Anda di halaman 1dari 12

European Journal of Medicinal Chemistry 145 (2018) 594e605

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Design, synthesis, biological evaluation and docking studies of new


3-(4,5-dihydro-1H-pyrazol/isoxazol-5-yl)-2-phenyl-1H-indole
derivatives as potent antioxidants and 15-lipoxygenase inhibitors
Haydi Saher ElBordiny a, Mostafa Mahmoud El-Miligy b, Shaymaa Emam Kassab a, *,
Hoda Daabees a, Waleed Ali Mohamed Ali c, Soad Abdelhamid Mohamed El-Hawash b
a
Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira, 22516, Egypt
b
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21526, Egypt
c
Biochemistry Department, General Hospital, Cairo, 11753, Egypt

a r t i c l e i n f o a b s t r a c t

Article history: New candidates of 3-(4,5-dihydro-1H-pyrazol/isoxazol-5-yl)-2-phenyl-1H-indole derivatives (4e7) were


Received 31 October 2017 designed combining the pyrazoline/isoxazoline heterocycles and 2-phenylindole to explore its potential
Received in revised form as 15-lipoxygenase (15-LOX) inhibitors. The design of the new derivatives was based on utilizing the
22 December 2017
antioxidant properties of pyrazoline, 2-phenylindole and the good 15-LOX inhibition properties of
Accepted 8 January 2018
Available online 10 January 2018
indolylpyrazoline. The derivatives were synthesized adopting simple and laboratory friendly reaction
conditions to give the target compounds in quantitative yields. The resulting indolylpyrazolines/iso-
xazolines were evaluated as antioxidants against 2,2-diphenyl-1-picrylhydrazyl (DPPH), nitric oxide (NO)
and superoxide dismutase (SOD); indolylpyrazoline (4b) was the most potent antioxidant against SOD
assay (IC50 ¼ 1.78 mM) to be superior to ascorbic by 2 folds. Consistently, (4b) was the most potent in-
hibitor when tested against Soybean 15-LOX (IC50 ¼ 3.84 mM) excelling quercetin as standard inhibitor by
1.8 folds. Some of the new derivatives were docked into the active binding site of human 15-LOX (PDB
entry 4NRE) emphasizing the most potent derivative (4b) and the least potent one (4c). Docking solu-
tions of compounds (4b), (4c), (5b) and (6c) revealed that (4b) was the only compound that got stabilized
into the catalytic pocket of enzyme by p-cation interaction with the catalytic Feþ and formation of one
hydrogen bond with Ile 676 amino acid. Other derivatives including the least potent one variably got
stabilized into the active binding pocket by p-cation interaction with the catalytic Feþ but failed to form
hydrogen bond with Ile 676. For the future optimization of the generated inhibitors, (i) antioxidant
activity against SOD, (ii) the inhibitor stabilization by p-cation interaction with the catalytic Feþ3 and (iii)
formation of hydrogen bond with Ile 676 should be regarded.
© 2018 Elsevier Masson SAS. All rights reserved.

1. Introduction isoforms of LOXs; 5-LOX, 12-LOX and 15-LOX were found to be


involved in the etiology and progression of various human diseases
Lipoxygenases (LOXs) constitute a family of dioxygenases, that that attracted the attention to the design and emerge of new drug
catalyze dioxygenation of 1,4-cis,cis-pentadiene-containing poly- candidates to work as potential lipoxygenase inhibitors [4]. Inter-
unsaturated fatty acids such as arachidonic (AA) and linoleic acids estingly, it has been proved that 15-LOX participated in cardiovas-
to related hydroperoxides [1,2]. They are class of non-heme iron cular complications, astherosclerosis as it has a role in the oxidative
containing enzyme found in both plant and animals. Three main modification of low-density lipoproteins (LDL) [5]. Moreover, 15-
LOXs including 5, 12 and 15-lipoxygenases have been distinguished LOX has been implicated in the pathophysiology of neurodegen-
based on the peroxidation site of AA [3]. Different mammalian erative diseases like Alzheimer's disease [6], progression of certain
cancer cell types [7e9], and formation of pro-inflammatory and
thrombotic mediators that result from oxidation of arachidonic and
* Corresponding author. linoleic acids [10,11]. Thus, 15-LOX is definitely a considerable
E-mail addresses: shaymaakassab@yahoo.com, shaymaa.kassab@pharm.dmu. medicinal target for therapeutic intervention [12].
edu.eg (S.E. Kassab).

https://doi.org/10.1016/j.ejmech.2018.01.026
0223-5234/© 2018 Elsevier Masson SAS. All rights reserved.
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 595

Regarding the crystal structure of different LOX members both 2-phenylindole and pyrazoline as antioxidants to utilize the
whether prokaryotic, plant, fungal, or vertebrate/mammalian, all good LOX inhibition activity of indolylpyrazoline for investigation
have the same catalytic domain that contains the non-heme iron of the new hybrids as potent small molecule inhibitors against
atom with conserved amino acids [13e17]. More careful investi- Soybean 15-LOX enzymatic assay. Isoxazoline ring have been cho-
gation about the sequence identity of different LOXs; those are from sen to represent isosteric replacement of pyrazoline ring of the new
plant origin and those are mammalian revealed that the highest hybrid structures in a way to examine the role of such isostere in
sequence identity between LOXs of both lies in the area of catalytic the potential activity of new derivatives against the target enzyme.
domain [18,19]. These important findings made the researchers Moreover, it is planned to evaluate the potential of the generated
confidently depend on the Soybean LOX which is more accessible target inhbitors as antioxidants against DPPH, NO, and SOD assays.
than human LOX for designing, testing, characterizing, defining the Testing the antioxidant activity of the new candidates against
inhibition mechanism and discovery of small molecule enzyme multiple antioxidant assays is targeted aiming at characterizing the
inhibitors [20]. antioxidant performance of the compounds that to us, is as
Most of publications that reported elaboration of new small important as 15-LOX inhibition activity. Because this would help us
molecule 15-LOX inhibitors adopted two strategeies [21e25] in the identify the antioxidant parameter that is most reliable in the
molecular design to alter the enzyme activity pathways as the emerge of redox 15-LOX inhibitors for the present study and for the
following: (i) antioxidants to interfer with the redox cycle of 15- future studies as well.
LOX and (ii) iron-chelators to stop the enzyme catalysis via coor-
dination with the metal. 2. Results and discussion
In pursuing the antioxidant potential of various heterocycles, we
have noticed that some interesting pyrazolines solely (Fig. 1-II) and/ 2.1. Chemistry
or in conjunction to other heterocycles exhibited potential anti-
oxidant activity [26e28]. On the other hand, 2-phenylindole de- In the present study, the key intermediate (3) that would be
rivatives (Fig. 1-I) showed promising antioxidant activity in utilized in synthesizing the target indolylpyrazolines (4) and
comparison to melatonin when tested against 2,2-diphenyl-1- indolylisoxazolines (5); wlas prepared as shown in Scheme 1. 2-
picrylhydrazyl (DPPH), superoxide radical scavenging and ferric phenylindole-3-carboxaldehyde (1) was prepared as reported
ions reducing antioxidant (FRAP) assays [29,30]. Interestingly, a previously adopting Vilsmeier-Haack reaction [32]. 4-/3-
report confirmed the good to moderate 15-LOX inhibition activity acetylpyridines and 2-acetylthiophene were reacted with 2-
of some indolylpyrazoline derivatives (Fig. 1-III) and emphasized phenyl-1H-indole-3-carboxaldehyde (1) by fusion in the presence
the importance of pyrazoline ring for the LOX inhibition activity of catalytic amount of piperidine [33] to afford the corresponding
that was lost when it got aromatized [31]. chalcones (3a-c) in very quantitative yields. All trials to synthesize
Accordingly, we describe herein the design of a hybrid scaffold the N-acetylchalcone counterparts of (3a-c) from 1-acetyl-2-
in which 2-phenylindole is substituted with pyrazoline ring at phenyl-1H-indole-3-carboxaldehyde (2) [34] adopting the same
position 3 to ultimately formulate new candidates of 3-(4,5- reaction conditions varying the reaction time, temperature, and/or
dihydro-1H-pyrazol-5-yl)-2-phenyl-1H-indole derivatives (Fig. 1- relative amount of the reagents were unsuccessful and deacetyla-
target 15-LOX inhibitors) that combines the beneficial effects of tion occurred to eventually afford the N-deacetylchalcones (3a-c).

Fig. 1. Rational design of the target molecular hybrid of 15-LOX inhibitors.


596 H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605

Scheme 1. Synthesis of indoly-3-pyrazolines (4) and indoly-3-isoxazolines (5).

This was confirmed by the m.p, IR and 1H-NMR of the products. to obtain the corresponding indolyl-3-(1-acetyl/propionylpyrazo-
Cyclization of chalcones (3a-c) into the corresponding indo- line) [38] failed to achieve the target products but it gave 2-
lylpyrazolines (4a-c) was conducted by treating the chalcones with phenylindole.
slight excessive equivalent amount of hydrazine hydrate in Compounds (6) and (7) were confirmed by IR and 1H-NMR, 13C-
refluxing ethanol [35,36] to give the target derivatives in quanti- NMR, Mass spectroscopy and Microanalysis.
tative yields. While indolylisoxazolines (5a-c) were synthesized by
reacting the respective chalcones (3a-c) with 1.4 equivalent 2.2. Antioxidant activity
amount of hydroxylamine hydrochloride in refluxing ethanol con-
taining catalytic amount of glacial acetic acid [37]. The resulting
compounds (3), (4), and (5) were confirmed by IR and 1H-NMR, 13C-
NMR, Mass spectroscopy and Microanalysis.
The preparation of the second-line of target compounds,
indolyl-3-(1-acylpyrazolines) (6) and indolyl-3-(1-
methylpyrazoline) (7) as shown in Scheme 2 was achieved by
adopting a procedure in which the indolyl-3-pyrazolines (4a,b)
were subjected to acetyl chloride or benzoyl chloride or methyl
iodide while cooling the starting indolylpyrazoline solution in dry
acetone in ice-bath; in the presence of triethylamine (TEA) [36]. (5) (4, 6, 7)
After complete addition of acyl chloride or methyl iodide, the re-
action temperature was raised to room temperature and the reac-
tion mixture was left stirring until judged complete to give the
respective indolyl-3-(1-acylpyrazolines) (6a-d) and indolyl-3-(1- 2.2.1. DPPH scavenging activity
methylpyrazoline) (7). It is important to report that refluxing a DPPH assay is used to determine the antioxidant potential of the
solution of indolylpyrazoline (4a) in either acetic or propionic acid new candidates (4e7) implementing Nahar et al.'s method [39]. The
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 597

Scheme 2. Synthesis of indoly-3-acylpyrazolines (6) and indoly-3-(1-methylpyrazoline) (7).

data as shown in Table 1 revealed the potential antioxidant activity ascorbic acid (IC50 ¼ 1.78, 1.87 and 2.57 mM respectively). Indo-
of indolylpyrazolines (4a-c) and the modest antioxidant property of lylpyrazoline (4b) was the most potent derivative (IC50 ¼ 1.78 mM)
the isoxazoline counterparts (5a-c) when compared to that of the with 2 folds that of ascorbic acid (IC50 ¼ 3.54 mM). In addition,
reference standard ascorbic acid (IC50 ¼ 23.40 mg/mL). Indolylpyr- indolylpyrazoline (4a) (IC50 ¼ 3.24 mM) exhibited comparable po-
azoline (4b) was the most potent antioxidant derivative tency to ascorbic acid and indolylisoxazoline (5b) (IC50 ¼ 3.54 mM)
(IC50 ¼ 13.61 mg/mL) even with 1.7 folds that of ascorbic acid. In was equipotent to ascorbic acid. The rest of compounds displayed
addition, indolyl-3-(1-acetylpyrazoline) derivative (6c) good SOD inhibition activities. The resulted activities of the tested
(IC50 ¼ 37.97 mg/mL) and indolyl-3-(1-methylpyrazoline) derivative compounds (4e7) as antioxidants against SOD emphasized the
(7) (IC50 ¼ 33.10 mg/mL) displayed good antioxidant activity but important role of 2-phenylindole in the antioxidant activity in this
lower than ascorbic acid. The lower antioxidant activity of indoly- enzymatic assay regardless the derivative was either indolylpyr-
lisoxazolines (5a-c), N-acylated indolylpyrazolines (6a-d) and N- azoline (4b) or indolylisoxazoline (5b) or pyrazoline-N-substituted
methyl indolylpyrazoline (7) made us infer that the free pyrazoline- counterpart (6c). Those dertivatives were even superior to ascorbic
NH might play a significant role in the redox reaction process. acid in the antioxidant potential. But the contribution of pyrazoline
and isoxazoline rings in controlling the activity of the generated
2.2.2. NO scavenging activity hybrids couldn't be neglected. So, the lower activity upon removal
NO assay is used to estimate the scavenging power of the target of isoxazoline and pyrazoline rings for compounds (4b), (5a) and
compounds (4e7) to NO. The results recorded in Table 1 showed (6c) is highly expected according to the IC50 values reported for the
that indolylpyrazolines (4a-c) displayed higher NO scavenging compounds. This is because the antioxidant activity appeared to be
potential than the indolylisoxazoline counterparts (5a-c) when (4b) > (5a) > (6c) to confirm the excel of indolylpyrazoline over
compared to the ascorbic acid as a reference standard. Indolylpyr- both indolylisoxazoline and N-acetylindolylpyrazoline and infer
azoline (4a) was the most potent NO scavenger (IC50 ¼ 20.99 mg/ the role of pyrazoline and isoxazoline in discriminating the po-
mL) with 1.2 folds that of ascorbic acid (IC50 ¼ 24.50 mg/mL). tential antioxidant properties among potent compounds.
Additionally, indolylpyrazolines (6a,c) showed comparable NO
scavenging activity (IC50 ¼ 29.14, and 29.80 mg/mL respectively) to 2.3. In vitro 15-lipoxygenase inhibition activity
ascorbic acid. The significant lower antioxidant activity of indoly-
lisoxazolines (5a-c) confirms that isoxazoline ring was not favour- New target inhibitors (4e7) were tested against Soybean 15-LOX
able substitution over pyrazoline ring for the antioxidant potaential enzyme and the results were expressed as IC50 values measured in
of the tested compounds against NO assay. mM as shown in Table 1. Compounds 4a,b; 5a,b; and 6b,c showed
potential 15-LOX inhibition activity at concentration lower than
2.2.3. SOD inhibition activity that of the standard inhibitor quercetin (IC50 ¼ 6.87 mM). Indo-
New target compounds (4e7) were evaluated against SOD lylpyrazoline (4b) in which the pyrazoline ring is substituted with
enzyme to examine their antioxidant potencies and the corre- 3-pyridyl moiety, was the most potent compound (IC50 ¼ 3.84 mM)
sponding capability to prevent the generation of elemental oxygen. that gave potency 1.8 times that of quercetin. Among the tested
The resulted IC50 values measured in mM as shown in Table 1. indolylisoxazolines (5a-c), only derivatives (5a,b) showed potential
Ascorbic acid was used as reference standard for the enzymatic activity against the target enzyme. On contrary, 2-thienyl de-
assay. rivatives of both indolylpyrazoline (4c) (IC50 ¼ 9.41 mM) and indo-
Based on the data listed in Table 1, the tested compounds (4b), lylisoxazoline (5c) (IC50 ¼ 7.51 mM) gave lower potency when
(5a), and (6c) showed SOD inhibition activity higher than that of compared to 4-pyridyl derivatives (4a) (IC50 ¼ 6.54 mM); (5a)
598 H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605

Table 1
15-LOX inhibition activity and antioxidant potential of compounds (4e7).

Compound R Ar Soybean 15-LOX IC50a (mM) ±SEM DPPH IC50b (mg/mL) ±SD NO IC50c (mg/mL) ±SD SOD IC50d (mM) ±SEM

4a H 06.54 ± 0.032 15.83 ± 0.63 20.99 ± 2.50 3.24 ± 0.083

4b H 03.84 ± 0.091 13.61 ± 0.52 35.01 ± 1.64 01.78 ± 0.125

4c H 09.41 ± 0.021 18.30 ± 0.913 24.04 ± 1.82 06.51 ± 0.030

5a e 04.89 ± 0.092 59.94 ± 0.44 70.50 ± 0.54 01.87 ± 0.116

5b e 06.71 ± 0.023 60.70 ± 1.214 116.70 ± 1.98 03.54 ± 0.111

5c e 07.51 ± 0.037 117.5 ± 1.92 233.40 ± 2.44 04.26 ± 0.081

6a CH3CO 08.74 ± 0.051 98.54 ± 0.71 29.140 ± 0.93 05.47 ± 0.055

6b C6H5CO 05.14 ± 0.062 107.32 ± 3.00 52.54 ± 1.23 04.27 ± 0.061

6c CH3CO 04.56 ± 0.053 37.97 ± 0.84 29.80 ± 0.72 02.57 ± 0.077

6d C6H5CO 07.23 ± 0.055 48.21 ± 1.11 50.33 ± 0.29 04.62 ± 0.074

7 CH3 09.32 ± 0.027 33.10 ± 0.66 37.10 ± 0.85 05.41 ± 0.050

Quercetin e e 06.87 ± 0.046 NDe ND ND


Ascorbic acid e e ND 23.40 ± 1.18 24.50 ± 3.94 03.54 ± 0.091
a
IC50 values are expressed as a mean ± SEM of three experiments in mM.
b
IC50 of DPPH values are expressed as a mean ± SD of three experiments in mg/mL.
c
IC50 of NO values are expressed as a mean ± SD of three experiments in mg/mL.
d
IC50 of SOD values are expressed as a mean value ± SEM of three experiments in mM.
e
Not determined.

(IC50 ¼ 4.89 mM) and 3-pyridyl derivatives (4b) (IC50 ¼ 3.84 mM); generated LOX inhibitors.
(5b) (IC50 ¼ 6.71 mM). It was also noticed that substitution of pyr-
azoline ring at position 1 with methyl, or acetyl, or benzoyl didn't
significantly improve the potency of the corresponding non- 2.4. Docking study
substituted derivative (4a) and reduced the potency of derivatives
(4b,c). This might be attributed to the role of the larger volume of 3/ In order to investigate the orientation of the most potent com-
4-pyridyl at position 3 of pyrazoline/isoxazoline ring in imparting pound (4b) into the active binding site pocket of the human 15-LOX
better fitting of the small molecule inhibitor into the catalytic (PDB entry 4NRE) [40] and to view the inhibitor-receptor in-
pocket of target enzyme. The data recorded for the tested com- teractions, docking study was performed using (MOLSOFT ICM
pounds shows that aryl substitution at positin 3 of pyrazoline/iso- 3.4e8C) software. Analysis of the docking solution of (4b) (Fig. 2A)
xazoline ring might be having a key role in improving the potency into the active binding pocket revealed that the benzene ring of
of the new inhibitors for futur optimization of the compounds. indole inhibitor was laid into the hydrophobic cavity of the active
In summary, the non-substituted pyrazoline ring played a sig- binding pocket with pyrazoline ring and stabilized by the p-cation
nificant role in letting the target compounds perform from excel- interaction with the catalytic Feþ3. Only one hydrogen bond was
lent to good antioxidant activity against DPPH, NO and SOD assays. formed between the amino acid Ile 676 and pyrazoline-NH.
Interestingly, the antioxidant potency values of the tested com- The docking solution of the lowest potent candidate; 3-
pounds (4e7) against only SOD assay almost went aligned with the thienylindolylpyrazoline (4c) (IC50 ¼ 9.41 mM) confirmed the
15-LOX inhibition potency values for the same compounds but it implication of p-cation interaction with the catalytic Feþ3 in
went conflicted for some compounds like (4c), (5a-c), and (6a) with inhibiting the target enzyme by both the non-substituted pyrazo-
the other antioxidant assays. Thus, the SOD as a type of antioxidant line-NH and indole-benzene ring. This is because the orientation of
assays is that should be regarded in the future studies on the (4c) into the active binding pocket (Fig. 2B) showed the difficulty of
the inhibitor to get stabilized by p-cation interaction with the
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 599

Fig. 2. Docking solutions of 15-LOX inhibitors A) 4b, B) 4c, C) 5b, D) 6c (colored by element), into the active binding site of human 15-LOX (PDB entry 4NRE), tagging the protein
residues that coordinate with Fe3þ catalytic metal (light green ball) and that interacted with the inhibitors. Hydrogen bonds are represented as white dots. Hydrogen bond distance
is indicated by numbers in Å. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

catalytic metal specifically via pyrazoline-NH that almost directed into the active binding pocket and the docking solutions revealed
away but it was possible via indole-benzene ring. Moreover, the that all of them were variably stabilized by p-cation interaction but
inability of the compound to form hydrogen bond with Ile 676 the failed to form hydrogen bond with Ile 676. Moreover, the docking
same way as indolylpyrazoline (4b). solution of indolyl-3-(1-acetylpyrazoline) derivative (6c) (Fig. 2D)
Considering the docking solution of the most potent inhibitor provided us with a convenient justification to the decrease in the
(4b) as a reference simulation to the inhibitor-receptor interaction potency of 15-LOX inhibition (IC50 ¼ 4.56 mM) when compared to
pattern to which the other inhibitors' docking solutions are the none-substituted indolylpyrazoline counterpart (4b)
compared, indolylisoxazoline (5b) (Fig. 2C) was chosen as a direct (IC50 ¼ 3.84 mM). The N-COCH3 enabled the compound (6c) to get
counterpart to (4b) to be docked into the active binding pocket of stabilized by p-cation interaction with the catalytic metal via
15-LOX. This is an attempt to realize the structural requirements carbonyl-oxygen but failed via indole ring. Also, absence of
among the new candidates to perform potent inhibition to the hydrogen bond donor of pyrazoline-NH made the compound un-
target enzyme. The docking solution and the orientation of indo- able to interact with Ile 676 via formation of hydrogen.
lylisoxazoline (5b) into the binding pocket showed the possibility
of the compound to get stabilized by p-cation interaction with the
catalytic metal via both isoxazolineeoxygen and indole-benzene 3. Conclusion
ring without any possible formation of hydrogen bond with Ile
676 due to absence of hydrogen bond donor NH group. Interest- The present study introduced new candidates of indolylpyr-
ingly, the binding mode of (5b) combined with the binding mode of azolines and indolylisoxazolines in which 2-phenylindole was
(4c) indicated that the formation of hydrogen bond with amino acid combined with pyrazoline/isoxazoline heterocycles to get
Ile 676 via either hydrogen bond donor or acceptor might be benefited of both as antioxidants and the good LOX inhibition ac-
essential for potent inhibition of the target enzyme. Thus, the lower tivity of indolylpyrazolines. The 15-LOX inhibition potential of the
potency of all the candidates other than (4b) might be attributed to new hybrids was verified upon testing the designed compounds
the inability of the compounds to form hydrogen bond with Ile 676. against 15-LOX enzyme. Among the target compounds tested
All the other candidates generated in this study were docked against 15-LOX, indolylpyrazoline (4b) was the most potent in-
hibitor and showed 1.8 times more potency than quercetin
600 H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605

standard inhibitor. The same compound exhibited the most anti- 115.54 (indole-C5), 121.26 (indole-C6), 121.50 (pyridine-C3,5), 121.88
oxidant potential when tested against SOD with 2 times more po- (indole-C4), 123.45 (2-phenyl-C4), 125.71 (indole-C2), 128.99 (2-
tency than ascorbic acid as standard antioxidant to infer that SOD phenyl-C2,6), 129.41 (C¼C-C¼O), 129.75 (2-phenyl-C3,5), 130.70
was the most reliable antioxidant parameter to depend on in the (indole-C3a), 137.00 (2-phenyl-C1), 140.58 (C¼C-C¼O), 144.70
future studies when compared with the data obtained from other (indole-C7a),146.20 (pyridine-C4), 150.62 (pyridine-C2,6), 188.56
antioxidant parameters. Docking solutions of the most potent (CO). MS EIMS: m/z (calcd) 324.13 (found) 323.90 (Mþ). Anal.
candidate (4b), the less potent candidates (4c), (5b) and (6c) Calcd for C22H16N2O: C, 81.46; H, 4.97; N, 8.64. Found: C, 81.63; H,
revealed that stabilization of the ligand inhibitor via p-cation 5.15; N 8.79.
interaction with the catalytic Feþ3 and formation of hydrogen bond
with the amino acid Ile 676 might be required for potential inter- 4.1.3. (E)-3-(2-Phenyl-1H-indol-3-yl)-1-(pyridin-3-yl) prop-2-en-
ference with the catalytic process of the target enzyme. Conclu- 1-one (3b)
sively, the study of the new hybrids generated as potential 15-LOX Yield: 90%. Yellow crystals; m.p.: 224e225  C, IR (KBr) ʋmax/
inhibitors coupled with the significant antioxidant activity high- cm1: 3265 (NH), 1645 (CO), 1H-NMR (DMSO-d6, 400 MHz):
lights some important fundamentals that should be regarded in the d 7.31e7.78 (m, 10H, indole-C4,5,6,7eH, 2-phenyl-C2,3,4,5,6-H,
future studies for further optimization of the generated candidates: CH¼CHCO), 8.09 (d, J ¼ 15.00 Hz, 1H, CH¼CH-CO), 8.21 (d,
i) SOD inhibition activity is the most reliable parameter for testing J ¼ 8.00 Hz, 1H, pyridine-C6-H), 8.28 (d, J ¼ 7.00 Hz, 1H, pyridine-C4-
the antioxidant activity of the inhibitors ii) Ligand inhibitor stabi- H), 8.79 (m, 1H, pyridine-C5-H), 9.26 (s, 1H, pyridine-C2-H), 12.3 (s,
lization by p-cation interaction with the catalytic Feþ3 and iii) 1H, NH, D2O exchangeable). 13C-NMR (DMSO-d6, 100 MHz):
formation of hydrogen bond with Ile 676 amino acid contribute to d 109.39 (indole-C3), 112.23 (indole-C7), 115.92 (indole-C5), 121.37
the potential activity of the inhibitors. (indole-C6), 121.78 (indole-C4), 123.34 (indole-C2), 123.83 (indole-
C3a), 125.73 (C¼C-CO), 128.99 (2-phenyl-C2,6), 129.34 (2-phenyl-C4),
4. Experimental 129.73 (2-phenyl-C3,5), 130.81 (pyridine-C3), 133.63 (2-phenyl-C1),
135.68 (C4-pyridine), 136.92 (indole-C7a), 139.58 (C5-pyridine),
4.1. Chemistry 145.71 (C¼C-CO), 149.25 (pyridine-C2), 152.67 (pyridine-C6), 187.89
(CO). MS EIMS: m/z (calcd) 324.13 (found) 323.98 (Mþ). Anal.
Melting points were determined on digital Gallen-Kamp MFB- Calcd for C22H16N2O: C, 81.46; H, 4.97; N 8.64. Found: C, 81.58; H,
595 instrument using open capillary tubes and are uncorrected. IR 4.79; N, 8.49.
spectra were recorded as potassium bromide discs on Schimadzu
FT-IR 440 spectrometer. 1H-NMR spectra were recorded on Bruker 4.1.4. (E)-3-(2-Phenyl-1H-indol-3 -yl)-1-(thiophen-2-yl) prop-2-
spectrophotometer at 300 MHz in DMSO-d6, Varian Mercury VX at en-1-one (3c)
400 MHz in DMSO-d6; values (d) are given in parts per million Yield: 70%. Orange crystals; m.p.: 254e255  C. IR (KBr) ʋmax/
1
(ppm) downfield from tetramethylsilane (TMS) as internal refer- cm : 3215 (NH), 1626 (CO), 1H-NMR (DMSO-d6, 400 MHz):
ence standard. 13C-NMR spectra were recorded using the same d 7.22e7.31 (m, 3H, indole-C5,6eH, 2-phenyl-C4-H),7.49 (d,
spectrophotometers that used for recording 1H NMR. Mass spectra J ¼ 7.50 Hz, 1H, C7-indole-H), 7.55e7.64 (m, 5H, C4,5-thiophen-H, 2-
were recorded on Shimadzu GCMS/QP-2010 plus (70eV) mass phenyl- C3,5-H, -CH¼CH-CO), 7.78 (d, J ¼ 8Hz, 2H, 2-phenyl C-2,6-H),
spectrometer. The elemental analyses were performed at the 8.02 (d, J ¼ 15.00 Hz, 1H, -CH¼CH-CO,), 8.20e8.26 (m, 2H, indole-
Microanalytical Center, Cairo University, Cairo, Egypt. Reactions C4-H, thiophen-C3-H), 12.35 (s, 1H, NH, D2O exchangeable). 13C-
were followed up by thin layer chromatography (TLC) using Merck NMR (DMSO-d6, 100 MHz): d 112.02 (indole-C3), 116.06 (indole-C7),
Silica gel/TLC cards with fluorescent indicator UV254, the spots 121.07 (indole-C5), 121.65 (indole-C6), 122.45 (indole-C4), 123.27
were visualized using Spectroline E series dual wavelength UV (indole-C2), 123.71 (indole-C3a), 125.77 (2-phenyl-C2,6), 128.76
lamp at l ¼ 254 nm. (C¼C-CO), 128.98 (2-phenyl-C4), 129.25 (2-phenyl-C3,5), 129.69
(thiophen-C4), 129.85 (thiophen-C3), 129.91 (2-phenyl-C1), 132.18
4.1.1. General procedure for (E)-3-(2-Phenyl-1H-indol-3-yl)-1- (thiophen-C5), 134.22 (indole-C7a), 135.91 (thiophen-C2), 137.85
(aryl) prop-2-en-1-ones (3a-c) (C¼C-CO), 185.51 (CO). MS EIMS: m/z (calcd) 329.09 (found) 329.22
A mixture of 2-phenyl-1H-indole-3-carboxaldehyde (1) (Mþ). Anal. Calcd for C21H15NOS: C,76.57; H, 4.59; N, 4.25. Found:
(1.0 mmol, 0.22 g) and 3- or 4-acetylpyridine (1.0 mmol, 0.1 mL) or C, 76.38; H, 4.77; N, 4.56.
2-acetylthiophen (1.3 mmol) and piperidine (0.2 mL) was heated
for 1 h, then ethanol (1.5 mL), glacial acetic acid and water (1:1) 4.1.5. General procedure for 2-Phenyl-3-(3-aryl-4,5-dihydro-1H-
were added dropwise to the resulting red solution until first pyrazol-5-yl)-1H-indoles (4a-c)
appearance cloudiness. The resulting product was filtered off, A solution (3a-c) (1.0 mmol), and hydrazine hydrate 99%
washed with water and recrystallized from ethanol. In case of 2- (1.1 mmol, 0.06 g) in absolute ethanol (10 mL) was refluxed for
thienyl derivative of chalcone, the product obtained was sticky 8e10 h. The resulting solution was concentrated, left to cool, the
mass, triturated with a mixture of petroleum ether and diethyl solid obtained was filtered off and recrystallized from the appro-
ether to afford yellow precipitate, then recrystallized from ethanol. priate solvent.

4.1.2. (E)-3-(2-Phenyl-1H-indol-3-yl)-1-(pyridin-4-yl) prop-2-en- 4.1.6. 2-Phenyl-3-[3-(pyridin-4-yl)-4,5-dihydro-1H-pyrazol-5-yl]-


1-one (3a) 1H-indole (4a)
Yield: 85%. Orange crystals; m.p.: 278e280  C. IR (KBr) ʋmax/ Yield: 60.0%. Recrystallized from ethanol; pale yellow crystals;
cm1: 3015 (NH), 1645 (CO), 1H-NMR (DMSO-d6, 400 MHz): m.p.: 209e210  C. IR (KBr) ʋmax/cm1: 3359 (NH). 1H-NMR (DMSO-
d 7.28e7.31 (m, 2H, indole-C5,6-H), 7.49e7.56 (m, 2H, indole-C4-H,- d6, 300 MHz): d 11.35 (s, 1H, NH, D2O exchangeable), 8.55 (d,
CH¼CH-C¼O), 7.58e7.63 (m, 5H, 2-phenyl-C2,3,4,5,6-H), 7.92 (d, J ¼ 4.7 Hz, 2H, pyridine-C2,6eH), 8.01 (s, 1H, NH-pyrazoline, D2O
J ¼ 5.0 Hz, 2H, pyridine-C3,5-H), 8.05 (d, J ¼ 15.45 Hz, 1H, -CH¼CH- exchangeable), 7.63 (d, J ¼ 7.7 Hz, 2H, 2-phenyl-C2,6eH), 7.54 (dd,
C¼O), 8.20 (d, J ¼ 7.00 Hz, 1H, indole-C4-H), 8.78 (d, J ¼ 5.0 Hz, 2H, J ¼ 11.5, 5.4 Hz, 4H, indole-C4,7, pyridine-C3,5eH), 7.42 (dd, J ¼ 12.6,
pyridine-C2,6-H), 12.34 (s, 1H, NH, D2O exchangeable). 13C-NMR 6.0 Hz, 3H, 2-phenyl-C3,4,5-H), 7.10 (t, J ¼ 7.5 Hz, 1H, indole-C6-H),
(DMSO-d6, 100 MHz): d 109.44 (indole-C3), 112.35 (indole-C7), 6.92 (t, J ¼ 7.4 Hz, 1H, indole-C5-H), 5.26 (t, J ¼ 11.6 Hz, 1H,
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 601

pyrazoline-C5eH), 3.50e3.41 (m, 1H, pyrazoline-C4eH), 3.20 (dd, phenyl-C2,4,6), 7.46 (t, J ¼ 8.1 Hz, 2H, indole-C5,6-H), 7.33 (d,
J ¼ 16.6, 11.1 Hz, 1H, pyrazoline-C4eH). 13C-NMR (DMSO-d6, J ¼ 7.7 Hz, 1H, indole-C7-H), 7.15 (t, J ¼ 7.5 Hz, 1H, 2-phenyl-C3-H),
75 MHz): d 37.59 (pyrazoline-C4), 56.85 (pyrazoline-C5), 111.630 6.99 (t, J ¼ 7.4 Hz, 1H, 2-phenyl-C5-H), 6.08 (t, J ¼ 10.9 Hz, 1H, iso-
(indole-C3), 111.50 (indole-C7), 118.80 (indole-C4), 119.27 (indole- xazoline-C5eH), 3.89 (dd, J ¼ 17.3, 11.8 Hz, 1H, isoxazoline-C4eH),
C5), 120.00 (indole-C6), 121.56 (indole-C2), 126.15 (indole-C3a), 3.66 (dd, J ¼ 17.4, 10.2 Hz, 1H, isoxazoline-C4eH). 13C-NMR (DMSO-
127.85 (2-phenyl-C4), 128.68 (pyridine-C2,6), 128.83 (2-phenyl-C2,6), d6, 75 MHz): d 78.78 (isoxazoline-C5), 109.00 (indole-C3), 112.42
132.26 (2-phenyl-C3,5), 135.92 (2-phenyl-C1), 136.45 (indole-C7a), (indole-C7), 119.61 (indole-C4), 120.16 (indole-C5), 121.20 (indole-
140.51 (pyridine-C1), 145.44 (pyrazoline-C3), 149.83 (pyridine-C3,5). C6), 122.54 (indole-C2), 126.0 (indole-C3a), 128.90 (pyridine-C3,5),
MS EIMS: m/z (calcd) 338.15 (found) 337.95 (Mþ). Anal. Calcd for 129.4 (2-phenyl-C2,6), 132.13 (2-phenyl-C1), 136.9 (2-phenyl-C3,5),
C22H18N4: C, 78.08; H, 5.36; N, 16.56. Found: C, 78.27; H, 5.54; N, 137.40 (indole-C7a), 138.17 (pyridine-C4), 150.87 (pyridine-C2,6),
16.37. 156.24 (isoxazoline-C3). MS EIMS: m/z (calcd) 339.14 (found)
339.00 (Mþ). Anal. Calcd for C22H17N3O: C, 77.86; H, 5.05; N, 12.38.
4.1.7. 2-Phenyl-3-[3-(pyridin-3-yl)-4,5-dihydro-1H-pyrazol-5-yl]- Found: C, 78.06; H, 4.90; N, 12.56.
1H-indole (4b)
Yield 57.0%. Recrystallized from ethanol; yellow powder; m.p. 4.1.11. 5-(2-Phenyl-1H-indol-3-yl)-3-(pyridin-3-yl)-4,5-
179e180  C. (KBr) ʋmax/cm1: 3056, 3318 (NH). 1H-NMR (DMSO- dihydroisoxazole (5b)
d6, 300 MHz): d 11.33 (s, 1H, NH, D2O exchangeable), 8.86 (s, 1H, Yield: 70%. Buff powder; m.p.119e120  C. IR (KBr) ʋmax/cm1:
pyridine-C2-H), 8.51 (d, J ¼ 2.2 Hz, 1H, pyridine-C4-H), 8.04 (d, 3386 (NH),1H-NMR (DMSO-d6, 300 MHz): d 11.58 (s, 1H, NH, D2O
J ¼ 8.0 Hz, 1H, pyridine-C6-H), 7.68 (s, 1H, NH-pyrazoline, D2O exchangeable), 8.94 (s, 1H, pyridine-C2-H), 8.68 (d, J ¼ 4.7 Hz, 1H,
exchangeable), 7.64 (d, J ¼ 7.4 Hz, 2H, indole-C4,7-H), 7.52 (dd, pyridine-C4-H), 8.18 (d, J ¼ 7.9 Hz, 1H, pyridine-C6-H), 7.65e7.50 (m,
J ¼ 15.2, 7.7 Hz, 3H, pyridine-C5-H, 2-phenyl-C2,6eH), 7.46e7.37 (m, 5H, pyridine-C5-H, indole-C4-H, 2-phenyl-C3,4,5-H), 7.51e7.41 (m,
3H, 2-phenyl-C3,4,5eH), 7.10 (t, J ¼ 7.5 Hz, 1H, indole-C6-H), 6.93 (t, 2H, 2-phenyl-C2,6-H), 7.37 (d, J ¼ 8.0 Hz, 1H, indole-C7-H), 7.15 (t,
J ¼ 7.5 Hz, 1H, indole-C5-H), 5.22 (t, J ¼ 11.3 Hz, 1H, pyrazoline-C5- J ¼ 7.5 Hz, 1H, indole-C6-H), 6.99 (t, J ¼ 7.5 Hz, 1H, indole-C5-H), 6.03
H), 3.47 (dd, J ¼ 16.3, 11.9 Hz, 1H, pyrazoline-C4-H), 3.24 (dd, (t, J ¼ 10.9 Hz, 1H, isoxazoline-C5-H), 3.92 (dd, J ¼ 17.2, 11.6 Hz, 1H,
J ¼ 16.6, 11.4 Hz, 1H, pyrazoline-C4-H). 13C-NMR (DMSO-d6, isoxazoline-C4eH), 3.69 (dd, J ¼ 17.4, 10.4 Hz, 1H, isoxazoline-
75 MHz): d 38.20 (pyrazoline-C4), 56.57 (pyrazoline-C5), 111.45 C4eH). 13C-NMR (DMSO-d6, 75 MHz): d 77.68 (isoxazoline-C5),
(indole-C3), 111.68 (indol-C7), 118.72 (indole-C4), 120.22 (indole-C5), 109.39 (indole-C3), 111.72 (indole-C7), 119.17 (indole-C4), 119.51
121.52 (indole-C6), 123.66 (indole-C2), 127.81 (indol-C3a), 128.67 (indole-C5) 121.93 (pyridine-C5), 124.02 (indole-C6), 125.82 (indole-
(pyridine-C2,6), 128.83 (2-phenyl-C2,6), 129.36 (2-phenyl-C4), 132.14 C2), 128.27 (2-phenyl-C2,6), 128.84 (indole-C3a), 131.65 (2-phenyl-
(2-phenyl-C3,5), 135.90 (2-phenyl-C1), 136.46 (indol-C7a), 145.94 C4), 133.84 (2-phenyl-C3,5), 136.38 (2-phenyl-C1, pyridine-C3),
(pyridine-C1), 146.48 (pyridine-C3,5), 148.54 (pyrazoline-C3). MS 137.49 (indole-C7a, pyridine-C4), 147.82 (pyridine-C2), 152.10 (pyri-
EIMS: m/z (calcd) 338.15 (found) 338.44 (Mþ). Anal. Calcd for dine-C6), 155.30 (isoxazoline-C3). MS EIMS: m/z (calcd) 339.14
C22H18N4: C, 78.08; H, 5.36; N, 16.56. Found: C, 78.28; H, 5.54; N, (found) 339.29 (Mþ). Anal. Calcd for C22H17N3O: C,77.86; H, 5.05;
16.73. N,12.38. Found: C, 78.04; H, 5.19; N, 12.20.

4.1.8. 2-Phenyl-3-[3-(thiophen-2-yl)-4, 5-dihydro-1H-pyrazol-5- 4.1.12. 5-(2-Phenyl-1H-indol-3-yl)-3-(thien-2-yl)-4,5-


yl]-1H-indole (4c) dihydroisoxazole(5c)
Yield: 55.0%. Recrystallized from benzene; mustard yellow Yield 80%. Yellow powder; m.p.188e190  C. IR (KBr) ʋmax/cm1:
powder; m.p. 124e125  C. IR (KBr) ʋmax/cm1: 3388, 3213 (NH). 1H- 3345 (NH). 1H-NMR (DMSO-d6, 400 MHz): d 12.59 (s, 1H, NH, D2O
NMR (DMSO-d6, 400 MHz): d 11.29 (s, 1H, NH, D2O exchangeable), exchangeable), 7.98 (d, J ¼ 7.5 Hz, 1H, thiophen-C3-H), 7.78 (d,
7.59 (s,1H, NH-pyrazoline), 7.52 (m, 3H, 2-phenyl-C3,4,5-H), 7.43 (m, J ¼ 8.0 Hz, 2H, 2-phenyl-C-2,6-H), 7.48e7.66 (m, 6H, thiophen-C4,5-
5H, indole-C4,7-H, thiophen-C5eH, 2-phenyl-C2,6-H), 7.07 (m, 3H, H, indole-C4,7-H, 2-phenyl- C3,5-H), 7.27e7.37 (m, 2H, indole-C6eH,
thiophen-C3eH, indole-C5,6-H), 6.96 (m, 1H, indole-C5-H), 5.24 (t, 2-phenyl-C4-H), 7.10e7.11 (m, 1H, indole-C5-H), 5.93 (t, J ¼ 11.0 Hz,
J ¼ 11.4 Hz, 1H, pyrazoline-C5-H), 3.40 (dd, J ¼ 16.4, 11.5 Hz, 1H, 1H, isoxazoline-C5-H), 3.90 (dd, J ¼ 17.5,11.0 Hz, 1H, isoxazoline-C4-
pyrazoline-C4-H), 3.20 (dd, J ¼ 16.4, 11.6 Hz, 1H, pyrazoline-C4-H), H), 3.70 (dd, J ¼ 17.5, 11.3 Hz, 1H, isoxazoline-C4-H). MS EIMS: m/z
MS EIMS: m/z (calcd) 343.11 (found) 343.32 (Mþ). Anal. Calcd for (calcd) 344.10 (found) 344.35 (Mþ). Anal. Calcd for C21H16N2OS: C,
C21H17N3S: C,73.44; H, 4.99; N, 12.24. Found: C, 73.62; H, 4.78; N, 73.23; H, 4.68; N, 8.13. Found: C, 73.50; H, 4.79; N, 8.27.
12.43.
4.1.13. General procedure for synthesis of 1-[5-(2-Phenyl-1H-indol-
4.1.9. General procedure for 5-(2-Phenyl-1H-indol-3-yl)-3-(aryl)-4, 3-yl)-3-(pyridin-3-/4-yl)-4,5-dihydro-1H-pyrazol-1-yl]ethan-1-
5-dihydroisoxazoles (5a-c) ones (6a,c) and Phenyl [5-(2-phenyl-1H-indol-3-yl)-3-(pyridin-3-/
A solution of 3a-c (1.0 mmol) and hydroxylamine hydrochloride 4-yl)-4,5-dihydro-1H-pyrazol-1-yl]methanones (6b,d)
(1.4 mmol, 0.1 g) and a catalytic amount of glacical acetic acid A solution of indolylpyrazolines 4a,b (1.0 mmol) and TEA
(0.5 mL) in absolute ethanol (10 mL), was heated under reflux from (1.0 mmol, 0.1 g) in dry acetone (15 mL) was cooled to 0  C in an ice
8 to 10 h until the reaction was judged complete, the reaction bath with stirring for 30 min, then acetyl- or benzoyl chloride
mixture left to cool down then poured onto crushed ice. The pre- (1.0 mmol) was added dropwise. After complete addition, the re-
cipitate obtained was filtered off, washed with water and recrys- action mixture was stirred for 6 h at room temperature until the
tallized from ethanol. reaction judged complete. The resulting solution was poured onto
ice-cold water and the separated solid was filtered off, washed with
4.1.10. 5-(2-Phenyl-1H-indol-3-yl)-3-(pyridin-4-yl)-4,5- water and then recrystallized from ethanol.
dihydroisoxazole(5a)
Yield: 81%. Off-white powder. m.p.: 224e225  C. IR (KBr) ʋmax/ 4.1.14. 1-[5-(2-Phenyl-1H-indol-3-yl)-3-(pyridin-4-yl)-4,5-
1
cm : 3201 (NH), 1H-NMR (DMSO-d6, 300 MHz): d 11.61 (s, 1H, NH, dihydro-1H-pyrazol-1-yl]ethan-1-one (6a)
D2O exchangeable), 8.71 (d, J ¼ 4.4 Hz, 2H, C2,6-pyridine-H), 7.71 (d, Yield: 55%. Pale yellow powder; m.p. 184e185  C. IR (KBr) ʋmax/
J ¼ 4.7 Hz, 2H, C3,5-pyridineeH), 7.66e7.51 (m, 4H, indole-C4-H, 2- cm1: 3390 (NH), 1678 (CO).1H-NMR (DMSO-d6, 300 MHz): d 11.43
602 H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605

(s, 1H, NH, D2O exchangeable), 8.80 (d, J ¼ 5.0 Hz, 2H, pyridine- 4.1.17. Phenyl[5-(2-phenyl-1H-indol-3-yl)-3-(pyridin-3-yl)-4,5-
C2,6eH), 8.03 (d, J ¼ 5.0 Hz, 2H, pyridine-C3,5eH), 7.80 (d, J ¼ 7.5 Hz, dihydro-1H-pyrazol-1-yl]methanone (6d)
2H, 2-phenyl-C2,6-H), 7.06e7.57 (m, 6H, indole-C4,6,7-H, 2-phenyl- Yield 65%. Yellowish brown powder; m.p.189e190  C. IR (KBr)
C3,4,5eH), 6.88e6.93 (m,1H, indole-C5-H), 5.24 (dd, J ¼ 12.0, ʋmax/cm1: 3268 (NH), 1699 (CO). 1H-NMR (DMSO-d6, 300 MHz):
12.60 Hz, 1H, pyrazoline-C5-H), 3.90 (dd, J ¼ 18.0, 12.0 Hz, 1H, pyr- 11.39 (s,1H, NH, D2O exchangeable), 8.92 (s,1H, pyridine-C2-H), 8.66
azoline-C4-H), 3.30 (dd, J ¼ 18.0, 12.40 Hz, 1H, pyrazoline-C4-H), (d, J ¼ 5.0 Hz, 1H, pyridine-C6-H), 8.13 (d, J ¼ 7.3 Hz,1H, pyridine-C4-
2.27 (s, 3H, CH3). 13C-NMR (DMSO-d6, 75 MHz): d 22.34 (CH3CO), H), 7.90 (t, J ¼ 7.0 Hz, 1H, pyridine-C5-H), 7.70 (d, J ¼ 7.0 Hz, 1H,
41.30 (pyrazoline-C4), 55.40 (pyrazoline-C5), 111.80 (indole-C3), indole-C4-H), 7.31e7.57 (m, 11H, indole-C7-H, 2-phenyl-C2,3,4,5,6-H,
112.36 (indole-C7), 118.20 (indole-C4), 120.0 (indole-C5), 122.90 benzoyl-C2,3,4,5,6), 7.10 (m,1H, indole-C6-H), 6.90 (m,1H, indole-C5-
(indole-C6), 123.15 (pyridine-C3,5), 125.65 (indole-C2), 128.52 H), 6.18 (dd, J ¼ 11.7, 6.3 Hz, 1H, pyrazoline-C5-H), 4.0 (dd, J ¼ 17.0,
(indole-C3a), 129.26 (2-phenyl-C2,6), 129.49 (2-phenyl-C3,5), 132.80 11.7 Hz, 1H, pyrazoline-C4-H), 3.41 (m, 1H, pyrazoline-C4-H). 13C-
(2-phenyl-C4), 135.90 (2-phenyl-C1), 136.79 (indole-C7a), 144.90 NMR (DMSO-d6, 75 MHz): 49.06 (pyrazoline-C4), 55.20 (pyrazo-
(pyridine-C2,6), 145.10 (pyridine-C1), 151.37 (pyrazoline-C3), 169.15 line-C5), 111.98 (indole-C3), 112.25 (indole-C7), 118.34 (indole-C4),
(C¼O). MS EIMS: m/z (calcd) 380.16 (found) 380.42 (Mþ). Anal. 119.95 (indole-C5), 122.07 (indole-C6), 125.85 (indole-C2), 128.21
Calcd for C24H20N4O: C,75.77; H, 5.30; N,14.73. Found: C,76.04; H, (pyridine-C5),128.39 (pyridine-C1), 129.02 (indole-C3a), 129.21 (2-
5.48; N, 14.94. phenyl-C2,6), 129.41 (benzoyl-C2,6), 129.71 (benzoyl-C3,5), 129.79
(2-phenyl-C3,5), 131.2 (2-phenyl-C4), 132.94 (benzoyl-C4), 133.31 (2-
phenyl-C1), 134.25 (indole-C7a), 135.89 (benzoyl-C1), 148.19 (pyri-
dine-C2), 151.43 (pyridine-C6), 153.81 (pyrazoline-C3), 166.35 (CO).
4.1.15. Phenyl[5-(2-phenyl-1H-indol-3-yl)-3-(pyridin-4-yl)-4,5-
MS EIMS: m/z (calcd) 442.18 (found) 441.87 (Mþ). Anal. Calcd for
dihydro-1H-pyrazol-1-yl]methanone (6b)
C29H22N4O: C,78.71; H, 5.01; N, 12.66. Found C,78.97; H, 5.08; N,
Yield 65%. White crystals; m.p.: 199e200  C. IR (KBr) ʋmax/
12.94.
cm1: 3401 (NH), 1636 (CO). 1H-NMR (DMSO-d6, 300 MHz): d 11.46
(s, 1H, NH, D2O exchangeable), 8.69 (d, J ¼ 4.6 Hz, 2H, pyridine-
C2,6eH), 7.92 (d, J ¼ 7.4 Hz, 2H, benzoyl-C2,6-H), 7.76 (d, J ¼ 6.9 Hz, 4.1.18. 3-[1-Methyl-3-(pyridin-4-yl)-4,5-dihydro-1H-pyrazol-5-yl]-
2H, 2-phenyl-C2,6-H), 7.66 (d, J ¼ 4.7 Hz, 2H, pyridine-C3,5eH), 7.58 2-phenyl-1H-indole (7)
(t, J ¼ 7.5 Hz, 2H, benzoyl-C3,5-H), 7.53e7.38 (m, 5H, 2-phenyl-C3,4,5- A solution of indolylpyrazoline 4a (1.0 mmol, 0.3 g) and TEA
H, benzoyl-C4-H, indole-C4-H), 7.29 (d, J ¼ 7.9 Hz, 1H, indole-C7-H), (1 mmol, 0.1 g) in dry acetone (15 mL) was cooled in an ice bath
7.09 (t, J ¼ 7.5 Hz, 1H, indole-C6-H), 6.95 (t, J ¼ 7.4 Hz, 1H, indole-C5- with stirring for 30 min and then methyl iodide (1 mmol, 0.14 g)
H), 6.17 (dd, J ¼ 12.2, 6.4 Hz, 1H, pyrazoline-C5-H), 3.98 (dd, J ¼ 18.2, was added dropwise. The stirring was continued for 9 h at room
12.5 Hz, 1H, pyrazoline-C4-H), 3.29 (m, 1H, pyrazoline-C4-H). 13C- temperature, then the reaction mixture was poured onto ice-cold
NMR (DMSO-d6, 75 MHz): d 45.35 (pyrazoline-C4), 55.09 (pyrazo- water, the solid separated was filtered off, washed with water
line-C5), 111.30 (indole-C3), 111.81 (indole-C7), 117.72 (indole-C4), and recrystallized from ethanol.
119.46 (indole-C5), 120.52 (indole-C6), 121.58 (pyridine-C3,5), 125.31 Yield 50%. Dark orange powder; m.p.145e146  C. IR (KBr) ʋmax/
(indole-C2), 127.76 (2-phenyl-C2,6), 127.92 (indole-C3a), 128.71 cm1: 3208 (NH), 1H-NMR (DMSO-d6, 300 MHz): 11.48 (s, NH, D2O
(benzoyl-C2,6), 128.91 (benzoyl-C3,5), 129.27 (2-phenyl-C3,5), 130.82 exchangeable), 8.75 (d, J ¼ 6.9 Hz, 1H, indole-C4eH), 8.67 (d,
(2-phenyl-C4), 132.40 (benzoyl-C4), 134.56 (2-phenyl-C1), 135.46 J ¼ 4.5 Hz, 2H, pyridine-C2,6eH), 8.55 (d, J ¼ 8.0 Hz, 1H, indole-
(indole-C7a), 136.32 (benzoyl-C1), 138.21 (pyridine-C5), 150.36 C7eH), 7.96 (d, J ¼ 4.2 Hz, 2H, pyridine-C3,5eH), 7.38e7.65 (m, 5H,
(pyridine-C2,6), 153.61 (pyrazoline-C3), 166.10 (CO). MS EIMS: m/z 2-phenyl-C2,3,4,5,6-H), 7.1e7.22 (m, 1H, indole-C5eH), 6.94e6.97
(calcd) 442.18 (found) 442.25 (Mþ). Anal. Calcd for C29H22N4O: C, (m,1H, indole-C6eH), 5.5e5.6 (m, 1H, pyrazoline-C5-H), 4.20 (s, 3H,
78.71; H, 5.01; N, 12.66. Found: C, 78.96; H, 5.22; N,12.89. N-CH3), 3.45e3.6 (m, 1H, pyrazoline-C4-H), 3.15e3.22 (m, 1H, pyr-
azoline-C4-H). 13C-NMR (DMSO-d6, 100 MHz): 31.15 (CH3), 46.15
(pyrazoline-C4), 57.35 (pyrazoline-C5), 112.03 (indole-C3), 112.82
(indole-C7), 119.32 (indole-C4), 119.79 (indole-C5), 120.53 (indole-
4.1.16. 1-[5-(2-Phenyl-1H-indol-3-yl)-3-(pyridin-3-yl)-4,5-
C6), 122.08 (indole-C2), 126.18 (indole-C3a), 128.37 (2-phenyl-C4),
dihydro-1H-pyrazol-1-yl]ethan-1-one (6c)
129.35 (pyridine-C2,6), 129.89 (phenyl-C2,6), 132.77 (2-phenyl-C3,5),
Yield 60%. White powder; m.p.124e125  C. IR (KBr) ʋmax/cm1:
136.44 (2-phenyl-C1), 136.97 (indole-C7a), 141.01 (pyridine-C1),
3427 (NH), 1656 (CO). 1H-NMR (DMSO-d6, 300 MHz): d 11.40 (s, 1H,
145.97 (pyrazoline-C3), 150.35 (pyridine-C3,5). MS EIMS: (calc)
NH, D2O exchangeable), 8.99 (s, 1H, pyridine-C2-H), 8.66 (d,
352.17 (found) 352.28 (Mþ). Anal. Calcd for C23H20N4: C,78.38; H,
J ¼ 4.3 Hz, 1H, pyridine-C6eH), 8.22 (d, J ¼ 7.8 Hz, 1H, pyridine-
5.72; N,15.90. Found: C,78.56; H, 5.94; N, 16.13.
C4eH), 7.87 (d, J ¼ 7.5 Hz, 2H, indole-C4,7-H), 7.54 (t, J ¼ 7.10 Hz, 3H,
2-phenyl-C3,4,5-H), 7.42 (dd, J ¼ 15.7, 7.7 Hz, 2H, indole-C6-H, pyri-
dine-C5), 7.21e6.97 (m, 2H, 2-phenyl-C2,6-H), 6.90 (t, J ¼ 7.6 Hz, 1H, 4.2. Antioxidant activity
indole-C5-H), 5.90 (dd, J ¼ 12.5, 5.7 Hz, 1H, pyrazoline-C5-H), 3.94
(dd, J ¼ 18.5, 12.7 Hz, 1H, pyrazoline-C4-H), 3.26 (m, 1H, pyrazoline- 4.2.1. DPPH method
C4-H), 2.24 (s, 3H, CH3CO). 13C-NMR (DMSO-d6, 75 MHz): 21.76 The DPPH scavenging activity of tested compounds was
(CH3CO), 48.52 (pyrazoline-C4), 53.49 (pyrazoline-C5), 111.68 measured according to the method described by Nahar et al. [39]
(indole-C3), 111.87 (indole-C7), 117.81 (indole-C4), 119.29 (indole- with some modifications. Briefly, 100 mL of different concentrations
C5), 121.45 (indole-C6), 123.91 (pyridine-C3), 125.16 (pyridine-C5), of the tested compounds (12.5, 25, 50, 100 and 200 mg/ml) were
127.21 (indole-C2), 127.79 (indole-C3a), 128.61 (2-phenyl-C2,6), pipetted into a 96-well flat-bottomed plate. Next, 100 mL of 100 mM
128.92 (2-phenyl-C3,5), 132.42 (2-phenyl-C4), 133.64 (2-phenyl-C1), DPPH methanolic solution were added to each well and the plate
134.99 (indole-C7a), 136.26 (pyridine-C4), 147.57 (pyridine-C2), was incubated protected from light at room temperature for
150.82 (pyrazol-C3), 152.18 (pyridine-C6), 167.81 (CO). MS EIMS: 30 min. The absorbance of the solution was measured at l 517 nm
(calcd) m/z 380.16 (found) 380.33 (Mþ). Anal. Calcd for [41]. Ascorbic acid and DMSO were used as the positive control and
C24H20N4O: C,75.77; H, 5.30; N,14.73. Found: C,76.01; H, 5.54; N, blank respectively. The percentage of DPPH scavenging activity was
14.94. calculated according to the following equation:
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 603

% of DPPH scavenging ¼ [(A blank eA sample) / A blank]  100


Dose-response curve was plotted between % inhibition of the
Where A blank is the absorbance of the control reaction (containing rate of increase of absorbance at 550 nm due to the reduction of
all reagents except the test compound), and A sample is the NBT to NBT-diformazan by the superoxide radical and the drug
absorbance of the test sample. concentration. The non-linear dose-response curve was used for
Dose-response curve was plotted between %free radical scav- calculating drug concentration showing 50% free radical inhibition
enging activity and the drug concentration. Linear regression activity (IC50).
analysis was carried out for calculating drug concentration showing
50% free radical inhibition activity (IC50). All tests were undertaken 4.3. In-vitro lipoxygenase inhibition activity
on three replicates and the results were averaged.
The assay was performed using Abnova Lipoxygenase inhibitor
4.2.2. NO scavenging method screening assay kit (Catalog#KA1329). In this method [44], 90 mL of
The nitric oxide (NO) scavenging activity of tested compounds soybean LOX was pipetted into a 96-well flat-bottomed plate. Next,
was measured according to the method described by Ho et al. [42]. 10 mL of test compound at concentrations (2.5 mM, 5.0 mM and
In this method 50 mL of different concentrations of the tested 10 mM) dissolved in DMSO were added to each well. The reaction
compounds (12.5, 25, 50,100 and 200 mg/ml) were pipetted into a 96- was initiated by adding 10 mL substrate (arachidonic acid or linoleic
well flat-bottomed plate. Next, 50 mL of 10 mM sodium nitroprusside acid) and the plate was placed on a shaker for at least five minutes.
dissolved in Phosphate buffered saline PBS (pH 7.4) were added to Finally, an equal volume 100 mL of chromogen was added to each
each well and the plate was incubated at room temperature for well to stop enzyme catalysis and develop the reaction. The
90 min. Finally, an equal volume of Griess reagent (1% of sulphani- absorbance of the solution was measured at l 490e500 nm. Quer-
lamide and 0.1% of naphthyl ethylene diamine in 2.5% H3PO3) was cetin, assay buffer and DMSO were used as the positive control,
added to each well to measure the nitrite content [41]. A pink-colored blank and 100% initial activity respectively.
chromophore was formed in diffused light. The absorbance of the The percentage inhibition was calculated according to the
solution was measured at l 546 nm. Ascorbic acid and DMSO were following equation:
used as the positive control and blank respectively. All tests were
undertaken on three replicates and the results were averaged. % inhibition ¼ [(IA -A sample) / IA]  100
The percentage of NO scavenging activity was calculated ac-
cording to the following equation: Where IA is the 100% initial activity (containing all reagents except
the test compound), and A sample is the absorbance of the test
% of NO scavenging ¼ [(A blank -A sample) / A blank]  100 sample.
Dose-response curve was plotted between % inhibition and the
Where A blank is the absorbance of the control reaction (containing drug concentration. The non-linear dose-response curve was used
all reagents except the test compound), and A sample is the for calculating drug concentration showing 50% enzyme inhibition
absorbance of the test sample. activity (IC50).
Dose-response curve was plotted between % free radical scav-
enging activity and the drug concentration. Linear regression 4.4. Docking study
analysis was carried out for calculating drug concentration showing
50% free radical inhibition activity (IC50). All docking studies were performed using ‘Internal Coordinate
Mechanics (MOLSOFT ICM 3.4e8C)’. ICM docking is probably one of
the most accurate and predictive tools of binding geometry today
4.2.3. SOD assay [45e48].
The assay was performed at room temperature using superoxide
Dismutase assay Kit (Catalog#7500-100-k). In this method, in a
4.4.1. Preparation of small molecule
disposable cuvette, the following component were added in order:
Regarding the synthesized compounds (4b), (4c), (5b) and (6c)
60 mL of reaction buffer, 7.5 mL xanthine solution, 30 mL of Nitroblue
are to be docked as 15-LOX inhibitors; ChemDraw 3D structures
Tetrazolium (NBT) solution, test compound at concentrations
were constructed using Chem 3D ultra 15.1 software [Molecular
(2.5 mM, 5.0 mM and 10 mM), 10 mL Xanthine Oxidase (XOD) solution,
Modeling and Analysis; Cambridge Soft Corporation, USA (2015)],
read the absorbance at 550 nm, start a timer or stopwatch and record
and then they were energetically minimized by using MOPAC
the absorbance reading every 30 s for 5 min [43]. The first time will
(semi-empirical quantum mechanics), Jop Type with 100 iterations
be at 30 s and the final time point will be at 5 min 30 secconds.
and minimum RMS gradient of 0.01, and saved as MDL MolFile
(*.mol).
1. Determine the rate of increase in absorbance units (A) per
minute for the negative control and for the test sample(s):
4.4.2. Generation of enzyme structure
A550 @ 5 min:30 sec:  A550 @30 sec: The crystal structure of target protein (15-LOX) is a human
D A550n =minute ¼ isoform of lipoxygenase in complex with ligand substrate (arach-
5min
idonate) was retrieved from the Protein Data Bank (http://www.
rcsb.org/pdb/welcome.- do). All bound water, substrate and co-
2. Determine the % inhibition for the test sample(s): factors were removed from the protein. The amino acids of the

½ðDA550n =minuteÞnegative control  ðDA550n =minuteÞtest


%Inhibition ¼  100
ðDA550 =minuteÞnegative control
604 H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605

binding site where defined using data in pdbsum (http://www.ebi. lipoxygenase catalytic-domain-inhibitor complex identifies a substrate-
binding channel for catalysis, Structure 20 (2012) 1490e1497.
ac.uk/thoronton-srv/databases/pdbsum/).
[18] W. Minor, J. Steczko, B. Stec, Z. Otwinowski, J.T. Bolin, R. Walter, B. Axelrod,
Crystal structure of soybean lipoxygenase L-1 at 1.4 A resolution, Biochem-
istry 35 (1996) 10687e10701.
4.4.3. Docking using MOLSOFT ICM 3.4e8C program [19] S.T. Prigge, J.C. Boyington, M. Faig, K.S. Doctor, B.J. Gaffney, L.M. Amzel,
Structure and mechanism of lipoxygenases, Biochimie 79 (1997) 629e636.
1. Convert our PDB file into an ICM object: This conversion in- [20] E. Skrzypczak-Jankun, K. Zhou, N.P. McCabe, S.H. Selman, J. Jankun, Structure
of curcumin in complex with lipoxygenase and its significance in cancer, Int. J.
volves addition of hydrogen bonds, assignment of atoms types,
Mol. Med. 12 (2003) 17e24.
and charges from the residue templates. [21] M. Mahdavi, M.S. Shirazi, R. Taherkhani, M. Saeedi, E. Alipour, F.H. Moghadam,
2. To perform ICM small molecule docking: A. Moradi, H. Nadri, S. Emami, L. Firoozpour, A. Shafiee, A. Foroumadi, Syn-
thesis, biological evaluation and docking study of 3-aroyl-1-(4-
a) Setup docking project:
sulfamoylphenyl)thiourea derivatives as 15-lipoxygenase inhibitors, Eur. J.
1) Set project name: Med. Chem. 82 (2014) 308e313.
2) Setup the receptor: [22] M.B. Tehrani, S. Emami, M. Asadi, M. Saeedi, M. Mirzahekmati, S.M. Ebrahimi,
3) Review and adjust binding site: M. Mahdavi, H. Nadri, A. Moradi, F.H. Moghadam, S. Farzipour, M. Vosooghi,
A. Foroumadi, A. Shafiee, Imidazo[2,1-b]thiazole derivatives as new inhibitors
4) Make receptor maps: of 15-lipoxygenase, Eur. J. Med. Chem. 87 (2014) 759e764.
b) Start docking simulation: [23] M. Roussaki, K. Zelianaios, E. Kavetsou, S. Hamilakis, D. Hadjipavlou-Litina,
3. Display the result: C. Kontogiorgis, T. Liargkova, A. Detsi, Structural modifications of coumarin
derivatives: Determination of antioxidant and lipoxygenase (LOX) inhibitory
activity, Bioorg. Med. Chem. 22 (2014) 6586e6594.
ICM stochastic global optimization algorithm attempts to find [24] S. Dianat, S. Moghimi, M. Mahdavi, H. Nadri, A. Moradi, L. Firoozpour,
the global minimum of the energy function that include five grid S. Emami, A. Mouradzadegun, A. Shafiee, A. Foroumadi, Quinoline-based
imidazole-fused heterocycles as new inhibitors of 15-lipoxygenase, J. Enzym.
potentials describing interaction of the flexible ligand with the Inhib. Med. Chem. 31 (2016) 205e209.
receptor and internal conformational energy of the ligand, during [25] A. Saeed, S.U. Khan, P.A. Mahesar, P.A. Channar, G. Shabir, J. Iqbal, Substituted
this process a stack of alternative low energy conformations is (E)-2-(2-benzylidenehydrazinyl)-4-methylthiazole-5-carboxylates as dual
inhibitors of 15-lipoxygenase & carbonic anhydrase II: synthesis, biochemical
saved. The binding pocket of the substrate co-crystallized with the
evaluation and docking studies, Biochem. Biophy. Res. Commun. 482 (2017)
enzyme that was chosen for docking the inhibitors into it. 176e181.
[26] S. Ono, K. Okazaki, M. Sakurai, Y. Inoue, Density functional study of the radical
reactions of 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186): implication for
References the biological function of MCI-186 as a highly potent antioxidative radical
scavenger, J. Phys. Chem. 101 (1997) 3769e3775.
[1] A.R. Brash, Lipoxygenases: occurrence, functions, catalysis, and acquisition of [27] T. Taj, R.R. Kamble, S.B. Marganakop, Facile synthesis of pyrazoline derivatized
substrate, J. Biol. Chem. 274 (1999) 23679e23682. amides viasydnone ring cleavage, J. Heterocycl. Chem. 51 (2014) E323eE328.
[2] E. Pontiki, D. Hadjipavlou-Litina, Lipoxygenases (LOs): an heterogenous family [28] P.C. Jagadish, N. Soni, A. Verma, Design, synthesis, and in vitro antioxidant
of lipid peroxidizing enzymes implicated in cell differentiation, inflammation, activity of 1,3,5-trisubstituted-2-pyrazolines derivatives, J. Chem. 2013 (2013)
asthma, carcinogenesis, atherogenesis-an interesting target for the develop- 6.
ment of promising drugs, Curr. Enzym. Inhib. 1 (2005) 309e327. [29] S. Suzen, P. Bozkaya, T. Coban, D. Nebiogu, Investigation of the in vitro anti-
[3] J.S. Larsen, E.P. Acosta, Leukotriene-receptor antagonists and 5-lipoxygenase oxidant behaviour of some 2-phenylindole derivatives: discussion on possible
inhibitors in asthma, Ann. Pharmacother. 27 (1993) 898e903. antioxidant mechanisms and comparison with melatonin, J. Enzym. Inhib.
[4] G.P. Pidgeon, J. Lysaght, S. Krishnamoorthy, J.V. Reynolds, K. O'Byrne, D. Nie, Med. Chem. 21 (2006) 405e411.
K.V. Honn, Lipoxygenase metabolism: roles in tumor progression and survival, [30] S. Suzen, N. Das-Evcimen, P. Varol, M. Sarıkaya, Preliminary evaluation of rat
Canc. Metastasis Rev. 26 (2007) 503e524. kidney aldose reductase inhibitory activity of 2-phenylindole derivatives:
[5] L. Zhao, C.D. Funk, Lipoxygenase pathways in atherogenesis, Trends Car- affiliation to antioxidant activity, Med. Chem. Res. 16 (2007) 112e118.
diovasc. Med. 14 (2004) 191e195. [31] M.V. Reddy, V.K. Billa, V.R. Pallela, M.R. Mallireddigari, R. Boominathan,
[6] D. Pratico  , V. Zhukareva, Y. Yao, K. Uryu, C.D. Funk, J.A. Lawson, J.L. Gabriel, E.P. Reddy, Design, synthesis, and biological evaluation of 1-(4-
J.Q. Trojanowski, V.M.Y. Lee, 12/15-Lipoxygenase is increased in Alzheimer's sulfamylphenyl)-3-trifluoromethyl-5-indolyl pyrazolines as cyclooxygenase-
disease: possible involvement in brain oxidative stress, Am. J. Pathol. 164 2 (COX-2) and lipoxygenase (LOX) inhibitors, Bioorg. Med. Chem. 16 (2008)
(2004) 1655e1662. 3907e3916.
[7] D. Nie, K.V. Honn, Cyclooxygenase, lipoxygenase and tumor angiogenesis, [32] A.I. Vogel, B.S. Furniss, A.I. Vogel, Vogel's Textbook of Practical Organic
Cellular and Molecular Life Sciences CMLS 59 (2002) 799e807. Chemistry, fifth ed., Longman Scientific & Technical; Wiley, London New York,
[8] U. Kelavkar, W. Glasgow, T.E. Eling, The effect of 15-lipoxygenase-1 expression 1989.
on cancer cells, Curr. Urol. Rep. 3 (2002) 207e214. [33] R.C. Blume, H.G. Lindwall, 2-phenylindole-3-aldehyde and certain of its
[9] U.P. Kelavkar, C. Cohen, H. Kamitani, T.E. Eling, K.F. Badr, Concordant induction condensation products, J. Org. Chem. 11 (1946) 185e188.
of 15-lipoxygenase-1 and mutant p53 expression in human prostate adeno- [34] A. Bourlot, E. Desarbre, J. Merour, A convenient synthesis of 1, 2-dihydro-3H-
carcinoma: correlation with Gleason staging, Carcinogenesis 21 (2000) indol-3-ones and 1, 2-dihydro-2H-indol-2-ones by Baeyer-Villiger oxidation,
1777e1787. Synthesis 1994 (1994) 411e416.
[10] C. Sultana, Y. Shen, V. Rattan, V.K. Kalra, Lipoxygenase metabolites induced [35] V.D. Joshi, M.D. Kshirsagar, S. Singhal, Synthesis and antimicrobial activities of
expression of adhesion molecules and transendothelial migration of various pyrazolines from chalcones, International J. ChemTech Res. 4 (2012)
monocyte-like HL-60 cells is linked to protein kinase C activation, J. Cell. 971e975.
Physiol. 167 (1996) 477e487. [36] M.A. Abdel-Sayed, S.M. Bayomi, M.A. El-Sherbeny, N.I. Abdel-Aziz, K.E. ElTahir,
[11] B.N. Setty, M.H. Werner, Y.A. Hannun, M.J. Stuart, 15-Hydroxyeicosatetraenoic G.S. Shehatou, A.A. Abdel-Aziz, Synthesis, anti-inflammatory, analgesic, COX-
acid-mediated potentiation of thrombin-induced platelet functions occurs via 1/2 inhibition activities and molecular docking study of pyrazoline de-
enhanced production of phosphoinositide-derived second messengers-sn- rivatives, Bioorg. Med. Chem. 24 (2016) 2032e2042.
1,2-diacylglycerol and inositol-1,4,5-trisphosphate, Blood 80 (1992) [37] P.O. Patil, S.B. Bari, Synthesis and antidepressant activity of some new 5-(1H-
2765e2773. Indol-3-yl)-3-(substituted aryl)-4,5-dihydroisoxazoline derivatives, J. Chem.
[12] T. Schewe, 15-lipoxygenase-1: a prooxidant enzyme, Biol. Chem. 383 (2002) 2013 (2013) 7.
365e374. [38] S. Viveka, Dinesha, P. Shama, G.K. Nagaraja, S. Ballav, S. Kerkar, Design and
[13] J.C. Boyington, B.J. Gaffney, L.M. Amzel, The three-dimensional structure of an synthesis of some new pyrazolyl-pyrazolines as potential anti-inflammatory,
arachidonic acid 15-lipoxygenase, Science 260 (1993) 1482e1486. analgesic and antibacterial agents, Eur. J. Med. Chem. 101 (2015) 442e451.
[14] A. Garreta, S.P. Val-Moraes, Q. Garcia-Fernandez, M. Busquets, C. Juan, [39] L. Nahar, W.R. Russell, M. Middleton, M. Shoeb, S.D. Sarker, Antioxidant
A. Oliver, A. Ortiz, B.J. Gaffney, I. Fita, A. Manresa, X. Carpena, Structure and phenylacetic acid derivatives from the seeds of Ilex aquifolium, Acta Phar-
interaction with phospholipids of a prokaryotic lipoxygenase from Pseudo- maceutica 55 (2005) 187e193.
monas aeruginosa, Faseb. J. 27 (2013) 4811e4821. [40] M.J. Kobe, D.B. Neau, C.E. Mitchell, S.G. Bartlett, M.E. Newcomer, The structure
[15] N.C. Gilbert, S.G. Bartlett, M.T. Waight, D.B. Neau, W.E. Boeglin, A.R. Brash, of human 15-lipoxygenase-2 with a substrate mimic, J. Biol. Chem. 289 (2014)
M.E. Newcomer, The structure of human 5-lipoxygenase, Science 331 (2011) 8562e8569.
217e219. [41] A. Padmaja, C. Rajasekhar, A. Muralikrishna, V. Padmavathi, Synthesis and
[16] S.A. Gillmor, A. Villasenor, R. Fletterick, E. Sigal, M.F. Browner, The structure of antioxidant activity of oxazolyl/thiazolylsulfonylmethyl pyrazoles and iso-
mammalian 15-lipoxygenase reveals similarity to the lipases and the de- xazoles, Eur. J. Med. Chem. 46 (2011) 5034e5038.
terminants of substrate specificity, Nat. Struct. Biol. 4 (1997) 1003e1009. [42] S.-C. Ho, Y.-L. Tang, S.-M. Lin, Y.-F. Liew, Evaluation of peroxynitrite-
[17] S. Xu, T.C. Mueser, L.J. Marnett, M.O. Funk Jr., Crystal structure of 12- scavenging capacities of several commonly used fresh spices, Food Chem.
H.S. ElBordiny et al. / European Journal of Medicinal Chemistry 145 (2018) 594e605 605

119 (2010) 1102e1107. Comput. 11 (2015) 2167e2186.


[43] D. Hadjipavlou-Litina, A. Samadi, M. Unzeta, J. Marco-Contelles, Analysis of the [46] A. Guspiel, J. Ziemska, A. Czescik, R. Kawecki, J. Solecka, Intracellular antiox-
antioxidant properties of differently substituted 2- and 3-indolyl carbohy- idant activity of a streptomyces Sp. 8812 secondary metabolite, 6,7-
drazides and related derivatives, Eur. J. Med. Chem. 63 (2013) 670e674. dihydroxy-3,4-dihydroisoqino- line-3-carboxylic acid, and its synthetic de-
[44] P. Plackova, M. Sala, M. Smidkova, M. Dejmek, H. Hrebabecky, R. Nencka, rivatives, Acta Poloniae Pharmaceutica 73 (2016) 645e651.
H.J. Thibaut, J. Neyts, H. Mertlikova-Kaiserova, 9-Norbornyl-6-chloropurine [47] T. James, M.L. Hsieh, L. Knipling, D. Hinton, Determining the architecture of a
(NCP) induces cell death through GSH depletion-associated ER stress and protein-DNA complex by combining FeBABE cleavage analyses, 3-D printed
mitochondrial dysfunction, Free Radical Biology & Med. 97 (2016) 223e235. structures, and the ICM Molsoft Program, Meth. Mol. Biol. 1334 (2015) 29e40.
[45] Y.A. Arnautova, R. Abagyan, M. Totrov, All-atom internal coordinate me- [48] R. Abagyan, M. Totrov, High-throughput docking for lead generation, Curr.
chanics (ICM) force field for hexopyranoses and glycoproteins, J. Chem. Theor. Opin. Chem. Biol. 5 (2001) 375e382.

Anda mungkin juga menyukai