You are on page 1of 12

Received: 5 April 2021 Revised: 7 June 2021 Accepted: 17 June 2021

DOI: 10.1002/jhet.4331

ARTICLE

Synthesis and bioactivity evaluation of eugenol


hybrids obtained by Mannich and 1,3 dipolar
cycloaddition reactions

Komuraiah Buduma1 | Niranjana Kumar A1 | Satya Srinivas KVN1 |


Kotesh Kumar J1 | Srinivas Chinde2 | Anand Kumar Domatti3 |
Yogesh Kumar4 | Paramjit Grover2 | Ashok Tiwari2 | Feroz Khan4

1
Natural Product Chemistry Division,
CSIR-Central Institute of Medicinal and
Abstract
Aromatic Plants, Research Centre, Design, synthesis, and bioactivity evaluation of novel mannich bases (2a-2j)
Hyderabad, India and triazole-chalcone derivatives (7a-7k) of Eugenol 1 were reported. Among
2
Toxicology Unit, Biology Division,
all the derivatives tested for antiproliferative activity, di-amine manich deriva-
CSIR-Indian Institute of Chemical
Technology, Hyderabad, India tive 2b (32.92 μM), and 4-methoxy chalcone triazole derivative 7d (33.05 μM)
3
Medicinal Chemistry and Pharmacology significantly inhibited HepG2 cell lines when compared to the standard doxo-
Division, CSIR-Indian Institute of rubicin (37.29 μM). Whereas most of the compounds such as diethylamine 2a
Chemical Technology, Hyderabad, India
4
(17.75 μM), (aminomethyl) methane diamine 2b (17.02 μM), and bis
Metabolic and Structural Biology
Department, CSIR-Central Institute of (chloromethyl) amine 2c (20.12 μM) showed moderate to better inhibition
Medicinal and Aromatic Plants, Lucknow, towards MCF-7 cell lines. The synthesized analogues were also tested for anti-
India
diabetic and antiobesity potentials. Compounds 2f (55.50%), 2c (54.34%), 7g
Correspondence (55.5%), and 2a (55.5%) have shown moderate inhibitory potentials toward
K. V. N. Satya Srinivas, Natural Product intestinal α-glucosidase enzyme when compared to the standard Acarbose
Chemistry Division, CSIR-Central
Institute of Medicinal and Aromatic
(72.86%). Likewise, compounds 7d (82.95%), 7f (76.19%), 7g (74.81%), 7e
Plants, Research Centre, Boduppal, (74.81%), and 2g (72.50%) have shown significant to moderate inhibitory
Hyderabad-500 092, India. potentials toward Pancreatic lipase enzyme when compared to the standard
Email: kvn.satyasrinivas@cimap.res.in
orlistat (91.10%). ROS induces life-threatening diseases like diabetes, cancer,
J. Kotesh Kumar, Natural Product
Chemistry Division, CSIR-Central
etc., and antioxidants play a major role in controlling their production. Com-
Institute of Medicinal and Aromatic pounds 2c (99.81%), 2i (99.80%), 2d (99.26%), 2g (98.79%), and 2f (98.42%) have
Plants, Research Centre, Boduppal, shown significant antioxidant profiles in ABTS assay when compared to the
Hyderabad-500 092, India.
Email: koteshkumarj@yahoo.com standard Trolox (99.07%). Further, In silico Molecular docking and pharmaco-
kinetic screening of the eugenol derivatives complemented the in vitro results
indicating the drug likeness of the obtained active compounds.

KEYWORDS
bioactivity screening, eugenol, Mannich bases, molecular docking, triazolechalcones

1 | INTRODUCTION

Komuraiah Buduma and A. Niranjana Kumar have equal contribution The discovery, development, and application of natural
and considered as first authors. drugs have been done for thousands of years. A recent

J Heterocyclic Chem. 2021;1–12. wileyonlinelibrary.com/journal/jhet © 2021 Wiley Periodicals LLC. 1


2 BUDUMA ET AL.

review by Newman and Cragg shown that 74.8% of small The aim of this research was to develop novel library
molecules used for cancer are from nonsynthetic origin [1]. of compounds such as manich and triazole chalcone
While semi-synthetic modification of naturally occurring bio- derivatives from Eugenol to get biologically active Euge-
active phytochemicals leads to the generation of most suc- nol hybrids.
cessful anticancer drugs such as paclitaxel I from 10-
deacetylbaccatin, etoposide II from podophyllotoxin,
irinotecan/topotecan from camptothecin and many more [2]. 2 | RESULTS A ND DISCUSSION
Natural products proved to be an inexhaustible resource of
novel and effective anticancer agents and remain 2.1 | Chemistry
unchallenged by other alternative drug discovery approaches
[3]. Due to the unique and vast chemical diversity, natural 2.1.1 | Synthesis of Manich bases
products are recognized as privileged scaffolds because they
interact with biological macromolecules, especially proteins Aminoalkyl derivative of Eugenol 1 was synthesized by
[4,5]. Eugenol or 4-allyl-2-methoxyphenol, majorly found in Mannich reaction via condensation of substituted second-
the essential oil of Ocimum Species, used as raw material in ary amine in the presence of formaldehyde to obtain com-
the manufacture of several drugs has attracted attention due pounds (2a–2j) (Scheme 1). The bases were characterized
to its wide range of biological activities [6] such as hypoten- by spectroscopy. The sharp bands in IR at 3280 and
sive [7], antioxidant [8,9], antitumoral [10], etc. As an essen- 3350 cm1 indicated the presence of N─H group, 3050
tial oil and antimicrobial agent, Eugenol has wider and 3034 cm1 for C─H, 2925, and 2923 cm1for O─H
applications in fragrance and flavoring industry cosmetic and functional groups.1H NMR spectral data of all the deriva-
food industry [11,12]. Furthermore, many recent studies have tives (2a–2j) showed characteristic signal between δ 3.57
explored the anticancer potential of eugenol and its ability to and 3.68 ppm and in 13C NMR, a new signal appeared
induce apoptosis in diverse cancer cell lines as well as in between δ55.39 and 58.71 ppm due to the presence of
in vivo tumor models [13,14]. methelene (CH2), which confirm insertion of aminoalkyl
1,2,3-Triazole and its derivatives enhanced consider- moiety in 1.
able attention for the past few decades due to their
chemotherapeutical value. Many 1,2,3-triazoles are found
to be more potent antimicrobial [15], antiinflammatory 2.1.2 | Synthesis of triazole chalcones
[16], antimalarial [17], anti-HIV [18], and anticancer
activities [19]. Some of the 1,2,3-triazoles were used as In the second series, the triazole chalcones were synthe-
DNA cleaving agents [20] and potassium channel activa- sized by selective O-propargylation at the hydroxyl group
tors [21,22]. These moieties have been widely used in the of 1 to get the intermediate 3 which were further coupled
synthetic intermediates and industrial applications, such with different substituted azides by employing facile 1,3
as dyes, anticorrosive agents, photo stabilizers, photo- dipolar cycloaddition [35,36] to get distinct derivatives of
graphic materials, and agrochemicals [23]. For example, 1,2,3-triazol-4-yl-eugenol (7a–7k) (Scheme 2).
carboxyamidotriazole III is shown as an angiogenesis The 1H NMR signals appeared at δ 5.35–5.39 ppm
inhibitor in cancer therapy [24], rufinamide IV exhibits confirmed the condensation of propargyl bromide. The
anticonvulsant activity and has been used to treat child- signal at δ 63.34–63.38 ppm attributed to carbon of
hood mental impairment of the Lennox-syndrome [25]. the active methylene inserted after O-propargylation of 1.
1
Mannich reaction is the most fundamental way to H NMR of all the triazole chalcone derivatives (7a–7k)
introduce the C─C bond formation in organic synthesis has shown a characteristic singlet for triazole proton in
[26]. Mannich base is a three-component condensation the range δ8.16–8.19 ppm. In addition to that, 13C NMR
product of active hydrogen-containing compound, alde- spectra exhibited characteristic signals for triazole ring
hyde, and a secondary amine. The formation of mannich carbon at δ119.8–121.6 ppm and 144.2–145.8 ppm,
base depends on the nucleophilicity of substrate and pH respectively.1H NMR of all the triazole chalcone deriva-
of the reaction medium [27]. The Mannich bases are tives (7a–7k) has shown a characteristic signal for
interested pharmacologically because of their antimicro- chalcone protons in the range δ7.60–7.91 ppm and 8.10–
bial [28], antimalarial [29], anti-HIV [30], antitubercular 8.26 ppm. In addition to that, 13C NMR spectra exhibited
[31], anticancer V and VI [32], antiinflammatory [33], characteristic signals for chalcone carbons at δ119.8–
and antioxidant properties [34]. Few important com- 121.6 ppm and 143.8–146.2 ppm, respectively. In addition
pounds, belonging to the above said pharmacophore cate- to these, 13C NMR spectra exhibit characteristic signals
gories, which gained attention in drug discovery are for carbonyl group of chalcone at δ189.20–196.2 ppm
shown in Figure 1. clearly indicates the formation of triazole chalcone
BUDUMA ET AL. 3

OAC O O
O O H
HO
NH OH
H
O O
HO O
H H O
HO O HO O
H O Cl O
O NH2
OAC O H2N
O
N N
O O OH Cl Cl N
O O
I II III

OH
F
O
N N
N
N N NH2 N OH
F OH
OH
OH
IV V VI

FIGURE 1 Structures of some biologically active derivatives

S C H E M E 1 Synthesis of Mannich OH
OH
derivatives (2a–2j) [Reagents and O
i O
conditions: Formaldehyde, derivatives of R
secondary amine, dry methanol, RT, 2–4 h]

1 2a-2j

Where R =

N N N
N N Cl OH
NH2
OH NH
NH2 Cl
2a 2c 2d 2e
2b

N N N N
N
N N O
COOH
O
2f HO 2g 2h 2i 2j

analogs. The synthetic yields, melting points, molecular evaluated in in vitro mode using 3-(4,5-dimethylthiazol-
weights, and NMR spectra of all the compounds are 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay [37]
described in the experimental section. on the selected human cancer cell lines A-549 (lung ade-
nocarcinoma), Hep-G2 (hematoma), MCF-7 (breast ade-
nocarcinoma), and SKOV3 (ovarian carcinoma). The
2.2 | Pharmocology assay was dependent on the reduction of tetrazolium salt
by the mitochondrial dehydrogenase of viable cells to
2.2.1 | Antiproliferative activity form a blue formazan product dissolved in DMSO and
measured at 570 nm. The results of cytotoxic activity
Antiproliferative activity of the synthesized mannich were expressed as the IC50 (μM), and doxorubicin was
bases and chalcone-triazole derivatives of Eugenol was used as positive control (Table 1). Compounds di-amine
4 BUDUMA ET AL.

Step-I Propargylation S C H E M E 2 Synthesis of Triazole


chalcone derivatives (7a–7k). [Reagents
O O and conditions: i. K2CO3, DMF, RT
OH i O 30 min, ii. a. ii) H2O:HCl (1:1), 0 C,
aq. NaNO2, aq. NaN3, 2 h (5a–5k).ii. b.
R-CHO, EtOH, KOH, RT 1 h, iii. CuI,
THF, RT, 12 h]
1 3

Step-II Chalcone formation

NH2 N3 N3
ii ii
a
b

O O O

R
4
5a-5k 6a-6k
Step-III Triazole formation

O N3 O N N
O O N
iii O

O R
7a-7k
3 6a-6k R

Where R=

F O Cl Cl
7a 7b 7c 7d 7e Cl 7f

O S
O
O 2N
O
7g 7i 7j 7k
7h

manich derivative 2b (32.92 μM) and 4-methoxy chalcone pancreas Type II (Sigma product L3126), was dissolved in
triazole derivative 7d (33.05 μM) have significantly inhibited ultrapure water at 10 mg/ml and then the supernatant was
HepG2 cell lines when compared to the standard doxorubi- used after centrifugation at 16,000 rpm for 5 min. The assay
cin (37.29 μM). Compound 2b (16.43 μM) was moderately buffer consisting of 100 mM Tris buffer (pH 8.2) and
active against A549 cell lines when compared to the stan- p-nitrophenyl laurate (pNP laurate) was used as the sub-
dard doxorubicin (7.22 μM).While 2b (19.06 μM) and 2a strate. The substrate stock 0.08% w/v pNP laurate was dis-
(21.78 μM) were moderately active against KVO3 cell lines solved in 5 mM sodium acetate (pH 5.0) containing 1%
when compared to the doxorubicin (11.34 μM). Among the Triton X-100 and was heated in boiling water for 1 min to
four cell lines, the majority of derivatives showed inhibitory aid dissolution, mixed well, and then cooled to room tem-
property toward MCF-7 cell line. perature. The control assay contained 400 μl assay buffer,
450 μl substrate solution, and 150 μl lipase. The synthesized
compounds were dissolved in DMSO (1 mg/ml) and added
2.2.2 | Antilipase activity in 50 μl total volume. The buffer, enzyme, and compounds
were added and then substrate was added to start the reac-
Synthesized eugenol derivatives were evaluated for inhibi- tion. The samples were incubated at 37 C for 2 h. Then
tory potentials of pancreatic lipase enzyme in in vitro mode samples were centrifuged at 16,000 rpm for 2.5 min and
as per the previous reports [40,41]. Lipase, from porcine read at 400 nm in a UV spectrophotometer. An inhibitor
BUDUMA ET AL. 5

TABLE 1 Pharmacological activities of triazole chalcone derivatives (7a–7k)

Anticancer activity (IC50 in μM/ml) Antioxidant activity

DPPH % ABTS %
Inhibition at Inhibition at α-Glucosidase LIPASE %
25 μg/ml (SC50 10 μg/ml Inhibition (%) Inhibition at
Compound A549 HepG2 MCF-7 SKOV3 μg /m l) (SC50 μg/ml) at 20 μg/ml 50 μg/ml
2a > 100 77.72 17.75 21.78 1.24 4.82 51.13 61.29
2b 16.43 32.92 17.02 19.06 54.07 NV 48.49 60.20
2c > 100 > 100 20.12 > 100 30.67 99.81 54.34 NV
2d > 100 > 100 22.15 > 100 46.42 99.26 34.66 45.93
2e > 100 > 100 23.64 > 100 42.10 98.79 42.83 NV
2f > 100 > 100 21.80 > 100 46.52 98.42 55.50 69.12
2g > 100 > 100 33.38 > 100 64.15 98.79 41.35 72.50
2h > 100 77.70 28.60 > 100 54.18 NV 14.41 NV
2i > 100 > 100 36.60 > 100 24.69 99.80 38.59 29.80
2j > 100 > 100 24.98 > 100 60.34 NV 42.77 NV
7a > 100 > 100 33.87 > 100 38.28 NV NV NV
7b > 100 > 100 42.06 > 100 00.86 09.83 26.82 41.01
7c > 100 > 100 32.36 > 100 04.47 05.01 15.37 55.91
7d > 100 33.05 32.77 > 100 05.18 05.38 17.23 82.95
7e > 100 > 100 37.40 > 100 09.54 04.73 32.15 74.81
7f > 100 > 100 31.59 > 100 03.99 04.17 20.77 76.19
7g > 100 > 100 30.34 > 100 15.42 05.75 54.86 74.81
7h > 100 > 100 20.07 > 100 NV 08.07 37.62 NV
7i > 100 > 100 24.00 > 100 08.73 12.80 23.73 43.78
7j > 100 > 100 20.08 > 100 29.87 07.05 07.27 NV
7k > 100 > 100 28.87 > 100 20.75 10.85 NV 14.90
Doxo 07.22 37.29 02.96 11.34 — — — —
Ascorbic Acid — — — — 86.52 — — —
Trolox — — — — — 99.07 — —
Acarbose — — — — — 72.86
Orlistst — — — — — — — 91.1

The bold values are relatively showing significant activities.

blank was prepared for each sample and all were assayed in properly, and the supernatant was used as a source of
triplicate. According to the results given in Table 1, com- crude intestinal α-glucosidase after centrifugation. In
pound 7d (82.95%) inhibited the enzyme more potentially, brief, 10 ml of test samples (5 mg/ml DMSO solution)
whereas compounds 7f (76.19%), 7e (74.81%), 7g (74.81%), were reconstituted in 100 ml of 100 mM phosphate buffer
and 2g (72.50%) inhibited moderately when compared to (pH 6.8) in 96-well microplate and incubated with 50 ml
the standard orlistat (91.10%). yeast α-glucosidase (0.76 U/ml in same buffer) or crude
intestinal α-glucosidase for 5 min before 50 ml substrate
(5 μM, p-nitrophenyl-α-D-glucopyranoside prepared in
2.2.3 | Anti α-glucosidase activity same buffer) was added. Release of p-nitrophenol was
measured at 405 nm spectrophotometrically 5 min after
The inhibitory potentials of the synthesized compounds incubation with substrate. Individual blanks for test sam-
against intestinal α-glucosidase enzyme were determined ples were prepared to correct background absorbance
as per the reported methods [42,43]. Rat intestinal ace- where substrate was replaced with 50 ml of buffer. Con-
tone powder in normal saline (100:1; w/v) was sonicated trol sample contained 10 ml DMSO in place of test
6 BUDUMA ET AL.

samples. Acarbose was taken as standard reference for mode on different anticancer targets of selected human
α-glucosidase inhibition. All the samples were assayed in cancer cell lines, that is, lung cancer cell line (A549) tar-
triplicate. As shown in Table 1, among all the synthesized get histone deacetylase (PDB: 1C3S) [46], liver cancer cell
derivatives, compound 2f (55.50%) exhibited better inhib- line (HepG2) target DNA topoisomerase II (PDB: 1ZXM)
itory activity, whereas compounds 2c (54.34%), 7g [47], ovarian cancer cell line (SKOV3) target STAT3
(54.86%), 2a (51.13%) showed moderate inhibition when (Signal Transducer and Activator of Transcription) (PDB:
compared to the standard drug Acarbose (72.86%). 3CWG) [48], and breast cancer cell line (MCF7) target
Beta Tubulin (PDB: 1SAO) [49]. Out of all the tested
derivatives, compounds 2b, 2a, 7d, 7h, 7i, 7j, 2h, 2j, 2c,
2.2.4 | Antioxidant activity 2d, 2e, 2g, 2i, 7g, 7e, and 7f have shown higher docking/
binding energy for the mentioned targets of given cell
The antioxidant and free radical scavenging activity of the lines when compared to the reported binding affinity of
Mannich bases and chalcone triazoles were measured by standard anticancer drug Doxorubicin [50]. According to
using DPPH and ABTS assays. The 2,2-diphenyl- the results given in Table S1, the compound 2b showed
1-picrylhydrazyl (DPPH) is a stable free radical, which is the higher docking/binding energy (15.8365 kJ/mol)
widely used for estimating free radical scavenging activities against target 1C3S of A549 cell line. Compounds 2b and
of antioxidants. DPPH-based free radical scavenging activ- 7d showed significant docking/binding energy (22.8615
ity usually involves hydrogen atom transfer reaction [38]. and 22.6069 kJ/mol) against target 1ZXM of HEPG2
The reducing power of Mannich bases was determined by cell line. Compounds 2a (11.5349 kJ/mol), 2b
using the standard method [39]. For the above studies, (27.2327 kJ/mol), 7h (27.4181 kJ/mol), 7i (18.5166
ascorbic acid was used as reference material. Antioxidant kJ/mol), and 7j (18.4119 kJ/mol) showed higher dock-
activity of all the compounds was measured by using ABTS ing/binding energy, respectively, on docking with targe
assay. As per the Table 1, compounds 2c (99.81%), 2i 1SAO of MCF7 cell line. Similarly, compounds 2b-
(99.80%), 2d (99.26%), 2g (98.79%), and 2f (98.42%) have (23.9803 kJ/mol) and 2a (14.4954 kJ/mol) showed
shown potential antioxidant activity when compared to the higher docking/binding energy on docking with targe
standard Trolox (99.07). Whereas DPPH assay showed that 3CWG of SKVO3 cell line, respectively, and formed
2g (64.15%), 2j (60.34%) were moderately active when com- hydrogen bonds with the interacting residues within the
pared to the standard ascorbic acid (86.52%). radius of 4 Å. All the above results complement the
reported in vitro results and are also comparable with the
docking/binding energy of Doxorubicin with respective
2.3 | In silico molecular docking targets of A549 (26.0596 kJ/mol), HepG2 (27.7295
kJ/mol), MCF7 (32.5618 kJ/mol), and SKOV3
The molecular docking and visualization studies of eugenol (30.0418 kJ/mol) cell lines, respectively. The docking
derivatives were performed through FlexX [44] docking pro- pose of all the compounds showed significant interaction
gram available in Lead IT 2.1.8 version of (Bio Solve IT Ger- within the active site of respective targets. Moreover,
many). FlexX is a fast algorithm for flexible docking of small potential hydrophobic contacts were found at the active
ligands into the active protein-binding site using an incremen- site of receptor (Tables S1 and S3).
tal construction process. During docking standard parameters
were kept in the FlexX program as implemented in Lead IT
v2.1.8. Chemical structure designing, energy minimization, 2.3.2 | In silico molecular docking studies for
and geometry cleaning were done through Chem Draw [45] antidiabetic and antilipase targets
and Chem 3D 15.0.0.16 version (Perkin Elmer Informatics).
The protein 3D crystallographic structures for docking were The synthesized Eugenol derivatives were tested for anti-
retrieved through RCSB Protein Data Bank (PDB) (www. diabetic activity by docking with target human intestinal
rcsb.org/). To find the possible bioactive conformations of maltase-glucoamylase (PDB: 2QMJ), for pancreatic anti-
eugenol derivative compounds, these softwares were used. lipase activity by docking with target colipase (PDB:
1LPB) and for antioxidant activity by docking with target
hematopoietic cell kinase Hck (PDB: 2HCK), respectively.
2.3.1 | In silico molecular docking study for The compounds 2a, 2c, 2g, and 7g which were experimen-
cancer targets tally validated as α-glucosidase inhibitors have docked in
the active pocket of 2QMJ and showed the higher binding
Eugenol derivatives synthesized in the above study were energy of 12.4824, 13.5502, 24.997, and 27.8332
evaluated for their docking/binding efficiency in in silico kJ/mol, respectively, and formation of hydrogen bonds.
BUDUMA ET AL. 7

The standard Acarbose showed comparable docking/bind- nonmutagenic except standard orlistat which is
ing energy of 28.3427 kJ/mol (Tables S2 and S3). predicted as mutagenic. Drug likeness evaluation was
Similarly docking studies for human pancreatic lipase done with the help of bi-plot between AlogP98 and
colipase (PDB: 1LPB) with compounds 2h (18.2392 PSA_2D (polar surface area) (Table S6). The bi-plot
kJ/mol), 7d (23.4847 kJ/mol), 7e (23.214 kJ/mol), 7f showed confidence ellipses of 95% and 99% for the BBB
(25.5355 kJ/mol), and 7g (25.4344 kJ/mol) have penetration and human intestinal absorption models
shown higher docking/binding energy and, respectively, (Figure S1). Most of the compounds were found inside
as compared to the control drug Orlistat (6.3146 the plot, which represent good bioavailability and
kJ/mol). This study showed that all the studied com- permeability, whereas the compounds shown outside
pounds were showing almost similar binding affinity having poor bioavailability and permeability. The toxic
with the targeted proteins. effect of the studied compounds was calculated in com-
parison to the standard control drugs (Doxorubicin,
Orlistat, Ascorbic acid, Acarbose, and Trolox) (Table S5).
2.3.3 | In silico pharmacokinetic parameters The computed toxicity results showed that all the com-
screening pounds were nonmutagenic, except Doxorubicin, most of
the compounds showed mild type of skin irritancy and
The pharmacokinetics (PK) is important to determine severe type of ocular irritancy.
the fate of substances administered externally to living
organism. PK is composed of important parameters as
ADMET (Absorption, Distribution, Metabolism, Excre- 3 | CONCLUSION
tion, and Toxicity). All these parameters will determine
the levels and kinetics of the drug exposed to the tissue The synthesis of novel derivatives of Eugenol 1 was car-
to influence the performance and pharmacological ried out by two distinct reactions, that is, by Mannich
action.Pharmacokinetic screening of eugenol deriva- reaction via condensation of substituted secondary amine
tives 2b, 2a, 7d, 7h, 7i, 7j, 2h, 2j, 2c, 2d, 2e, 2g, 2i, 7g, in the presence of formaldehyde to yield 2a–2j and tri-
7e, and 7f with controls Orlistat, Acarbose, Trolox, azole chalcones by selective O-propargylation at the
Ascorbic Acid, and Doxorubicin was performed by Dis- hydroxyl group of 1 afforded the intermediate 3 which
covery Studio v3.5 software (Accelrys), ADMET mod- was further, coupled with different substituted chalcone
ule. Mathematical predictive ADMET QSAR models azides by employing facile click chemistry to get distinct
were used for PK parameters such as aqueous solubil- derivatives of 1,2,3-triazol-4-yl-eugenol (7a–7k).All the
ity, blood–brain barrier (BBB) penetration, cytochrome synthesized compounds were screened for antip-
P4502D6 inhibition, hepatotoxicity, human intestinal roliferative, antidiabetic, antilipase, and antioxidant
absorption, and plasma protein binding. The studied activity in in-vitro mode. Under antiproliferative activity,
compounds were also evaluated against the Lipinski's compounds 2b and 7d were potent against HepG2 cancer
rule of five [51]. The toxicity that is often used in drug cell lines. α-Glucosidase inhibition results showed that
development, was calculated for all designed com- compounds 2f, 2c, 7g, and 2a were significant. Under
pounds by DS_TOPKAT module of Discovery Studio lipase inhibition activity compound 7d is potent. Three
3.5 software (Accelrys). Results showed that all the compounds 2c, 2d, and 2i shown more potent in vitro
compounds including standard drugs except Trolox, 2e, antioxidant activity than the standard Trolox. Further, in
2g, 2h, 2i, 2d, 2a, 2b, 2c, and ascorbic acid violated the silico molecular docking and pharmacokinetic screening
Lipinski rule of five, therefore seems to be low oral bio- of the eugenol derivatives complemented the invitro
available (Table S4). Evaluation through standard results indicating the drug likeliness of active
ADMET parameters showed that most of the studied compounds.
compounds have very low aqueous solubility level and
BBB penetration level similar to standard drugs. Most
of the compounds showed noninhibitor activity toward 4 | EXPERIMENTAL SECTION
Cytochrome (CYP-2D6), which is an important enzyme
of drug metabolism. Most of the compounds showed 4.1 | Chemistry
non hepatotoxicity (liver toxicity) except 7h, 7i, 7e, 2d,
2a, 2c, Doxorubicin, and Trolox, respectively. Plasma Melting points of all the compounds were recorded on
protein binding (PPB) was high for most of the com- Casia-Siamia (VMP-AM) melting point apparatus and
pounds thus seems to be easily distributed in the body. uncorrected. IR spectra were recorded on a Perkin–
All the studied compounds were predicted as Elmer FT-IR 240-C spectrometer using KBr optics.
8 BUDUMA ET AL.

NMR spectra were recorded on Burker Avance ([6-Allyl-2-hydroxy-3-methoxybenzyl]azanediyl)


500 MHz in CDCl3 and DMSO-d6 using TMS as internal dimethanol (2d)
standard. Electron impact (EI) and chemical ionization Viscous liquid, yield: 87%, 1H NMR (CDCl3, 500 MHz) δ
mass spectra were recorded on a VG Micro mass model (ppm): 3.34 (s, 2H), 3.64 (s, 2H), 3.65 (s, 3H), 4.77 (s, 4H),
7070H instrument. All the reactions were monitored on 4.91 (s, 2H),5.80 (m, 2H), 6.69 (m, 1H), 7.23 (d, 1H), 7.41
silica gel percolated TLC plates of Merck and spots (d, 1H), 8.71 (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ
were visualized with UV light. Silica gel (100–200 (ppm): 39.82, 58.71, 63.07, 86.72, 112.02, 115.88, 121.14,
mesh) used for column chromatography was procured 123.48, 130.22, 138.49, 145.17, 147.90. ESIMS (m/z)
from Merck. 254 (M+ 1) observed for C13H19NO4.

3-Allyl-6-methoxy-2-(piperazin-1-ylmethyl)phenol (2e)
4.2 | General procedure and spectral White solid, yield: 93%, m.p.145–147 C, IR (KBr): 2930,
data of synthesized compounds 2831, 2788, 1740, 1639, 1597, 1493, 1457, 1411, 1352,
1242, 1151, 1088, 1003, 908, 830, 795, 704 cm1; 1H NMR
4.2.1 | Synthetic procedures for amino alkyl (CDCl3, 500 MHz) δ (ppm): 1.24 (s, 1H), 2.85 (t, 4H), 3.24
derivatives (2a–2j) (t, 4H), 3.34 (d, 2H), 3.58 (s, 2H), 3.72 (s, 3H), 5.02 (m,
2H), 5.92 (m, 1H), 6.48 (s, 1H), 6.65 (s, 1H). 13C NMR
Compound 1 (1.0 equiv) was dissolved in anhydrous (125 Hz, DMSO-d6) δ (ppm): 39.85, 53.21, 55.53, 60.16,
methanol (5 ml) and added 37% formaldehyde solution 111.59, 115.32, 120.57, 121.99, 129.45, 138.14, 144.77,
50 ml after stirring 30 min at room temperature added 147.21. ESIMS (m/z) 263 (M+ 1) observed for
1.4 equivalent distinct derivatives of secondary amines. C15H22N2O2.
The reaction mixture was stirred at room temperature for
2–3 h. After reaction completion, the mixture was 6,60 -(Piperazine-1,4-diylbis[methylene])bis(5-allyl-
quenched with water and extracted with ethyl acetate 2-methoxyphenol) (2f )
three times. The solvent was evaporated in vacuum, and Colorless crystal, yield: 89%, m.p.141–143 C, IR (KBr):
the crude product was purified by column chromatogra- 2948, 2820, 1749, 1597, 1494, 1459, 1408, 1343, 1236,
phy using silica gel (100–200 mesh) as a stationary phase 1150, 1085, 1003, 909, 851, 792, 702 cm1; 13C NMR
to get pure products (2a–2j) with good yields. (125 Hz, DMSO-d6) δ (ppm): 39.84, 52.18, 55.76, 58.64,
111.64, 115.61, 121.16, 122.24, 129.92, 138.29, 144.30,
3-allyl-2-([diethylamino]methyl)-6-methoxyphenol (2a) 147.43. ESIMS (m/z) 439 (M+ 1) observed for
Colorless crystal, yield: 92%, m.p.149–151 C, 1H NMR C26H34N2O4.
(CDCl3, 500 MHz) δ (ppm): 1.16 (t, 6H), 2.47 (q, 4H), 3.29
(d, 2H), 3.51 (t, 4H), 3.57 (s, 2H), 3.70 (s, 3H), 4.94 (m, 3-Allyl-6-methoxy-2-([4-methylpiperazin-1-yl]methyl)
2H), 5.85 (m, 1H), 6.53 (s, 1H), 6.66 (s, 1H). ESIMS (m/z) phenol (2g)
250 (M+ 1) observed for C15H23NO2. White solid, yield: 96%, m.p. 153–155 C, IR (KBr): 3012,
2951, 2835, 2804, 1740, 1599, 1496, 1455, 1358, 1285,
3-Allyl-2-((bis(aminomethyl)amino)methyl)- 1222, 1150, 1087, 999, 817, 776, 706 cm1; 1H NMR
6-methoxyphenol (2b) (CDCl3, 500 MHz) δ (ppm): 2.09 (s, 3H), 2.51 (t, 4H), 3.73
Viscous liquid, yield: 84%, 1H NMR (CDCl3, 500 MHz) δ (t, 4H), 3.29 (d, 2H), 3.60 (s, 2H), 3.77 (s, 3H), 4.79 (m,
(ppm): 3.33 (s, 2H), 3.53 (s, 2H), 3.68 (s, 3H), 3.80 (m, 2H), 5.04 (m, 1H), 5.92 (s, 1H), 6.66 (s, 1H). 13C NMR
4H), 4.98 (m, 2H), 5.90 (m, 1H), 6.48 (d, 1H), 6.62 (d, 1H). (125 Hz, DMSO-d6) δ (ppm): 39.84, 49.56, 53.58, 55.77,
13
C NMR (125 Hz, DMSO-d6) δ (ppm): 39.59, 55.90, 56.51, 110.31, 111.37, 115.30, 120.91, 123.30, 129.47,
75.73, 110.64, 111.76, 112.00, 115.83, 119.24, 120.94, 138.13, 144.03, 147.39. ESIMS (m/z) 277 (M+ 1) observed
122.05, 123.62, 138.22, 144.34, 147.80. ESIMS (m/z) for C16H24N2O2.
252 (M+ 1) observed for C13H21N3O2.
3-Allyl-6-methoxy-2-(morpholinomethyl)phenol (2h)
3-Allyl-2-((bis(chloromethyl)amino)methyl)- Brown solid, yield: 90%, m.p.39–41 C, IR (KBr): 2961,
6-methoxyphenol (2c) 2822, 1740, 1596, 1493, 1457, 1399, 1234,1113, 1079,
Viscous liquid, yield: 90%, 1H NMR (CDCl3, 500 MHz) δ 990, 864, 796, 701 cm1; 1H NMR (CDCl3, 500 MHz) δ
(ppm): 3.29 (s, 2H), 3.63 (s, 2H), 3.69 (s, 3H), 4.24 (s, 4H), (ppm): 2.38 (t, 4H), 3.33 (d, 2H), 3.52 (t, 4H), 3.55 (s, 2H),
4.97 (m, 2H), 5.83 (m, 1H), 6.46 (d, 1H), 6.62 (d, 1H), 9.67 3.68 (s, 3H), 4.96 (m, 2H), 5.90 (m, 1H), 6.48 (s, 1H), 6.63
(s, 1H). ESIMS (m/z) 290 (M+ 1) observed for (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ (ppm): 39.81,
C13H17Cl2NO2. 52.86, 55.96, 59.28, 66.46, 111.85, 112.89, 115.73, 115.90,
BUDUMA ET AL. 9

120.99, 122.07, 130.46, 138.54, 144.34, 147.64. ESIMS (m/ 4.2.4 | Procedure for the synthesis of azido-
z) 264 (M+ 1) observed for C15H21NO3. chalcones (6a–6k)

1-(6-Allyl-2-hydroxy-3-methoxybenzyl)piperidine- To a solution of azido derivatives (5a–5k) in THF KOH


4-carboxylic acid (2i) (1.4 equiv) was added and a solution of 3 (1 equiv) in
White solid, yield: 94%, m.p.145–147 C, IR (KBr): 3363 methanol (5 ml), and the mixture was stirred at RT for
(br), 2963, 2933, 1739, 1568, 1501, 1437, 1393, 1342, 1 h. After dilution with water (100 ml), the resulting solid
1291, 1243, 1090, 921, 864, 778, 699 cm1; 1H NMR filtered off, washed with water, and recrystallized from
(CDCl3, 500 MHz) δ (ppm): 2.55 (t, 4H), 2.88 (t, 4H), petroleum ether to get yellow needles 6a–6k.
3.26 (d, 2H), 3.63 (s, 2H), 3.77 (s, 3H), 5.06 (m, 2H),
5.93 (m, 1H), 6.49 (s, 1H), 6.66 (s, 1H), 8.28 (s, 1H). 13C
NMR (125 Hz, DMSO-d6) δ (ppm): 27.76, 40.01, 51.68, 4.2.5 | Synthesis of chalcones triazole
55.39, 111.40, 115.05, 120.46, 129.43, 137.77, 144.60, derivatives (7a–7k)
147.05, 175.56. ESIMS (m/z) 306 (M+ 1) observed for
C17H23NO4. Intermediate 4-allyl-1-methoxy-2-(prop-2-yn-1-yloxy)ben-
zene 3 (1.0 equiv) was dissolved in dry THF (10 ml) and
3-Allyl-2-((sec-butyl[pentan-2-yl]amino)methyl)- catalytic amount of copper iodide was added. To this,
6-methoxyphenol (2j) azido- chalcones (6a–6k) (1.0 equiv), in dry THF, were
White solid, yield: 93%, m.p.147–149 C, 1H NMR (CDCl3, added slowly while stirring at room temperature under
500 MHz) δ (ppm): 1.15–1.58 (m, Cyclohexane ring nitrogen atmosphere after 12 h after the monitoring of
hydrogens), 2.88 (t, 4H), 3.22 (d, 2H), 3.59 (s, 2H), 3.82 (s, the reaction the solvent was removed under reduced
3H), 5.08 (m, 2H), 5.91 (m, 1H), 6.50 (s, 1H), 6.64 (s, 1H). pressure and the residue was diluted with distilled water
ESIMS (m/z) 358 (M+ 1) observed for C23H35NO2. and extracted thrice with ethyl acetate. The combined
organic layers were dried over anhydrous Na2SO4 and
concentrated to get the product. The crude product was
4.2.2 | Synthetic procedure for purified by column chromatography with ethyl acetate in
propargylation (3) hexane.

Eugenol 1 (1.0 equiv) along with 1.5 mol of potassium (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-


carbonate was taken in dimethyl formamide (DMF) in 1,2,3-triazol-1-yl)phenyl)-3-phenylprop-2-en-1-one (7a)
clean rb flask and added propargyl bromide (1.5 equiv) to Yellow solid, yield: 94%, m.p. 90–92 C, IR (KBr): 3138,
this solution. Reaction mixture was stirred at RT for 3069, 3002, 2936, 2834, 1738, 1659, 1603, 1511, 1453,
30 min to afford crude propargyl derivative. This crude 1415, 1336, 1260, 1222, 1137, 1022, 986, 913, 834, 765,
compound was column chromatographed over silica gel 694, 666 cm1; 1H NMR (CDCl3, 300 MHz) δ (ppm): 3.33
(100–200 mesh) and eluted with 10% ethyl acetate in n- (d, 2H), 3.93 (s, 3H), 5.10 (m, 2H), 5.36 (s, 2H), 5.96 (m,
hexane to obtain pure compound 4-allyl-1-methoxy- 1H), 6.73 (d, 2H), 7.00 (d, 1H), 7.43 (m, 2H), 7.55 (m, 2H),
2-(prop-2-yn-1-yloxy)benzene 3. 7.84 (m, 2H), 7.89 (m, 2H), 7.91 (m, 1H), 8.17 (s,
2H), 8.19 (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ (ppm):
39.84, 55.89, 63.37, 112.41, 114.52, 115.78, 120.20, 120.60,
4.2.3 | Synthetic procedures for 1-azido 120.89, 121.44, 128.60, 129.06, 130.27, 130.27, 130.90,
derivatives (5a–5k) 134.16, 134.64, 137.43, 138.13, 139.79, 145.78, 145.81,
149.59, 188.95. ESI-MS (m/z) 452 (M+ 1) observed for
Eleven aromatic ring substituted 1-azido derivatives C28H25N3O3.
were synthesized by dissolving 4-amino aetophenone
in H2O:HCl (1:1) and stirred at 0–5 C for 30 min. The (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-
aqueous NaNO2 was added drop wise to diazotize 1,2,3-triazol-1-yl)phenyl)-3-(4-fluorophenyl)prop-2-en-
amine hydrochloride. After stirring at 0–5 C, the solu- 1-one (7b)
tion was treated with aqueous NaN3 and further stirred Pale yellow solid, yield: 81%–83%, m.p. 109–111 C, IR
for 1 h. Finally, the reaction mixture was extracted (KBr): 3135, 3072, 2934, 1740, 1659, 1600, 1510, 1463,
with EtOAc, dried over Na2SO4 and concentrated in 1415, 1331, 1223, 1138, 1023, 986, 912, 820, 784 cm1; 1H
vacuo afforded eleven different substituted derivatives NMR (CDCl3, 300 MHz) δ (ppm): 3.33 (d, 2H), 3.93 (s,
of 1-azido compound (5a–5k in Scheme 2) in quantita- 3H), 5.10 (m, 2H), 5.36 (s, 2H), 5.97 (m, 1H), 6.74 (d, 2H),
tive yields. 7.00 (d, 1H), 7.44 (s, 1H), 7.48 (s, 1H), 7.67 (m, 2H), 7.83
10 BUDUMA ET AL.

(m, 2H), 7.91 (m, 2H), 8.16 (m, 2H), 8.18 (s, 1H). 13C (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-
NMR (125 Hz, DMSO-d6) δ (ppm): 39.83, 55.89, 63.37, 1,2,3-triazol-1-yl)phenyl)-3-(4-chlorophenyl)prop-2-en-
112.40, 114.49, 115.81, 116.18, 116.36, 120.20, 120.59, 1-one (7f )
120.85, 121.11, 130.24, 130.50, 130.57, 130.90, 134.15, Pale yellow solid, yield: 84%, m.p. 121–123 C, IR (KBr):
137.46, 138.01, 139.85, 144.41, 145.82, 145.87, 149.58, 3158, 3072, 3003, 2937, 1740, 1665, 1609, 1511, 1254,
163.28, 188.69. ESI-MS (m/z) 470 (M+ 1) observed for 1219, 1137, 1014, 912, 847, 806, 660 cm1; 1H NMR
C28H24 FN3O3. (CDCl3, 300 MHz) δ (ppm): 3.33 (d, 2H), 3.88 (s, 3H),
5.07 (m, 2H), 5.36 (s, 2H), 5.96 (m, 1H), 6.73 (d, 2H), 6.99
(E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H- (d, 2H), 7.52 (m, 4H), 7.83 (m, 1H), 7.88 (m, 2H), 8.15 (m,
1,2,3-triazol-1-yl)phenyl)-3-(p-tolyl)prop-2-en-1-one (7c) 2H), 8.23 (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ (ppm):
Yellow solid, yield: 95%, m.p.135–137 C, IR (KBr): 39.83, 55.89, 63.37, 112.41, 114.50, 115.81, 120.21, 120.60,
3137, 3070, 3002, 2918, 1741, 1656, 1600, 1512, 1460, 120.84, 121.89, 125.22, 129.91, 130.27, 132.33, 133.55,
1412, 1258, 1226, 1135, 1026, 988, 842, 810, 668 cm1; 134.16, 137.45, 137.89, 139.91, 144.27, 145.82, 145.89,
1
H NMR (CDCl3, 500 MHz) δ (ppm): 2.43 (s, 3H), 3.37 149.59, 188.61. ESI-MS (m/z) 486 (M+ 1) observed for
(d, 2H), 3.90 (s, 3H), 5.10 (m, 2H), 5.39 (s, 2H), 5.98 C28H24ClN3O3.
(m, 1H), 6.76 (d, 2H), 7.03 (d, 1H), 7.28 (s, 1H), 7.50
(m, 2H), 7.58 (m, 2H), 7.88 (m, 2H), 7.93 (m, 2H), 8.19 (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-
(s, 1H), 8.21 (s, 1H), 13C NMR (125 Hz, DMSO-d6) δ 1,2,3-triazol-1-yl)phenyl)-3-(2-nitrophenyl)prop-2-en-
(ppm): 21.59, 39.84, 55.90, 63.39, 112.41, 114.52, 1-one (7g)
115.79, 120.18, 120.44, 120.61, 120.88, 128.65, 129.82, yellow solid, yield: 86%, m.p. 97–99 C, IR (KBr): 3144,
130.23, 131.93, 134.15, 137.46, 138.31, 141.55, 145.89, 3080, 2937, 1658, 1603, 1513, 1418, 1341, 1258, 1221,
145.88, 149.60, 189.04. ESI-MS (m/z) 466 (M+ 1) 1138, 1017, 798, 753, 703, 667 cm1; 1H NMR (CDCl3,
observed for C29H27N3O3. 300 MHz) δ (ppm): 3.35 (d, 2H), 3.90 (s, 3H), 5.10 (m,
2H), 5.39 (s, 2H), 5.96 (m, 1H), 6.74 (d, 2H), 6.98 (d, 1H),
(E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H- 7.60 (m, 4H), 7.79 (m, 1H), 7.93 (m, 2H), 8.13 (m, 2H),
1,2,3-triazol-1-yl)phenyl)-3-(4-methoxyphenyl)prop-2-en- 8.20 (s, 1H) 8.22 (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ
1-one (7d) (ppm): 39.83, 55.89, 63.37, 112.44, 114.54, 115.80, 120.24,
Yellow solid, yield: 96%, m.p.125–127 C, 1H NMR 120.60, 120.83, 125.11, 126.52, 126.82, 129.30, 130.45,
(CDCl3, 300 MHz) δ (ppm): 3.37 (d, 2H), 3.77 (s, 3H), 130.61, 131.16, 133.41, 133.66, 134.17, 137.46, 140.06,
3.88 (s, 3H), 5.10 (m, 2H), 5.36 (s, 2H), 5.96 (m, 1H), 6.75 141.11, 145.82, 145.91, 148.58, 149.61, 189.03. ESI-MS (m/
(d, 2H), 7.00 (d, 1H), 7.51 (m, 2H), 7.62 (m, 2H), 7.88 (m, z) 497 (M+ 1) observed for C28H24N4O5.
2H), 7.90 (m, 2H), 8.11 (m, 1H), 8.17 (m, 1H), 8.18 (s,
1H), 13C NMR (125 Hz, DMSO-d6) δ (ppm): 40.11, 55.55, (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-
55.74, 63.65, 112.68, 113.98, 114.51, 114.81, 116.07, 1,2,3-triazol-1-yl)phenyl)-3-(benzo[d][1,3]dioxol-4-yl)
119.38, 120.29, 120.43, 120.59, 120.87, 121.13, 123.07, prop-2-en-1-one (7h)
127.66, 130.15, 130.43, 130.71, 130.90, 137.74, 138.46, Yellow solid, yield: 94%, m.p. 119–121 C, 1H NMR
139.64, 145.90, 149.59, 162.27, 189.22. ESI-MS (m/z) (CDCl3, 300 MHz) δ (ppm): 3.33 (d, 2H), 3.88 (s, 3H),
482 (M+ 1) observed for C29H27N3O4. 5.10 (m, 2H), 5.36 (s, 2H), 5.98 (m, 1H), 6.04 (d, 2H),
6. 74 (m, 2H), 6.98 (d, 1H), 7.23 (m, 2H), 7.35 (m, 4H),
(E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H- 7.79 (m, 1H), 8.16 (m, 2H), 8.17 (s, 1H). 13C NMR
1,2,3-triazol-1-yl)phenyl)-3-(2,3-dichlorophenyl)prop- (125 Hz, DMSO-d6) δ (ppm): 39.83, 55.89, 63.38, 101.74,
2-en-1-one (7e) 106.72, 108.77, 112.41, 114.52, 115.80, 119.42, 120.17,
Yellow solid, yield: 88%, m.p. 149–151 C, 1H NMR 120.60, 120.85, 125.58, 129.13, 130.16, 134.14, 137.47,
(CDCl3, 300 MHz) δ (ppm): 3.35 (d, 2H), 3.90 (s, 3H), 138.33, 139.70, 145.60, 145.82, 148.53, 149.60, 150.26,
5.10 (m, 2H), 5.39 (s, 2H), 5.96 (m, 1H), 6.75 (d, 2H), 188.77. ESI-MS (m/z) 496 (M+ 1) observed. for
7.01 (d, 1H), 7.50 (m, 2H), 7.68 (m, 2H), 7.93 (m, 2H), C28H25N3O5.
8.19 (m, 2H), 8.22 (m, 1H), 8.25 (s, 1H). 13C NMR
(125 Hz, DMSO-d6) δ (ppm): 39.83, 55.89, 63.36, (E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H-
112.41, 114.49, 115.81, 120.23, 120.59, 120.83, 125.28, 1,2,3-triazol-1-yl)phenyl)-3-(naphthalen-1-yl)prop-2-en-
125.97, 127.47, 130.43, 131.92, 133.64, 134.16, 134.26, 1-one (7i)
135.41, 137.45, 137.56, 140.02, 141.37, 145.82, 145.91, Yellow solid, yield: 89%, m.p. 87–89 C, 1H NMR (CDCl3,
149.59, 188.61. ESI-MS (m/z) 520 (M+ 1) observed for 300 MHz) δ (ppm): 3.32 (d, 2H), 3.87 (s, 3H), 5.10 (m,
C28H23Cl2N3O3. 2H), 5.35 (s, 2H), 5.95 (m, 1H), 6.73 (m, 3H), 6.99
BUDUMA ET AL. 11

(m, 4H), 7.85 (m, 3H), 8.23 (m, 6H), 8.25 (s, 1H). 13C NMR v3.5 software (Accelrys), ADMET module. Mathematical
(125 Hz, DMSO-d6) δ (ppm): 39.83, 55.88, 63.35, 112.41, predictive ADMET QSAR models for different pharmaco-
114.49, 115.80, 120.10, 120.59, 120.83, 130.10, 134.15, 136.92, kinetic parameters were used, namely, aqueous solubil-
137.45, 140.04, 145.82, 145.85, 149.58, 196.54. ESI-MS (m/z) ity, BBB penetration, cytochrome P450 2D6 inhibition,
502 (M+ 1) observed for C32H27N3O3. hepatotoxicity, human intestinal absorption, and plasma
protein binding. These ADMET descriptors allow us to
(E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H- eliminate compounds with unfavorable ADMET charac-
1,2,3-triazol-1-yl)phenyl)-3-(furan-2-yl)prop-2-en- teristics early, to avoid expensive reformulation, prefera-
1-one (7j) bly before synthesis and also help to evaluate proposed
Pale yellow solid, yield: 94%, m.p. 111–113 C, IR (KBr): structural refinements that are designed to improve
3134, 3002, 2918, 2863, 1738, 1654, 1601, 1513, 1259, ADMET properties. The studied compounds were also
1229, 1137, 1021, 911, 834, 743, 668 cm1; ESI-MS (m/z) evaluated against the Lipinski's rule of five. The toxicity
442 (M+ 1) observed for C26H23N3O4. that is often used in drug development was calculated for
all designed compounds, all the predictions were calcu-
(E)-1-(4-(4-([5-allyl-2-methoxyphenoxy]methyl)-1H- lated by DS_TOPKAT module of Discovery Studio 3.5
1,2,3-triazol-1-yl)phenyl)-3-(thiophen-2-yl)prop-2-en- software (Accelrys).
1-one (7k)
Yellow solid, yield: 84%, m.p. 117–119 C, IR (KBr): 3142, ACKNOWLEDGMENTS
3077, 2999, 2936, 2834, 1652, 1585, 1511, 1416, 1256, 1224, The authors thank the director, CSIR-CIMAP, Lucknow,
1140, 1019, 819, 706 cm1; 1H NMR (CDCl3, 300 MHz) δ for his constant encouragement and support. Author
(ppm): 3.35 (d, 2H), 3.90 (s, 3H), 5.08 (m, 2H),5.37 (s, 2H) Komuraiah Buduma is thankful to CSIR, New Delhi, for
5.94 (m, 1H), 6.7 (m, 1H), 7.01 (m, 2H), 7.33 (m, 1H), 7.41 providing financial assistance in the form of a Senior
(m, 2H), 7.47 (m, 1H), 7.90 (m, 2H), 7.99 (s, 1H), 8.19 (m, Research Fellowship.
2H), 8.21 (s, 1H). 13C NMR (125 Hz, DMSO-d6) δ (ppm):
39.84, 55.89, 63.37, 112.41, 114.51, 115.80, 120.07, 120.18, DA TA AVAI LA BI LI TY S T ATE ME NT
120.60, 120.85, 128.50, 129.30, 130.10, 130.16, 132.60, 134.14, Data available in article supplementary material
137.47, 138.07, 139.79, 140.17, 145.83, 149.59, 188.26. ESI-
MS (m/z) 458 (M+ 1) observed for C26H23N3O3S.
ORCID
Kotesh Kumar J https://orcid.org/0000-0001-8294-2234

4.3 | In silico molecular docking


RE FER EN CES
[1] D. J. Newman, G. M. Cragg, J. Nat. Prod. 2012, 75, 311.
Molecular docking and visualization studies of eugenol
[2] S. Nobili, D. Lippi, E. Witort, M. Donnini, L. Bausi, E. Mini, S.
derivatives series were performed through FlexX docking Capaccioli, Pharm. Res. 2009, 59, 365.
program available in Lead IT 2.1.8 version of (Bio Solve IT [3] M. S. Butler, J. Nat. Prod. 2004, 67, 2141
Germany). FlexX is a fast algorithm for flexible docking of [4] J. Clardy, C. Walsh, Nature 2004, 432, 829837
small ligands into the active protein-binding site using an [5] S. Y. Wang, P. F. Chen, S. T. Chang, Bioresour. Technol. 2005,
incremental construction process. During docking standard 96, 813
parameters were kept in the FlexX program as implemented [6] A. M. Piggott, P. Karuso, Comb. Chem. High Throughput
Screen. 2004, 7, 607.
in LeadIT v2.1.8. Chemical structure designing, energy min-
[7] S. Lahloua, A. F. Figueiredo, P. J. C. Magalh~aes, J. H. Leal
imization, and geometry cleaning were done through Cardoso, P. D. Gloria, Life Sci. 2004, 74, 2401.
ChemDraw and Chem3D 15.0.0.16 version (PerkinElmer [8] M. E. Hidalgo, C. De la Rosa, H. Carrasco, W. Cardona, C.
Informatics). The protein 3D crystallographic structures for Gallardo, L. Espinoza, Quim. Nova 2009, 32, 1467.
docking were retrieved through RCSB Protein Data Bank [9] S. Fujisawa, T. Atsumi, Y. Kadoma, H. Sakagami, Toxicology
(PDB) (www.rcsb.org/). To find the possible bioactive con- 2002, 177, 39.
formations of eugenol derivatives series of compounds, these [10] P. Manikandan, R. M. Senthil, R. V. Priyadarsini, G.
softwares were used. Vinothini, S. Nagini, Life Sci. 2010, 86, 936.
[11] K. Chaieb, H. Hajlaoui, T. Zmanta, A. B. Kahla Nakbi, M.
Rouabhia, K. Mahdouani, A. Bakhrouf, Phytother. Res. 2007,
21, 501.
4.4 | In silico pharmacokinetic screening [12] (a) N. V. Yanishlieva, E. M. Marinova, Eur. J. Lipid Sci.
Technol. 2001, 103, 752. (b) P. Suppakul, J. Miltz, K.
Pharmacokinetic screening of eugenol derivatives with Sonneveld, S. W. Bigger, J. Agric. Food Chem. 2003, 51, 3197.
control molecules was performed by Discovery Studio [13] S. K. Jaganathan, E. Supriyanto, Molecules 2012, 17, 6290.
12 BUDUMA ET AL.

[14] H. Majeed, J. Antoniou, Z. Fang, Asian Pac. J. Cancer Prev. [33] A. Mostafa, Hussein, M. Refaat Shaker, A. Mohammed, A.
2014, 15, 9159. Meen, F. Mohammed, Arch. Pharm. Res. 2011, 34, 1239.
[15] M. J. Genin, D. A. Allwine, D. J. Anderson, M. R. Barbachyn, [34] K. Parameswaran, P. Sivaguru, A. Lalitha, Bioorg. Med. Chem.
D. E. Emmert, S. A. Garmon, D. R. Graber, K. C. Grega, J. B. Lett. 2013, 23, 3873.
Hester, D. K. Hutchinson, J. Morris, R. J. Reischer, C. W. [35] P. Thirumurugan, D. Matosiuk, K. Jozwiak, Chem. Rev. 2013,
Ford, G. E. Zurenko, J. C. Hamel, R. D. Schaadt, D. Stapert, 113, 4905.
B. H. Yagi, J. Med. Chem. 2000, 43, 953. [36] S. Brase, C. Gil, K. Knepper, V. Zimmermann, Angew. Chem.
[16] L. Savini, P. Massarelli, P. Corti, L. Chiasserini, C. Pellerano, Int. Ed. 2005, 44, 5188.
G. Bruni, I Farmaco 1994, 49, 363. [37] T. Mossman, J. Immunol. Methods 1983, 65, 55.
[17] M. Jilino, F. Malcom, G. Stevens, J. Chem. Soc. Perkin Trans. [38] I. Gulcin, A. H. Alici, M. Cesur, Chem. Pharm. Bull. 2005,
1998, 10, 1677. 53, 281.
[18] W. T. Christian, C. Caspar, M. Morten, J. Org.Chem. 2002, 67, [39] M. Oyaizu, Jpn. J. Nutr. 1986, 44, 307.
3057. [40] M. D. Gilham, R. Lehner, Methods 2005, 36, 139.
[19] Fan, W. Q.; Katritzky, A. R., Comprehensive Heterocyclic [41] S. F. Lin, C. M. Chiou, C. M. Yeh, Y. C. Tsai, Appl. Environ.
Chemistry II 1996, 4, 1–126. Microbiol. 1996, 62, 1093.
[20] M. Stefano, Chiara, V. Beatrice, M. Maurizio, B. Nicoletta, R. [42] K. S. Babu, A. K. Tiwari, P. V. Srinivas, A. Z. Ali, B. C. Raju,
Cristina, M. Carlo, G. Roberto, Bioorg. Med. Chem. 2000, 8, J. M. Rao, Bioorg. Med. Chem. Lett. 2004, 14, 3841.
2343. [43] G. J. McDougall, F. Shpiro, P. Dobson, P. Smith, A. Blake, D.
[21] K. M. Banu, A. Dinakar, C. Ananthanarayanan, Indian Stewart, J. Agric. Food Chem. 2005, 53, 2760.
J. Pharm. Sci. 1999, 61, 202. [44] M. Flex X-Rarey, B. Kramer, T. Lengauer, G. Klebe, J. Mol.
[22] S. Danoun, G. Baziard Mouysset, J. L. Stigliani, M. Payard, Biol. 1996, 261, 470.
M. Selkti, B. Viossat, A. Tomas, Heterocycl. Commun. 1998, [45] Z. Li, J. Chem. Inf. Comput. sci. 2004, 44, 1886.
4, 45. [46] M. S. Finnin, Nature 1999, 401, 188.
[23] B. Giuliana, C. Vincenzo, G. Irene, L. Oreste, M. Enrica, M. [47] H. Wei, J. Biol. Chem. 2005, 280, 37041.
Alma, N. Antonio, II Farmaco 2004, 59, 397. [48] Z. Ren, Biochem. Biophys. Res. Commun. 2008, 374, 1.
[24] J. K. Luzzi, J. H. Varghese, C. I. Mac Donald, E. E. Schmidt, C. [49] S. B. Jang, Biol. Crystallograph. 2004, 60, 829.
Kohn, L. V. Morris, E. K. Marshall, F. A. Chamber, C. A. [50] O. Tacar, P. Sriamornsak, C. R. Dass, J. Pharm. Pharmacol.
Groom, Angiogenesis 1999, 2, 373. 2013, 65, 157.
[25] S. Hakimian, A. Cheng-Hakimian, G. D. Anderson, J. W. [51] C. A. Lipinski, F. Lombardo, B. W. Dominy, P. J. Feeney, Adv.
Miller, Expert Opin. Pharmacother. 2007, 12, 1931. Drug Deliv. Rev. 2001, 46, 326.
[26] C. Mannich, W. Krosche, Archiv. Pharm. Med. Chem. 1912,
250, 647.
[27] T. F. Cummings, J. R. Shelton, J. Org. Chem. 1960, 25 SU PP O R TI N G I N F O RMA TI O N
(3), 419. Additional supporting information may be found online
[28] T. Plech, M. Wujec, A. Siwek, U. Kosikowska, A. Malm, Eur. in the Supporting Information section at the end of this
J. Med. Chem. 2011, 46, 241. article.
[29] B. Singh, D. Chetia, S. K. Puri, K. Srivastava, A. Prakash, Med.
Chem. Res. 2011, 20, 1523.
[30] N. Surendra, N. Pandeya, D. Sriram, G. Nath, E. De Clercq, How to cite this article: K. Buduma,
Eur. J. Med. Chem. 2000, 35, 249. N. Kumar A, S. Srinivas KVN, K. Kumar J,
[31] S. Joshi, N. Khosla, P. Tiwari, Bioorg. Med. Chem. 2004, S. Chinde, A. K. Domatti, Y. Kumar, P. Grover,
12, 571.
A. Tiwari, F. Khan, J Heterocyclic Chem 2021, 1.
[32] A. Y. Shaw, C.-Y. Chang, M.-Y. Hsu, P.-J. Lu, C.-N. Yang, H.-
L. Chen, C.-W. Lo, C.-W. Shiau, M.-K. Chern, Eur. J. Med.
https://doi.org/10.1002/jhet.4331
Chem., 2010, 45, 2860

You might also like