Hz), 149.08 (d, JCF = 10.2 Hz), 136.50, 130.33, 127.65, 126.35 (d, JCF = 6.7 Hz), 124.85 (d, JCF = 2.5 Hz), 124.31, 122.51, 116.28, 114.96 (d, JCF = 20.2 Hz), 62.54 (OCH2). Anal.calcd. for C16H10ClFN4OS (Mr = 360.79): C 53.27, H 2.79, N 15.53; found: C 53.08, H 2.79, N 15.46.
6-Chloro-2-(3-methoxy-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 21c. Compound 21c was prepared using the above-mentioned procedure from 12c (200 mg, 0.61 mmol) and trimethylsilyl azide (121 µL, 0.92 mmol) to obtain 21c as a beige powder (49.4 mg, 21%; m.p. 183–188 °C). 1H NMR (300 MHz, DMSO) δ 15.14 (1H, s, NH), 8.28 (1H, d, J = 2.1 Hz), 8.02 (2H, d, J = 8.7 Hz), 7.66–7.58 (2H, m), 7.55 (1H, dd, J = 8.7, 2.2 Hz), 7.33 (1H, d, J = 8.3 Hz), 5.29 (2H, s, OCH2), 3.88 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 168.63, 152.83, 150.88, 149.84, 136.36, 130.03, 127.47, 126.15, 124.10, 122.35, 121.35, 114.01, 110.14, 62.02 (OCH2), 56.10 (OCH3). Anal.calcd. for C17H13ClN4O2S (Mr = 372.83): C 54.77, H 3.51, N 15.03; found: C 54.58, H 3.51, N 14.96.
6-Fluoro-2-(4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 22a. Compound 22a was prepared using the above-mentioned procedure from 13a (200 mg, 0.71 mmol) and trimethylsilyl azide (140 µL, 1.07 mmol) to obtain 22a as a white powder (45.7 mg, 19%; m.p. 212–215 °C). 1H NMR (600 MHz, DMSO) δ 15.12 (1H, s, NH), 8.08–7.98 (4H, ), 7.39 (1H, td, J = 9.0, 2.7 Hz), 7.23 (2H, d, J = 8.8 Hz), 5.31 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 167.62 (d, JCF = 2.8 Hz), 160.96, 160.08 (d, JCF = 242.6 Hz), 150.93, 135.96 (d, JCF = 11.9 Hz), 129.29, 126.11, 124.14 (d, JCF = 9.5 Hz), 116.04, 115.40 (d, JCF = 24.8 Hz), 109.10 (d, JCF = 27.3 Hz), 61.59 (OCH2). Anal.calcd. for C16H11FN4OS (Mr = 326.35): C 58.89, H 3.40, N 17.17; found: C 58.65, H 3.40, N 17.08.
6-Fluoro-2-(3-fluoro-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 22b. Compound 22b was prepared using the above-mentioned procedure from 13b (200 mg, 0.66 mmol) and trimethylsilyl azide (130 µL, 0.99 mmol) to obtain 22b as a brown powder (48.3 mg, 21%; m.p. 177–180 °C). 1H NMR (300 MHz, DMSO) δ 15.19 (1H, s, NH), 8.11–8.00 (2H, m), 7.96–7.84 (2H, m), 7.55 (1H, t, J = 8.6 Hz), 7.41 (1H, td, J = 9.1, 2.7 Hz), 5.40 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 166.42, 160.23 (d, JCF = 243.1 Hz), 150.77, 152.20 (d, JCF = 245.8 Hz), 148.82, 136.20 (d, JCF = 12.1 Hz), 126.43, 124.67 (d, JCF = 3.2 Hz), 124.41 (d, JCF = 9.6 Hz), 116.29, 115.61 (d, JCF = 24.8 Hz), 114.83 (d, JCF = 20.3 Hz), 109.22 (d, JCF = 27.5 Hz), 62.54 (OCH2). Anal.calcd. for C16H10F2N4OS (Mr = 344.34): C 55.81, H 2.93, N 16.27; found: C 55.59, H 2.92, N 16.19.
6-Fluoro-2-(3-methoxy-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 22c. Compound 22c was prepared using the above-mentioned procedure from 13c (200 mg, 0.64 mmol) and trimethylsilyl azide (126 µL, 0.96 mmol) to obtain 22c as a beige powder (50.6 mg, 22%; m.p. 196–199 °C). 1H NMR (300 MHz, DMSO) δ 15.10 (1H, s, NH), 8.04 (3H, dd, J = 9.0, 5.2 Hz), 7.63 (1H, s), 7.59 (1H, dd, J = 8.4, 1.8 Hz), 7.39 (1H, td, J = 9.1, 2.6 Hz), 7.32 (1H, d, J = 8.4 Hz), 5.28 (2H, s, OCH2), 3.87 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 167.23 (d, JCF = 3.1 Hz), 159.61 (d, JCF = 242.7 Hz), 150.41, 150.20, 149.36, 135.55 (d, JCF = 11.8 Hz), 128.79, 125.85, 123.66 (d, JCF = 9.3 Hz), 120.68, 114.88 (d, JCF = 24.9 Hz), 113.54, 109.55, 108.59 (d, JCF = 27.0 Hz), 61.51 (OCH2), 55.60 (OCH3). Anal.calcd. for C17H13FN4O2S (Mr = 356.37): C 57.30, H 3.68, N 15.72; found: C 57.09, H 3.67, N 15.65.
2-(4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 23a. Compound 23a was prepared using the above-mentioned procedure from 14a (200 mg, 0.75 mmol) and trimethylsilyl azide (149 µL, 1.13 mmol) to obtain 23a as a beige powder (53.6 mg, 23%; m.p. 204–207 °C). 1H NMR (400 MHz, DMSO) δ 15.04 (1H, s, NH), 8.12 (1H, d, J = 7.7 Hz), 8.05 (2H, d, J = 8.8 Hz), 8.02 (1H, d, J = 8.4 Hz), 7.53 (1H, t, J = 8.2 Hz), 7.44 (1H, t, J = 8.1 Hz), 7.24 (2H, d, J = 8.8 Hz), 5.31 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 167.43, 160.97, 154.13, 134.73, 129.34, 127.00, 126.34, 125.62, 122.97, 122.68, 116.01, 61.65 (OCH2). Anal.calcd. for C16H12N4OS (Mr = 308.36): C 62.32, H 3.92, N 18.17; found: C 62.06, H 3.91, N 18.07
2-(3-Fluoro-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 23b. Compound 23b was prepared using the above-mentioned procedure from 14b (200 mg, 0.71 mmol) and trimethylsilyl azide (139 µL, 1.06 mmol) to obtain 23b as a grey powder (37.3 mg, 16%; m.p. 185–189 °C). 1H NMR (300 MHz, DMSO) δ 15.16 (1H, s, NH), 8.14 (1H, d, J = 7.3 Hz), 8.07 (1H, s), 8.04 (1H, d, J = 7.7 Hz), 7.98–7.85 (2H, m, J = 7.7, 7.1, 1.6 Hz), 7.60–7.51 (2H, m, J = 9.6, 6.1, 2.0 Hz), 7.46 (1H, t, J = 7.0 Hz), 5.40 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 166.27 (d, JCF = 2.7 Hz), 162.82, 153.90, 152.20 (d, JCF = 245.8 Hz), 148.83 (d, JCF = 10.6 Hz), 134.90, 127.16, 126.72 (d, JCF = 7.0 Hz), 125.93, 124.69 (d, JCF = 3.3 Hz), 123.16, 122.77, 116.26 (d, JCF = 1.6 Hz), 114.87 (d, JCF = 20.2 Hz), 62.38 (OCH2). Anal.calcd. for C16H11FN4OS (Mr = 326.35): C 58.89, H 3.40, N 17.17; found: C 58.63, H 3.39, N 17.07.
2-(3-Methoxy-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 23c. Compound 23c was prepared using the above-mentioned procedure from 14c (200 mg, 0.68 mmol) and trimethylsilyl azide (134 µL, 1.02 mmol) to obtain 23c as a beige powder (32.2 mg, 13%; m.p. 177–180 °C). 1H NMR (400 MHz, DMSO) δ 15.10 (1H, s, NH), 8.12 (1H, d, J = 7.7 Hz), 8.04 (1H, d, J = 8.0 Hz), 7.67 (1H, d, J = 2.0 Hz), 7.62 (1H, dd, J = 8.3, 2.0 Hz), 7.53 (1H, t, J = 7.1 Hz), 7.44 (1H, t, J = 7.1 Hz), 7.33 (1H, d, J = 8.4 Hz), 5.29 (2H, s, OCH2), 3.89 (3H, s, OCH3). 13C NMR (101 MHz, DMSO) δ 167.61, 154.06, 150.68, 149.85, 134.80, 127.03, 126.56, 125.67, 123.00, 122.69, 121.22, 114.03, 110.13, 62.00 (OCH2), 56.10 (OCH3). Anal.calcd. for C17H14N4O2S (Mr = 338.38): C 60.34, H 4.17, N 16.56; found: C 60.09, H 4.16, N 16.47.

3.2.4. General Procedure for Preparation of Target 1-Benzyl-1,2,3-triazole-substituted Benzothiazole Analogs 24a24c, 25a25c and 26a26c

Stir a solution of benzyl chloride (1.2 eq), NaN3 (1.5 eq) and triethylamine (1.5 eq) in a mixture of t-BuOH:H2O = 1:1 (2 mL) at room temperature for 2 h. To reaction mixture, add corresponding propargylated compounds 12a12c, 13a13c, 14a14c (1 eq) and Cu(OAc)2 (0.2 eq). The reaction mixture was stirred at room temperature for 12 h. The solvent was removed under reduced pressure and purified by column chromatography with CH2Cl2.
6-Chloro-2-(4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 24a. Using the above-mentioned procedure from 12a (100 mg, 0.33 mmol) and benzyl chloride (46 µL, 0.39 mmol), compound 24a was obtained as an orange powder (82.4 mg, 57%; m.p. 201–205 °C). 1H NMR (600 MHz, DMSO) δ 8.36 (1H, s), 8.31 (1H, d, J = 2.1 Hz), 8.06 (2H, d, J = 8.9 Hz), 8.03 (1H, d, J = 8.7 Hz), 7.58 (1H, dd, J = 8.6, 2.2 Hz), 7.44–7.39 (2H, m), 7.37 (3H, dd, J = 10.1, 4.5 Hz), 7.26 (2H, d, J = 8.9 Hz), 5.65 (2H, s, NCH2), 5.29 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 167.98, 160.69, 152.42, 142.51, 135.95, 135.80, 129.48, 128.93, 128.75, 128.15, 127.94, 126.97, 125.43, 124.85, 123.59, 121.89, 115.55, 61.34 (OCH2), 52.83 (NCH2). Anal.calcd. for C23H17ClN4OS (Mr = 432.93): C 63.81, H 3.96, N 12.94; found: C 63.60, H 3.95, N 12.89.
6-Chloro-2-(3-fluoro-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 24b. Using the above-mentioned procedure from 12b (100 mg, 0.31 mmol) and benzyl chloride (43 µL, 0.37 mmol), compound 24b was obtained as a beige powder (53.4 mg, 38%; m.p. 181–186 °C). 1H NMR (300 MHz, DMSO) δ 8.36 (1H, s), 8.31 (1H, d, J = 2.1 Hz), 8.02 (1H, d, J = 8.7 Hz), 7.90 (2H, t, J = 10.1 Hz), 7.61–7.50 (2H, m), 7.43–7.27 (5H, m), 5.63 (2H, s, NCH2), 5.35 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 167.33, 152.70, 152.18 (d, JCF = 245.9 Hz), 149.07 (d, JCF = 10.6 Hz), 142.54, 136.45 (d, JCF = 5.6 Hz), 134.63, 130.31, 129.25, 128.65, 128.44, 127.64, 126.32 (d, JCF = 7.0 Hz), 125.65, 124.83 (d, JCF = 3.1 Hz), 124.30, 122.50, 116.31 (d, JCF = 1.5 Hz), 114.94 (d, JCF = 20.2 Hz), 62.6 (OCH2), 53.34 (NCH2). Anal.calcd. for C23H16ClFN4OS (Mr = 450.92): C 61.26, H 3.58, N 12.43; found: C 61.07, H 3.57, N 12.37.
6-Chloro-2-(3-methoxy-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 24c. Using the above-mentioned procedure from 12c (100 mg, 0.30 mmol) and benzyl chloride (41 µL, 0.36 mmol), compound 24c was obtained as a beige powder (51.7 mg, 37%; m.p. 190–193 °C). 1H NMR (300 MHz, DMSO) δ 8.32 (1H, s), 8.27 (1H, d, J = 2.1 Hz), 8.01 (1H, d, J = 8.7 Hz), 7.66–7.57 (2H, m), 7.55 (1H, dd, J = 8.7, 2.2 Hz), 7.42–7.28 (6H, m), 5.62 (2H, s, NCH2), 5.23 (2H, s, OCH2), 3.85 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 168.64, 152.84, 150.88, 149.82, 142.96, 136.44, 136.36, 130.02, 129.24, 128.65, 128.47, 127.47, 126.12, 125.50, 124.10, 122.36, 121.34, 114.02, 110.06, 62.18(OCH2), 56.05(OCH3), 53.32 (NCH2). Anal.calcd. for C24H19ClN4O2S (Mr = 462.95): C 62.27, H 4.14, N 12.10; found: C 62.07, H 4.13, N 12.05.
6-Fluoro-2-(4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 25a. Using the above-mentioned procedure from 13a (100 mg, 0.35 mmol) and benzyl chloride (48 µL, 0.42 mmol), compound 25a was obtained as a white powder (18.3 mg, 12%; m.p. 176–180 °C). 1H NMR (600 MHz, DMSO) δ 8.33 (1H, s), 8.06–7.99 (4H, m), 7.42–7.36 (3H, m), 7.36–7.30 (3H, m), 7.22 (2H, d, J = 8.8 Hz), 5.63 (2H, s, NCH2), 5.26 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 167.09 (d, JCF = 3.1 Hz), 160.51, 159.58 (d, JCF = 242.6 Hz), 150.48 (d, JCF = 1.0 Hz), 142.53, 135.94, 135.48 (d, JCF = 11.9 Hz), 128.78, 128.75, 128.15, 127.94, 125.60, 124.84, 123.65 (d, JCF = 9.5 Hz), 115.53, 114.89 (d, JCF = 24.7 Hz), 108.64 (d, JCF = 27.3 Hz), 61.32 (OCH2), 52.83 (NCH2). Anal.calcd. for C23H17FN4OS (Mr = 416.47): C 66.33, H 4.11, N 13.45; found: C 66.11, H 4.10, N 13.39.
6-Fluoro-2-(3-fluoro-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 25b. Using the above-mentioned procedure from 13b (100 mg, 0.33 mmol) and benzyl chloride (46 µL, 0.39 mmol), compound 25b was obtained as a white powder (69.5 mg, 48%; m.p. 205–208 °C). 1H NMR (300 MHz, DMSO) δ 8.34 (1H, s), 8.08–7.99 (2H, m), 7.91–7.80 (2H, m), 7.54 (1H, t, J = 8.5 Hz), 7.43–7.28 (6H, m), 5.61 (2H, s, NCH2), 5.33 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 160.23 (d, JCF = 243.0 Hz), 150.79 (d, JCF = 1.3 Hz), 152.20 (d, JCF = 245.8 Hz), 148.89 (d, JCF = 10.6 Hz), 142.57, 136.41, 136.21 (d, JCF = 12.0 Hz), 129.24, 128.65, 128.43, 126.55, 125.64, 124.66 (d, JCF = 3.1 Hz), 124.41 (d, JCF = 9.7 Hz), 116.33 (d, JCF = 1.7 Hz), 115.61 (d, JCF = 24.9 Hz), 114.82 (d, JCF = 20.2 Hz), 109.23 (d, JCF = 27.5 Hz), 62.68 (OCH2), 53.35 (NCH2). Anal.calcd. for C23H16F2N4OS (Mr = 434.46): C 63.58, H 3.71, N 12.90; found: C 63.39, H 3.71, N 12.84.
6-Fluoro-2-(3-methoxy-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 25c. Using the above-mentioned procedure from 13c (100 mg, 0.33 mmol) and benzyl chloride (44 µL, 0.38 mmol), compound 25c was obtained as white powder (50.4 mg, 35%; m.p. 180–183 °C). 1H NMR (600 MHz, DMSO) δ 8.29 (1H, s), 8.04–7.99 (2H, m), 7.60 (1H, d, J = 2.0 Hz), 7.56 (1H, dd, J = 8.4, 2.1 Hz), 7.36 (3H, m), 7.33–7.28 (4H, m), 5.60 (2H, s, NCH2), 5.21 (2H, s, OCH2), 3.83 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 167.75 (d, JCF = 3.3 Hz), 160.11 (d, JCF = 242.7 Hz), 150.90 (d, JCF = 1.1 Hz), 150.69, 149.83, 142.98, 136.44, 136.04 (d, JCF = 12.1 Hz), 129.24, 128.65, 128.47, 126.30, 125.49, 124.16 (d, JCF = 9.6 Hz), 121.18, 115.40 (d, JCF = 24.7 Hz), 114.04, 109.96, 109.12 (d, JCF = 27.3 Hz), 62.17 (OCH2), 56.04 (OCH3), 53.31 (NCH2). Anal.calcd. for C24H19FN4O2S (Mr = 446.50): C 64.56, H 4.29, N 12.55; found: C 64.37, H 4.28, N 12.50.
2-(4-((1-Benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 26a. Using the above-mentioned procedure from 14a (100 mg, 0.38 mmol) and benzyl chloride (53 µL, 0.46 mmol), compound 26a was obtained as a white powder (50.2 mg, 33%; m.p. 210–214 °C). 1H NMR (300 MHz, DMSO) δ 8.33 (1H, s), 8.11 (1H, d, J = 7.9 Hz), 8.03 (3H, t, J = 7.8 Hz), 7.57–7.48 (1H, m), 7.46–7.40 (1H, m), 7.40–7.29 (5H, m), 7.22 (2H, d, J = 8.9 Hz), 5.63 (2H, s, NCH2), 5.26 (2H, s, OCH2). 13C NMR (151 MHz, DMSO) δ 166.94, 160.49, 153.63, 142.55, 135.94, 134.22, 128.83, 128.75, 128.15, 127.94, 126.51, 125.79, 125.12, 124.84 (Tr), 122.47, 122.19, 115.50, 61.32 (OCH2), 52.83 (NCH2). Anal.calcd. for C23H18N4OS (Mr = 398.48): C 69.33, H 4.55, N 14.06; found: C 69.09, H 4.56, N 14.00.
2-(3-Fluoro-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 26b. Using the above-mentioned procedure from 14b (100 mg, 0.35 mmol) and benzyl chloride (48 µL, 0.42 mmol), compound 26b was obtained as white powder (40.5 mg, 27%; m.p. 198–201 °C). 1H NMR (300 MHz, DMSO) δ 8.36 (1H, s), 8.14 (1H, d, J = 7.8 Hz), 8.04 (1H, d, J = 8.0 Hz), 7.96–7.84 (2H, m), 7.61–7.50 (2H, m), 7.45 (1H, t, J = 7.3 Hz), 7.42–7.27 (5H, m), 5.64 (2H, s, NCH2), 5.35 (2H, s, OCH2). 13C NMR (75 MHz, DMSO) δ 165.74, 153.43, 151.70 (d, JCF = 245.8 Hz), 148.35 (d, JCF = 10.5 Hz), 142.08, 135.91, 134.43, 128.73, 127.92, 126.64, 126.22 (d, JCF = 7.0 Hz), 125.41, 125.12, 124.16, 123.92, 122.67, 122.28, 115.81, 114.36 (d, JCF = 19.6 Hz), 62.18 (OCH2), 52.85 (OCH3). Anal.calcd. for C23H17FN4OS (Mr = 416.47): C 66.33, H 4.11, N 13.45; found: C 66.12, H 4.10, N 13.40.
2-(3-Methoxy-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)benzothiazole 26c. Using the above-mentioned procedure from 14c (100 mg, 0.34 mmol) and benzyl chloride (47 µL, 0.41 mmol), compound 26c was obtained as white powder (43.7 mg, 33%; m.p. 170–173 °C). 1H NMR (300 MHz, DMSO) δ 8.32 (1H, s), 8.12 (1H, d, J = 7.6 Hz), 8.03 (1H, d, J = 7.9 Hz), 7.66 (1H, d, J = 2.0 Hz), 7.61 (1H, dd, J = 8.3, 2.0 Hz), 7.53 (1H, t, J = 7.0 Hz), 7.47–7.41 (1H, m), 7.41–7.29 (5H, m), 5.63 (2H, s, NCH2), 5.24 (2H, s, OCH2), 3.87 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 167.11, 153.56, 150.17, 149.33, 142.51, 135.95, 134.29, 128.74, 128.15, 127.97, 126.51, 126.03, 125.15, 124.98, 122.48, 122.17, 120.70, 113.57, 109.58, 61.69 (OCH2), 55.56 (OCH3), 52.82 (NCH2). Anal.calcd. for C24H20N4O2S (Mr = 428.51): C 67.27, H 4.70, N 13.08; found: C 67.07, H 4.70, N 13.02.

3.2.5. General Procedure for the Synthesis of Target 6-Amidino-substituted Benzothiazole Analogs 34a34c, 35a, 35c, 36a36c, and 37a37c

To a stirred solution of amidino-substituted 2-aminobenzenethiolate 32 or 33 (1 eq) in glacial acetic acid (3 mL), a corresponding benzaldehyde (1 eq) was added. The reaction mixture was stirred and heated under nitrogen for 3 h, then poured onto ice and made alkaline (pH 10–11) with 20% NaOH. Resulting free base was filtered, washed with water and dried. The free base was suspended in ethanol/HCl(g) (10 mL), and stirred at room temperature for 24 h. The addition of ether resulted in precipitation of products. Solid was collected by filtration, washed with anhydrous ether, and dried under vacuum.
2-(4-(2-Oxo-2-phenylethoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 34a. Compound 34a was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 28a (67.3 mg, 0.28 mmol) to obtain 34a as beige powder (17.5 mg, 12%; m.p. > 250 °C). 1H NMR (300 MHz, DMSO) δ 10.87 (2H, s, CNH), 8.86 (1H, d, J = 10.5 Hz), 8.23 (1H, dd, J = 13.2, 8.6 Hz), 8.15–7.98 (4H, m), 7.73 (1H, t, J = 7.4 Hz), 7.60 (2H, t, J = 7.5 Hz), 7.20 (2H, d, J = 8.9 Hz), 6.99 (1H, d, J = 8.7 Hz), 5.78 (2H, s, OCH2), 4.04 (4H, s, NCH2).13C NMR (75 MHz, DMSO) δ 194.47 (C=O), 172.26 (CNH), 165.07, 162.07, 161.84, 157.57, 135.24, 134.43, 129.90, 129.36, 128.38, 127.09, 125.58, 124.09, 123.37, 118.94, 116.14, 70.83 (OCH2), 44.92 (NCH2). Anal.calcd. for C24H19N3O2S × HCl × 1.75H2O (Mr = 481.48): C 59.87. H 4.92. N 8.73; found: C 59.98. H 4.83. N 8.86.
2-(3-Fluoro-4-(2-oxo-2-phenylethoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 34b. Compound 34b was prepared using the above-mentioned procedure from 32(60.0 mg, 0.28 mmol) and 28b (72.3 mg, 0.28 mmol) to obtain 34b as brown powder (19.8 mg, 14%; m.p. >240 °C). 1H NMR (400 MHz, DMSO) δ 11.30 (2H, s, CNH), 9.12 (1H, s), 8.32 (1H, d, J = 9.9 Hz), 8.23 (1H, d, J = 8.6 Hz), 8.09–8.00 (3H, m), 7.95–7.87 (1H, m), 7.73 (1H, t, J = 7.4 Hz), 7.61 (2H, t, J = 7.7 Hz), 7.37 (1H, t, J = 8.7 Hz), 5.91 (2H, OCH2), 4.03 (4H, s, NCH2). 13C NMR (151 MHz, DMSO) δ 193.56 (C=O), 170.49 (CNH), 164.11, 156.73, 151.48 (d, JCF = 245.9 Hz), 149.30 (d, JCF = 10.7 Hz), 134.80, 134.04, 133.99, 128.86, 127.90, 126.95, 125.38 (d, JCF = 6.4 Hz), 124.74 (d, JCF = 2.3 Hz), 124.04, 122.93, 118.77, 115.74, 114.93 (d, JCF = 20.2 Hz), 70.92 (OCH2), 44.31 (NCH2). Anal.calcd. for C24H18FN3O2S × HCl × 1.5H2O (Mr = 494.97): C 58.24. H 4.48. N 8.49; found: C 58.32. H 4.56. N 8.37.
2-(3-Methoxy-4-(2-oxo-2-phenylethoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 34c. Compound 34c was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 28c (75.7 mg, 0.28 mmol) to obtain 34c as beige powder (40.5 mg, 26%; m.p. >240 °C). 1H NMR (400 MHz, DMSO) δ 10.74 (2H, s, CNH), 8.81 (1H, s), 8.28 (1H, d, J = 8.6 Hz,), 8.11–8.03 (3H, m), 7.76–7.70 (2H, m), 7.66 (1H, dd, J = 8.4, 2.1 Hz), 7.60 (2H, t, J = 7.7 Hz), 7.11 (1H, d, J = 8.6 Hz), 5.78 (2H, s, OCH2), 4.06 (4H, s, NCH2), 3.96 (3H, s, OCH3). 13C NMR (101 MHz, DMSO) δ 194.44 (C=O), 172.42 (CNH), 170.96, 165.16, 157.53, 151.59, 149.71, 135.34, 134.71, 134.41, 129.35, 128.39, 127.04, 125.69, 123.99, 123.41, 121.91, 118.96, 113.96, 110.68, 71.08 (OCH2), 56.32 (OCH3), 44.96 (NCH2). Anal.calcd. for C25H21N3O3S × HCl × 3.5H2O (Mr = 538.53): C 55.76. H 5.33. N 7.80; found: C 55.83. H 5.26. N 7.69.
2-(4-(Pyridin-2-ylmethoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 35a. Compound 35a was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 29a (59.7 mg, 0.28 mmol) to obtain 35a as yellow powder (19.2 mg, 15%; m.p. > 250 °C). 1H NMR (600 MHz, DMSO) δ 10.99 (2H, s, NCH), 8.94 (1H, d, J = 1.6 Hz), 8.84 (1H, d, J = 4.8 Hz), 8.35 (1H, t, J = 7.6 Hz), 8.24 (1H, d, J = 8.6 Hz), 8.17 (3H, dd, J = 13.6, 5.2 Hz), 7.96 (1H, d, J = 7.8 Hz), 7.83–7.78 (1H, m), 7.31 (2H, d, J = 8.8 Hz), 5.55 (2H, s, OCH2), 4.04 (4H, s, NCH2). 13C NMR (151 MHz, DMSO) δ 171.56 (NCH), 164.44, 160.82, 157.00, 152.92, 145.14, 142.49, 134.73, 129.58, 126.68, 125.63, 125.09, 124.12, 123.72, 122.89, 118.53, 115.82, 67.63 (OCH2), 44.39 (NCH2). Anal.calcd. for C22H18N4OS × HCl × 1.25H2O (Mr = 445.45): C 59.32. H 4.86. N 12.58; found: C 58.58. H 4.72. N 12.73.
2-(3-Methoxy-4-(pyridin-2-ylmethoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 35c. Compound 35c was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 29c (68.1 mg, 0.28 mmol) to obtain 35c as yellow powder (24.1 mg, 19%; m.p. 219–223 °C). 1H NMR (300 MHz, DMSO) δ 10.93 (2H, s, CNH), 8.91 (1H, d, J = 1.6 Hz), 8.74 (1H, d, J = 4.4 Hz), 8.27 (1H, d, J = 8.6 Hz), 8.15 (2H, d, J = 8.8 Hz), 7.82–7.68 (3H, m), 7.68–7.57 (1H, m), 7.29 (1H, d, J = 8.3 Hz), 5.43 (2H, s, OCH2), 4.05 (4H, s, NCH2), 3.95 (3H, s, OCH3). 13C NMR (75 MHz, DMSO) δ 172.27 (CNH), 165.00, 157.45, 154.68, 151.29, 149.96, 147.20, 140.96, 135.31, 127.14, 126.19, 124.87, 124.15, 123.80, 123.40, 122.07, 119.05, 114.34, 110.60, 69.75 (OCH2), 56.33 (OCH3), 44.91 (NCH2). Anal.calcd. for C23H20N4O2S × HCl × H2O (Mr = 470.97): C 58.65, H 4.92, N 11.90; found: C 58.43, H 4.80, N 12.09.
2-(4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 36a. Compound 36a was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 30a (56.9 mg, 0.28 mmol) to obtain 36a as an orange powder (36.5 mg, 29%; m.p. 218–221 °C). 1H NMR (600 MHz, DMSO) δ 11.00 (2H, s, CNH), 8.94 (1H, d, J = 1.7 Hz), 8.23 (1H, d, J = 8.6 Hz), 8.18 (1H, dd, J = 8.7, 1.8 Hz), 8.12 (2H, d, J = 8.9 Hz), 8.04 (1H, s), 7.30–7.25 (2H, d, J = 8.9 Hz), 5.33 (s, 3H), 4.04 (s, 4H). 13C NMR (151 MHz, DMSO) δ 171.70 (CNH), 164.43, 161.27, 157.04, 134.69, 129.47, 126.67, 125.09, 123.69, 122.81, 118.45, 115.70, 61.15 (OCH2), 44.38 (NCH2). Anal.calcd. for C19H16N6OS × HCl × 1.5H2O (Mr = 439.92): C 51.87. H 4.58. N 19.10; found: C 51.68. H 4.65. N 18.98.
2-(3-Fluoro-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 36b. Compound 36b was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 30b (61.9 mg, 0.28 mmol) to obtain 36b as a brown powder (22.5 mg, 17%; m.p. 234–237 °C). 1H NMR (600 MHz, DMSO) δ 10.98 (1H, s, CNH), 10.96 (1H, s, CNH), 8.94 (1H, d, J = 5.7 Hz), 8.25 (1H, d, J = 8.6 Hz), 8.19–8.13 (1H, m), 8.08 (1H, s), 8.00 (1H, dd, J = 11.7, 2.1 Hz), 7.98 (1H, dd, J = 8.6, 1.9 Hz), 7.60 (1H, t, J = 8.6 Hz), 5.42 (2H, s, OCH2), 4.04 (4H, s, NCH2). 13C NMR (101 MHz, DMSO) δ 171.05 (CNH), 164.94, 157.28, 152.18 (d, JCF = 246.2 Hz), 149.69 (d, JCF = 10.7 Hz), 135.40, 127.24, 125.91 (d, JCF = 6.7 Hz), 125.45 (d, JCF = 2.5 Hz), 124.31, 123.57, 119.26, 116.32, 115.35 (d, JCF = 20.2 Hz), 62.46 (OCH2), 44.93 (NCH2). Anal.calcd. for C19H15FN6OS × HCl × 1.5H2O (Mr = 457.91): C 49.84. H 4.18. N 18.35; found: C 49.95. H 4.10. N 18.23.
2-(3-Methoxy-4-((1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 36c. Compound 36c was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 30c (65.3 mg, 0.28 mmol) to obtain 36c as beige powder (12.7 mg, 9%; m.p. >240 °C). 1H NMR (600 MHz, DMSO) δ 10.66 (2H, s, CNH), 8.77 (1H, s), 8.28 (1H, d, J = 8.6 Hz), 8.05 (1H, dd, J = 8.6, 1.4 Hz), 7.91 (1H, s), 7.73 (1H, dd, J = 8.4, 2.1 Hz), 7.70 (1H, d, J = 1.6 Hz), 7.38 (1H, t, J = 13.0 Hz), 5.31 (2H, s, OCH2), 4.05 (4H, s, NCH2), 3.89 (3H, s, OCH3). 13C NMR (151 MHz, DMSO) δ 171.37 (CNH), 164.19, 156.44, 148.80, 135.28, 134.29, 125.90, 124.35, 123.75, 122.84, 122.37, 120.97, 117.90, 112.93, 109.31, 61.00 (OCH2), 55.09 (OCH3), 43.91 (NCH2). Anal.calcd. for C19H18N6O2S × HCl × H2O (Mr = 460.94): C 52.11. H 4.59. N 18.23; found: C 51.99. H 4.67. N 18.13.
2-(4-((1-Benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 37a. Compound 37a was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 31a (82.1 mg, 0.28 mmol) to obtain 37a as a yellow powder (32.5 mg, 21%; m.p. 153–156 °C). 1H NMR (600 MHz, DMSO) δ 10.89 (2H, s, NCH), 8.89 (1H, d, J = 1.6 Hz), 8.35 (1H, s), 8.24 (1H, d, J = 8.6 Hz), 8.15–8.10 (3H, m), 7.41–7.36 (2H, m), 7.34 (3H, dd, J = 7.1, 5.0 Hz), 7.26 (2H, d, J = 8.9 Hz), 5.63 (2H, s, NCH2), 5.28 (2H, s, OCH2), 4.04 (4H, s, NCH2). 13C NMR (151 MHz, DMSO) δ 171.74 (CNH), 164.55, 161.30, 157.07, 142.42, 135.94, 134.72, 129.46, 128.75, 128.15, 127.95, 126.60, 125.06, 124.90, 123.61, 122.86, 118.45, 115.69, 61.40 (OCH2), 52.84 (NCH2), 44.42 (NCH2). Anal.calcd. for C26H22N6OS × HCl × 2.25H2O (Mr = 543.55): C 57.45. H 5.10. N 15.46; found: C 57.32. H 5.19. N 15.32.
2-(3-Fluoro-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 37b. Compound 37b was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 31b (87.2 mg, 0.28 mmol) to obtain 37b as a yellow powder (44.9 mg, 28%; m.p. >240 °C). 1H NMR (400 MHz, DMSO) δ 10.97 (2H, s, CNH), 8.93 (1H, d, J = 1.6 Hz), 8.39 (1H, s), 8.25 (1H, d, J = 8.6 Hz), 8.16 (1H, dd, J = 8.7, 1.8 Hz), 8.01–7.94 (2H, m), 7.60 (1H, t, J = 8.8 Hz), 7.43–7.27 (6H, m), 5.64 (2H, s, NCH2), 5.37 (2H, s, OCH2), 4.04 (4H, s, NCH2). 13C NMR (75 MHz, DMSO) δ 171.07 (CNH), 165.04, 157.29, 152.24 (d, JCF = 245.1 Hz), 149.70 (d, JCF = 10.7 Hz), 142.46, 136.40, 135.42, 129.25, 128.66, 128.45, 127.17, 125.70, 125.44, 124.23, 123.59, 119.25, 116.37 (d, J = 1.3 Hz), 115.34 (d, JCF = 20.4 Hz), 62.71 (OCH2), 53.35 (NCH2), 44.94 (NCH2). Anal.calcd. for C26H21FN6OS × HCl × 2H2O (Mr = 557.04): C 56.06. H 4.70. N 15.09; found: C 55.93. H 4.79. N 15.00.
2-(3-Methoxy-4-((1-benzyl-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-6-(4,5-dihydro-1H-imidazol-2-yl)benzothiazole hydrochloride 37c. Compound 37c was prepared using the above-mentioned procedure from 32 (60.0 mg, 0.28 mmol) and 31c (90.5 mg, 0.28 mmol) to obtain 37c as a yellow powder (32.5 mg, 19%; m.p. >240 °C). 1H NMR (400 MHz, DMSO) δ 10.74 (2H, s, CNH), 8.81 (1H, d, J = 1.7 Hz), 8.34 (1H, s), 8.28 (1H, d, J = 8.6 Hz), 8.08 (1H, dd, J = 8.6, 1.8 Hz), 7.73 (1H, dd, J = 8.4, 2.1 Hz), 7.69 (1H, d, J = 2.0 Hz), 7.43–7.29 (6H, m), 5.64 (2H, s, NCH2), 5.27 (2H, s, OCH2), 4.06 (4H, s, NCH2), 3.88 (3H, s, OCH3). 13C NMR (101 MHz. DMSO) δ 172.43 (CNH), 165.19, 151.55, 142.86, 136.43, 135.34, 129.26, 128.49, 127.03, 125.56, 123.97, 123.42, 122.04, 118.97, 114.04, 110.33, 62.20 (OCH2), 56.12 (OCH3), 53.33 (NCH2), 44.96 (NCH2). Anal.calcd. for C27H24N6O2S × HCl × 3H2O (Mr = 587.09): C 55.24, H 5.32, N 14.31; found: C 55.36, H 5.38, N 4.17.

3.3. Antiproliferative Activity In Vitro

The growth inhibition activity was assessed according to the slightly modified procedure performed at the National Cancer Institute, Developmental Therapeutics Program [56].

3.3.1. Cell Lines

Examined compounds were dissolved in DMSO (1 × 10−2 M). The experiments were carried out on seven human tumor cell lines and two normal cell lines. The following cell lines were used: HeLa (human cervical adenocarcinoma; purchased from ATCC), CaCo-2 (human colorectal adenocarcinoma), HuT78 (T-cell lymphoma), THP-1 (acute monocytic leukemia), SW620 (colorectal adenocarcinoma, metastatic), MDA-MB-231 (human breast adenocarcinoma), HL60 (promyelocytic leukemia cell line), foreskin fibroblast cells (BJ) and MDCK1 (Madine–Darby canine kidney fibroblast like cells). MDCK1 cells were used between 24 and 26 passages.

3.3.2. Cell Culturing

Adherent cells were cultured in the Dulbecco’s modified Eagle medium—DMEM (Gibco, EU) supplemented with 10 % heat-inactivated fetal bovine serum (FBS, Gibco, EU), 2 mM glutamine, and 100 U/0.1 mg penicillin/streptomycin. Cells on suspension were cultured in RPMI 1640 (Gibco, EU) medium supplemented with 10 % FBS (Gibco, EU), 2 mM glutamine, 1 mM sodium pyruvate, 10 mM HEPES. Cells were grown in humidified atmosphere under the conditions of 37 °C/5% of CO2 gas in the CO2 incubator (IGO 150 CELLlifeTM, JOUAN, Thermo Fisher Scientific, Waltham, MA, USA). A erythrosin B (Sigma-Aldrich, St. Louis, MO, USA) dye exclusion method was used to assess cell viability before plating.

3.3.3. Proliferation Assay

Adherent cells (HeLa, CaCo-2, MCF-7 and MDCK-1) were plated in 96-well flat bottom plates (Greiner, Frickenhausen, Austria) at a concentration of 2 × 104 cells/mL. Suspension cells (THP-1 and HuT78) were plated in 96-well microtiter plates (Sarstead, Newton, USA) at a concentration of 1 × 105 cells/mL. Twenty-four hours later, cells were treated with test agents in five 10-fold dilutions (10−7 to 10−4 M) and incubated for further 72 h. Working dilutions were freshly prepared on the day of testing. The solvent was also tested for eventual inhibitory activity by adjusting its concentration to be the same as in working concentrations. After 72 h of incubation, the cell growth rate was evaluated by performing the MTT assay, which detects dehydrogenase activity in viable cells [57]. For this purpose, upon completion of the incubation period, growth medium was discarded and 50 μL of MTT was added to each well at a concentration of 5 mg/mL. After four hours of incubation at 37 °C, water insoluble MTT-formazan crystals were dissolved in 150 μL of dimethyl-sulfoxide (DMSO) for adherent cells, and in 10 % SDS with 0.01 M/L HCl for cells grown in suspension. The absorbance (OD, optical density) was measured on a microplate reader (iMark, BIO RAD, Hercules, CA, USA) at 595 nm.
Percent of life cells was calculated as follows: % = OD (sample)–OD (background)/OD (control)–OD (background) × 100.
Optical density (OD) of background for adherent cells is the OD of MTT solution and DMSO; OD (background) for suspension cells is OD of the culture medium with MTT and 10% SDS with 0.01 M/L HCl; OD (control) is the OD of the cells growth without tested compounds.
The results were expressed as GI50, a concentration necessary for 50% of inhibition. Calculation of GI50 value curves and QC analysis is performed by using the Excel tools and GraphPadPrism software (La Jolla, CA), v. 5.03. Briefly, individual concentration effect curves are generated by plotting the logarithm of the concentration of tested compounds(X) vs. corresponding percent inhibition values (Y) using least squares fit. The best fit GI50 values are calculated using Log (inhibitor) versus normalized response—Variable slope equation, where Y Ľ 100/(1 ţ 10 ((LogIC50 _ X) * HillSlope)). QC criteria parameters (Z0, S:B, R2, HillSlope) were checked for every GI50 curve.

3.3.4. Cell Cycle Analysis

The HuT78 cells were plated in 6-well plates at a concentration of 5 × 105 cells per well and treated 24 h and 48 h with selected compounds 36c, 42a, 42c, 45a, 45b, 45c and 46c at a concentration of 5 μM. After drug treatment, the cells were fixed with ice-cold 70% ethanol in phosphate-buffered saline (PBS) and incubated with 0.3 μg/mL propidium iodide for 30 min at room temperature. Before being analyzed by flow cytometry (BD FACSCalibur, Becton Dickinson, San Jose, CA, SAD), samples were treated with 0.4 μg/mL RNase A for 5 min at room temperature. The resultant DNA histograms were generated and analyzed using FlowJo 7.6 software (Treestar, Inc, Ashland, OR, USA). Experiments were done in duplicate and the quantitative data are reported as average value ± standard deviation. Comparisons between control (non-treated) and treated groups were done using one-way analysis of variance (ANOVA) with Tukey–Kramer’s post hoc test with MedCalc statistical program. P-value less than 0.05 was considered statistically significant.

3.3.5. Measurement of Mitochondrial Membrane Potential (∆Ψm)

Changes in the (∆Ψm) were measured using TMRE (Tetramethylrhodamine, Ethyl Ester, Perchlorate) dye. In brief, tested cells (HuT78) were plated in 6-well plates at a concentration of 5 × 105 cells per well and treated with 5 μM of compounds 36c, 42a, 42c, 45a, 45b, 45c, and 46c. After 48 h of treatment, cells were collected, centrifuged 6 min at 1100 rpm, and stained with 200 nM TMRE dye according to the kit protocol (TMRE Mitochondrial Membrane Potential Assay Kit, abcam, Cambridge, UK). Positive control cells were treated with 20 μM FCCP (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone) for 10 min. Cells were analyzed by flow cytometry (BD FACSCalibur, Becton Dickinson, San Jose, CA, SAD) and FlowJo software (FlowJo, LLC, Ashland, OR, USA).

3.3.6. Determination of Apoptosis

Proapoptotic potential of compounds was tested on HuT78 cells using Alexa Fluor 488 annexin V and propidium iodide (Alexa Fluor 488 annexin V/Dead Cell Apoptosis Kit, Invitrogen, Thermo Fisher Scientific, Inc., Waltham, MA, USA). Cells were plated in 6-well plates at a concentration 5 × 105 cells/well and treated for 24 and 48 h with 5 µM 36c, 42c, 45a, 45b, 45c, and 46c. After incubation, cells were collected and centrifuged at 1100 rpm for 6 min, stained according to the manufacturer’s protocol and analyzed by flow cytometry (BD FACSCalibur, Becton Dickinson, San Jose, CA, USA) using FlowJo software (FlowJo, LLC, Ashland, OR, USA).

3.4. QSAR

QSAR analysis was performed on the anticancer activity against the MDCK-1 cell and Hut-78 cell line. Anticancer activities were converted in the form of the logarithm (logIC50). For the inactive compounds, whose IC50 values were estimated as 100, logIC50 was set to 2.
The 3D structures were optimized using molecular mechanics force fields (MM+) [58] using the HyperChem 8.0 (HyperCube, Inc., Gainesville, FL, USA). Subsequently, all structures were submitted to geometry optimization using the semi-empirical AM1 method [59]. The 2D and 3D molecular descriptors used in this study were calculated using ADMEWORKS ModelBuilder 7.9.1.0 (Fujitsu Kyushu Systems Limited, Fukuoka, Japan). Employing the QSARINS-Chem 2.2.1 (University of Insubria, Varese, Italy) [60], descriptors with a constant value for more than 80%, and descriptors that were too inter-correlated (>70%) were excluded. The final number of descriptors selected for the generation of models was 455. Generation of QSAR models was obtained by the Genetic Algorithm (GA) using QSARINS. The models were assessed by fitting criteria; internal cross-validation using the leave-one out (LOO) method; and external validation. The robustness of QSAR models was tested by the Y-randomisation test. Investigation of the applicability domain of the prediction model was performed by Williams plots (plotting residuals vs. leverage of training compounds) in order to identify the outliers and influential chemicals. The predicted data for chemicals with leverage values higher than the warning leverage (h*) must be considered with caution. The warning leverage h* is defined as 3p′/n, where n is the number of training compounds and p′ is the number of model parameters [43].

4. Conclusions

6-Halogen-substituted and 6-unsubstituted benzothiazoles were prepared by condensation of corresponding 4-hydroxybenzaldehydes and 2-aminotiophenoles and subsequent O-alkylation with halides to synthesize benzothiazoles 15a20a, 15b20b, and 15c20c linked via phenoxymethylene to the aromatic units. 1,2,3-Triazole-substituted benzothiazoles 21a26a, 21b26b and 21c26c were prepared by regioselective copper(I) catalyzed cycloaddition from corresponding propargylated benzothiazole intermediates and azides. 6-Imidazolyl benzothiazoles 34a34c, 35a, 35c, 36a36c, 37a37c and 6-pyrimidinyl benzothiazoles 38a, 38b, 39c, 40a40c, and 41a, 41c were prepared by cyclocondensation of 5-amidino-2-aminothiophenoles and corresponding benzaldehydes.
We found that the antiproliferative capacity of the tested compounds varied (after 72 h of exposure, IC50 ranged from 1.4 × 10−6 M to >100 × 10−6 M). The majority of compounds from the non-substituted and halogen-substituted benzothiazole series did not exhibit antiproliferative activity on tested tumor cell lines. From the amidine series, 6-imidazolyl benzothiazole analogs showed strong antiproliferative activity on tested tumor cell lines; however, they were also toxic on normal cells, except for 36c. The introduction of the 1H-1,2,3-triazole substituent in the benzothiazoles 36a36c resulted in reduced cytotoxicity against both MDCK1 and BJ control cell lines, while maintaining excellent growth-inhibitory effect on HuT78 cells with IC50 values of 4.4 µM for 36a, 1.8 µM for 36b and 1.6 µM for 36c and selectivity index (SI) of 9, 18 and 94, respectively. Among benzimidazole amidines, 45a (IC50 = 4.8 µM), 45b (IC50 = 5.5 µM), and 45c (IC50 = 4.1 µM) with 1-benzyl-1,2,3-triazole substituent, as well as 46c (IC50 = 5.1 µM) containing morpholinoethyl-1,2,3-triazole, demonstrated the strongest antiproliferative activity, with SI of 12, 15, 24 and 20, respectively.
The predictive quantitative structure–activity relationship (QSAR) models have been obtained for cytotoxic effects on non-tumor MDCK-1 cells and T-cell lymphoma (HuT78) cells. QSAR analysis showed that the stronger inhibition against MDCK-1 cells depended on larger substituents, the higher 3D distribution of atomic mass at the 6-position of benzothiazoles, the presence of a sulphur atom in the benzothiazole instead of a nitrogen atom in the benzimidazole, and a sulphur atom at topological distance 4 from the fluorine atom of benzothiazoles. The presence of atoms with higher atomic mass and polarizability, such as the sulphur atom and the absence of atoms with higher van der Waals volume at topological distances 8 from the atom at position 6 of the benzothiazole, implied greater activity against HuT78.
Cell cycle perturbation assays on the HuT78 cells treated with 36c, 42c, 45a45c, and 46c showed accumulation of cells in the G2/M and subG0/G1 phase compared to non-treated cells. Annexin-V binding flow cytometry evaluations showed that 48 h post-treatment with 36c, 42c, 45a45c, and 46c the number of apoptotic cells increased. Flow cytometric analysis showed changes in mitochondrial membrane potential, suggesting that the disruption of mitochondrial membrane potential produced by 36c, 42c, 45a45c, and 46c can lead to cytotoxicity and cell death by apoptosis and/or necrosis.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232415843/s1.

Author Contributions

Conceptualization, S.R.-M. and L.G.-O.; synthesis, V.R.K. and L.R.; antiproliferative evaluations, M.L. and L.G.-O.; QSAR analysis, V.R. and D.Š.; writing—original draft preparation, V.R.K., M.L., S.R.-M., L.G.-O. and V.R.; writing—review and editing, V.R.K., M.L., S.R.-M., L.G.-O. and V.R.; supervision, S.R.-M., L.G.-O. and V.R.; project administration, S.R.-M.; funding acquisition, S.R.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Croatian Science Foundation (project No. IP-2018-01-4682).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Nussinov, R.; Tsai, C.-J.; Jang, H. Anticancer Drug Resistance: An Update and Perspective. Drug Resist. Update 2021, 59, 100796. [Google Scholar] [CrossRef] [PubMed]
  4. Torre, B.G.; Albericio, F. The Pharmaceutical Industry in 2020. An Analysis of FDA Drug Approvals from the Perspective of Molecules. Molecules 2021, 26, 627. [Google Scholar] [CrossRef] [PubMed]
  5. Akhtar, J.; Khan, A.A.; Ali, Z.; Haider, R.; Yar, M.S. Structure-Activity Relationship (SAR) Study and Design Strategies of Nitrogen-Containing Heterocyclic Moieties for Their Anticancer Activities. Eur. J. Med. Chem. 2017, 125, 143–189. [Google Scholar] [CrossRef]
  6. Alzhrani, Z.M.M.; Alam, M.M.; Nazreen, S. Recent Advancements on Benzimidazole: A Versatile Scaffold in Medicinal Chemistry. Mini Rev. Med. Chem. 2022, 22, 365–386. [Google Scholar] [CrossRef]
  7. Vasava, M.S.; Bhoi, M.N.; Rathwa, S.K.; Jethava, D.J.; Acharya, P.T.; Patel, D.B.; Patel, H.D. Benzimidazole: A Milestone in the Field of Medicinal Chemistry. Mini Rev. Med. Chem. 2020, 20, 532–565. [Google Scholar] [CrossRef]
  8. Pathak, N.; Rathi, E.; Kumar, N.; Kini, S.G.; Rao, C.M. A Review on Anticancer Potentials of Benzothiazole Derivatives. Mini Rev. Med. Chem. 2020, 20, 12–23. [Google Scholar] [CrossRef]
  9. Haider, K.; Rehman, S.; Pathak, A.; Najmi, A.K.; Yar, M.S. Advances in 2-substituted Benzothiazole Scaffold-based Chemotherapeutic Agents. Arch. Pharm. 2021, 354, e2100246. [Google Scholar] [CrossRef]
  10. Irfan, A.; Batool, F.; Naqvi, S.A.Z.; Islam, A.; Osman, S.M.; Nocentini, A.; Alissa, S.A.; Supuran, C.T. Benzothiazole Derivatives as Anticancer Agents. J. Enzyme Inhib. Med. Chem. 2020, 35, 265–279. [Google Scholar] [CrossRef]
  11. Feng, L.; Su, W.; Cheng, J.; Xiao, T.; Li, H.; Chen, D.; Zhang, Z. Benzimidazole Hybrids as Anticancer Drugs: An Updated Review on Anticancer Properties, Structure–Activity Relationship, and Mechanisms of Action (2019–2021). Arch. Pharm. 2022, 355, e2200051. [Google Scholar] [CrossRef]
  12. Arya, C.G.; Gondru, R.; Li, Y.; Banothu, J. Coumarin–Benzimidazole Hybrids: A Review of Developments in Medicinal Chemistry. Eur. J. Med. Chem. 2022, 227, 113921. [Google Scholar]
  13. Akhtar, J.; Yar, M.S.; Sharma, V.K.; Khan, A.A.; Ali, Z.; Haider, R.; Pathak, A. Recent Progress of Benzimidazole Hybrids for Anticancer Potential. Curr. Med. Chem. 2020, 27, 5970–6014. [Google Scholar] [CrossRef]
  14. Philoppes, J.N.; Lamie, P.F. Design and Synthesis of New Benzoxazole/Benzothiazole-Phthalimide Hybrids as Antitumor-Apoptotic Agents. Bioorg. Chem. 2019, 89, 102978. [Google Scholar] [CrossRef]
  15. Reddy, V.G.; Reddy, T.S.; Jadala, C.; Reddy, M.S.; Sultana, F.; Akunuri, R.; Bhargava, S.K.; Wlodkowic, D.; Srihari, P.; Kamal, A. Pyrazolo-Benzothiazole Hybrids: Synthesis, Anticancer Properties and Evaluation of Antiangiogenic Activity Using in Vitro VEGFR-2 Kinase and in Vivo Transgenic Zebrafish Model. Eur. J. Med. Chem. 2019, 182, 111609. [Google Scholar] [CrossRef]
  16. Aouad, M.; Soliman, M.; Alharbi, M.; Bardaweel, S.; Sahu, P.; Ali, A.; Messali, M.; Rezki, N.; Al-Soud, Y. Design, Synthesis and Anticancer Screening of Novel Benzothiazole-Piperazine-1,2,3-Triazole Hybrids. Molecules 2018, 23, 2788. [Google Scholar] [CrossRef] [Green Version]
  17. Bradshaw, T. Phortress: The Smart Antitumour Agent which Induces its own Metabolism. Pharm. J. 2010, 284, 23–24. [Google Scholar]
  18. Kamal, A.; Tamboli, J.R.; Nayak, V.L.; Adil, S.F.; Vishnuvardhan, M.V.P.S.; Ramakrishna, S. Synthesis of Pyrazolo[1,5-a]Pyrimidine Linked Aminobenzothiazole Conjugates as Potential Anticancer Agents. Bioorg. Med. Chem. Lett. 2013, 23, 3208–3215. [Google Scholar] [CrossRef]
  19. Hong, S.K.; Starenki, D.; Johnson, O.T.; Gestwicki, J.E.; Park, J.I. Analogs of the Heat Shock Protein 70 Inhibitor MKT-077 Suppress Medullary Thyroid Carcinoma Cells. Int. J. Mol. Sci. 2022, 23, 1063. [Google Scholar] [CrossRef]
  20. Mondal, J.; Sreejith, S.; Borah, P.; Zhao, Y. One-Pot Synthesis of Antitumor Agent PMX 610 by a Copper(II)-Incorporated Mesoporous Catalyst. ACS Sustain. Chem. Eng. 2014, 2, 934–941. [Google Scholar] [CrossRef]
  21. Kadri, H.; Matthews, C.S.; Bradshaw, T.D.; Stevens, M.F.G.; Westwell, A.D. Synthesis and Antitumour Evaluation of Novel 2-Phenylbenzimidazoles. J. Enzyme Inhib. Med. Chem. 2008, 23, 641–647. [Google Scholar] [CrossRef] [PubMed]
  22. Aiello, S.; Wells, G.; Stone, E.L.; Kadri, H.; Bazzi, R.; Bell, D.R.; Stevens, M.F.G.; Matthews, C.S.; Bradshaw, T.D.; Westwell, A.D. Synthesis and Biological Properties of Benzothiazole, Benzoxazole, and Chromen-4-One Analogues of the Potent Antitumor Agent 2-(3,4-Dimethoxyphenyl)-5-Fluorobenzothiazole (PMX 610, NSC 721648). J. Med. Chem. 2008, 51, 5135–5139. [Google Scholar] [CrossRef] [PubMed]
  23. Stone, E.L.; Citossi, F.; Singh, R.; Kaur, B.; Gaskell, M.; Farmer, P.B.; Monks, A.; Hose, C.; Stevens, M.F.G.; Leong, C.O.; et al. Antitumour Benzothiazoles. Part 32: DNA Adducts and Double Strand Breaks Correlate with Activity; Synthesis of 5F203 Hydrogels for Local Delivery. Bioorg. Med. Chem. 2015, 23, 6891–6899. [Google Scholar] [CrossRef] [PubMed]
  24. Callero, M.A.; Luzzani, G.A.; Dios, D.O.D.; Bradshaw, T.D.; Perez, A.I.L. Biomarkers of Sensitivity to Potent and Selective Antitumor 2-(4-Amino-3-Methylphenyl)-5-Fluorobenzothiazole (5F203) in Ovarian Cancer. J. Cell. Biochem. 2013, 114, 2392–2404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bistrović, A.; Krstulović, L.; Harej, A.; Grbčić, P.; Sedić, M.; Koštrun, S.; Kraljević Pavelić, S.; Bajić, M.; Raić-Malić, S. Design, Synthesis and Biological Evaluation of Novel Benzimidazole Amidines as Potent Multi-Target Inhibitors for the Treatment of Non-Small Cell Lung Cancer. Eur. J. Med. Chem. 2018, 143, 1616–1634. [Google Scholar] [CrossRef]
  26. Bistrović, A.; Grbčić, P.; Harej, A.; Sedić, M.; Kraljević Pavelić, S.; Koštrun, S.; Plavec, J.; Makuc, D.; Raić-Malić, S. Small Molecule Purine and Pseudopurine Derivatives: Synthesis, Cytostatic Evaluations and Investigation of Growth Inhibitory Effect in Non-Small Cell Lung Cancer A549. J. Enzyme Inhib. Med. Chem. 2018, 33, 271–285. [Google Scholar] [CrossRef]
  27. Racané, L.; Rep, V.; Kraljević Pavelić, S.; Grbčić, P.; Zonjić, I.; Radić Stojković, M.; Taylor, M.C.; Kelly, J.M.; Raić-Malić, S. Synthesis, Antiproliferative and Antitrypanosomal Activities, and DNA Binding of Novel 6-Amidino-2-Arylbenzothiazoles. J. Enzyme Inhib. Med. Chem. 2021, 36, 1952–1967. [Google Scholar] [CrossRef]
  28. Rep, V.; Štulić, R.; Koštrun, S.; Kuridža, B.; Crnolatac, I.; Radić Stojković, M.; Čipčić Paljetak, H.; Perić, M.; Matijašić, M.; Raić-Malić, S. Novel Tetrahydropyrimidinyl-Substituted Benzimidazoles and Benzothiazoles: Synthesis, Antibacterial Activity, DNA Interactions and ADME Profiling. RSC Med. Chem. 2022. advance article. [Google Scholar] [CrossRef]
  29. Racané, L.; Tralić-Kulenović, V.; Mihalić, Z.; Pavlović, G.; Karminski-Zamola, G. Synthesis of New Amidino-Substituted 2-Aminothiophenoles: Mild Basic Ring Opening of Benzothiazole. Tetrahedron 2008, 64, 11594–11602. [Google Scholar] [CrossRef]
  30. Racané, L.; Cindrić, M.; Zlatar, I.; Kezele, T.; Milić, A.; Brajša, K.; Hranjec, M. Preclinical in Vitro Screening of Newly Synthesised Amidino Substituted Benzimidazoles and Benzothiazoles. J. Enzyme Inhib. Med. Chem. 2021, 36, 163–174. [Google Scholar] [CrossRef]
  31. Racané, L.; Ptiček, L.; Fajdetić, G.; Tralić-Kulenović, V.; Klobučar, M.; Kraljević Pavelić, S.; Perić, M.; Paljetak, H.Č.; Verbanac, D.; Starčević, K. Green synthesis and biological evaluation of 6-substituted-2-(2-hydroxy/methoxy phenyl)benzothiazole derivatives as potential antioxidant, antibacterial and antitumor agents. Bioorg. Chem. 2020, 95, 103537. [Google Scholar] [CrossRef]
  32. Darzynkiewicz, Z.; Halicka, H.D.; Zhao, H. Analysis of Cellular DNA Content by Flow and Laser Scanning Cytometry. Adv. Exp. Med. Biol. 2010, 676, 137–147. [Google Scholar]
  33. Matsumoto, T.; Wakefield, L.; Peters, A.; Peto, M.; Spellman, P.; Grompe, M. Proliferative Polyploid Cells Give Rise to Tumors via Ploidy Reduction. Nat. Commun. 2021, 12, 646. [Google Scholar] [CrossRef]
  34. Was, H.; Borkowska, A.; Olszewska, A.; Klemba, A.; Marciniak, M.; Synowiec, A.; Kieda, C. Polyploidy Formation in Cancer Cells: How a Trojan Horse Is Born. Semin. Cancer Biol. 2022, 81, 24–36. [Google Scholar] [CrossRef]
  35. Ma, K.; Chen, G.; Li, W.; Kepp, O.; Zhu, Y.; Chen, Q. Mitophagy, Mitochondrial Homeostasis, and Cell Fate. Front. Cell Dev. Biol. 2020, 8, 467. [Google Scholar] [CrossRef]
  36. Gharbaran, R.; Shi, C.; Onwumere, O.; Redenti, S. Plumbagin Induces Cytotoxicity via Loss of Mitochondrial Membrane Potential and Caspase Activation in Metastatic Retinoblastoma. Anticancer Res. 2021, 41, 4725–4732. [Google Scholar] [CrossRef]
  37. Li, M.H.; Yang, P.; Yang, T.; Zhang, K.; Liu, Y.; Liu, J.; Li, L.M.; Luo, X.Y.; Yang, S.X.; Zou, Q.; et al. A Novel Water-Soluble Benzothiazole Derivative BD926 Triggers ROS-Mediated B Lymphoma Cell Apoptosis via Mitochondrial and Endoplasmic Reticulum Signaling Pathways. Int. J. Oncol. 2016, 49, 2127–2134. [Google Scholar] [CrossRef] [Green Version]
  38. Todeschini, R.; Consonni, V.; Maiocchi, A. The K Correlation Index: Theory Development and Its Application in Chemometrics. Chemom. Intell. Lab. Syst. 1999, 46, 13–29. [Google Scholar] [CrossRef]
  39. Kiralj, R.; Ferreira, M.M.C. Basic Validation Procedures for Regression Models in QSAR and QSPR Studies: Theory and Application. J. Braz. Chem. Soc. 2009, 20, 770–787. [Google Scholar] [CrossRef] [Green Version]
  40. Roy, P.P.; Paul, S.; Mitra, I.; Roy, K. On Two Novel Parameters for Validation of Predictive QSAR Models. Molecules 2009, 14, 1660–1701. [Google Scholar]
  41. Masand, V.H.; Mahajan, D.T.; Nazeruddin, G.M.; Hadda, T.B.; Rastija, V.; Alfeefy, A.M. Effect of Information Leakage and Method of Splitting (Rational and Random) on External Predictive Ability and Behavior of Different Statistical Parameters of QSAR Model. Med. Chem. Res. 2015, 24, 1241–1264. [Google Scholar] [CrossRef]
  42. Chirico, N.; Gramatica, P. Real External Predictivity of QSAR Models: How to Evaluate It? Comparison of Different Validation Criteria and Proposal of Using the Concordance Correlation Coefficient. J. Chem. Inf. Model. 2011, 51, 2320–2335. [Google Scholar] [CrossRef] [PubMed]
  43. Eriksson, L.; Jaworska, J.; Worth, A.P.; Cronin, M.T.D.; McDowell, R.M.; Gramatica, P. Methods for Reliability and Uncertainty Assessment and for Applicability Evaluations of Classification- and Regression-Based QSARs. Environ. Health Perspect. 2003, 111, 1361–1375. [Google Scholar] [CrossRef] [Green Version]
  44. González, M.P.; Terán, C.; Teijeira, M.; Besada, P.; González-Moa, M.J. BCUT Descriptors to Predicting Affinity toward A3 Adenosine Receptors. Bioorg. Med. Chem. Lett. 2005, 15, 3491–3495. [Google Scholar] [CrossRef] [PubMed]
  45. Consonni, V.; Todeschini, R.; Pavan, M. Structure/Response Correlations and Similarity/Diversity Analysis by GETAWAY Descriptors. 1. Theory of the Novel 3D Molecular Descriptors. J. Chem. Inf. Comput. Sci. 2002, 42, 682–692. [Google Scholar] [CrossRef] [PubMed]
  46. González, M.P.; Caballero, J.; Helguera, A.M.; Garriga, M.; González, G.; Fernández, M. 2D Autocorrelation Modelling of the Inhibitory Activity of Cytokinin-Derived Cyclin-Dependent Kinase Inhibitors. Bull. Math. Biol. 2006, 68, 735–751. [Google Scholar] [CrossRef] [PubMed]
  47. Basak, S.C.; Majumdar, S.; Lungu, C. Applications of Multidimensional Space of Mathematical Molecular Descriptors in Large-Scale Bioactivity and Toxicity Prediction-Applications to Prediction of Mutagenicity and Blood-Brain Barrier Entry of Chemicals. Croat. Chem. Acta 2020, 93, 261–272. [Google Scholar] [CrossRef]
  48. Devinyak, O.; Havrylyuk, D.; Lesyk, R. 3D-MoRSE Descriptors Explained. J. Mol. Graph. Model. 2014, 54, 194–203. [Google Scholar] [CrossRef]
  49. Gilani, S.J.; Khan, S.A.; Siddiqui, N.; Verma, S.P.; Mullick, P.; Alam, O. Synthesis and in Vitro Antimicrobial Activity of Novel N-(6-Chlorobenzo[d]Thiazol-2-Yl) Hydrazine Carboxamide Derivatives of Benzothiazole Class. J. Enzyme Inhib. Med. Chem. 2011, 26, 332–340. [Google Scholar] [CrossRef]
  50. Shashank, D.; VIshawanth, T.; Pasha, M.A.; Balasubramaniam, V.; Nagendra, A.; Perumal, P.; Suthakaran, R. Synthesis of Some Substituted Benzothiazole Derivaties and Its Biological Activities. Int. J. Chemtech Res. 2009, 1, 1224–1231. [Google Scholar]
  51. Kumar, M.; Sharma, K.; Fogla, A.K.; Sharma, K.; Rathore, M. Synthesis and Antimicrobial Activity of 2,4-Diaryl-2,3-Dihydrobenzo[b][1,4]Thiazepines. Res. Chem. Intermed. 2013, 39, 2555–2564. [Google Scholar] [CrossRef]
  52. Zheng, M.Q.; Yin, D.-Z.; Qiao, J.P.; Zhang, L.; Wang, Y.X. Syntheses and Evaluation of Fluorinated Benzothiazole Anilines as Potential Tracers for β-Amyloid Plaques in Alzheimer’s Disease. J. Fluor. Chem. 2008, 129, 210–216. [Google Scholar] [CrossRef]
  53. Chandrika, N.T.; Shrestha, S.K.; Ngo, H.X.; Garneau-Tsodikova, S. Synthesis and Investigation of Novel Benzimidazole Derivatives as Antifungal Agents. Bioorg. Med. Chem. 2016, 24, 3680–3686. [Google Scholar] [CrossRef] [Green Version]
  54. Bistrović Popov, A.; Krstulović, L.; Koštrun, S.; Jelić, D.; Bokulić, A.; Radić Stojković, M.; Zonjić, I.; Taylor, M.C.; Kelly, J.M.; Bajić, M.; et al. Design, Synthesis, Antitrypanosomal Activity, DNA/RNA Binding and in Vitro ADME Profiling of Novel Imidazoline-Substituted 2-Arylbenzimidazoles. Eur. J. Med. Chem. 2020, 207, 112802. [Google Scholar] [CrossRef]
  55. Greenidge, P.A.; Kramer, C.; Mozziconacci, J.-C.; Wolf, R.M. MM/GBSA Binding Energy Prediction on the PDBbind Data Set: Successes, Failures, and Directions for Further Improvement. J. Chem. Inf. Model. 2013, 53, 201–209. [Google Scholar] [CrossRef]
  56. Boyd, M.R.; Paull, K.D. Some Practical Considerations and Applications of the National Cancer Institute In Vitro Anticancer Drug Discovery Screen. Drug Dev. Res. 1995, 34, 91–109. [Google Scholar] [CrossRef]
  57. Mosmann, T. Rapid Colorimetric Assay for Cellular Growth and Survival: Application to Proliferation and Cytotoxicity Assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  58. Hocquet, A.; Langgård, M. An Evaluation of the MM+ Force Field. J. Mol. Model. 1998, 4, 94–112. [Google Scholar] [CrossRef]
  59. Stewart, J.J.P. Optimization of Parameters for Semiempirical Methods I. Method. J. Comput. Chem. 1989, 10, 209–220. [Google Scholar] [CrossRef] [Green Version]
  60. Gramatica, P.; Chirico, N.; Papa, E.; Cassani, S.; Kovarich, S. QSARINS: A New Software for the Development, Analysis, and Validation of QSAR MLR Models. J. Comput. Chem. 2013, 34, 2121–2132. [Google Scholar] [CrossRef]
Figure 1. Examples of benzothiazole-based derivatives as anticancer agents.
Figure 1. Examples of benzothiazole-based derivatives as anticancer agents.
Ijms 23 15843 g001
Figure 2. Design of novel 2-aryl-substituted benzothiazole and benzimidazole derivatives.
Figure 2. Design of novel 2-aryl-substituted benzothiazole and benzimidazole derivatives.
Ijms 23 15843 g002
Scheme 1. Reagents and conditions: (i) KSCN, Br2, glacial HOAc, 0 °C, rt, 24 h (ii) 10 N NaOH/KOH, reflux, 12 h; (iii) Na2S2O5, DMF, 100 °C, 2 h.
Scheme 1. Reagents and conditions: (i) KSCN, Br2, glacial HOAc, 0 °C, rt, 24 h (ii) 10 N NaOH/KOH, reflux, 12 h; (iii) Na2S2O5, DMF, 100 °C, 2 h.
Ijms 23 15843 sch001
Scheme 2. Reagents and conditions: (i) RCl/RBr, CH3CN, K2CO3, rt, 12 h; (ii) CuI, TMSN3, DMF:H2O = 9:1, 100 °C, 12 h; (iii) benzyl chloride, NaN3, Et3N, 2 h, rt, Cu(OAc)2, t-BuOH:H2O = 1:1, rt, 12 h.
Scheme 2. Reagents and conditions: (i) RCl/RBr, CH3CN, K2CO3, rt, 12 h; (ii) CuI, TMSN3, DMF:H2O = 9:1, 100 °C, 12 h; (iii) benzyl chloride, NaN3, Et3N, 2 h, rt, Cu(OAc)2, t-BuOH:H2O = 1:1, rt, 12 h.
Ijms 23 15843 sch002
Scheme 3. Reagents and conditions: (i) RCl/RBr, CH3CN, K2CO3, reflux, 8 h; (ii) CuI, TMSN3, DMF:H2O = 9:1, reflux, 6 h; (iii) benzyl chloride, NaN3, Et3N, 30 min, rt, Cu(OAc)2, t-BuOH:H2O = 1:1, 24 h.
Scheme 3. Reagents and conditions: (i) RCl/RBr, CH3CN, K2CO3, reflux, 8 h; (ii) CuI, TMSN3, DMF:H2O = 9:1, reflux, 6 h; (iii) benzyl chloride, NaN3, Et3N, 30 min, rt, Cu(OAc)2, t-BuOH:H2O = 1:1, 24 h.
Ijms 23 15843 sch003
Scheme 4. Reagents and conditions: (i) HOAc, reflux, 3 h; H2O/NaOH pH 12; EtOH/HCl(g), 24 h, rt.
Scheme 4. Reagents and conditions: (i) HOAc, reflux, 3 h; H2O/NaOH pH 12; EtOH/HCl(g), 24 h, rt.
Ijms 23 15843 sch004
Figure 3. Cell cycle distribution. The HuT 78 cells were treated with compounds 36c, 42c, 45a45c and 46c (5 µM) for 24 and 48 h, stained with PI and analyzed by flow cytometry. Changes in the cell cycle in treated compared to control (untreated) cells (a) after 24 h of treatment and (b) after 48 h of treatment. Data represent mean value of two independent experiments. Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Figure 3. Cell cycle distribution. The HuT 78 cells were treated with compounds 36c, 42c, 45a45c and 46c (5 µM) for 24 and 48 h, stained with PI and analyzed by flow cytometry. Changes in the cell cycle in treated compared to control (untreated) cells (a) after 24 h of treatment and (b) after 48 h of treatment. Data represent mean value of two independent experiments. Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Ijms 23 15843 g003
Figure 4. Detection of apoptosis in HuT78 cells after 48 h treatment with 5 µM 36c, 42c, 45a45c and 46c. Control cells were nontreated cells. Apoptosis was assessed by Alexa Fluor 488 annexin V/propidium iodide staining. (a) Cells in Q1, Q2, Q3, and Q4 quadrants are necrotic cells, late apoptotic cells, early apoptotic cells, and normal cells, respectively. (b) Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Figure 4. Detection of apoptosis in HuT78 cells after 48 h treatment with 5 µM 36c, 42c, 45a45c and 46c. Control cells were nontreated cells. Apoptosis was assessed by Alexa Fluor 488 annexin V/propidium iodide staining. (a) Cells in Q1, Q2, Q3, and Q4 quadrants are necrotic cells, late apoptotic cells, early apoptotic cells, and normal cells, respectively. (b) Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Ijms 23 15843 g004
Figure 5. Detection of changes in the mitochondrial membrane potential (∆Ψm) in HuT78 cells after 48 h treatment with 5 µM 36c, 42c, 45a, 45b, 45c and 46c. Control cells were nontreated cells. Changes in the (∆Ψm) were measured using TMRE (Tetramethylrhodamine, Ethyl Ester, Perchlorate) dye. Positive control cells were treated with 20 μM FCCP (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone) (a) High MMP—undamaged mitochondria with high ΔΨm; Low MMP—damaged mitochondria with low ΔΨm. (b) Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Figure 5. Detection of changes in the mitochondrial membrane potential (∆Ψm) in HuT78 cells after 48 h treatment with 5 µM 36c, 42c, 45a, 45b, 45c and 46c. Control cells were nontreated cells. Changes in the (∆Ψm) were measured using TMRE (Tetramethylrhodamine, Ethyl Ester, Perchlorate) dye. Positive control cells were treated with 20 μM FCCP (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone) (a) High MMP—undamaged mitochondria with high ΔΨm; Low MMP—damaged mitochondria with low ΔΨm. (b) Quantitative data are reported as average value  ±  SD. An asterisk (*) denotes values statistically significantly different when compared to the control (p < 0.05).
Ijms 23 15843 g005
Figure 6. Williams plot of applicability domain of the QSAR model for antiproliferative activity against MDCK-1 cells calculated by model (1a). h* = warning leverage.
Figure 6. Williams plot of applicability domain of the QSAR model for antiproliferative activity against MDCK-1 cells calculated by model (1a). h* = warning leverage.
Ijms 23 15843 g006
Figure 7. Williams plot of applicability domain of the QSAR model for antiproliferative activity against HuT78 cells calculated by model (2a). h* = warning leverage.
Figure 7. Williams plot of applicability domain of the QSAR model for antiproliferative activity against HuT78 cells calculated by model (2a). h* = warning leverage.
Ijms 23 15843 g007
Table 1. In vitro growth-inhibitory effects of compounds 15a26a, 15b26b, and 15c26c on selected tumor cell lines.
Table 1. In vitro growth-inhibitory effects of compounds 15a26a, 15b26b, and 15c26c on selected tumor cell lines.
Ijms 23 15843 i001
CpdR1R2R3 IC50 (µM) 1
MDCK1BJHeLaCaCo-2HuT78
15a HIjms 23 15843 i002>100>100>100>100>100
15bClF>100>100>100>100>100
15c OMe79.3 ± 14.4100>100>100>100
16a H87.3 ± 6.9>100>100>100>100
16bFF25.8 ± 9.4>100>10089.3 ± 15.869.8 ± 15.5
16c OMe6.8 ± 6.2>100>100>100>100
17a H100>100>100>100>100
17bHF>100>100>100>100>100
17c OMe78.0 ± 27.8>100>100>100>100
18a HIjms 23 15843 i00317.6 ± 5.7>10042.4 ± 7.510013.2 ± 1.2
18bClF29.8 ± 17.7>100>100>100100
18c OMe41.4 ± 8.1>10067.3 ± 7.8>10055.6 ± 34.6
19a H1.1 ± 0.2>100>100>100>100
19bFF16.3 ± 11.5>100>100>100100
19c OMe1.1 ± 0.2>100>100>100>100
20a H65.7 ± 11.8>100>100>100>100
20bHF94.2 ± 18.4>100>10028.7 ± 16.7>100
20c OMe85.9 ± 15.7100>10089.2 ± 16.864.7 ± 11.3
21a HIjms 23 15843 i00412.6 ±4.8>100100>1006.8 ± 6.2
21bClF10.1 ±4.584.1 ±24.7100>1003.6 ± 3.1
21c OMe43.2 ±12.4>100100>10024.3 ± 6.5
22a H1.1 ± 0.2>10054.5 ± 29.473.5 ±4.514.9 ± 10.8
22bFF1.0 ± 0.086.4 ± 20.950.6 ± 19.31009.1 ± 2.8
22c OMe0.7 ± 0.3>100100>10038.1 ± 23.4
23a H40.7 ± 5.7>10052.4 ± 18.495.6 ± 6.619.0 ± 4.0
23bHF27.4 ± 4.240.5 ± 17.234.9 ± 14.856.0 ± 14.617.1 ± 7.7
23c OMe29.5 ± 7.210045.7 ± 16.396.4 ± 13.320.0 ± 8.7
24a HIjms 23 15843 i00511.4 ± 14.5>100>100>100>100
24bClF8.8 ± 8.3>100>100>100>100
24c OMe41.1 ± 17.2>10082.8 ± 32.3>10049.6 ± 38.0
25a H8.2 ± 5.810 ± 0.01>100>100>100
25bFF51.3 ± 4.6>100>100>10019.7 ± 10.6
25c OMe22.8 ± 4.9>100>10077.6 ±43.3>100
26a H36.7 ± 18.0>100>100>100>100
26bHF64.8 ± 38.5>100>100>100>100
26c OMe8.5 ± 6.476.7 ± 27.723.3 ± 15.310023.7 ± 5.2
5-FU 55.0± 8.774.0± 3.18.2 ± 1.95.9 ± 0.7>100
1 IC50—Compound concentration that inhibited cell growth by 50%. Data represent mean IC50 (μM) values ± standard deviation (SD) of three independent experiments. Exponentially growing cells were treated with compounds during 72 h. Cytotoxicity was analyzed using MTT survival assay.
Table 2. In vitro growth-inhibitory effects of benzothiazoles 34a38a, 40a, 41a, 34b, 36b38b, 40b, 34c37c, 39c41c and benzimidazoles 42a49a, 42b45b, 47b49b, 42c46c, 48c, and 49c on selected tumor and normal cell lines.
Table 2. In vitro growth-inhibitory effects of benzothiazoles 34a38a, 40a, 41a, 34b, 36b38b, 40b, 34c37c, 39c41c and benzimidazoles 42a49a, 42b45b, 47b49b, 42c46c, 48c, and 49c on selected tumor and normal cell lines.
Ijms 23 15843 i006
CpdR1R2R3X IC50 (µM) 1
MDCK1BJHeLaCaCo-2HuT78
34aIjms 23 15843 i007HIjms 23 15843 i002S2.8 ± 0.33.9 ± 0.33.4 ± 0.24.0 ± 0.22.2 ± 0.3
34bFS2.2 ± 0.311.2 ± 6.04.5 ± 4.37.0 ± 5.42.1 ± 0.2
34cOMeS2.1 ± 0.34.0 ± 0.13.1 ± 0.26.3 ± 3.01.8 ± 0.7
35aIjms 23 15843 i007HIjms 23 15843 i003S2.7 ± 0.33.2 ± 0.318.9 ± 3.626.7 ± 2.11.4 ± 0.2
35cOMeS7.5 ± 3.56.7 ± 4.921.8 ± 5.518.2 ± 3.36.2 ± 2.7
36aIjms 23 15843 i007HIjms 23 15843 i004S31.8 ± 4.038.5 ± 4.662.6 ± 22.024.8 ± 9.24.4 ± 3.9
36bFS25.2 ± 1.332.6 ± 3.430.0 ± 1.038.3 ± 1.21.8 ± 0.4
36cOMeS100>100>100>1001.6 ± 0.8
37aIjms 23 15843 i007HIjms 23 15843 i005S1.5 ± 0.34.3 ± 3.63.6 ± 0.13.7 ± 1.51.6 ± 0.8
37bFS2.4 ± 0.35.3 ± 2.94.0 ± 0.46.1 ± 2.72.1 ± 0.2
37cOMeS2.6 ± 0.14.8 ± 3.53.4 ± 0.24.5 ± 2.73.0 ± 1.0
38aIjms 23 15843 i007HIjms 23 15843 i002S27.1 ± 3.422.6 ± 4.619.7 ± 3.125.4 ± 3.516.2 ±5.5
38bFS26.6 ± 3.132.2 ± 2.822.1 ± 1.219.4 ± 4.74.0 ±0.9
39cIjms 23 15843 i007OMeIjms 23 15843 i003S10038.1 ±1.4>10034.7 ±2.812.7 ±0.9
40aIjms 23 15843 i007HIjms 23 15843 i004S59.4 ± 18.8>100100100100
40bFS>100>100>100>10037.3 ± 12.8
40cOMeS>100100>100>100100
41aIjms 23 15843 i007HIjms 23 15843 i005S58.2 ± 17.327.3 ± 6.025.8 ± 0.829.2 ± 4.24.1 ± 3.6
41cOMeS>100>100>100>100>100
42aIjms 23 15843 i007HIjms 23 15843 i002NH100100>100>1007.0 ± 5.2
42bFNH67.3 ± 12.747.0 ± 3.310010011.9 ± 3.6
42cOMeNH10032.8 ± 4.1>100>10024.9 ± 4.6
43aIjms 23 15843 i007HIjms 23 15843 i003NH10033.0 ±20.3>100>10028.6 ± 3.9
43bFNH100>100>100>10043.3 ± 4.4
43cOMeNH63.4 ± 8.930.9 ± 8.7>100>10017.7 ± 6.1
44aIjms 23 15843 i007HIjms 23 15843 i004NH10073.3 ± 3.2100>10089.2 ± 20.6
44bFNH>10045.1 ± 20.9>100>10065.6 ± 17.3
44cOMeNH100>100>100>100100
45aIjms 23 15843 i007HIjms 23 15843 i005NH10055.6 ± 13.4>1001004.8 ± 3.6
45bFNH46.1 ± 14.882.1 ± 0.4>1001005.5 ± 4.0
45cOMeNH68.5 ± 22.7>100>100>1004.1 ± 3.3
46aIjms 23 15843 i007HIjms 23 15843 i008NH100100>100>10063.0 ± 15.1
46cOMeNH>100>100>100>1005.1 ± 4.2
47aIjms 23 15843 i007HIjms 23 15843 i009NH100>100>100>100100
47bFNH>100100>100>100100
48aIjms 23 15843 i007HIjms 23 15843 i010NH100>100>100>100100
48bFNH>100100>100>100100
48cOMeNH>100100>100>10069.6 ± 25.9
49aIjms 23 15843 i007HIjms 23 15843 i011NH10080.4 ± 9.9100>100100
49bFNH82.9 ± 6.664.8 ± 9.2100>100100
49cOMeNH>100>100100>100100
1 IC50—Compound concentration that inhibited cell growth by 50 %. Data represent mean IC50 (μM) values ± standard deviation (SD) of three independent experiments. Exponentially growing cells were treated with compounds during 72 h. Cytotoxicity was analyzed using MTT survival assay.
Table 3. Additional antiproliferative testing of selected compounds.
Table 3. Additional antiproliferative testing of selected compounds.
CpdStructure IC50 (µM) 1
SW620MDA-MB-231HL60THP1
36aIjms 23 15843 i0125.6 ± 1.233.0 ± 4.918.8 ± 1.120.4 ± 3.3
36cIjms 23 15843 i013100>100100>100
38bIjms 23 15843 i01430.5 ± 9.242.4 ± 7.215.8 ± 2.314.7 ± 4.1
39cIjms 23 15843 i01534.2 ± 7.138.5 ± 1.631.1 ± 3.069.7 ± 6.3
42aIjms 23 15843 i01610076.3 ± 11.833.2 ± 1.844.5 ± 5.3
42bIjms 23 15843 i01792.7 ± 34.384.5 ± 18.344.4 ± 12.6100
42cIjms 23 15843 i01829.8 ± 3.038.9 ± 4.219.5 ± 0.819.7 ± 2.4
43aIjms 23 15843 i019>10010040.4 ± 12.029.7 ± 8.6
45aIjms 23 15843 i020>10010048.1 ± 16.40.8 ± 0.1
45bIjms 23 15843 i02110010030.0 ± 12.32.5 ± 1.5
45cIjms 23 15843 i02210010025.1 ± 16.0>100
46cIjms 23 15843 i023>100>10050.7 ± 20.5100
1 IC50—Compound concentration that inhibited cell growth by 50 %. Data represent mean IC50 (μM) values ± standard deviation (SD) of three independent experiments. Exponentially growing cells were treated with compounds during 72 h. Cytotoxicity was analyzed using MTT survival assay.
Table 4. The statistical results of QSAR models 1 and 2.
Table 4. The statistical results of QSAR models 1 and 2.
Statistical ParametersModel 1aModel 1bModel 2aModel 2b
Ntr60594745
Nex17171211
R20.710.740.800.87
R2adj0.680.720.780.85
s0.350.330.290.25
F26.7230.7642.4866.48
Kxx0.340.340.220.19
ΔK0.070.080.080.12
RMSEtr0.330.310.280.23
MAEtr0.270.260.220.20
CCCtr0.830.850.890.93
Q2LOO0.640.680.750.83
RMSEcv0.370.350.310.26
MAEcv0.300.290.250.22
CCCcv0.800.810.860.91
R2Yscr0.080.090.090.09
Q2Yscr−0.13−0.13−0.16−0.15
RMSEAV Yscr0.580.590.590.61
RMSEext0.360.340.360.31
MAEext0.300.290.300.24
R2ext0.810.830.880.87
CCCext0.820.850.860.85
Q2F10.770.790.640.68
Q2F20.740.770.640.67
Q2F30.640.690.660.77
r2m average0.450.510.830.75
Δr2m0.290.250.040.13
Applicability domain
N compounds outlier25b-25b, 39c, 47b-
N compounds out of app.dom.----
LOO (the leave-one out); R2 (coefficient of determination); R2adj (adjusted coefficient of determination); s (standard deviation of regression); F (Fisher ratio); Kxx (multivariate correlation index); ΔK (global correlation among descriptors); RMSEtr (root-mean-square error of the training set); MAEtr (mean absolute error of the training set); CCCtr (concordance correlation coefficient of the training set); Q2LOO (cross-validated explained variance); RMSEcv (root-mean-square error of the training set determined through the cross validated method; MAEcv (mean absolute error of the internal validation set); CCCcv (concordance correlation coefficient test set using cross validation); R2Yscr (Y-scramble correlation coefficients); Q2Yscr (Y-scramble cross-validation coefficients); RMSEAV Yscr (root-mean-square error of Y-randomization); RMSEex (root-mean-square error of the external validation set); MAEex (mean absolute error of the external validation set); R2ext (coefficient of determination of validation set); Q2F1, Q2F2, Q2F3 (predictive squared correlation coefficients); CCCext (concordance correlation coefficient of the test set); r2m average (average value of squared correlation coefficients between the observed and (leave-one-out) predicted values of the compounds); Δr2m (absolute difference between the observed and leave-one-out predicted values of the compounds).
Table 5. Correlation matrix between descriptors included in the model 1a.
Table 5. Correlation matrix between descriptors included in the model 1a.
SIC1GATS4pBEHv6BELp1R7m
SIC11.00
GATS4p−0.601.00
BEHv6−0.220.031.00
BELp1−0.070.070.64
R7m0.040.161.000.021.00
Table 6. Correlation matrix between descriptors included in the model 2a.
Table 6. Correlation matrix between descriptors included in the model 2a.
MAT8vMor30mMor09pE2u
MATS8v1.00
MOR30m−0.051.00
Mor09p0.180.051.00
E2u−0.02−0.210.241.00
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rep Kaulić, V.; Racané, L.; Leventić, M.; Šubarić, D.; Rastija, V.; Glavaš-Obrovac, L.; Raić-Malić, S. Synthesis, Antiproliferative Evaluation and QSAR Analysis of Novel Halogen- and Amidino-Substituted Benzothiazoles and Benzimidazoles. Int. J. Mol. Sci. 2022, 23, 15843. https://doi.org/10.3390/ijms232415843

AMA Style

Rep Kaulić V, Racané L, Leventić M, Šubarić D, Rastija V, Glavaš-Obrovac L, Raić-Malić S. Synthesis, Antiproliferative Evaluation and QSAR Analysis of Novel Halogen- and Amidino-Substituted Benzothiazoles and Benzimidazoles. International Journal of Molecular Sciences. 2022; 23(24):15843. https://doi.org/10.3390/ijms232415843

Chicago/Turabian Style

Rep Kaulić, Valentina, Livio Racané, Marijana Leventić, Domagoj Šubarić, Vesna Rastija, Ljubica Glavaš-Obrovac, and Silvana Raić-Malić. 2022. "Synthesis, Antiproliferative Evaluation and QSAR Analysis of Novel Halogen- and Amidino-Substituted Benzothiazoles and Benzimidazoles" International Journal of Molecular Sciences 23, no. 24: 15843. https://doi.org/10.3390/ijms232415843

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop