ArticlePDF Available

Synthesis and evaluation of antibacterial and antioxidant activity of novel 2-phenyl-quinoline analogs derivatized at position 4 with aromatically substituted 4 H -1,2,4-triazoles

Taylor & Francis
Journal of Enzyme Inhibition and Medicinal Chemistry
Authors:

Abstract and Figures

A set of novel quinolone–triazole conjugates (12–31) were synthesized in three steps in good yields starting from 2-phenylquinoline-4-carboxylic acid. All the intermediates, as well as the final 1,2,4-triazolyl quinolines were fully characterized by their detailed spectral analysis utilizing different techniques such as IR, 1H NMR, 13C NMR, and finally mass spectrometry. All the synthesized compounds were evaluated in vitro for their potential antibacterial activity and their preliminary safety profile was assessed through cytotoxicity assay. Additionally, six selected conjugates were evaluated for their antioxidative properties on the basis of density functional theory calculations, using radical scavenging assay (DPPH) and cellular antioxidant assay. The reported results encourage further investigation of selected compounds and are shading light on their potential pharmacological use.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=ienz20
Download by: [Zagreb University] Date: 20 June 2016, At: 00:29
Journal of Enzyme Inhibition and Medicinal Chemistry
ISSN: 1475-6366 (Print) 1475-6374 (Online) Journal homepage: http://www.tandfonline.com/loi/ienz20
Synthesis and evaluation of antibacterial and
antioxidant activity of novel 2-phenyl-quinoline
analogs derivatized at position 4 with aromatically
substituted 4H-1,2,4-triazoles
Donatella Verbanac, Ritu Malik, Mahesh Chand, Khushbu Kushwaha, Monika
Vashist, Mario Matijašić, Višnja Stepanić, Mihaela Perić, Hana Čipčić Paljetak,
Luciano Saso & Subhash C. Jain
To cite this article: Donatella Verbanac, Ritu Malik, Mahesh Chand, Khushbu Kushwaha,
Monika Vashist, Mario Matijašić, Višnja Stepanić, Mihaela Perić, Hana Čipčić Paljetak, Luciano
Saso & Subhash C. Jain (2016): Synthesis and evaluation of antibacterial and antioxidant
activity of novel 2-phenyl-quinoline analogs derivatized at position 4 with aromatically
substituted 4H-1,2,4-triazoles, Journal of Enzyme Inhibition and Medicinal Chemistry, DOI:
10.1080/14756366.2016.1190714
To link to this article: http://dx.doi.org/10.1080/14756366.2016.1190714
View supplementary material
Published online: 19 Jun 2016.
Submit your article to this journal
View related articles
View Crossmark data
www.tandfonline.com/ienz
ISSN: 1475-6366 (print), 1475-6374 (electronic)
J Enzyme Inhib Med Chem, Early Online: 1–7
!2016 Informa UK Limited, trading as Taylor & Francis Group. DOI: 10.1080/14756366.2016.1190714
RESEARCH ARTICLE
Synthesis and evaluation of antibacterial and antioxidant activity of
novel 2-phenyl-quinoline analogs derivatized at position 4 with
aromatically substituted 4H-1,2,4-triazoles
Donatella Verbanac
1
, Ritu Malik
2
, Mahesh Chand
2
, Khushbu Kushwaha
2
, Monika Vashist
2
, Mario Matijas
ˇic
´
1
,
Vis
ˇnja Stepanic
´
3
, Mihaela Peric
´
1
, Hana C
ˇipc
ˇic
´Paljetak
1
, Luciano Saso
4
, and Subhash C. Jain
2
1
University of Zagreb School of Medicine, Center for Translational and Clinical Research, Zagreb, Croatia,
2
Department of Chemistry, University of
Delhi, Delhi, India,
3
Laboratory for Epigenomics, Division of Molecular Medicine, Rud
er Bos
ˇkovic
´Institute, Zagreb, Croatia, and
4
Department of
Physiology and Pharmacology ‘Vittorio Ersparmer’, Sapienza University of Rome, Rome, Italy
Abstract
A set of novel quinolone–triazole conjugates (1231) were synthesized in three steps in good
yields starting from 2-phenylquinoline-4-carboxylic acid. All the intermediates, as well as the
final 1,2,4-triazolyl quinolines were fully characterized by their detailed spectral analysis utilizing
different techniques such as IR,
1
H NMR,
13
C NMR, and finally mass spectrometry. All the
synthesized compounds were evaluated in vitro for their potential antibacterial activity and
their preliminary safety profile was assessed through cytotoxicity assay. Additionally, six
selected conjugates were evaluated for their antioxidative properties on the basis of density
functional theory calculations, using radical scavenging assay (DPPH) and cellular antioxidant
assay. The reported results encourage further investigation of selected compounds and are
shading light on their potential pharmacological use.
Keywords
Cytotoxicity assays, DFT calculations using
DPPH and CAA, 1,2,4-triazole–quinoline
conjugates
History
Received 17 April 2016
Revised 7 May 2016
Accepted 8 May 2016
Published online 9 June 2016
Introduction
The development of resistance to currently used antibacterial
therapy has obliged the scientists, chemists, and biologists, to
further search for more effective agents with less or no side
effects. This is even more relevant to the present day scenario as
the primary and opportunistic bacterial and/or fungal infections
still continue to escalate with the increased number of immune
compromised patients (mostly due to the diseases such as AIDS,
cancer, and also as a consequence of long-term therapy after
transplants)
1
. This is the reason why extensive research is going
on around the world, to discover novel molecules to fight such
infections.
Despite plenty of research on different heterocyclic molecules,
the azole ring still remains interesting chemical fragment for the
development of novel molecules especially in the antibacterial
and antifungal therapeutic area. This is due to the fact that most of
the existing azole derivatives possessed both bacterial and fungal
static action and can be orally applied owing to favorable
bioavailability. They possess broad spectrum of activities against
most of the yeasts and filamentous fungi
2
.
Among azoles, 1H-1,2,4-triazole derivatives are considered to
be more interesting as they possess important pharmacological
activities such as antifungal
3,4
, antiviral
5
, antioxidant
6–8
, anti-
asthmatic
9
, anticonvulsant
10
, antidepressant
11
, antithyroid
12
, anti-
HIV
13
, anti-inflammatory
14
and anticancer
15–17
.
On the other hand, quinoline derivatives are known to possess
diverse pharmacological properties such as antioxidant
18,19
,
antibiotic
20,21
, cardiovascular
22
, anti-TB
23
, antiplatelet
24
, antic-
ancer
25,26
, receptor antagonists
27
, NK3 receptor antagonists-II
28
,
anti-inflammatory
29
, antimicrobial
30,31
, selective estrogen recep-
tor modulators (SERMs)
32
and protein kinase inhibitor
33
.
Keeping above in mind, we decided to synthesize conjugates of
the above-mentioned moieties to study the biological profile of the
resulting product. Genesis of our chemistry is derived from the fact
that few earlier conjugates of 1,2,4-triazole and quinoline, where
these molecules are linked via oxygen- or amide-containing linkers
in a single frame, are known to possess biological activities like
antimalarial
34
, antimicrobial
35
, anti tubercular
36
, antitumor
37,38
and antiviral
39
. However, the literature data about molecules that
contain both of these bioactive ligands directly linked/bonded to
each other in a single molecular frame are unknown.
As a result of our research programs involving the synthesis of
new bioactive molecules
40–47
, we report herein for the first time
synthesis of a new set of directly coupled quinolone–triazole
conjugates. We also assessed biological activity profiles of these
conjugates, with particular emphasis on their antioxidative
properties.
Methods
Chemistry
All chemicals were purchased from commercial suppliers (Merck,
SD Fine, and Spectrochem) and used as such. Solvents used in
extraction and purification were distilled prior to use. Products
Address for correspondence: Subhash C. Jain. E-mail: jainsc48@
hotmail.com
Downloaded by [Zagreb University] at 00:29 20 June 2016
were purified by column chromatography using silica gel as an
adsorbent. Melting points were determined on an electronic
apparatus and are uncorrected.
1
H and
13
C NMR spectra were
recorded at 400 MHz on a JEOL spectrometer (Tokyo, Japan) and
at 300 MHz on a Bruker spectrometer (Billerica, MA) using
CDCl
3
/DMSO-d
6
as solvent and tetramethylsilane (TMS) as
internal standard. TMS was used as a reference for both
1
H and
13
C NMR spectra. In
1
H NMR abbreviations s, d, dd, t, q, and m
represents singlet, doublet, double doublet, triplet, quartet, and
multiplet respectively. Coupling constants Jvalues are given in
hertz and the chemical shifts are given in . Elemental analyses
were performed on a PerkinElmer Series II, CHNS/O Analyzer
2400 (Waltham, MA). Mass spectra were recorded on JEOL-JMS-
DX303 mass spectrometer (Tokyo, Japan).
General procedure for the synthesis of compounds 3–5
Indol-2,3-dione/5-fluoro-indol-2,3-dione/5-methyl-indol-2,3-dione
(10 g) and NaOH (8.16 g) were stirred together in water (80 mL) in
a round bottom flask. To the reaction mixture, acetophenone
(8.16g) was added and contents refluxed. Reaction was monitored
on thin layer chromatography (TLC) and after its completion the
reaction mixture was cooled and acidified with conc. HCl solution.
The precipitate obtained was collected, washed, and dried to
afford 35.
2-Phenylquinoline-4-carboxylic acid (3). White solid. Yield:
14.72 g, 87%, m.p. 208–210 C. IR (KBr)
max
: 3442, 2465, 1953,
1705, 1601, 1550, 1448, 1354, 1259, 1204, 1082, 894, 781, 760,
732, 699 cm
1
.
1
H NMR (, DMSO-d
6
, 400 MHz): 11.38 (brs,
1H, –COOH), 8.79 (d, 1H, J¼8.80 Hz), 8.47 (s, 1H), 8.29–8.27
(m, 2H), 8.19 (d, 1H, J¼8.80 Hz), 7.85–7.66 (m, 2H), 7.59–7.52
(m, 3H).
13
C NMR (, DMSO-d
6
, 100 MHz): 167.50, 155.73,
148.50, 138.01, 137.03, 129.64, 129.56, 128.64, 127.28, 126.97,
125.39, 123.61, 119.28. Mass Spectral data, TOF MS ES+ m/z
(%): 250 (M
+
+1). Anal. Calcd for C
16
H
11
NO
2
: C, 77.10; H, 4.45;
N, 5.62. Found: C, 77.14; H, 4.42; N, 5.60.
General procedure for the synthesis of compounds 6–8
Prepared compound 3/4/5 (10 g) was added to abs. ethanol
(150 mL) in a flask, followed by conc. H
2
SO
4
(5 mL). The
resulting reaction mixture was refluxed and completion of
reaction was monitored by TLC. The reaction mixture was
cooled and poured over crushed ice in a beaker. The resulting
contents were rendered alkaline by adding sufficient amount of
ammonia solution. The mixture was then extracted thrice with
diethyl ether. The combined ethereal solution was dried over
anhydrous sodium sulfate and the solvent was removed by
distillation to get the desired compounds 6/7/8.
Ethyl 2-phenylquinoline-4-carboxylate (6). Yellow color oil.
Yield: 9.90 g, 89%. IR (film)
max
: 2983, 1724, 1623, 1594, 1513,
1338, 1248, 1230, 1193, 1143, 1119, 1037, 827, 749 cm
1
.
1
H
NMR (, CDCl
3
, 400 MHz): 8.73 (d, 1H, J¼8.72 Hz), 8.38 (s,
1H), 8.23–8.19 (m, 3H), 7.76 (t, 1H, J¼7.32 Hz), 7.62 (t, 1H,
J¼7.32 Hz), 7.56–7.52 (m, 3H), 4.53 (q, 2H, –COOCH
2
CH
3
),
1.50 (t, 3H, –COOCH
2
CH
3
).
13
C NMR (, CDCl
3
, 100 MHz):
166.45, 156.72, 149.22, 138.84, 136.07, 130.28, 129.85, 129.68,
128.91, 127.71, 127.46, 125.38, 123.98, 120.19, 61.90, 14.32.
Mass Spectral data, TOF MS ES+ m/z (%): 278 (M
+
+1). Anal.
Calcd for C
18
H
15
NO
2
: C, 77.96; H, 5.45; N, 5.05. Found: C,
78.02; H, 5.48; N, 5.02.
General procedure for the synthesis of compounds 9–11
A mixture of compounds 68(5 g) and hydrazine hydrate
(1.31 mL) was heated at 50–60 C temperature with constant
stirring. The solid that separated out on cooling was filtered and
crystallized to give their corresponding carbohydrazide 911.
2-Phenylquinoline-4-carbohydrazide (9). White crystalline
solid. Yield: 4.46 g, 94%, m.p. 214–216 C. IR (KBr)
max
:
3385, 2917, 2366, 1701, 1606, 1483, 1449, 1360, 1287, 1246,
1229, 1149, 1039, 932, 889, 830, 754 cm
1
.
1
H NMR (, DMSO-
d
6
, 400 MHz): 10.03 (s, 1H, –NH), 8.31 (d, 1H, J¼7.8 Hz), 8.25–
8.23 (m, 2H), 8.14 (m, 1H), 8.03 (m, 1H), 7.78–7.74 (m, 2H),
7.60–7.48 (m, 3H), 4.52 (brs, 2H, –NH
2
).
13
C NMR (, DMSO-
d
6
, 100 MHz): 166.35, 155.71, 147.92, 140.76, 138.24, 129.55,
129.47, 129.43, 128.36, 127.02, 126.69, 125.05, 123.36, 116.84.
Mass Spectral data, TOF MS ES+ m/z (%): 264 (M
+
+1). Anal.
Calcd for C
16
H
13
N
3
O: C, 72.99; H, 4.98; N, 15.96. Found: C,
73.04; H, 5.01; N, 15.97.
General procedure for the synthesis of compounds 12–26
The synthesized carbohydrazides 911 (200 mg) and substituted
benzaldehyde (92 mg) were dissolved in 10 mL of glacial acetic
acid in a round bottom flask. To the mixture, ammonium acetate
(84 mg) was added. The reaction mixture was stirred for a period
of 6–8 h at room temperature. The progress of the reaction was
monitored by TLC. After completion, the reaction mixture was
poured into ice-cold water and neutralized with ammonia. The
precipitated product was filtered, washed with water, and
crystallized from chloroform/methanol to give the desired
product.
2-Phenyl-4-[5-(4-hydroxyphenyl)-4H-[1,2,4]-triazol-3-
yl]quinoline (13). Cream-colored solid. Yield: 254 mg, 92%, m.p.
270–272 C. IR (KBr)
max
: 3257, 2925, 1654, 1606, 1585, 1545,
1513, 1365, 1267, 1235, 1166, 845, 766 cm
1
.
1
H NMR
(, DMSO-d
6
, 400 MHz): 12.08 (s, 1H, 4NH,D
2
O exchangeable),
10.06 (s, 1H, –OH,D
2
O exchangeable), 8.36–8.33 (m, 2H), 8.27–
8.25 (m, 2H), 8.20–8.16 (m, 3H), 7.85 (m, 1H), 7.67 (d, 2H,
J¼7.1 Hz), 7.57 (d, 2H, J¼7.5 Hz), 6.88 (d, 2H, J¼7.8 Hz).
13
C
NMR (, DMSO-d
6
, 100 MHz): 162.56, 159.71, 155.77, 149.14,
147.89, 145.17, 143.53, 141.46, 138.05, 130.34, 129.96, 129.60,
128.91, 127.15, 125.13, 123.47, 117.14, 115.79. Mass spectral
data, TOF MS ES+ m/z (%): 365 (M
+
+1). Anal. Calcd for
C
23
H
16
N
4
O: C, 75.81; H, 4.43; N, 15.38. Found: C, 75.94; H,
4.48; N, 15.39.
General procedure for the synthesis of compounds 27–31
The synthesized carbohydrazides 911 (200 mg) and heterocyclic
aldehyde (78 mg) were dissolved in 10 mL of glacial acetic acid in
a round bottom flask. To the mixture, ammonium acetate (84 mg)
was added. The reaction mixture was stirred for a period of 6–8 h
at room temperature. The progress of the reaction was monitored
by TLC. After completion of the reaction, the reaction mixture
was poured into ice-cold water and neutralized with ammonia.
The precipitated product was filtered, washed with water, and
crystallized from chloroform/methanol to give the desired
product.
2-Phenyl-4-[5-(furan-2-yl)-4H-[1,2,4]-triazol-3-yl]quinoline
(27). White solid. Yield: 230 mg, 88%, m.p. 198–200 C. IR
(KBr)
max
: 3182, 3052, 2925, 1655, 1623, 1591, 1544, 1349,
1284, 1270, 1158, 938, 765 cm
1
.
1
H NMR (, DMSO-d
6
,
400 MHz): 12.16 (s, 1H, 4NH,D
2
O exchangeable), 8.34–8.30
(m, 2H), 8.25–8.22 (m, 2H), 8.15 (m, 1H), 7.85–7.82 (m, 2H),
7.67 (m, 1H), 7.56–7.52 (m, 3H), 6.97 (m, 1H), 6.56 (m, 1H).
13
C NMR (, DMSO-d
6
, 100 MHz): 162.94, 155.65, 148.96,
147.98, 144.48, 140.65, 138.37, 129.77, 129.45, 128.48, 126.88,
126.77, 123.67, 123.26, 124.87, 119.21, 116.87. Mass spectral
data, TOF MS ES+ m/z (%): 339 (M
+
+1). Anal. Calcd for
C
21
H
14
N
4
O: C, 74.54; H, 4.17; N, 16.56. Found: C, 74.60; H,
4.20; N, 16.58.
2D. Verbanac et al. J Enzyme Inhib Med Chem, Early Online: 1–7
Downloaded by [Zagreb University] at 00:29 20 June 2016
In vitro biological screening
Antibacterial activity assay
Bacterial strains, Staphylococcus aureus (American Type Culture
Collection (Manassas, VA; ATCC), 29213), Streptococcus
pneumoniae (ATCC, 49619), Streptococcus pyogenes (ATCC,
700294), Haemophilus influenzae (ATCC, 49247), and
Escherichia coli (ATCC, 25922), were purchased from ATCC
and utilized to evaluate antibacterial activity of compounds.
Antibacterial activity was determined by the standard broth
microdilution method with azithromycin as comparator.
Minimum inhibitory concentrations (MICs) were established
according to guidelines of the Clinical Laboratory Standards
Institute
48
with the exception that lysed blood was substituted by
5% horse serum for Streptococcus medium. Double dilutions of
tested compounds were prepared in 128–0.5 mg/mL concentration
range within microplate wells. Bacteria were grown on appropri-
ate agar plates (Becton Dickinson, Franklin Lakes, NJ). Inocula
were prepared by direct colony suspension method and micro-
plates were inoculated with 5 104 CFU/well. Results were
determined by visual inspection after 20-h incubation at 37 C.
Cytotoxicity assays
A549 human lung adenocarcinoma cell line (ATCC, CCL-185),
HepG2 human hepatocellular carcinoma cell line (ATCC, HB-
8065), MDA-MB-231 human breast adenocarcinoma cell line
(ATCC, HTB-26), PC-3 human prostate adenocarcinoma cell line
(ATCC, CRL-1435), and THP-1 human acute monocytic leuke-
mia cell line (ATCC, TIB-202) were purchased from ATCC. Cell
lines were maintained in complete DMEM/F12 medium (Sigma-
Aldrich Chemie GmbH, Taufkirchen, Germany; D8437), or
complete RPMI1640 (Sigma, R7388) for THP-1 cells, supple-
mented with 10% Fetal Bovine Serum (Sigma, F7524) at 37 Cin
5% CO
2
atmosphere.
Cytotoxicity assay was performed using MTS Cell Titer 96
AQueous One Solution Cell Proliferation Assay (Promega
Corporation, Madison, WI; G3580)
49
. Double dilutions of tested
compounds were prepared in 100–0.2 mM concentration range
within microplate wells. 5 104 cells were added per well and
incubated overnight at 37 Cin5%CO
2
atmosphere. 15 mL of MTS
reagent was dispensed per well and plates incubated for 1–4 h at
37 Cin5%CO
2
atmosphere. The absorbance was recorded at
490 nm using Wallac Victor2 microplate reader (PerkinElmer Life
and Analytical Sciences, Turku, Finland). Results were analyzed in
GraphPad Prism software (GraphPad Software, Inc., La Jolla, CA).
DPPH-free radical scavenging assay
The DPPH (1,1-diphenyl-1,2-picryl hydrazyl) (Sigma-Aldrich
Chemie GmbH, Taufkirchen, Germany) method was used to
determine the free radical scavenging activity of compounds
50
.
Dilutions of tested compounds and ascorbic acid as a standard
antioxidant comparator were prepared in 1000–1 mg/mL con-
centration range. 1 mL of compound solution was added to
1 mL of freshly prepared DPPH solution (3.9 mg/50 mL
ethanol) and the reaction mixture incubated in the dark at
room temperature for 20 min. Absorbance (A) was measured at
517 nm using Analytik Jena UV Winaspect Specrod PC
250 spectrophotometer (Analytic Jena AG, Jena, Germany).
Inhibition of the DPPH radical by the compounds was
calculated according to the following formula:
DPPH scavenging activity %ðÞ¼A0 A1ðÞ=A0½100
Where A0 is the absorbance of the control and A1 is the
absorbance of the sample. The results are averages of three
measurements. The EC
50
value, compound concentration to reduce
50% of the DPPH, was calculated using GraphPad Prism software.
Cellular antioxidant activity (CAA) assay
OxiSelect Cellular Antioxidant Assay Kit (Cell Biolabs Inc., San
Diego, CA; STA-349) was used to assess antioxidant activity of
the compounds within a cell in a standard cell culture environ-
ment. The assay employs cell-permeable fluorogenic probe 20,70-
dichlorodihydrofluorescein diacetate (DCFH-DA), which is
diffused into cells, deacetylated by cellular esterases and oxidized
by free radicals to fluorescent 20,70-dichloro-dihydrofluorescein
(DCF), with the fluorescence intensity being proportional to the
reactive oxygen species levels within the cell cytosol
51
. The assay
was performed according to manufacturer’s instructions. In brief,
HepG2 cells were seeded at 5 104 per well in 96-well black
microplates and incubated overnight at 37 Cin5%CO
2
atmosphere. The medium was removed and wells washed with
sterile phosphate-buffered saline (PBS). Double dilutions of
compounds were prepared in 500–8 mM concentration range in
cell medium (50 mL) and DCFH-DA probe was added to wells
(50 mL). The cells were incubated 1 h at 37 Cin5%CO
2
atmosphere. After the medium was removed and cells washed
with sterile PBS, free radical initiator solution was added to all
wells (100 mL) and fluorescence read using a fluorescence
microplate reader (Victor 2 Wallac, PerkinElmer Life and
Analytical Sciences, Turku, Finland) (excitation 480 nm/emission
530 nm). The readouts were saved in increments of 15 min for a
total of 180 min. Results were analyzed in GraphPad Prism
software and quantified as IC
50
values.
Density functional theory (DFT) calculations
The reaction enthalpies BDE, IP, PA, and ETE and corresponding
free energies (Table 2 in Supplementary Material) were calculated
by a known method
52
. These reaction parameters were calculated
by applying the DFT model (U)B3LYP/6-31 + G(d, p) as
implemented in software Gaussian 03 (Gaussian, Inc.,
Wallingford, CT)
53
. Calculations were performed for the gas
and aqueous phases. Equilibrium geometries in unionized and
anionic closed-shell ground electronic states as well as of
corresponding radical and radical cation open-shell doublet
ground electronic states were fully optimized in the gas phase.
The minima were confirmed by no imaginary vibrational
frequencies at temperature of 298.15 K and pressure of 1 atm.
The free energies of solvation DG*hyd for all studied molecular
species at 1 M standard state in water were determined at the gas
phase equilibrium geometries by using integral equation formal-
ism of polarizable continuum model (IEFPCM) of solvation with
Bondi radii and tight SCF convergence criterion
54
. The free
energies (Table 2 in Supplementary Material) were calculated by
employing corresponding thermodynamic cycles
55
. Lipophilicity
values logP were calculated by OpenBabel
56
.
Results and discussion
Chemistry
The synthetic route employed for the preparation of a set of novel
quinoline-triazole conjugates (1231) is shown in Scheme 1.
2-Phenylquinoline-4-carboxylic acid (3) was converted into its
ethyl ester 6by using absolute ethanol in the presence of
concentrated sulfuric acid. The ester was purified and characterized
by its IR spectrum which showed characteristic absorption band at
1724 cm
1
for ester group. Its
1
H NMR spectrum displayed signals
at 1.50 (t, 3H) for –COOCH
2
CH
3
and at 4.53 (q, 2H) for
–COOCH
2
CH
3
. Compound 6on treatment with hydrazine hydrate
gave 2-phenylquinoline-4-carbohydrazide (9), which exhibited
DOI: 10.1080/14756366.2016.1190714 Antibacterial and antioxidant activity of novel 2-phenyl-quinoline analogs 3
Downloaded by [Zagreb University] at 00:29 20 June 2016
characteristic absor ption band at 3385 cm
1
for NHNH
2
stretching
in its IR spectrum. Further its
1
H NMR did not show any signal
corresponding to the ester group indicating thereby complete
conversion. Final confirmation of structure of compound 9came
from its mass spectrum, which showed M
+
+1 signal at m/z 264. The
hydrazide obtained was cyclocondensed with 4-hydroxybenzalde-
hyde in the presence of ammonium acetate in glacial acetic acid at
room temperature to finally give a cream-colored solid compound
characterized as 13 (Scheme 1). Its
1
H NMR spectrum showed two
characteristic D
2
O exchangeable broad signals at 12.08 and
10.06 integrating for one proton each, indicating the presence4NH
and –OHprotons, respectively. In IR spectrum, absorption band at
1654 cm
1
indicated the presence of imido bond (C¼N) which was
further supported by the absence of –CH¼N-proton in
1
H NMR
spectrum thus confirming that cyclization has taken place. The other
protons of the phenyl group and quinoline skeleton were observed at
8.36–8.33 (m, 2H), 8.27–8.25 (m, 2H), 8.20–8.16 (m, 3H), 7.85
(m, 1H), 7.67 (d, 2H, J¼7.1 Hz), 7.57 (d, 2H, J¼7.5 Hz), 6.88 (d,
2H, J¼7.8 Hz). The
13
C NMR spectrum showed all expected
characteristic signals. On the basis of above analysis, and mass
spectrum compound 13 was characterized as 2-phenyl-4-[5-(4-
hydroxyphenyl)-4H-[1,2,4]-triazol-3-yl]quinoline. Similar set
of the above-described reactions were repeated with substituted
aryl and hetero aromatic ring aldehydes to obtain corresponding
desired compounds.
Biological activity
All final products (1231) have been profiled in vitro, in terms of
their antibacterial activity and cytotoxicity. The cytotoxicity of a
compound is closely associated with potential adverse effects on
particular cells, tissues, or organs of drugs intended for human use.
The antibacterial activity was determined against five different
bacterial species: two gram-negative (E. coli,H. influenzae) and
three gram-positive (S. aureus,S. pneumoniae,S. pyogenes).
However, neither of the compounds showed substantial and
significant antibacterial activity (Supplementary Table S1).
Although molecules containing quinoline and 4H-1,2,4-triazole
fragments are often recognized in vitro as potent antibacterials
57
this has not to be general case
58
. In our case for 2-phenyl 4H-
1,2,4-triazole substituted quinolines, no antibacterial activity has
been detected. This may be either due to the specific unfavorable
structural factors such as site of substitution
57
or unfavorable
physicochemical properties
59
.
N
H
O
NN
N
COOH
COCH3COOC2H5
Ph
CONHNH2
Ph
Ph
+
RRR
R
O
ab
c
3 R = H
4 R = F
5 R = CH3
6 R = H
7 R = F
8 R = CH3
9 R = H
10 R = F
11 R = CH3
12
NPh
R
12 R = H; R1 = H; R2 = OH; R3 = OCH3
13 R = H; R1 = R3 = H; R2 = OH
14 R = H; R1 = R3 = H; R2 = F
15 R = R1 = R2 = R3 = H
16 R = F; R1 = H; R2 = OH; R3 = OCH3
17 R = F; R1 = R3 = H; R2 = OH
18 R = F; R1 = R3 = H; R2 = F
19 R = F; R1 = R2 = R3 = H
20 R = CH3; R1 = H; R2 = OH; R3 = OCH3
21 R = CH3; R1 = R3 = H; R2 = OH
22 R = CH3; R1 = R3 = H; R2 = F
23 R = CH3; R1 = R2 = R3 = H
24 R = H; R1 = H; R2 = R3 = OH
25 R = F; R1 = H; R2 = R3 = OH
26 R = CH3; R1 = H; R2 = R3 = OH
12-26
NH
N
N
R1R2
R3
dd
NPh
R
27 R = H; X = O
28 R = H; X = S
29 R = F; X = O
30 R = F; X = S
31 R = CH3; X = O
NH
N
N
X
27-31
Scheme 1. Synthesis of quinolone–triazole conjugates (12nju). Reagents and conditions: ai ¼NaOH, H
2
O, ref lux, 4–6 h; b–¼abs. C
2
H
5
OH, conc.
H
2
SO
4
, ref lux, 18–20 h; c8 ¼N
2
H
4
H
2
O, heat, 50–60 C, 4–6 h; d–¼aldehyde, glc. CH
3
COOH, CH
3
COONH
4
, 6–8 h at room temperature.
4D. Verbanac et al. J Enzyme Inhib Med Chem, Early Online: 1–7
Downloaded by [Zagreb University] at 00:29 20 June 2016
The antibacterials are generally hydrophilic molecules, while
these derivatives are very lipophilic with clogP values around 5 or
higher. Absence or very weak of antibacterial activity generally
indicates the potential of specific compounds for their long-term
use, for example as anti-inflammatory agents in the treatment of
chronic disease, without potential risks to induce resistance.
The cytotoxic effect of compounds was evaluated on five
different human cell lines (A549, HepG2, MDA-MB-231, PC-3,
and THP-1) using MTS test (Table 1). The test quantifies
metabolic activity of cells by measuring their metabolism through
the released NADH levels, thus indicating whether a compound
impairs any of the key cellular metabolic pathways.
The cytotoxicity of a compound is closely associated with its
potential adverse drug effects. In addition, MTS test is very often
used for the estimation of antiproliferative capacity of a
compound when followed through longer period of time. The
results of compound cytotoxicity screening, here performed
during 24 h, are shown in Table 1. 5-Aryl 4H-1,2,4-triazolyl
compounds 13,17,21, and 26 displayed significant cytotoxic
effect on THP-1 cell line. Derivatives 25 as well as 2730 showed
weak cytotoxic effect on THP-1 cells. Only compounds 17 and 26
were additionally cytotoxic for HepG2 cell line. With respect to
these data, all the other compounds could be considered suitable
for further in vitro profiling in cellular assays.
The cytotoxicity of the active compounds toward the most
sensitive THP-1 cell-line is determined by substituents at position
5 of the triazole ring. The observed cytotoxic activity may be
attributed to the presence of hydrogen-bond accepting centers at
specific positions in these substituents, the chemical feature that is
absent in the inactive analogs.
In addition, 5-aryl 4H-1,2,4-triazolyl derivatives which mod-
erately inhibited metabolic activity in the most sensitive cell line
(THP-1) are (poly)phenolic compounds. It is also well known that
many polyphenols possess antioxidant features since they can
directly scavenge-free radicals by donating H-atom and/or
modulate activities of various proteins included directly or
indirectly in free radical production
60
. Therefore, some of the 5-
aryl 4H-1,2,4-triazolyl derivatives have been tested for their
antioxidant activity.
At first, assuming that free hydroxyl (OH) group of the active
compounds (Table 2) can donate hydrogen atom to a free radical,
the radical scavenging activity of these derivatives was estimated
in silico by using common approach
52
. Derivatives 12,16, and 20
containing guaiacyl-like group were also included in computa-
tions. Values of the calculated parameters gas-phase bond
dissociation enthalpy (BDEg) and aqueous bond dissociation
free energy (BDFEaq) were used for comparing radical scaven-
ging capacities of our polyphenolic derivatives mutually as well
as with corresponding natural polyphenols (Table 2). As expected,
compounds 24,25, and 26 with catechol fragment have stronger
radical scavenging ability than compounds 13,17, and 21 with
para-phenyl substituent and compounds 12,16, and 20 with
guaiacyl-like moiety
52
. Compounds within each of these three
subgroups have mutually similar radical scavenging capacities.
Additionally, the order of radical scavenger capacities of the three
subgroups corresponds to the order of the natural polyphenols
quercetin, apigenin, and tamarixetin containing analogous poly-
phenolic fragments (Table 2). The obtained density functional
theory (DFT) results indicate that for our compounds, the radical
scavenging activities of (poly)phenolic fragments are quite
independent on the rest of their structures.
The DPPH assay provides an easy and rapid in vitro method
commonly used to evaluate antioxidant activity of natural plant
Table 2. Comparison of calculated gas-phase (g) bond dissociation enthalpies (BDE) and corresponding aqueous
(aq) free energies (all given in kJ/mol) as well as estimated lipophilicity values logP of the 5-aryl-4H-1,2,4-triazolyl
derivatives and natural polyphenols. Experimentally determined radical scavenging (DPPH assay) and cellular
antioxidant (CAA) activities are also presented.
Compounds BDE
g
BDFE
aq
logP IC
50
(DPPH) (mg/mL) IC
50
(CAA) (mmol/mL)
12 342.7 335.8 5.07 198.1 4500
16 (F) 343.6 334.8 5.21 77.68 4500
20 (Me) 340.6 341.0 5.38 9.35 4500
13 342.2 324.0 5.06 NA NA
17 (F) 343.2 323.7 5.20 NA NA
21 (Me) 341.6 322.9 5.37 NA NA
24
a
305.9 306.9 4.77 5.61 114.4
25 (F) 306.8 307.8 4.90 3.57 100.9
26 (Me) 305.4 306.6 5.07 6.00 405.4
Tamarixetin 346.9 332.8 2.29
Apigenin 349.3 327.7 2.58
Quercetin 310.3 294.4 1.99 31
Ascorbic acid 9.67
a
For compounds with the catechol moiety, the calculated data for more active para-OH group are listed.
Table 1. The results of compound cytotoxicity screening expressed as
IC
50
values in mM.
Compounds HepG2 THP-1 A549 MDA-MB-231 PC-3
12 4100 4100 4100 4100 4100
13 4100 37 / / /
14 4100 4100 / / /
15 4100 4100 / / /
16 4100 4100 4100 4100 4100
17 95 37 / / /
18 4100 4100 / / /
19 4100 4100 / / /
20 4100 4100 4100 4100 4100
21 4100 45 / / /
22 4100 4100 / / /
23 4100 4100 / / /
24 4100 4100 4100 4100 4100
25 4100 78 4100 4100 4100
26 55 39 4100 4100 4100
27 4100 87 / / /
28 4100 95 / / /
29 4100 76 / / /
30 4100 70 / / /
31 4100 4100 / / /
Staurosporine 3,18 0,2 / / /
DOI: 10.1080/14756366.2016.1190714 Antibacterial and antioxidant activity of novel 2-phenyl-quinoline analogs 5
Downloaded by [Zagreb University] at 00:29 20 June 2016
extracts and chemical compounds which act as free radical
scavengers. The activity of six selected compounds assessed using
the DPPH assay demonstrates considerable radical scavenging
activity (Table 2). Ascorbic acid was used as a comparator in this
assay, yielding IC
50
value of 9.67 mg/mL. Four compounds
displayed radical scavenging capabilities comparable to ascorbic
acid, with the highest DPPH-scavenging activity shown by
compound 25 (IC
50
value of 3.57 mg/mL).
There are many chemical assays used to quantify radical
scavenging activity of compounds. However, their efficacy to
predict in vivo antioxidant activity is modest since they do not
address critical issues such as uptake, distribution, and metabol-
ism of antioxidants which may significantly impact their
bioavailability, stability, and tissue retention. In addition, in
such a way, it is not possible to assess indirect antioxidant
activities of compounds attainable in the cell
60
. With respect to all
this, cellular antioxidant assay (CAA) serves as a more biologic-
ally relevant method for assessing the antioxidant activity of
compounds in vivo. CAA was employed to test antioxidant
activity of 6 selected compounds in HepG2 cell line (Table 2).
Catecholic compounds 24 and 25 displayed considerable antioxi-
dant activity in cellular system with IC
50
values of 114 mM and
101 mM, respectively. Quercetin was used as a comparator in this
assay exhibiting IC
50
value of 31 mM.
Conclusions
The novel quinolone–triazole conjugates were synthesized in
three steps starting from 2-phenylquinoline-4-carboxylic acid.
The acid was converted into carbohydrazide derivative via its
ethyl ester. Carbohyrazide on cyclocondensation using ammo-
nium acetate in acetic acid in the presence of aryl/hetero aryl
aldehyde gave finally the target molecules. These novel
compounds were evaluated in vitro for their potential antibac-
terial activity. No significant antibacterial activity has been
observed, qualifying these compounds for additional develop-
ment as promising leads in other therapeutic areas where
chronic, long-term application is required. Moreover, when
their preliminary safety profile was assessed through cytotox-
icity assays on five different cell lines, we have seen potential
for further cell-based assays profiling since only moderate
inhibition of cell-metabolism activity has been observed on the
most sensitive THP-1 cell line. And finally, based on DFT
calculations, six selected conjugates were evaluated for their
antioxidative properties on radical scavenging assay (DPPH)
and CAA. A plausible way to increase antibacterial activity
would be to decrease lipophilicity. For example, by synthetizing
derivatives without phenyl ring at position 2 (which will reduce
clogP by 2 or 3 units). However, his approach needs to be
further investigated.
Nevertheless, the obtained results in all these assays are
advocating in terms that additional synthesis of new derivatives
and further investigations in this therapeutic area might provide
interesting and potentially promising results that can finally be
applied for enriching our knowledge and experience in the
development of new chemical leads with this specific biological
activity.
Acknowledgements
M.C. and M.V. are thankful to UGC and K.K. to CSIR New Delhi (India)
for research fellowships. Additionally, V.S. is grateful to the University of
Zagreb Computing Centre SRCE for supporting the computer cluster
where computations were done.
Declaration of interest
The authors declare no conflict of interest.
This work was supported by the research grants received from
the University of Delhi, India (number DRCH/R&D/2010-13) and
from the Croatian Science Foundation, Croatia (number 5467).
References
1. Zitouni GT, Kaplancıklı ZA, Y ıldız MT, et al. Synthesis and
antimicrobial activity of 4-phenyl/cyclohexyl-5-(1-phenoxyethyl)-3-
[N-(2-thiazolyl)acetamido]thio-4H-1,2,4-triazole derivatives. Eur J
Med Chem 2005;40:607–13.
2. Aher NG, Pore VS, Mishra NN, et al. Synthesis and antifungal
activity of 1,2,3-triazole containing fluconazole analogues. Bioorg
Med Chem Lett 2009;19:759–63.
3. Tsukuda Y, Shiratori M, Watanabe H, et al. Modeling, synthesis and
biological activity of novel antifungal agents. Bioorg Med Chem
Lett 1998;8:1819–24.
4. Roberts J, Schock K, Marino S, Andriole VT. Efficacies of two new
antifungal agents, the triazole Ravuconazole and the Echinocandin
LY-303366, in an experimental model of invasive aspergillosis.
Antimicrob Agents Chemother 2000;44:3381–8.
5. Kini GD, Robins RK, Avery TL. Synthesis and antitumor activity of
ribavirin imidates. A new facile synthesis of ribavirin amidine (1-
b-D-ribofuranosyl-1,2,4-triazole-3-carboxamidine hydrochloride).
J Med Chem 1989;32:1447–9.
6. Aswathanarayanappa C, Bheemappa E, Bodke YD, et al. Synthesis
and evaluation of antioxidant properties of novel 1,2,4-triazole-
based Schiff base heterocycles. Arch Pharm 2013;346:922–30.
7. Pokuri S, Singla RK, Bhat VG, Shenoy GG. Insights on the
antioxidant potential of 1, 2, 4-triazoles: synthesis, screening &
QSAR studies. Curr Drug Metab 2014;15:389–97.
8. Maddila S, Kumar AS, Gorle S, et al. Synthesis and antioxidant
activity of 1,2,4-triazole linked thieno[2,3-d]pyrimidine derivatives.
Lett Drug Des Discov 2014;10:186–93.
9. Naito Y, Akahoshi F, Takeda S, et al. Synthesis and pharmacological
activity of triazole derivatives inhibiting Eosinophilia. J Med Chem
1996;39:3019–29.
10. Kamboj VK, Verma PK, Danda A, Ranjan S. 1,2,4-Triazole
derivatives as potential scaffold for anticonvulsant activity. Cent
Nerv Syst Agents Med Chem 2015;15:17–22.
11. Deng XQ, Song MX, Zheng Y, Quan ZS. Design, synthesis and
evaluation of the antidepressant and anticonvulsant activities of
triazole containing quinolinones. Eur J Med Chem 2014;73:217–24.
12. Takaoka M, Manabe S, Yamoto T, et al. Comparative study of
goitrogenic actions of 3-substituted 1,2,4-triazoles in rats. J Vet Med
Sci 1994;56:341–6.
13. Patel NB, Khan IH, Pannecouque C, Clercq ED. Anti-HIV,
antimycobacterial and antimicrobial studies of newly synthesized
1,2,4-triazole clubbed benzothiazoles. Med Chem Res 2013;22:
1320–9.
14. Sarigol D, Baran AU, Tel BC, et al. Novel thiazolo[3,2-b]-1,2,4-
triazoles derived from naproxen with analgesic/anti-inflammatory
properties: synthesis, biological evaluation and molecular modeling
studies. Bioorg Med Chem 2015;23:2518–28.
15. Goss PE, Strasser-Weippl K. Aromatase inhibitors for chemo-
prevention. Best Pract Res Clin Endocrinol Metab 2004;18:113–30.
16. Santen JR. Inhibition of aromatase: insights from recent studies.
Steroids 2003;68:559–67.
17. Clemons M, Coleman RE, Verma S. Aromatase inhibitors in the
adjuvant setting: bringing the gold to a standard. Cancer Treat Rev
2004;30:325–32.
18. Orhan PM, Tekiner B, Suzen S. Recent studies of antioxidant
quinoline derivatives. Mini Rev Med Chem 2013;13:365–72.
19. Savegnago L, Vieira AI, Seus N, et al. Synthesis and antioxidant
properties of novel quinolone-chalcogenium compounds. Tet Lett
2013;54:40–4.
20. Lam KH, Gambari R, Lee KKH, et al. Preparation of 8-
hydroxyquinoline derivatives as potential antibiotics against
Staphylococcus aureus. Bioorg Med Chem Lett 2014;24:367–70.
21. Bringmann G, Reichert Y, Kane V. The total synthesis of
streptonigrin and related antitumor antibiotic natural products.
Tetrahedron 2004;60:3539–74.
22. Sircar I, Haleen SJ, Burke SE, Barth H. Synthesis and biological
activity of 4-(diphenylmethyl)-a-[(4-quinolinyloxy)methyl]-1-piper-
azineethanol and related compounds. J Med Chem 1992;35:4442–9.
6D. Verbanac et al. J Enzyme Inhib Med Chem, Early Online: 1–7
Downloaded by [Zagreb University] at 00:29 20 June 2016
23. Senthilkumar P, Dinakaran M, Yogeeswari P, et al. Synthesis and
antimycobacterial activities of novel 6-nitroquinolone-3-carboxylic
acids. Eur J Med Chem 2009;44:345–58.
24. Ko TC, Hour MJ, Lien JC, et al. Synthesis of 4-alkoxy-2-
phenylquinoline derivatives as potent antiplatelet agents. Bioorg
Med Chem Lett 2001;11:279–82.
25. Mikata Y, Mika Y, Shun-ichiro O, et al. Effect of side chain location
in (2-aminoethyl)aminomethyl-2-phenylquinolines as antitumor
agents. Bioorg Med Chem Lett 1998;8:1243–8.
26. Zhang L, Sun F, Li Y, et al. Rapid synthesis of iminosugar
derivatives for cell based in situ screening: discovery of ‘‘Hit’’
compounds with anticancer activity. Chem Med Chem 2007;2:
1497–594.
27. Bromidge SM, Bertani B, Borriello M, et al. 6-[2-(4-Aryl-1-
piperazinyl)ethyl]-2H-1,4-benzoxazin-3(4H)-ones: dual-acting 5-
HT
1
receptor antagonists and serotonin reuptake inhibitors. Bioorg
Med Chem Lett 2008;18:5653–6.
28. Zhang J, Chiang FI, Wu L, et al. Surprising alteration of
antibacterial activity of 500-modified neomycin against resistant
bacteria. J Med Chem 2008;51:7563–73.
29. Elliott JM, Carling RW, Chicchi GG, et al. N0,2-Diphenylquinoline-
4-carbohydrazide based NK
3
receptor antagonists II. Bioorg Med
Chem Lett 2006;16:5752–6.
30. Cuny GD, Robin M, Ulyanova NP, et al. Structure–activity
relationship study of acridine analogs as haspin and DYRK2
kinase inhibitors. Bioorg Med Chem Lett 2010;20:3491–4.
31. Mathew V, Keshavayya J, Vaidya VP, Giles D. Studies on synthesis
and pharmacological activities of 3,6-disubstituted-1,2,4-tria-
zolo[3,4-b]-1,3,4-thiadiazoles and their dihydro analogues. Eur J
Med Chem 2007;42:823–40.
32. Rizk OH, Mahran MA, El-Khawass SM, et al. Synthesis of some
new antimicrobial thiadiazolyl and oxadiazolyl quinoline deriva-
tives. Med Chem Res 2005;14:260–73.
33. Hoekstra WJ, Patel HS, Liang X, et al. Discovery of novel quinoline-
based estrogen receptor ligands using peptide interaction profiling.
J Med Chem 2005;48:2243–7.
34. Havaldar FH, Patil AR. Syntheses of 1, 2, 4 triazole derivatives and
their biological activity. Eur J Chem 2008;5:347–54.
35. Pandey SK, Singh A, Nizamuddin A. Antimicrobial studies of some
novel quinazolinones fused with [1,2,4]-triazole, [1,2,4]-triazine and
[1,2,4,5]-tetrazine rings. Eur J Med Chem 2009;44:1188–97.
36. Upadhayaya RS, Kulkarni GM, Vasireddy NR, et al. Design,
synthesis and biological evaluation of novel triazole, urea and
thiourea derivatives of quinoline against Mycobacterium tubercu-
losis. Bioorg Med Chem 2009;17:4681–92.
37. Rashad AE, El-Sayed WA, Mohamed AM, Ali MM. Synthesis of
new quinoline derivatives as inhibitors of human tumor cells growth.
Arch Pharm (Weinheim) 2010;343:440–8.
38. Upadhayaya RS, Vandavasi JK, Kardile RA, et al. Novel quinoline
and naphthalene derivatives as potent antimycobacterial agents. Eur
J Med Chem 2010;45:1854–67.
39. Wang Z, Wu B, Kuhen KL, et al. Synthesis and biological evalu-
ations of sulfanyltriazoles as novel HIV-1 non-nucleoside reverse
transcriptase inhibitors. Bioorg Med Chem Lett 2006;16:4174–7.
40. Panda SS, Jain SC. Synthesis and QSAR studies of some novel
disubstituted 1,2,4-triazoles as antimicrobial agents. Med Chem Res
2014;23:848–61.
41. Panda SS, Jain SC. New trif luoromethyl quinolone derivatives:
synthesis and investigation of antimicrobial properties. Bioorg Med
Chem Lett 2013;23:3225–9.
42. Vashist M, Kushwaha K, Kaushik R, Jain SC. Synthesis of medicin-
ally important quinazolines decorated with 1,4-disubstituted-1,2,3-
triazoles using CuSO
4
5H
2
O-Et
3
N catalytic system. RSC Adv 2014;
4:23679–84.
43. Kushwaha K, Kaushik N, Lata Jain SC. Design and synthesis
of novel 2H-chromen-2-one derivatives bearing 1,2,3-triazole
moiety as lead antimicrobials. Bioorg Med Chem Lett 2014;27:
1795–801.
44. Kushwaha K, Sakhuja R, Jain SC. Synthesis and antimicrobial
activity of novel bis-azaphenothiazines. Med Chem Res 2013;22:
4459–67.
45. Verbanac D, Jain SC, Jain N, et al. An efficient and convenient
microwave-assisted chemical synthesis of (thio)xanthones with
additional in vitro and in silico characterization. Bioorg Med
Chem 2012;20:3180–5.
46. Panda SS, Malik R, Chand M, Jain SC. Synthesis and antimicrobial
activity of some new 4-triazolylmethoxy-2H-chromen-2-one deriva-
tives. Med Chem Res 2012;21:3750–6.
47. Sakhuja R, Panda SS, Khanna L, et al. Design and synthesis
of spiro[indole-thiazolidine]spiro[indole-pyrans] as antimicrobial
agents. Bioorg Med Chem Lett 2011;21:5465–9.
48. Clinical and Laboratory Standards Institute (CLSI) (formerly
known as National Committee on Clinical Laboratory Standards–
NCCLS) (http://www.clsi.org/) 2009;11:32–40. Available from:
http://www.cdc.gov/meningitis/lab-manual/chpt11-antimicrobial-
suscept-testing.pdf.
49. Mosmann T. Rapid colorimetric assay for cellular growth and
survival: application to proliferation and cytotoxicity assays.
J Immunol Methods 1983;65:55–63.
50. Brand-Williams W, Cuvelier ME, Berset C. Use of a free radical
method to evaluate antioxidant activity. LWT Food Sci Technol
1995;28:25–30.
51. Wolfe KL, Liu RH. Cellular antioxidant activity (CAA) assay for
assessing antioxidants, foods, and dietary supplements. J Agric Food
Chem 2007;55:8896–907.
52. Stepanic
´V, Gall TK, Luc
ˇic
´B, et al. Bond dissociation free energy as
a general parameter for flavonoid radical scavenging activity. Food
Chem 2013;141:1562–70.
53. Frisch MJ, Trucks GW, Schlegel HB, et al. Gaussian 03, Revision
C.02. Wallingford (CT): Gaussian Inc.; 2004. Available from: http://
www.gaussian.com/g_misc/g03/citation_g03.htm.
54. Cance
`s MT, Mennucci B, Tomasi J. A new integral equation
formalism for the polarizable continuum model: theoretical back-
ground and applications to isotropic and anisotropic dielectrics.
J Chem Phys 1997;107:3032–41.
55. Alongi KS, Shields GC. Theoretical calculations of acid
dissociation constants: a review. Annu Rep Comput Chem 2010;6:
113–38.
56. O’Boyle NM, Banck M, James CA. Open Babel: an open chemical
toolbox. J Cheminform 2011;3:33–46.
57. Eswaran S, Adhikari AV, Shetty NS. Synthesis and antimicrobial
activities of novel quinoline derivatives carrying 1,2,4-triazole
moiety. Eur J Med Chem 2009;44:4637–47.
58. Patel RV, Park SW. Access to a new class of biologically active quin-
oline based 1,2,4-triazoles. Eur J Med Chem 2014;71:24–30.
59. O’Shea R, Moser HE. Physicochemical properties of antibacterial
compounds: implications for drug discovery. J Med Chem 2008;51:
2878–1.
60. Lo
´pez-Alarco
´n C, Denicola A. Evaluating the antioxidant capacity
of natural products: a review on chemical and cellular-based assays.
Anal Chim Acta 2013;763:1–10.
Supplementary material available online
DOI: 10.1080/14756366.2016.1190714 Antibacterial and antioxidant activity of novel 2-phenyl-quinoline analogs 7
Downloaded by [Zagreb University] at 00:29 20 June 2016
... Compound II was reported to exhibit promising anti-oxidant and anti-inflammatory activities. The IC 50 value for free radical scavenging activity was 7.75 μg/mL compared to ascorbic acid as reference standard with IC 50 value of 18.65 μg/mL. Additionally, compound II exerted significant antiinflammatory activity via inhibition of proteinase activity with IC 50 value of 123.4 μg/mL compared to Diclofenac sodium as reference standard with IC 50 value of 0.88 μg/mL [49] ( Fig. 1 ). ...
... Compound III was found to display remarkable DPPH-scavenging activity with an IC 50 value of 3.57 μg/mL compared to ascorbic acid as reference standard (IC 50 value of 9.67 μg/mL). Moreover, compound III exhibited considerable anti-oxidant activity in cellular system (IC 50 value of 101 μg/mL) compared to Quercetin as reference standard (IC 50 value of 31 μM) [50] ( Fig. 1 ). ...
... COX 50 , nM) is the mean of two determinations acquired using an ovine COX-1/COX-2 assay Kits (Cayman Chemicals Inc., Ann Arbor, MI, USA) and the deviation from the mean is < 10% of the mean value. ...
Article
A novel series of 19 quinoline/1,2,4-triazole hybrid 6a-i and 7a-j was synthesized and evaluated in vitro as dual COX-2/5-LOX inhibitors. Compounds 6e, 6i, and 7e displayed the highest potency and selectivity for inhibiting COX-2 activity (IC50 = 7.25, 8.13, and 8.48 nM, respectively; selectivity index (COX-1/COX-2) = 44.89, 30.30, and 33.47, respectively) in comparison to celecoxib (COX-2 IC50 = 42.60 nM; selectivity index (SI) = 8.05). The anti-inflammatory activity of the newly synthesized compounds was further examined in vivo using a carrageenan induced paw edema assay. Interestingly, the in vitro findings of the COX inhibitory assay were consistent with the in vivo assay. Moreover, 6e, 6i, and 7e showed a substantial reduction in serum concentrations of PGE2, TNF-α, IL-6. Molecular docking analysis of compounds 6e, 6f, 6i, 7e, and 7f revealed high binding affinities toward COX-2 compared to COX-1, which was matched with the experimental results. In addition, these compounds exhibited different binding orientations into the active site of COX-2, which were dependent on the type of substitutions on N4 of the triazole ring. Among the tested derivatives, compounds 6e, 6i and 7e which showed high selectivity to COX-2, exhibited hydrogen bonding interactions with key amino acids in COX-2 such as Arg120, Arg513, and/or Glu524. In addition, the tested compounds also showed multiple hydrogen bonds with the Arg101, Val110, Arg138 or His130 in 5-LOX. These findings show, taken together, that those derivatives are good leads to potential anti-inflammatory agents with lowest gastric damage.
... antimicrobial, analgesic, anticonvulsant, antimalarial, antiviral, anticancer, and antioxidant activities. [2][3][4][5][6][7][8][9] Though many triazole-based derivatives are available as medicines, considerable attention has been received by the chemistry of triazoles and their fused heterocyclic derivatives because of their synthetic and effective biological importance. [10][11][12][13][14] Besides, a number of 1,2,4-triazole derivatives are commercialized as important fungicides, herbicides, and plant growth regulators. ...
... After incubating the mixtures at 37 ± 0.1 °C for 30 min, the absorbance was evaluated with a spectrophotometer (540 nm). The DPPH radical scavenging rate of each sample was calculated as follows (Verbanac et al., 2016;Zeleke et al., 2020). ...
Article
Full-text available
Cancer is one of the most common pathologies in the world, leading to a reduced standard of living and even death for centuries. Despite promising developments in treatment methods in recent years, the expected level of treatment and success hasn’t yet been achieved due to the side effects and cost of treatment methods and the fact that some drugs are still in the trial phase. This situation has encouraged the scientific community to search for natural agents with lower costs and limited side effects. Abies cilicica, also known as fir, and Aloe vera has been used in both food and traditional medicine from the past to the present. In the literature review, it was found that both A. vera and A. cilicica have many beneficial effects, especially anti-inflammatory, antifungal, and wound-healing properties. This study aimed to investigate the antimicrobial, antioxidant, cytotoxic, and apoptotic/necrotic effects of extracts of A. vera from Asphodelaceae and A. cilicica (Ant. Et Kotschy.) subsp. cilicica Carr. from Pinaceae. The best antimicrobial activity was observed against Escerichia coli with a zone diameter of 20.00 ± 3.59 mm and Klebsiella pneumoniae with a zone diameter of 21 ± 5.35 mm. 2KA showed the best effect on antioxidant activity. 2MA + 2KSA showed significant cytotoxic activity on MDA-MB-231 cancer cells. IC50 values of 1EA + 1KA extract (whole A. cilicica and A. vera dissolved in ethanol) against MDA-MB-231 cell line (IC50 458.29 ± 19.01 µg/ml) and MCF-7 cell line (IC50 596.03 ± 5.56 µg/ml) were determined. According to the data obtained from the study, A. vera and A. cilicica were found to have antimicrobial, antioxidant, and cytotoxic effects both alone and synergistically. It is predicted that they can be used especially in cancer treatment.
... 2-Phenylquinoline, an alkaloid, has potential antioxidant activity owing to its ability to inhibit detoxification enzyme depletion and as well as increase antioxidant levels [59][60][61]. Meanwhile, 2,9-dimethyl-1, 10-phenanthroline has been described as a neuronal cell protector in oxidative stress, as it selectively chelates oxidative stress inducers such as copper ion [62]. ...
Article
Full-text available
Hepatotoxicity caused by an overdose of acetaminophen (APAP) is the leading reason for acute drug-related liver failure. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a protein that helps to regulate redox homeostasis and coordinate stress responses via binding to the Kelch-like ECH-associated protein 1 (Keap1). Targeting the Keap1-Nrf2 interaction has recently emerged as a potential strategy to alleviate liver injury caused by APAP. Here, we designed and synthesized a number of iridium(III) and rhodium(III) complexes bearing ligands with reported activity against oxidative stress, which is associated with Nrf2 transcriptional activation. The iridium(III) complex 1 bearing a bioactive ligand 2,9-dimethyl-1,10-phenanthroline and 4-chloro-2-phenylquinoline, a derivative of the bioactive ligand 2-phenylquinoline, was identified as a direct small-molecule inhibitor of the Keap1–Nrf2 protein-protein interaction. 1 could stabilize Keap1 protein, upregulate HO-1 and NQO1, and promote Nrf2 nuclear translocation in normal liver cells. Moreover, 1 reversed APAP-induced liver damage by disrupting Keap1–Nrf2 interaction and without inducing organ damage and immunotoxicity in mice. Our study demonstrates the identification of a selective and efficacious antagonist of Keap1–Nrf2 interaction possessed good cellular permeability in cellulo and ideal pharmacokinetic parameters in vivo, and, more importantly, validates the feasibility of conjugating metal complexes with bioactive ligands to generate metal-based drug leads as non-toxic Keap1–Nrf2 interaction inhibitors for treating APAP-induced acute liver injury.
... These oxidative injuries are associated with radiation [18], food components, or pollution or which are produced endogenously by metabolic reactions in the human body [19] accountable for oxidative damage to proteins, DNA, and lipids [20]. Many experiments showed that antioxidants could be potent in inhibiting or overcoming such disorders [21]. Hence, the synthesized imidazoquinolines-based free radical scavenging activity occurs either from phenolic -OH or -SH groups of the quinoline moiety. ...
Article
Full-text available
Quinoline and imidazole derivatives have been playing a significant role in functional bioactivities and were potentially used as antibacterial, antifungal, anticancer, and anti-inflammatory drugs. Owing to the limitation of drug resistance, herein we synthesized thio-, chloro-, and hydroxyl-functionalized various imidazoquinolines by molecular hybridization approach. All the imidazoquinoline derivatives were examined for their antibacterial activity against selected bacterial pathogens by the agar well diffusion method. In addition, the anti-oxidant efficacy of imidazoquinolines was also tested using ferric reducing antioxidant power (FRAP). Among them, electron-withdrawing (-Cl) substituent containing imidazoquinoline 5f showed higher antibacterial and anti-oxidant activities than other imidazoquinolines and reached the effectiveness of the standard. In addition, compounds 4f, 5e, and 3f showed moderate antibacterial activity and other derivatives displayed weak activity against various pathogens. Molecular docking studies were also performed on selected imidazoquinoline derivatives (3f, 4f, and 5f), which showed high docking score and strong binding energy values. These results revealed that thio-imidazoquinoline could assist as a prototype for the designing of multidrug-resistant antibiotics against various microbial organisms.
Article
Novel antioxidants based on polymerized 2,2,4-trimethyl-1,2-dihydroquinoline (TMQ) are synthesized. TMQ is chemically modified to obtain new derivatives, namely; Ester, Hydarzide, Oxadiazole and Triazole. The synthesized polymers are characterized by Fourier transforms infrared spectroscopy (FTIR) and proton-nuclear magnetic resonance (1HNMR). The formed polymers are evaluated as antioxidants for styrene-butadiene rubber (SBR) composites. TMQ and the prepared polymers are added to SBR by a concentration of 1 phr and their antioxidant activity is compared. The rheometric characteristics and the mechanical properties including tensile strength, elongation at break, modulus at 100% elongation and hardness are studied. The results show that the prepared polymer vulcanizates are characterized by high mechanical properties compared to the commercial TMQ. The tensile strength equals 14.92 MPa, 18.3 MPa, 20.43 MPa, 21.59 MPa, 19.5 MPa and 18.92 MPa for blank, TMQ, Ester, Hydrazide, Oxadiazole and triazole vulcanizates, respectively. The prepared vulcanizates were subjected to thermo-oxidative aging for two, four, six and seven days. Mechanical properties are measured after aging and their retained values are estimated. The results show that the prepared compounds have high antioxidant activity compared to commercial TMQ. The retained values of tensile strength after aging for seven days are 50.27%, 71.97%, 80.90%, 86.66%, 80.15%, 72.99%, for blank, TMQ, Ester, Hydrazide, Oxadiazole and triazole vulcanizates, respectively. The hydrazide polymer exhibits the highest antioxidant efficiency among the prepared polymers. It has retained tensile strength of 86.66% after aging for seven days. It is also found that the rubber vulcanizates containing the synthesized polymers have the lowest swelling values and consequently higher crosslink density leading to high mechanical properties and high antioxidant efficiency.
Article
Full-text available
The present work focuses on the synthesis of novel heterocycles 2-(aryloxy)-3-(4,5-diaryl-1H-imidazol-2-yl)quinolines (6k-v) by an effective condensation reaction. These molecules exhibited fluorescent properties and hence for the proper understanding of their optical behavior and quantum yields, solvatochromic studies have been carried out. Further, frontier molecular orbitals, molecular electrostatic potential (MEP), and geometrical structure optimization have been investigated using the B3LYP/6-311G ++ (d, p) method. The energy gap between the HOMO, LUMO of the optical and energy band gap is determined by DFT and UV–visible spectra for TD-DFT studies are done. The screening of these compounds for in vitro COX-1 and COX-2 inhibition and DPPH free radical scavenging ability assays produced promising results. The binding interactions of these molecules against the COX-2 enzyme (PDB: 5IKR) were validated by docking studies. Graphical Abstract
Article
Aim: A series of new hybrid molecules with two iodine atoms on the sides were synthesized. Methods: A one-pot, two-component method with trifluoroacetic acid as an effective catalyst to obtain dihydro-pyrrol-2-one compounds was developed. Short reaction times, a cheap catalyst, high yields and clean work-up are benefits of this method. Results: The chemical structures of the newly synthesized compounds were verified through spectroscopic techniques. Their antimicrobial activity against S. aureus, P. aeruginosa and C. albicans was tested in vitro. Conclusion: NC- and OH- radicals confer broad-spectrum antimicrobial activity, including against Gram-positive and Gram-negative bacteria and yeasts. Compounds 3g >7 and >9 were most active on the two bacterial species, while 3l >9 and >3i were most active against the fungal strain.
Article
Full-text available
The search for new anticonvulsant agent with more selectivity and lower toxicity continues to be an area of rigorous investigation in medicinal chemistry. Epilepsy is a chronic disease whose treatment consists of controlling seizures with antiepileptics that very often associated with side-effects which in rare circumstances can be potentially life-threatening. Triazolam and Alprazolam are established drugs used in epilepsy which have triazole moiety. The potency and broad spectrum of the pharmacological response of triazole moiety as anticonvulsant agent have attracted the attention of many researchers to explore this framework for its potential. The literature shows that different substitution on triazole ring exhibit potent antiepileptic activity with no or lesser neurotoxicity. It is a sincere attempt to compile the reported potent triazole derivatives with significant anticonvulsant action.
Article
Full-text available
The aligned manuscript reports synthesis, screening and QSAR analysis of twenty six 1, 2, 4-triazole analogues from their respective aromatic carboxylic acids. The structures of synthesized analogues were characterized using physical and spectral analysis. 1,2,4-Triazole analogs antioxidant capacity was determined using DPPH radical scavenging assay. Results revealed that out of L,T & VRT series, VRT series of 1,2,4-triazoles have significant antioxidant activities when compared with standard ascorbic acid. To obtain structural insights for development of new antioxidants a 2D-QSAR analysis of this dataset of 26 molecules was performed. The 2D-QSAR models correlate with the in vitro results and explain the salient structural features predominant in the molecules responsible for antioxidant activity.
Article
Full-text available
Disubstituted 1,2,4-triazoles 3a–k, 4a–k, and 6a–k have been synthesized from anthranilic acid and nicotinic acid, respectively, through a multi-step reaction sequence via their hydrazides. Synthesized compounds were evaluated for their in vitro antimicrobial activity against two gram-positive bacteria (S. aureus and B. subtilis), three gram-negative bacteria (E. coli, S. typhi, and K. pneumonia) as well as four fungi (A. niger, A. fumigatus, A. flavus, and C. albicans). To explore computational approach, structure–activity relationships were generated statistically using the synthesized compounds and their respective quantitative values of biological activities. These models can be used in future for predicting antimicrobial activity on similar class of compounds.
Article
3-Substituted-1,2,4-triazole-5-thiones are versatile synthetic intermediates for the preparation of several biologically active N-bridged heterocyclic compounds, given that they have two reactive sites, thiocarbonyl and an amine nitrogen (N1/N4). For several years, our interest has focused on the synthesis of novel heterocyclic systems derived from 3-substituted-1,2,4-triazole-5-thiones having analgesic/anti-inflammatory activity. In this study, a series of novel thiazolo[3,2-b]-1,2,4-triazole-6(5H)-one derivatives bearing naproxen was synthesized and evaluated for their in vivo analgesic and anti-inflammatory properties in acute experimental pain and inflammation models. The compounds were also tested for their ulcerogenic potential. Our findings showed that all the newly synthesized derivatives attenuate nociception and inflammation compared with a control. All the synthesized compounds exhibited much lower ulcerogenic risk than the standard drugs indomethacin and naproxen. Some compounds with significant analgesic and/or anti-inflammatory activities as well as low ulcer scores were further evaluated for in vitro COX-1 and COX-2 inhibitory potential in a COX-catalyzed prostaglandin biosynthesis assay. Among the tested compounds, compound 1q showed the highest selectivity index (SI) of 4.87. The binding mode for some of the tested compounds to the cyclooxygenase (COX) enzymes was predicted using docking studies.
Article
The direct use of Cu (II) sulfate pentahydrate in presence of triethylamine has resulted into 1,4-disubstituted-1,2,3-triazoles via 1,3-dipolar cycloaddition of terminal alkyne(s) to azide(s) at room temperature. The study of additive effect of triethylamine in presence of Cu(II) sulfate pentahydrate revealed that it is essential for activation of copper catalyst and is responsible for the reaction between aliphatic/aromatic heterocyclic alkyne (s) and azide (s) which otherwise did not work under standard reaction conditions often used in click chemistry.