Next Article in Journal
Electron-Deficient Acetylenes as Three-Modal Adjuvants in SNH Reaction of Pyridinoids with Phosphorus Nucleophiles
Next Article in Special Issue
Development of Functional Composite Cu(II)-Polyoxometalate/PLA with Antimicrobial Properties
Previous Article in Journal
Linnaea borealis L. var. borealis—In Vitro Cultures and Phytochemical Screening as a Dual Strategy for Its Ex Situ Conservation and a Source of Bioactive Compounds of the Rare Species
Previous Article in Special Issue
A New Dimeric Copper(II) Complex of Hexyl Bis(pyrazolyl)acetate Ligand as an Efficient Catalyst for Allylic Oxidations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mild Copper-Catalyzed, l-Proline-Promoted Cross-Coupling of Methyl 3-Amino-1-benzothiophene-2-carboxylate

by
Vilija Kederienė
1,*,
Indrė Jaglinskaitė
1,
Paulina Voznikaitė
1,
Jolanta Rousseau
2,
Patrick Rollin
3,
Algirdas Šačkus
1 and
Arnaud Tatibouët
3,*
1
Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų pl. 19, LT-50254 Kaunas, Lithuania
2
Univ. Artois, CNRS, Centrale Lille, Univ. Lille, UMR 8181—UCCS—Unité de Catalyse et Chimie du Solide, F-62300 Lens, France
3
Institut de Chimie Organique et Analytique (ICOA), Université d’Orléans et CNRS, UMR 7311, BP 6759, F-45067 Orléans, France
*
Authors to whom correspondence should be addressed.
Submission received: 30 September 2021 / Revised: 7 November 2021 / Accepted: 8 November 2021 / Published: 11 November 2021
(This article belongs to the Special Issue Copper in Synthesis and Catalysis)

Abstract

:
Cu-catalyzed N-arylation is a useful tool for the chemical modification of aromatic heterocycles. Herein, an efficient carbon–nitrogen cross-coupling of methyl 3-amino-1-benzothiophene-2-carboxylate with a range of (hetero)aryl iodides using CuI, l-proline and Cs2CO3 in dioxane at moderate temperature is described. The procedure is an extremely general, relatively cheap, and experimentally simple way to afford the N-substituted products in moderate to high yields. The structures of the new heterocyclic compounds were confirmed by NMR spectroscopy and HRMS investigation.

1. Introduction

Compounds containing the benzothiophene frame are important building blocks in organic synthesis and are used in a broad range of applications [1]. The benzothiophene skeleton is present in many organic materials, such as solar cells, light-emitting diodes (OLEDs), and field-effect transistors [2,3,4]. In the medical field, synthetic benzothiophene-based derivatives show various biological activities [5]. Recently, they were evaluated as inhibitors of tubulin polymerization [6] and as candidates for the treatment of human cancer [6]; furthermore, they can be used for anti-HCV (hepatitis C virus) applications [7] as anti-inflammatory agents [8]. Moreover, a number of aminobenzothiophene derivatives are recognized as antioxidant I [9,10], photochemotherapeutic II [11], antimitotic III [12], antimicrobial IV [13], antiproliferative V [14], and anti-inflammatory VI agents [9,15] (Figure 1).
The importance of the aforementioned class of compounds supported our interest in the synthesis of the anthranilic acid analogues in which the thiophene moiety is present as a benzene ring bioisostere [16]. Moreover, N-substituted anthranilic acids are broadly used in the treatment of various diseases, such as arthritis, atypical dermatitis, and bronchial asthma, and they are also described as anti-allergic, analgesic, and anticancer agents [17,18,19,20,21].
Many methods have been developed for the synthesis of anthranilic acids. Copper-catalyzed arylation is one of the most powerful tools for the synthesis of N-arylanthranilic acids, which are important precursors for the synthesis of substituted acridines and acridones [22,23,24,25,26,27].
The traditional Ullmann-type reaction requires specific reaction conditions, such as strong base and high temperature, and offers a limited substrate scope [28]. Following important earlier breakthroughs, a major improvement came in 2001 with the discovery of new versatile and efficient copper/ligand systems. For example, Buchwald and co-workers reported that diamine ligands enable the formation of the C–N bond under much milder Cu-catalyzed conditions [29,30,31]. The same year, Venkataraman and co-workers discovered that the coupling of aryl halides with various nucleophiles could be successfully performed with 1,10-phenanthroline as ligand [32]. Ma and co-workers described that, by using l-proline as an additive, the copper-catalyzed cross-coupling reactions under mild conditions gave N-arylazoles in excellent yields [33,34]. This discovery was followed by several studies to improve C–N coupling conditions with the effect of l-proline as a ligand [35,36,37]. In view of these research studies, copper-catalyzed N-arylation of amines has become one of the most attractive and highly efficient methods to synthesize these compounds due to excellent catalytic selectivity and high functional group compatibility, as well as low toxicity and economic attractiveness [36].
To date, N-substituted benzo[b]thiophene derivatives have been synthesized using palladium-ligand complexes by the C–N coupling method of 3-halobenzo[b]thiophenes with anilines and aminopyridines [38,39,40,41,42] and the substitution of aminobenzo[b]thiophenes with bromobenzenes and bromopyridines [43,44]. Coupling reactions were successful; however, this synthetic pathway requires relatively expensive, air-sensitive, and highly toxic Pd catalysts systems [45]. Moreover, there exist difficulties with removing palladium traces during the purification stage, which is usually time-demanding and expensive; for this reason, this poses a serious practical problem in the pharmaceutical industry [46,47].
For the above reasons, we wish to report mild copper-catalyzed cross-coupling of aminobenzo[b]thiophene derivatives with (hetero)aryl iodides bearing various functional groups. The application of an Ullmann-type methodology to heteroaromatic systems is still relatively rare.

2. Results and Discussion

Here, we report an efficient catalytic system, which makes the methodology environmentally benign and economical, using commercially available copper(I) iodide and natural amino acid. Many copper complexes are effective catalyst precursors for N-arylation reactions. However, copper(I) salts give superior results, compared to copper(II), as they are a more active species [48]. We chose to focus on the use of CuI owing to its stability in air and lower strictness of the reaction conditions.
The precursor methyl 3-amino-1-benzothiophene-2-carboxylate 1 was synthesized under modified conditions by treatment of 2-fluorobenzonitrile with methyl thioglycolate in DMF [49,50].
The coupling reaction between methyl 3-amino-1-benzothiophene-2-carboxylate 1 and 4-iodoanisole was chosen as a model to optimize the reaction conditions with regard to bases, solvents, and ligands (Scheme 1, Table 1). Several details are worth commenting on. Firstly, a strongly electron-donating agent, N,N’-dimethylethylenediamine (DMEDA), was initially investigated as an additive for the present reaction because of its known and excellent activity in CuI-catalyzed cross-coupling [51]. Since a sensitive carboxylate group is present in substrate 1 [49], we used milder standard bases (K3PO4, Cs2CO3, and K2CO3), which are often used for catalytic arylation. The coupling reaction proceed well in the presence of Cs2CO3 as a base in dioxane at a reflux temperature for 24 h; the expected N-arylated coupling product 3a was obtained in 31% yield. Other bases commonly employed for these couplings, such as K2CO3 and K3PO4, were inferior to Cs2CO3, affording 3a in 5 and 15% yield, respectively (Table 1, entries 1–3). Therefore, Cs2CO3 was selected as the base of choice for further screening reactions.
In the assessment of the effect of the solvent, the coupling results showed that dioxane was the most effective solvent, while DMSO and DMF gave only traces of the desired coupling product (Table 1, compare entries 3–6). On the other hand, dioxane is easily removable from reaction mixtures. Several bidentate N,N-ligands (DMEDA [51] and 10-phenanthroline [52]), which are commonly used as standard ligands in Cu-catalyzed cross-coupling reactions, were examined and significant differences in yield were observed (Table 1, entries 3 and 7). Conversely, amino acids can also act as effective bidentate N,O-chelators in the C–N coupling [53]; therefore, we focused our attention on employing l-proline as the additive. As expected, when N,O-donor l-proline was used, the reaction provided the coupling product in a much higher yield, 58% (Table 1, entry 8). l-proline capability for promoting Cu-catalyzed C–N coupling reactions might be dependent on its reactivity as the coupling agent and coordination ability as a bidentate additive [53]. In the literature has been reported an accelerating effect induced by the structure of amino acids and possible catalytic mechanisms for Cu-catalyzed amino acids promoted coupling reactions [33,34,53].
To compare the reactivity, we also studied the copper-catalyzed amination using methyl 3-iodo-1-benzothiophene-2-carboxylate 2 and 4-anisidine as coupling reagent (Table 1, entry 9). The precursor 2 was already known [54] and was synthesized, here, in a higher yield, 73%, via a diazotization–iodination reaction of compound 1 (Scheme 1). However, to our surprise, cross-coupling was not successful, as compound 2 afforded only 12% of product 3a, which, we suppose, was possibly because of a radical reaction mechanism [55]. The literature has described several speculative mechanisms for Ullmann-type coupling reactions for the aryl halide activation step and one of them is a possible radical pathway-atom transfer mechanism involving aryl radical, which causes the transfer of the halide atom from the aryl halide [56]. Methyl 3-iodo-1-benzothiophene-2-carboxylate 2 may form an inactive radical, which leads to the formation of 3a in a very low yield.
The coupling reaction of methyl 3-amino-1-benzothiophene-2-carboxylate (1) with 4-bromoanisole was also tested. Under these conditions, 4-bromoanisole was less reactive, affording coupled product 3a in a 38% yield (Table 1, entry 10), while 4-iodoanisole exhibited greater reactivity. These results established the reactivity order of the aryl halides: iodides > bromides > chlorides [57].
It also seems that the reaction time played an important role. After 6 h of heating using 4-iodoanisole, the yield of the coupled product 3a was only 28%, while, upon allowing the reaction to continue for 24 h, the yield was increased to 58% (Table 1, entries 8, 11–12). A longer reaction time (48 h) did not improve the efficiency of the coupling transformation.
For a copper-catalyzed N-arylation, the choice of a suitable stoichiometric quantity of copper catalyst and ligand is very important. For further optimization of the reaction conditions, the increase in the yields of the coupling products was observed by changing the molar ratio of CuI:l-proline (Scheme 2). As summarized in Table 2, it was found that the ratio of 1:1 for CuI and l-proline was necessary to ensure a satisfactory result. It was noticed that, when different ratios of reagents were used, lower yields of reaction products were obtained (Table 2, compare entries 2–5). Moreover, the ratio of Cu(I) to l-proline used in this study was only equal to 1:1, whereas the ratio generally used in most of the other copper-catalyzed cross-coupling reactions using amino acids as ligands is 1:2 [53]. When 20 mol% of CuI and l-proline each was used, the reaction was complete in 24 h and gave methyl 3-[(4-methoxyphenyl)amino]-1-benzothiophene-2-carboxylate 3a in excellent yield (Table 2, entry 2). Reducing the amounts of copper catalyst and l-proline as a promoter to 10 mol% gave a similar yield (Table 2, entry 6), while 5 mol% afforded slightly worse results (Table 2, entry 7). Thus, the appropriate ratio of CuI:l-proline is essential for the optimal reaction outcome. More importantly, we were pleased to observe that the control experiment with methyl 3-amino-1-benzothiophene-2-carboxylate 1 without ligand was also successful (Table 2, entry 1).
To improve the reaction yield, as well as to shorten the reaction time, an attempt was made to use microwave (MW) irradiation heating (Scheme 3, Table 3). Our previous results showed that dioxane and acetonitrile were the more suitable solvents, while THF, toluene, or DMF gave poor yields of coupling products under MW heating. According to the literature, dioxane is microwave-transparent, and the reaction in this solvent can be thermally activated only if other components in the reaction mixture respond to microwave energy, i.e., if the reaction mixture contains either polar reactants or ions [58]. However, in our case, despite the presence of polar amine and Cs2CO3 in the reaction mixture, MW activation was not very effective. MW heating using acetonitrile did not increase the yields of the coupling products either, contrary to using conventional heating (Table 3). For this reason, further experiments were carried out using conventional heating in dioxane for 24 h.
Thus, the optimized reaction conditions utilized 10 mol% of Cu(I), 10 mol% of l-proline, and Cs2CO3 (2 equiv) in dioxane at reflux under argon. These reaction conditions were applied to the coupling of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with various functionalized aryl iodides to synthesized coupling compound 3as (Figure 2, Table 3 and Table 4). To study the steric and electronic effects on the product selectivity, reactions with aryl iodides having electron-donating and electron-withdrawing groups such as NO2, OCH3, CF3, F, OH, and NH2 were further examined. Table 3 and Table 4 show that all para-, meta-, and ortho-substitutions on the aryl halide were tolerated under these reaction conditions. For example, the palladium-catalyzed C–N coupling of ortho-substituted aryl halides is difficult and is the major drawback of this method [59]. We showed that N-substituted methyl 3-amino-1-benzothiophene-2-carboxylates 3ao can be obtained in moderate to high yields using activated aryl iodides as starting reagents.
The steric hindrance did not seem to influence cross-coupling, as the reaction of aminobenzothiophene 1 with ortho-, meta-, or para-substituted iodides gave products in similar yields. It is noteworthy that activated iodides bearing electron-withdrawing groups such as iodoanisoles gave good yields (Table 3, entries 1–4, and Table 4, entry 1). The coupling of non-substituted phenyl iodide with compound 1 gave a lower yield of the desired product 3l (Table 4, entry 6).
A variety of functional groups can be present on aryl halides; nonetheless, for electron-rich aryl iodides bearing free hydroxy and amino functional groups, low conversion was observed under the above-described standard reaction conditions (Table 4, entries 7 and 9). After some experimentation, we found out that this problem could be easily solved by the protection of those functional groups (Table 4, entries 8 and 10). For that purpose, N-(4-iodophenyl)acetamide and 4-iodophenyl acetate were prepared from 4-iodophenol and 4-iodoaniline, respectively, following known procedures [60]. Acylation of the –OH and –NH2 groups proved to be crucial and resulted in the formation of the products 3m and 3o in higher yield (Table 4, entries 8, 10). The cross-coupling approach of compound 1 with 4-iodophenyl acetate formed the same final product 3m as coupling with 2-iodophenol (Table 4, entries 7–8). Coupling reactions of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with 2-iodoaniline, N-(2-iodophenyl)acetamide and 2-iodophenol were unsuccessful and gave traces of the products (Table 4, entries 11–13).
However, the Ullmann-type reaction between 2-iodoaniline and compound 1 after additional heating for 72 h gave an interesting tetracyclic derivative 4 in a 41% yield (Scheme 4). This new diazepinone 4 was the result of a C–N coupling, followed by spontaneous intramolecular condensation in one step, with loss of methanol. Careful NMR analysis permitted the elucidation of structure 4. The combined application of HSQC, HMBC, and COSY experiments finally enabled the unambiguous assignment of 5a,7-dihydro-6H-benzothieno[3,2-b][1,5]benzodiazepin-6-one 4.
The study of the literature revealed only a few recently described examples via the C–N cross-coupling of diazepinone compounds. For example, recently reported synthetic pathways to dibenzodiazepine analogues required catalytic quantities of palladium [61]. Typically, several steps are required to prepare dibenzodiazepinones, beginning with the copper-catalyzed coupling reactions of anthranilic acids or substituted benzoates [62]. On the other hand, double amination of ortho-substituted aryl bromides has been reported, which revealed a strong ortho-substituent effect caused by N-aryl aminocarbonyl groups during copper-catalyzed aryl amination [63]. To our knowledge, only one example of a tricyclic benzothieno[3,2-b]diazepinone derivative has been described and was studied as an antiherpetic agent [64]. For this reason, we are currently working on the improved synthesis of benzothieno[3,2-b][1,5]benzodiazepinones via copper-catalyzed cross-coupling, whose results will be reported in due course.
The reaction of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with 2-iodobenzoic acid using the above conditions failed to give any conversion to the desired product. The possible explanation could be that coordination of the resulting aryl carboxylate to copper resulted in the inactivation of the catalytic species and that the aryl carboxylate might have limited solubility in dioxane [51].
The optimized Cu-catalyzed reaction conditions also were applied to the coupling of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with hetero iodide 2 to synthesized coupling compound 5 (Scheme 5).
The result indicated that methyl 3-amino-1-benzothiophene-2-carboxylates 1 must play an important role in accelerating the Cu(I)-catalyzed cross-coupling reactions. This effect might be influenced by a possible chelating effect of the aminobenzo[b]thiophene N atom, the Cu complex, and the oxygen atom of the 2-carbonyl unit. For example, Liebeskind and co-workers reported Ullmann-type cross-coupling reactions of substituted aryl, heteroaryl, and alkenyl halides using copper(I)-thiophene-2-carboxylate (CuTC) as a promoter [65]. According to the literature, Liebeskind’s catalyst CuTC is superior to other catalysts in various cross-couplings [66].

3. Materials and Methods

3.1. General Information

Reactions in anhydrous conditions were performed under argon atmosphere in pre-dried flasks, using anhydrous solvents (distilled when necessary according to D. D. Perrin, W. L. F. Armarego and D. R. Perrin in Purification of Laboratory Chemicals, Pergamon, Oxford, 1986). All reagents and solvents were purchased from commercial chemical suppliers and used without further purification. The course of the reactions was monitored by initial TLC analysis on aluminum foil-backed plates (Merck Kieselgel 60 F254, Darmstadt, Germany), which were developed using standard visualization techniques or agents: UV fluorescence (254 nm) and by staining with a 1% aq KMnO4 solution. Flash column chromatography was performed with Silica Gel 60 Å (230–400 µm, Merck KGaA, Darmstadt, Germany). Melting points [°C] were measured with a Thermo Scientific 9200 capillary apparatus, Büchi Melting Point M-560 (Büchi Labortechnik AG, Flawil, Switzerland) (for compounds 3c, 3e, 3f), and are uncorrected. NMR spectra were recorded on a 400 MHz Bruker Avance 2 400 and Bruker Avance III 400 (for compounds 3c, 3e, 3f) spectrometers (Bruker BioSpin AG, Fallanden, Switzerland) (400 MHz (1H), 100 MHz (13C), 376 MHz (19F)). Chemical shifts are expressed in parts per million (ppm) downfield from TMS internal standard. Coupling patterns for 1H-NMR are designated as s = singlet, d = doublet, t = triplet, m = multiplet, (some 13C-NMR are designated as d = doublet, q = quartet), and coupling constants are given in Hz. NMR peak assignments were elucidated via DEPT, COSY, and HSQC techniques when necessary. IR spectra from samples in neat form were measured with a Thermo Scientific Nicolet iS10 FT-IR spectrophotometer (Thermo Fisher Scientific Inc., Waltham, MA, USA) and Bruker Tensor 27 (Bruker Optic GmbH, Ettlingen, Germany) (KBr pellets, for compounds 3c, 3e, 3f). IR absorption frequencies are given in cm−1. Low-resolution mass spectra were recorded with Perkin–Elmer Sciex API 300 spectrometer (PerkinElmer Inc., Waltham, MA, USA) and Shimadzu LCMS-2020 (Shimadzu Corporation, Kyoto, Japan) (for compounds 3c, 3e, 3f). High-resolution mass spectra (HRMS) were recorded with a Bruker MaXis spectrometer (Bruker Daltonik GmbH, Bremen, Germany) in the electrospray ionization (ESI) mode. 1H-, 13C-, 19F-, DEPT, and some COSY, HSQC NMR spectra, and HRMS data of all new compounds are provided in Supplementary Materials as Figures S1–S61.

3.2. Synthesis of Methyl 3-Amino-1-benzothiophene-2-carboxylate (1)

To a solution of 2-fluorobenzonitrile (0.45 mL, 4.15 mmol) in dry DMF (2 mL) at room temperature methyl thioglycolate (0.41 mL, 4.58 mmol) was added dropwise. The reaction mixture was stirred at room temperature for 10 min, and K2CO3 (680 mg, 4.92 mmol) was added. After stirring at 105 °C for 2 h, the reaction mixture was poured into crushed ice-water (50 mL). The resulting precipitate was collected by filtration, rinsed with cold water, and air dried to give compound 1 as a pale beige solid; yield: 840 mg, 98%; m.p. 107–108 °C (lit. [67] m.p. 110–111 °C). MS (ESI): m/z = 208 [M + H]+.

3.3. Synthesis of Methyl 3-Iodo-1-benzothiophene-2-carboxylate (2)

Methyl 3-amino-1-benzothiophene-2-carboxylate (601 mg, 2.90 mmol) was gradually added to a vigorously stirred 6 M hydrochloric acid solution (2 mL). The reaction mixture was stirred for 20 min at 60 °C. Then mixture was cooled below 0 °C (ice and NaCl bath), and slowly diazotized with sodium nitrite (219 mg, 3.186 mmol). The resulting diazonium salt was stirred for 1 h at this temperature and was poured at once into a well stirred solution of potassium iodide (721 mg, 4.343 mmol) in water (2 mL). The reaction mixture was heated at 60 °C and once evolution of nitrogen had finished, resulting reaction mixture was cooled. Then, mixture extracted with chloroform (3 × 30 mL) and washed with water (50 mL), brine, and finally dried over MgSO4. After filtration, the solvent was removed under reduced pressure. The obtained residue was purified by column chromatography, Rf = 0.24 (PE/EtOAc 97:3). Yield: 700 mg, 73%; pale yellow solid; m.p. 92–93 °C (lit. [54] m.p. 92–93.5 °C). 1H-NMR (400 MHz, CDCl3): δ = 3.98 (s, 3H, OCH3), 7.47–7.54 (m, 2H, 5-H, 6-H), 7.80–7.83 (m, 1H, 7-H), 7.96–7.98 (m, 1H, 4-H) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.8 (OCH3), 88.4 (Cq), 122.8 (C-4), 126.1 (C-6), 128.3 (C-5, C-7), 130.2 (Cq), 140.4 (Cq), 142.0 (Cq), 162.2 (C=O) ppm. IR (neat): νmax = 1713 (C=O), 1497, 1218 (C-O), 1082, 1052 (C=C) cm−1. MS (ESI): m/z = 319 [M + H]+. HRMS (ESI): m/z [M + H]+ calcd. for C10H8IO2S 318.92842, found 318.92847. HRMS (ESI): m/z [M + Na]+ calcd. for C10H7INaO2S 340.91036, found 340.91040. Compound 2 was previously prepared using a different synthetic pathway. The 1H- and 13C-NMR spectra are in good agreement with the literature data [54].

3.4. Synthesis Procedure for the Preparation of Compounds 3ao

Under argon atmosphere, a mixture of methyl 3-amino-1-benzothiophene-2-carboxylate (1 equiv), aryl iodide (1.5 equiv), Cs2CO3 (2 equiv), CuI (0.1 equiv), and l-proline (0.1 equiv) in dry dioxane (2 mL) was stirred under reflux for 24 h. After the mixture was cooled to room temperature and diluted with water and extracted with ethyl acetate. The combined organic layer was washed with brine, dried over anhydrous Na2SO4, and the solvent was evaporated under reduced pressure. The resulting residue was purified by column chromatography using PE/EtOAc as eluent to afford the corresponding compounds. The data for selected compounds is described below.

3.4.1. Methyl 3-[(4-Methoxyphenyl)amino]-1-benzothiophene-2-carboxylate (3a)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3a (260 mg, 86%); Rf = 0.30 (PE/EtOAc 97:3); yellow solid, m.p. 103–104 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.83 (s, 3H, OCH3), 3.91 (s, 3H, COOCH3), 6.86 (d, J = 8.9 Hz, 2H, 2-HPh, 6-HPh), 7.06–7.10 (m, 3H, 3-HPh, 5-HPh, 6-H), 7.20 (d, J = 8.2 Hz, 1H, 7-H), 7.35–7.39 (m, 1H, 5-H), 7.72 (d, J = 8.2 Hz, 1H, 4-H), 8.79 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 51.9 (COOCH3), 55.7 (OCH3), 103.5 (Cq), 114.5 (CPh-3, CPh-5), 123.4 (C-4, C-7), 125.2 (CPh-2, CPh-6), 125.8 (C-6), 127.7 (C-5), 131.7 (Cq), 135.1 (Cq), 140.5 (Cq), 148.0 (Cq), 157.0 (Cq), 166.2 (C=O) ppm. IR (neat): νmax = 3297 (N-H), 1666 (C=O), 1577, 1508, 1440, 1397, 1230 (C–O), 1146, 1031 (C=C, C–N) cm−1. MS (IS): m/z = 282 [M–OCH3]+, 314 [M + H]+, 336 [M + Na]+. HRMS (ESI): m/z [M + H]+ calcd. for C17H16NO3S 314.08454, found 314.08440. HRMS (ESI): m/z [M + Na]+ calcd. for C17H15NNaO3S 336.06649, found 336.06626.H], 95%). HRMS (ESI+) for C21H29N3NaO5 ([M + Na]+) calcd 426.1999, found 426.2001.

3.4.2. Methyl 3-[(2-Methoxyphenyl)amino]-1-benzothiophene-2-carboxylate (3b)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3b (218 mg, 72%); Rf = 0.22 (PE/EtOAc 98:2); yellow solid; m.p. 123–124 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.89 (s, 3H, COOCH3), 3.91 (s, 3H, OCH3), 6.76–6.80 (m, 1H, HPh), 6.88 (dd, J = 0.9 Hz, J = 7.8 Hz, 1H, 6-HPh), 6.93 (dd, J = 1.1 Hz, J = 8.0 Hz, 1H, 3-HPh), 6.97–7.02 (m, 1H, HPh), 7.14–7.18 (m, 1H, 6-H), 7.40 (td, J = 0.95 Hz, J = 8.1 Hz, 1H, 5-H), 7.52 (d, J = 8.3 Hz, 1H, 7-H), 7.74 (d, J = 8.1 Hz, 1H, 4-H), 8.56 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.1 (COOCH3), 55.9 (OCH3), 108.0 (Cq), 111.0 (CPh-3), 119.7 (CPh-6), 120.5 (CPh), 123.0 (C-4), 123.4 (CPh), 123.6 (C-6), 125.9 (C-7), 127.8 (C-5), 131.8 (Cq), 132.6 (Cq), 140.1 (Cq), 145.6 (Cq), 150.7 (Cq), 165.6 (C=O) ppm. IR (neat): νmax = 3378 (N–H), 1684 (C=O), 1563, 1530, 1502, 1437, 1274, 1239 (C-O), 1218, 1109, 1025 (C=C, C–N) cm−1. MS (ESI): m/z = 282 [M–OCH3]+, 314 [M + H]+, 336 [M + Na]+, 352 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C17H16NO3S 314.08454, found 314.08405.

3.4.3. Methyl 3-[(4-Flourophenyl)amino]-1-benzothiophene-2-carboxylate (3c)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3c (273 mg, 94%); Rf = 0.22 (PE/EtOAc 98:2); yellowish crystals; m.p. 92–93 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.95 (s, 3H, OCH3), 6.99–7.05 (m, 2H, HAr), 7.06–7.11 (m, 2H, HAr), 7.14–7.18 (m, 1H, HBT), 7.26–7.32 (m, 1H, HBT), 7.43–7.49 (m, 1H, HBT), 7.77 (d, J = 8.2 Hz, 1H, 4-HBT), 8.75 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.0 (OCH3), 105.9 (Cq), 115.9 (d, 2JC,F = 23.0 Hz, 2xCH), 123.5 (d, 3JC,F = 14.0 Hz, CH), 124.1 (d, 3JC,F = 8.0 Hz, CH), 125.6 (CH), 127.8 (CH), 131.7 (Cq), 138.4 (d, 3JC,F = 2.0 Hz, Cq), 140.3 (Cq), 146.8 (Cq), 159.7 (d, 1JC,F = 242.0 Hz, Cq), 166.1 (C=O) ppm. 19F-NMR (376 MHz, CDCl3): δ = −119.03 (s, 1F) ppm. IR (KBr): νmax = 3313 (N–H), 1674 (C=O), 1674, 1433, 1278, 1253, 1242, 1147, (C–O, C–F, C=C, C–N) cm−1. MS (ESI): m/z = 302 [M + H]+.

3.4.4. Methyl 3-[(2-Fluorophenyl)amino]-1-benzothiophene-2-carboxylate (3d)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3d (230 mg, 79%); Rf = 0.45 (PE/EtOAc 98:2); yellowish crystals; m.p. 126–127 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.93 (s, 3H, OCH3), 6.96–7.06 (m, 3H, 4-HPh, 5-HPh, 6-HPh), 7.12–7.21 (m, 2H, 3-HPh, 6-H), 7.41–7.45 (m, 2H, 5-H, 7-H), 7.78 (d, J = 8.3 Hz, 1H, 4-H), 8.53 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.1 (OCH3), 108.4 (Cq), 116.0 (d, 2JC,F = 19.1 Hz, CPh-3), 122.2 (d, 4JC,F = 1.4 Hz, CPh-5), 123.4 (C-4), 123.8 (C-6), 123.8 (d, 3JC,F = 7.2 Hz, CPh-4), 124.1 (d, 3JC,F = 3.8 Hz, CPh-6), 125.2 (C-7), 127.8 (C-5), 130.6 (d, 2JC,F = 11.3 Hz, CPh-1), 132.2 (Cq), 140.0 (Cq), 145.0 (Cq), 155.0 (d, 1JC,F = 245.5 Hz, CPh-2), 165.6 (C=O) ppm. 19F-NMR (376 MHz, CDCl3): δ = −127.42 (s, 1F) ppm. IR (neat): νmax = 3324 (N–H), 1675 (C=O), 1433, 1277, 1243 (C–O), 1190 (C–F, C=C, C–N) cm−1. MS (ESI): m/z = 270 [M–OCH3]+, 302 [M + H]+, 324 [M + Na]+, 340 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H13FNO2S 302.06455, found 302.06373.

3.4.5. Methyl 3-{[4-(Trifluoromethyl)phenyl]amino}-1-benzothiophene-2-carboxylate (3e)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3e (297 mg, 97%); Rf = 0.43 (PE/EtOAc 98:2); yellowish crystals; m.p. 125–126 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.96 (s, 3H, OCH3), 7.08 (d, J = 8.4 Hz, 2H, 2xHAr), 7.28–7.31 (m, 1H, HAr), 7.48–7.54 (m, 4H, HAr, HBT), 7.82–7.85 (m, 1H, HBT), 8.68 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.3 (OCH3), 110.7 (Cq), 119.3 (2 × CH), 123.5 (CH), 124.0 (CH), 124.3 (q, 2JC,F = 33.0 Hz, Cq), 125.4 (CH), 126.5 (q, 3JC,F = 4.0 Hz, 2 × CH), 128.0 (CH), 132.2 (Cq), 140.0 (Cq), 143.9 (Cq), 145.8 (Cq), 165.5 (C=O) ppm. 19F-NMR (376 MHz, CDCl3): δ = −61.71 (s, 3F) ppm. IR (KBr): νmax = 3317 (N–H), 1668 (C=O), 1328, 1280, 1260, 1244, 1117, 1110 (C–F, C–O–C, C=C, C–N) cm−1. MS (ESI): m/z = 352 [M + H]+.

3.4.6. Methyl 3-{[3-(Trifluoromethyl)phenyl]amino}-1-benzothiophene-2-carboxylate (3f)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3f (248 mg, 73%); Rf = 0.45 (PE/EtOAc 98:2); yellowish crystals; m.p. 133–134 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.93 (s, 3H, OCH3), 7.16–7.23 (m, 2H, HAr), 7.28–7.31 (m, 2H, HAr), 7.37 (dd, J = 8.0 Hz, J = 11.5 Hz, 2HBT), 7.43–7.49 (m, 1H, HBT), 7.80 (d, J = 8.1 Hz, 1HBT), 8.72 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.3 (OCH3), 109.4 (Cq), 117.2 (q, 3JC–F = 4.0 Hz, CH), 119.4 (q, 3JC–F = 4.0 Hz, CH), 123.5 (CH), 123.6 (CH), 123.9 (CH), 124.0 (q, 1JC–F = 272.0 Hz, Cq), 125.3 (CH), 128.0 (CH), 129.7 (CH), 131.7 (q, 2JC–F = 32.0 Hz, Cq), 131.9 (Cq), 140.1 (Cq), 143.2 (Cq), 144.6 (Cq), 165.7 (C=O) ppm. 19F-NMR (376 MHz, CDCl3): δ = −62.86 (s, 3F) ppm. IR (KBr): νmax = 3312 (N–H), 1680 (C=O), 1337, 1275, 1165, 1143, 1120 (C–F, C–O–C, C=C, C–N) cm−1. MS (ESI): m/z = 352 [M + H]+.

3.4.7. Methyl 3-{[2-(Trifluoromethyl)phenyl]amino}-1-benzothiophene-2-carboxylate (3g)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3g (224 mg, 72%); Rf = 0.45 (PE/EtOAc 98:2); pale pink needles; m.p. 152–153 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.92 (s, 3H, OCH3), 6.94 (d, J = 8.1 Hz, 1H, 6-HPh), 7.11 (t, J = 7.6 Hz, 1H, 4-HPh), 7.14–7.18 (m, 1H, 5-HPh), 7.27–7.33 (m, 2H, 6-H, 7-H), 7.41 (td, J = 1.1 Hz, J = 8.2 Hz, 1H, 5-H), 7.67 (d, J = 7.8 Hz, 1H, 3-HPh), 7.77 (d, J = 8.2 Hz, 1H, 4-H), 8.82 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.4 (OCH3), 110.4 (Cq), 121.3 (q, 2JC,F = 29.8 Hz, CPh-2), 122.3 (CPh-6), 122.7 (CPh-4), 123.6 (C-4), 124.0 (CPh-5), 124.5 (q, 1JC,F = 272.8 Hz, CF3), 125.4 (C-7), 126.9 (q, 3JC,F = 5.3 Hz, CPh-3), 127.9 (C-5), 132.1 (Cq), 132.5 (C-6), 140.1 (Cq), 141.0 (Cq), 144.2 (Cq), 165.6 (C=O) ppm. 19F-NMR (376 MHz, CDCl3): δ = −62.01 (s, 3F) ppm. IR (neat): νmax = 3316 (N–H), 1678 (C=O), 1588, 1463, 1321, 1293, 1274, 1251 (C–O), 1115 (C–F), 1102, 1033 (C=C, C–N) cm−1. MS (ESI): m/z = 320 [M–OCH3]+, 352 [M + H]+, 374 [M + Na]+, 390 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C17H13F3NO2S 352.06136, found 352.06111. HRMS (ESI): m/z [M + Na]+ calcd. for C17H12F3NNaO2S 374.04331, found 374.04331. HRMS (ESI): m/z [M + K]+ calcd. for C17H12F3KNO2S 390.01724, found 390.01729.

3.4.8. Methyl 3-[(3-Methoxyphenyl)amino]-1-benzothiophene-2-carboxylate (3h)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3h (230 mg, 76%); Rf = 0.25 (PE/EtOAc 98:2); pale beige solid; m.p. 89–90 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.74 (s, 3H, OCH3), 3.92 (s, 3H, COOCH3), 6.62–6.66 (m, 3H, HPh), 7.15–7.20 (m, 2H, HPh, 6-H), 7.39–7.43 (m, 1H, 5-H), 7.47 (d, J = 8.3 Hz, 1H, 7-H), 7.75 (d, J = 8.2 Hz, 1H, 4-H), 8.71 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.0 (COOCH3), 55.4 (OCH3), 107.1 (Cq), 107.2 (CPh), 109.3 (CPh), 114.1 (CPh), 123.3 (CPh), 123.6 (C-4), 125.9 (C-7), 127.8 (C-5), 129.9 (C-6), 132.2 (Cq), 140.1 (Cq), 143.7 (Cq), 146.1 (Cq), 160.5 (Cq), 165.9 (C=O) ppm. IR (neat): νmax = 3310 (N–H), 1669 (C=O), 1574, 1536, 1435, 1391, 1274, 1238 (C–O), 1196, 1036 (C=C, C–N) cm−1. MS (ESI): m/z = 282 [M–OCH3]+, 314 [M + H]+. HRMS (ESI): m/z [M + H]+ calcd. for C17H16NO3S 314.08454, found 314.08484. HRMS (ESI): m/z [M + Na]+ calcd. for C17H15NNaO3S 336.06649, found 336.06691.

3.4.9. Methyl 3-[(4-Nitrophenyl)amino]-1-benzothiophene-2-carboxylate (3i)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3i (190 mg, 60%); Rf = 0.20 (PE/EtOAc 9:1); yellow solid; m.p. 139–140 °C (lit. [43] 133–135 °C). 1H-NMR (400 MHz, CDCl3): δ = 3.94 (s, 3H, OCH3), 6.98 (d, J = 8.9 Hz, 2H, 2-HPh, 6-HPh), 7.31 (t, J = 7.6 Hz, 1H, 6-H), 7.49–7.52 (m, 2H, 5-H, 7-H), 7.84 (d, J = 8.3 Hz, 1H, 4-H), 8.14 (d, J = 8.9 Hz, 2H, 3-HPh, 5-HPh), 8.65 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.5 (OCH3), 113.6 (Cq), 117.7 (CPh-2, CPh-6), 123.7 (C-4), 124.5 (C-6), 125.1 (C-5), 125.7 (CPh-3, CPh-5), 128.2 (C-7), 132.3 (Cq), 139.8 (Cq), 141.9 (Cq), 142.0 (Cq), 148.9 (Cq), 165.1 (C=O) ppm. IR (neat): νmax = 3340 (N-H), 1686 (C=O), 1589 (N–O), 1507, 1318 (N–O), 1238 (C–O), 1111 (C=C, C–N) cm−1. MS (ESI): m/z = 329 [M + H]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H13N2O4S 329.05905, found 329.05911. HRMS (ESI): m/z [M + Na]+ calcd. for C16H12N2NaO4S 351.04100, found 351.04112. 1H- and 13C-NMR spectra are in good agreement with the literature data [43].

3.4.10. Methyl 3-[(3-Nitrophenyl)amino]-1-benzothiophene-2-carboxylate (3j)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3j (241 mg, 79%); Rf = 0.33 (PE/EtOAc 9:1); yellow solid; m.p. 141–142 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.94 (s, 3H, OCH3), 7.23 (dd, J = 1.0 Hz, J = 8.2 Hz, 1H, 6-HPh), 7.29 (dd, J = 1.4 Hz, J = 8.1 Hz, 1H, 4-HPh), 7.39–7.43 (m, 2H, 5-H, HPh), 7.47 (td, J = 1.0 Hz, J = 8.1 Hz, 1H, 6-H), 7.81–7.83 (m, 2H, 7-H, HPh), 7.87 (dd, J = 1.0 Hz, J = 8.1 Hz, 1H, 4-H), 8.70 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.3 (OCH3), 111.0 (Cq), 114.4 (C-7), 117.2 (C-4), 123.7 (CPh), 124.2 (CPh-6), 125.0 (CPh), 125.5 (CPh-4), 128.1 (C-6), 129.9 (C-5), 131.9 (Cq), 140.0 (Cq), 143.6 (Cq), 144.1 (Cq), 149.1 (Cq), 165.5 (C=O) ppm. IR (neat): νmax = 3321 (N–H), 1680 (C=O), 1573 (N–O), 1465, 1343 (N–O), 1281, 1240 (C–O) 1064 (C=C, C–N) cm−1. MS (ESI): m/z = 297 [M–OCH3]+, 329 [M + H]+, 351 [M + Na]+, 367 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H13N2O4S 329.05905, found 329.05903. HRMS (ESI): m/z [M + Na]+ calcd. for C16H12N2NaO4S 351.04100, found 351.04092.

3.4.11. Methyl 3-[(2-Nitrophenyl)amino]-1-benzothiophene-2-carboxylate (3k)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3k (238 mg, 75%); Rf = 0.41 (PE/EtOAc 9:1); orange needles; m.p. 209–210 °C (lit. [43] 207–209 °C). 1H-NMR (400 MHz, CDCl3): δ = 3.92 (s, 3H, OCH3), 6.84 (d, J = 8.5 Hz, 1H, HPh), 6.91 (td, J = 1.0 Hz, J = 8.3 Hz, 1H, 6-H), 7.27–7.33 (m, 2H, HPh), 7.46–7.49 (m, 2H, 5-H, HPh), 7.83 (d, J = 8.3 Hz, 1H, 4-H), 8.21 (dd, J = 1.3 Hz, J = 8.5 Hz, 1H, 7-H), 10.29 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.7 (OCH3), 118.6 (Cq), 118.9 (CPh), 119.7 (C-6), 123.7 (C-4), 124.7 (CPh), 124.8 (C-5), 126.6 (C-7), 128.0 (CPh), 133.5 (C), 135.0 (CPh), 135.9 (Cq), 139.1 (Cq), 139.7 (Cq), 140.5 (Cq), 163.8 (C=O) ppm. IR (neat): νmax = 3319 (N–H), 1678 (C=O), 1575 (N–O), 1490, 1435, 1339 (N–O), 1269, 1233 (C–O), 1150 (C=C, C–N) cm−1. MS (ESI): m/z = 329 [M + H]+, 351 [M + Na]+, 367 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H13N2O4S 329.05905, found 329.05894. HRMS (ESI): m/z [M + Na]+ calcd. for C16H12N2NaO4S 351.04100, found 351.04099. HRMS (ESI): m/z [M + K]+ calcd for C16H12KN2O4S 367.01494, found 367.01501. 1H- and 13C-NMR spectra are in good agreement with the literature data [43].

3.4.12. Methyl 3-(Phenylamino)-1-benzothiophene-2-carboxylate (3l)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3l (145 mg, 55%); Rf = 0.45 (PE/EtOAc 98:2); pale beige solid; m.p. 116–117 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.91 (s, 3H, CH3), 7.06–7.15 (m, 4H, HPh, 6-H), 7.25–7.30 (m, 2H, HPh), 7.37 (d, J = 7.9 Hz, 1H, 7-H), 7.41 (d, J = 8.1 Hz, 1H, 5-H), 7.75 (d, J = 8.1 Hz, 1H, 4-H), 8.74 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.0 (OCH3), 106.7 (Cq), 121.8 (2 × CPh), 123.4 (CPh), 123.5 (C-6), 123.6 (C-4), 125.8 (C-7), 127.8 (C-5), 129.2 (2 × CPh), 132.1 (Cq), 140.2 (Cq), 142.4 (Cq), 146.3 (Cq), 165.9 (C=O) ppm. IR (neat): νmax = 3320 (N–H), 1666 (C=O), 1567, 1532, 1235 (C–O), 1061 (C=C, C–N) cm−1. MS (ESI): m/z = 252 [M–OCH3]+, 284 [M + H]+, 306 [M + Na]+, 322 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H14NO2S 284.07398, found 284.07412.

3.4.13. Methyl 3-[(4-Hydroxyphenyl)amino]-1-benzothiophene-2-carboxylate (3m)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3m (73 mg, 25%) using 4-iodophenol as coupling reagent; to yield 3m (145 mg, 50%) using 4-iodophenyl acetate as coupling reagent; Rf = 0.29 (PE/EtOAc 8:2); yellow solid; m.p. 166–167 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.92 (s, 3H, OCH3), 4.97 (s, 1H, OH), 6.79 (d, J = 8.7 Hz, 2H, 2-HPh, 6-HPh), 7.01–7.04 (m, 2H, 3-HPh, 5-HPh), 7.06–7.10 (m, 1H, H-6), 7.21 (d, J = 8.3 Hz, 1H, 7-H), 7.37 (td, J = 1.0 Hz, J = 8.1 Hz, 1H, 5-H), 7.72 (d, J = 8.1 Hz, 1H, 4-H), 8.75 (s, 1H, NH). 13C-NMR (100 MHz, CDCl3): δ = 51.9 (OCH3), 103.5 (Cq), 116.0 (CPh-2, CPh-6), 123.4 (C-6), 123.4 (C-4), 125.4 (CPh-3, CPh-5), 125.8 (C-7), 127.8 (C-5), 131.7 (Cq), 135.2 (Cq), 140.5 (Cq), 148.0 (Cq), 153.0 (Cq), 166.3 (C=O). IR (neat): νmax = 3453 (O–H), 3307 (N–H), 1650 (C=O), 1512, 1441, 1280, 1240, 1191, 1068 (C–O, C=C, C–N, C–O–H) cm−1. MS (ESI): m/z = 268 [M–OCH3]+, 300 [M + H]+, 322 [M + Na]+, 338 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H14NO3S 300.06889, found 300.06887. HRMS (ESI): m/z [M + Na]+ calcd. for C16H13NNaO3S 322.05084, found 322.05069.

3.4.14. Methyl 3-[(4-Aminophenyl)amino]-1-benzothiophene-2-carboxylate (3n)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3n (43 mg, 15%); Rf = 0.17 (PE/EtOAc 8:2); brown solid; m.p. 169–172 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.66 (br s, 2H, NH2), 3.91 (s, 3H, OCH3), 6.66 (d, J = 8.5 Hz, 2H, 2-HPh, 6-HPh), 6.99 (d, J = 8.5 Hz, 2H, 3-HPh, 5-HPh), 7.03–7.07 (m, 1H, 6-H), 7.21 (d, J = 8.3 Hz, 1H, 7-H), 7.35 (td, J = 1.0 Hz, J = 8.1 Hz, 1H, 5-H), 7.70 (d, J = 8.1 Hz, 1H, 4-H), 8.79 (br s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 51.8 (OCH3), 102.1 (Cq), 115.8 (CPh-2, CPh-6), 123.3 (C-4, C-6), 126.0 (CPh-3, CPh-5), 126.0 (C-7), 127.7 (C-5), 131.7 (Cq), 133.1 (Cq), 140.6 (Cq), 144.0 (Cq), 148.6 (Cq), 166.3 (C=O) ppm. IR (neat): νmax = 3472 (N–H), 3375 and 3312 (NH2), 1630 (C=O), 1572, 1514, 1439, 1390, 1237 (C–O), 1064 (C=C, C–N) cm−1. MS (ESI): m/z = 267 [M–OCH3]+, 299 [M + H]+. HRMS (ESI): m/z [M + H]+ calcd. for C16H15N2O2S 299.08487, found 299.08533.

3.4.15. Methyl 3-{[4-(Acetylamino)phenyl]amino}-1-benzothiophene-2-carboxylate (3o)

The representative experimental procedure was applied to compound 1 (200 mg, 0.965 mmol) to yield 3o (180 mg, 55%); Rf = 0.18 (PE/EtOAc 7:3); yellow solid; m.p. 240–241 °C. 1H-NMR (400 MHz, DMSO-d6): δ = 2.03 (s, 3H, COCH3), 3.82 (s, 3H, COOCH3), 7.00 (d, J = 8.7 Hz, 2H, 2-HPh, 6-HPh), 7.22–7.28 (m, 2H, 6-H, 7-H), 7.46–7.51 (m, 3H, 3-HPh, 5-HPh, 5-H), 7.95 (d, J = 8.2 Hz, 1H, 4-H), 8.72 (s, 1H, NH), 9.90 (s, 1H, NHCOCH3) ppm. 13C-NMR (100 MHz, DMSO-d6): δ = 23.9 (COCH3), 51.9 (COOCH3), 106.1 (Cq), 119.7 (CPh-3, CPh-5), 121.5 (CPh-2, CPh-6), 123.6 (C-4), 123.9 (C-6), 124.8 (C-7), 128.0 (C-5), 132.0 (Cq), 135.0 (Cq), 137.4 (Cq), 139.1 (Cq), 145.1 (Cq), 164.3 (C=O), 168.0 (HN-C=O) ppm. IR (neat): νmax = 3279 (N–H), 3275 (N–H), 1670 (C=O), 1538, 1241 (C–O, C=C, C–N) cm−1. MS (ESI): m/z = 309 [M–OCH3]+, 341 [M + H]+, 363 [M + Na]+. HRMS (ESI): m/z [M + H]+ calcd. for C18H17N2O3S 341.09544, found 341.09575. HRMS (ESI): m/z [M + Na]+ calcd. for C18H16N2NaO3S 363.07738, found 363.07767.

3.5. Synthesis of 5a,7-Dihidro-6H-[1]benzothieno[2,3-b][1,5]benzodiazepin-6-one (4)

Compound 1 (100 mg, 0.482 mmol) was dissolved in dry dioxane (2 mL) and treated with 2-iodoaniline (160 mg, 0.724 mmol), Cs2CO3 (315 mg, 0.965 mmol), CuI (10 mg, 0.050 mmol), and l-proline (6 mg, 0.048 mmol), under argon atmosphere. Reaction mixture was stirred under reflux for 72 h. After the mixture was cooled to room temperature and diluted with water (100 mL) and extracted with ethyl acetate (2 × 50 mL). The combined organic layer was washed with brine, dried over Na2SO4, and the solvent was evaporated under reduced pressure. The resulting residue was purified by flash column chromatography (SiO2, eluent: petroleum ether/ethyl acetate, 7:3, Rf = 0.17) to provide compound 4 as white solid. Yield 53 mg (41%), m.p. 214–215 °C. 1H-NMR (400 MHz, DMSO-d6): δ = 6.72 (d, J = 7.7 Hz, 1H, HBT), 6.95–6.99 (m, 1H, HBT), 7.06 (d, J = 7.7 Hz, 1H, HBT), 7.09–7.13 (m, 1H, HBT), 7.38–7.49 (m, 3H, HAr), 8.08 (s, 1H, CH), 8.11 (d, J = 8.0 Hz, 1H, HAr), 11.23 (br s, 1H, NH) ppm. 13C-NMR (100 MHz, DMSO-d6): δ = 108.3 (CBT), 109.2 (CBT), 121.0 (CBT), 121.6 (CAr), 121.8 (CBT), 123.5 (CAr), 124.6 (CAr), 125.1 (CAr), 125.3 (CAr), 126.6 (Cq), 128.6 (Cq), 130.8 (Cq), 134.5 (Cq), 138.1 (Cq), 153.3 (C=O) ppm. IR (neat): νmax = 3126 (N–H), 3072, 1686 (C=O), 1480, 1389, 1289, 752 (C=C, C–N, C=N) cm−1. MS (ESI): m/z = 267 [M + H]+, 389 [M + Na]+, 305 [M + K]+. HRMS (ESI): m/z [M + H]+ calcd. for C15H11N2OS 267.05866, found 267.05844. HRMS (ESI): m/z [M + Na]+ calcd. for C15H10N2NaOS 289.04060, found 289.04039. HRMS (ESI): m/z [M + K]+ calcd. for C15H10KN2OS 305.01454, found 305.01432.

3.6. Synthesis of Dimethyl 3,3′-Iminobis(benzo[b]thiophene-2-carboxylate) (5)

Compound 1 (100 mg, 0.482 mmol) was dissolved in dry dioxane (2 mL) and treated with compound 2 (230 mg, 0.724 mmol), Cs2CO3 (315 mg, 0.965 mmol), CuI (10 mg, 0.050 mmol), and l-proline (6 mg, 0.048 mmol) under argon atmosphere. Reaction mixture was stirred under reflux for 24 h. After the mixture was cooled to room temperature, it was diluted with water (100 mL) and extracted with ethyl acetate (2 × 50 mL). The combined organic layer was washed with brine, dried over Na2SO4, and the solvent was evaporated under reduced pressure. The resulting residue was purified by flash column chromatography (SiO2, eluent: petroleum ether/ethyl acetate, 95:5, Rf = 0.26) to provide compound 5 as yellow solid. Yield 32 mg (25%), m.p. 236–237 °C. 1H-NMR (400 MHz, CDCl3): δ = 3.90 (s, 6H, 2 × OCH3), 7.00–7.04 (m, 2H, 2 × 6-H), 7.16 (d, J = 8.3 Hz, 2H, 2 × 7-H), 7.34–7.38 (m, 2H, 2 × 5-H), 7.77 (d, J = 8.2 Hz, 2H, 2 × 4-H), 9.84 (s, 1H, NH) ppm. 13C-NMR (100 MHz, CDCl3): δ = 52.3 (2 × OCH3), 110.6 (2 × Cq), 123.3 (2 × C-4), 124.2 (2 × C-6), 125.0 (2 × C-7), 127.8 (2 × C-5), 133.5 (2 × Cq), 139.5 (2 × Cq), 142.5 (2 × Cq), 164.7 (2 × C=O) ppm. IR (neat): νmax = 3323 (N–H), 1677 (C=O), 1574, 1438, 1277, 1228 (C–O), 1166, 1065 (C=C, C–N) cm−1. MS (ESI): m/z = 398 [M + H]+, 420 [M + Na]+. HRMS (ESI): m/z [M + H]+ calcd. for C20H16NO4S2 398.05153, found 398.05196.

4. Conclusions

In summary, we demonstrated that the methyl 3-amino-1-benzothiophene-2-carboxylate 1 is a suitable precursor for modern Ullmann-type cross-coupling reactions. We successfully developed an efficient and mild CuI-catalyzed N-arylation reaction of functionalized aminobenzo[b]thiophene with a broad selection of aromatic iodides using l-proline as N,O-donor ligand, providing the coupling products 3ao in moderate to good yields. A tetracyclic diazepinone 4 was obtained from the coupling of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with 2-iodoaniline in a one-pot via C–N coupling followed by immediate intramolecular cyclization. The structures of all synthesized compounds were confirmed by detailed NMR spectroscopy and HRMS investigations.
It is noteworthy that l-proline is significantly inexpensive and readily available as a promoter, and it can be easily removed from the crude reaction mixtures by simply washing them with water.

Supplementary Materials

The following are available online. Figure S1: 1H-NMR (400 MHz, CDCl3) spectrum of compound 2, Figure S2: 13C-NMR (100 MHz, CDCl3) spectrum of compound 2, Figure S3: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3a, Figure S4: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3a, Figure S5: HRMS (ESI) of compound 3a, Figure S6: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3b, Figure S7: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3b, Figure S8: HRMS (ESI) of compound 3b, Figure S9: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3c, Figure S10: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3c, Figure S11: 19F-NMR (100 MHz, CDCl3) spectrum of compound 3c, Figure S12: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3d, Figure S13: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3d, Figure S14: 19F-NMR (100 MHz, CDCl3) spectrum of compound 3d, Figure S15: HRMS (ESI) of compound 3d, Figure S16: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3e, Figure S17: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3e, Figure S18: 19F-NMR (100 MHz, CDCl3) spectrum of compound 3e, Figure S19: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3f, Figure S20: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3f, Figure S21: 19F-NMR (100 MHz, CDCl3) spectrum of compound 3f, Figure S22: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3g, Figure S23: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3g, Figure S24: 19F-NMR (100 MHz, CDCl3) spectrum of compound 3g, Figure S25: HRMS (ESI) of compound 3g, Figure S26: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3h, Figure S27: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3h, Figure S28: DEPT135 (100 MHz, CDCl3) spectrum of compound 3h, Figure S29: HRMS (ESI) of compound 3h, Figure S30: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3i, Figure S31: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3i, Figure S32: HRMS (ESI) of compound 3i, Figure S33: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3j, Figure S34: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3j, Figure S35: HRMS (ESI) of compound 3j, Figure S36: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3k, Figure S37: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3k, Figure S38: HRMS (ESI) of compound 3k, Figure S39: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3l, Figure S40: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3l, Figure S41: HRMS (ESI) of compound 3l, Figure S42: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3m, Figure S43: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3m, Figure S44: HRMS (ESI) of compound 3m, Figure S45: 1H-NMR (400 MHz, CDCl3) spectrum of compound 3n, Figure S46: 13C-NMR (100 MHz, CDCl3) spectrum of compound 3n, Figure S47: DEPT135 (100 MHz, CDCl3) spectrum of compound 3n, Figure S48: HRMS (ESI) of compound 3n, Figure S49: 1H-NMR (400 MHz, DMSO-d6) spectrum of compound 3o, Figure S50: 13C-NMR (100 MHz, DMSO-d6) spectrum of compound 3o, Figure S51: HRMS (ESI) of compound 3o, Figure S52: 1H-NMR (400 MHz, DMSO-d6) spectrum of compound 4, Figure S53: 13C-NMR (100 MHz, DMSO-d6) spectrum of compound 4, Figure S54: DEPT135 (100 MHz, CDCl3) spectrum of compound 4, Figure S55: COSY spectrum of compound 4, Figure S56: HSQC spectrum of compound 4, Figure S57: HRMS (ESI) of compound 4, Figure S58: 1H-NMR (400 MHz, CDCl3) spectrum of compound 5, Figure S59: 13C-NMR (100 MHz, CDCl3) spectrum of compound 5, Figure S60: DEPT135 (100 MHz, CDCl3) spectrum of compound 5, Figure S61: HRMS (ESI) of compound 5.

Author Contributions

Conceptualization, A.T. and A.Š.; Methodology, J.R.; Validation, V.K., I.J. and P.V.; Formal analysis, V.K. and I.J.; Investigation, V.K., I.J. and P.V.; Data curation, J.R.; Writing—original draft preparation, V.K., A.Š., P.R. and P.V.; Writing—review and editing, J.R., P.R. and A.T.; Visualization V.K. and I.J.; Resources, A.T. and A.Š., Supervision, A.T. and A.Š. All authors have read and agreed to the published version of the manuscript.

Funding

This project has received funding from the Research Council of Lithuania (LMTLT), agreement No [SDS-2010-022].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Acknowledgments

The authors express deep gratitude to Aneta Juknaitė for her valuable contribution.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Duc, D.X. Recent Progress in the Synthesis of Benzo[b]Thiophene. Curr. Org. Chem. 2020, 24, 2256–2271. [Google Scholar] [CrossRef]
  2. Hu, L.; Liu, S.; Xie, G.; Yang, W.; Zhang, B. Bis(benzothiophene-S,S-dioxide) fused small molecules realize solution-processible, high-performance and non-doped blue organic light-emitting diodes. J. Mater. Chem. C 2020, 8, 1002–1009. [Google Scholar] [CrossRef]
  3. Cho, W.; Kim, H.; Gal, Y.S.; Jin, S.H. Highly reliable benzothiophene-phenylquinoline based heteroleptic Ir(III) complexes; The solution process NIR phosphorescence organic light-emitting diodes. Mol. Cryst. Liq. Cryst. 2017, 654, 62–72. [Google Scholar] [CrossRef]
  4. Seo, C.; Choi, J.M.; Hong, S.S.; Lee, J.Y.; Seo, S.Y. Synthesis of novel benzothiophene derivative as a host material for blue phosphorescent organic light-emitting diodes. Dyes Pigm. 2017, 136, 145–149. [Google Scholar] [CrossRef]
  5. Keri, R.S.; Chand, K.; Budagumpi, S.; Somappa, S.B.; Patil, S.A.; Nagaraja, B.M. An Overview of Benzo[b]Thiophene-Based Medicinal Chemistry. Eur. J. Med. Chem. 2017, 138, 1002–1033. [Google Scholar] [CrossRef] [PubMed]
  6. Romagnoli, R.; Baraldi, P.G.; Salvador, M.K.; Preti, D.; Tabrizi, M.A.; Bassetto, M.; Brancale, A.; Hamel, E.; Castagliuolo, I.; Bortolozzi, R.; et al. Synthesis and Biological Evaluation of 2-(Alkoxycarbonyl)-3-anilinobenzo[b]thiophenes and Thieno[2,3-b]pyridines as New Potent Anticancer Agents. J. Med. Chem. 2013, 56, 2606–2618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Ai, T.; Xu, Y.; Qiu, L.; Geraghty, R.J.; Chen, L. Hydroxamic Acids Block Replication of Hepatitis C Virus. J. Med. Chem. 2015, 58, 785–800. [Google Scholar] [CrossRef] [PubMed]
  8. Fakhr, I.M.I.; Radwan, M.A.A.; El-Batran, S.; El-Salam, O.A.M.E.; El-Shenawy, S.M. Synthesis and pharmacological evaluation of 2-substituted benzo[b]thiophenes as anti-inflammatory and analgesic agents. Eur. J. Med. Chem. 2009, 44, 1718–1725. [Google Scholar] [CrossRef]
  9. Metwally, M.A.; Khalifa, M.E.; El-Hiti, G.A. Recent trends in the chemistry of aminobenzo[b]thiophenes. J. Sulfur Chem. 2010, 31, 205–229. [Google Scholar] [CrossRef]
  10. Gouda, M.A.; Eldien, H.F.; Girges, M.M.; Berghot, M.A. Synthesis and Antioxidant Activity of Novel Series of Naphthoquinone Derivatives Attached to Benzothiophene Moiety. Med. Chem. 2013, 3, 2228–2232. [Google Scholar] [CrossRef] [Green Version]
  11. Fossa, P.; Mosti, L.; Menozzi, G.; Marzano, C.; Baccichetti, F.; Bordin, F. Novel angular furo and thieno-quinolinones: Synthesis and preliminary photobiological studies. Bioorg. Med. Chem. 2002, 10, 743–751. [Google Scholar] [CrossRef]
  12. Romagnoli, R.; Baraldi, P.G.; Lopez-Cara, C.; Preti, D.; Tabrizi, M.A.; Balzarini, J.; Bassetto, M.; Brancale, A.; Fu, X.H.; Gao, Y.; et al. Concise Synthesis and Biological Evaluation of 2-Aroyl-5-Amino Benzo[ b ]Thiophene Derivatives As a Novel Class of Potent Antimitotic Agents. J. Med. Chem. 2013, 56, 9296–9309. [Google Scholar] [CrossRef] [Green Version]
  13. Ferreira, I.C.F.R.; Calhelha, R.C.; Estevinho, L.M.; Queiroz, M.J.R.P. Screening of antimicrobial activity of diarylamines in the 2,3,5-trimethylbenzo[b]thiophene series: A structure-activity evaluation study. Bioorg. Med. Chem. Lett. 2004, 14, 5831–5833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Romagnoli, R.; Baraldi, P.G.; Cara, C.L.; Hamel, E.; Basso, G.; Bortolozzi, R.; Viola, G. Synthesis and biological evaluation of 2-(3′,4′,5′- trimethoxybenzoyl)-3-aryl/arylaminobenzo[b]thiophene derivatives as a novel class of antiproliferative agents. Eur. J. Med. Chem. 2010, 45, 5781–5791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Radwan, M.A.A.; Shehab, M.A.; El-Shenawy, S.M. Synthesis and biological evaluation of 5-substituted benzo[b]thiophene derivatives as anti-inflammatory agents. Monatsh. Chem. 2009, 140, 445–450. [Google Scholar] [CrossRef]
  16. Moreira-Lima, L.; Barreiro, E.J. Bioisosterism: A useful strategy for molecular modification and drug design. Curr. Med. Chem. 2005, 12, 23–49. [Google Scholar] [CrossRef]
  17. Cimolai, N. The potential and promise of mefenamic acid. Expert Rev. Clin. Pharmacol. 2013, 6, 289–305. [Google Scholar] [CrossRef]
  18. Osman, S.; Raza, A.; Al-Zaidan, L.; Inchakalody, V.P.; Merhi, M.; Prabhu, K.S.; Abdelaziz, N.; Hydrose, S.; Uddin, S.; Dermime, S. Anti-cancer effects of Tranilast: An update. Biomed. Pharmacother. 2021, 141, 111844. [Google Scholar] [CrossRef]
  19. Prasher, P.; Sharma, M. Medicinal chemistry of anthranilic acid derivatives: A mini review. Drug Dev. Res. 2021, 1–14. [Google Scholar] [CrossRef]
  20. Inglis, J.J.; Criado, G.; Andrews, M.; Feldmann, M.; Williams, R.O.; Selley, M.L. The anti-allergic drug, N-(3′,4′-dimethoxycinnamonyl) anthranilic acid, exhibits potent anti-inflammatory and analgesic properties in arthritis. Rheumatology 2007, 46, 1428–1432. [Google Scholar] [CrossRef] [Green Version]
  21. Congiu, C.; Cocco, M.T.; Lilliu, V.; Onnis, V. New Potential Anticancer Agents Based on the Anthranilic Acid Scaffold. Synthesis and Evaluation of Biological Activity. J. Med. Chem. 2005, 48, 8245–8252. [Google Scholar] [CrossRef] [PubMed]
  22. Hikawa, H.; Mori, Y.; Kikkawa, S.; Azumaya, I. A Radical Pathway for Direct Substitution of Benzyl Alcohols with Water-Soluble Copper Catalyst in Water. Adv. Synth. Catal. 2016, 358, 765–773. [Google Scholar] [CrossRef]
  23. Bueno, M.A.; Silva, L.R.S.P.; Corrêa, A.G. Microwave-Promoted Synthesis of Novel N-Aryl Anthranilic Acids. J. Braz. Chem. Soc. 2008, 19, 1264–1269. [Google Scholar] [CrossRef] [Green Version]
  24. Mei, X.; August, A.T.; Wolf, C. Regioselective Copper-Catalyzed Amination of Chlorobenzoic Acids: Synthesis and Solid-State Structures of N-Aryl Anthranilic Acid Derivatives. J. Org. Chem. 2006, 71, 142–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Singh, P.; Kumar, A.; Sharma, A.; Kaur, G. Identification of amino acid appended acridines as potential leads to anti-cancer drugs. Bioorg. Med. Chem. Lett. 2015, 25, 3854–3858. [Google Scholar] [CrossRef] [PubMed]
  26. Kaur, M.; Singh, P. Targeting Tyrosine kinase: Development of acridone–pyrrole–oxindole hybrids against human breast cancer. Bioor. Med. Chem. Lett. 2019, 29, 32–35. [Google Scholar] [CrossRef]
  27. Velingkar, V.S.; Dandekar, V.D. Microwave-Assisted Synthesis and Evaluation of Substituted Aryl Propyl Acridone-4-Carboxamides as Potential Chemosensitizing Agents for Cancer. Lett. Drug Des. Discov. 2011, 8, 268–275. [Google Scholar] [CrossRef]
  28. Sambiagio, C.; Marsden, S.P.; Blacker, A.J.; McGowan, P.C. Copper catalysed Ullmann type chemistry: From mechanistic aspects to modern development. Chem. Soc. Rev. 2014, 43, 3525–3550. [Google Scholar] [CrossRef]
  29. Kiyomori, A.; Marcoux, J.F.; Buchwald, S.L. An efficient copper-catalyzed coupling of aryl halides with imidazoles. Tetrahedron Lett. 1999, 40, 2657–2660. [Google Scholar] [CrossRef]
  30. Klapars, A.; Antilla, J.C.; Huang, X.; Buchwald, S.L. A General and Efficient Copper Catalyst for the Amidation of Aryl Halides and the N-Arylation of Nitrogen Heterocycles. J. Am. Chem. Soc. 2001, 123, 7727–7729. [Google Scholar] [CrossRef]
  31. Wolter, M.; Klapars, A.; Buchwald, S.L. Synthesis of N-Aryl Hydrazides by Copper-Catalyzed Coupling of Hydrazides with Aryl Iodides. Org. Lett. 2001, 3, 3803–3805. [Google Scholar] [CrossRef]
  32. Gujadhur, R.K.; Bates, C.G.; Venkataraman, D. Formation of Aryl-Nitrogen, Aryl-Oxygen, and Aryl-Carbon Bonds Using Well-Defined Copper(I)-Based Catalysts. Org. Lett. 2001, 3, 4315–4317. [Google Scholar] [CrossRef]
  33. Ma, D.; Zhang, Y.; Yao, J.; Wu, S.; Tao, F. Accelerating Effect Induced by the Structure of α-Amino Acid in the Copper-Catalyzed Coupling Reaction of Aryl Halides with α-Amino Acids. Synthesis of Benzolactam-V8. J. Am. Chem. Soc. 1998, 120, 12459–12467. [Google Scholar] [CrossRef]
  34. Ma, D.; Xia, C. CuI-Catalyzed Coupling Reaction of β-Amino Acids or Esters with Aryl Halides at Temperature Lower than That Employed in the Normal Ullmann Reaction. Facile Synthesis of SB-214857. Org. Lett. 2001, 3, 2583–2586. [Google Scholar] [CrossRef]
  35. Yoo, J.M.; Dao, P.D.Q.; Cho, C.S. Copper-Catalyzed C–N Coupling and Cyclization of 2-(2-Bromophenyl)-1H-Indoles with Primary Amides Leading to Indolo[1,2-c]Quinazolines. Bull. Korean Chem. Soc. 2018, 39, 1105–1108. [Google Scholar] [CrossRef]
  36. Ding, Z.; Nie, N.; Chen, T.; Meng, L.; Wang, G.; Chen, Z.; Hu, J. L-Proline N-oxide dihydrazides as an efficient ligand for cross-coupling reactions of aryl iodides and bromides with amines and phenols. Tetrahedron 2021, 79, 131826. [Google Scholar] [CrossRef]
  37. Xu, L.; Peng, Y.; Pan, Q.; Jiang, Y.; Ma, D. Assembly of Substituted 3-Aminoindazoles from 2-Bromobenzonitrile via a CuBr-Catalyzed Coupling/Condensation Cascade Process. J. Org. Chem. 2013, 78, 3400–3404. [Google Scholar] [CrossRef]
  38. Gaber, H.M.; Bagley, M.C. Regioselective synthesis and biological evaluation of some novel thiophene-containing heterocyclic scaffolds as potential chemotherapeutic agents. Eur. J. Chem. 2011, 2, 214–222. [Google Scholar] [CrossRef] [Green Version]
  39. Queiroz, M.J.R.P.; Begouin, A.; Ferreira, I.C.F.R.; Kirsch, G.; Calhelha, R.C.; Barbosa, S.; Estevinho, L.M. Palladium-Catalysed Amination of Electron-Deficient or Relatively Electron-Rich Benzo[b]Thienyl Bromides—Preliminary Studies of Antimicrobial Activity and SARs. Eur. J. Org. Chem. 2004, 17, 3679–3685. [Google Scholar] [CrossRef] [Green Version]
  40. Lamanna, G.; Menichetti, S. 2,3-Disubstituted Benzo[b]Thiophenes from Diarylalkynes via Electrophilic Addition-Cyclization and Palladium-Catalyzed Cross-Coupling. Adv. Synth. Catal. 2007, 349, 2188–2194. [Google Scholar] [CrossRef]
  41. Ferreira, I.C.F.R.; Queiroz, M.J.R.P.; Kirsch, G. Palladium-catalyzed amination and cyclization to heteroannellated indoles and carbazoles. Tetrahedron 2003, 59, 3737–3743. [Google Scholar] [CrossRef] [Green Version]
  42. Salman, G.A.; Hussain, M.; Iaroshenko, V.; Villinger, A.; Langer, P. Thienopyrroles and Benzothienoquinolines via Palladium-Catalyzed Domino Reactions. Synfacts 2011, 5, 0475. [Google Scholar] [CrossRef]
  43. Queiroz, M.J.R.P.; Calhelha, R.C.; Kirsch, G. Reactivity of several deactivated 3-aminobenzo[b]thiophenes in the Buchwald-Hartwig C-N coupling. Scope and limitations. Tetrahedron 2007, 63, 13000–13005. [Google Scholar] [CrossRef]
  44. Calhelha, R.C.; Queiroz, M.J.R.P. Synthesis of new thieno[3,2-b]pyridine derivatives by palladium-catalyzed couplings and intramolecular cyclizations. Tetrahedron Lett. 2010, 51, 281–283. [Google Scholar] [CrossRef]
  45. Egorova, K.S.; Ananikov, V.P. Toxicity of Metal Compounds: Knowledge and Myths. Organometallics 2017, 36, 4071–4090. [Google Scholar] [CrossRef] [Green Version]
  46. Chen, Y.J.; Chen, H.H. 1,1,1-Tris(Hydroxymethyl)Ethane as a New, Efficient, and Versatile Tripod Ligand for Copper-Catalyzed Cross-Coupling Reactions of Aryl Iodides with Amides, Thiols, and Phenols. Org. Lett. 2006, 8, 5609–5612. [Google Scholar] [CrossRef]
  47. Senra, J.D.; Aguiar, L.C.S.; Simas, A.B.C. Recent Progress in Transition-Metal-Catalyzed C-N Cross-Couplings: Emerging Approaches towards Sustainability. Curr. Org. Synth. 2011, 8, 53–78. [Google Scholar] [CrossRef]
  48. Jiao, J.; Zhang, X.R.; Chang, N.H.; Wang, J.; Wei, J.F.; Shi, X.Y.; Chen, Z.G. A Facile and Practical Copper Powder-Catalyzed, Organic Solvent-and Ligand-Free Ullmann Amination of Aryl Halides. J. Org. Chem. 2011, 76, 1180–1183. [Google Scholar] [CrossRef]
  49. Kriščiūnienė, V.; Matulevičiūtė, G.; Paliulis, O.; Rollin, P.; Šačkus, A. Conversion of 2-Thioxo-2,3-Dihydroquinazolin-4(1H)-Ones to N(3)-Unsubstituted 2-(Het)Arylquinazolin-4(3H)-Ones by Copper-Mediated Pd-Catalysed Cross-Coupling Reactions. Heterocycles 2016, 93, 150–163. [Google Scholar] [CrossRef]
  50. Sun, J.; Yoo, S.E.; Yi, K.Y.; Kim, N.; Kim, E.; Jung, E.; Lee, Y.S.; Suh-Kim, H. Benzofuran and Benzothiophene Derivatives Substituted with Amide, Process for the Preparation Thereof, and Pharmaceutical Compositions Containing the Same. WO Patent WO2009/048274A2, 16 April 2009. [Google Scholar]
  51. Liu, Y.; Bai, Y.; Zhang, J.; Li, Y.; Jiao, J.; Qi, X. Optimization of the Conditions for Copper-Mediated N-Arylation of Heteroarylamines. Eur. J. Org. Chem. 2007, 36, 6084–6088. [Google Scholar] [CrossRef]
  52. Liu, Z.; Chen, Y. Efficient synthesis of 2,3-dihydro-1,4-benzoxazines via intramolecular copper-catalyzed O-arylation. Tetrahedron Lett. 2009, 50, 3790–3793. [Google Scholar] [CrossRef]
  53. Zhang, H.; Cai, Q.; Ma, D. Amino Acid Promoted CuI-Catalyzed C-N Bond Formation between Aryl Halides and Amines or N-Containing Heterocycles. J. Org. Chem. 2005, 70, 5164–5173. [Google Scholar] [CrossRef]
  54. Rejňák, M.; Klíma, J.; Svoboda, J.; Ludvík, J. Synthesis and Electrochemical Reduction of Methyl 3-Halo-1-Benzothiophene-2-Carboxylates. Collect. Czech. Chem. Commun. 2004, 69, 242–260. [Google Scholar] [CrossRef]
  55. Lindley, J. Tetrahedron report number 163: Copper assisted nucleophilic substitution of aryl halogen. Tetrahedron 1984, 40, 1433–1456. [Google Scholar] [CrossRef]
  56. Jones, G.O.; Liu, P.; Houk, K.N.; Buchwald, S.L. Computational Explorations of Mechanisms and Ligand-Directed Selectivities of Copper-Catalyzed Ullmann-Type Reactions. J. Am. Chem. Soc. 2010, 132, 6205–6213. [Google Scholar] [CrossRef]
  57. Rao, H.; Jin, Y.; Fu, H.; Jiang, Y.; Zhao, Y. A Versatile and Efficient Ligand for Copper-Catalyzed Formation of C-N, C-O, and P-C Bonds: Pyrrolidine-2-Phosphonic Acid Phenyl Monoester. Chem. Eur. J. 2006, 12, 3636–3646. [Google Scholar] [CrossRef]
  58. Kappe, C.O. Controlled Microwave Heating in Modern Organic Synthesis. Angew. Chem. Int. Ed. 2004, 43, 6250–6284. [Google Scholar] [CrossRef]
  59. Hu, Y.L.; Wanga, P.C.; Chen, T.; Lu, M. Facile and Efficient Amination of Organic Halides Catalyzed by Copper Sulfate in PEG1000-DIL/Methylcyclohexane Temperature-Dependent Biphasic System. J. Chin. Chem. Soc. 2010, 57, 604–611. [Google Scholar] [CrossRef]
  60. Hoshino, J.; Park, E.J.; Kondratyuk, T.P.; Marler, L.; Pezzuto, J.M.; Van Breemen, R.B.; Mo, S.; Li, Y.; Cushman, M. Selective Synthesis and Biological Evaluation of Sulfate-Conjugated Resveratrol Metabolites. J. Med. Chem. 2010, 53, 5033–5043. [Google Scholar] [CrossRef] [Green Version]
  61. Tsvelikhovsky, D.; Buchwald, S.L. Concise Palladium-Catalyzed Synthesis of Dibenzodiazepines and Structural Analogues. J. Am. Chem. Soc. 2011, 133, 14228–14231. [Google Scholar] [CrossRef] [Green Version]
  62. Al-Tel, T.H.; Al-Qawasmeh, R.A.; Schmidt, M.F.; Al-Aboudi, A.; Rao, S.N.; Sabri, S.S.; Voelter, W. Rational Design and Synthesis of Potent Dibenzazepine Motifs as β-Secretase Inhibitors. J. Med. Chem. 2009, 52, 6484–6488. [Google Scholar] [CrossRef] [PubMed]
  63. Diao, X.; Xu, L.; Zhu, W.; Jiang, Y.; Wang, H.; Guo, Y.; Ma, D. The N-Aryl Aminocarbonyl Ortho-Substituent Effect in Cu-Catalyzed Aryl Amination and Its Application in the Synthesis of 5-Substituted 11-Oxo-Dibenzodiazepines. J. Am. Chem. Soc. 2011, 13, 6422–6425. [Google Scholar] [CrossRef]
  64. Hamilton, H.W.; Nishiguchi, G.; Hagen, S.E.; Domagala, J.D.; Weber, P.C.; Gracheck, S.; Boulware, S.L.; Nordby, E.C.; Cho, H.; Nakamura, T.; et al. Novel benzthiodiazepinones as antiherpetic agents: SAR improvement of therapeutic index by alterations of the seven-membered ring. Bioorg. Med. Chem. Lett. 2002, 12, 2981–2983. [Google Scholar] [CrossRef]
  65. Zhang, S.; Zhang, D.; Liebeskind, L.S. Ambient Temperature, Ullmann-like Reductive Coupling of Aryl, Heteroaryl, and Alkenyl Halides. J. Org. Chem. 1997, 62, 2312–2313. [Google Scholar] [CrossRef]
  66. Wang, M.; Lin, Z. Stille Cross-Coupling Reactions of Alkenylstannanes with Alkenyl Iodides Mediated by Copper(I) Thiophene-2-Carboxylate: A Density Functional Study. Organometallics 2010, 29, 3077–3084. [Google Scholar] [CrossRef]
  67. Beck, J.R. A Direct Synthesis of Benzo[b]Thiophene-2-Carboxylate Esters Involving Nitro Displacement. J. Org. Chem. 1972, 37, 3224–3226. [Google Scholar] [CrossRef]
Figure 1. Biologically active building blocks IVI with an aminobenzothiophene frame in drug discovery.
Figure 1. Biologically active building blocks IVI with an aminobenzothiophene frame in drug discovery.
Molecules 26 06822 g001
Scheme 1. Reagents and conditions: (1) 6 M aq. HCl, 65 °C, 20 min; then, keeping at −18 °C, 1 h; (2) NaNO2 (1.1 equiv), –10 °C, 30 min; (3) KI (1.5 equiv), from 0 to 70 °C, 30 min.
Scheme 1. Reagents and conditions: (1) 6 M aq. HCl, 65 °C, 20 min; then, keeping at −18 °C, 1 h; (2) NaNO2 (1.1 equiv), –10 °C, 30 min; (3) KI (1.5 equiv), from 0 to 70 °C, 30 min.
Molecules 26 06822 sch001
Scheme 2. Synthesis of compound 3a.
Scheme 2. Synthesis of compound 3a.
Molecules 26 06822 sch002
Scheme 3. Synthesis of compound 3as.
Scheme 3. Synthesis of compound 3as.
Molecules 26 06822 sch003
Figure 2. Synthesized compounds 3ao.
Figure 2. Synthesized compounds 3ao.
Molecules 26 06822 g002
Scheme 4. Ullmann-type reaction between 2-iodoaniline and compound 1.
Scheme 4. Ullmann-type reaction between 2-iodoaniline and compound 1.
Molecules 26 06822 sch004
Scheme 5. Synthesis of compound 5.
Scheme 5. Synthesis of compound 5.
Molecules 26 06822 sch005
Table 1. CuI-catalyzed N-arylation of 3-aminobenzothiophene derivative 1—optimization of the catalysis conditions.
Table 1. CuI-catalyzed N-arylation of 3-aminobenzothiophene derivative 1—optimization of the catalysis conditions.
EntryBase (B)Solvent (S)Ligand (L)3a, Yield (%) [a]
1K2CO3DioxaneDMEDA5 [b]
2K3PO4DioxaneDMEDA15 [b]
3Cs2CO3DioxaneDMEDA31 [b]
4Cs2CO3DMSODMEDAtraces [b]
5Cs2CO3DMFDMEDAtraces [b]
6Cs2CO3TolueneDMEDA20 [c]
7Cs2CO3Dioxane1,10-phenanthroline9 [b]
8Cs2CO3Dioxanel-proline58 [b]
9Cs2CO3Dioxanel-proline12 [d]
10Cs2CO3Dioxanel-proline38 [e]
11Cs2CO3Dioxanel-proline28 [f]
12Cs2CO3Dioxanel-proline61 [g]
[a] After purification by column chromatography. [b] Reactions were carried out using methyl 3-amino-1-benzothiophene-2-carboxylate 1 (1.0 equiv), 4-iodoanisole (1.5 equiv), base (2.0 equiv), CuI (0.2 equiv) and ligand (0.6 equiv) at 100 °C for 24 h, unless stated otherwise. [c] At 110 °C. [d] Using methyl 3-iodo-1-benzothiophene-2-carboxylate 2 (1.0 equiv) as starting compound and p-anisidine (1.5 equiv) as coupling partner. [e] Using 4-bromoanisole (1.5 equiv) as coupling partner. [f] For 6 h. [g] For 48 h.
Table 2. Comparative studies utilizing various ratios of catalyst and ligand.
Table 2. Comparative studies utilizing various ratios of catalyst and ligand.
EntryCuI:Ligand (Ratio)CuIl-Proline3a, Yield (%) [a], [b]
11:00.2-54
21:10.20.287
31:20.20.465
41:30.20.658
51:40.20.860
61:10.10.186
71:10.050.0561
[a] Reagents and conditions: methyl 3-amino-1-benzothiophene-2-carboxylate 1 (1.0 equiv); 4-iodoanisole (1.5 equiv); Cs2CO3 (2.0 equiv); CuI; l-proline; dioxane; Δ; 24 h. [b] After purification by column chromatography.
Table 3. Comparative studies utilizing MW and conventional heating [a].
Table 3. Comparative studies utilizing MW and conventional heating [a].
EntrySubstituent (Ar)ProductYield (%) [b]Yield (%) [c]Yield (%) [d]Yield (%) [e]
14-OCH3C6H53a86103561
22-OCH3C6H53b72103252
34-FC6H53c94537529
42-FC6H53d79202650
54-CF3C6H53e97752351
64-CF3C6H53f73184530
[a] After purification by column chromatography. Reagents and conditions: methyl 3-amino-1-benzothiophene-2-carboxylate 1 (1.0 equiv); aryl iodide (1.5 equiv); Cs2CO3 (2.0 equiv); CuI (0.1 equiv); l-proline (0.1 equiv). [b] Dioxane; Δ; 24 h. [c] Dioxane; MW; 1 h. [d] Acetonitrile; MW; 1 h. [e] Acetonitrile; Δ; 24 h.
Table 4. Cu(I)-catalyzed cross-coupling of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with aryl iodides. [a].
Table 4. Cu(I)-catalyzed cross-coupling of methyl 3-amino-1-benzothiophene-2-carboxylate 1 with aryl iodides. [a].
EntrySubstituent (Ar)ProductYield (%) [b]
12-CF3C6H53g72
23-OCH3C6H53h76
34-NO2C6H53i60
43-NO2C6H53j79
52-NO2C6H53k75
6C6H53l55
74-OHC6H53m25
84-OCOCH3C6H53m50
94-NH2C6H53n15
104-NHCOCH3C6H53o55
112-OHC6H53ptrace
122-NH2C6H53rtrace
132-NHCOCH3C6H53strace
[a] Reagents and conditions: methyl 3-amino-1-benzothiophene-2-carboxylate 1 (1.0 equiv); aryl iodide (1.5 equiv); Cs2CO3 (2.0 equiv); CuI (0.1 equiv); and l-proline (0.1 equiv) in dioxane at reflux for 24 h. [b] After purification by column chromatography.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kederienė, V.; Jaglinskaitė, I.; Voznikaitė, P.; Rousseau, J.; Rollin, P.; Šačkus, A.; Tatibouët, A. Mild Copper-Catalyzed, l-Proline-Promoted Cross-Coupling of Methyl 3-Amino-1-benzothiophene-2-carboxylate. Molecules 2021, 26, 6822. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26226822

AMA Style

Kederienė V, Jaglinskaitė I, Voznikaitė P, Rousseau J, Rollin P, Šačkus A, Tatibouët A. Mild Copper-Catalyzed, l-Proline-Promoted Cross-Coupling of Methyl 3-Amino-1-benzothiophene-2-carboxylate. Molecules. 2021; 26(22):6822. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26226822

Chicago/Turabian Style

Kederienė, Vilija, Indrė Jaglinskaitė, Paulina Voznikaitė, Jolanta Rousseau, Patrick Rollin, Algirdas Šačkus, and Arnaud Tatibouët. 2021. "Mild Copper-Catalyzed, l-Proline-Promoted Cross-Coupling of Methyl 3-Amino-1-benzothiophene-2-carboxylate" Molecules 26, no. 22: 6822. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26226822

Article Metrics

Back to TopTop