Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter July 18, 2016

Design, synthesis and cytotoxicity evaluation of novel (E)-3-(3-aryl-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-ones as anticancer agents

  • Raquib Alam , Md. Aftab Alam , Amulya K. Panda and Rahis Uddin EMAIL logo

Abstract

(E)-3-(3-Aryl-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-ones 4a–i have been synthesized and evaluated for their in vitro cytotoxicity against a panel of three human cancer cell lines Caco-2, MIA PaCa-2, MCF-7 and a normal NIH-3T3 cell line. Compound 4g is cytotoxic with the IC50 value of 15.32±0.62 μm against the Caco-2 cell line.

Although there has been considerable progress in reducing cancer incidence in the United States, the number of cancer patients continues to increase [1]. Chemotherapy is one of the most effective approaches used for treating cancer patients. However, the lack of selectivity and development of drug-resistance reduces the efficacy of cancer chemotherapy [2]. Therefore development of effective and safe anticancer agents with high potency and less toxicity is a major focus for researchers across the world. The heterocyclic compounds containing a pyrazole ring have received considerable attention owing to their diverse chemotherapeutic potential [3], [4], [5]. Important pyrazole-based antitumor drugs available in the market include ruxolitinib and crizotinib [6]. Celecoxib is a typical model of pyrazole-based diaryl heterocyclic small molecule [7] with antitumor activity against prostate tumors in experimental models [8], [9], [10]. Chalcones show anti-cancer activity [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28] apparently due to their inhibition of tubulin [15], thioredoxin reductase [17], VEGF [18], mTOR [19] topoisomerase-I/II [20], 5α-reductase [21], sirtuin-1 [22], JAK/STAT signaling pathways [23], MMP-2 [24], cathepsin-K [25], Wnt [26], B-Raf [27] and NF-κB [28], among others.

On the basis of the interesting biological activity profiles of pyrazoles and chalcones, we were inspired to synthesize some pyrazolic chalcones as potential anticancer agents. Synthesis is outlined in Scheme 1. Pyrazolic chalcones were prepared from the corresponding 3-aryl-1-phenylpyrazol-4-carboxaldehydes 3a–i [29], [30], [31], [32], [33] which, in turn, were synthesized from the heterocyclic substrates 2a–i [30] (see Supplementary Material). The Claisen-Schmidt condensation of compounds 3a–i with 3-acetylpyridine in methanolic NaOH afforded the desired (E)-3-(3-(aryl)-1-phenyl-1H-pyrazol-4-yl)-pyridin-3-yl)prop-2-en-1-ones 4a–i. Compounds 3a–i and 4a–i were characterized by spectral methods and elemental analysis.

Scheme 1 Conditions: (i) EtOH, H2SO4, reflux; (ii) POCl3/DMF, 80°C, then NaHCO3/H2O; (iii) 3-acetylpyridine, MeOH, NaOH, r.t.
Scheme 1

Conditions: (i) EtOH, H2SO4, reflux; (ii) POCl3/DMF, 80°C, then NaHCO3/H2O; (iii) 3-acetylpyridine, MeOH, NaOH, r.t.

In vitro cytotoxicity of compounds 4ai was measured by an MTT [(3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide)] assay against a panel of three different human cancer cell lines, namely Caco-2 (human intestinal), MIA PaCa-2 (human pancreatic) and MCF-7 (human breast) and one normal cell line (mouse embryo fibroblasts NIH-3T3) [34]. Etoposide was taken as the reference drug and the results are summarized in terms of IC50 values (Table 1). Most of the compounds show moderate to good cytotoxicity against intestinal, pancreatic and breast cancer cell lines and very weak toxicity towards NIH-3T3 normal cell line. Analogs 4fh show significant cytotoxicity as compared to standard drug etoposide. Out of 9 pyrazolic chalcones synthesized, compound 4g displays the most potent cytotoxicity against Caco-2 cell line. SAR analysis of these pyrazolic chalcones indicate that compounds with a para substituted NO2 group in the benzene ring are more cytotoxic. Compounds with F, Cl, Br, CH3 or OCH3 substituent show more moderate effects, which is clearly seen from Table 1. The cytotoxic activity against all tested cancer cell lines shows that the strength order is NO2>F>Br>Cl for compounds with a para substituted electron-withdrawing group present in the benzene ring. Among compounds with meta substituted electron-withdrawing group, the cytotoxic activity order is NO2>Br. Finally, cytotoxic activity against Caco-2 and MIA PaCa-2 shows that the strength order is CH3>OCH3 for compounds with electron-donating substituent present in para position of the benzene ring. Furthermore, cytotoxic activity against MCF-7 shows the strength order is OCH3>CH3 for compounds with para electron-donating substituent.

Table 1

In vitro cytotoxicity evaluation of the synthesized compounds against a panel of human cancer cell lines and a normal cell line in terms of IC50 value in μm.

CompoundsRCaco-2MIA PaCa-2MCF-7NIH-3T3
4aH32.30±0.6538.52±2.3156.65±2.75>100
4b4-F23.09±0.9328.28±2.1734.38±3.8391.46±2.99
4c4-Cl38.93±2.4946.57±2.3054.53±1.1092.58±2.52
4d3-Br30.28±2.0124.80±1.4646.67±2.0196.56±1.55
4e4-Br27.95±0.2930.40±0.8439.79±1.0796.13±2.09
4f3-NO219.62±0.7619.70±0.3229.78±0.8487.58±1.96
4g4-NO215.32±0.6218.89±0.4429.59±2.1887.39±0.44
4h4-CH337.83±2.8521.94±5.1068.78±1.16>100
4i4-OCH349.12±8.3732.05±1.2344.36±1.08>100
Etoposide17.51±0.2423.66±0.3332.31±1.4890.53±4.6

Experimental

All starting materials and solvents were purchased from commercial sources and used without further purification. Melting points were determined in open capillaries using an electro-thermal melting point apparatus and are uncorrected. The progress of the reactions was monitored by TLC using precoated aluminum sheets (Silica gel 60 F254, Merck) and spots were visualized under UV light. IR spectra were recorded on an Agilent Cary 630 FT-IR spectrometer. The 1H NMR (400 MHz) and 13C NMR (100 MHz) spectra were recorded on a Bruker Avance 400 spectrometer using CDCl3 or DMSO-d6 as a solvent and TMS as an internal standard. Elemental analysis were performed on an Elementar Vario analyzer. Mass spectra (ESI-MS) were recorded on an AB-Sciex 2000 spectrometer.

General synthesis of compounds 4a–i

A mixture of 3-aryl-1-phenylpyrazol-4-carboxaldehyde 3a–i (10 mmol) and 3-acetylpyridine (10 mmol) in methanolic solution of sodium hydroxide was stirred for 24 h at room temperature. The resultant precipitate of 4a–i was filtered off, washed with water, dried, and crystallized from ethanol (4a–e and 4h,i) or N,N-dimethylformamide (4f,g).

(E)-3-(1,3-Diphenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4a)

Yellow solid; yield 64%; mp 168–170°C; IR (neat, νmax): 3132, 1665, 1585, 1415 cm-1; 1H NMR (CDCl3): δ 9.13 (s, 1H), 8.73–8.75 (m, 1H), 8.37(s, 1H), 8.21 (m, 1H), 7.91 (d, J = 15.6 Hz, 1H), 7.26–7.78 (m, 12H); 13C NMR (CDCl3): δ 188.5, 154.0, 153.0, 149.6, 139.3, 136.5, 135.7, 133.5, 132.1, 129.6, 128.9, 128.8, 128.8, 127.3, 127.2, 123.6, 120.5, 119.3, 117.9. ESI-MS. Calcd for C23H17N3O, [M + H]+: m/z 352.1. Found: m/z 352.2. Anal. Calcd for C23H17N3O: C, 78.61; H, 4.55; N, 11.96. Found: C, 78.33; H, 4.84; N, 11.94.

(E)-3-(3-(4-Fluorophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4b)

Yellow solid; yield: 70%; mp 170–172°C; IR (neat, νmax): 3119, 1667, 1596, 1525, 1419 cm-1; 1H NMR (CDCl3): δ 9.20 (s, 1H), 8.80 (d, J = 3.6 Hz, 1H), 8.40 (s, 1H), 8.28 (d, J = 8 Hz, 1H), 7.91 (d, J = 15.6 Hz, 1H), 7.80 (d, J = 8 Hz, 2H), 7.33–7.70 (m, 7H), 7.20 (t, J = 8.4 Hz, 2H); 13C NMR (CDCl3): δ 188.4, 164.4, 162.0, 153.1, 149.6, 139.2, 136.2, 135.8, 133.4, 130.6, 130.5, 129.6, 128.2, 127.5, 127.1, 123.7, 120.6, 119.4, 117.9, 116.0, 115.8. ESI-MS. Calcd for C23H16FN3O, [M + H]+: m/z 370.1. Found: m/z 370.5. Anal. Calcd for C23H16FN3O: 74.78; H, 4.37; N, 11.38. Found: C, 74.73; H, 4.39; N, 11.41.

(E)-3-(3-(4-Chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4c)

Yellow solid; yield 64%; mp 162–164°C; IR (neat, νmax): 3134, 1665, 1583, 1501, 1404 cm-1; 1H NMR (CDCl3): δ 9.16 (s, 1H), 8.76 (d, J = 4.4 Hz, 1H), 8.37 (s, 1H), 8.21 (d, J = 8 Hz, 1H), 7.85 (d, J = 15.2 Hz, 1H), 7.76 (d, J = 8 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.29–7.50 (m, 7H); 13C NMR (CDCl3): δ 188.8, 153.1, 152.8, 149.6, 139.2, 136.0, 135.7, 134.9, 133.4, 130.6, 130.0, 129.6, 129.0, 127.5, 127.1, 123.7, 120.8, 119.4, 117.9. ESI-MS. Calcd for C23H16ClN3O, [M + H]+: m/z 386.1. Found: m/z 386.4. Anal. Calcd for C23H16ClN3O: C, 71.59; H, 4.18; N, 10.89. Found: C, 71.65; H, 4.16; N, 10.85.

(E)-3-(3-(3-Bromophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4d)

Yellow solid; yield 82%; mp 172–174°C; IR (neat, νmax): 3136, 1663, 1596, 1493, 1404 cm-1; 1H NMR (CDCl3): δ 9.17 (s, 1H), 8.80 (d, J = 3.2 Hz, 1H), 8.39 (s, 1H), 8.26 (d, J = 7.6 Hz, 1H), 7.89 (d, J = 15.2 Hz, 2H), 7.80 (d, J = 8 Hz, 2H), 7.36–7.62 (m, 8H); 13C NMR (CDCl3): δ 188.5, 153.1, 152.3, 149.6, 139.1, 135.8, 135.8, 134.2, 133.4, 131.8, 131.6, 130.3, 129.6, 127.6, 127.4, 127.3, 123.6, 122.9, 121.0, 119.4, 118.0. ESI-MS. Calcd. for C23H16BrN3O, [M + H]+: m/z 430.05. Found: m/z 430.04. Anal. Calcd for C23H16BrN3O: C, 64.20; H, 3.75; N, 9.77. Found: C, 64.26; H, 3.71; N, 9.75.

(E)-3-(3-(4-Bromophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4e)

Yellow solid; yield 75%; mp 158–160°C; IR (neat, νmax ): 3136, 1663, 1596, 1501, 1400 cm-1; 1H NMR (CDCl3): δ 9.20 (s, 1H), 8.80 (d, J = 4.4 Hz, 1H), 8.40 (s, 1H), 8.27 (d, J = 7.6 Hz, 1H), 7.90 (d, J = 15.6 Hz, 1H), 7.80 (d, J = 8 Hz, 2H), 7.64 (d, J = 8 Hz, 2H), 7.58 (d, J = 8 Hz, 2H), 7.33–7.53 (m, 5H); 13C NMR (CDCl3): δ 188.3, 153.1, 152.8, 149.6, 139.1, 136.0, 135.8, 133.4, 132.0, 131.0, 130.2, 129.6, 127.5, 127.1, 123.7, 123.2, 120.8, 119.3, 117.9. ESI-MS. Calcd for C23H16BrN3O, [M + H]+: m/z 430.05. Found: 430.04. Anal. Calcd for C23H16BrN3O: C, 64.20; H, 3.75; N, 9.75. Found: C, 64.14; H, 3.77; N, 9.79.

(E)-3-(3-(3-Nitrophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4f)

Yellow solid; yield 69%; mp 206–208°C; IR (neat, νmax): 3136, 1665, 1596, 1533, 1423, 1344 cm-1; 1H NMR (DMSO-d6): δ 9.40 (s, 1H), 9.24 (d, J = 1.6 Hz, 1H), 8.78–8.80 (m, 1H), 8.40 (s, 1H), 8.32 (d, J = 8 Hz, 1H), 8.27 (d, J = 8 Hz, 1H), 8.06 (d, J = 8 Hz, 1H), 7.77–7.89 (m, 4H), 7.65 (d, J = 15.2 Hz, 1H), 7.37–7.58 (m, 4H); 13C NMR (DMSO-d6): δ 188.1, 153.7, 150.8, 149.9, 148.5, 139.1, 136.0, 134.9, 134.3, 133.8, 133.1, 131.0, 130.1, 129.9, 127.9, 124.4, 123.8, 122.9, 122.2, 119.2, 118.4. ESI-MS. Calcd. for C23H16N4O3, [M + H]+: m/z 397.2. Found: m/z 397.2. Anal. Calcd for C23H16N4O3: C, 69.69; H, 4.07; N, 14.13. Found: C, 69.65; H, 4.08; N, 14.16.

(E)-3-(3-(4-Nitrophenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4g)

Yellow solid; yield 76%; mp 220–222°C; IR (neat, νmax): 3131, 1669, 1596, 1542, 1512, 1421, 1346 cm-1; 1H NMR (DMSO-d6): δ 9.38 (s, 1H), 9.23 (s, 1H), 8.78 (d, J = 3.6 Hz, 1H), 7.80–8.32 (m, 8H), 7.63 (d, J = 15.2 Hz, 1H), 7.34–7.57 (m, 4H, 1H); 13C NMR (DMSO-d6): δ 187.9, 153.7, 150.8, 149.9, 147.6, 139.0, 138.6, 136.0, 134.3, 133.1, 130.1, 129.9, 129.6, 127.9, 124.4, 124.4, 122.2, 119.2, 118.7. ESI-MS. Calcd for C23H16N4O3, [M + H]+: m/z 397.2. Found: m/z 397.2. Anal. Calcd for C23H16N4O3: C, 69.69; H, 4.07; N, 14.13. Found: C, 69.67; H, 4.08; N, 14.14.

(E)-3-(1-Phenyl-3-p-tolyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4h)

Yellow Solid; yield 61%; mp 146–148°C; IR (neat, νmax): 3119, 1667, 1596, 1538, 1499, 1419 cm-1; 1H NMR (CDCl3): δ 9.15 (s, 1H), 8.76 (d, J = 3.2 Hz, 1H), 8.36 (s, 1H), 8.21 (d, J = 8Hz, 1H), 7.91 (d, J = 15.6 Hz, 1H), 7.77 (d, J = 7.6 Hz, 2H), 7.57 (d, J = 8Hz, 2H), 7.26–7.49 (m, 7H), 2.41 (s, 3H); 13C NMR (CDCl3): δ 188.5, 154.1, 152.9, 149.6, 139.3, 138.8, 136.7, 135.8, 133.6, 129.5, 129.5, 129.2, 128.6, 127.3, 127.1, 123.6, 120.4, 119.3, 117.9, 21.3. ESI-MS. Calcd for C24H19N3O, [M + H]+: m/z 366.1. Found: m/z 366.2. Anal. Calcd for C24H19N3O: C, 78.88; H, 5.24; N, 11.50. Found: C, 78.85; H, 5.25; N, 11.52.

(E)-3-(3-(4-Methoxyphenyl)-1-phenyl-1H-pyrazol-4-yl)-1-(pyridin-3-yl)prop-2-en-1-one (4i)

Yellow solid; yield 61%; mp 158–160°C; IR (neat, νmax): 3119, 1665, 1596, 1525, 1503, 1419 cm-1; 1H NMR (CDCl3): δ 9.15 (s, 1H), 8.74 (d, J = 4.8 Hz, 1H), 8.35 (s, 1H), 8.21 (d, J = 8Hz, 1H), 7.89 (d, J = 15.6 Hz, 1H), 7.76 (d, J = 8 Hz, 2H), 7.60 (d, J = 8.4 Hz, 2H), 7.26–7.47 (m, 5H), 6.99 (d, J = 8.8 Hz, 2H), 3.83 (s, 3H); 13C NMR (CDCl3): δ 188.5, 160.2, 153.9, 153.0, 149.6, 139.3, 136.7, 135.7, 133.5, 130.0, 129.5, 127.2, 127.0, 124.5, 123.6, 120.2, 119.3, 117.8, 114.3, 55.3. ESI-MS. Calcd for C24H19N3O2, [M + H]+: m/z 382.1. Found: m/z 382.2. Anal. Calcd for C24H19N3O2: C, 75.57; H, 5.02; N, 11.02. Found: C, 75.54; H, 5.03; N, 11.04.

In vitro cytotoxic activity

The MTT [(3-(4, 5-dimethyl-2-thiazolyl) 2,5-diphenyl-2H-tetrazolium bromide)] assay is based on conversion of yellow, water soluble tetrazolium dye to a water-insoluble purple formazan by living cells. The amount of formazan crystals generated is directly proportional to the number of viable cells. The Caco-2, MIA PaCa-2, MCF-7 and NIH-3T3 cells were grown (37°C, 5% CO2 in water jacketed incubator shell) using DMEM media with 10% FBS (fotal bovine serum), seeded on a single 96 well plate and allowed to adhere for MTT assays. The plate was treated with increasing concentrations of 1, 12.5, 25, 50 and 100 μm of the compounds. These concentrations were used in triplicate to the single 96 well tissue culture plate. After 24 h of treatment, the MTT assay was performed to check cell viability. For the MTT assay, the media were removed from all the wells, 10 μL of MTT reagent per well from a working stock (5 mg/mL) was added and the plates were incubated (37°C and 5% CO2) for 2–3 h, and then the reagent was removed and the crystals were dissolved in dimethyl sulfoxide. The absorbance was measured at a test wavelength of 570 nm using an ELISA plate reader, LMR-340 M with a microplate reader. The percentage inhibition was calculated by the formulae:

% Inhibition=100-Mean OD of treated cellsMean OD of the vehicle control cells (negative control)×100

Each assay was repeated three times. The IC50 values were calculated from the dose effect curve (Figure S1) and expressed as concentration (μm) of drug [34].

Acknowledgments

The authors thank the Head, Department of Chemistry, JMI, New Delhi, for providing research facilities, Prof. Vibha Tandon and Dr. Mohammad Abid for helpful discussions, and UGC, New Delhi, for research fellowship for Raquib Alam.

References

[1] Siegel, R.; DeSantis, C.; Virgo, K.; Stein, K.; Mariotto, A.; Smith, T.; Cooper, D.; Gansler, T.; Lerro, C.; Fedewa, S.; et al. Cancer treatment and survivorship statistics. CA Cancer J. Clin.2012, 62, 220–241.10.3322/caac.21149Search in Google Scholar PubMed

[2] Wu, Q.; Yang, Z.; Nie, Y.; Shi, Y.; Fan, D. Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches. Cancer Lett.2014, 347, 159–166.10.1016/j.canlet.2014.03.013Search in Google Scholar PubMed

[3] Fustero, S.; Sanchez-Rosello, M.; Barrio, P.; Simon-Fuentes, A. From 2000 to Mid-2010: a fruitful decade for the synthesis of pyrazoles. Chem. Rev.2011, 111, 6984–7034.10.1021/cr2000459Search in Google Scholar PubMed

[4] Raffa, D.; Maggio, B.; Raimondi, M. V.; Cascioferro, S.; Plescia, F.; Cancemi, G.; Daidone, G. Recent advanced in bioactive systems containing pyrazole fused with a five membered heterocycle. Eur. J. Med. Chem.2015, 97, 732–746.10.1016/j.ejmech.2014.12.023Search in Google Scholar PubMed

[5] Küçükgüzel, S. G.; Senkardes, S. Recent advances in bioactive pyrazoles. Eur. J. Med. Chem.2015, 97, 786–815.10.1016/j.ejmech.2014.11.059Search in Google Scholar PubMed

[6] Bronson, J.; Dhar, M.; Ewing, W.; Lonberg, N. Chapter thirty-one-to market, to market-2011. Annu. Rep. Med. Chem.2012, 47, 499–569.10.1016/B978-0-12-396492-2.00031-XSearch in Google Scholar

[7] Palomer, A.; Cabre, F.; Pascual, J.; Campos, J.; Trujillo, M. A.; Entrena, A.; Gallo, M. A.; Garcia, L.; Mauleon, D.; Espinosa, A. Identification of novel cyclooxygenase-2 selective inhibitors using pharmacophore models. J. Med. Chem.2002, 45, 1402–1411.10.1021/jm010458rSearch in Google Scholar PubMed

[8] Hsu, A. L.; Ching, T. T.; Wang, D. S.; Song, X.; Rangnekar, V. M.; Chen, C. S. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2. J. Biol. Chem.2000, 275, 11397–11403.10.1074/jbc.275.15.11397Search in Google Scholar PubMed

[9] Williams, C. S.; Watson, A. J.; Sheng, H.; Helou, R.; Shao, J.; DuBois, R. N. Celecoxib prevents tumor growth in vivo without toxicity to normal gut: lack of correlation between in vitro and in vivo models. Cancer Res.2000, 60, 6045–6051.Search in Google Scholar

[10] Kulp, S. K.; Yang, Y. T.; Hung, C. C.; Chen, K. F.; Lai, J. P.; Tseng, P. H.; Fowble, J. W.; Ward, P. J.; Chen, C. S. 3-Phosphoinositide-dependent protein kinase-1/Akt signaling represents a major cyclooxygenase-2-independent target for celecoxib in prostate cancer cells. Cancer Res.2004, 64, 1444–1451.10.1158/0008-5472.CAN-03-2396Search in Google Scholar PubMed

[11] Arasavelli, A. M.; Ganapavarapu, V.; Vidavalur, S. Design, synthesis, and anti-cancer activity of novel aryl/heteroaryl chalcone derivatives. Heterocycl. Commun.2016, 22, 1–5.10.1515/hc-2015-0271Search in Google Scholar

[12] Lee, D. H.; Jung, Y. J.; Koh, D.; Lim, Y.; Lee, Y. H.; Shin, S. Y. A synthetic chalcone, 2′-hydroxy-2,3,5′-trimethoxychalcone triggers unfolded protein response-mediated apoptosis in breast cancer cells. Cancer Lett.2016, 372, 1–9.10.1016/j.canlet.2015.12.017Search in Google Scholar PubMed

[13] Winter, E.; Neuenfeldt, P. D.; Chiaradia-Delatorre, L. D.; Gauthier, C.; Yunes, R. A.; Nunes, R. J.; Creczynski-Pasa, T. B.; Pietro, A. D. Symmetric bis-chalcones as a new type of breast cancer resistance protein inhibitors with a mechanism different from that of chromones. J. Med. Chem.2014, 57, 2930–2941.10.1021/jm401879zSearch in Google Scholar

[14] Nelson, G.; Alam, M. A.; Atkinson, T.; Gurrapu, S.; Kumar, J. S.; Bicknese, C.; Williams, J. L. Synthesis and evaluation of p-N,N-dialkyl substituted chalcones as anti-cancer agents. Med. Chem. Res.2013, 22, 4610–4614.10.1007/s00044-013-0469-8Search in Google Scholar

[15] Yang, Z.; Wu, W.; Wang, J.; Liu, L.; Li, L.; Yang, J.; Wang, G.; Cao, D.; Zhang, R.; Tang, M.; et al. Synthesis and biological evaluation of novel millepachine derivatives as a new class of tubulin polymerization inhibitors. J. Med. Chem.2014, 57, 7977–7989.10.1021/jm500849zSearch in Google Scholar

[16] Insuasty, B.; Tigreros, A.; Orozco, F.; Quiroga, J.; Abonia, R.; Nogueras, M.; Sanchez, A.; Cobo, J. Synthesis of novel pyrazolic analogues of chalcones and their 3-aryl-4-(3-aryl-4,5-dihydro-1H-pyrazol-5-yl)-1-phenyl-1H-pyrazole derivatives as potential antitumor agents. Bioorg. Med. Chem.2010, 18, 4965–4974.10.1016/j.bmc.2010.06.013Search in Google Scholar

[17] Zhang, B.; Duan, D.; Ge, C.; Yao, J.; Liu, Y.; Li, X.; Fang, J. Synthesis of xanthohumol analogues and discovery of potent thioredoxin reductase inhibitor as potential anticancer agent. J. Med. Chem.2015, 58, 1795–1805.10.1021/jm5016507Search in Google Scholar

[18] Wang, Z.; Wang. N.; Han, S.; Wang, D.; Mo. S.; Yu, L.; Huang, H.; Tsui, K.; Shen, J.; Chen, J. Dietary compound isoliquiritigenin inhibits breast cancer neoangiogenesis via VEGF/VEGFR-2 signaling pathway. PLoS One2013, 8, e68566.10.1371/journal.pone.0068566Search in Google Scholar

[19] Yo, Y.; Shieh, G.; Hsu, K.; Wu, C.; Shiau, A.; Licorice and Licochalcone-A induce autophagy in LNCaP prostate cancer cells by suppression of Bcl-2 expression and the mTOR pathway. J. Agric. Food Chem.2009, 57, 8266–8273.10.1021/jf901054cSearch in Google Scholar

[20] Abdel-Aziz, M.; Park, S.; El-Din, G.; Abuo-Rahma, A. A.; Sayed, M. A.; Kwon, Y. Novel N-4-piperazinyl-ciprofloxacin-chalcone hybrids: synthesis, physicochemical properties, anticancer and topoisomerase I and II inhibitory activity. Eur. J. Med. Chem. 2013, 69, 427–438.10.1016/j.ejmech.2013.08.040Search in Google Scholar

[21] Shimizu, K.; Kondo, R.; Sakai, K.; Buabarn, S.; Dilokkunanant, U. A geranylated chalcone with 5α-reductase inhibitory properties from Artocarpus incises. Phytochem.2000, 54, 737–739.10.1016/S0031-9422(00)00187-4Search in Google Scholar

[22] Kahyo, T.; Ichikawa, S.; Hatanaka, T.; Yamada, M. K.; Setou, M. A novel chalcone polyphenol inhibits the deacetylase activity of SIRT1 and cell growth in HEK293T cells. J. Pharmacol. Sci.2008, 108, 364–371.10.1254/jphs.08203FPSearch in Google Scholar

[23] Pinz, S.; Unser, S.; Brueggemann, S.; Besl, E.; Al-Rifai, N.; Petkes, H.; Amslinger, S.; Rascle, A. The synthetic α-bromo-20,3,4,4′-tetramethoxychalcone (α-Br-TMC) inhibits the JAK/STAT signaling pathway. PLoS One2014, 9, e90275.10.1371/journal.pone.0090275Search in Google Scholar

[24] Ngameni, B.; Touaibia, M.; Patnam, R.; Belkaid, A.; Sonna, P.; Ngadjui, B. T.; Annabi, B.; Roy, R. Inhibition of MMP-2 secretion from brain tumor cells suggests chemopreventive properties of a furanocoumarin glycoside and of chalcones isolated from the twigs of Dorstenia turbinate. Phytochem.2006, 67, 2573–2579.10.1016/j.phytochem.2006.09.017Search in Google Scholar

[25] Ramalho, S. D.; Bernades, A.; Demetrius, G.; Noda-Perez, C.; Vieira, P. C.; dos Santos, C. Y.; da Silva, J. A.; de Moraes, M. O.; Mousinho, K.C. Synthetic chalcone derivatives as inhibitors of cathepsins K and B, and their cytotoxic evaluation. Chem. Biodivers. 2013, 10, 1999–2006.10.1002/cbdv.201200344Search in Google Scholar

[26] Cho, M.; Ryu, M.; Jeong, Y.; Chung, Y. H.; Kim, D. E.; Cho, H. S.; Kang, S.; Han, J. S.; Chang, M. Y.; Lee, C. K.; et al. Cardamonin suppresses melanogenesis by inhibition of Wnt/beta-catenin signalling. Biochem. Biophys. Res. Commun.2009, 3, 500–505.10.1016/j.bbrc.2009.09.124Search in Google Scholar

[27] Li, Q.; Li, C.; Lu, X.; Zhang, H.; Zhu, H. Design, synthesis and biological evaluation of novel (E)-α-benzylsulfonyl chalcone derivatives as potential BRAF inhibitors. Eur. J. Med. Chem.2012, 50, 288–295.10.1016/j.ejmech.2012.02.007Search in Google Scholar

[28] Orlikova, B.; Schnekenburger, M.; Zloh, M.; Golais, F.; Diederich, M.; Tasdemir, D. Natural chalcones as dual inhibitors of HDACs and NF-κB. Oncol. Reports2012, 28, 797–805.10.3892/or.2012.1870Search in Google Scholar

[29] Kira, M. A.; Adbel-Raeman, M. O.; Gadalla, K. Z. The vilsmeier-haack reaction-III Cyclization of hydrazones to pyrazoles. Tetrahedron Lett.1969, 10, 109–110.10.1016/S0040-4039(01)88217-4Search in Google Scholar

[30] Yadlapalli, R. K.; Chourasia, O. P.; Vemuri, K.; Sritharan, M.; Perali, R. S. Synthesis and in vitro anticancer and antitubercular activity of diarylpyrazole ligated dihydropyrimidines possessing lipophilic carbamoyl group. Bioorg. Med. Chem. Lett.2012, 22, 2708–2711.10.1016/j.bmcl.2012.02.101Search in Google Scholar PubMed

[31] Desai, N. C.; Joshi, V. V.; Rajpara, K. M.; Vaghani, H. V.; Satodiya, H. M. Facile synthesis of novel fluorine containing pyrazole based thiazole derivatives and evaluation of antimicrobial activity. J. Fluorine Chem.2012, 142, 67–78.10.1016/j.jfluchem.2012.06.021Search in Google Scholar

[32] Elkady, M.; Nieß, R.; Schaible, A. M.; Bauer, J.; Luderer, S.; Ambrosi, G.; Werz, O.; Laufer, S. A. Modified acidic nonsteroidal anti-inflammatory drugs as dual inhibitors of mPGES-1 and 5-LOX. J. Med. Chem.2012, 55, 8958–8962.10.1021/jm3010543Search in Google Scholar PubMed

[33] Prakash, O.; Pannu, K.; Kumar, A. Synthesis of some new 2-(3-aryl-1-phenyl-4-pyrazolyl)-benzoxazoles using hypervalent iodine mediated oxidative cyclization of Schiff’s bases. Molecules2006, 11, 43–48.10.3390/11010043Search in Google Scholar

[34] Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods1983, 65, 55–63.10.1016/0022-1759(83)90303-4Search in Google Scholar


Supplemental Material:

The online version of this article (DOI: 10.1515/hc-2016-0042) offers supplementary material, available to authorized users.


Received: 2016-3-16
Accepted: 2016-6-1
Published Online: 2016-7-18
Published in Print: 2016-8-1

©2016 Walter de Gruyter GmbH, Berlin/Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 5.5.2024 from https://www.degruyter.com/document/doi/10.1515/hc-2016-0042/html
Scroll to top button