Accepted Manuscript Synthesis, docking and ADMET studies of novel chalcone triazoles for anti-cancer and anti-diabetic activity Yakaiah Chinthala, Sneha Thakur, Shalini Tirunagari, Srinivas Chinde, Anand kumar Domatti, Niranjana Kumar Arigari, K.V.N.S. Srinivas, Sarfaraz Alam, J. Kotesh Kumar, Feroz Khan, Ashok Tiwari, Paramjit Grover PII:

S0223-5234(15)00123-3

DOI:

10.1016/j.ejmech.2015.02.027

Reference:

EJMECH 7709

To appear in:

European Journal of Medicinal Chemistry

Received Date: 1 December 2014 Revised Date:

22 January 2015

Accepted Date: 18 February 2015

Please cite this article as: Y. Chinthala, S. Thakur, S. Tirunagari, S. Chinde, A.k. Domatti, N.K. Arigari, K.V.N.S. Srinivas, S. Alam, J.K. Kumar, F. Khan, A. Tiwari, P. Grover, Synthesis, docking and ADMET studies of novel chalcone triazoles for anti-cancer and anti-diabetic activity, European Journal of Medicinal Chemistry (2015), doi: 10.1016/j.ejmech.2015.02.027. 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

RI PT

Leave this area blank for abstract info.

Synthesis, docking and ADMET studies of novel chalcone triazoles for anti-cancer and anti-diabetic activity

AC C

EP

TE D

M AN U

SC

Yakaiah Chinthala, Sneha Thakur, Shalini Tirunagari, Srinivas Chinde, Anand kumar Domatti, Niranjana Kumar Arigari, Srinivas K.V.N.S., Sarfaraz Alam, J. Kotesh Kumar*, Feroz Khan, Ashok Tiwari, Paramjit Grover

ACCEPTED MANUSCRIPT

Synthesis, docking and ADMET studies of novel chalcone triazoles for anti-cancer and anti-diabetic activity Yakaiah Chinthalaa, Sneha Thakura, Shalini Tirunagaria, Srinivas Chindeb, Anand kumar

RI PT

Domattic, Niranjana Kumar Arigaria, Srinivas K.V.N.S. a, Sarfaraz Alamd, J. Kotesh Kumara*, Feroz Khand, Ashok Tiwaric, Paramjit Groverb

Natural Product Chemistry, CSIR-Central Institute of Medicinal and Aromatic PlantsResearch Centre, Boduppal, Hyderabad-500092, India b Toxicology Unit, Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India. c Medicinal chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad – 500007, India. d Metabolic and Structural Biology Department, CSIR-CIMAP, Lucknow-226015, UP

*

M AN U

SC

a

Corresponding: Telefax: +91-40-27202602; Email: [email protected]

Abstract

AC C

EP

TE D

A series of novel chalcone-triazole derivatives were synthesized and screened for in vitro anticancer activity on the human cancer cell lines IMR32 (neuroblastoma), HepG2 (Human hepatoma) and MCF-7 (Human breast adenocarcinoma), DU-145 (Human prostate carcinoma), and A549 (Human Lung adenocarcinoma). Among the tested compounds, 4r showed the most promising anticancer activity in all the cell lines whereas, compounds 4c (IC50 65.86 µM), 4e (IC50 66.28 µM), 4o (IC50 35.81 µM), 4q (IC50 50.82 µM) and 4s (IC50 48.63 µM) showed better activity than the standard doxorubicin (IC50 69.33 µM) in A549 cell line alone. Rat intestinal αglucosidase inhibitory activity of the synthesized derivatives showed 4m (IC50 67.77 µM), 4p (IC50 74.94 in µM) and 4s (IC50 102.10 µM) as most active compared to others. The in silico docking of synthesized derivatives 4a-4t with DNA topoisomerase IIα revealed the LibDock score in the range of 71.2623 to 118.29 whereas, compounds 4h, 4m, 4p and 4s with docking target α-glucosidase were in the range of 100.372 to 107.784. Key words:

Chromanochalcones, 1,2,3-triazoles, Anticancer activity, α-Glucosidase inhibition, Molecular docking. 1. Introduction The World Health Organization’s cancer agency warns that there will be 22 million new cases of cancer every year within the next two decades. Report from the International Agency for 1

ACCEPTED MANUSCRIPT

Research on Cancer (IARC) estimated in 2012 that there were 14 million new cases but predicted that the figure would jump significantly due to global ageing and the spread of cancers to developing countries. Cancer, a diverse group of diseases characterized by uncontrolled growth of abnormal cells and it is a fatal disease standing next to the cardiovascular disease in

RI PT

terms of morbidity and mortality. Although the cancer research has led to a number of new and effective solutions, the medicines used as treatments have clear limitations and unfortunately cancer is projected as the primary cause of death in the future [1, 2]. Currently there is a huge scientific and commercial interest in the discovery of potent, safe and selective anticancer drugs.

SC

Diabetes Mellitus (DM) is also a growing health problem and according to WHO reports, around 250 million people are currently living with diabetes and this number is expected to be

M AN U

more than 366 million by 2030 [3]. Treatments of type 2 diabetes mellitus (T2DM) include improvement of insulin sensitivity or reducing the rate of carbohydrate absorption from the gastrointestinal tract. So far, the drugs used to treat T2DM have several side effects, especially for those patients with liver and renal functional disorders [4]. α-glucosidase is a membranebound enzyme at the epithelium of the small intestine and plays a key role in carbohydrate digestion. Inhibition of α-glucosidase leads to the delay or reduction of increased postprandial

TE D

blood glucose levels. Thus, α-glucosidase inhibitors have been proposed as a potential therapeutic target for drug discovery in the treatment of T2DM. Chalcones (1,3-diaryl-2-propen-1-ones) constitute an important class of natural products belonging to the flavonoid family, which display interesting biological activities including anti-

EP

inflammatory [5], antibacterial [6], antioxidant [7], antimalarial [8] and anticancer [9]. Due to their abundance in plants and ease of synthesis, this class of compounds has generated great

AC C

interest for possible therapeutic uses. They are also effective in vivo as cell proliferating inhibitors, anti-tumor promoting and chemo preventing agents. Since a number of clinically useful anticancer drugs have genotoxic effects due to interaction with the amino groups of nucleic acids, chalcones may be devoid of this important side effect [10]. Despite the comprehensive biological studies on chalcones, reports on their anti-diabetic activity are scarce [11]. On the other hand, the 1,2,3-Triazoles have occupied an important role not only in organic chemistry but also in medicinal chemistry due to their easy synthesis by click chemistry and attractive features as well as numerous biological activities [12-14]. 1,2,3-triazoles are highly 2

ACCEPTED MANUSCRIPT

stable under acid/base hydrolysis and oxidative/reductive conditions, indicating its high aromatic stabilization [15,16]. Moreover, these heterocyclic compounds have a high dipole moment and is capable of forming hydrogen bonding, which could be favorable in the binding of biomolecular targets [17]. 1,2,3-Triazole is one of the key structural units found in a large variety of bioactive

RI PT

molecules as antifungal [18], antibacterial [19,20], antiallergic [21], antiHIV [22,23], antitubercular [24,25] and antiinflammatory [26] agents. Several 1,2,3-triazole containing drug molecules including tazobactum [27], carboxyamidotriazole [28] are now available in the

SC

market. In recent year, researchers are increasingly focusing on their anticancer activity [29-37].

In the design of new drugs, the development of hybrid molecules through the combination of

M AN U

different pharmacophores may lead to compounds with interesting biological profiles. These agents show different mechanisms of action and a single molecule containing more than one pharmacophore, each with different mode of action could be beneficial for the treatment. Hybrid chalcones designed by chemically linking chalcones to other prominent anticancer scaffolds such as pyrrol[2,1-c][1,4]benzodiazepines [38], benzopyran [39] have demonstrated synergistic or additive pharmacological activities. By combining 1,2,3-triazole with other pharmacophores via.

TE D

click chemistry, potential bioactive compounds could be synthesized [40]. Thus, a series of 1,2,3-triazoles hooked to β-lactam chalcones were designed and synthesized to yield novel bioactive hybrids. The synthesized derivatives were screened for their anticancer and antidiabetic

EP

properties in in vitro modes. 2. Results and Discussion Chemistry

AC C

2.1.

The synthetic strategy followed for the synthesis of different chalcone triazoles is outlined in

(schem-1). A series of derivatives 4a-4t were synthesized in four steps. Chromans 2a, 2b were prepared by reaction between isoprene and 2,4-dihydroxy Acetophenone. These intermediates 2a, 2b were common to all molecules being synthesized. In the second step, 4-hydroxy benzaldehyde was reacted with propargylbromide in presence of potassium carbonate at reflux temperatures to obtain compound 3. In the third step, chromans were condensed with 4-(prop-2ynylony)-benzaldehyde 3 in the basic medium to give respective chalcones 3a and 3b. In the final step, the chalcones 3a and 3b were further reacted with appropriately substituted aromatic 3

ACCEPTED MANUSCRIPT

azides by a click reaction to afford compounds 4a-4t. All the synthesized compounds were characterized by 1H,

13

C-NMR, Mass and IR spectroscopy. In the 1H-NMR the presence of

characteristic singlet at δ 2.73 (t, 2H, J = 6.7 Hz) for aliphatic proton provided evidence for formation of 2a, 2b. Formation of the chalcones was characterized by a peculiar doublet for

RI PT

olefin protons resonating at δ 7.47-7.50 ppm. Also, formation of chalcones-triazoles hybrids 4a4t was confirmed by presence of a singlet for O-CH2 protons resonating at δ 5.3 ppm. The IR spectra of compounds 4a-4t showed the enolate protons and C=O stretching bands at 3460-3446

SC

cm-1 and 1735-1670 cm-1 respectively.

2.2.

Anticancer activity

M AN U

Scheme 1: Synthesis of Chalcone 1,2,3-triazole derivatives

Anti-cancer activity of the synthesized chalcone-triazole derivatives was evaluated in in vitro mode using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay [41] on the human cancer cell lines MCF-7 (Human breast adenocarcinoma), DU-145 (Human prostate carcinoma), IMR-32 (neuroblastoma), A-549 (Human lung adenocarcinoma) and Hep-G2

TE D

(Human hematoma). The assay was dependent on the reduction of tetrazolium salt by the mitochondrial dehydrogenase of viable cells to form a blue formazan product dissolved in DMSO and measured at 570 nm. The results of cytotoxic activity were expressed as the IC50

EP

(µM) and doxorubicin was used as positive control Table 1. The structural diversity in all the derivatives was introduced by varying substitution at 3 position of the triazole ring while keeping the chalcone-triazole moiety intact. As given in the Table 1, most of the compounds showed

AC C

moderate anticancer activity but, compound 4r significantly inhibited the growth of all the cell lines when compared to its positional isomer 4o. Similarly, compounds 4c (IC50 65.86 µM), 4e (IC50 66.28 µM), 4o (IC50 35.81 µM), 4q (IC50 50.82 µM) and 4s (IC50 48.63 µM) were behaved better than the standard doxorubicin (IC50 37.65 µM) in only A549 cell lines.

2.3. α-glucosidase inhibition Rat Intestinal α-glucosidase inhibitory activity was determined as per earlier reported methods [42,43]. Rat intestinal acetone powder in normal saline (100:1; w/v) was sonicated 4

ACCEPTED MANUSCRIPT

properly and the supernatant was used as a source of crude intestinal α-glucosidase after centrifugation. In brief, 10 mL of test samples (5 mg/mL DMSO solution) were reconstituted in 100 mL of 100 mM phosphate buffer (pH 6.8) in 96-well microplate and incubated with 50 mL yeast α-glucosidase (0.76 U/Ml in same buffer) or crude intestinal α-glucosidase for 5 min before

RI PT

50 mL substrate (5 mM, p-nitrophenyl-α-D-glucopyranoside prepared in same buffer) was added. Release of p-nitrophenol was measured at 405 nm spectrophotometrically (SpectraMAx Plus384, Molecular Devices Corporation, Sunnyvale, CA, USA) 5 min after incubation with substrate. Individual blanks for test samples were prepared to correct back-ground absorbance

SC

where substrate was replaced with 50 µL of buffer. Control sample contained 10 µL DMSO in place of test samples. Acarbose was taken as standard reference for α-glucosidase inhibition. All

M AN U

the samples were studied in triplicate. Percentage of enzyme inhibition was calculated as (1B/A) x 100 where A represents absorbance of control without test samples, and B represents absorbance in presence of test samples. All the tests were run in duplicate. The IC50 values were calculated applying suitable regression analysis from the mean inhibitory values. As shown in Table 1, amongst all the synthesized derivatives, compounds 4m (IC50 67.77 µM), 4p (IC50 74.94 in µM) and 4s (IC50 102.10 µM) have shown better α-glucosidase enzyme inhibitory activity.

TE D

Comparing the α-glucosidase inhibitory activity of compounds it is inferred that the presence of aliphatic chain length up to five carbons attached to the triazole moiety may be ideal to generate activity.

EP

Table 1: Anticancer and α-glucosidase inhibitory activity of compound 4a-4t.

AC C

2.4. Molecular docking and in silico pharmacology studies Protein–ligand docking studies The docking study of chalcone derivatives 4a-4t with DNA topoisomerase IIα (PDB:

1ZXM) revealed the high docking score (LibDock) and binding affinities [44], in the range of 71.2623 to 118.29, as compared to Doxorubicine 125.857 (Fig. 1, Table 2 and Fig S1 (in Supplementary)). Similarly, chalcone triazoles, which were active in vitro, have shown docking scores in the range of 100.372 to 107.784 (Fig. 2, Table 3 and Fig. S2 (in Supplementary)) when docked with α-Glucosidase (PDB: 2QMJ). These results indicate that most of the compounds 5

ACCEPTED MANUSCRIPT

bound within the binding site pocket of doxorubicin and similar binding pattern with binding site amino acid residues. Few compounds bound at different binding site of DNA topoisomerase IIα. In case of docking with α-glucosidase, compound 4m showed the similar binding pattern as acarbose, a known antidiabetic drug. On the other hand compounds 4d, 4h, and 4s showed

RI PT

diverse binding site locations. The significant binding affinity on respective targets infers that these compounds are very active and can be potential leads against cancer and diabetes. These results indicate that the compounds are bound well within the binding site pocket of doxorubicin and almost similar binding pattern were identified. In case of docking with α-

SC

glucosidase, all the studied compounds showed similar docking results quantitatively as compared to in vitro activity. Detailed analysis of the results showing various interactions, such

M AN U

as hydrogen bonds, atomic charge interactions and Pi interactions between the surrounding residues and the ligand were mapped. These interactions are displayed with 2D diagram and represented by different colors like pink indicates electrostatic interaction; purple indicates covalent bond and green indicates van der–Waals molecular interaction. Solvent accessibility of the ligand atom and the amino acid residues are shown in light blue shading surrounding the atom or residue. High shading indicates more exposure to solvent. The significant docking score

TE D

on the respective targets infers that these compounds are promiscuous and can be potential leads against cancer and diabetes.

EP

Fig. 1 Structural model of DNA topoisomerase II α (PDB: 1ZXM) with doxorubicin binding site (sphere); (B) Binding site and binding pattern of candidate compound along with control.

AC C

Fig. 2 Structural model of α-glucosidase (PDB: 2QMJ) with Acarbose binding site (sphere). (B) Binding site and binding pattern of candidate compound along with control. Table 2: Details of LibDock score, hydrogen bonds, Pi interactions and active site pocket residues revealed through molecular docking of Chalcone derivatives and Doxorubicin on DNA Topoisomerase IIα (PDB: 1ZXM). Table 3: Details of LibDock score, hydrogen bonds, Pi interactions and active site pocket residues revealed through molecular docking of Chalcone derivatives and Acarbose on α-glucosidase (PDB: 2QMJ)

6

ACCEPTED MANUSCRIPT

Bioavailability and drug likeness screening Different chemical descriptors for the pharmacokinetics properties were calculated to check the compliance of studied compound with the standard range. For this the aqueous

RI PT

solubility, blood–brain barrier penetration, cytochrome P450 2D6 binding, hepatotoxicity, intestinal absorption, and plasma protein binding were calculated [45]. The calculation of these chemical properties was intended as the first step toward analyzing the novel chemical entities, in order to check the failure of lead candidates which may cause toxicity or metabolized by the body in to an inactive form or one unable to cross the intestinal membranes. The

SC

pharmacokinetic profiles of all the compounds under investigation were predicted by means of six pre-calculated ADMET models and result provided in Table S1 in supplementary. The

M AN U

aqueous solubility prediction (defined in water at 25°C) indicated that all the compounds are soluble in water. The compounds are found to be non-inhibitors of cytochrome P450 2D6 (CYP2D6). The CYP2D6 enzyme is one of the important enzymes involved in drug metabolism. During ADME screening, the predictive hepatotoxicity was observed for some of the candidate compounds in comparison to acarbose (non-toxic) and doxorubicin (toxic). The candidate

and doxorubicin.

TE D

compounds are found to be highly bound with plasma protein binding in comparison to acarbose

Prediction of toxic effect

The safety of the compounds is absolutely crucial for a successful drug. To grab this, the

EP

different toxicity risk screening such as skin irritancy, ocular irritancy, mutagenicity, aerobic biodegradability and developmental toxicity potential was checked for all the compounds (Table

AC C

S2, in supplementary). All the compounds are found to be non-mutagenic and either mild or no irritancy (Ocular & Skin) when compared to doxorubicin which is mutagenic and acarbose which shows severe ocular irritancy. The dose dependent toxicity such as Rat Oral LD50, Rat Inhalational LC50, Carcinogenic Potency TD50, Rat Maximum Tolerated Dose feed, Daphnia EC50 (mg/l), Rat Chronic LOAEL along with effective concentration extent are provided for each compound in Table S2. These results will help to set dose-ranges for animal assays. 3. Conclusion

7

ACCEPTED MANUSCRIPT

Synthesis of a series of novel hybrid molecules consisting of biologically important pharmacophores Chromano chalcones tagged with substituted 1,2,3-triazoles showed potential anticancer and antidiabetic activity. Among the synthesized compounds, 4r showed the most promising anticancer activity in all the cell lines and about nine compounds out twenty

RI PT

derivatives were active against A-549 cell line. Compounds 4m, 4p and 4s showed promising αglucosidase inhibitory activity. The in silico docking with DNA topoisomerase IIα and αglucosidase targets and toxicity studies revealed high binding affinities and within toxicity limits

4.

SC

required in the drug discovery procedure.

Experimental

M AN U

4.1. Chemistry

Melting points of all the compounds were recorded on Casia-Siamia (VMP-AM) melting point apparatus and uncorrected. IR spectra were recorded on a Perkin–Elmer FT-IR 240-C spectrometer using KBr optics. NMR spectra were recorded on Burker Avance 300 MHz in CDCl3 and DMSO-d6 using TMS as internal standard. Electron impact (EI) and chemical ionization mass spectra were recorded on a VG Micro mass model 7070H instrument. All the

TE D

reactions were monitored on silica gel percolated TLC plates of Merck and spots were visualized with UV light. Silica gel (100-200 mesh) used for column chromatography was procured from Merck.

4.1.1. Synthesis of 1-(5-hydroxy-2, 2-dimethyl-3, 4-dihydro-2H-chromen-6-yl) ethanone (2a)

procedure

EP

and 1-(7-hydroxy-2, 2-dimethyl-3, 4-dihydroxy-2H-chromen-6-yl) ethanone (2b) General

AC C

1-(5-hydroxy-2, 2-dimethyl-3, 4-dihydro-2H-chromen-6-yl) ethanone (2a) and 1-(7hydroxy-2,2-dimethyl-3,4-dihydroxy-2H-chromen-6-yl)ethanone(2b) to a stirred solution of Amberlyst-15 (6.2 gm) and 1-(2,4-dihydroxyphenyl)-ethanone 1 (4.56 gm, 30 mmol) in THF (10 ml) at 65-70 °C, isoprene (3.2 ml, 4.7 mmol) in heptane (10 ml) was added drop wise over a period of 2h. The reaction mixture was filtered and washed with hot acetone (2 × 50 ml) and separated by column chromatography using hexane/ethyl acetate (8:2 and 6:4) as eluent to afford 2a (2.8 g, 43%) and 2b (0.95 g, 15%) in pure state. 1-(5-hydroxy-2, 2-dimethylchroman-6-yl) ethanone 2a White solid, m.p. 70 °C, IR (KBr): 3429, 2967, 2925, 2855, 1629, 1489, 1427, 1363, 1263, 1154, 1067, 614 cm-1; 1H-NMR (CDCl3, 200 8

ACCEPTED MANUSCRIPT

MHz) δ (ppm): 13.11(s, 1H, chelated OH), 7.49 (d, 1H, J = 8.9 Hz,), 6.33 (d, 1H, J = 8.9 Hz), 2.68 (t, 2H, J = 6.7 Hz), 2.54 (s, 3H, COCH3), 1.80 (t, 2H, J = 6.7 Hz), 1.34 (s, 6H). 1-(7-hydroxy-2, 2-dimethylchroman-6-yl) ethanone 2b White solid, m.p. 118 °C, IR (KBr): 3429, 2927, 2856, 1642, 1494,1365, 1281, 1159, 1056, 811, 655 cm-1; 1H-NMR (CDCl3, 200

2.53 (s, 3H, COCH3), 1.82 (t, 2H, J = 6.7 Hz), 1.34 (s, 6H).

RI PT

MHz) δ (ppm): 12.32 (s, 1H, chelated OH), 7.43 (s,1H), 6.31 (s,1H), 2.73 (t, 2H, J = 6.7 Hz),

4.1.2. Procedure for propargylation for synthesis of 4-(prop-2-ynylony) benzaldehyde

4-hydroxy benzaldehyde (1.0 mol) along with 1.5 mol of potassium carbonate was taken

SC

in dimethyl formamide (DMF) and added to this solution of propargyl bromide (1.5 mol) in DMF. Reaction mixture was stirred (reflux) to afford crude 4-(prop-2-ynylony) benzaldehyde.

M AN U

This crude compound was column chromatographed over silica gel (100-200 mesh) and eluted with 15 % ethyl acetate in n-hexane to obtain pure compound 3.

4-(prop-2-ynylony) benzaldehyde 3 White solid, yield: 92%; m.p.76-78 °C, 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.25 (s, 2H), 2.57 (s, 1H), 4.29-4.79 (s, 2H), 7.09-7.11 (d, 1H), 7.86-7.88 (d, 1H), 9.99 (s, CHO).

4.1.3. Procedure for the synthesis of chalcones 3b-(E)-1-(7-hydroxy-2, 2-dimethylchroman-6-

TE D

yl)-3-(4-(prop-2-ynyloxy) phenyl) prop-2-en-1-one

To a solution of chromanes 2a/2b (5 g, 22.7 mol) was added KOH (3.63 g, 1 mol) and a solution of 3 (1.9 g, 1.5 mol) in ethanol (5 mL), and the mixture was stirred at 35-40 °C for 6h. After dilution with water (100 mL) and acidification with cold diluted hydrochloric acid (25 mL)

EP

the resulting solid filtered off, washed with water and recrystallized form petroleum ether to give yellow needles 3a/3b.

AC C

(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-(prop-2-ynyloxy)phenyl)prop-2-en-1-one (3a) Yellow solid, yield: 85%; m.p.124-125 °C; IR (KBr) 3450, 2976, 2934, 1630, 1561, 1363, 1216, 1170, 1021 cm-1; 1H NMR (CDCl3, 300 MHz) δ (ppm): 1.365 (s, 6H), 1.812-1.846 (t, 2H), 2.55-2.565 (t, 1H), 2.704-2.740 (t, 2H), 4.74-4.74 (d, 2H, J = 2.510 Hz), 6.369-6.392 (d, 1H, J = 9.035 Hz), 7.00-7.03 (d, 2H, J = 9.035 Hz), 7.466-7.50 (d, 1H, J = 15.53 Hz), 7.60-7.62 (d, 2H, J = 9.288 Hz), 7.68-7.70 (d, 1H, J = 9.288 Hz), 7.82-7.86 (d, 1H, J = 15.560 Hz), 13.92 (s, 1H). 13

C NMR (75 Hz, CDCl3) δ (ppm): 16.37, 26.74, 31.87, 55.86, 75.81, 76.02, 76.71, 78.01,

106.14, 109.34, 112.86, 115.33, 118.66, 128.46, 128.54, 130.17, 143.41, 159.45, 160.77, 164.11, 191.81. ESI-MS (m/z) 363 (M+ 1) observed for C23H22O4. 9

ACCEPTED MANUSCRIPT

(E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-(prop-2-ynyloxy)phenyl)prop-2-en-1-one (3b) Yellow solid, yield: 85%; m.p.130-132 °C; IR (KBr) 3416, 3231, 2923, 1638, 1553, 1365, 1224, 1149, 1027 cm-1; 1H NMR (CDCl3, 300 MHz) δ (ppm): 1.36 (s, 6H), 1.82-1.86 (t, 2H), 2.55-2.57 (t, 1H), 2.75-2.79 (t, 2H), 4.73-4.74 (d, 2H, J = 2.510 Hz), 6.35 (s, 1H), 7.00-7.02 (d,

RI PT

2H, J = 9.035 Hz), 7.44-7.48 (d, 1H, J = 15.81 Hz), 7.61-7.63 (d, 3H, J = 9.53 Hz), 7.80-7.84 (d, 1H, J = 15.560 Hz), 13.06 (s, 1H). 13C NMR (75 Hz, CDCl3) δ (ppm): 21.81, 26.99, 32.78, 55.86, 75.95, 76.05, 76.73, 78.01, 104.89, 112.68, 114.23, 115.33, 118.60, 128.50, 130.21, 131.06, 143.56, 159.48, 161.41, 164.13, 191.68. ESI-MS (m/z) 363 (M+ 1) observed for C23H22O4.

SC

4.1.4. Synthesis of Synthesis of Chalcones-Triazole derivatives (4a-4t) Intermediate,

(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-(prop-2-ynyloxy)

M AN U

phenyl)-prop-2-en-1-one, 3a/3b (1.0 mmol) was dissolved in dry THF (10 mL) and catalytic amount of copper iodide was added. To this, substituted aromatic and aliphatic azides (1.0 mmol), in dry THF, were added slowly while stirring at room temperature under nitrogen atmosphere for 12 h. Later, the solvent was removed under reduced pressure and the residue was diluted with distilled water and extracted thrice with dichloromethane. The combined organic layers were dried over anhydrous Na2SO4 and concentrated to get the product. The crude product

TE D

was purified by column chromatography with ethyl acetate in hexane.

4.1.5.(E)-3-(4-((1-(4-chlorophenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(7-hydroxy-2,2dimethylchroman-6-yl)prop-2-en-1-one (4a) Yellow solid, yield: 92%, m.p.171-172 °C, IR

EP

(KBr) 3446, 2927, 2866, 1603, 1508,1385, 1247, 1172, 1021, 811, 653 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.381 (s, 6H), 1.821-1.866 (t, 2H), 2.714-2.756 (t, 2H), 5.35 (s, 2H), 6.380-

AC C

6.410 (d, 1H, J = 9.042 Hz), 7.060-7.086 (d, 2H, J = 8.65 Hz), 7.468-7.535 (m, 3H), 7.618-7.647 (d, 2H, J = 8.655 Hz), 7.687-7.697 (d, 2H, J = 3.20 Hz) , 7.71 -7.72 (d, 1H, J = 23.20 Hz) , 7.827.87 (d, 1H, J = 15.447 Hz), 8.068 (s, 1H) ,13.92 (s, 1H).

13

C-NMR (CDCl3,75 Hz) δ (ppm):

16.36, 26.74, 29.70, 31.86, 62.00, 75.81, 109.15, 109.34, 112.84, 115.22, 118.62, 120.91, 121.76, 128.41, 128.46, 130.01, 130.32, 134.82, 135.38, 143.36, 144.73, 160.05, 160.79, 164.11. ESIMS (m/z) 518 (M+ 2) observed for C29H26ClN3O4. 4.1.6.

(E)-3-(4-((1-(4-bromophenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(7-hydroxy-2,2-

dimethylchroman-6-yl)prop-2-en-1-one

(4b) Yellow solid, yield: 85%, m.p.199-200 °C, IR

(KBr) 3447, 2927, 1636, 1559, 1348, 1247, 1144, 1023, 823, 620 cm-1; 1H-NMR (CDCl3, 300 10

ACCEPTED MANUSCRIPT

MHz) δ (ppm): 1.369 (s, 6H), 1.818-1.85 (t, 2H), 2.7-2.74 (t, 2H), 5.35 (s, 2H), 6.37-6.39 (d, 1H, J = 9.035 Hz), 7.05-7.079 (d, 2H, J = 9.28 Hz), 7.47-7.512 (d, 1H, J = 15.309 Hz), 7.62-7.63 (d, 1H, J = 4.26 Hz), 7.6-7.64 (d, 1H, J = 2.00 Hz), 7.65-7.67 (m, 3H), 7.68-7.70 (d, 2H, J = 9.28 Hz), 7.8-7.86 (d, 1H, J = 15.309 Hz), 8.06 (s, 1H), 13.921 (s, 1H). ESI-MS (m/z) 561 (M+ 1)

4.1.7.

RI PT

observed for C29H26BrN3O4.

(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(2-methyl-3-nitrophenyl)-1H-

1,2,3-triazol-4-yl)methoxy)phenyl)prop-2-en-1-one (4c) Yellow solid, yield: 80%, m.p.174-176 °

C, IR (KBr) 3448, 2923, 2853, 1632, 1368, 1223, 1174, 1039 cm-1; 1H-NMR (CDCl3, 300 MHz)

SC

δ (ppm): 1.36 (s, 6H), 1.83-1.86 (t, 2H), 2.313 (s, 3H), 2.76-2.80 (t, 2H), 5.39 (s, 2H), 6.36 (s, 1H), 7.07-7.09 (d, 2H, J = 9.03 Hz), 7.46-7.50 (d, 1H, J = 15.56 Hz), 7.52-7.56 (t, 1H), 7.6 (s,

M AN U

2H), 7.62-7.66 (d, 2H, J = 9.03 Hz), 7.8-7.86 (d, 1H, J = 15.3 Hz), 7.89 (s, 1H), 8.04-8.07 (d, 1H, J = 9.78 Hz), 13.05 (s, 1H). ESI-MS (m/z) 541 (M+ 1) observed for C30H28N4O6. 4.1.8.(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(4-methoxy-2-nitrophenyl)-1H1,2,3-triazol-4-yl)methoxy)phenyl)prop-2-en-1-one (4d) Yellow solid, yield: 83%, m.p.1971980C, IR (KBr): 3446, 2927, 2866, 1603, 1508, 1385, 1247, 1172, 1021, 811, 653 cm-1; 1HNMR (CDCl3, 300 MHz): δ 1.368 (s, 6H), 1.817-1.851 (t, 2H), 2.708-2.743 (t, 2H), 3.874 (s,

TE D

3H), 5.347 (s, 2H), 6.372-6.395 (d, 1H, J = 9.035 Hz), 7.016 -7.039 (d, 2H, J = 9.285 Hz), 7.063 -7.086 (d, 2H, J = 9.035 Hz), 7.471 -7.510 (d, 1H, J =15.550 Hz),7.617 -7.626 (d, 1H, J = 3.514 Hz), 7.640 -7.648 (d, 2H, J = 3.54 Hz), 7.687 -7.709 (d, 1H, J = 9.035 Hz), 7.829-7.868 (d, 1H, J = 15.560 Hz), 7.988 (s, 1H), 13.925 (s, 1H). ESI-MS (m/z) 557 (M+ 1) observed for

4.1.9.

EP

C30H28N4O7.

(E)-3-(4-((1-(4-acetylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(7-hydroxy-2,2-

dimethylchroman-6-yl)prop-2-en-1-one (4e) Yellow solid, yield: 93%, m.p.179-180 °C, IR

AC C

(KBr) 3426, 2930, 1631, 1512,1364, 1223, 1175, 1045 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.36 (s, 6H), 1.81-1.85 (t, 2H), 2.66 (s, 3H), 2.70-2.74 (t, 2H), 5.36 (s, 2H), 6.34-6.39 (d, 1H, J = 9.03 Hz), 7.06-7.08 (d, 2H, J = 9.28 Hz), 7.47-7.51(d, 1H, J = 15.30 Hz), 7.62-7.64 (d, 2H, J = 9.03 Hz), 7.68-7.70 (d, 1H, J = 9.28 Hz), 7.82-7.86 (d, 1H, J = 15.30 Hz), 7.88-7.90 (d, 1H, J = 9.03 Hz), 8.21 (s, 1H), 8.15-8.16 (d, 2H, J = 5.01 Hz), 13.91 (s, 1H). ESI-MS (m/z) 524 (M+ 1) observed for C31H29N3O5. 4.1.10.

(E)-3-(4-((1-(2-ethylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(7-hydroxy-2,2-

dimethylchroman-6-yl)prop-2-en-1-one (4f) Yellow solid, yield: 70%, m.p.120-122 °C, IR 11

ACCEPTED MANUSCRIPT

(KBr) 3446, 2927, 2866, 1603, 1508, 1385, 1247, 1172, 1021, 811, 653 cm-1. ESI-MS (m/z) 510 (M+ 1) observed for C31H31N3O4. 4.1.11.(E)-3-(4-((1-(3-bromo-4-methylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(7hydroxy-2,2-dimethylchroman-6-yl)prop-2-en-1-one (4g) Yellow solid, yield: 90%, m.p. 151-

RI PT

152 °C, IR (KBr) 3479, 2924, 2854, 1626, 1506, 1365, 1223, 1109, 1042, 871, 796, 694, 600 cm1 1

; H-NMR (CDCl3, 300 MHz) δ (ppm): 1.368 (s, 6H), 1.816-1.850 (t, 2H), 2.467 (s, 3H), 2.707-

2.743 (t, 2H), 5.347 (s, 2H), 6.372-6.394 (d, 1H, J = 9.035 Hz), 7.054-7.077 (d, 2H, J = 9.035 Hz), 7.376-7.399 (d, 1H, J = 8.282 Hz), 7.471-7.510 (d, 1H, J = 15.560 Hz), 7.591-7.597 (d, 1H,

SC

J = 2.259 Hz), 7.612-7.619 (d, 1H, J = 2.761 Hz), 7.641(s, 1H), 7.685-7.707 (d, 1H, J = 9.035 Hz), 7.826-7.865 (d, 1H, J = 15.560 Hz), 7.949-7.955 (d, 1H, J = 2.510 Hz), 8.039(s, 1H),

M AN U

13.952 (s, 1H). ESI-MS (m/z) 575 (M+ 1) observed for C30H28BrN3O4.

4.1.12. (E)-3-(4-((1-(4-chlorophenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(5-hydroxy-2,2dimethylchroman-6-yl)prop-2-en-1-one (4h) Yellow solid, yield: 92%, m.p.196-197 °C, IR (KBr) 3415, 2924, 2850, 1637, 1560, 1349, 1115, 1012 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.381 (s, 6H), 1.821-1.866 (t, 2H), 2.714-2.756 (t, 2H), 5.355 (s, 2H), 6.380- 6.410 (d, 1H, J = 9.042 Hz) ,7.060-7.086 (d, 2H, J = 8.655 Hz), 7.468-7.535 (m, 3H) 7.618-7.647 (d, 2H, J

TE D

= 8.655 Hz), 7.687-7.697 (d, 2H, J = 3.202 Hz) , 7.717 -7.728 (d, 1H, J = 23.202 Hz) , 7.8247.876 (d, 1H, J = 15.447 Hz), 8.068 (s, 1H), 13.920 (s, 1H); 13C-NMR (75 Hz, CDCl3) δ (ppm): 16.36, 26.74, 29.70, 31.86, 62.00, 75.81,109.15, 109.34, 112.84, 115.22, 118.62, 120.91, 121.76, 128.41, 128.46, 130.01, 130.32, 134.82, 135.38, 143.36, 144.73, 160.05, 160.79, 164.11. ESI-

EP

MS (m/z) 517 (M+ 1) observed for C29H26ClN3O4. 4.1.13. (E)-3-(4-((1-(4-bromophenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(5-hydroxy-2,2dimethylchroman-6-yl)prop-2-en-1-one (4i) Yellow solid, yield: 85%, m.p.142-143 °C; IR

AC C

(KBr) 3414, 2925, 1636, 1559, 1350, 1247, 1145, 1023, 825 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.367 (s, 6H), 1.82-1.86 (t, 2H), 2.76-2.79 (t, 2H), 5.35 (s, 2H), 6.36 (s, 1H) ,7.06-7.08 (d, 2H, J = 9.035 Hz), 7.45-7.49 (d, 1H, J = 5.5 Hz), 7.61-7.63 (d, 2H, J = 5.77 Hz), 7.65-7.66 (d, 5H, J = 5.019 Hz), 7.8-7.85 (d, 1H, J = 15.309 Hz), 8.06 (s, 1H), 13.06 (s, 1H). ESI-MS (m/z) 561 (M+ 1) observed for C29H26BrN3O4. 4.1.14.

(E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(2-methyl-3-nitrophenyl)-1H-

1,2,3 triazol-4-yl)methoxy)phenyl)prop-2-en-1-one (4j) Yellow solid, yield: 82%, m.p. 172-173 °

C; IR (KBr) 3414, 2929, 2859, 1637, 1565,1346, 1252, 1149, 1034 cm-1; 1H-NMR (CDCl3, 300 12

ACCEPTED MANUSCRIPT

MHz) δ (ppm): 1.37 (s, 6H), 1.82-1.85 (t, 2H), 2.31 (s, 3H) 2.71-2.75 (t, 2H), 5.38 (s, 2H), 6.376.39 (d, 1H, J = 9.03 Hz), 7.062-7.08 (d, 2H, J = 9.03 Hz),7.48-7.52 (d, 1H, J = 15.81 Hz), 7.547.56 (d, 1H, J = 8.53 Hz), 7.63 (s, 1H), 7.62-7.63 (d, 1H, J = 3.51 Hz), 7.69-7.71 (d, 2H, J = 9.03 Hz), 7.82-7.9 (m, 2H), 8.05-8.07 (d, 1H, J = 7.78 Hz), 13.91 (s, 1H). ESI-MS (m/z) 541

RI PT

(M+ 1) observed for C30H28N4O6.

4.1.15.(E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(4-methoxy-2-nitrophenyl)-1H1,2,3-triazol-4-yl)methoxy)phenyl)prop-2-en-1-one (4k) Yellow solid, yield: 80%, m.p. 192193 °C, IR (KBr) 3416, 2932, 2860, 1637, 1541, 1509,1359, 1239, 1150, 1039 cm-1; 1H-NMR

SC

(CDCl3, 300 MHz) δ (ppm): 1.368 (s, 6H), 1.817-1.851 (t, 2H), 2.708-2.743 (t, 2H), 3.874 (s, 3H), 5.347 (s, 2H), 6.372-6.395 ( d, 1H, J = 9.035 Hz), 7.016 -7.039 (d, 2H, J = 9.285 Hz), 7.063

M AN U

-7.086 (d, 2H, J = 9.035 Hz), 7.471 -7.510 (d, 1H, J = 15.550 Hz), 7.617 -7.626 (d, 1H, J = 3.514 Hz), 7.640 -7.648 (d, 2H, J = 3.54 Hz), 7.687 -7.709 (d, 1H, J = 9.035 Hz), 7.829-7.868 (d, 1H, J = 15.560 Hz), 7.988 (s, 1H), 13.925 (s, 1H). ESI-MS (m/z) 557 (M+ 1) observed for C30H28N4O7.

4.1.16. (E)-3-(4-((1-(4-acetylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(5-hydroxy-2,2dimethylchroman-6-yl)prop-2-en-1-one (4l) Yellow solid, yield: 85%, m.p. 189-190 °C; IR

TE D

(KBr) 3415, 2927, 2856, 1679, 1636, 1509, 1361, 1246, 1150, 1015 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.36 (s, 6H), 1.81-1.85 (t, 2H), 2.66 (s, 3H), 2.70-2.74 (t, 2H), 5.36 (s, 2H), 6.346.39 (d, 1H, J = 9.03 Hz), 7.06-7.08 (d, 2H, J = 9.28 Hz), 7.47-7.51 (d, 1H, J = 15.30 Hz), 7.627.64 (d, 2H, J = 9.03 Hz), 7.68-7.70 (d, 1H, J = 9.28 Hz), 7.82-7.86 (d, 1H, J = 15.30 Hz), 7.88-

EP

7.90 (d, 1H, J = 9.03 Hz), 8.21 (s, 1H), 8.15-8.16 (d, 2H, J = 5.01 Hz), 13.91 (s, 1H). ESI-MS (m/z) 524 (M+ 1) observed for C31H29N3O5. 4.1.17.

(E)-3-(4-((1-(2-ethylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(5-hydroxy-2,2-

AC C

dimethylchroman-6-yl)prop-2-en-1-one (4m) Yellow solid, yield: 70%, m.p. 99-100 °C; 1HNMR (CDCl3, 300 MHz) δ (ppm): 1.09-1.13 (t, 3H), 1.36 (s, 6H), 1.83-1.86 (t, 2H), 2.47-2.53 (q, 2H), 2.76-2.80 (t, 2H), 5.37 (s, 2H), 6.37 (s, 1H), 7.08-7.10 (d, 2H, J = 9.035 Hz), 7.29-7.37 (m, 2H), 7.41-7.43 (d, 1H, J = 7.278 Hz), 7.46-7.50 (m, 2H), 7.62-7.63 (d, 2H, J = 4.51Hz), 7.66 (s, 1H), 7.82-7.86 (d, 2H, J = 16.313 Hz), 13.07 (s, 1H). ESI-MS (m/z) 510 (M+ 1) observed for C31H31N3O4. 4.1.18.(E)-3-(4-((1-(3-bromo-4-methylphenyl)-1H-1,2,3-triazol-4-yl)methoxy)phenyl)-1-(5hydroxy-2,2-dimethylchroman-6-yl)prop-2-en-1-one (4n) Yellow solid, yield: 90%, m.p. 20013

ACCEPTED MANUSCRIPT

202 °C; IR (KBr) 3415, 2933, 1636, 1554, 1358, 1297, 1149, 1010 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.368 (s, 6H), 1.816-1.850 (t, 2H), 2.467 (s, 3H), 2.707-2.743 (t, 2H), 5.347 (s, 2H), 6.372-6.394 (d, 1H, J = 9.035 Hz), 7.054-7.077 (d, 2H, J = 9.035 Hz), 7.376-7.399 (d, 1H, J = 8.282 Hz), 7.471-7.510 (d, 1H, J = 15.560 Hz), 7.591-7.597 (d, 1H, J = 2.259 Hz), 7.612-7.619

RI PT

(d, 1H, J = 2.761 Hz), 7.641 (s, 1H), 7.685-7.707 (d, 1H, J = 9.035 Hz), 7.826-7.865 (d, 1H, J = 15.560 Hz), 7.949-7.955 (d, 1H, J = 2.510 Hz), 8.039 (s, 1H), 13.952 (s, 1H). ESI-MS (m/z) 575 (M+ 1) observed for C30H28BrN3O4.

4.1.19. (E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(4-nitrobenzyl)-1H-1,2,3-triazol-

SC

4-yl)methoxy)phenyl)prop-2-en-1-one (4o) Yellow solid, yield: 75%, m.p. 156-157 °C; IR (KBr) 3450, 2930, 1630, 1518, 1350, 1221, 1111, 1048, 833 cm-1; 1H-NMR (CDCl3, 300 MHz) δ

M AN U

(ppm): 1.368 (s, 6H), 1.817-1.852 (t, 2H), 2.707-2.743 (t, 2H), 5.27 (s, 2H), 5.67 (s, 2H), 6.3726.394 (d, 1H, J = 9.035 Hz), 7.054-7.077 (d, 2H, J = 9.035 Hz), 7.41-7.43 (d, 1H, J = 9.035 Hz), 7.46-7.510 (d, 1H, J = 15.30 Hz), 7.591-7.597 (d, 1H, J = 9.035 Hz), 7.63 (s, 1H), 7.685-7.707 (d, 2H, J = 9.28 Hz), 7.826-7.865 (d, 2H, J = 15.81 Hz), 8.23-8.26 (d, 2H, J = 9.035 Hz), 13.952 (s, 1H). ESI-MS (m/z) 541 (M+ 1) observed for C30H28N4O6. 4.1.20.

(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-pentyl-1H-1,2,3-triazol-4-

TE D

yl)methoxy)phenyl)prop-2-en-1-one (4p) Yellow solid, yield: 85%, m.p. 104-105 °C; IR (KBr) 3449, 2932, 1622, 1554, 1365, 1224, 1108 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 0.8830.919 (t, 3H), 1.36 (s, 4H), 1.57 (s, 6H), 1.81-1.85 (t, 2H), 1.88-1.963 (m, 2H), 2.70-2.74 (t, 2H), 4.34-4.38 (t, 2H), 5.27 (s, 2H), 6.37-6.39 (d, 1H, J = 9.03 Hz), 7.02-7.05 (d, 2H, J = 9.03 Hz),

EP

7.46-7.50 (d, 1H, J = 15.56 Hz), 7.60-7.61 (d, 2H, J = 4.26 Hz), 7.62 (s, 1H) 7.68- 7.70 (d, 1H, J = 9.28 Hz), 7.82-7.86 (d, 1H, J = 15.30 Hz ), 13.93 (s, 1H). ESI-MS (m/z) 476 (M+ 1)

AC C

observed for C28H33N3O4. 4.1.21.

(E)-1-(7-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-octyl-1H-1,2,3-triazol-4-

yl)methoxy)phenyl)prop-2-en-1-one (4q) Yellow solid, yield: 75%, m.p. 101-102 °C; IR (KBr): 3417, 2928, 2856, 1630, 1367, 1221, 1047 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 0.873 (s, 3H), 1.251-1.322 (m, 10H), 1.367 (s, 6H), 1.59 (s, 2H), 1.817-1.85 (t, 2H), 2.70-2.74 (t, 2H), 4.34-4.36 (t, 2H), 5.27 (s, 2H), 6.37-6.39 (d, 1H, J = 9.03 Hz), 7.02-7.04 (d, 2H, J = 9.03 Hz), 7.46-7.50 (d, 1H, J = 15.56 Hz), 7.60-7.62 (m, 3H, J = 9.53 Hz), 7.68-7.70 (d, 1H, J = 9.28 Hz), 7.82-7.86 (d, 1H, J = 15.56 Hz), 13.93 (s, 1H). ESI-MS (m/z) 518 (M+ 1) observed for C31H39N3O4. 14

ACCEPTED MANUSCRIPT

4.1.22. (E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-(4-nitrobenzyl)-1H-1,2,3-triazol4-yl)methoxy)phenyl)prop-2-en-1-one (4r) Yellow solid, yield: 75%, m.p. 148-150 °C; IR (KBr) 3415, 2930, 2852, 1636, 1512, 1352, 1299, 1150, 836, 796, 678 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 1.368 (s, 6H), 1.817-1.852 (t, 2H), 2.707-2.743 (t, 2H), 5.27 (s, 2H), 5.67 (s, 2H),

RI PT

6.37 (s, 1H), 7.054-7.077 (d, 2H, J = 9.28 Hz), 7.41-7.43 (m, 2H, J = 13.80 Hz), 7.46-7.510 (d, 2H, J = 13.30 Hz), 7.61-7.64 (d, 2H, J = 10.54 Hz), 7.826-7.865 (d, 2H, J = 15.51 Hz), 8.23-8.26 (d, 2H, J = 9.285 Hz), 13.952 (s, 1H). ESI-MS (m/z) 541 (M+ 1) observed for C30H28N4O6. 4.1.23.

(E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-pentyl-1H-1,2,3-triazol-4-

SC

yl)methoxy)phenyl)prop-2-en-1-one (4s) Yellow solid, yield: 85%, m.p. 110-111 °C; IR (KBr) 3415, 2927, 2859, 1638, 1555, 1360, 1247, 1149, 1050 cm-1; 1H-NMR (CDCl3, 300 MHz) δ

M AN U

(ppm): 0.883-0.919 (t, 3H), 1.25-1.30 (m, 4H), 1.36 (s, 6H), 1.82-1.86 (t, 2H), 1.90-1.96 (m, 2H), 2.76-2.80 (t, 2H), 4.34-4.38 (t, 2H), 5.27 (s, 2H), 6.37 (s, 1H), 7.054-7.077 (d, 2H, J = 9.035 Hz), 7.26 (s, 1H), 7.41-7.43 (d, 1H, J = 15.56 Hz), 7.62-7.63 (d, 2H, J = 7.027 Hz), 7.826-7.865 (d, 1H, J = 15.56 Hz), 13.07 (s, 1H). ESI-MS (m/z) 476 (M+ 1) observed for C28H33N3O4. 4.1.24.

(E)-1-(5-hydroxy-2,2-dimethylchroman-6-yl)-3-(4-((1-octyl-1H-1,2,3-triazol-4-

yl)methoxy)phenyl)prop-2-en-1-one (4t) Yellow solid, yield: 86%, m.p. 88-89 °C; IR (KBr)

TE D

3415, 2924, 2855, 1634, 1554, 1362, 1245, 1150 cm-1; 1H-NMR (CDCl3, 300 MHz) δ (ppm): 0.874 (s, 3H), 1.25-1.32 (m, 10H, J = 7.278 Hz), 1.368 (s, 6H), 1.59 (s, 2H), 1.816-1.850 (t, 2H), 2.467 (s, 3H), 2.707-2.743 (t, 2H), 5.347 (s, 2H), 6.372-6.394 (d, 1H, J = 9.035 Hz), 7.0547.077 (d, 2H, J = 9.035 Hz), 7.376-7.399 (d, 1H, J = 8.28 Hz), 7.471-7.510 (d, 1H, J = 15.56

EP

Hz), 7.591-7.597 (d, 1H, J = 2.25 Hz), 7.612-7.619 (d, 1H, J = 2.761 Hz), 7.641 (s, 1H), 7.617.62 (d, 2H, J = 2.8 Hz), 7.63-7.64 (d, 2H, J = 2.259 Hz), 7.81-7.85 (d, 1H, J = 15.56 Hz),

AC C

13.072 (s, 1H). ESI-MS (m/z) 518 (M+ 1) observed for C31H39N3O4.

Supplementary

Tables S1 and S2 consisting of in silico parameters of oral bioavailability, drug likeness properties, toxicity risk assessment and various NMR and mass spectra of all the synthesized derivatives were provided in supplementary.

15

ACCEPTED MANUSCRIPT

Acknowledgments Authors thank Director, CSIR-CIMAP, Lucknow and Scientist-In-Charge, CSIRCIMAP-Research Centre, Hyderabad, India for their constant encouragement and support.

RI PT

Author Yakaiah thanks CSIR-New Delhi for granting Senior Research Fellowship.

References

[1] J.B. Gibbs, Mechanism-Based Target Identification and Drug Discovery in Cancer Research, Science. 287 (2000) 1969-1973.

SC

[2] H. Varmus, The New Era in Cancer Research, Science. 312 (2006) 1162.

[3] C.T. Hsieh, T.J. Hsieh, M. El-Shazly, D.W. Chuang, Y.H. Tsai, C.T. Yen, S.F. Wu, Y.C. Wu,

Chem. Let. 22 (2012), 3912-5.

M AN U

F.R. Chang, Synthesis of chalcone derivatives as potential anti-diabetic agents. Bioorg. Med.

[4] Z.H. Israili, Advances in the treatment of type 2 Diabetes Mellitus. Am. J. Therap. 18 (2011) 117-52.

[5] J. H. Cheng, C. F. Hung, S. C.Yang, J. P. Wang, S. J. Won, C. N. Lin, Synthesis and cytotoxic, anti-inflammatory, and anti-oxidant activities of 2′,5′-dialkoxylchalcones as cancer

TE D

chemopreventive agents, Bioorg. Med. Chem. 16 (2008), 7270- 7276. [6] S. F. Nielsen, T. Boesen, M. Larsen, K. Schonning, H. Kromann, Antibacterial chalcones–– bioisosteric replacement of the 4′-hydroxy group, Bioorg. Med. Chem. 12 (2004) 30473054.

EP

[7] C. Zhan, J. Yang, Protective effects of isoliquiritigenin in transient middle cerebral artery occlusion-induced focal cerebral ischemia in rats, Pharmacol. Res. 53 (2006) 303. [8] M. Liu, P. Wilairat, M. L. Go, Antimalarial alkoxylated and hydroxylated chalcones

AC C

[corrected]: structure-activity relationship analysis. J. Med. Chem. 44 (2001) 4443-52.

[9] M.L. Go, X. Wu, X. Liu, Chalcones: an update on cytotoxic and chemoprotective properties L. Curr. Med. Chem.12 (2005) 481-99.

[10] (a) O. Sabzevari, G. Galati, M.Y. Moridani, A. Siraki, P. O’Brien, J. Chem. Biol. Interact. 148 (2004) 57. (b) Y. S. Lee, S. S. Lim, K. H. Shin, Y. S. Kim, K. Ohuchi, S. H. Jung, Biol. Pharm. Bull. 29 (2006) 1028. (c) Z. Rozmer, T. Berki, P. Perjési, Toxicol. In Vitro 20 (2006) 1354.

16

ACCEPTED MANUSCRIPT

[11] C.T. Hsieh, T.J. Hsieh, M. El Shazly, D.W. Chuanq, Y.H. Tsai, C.T. Yen, S.F. Wu, Y.C. Wu, F.R. Chang, Synthesis of chalcone derivatives as potential anti-diabetic agents. Bioorg. Med. Chem. Lett. 22 (2012) 3912-5. [12] H.C. Kolb, K.B. Sharpless, The growing impact of click chemistry on drug discovery, Drug

RI PT

Discov. Today 8 (2009) 1128-1137.

[13] S.G. Agalave, S.R. Maujan, V.S. Pore, Click chemistry: 1,2,3-triazoles as pharmacophores, Chem. Asian J. 6 (2011) 2696-2718.

[14] V.V. Rostovtsev, K.B. Sharpless, A stepwise Huisgen cycloaddition process: copper (I)-

SC

catalyzed regioselective “Ligation” of azides and terminal alkynes, Angew. Chem. Int. Ed. 41 (2002) 2596-2599.

M AN U

[15] N.S. Vatmurge, B.G. Hazra, V.S. Pore, F. Shirazi, P.S. Chavan, M.V. Deshpande, Synthesis and antimicrobial activity of b-lactam-bile acid conjugates linked via triazole, Bioorg. Med. Chem. Lett. 18 (2008) 2043-2047.

[16] M. Whiting, J.Muldoon, Y.C. Lin, S.M. Silverman,W. Lindstron, A.J. Olson, H.C. Kolb, M.G. Finn, K.B. Sharpless, J.H. Elder, V.V. Fokin, Inhibitors of HIV-1 protease by using in situ click chemistry, Angew. Chem. Int. Ed. 45 (2006) 1435-1439.

TE D

[17] V.D. Bock, D. Speijer, H. Hiemstra, J.H.V. Maarseveen, 1,2,3-Triazoles as peptide bond isosteres: synthesis and biological evaluation of cyclotetrapeptide mimics, Org. Biomol. Chem. 5 (2007) 971-976.

[18] N.G. Aher, V.S. Pore, N.N. Mishra, A. Kumar, P.K. Shukla, A. Sharma, M.K. Bhat,

EP

Synthesis and antifungal activity of 1,2,3-triazole containing fluconazole analogues, Bioorg. Med. Chem. Lett. 19 (2009) 759-763. J.A.

Demaray,

J.E.

Thuener,

M.N.

Dawson,

S.J.

Sucheck,

Synthesis

of

AC C

[19]

triazoleoxazolidinones via a one-pot reaction and evaluation of their antimicrobial activity, Bioorg. Med. Chem. Lett. 18 (2008) 4868-4871.

[20] X.L. Wang, K. Wan, C.H. Zhou, Synthesis of novel sulfanilamide-derived 1,2,3-triazoles and their evaluation for antibacterial and antifungal activities, Eur. J. Med. Chem. 45 (2010) 4631-4639. [21] D.R. Buckle, D.J. Outred, C.J.M. Rockell, H. Smith, B.A. Spicer, Studies on v-triazoles. 7. Antiallergic 9-oxo- 1H,9H-benzopyrano[2,3-d]-v-triazoles, J. Med. Chem. 26 (1983) 251254. 17

ACCEPTED MANUSCRIPT

[22] M. Whiting, J.C. Tripp, Y.C. Lin, W. Lindstrom, A.J. Olson, J.H. Elder, K.B. Sharpless, V.V. Fokin, Rapid discovery and structure-activity profiling of novel inhibitors of human immunodeficiency virus type 1 protease enabled by the copper(I)-catalyzed synthesis of 1,2,3-triazoles and their further functionalization, J. Med. Chem. 4 (2006) 7697-7710.

RI PT

[23] M.J. Giffin, H. Heaslet, A. Brik, Y.C. Lin, G. Cauvi, C.H. Wong, D.E. McRee, J.H. Elder, C.D. Stout, B.E. Torbett, A copper(I)-catalyzed 1,2,3-triazole azidealkyne click compound is a potent inhibitor of a multidrug-resistant HIV-1 protease variant, J. Med. Chem. 51 (2008) 6263-6270.

SC

[24] M.S. Costa, N. Boechat, E.A. Rangel, F.D.C.D. Silva, A.M.T.D. Souza, C.R. Rodrigues, H.C. Castro, I.N. Junior, M.C.S. Lourenc, S.M.S.V. Wardell, V.F. Ferreirab, Synthesis,

M AN U

tuberculosis inhibitory activity, and SAR study of N-substituted-phenyl- 1,2,3-triazole derivatives, Bioorg. Med. Chem. 14 (2006) 8644-8653.

[25] S.R. Patpi, L. Pulipati, P. Yogeeswari, D. Sriram, N. Jain, B. Sridhar, R. Murthy, D.T. Anjana, S.V. Kalivendi, S. Kantevar, Design, Synthesis, and structureactivity correlations of novel dibenzo [b, d] furan, dibenzo [b, d] thiophene, and N-methylcarbazole clubbed 1,2,3-triazoles as potent inhibitors of mycobacterium tuberculosis, J. Med. Chem. 55

TE D

(2012) 3911-3922.

[26] R.D. Simone, M.G. Chini, I. Bruno, R. Riccio, D. Mueller, O. Werz, G. Bifulco, Structurebased discovery of inhibitors of microsomal prostaglandin E2 synthase-1,5-lipoxygenase and 5-lipoxygenase-activating protein: promising hits for the development of new anti-

EP

inflammatory agents, J. Med. Chem. 54 (2011) 1565-1575. [27] I.S. Bennet, G. Brooks, N.J.P. Broom, S.H. Calvert, K. Coleman, I. Francois, 6-(substituted

AC C

methylene) penems, potent broad spectrum inhibitors of bacterial b-Lactamse. V. chiral 1,2,3-triazolyl derivatives, J. Antibiot. 44 (1991) 969-978.

[28] M.J. Soltis, H.J. Yeh, K.A. Cole, N. Whittaker, R.P. Wersto, E.C. Kohn, Identification and characterization of human metabolites of CAI [5-amino-1- 1(4’-chlorobenzoyl-3,5dichlorobenzyl)-1,2,3-triazole-4-carboxamide), Drug Metab. Dispos 24 (1996) 799-806.

[29] M.J. Fray, D.J. Bull, C.L. Carr, E.C.L. Gautier, C.E. Mowbray, A. Stobie, Structureactivity Relationships of 1,4-dihydro-(1H,4H)-quinoxaline-2,3-dionesas N-methyl-D-aspartate (glycine site) receptor antagonists. 1. Heterocyclic substituted 5-alkyl derivatives, J. Med. Chem. 24 (2001) 1951-1962. 18

ACCEPTED MANUSCRIPT

[30] D. Imperio, T. Pirali, U. Galli, F. Pagliai, L. Cafici, P.L. Canonico, G. Sorba, A.A. Genazzani, G.C. Tron, Replacement of the lactone moiety on podophyllotoxins and steganacin analogues with a 1,5-disubstituted 1,2,3-triazole via ruthenium-catalyzed click chemistry, Bioorg. Med. Chem. 15 (2007) 6748-6757.

RI PT

[31] L.S. Kallander, Q. Lu, W. Chen, T. Tomaszek, G. Yang, D. Tew, T.D. Meek, G.A. Hofmann, C.K.S. Pritchard, W.W. Smith, T.F. Ho, P.W. Fisher, M.R. Mattern, R.K. Johnson, M.J. Hansbury, J.D. Winkler, K.W. Ward, D.F. Veber, S.K. Thompson, 4-Aryl1,2,3-triazole: a novel template for a reversible methionine aminopeptidase 2 inhibitor,

SC

optimized to inhibit angiogenesis in vivo, J. Med. Chem. 48 (2005) 5644-5647.

[32] F. Pagliai, T. Pirali, E.D. Grosso, R.D. Brisco, G.C. Tron, G. Sorba, A.A. Genazzani, Rapid

M AN U

synthesis of triazole-modified resveratrol analogues via click chemistry, J. Med. Chem. 49 (2006) 467-470.

[33] L. Cafici, T. Pirali, F. Condorelli, E.D. Grosso, A. Massarotti, G. Sorba, P.L. Canonico, G.C. Tron, A.A. Genazzani, Solution-phase parallel synthesis and biological evaluation of combretatriazoles, J. Comb. Chem. 10 (2008) 732-740.

[34] C.B. Yim, I. Dijkgraaf, R. Merkx, C. Versluis, A. Eek, G.E. Mulder, D.T.S. Rijkers, O.C.

TE D

Boerman, R.M.J. Liskamp, Synthesis of DOTA-conjugated multimeric [Tyr3] Octreotide peptides via a combination of Cu(I)- catalyzed "Click" cycloaddition and thio acid/sulfonyl azide "sulfo-click" amidation and their in vivo evaluation, J. Med.Chem. 53 (2010) 39443953.

EP

[35] J. Yoon, J.S. Ryu, A rapid synthesis of lavendustin-mimetic small molecules by click fragment assembly, Bioorg. Med. Chem. Lett. 20 (2010) 3930-3935.

AC C

[36] J. Vantikommu, S. Palle, P.S. Reddy, V. Ramanatham, M. Khagga, V.R. Pallapothula, Synthesis and cytotoxicity evaluation of novel 1,4-disubstituted 1,2,3-triazoles via CuI catalysed 1,3-dipolar cycloaddition, Eur. J. Med. Chem. 45 (2010) 5044-5050.

[37] X.L. Li, Y.J. Lin, Q.Q. Wang, Y.K. Yuan, H. Zhang, X.H. Qian, The novel anti-tumor agents of 4-triazol-1,8- naphthalimides: synthesis, cytotoxicity, DNA intercalation and photocleavage, Eur. J. Med. Chem. 45 (2011) 1274-1279. [38] A. Kamal, S. Prabhakar, M.J. Ramaiah, P.V. Reddy, C.R. Reddy, A. Mallareddy, N. Shankaraiah, T.L.N. Reddy, S.N.C.V.L. Pushpavalli, M.P. Bhadra,

19

Synthesis and

ACCEPTED MANUSCRIPT

anticancer activity of chalcone-pyrrolobenzodiazepine conjugates linked via 1,2,3-triazole ring side-armed with alkane spacers, Eur J Med Chem 46 (2011) 3820-3831. [39] G. Wang, C. Li, L. He, K. Lei, F. Wang, Y. Pu, Z. Yang, D. Cao, L. Ma, J. Chen, Y. Sang, X. Liang, M. Xiang, A. Peng, Y. Wei, L. Chen, Design, synthesis and biological evaluation

agents, Bioorg. Med. Chem. 22 (2014) 2060–2079.

RI PT

of a series of pyranochalcone derivatives containing indole moiety as novel anti-tubulin

[40] Ch. Yakaiah, D. Anand Kumar, Alam Sarfaraz, S.P. Shailendra, A. Niranjan Kumar, G. Namita, K.V.N.S. Srinivas, J. Kotesh Kumar, Feroz Khan, K. Ashok.Tiwari, Grover

SC

Paramjit, Synthesis, biological evaluation and molecular modeling studies of some novel thiazolidinediones with triazole ring, Eur. J. Med. Chem. 70 (2013) 308-314

M AN U

[41] T. Mossman, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays, J. Immunol. Methods 65 (1983) 55-63. [42] K.S. Babu, A.K. Tiwari, P.V. Srinivas, A.Z. Ali, B.C. Raju, J.M. Rao, Yeast and mammalian α -glucosidase inhibitory constituents from Himalayan rhubarb Rheum emodi Wall. Ex Meisson, Bioorg. Med. Chem. Lett. 14 (2004) 3841- 3845. [43] G.J. McDougall, F. Shpiro, P. Dobson, P. Smith, A. Blake, D. Stewart, Different

TE D

polyphenolic components of soft fruits inhibit a-amylase and α-glucosidase, J. Agric. Food Chem. 53 (2005) 2760-2766.

[44] K. Kalani, J. Agarwal, S. Alam, F. Khan, A. Pal, S.K. Srivastava, In Silico and In Vivo Anti-Malarial Studies of 18β Glycyrrhetinic Acid from Glycyrrhiza glabra. PLoS ONE

EP

(2013) 8 e74761.

[45] S. Alam, F. Khan, QSAR and docking studies on xanthone derivatives for anticancer

AC C

activity targeting DNA topoisomerase IIα, Drug Design, Development and Therapy 8 (2014) 183-195.

Figures & Legends:

Fig. 1 Structural model of topoisomerase II α (PDB: 1ZXM, chain A and B) showing binding pattern of candidate compound along with control (yellow colour).

20

ACCEPTED MANUSCRIPT

Fig. 2 Structural model of α-glucosidase (PDB: 2QMJ) showing binding pattern of candidate compound along with control (red colour).

RI PT

Scheme 1: Synthesis of Chalcone 1,2,3 tiazole derivatives

Tables:

Table 1: Anticancer and α-glucosidase inhibitory activity of compound 4a-4t.

SC

Table 2: Details of Docking energy, active site pocket residues and hydrogen bonds revealed through molecular docking of Chalcone derivatives and Doxorubicin on Topoisomerase II (PDB: 1ZXM)

AC C

EP

TE D

M AN U

Table 3: Details of binding affinity, active site pocket residues and hydrogen bonds revealed through molecular docking of Chalcone derivatives and Acarbose on α-Glucosidase (PDB: 2QMJ)

21

ACCEPTED MANUSCRIPT

Table 1: Anticancer and α-glucosidase inhibitory activity of compound 4a-4t.

NA-Not Active

A-549 IC50 in µM 75.46 79.99 65.86 99.48 66.28 137.35 125.37 NA 128.59 NA NA 149.27 111.60 NA 35.81 91.29 50.82 69.88 48.63 NA NA 69.33

Hep G2 IC50 in µM NA NA 108.89 NA NA 114.73 116.79 102.16 135.86 115.46 119.96 91.39 67.77 110.41 108.89 95.81 127.46 69.90 9.82 74.11 NA 1.05

RI PT

1MR-32 IC50in µM 92.13 69.42 NA NA NA NA NA NA 117.19 73.53 132.08 89.55 NA NA 107.82 NA 7.09 17.34 NA NA NA 44.40

SC

4a 4b 4c 4d 4e 4f 4g 4h 4i 4j 4k 4l 4m 4n 4o 4p 4q 4r 4s 4t Acarbose Doxorubicin

α-glucosidase Anticancer activity inhibition MCF-7 DU-145 IC50 in µM IC50in µM IC50 in µM NA NA NA NA NA NA NA 176.03 NA NA NA NA NA 181.54 NA NA NA NA NA NA NA 127.90 NA NA NA NA NA NA 145.09 NA NA 73.08 NA NA 171.39 NA 67.77 NA NA NA NA NA NA NA NA NA 74.94 NA NA NA NA NA 17.11 29.88 102.10 190.61 NA NA NA NA 23.87 NA NA NA 2.63 17.67

M AN U

Compound

Com poun d 4a

LibDo ck sco re 114.67 8

-H bond

4b

112.13 5

TYR 186

ILE125

71.262 3

LYS 157 (2)

NO

76.324 2

NO

NO

4e

101.44 5

NO

NO

4f

112.69 3

NO

NO

4d

Active site pocket residues

ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, ASN120, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, SER148, SER149, THR215, ILE217 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, ASN120, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, SER149,ALA167, THR215, ILE217 ASN91, ASP94, ASN95, ARG98, ILE125, PRO126, VAL137, ILE141, PHE142, THR147, SER148, SER149, ASN150, GLU155, LYS157, THR159, GLY161, GLY164, ALA167, LYS168 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, TRP119, ASN120, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, SER148, SER149, ALA167, LYS168, THR215, ILE217 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, ASN120, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, SER149. THR215, ILE217 ASN91, ALA92, ASP94, ASN95, ARG98, ASN120, LYS123, GLY124, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, THR147, SER148, SER149, GLY161, GLY164, LYS168, THR215, ILE217

EP

Pi interact ion NO

AC C

4c

NO

TE D

Table 2: Details of LibDock score, hydrogen bonds, Pi interactions and active site pocket residues revealed through molecular docking of Chalcone derivatives and Doxorubicin on DNA Topoisomerase IIα (PDB: 1ZXM).

ACCEPTED MANUSCRIPT

4h

99.424 6

NO

ARG98 (2)

4i

103.28 1

NO

4j

75.655 9

ARG 98

ARG98 (2) LYS123 LYS157 ARG98

4k

73.454

NO

NO

4l

104.48

HIS 130

ILE125

4m

79.411 7

NO

LYS157

4n

111.32 3

SER 183

4o

91.525 2

4P

100.69

ARG 98 (3) TYR 165 LYS 168 NO

ARG98 (3) ILE141 NO

NO

4q

118.29

NO

NO

4r

95.601 8

ASN 120

PHE142

4s

99.869 9

TYR 186

ILE125

109.87

NO

ILE141

125.85 7

ASN 91 LYS 168 (2)

NO

Doxo rubici n

ILE88, ASN91, ALA92, ASN95, GLN97, ARG98, ASN120, LYS123, ILE125, PRO126, VAL137, ILE141, PHE142, THR147, SER148, SER149, GLU155, LYS157, ALA167, THR215, ILE217 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, ASN120, ILE125, PRO126, VAL137, ILE141, PHE142, SER149, THR215, ILE217 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, TRP119, ASN120, ILE125, PRO126, HIS130, VAL137, LEU140, ILE141, PHE142, SER149, THR215, ILE217 ASN91, ALA92, ASN95, ARG98, ASN120, ILE125, VAL137, ILE141, PHE142, THR147, SER148, SER149, GLU155, LYS157, THR215 ASN91, ARG98, LYS123, GLY124, ILE125, PRO126, ILE141, PHE142, THR147, SER148, SER149, GLY164, TYR165, GLY166, ALA167, LYS168 ASN91, ARG98, LYS123, ILE125, PRO126, VAL137, ILE141, SER148, SER149 , ASN150, LYS157, GLY161, GLY164, TYR165, GLY166, ALA167, LYS168

ILE88, ASN91, ALA92, ASP94, ASN95, ARG98, ILE118, TRP119, ASN120, ILE125, PRO126, VAL137, ILE141, PHE142, SER148, SER149, THR215, ILE217 ASN91, ASP94, ARG98, LYS123, GLY124, ILE125, PRO126, VAL137, ILE141, PHE142, THR147, SER148, SER149, GLY161, ARG162, ASN163, GLY164, TYR165, GLY166, ALA167, LYS168 ASN91, ALA92, ASN95, ARG98, ASN120, LYS123, GLY124, ILE125, PRO126, VAL137, ILE141, PHE142, SER148, SER149, GLU155, LYS157, LYS168, THR215 ILE88, ASN91, ALA92, ASN95, ARG98, ILE118, TRP119, ASN120, ILE125, PRO126, HIS130, VAL137, ILE141, PHE142, SER149, THR215, CYS216, ILE217 ASN91, GLN97, ARG98, LYS123, ILE125, PRO126, VAL137, ILE141, PHE142, THR147, SER148, SER149, GLU155, LYS157, THR159, GLY164, TYR165, GLY166, ALA167, LYS168 ASN91, ALA92, ASP94, ASN95, ARG98, ASN120, ILE125, PRO126, ILE141, PHE142, THR147, SER148, SER149, GLY164, ALA167, LYS168, THR215

EP

AC C

4t

ILE88, ASN91, ALA92, ASN95, ARG98, ASN120, LYS123, ILE125, PRO126, VAL137, ILE141, PHE142, SER148, SER149, GLU155, LYS157, ALA167, THR215, ILE217 ASN91, ASP94, ASN95, ARG98, LYS123, ILE125, PRO126, ILE141, THR147, SER148, SER149, ASN150, LYS157, GLY161, ARG162, GLY164, ALA167, LYS168 ILE88, ASN91, ALA92, ASP94, ASN95, GLN97, ARG98, ASN120, ILE125, PRO126, ILE141, PHE142, THR147, SER148, SER149, LYS157, THR159, ALA167, LYS168, THR215, ILE217

RI PT

LYS157

SC

ARG 98

M AN U

95.239 6

TE D

4g

ACCEPTED MANUSCRIPT

Table 3: Details of LibDock score, hydrogen bonds, Pi interactions and active site pocket residues revealed through molecular docking of Chalcone derivatives and Acarbose on αglucosidase (PDB: 2QMJ)

100.372

4m

104.681

ARG 526 ASP542 ARG 526 (2)

4p

106.353

4s

107.784

Acar bose

123.536

Pi interact ion PHE450

TRP406 HIS600

ASN 207 ARG 526 ARG 526

TRP406

THR 205 ASN 207 ASP 327 (2) ARG 526 ASP 542 (2)

NO

ASP203, TYR299, ASP327, ILE328, ARG334, ILE364, GLU404, TRP406, TRP441, ASP443, MET444, PHE450, LYS480, ARG526, ASP542, PHE575, HIS600 ASP203, THR204, THR205, PRO206, ASP327, ILE328, ILE364, TRP406, TRP441, ASP443, MET444, PHE450, ARG526, TRP539, ASP54, THR544, PHE575, ALA576, LEU577, HIS600, TYR605 THR205, ASN207, ASN209, TYR299, ASP327, ILE328, ILE364, TRP406, TRP441, ASP443, MET444, ARG526, TRP539, ASP542, THR544, PHE575, ALA576, LEU577, ARG598, HIS600, TYR605 THR205, TYR299, ASP327, ILE364, TRP406, TRP441, ASP443, ARG526, TRP539, ASP542, THR544, ASP571, PHE575, ALA576, LEU577, ARG598, HIS600, TYR605 ASP203, THR205, ASN207, TYR299, ASP327, ILE328, ILE364, TRP406, TRP441, ASP443, MET444, ARG526, TRP539, ASP542, THR544, PHE575, ALA576, HIS600

AC C

EP

TE D

TRP406

Active site pocket residues

RI PT

-H bond

SC

LibDoc k score

M AN U

Com poun d 4h

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

Fig. 1 Structural model of DNA topoisomerase II α (PDB: 1ZXM) with doxorubicin binding site (sphere); (B) Binding site and binding pattern of candidate compound along with control.

Fig. 2 Structural model of α-glucosidase (PDB: 2QMJ) with Acarbose binding site (sphere). (B) Binding site and binding pattern of candidate compound along with control.

1

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

Scheme 1: Synthesis of Chalcone 1,2,3 triazole derivatives

2

ACCEPTED MANUSCRIPT

 Dimethyl chromano chalcones were tagged with substituted 1,2,3-triazoles.  Chalcone-triazoles were synthesized and screened for in vitro anticancer activity.

RI PT

 Compounds showed good α-glucosidase inhibition.

AC C

EP

TE D

M AN U

SC

 Molecular docking, bioavailability and toxicity risk assessment were mapped.

ACCEPTED MANUSCRIPT

SUPPORTING INFORMATION Synthesis, docking and ADMET studies of novel chalcone triazoles for

RI PT

anti-cancer and anti-diabetic activity

Yakaiah Chinthalaa, Sneha Thakura, Shalini Tirunagaria, Srinivas Chindeb, Anand kumar Domattic, Niranjana Kumar Arigaria, Srinivas K.V.N.S. a, Sarfaraz Alamd, J. Kotesh Kumara*, Feroz Khand, Ashok Tiwaric, Paramjit Groverb

Natural Product Chemistry, CSIR-Central Institute of Medicinal and Aromatic PlantsResearch Centre, Boduppal, Hyderabad-500092, India b Toxicology Unit, Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India. c Medicinal chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad – 500007, India. d Metabolic and Structural Biology Department, CSIR-CIMAP, Lucknow-226015, UP

*

M AN U

SC

a

Corresponding: Telefax: +91-40-27202602; Email: [email protected]

TE D

Abstract

AC C

EP

A series of novel chalcone-triazole derivatives were synthesized and screened for in vitro anticancer activity on the human cancer cell lines IMR32 (neuroblastoma), HepG2 (Human hepatoma) and MCF-7 (Human breast adenocarcinoma), DU-145 (Human prostate carcinoma), and A549 (Human Lung adenocarcinoma). Among the tested compounds, 4r showed the most promising anticancer activity in all the cell lines whereas, compounds 4c (IC50 65.86 µM), 4e (IC50 66.28 µM), 4o (IC50 35.81 µM), 4q (IC50 50.82 µM) and 4s (IC50 48.63 µM) showed better activity than the standard doxorubicin (IC50 69.33 µM) in A549 cell line alone. Rat intestinal α-glucosidase inhibitory activity of the synthesized derivatives showed 4m (IC50 67.77 µM), 4p (IC50 74.94 in µM) and 4s (IC50 102.10 µM) as most active compared to others. The in silico docking of synthesized derivatives 4a-4t with DNA topoisomerase IIα revealed the LibDock score in the range of 71.2623 to 118.29 whereas, compounds 4h, 4m, 4p and 4s with docking target α-glucosidase were in the range of 100.372 to 107.784. Key words:

Chromanochalcones, 1,2,3-triazoles, Anticancer activity, α-Glucosidase inhibition, Molecular docking.

1

ACCEPTED MANUSCRIPT

Molecular docking and in silico pharmacology studies Table S1: Compliance of compounds to the theoretical parameters of oral bioavailability and drug likeness properties. Cytochrome P450 2D6 Binding non inhibitor

Hepatotoxici ty

4d

very low, but possible very low, but possible very low, but possible yes, low

undefined

non inhibitor

Toxic

undefined

non inhibitor

Toxic

undefined

non inhibitor

Toxic

4e

yes, low

undefined

non inhibitor

Toxic

poor

4f

undefined

non inhibitor

Toxic

very poor

non inhibitor

Toxic

very poor

non inhibitor

Toxic

poor

non inhibitor

Toxic

poor

non inhibitor

Toxic

poor

4k

very low, but possible very low, but possible very low, but possible very low, but possible very low, but possible yes, low

non inhibitor

Toxic

poor

4l

yes, low

4m

4o

very low, but possible very low, but possible yes, low

4p 4q

4b 4c

4g 4h 4i 4j

4s 4t

undefined undefined undefined

Doxorubic in Acarbose

poor

poor

poor

undefined

non inhibitor

Toxic

poor

undefined

non inhibitor

Non-Toxic

very poor

undefined

non inhibitor

Toxic

very poor

undefined

non inhibitor

Toxic

poor

yes, low

undefined

non inhibitor

Toxic

poor

very low, but possible yes, low

undefined

non inhibitor

Non-Toxic

very poor

undefined

non inhibitor

Toxic

poor

yes, low

undefined

non inhibitor

Toxic

poor

very low, but possible yes, low

undefined

non inhibitor

Non-Toxic

very poor

undefined

non inhibitor

Toxic

very poor

very low, but possible

undefined

non inhibitor

Non-Toxic

very poor

AC C

4r

undefined

EP

4n

undefined

Toxic

SC

4a

Intestinal absorptio n poor

2

Plasma protein binding highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded highly bounded poorly bounded poorly bounded

RI PT

Blood brain barrier penetration undefined

M AN U

Aqueous solubility

TE D

Compoun d

ACCEPTED MANUSCRIPT

Table S2: In-silico screening of chalcone 1,2,3 triazole derivatives for toxicity risk assessment 4b

4c

4d

4e

4f

4g

4h

4i

4j

4k

4l

4 m

4n

4o

Rat Oral LD50 (g/kg_body_we ight) Rat Inhalational LC50 (mg/m3/h) M Carcinog o enic u Potency s TD50 e (mg/kg_b ody_weig R ht/day) a t Rat Maximum Tolerated Dose feed (g/kg_body_we ight) Developmental Toxicity Potential

1. 26

1. 82

1. 07

1. 09

1. 24

1. 40

0. 60

0. 81

1. 17

0. 69

0. 82

0. 81

0. 91

0. 46

2. 19

2. 13

1. 29

1. 43

1. 06

1. 53

4. 5

1. 4

1. 36

0. 82

0. 91

1. 06

0. 98

3. 19

1. 4

1. 01

3. 02

3. 57

11 .1

3. 89

7. 56

10 .9

3. 71

1. 85

2. 19

6. 86

3. 88

4. 64

6. 71

3. 71

3. 41 7

22 .6

24 .6

48 .8

2. 85

56 .4

22 .1

22 .6

24 .6

48 .8

2. 85

56 .4

22 .8

0. 19 2

0. 14 6

0. 11 4

0. 18 5

0. 15 6

0. 29 6

0. 19 2

0. 14 6

0. 11 4

0. 18 5

11 0. 8 0. 24 6

22 .1

0. 29 6

11 0. 78 0. 24 6

0. 15 6

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 10 .8 54 5 St ro ng 0. 00 1

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 6. 01 07 4 St ro ng 0. 00 2

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n M ult iCa rci no ge n N on M ut ag en 6. 94 28 8 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 4. 68 21 7 St ro ng 0. 00 2

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 12 .9 56 9 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 8. 56 03 8 St ro ng 0. 00 4

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 4. 48 48 2 St ro ng 0. 00 1

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 8. 90 27 3 St ro ng 0. 00 1

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 4. 92 99 4 St ro ng 0. 00 1

N on To xi c N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n M ult iCa rci no ge n N on M ut ag en 5. 69 44 7 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 3. 24 42 1 St ro ng 0. 00 2

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 10 .6 27 1 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 7. 02 11 2 St ro ng 0. 00 4

Mou se male

Rat fema le

AC C

Rat Male

Ames Mutagenicity

Daphnia EC50 (mg/l)

Skin Sensitization Rat Chronic LOAEL (g/kg_body_we ight)

3

4q

4r

4s

4t

1. 22

1. 43

1. 41

0. 79

0. 92

Do xo ru bic in 0.3 1

Ac ar bo se

7. 94

6. 48

0. 64

5. 07

4. 14

0.0 8

0. 00 1

35 .6

39 .7

2. 1

21 .9

24 .4

6.9 7

0. 65

44 6. 5 0. 34 6

0.6 6

0. 1

0.2 77

1. 30 2

11 .1

44 6. 5 0. 34 6

22 .8

0. 17 4

46 9. 9 0. 29 3

0. 17 4

46 9. 9 0. 29 3

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xi c

To xic

To xi c

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 3. 10 72 5 St ro ng 0. 00 1

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n M ult iCa rci no ge n N on M ut ag en 2. 44 06 5 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 1. 22 62 9 St ro ng 0. 01 1

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 0. 25 67 85 St ro ng 0. 01 0

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n M ult iCa rci no ge n N on M ut ag en 2. 00 17 9 St ro ng 0. 00 3

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 1. 00 57 9 St ro ng 0. 01

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 0. 21 06 12 St ro ng 0. 00 9

No nCa rci no ge n

N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on Ca rci no ge n N on M ut ag en 3. 28 98 8 W ea k 0. 02 4

M AN U

TE D

Mou se fema le

EP

FDA Rode nt Carci nogen icity

4p

RI PT

4a

SC

Compound

No nCa rci no ge n No nCa rci no ge n No nCa rci no ge n M uta ge n

9.7 79 97 W ea k 0.0 13

ACCEPTED MANUSCRIPT

Ocular Irritancy Skin Irritancy

N on D eg ra da bl e M ild

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

De gr ad ab le

N on De gr ad ab le

N on De gr ad ab le

De gr ad ab le

No nDe gr ad abl e

De gr ad ab le

M ild

M ild

M ild

M ild

M ild

M ild

M ild

M ild

M ild

M ild

M ild

N on e

M ild

N on e M ild

N on e M ild

N on e M ild

Mi ld

N on e

N on e M ild

M ild

M ild

N on e M ild

M ild

M ild

N on e M ild

M ild

N on e

N on e M ild

M ild

N on e

N on e M ild

Se ve re M ild

M ild

No ne

RI PT

Aerobic Biodegradabili ty

Abbreviations: EC50, effective concentration 50%; US FDA, United States Food and Drug administration; LC50, lethal concentration 50%; LD50, lethal dose 50%; LOAEL, lowest observed adverse effect level; TD50, tumorigenic dose 50%.

4.2 Molecular doking studies

SC

Protein preparation

The protein preparation protocol is used to perform different tasks such as

M AN U

inserting missing atoms in incomplete residues, deleting alternate conformations (disorder), removing waters, modeling missing loop regions, and protonating titratable residues by using predicted pKs (negative logarithmic measure of acid dissociation constant). Chemistry at HARvard Macromolecular Mechanics (CHARMM); Cambridge, MA, USA) is used for protein preparation. The hydrogen atoms were added before the processing. Protein coordinates from the crystal structure of Topoisomerase IIα (PDB

TE D

[Protein Data Bank] ID: 1ZXM) Chain A determined at a resolution of 1.87 Å were used (Fig. 1 in the main manuscript) for anticancer studies and crystal structure of αglucosidase (PDB ID 2QMJ) at a resolution of 1.90 Å was used for antidiabetic studies

EP

(Fig. 2 in the main manuscript). Protein–ligand docking

AC C

Discovery Studio, version 3.5 (Accelrys, San Diego, CA, USA) software was used for molecular docking studies, 3D visualization and post docking analysis. In this study the ligand poses are placed into the polar and apolar receptor interactions site. The Merck Molecular Force Field was used for energy minimization of the ligands and Conformer Algorithm, based on Energy Screening And Recursive build-up (CAESAR) was used for generating conformations. All other docking and consequent scoring parameters used were kept at their default settings. Protein ligand complexes were also analyzed to better understand the interactions between protein residues and bound ligands, along with the binding site residues of the defined receptor. 4

ACCEPTED MANUSCRIPT

Bioavailability and drug likeness screening The pharmacokinetic properties were calculated by using different standard

RI PT

descriptors to check the compliance of chalcone derivatives. For this aqueous solubility, intestinal absorption, hepatotoxicity, cytochrome P450 2D6 binding, blood brain barrier penetration and plasma protein binding were evaluated through Discovery Studio v3.5. Toxicity Studies

SC

The toxicities that are oftentimes utilized in the drug development process are calculated through TOPKAT protocols of Accelrys DS 3.5. These predictions help in

M AN U

optimizing therapeutic ratios of lead compounds for further growth and evaluating their different potential safety concerns. These predictions also used in evaluating intermediate metabolites, along with setting a dose range of animal assays.

EP

4b

AC C

4a

TE D

Figure S1: 2D docking diagrams of Topoisomerase IIα–Chalcone triazole interactions:

4c

4d 5

4f

M AN U

4e

SC

RI PT

ACCEPTED MANUSCRIPT

TE D

4h

4i

AC C

EP

4g

4j

6

4l

4n

4o

4q

AC C

EP

4m

TE D

M AN U

SC

4k

RI PT

ACCEPTED MANUSCRIPT

4p

4r 7

4t

M AN U

SC

4s

RI PT

ACCEPTED MANUSCRIPT

TE D

Doxorubicn pink indicates electrostatic interaction; purple indicates covalent bond;and green indicates van der–Waals molecular interaction; High shading indicates more exposure to solvent

AC C

EP

Figure S2: 2D docking diagrams of α-glucosidase –Chalcone triazole interactions:

4h

4m

8

4s

Acarbose

TE D

M AN U

SC

4p

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

pink indicates electrostatic interaction; purple indicates covalent bond;and green indicates van der–Waals molecular interaction; High shading indicates more exposure to solvent

9

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

NMR and Mass Spectra of Chalcone triazole derivatives

10

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

11

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

12

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

13

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

14

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

15

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

16

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

17

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

18

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

19

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

20

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

21

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

22

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

23

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

24

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

25

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

26

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

27

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

28

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

29

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

30

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

31

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

32

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

33

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

34

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

35

Synthesis, docking and ADMET studies of novel chalcone triazoles for anti-cancer and anti-diabetic activity.

A series of novel chalcone-triazole derivatives were synthesized and screened for in vitro anticancer activity on the human cancer cell lines IMR32 (n...
6MB Sizes 0 Downloads 6 Views