Next Article in Journal
9G DNAChip Technology: Self-Assembled Monolayer (SAM) of ssDNA for Ultra-Sensitive Detection of Biomarkers
Previous Article in Journal
Development and Validation of Single Nucleotide Polymorphisms (SNPs) Markers from Two Transcriptome 454-Runs of Turbot (Scophthalmus maximus) Using High-Throughput Genotyping
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of Thieno[2,3-b]thiophene Containing Bis-Heterocycles-Novel Pharmacophores

by
Yahia Nasser Mabkhot
1,*,†,
Assem Barakat
1,2,*,†,
Abdullah Mohammed Al-Majid
1,† and
Muhammad Iqbal Choudhary
1,3,†
1
Department of Chemistry, Faculty of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
2
Department of Chemistry, Faculty of Science, Alexandria University, P. O. Box 426, Ibrahimia, Alexandria 21321, Egypt
3
Research Institute of Chemistry, International Center for Chemical Sciences, University of Karachi, Karachi 75270, Pakistan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2013, 14(3), 5712-5722; https://doi.org/10.3390/ijms14035712
Submission received: 27 November 2012 / Revised: 1 February 2013 / Accepted: 18 February 2013 / Published: 12 March 2013

Abstract

:
Thioenethiophene derivatives represent an important class of compounds with diverse biological activities. We describe here the synthesis of a new series of thieno[2,3-b]thiophene containing bis-heterocyclic compounds 37. All the compounds were evaluated for their in vitro antioxidant potential, α-glucosidase and β-glucuronidase inhibiton and anticancer activity against PC-3 cell lines. Compounds 2b (IC50 = 1.3 ± 0.2 μM), 5a (IC50 = 2.3 ± 0.4 μM) and 5b (IC50 = 8.7 ± 0.1 μM) showed a potent inhibition of β-glucuronidase enzyme, more active than the standard d-saccharic acid 1,4-lactone (IC50 = 45.8 ± 2.5 μM). Compounds 5a (IC50 = 22.0 ± 0.3 μM) and 5b (IC50 = 58.4 ± 1.2 μM) were also found to be potent α-glucosidase inhibitors as compared to standard drug (acarbose, IC50 = 841 ± 1.7 μM).

Graphical Abstract

1. Introduction

Thienothiophene derivatives possess diverse biological activities, such as antitumor, antiviral [17], and antibiotic properties. They are also used as antiglaucoma drugs and as inhibitors of platelet aggregation [812]. Recently, numerous studies have been conducted on biological profiles of thieno[2,3-b]thiophene scaffold. Chemically these compounds have also attracted considerable attention as moieties that offer significant synthetic advantages, such as centrosymmetry and higher rigidity. They are also used in the design of novel nonlinear optical (NLO) systems through incorporation within unsymmetrically functionalized cyclophane [2]. Thienothiophene derivatives have been vigorously investigated from various viewpoints, i.e., unusual physical, chemical, biological properties [1316]. On the other hand, imidazopyrimidine and triazolopyrimidine analogs can be suitably modified with the introduction of different heterocyclic moieties to confer a broad range of biological activities, such as antimicrobial effects [1721].
As part of our continuing efforts toward the synthesis of bisheterocycle system and convenient access to the diverse frameworks present in them, we have explored the synthetic potential and generality of a cyclization strategy to gain facile entry into the thieno[2,3-b]thiophene framework from a conveniently accessible precursor as shown in Scheme 1. Our new results on the thieno[2,3-b]thiophene derivatives, including chemical synthesis, their in vitro antioxidant potential, α-glucosidase and β-glucuronidase inhibition, and anticancer activity against PC-3 cell lines are presented herein.

2. Results and Discussion

2.1. Chemistry

The synthesis of the pyrimidine, pyrazole, triazolopyrimidine and imidazopyrimidine derivatives 47 were carried out by reacting commercially available benzoylacetone 1, dimethylformamide dimethylacetal (DMF-DMA), 4-amino-1,2,4-triazole and 2-aminobenzimidazole. Previously 1,1′-(3-methyl-4-phenylthieno[2,3-b]thiophene-2,5-diyl)diethanone 2 was synthesized starting from benzoylacetone 1. The method consists of sequential directed nucleophilic addition, side chain deprotonation, nucleophilic addition, and cyclization using a nitrogen or sulfur moiety as internal nucleophile. Condensation of 2 with dimethylformamide dimethyl acetal (DMF-DMA) under reflux for ten hours in the presence of xylene furnished enaminone 3. Reaction of enaminone 3 with N-nucleophiles, such as urea derivatives in dioxane or EtOH/DMF mixture under reflux for four to six hours in the presence of a catalytic amount of ZnCl2 as a Lewis acid afforded 4ac. The formation of compounds 4ac would involve an initial addition of the amino group in urea to the activated double bond in enaminone derivative 3, followed by deamination to an intermediate, which then undergoes cyclization and aromatization via loss of water, affording the final isolable product. Similarly, enaminone derivative 3 cyclized with hydrazine compounds by refluxing with absolute ethanol for six hours. The novel bispyrazole 5a was assumed to be formed via addition of the amino group in the hydrazine to the activated double bond of the enamine derivative, followed by deamination, dehydration, and subsequently nucleophilic cyclization to afford the final product. The utility of enaminone 3 in the synthesis of annelated heterocycles was further explored via its reaction with 4-amino-1,2,4-triazole in absolute ethanol under reflux for seven hours in the presence of a catalytic amount of ZnCl2 affording compound 6. The study was extended to investigate the behavior of enaminone derivatives 3 with different nucleophiles such as 2-aminobenzimidazole in order to synthesize various heterocyclic ring systems. Thus, the reaction of 3 with 2-iminobenzimidazole in refluxing ethanol in the presence of catalytic amount of ZnCl2 furnished the corresponding product 7. The structures of these compounds were determined by 1H-NMR, EI, IR and UV spectroscopic, and micro analyses for carbon, hydrogen and nitrogen [22].

2.2. Biological Activity Evaluation

Compounds 3, 4b, 5a, 5b and 7 were evaluated for a variety of biological activities including β-glucuronidase and α-glucosidase inhibiton, antioxidant, and anticancer properties by using non-physiologica and in vitro biochemical and mechanism-based assays. The results of the assays are presented in Table 1.
Compound 4b (IC50 = 1.3 ± 0.172 μM) was found to be most potent inhibitor of β-glucuronidase enzyme, more active than the standard, d-saccharic acid 1,4-lactone (IC50 = 45.8 ± 2.5 μM). This compound was found to be inactive in DPPH radical scavenging and α-glucosidase inhibition assays. However interesting results were obtained for pyrazole ring containing compounds 5a and 5b. Both compounds showed a potent inhibition of β-glucuronidase (5a, IC50 = 2.3 ± 0.4 and 5b IC50 = 8.7 ± 0.1 μM) and α-glucosidase enzymes (5a, IC50 = 22 ± 0.3 and 5b IC50 = 58.4 ± 1.2 μM) again more active than standard d-saccharic acid 1,4-lactone (IC50 = 45.8 ± 2.2 μM) and acarbose (IC50 = 841 ± 1.7 μM), respectively. Compound 5b also showed a significant antioxidant potential (IC50 = 165 ± 2.8 μM) in DPPH radical scavenging assay. However, phenyl substituted pyrazole ring containing compound 5a showed no antioxidant activity. These compounds were also evaluated for their anticancer activity against PC-3 cancer cell line. Again, compound 5a (IC50 = 22.5 ± 0.1 μM) showed significant antitumor activity, while all other compounds were largely inactive.
Compounds 37 were also evaluated for their inhibitory effects against other enzymes such as carbonic anhydrase, α-chymotrypsin, xanthine oxidase, and phosphodiesterase. However, no activity was observed. The results indicate that compounds 5a and 5b have specific and signifcant biological potential and deserve to be further investigated.

3. Experimental Section

All melting points were measured on a Gallenkamp melting point apparatus in open glass capillaries and are uncorrected. IR Spectra were measured as KBr pellets on a Perkin Elmer FT 1000 spectrophotometer. The NMR spectra were recorded on a Varian Mercury Jeol-400 NMR spectrometer (400 MHz) and 13C-NMR (100 MHz) spectra were measured in deuterated dimethylsulphoxide (DMSO-d6). Chemical shifts (δ) are referred in terms of ppm and J-coupling constants are given in Hz. Abbreviations for multiplicities are as follows: s (singlet), d (doublet), t (triplet), q (quartet) and m (multiplet). Mass spectra were recorded on a Shimadzu GCMS-QP 1000 EX mass spectrometer at 70 eV. Elemental analysis was carried out on an Elementar Vario EL analyzer.

3.1. 1,1′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Diethanone (2)

A mixture of benzoylacetone 1 (16.2 g, 0.1 mol) and anhydrous potassium carbonate (25 g) in DMF (30–40 mL) was stirred vigorously at room temperature for 5 min then carbon disulfide (7.6 mL, 0.1 mol) was added with continued stirring for 30 min. The resulting mixture was cooled in ice bath, then chloroacetone (18.5 mL, 0.2 mol) was added with continued stirring for 15 min, then cooling bath subsequently removed, and the mixture was stirred for further 30 min. The solid product was collected by filtration and washed with water, dried, and the crude product was recrystallized from glacial acetic acid to obtain pale green crystals. Yield: 27.35 gm, 87 mmol, 87%; m.p. 204–206 °C; IR (νmax): 1645 (C=O) cm−1; 1H-NMR δ (ppm): 1.84 (s, 6H, CH3), 1.96 (s, 3H, CH3), 7.551–7.61 (m, 5H, C6H5); 13C-NMR δ (ppm): 14.49 (CH3), 29.37–30.55 (COCH3), 192.2 (C=O), 129.23, 129.55, 129.87, 134.79, 138.82, 141.84, 147.68 (Ar–C); MS m/z (%): 314 [M+, 70%], 299 (100), 226 (37), 184 (14); Anal. calcd. for C17H14O2S2: C, 64.94; H, 4.49; S, 20.40; Found: C, 64.95; H, 4.44; S, 20.43.

3.2. 1,1′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Bis(3-(Dimethylamino)Prop-2-En-1-One) (3)

A mixture of compound 2 (1.75 g, 5 mmol), DMF-DMA (1.19 mL, 0.01 mol) was refluxed in m-xylene (15 mL) for 10 h. After cooling, the resulting solid product was collected by filtration to obtain dark yellow crystals. Yield: 1.55 g, 3.75 mmol, 73%; m.p. 250 °C; IR (νmax): 1622 (C=O) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 2.99 (s, 12H, CH3), 4.53 (d, 1H, J = 12.0 Hz, CH), 5.38 (d, 1H, J = 12.0 Hz, CH), 7.41–7.65 (m, 5H, C6H5); 13C-NMR δ (ppm): 14.9 (−CH3), 44.79 (−N=(CH3)2), 109.8 (−CO–CH=), 153.9 (=CH–N), 180 (C=O); MS m/z (%): 424 [M+, 57%], 380(51), 336 (18), 309 (18), 98 (100); Anal. calcd. for C23H24N2O2S2: C, 65.06; H, 5.70; N, 6.60; S, 15.10; Found: C, 65.10; H, 5.68; S, 15.07.

3.3. General Procedure for the Synthesis of Compounds 4a–c

A mixture of compound 3 (0.212 g, 0.5 mmol), urea dervitives (2 equiv., 1 mmol) refluxed in dioxane (20 mL) for 4–6 h after in the presence of 0.5 mL of TEA and catalytic amount of ZnCl2. After cooling, the resulting solid products were filtered off, washed with ethanol, dried, and recrystallized from DMF/EtOH, afford the corresponding derivatives 4ac.

3.3.1. 4,4′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Dipyrimidin-2-Ol (4a)

Compound 4a was prepared from urea, following general procedure Section 3.3, as a pale yellow crystals powder. Yield: 0.14 g, 0.33 mmol, 67%; m.p. 248 °C; IR (νmax): 3444 (OH), 1624 (C=N) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 5.38 (d, 1H, J = 7.8 Hz, CH), 6.5 (s, 1H, O–H), 7.65 (d, 1H, J = 7.8 Hz, CH), 7.41–7.65 (m, 5H, C6H5); MS m/z (%): 418 [M+, 2%]; Anal. calcd. for C21H14N4O2S2: C, 60.27; H, 3.37; N, 13.39; O, 7.65; S, 15.32; Found: C, 60.24; H, 3.31; N, 13.38; S, 15.32.

3.3.2. 4,4′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Dipyrimidin-2-Thiol (4b)

Compound 4b was prepared from thiourea following general procedure Section 3.3, as a pale yellow crystals powder. Yield: 0.15 g, 0.34 mmol, 68%; m.p. 247 °C; IR (νmax): 1625 (C=N) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 5.36 (d, 1H, J = 7.8 Hz, CH), 6.5 (s, 1H, S–H), 7.41–7.65 (m, 5H, C6H5), 7.62 (d, 1H, J = 7.8 Hz, CH); MS m/z (%): 450 [M+, 2%]; Anal. calcd. for C21H14N4S4: C, 55.97; H, 3.13; N, 12.43; S, 28.46; Found: C, 55.98; H, 3.12; N, 12.41; S, 28.41.

3.3.3. 4,4′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Dipyrimidin-2-Amine (4c)

Compound 4c was prepared from guanidine following general procedure Section 3.3, as a yellow crystals powder. Yield: 0.15 g, 0.36 mmol, 72%; m.p. 246 °C; IR (νmax): 3419 (NH2), 1624 (C=N) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 4.50–4.53 (d, 2H, NH2), 5.39 (d, 1H, J = 11.7 Hz, CH), 7.41–7.52 (m, 5H, C6H5), 7.66 (d, 1H, J = 11.7 Hz, CH); 13C-NMR δ (ppm): 14.99, 19.12, 56.58, 94.12, 108, 128, 129, 130, 136, 154, 179; MS m/z (%): 416 [M+, 2%], 336 (100), 324 (47), 153 (8); Anal. calcd. for C21H16N6S2: C, 60.55; H, 3.87; N, 20.18; S, 15.40; Found: C, 60.58; H, 3.85; N, 20.15; S, 15.38.

3.4. General Procedure for the Synthesis of Compounds 5a,b

A mixture of compound 3 (1 mmol) and an excess of hydrazine derivatives (1 mL) were refluxed in EtOH (20 mL) for 6 h. After cooling, the resulting solid products were filtered off, washed with ethanol, dried, and recrystallized from MeOH, to obtain corresponding derivatives 5a,b.

3.4.1. 3,3′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Bis(1H-Pyrazole) (5a)

Compound 5a was prepared from hydrazine hydrate, following general procedure Section 3.4, as white crystals. Yield: 0.22 g, 0.62 mmol, 62%; m.p. 177 °C; IR (νmax): 3402 (NH), 1624 (C=N) cm−1; 1H-NMR δ (ppm): 1.87 (s, 3H, CH3), 6.45 (d, 1H, J = 4.5 Hz, CH), 7.53–7.40 (m, 5H, C6H5), 7.81 (d, 1H, J = 4.5 Hz, CH), 13.01 (s, 1H, NH); 13C-NMR δ (ppm): 14.03 (CH3), 103 (CH), 145 (N=CH), 128.62, 129.13, 129.96, 130.35, 130.54, 136.37, 147.33 (Ar–C); MS m/z (%): 362 [M+, 43%]; Anal. calcd. for C19H14N4S2: C, 62.96; H, 3.89; N, 15.46; S, 17.69; Found: C, 62.98; H, 3.86; N, 15.45; S, 15.72.

3.4.2. 3,3′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Bis(1-Phenyl-1H-Pyrazole) (5b)

Compound 5b was prepared from phenyl hydrazine, following general procedure Section 3.4, as brown crystals. Yield: 0.33 g, 0.64 mmol, 64%; m.p. 199 °C; IR (νmax): 1606 (C=N) cm−1; 1H-NMR δ (ppm): 1.86 (s, 3H, CH3), 6.48 (d, 1H, J = 4.5 Hz, CH), 6.53–7.20 (m, 15H, C6H5), 7.55 (d, 1H, J = 4.5 Hz, CH); 13C-NMR δ (ppm): 14.03 (CH3), 102 (CH), 143 (N=CH), 128.52, 129.23, 129.94, 130.38, 130.54, 136.38, 147.33, 152.84 (Ar–C); MS m/z (%): 514 [M+, 1%]; 169 (5), 107 (100), 92 (55), 90 (35); Anal. calcd. for C31H22N4S2: C, 72.34; H, 4.31; N, 10.89; S, 12.46; Found: C, 72.36; H, 4.29; N, 10.86; S, 12.43.

3.5. General Procedure for the Synthesis of Compounds 6 and 7

To a solution of compound 3 (212 mg, 0.5 mmol) in DMF (2 mL), substituted amine (2 equiv., 1 mmol) in EtOH (20 mL, 99.9%) was added, and then the resulting reaction mixture was heated under reflux for 7 h in the presence of a catalytic amount of ZnCl2. After cooling, the solid product was collected by filtration, washed with ethanol, dried, and recrystallized from DMF/EtOH to afford the corresponding derivatives 6 and 7.

3.5.1. 7,7′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Di-[1,2,4]Triazolo[1,5-a]Pyrimidine (6)

According to GP3, 6 was obtained from 4-amino-1,2,4-triazole (84 mg) as yellow crystals. Yield: 156 mg, 0.34 mmol, 67%; m.p. 245 °C; IR (νmax): 1624 (C=N) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 7.41–7.56 (m, 5H, C6H5), 8.17 (d, 1H, J = 8.5 Hz, CH, pyrimidyl), 8.67 (s, 1H, CH, triazole), 8.99 (d, 1H, J = 12.5 Hz, CH, pyrimidyl); 13C-NMR δ (ppm): 14.29 (−CH3), 115.38, 125, 129.11, 129.98, 130.21, 132.81, 135.8, 152.8, 154.43, 159.98, 162.88 (Ar–C); MS m/z (%): 466 [M+, 45%]; Anal. calcd. for C23H14N8S2: C, 59.21; H, 3.02; N, 24.02; S, 13.75; Found: C, 59.19; H, 3.03; N, 24.04; S, 13.71.

3.5.2. 2,2′-(3-Methyl-4-Phenylthieno[2,3-b]Thiophene-2,5-Diyl)Bis(Benzo[4,5]Imidazo[1,2-a] Pyrimidine) (7)

According to general procedure Section 3.1.5, 7 was obtained from 2-aminobenzimidazole (133 mg) as dark yellow crystals. Yield: 175 mg, 0.31 mmol, 62%; m.p. 245 °C; IR (νmax): 1622 (C=N) cm−1; 1H-NMR δ (ppm): 1.96 (s, 3H, CH3), 7.42–7.65 (m, 8H, C6H5, benzimidazole), 8.16 (d, 1H, J = 12.5 Hz, CH, pyrimidyl), 8.39 (d, 1H, J = 8.5 Hz, CH, benzimidazole), 8.80 (d, 1H, J = 12.5 Hz, CH, pyrimidyl); 13C-NMR δ (ppm): 14.8 (−CH3), 112.0, 115.1, 122.5, 127.1, 128.1, 129.9, 131.31, 135.97, 139.1, 142.0, 148.4, 156 (Ar–C); MS m/z (%): 564 [M+, 1%], 488 (1.5), 380 (38), 336 (55), 324(67), 98 (100); Anal. calcd. for C33H20N6S2: C, 70.19; H, 3.57; N, 14.88; S, 11.36; Found: C, 70.21; H, 3.54; N, 14.85; S, 11.40.

3.6. Biological Activities

Results are presented here as means ± standard error mean from triplicate (n = 3) observation. IC50 Values were determined by using EZ-FIT, enzyme kinetics software by Perrella Scientific, Inc., USA.

3.6.1. Anticancer Activity

Cytotoxic activity of compounds was evaluated in 96-well flat-bottomed microplates by using the standard MTT (3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyl-tetrazolium bromide, MP) colorimetric assay [22]. For this purpose, PC3 cells (Prostrate Cancer) were cultured in Dulbecco’s Modified Eagle Medium, supplemented with 10% of fetal bovine serum (FBS, PAA), 100 IU/mL of penicillin and 100 μg/mL of streptomycin in 75 cm2 flasks, and kept in 5% CO2 incubator at 37 °C. Exponentially growing cells were harvested, counted with a haemocytometer, and diluted with a particular medium with 5% FBS. Cell culture with the concentration of 1 × 105 cells/mL was prepared and introduced (100 μL/well) into 96-well plates. After overnight incubation, medium was removed and 200 μL of fresh medium was added with different concentrations of compounds (1–30 μM). Stock solution, 20 mM of compounds were prepared in 100% DMSO, and the final concentration of DMSO at 30 μM is 0.15%. After 48 h, 200 μL MTT (0.5 mg/mL) was added to each well and incubated further for four hours. Subsequently, 100 μL of DMSO was added to each well. The extent of MTT reduction to formazan within cells was calculated by measuring the absorbance at 570 nm, using a microplate reader (SpectraMax plus, Molecular Devices, Sunnyvale, CA, USA). The cytotoxicity was recorded as concentration causing 50% growth inhibition (IC50) for PC3 cells. The percent inhibition was calculated by using the following formula:
% Inhibition = 100 - ( ( mean of OD of test compound - mean of OD of negative control ) / ( mean of OD of positive control - mean of OD of negative control ) × 100 ) .
The results (% inhibition) were processed by using SoftMax Pro software (Molecular Device, USA).

3.6.2. In Vitro Antioxidant Activity

Test samples were allowed to react with stable free radical, 1,1-diphenyl-2-picrylhydrazyl radical (DPPH, Wako Chemicals, Shanghai, China) for half an hour at 37 °C. Various concentrations of test samples (prepared in DMSO) were incubated with DPPH (300 μM; prepared in ethanol). After incubation, decrease in absorption was measured at 515 nm by using a microplate reader (SpectraMax plus 384). Percentage radical scavenging activity (% RSA) of samples was determined in comparison with a DMSO-treated control group. Percentage of radical scavenging activity was calculated by using the formula given in statistical analysis section [23].

3.6.3. In Vitro β-Glucuronidase Inhibition Assay

β-Glucuronidase inhibitory activity was determined by the spectrophotometric method by measuring the absorbance at 405 nm of p-nitrophenol formed from the substrate p-nitrophenyl-β-d-glucuronide N1627-250 mg (Sigma Aldrich Co., 3050 spruce street, St. Louis, MO, USA). The total reaction volume was 250 μL. The compound (5 μL) was dissolved in DMSO (100%), which became 2% in the ultimate assay (250 μL) and similar conditions were used for standard (d-saccharic acid 1,4-lactone, Sigma Aldrich Co.). The reaction mixture contained 185 μL of 0.1 M acetate buffer, 5 μL of test compound solution, 10 μL of (1U) enzyme solution (G7396-25KU, Sigma Aldrich) was incubated at 37 °C for 30 min. The plates were read on a multiplate reader (SpectraMax plus 384) at 405 nm after the addition of 50 μL of 0.4 mM p-nitrophenyl-β-d-glucuronide. All assays were performed in triplicate. IC50 Values were calculated by using EZ-Fit software (Perrella Scientific Inc., Amherst, MA, USA). These values are the mean of three independent readings [24].

3.6.4. In Vitro α-Glucosidase Inhibition Assay

α-Glucosidase inhibition assay was performed spectrophotometrically. α-Glucosidase from Saccharomyces cerevisiae (G0660-750UN, Sigma Aldrich), was dissolved in phosphate buffer (pH 6.8, 50 mM). Test compounds were dissolved in 70% DMSO. In 96-well plates, 20 μL of test sample, 20 μL of enzyme and 135 μL of buffer were added and incubated for 15 min at 37 °C. After incubation, 25 μL of p-nitrophenyl-α-d-glucopyranoside (0.7 mM, Sigma Aldrich) was added and change in absorbance was monitored for 30 min at 400 nm. Test compound was replaced with DMSO (7.5% final) in control. Acarbose (Sigma Aldrich) was used as a standard inhibitor [25,26].

3.6.5. Statistical Analysis

All reactions were performed in triplicate in a final volume of 200 μL. The results were processed using SoftMax Pro 4.8 software (Molecular Devices, Sunnyvale, CA, USA) and then using MS Excel. The percentage of inhibition was calculated by the following formula:
% Inhibition = 100 - ( OD of test sample / OD of the control ) × 100

4. Conclusions

In summary, the present investigation describes an efficient method for the synthesis of novel bisheterocycles, many of which may display interesting biological activities in the field of medicinal chemistry. Compounds 4b, 5a,b showed the inhibitory effect against β-glucuronidase enzyme, which is highly expressed in cancer, rheumatoid arthritis, and AIDS. Compounds 5a and 5b also showed a potent inhibition of α-glucosidase enzyme and can serve as potential lead molecules for further investigation.

Acknowledgments

This project was supported by the King Saud University, Deanship of Scientific Research, College of Science Research Center. Thanks are also extended to Nimra Naveed Shaikh, Kulsoom Javaid, Shahida Perveen and Rizwana Malik from International Center for Chemical and Biological Sciences, University of Karachi, for the evaluation of β-glucuronidase and α-glucosidase inhibition, DPPH radical scavenging and anticancer activities, respectively of various compounds.

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Heeney, M.; Bailey, C.; Genevicius, K.; Shkunov, M.; Sparrowe, D.; Tierney, S.; Mculloch, I. Stable polythiophene semiconductors incorporating thieno[2,3-b]thiophene. J. Am. Chem. Soc 2005, 127, 1078–1079. [Google Scholar]
  2. Mashraqui, S.H.; Sangvikar, Y.S.; Meetsma, A. Synthesis and structures of thieno[2,3-b]thiophene incorporated [3,3]dithiacyclophanes. Enhanced first hyperpolarizability in an unsymmetrically polarized cyclophane. Tetrahedron Lett 2006, 47, 5599–5602. [Google Scholar]
  3. Mashraqui, S.H.; Sangvikar, Y.S.; Ashraf, M.; Kumar, S.; Daub, E. Dipyridyl/Pyridinium thieno[2,3-b]thiophenes as new atropisomeric systems. Synthesis, conformat-ional analysis and energy minimization. Tetrahedron 2005, 61, 3507–3513. [Google Scholar]
  4. Leriche, P.R.J.; Turbiez, M.M.; Monroche, V.; Allain, M.; Sauvage, F.X.; Roncali, J.; Frere, P.; Skabara, P.J. Linearly extended tetrathiafulvalene analogues with fused thiophene units as π conjugated spacers. J. Mater. Chem 2003, 13, 1324–1327. [Google Scholar]
  5. Lee, B.; Seshadri, V.; Palko, H.; Sotzing, G.A. Ring-Sulfonated poly(thienothiophene). J. Adv. Mater 2005, 17, 1792–1795. [Google Scholar]
  6. Lim, E.; Jung, B.J.; Lee, J.; Shim, H.K.; Lee, J.I.; Yang, Y.S.; Do, L.M. Thin-Film morphologies and solution-processable field-effect transistor behavior of a fluorine-thieno[3,2-b]thiophene-based conjugated copolymer. Macromolecules 2005, 38, 4531–4535. [Google Scholar]
  7. Kim, H.S.; Kim, Y.H.; Kim, T.H.; Noh, Y.Y.; Pyo, S.; Yi, M.H.; Kim, D.Y.; Kwon, S.K. Synthesis and studies on 2-hexylthieno[3,2-b]thiophene end-capped oligomers for OTFTs. Chem. Mater 2007, 19, 3561–3567. [Google Scholar]
  8. Jarak, I.; Kralj, M.; Piantanida, I.; Suman, L.; Zinic, M.; Pavelic, K.; Karminski-Zamola, G. Novel cyano- and amidino-substituted derivatives of thieno[2,3-b]- and thien-o[3,2-b]thiophene-2-carboxanilides and thieno[30,20:4,5]thieno- and thieno[20,30:4,5]thieno[2,3-c]quinolones: Synthesis, photochemical synthesis, DNA binding, and antitumor evaluation. Bioorg. Med. Chem 2006, 14, 2859–2868. [Google Scholar]
  9. Peters, D.; Hornfeldt, A.B.; Gronowitz, S. Synthesis of various 5-substituted uracils. J. Heterocycl. Chem 1990, 27, 2165–2173. [Google Scholar]
  10. Kukolja, S.; Draheim, S.E.; Graves, B.J.; Hunden, D.C.; Pfeil, J.L.; Cooper, R.D.G.; Ott, J.L.; Couter, F.T. Orally absorbable cephalosporin antibiotics. 2. Structure-Activity studies of bicyclic glycine derivatives of 7-aminodeacetoxycephalosporanic acid. J. Med. Chem 1985, 28, 1896–1903. [Google Scholar]
  11. Prugh, J.D.; Hartman, G.D.; Mallorga, P.J.; McKeever, B.M.; Michelson, S.R.; Murcko, M.A.; Schwam, H.; Smith, R.L.; Sondey, J.M.; Springer, J.P.; et al. New isomeric classes of topically active ocular hypotensive carbonic anhydrase inhibitors: 5-Substituted thieno[2,3-b]thiophene-2-sulfonamides and 5-substituted thieno[3,2-b]thiophene-2-sulfonamides. J. Med. Chem 1991, 34, 1805–1818. [Google Scholar]
  12. Egbertson, M.S.; Cook, J.J.; Bednar, B.; Prugh, J.D.; Bednar, R.A.; Gaul, S.L.; Gould, R.J.; Hartman, G.D.; Homnick, C.F.; Holahan, M.A.; et al. Centrally constrained thienothiophene α-sulfonamides are potent, long acting in vivo inhibitors of platelet aggregation. J. Med. Chem 1999, 42, 2409–2421. [Google Scholar]
  13. Allard, S.; Forster, M.; Souharce, B.; Thiem, H.; Scherf, U. Organic semiconductors for solution-processable field-effect transistors (OFETs). Angew. Chem. Int. Ed 2008, 47, 4070–4098. [Google Scholar]
  14. Wu, Z.-Q.; Ono, R.J.; Chen, Z.; Bielawski, C.W. Synthesis of poly(3-alkylthiophene)-block-poly( arylisocyanide): Two sequential, mechanistically distinct polymerizations using a single catalyst. J. Am. Chem. Soc 2010, 132, 14000–14001. [Google Scholar]
  15. Li, Z.; Ono, R.J.; Wu, Z.-Q.; Bielawski, C.W. Synthesis and self-assembly of poly(3-hexylthiophene)-block-poly(acrylic acid). Chem. Commun 2011, 47, 197–199. [Google Scholar]
  16. Wu, Z.-Q.; Ono, R.J.; Chen, Z.; Li, Z.; Bielawski, C.W. Polythiophene-block-poly(γ-benzyl l-glutamate): Synthesis and study of a new rod-rod block copolymer. Polym. Chem 2011, 2, 300–302. [Google Scholar]
  17. Sirringhaus, H.; Brown, P.J.; Friend, R.H.; Nielsen, M.M.; Bechgaard, K.; Langeveld-Voss, B.M.W.; Spiering, A.J.H.; Janssen, R.A.J.; Meijer, E.W.; Herwig, P.; et al. Two-Dimensional charge transport in self-organized, high-mobility conjugated polymers. Nature 1999, 401, 685–688. [Google Scholar]
  18. Schuett, W.; Raoort, H.S. The paralytic shellfish poison. Degradation to a pyrrolopyrimidine. J. Am. Chem. Soc 1962, 84, 2266–2267. [Google Scholar]
  19. Ho, Y.W. 5-(1-Pyrrolyl)-2-phenylthieno[2,3-d]pyrimidine as building block in heterocyclic synthesis: Novel synthesis of some pyrazoles, pyrimidines, imidazo[1,2-a]pyrimidines, pyrazolo[1,5-a]pyrimidines, pyrido-(pyrimido)pyrazolo[1,5-a]pyrimidines, 1,2,4-triazolo[1,5-a]pyrimidine and a 1,2,3,4-tetrazolo[1,5-a]pyrimidine derivative. J. Chin. Chem. Soc 2007, 54, 1075–1085. [Google Scholar]
  20. Alexander, S.K.; Smith, L. Novel one pot synthesis of polysubstituted pyrazolo[1,5-a] and imidazo[1,2-a]pyrimidines. Tetrahedron Lett 2006, 47, 2611–2614. [Google Scholar]
  21. Riyadh, S.M. Enaminones as building blocks for the synthesis of substituted pyrazoles with antitumor and antimicrobial activities. Molecules 2011, 16, 1834–1853. [Google Scholar]
  22. Mabkhot, Y.N.; Al-Majid, A.M.; Barakat, A.; Alshahrani, S.; Yamin, S. 1,1′-(3-Methyl-4-phenylthieno[2,3-b]thiophene-2,5-diyl)diethanone as a building block in heterocyclic synthesis. Novel synthesis of some pyrazole and pyrimidine derivatives. Molecules 2011, 16, 6502–6511. [Google Scholar]
  23. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Meth 1983, 65, 55–63. [Google Scholar]
  24. Khan, K.M.; Shah, Z.; Ahmad, V.U.; Khan, M.; Taha, M.; Rahim, F.; Ali, S.; Ambreen, N.; Perveen, S.; Choudhary, M.I.; Voelter, W. 2,4,6-Trichlorophenylhydrazine Schiff bases as DPPH radical and super oxide anion scavengers. Med. Chem 2012, 8, 452–461. [Google Scholar]
  25. Khan, K.M.; Rahim, F.; Halim, S.A.; Taha, M.; Khan, M.; Perveen, S.; Zaheer-ul-Haq; Mesaik, M.A.; Choudhary, M.I. Synthesis of novel inhibitors of β-glucuronidase based on benzothiazole skeleton and study of their binding affinity by molecular docking. Bioorg. Med. Chem. 2011, 19, 4286–4294. [Google Scholar]
  26. Choudhary, M.I.; Adhikari, A.; Rasheed, S.; Marasini, B.P.; Hussain, N.; Ahmad Kaleem, W.; Atta-ur-Rahman. Cyclopeptide alkaloids of Ziziphus oxyphylla Edgw as novel inhibitors of α-glucosidase enzyme and protein glycation. Phytochem. Lett. 2011, 4, 404–406. [Google Scholar]
Scheme 1. Synthesis of compounds 27.
Scheme 1. Synthesis of compounds 27.
Ijms 14 05712f1
Table 1. Results of various biological assays on compounds 37.
Table 1. Results of various biological assays on compounds 37.
CompoundsIC50 ± SEM [μM]

Anticancer Activity (PC-3 Cell line)DPPH Radical Scavenging Assayβ-Glucuronidase Inhibitionα-Glucosidase Inhibition
3>30NANANA
4a>30---
4b>30NA1.3 ± 0.2-
4c----
5a22.5 ± 0.1NA2.3 ± 0.422.0 ± 0.3
5b>30165 ± 2.88.7 ± 0.158.4 ± 1.2
6>30---
7>30-NANA
Std.DoxorubicinN-Acetylcysteined-Saccharic acidAcarbose
0.91 ± 0.1106 ± 1.11,4-lactone 45.8 ± 2.2841 ± 1.7
SEM = standard error of mean; NA = not active; failed to show 50% or more inhibition at 500 μM or more.

Share and Cite

MDPI and ACS Style

Mabkhot, Y.N.; Barakat, A.; Al-Majid, A.M.; Choudhary, M.I. Synthesis of Thieno[2,3-b]thiophene Containing Bis-Heterocycles-Novel Pharmacophores. Int. J. Mol. Sci. 2013, 14, 5712-5722. https://doi.org/10.3390/ijms14035712

AMA Style

Mabkhot YN, Barakat A, Al-Majid AM, Choudhary MI. Synthesis of Thieno[2,3-b]thiophene Containing Bis-Heterocycles-Novel Pharmacophores. International Journal of Molecular Sciences. 2013; 14(3):5712-5722. https://doi.org/10.3390/ijms14035712

Chicago/Turabian Style

Mabkhot, Yahia Nasser, Assem Barakat, Abdullah Mohammed Al-Majid, and Muhammad Iqbal Choudhary. 2013. "Synthesis of Thieno[2,3-b]thiophene Containing Bis-Heterocycles-Novel Pharmacophores" International Journal of Molecular Sciences 14, no. 3: 5712-5722. https://doi.org/10.3390/ijms14035712

Article Metrics

Back to TopTop