Accepted Manuscript Synthesis and anticancer activity of some 8-substituted-7-methoxy-2H-chromen-2one derivatives toward hepatocellular carcinoma HepG2 cells Kamilia M. Amin , Sahar M. Abou-Seri , Fadi M. Awadallah , Amal A.M. Eissa , Ghaneya S. Hassan , Mohamed M. Abdulla PII:

S0223-5234(14)01059-9

DOI:

10.1016/j.ejmech.2014.11.027

Reference:

EJMECH 7520

To appear in:

European Journal of Medicinal Chemistry

Received Date: 1 October 2014 Revised Date:

4 November 2014

Accepted Date: 14 November 2014

Please cite this article as: K.M. Amin, S.M. Abou-Seri, F.M. Awadallah, A.A.M. Eissa, G.S. Hassan, M.M. Abdulla, Synthesis and anticancer activity of some 8-substituted-7-methoxy-2H-chromen-2-one derivatives toward hepatocellular carcinoma HepG2 cells, European Journal of Medicinal Chemistry (2014), doi: 10.1016/j.ejmech.2014.11.027. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Graphical abstract:

H3CO

O

O

H3CO

O

N

N N R

O

NH

Ar NH2

O

Ar

SC

R= H, C6H5, COCH3, CONH2

RI PT

Four series of coumarin-pyrazoline hybrids, in addition to a series of coumarins bearing non-cyclic isosteres of pyrazolines, were synthesized. Cytotoxicity against HepG2 cell line was evaluated. Telomerase inhibition and apoptosis induction were examined for the most active compounds.

Ar= thiophen-2-yl, phenyl, p-(methyl)phenyl, p-(trifluoromethyl)phenyl, p-(methylthio)phenyl.

7a-e - 10a-e

AC C

EP

TE D

M AN U

2% G2 p. e6 H8 t7 s= n ie as gr a ae Mm no l e t f o . h n i

7e: IC50 = 10

11a-e

Synthesis and anticancer activity of some 8-substituted-7-methoxy-2HACCEPTED MANUSCRIPT chromen-2-one derivatives toward hepatocellular carcinoma HepG2 cells. Kamilia M. Amin,a Sahar M. Abou-Seri,a Fadi M. Awadallah,*a Amal A.M. Eissa,a Ghaneya S. Hassan,a Mohamed M. Abdullab a

Pharmaceutical Chemistry department, Faculty of Pharmacy, Cairo University, Kasr El-Aini street 11562,

b

Research Unit, Saco Pharm. Co., 6th October City, Egypt

SC

Abstract.

RI PT

Cairo Egypt.

Based on the reported anticancer activity of coumarin and pyrazoline derivatives, the present investigation dealt with the design and synthesis of coumarin derivatives bearing

M AN U

diversely substituted pyrazoline moieties 7-10. The non-cyclic isosteres 11a-e of compounds 10a-e were synthesized for comparative reasons. The target compounds were synthesized from 8-acetyl-7-methoxycoumarin that underwent Claisen-Schmidt condensation with various aldehydes to give the chalcones 6a-e, followed by reaction with hydrazine hydrate, phenyl hydrazine or semicarbazide under the appropriate conditions. Cytotoxicity of the synthesized

TE D

compounds was evaluated in vitro against liver HepG2 cell lines. Compounds were active in the nanomolar range. The most active compounds were investigated for their telomerase inhibition and proapoptotic activities.

AC C

EP

Keywords: Coumarin; pyrazoline; synthesis; anticancer activity; liver cancer.

*

Corresponding author: [email protected] Tel: 002-02-25353400, 002-012-23483941 Fax:002-02-23628426

1

ACCEPTED MANUSCRIPT 1. Introduction. It is observed that telomerase is up-regulated in 80−90% of all cancer cells isolated from primary human tumors but is absent in neighboring normal cells. This resulted in significant efforts to validate telomerase as an anticancer drug target and to develop effective approaches toward its inhibition. When considering anticancer drug targets, several

RI PT

advantages for targeting telomerase and telomere maintenance exist when compared to other targets and pathways. Most important is the apparently universal requirement for telomere maintenance in cancer cells including putative cancer stem cells [1]. Telomerase remains active in the early stages of life maintaining telomere length and the chromosomal integrity of

SC

frequently dividing cells. It turns dormant in most somatic cells during adulthood [2,3]. However telomerase gets reactivated in the vast majority of invasive cancers. Extension of

M AN U

telomeres by telomerase is required for un-limited proliferation and it is well accepted that human cancer cells achieve immortalization in large part through the illegitimate activation of telomerase expression [4,5]. In addition to its telomere maintenance function, telomerase also has a pro-survival function resulting in an increased resistance against DNA damage and decreased apoptosis induction [6]. Thus, the inhibition of telomerase activity is a potential target for cancer therapeutic discovery which might have minimal side effects.

TE D

In the course of identifying various chemical substances which may serve as leads for designing new anticancer agents, the present work particularly emphasizes on the coumarin and pyrazoline derivatives which have been identified as cores for chemotherapeutic agents

EP

with significant therapeutic efficacy against tumors. Coumarin comprised the nucleus of a wide variety of naturally occurring compounds.

AC C

They were extremely variable in structure, due to various types of substitutions in their basic structure, which could influence their biological activity. Literature survey revealed their wide spectrum and diversity of biological activities [7-16], particularly their cytotoxic activity against numerous types of cancers [17- 24]. Several studies revealed the mechanism of anticancer activity of coumarin derivatives which might include induction of apoptosis [2531]. Osthole I, a bioactive simple coumarin derivative extracted from Cnidium monnieri (L) Cusson, proved effective against human hepatocellular carcinoma (HCC) through induction of apoptosis [32]. Additionally, the coumarin-monastrol hybrid II demonstrated anticancer efficacy against many cancer cell lines mediated by apoptotic activity [33] (Figure 1). 2

On the other hand, pyrazoline-based compounds received considerable attention

ACCEPTED MANUSCRIPT

owing to their high chemotherapeutic potential. Diversely substituted pyrazoline derivatives showed good cytotoxic and antiproliferative activities against a wide range of human tumor cell lines [34-40]. This could be exemplified by the 3,5-diarylsubstituted pyrazolines III [41]



RI PT

and IV [42]. (Figure 2).

Moreover, it was pointed out that introducing the dihydropyrazole moiety in the coumarin skeleton, as illustrated by compounds V [43] and VI [44], gave highly potent

of apoptosis. (Figure 3).

M AN U



SC

cytotoxic compounds that were believed to act through inhibition of telomerase and induction

The current strategy in developing new anticancer drugs shifted toward the multiple mechanistic approach and several drugs have been validated and developed. Inspired by the inherent biological relevance of the coumarin and pyrazoline rings, the present investigation pertained to the hybridization of these two pharmacophoric moieties, in a single molecule. Expectedly, the additive effect of this combination might produce compounds of high

TE D

anticancer activity that might act by dual mechanism through induction of apoptosis and inhibition of telomerase. The target compounds had the general structure 7a-e - 9a-e, featuring a 7-methoxycoumarin nucleus substituted at position 8 with a 5-arylsubstitutedpyrazoline group. The pyrazoline ring was either unsubstituted at position 1, or carrying a

EP

phenyl group or an acetyl group. Isosteric replacement of the N-acetyl group with the carboxamide function gave compounds of general formula 10a-e. A cyclic versus noncyclic

AC C

bioisosteric modification of compounds 10a-e was also adopted to obtain the noncyclic bioisosteres 11a-e (Figure 4).

Antiproliferative activity of the synthesized compounds was evaluated in vitro against

human liver cell line HepG2. Telomerase inhibition and apoptosis induction of the most active compounds were also investigated. 2. Results and Discussions 2.1. Chemistry 3

The synthesis of the target compounds 7-11 was depicted in Schemes 1 and 2. 8-

ACCEPTED MANUSCRIPT

Acetyl-7-hydroxy-2H-chromen-2-one 4 was obtained together with its regioisomer 3 through a Fries rearrangement of the intermediate acetoxy derivative 2, which was obtained by acetylation of the commercially available 7-hydroxycoumarin 1 with acetic anhydride. The regioisomers 3 and 4 were separated by fractional crystallization from aqueous ethanol [46]. Methylation of 4 with methyl iodide in the presence of potassium carbonate in dry acetone gave the corresponding 8-acetyl-7-methoxy-2H-chromen-2-one 5 [47-49]. The chalcones 6

RI PT

were prepared by conventional Claisen-Schmidt condensation reaction of 5 with (un)substituted-arylaldehydes in the presence of 10 % sodium hydroxide in ethanol (Scheme 1) [50].

SC



The synthesis of the novel coumarin-pyrazoline hybrids 7-10 was accomplished

M AN U

through 1,4-addition of hydrazine hydrate, phenyl hydrazine or semicarbazide to the α,βunsaturated carbonyl system of the precursor chalcones 6, followed by dehydration and rearrangement (Scheme 2). The N1 unsubstituted and N1 phenyl pyrazolines 7 and 8 were produced by cyclocondensation of the chalcones 6 with hydrazine hydrate 99% or phenyl hydrazine in absolute ethanol, respectively. Alternatively, condensation of 6 with hydrazine hydrate in absolute ethanol in the presence of glacial acetic acid furnished the N1 acetyl

TE D

pyrazoline derivatives 9 as a result of in situ acetylation of the initially formed pyrazolines 7. In addition, base catalyzed cyclization of chalcones 6 into the corresponding pyrazoline-1carboxamide 10 was achieved through reaction with semicarbazide HCl in ethanolic sodium hydroxide. On the other hand, reacting 6 with semicarbazide HCl in absolute ethanol yielded

AC C

EP

the uncyclic allylidene semicarbazide derivatives 11.

The structure elucidation of the newly synthesized compounds was based on the

analytical and spectral data (IR, 1H NMR,

13

C NMR, and mass spectrometry). 1H NMR

spectra of most pyrazolines 7-10 exhibited an AMX pattern for the presence of two diastereotopic protons at C-4 (HA and HM) and one single proton at the C-5 positions (HX). These protons appeared as three doublets of doublets, respectively, at δ 2.90-3.35, 3.46-3.89, 4.91-5.63 ppm regions, each integrating for one proton. In case of compound 8a simple splitting patterns was observed with pyrazoline protons, which appeared as two signals, one doublet equivalent to 2H at δ 3.73 ppm and a multiplet equivalent to 1H at δ 5.75 ppm. Moreover, the 1H NMR spectra of compounds 7 and 10 displayed singlet exchangeable signal at δ 4.20-5.37 or δ 10.20-11.05 ppm corresponding to the pyrazoline (NH) or the N1 4

carboxamide (NH2), respectively. Meanwhile, the analogs 9 showed singlet signal derived

ACCEPTED MANUSCRIPT

from the N1 acetyl (CH3) at δ 2.18-2.37 ppm. On the other hand, the 1H NMR spectra of the allylidene semicarbazide derivatives 11 revealed the presence of two exchangeable singlet signals resonating at δ 6.47-6.54 and 8.88-9.10 ppm for the semicarbazide (NH2 and NH) protons and two doublets at δ 6.05-6.74 and 6.85-7.29 ppm (J= 15.0-16.9 Hz) representing the allylidene (CH=CH) protons, which asserted the production of the uncyclized derivatives 11. The protons belonging to the aromatic system and phenyl substituents were observed at the

RI PT

expected chemical shifts and integral values. 2.2. Anticancer activity. 2.2.1. Antiproliferative activity study.

SC

The in vitro anti-hepatocellular carcinoma (anti-HCC) activity of the newly synthesized compounds was examined against HepG2 cell line using the MTT method [51].

M AN U

Doxorubicin which is one of the most effective anticancer agents was used as positive control. The activity was expressed by median growth inhibitory concentration (IC50) and was provided in Table 1. The reported antiproliferative activity of Osthole I [32] and the lead compound VI [44] was included for comparison. The results revealed that, all the new coumarin-pyrazoline hybrids exhibited potent antiproliferative activity with IC50 values ranging from 10 ̶ 99 nM, which were far more potent than Osthole I (IC50= 161.4µM) and

TE D

compound VI (IC50= 6.64µM). In particular compounds 7a, 7d, 7e, 8a and 10e possessed superior activity at low nanomolar levels (IC50 = 11, 13, 10, 15 and 18 nM, respectively) The N1-unsubstituted pyrazoline derivatives 7a-e displayed excellent growth

EP

inhibitory activity (IC50 = 10 ̶ 48 nM). Compound 7b having phenyl substituent at position 5 of the pyrazoline ring was the least active member of this group (IC50 = 48 nM). Appending a

AC C

lipophilic substituent to the p-position of the phenyl in 7d-e contributed to an increase in potency, where the highest activity was associated with the 4-trifluoromethylphenyl 7d and 4methylthiophenyl 7e congeners (IC50 = 13 and 10 nM, respectively). Similarly, isosteric replacement of the phenyl ring with the thienyl rest in 7a resulted in about 4 fold increase in growth inhibition.

Substitution at N1 of the pyrazoline ring with a phenyl group, 8a-e, led to significant drop in activity (IC50 = 15 ̶ 78 nM), with the 5-thienylpyrazoline derivative, 8a, having the highest activity in this series. Likewise, compounds bearing the N1-acetyl group, 9a-e and the N1-carboxamide derivatives 10a-e revealed a decrease in activity compared to the N1unsubstituted pyrazolines 7a-e. Moreover, the allylidene semicarbazides 11a-e, which could 5

be considered as the non-cyclic isosteres of 10a-e, showed reduced growth inhibitory effect

ACCEPTED MANUSCRIPT

(IC50 = 75 ̶ 99 nM). All compounds in this series demonstrated the lowest activity compared to their N1-un/substituted pyrazoline counterparts 7-10. In summary, the antiproliferative activity of the novel coumarin-pyrazoline hybrids was influenced by the substitution pattern on the pyrazoline ring. Mostly, the introduction of a more lipophilic substituent to position 5 of the pyrazoline ring produced compounds with enhanced potency as shown by the 4-trifluoromethylphenyl 7d and 4-methylthiophenyl

RI PT

analogs 7e and 10e. Also, isosteric replacement of the phenyl ring with the thienyl group as in 7a and 8a enhanced the activity. However, the activity was not favored by the presence of N1substitutions as in 8a-e, 9a-e and 10a-e, which was less active than the N1-unsubstituted derivatives, 7a-e. Furthermore, a noticeable drop in potency was observed with the coumarin

2.2.2. Telomerase inhibitory activity.

M AN U



SC

derivatives bearing at position 8 an open-chain substitution, 11a-e.

The most potent cytotoxic compounds 7a, 7d, 7e, 8a and 10e were selected to investigate the mechanism underlying their anti-HCC effect. To investigate the effects of these compounds on telomerase activity, HepG2 cells were cultured with 1, 2 and 4 µM of the

TE D

selected compounds. Telomerase activity was measured by TRAP-PCR-ELISA assay after 1, 2 and 3 days of exposure to tested compounds and the results are summarized in Table 2. The pan kinase inhibitor, Staurosporine, was used as positive control [52]. The results revealed that all the tested compounds inhibited telomerase activity in HepG2cells in a dose-dependent

EP

manner. The maximal repression was observed on day 3 at 4 µM (Reduction % =61.7 ̶ 78.6 %). The order of reduction in telomerase activity was 7e > 7a > 7d > 8a > 10e, which is in

AC C

accordance with the order of their growth inhibition effect, suggesting good correlation between both activities.



2.2.3. Apoptosis study.

Suppression of telomerase activity by the new coumarin-pyrazoline hybrids is expected to be associated with induction of apoptosis in HepG2 cells. To examine the involvement of apoptosis in the antiproliferative action of the new hybrids, HepG2 cells were incubated with different concentrations of the most potent compound 7e (0.01, 0.02, 0.04 or 0.08 µmol/ml) for 24 h. The percentage of apoptotic cells were determined by Annexin V6

FITC/PI staining and the stained cells were detected by flow cytometry (Figure 5). The results

ACCEPTED MANUSCRIPT

showed that the 7e induced apoptosis in HepG2 cells in a dose-dependent manner. The percentage of early apoptotic cells was markedly elevated as a function of concentration of the tested agent from 0.54 % to 45.76%. 3. Conclusions.

RI PT

Four series of coumarin-pyrazoline hybrids 7a-e, 8a-e, 9a-e and 10a-e, in addition to the non-cyclic isosteres 11a-e, were synthesized for anticancer activity evaluation. Target compounds were screened for their antiproliferative activity against hepatocellular carcinoma cell line HepG2 using the MTT assay. All compounds demonstrated IC50 values in the

SC

nanomolar range. Compounds bearing lipophilic moieties and unsubstituted at N1 of pyrazoline ring were the most active. To investigate the mechanism of action underlying this

M AN U

activity, the most active compounds 7a, 7d, 7e, 8a and 10e were studied for their telomerase inhibitory activity where the percent reduction in telomerase activity ranged from 61.7-78.6 %. Furthermore, the most potent inhibitor 7e was tested for its apoptosis induction where it enhanced apoptosis in a dose dependent manner. Comparing the antiproliferative activity against HepG2 of the new compounds with Osthole I or the coumarin-pyrazoline derivative VI revealed that the design of the new hybrid molecules made a breakthrough in activity,

4. Experimental.

EP

4.1. Chemistry.

TE D

especially compound 7e, which could be a candidate for future investigation.

Unless otherwise noted, all materials were obtained from commercial suppliers and

AC C

used without further purification. TLC was monitored on FLUKA silica gel TLC aluminium cards (0.2 mm thickness) with fluorescent indicator 254 nm using chloroform: methanol (9:1) as eluent. Melting points were performed on Stuart SMP3 version 5 digital melting point apparatus and were uncorrected. Elemental microanalyses were performed at the Microanalytical Center, Faculty of Science, Cairo University and the Regional Center for Microbiology and Biotechnology, Al-Azhar University. NMR spectra were recorded on Varian mercury 300BB at 300 MHz for 1H NMR, and at 75.45 MHz for

13

C NMR; using

tetramethylsilane (TMS) as internal reference. Chemical shift values were given in ppm. Mass spectra were performed on Fennigan MAT, SSQ 7000 mass spectrophotometer at 70eV. Infrared Spectra were recorded on Schimadzu FT-IR 8400S spectrophotometer (Shimadzu, Kyoto, Japan), and expressed in wave number (cm-1), using potassium bromide discs. 7

4.1.1. 7-Acetoxy-2H-chromen-2-one (2).

ACCEPTED MANUSCRIPT

Compound 2 was prepared according to the literature procedure (m.p.147oC, as reported) [46].

4.1.2. 8-Acetyl-7-hydroxy-2H-chromen-2-one (4).

reported) [46].

SC

4.1.3. 8-Acetyl-7-methoxy-2H-chromen-2-one (5).

RI PT

Compound 4 was prepared according to the literature procedure (m.p.167oC, as

Compound 5 was prepared according to the literature procedure (m.p.123oC, as

M AN U

reported) [47-49].

4.1.4. General procedure for synthesis of 7-Methoxy-8-[arylacryloyl]-2H-chromen-2-ones (6a-e).

TE D

Compounds of this series (6a-e) were prepared according to the literature procedure [50].

EP

4.1.5. General procedure for synthesis of 8-(5-substituted-4,5-dihydro-1H-pyrazol-3-yl)-7methoxy-2H-chromen-2-one (7a-e).

AC C

Hydrazine hydrate 99% (0.5 ml, 10 mmol) was added to a solution of the appropriate

chalcone 6a-e (10 mmol) in absolute ethanol (20 ml) and the reaction mixture was heated under reflux for 8 h. The solution was left to cool at room temperature and the solid formed was filtered off, washed with water, dried and recrystallized from absolute ethanol. 4.1.5.1.

7-Methoxy-8-[5-(thiophen-2-yl)-4,5-dihydro-1H-pyrazol-3-yl]-2H-chromen-2-one

(7a): Yield 55%; m.p. 212-214oC; IR υmax/cm-1: 3336 (NH), 3113, 3005 (CH aromatic), 2939, 2904, 2839 (CH aliphatic), 1674 (C=O), 1604, 1560 (C=N, C=C). 1H NMR (CDCl3) δ: 3.25 (dd, 1H, C4-HA pyrazoline, JAM= 25.8 Hz and JAX= 8.4), 3.50 (dd, 1H, C4-HM pyrazoline, JMA= 25.8 and JMX= 10.2 Hz), 3.87 (s, 3H, OCH3), 4.69 (s, 1H, NH, D2O exch.), 8

5.09 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 and JXM= 10.2 Hz), 6.29 (d, 1H, C3-H chromene,

ACCEPTED MANUSCRIPT

J= 9.6 Hz), 6.89 (d, 1H, C6-H chromene, J= 8.7 Hz), 6.95 (d, 1H, C3-H thiophene, J= 3.9 Hz), 7.22 (m, 2H, C4-H and C5-H thiophene), 7.40 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.81 (d, 1H, C4-H chromene, J= 9.6 Hz). Anal. Calcd. for C17H14N2O3S (326.37): C, 62.56; H, 4.32; N, 8.58. Found: C, 62.66; H, 4.41; N, 8.36. 4.1.5.2. 7-Methoxy-8-(5-phenyl-4,5-dihydro-1H-pyrazol-3-yl)-2H-chromen-2-one (7b):

RI PT

Yield 60%; m.p. 172-174oC. IR υmax/cm-1: 3298 (NH), 3082, 3024 (CH aromatic), 2970, 2939, 2889 (CH aliphatic), 1716 (C=O), 1600, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 3.19 (dd, 1H, C4-HA pyrazoline, JAM= 21.0 and JAX= 8.4 Hz), 3.62 (dd, 1H, C4-HM pyrazoline, JMA= 21.0 and JMX= 10.5 Hz), 3.93 (s, 3H, OCH3), 4.80 (s, br, 1H, NH, D2O

SC

exch.), 5.05 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM = 10.5), 6.28 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.91 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.29-7.40 (m, 3H, ArH), 7.46 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.55 (d, 2H, ArH, J= 8.1 Hz), 7.65 (d, 1H, C4-H

Found: C, 71.11; H, 5.09; N, 8.85. 4.1.5.3.

M AN U

chromene, J = 9.6 Hz). Anal. Calcd. for C19H16N2O3 (320.34): C, 71.24; H, 5.03; N, 8.74.

7-Methoxy-8-[5-(4-methylphenyl)-4,5-dihydro-1H-pyrazol-3-yl]-2H-chromen-2-one

(7c) :

TE D

Yield 70%; m.p. 194-196oC. IR υmax/cm-1: 3294 (NH), 3080, 3012 (CH aromatic), 2981, 2947, 2920 (CH aliphatic), 1720 (C=O), 1597, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 2.30 (s, 3H, CH3), 3.14 (dd, 1H, C4-HA pyrazoline, JAM= 21.0 and JAX= 8.4 Hz), 3.57 (dd, 1H, C4-HM pyrazoline, JMA= 21.0 Hz and JMX= 10.5 Hz), 3.92 (s, 3H, OCH3), 4.98 (dd, 1H, C5-

EP

HX pyrazoline, JXA= 8.4 Hz and JXM= 10.5 Hz), 5.37 (s, br, 1H, NH, D2O exch.), 6.20 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.84 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.17 (d, 2H, ArH , J=

Hz).

AC C

8.1 Hz), 7.40-7.46 (m, 3H, ArH and C5-H chromene), 7.64 (d, 1H, C4-H chromene, J= 9.3 13

C NMR(CDCl3): 21.0 (CH3), 44.7 (C-4 pyrazoline), 56.3 (OCH3), 63.9 (C-5

pyrazoline), 107.6 (C-6 chromene), 111.2 (C-3 chromene), 112.9 (C-4a chromene), 113.4 (C8 chromene), 126.5-129.3 (aromatic Cs), 137.4 (C-4 of C6H4CH3), 139.4 (C-1 of C6H4CH3), 143.2 (C-4 chromene), 145.9 (C-8a chromene), 153.2 (C=N pyrazoline), 160.3 (C-7 chromene), 160.7 (C=O). MS m/z: 334 [M+]. Anal. Calcd. for C20H18N2O3 (334.37): C, 71.84; H, 5.43; N, 8.38. Found: C, 71.55; H, 5.70; N, 8.30. 4.1.5.4.

7-Methoxy-8-{5-[4-(trifluoromethyl)phenyl]-4,5-dihydro-1H-pyrazol-3-yl}-2H-

chromen-2-one (7d):

9

Yield 62%; m.p. 200-202oC. IR υmax/cm-1: 3294 (NH), 3080, 3012 (CH aromatic),

ACCEPTED MANUSCRIPT

2985, 2951, 2912 (CH aliphatic), 1728 (C=O), 1600, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 3.17 (dd, 1H, C4-HA pyrazoline, JAM= 21.6 and JAX= 8.4 Hz), 3.68 (dd, 1H, C4-HM pyrazoline, JMA= 21.6 and JMX= 10.5 Hz), 3.94 (s, 3H, OCH3), 4.20 (s, br, 1H, NH, D2O exch.), 5.10 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 and JXM= 10.5 Hz), 6.29 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.92 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.48 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.62-7.80 (m, 5H, ArH and C4-H chromene). MS m/z: 388 [M+]. Anal. Calcd. for

4.1.5.5.

RI PT

C20H15F3N2O3 (388.34): C, 61.86; H, 3.89; N, 7.21. Found: C, 61.69; H, 4.19; N, 7.00.

7-Methoxy-8-{5-[4-(methylthio)phenyl]-4,5-dihydro-1H-pyrazol-3-yl}-2H-chromen-

SC

2-one (7e):

Yield 60%; m.p. 208-210oC. IR υmax/cm-1: 3294 (NH), 3047, 3008 (CH aromatic),

M AN U

2970, 2900, 2839 (CH aliphatic), 1720 (C=O), 1600, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 2.48 (s, 3H. S-CH3), 3.18 (dd, 1H, C4-HA pyrazoline, JAM= 25.2 and JAX= 8.4 Hz), 3.63 (dd, 1H, C4-HM pyrazoline, JMA= 25.2 and JMX= 10.5 Hz), 3.94 (s, 3H, OCH3), 4.80 (s, br, 1H, NH, D2O exch.), 5.04 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 and JXM= 10.5 Hz), 6.28 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.89 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.27 (d, 2H, ArH, J= 8.1 Hz), 7.47 (m, 3H, ArH and C5-H chromene), 7.65 (d, 1H, C4-H chromene, J= 9.6 Hz). 13

TE D

C NMR (CDCl3): 15.8 (SCH3), 44.8 (C-4 pyrazoline), 56.5 (OCH3), 63.4 (C-5 pyrazoline),

107.7 (C-6 chromene), 113.0 (C-3 chromene), 113.5 (C-4a and C-8 chromene), 126.9-129.3 (aromatic Cs), 138.2 (C-1 of C6H4SCH3), 138.4 (C-4 of C6H4SCH3), 143.2 (C-4 chromene), 149.0 (C-8a chromene), 153.2 (C=N pyrazoline), 160.2 (C-7 chromene), 160.7 (C=O). MS

EP

m/z: 366 [M+]. Anal. Calcd. for C20H18N2O3S (366.43): C, 65.55; H, 4.95; N, 7.64. Found: C,

AC C

65.72; H, 4.81; N, 8.05.

4.1.6. General procedure for synthesis of 8-(5-substituted-1-phenyl-4,5-dihydro-1H-pyrazol3-yl)-7-methoxy-2H-chromen-2-one (8a-e) To a solution of the appropriate chalcone 6a-e (10 mmol) in absolute ethanol (20 ml), phenylhydrazine 97% (1.0 ml, 10 mmol) was added and the reaction mixture was heated under reflux for 48 h. The reaction mixture was concentrated and poured on ice-water. The crude product was filtered off, left to dry. The obtained solid was recrystallized from DMF/ water.

10

4.1.6.1. 7-Methoxy-8-[1-phenyl-5-(thiophen-2-yl)-4,5-dihydro-1H-pyrazol-3-yl]-2H-chromen-

ACCEPTED MANUSCRIPT

2-one (8a):

Yield 98 %, m.p. 167-168oC. IR υmax/cm-1: 3050 (CH aromatic), 2920, 2845 (CH aliphatic), 1728 (C=O), 1601, 1562, 1543 (C=C, C=N). 1H NMR (DMSO-d6) δ: 3.73 (d, 2H, CH2 pyrazoline, overlapped), 3.91 (s, 3H, OCH3), 5.75 (m, 1H, C5-H pyrazoline), 6.30 (d, 1H, C3-H chromene, J= 9.0 Hz), 6.67 (d, 1H, C3-H thiophene J= 5.7 Hz), 6.96- 7.39 (m, 7H, ArH, C6-H chromene and C4-H thiophene), 7.45 (d, 1H, C5-H thiophene J= 5.7 Hz), 7.73 (d, 13

C NMR

RI PT

1H, C5-H chromene, J= 8.4 Hz), 8.00 (d, 1H, C4-H chromene, J= 9.0 Hz).

(DMSO-d6) δ: 46.7 (C-4 pyrazoline), 56.6 (C-5 pyrazoline), 59.2 (OCH3), 108.6 (C-6 chromene), 112.8 (C-4a chromene), 113.8 (C-3 and C-8 chromene), 119.2-130.0 (aromatic Cs), 142.0 (C-4 chromene), 144.3 (C-1 of C6H5), 146.0 (C-8a chromene), 152.0 (C=N), 156.0

M AN U

68.64; H, 4.51; N, 6.96. Found: C, 68.90 H, 4.69 N, 6.98.

SC

(C-7 chromene), 161.0 (C=O). MS m/z: 402 [M+]. Anal. Calcd. for C23H18N2O3 S (402.47): C,

4.1.6.2. 8-(1,5-Diphenyl-4,5-dihydro-1H-pyrazol-3-yl)-7-methoxy-2H-chromen-2-one (8b): Yield 98.5%, m.p. 112-113oC. IR υmax/cm-1: 3036 (CH aromatic), 2924 (CH aliphatic), 1728 (C=O), 1601, 1562, (C=N, C=C). 1H NMR (DMSO-d6) δ: 3.00 (dd, 1H, C4-HA

TE D

pyrazoline, JAM= 21.6 and JAX= 8.4 Hz), 3.85 (dd, 1H, C4-HM pyrazoline, JMA= 21.6 and JMX = 10.8 Hz), 3.89 (s, 3H, OCH3), 5.50 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.8), 6.30 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.94 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.09-7.44 (m, 10H, ArH), 7.76 (d, 1H, C5-H chromene, J= 8.7 Hz), 8.3 (d, 1H, C4-H chromene, J= 9.6 13

C NMR (DMSO-d6) δ: 46.5 (C-4 pyrazoline), 56.3 (OCH3), 62.9 (C-5 pyrazoline),

EP

Hz).

108.5 (C-6 chromene), 109.3 (C-3 chromene), 112.6 (C-4a chromene), 112.8 (C-8 chromene),

AC C

113.0-129.9 (aromatic Cs), 142.6 (C-1 of C6H5), 141.3 (C-1 of C6H5), 144.3 (C-4 chromene), 144.5 (C-8a chromene), 152.5 (C=N pyrazoline), 159.7 (C-7 chromene), 160.5 (C=O). MS m/z: 396 [M+]. Anal. Calcd. for C25H20N2O3 (396.44): C, 75.74; H, 5.08; N, 7.07. Found: C, 76.00 H, 5.10 N, 6.96.

4.1.6.3. 7-Methoxy-8-[5-(4-methylphenyl)-1-phenyl-4,5-dihydro-1H-pyrazol-3-yl]-7-methoxy2H-chromen-2-one (8c): Yield 95 %, m.p. 233-234oC. IR υmax/cm-1: 3024 (CH aromatic), 2941, 2922, 2843 (CH aliphatic), 1728 (C=O), 1600, 1560 (C=N, C=C). 1H NMR (CDCl3) δ: 2.34 (s, 3H, CH3), 11

3.08 (dd, 1H, C4-HA pyrazoline, JAM= 24.6 and JAX= 6.9 Hz), 3.89 (dd, 1H, C4-HM

ACCEPTED MANUSCRIPT

pyrazoline, JMA= 24.0 and JMX= 12.0 Hz), 3.91 (s, 3H, OCH3), 5.23 (dd, 1H, C5-HX pyrazoline, JXA= 7.2 Hz and JXM= 12.0), 6.25 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.88 (d, 1H, C6-H chromene, J= 8.4 Hz), 7.09-7.44 (m, 9H, ArH), 7.42 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.62 (d, 1H, C4-H chromene, J= 9.6 Hz). MS m/z: 410 [M+]. Anal. Calcd. for

RI PT

C26H22N2O3 (410.46): C, 76.08; H, 5.40; N, 6.83. Found: C, 76.29; H, 5.52; N, 6.68.

4.1.6.4. 7-Methoxy-8-{1-phenyl-5-[4-(trifluoromethyl)phenyl]-4,5-dihydro-1H-pyrazol-3-yl}2H-chromen-2-one (8d):

Yield 89 %, m.p. 178-180oC. IR υmax/cm-1: 3078 (CH aromatic), 2931, 2825 (CH

SC

aliphatic), 1732 (C=O), 1601, 1543 (C=N, C=C). 1H NMR (DMSO-d6) δ: 3.00 (dd, 1H, C4HA pyrazoline, JAM= 21.6 and JAX= 8.4 Hz), 3.77 (dd, 1H, C4-HM pyrazoline, JMA= 21.6 and

M AN U

JMX= 10.8 Hz), 3.88 (s, 3H, OCH3), 5.60 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.8 Hz), 6.30 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.93 (d, 2H, ArH, J= 7.8 Hz), 7.06-7.21 (m, 6H, ArH and C6-H chromene), 7.21 (d, 2H, ArH, J= 8.1 Hz),

7.9 (d, 1H, C5-H

chromene, J= 8.7 Hz), 8.20 (d, 1H, C4-H chromene, J= 9.6 Hz). MS m/z: 464 [M+]. Anal. Calcd. for C26H19F3N2O3 (464.44): C, 67.24; H, 4.12; N, 6.03. Found: C, 67.44; H, 4.29; N,

4.1.6.5.

TE D

6.26.

7-Methoxy-8-{5-[4-(methylthio)phenyl]-1phenyl-4,5-dihydro-1H-pyrazol-3-yl}-2H-

EP

chromen-2-one (8e):

Yield 94%, m.p. 175-176oC. IR υmax/cm-1: 3078 (CH aromatic), 2924, 2851 (CH

AC C

aliphatic), 1732 (C=O), 1601,1566 (C=N, C=C). 1H NMR (DMSO-d6) δ: 2.44 (s, 3H, S-CH3), 2.98 (dd, 1H, C4-HA pyrazoline, JAM= 25.2 and JAX= 8.4 Hz), 3.78 (dd, 1H, C4-HM pyrazoline, JMA= 25.2 and JMX= 10.5 Hz), 3.90 (s, 3H, OCH3), 5.40 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 and JXM= 10.5 Hz), 6.32 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.70 (t, 1H, ArH, J= 9.0 Hz), 6.93 (d, 2H, ArH , J= 7.8 Hz), 7.10-7.23 (m, 5H, ArH and C6-H chromene), 7.36 (d, 2H, ArH, J= 8.4 Hz), 7.75 (d, 1H, C5-H chromene, J= 9.0 Hz), 8.02 (d, 1H, C4-H chromene, J= 9.6 Hz). MS m/z: 442 [M+]. Anal. Calcd. for C26H22N2O3S (442.54): C, 70.57; H, 5.01; N, 6.33. Found: C, 70.82; H, 5.31; N, 6.31.

12

4.1.7. General procedure for synthesis of 8-(1-Acetyl-5-substituted-4,5-dihydro-1H-pyrazol-3-

ACCEPTED MANUSCRIPT

yl)-7-methoxy-2H-chromen-2-one (9a-e)

Hydrazine hydrate 99% (1 ml, 20 mmol) was added to a solution of the appropriate chalcone 6a-e (20 mmol) in absolute ethanol (10 ml) and glacial acetic acid (1 ml). The reaction mixture was heated under reflux for 18 h. The solution was poured into ice water. The crude product was filtered off, washed several times with water, dried and crystallized

RI PT

from aqueous ethanol. 4.1.7.1. 8-[1-Acetyl-5-(thiophen-2-yl)-4,5-dihydro-1H-pyrazol-3-yl]-7-methoxy-2H-chromen2-one (9a):

Yield 68%; m.p. 195-197oC. IR υmax/cm-1: 3086, 3047 (CH aromatic), 2927, 2846 (CH

SC

aliphatic), 1736 (C=O), 1659 (C=O), 1605, 1562 (C=N, C=C). 1H NMR (DMSO-d6) δ: 2.19 (s, 3H, COCH3), 2.95 (dd, 1H, C4-HA pyrazoline, JAM= 21.3 and JAX= 8.4 Hz), 3.72 (dd, 1H,

M AN U

C4-HM pyrazoline, JMA= 21.3 and JMX= 10.5 Hz), 3.93 (s, 3H, OCH3), 5.10 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 and JXM= 10.5 Hz), 6.34 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.99 (d, 1H, C3-H thiophene, J= 4.2 Hz), 7.11-7.29 (m, 2H, C4-H thiophene and C6-H chromene), 7.43 (d, 1H, C5-H thiophene, J= 5.1 Hz), 7.80 (d, 1H, C5-H chromene, J= 9.0), 8.06 (d, 1H, C4-H chromene, J= 9.3 Hz). MS m/z: 368 [M+]. Anal. Calcd. for C19H16N2O4S (368.41): C,

4.1.7.2.

TE D

61.94; H, 4.38; N,7.60. Found C, 62.12; H, 4.29; N, 7.96.

8-(1-Acetyl-5-phenyl-4,5-dihydro-1H-pyrazol-3-yl)-7-methoxy-2H-chromen-2-one

EP

(9b):

Yield 62%; m.p. 126-128oC. IR υmax/cm-1: 3082, 3059 (CH aromatic), 2924, 2851 (CH

AC C

aliphatic), 1732 (2C=O), 1600, 1562 (C=N, C=C). 1H NMR (DMSO-d6) δ: 2.20 (s, 3H, COCH3), 3.05 (dd, 1H, C4-HA pyrazoline, JAM= 20.2 and JAX= 8.4 Hz), 3.85 (dd, 1H, C4-HM pyrazoline, JMA= 20.2 and JMX= 10.5 Hz), 3.91 (s, 3H, OCH3), 5.55 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.5 Hz), 6.34 (d, 1H, C3-H chromene, J= 9.3 Hz), 7.137.54 (m, 6H, Ar-H and C6-H chromene), 7.83 (d, 1H, C5-H chromene, J= 8.7 Hz), 8.06 (d, 1H, C4-H chromene, J= 9.3 Hz). Anal. Calcd. for C21H18N2O4 (362.38): C, 69.60; H, 5.01; N, 7.73. Found C, 69.42; H, 5.17; N, 8.08.

4.1.7.3.

8-[1-Acetyl-5-(4-methylphenyl)-4,5-dihydro-1H-pyrazol-3-yl]-7-methoxy-2H-

chromen-2-one (9c): 13

Yield 75%; m.p. 140-142oC. IR υmax/cm-1: 3082 (CH aromatic), 2924, 2846 (CH

ACCEPTED MANUSCRIPT

aliphatic), 1654 (2C=O), 1600, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 2.35 (s, br, 6H, CH3 and COCH3), 3.05 (dd, 1H, C4-HA pyrazoline, JAM= 20.1 and JAX= 8.4 Hz), 3.46 (dd, 1H, C4HM pyrazoline, JMA= 20.1 and JMX= 10.5 Hz), 3.91 (s, 3H, OCH3), 4.91 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.5 Hz), 6.23 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.87 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.17-7.34 (m, 3H, ArH and C5-H chromene), 7.43 (d, 2H, ArH , J= 7.5 Hz), 7.61 (d,1H, C4-H chromene, J= 9.0 Hz).

13

C NMR(CDCl3): 21.0 (CH3),

RI PT

29.6 (COCH3), 44.6 (C-4 pyrazoline), 56.3 (OCH3), 64.0 (C-5 pyrazoline), 107.6 (C-6 chromene), 111.3 (C-3 chromene), 112.9 (C-4a chromene), 113.3 (C-8 chromene), 126.4129.3 (aromatic Cs), 137.3 (C-4 of C6H4CH3), 139.7 (C-1 of C6H4CH3), 143.3 (C-4 chromene), 145.0 (C-8a chromene), 153.2 (C=N pyrazoline), 160.4 (C-7 and C=O chromene),

SC

160.6 (COCH3). MS m/z: 376 [M+]. Anal. Calcd. for C22H20N2O4 (376.41): C, 70.20; H, 5.36;

4.1.7.4.

M AN U

N, 7.44. Found C, 70.24; H, 5.29; N, 7.02.

8-{1-Acetyl-5-[4-(trifluoromethyl)phenyl]-4,5-dihydro-1H-pyrazol-3-yl}-7-methoxy-

2H-chromen-2-one (9d):

Yield 60%; m.p. 187-189oC. IR υmax/cm-1: 3076, 3049 (CH aromatic), 2943, 2848 (CH aliphatic), 1732 (C=O), 1666 (C=O), 1603, 1562 (C=N, C=C). 1H NMR (CDCl3) δ: 2.37 (s,

TE D

3H, COCH3), 3.11 (dd, 1H, C4-HA pyrazoline, JAM= 21.0 and JAX= 8.4 Hz), 3.78 (dd, 1H, C4HM pyrazoline, JMA= 21.0 and JMX= 10.5 Hz), 3.94 (s, 3H, OCH3), 5.62 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.5 Hz), 6.29 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.92 (d, 1H, C6-H chromene, J= 8.7 Hz), 7.34 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.45-7.56 (m, 4H,

EP

Ar-H), 7.68 (d, 1H, C4-H chromene, J= 9.6 Hz). MS m/z: 430 [M+]. Anal. Calcd. for

AC C

C22H17F3N2O4 (430.38): C, 61.40; H, 3.98; N, 6.51. Found C, 61.58; H, 4.05; N, 6.25.

4.1.7.5.

8-{1-Acetyl-5-[4-(methylthio)phenyl]-4,5-dihydro-1H-pyrazol-3-yl}-7-methoxy-2H-

chromen-2-one (9e):

Yield 55%; m.p. 137-139oC. IR υmax/cm-1: 3078 (CH aromatic), 2920, 2843 (CH aliphatic), 1732 (C=O), 1662 (C=O), 1605, 1562 (C=N, C=C). 1H NMR (DMSO-d6) δ: 2.18 (s, 3H, COCH3), 2.44 (s, 3H, SCH3), 2.95 (dd, 1H, C4-HA pyrazoline, JAM= 21.0 and JAX= 8.4 Hz), 3.75 (dd, 1H, C4-HM pyrazoline, JMA= 21.0 and JMX= 10.5 Hz), 3.91 (s, 3H, OCH3), 5.48 (dd, 1H, C5-HX pyrazoline, JXA= 8.4 Hz and JXM= 10.5 Hz), 6.34 (d, 1H, C3-H chromene, J= 9.9 Hz), 7.19-7.31 (m, 5H, Ar-H and C6-H chromene), 7.80 (d, 1H, C5-H chromene, J= 9.0 14

Hz), 8.04 (d, 1H, C4-H chromene, J= 9.9 Hz). Anal. Calcd. for C22H20N2O4S (408.47): C,

ACCEPTED MANUSCRIPT

64.69; H, 4.94; N, 6.86. Found C, 64.80; H, 5.10; N, 6.72.

4.1.8. General procedure for synthesis of 3-(7-methoxy-2-oxo-2H-chromen-8-yl)-5substituted-4,5-dihydropyrazole-1-carboxamide (10a-e): To a solution of the appropriate chalcone 6a-e (1.6 mmol) in absolute ethanol (10 ml),

RI PT

semicarbazide hydrochloride (0.4 g; 3.66 mmol) and sodium hydroxide (0.3 g; 7.32 mmol) were added and the solution was refluxed for 12 h. The reaction mixture was poured onto ice water and the obtained solid was filtered off, washed with water and dried. The crude product

4.1.8.1.

SC

was crystallized from ethanol.

3-(7-methoxy-2-oxo-2H-chromen-8-yl)-5-(thiophen-2-yl)-4,5-dihydropyrazole-1-

M AN U

carboxamide (10a):

Yield 76%; m.p. 165-167 oC. IR υmax/cm-1: 3468, 3325 (NH2), 3109 (CH aromatic), 2920, 2850 (CH aliphatic), 1697 (2C=O, br.), 1604, 1570 (C=N, C=C). 1H NMR (DMSO-d6): δ 3.17 (dd, 1H, C4-HA pyrazoline, JAM= 21.3 and JAX= 7.2 Hz), 3.75 (dd, 1H, C4-HM pyrazoline, JMA= 21.3 and JMX= 16.5 Hz), 3.84 (s, 3H, OCH3), 5.63 (dd, 1H, C5-HX pyrazoline, JXA= 7.2 and JXM= 16.5 Hz), 6.34 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.93-6.99

TE D

(m, 2H, C3-H and C4-H thiophene), 7.21 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.42 (d, 1H, C5-H thiophene, J= 5.7 Hz), 7.69 (d, 1H, C5-H chromene, J= 8.7 Hz), 8.03 (d, 1H, C4-H chromene, J= 9.6 Hz), 11.02 (s, 2H, NH2, D2O exch.). Anal. Calcd. for C18H15N3O4S

4.1.8.2.

EP

(369.39): C, 58.53; H, 4.09; N, 11.38. Found: C, 58.61; H, 4.13; N, 11.59. 3-(7-methoxy-2-oxo-2H-chromene-8-yl)-5-phenyl-4,5-dihydropyrazole-1-

AC C

carboxamide (10b):

Yield 74%; m.p. 158-160 oC. IR υmax/cm-1: 3479, 3317 (NH2), 3032 (CH aromatic),

2924, 2846 (CH aliphatic), 1685 (2C=O, br.), 1604, 1570 (C=N, C=C).

1

H NMR (DMSO-

d6): δ 2.90 (dd, 1H, C4-HA pyrazoline, JAM= 21.9 and JAX= 7.2 Hz), 3.73 (dd, 1H, C4-HM pyrazoline, JMA= 21.9 and JMX= 12.3 Hz), 3.89 (s, 3H, OCH3), 5.40 (dd, 1H, C5-HX pyrazoline, JXA= 7.2 and JXM= 12.3 Hz), 6.33 (d, 1H, C3-H chromene, J= 9.6 Hz), 7.16-7.30 (m, 5H, ArH), 7.35 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.77 (d, 1H, C5-H chromene, J= 9.0 Hz), 8.03 (d, 1H, C4-H chromene, J= 9.3 Hz), 11.05 (s, 2H, NH2, D2O exch.). Anal. Calcd. for C20H17N3O4 (363.37): C, 66.11; H, 4.72; N, 11.56. Found: C, 66.19; H, 4.84; N, 11.72.

15

4.1.8.3.

3-(7-methoxy-2-oxo-2H-chromen-8-yl)-5-(4-methylphenyl)-4,5-dihydropyrazole-1-

ACCEPTED MANUSCRIPT

carboxamide (10c):

Yield 69%; m.p. 155-156 oC. IR υmax/cm-1: 3491, 3336 (NH2), 3047 (CH aromatic), 2920, 2850 (CH aliphatic), 1678 (2C=O, br.), 1604, 1581 (C=N, C=C). 1H NMR (DMSO-d6): δ 2.31 (s, 3H, CH3), 3.10 (dd, 1H, C4-HA pyrazoline, JAM= 21.9 and JAX = 4.8 Hz), 3.67 (dd, 1H, C4-HM pyrazoline, JMA= 21.9 and JMX= 17.1 Hz), 3.89 (s, 3H, OCH3), 5.25 (dd, 1H, C5HX pyrazoline, JXA= 4.8 and JXM= 17.1 Hz), 6.33 (d, 1H, C3-H chromene, J= 9.6 Hz), 7.08-

RI PT

7.25 (m, 4H, ArH), 7.54 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.81 (d, 1H, C5-H chromene, J= 8.7 Hz), 8.06 (d, 1H, C4-H chromene, J= 9.6 Hz), 10.2 (s, 2H, NH2, D2O exch.). MS, m/z: 377 [M+]. Anal. Calcd. For C21H19N3O4 (377.39): C, 66.83; H, 5.07; N, 11.13. Found: C,

4.1.8.4.

SC

66.95; H, 5.18; N, 11.41.

3-(7-methoxy-2-oxo-2H-chromen-8-yl)-5-[(4-trifluoromethyl)phenyl]-4,5-

M AN U

dihydropyrazole-1-carboxamide (10d):

Yield 70%; m.p. 144-146 oC. IR υmax/cm-1: 3479, 3352 (NH2), 3100 (CH aromatic), 2927, 2850 (CH aliphatic), 1685 (2C=O, br.), 1604, 1573 (C=N, C=C). 1H NMR (DMSO-d6): δ 2.95 (dd, 1H, C4-HA pyrazoline, JAM= 21.3 and JAX= 4.8 Hz), 3.73 (dd, 1H, C4-HM pyrazoline, JMA= 21.3 and JMX= 14.1 Hz), 3.89 (s, 3H, OCH3), 5.45 (dd, 1H, C5-HX pyrazoline, JXA= 4.8 and JXM= 14.1 Hz), 6.33 (d, 1H, C3-H chromene, J= 9.3 Hz), 7.19 (d,

TE D

1H, C6-H chromene, J= 8.7 Hz), 7.55-7.70 (m, 4H, ArH), 7.86 (d, 1H, C5-H chromene, J= 9.0 Hz), 8.05 (d, 1H, C4-H chromene, J= 9.6 Hz), 10.40 (s, 2H, NH2, D2O exch.). 13C NMR (DMSO-d6): δ 45.4 (C-4 pyrazoline), 56.5 (OCH3), 59.1 (C-5 pyrazoline), 108.3 (C-6 chromene), 108.7 (C-3 chromene), 112.6 (C-4a chromene), 112.8 (C-8 chromene), 124.9-

EP

137.5 (aromatic Cs and CF3), 144.3 (C-4 chromene), 145.5 (C-1 of C6H4CF3), 148.0 (C-8a chromene), 152.4 (C=N pyrazoline), 159.6 (CONH2), 160.2 (C-7 chromene), 166.1 (C=O).

AC C

MS, m/z: 431 [M+]. Anal. Calcd. for C21H16F3N3O4 (431.36): C, 58.47; H, 3.74; N, 9.74. Found: C, 58.53; H, 3.82; N, 9.82. 4.1.8.5.

3-(7-methoxy-2-oxo-2H-chromen-8-yl)-5-[(4-methylthio)phenyl]-4,5-

dihydropyrazole-1-carboxamide (10e): Yield 73%; m.p. 161-163 oC. IR υmax/cm-1: 3464, 3278 (NH2), 3070 (CH aromatic), 2974, 2920 (CH aliphatic), 1685 (2C=O, br.), 1600,1570 (C=N, C=C). 1H NMR (DMSO-d6): δ 2.46 (s, 3H, SCH3), 3.35 (dd, 1H, C4-HA pyrazoline, JAM= 19.8 and JAX= 6.9 Hz), 3.78 (dd, 1H, C4-HM pyrazoline, JMA= 19.8 and JMX= 12.1 Hz), 3.89 (s, 3H, OCH3), 5.43 (dd, 1H, C5HX pyrazoline, JXA= 6.9 and JXM= 12.1 Hz), 6.45 (d, 1H, C3-H chromene, J= 9.3 Hz), 7.2316

7.33 (m, 5H, ArH and C6-H chromene), 7.65 (d, 1H, C5-H chromene, J= 8.7 Hz), 7.80 (d,

ACCEPTED MANUSCRIPT

1H, C4-H chromene, J= 9.6 Hz), 10.20 (s, 2H, NH2, D2O exch.). Anal. Calcd. for C21H19N3O4S (409.46): C, 61.60; H, 4.68; N, 10.26. Found: C, 61.72; H, 4.76; N, 10.43. 4.1.9. General procedure for synthesis of 1-[1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3substituted allylidene]semicarbazide (11a-e). The appropriate chalcone 6a-e (1.6 mmol) was dissolved in absolute ethanol (10 ml),

RI PT

semicarbazide hydrochloride (0.4 g; 3.66 mmol) was added and the solution was refluxed for 24 h. The reaction mixture was poured on ice water and the crude product obtained was filtered off, washed with water and left to dry then crystallized from ethanol. 4.1.9.1.

1-[1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3-(thiophen-2-

SC

yl)allylidene]semicarbazide (11a):

Yield 80%; m.p. 128-130 oC. IR υmax/cm-1: 3471, 3348 (NH, NH2), 3086 (CH

M AN U

aromatic), 2927, 2846 (CH aliphatic), 1732 (C=O), 1701 (C=O), 1604, 1562 (C=N, C=C). 1H NMR (DMSO-d6): δ 3.85 (s, 3H, OCH3), 6.31 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.50 (s, 2H, NH2, D2O exch.), 6.74 (d, 1H, N=C-CH=CH, J= 15.9 Hz), 6.85 (d, 1H, N=C-CH=CH, J= 15.9 Hz), 7.15 (d, 1H, C3-H thiophene, J= 5.7 Hz), 7.22 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.42 (d, 1H, C4-H thiophene, J= 5.7 Hz), 7.58 (d, 1H, C5-H thiophene, J= 5.7 Hz), 7.81 (d,

D2O exch.).

13

TE D

1H, C5-H chromene, J= 8.7 Hz), 8.07 (d, 1H, C4-H chromene, J= 9.9 Hz), 8.88 (s, 1H, NH, C NMR (DMSO-d6): δ 56.5 (OCH3), 108.7 (C-8 chromene), 112.7 (C-4a

chromene), 113.0 (C-3 chromene), 113.5 (C-6 chromene), 120.6-139.6 (aromatic Cs), 144.2 (C-8a chromene), 151.1 (C=N), 158.8 (C-7 chromene), 159.5 (CONH2), 160.0 (C=O). MS,

EP

m/z: 369 [M+]. Anal. Calcd. for C18H15N3O4S (369.39): C, 58.53; H, 4.09; N, 11.38. Found: C, 58.86; H, 4.23; N, 11.69.

AC C

4.1.9.2. 1-[1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3-phenylallylidene]semicarbazide (11b): Yield 73%; m.p. 125-127 oC. IR υmax/cm-1: 3471, 3348 (NH, NH2), 3082 (CH

aromatic), 2924, 2850 (CH aliphatic), 1732 (C=O), 1697 (C=O), 1604, 1573 (C=N, C=C). 1H NMR (DMSO-d6): δ 3.88 (s, 3H, OCH3), 6.16 (d, 1H, N=C-CH=CH, J= 16.5 Hz), 6.31 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.47 (s, 2H, NH2, D2O exch.), 7.12-7.33 (m, 6H, ArH and N=C-CH=CH), 7.43 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.87 (d, 1H, C5-H chromene, J= 9.0 Hz), 8.08 (d, 1H, C4-H chromene, J= 9.6 Hz), 8.90 (s, 1H, NH, D2O exch.).

13

C NMR

(DMSO-d6): δ 56.5 (OCH3), 107.2 (C-8 chromene), 108.7 (C-4a chromene), 112.8 (C-3 chromene), 113.4 (C-6 chromene), 126.6-140.2 (aromatic Cs), 144.5 (C-8a chromene), 152.2

17

(C=N), 156.1 (C-7 chromene), 159.4 (CONH2), 160.0 (C=O). Anal. Calcd. for C20H17N3O4

ACCEPTED MANUSCRIPT

(363.37): C, 66.11; H, 4.72; N, 11.56. Found: C, 66.32; H, 4.88; N, 11.91. 4.1.9.3.

1-{1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3-[(4-

methyl)phenyl]allyliden}semicarbazide (11c): Yield 75%; m.p. 210-211 oC. IR υmax/cm-1: 3475, 3340 (NH, NH2), 3086 (CH aromatic), 2924, 2850 (CH aliphatic), 1728 (C=O), 1697 (C=O), 1604, 1566 (C=N, C=C). 1H

RI PT

NMR (DMSO-d6): δ 2.26 (s, 3H, CH3), 3.83 (s, 3H, OCH3), 6.05 (d, 1H, N=C-CH=CH, J= 16.8 Hz), 6.29 (d, 1H, C3-H chromene, J= 9.6 Hz), 6.47 (s, 2H, NH2, D2O exch.), 7.06 (d, 1H, N=C-CH=CH, J= 15.0 Hz), 7.22 (d, 2H, ArH, J= 7.2 Hz), 7.29 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.61 (d, 2H, ArH, J= 7.8 Hz), 7.81 (d, 1H, C5-H chromene, J= 7.8 Hz), 8.05 (d,

SC

1H, C4-H chromene, J= 9.6 Hz), 8.89 (s, 1H, NH, D2O exch.). Anal. Calcd. for C21H19N3O4 (377.39): C, 66.83; H, 5.07; N, 11.13. Found: C, 67.11; H, 5.19; N, 11.54.

1-{1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3-[(4-

M AN U

4.1.9.4.

trifluoromethyl)phenyl]allylidene}semicarbazide (11d):

Yield 68%; m.p. 138-140 oC. IR υmax/cm-1: 3479, 3352 (NH, NH2), 3086 (CH aromatic), 2927, 2850 (CH aliphatic), 1732 (C=O), 1701 (C=O), 1604, 1558 (C=N, C=C). 1H NMR (DMSO-d6): δ 3.83 (s, 3H, OCH3), 6.19 (d, 1H, N=C-CH=CH, J= 15.9), 6.30 (d, 1H,

TE D

C3-H chromene, J= 9.9 Hz), 6.54 (s, 2H, NH2, D2O exch.), 7.21 (d, 1H, C6-H chromene, J= 9.0 Hz), 7.29 (d, 1H, N=C-CH=CH, J= 16.2 Hz), 7.44-7.65 (m, 4H, ArH), 7.85 (d, 1H, C5-H chromene, J= 9.0 Hz), 8.05 (d, 1H, C4-H chromene, J= 9.3 Hz), 9.10 (s, 1H, NH, D2O exch.). MS, m/z: 431 [M+]. Anal. Calcd. for C21H16F3N3O4 (431.36): C, 58.47; H, 3.74; N, 9.74.

4.1.9.5.

EP

Found: C, 58.43; H, 3.81; N, 9.18.

1-{1-(7-Methoxy-2-oxo-2H-chromen-8-yl)-3-[(4-methylthio)phenyl]allylidene}

AC C

semicarbazide (11e):

Yield 83%; m.p. 236-237 oC. IR υmax/cm-1: 3479, 3344 (NH, NH2), 3090 (CH

aromatic), 2924, 2850 (CH aliphatic), 1732 (C=O), 1685 (C=O), 1600, 1573 (C=N, C=C). 1H NMR (DMSO-d6): δ 2.50 (s, 3H, SCH3), 3.84 (s, 3H, OCH3), 6.05 (d, 1H, N=C-CH=CH, J= 16.5 Hz), 6.30 (d, 1H, C3-H chromene, J= 9.3 Hz), 6.49 (s, 2H, NH2, D2O exch.), 7.09 (d, 1H, N=C-CH=CH, J= 15.9 Hz), 7.17-7.26 (m, 4H, ArH), 7.35 (d, 1H, C6-H chromene, J= 8.4 Hz), 7.84 (d, 1H, C5-H chromene, J= 8.7 Hz), 8.06 (d, 1H, C4-H chromene, J= 9.6 Hz), 8.93 (s, 1H, NH, D2O exch.). Anal. Calcd. for C21H19N3O4S (409.46): C, 61.60; H, 4.68; N, 10.26. Found: C, 61.72; H, 4.74; N, 10.48.

18

4.2. Anticancer activity study

ACCEPTED MANUSCRIPT

4.2.1. MTT assay for cell viability HepG2 cells were trypsinized and washed with Ca2+/Mg2+-free PBS (pH 7.2). Cells were adjusted to 4×104 cells/ml with DMEM supplemented with 10% fetal calf serum (Hyclone, Logan, UT,USA) and plated (50 µl/well) in 96-well cell culture plate (Corning, Corning, NY, USA) overnight at 37˚C with 5% CO2 and 95% humidity. Fifty microliters of

RI PT

serial 10-fold diluted sterile tested compounds were added to final concentrations of 0-10µM. Culture medium was used as negative control. Cultures were incubated for 72 h. Supernatants were discarded, 20 µl/well of methylthiazolyldiphenyl-tetrazolium bromide (MTT) reagent (Promega, Madison, WI, USA) was added and incubated for 4 h at 37˚C with 5% CO2. Sterile

SC

sodium dodecyl sulfate (10% v/v in PBS) was added (25 µl/well) and the plate was kept at room temperature for 18 h before measuring optical density (OD) at 490 nm. DMEM was used as a blank control. Results from three separate experiments were recorded and the

M AN U

percentage of viable cells was calculated as [(O.D. of cell control–O.D. of treated cells)/(O.D. of cell control–O.D. of initiated cells)] ×100. 4.2.2. Telomerase inhibition assay

Telomeric repeat amplification protocol-enzyme-linked immunosorbent assay (TRAP-

TE D

ELISA) was performed using the telomerase kit Telo TAGGG Telomerase PCR ELISA PLUS (Roche Diagnostics, Mannheim, Germany) according to the manufacturer’s instructions. In brief, cells were treated with the tested compounds and harvested; approximately 1x106 cells were lysed in 200 µl of lysis reagent and incubated on ice for 30

EP

min. For the TRAP reaction, 2 µl of cell extract (containing 2 µg of protein) was added to 25 µl of reaction mixture with the appropriate amount of sterile water to make a final volume of

AC C

50 ml. PCR was performed in a Mastercycler as follows: primer elongation (30 min, 25 oC), telomerase inactivation (5 min, 94 oC), and product amplification by the repeat of 30 cycles (94 oC for 30 s, 50 oC for 30 s, 72 oC for 90 s). Hybridization and the ELISA reaction were carried out following the manufacturer’s instructions. Staurosporine was used as positive control [52]. Results from three separate experiments were recorded and average percent inhibition was calculated. 4.2.3. Apoptosis study HepG2 cells (1×105 cells/ml) were plated in a 24-well culture plate (Corning) for 24 h at 37˚C with 5% CO2 and 95% humidity. Different concentrations (0- 32 µg/ml) of tested compound 7e were added. Cells were incubated for another 24 h. Cells were trypsinized with 19

0.25% trypsin in Ca2+/Mg2+-free 2% BSA-PBS (Sigma, St. Louis, MO,USA) and washed

ACCEPTED MANUSCRIPT

twice with Ca2+/Mg2+-free PBS. Cells were stained and adjusted to 1×106 cells/ml with annexin V-FITC/PI. The reaction was allowed to proceed in the dark for 30 min at room temperature. Finally, 400 µl of 1× binding buffer was added and flow cytometry carried out within 1 h. References.

RI PT

[1] V. Sekaran, J. Soares, M.B. Jarstfer, J. Med. Chem., 2014 (57) 521–538. [2] W.E. Wright, J.W. Shay, Curr. Opin. Genet. Dev. 11 (2001) 98-103.

[3] X-H. Liu, J. Li, J.B. Shi, B-A. Song, X-B. Qi, Eur. J. Med. Chem., 51 (2012) 294-299.

M AN U

Y. R. Pan, J. J. Lin, J. Med. Chem. 2003 (46) 3300-3307.

SC

[4] H. S. Huang, J. F. Chiou, Y. Fong, C. C. Hou, Y. C. Lu, J. Y. Wang, J. W. Shih,

[5] A. G. Bodnar, M. Ouellette, M. Frolkis, S. E. Holt, C. P. Chiu, G. B. Morin, C. B. Harley, J. W. Shay, S. Lichtsteiner, W. E. Wright, Science, 1998 (279) 349-352. [6] C. Singhapol, D. Pal, R. Czapiewski, M. Porika, G. Nelson, G.C. Saretzki, PLoS ONE 8

TE D

(2013) e52989.

[7] A. Behrenswerth, N. Volz, J. Toräng, S. Hinz, S. Bräse, C.E. Müller, Bioorg. Med. Chem. 17 (2009) 2842-2851.

EP

[8] X. Zhou, X. B. Wang, T. Wang, L.Y. Kong, Bioorg. Med. Chem. 16 (2008) 8011-8021. [9] B.C. Raju, A.K. Tiwari, J.A. Kumar, A.Z. Ali, S.B. Agawane, G. Saidachary, K.

AC C

Madhusudana, Bioorg. Med. Chem. 18 (2010) 358-365. [10] M. Ghate, R.A. Kusanur, M.V. Kulkarni, Eur. J. Med. Chem. 40 (2005) 882-887. [11] Q. Shen, Q. Peng, J. Shao, X. Liu, Z. Huang, X. Pu, L. Ma, Y-M. Li, A.S.C. Chan, L. Gu, Eur. J. Med. Chem. 40 (2005) 1307-1315. [12] G. Sandeep, Y.S. Ranganath, S. Bhasker, N. Rajkumar, Asian J. Res. Chem. 2 (2009) 4648. [13] H. Nie, L.Z. Meng, J.Y. Zhou, X.F. Fan, Y. Luo, G.W. Zhang, Chin. J. Integr. Med. 15 (2009) 442-447. [14] R.S. Thakur, S.C. Bagadia, M.L. Sharma, Experientia 34 (1978) 158-159. 20

[15] E. Quezada, G. Delogu, C. Picciau, L. Santana, G. Podda, F. Borges, V. García-Morales,

ACCEPTED MANUSCRIPT

D. Viña, F. Orallo, Molecules 15 (2010) 270-279.

[16] M. Campos-Toimil, F. Orallo, L. Santana, E. Uriarte, Bioorg. Med. Chem. Lett. 12 (2002) 783-786. [17] E. Budzisz, E. Brzezinska, U. Krajewska, M. Rozalski, Eur. J. Med. Chem. 38 (2003) 597-603.

RI PT

[18] F. Chimenti, B. Bizzarri, A. Bolasco, D. Secci, P. Chimenti, A. Granese, S. Carradori, D. Rivanera, A. Zicari, M. M. Scaltrito, F. Sisto, Bioorg. Med. Chem. Lett. 20 (2010) 49224926.

SC

[19] M. Kawase, H. Sakagami, N. Motohashi, H. Hauer, S.S. Chatterjee, G. Spengler, V.A. Vigyikanne, A. Molnar, J. Molnar, In Vivo 19 (2005) 705-711.

M AN U

[20] I. Kostova, Curr. Med. Chem. 5 (2005) 29-46.

[21] M. Ishihara, Y. Yokote, H. Sakagami, Anticancer Res. 26 (2006) 2883-2886. [22] X. Liu, H. Liu, J. Chen, Y. Yang, B. Song, L. Bai, J. Liu, H. Zhu, X. Qi, Bioorg. Med. Chem. Lett. 20 (2010) 5705-5708.

1035-1044.

TE D

[23] D. Yim, P. R. Singh, C. Agarwal, S. Lee, H. Chi, R. Agarwal, Cancer Res. 65 (2005)

[24] J. Chuang, Y. Huang, H. Lu, H. Ho, J. Yang, T. Li, N. Chang, J. Chung, In Vivo 21

EP

(2007) 1003-1010.

[25] G. Viola, D. Vedaldia, F. dall' Acquaa, G. Bassob, S. Disaro, M. Spinellib, B.

AC C

Cosimellic, M. Boccalinid, S. Chimichid, Chem. Biodivers. 1 (2004) 1265-1280. [26] J.S. Lopez-Gonzaleza, H. Prado-Garciaa, D. Aguilar-Cazaresa, J.A. Molina-Guarnerosc, J. Morales-Fuentesb, J.J. Mandokic, Lung Cancer 43 (2004) 275-283.

[27] J.-Y. Chuang, Y.-F. Huang, H.-F. Lu, H.-C. Ho, J.-S. Yang, T.-M. Li, N.-W. Chang, J.G. Chung, In vivo 21 (2007) 1003-1010.

[28] X. Xu, Y. Zhang, D. Qu, T. Jiang, S. Li, J. Exp. Clin. Canc. Res., 30 (2011) 33.

21

[29] R. Vazquez, M. E. Riveiro, M. Vermeulen, E. Alonso, C. Mondillo, G. Facorro, L. Piehl,

ACCEPTED MANUSCRIPT

N. Gomez, A. Moglioni, N. Fernandez, A. Baldi, C. Shayo, C. Davio, Bioorg. Med. Chem., 20 (2012) 5537-5549.

[30] B.R.V. Avin, P. Thirusangu, V.L. Ranganatha, A. Firdouse, B.T. Prabhakar, S.A. Khanum, Eur. J. Med. Chem. 75 (2014) 211-221.

RI PT

[31] F. Belluti, G. Fontana, L.D. Bo, N. Carenini, C. Giommarelli, F. Zunino, Bioorg. Med. Chem., 18 (2010) 3543-3550.

[32] L. Zhang, G. Jiang, F. Yao, Y. He, G. Liang, Y. Zhang, B. Hu, Y. Wu, Y. Li, H. Liu,

SC

PLOS One 7 (2012) e37865.

60 (2013) 120-127.

M AN U

[33] K.V. Sashidhara, S.R. Avula, K. Sharma, G.R. Palnati, S.R. Bathula, Eur. J. Med. Chem.,

[34] Y. Xia, Z.W. Dong, B.X. Zhao, X. Ge, N. Meng, D.S. Shin, J.Y. Miao, Bioorg. Med. Chem. 15 (2007) 6893-6899.

(2008) 2347-2353.

TE D

[35] Y. Xia, C.D. Fan, B.X. Zhao, J. Zhao, D.S. Shin, J.Y. Miao, Eur. J. Med. Chem. 43

[36] A.M. Farag, A.S. Mayhoub, S.E. Barakat, A.H. Bayomi, Bioorg. Med. Chem. 16 (2008) 881-889.

EP

[37] N.C. Warshakoon, S. Wu, A. Boyer, R. Kawmoto, S. Renock, K. Xu, M. Pokross, A.G. Evdokimov, S. Zhou, C. Winter, R. Walter, M. Mekel, Bioorg. Med. Chem. Lett. 16 (2006)

AC C

5687-5690.

[38] N. Zhang, S. Ayral-Kaloustian, T.J. Anderson, T. Nguyen, S. Das, M.A. Venkatesan, N. Brooijmans, J. Lucas, K. Yu. I. Hollander, R. Mallon, Bioorg. Med. Chem. Lett. 20 (2010) 3526-3529.

[39] M. Shaharyar, M. M. Abdullah, M. A. Bakht, J. Majeed, Eur. J. Med. Chem. 45 (2010) 114–119. [40] D. Havrylyuk, B. Zimenkovsky, O. Vasylenko, L. Zaprutko, A. Gzella, R. Lesyk, Eur. J. Med. Chem. 44 (2009) 1396–1404.

22

[41] M.O. Duffey, R. Adams, C. Blackburn, R.W. Chau, S. Chen, K.M. Galvin, K. Garcia,

ACCEPTED MANUSCRIPT

A.E. Gould, P.D. Greenspan, S. Harrison, S-C. Huang, M-S. Kim, B. Kulkarni, S. Langston, J. X. Liu, L-T. Ma, S. Menon, M. Nagayoshi, R.S. Rowland, T.J. Vos, T. Xu, J.J. Yang, S. Yu, Q. Zhang, Bioorg. Med. Chem. Lett. 20 (2010) 4800–4804. [42] C. Blackburn, O.M. Duffey, E.A. Gould, B. Kulkarni, X.J. Liu, S. Menon, M. Nagayoshi, J.T. Vos, J. Williams, Bioorg. Med. Chem. Lett. 20 (2010) 4795-4799.

RI PT

[43] X. H. Liu, H. F. Liu, J. Chen, Y. Yang, B.A. Song, L.S. Bai, J.X. Liu, H.L. Zhu, X.B. Qi, Bioorg Med. Chem Lett. 20 (2010) 5705-5708.

[44] X.-Q. Wu, C. Huang, Y.-M. Jia, B.-A. Song, J. Li, X.-H. Liu, Eur. J. Med. Chem., 74

SC

(2014) 717-725.

[46] P. Barbier, C.J. Benezra, J. Org. Chem. 48 (1983) 2705-2709.

M AN U

[47] A. Maresca, A. Scozzafava, C.T. Supuran, Bioorg. Med. Chem. Lett. 20 (2010) 72557258.

[48] D. Lakshmi Narayan, S. Nitai Chand, S. Asoke Kumar, Ind. J. Chem. 27 B (1988) 801824.

TE D

[49] V.K. Ahluwalia, N. Lalita, A.K. Tehim, Ind. J. Chem. 27 B (1988) 70-71. [50] K.M. Amin, F.M. Awadallah, A.M. Eissa, S.M. Abou-Seri, G.S. Hassan, Bioorg. Med. Chem. 19 (2011) 6087-6097.

EP

[51] T. Mosmann, J. Immunol. Methods, 65 (1983) 55-63. [52] Y.-B. Zhang, X.-L. Wang, W. Liu, Y.-S. Yang, J.-F. Tang, H.-L. Zhu, Bioorg. Med.

AC C

Chem. 20 (2012) 6356-6365.

23

ACCEPTED MANUSCRIPT

Figure captions:

Figure 1. Examples of some coumarin-based anticancer agents. Figure 2. Examples of some pyrazoline-based anticancer agents. Figure 3. Examples of coumarin-pyrazoline hybrids of anticancer activity.

RI PT

Figure 4. Structures of target compounds 7a-f-11a-f. Figure 5. Flow cytometry histograms of HepG2 cells after 24 h treatment with different concentrations of compound 7e.

Scheme 1. Preparation of the key intermediates 6a-f.

SC

Scheme captions:

Table captions:

M AN U

Scheme 2. Synthesis of the target compounds 7a-f – 11a-f.

Table 1: Cytotoxic activity of the newly synthesized compounds against HepG2 hepatocellular carcinoma cells.

AC C

EP

TE D

Table 2. Telomerase inhibitory effect of compounds7a, 7d, 7e, 8a and 10e.

24

ACCEPTED MANUSCRIPT

Table 1.

N N R

O

H3CO N NH

O Ar

R

7a

H

7b

H

7c

H

7d

H

7e

H

8a

-C6H5

8e 9a

IC50 (nM) ± SEM*

Ar S

M AN U

11±0.6

48±1.1

CH3

TE D

CF3

SCH3

EP

S

-C6H5

43±1.5

13±0.8 10±0.5 15±0.9 73±2.1

-C6H5

AC C

8d

Ar

11a-e

Compound

8c

O

NH2

7a-e - 10a-e

8b

O

RI PT

O

SC

H3CO

CH3

51±1.5

-C6H5

CF3

38±1.1

-C6H5

SCH3

78±2.6

COCH3

S

75±2.8

ACCEPTED MANUSCRIPT

9b

COCH3

9c

COCH3

9d

COCH3

CF3

80±3.4

9e

COCH3

SCH3

42±2.3

10a

CONH2

10b

CONH2

10c

CONH2

10d

CONH2

10e

CONH2

11a

-

11b

-

11c

-

S

50±1.5

RI PT

CH3

SC

26±1.4

90±4.3

46±2.4

CF3

96±6.3

SCH3

18±1.3

M AN U CH3

S

TE D

75±3.8

EP

CH3

-

AC C

11d

52±2.0

82±6.7 93±5.9

CF3

96±7.2

SCH3

99±8.6

11e

-

DOX

-

-

630±34.6

Osthole I [32]

-

-

161.4 (µM)

VI [44]

-

-

6.64±0.4 (µM)

*IC50 values were calculated from the mean values of data from three separate experiments

ACCEPTED MANUSCRIPT

Table 2. % reduction in telomerase activity at conc. 1 µM over time in days ± SEM*

% reduction in telomerase activity at conc. 2 µM over time in days ± SEM

% reduction in telomerase activity at conc. 4 µM over time in days ± SEM

1

2

3

1

2

3

1

7a

18.6±1.2

21.5±2.1

28.9±2.5

22.3±2.1

33.1±2.9

44.6±3.7

7d

17.5±1.5

20.2±1.9

27.2±2.4

21.0±1.7

31.2±2.6

42.0±3.8

7e

19.4±1.3

22.4±2.1

30.1±2.8

23.2±2.1

34.5±2.9

46.45±3.5

8a

16.8±0.9

19.4±1.6

26.1±2.3

20.1±1.7

30.0±2.1

40.3±3.6

10e

15.2±1.1

17.6±1.4

23.6±1.8

18.2±1.3

27.1±2.4

36.5±2.8

Staurosporine

10.7±1.0

11.3±0.9

15.4±1.2

12.7±1.1

18.9±1.3

RI PT

28.7±2.6

M AN U TE D EP

3

62.6±5.4

75.5±6.8

29.2±2.4

59.0±4.6

71.1±6.9

32.3±3.1

65.23±5.6

78.62±6.4

28.0±2.5

56.6±5.2

68.3±6.4

25.3±2.1

51.2±4.8

61.7±5.8

30.1±2.5

40.1±3.7

56.5±5.1

* Values were calculated from the mean values of data from three separate experiments.

AC C

2

31.0±2.7

SC

Compound

ACCEPTED MANUSCRIPT

CH3 H3CO

O

HN

O

S

N H

OCH3

Osthole I

II

N N

OH

NH2

N

M AN U

F

SC

Figure 1. N

N N

O

O

S

O

N

IV

TE D

III

AC C

EP

Figure 2.

O

O CH3

N N

N N

O

O

V Figure 3.

O

RI PT

O

OH

OCH3 OCH3

COOC2H5

O

O

VI

S

ACCEPTED MANUSCRIPT

O N N R

O

H3CO

O

O

O Ar

O

10a-f

NH

Ar

11a-f

EP

TE D

M AN U

SC

Figure 4.

AC C

O

NH2

NH2

3

H C O C , 5 H 6 C , H = R 7a-f - 9a-f

H3CO N

N N Ar

O

RI PT

H3CO

Control

PI

ACCEPTED MANUSCRIPT

Annexin FITC 0.01 µmol/ml

Annexin FITC

TE D

AC C

40.96%

0.08 µmol/ml

PI

EP

PI

Figure 5.

35.00%

Annexin FITC

0.04 µmol/ml

Annexin FITC

SC

PI

0.02 µmol/ml

M AN U

PI

25.45%

RI PT

0.54%

46.85%

Annexin FITC

SC

RI PT

ACCEPTED MANUSCRIPT

Scheme 1.

AC C

EP

TE D

M AN U

Reagents and conditions: (i) acetic anhydride / reflux 5h; (ii) AlCl3 / heat 145oC 1h; (iii) CH3I / dry actone / reflux 24h; (iv) Ar-CHO, 10% NaOH / ethanol / r.t. 24 h.

Scheme 2. Reagents and conditions: (i) hydrazine hydrate / absolute ethanol / reflux 8h; (ii) phenyl hydrazine /absolute ethanol / reflux 48h; (iii) hydrazine hydrate/ absolute ethanol / gl. acetic acid/ reflux 18h; (iv) semicarbazide HCl /NaOH/ absolute ethanol/ reflux 12h; (v) semicarbazide HCl/ absolute ethanol/ reflux 24h.

ACCEPTED MANUSCRIPT Highlights: • •

AC C

EP

TE D

M AN U

SC

RI PT

• •

Coumarin-pyrazoline hybrids were synthesized. Compounds were evaluated for their anticancer activity on HepG2 human cell line. Compounds inhibited telomerase up to 78.6 %. The most active compound, 7e, induced apoptosis in a dose dependent manner.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Synthesis and anticancer activity of some 8-substituted-7-methoxy-2H-chromen-2-one derivatives toward hepatocellular carcinoma HepG2 cells.

Based on the reported anticancer activity of coumarin and pyrazoline derivatives, the present investigation dealt with the design and synthesis of cou...
2MB Sizes 0 Downloads 5 Views