DOI QR코드

DOI QR Code

Synthesis and Antidiabetic Evaluation of Benzothiazole Derivatives

  • Mariappan, G. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute) ;
  • Prabhat, P. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute) ;
  • Sutharson, L. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute) ;
  • Banerjee, J. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute) ;
  • Patangia, U. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute) ;
  • Nath, S. (Department of Medicinal Chemistry, Himalayan Pharmacy Institute)
  • Received : 2012.02.18
  • Accepted : 2012.03.27
  • Published : 2012.04.20

Abstract

A novel series of benzothiazole derivatives were synthesized and assayed in vivo to investigate their hypoglycemic activity by streptozotocin-induced diebetic model in rat. These derivatives showed considerable biological efficacy when compared to glibenclamide, a potent and well known antidiabetic agent as a reference drug. All the compounds were effective, amongst them 3d showed more prominent activity at 100 mg/kg p.o. The experimental results are statistically significant at p<0.01 and p<0.05 level.

Keywords

INTRODUCTION

Benzothiazole ring system is present in various marine and terrestrial natural compounds, which have useful biological activities.1-4 2-Aminobenzothiazoles are highly reactive compounds and extensively utilized as reactants or reaction intermediates since the NH2 and endocyclic N functions are suitably situated to enable reactions with various reactants to form a variety of fused heterocyclic compounds. Medicinal chemist attention was drawn to this series when the pharmacological profile of Riluzole5 was observed as clinically available anticonvulsant drug. These derivatives are reported in the literature for the treatment of epilepsy,6-12 inflammation, 13,14 analgesia,15,16 amyotrophic lateral sclerosis,17 and viral infections.18 They also exhibit antitumour,19-33 antitubercular,34,35 antibacterial,36 antifungal,37 antimalarial, 38 antihelmintic.39 2-aryl substituted benzothiazoles have emerged in recent years as an important pharmacophore in non-invasive diagnosis of alzheimer’s disease. 40 Benzothiazole derivatives have been evaluated as potential amyloid-binding diagnostic agents in neurodegenerative disease41,42 and selective fatty acid amide hydrolase inhibitors.43 They are broadly found in bio organic and medicinal chemistry with applications in drug discovery and development for the treatment of diabetes. 44-52 Diabetes has become an increasing concern to the world’s population. In view of these literatures, it was of considerable interest to synthesize the title compound with a hope to obtain potent biologically active and safe oral anti diabetic agents.

 

RESULT AND DISCUSSION

Chemistry

A congeneric series of benzothiazole derivatives were synthesized as illustrated in Scheme 1. The starting material 2-amino-5-chloro benzothiazole 1 was prepared by the reaction of 4-chloro aniline and potassium thiocyanate in glacial acetic acid. The compound 1 was refluxed with chloroacetyl chloride in presence of potassium carbonate and chloroform to yield 2-chloroacetamido-5-chloro-benzothiazole 2. The condensation of compound 2 with various primary and secondary amines in absolute alcohol and HCl afforded the final compounds 3 (a-j). Their structures have been elucidated from UV, IR, 1H NMR, mass spectral data and elemental analysis. The physicochemical data of the synthesized compounds are given in Table 1.

Scheme 1.Synthesis of benzothiazole derivatives.

Table 1.Physical data of the synthesized compounds 3(a-j)

Spectral Analysis

A sharp singlet at 2.52-3.40 ppm ascertained the presence of CH2 proton (aliphatic) and a characteristic signal at 7.14-7.60 ppm is assigned to NH proton (benzothiazole) in all the synthesized compounds. A multiplet observed at 6.52-8.15 ppm indicated the presence of aromatic proton in all the compounds. In addition to this, two OH proton of compound 3c exhibited a sharp singlet at 2.48 ppm. The two sharp singlet signals of morpholine were observed at 2.52 and 3.55 ppm corresponding to two types of CH2 proton in compound 3e. The appearance of signals at 1.53 ppm and 2.50 ppm ascertained the presence of CH2 of piperidine. A sharp singlet at 4.06 ppm, 4.46 ppm and 4.49 ppm demonstrated the existence of NH proton (benzene) for the compounds 3f, 3g and 3j respectively. The NHproton of pyridine ring in 3h and 3i are assigned by singlet at 3.37 and 4.47 ppm respectively. Hence, the compounds synthesized were in conformity with the structures postulated.

Antidiabetic Evaluation

The LD50 values of the synthesized compounds were estimated to be in the range of 100-1000 mg/kg b.w. STZ causes diabetes by the rapid depletion of β-cells and thereby bring about a reduction of insulin release4. The results summarized in Table 2 revealed that all the synthesized compounds exhibited anti diabetic response at the end of ten-day experimental period. From Table 2, it has been found that oral administration of synthesized compounds 3c, 3d, 3e and 3j caused a more significant reduction in blood glucose than other compounds in diabetic rats. However, the compound 3d at 100 mg/kg b.w. exerted maximum glucose lowering effects whereas 3g showed minimum glucose lowering effects. The maximum glucose lowering effects of compound 3d may be due to the presence of heterocyclic amine (morpholine).

Table 2.All the values were expressed as m±S.E.M (n=6). *p<0.05 and **p<0.01

The fundamental mechanism underlying hyperglycemia in diabetes mellitus involves over-production and decreased utilization of glucose by the tissues. The plausible mechanism by which benzothiazole derivatives brought about its hypoglycemic action in diabetic rats might be by potentiating the effect of insulin in plasma by increasing either the pancreatic secretion of insulin from existing beta cells or by its release from bound form. In STZ induced diabetes, induction of diabetes with STZ is associated with characteristic loss of body weight, which is due to increased muscle wasting in diabetes.5 The present study has indicated the fact that benzothiazole derivatives have anti diabetic property and further exploitation of benzothiazole core may afford a safe anti diabetic drug.

Pharmacological Evaluation

Acute toxicity studies:

Groups of six albino mice, weighing 20-25 g were fasted overnight and treated per orally with test compounds. The dosage was varied from 100 to 1000 mg kg-1 body weight. The animals were observed for 24 h for any signs of acute toxicity such as increased or decreased motor activity, tremors, convulsion, sedation, lacrimation etc. No such signs, symptoms and mortality were observed even after 24 h. Hence the LD50 cut off value of the test compounds was fixed at 100 mg/kg. b.w and the same dose was considered for evaluation of anti diabetic activity. All the animal experiments were conducted by the approval of Institutional Animal Ethics Committee, Himalayan Pharmacy Institute, East Sikkim, India. During the study period, guidelines of Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Institutional Animal Ethics Committee (IAEC) were followed for the maintenance of animals.

Antidiabetic Activity

Induction of experimental diabetes by Streptozotocin (STZ):

The rats were injected intraperitoneally with streptozotocin dissolved in sterile normal saline at a dose of 60 mg kg-1 b.w. The animals showing blood glucose range of 200-300 mg dL-1 were used for the experiment and the hyperglycemia was confirmed after 72 hours of streptozotocin injection.

Experimental Design

Animals were divided into 12 groups of 6 animals in each (n=6). Group 1 diabetic animals received 0.5% carboxy methyl cellulose (CMC) (1 ml); Group 2 diabetic animals received glibenclamide 20 mg/kg. Groups (3-12) diabetic animals received compounds 3a-3j in a single dose of 100 mg/kg b.w p.o respectively for 7 days continuously.

Blood Glucose Measurement

Blood was withdrawn from the tail vein each time. At the end of 0, 3rd, 7th and 10th day, blood sample was withdrawn from a tail vein by snipping the tip of the tail and the blood glucose level was measured by Accu Sure Blood Glucose Monitoring System (Dr. Gene Health & Wellness).

Statistical Analysis

Values are expressed as mean ± SEM. Data were analyzed using analysis of variance and group means were compared with Tukey-Kramer Post ANOVA test. The values were considered to be significant at p<0.05 and p<0.01 level.

 

EXPERIMENTAL SECTION

All the chemicals were of synthetic grade and commercially procured from Qualigen, Mumbai, India. Melting points were determined in open capillary method and are uncorrected. IR spectra were recorded on FT-IR8400S, Fourier Transform (Shimadzu) Infrared Spectrophotometer using KBr disc method. The 1H-NMR were recorded on BRUKER ADVANCE-II 400 NMR spectrophotometer in DMSO-d6 as a solvent and TMS as an internal standard. Mass spectra were recorded on a PEP-SCIUX-APIQ pulsar (electron pre-ionisation) mass spectrometer. Elemental analyses were performed on Perkin-Elmer EAL-240 elemental analyzer.

General procedure for the synthesis of 6-chloro-1, 3-benzothiazol-2-amine 1

To a required amount of chilled glacial acetic acid, KSCN and 4-chloroaniline were added and placed in freezing mixture. The solution was stirred mechanically with dropwise addition of Br2 in glacial acetic acid at such a rate that temperature does not rise above 5 ℃. The stirring was continued for an additional 3 hr at 0-10 ℃ and the separated hydrochloride salt was filtered, washed with acetic acid and dried. It was dissolved in hot water and neutralized with aqueous ammonia solution (25%). The resulting precipitate was filtered, washed with water and recrystallized from methanol to obtain pure 6-chloro-1, 3-benzothiazol-2-amine.

Synthesis of 2-chloro-N-(6-chlorobenzothiazol-2-yl) acetamide 2

Equimolar quantity of compound 1 and chloroacetyl chloride in sufficient quantity of chloroform was refluxed in the presence of K2CO3 for about 10 hours. Excess solvent was removed in vacuum and the residue thus obtained was washed with 5% NaHCO3 and subsequently with water. The resulting crude product was dried and recrystallized from ethanol to furnish white crystal.

Synthesis of N-(6-chlorobenzothiazol-2-yl)-2-(substituted amino) acetamide 3(a-j)

To a solution of compound 2 (0.01 mol) in 25 ml of absolute alcohol was added different secondary and primary amine (0.01 mol). The mixture was refluxed on water bath for 4-6 hours and the completion of reaction was checked by TLC. The crude product thus obtained was filtered, dried and recrystallized from aqueous alcohol.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(dimethylamino) acetamide (3a):

Colorless crystal; UV (nm) 225; IR (KBr, cm-1) 3460 (N-H), 3063 (C-H str of CH2), 1531 (C=O), 1444(C=N); 1HNMR (400Hz, DMSO-d6): δ 7.61(s, 1H, NH), δ 2.50 (s, 2H, CH2), δ 3.37(s, 6H, (CH3)2), δ 7.19-7.76 (m, 3H, Ar-H); Mass (m/z) 269; Ana. Calcd. C11H12ClN3OS C, 48.98; H, 4.48; N, 15.58, found C, 49.38; H, 4.58; N, 15. 48%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(diethylamino) acetamide (3b):

Colorless crystal; UV (nm) 225; IR (KBr, cm-1) 3456 (N-H), 3091 (CH str of CH2), 1637 (C=O), 1533(C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.61 (s, IH, NH), δ 2.50 (s, 4H, (CH2)2), δ 3.38(s, 6H, (CH3)2) 7.19-7.76 (m, 3H, Ar-H); Mass (m/z) 298; Ana. Calcd. C13H16ClN3OS C, 52.43; H, 5.42; N, 14.11, found C, 52.83; H, 5.12; N, 14.51%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(bis (2-hydroxyethyl) amino) acetamide (3c):

Colorless crystal; UV (nm) 226; IR (KBr, cm-1) 3458 (O-H), 3271(N-H), 3093 (C-H str of CH2), 1637 (C=O), 1533(C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.60 (s, 1H, NH), δ 3.39 (s, 2H, CH2), δ 2.49 (s, 8H, 2×(CH2)2), δ 2.48 (s, 2H, 2×OH), δ 7.17-7.74 (m, 3H, Ar-H); Mass (m/z) 329; Ana. Calcd. C13H16ClN3O3S C, 47.34; H, 4.89; N, 12.74; found C, 47.74; H, 4.99; N, 12.64%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-morpholinoacetamide (3d):

Colorless crystal; UV (nm) 215; IR (KBr,cm-1) 3329 (N-H), 3080 (C-H str of CH2),1695 (C=O), 1599 (C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.14(s, 1H, NH), δ 3.62 (s, 2H, CH2), δ 2.52 (s, 4H, 2×CH2 of morpholine), δ 3.55 (s, 4H, 2×CH2 of morpholine), δ 7.42-8.12 (m, 3H, Ar-H); Mass (m/z) 312; Ana. Calcd. C13H14ClN3O2S C, 50.08; H, 4.53; N, 13.48; found C, 50.48; H, 4.83; N, 13.28%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(piperidin-1-yl) acetamide (3e):

Pale yellow powder; UV (nm) 224; IR (KBr, cm-1) 3460 (N-H), 3265 (C-H str of CH2), 1639 (C=O), 1433 (C=N), 1H NMR (400 Hz, DMSO-d6): δ 7.60 (s,1H, NH), δ 3.35 (s, 2H, CH2), δ 1.53 (s, 8H, 4×CH2 of piperidine), δ 2.50 (s, 2H, CH2 of piperidine), δ 7.19-7.77 (m, 3H, Ar-H); Mass (m/z) 309; Ana. Calcd. C14H16ClN3OS C, 54.27; H, 5.21; N, 13.56; found C, 54.67; H, 4.81; N, 13.76%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-fluorophenylamino) acetamide (3f):

Colorless powder; UV (nm) 218; IR (KBr, cm-1) 3365 (N-H), 3178 (C-H str of CH2), 1600 (C=O), 1521 (C=N); 1H NMR (400Hz, DMSO-d6): δ 8.13 (s, 1H, NH), δ 3.36 (s, 2H, CH2), 4.06 (s, 1H, Ar-NH), δ 6.55-8.13 (m, 3H, Ar-H), δ 7.73-7.43 (m, 4H, Ar-H); Mass (m/z) 336; Ana. Calcd. C15H11ClFN3OS C, 53.65; H, 3.30; N, 12.51; found C, 53.25; H, 3.6; N, 12.91%.

N-(6-chlorobenzo[d]thiazol-2-yl-2-(3-chlorophenylamino) acetamide (3g):

Colorless powder; UV (nm) 282; IR (KBr, cm-1), 3379 (N-H), 3176 (C-H str of CH2), 1668 (C=O), 1541 (C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.73 (s, 1H, NH), δ 4.04 (s, 2H, CH2), δ 4.46 (s, IH, Ar-NH), δ 6.52-8.14 (m, 7H, Ar-H); Mass (m/z) 352; Ana. Cald. C15H11Cl2N3OS C, 51.15; H, 3.15; N, 11.93; found C, 51.05; H, 3.45; N, 11.73%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(pyridin-4-ylamino) acetamide (3h):

Colorless crystal; UV (nm) 224; IR (KBr, cm-1), 3456 (N-H), 3269 (C-H str of CH2), 1637 (C=O), 1533 (C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.61 (s, 1H, NH), δ 2.51 (s, 2H, CH2), δ 3.37 (s, 1H, pyridine-NH), δ 7.19-7.22 (m, 3H, Ar-H), δ 7.28-7.77 (m, 4H, Het. Ar-H); Mass (m/z) 318; Ana. Cald. C14H11ClN4OS C, 52.75; H, 3.48; N, 17.58; found C, 51.43; H, 3.52; N, 17.43%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(pyridin-2-ylamino) acetamide (3i):

Brick red powder; UV (nm) 224; IR (KBr, cm-1), 3454 (N-H), 3416 (C-H str of CH2), 1699 (C=O), 1535 (C=N); 1H NMR (400 Hz, DMSO-d6): δ 7.60 (s, 1H, NH), δ 3.36 (s, 2H, CH2), δ 4.47 (s, 1H, pyridine-NH), δ 7.19-7.31 (m, 3H, Ar-H), δ 7.46-8.15 (m, 4H, Het. Ar-H); Mass (m/z) 318; Ana. Cald C14H11ClN4OS C, 52.75; H, 3.48; N, 17.58; found C, 52.43; H, 3.48; N, 17.43%.

N-(6-chlorobenzo[d]thiazol-2-yl)-2-(4-sulfanilido) acetamide (3j):

Colorless crystal; UV (nm) 281; IR (KBr, cm-1), 3180 (N-H), 2993 (C-H str of CH2), 1668 (C=O), 1541 (C=N); 1H NMR (400 Hz, DMSO-d6): δ 8.15 (s, 1H, NH), δ 3.38 (s, 2H, CH2), δ 4.49 (s, 1H, NH), δ 7.46 (m, 2H, SO2NH2), δ7.75-8.15 (m, 7H, Ar-H); Mass (m/z) 397; Ana. Cald C15H13ClN4O3S2 C, 45.40; H, 3.30; N, 14.12; found C, 45.43; H, 3.44; N, 14.22%.

References

  1. Geewananda, G. P.; Shigeo, K.; Sarath, P. G.; Oliver, J. M.; Frank, E. K. J. Am. Chem. Soc. 1988, 110(14), 4856. https://doi.org/10.1021/ja00222a071
  2. Geewananda, G. P.; Shigeo, K.; Neal, S.B. Tetrahedron Lett. 1989, 30, 4359. https://doi.org/10.1016/S0040-4039(00)99360-2
  3. Gunawardana, G. P.; Koehn, F. E.; Lee, A. Y.; Clardy, J.; He, H. Y.; Faulkenr, J. D. J. Org. Chem. 1992, 57(5), 523.
  4. Carroll, A. R.; Scheuer, P. J. J. Org. Chem. 1990, 55(14), 4426. https://doi.org/10.1021/jo00301a040
  5. Bryson, M.; Fulton, B.; Benfield, P. Drugs. 1996, 52, 549. https://doi.org/10.2165/00003495-199652040-00010
  6. Chopade, R. S.; Bahekar, R. H.; Khedekar, P. B.; Bhusari, K. P.; Rao, A.R.R. Arch. Pharm. Pharm. Med. Chem. 2002, 8, 381.
  7. Yogeeswari, P.; Srisam, D.; Suniljit, L.; Kumar, S.; Stables, J. Eur. J. Med. Chem. 2002, 37, 231. https://doi.org/10.1016/S0223-5234(02)01338-7
  8. Yogeeswari, P.; Sriram, D.; Mehta, S.; Nigam, D.; Kumar, M.; Murugesan, S. J. Stables II, Farmaco. 2005, 60, 1. https://doi.org/10.1016/j.farmac.2004.09.001
  9. Siddiqui, N.; Pandeya, S.; Khan, S.; Stables, J.; Rana, A.; Alam, M.; Arshad, M.; Bhat, M. Bioorg. Med. Chem. Lett. 2007, 17, 255. https://doi.org/10.1016/j.bmcl.2006.09.053
  10. Siddiqui, N.; Rana, A.; Khan, S.; Bhat, M.; Haque, S. Bioorg. Med. Chem. Lett. 2007, 17, 4178. https://doi.org/10.1016/j.bmcl.2007.05.048
  11. Hays, S. J.; Rice, M. J.; Ortwine, D. F.; Johnson, G.; Schwartz, R. D.; Boyd, D. K.; opeland, L. F.; Vartanian, M. G.; Boxer, P. A. J. Pharm. Sci. 1994, 83, 1425. https://doi.org/10.1002/jps.2600831013
  12. He, Y.; Benz, A.; Fu, T.; Wang, M.; Covey, D. F.; Zorumski, C. F.; Mennick, S. Neuropharmacology 2002, 42, 199. https://doi.org/10.1016/S0028-3908(01)00175-7
  13. Gurupadayya, B. M.; Gopal, M.; Padmashali, B.; Vaidya, V. P. Int. J. Heterocyclic Chem. 2005, 15, 169.
  14. Sawhney, S. N.; Arora, S. K.; Singh, J. V.; Bansal, O. P.; Singh, S. P. Indian J. Chem. 1978, 16B, 605.
  15. Foscolos, G.; Tsatsas, G.; Champagnac, A.; Pommier, M. Ann. Pharm. Fr. 1977, 35, 295.
  16. Siddiqui, N.; Alam, M.; Siddiqui, A. A. Asian. J. Chem. 2004, 16, 1005.
  17. Bensimon, G.; Lacomblez, L.; Meininger, V. New Engl. J. Med. 1994, 330, 585. https://doi.org/10.1056/NEJM199403033300901
  18. Bensimon, G.; Lacomblez, L.; Meininger, V. New Engl. J. Med. 1994, 330, 585. https://doi.org/10.1056/NEJM199403033300901
  19. Paget, C. J.; Kisner, K.; Stone, R. L.; Delong, D. C. J. Med. Chem. 1969, 12, 1016. https://doi.org/10.1021/jm00306a011
  20. Vicini, P.; Gernonikaki, A.; Incerti, M.; Busonera, B.; Poni, G.; Cabras, C. A.; Colla, P. L. Bioorg. Med. Chem. 2003, 11, 4785. https://doi.org/10.1016/S0968-0896(03)00493-0
  21. Caleta, I.; Kralj, M.; Branimir Bertosa, B.; Sanja Tomic, S.; Pavlovic, G.; Pavelic, K.; Karminski-Zamola, G. J. Med. Chem. 2009, 52, 1744. https://doi.org/10.1021/jm801566q
  22. Chung, Y.; Shin, Y. K.; Zhan, C. G.; Lee, S.; Cho, H. Arch. Pharmacol. Res. 2004, 27, 893. https://doi.org/10.1007/BF02975839
  23. Yoshida, M.; Hayakawa, I.; Hayashi, N.; Agatsuma, T.; Oda, Y.; Tanzawa, F.; Iwasaki, S.; Koyama, K.; Furukawa, H.; Kurakata, S. Bioorg. Med. Chem. Lett. 2005, 15, 3328. https://doi.org/10.1016/j.bmcl.2005.05.077
  24. Bradshaw, T. D.; Stevens, M. F. G.; Westwell, A. D. Curr. Med. Chem. 2001, 8, 203. https://doi.org/10.2174/0929867013373714
  25. Chua, M. S.; Shi, D. F.; Wrigley, S.; Bradshaw, T. D.; Hutchinson, I.; Shaw, P. N.; Barrett, D. A.; Stanley, L. A.; Stevens, M. F. G. J. Med. Chem. 1999, 42, 381. https://doi.org/10.1021/jm981076x
  26. O'Brien, S. E.; Browne, H. L.; Bradshaw, T. D.; Westwell, A. D.; Stevens, M. F. G.; Laughton, C. A. Org. Biomol. Chem. 2003, 1, 493. https://doi.org/10.1039/b209067h
  27. Bradshaw, T. D.; Wrigley, S.; Shi, D. F.; Schulz, R. J.; Paull, K. D.; Stevens, M. F. G. Br. J. Cancer 1998, 77, 745. https://doi.org/10.1038/bjc.1998.122
  28. Kashiyama, E.; Hutchinson, I.; Chua, M. S.; Stinson, S. F.; Phillips, L. R.; Kaur, G.; Sausville, E. A.; Bradshaw, T. D.; Westwell, A. D.; Stevens, M. F. G. J. Med. Chem. 1999, 42, 4172. https://doi.org/10.1021/jm990104o
  29. Hutchinson, I.; Chua, M. S.; Browne, H. L.; Trapani, V.; Bradshaw, T. D.; Westwell, A. D.; Stevens, M. F. G. J. Med. Chem. 2001, 44, 1446. https://doi.org/10.1021/jm001104n
  30. Shi, D. F.; Bradshaw, T. D.; Wrigley, S.; McCall, C. J.; Lelieveld, P.; Stevens, M. F. G. J. Med. Chem. 1996, 39, 3375. https://doi.org/10.1021/jm9600959
  31. Lion, C. J.; Matthews, C. S.; Wells, G.; Bradshaw, T. D.; Stevens, M. F. G.; Westwell, A. D. Bioorg. Med. Chem. Lett. 2006, 16, 5005. https://doi.org/10.1016/j.bmcl.2006.07.072
  32. Mortimer, C. S.; Wells, G.; Crochard, P. J.; Stone, E. L.; Bradshaw, T. D.; Stevens, A. D.; Westwell, M. F. G. J. Med. Chem. 2006, 49, 179. https://doi.org/10.1021/jm050942k
  33. Wells, G.; Berry, J. M.; Bradshaw, T. D.; Burger, A. M.; Seaton, A.; Wang, B.; Westwell, A. D.; Stevens, M. F. G. J. Med. Chem. 2003, 46, 532. https://doi.org/10.1021/jm020984y
  34. Hutchinson, I.; Jennings, S. A.; Vishnuvajjala, B. R.; Westwell, A. D.; Stevens, M. F. G. J. Med. Chem. 2002, 45, 744. https://doi.org/10.1021/jm011025r
  35. Khadse, B. G.; Sengupta, S. R. Indian J. Chem. 1993, Sec-B, 407.
  36. Palmer, F. J.; Trigg, R. B.; Warrington, J. V. J. Med. Chem. 1971, 14, 248. https://doi.org/10.1021/jm00285a022
  37. Gurupadaiah, B. M.; Jayachandran, E.; ShivaKumar, B.; Nagappa, A. N.; Nargund, L. V. G. Indian J. Heterocycl. Chem. 1998, 7, 213.
  38. Gopkumar, P.; Shivakumar, B.; Jayachandran, E.; Nagappa, A. N.; Nargund, L. V. G.; Gurupadaiah, B. M. Indian J. Heterocycl. Chem. 2001, 11, 39.
  39. Burger, A.; Sawhey, S. N. J. Med. Chem. 1968, 11, 270. https://doi.org/10.1021/jm00308a018
  40. Jayachandran, E.; Bhatia, K.; Naragud, L. V. G.; Roy, A. Indian Drugs 2003, 40, 408.
  41. Weekes, A. A.; Westwell, A. D. Curr. Med. Chem. 2009, 16, 2430. https://doi.org/10.2174/092986709788682137
  42. Henriksen, G.; Hauser, A. I.; Westwell, A. D.; Yousefi, B. H.; Schwaiger, M.; Drzega, A.; Wester, H. J. J. Med. Chem. 2007, 50, 1087. https://doi.org/10.1021/jm061466g
  43. Mathis, C. A.; Wang, Y.; Holt, D. P.; Haung, G. F.; Debnath, M. L.; Klunk, W. E. J. Med. Chem. 2003, 46, 2740. https://doi.org/10.1021/jm030026b
  44. Wang, X.; Sarris, K.; Zhang, K. Kage, D.; Brown, S. P.; Kolasa, T.; Surowy, C.; ElKouhen, O. F.; Muchmore, S. W.; Brioni, J. D.; Stewart, A. O. J. Med. Chem. 2009, 52, 170. https://doi.org/10.1021/jm801042a
  45. Suter, H.; Zutter, H. Helv. Chim. Acta 1967, 50, 1084. https://doi.org/10.1002/hlca.19670500415
  46. Diaz, H. M.; Molina, R. V.; Andrade, R. O.; Coutino, D. D.; Franco, L. M.; Webster, S. P.; Binnie, M.; Soto, S. E.; Barajas, M. I.; Rivera, I. L.; Vazquez, G. N. Bioorg. Med. Chem. Lett. 2008, 18, 2871. https://doi.org/10.1016/j.bmcl.2008.03.086
  47. Nitta, A.; Fujii, H.; Sakami, S.; Nishimura, Y.; Ohyama, T.; Satoh, M.; Nakaki, J.; Satoh, S.; Inada, C.; Kozono, H.; Kumagai, H.; Shimamura, M.; Fukazawa, T.; Kawai, H. Bioorg. Med. Chem. Lett. 2008, 15, 5435.
  48. Vazquez, G. N.; Paoli, P.; Rivera, I. L.; Molina, R. V.; Franco, J. L. M.; Andrade, R. O.; Soto, S. E.; Camici, G.; Coutino, D. D.; Ortiz, I. G.; Mayorga, K. M.; Díaz, H. M. Bioorg. Med. Chem. Lett. 2009, 17, 3332. https://doi.org/10.1016/j.bmc.2009.03.042
  49. Su, X.; Vicker, N.; Ganeshapillai, D.; Smith, A.; Purohit, A.; Reed, M. J.; Potter, B. V. Mol. Cell Endocrinol. 2006, 248, 214. https://doi.org/10.1016/j.mce.2005.10.022
  50. Barf, T.; Vallgarda, J.; Emond, R.; Haggstrom, C.; Kurz, G.; Nygren, A.; Larwood, V.; Mosialou, E.; Axelsson, K.; Olsson, R.; Engblom, L.; Edling, N.; Ronquist-Nii, Y.; Ohman, B.; Alberts, P.; Abrahmsen, L. J. Med. Chem. 2002, 45, 3813. https://doi.org/10.1021/jm025530f
  51. Fujieda, H.; Usui, S.; Suzuki, T.; Nakagawa, H.; Ogura, M.; Makishima, M.; Miyata, N. Bioorg. Med. Chem. Lett. 2007, 17, 4351. https://doi.org/10.1016/j.bmcl.2007.05.017
  52. Jeon, R.; Kim, Y. J.; Cheon, Y.; Ryu, J. H. Arch. Pharmacal Res. 2006, 29, 394. https://doi.org/10.1007/BF02968589
  53. Pattan, S. R.; Suresh, C.; Pujar, V. D.; Reddy, V. V. K.; Rasal, V. P.; Koti, B. C. Indian. J. Chem. 2005, 44B, 2404.

Cited by

  1. Antidiabetic potential and enzyme kinetics of benzothiazole derivatives and their non-bonded interactions with α-glucosidase and α-amylase vol.25, pp.4, 2016, https://doi.org/10.1007/s00044-016-1520-3
  2. Synthesis, crystal structure and antidiabetic activity of substituted (E)-3-(Benzo [d]thiazol-2-ylamino) phenylprop-2-en-1-one vol.59, 2013, https://doi.org/10.1016/j.ejmech.2012.11.020
  3. Visible-light-promoted synthesis of benzothiazoles from 2-aminothiophenols and aldehydes vol.58, pp.9, 2017, https://doi.org/10.1016/j.tetlet.2017.01.053
  4. A comprehensive review in current developments of benzothiazole-based molecules in medicinal chemistry vol.89, 2015, https://doi.org/10.1016/j.ejmech.2014.10.059
  5. Green and Highly Efficient Synthesis of Mono- and Bis-Benzothiazoles in the Presence of Fe(SD)3under Ultrasound Irradiation vol.64, pp.1, 2017, https://doi.org/10.1002/jccs.201600200
  6. Enhanced cytotoxicity by a benzothiazole-containing cisplatin derivative in breast cancer cells vol.41, pp.2, 2017, https://doi.org/10.1039/C6NJ02753A
  7. Synthesis and blood glucose and lipid-lowering effects of benzothiazole-substituted benzenesulfonylurea derivatives vol.146, pp.12, 2015, https://doi.org/10.1007/s00706-015-1471-2
  8. Novel cycloalkylthiophene–imine derivatives bearing benzothiazole scaffold: Synthesis, characterization and antiviral activity evaluation vol.23, pp.18, 2013, https://doi.org/10.1016/j.bmcl.2013.07.023
  9. Synthesis and Biological Activity of New 1,3-Benzothiazole Derivatives vol.48, pp.8, 2014, https://doi.org/10.1007/s11094-014-1139-y
  10. Efficiently functionalized oxacalix[4]arenes: Synthesis, characterization and exploration of their biological profile as novel HDAC inhibitors vol.26, pp.3, 2016, https://doi.org/10.1016/j.bmcl.2015.12.044
  11. Ruthenium Catalyzed Intramolecular C–S Coupling Reactions: Synthetic Scope and Mechanistic Insight vol.18, pp.3, 2016, https://doi.org/10.1021/acs.orglett.5b03185
  12. Recent Advances in the Chemistry and Biology of Benzothiazoles vol.348, pp.3, 2015, https://doi.org/10.1002/ardp.201400340
  13. Synthesis and antimicrobial activity of pyrimidinyl bis(benzazoles) vol.26, pp.2, 2017, https://doi.org/10.1007/s00044-016-1758-9
  14. Novel benzothiazole based sulfonylureas/sulfonylthioureas: design, synthesis and evaluation of their antidiabetic potential vol.40, pp.8, 2016, https://doi.org/10.1039/C5NJ03589A
  15. Contemporary progress in the synthesis and reactions of 2-aminobenzothiazole: a review vol.42, pp.2, 2018, https://doi.org/10.1039/C7NJ03776G
  16. Design, Synthesis and Anticancer Screening of Novel Benzothiazole-Piperazine-1,2,3-Triazole Hybrids vol.23, pp.11, 2018, https://doi.org/10.3390/molecules23112788
  17. A modern and practical laccase-catalysed route suitable for the synthesis of 2-arylbenzimidazoles and 2-arylbenzothiazoles vol.8, pp.69, 2012, https://doi.org/10.1039/c8ra07377e
  18. Facile Synthesis of a Series of Non-Symmetric Thioethers Including a Benzothiazole Moiety and Their Use as Efficient In Vitro anti-Trypanosoma cruzi Agents vol.24, pp.17, 2012, https://doi.org/10.3390/molecules24173077
  19. 1‐(Hydroxymethyl)‐1 H ‐benzotriazole: An Efficient Ligand for Copper‐Catalyzed Ullmann‐Type Coupling Reaction Leading to Expeditious Synthesis of Diverse Benzoxazole vol.4, pp.33, 2012, https://doi.org/10.1002/slct.201902420
  20. An Exploration about the Interaction of Mild Steel with Hydrochloric Acid in the Presence of N-(Benzo[d]thiazole-2-yl)-1-phenylethan-1-imines vol.123, pp.37, 2012, https://doi.org/10.1021/acs.jpcc.9b03994
  21. Isothioureas, Ureas, and Their N-Methyl Amides from 2-Aminobenzothiazole and Chiral Amino Acids vol.24, pp.18, 2012, https://doi.org/10.3390/molecules24183391
  22. Design, Synthesis and Antimicrobial Study of Novel 1-(1,3-benzothiazol-2- yl)-3-chloro-4H-spiro[azetidine-2,3'-indole]-2',4(1'H)-diones Through Ketene- imine Cycloaddition Reaction vol.17, pp.2, 2012, https://doi.org/10.2174/1570178616666190705153224
  23. Green microwave versus conventional synthesis, crystal structure of 1-(4-(Benzothiazol-2-yl)piperazin-1-yl)-2-(4-phenyl-1H-1,2,3-triazol-1-yl)ethenone and HS-Analysis vol.14, pp.1, 2020, https://doi.org/10.1080/16583655.2020.1751977
  24. Molecular docking studies of N-(benzo[d]thiazol-2-ylcarbamothioyl)-2/4-substituted benzamides as an anti-bacterial inhibitor for E. coli dihydroorotase vol.1913, pp.1, 2021, https://doi.org/10.1088/1742-6596/1913/1/012082