Anda di halaman 1dari 11

Chem Biol Drug Des 2014; 84: 409419

Research Article

Synthesis and Biologic Evaluation of Substituted


5-methyl-2-phenyl-1H-pyrazol-3(2H)-one Derivatives as
Selective COX-2 Inhibitors: Molecular Docking Study
Pritam N. Dube1,*, Shweta S. Bule2, Santosh N. ators (24). Both enzymes are sensitive to inhibition by
Mokale1, Manoj R. Kumbhare2, Pravin R. Dighe2 conventional non-steroidal anti-inflammatory drugs (NSA-
and Yogesh V. Ushir2 IDs). Observations that COX-1 was involved in several
homeostatic processes, while COX-2 expression was asso-
1 ciated with inflammation and other pathologies, such as
Department of Pharmaceutical Chemistry, Y. B. Chavan
College of Pharmacy, Dr. Rafiq Zakaria Campus, cancer proliferation, have led to the development of COX-2
Aurangabad-431001, Maharashtra India selective inhibitors to improve the therapeutic potency and
2
Department of Pharmaceutical Chemistry, S.M.B.T. to reduce the classical side-effects associated with the use
College of Pharmacy, Dhamangaon, Nashik-422403, of conventional NSAIDs (57).
Maharashtra India
*Corresponding author: Pritam N. Dube,
pritamdube@gmail.com Classical NSAIDs such as flurbiprofen (Figure 1A) non-
selectively inhibit both isoenzymes and cause gastric failure
The present study reported the synthesis and biologic like bleeding and ulcer. To prevent or decrease these side-
evaluation of new pyrazolone derivatives for COX-2 effects, a current strategy consists of designing selective
inhibitory activities and investigated in vivo for their COX-2 inhibitors with an improved gastric safety profile.
anti-inflammatory activities using carrageenan-induced The improved safety profile of COX-2 inhibitors may allow
rat paw edema model as well as in vitro using HRBC the use of these new agents for long-term prophylactic use
membrane stabilization and protein denaturation in certain chronic diseases. This has led intense efforts in
method. Eight derivatives showed pronounced COX-2 search for potent and selective COX-2 inhibitors, which
inhibition, and 5a, 5d, and 5f exhibited the highest could provide anti-inflammatory drugs with fewer risks.
COX-2 inhibition. The derivatives were further evalu- Several classes of compounds having selective COX-2
ated for antioxidant activity wherein 5a and 5b showed inhibitory activity have been reported in the literature such
potent free radical-scavenging activity against DPPH,
as SC-558 and celecoxib (Figure 1B and C, respectively).
nitric oxide, and hydrogen peroxide radicals. Molecular
docking study revealed the binding orientations of py-
It is interesting to note that most of the COX-2 inhibitors
razolone derivatives into the active sites of COX-2 and
bear a common structural feature, namely a central
thereby helps to design the potent inhibitors.
heterocyclic five member ring (pyrazole in celecoxib/Celeb-
rex, 3,4-dihydrofuran-2-one in rofecoxib/Vioxx, or
Key words: anti-inflammatory, antioxidants, molecular dock-
1,2-oxazole in valdecoxib/Bextra) and that several devel-
ing, pyrazolone
opments were based on that fact using, for example, tet-
Received 3 December 2013, revised 14 February 2014 and rahydrofuran (5) or 1H-pyrrole (6); moreover, all these
accepted for publication 12 March 2014 described compounds are vicinal substituted. From these,
the pyrazole ring is considered a privileged structure and
an attractive scaffold for drug discovery.

Cyclooxygenase is the key enzyme in the biosynthesis of A number of molecules based upon a monocyclic hetero-
prostanoids, biologically active substances that are involved cyclic template have been investigated for their COX-2
in several physiologic processes but also in pathologic con- inhibitory activity (810). However, the recent success of
ditions, such as inflammation (1). It is actually well known pyrazole COX-2 inhibitors has highlighted the importance
that this enzyme exists under two forms: cyclooxygenase-1 of these heterocyclics in medicinal chemistry (8). Thus, our
(COX-1) and cyclooxygenase-2 (COX-2). COX-1 is a con- main objective is to design novel heterocyclic compounds
stitutive enzyme and is responsible for the production of as specific inhibitors of COX-2 in the hope that these mol-
cytoprotective prostaglandins in the gastrointestinal tract ecules may be further explored as powerful and novel
(GI) and proaggregatory thromboxane in blood platelets. non-ulcerogenic anti-inflammatory lead candidates. Thus,
However, COX-2 is an inducible enzyme which is induced our main objective is to design novel pyrazolones as spe-
in response to the release of several proinflammatory medi- cific inhibitors of COX-2 in the hope that these molecules

2014 John Wiley & Sons A/S. doi: 10.1111/cbdd.12324 409


Dube et al.

Figure 1: Representative
examples of non-selective (A) and
selective COX-2 inhibitors (B and C)
and the designed pyrazolone
derivatives (3, 5a5g).

may be further explored as powerful anti-inflammatory lead Research-Lab Fine Chem Industries, Mumbai. Solvents
candidates. In view of the rationale and in aiming to finding and Chemicals were purified wherever required.
new structure leads with potential anti-inflammatory activi-
ties and selective COX-2 inhibition (11,12), new series of
5-methyl-2-phenyl-1H-pyrazol-3(2H)-one derivatives (3, Step-I: Synthesis of 5-methyl-2-phenyl-1,2-
5a5g) (Figure 1) have been synthesized and evaluated dihydro-3H-pyrazol-3-one (3)
the in vitro COX-1/COX-2 inhibition and in vivo and in vitro Redistilled ethyl acetoacetate 50 g (49 mL, 0.384 mol) and
assessment as anti-inflammatory activities. 40 g (36.5 mL, 0.37 mol) of phenylhydrazine in a 250-mL
RBF were mixed together and refluxed for 3 h. The heavy
Docking studies of this class of molecules have shown that reddish syrup was allowed to cool for sometime, and then,
they accommodate entirely in the binding site of COX-2 in about 100 mL of ether was added, and the mixture was stir-
positions very close to that of SC-558 in the crystal structure red vigorously. The syrup which was insoluble in ether got
of its complex with COX-2. So, we docked the most active solidified within 15 min. the solid was filtered at the pump
COX-2 inhibitors into COX-2 (1CX2) active site to predict and washed thoroughly with ether to remove colored impu-
their binding modes, their binding affinities, and orientation rities and crystallized from ethanol to give the corresponding
of these compounds at the active site of the COX-2 enzyme. 5-methyl-2-phenyl-1,2-dihydro-3H-pyrazol-3-one (13,14).

Colorless crystals; MP 126127 C; % yield: 80%; UV k


Materials and Methods max: 302.

Chemistry FT-IR (KBr) max/cm: 3132 (C-H aromatic); 2925 (C-H sat-
The melting points were determined by open capillary urated); 1728 (C=O lactam); 1620 (C=N); 1597, 1473 (C=C
method and were uncorrected. UV spectra were recorded aromatic); 1410, 1518 (N=O nitro group); 1340 (C-N);
on Thermo scientific spectrum 2600 spectrophotometer. 1026 (C-O); 1455, 1378 (C-H); 1357 (N-N); 1H NMR
The IR spectra were recorded on a Jasco FT-IR spectro- (400 MHz, DMSO-d6) d ppm: 7.76.9 (m, 5H, Ar-H), 5.1
photometer, model-4100 using KBr disk method. 1H and (s, 1H, Ar-CH), 2.3 (s, 3H, CH3), 1.9 (s, 1H, NH); 13C
13
C spectra were recorded on a Bruker AVANCE NMR (400 MHz, DMSO-d6) d ppm: 172.8 (C=O of Pyrazo-
400 MHz spectrometer at 25 C using tetramethylsilane as lone), 159.5 (C-N of pyrazolone), 139.6 (C-N of Ar),
an internal standard and DMSO-d6 as the solvent. The 129.3120.4 (5C, Ar), 91.7 (C4 of pyrazolone), 14.6 (CH3
chemical shifts are expressed in d ppm. Mass spectra of pyrazolone). MS: m/z = 175.0 [M+H].
were recorded on a Varian Inc, 410 Prostar Binary LC with
500 MS IT PDA Detectors. Purity of the compounds was
checked on TLC plates using silica gel G as stationary Step-II: General method of synthesis of 5-methyl-
phase and iodine vapors as visualizing agent. For TLC 2-phenyl-1,2-dihydro-3H-pyrazol-3-one derivative
plate development, the benzenemethanol system at vari- (5a5g)
ous ratios was used as eluent. The% yield was reported To a solution of compound 5-methyl-2-phenyl-1,2-dihy-
after the recrystallization. All chemicals used were of dro-3H-pyrazol-3-one (0.01 mol) in ethanol (30 mL),

410 Chem Biol Drug Des 2014; 84: 409419


Synthesis and Evaluation of COX-2 Inhibitors

formaldehyde (0.02 mol) and corresponding substituents (C-O); 1047 (C=S); 1H NMR (400 MHz, DMSO-d6) d ppm:
like urea, p-chlorophenyl urea, thiourea, N-phenyl thiourea, 8.06.9 (m, 10H, Ar-H), 4.2 (s, 1H, NH), 4.1, 3.5 (d, 2H,
guanidine, sulfonamide, semicarbazide were added. The CH2), 2.9 (s, 1H, CH), 2.2 (s, 1H, NH), 1.9 (s, 3H, CH3);
13
reaction mixture was refluxed for 610 h. The solvent was C NMR (400 MHz, DMSO-d6) d ppm: 181.9 (C=S),
distilled off, and the residue was poured into ice water/ 177.4 (C=O), 155.6 (C=N), 137.2121.7 (Ar-C), 20.1 (C5 of
purified water. The precipitated solid was filtered off, dried, pyrazolone). MS: m/z = 339.12 [M+H].
and recrystallized from ethanol.
1-((3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-4-
1-[(3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-4- yl)methyl)guanidine (5e). White powder; MP
yl)methyl]urea (5a). White powder; MP 235238 C; % 185187 C; % yield: 79.18%; UV k max: 311.
yield: 61.22%; UV k max: 313.
FT-IR (KBr) max/cm: 3395 (C=NH); 3062 (C-H aromatic);
FT-IR (KBr) max/cm: 3351 (C-H aromatic); 2969 (C-H sat- 2973 (C-H saturated); 1712 (C=O lactam); 1625 (C=N);
urated); 1630 (C=O lactam); 1620 (C=N); 1547 (C=C aro- 1591, 1477 (C=C aromatic); 1458, 1372 (C-H); 1324 (N-
matic); 1495, 1398 (C-H); 1370 (C-N); 1359 (N-N); 1037 N); 1330 (C-N); 1027 (C-O); 1H NMR (400 MHz, DMSO-
(C-O); 1H NMR (400 MHz, DMSO-d6) d ppm: 8.07.1 (m, d6) d ppm: 8.7 (s, 2H, NH2), 7.97.1 (m, 5H, Ar-H), 6.1 (s,
5H, Ar-H), 6.16.0 (s, 3H, NH), 3.7, 3.4 (m, 2H, CH2), 3.1 1H, NH), 3.7, 3.3 (d, 2H, CH2), 3.0 (s, 1H, CH), 2.0 (s, 1H,
(s, 1H, CH), 1.4 (s, 3H, CH3), 13C NMR (400 MHz, DMSO- NH), 1.6 (s, 3H, CH3); 13C NMR (400 MHz, DMSO-d6) d
d6) d ppm: 173.8 (C=O of pyrazolone), 163.2 (C=O of ppm: 174.4 (C=O), 162.7 (C=N), 155.6 (C-N of pyrazo-
urea), 156.1 (C-N), 132.6120.4 (Ar-C), 49.3 (C4 of lone), 134.9120.2 (Ar-C), 19.3 (C5 of pyrazolone). MS: m/
pyrazolone), 21.5 (CH3). MS: m/z = 246.20 [M] +. z = 245.1 [M] +.

1-(4-chlorophenyl)-3-[(3-methyl-5-oxo-1-phenyl-4,5- 4-{[(3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-
dihydro-1H-pyrazol-4-yl) methyl]urea (5b). White 4-yl)methyl]amino} benzene sulfonamide (5f). White
powder; MP 8082 C; % yield: 65.30%; UV k max: 328. powder; MP 220222 C; % yield: 63.60%; UV k max:
390.
FT-IR (KBr) max/cm: 3328 (C-H aromatic); 2976 (C-H sat-
urated); 1704 (C=O lactam); 1633 (C=N); 1594, 1492 (C=C FT-IR (KBr) max/cm: 2973 (C-H aromatic); 2900 (C-H sat-
aromatic); 1455, 1378 (C-H); 1400 (N=O nitro group); urated); 1702 (C=O lactam); 1595 (C=N); 1501, 1489 (C=C
1357 (N-N), 1340 (C-N); 1026 (C-O); 754 (C-Cl); 1H NMR aromatic); 1020 (C-O); 1452, 1377 (C-H); 1321 (sulfon-
(400 MHz, DMSO-d6) d ppm: 7.97.1 (m, 9H, Ar-H), 6.2, amide); 1321 (N-N); 1H NMR (400 MHz, DMSO-d6) d
6.1 (s, 2H, NH), 3.8, 3.4 (d, 2H, CH2), 3.1 (s, 1H, CH), 1.9 ppm: 8.17.0 (m, 9H, Ar-H), 4.4 (s, 1H, NH), 3.8, 3.2 (s,
(s, 3H, CH3); 13C NMR (400 MHz, DMSO-d6) d ppm: 2H, CH2), 2.8 (s, 1H, CH), 2.1 (s, 2H, NH2), 1.9 (s, 3H,
172.4 (C=O of pyrazolone), 168.1 (C=O of urea), 154.3 CH3); 13C NMR (400 MHz, DMSO-d6) d ppm: 178.1
(C-N), 138.3120.2 (Ar-C), 46.9 (C4 of pyrazolone), 19.3 (C=O), 156.3 (C-N of pyrazolone), 151.4 (Ar-N), 132.7
(-CH3). MS: m/z = 357.1 [M+H]. 116.2 (Ar-C), 22.5 (C5 of pyrazolone). MS: m/z = 359.12
[M+H].
1-[(3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-4-
yl)methyl]thiourea (5c). White powder; MP 98100 C; N-[(3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-
% yield: 69.46%; UV k max: 357. 4yl)methyl] hydrazine carboxamide (5g). Brown pow-
der; MP 188189 C; % yield: 59.74%; UV k max: 393.
FT-IR (KBr) max/cm: 3300 (C-H aromatic); 2969 (C-H satu-
rated); 1711 (C=O lactam); 1703 (C=N); 1594, 1499 (C=C FT-IR (KBr) max/cm: 3050 (C-H aromatic); 1668 (C=O lac-
aromatic); 1406 (N=O nitro group); 1455, 1378 (C-H); 1368 tam); 1630 (C=N); 1592, 1499 (C=C aromatic); 1448, 1373
(N-N), 1336 (C-N); 1026 (C-O); 1H NMR (400 MHz, DMSO- (C-H); 1354 (N-N); 1340 (C-N); 1021 (C-O); 1H NMR
d6) d ppm: 8.6 (s, 2H, NH2), 7.87.2 (m, 5H, Ar-H), 3.7, 3.1 (400 MHz, DMSO-d6) d ppm: 7.87.0 (m, 5H, Ar-H), 6.1,
(d, 2H, CH2), 2.9 (s, 1H, CH), 2.7 (s, 1H, NH), 1.8 (s, 3H, 6.0 (s, 2H, NH), 3.9, 3.4 (d, 2H, CH2), 3.1 (s, 1H, CH), 2.2
CH3); 13C NMR (400 MHz, DMSO-d6) d ppm: 182.1 (C=S), (s, 1H, NH), 1.8 (s, 3H, CH3); 13C NMR (400 MHz, DMSO-
171.4 (C=O of pyrazolone), 132.6120.9 (Ar-C), 42.1 (C5 of d6) d ppm: 173.8 (C=O of pyrazolone), 160.2 (C=O), 156.3
pyrazolone), 21.7 (CH3). MS: m/z = 263.10 [M+H]. (C-N of pyrazolone), 137.4121.7 (Ar-C), 20.5 (C5 of
pyrazolone). MS: m/z = 262.2 [M+H].
1-[(3-methyl-5-oxo-1-phenyl-4,5-dihydro-1H-pyrazol-4-
yl)methyl]-3-phenylthiourea (5d). White powder; MP
150152 C; % yield: 85.58%; UV k max: 335. Biologic screening

FT-IR (KBr) max/cm: 3190 (C-H aromatic); 2925 (C-H sat- In vitro cyclooxygenase (COX) inhibition assay
urated); 1759 (C=O lactam); 1611 (C=N); 1611, 1527 (C=C The ability of the test compounds listed in Table 2 to
aromatic); 1453, 1385(C-H); 1369 (N-N); 1341 (C-N); 1062 inhibit ovine COX-1 and COX-2 (IC50 values, lM) was

Chem Biol Drug Des 2014; 84: 409419 411


Dube et al.

determined using an enzyme immunoassay (EIA) kit (cat- activity (%) = (1 D/C) 100, where D represents the
alog number 560101, Cayman Chemical, Ann Arbor, MI, difference in paw volume before and after compounds
USA). Cyclooxygenase catalyzes the first step in the bio- were administered to the rats, and C stands for the differ-
synthesis of arachidonic acid (AA) to PGH2. PGF2a, pro- ence of volume in the control groups.
duced from PGH2 by reduction with stannous chloride, is
measured by enzyme immunoassay (ACETM competitive
EIA). Stock solutions of test compounds were dissolved In vitro anti-inflammatory activity
in a minimum volume of DMSO. Briefly, to a series of HRBC membrane stabilization. The test sample con-
supplied reaction buffer solutions (960 lL, 0.1 M TrisHCl sisted of stock erythrocyte (RBC) suspension 0.030 mL
pH 8.0 containing 5 mM EDTA and 2 mM phenol) with mixed with 5 mL of hypotonic solution (154 mM NaCl in
either COX-1 or COX-2 (10 lL) enzyme in the presence 10 mM sodium phosphate buffer at pH 7.4) containing
of heme (10 lL) was added 10 lL of various concentra- synthesized compound ranging from concentration 5,
tions of test drug solutions (0.01, 0.1, 1, 10, 50, and 10, 15, and 20 lg/mL (16). The control sample con-
100 lM in a final volume of 1 mL). These solutions were sisted of 0.030 mL RBC suspension mixed with hypo-
incubated for a period of 5 min at 37 C, after which tonic buffered solution alone. The standard drug Analgin
10 lL of AA (100 lM) solution was added and the COX was treated similar to test at 5, 10, 15, and 20 lg/mL
reaction was stopped by the addition of 50 lL of 1M concentrations. The experiment was carried out in tripli-
HCl after 2 min. PGF2a produced from PGH2 by reduc- cate. The mixtures were incubated at 10 min at room
tion with stannous chloride was measured by enzyme temperature, centrifuged for 10 min at 1077 9 g, and
immunoassay. This assay was based on the competition absorbance of the supernatant was measured spectro-
between PGs and a PG-acetylcholinesterase conjugate photometrically at 540 nm. The percentage inhibition of
(PG tracer) for a limited amount of PG antiserum. The hemolysis or membrane stabilization was calculated by
amount of PG tracer that is able to bind to the PG anti- following equation:
serum is inversely proportional to the concentration of
PGs in the wells because the concentration of PG tracer %Inhibition of hemolysis 100  A1  A2 =A1 ;
is held constant while the concentration of PGs varies.
This antibodyPG complex binds to a mouse anti-rabbit where A1 = absorbance of hypotonic buffered solution
monoclonal antibody that had been previously attached alone, and A2 = absorbance of test/standard sample in
to the well. The plate was washed to remove any hypotonic solution.
unbound reagents, and then, Ellmans reagent, which Effect on protein denaturation. Test solution consist-
contains the substrate to acetylcholine esterase, was ing of 1 mL of different concentrations of synthesized
added to the well. The product of this enzymatic reaction compounds ranging from 5, 10, 15, and 20 lg/mL or
produces a distinct yellow color that absorbs at 406 nm. standard Analgin 5, 10, 15, and 20 lg/mL was mixed with
The intensity of this color, determined spectrophotometri- 1 mL of egg albumin solution (1 mM) and incubated at
cally, is proportional to the amount of PG tracer bound 27  1 C for 15 min (17,18). Denaturation was induced
to the well, which is inversely proportional to the amount by keeping the reaction mixture at 70 C in a water bath
of PGs present in the well during the incubation: Absor- for 10 min. After cooling, the turbidity was measured
bance a [Bound PG Tracer] a1/PGs. Percent inhibition spectrophotometrically at 660 nm. Percentage inhibition of
was calculated by the comparison of compounds treated denaturation was calculated from control where no drug
to various control incubations. The concentration of the was added. Each experiment was carried out in triplicate,
test compounds causing 50% inhibition (IC50, lM) was and the average was taken.
calculated from the concentrationinhibition response
curve (duplicate determinations).
Antioxidant activity
Scavenging activity against 1,1-diphenyl-2-picryl
In vivo anti-inflammatory activity hydrazyl radical (DPPH). First, the antiradical activity
The test compounds were evaluated using in vivo rat car- was measured as the scavenging activity of the stable
rageenan-induced foot paw edema model reported previ- 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radical. In its
ously (15). Male Sprague Dawley rats (250 g) were fasted radical form, DPPH has an absorption band at 516 nm
with free access to water at least 16 h prior to experi- which disappears upon reduction by an antiradical com-
ments. Edema was produced by injecting 0.2 mL of a pound (19,20). The reaction mixture contained in 3 mL of
solution of 1% k-carrageenan in the hind paw. Paw vol- ethanol, 80 lM DPPH and test compounds at different
ume was measured by water displacement with a plethys- concentrations (10, 15, 20, 25 lg/mL). By the same pro-
mometer (UGO BASILE) before, 1, and 2 h after cedure, standard was also prepared. After 30 min at room
treatment. The compounds were administered intraperito- temperature, the absorbance was recorded at 516 nm on
neally with a 1 mL suspension of test compounds in vehi- an UV spectrophotometer. All experiments were carried
cle (0.5% methyl cellulose). The percentage was out in triplicate. The percentage of remaining DPPH (DPPH
calculated by the following equation: anti-inflammatory REM) was calculated as follows:

412 Chem Biol Drug Des 2014; 84: 409419


Synthesis and Evaluation of COX-2 Inhibitors

  using Polak-Ribier conjugate gradient and Truncated New-


Control  Sample
% DPPH  100: ton conjugate gradient algorithms. The convergence
Control
threshold used was RMS gradient of 0.01. Conformational
For all derivatives, IC50, that is, the concentration of test models of the ligands were generated.
compound needed to reduce DPPH absorption by 50% at
516 nm was determined.
Docking
Assay of nitric oxide (NO)-scavenging activity. The Docking of the ligands was carried out using extra preci-
absorbance of the chromophore formed during diazotiza- sion (XP) method called grid-based ligand docking with
tion of nitrite with sulfanilamide and on subsequent cou- energetics (GLIDE) for flexible ligand docking. The ligands
pling with N-naphthylene diamine was read at 546 nm were prepared for docking using LIGPREP from Schro-
(21,22). dingers molecular modeling softwares. The co-ordinate
for COX-2 enzyme (PDB ID 1CX2) was taken from RCSB
The different concentrations (10,15, 20, 25 lg/mL) of Protein Data Bank and prepared for docking using protein
tested compound were taken in volume of 200 lL. Nitric preparation wizard in Maestro v9.3.
oxide production was initiated with the addition of 200 lL
of phosphate buffer (0.1 M, pH 7.4) and 800 lL sodium ni- The receptor grid generation for docking was prepared
troprusside (10 ml), and the reaction mixture was incu- using both the centroid of selected active site residues
bated at 25 C for 150 min. A control tube was also and blind docking. The results of both the methods were
proceeding in the same way except for test compound similar. The different conformations of the compounds
ethanol was used. After incubation, 1.2 mL of Griess were docked flexibly, and 1000 poses per compound
reagent was added to the test-tubes, and they were kept were obtained. The poses, complexes, and the binding
at RT for 30 min for color development. By the same pro- affinities between the receptor and ligands were analyzed
cedure, standard was also prepared. The optical density using Schrodingers modeling software (24). All the molec-
was read at 540 nm. All experiments were carried out in ular modeling studies were performed on Schrodinger
triplicate. Maestro, and their binding affinities with the receptor were
analyzed.
A0  A1   100
% Inhibition of NO ;
A0
Result and Discussion
where A0 is the absorbance before reaction, and A1 is the
absorbance after reaction has taken place. The IC50 value Chemistry
was derived from% Inhibition at different concentrations.
Assay of hydrogen peroxide-scavenging activity. The Synthesis of compounds (3)
H2O2-scavenging ability of the synthesized compounds The preparation of target 5-methyl-2-phenyl-1H-pyrazol-3
5ag was determined spectrophotometrically according to (2H)-one (3) is shown in Scheme 1. The cyclization of ethyl
the method of Ruch et al (23). Briefly, a solution of hydrogen acetoacetate with phenylhydrazine occurred in the first step.
peroxide (2 mM) was prepared in 0.17 M phosphate buffer
(pH 7.4). Various concentrations of the samples (in ethanol)
were added to the reaction mixture containing 2 mM hydro- Synthesis of compounds 5a5g
gen peroxide. After 10 min incubation at room temperature, The addition of amine derivatives to compound 3 afforded
the absorbance was read against a blank at 230 nm. the corresponding 5-methyl-4-(methylamino)-2-phenyl-1H-
pyrazol-3(2H)-one derivatives (5a5g) as the major product
H2O2-scavenging activity (%) = [(A control A sample)/A (Table 1). The amino alkylation of an acidic proton was
blank] 9 100, where A control is the absorbance of the placed next to a carbonyl functional group by formalde-
control reaction (containing all reagents except the test hyde and amines. The reaction is known as Mannich reac-
compound), and A sample is the absorbance of the test tion. The structures of the isolated products 3, 5a5g
compound. were established on the basis of their elemental and spec-
tral analyses.

Molecular modeling
The compounds were modeled using BUILD application of Biologic activity
Maestro 9.3. The geometry was optimized by molecular
mechanics using IMPACT in a dynamic environment using In vitro COX inhibition study
standard TIP4P water model. The energy minimization was It was found that the structurally simple compounds, py-
done using optimized potentials for liquid simulations 2005 razolone derivatives 3 and 5a5g (Figure 1), were obtained
(OPLS 2005) force field. Energy minimization was done as a potent and slightly COX-2-selective inhibitor. Accord-

Chem Biol Drug Des 2014; 84: 409419 413


Dube et al.

Step -1
CH3

H 2N NH
NH O O O N
a

+ H 3C OC 2 H5

1 2
3
Step-2

HN CH 3
CH3 R
O NH
O NH N
N b
+ R NH2
Scheme 1: synthesis of designed
compounds. Reaction conditions:
4 5a-5g (A) reflux for 3 h, (B) reflux for
3 610 h.

ing to the above rationale, the compounds synthesized in the COX-inhibiting activity of the tested compounds. The
this work were evaluated for their ability to inhibit COX-1 phenylurea analog (5d and 5f) showed the most potent
and COX-2 using an ovine COX-1/COX-2 assay kit (Cata- activity, being 1.25 and 2 times more active than celecoxib
log No. 560101, Cayman Chemicals Inc.). IC50 (lM) was in COX-1-and COX-2-inhibiting activity, respectively.
determined and means of two determinations acquired,
and the deviation from the mean is <10% of the mean
value. The selectivity index (SI values) was defined as IC50 In vivo anti-inflammatory studies
(COX-1)/IC50 (COX-2). The known COX inhibitor celecoxib All the synthesized compounds were tested for anti-inflam-
was used as positive control, and the IC50 values of celec- matory activities. The results of anti-inflammatory activities
oxib on COX-1 and COX-2 were determined to be >100 against carrageenan-induced rat paw edema and ED50
and 0.40 lM, respectively, indicating that celecoxib is a (mg/kg) of all tested compounds are shown in Table 3. Of
selective COX-2 inhibitor [COX-2 (SI) > 250.0]. The results the eight compounds, three compounds (5a, 5d, and 5f)
showed that most of the compounds showed potent inhi- were found to possess potent anti-inflammatory activity
bition against COX-2 (IC50  0.12.5 lM) compared to the (8292% reduction in inflammation) and some of which
inhibition for COX-1 (IC50 > 100 lM) as listed in Table 2. have shown higher inhibition in comparison with reference
Nearly 8 of the compounds (5a, 5d, and 5f) were found to drug diclofenac. In general, pyrazolones which were
be potent and selectively similar to celecoxib against COX- substituted with chloro moiety at 3-phenyl ring (5b) and
2. Compound 5d was the most potent inhibitor in this ser- unsubstituted urea (5c, 5g, and 5e) have shown lower
ies with the activity (IC50 = 0.2 lM) twofold higher than cel- percentage of inhibition of edema when compared with
ecoxib (IC50 = 0.4 lM). The effects of substituents compounds containing urea-phenyl group at the same
introduced into the pyrazolone moiety of compound 3 position (5d and 5f). These findings may be attributed to
were revealed to be directional, being dependent on the the importance of sulfonyl group and thiourea derivative in
electronic nature of the substituents, that was introduction COX receptor binding site.
of a phenylthiourea group at the 3-position (5d) enhanced
both COX-1- and COX-2-inhibiting activity with higher
potency for the latter, resulting in a COX-2-selective inhibi- In vitro Anti-inflammatory activity
tor (SI = 400). Moreover, introduction of halogen such as It was found that the structurally simple compound, py-
chloro (5b) resulted in lowering the COX-2-inhibiting activ- razolone derivatives, was obtained as a potent and slightly
ity, while introduction of an electron-donating sulfonyl COX-2-selective inhibitor. According to the above ratio-
group (5f) showed just the opposite effects, resulting in a nale, the compounds synthesized in this work are evalu-
COX-2-selective inhibitor (SI= >100). The activities of syn- ated for their ability to inhibit COX-2 using hypotonic
thesized analogs (5a5g) are shown in Table 2. Among solution hemolysis and protein denaturation methods
them, compounds 5a and 5f are a COX-2-selective inhibi- (Table 4).
tor (SI = >40.0). No clear-cut structureactivity relation-
ships could be deduced at this stage. Interestingly, benzyl Initially, the compounds were tested at 30 lM and their
substituents on the N-pyrazolone ring play critical roles in effects on COX-2 inhibition. The study was further extended

414 Chem Biol Drug Des 2014; 84: 409419


Synthesis and Evaluation of COX-2 Inhibitors

Table 1: Physical characterization of synthesized compounds


CH3 HN CH3
R
O NH NH
N O N

3 5a-5g

Compound code R Molecular formula Molecular weight Rf valuea

3 C10H10N2O 174.19 0.50


5a O C12H14N4O2 246.10 0.68
H 2N

5b Cl C18H17ClN4O2 356.80 0.31


O

N
H

5c S C12H14N4OS 262.33 0.55


H2N

5d S C18H18N4OS 338.43 0.67

N
H

5e NH C12H15N5O 245.28 0.42


H2 N

5f O C17H18N4O3S 358.41 0.63


S NH2
O

5g O C12H15N5O2 261.28 0.72


H 2N
N
H

a
eluent used for TLC plates Benzene/Methanol (4:1).

Table 2: In vitro COX-1/COX-2 enzyme inhibition assay of the Table 3: In vivo anti-inflammatory activity against carrageenan-
designed compounds induced rat paw edema and ED50 (mg/kg) of the designed
compounds
IC50 (lM)a
% Inhibition of edemaa
Compound no COX-1 COX-2 SIb
Compound no After 1 h After 2 h ED50b(mg/kg)
3 >100 35.00 >2.86
5a >100 2.30 >43.48 3 69 77 137
5b >100 >100 5a 76 82 118
5c >100 85.00 >1.17 5b 51 75 185
5d 80.00 0.20 400.0 5c 33 59 193
5e >100 50.70 >1.97 5d 83 91 102
5f >100 1.00 >100.0 5e 48 69 162
5g >100 >100 5f 88 92 109
Celecoxib >100 0.40 >250.0 5g 27 38
Diclofenac 77 83 121
a
IC50 value is the compound concentration required to produce
50% inhibition of COX-1 or COX-2 for means of two determina- a
The carrageenan-induced rat paw edema assay was carried out
tions using an ovine COX-1/COX-2 assay kit (catalog no. 560101, using six animals (male Sprague Dawley rats)/group following IP of
Cayman Chemicals Inc.), and deviation from the mean is <10% of the test compound. The results are expressed as means  SEM
the mean value. (n = 46) following a 100 mg/kg IP of the test compounds.
b
Selectivity index = (COX-1 IC50/COX-2 IC50). b
ED50 was the effective dose calculated after 2 h.

Chem Biol Drug Des 2014; 84: 409419 415


Dube et al.

Table 4: In vitro anti-inflammatory activity evaluated by hypotonic highly relevant to the design of selective COX-2 inhibitors.
solution hemolysis and protein denaturation methods This COX-2 2-pocket arises due to a conformational
change at Tyr355, that is, attributed to the presence of
Compound % Protein
code Hemolysis IC50 denaturation IC50
Ile523 in COX-1 relative to Val523 having a smaller side
chain in COX-2. The level of COX-2 inhibition and anti-
3 61.43 6.76 78.00 1.75 inflammatory activities of synthesized compounds prompted
5a 60.02 36.36 77.04 4.74 us to perform molecular docking studies to understand the
5b 55.64 8.31 72.53 18.49 ligandprotein interactions in detail. The pdb entry (1CX2)
5c 56.59 15.98 32.78 71.95
was selected as a dataset to compare the binding of syn-
5d 66.27 0.61 45.62 40.17
thesized molecules with that of pyrazole derivative celecoxib
5e 63.95 22.50 36.34 53.68
5f 61.43 3.15 46.44 38.51 which is a well-established COX-2 antagonist. Docking of
5g 59.78 12.39 82.92 15.41 compounds was done with COX-2, and the results not only
Celecoxib 61.57 8.5 85.65 17.90 validated our docking protocol but also established the fol-
lowing findings.
IC50 values are reported as an average of three determinations.
Access of ligands to the pocket of COX-2 is controlled by
Leu352, Gln192, and Arg513. Interaction of Leu352 with the
to examine the concentrationactivity responses at different bound drug is a requirement for time-dependent inhibition of
concentrations to determine the IC50 values for COX-2. COX-2. With few exceptions, all the ligands showed similar
Four compounds (3, 5a, 5d, and 5f) showed better COX-2 orientation in the COX-2 active site, and the complex formed
inhibitory activity than celecoxib with IC50 values in the was stabilized by formation of classical and non-classical
range of 0.618.5 lM. Compounds 3 and 5d (3-phenylthio- hydrogen bonds (Figure 2). The compound 5d was found to
urea pyrazolone derivative) exhibited the highest COX-2 dock into the active site of COX-2 with interaction energy of
inhibition (IC50 0.61 lM) in protein denaturation model fol- 12.94 kcal/mol. It formed hydrogen bonds with Ser530
lowed by 5d (IC50 0.61 lM) and 5f (IC50 3.15 lM) in percent-
age hemolysis model (Table 4). The COX-2 inhibitory effect
Table 5: In vitro antioxidant activity of pyrazolone derivatives
of 5d was >10-fold as compared to celecoxib (IC50 8.5 lM).
Introduction of either urea or guanidine results in decrease % Inhibition  SDa
in COX-2 inhibitory activity. The substitution of pyrazolone Sr. Compound
with distal aryl ring results in reasonable COX-2 inhibition no. code DPPH NO H2O2
profile. The increased activity was also observed in thiourea
1 3 62.01  0.11 40.41  0.70 64.13  1.56
derivative than urea and guanidine. 2 5a 27.77  0.56 13.46  0.58 18.56  0.56
3 5b 29.14  0.56 11.54  1.56 22.13  0.06
4 5c 39.43  0.56 18.55  2.56 23.67  1.58
In vitro Antioxidant activity 5 5d 60.67  2.54 30.28  0.51 62.43  0.66
It has been reported that in inflammatory disorders, exces- 6 5e 53.76  0.54 29.97  0.22 59.67  2.54
sive free radical generation takes place and several NSA- 7 5f 55.24  2.56 23.97  0.22 25.77  0.76
8 5g 51.67  1.56 14.23  0.66 32.54  0.06
IDs and pyrazolone compounds with anti-inflammatory
9 Standard 22.13  0.06 18.56  0.56 15.65  0.33
activity are reported to act as radical scavengers (25).
Therefore, antioxidant property of the synthesized pyrazol- a
Average of three determinations.
ones was evaluated using DPPH, nitric oxide, and hydro-
gen peroxide radical-scavenging in vitro assays (26,27).
The compounds were tested at different concentrations,
Table 6: IC50 value of synthesized compounds and standard
and it was observed that all compounds have shown ascorbic acid
antioxidant property (Tables 5 and 6). The free OH group
on substituted distal aryl ring is responsible for antioxidant IC 50  SDa
activity as it is proved by testing compounds 5a, 5b, and Sr. Compound
5c that showed good activity. no. code DPPH NO H2O2

1 3 14.75  0.51 22.11  1.20 20.45  2.14


2 5a 14.01  0.14 15.50  1.34 15.64  2.41
Molecular docking analysis 3 5b 17.00  0.41 20.15  0.11 22.50  0.15
The docking study may offer more insight into the under- 4 5c 26.72  0.32 25.13  1.10 23.70  1.36
standing of the proteinligand interactions and the structural 5 5d 25.75  0.01 36.36  0.32 38.00  0.45
features of the active site. The compounds were modeled 6 5e 36.34  1.54 37.00  0.18 25.70  0.10
7 5f 21.95  1.07 40.00  0.23 32.34  0.72
using BUILD application of Schrodinger Maestro 9.3, and
8 5g 21.91  1.09 38.00  0.02 25.77  1.37
the binding affinities of the synthesized compounds with the 9 Standard 6.50  0.06 8.90  1.44 7.25  2.87
receptor were analyzed. The COX-2 binding site possesses
a
an additional 2-pocket, that is, absent in COX-1, which is Average of three determinations.

416 Chem Biol Drug Des 2014; 84: 409419


Synthesis and Evaluation of COX-2 Inhibitors

A
A

C C

Figure 2: 2D ligand interaction diagrams of (A) celecoxib, (B)


compound 5d, and (C) compound 5f. Figure 3: Overlap of binding pose of celecoxib (purple color) with
(A) compound 3 (red), (B) compound 5a (green), and (C)
compound 5f (orange). Hydrogen bonds are shown by yellow
(Figure 2b) into the active site of COX-2 and bonding dis- dotted line.
tance between NH of 5d and OH of Ser530 2.58  A (HO)
and pp stacking interaction between urea-substituted phe-
nyl ring with Trp387 and Tyr385 was observed. Similarly, 5d could dock successfully into the COX-2 active site as well.
compound 5f forms two hydrogen bonds with Gln192 and Our docking study showed that compounds 5b, 5d, and 5f
Ser353 as similar to that of celecoxib. were oriented so that their 4-substituted pyrazolone fragment
and phenyl moiety fill this adjunct pocket.
The urea-substituted phenyl ring of 5d was surrounded by
the active site amino acid residues Tyr385, Gly526, Phe381, The complex generated by docking studies of 3, 5a, and 5f
Leu384, Met522, and Trp387 (Figure 2B). The compound with COX-2 and superimposition with the structure of the

Chem Biol Drug Des 2014; 84: 409419 417


Dube et al.

selective inhibitor, celecoxib, co-crystallized with COX-2, derivatives as selective COX-2 inhibitors: synthesis, bio-
illustrated in Figure 3, shows that compound 5f can bind in logical evaluation, and docking studies. Eur J Med
the active site of this enzyme in approximately similar fashion Chem;57:149161.
as the pyrazolic prototype (celecoxib). Comparison of the 4. Cryer B., Feldman M. (1992) Effects of nonsteroidal
interactions performed by SC-558 in the crystal and the anti-inflammatory drugs on endogenous gastrointestinal
docked structure of 5f with COX-2 (Figure 3C) shows that prostaglandins and therapeutic strategies for preven-
the sulfonamide groups of 5f hydrogen bonded to the amino tion and treatment of nonsteroidal anti-inflammatory
acid residue Ser353, similarly to the sulfonyl moiety of the drug-induced damage. Arch Intern Med;152:1145
pharmacophoric sulfonamide group pertaining to SC-558 1155.
and celecoxib. 5. Dannhardt G., Kiefer W. (2001) Cyclooxygenase inhibi-
tors-current status and future prospects. Eur J Med
Chem;36:109126.
Conclusion 6. Smith W.L., Langenbach R. (2001) Why there are two
cyclooxygenase isozymes. J Clin Invest;107:14911495.
In conclusion, eight pyrazolone derivatives were synthesized 7. Dinarello C.A. (2010) Anti-inflammatory agents: present
and showed more pronounced COX-2 inhibition than natu- and future. Cell;140:935950.
rally occurring chromone, stellatin. The SAR study revealed 8. Zhong B., Cai X., Chennamaneni S., Yi X., Liu L., Pink
that a pyrazolone skeleton with C3 methyl group, C=O at 5- J.J., Dowlati A., Xu Y., Zhou A., Su B. (2012) From
position and urea substitution on 4-position is an essential COX-2 inhibitor nimesulide to potent anti-cancer agent:
feature for COX inhibition. Four compounds (3, 5a, 5d, and synthesis, in vitro, in vivo and pharmacokinetic evalua-
5f) exhibited greater anti-inflammatory activity than stan- tion. Eur J Med Chem;47:432444.
dard, and two compounds (5a and 5b) showed the potent 9. Chennamaneni S., Zhong B., Lama R., Su B. (2012)
radical-scavenging activity as an antioxidant agent. Molecu- COX inhibitors Indomethacin and Sulindac derivatives
lar docking study further helped in supporting the observed as antiproliferative agents: synthesis, biological evalua-
COX-2 selectivity. The findings of the study inferred that the tion, and mechanism investigation. Eur J Med
dual functioning of 5a as a COX inhibitor and antioxidant Chem;56:1729.
agent renders it as a lead molecule for further development 10. Singh P., Mittal A., Satwiderjeet K., Holzer W., Kumar
of new anti-inflammatory agents. S. (2008) 2, 3-Diaryl-5-ethyl sulfanyl methyl tetra-
hydrofurans as a new class of COX-2 inhibitors and
cytotoxic agents. Org Biomol Chem;6:27062712.
Acknowledgments 11. Biava M., Porretta G.C., Poce G., Battilocchio C., Ma-
netti F., Botta M., Forli S., Sautebin L., Rossi A., Pergola
The authors are thankful to Head of the Department, Phar- C., Ghelardini C., Galeotti N. (2010) Novel ester and acid
maceutical Chemistry; A.S. Dhake, Principal, S.M.B.T Col- derivatives of the 1,5-diarylpyrrole scaffold as anti-
lege of Pharmacy, Nashik, for providing necessary facilities inflammatory and analgesic agents. Synthesis and in vi-
to carry out this research work. This work was supported tro and in vivo biological evaluation. J Med Chem;53:
by the Department of Science and Technology of India 723733.
[Project File No. SR/FT/LS-132/2012 and Sanction Diary 12. Kurumbail R.G., Stevens A.M., Gierse J.K., McDonald
No. SERB/F/4302/2012-13]. J.J., Stegeman R.A., Pak J.P., Gildehaus D., Miyashiro
J.M., Penning T.D., Seibert K., Isakson P.C., Stallings
W.C. (1996) Structural basis for selective inhibition of
Conflict of Interest cyclooxygenase-2 by anti-inflammatory agents.
Nature;384:644648.
No. 13. El-Hawash S.A.M., Badawey E.S.A.M., El-Ashmawey
I.M. (2006) Nonsteroidal antiinflammatory agentspart
2 antiinflammatory, analgesic and antipyretic activity
References of some substituted 3-pyrazolin-5-ones and
1,2,4,5,6,7-3H-hexahydroindazol-3-ones. Eur J Med
1. Williams D.A., Lemke T.L. (2002) Non-Steroidal Anti- Chem;41:155165.
Inflammatory Drugs, Foyes Principles of Medicinal 14. Furniss B.S., Hannaford A.J. (2007) Textbook of
Chemistry, 5th edn. Baltimore, MD: Lippincott Williams Practical Organic Chemistry, 5th edn. Harlow: Long-
& Wilkins; p. 751793. man Pearson education; p. 11501154.
2. Frank M.M., Fries L.F. (1991) The role of complement 15. Kasahara Y., Hikino H., Tsurufuji S., Watanabe M.,
in inflammation and phagocytosis. Immunol Ohuhi K. (1985) Antiinflammatory actions of ephedrines
Today;12:322326. in acute inflammations. Planta Med;51:325331.

3. Basile L., Alvarez S., Blanco A., Santagati A., Granata 16. Ushir Y.V., Patel K.N. (2011) Microscope Evaluation
G., Di Pietro P., Guccione S., Mun ~oz-Ferna 
ndez M.A. sanctum and Ocimum basillicum Seeds. Inventi Rapid:
(2012) Sulfonilamidothiopyrimidone and thiopyrimidone Planta Activa;3:1926.

418 Chem Biol Drug Des 2014; 84: 409419


Synthesis and Evaluation of COX-2 Inhibitors

17. Padmanabhan P., Jangle S.N. (2012) Evaluation of and [15N]nitrate in biological fluids. Anal Biol
in-vitro anti-inflammatory activity of herbal preparation, Chem;2:131138.
a combination of four herbal plants. Int J Basic App 23. Ruch R.J., Cheng S.J., Klaunig J.E. (1989) Preven-
Med Sci;2:109116. tion of cytotoxicity and inhibition of intercellular
18. Lovly C.M., Heuckmann J.M., de Stanchina E., Chen communication by antioxidant catechins isolated
H., Thomas R.K., Liang C., Pao W. (2011) Insights into from Chinese green tea. Carcinogenesis;10:1003
ALK-driven cancers revealed through development of 1008.
novel ALK tyrosine kinase inhibitors. Cancer 24. Maestro, version 9.3, Schro dinger, LLC, New York,
Res;15:49204931. NY, 2012.
19. Cressier D., Prouillac C., Hernandez C., Amourette C., 25. Maffei Facino R.M., Cairini M., Saibene L. (1996)
Diserbo M., Lion C., Rima G. (2009) Synthesis, antioxi- Scavenging of free radicals by tenoxicam: a partici-
dant properties and radioprotective effects of new pating mechanism in the antirheumatic/antiinflamma-
benzothiazoles and thiadiazoles. Bioorg Med tory efficacy of the drug. Arch Pharm Med
Chem;17:52755284. Chem;329:457463.
20. Kus C.G., Ayhan-Klcgil F., Kaynak B., Melek K., Tulay 26. Gautam R., Srivastava A., Jachak S.M., Saklani A.
O., Can-Eke B. (2008) Synthesis and antioxidant proper- (2010) Anti-inflammatory, cyclooxygenase (COX)-2,
ties of novel N-methyl-1,3,4-thiadiazol-2-amine and 4- COX-1 inhibitory and antioxidant effects of Dysophylla
methyl-2H-1,2,4-triazole-3(4H)-thione derivatives of stellata Benth. Fitoterapia;81:4549.
benzimidazole class. Bioorg Med Chem;16:42944303. 27. Gautam R., Karkhile K.V., Bhutani K.K., Jachak S.M.
21. Galla R., Koganti P. (2011) Synthesis and Biological (2010) Anti-inflammatory, cyclooxygenase (COX)-2,
Evaluation of 3-Amino Pyrazolones. Asian J COX-1 inhibitory, and free radical scavenging effects of
Chem;23:684686. Rumex nepalensis. Planta Med;76:15641569.
22. Green L.C., Wagner D.A., Glogowski J., Sleipper P.L.,
Tannenbaum S.R. (1982) Analysis of nitrate, nitrite,

Chem Biol Drug Des 2014; 84: 409419 419

Anda mungkin juga menyukai