Accepted Manuscript Discovery of novel Methylsulfonyl phenyl derivatives as potent human Cyclooxygenase-2 inhibitors with effective anticonvulsant action: Design, synthesis, in-silico, in-vitro and in-vivo evaluation Chandra Bhushan Mishra, Shikha Kumari, Amresh Prakash, Rajesh Yadav, Ankit Kumar Tiwari, Preeti Pandey, Manisha Tiwari PII:
S0223-5234(18)30334-9
DOI:
10.1016/j.ejmech.2018.04.007
Reference:
EJMECH 10356
To appear in:
European Journal of Medicinal Chemistry
Received Date: 23 November 2017 Revised Date:
28 February 2018
Accepted Date: 2 April 2018
Please cite this article as: C.B. Mishra, S. Kumari, A. Prakash, R. Yadav, A.K. Tiwari, P. Pandey, M. Tiwari, Discovery of novel Methylsulfonyl phenyl derivatives as potent human Cyclooxygenase-2 inhibitors with effective anticonvulsant action: Design, synthesis, in-silico, in-vitro and in-vivo evaluation, European Journal of Medicinal Chemistry (2018), doi: 10.1016/j.ejmech.2018.04.007. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT 1
Discovery of Novel Methylsulfonyl Phenyl Derivatives as Potent Human
2
Cyclooxygenase-2 Inhibitors with Effective Anticonvulsant Action: Design,
3
Synthesis, In-silico, In-vitro and In-vivo evaluation
4
Chandra Bhushan Mishra1, Shikha Kumari1, Amresh Prakash2, Rajesh Yadav1, Ankit Kumar Tiwari1,
5
Preeti Pandey2, Manisha Tiwari1*
6
1
7
Delhi, Delhi-110007, India.
8
2
9
India.
RI PT
1
SC
Bio-Organic Chemistry Laboratory, Dr. B. R. Ambedkar Center for Biomedical Research, University of
M AN U
School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi,
10
*Corresponding Author
11
[email protected] (Dr. Manisha Tiwari)
12
Abstract
14
A novel series of methylsulfonyl phenyl derivatives has been designed and synthesized to
15
evaluate their COX-2 inhibitory activity along with anti-convulsant potential. In-vitro evaluation
16
revealed that two compounds MTL-1 and MTL-2 appeared as most potent and selective COX-2
17
inhibitors in the entire series. Anti-convulsant activity of both potent COX-2 inhibitors was
18
assessed in sc-PTZ induced seizure test and MTL-1 excellently protected animals against PTZ
19
induced seizure at the dose of 30 mg/kg. MTL-1 also indicates long duration of action in time
20
course study and displayed significant seizure protection up to 6h of drug administration.
21
Further, the anti-epileptogenic effect of MTL-1 has been examined in PTZ induced chronic
22
model of epilepsy. The results indicated that MTL-1 had a significant anti-epileptogenic effect in
23
PTZ kindled rats as compared to Etoricoxib (ETX) and PTZ alone treated group. Additionally,
24
MTL-1 successfully improved cognition deficit in PTZ kindled rats, which were confirmed by
25
social recognition, novel object recognition and light-dark chamber tests. Moreover, molecular
26
docking and molecular simulation (MD simulation) studies were also performed to elucidate the
AC C
EP
TE D
13
1
ACCEPTED MANUSCRIPT 2
interaction of MTL-1 with the active site of COX-2 and results showed that MTL-1 suitably
28
binds within active site of COX-2. To investigate the safety profile of MTL-1, a sub-acute
29
toxicity study was also performed and MTL-1 emerged as a new non-toxic chemical entity.
30
Thus, the present investigation discovered a potent and safe COX-2 inhibitor, which is endowed
31
with an effective anti-epileptic action.
32
1. Introduction
33
Epilepsy is a common and complex neurological disorder which affects approximately 65
34
million people worldwide [1]. The disease is symptomized by recurrent and spontaneous seizures
35
and caused due to uncontrolled synchronous and rhythmic firing of central nervous system (CNS)
36
neurons which leads to sensory and motor dysfunctions [2]. Clinical evidences concluded that
37
epilepsy is a group of more than 30 types of epileptic syndromes containing 15 different types of
38
seizures [3]. The clinical hallmark of epilepsy is the continual emergence of sudden and
39
unexpected seizures [4]. It is well studied that numerous neurotransmitters or neuromodulators
40
have been shown to play a crucial role in the development of epilepsy [5]. Cyclooxygenase
41
(COX) is also known as prostaglandin-endoperoxide synthase, exist in two isoform, COX-1 as
42
well as COX-2 and is the key enzyme that converts arachidonic acid to prostaglandin, which
43
impart crucial role in signaling and inflammation [6]. COX-1 is expressed in many tissues and
44
prostaglandins (PGs) produced by COX-1 mediate the “housekeeping” functions like platelet
45
aggregation, renal blood flow regulation and cytoprotection of gastric mucosa. On the other side,
46
COX-2 is not present in most normal tissues, but its expression is induced by various stimuli
47
such as lipopolysaccharides, proinflammatory cytokines (IL1b, TNFα), growth factors (fibroblast
48
growth factor, platelet-derived growth factor, epidermal growth factor), mitogens and oncogenes
49
(phorbol esters), hormones (luteinizing hormone) and disorders of water-electrolyte hemostasis,
50
resulting in increased synthesis of PGs in inflamed and neoplastic tissues.
51
In the brain, COX-2 isoform is predominantly expressed in glutamatergic neurons, particularly
52
within cortex and hippocampus, the main area prone to onset of epileptic seizures. It was
53
observed that expression of COX-2 is noticeably enhanced in the pyramidal cells of the
54
hippocampus of kindled animals as compared to control [7]. Additionally, elevated expression of
55
COX-2 and brain prostaglandin E2 (PGE2) levels was also noticed in lithium chloride and
AC C
EP
TE D
M AN U
SC
RI PT
27
2
ACCEPTED MANUSCRIPT 3
tacrine induced status epilepticus seizure models [8]. Dhir et al. studied the possible mechanism
57
and suggested that COX-2 inhibitors protect PTZ induced seizure probably through the
58
GABA/Benzodiazepine receptor mechanism [9]. Till date, several selective COX-2 inhibitors
59
have been developed and some of them such as Celecoxib, Rofecoxib, Valdecoxib and
60
Etoricoxib are being successfully used clinically [10]. These COX inhibitors also have shown
61
promising anticonvulsant activity in pentylenetetrazole (PTZ) induced acute and chronic model
62
of epilepsy [7]. However, some of them have low selectivity towards COX-2, poor
63
pharmacokinetic properties and severe liver toxicity after long time use [11]. Therefore, it is
64
mandatory to discover more potent and very selective COX-2 inhibitor with minimum toxicity.
65
Methylsulfonyl (−SO2CH3) containing molecules have a successful history to discover potent
66
COX-2 inhibitors and numerous clinically approved COX-2 inhibitors such as Rofecoxib,
67
Etoricoxib and SC57666 (Fig.1) are examples which contain this functional group [10, 12]. By
68
installing methylsulfonyl on the benzene ring numerous potent COX-2 inhibitors have been
69
synthesized and tested for their COX-2 inhibitory activity [10]. Additionally, urea and thiourea
70
groups appeared to be versatile, functional groups and widely used to develop various potent
71
biologically active molecules, including COX-2 inhibitors as well as anti-convulsants [13, 14].
72
Keeping these valuable properties of methylsulfonyl benzene, urea and thiourea functional
73
groups
74
(phenylcarbamothioyl)hydrazinecarboxamide derivatives where, methylsulfonyl benzene is
75
linked with substituted phenyl by urea and thiourea linker. Synthesized derivatives were
76
screened for their COX-2 inhibitory potential and their selectivity was assessed over COX-1
77
isoform. Further, the most potent and selective COX-2 inhibitors MTL-1 and MTL-2 were
78
selected for examining their anticonvulsant potential. Firstly, MTL-1 and MTL-2 were assessed
79
for its anti-convulsant activity against sc-PTZ induced seizure mice model. A time course study
80
was also performed to evaluate time depended anticonvulsant activity of MTL-1. Additionally,
81
MTL-1 was evaluated against PTZ induced chronic model (kindled model) of epilepsy to
82
confirm its anti-epileptogenic potential. Moreover, the toxicity profile of MTL-1 has been
83
observed in sub-acute toxicity rat model. Furthermore, cognition enhancing ability of MTL-1 has
84
been evaluated in social recognition, object recognition and light-dark test. The molecular
aimed
to
design
a
novel
series
of
N-(4-(methylsulfonyl)phenyl)-2-
AC C
EP
we
TE D
M AN U
SC
RI PT
56
3
ACCEPTED MANUSCRIPT 4
85
interaction of MTL-1 with COX-2 has been investigated by performing molecular docking and
86
molecular dynamic simulation (MD simulation) studies. 2. Result and discussion
RI PT
87 88 89 90 91
2.1. Synthesis Synthesis of the designed compounds 4-14 have been synthesized according to scheme 1. Briefly, an equimolar amount of 4-(methylsulfonyl) aniline 1 reacted with an equimolar amount
93
of phenylchloroformate which produced carbamate derivative 2. Further, carbamate derivative 2
94
was refluxed with hydrazine hydrate to give key intermediate N-(4-(methylsulfonyl) phenyl)
95
hydrazinecarboxamide 3. Finally, target compounds 4-14 were obtained by reacting key
96
intermediate 3 with substituted isothiocyanates in high yields. Synthesized compounds were fully
97
characterized by NMR spectroscopy, mass spectroscopy and elemental analysis.
98 99
R-Group Phenyl 4-Methylphenyl 2, 6 Dimethylphenyl 4-Methoxyphenyl 4-Fluorophenyl 2, 6 Difluorophenyl
AC C
EP
Compound Number 4 5 6 7 8 9
TE D
M AN U
SC
92
Compound Number 10 11 12 13 14
R-Group 4-Chlorophenyl 2, 3 Dichlorophenyl Benzyl Phenylethyl Ethyl
100 101 102 103 104
Scheme1: Reagent & conditions: A. Phenyl chloroformate, dryTHF, pyridine, RT, 14h; B. Hydrazine hydrate, 1, 4-dioxane, reflux, 6h. C, substituted isothiocyanate, dry ACN: ethanol, reflux, 8-10h
105
2.2. In-vitro COX inhibition study and Structure activity relationship (SAR) analysis
4
ACCEPTED MANUSCRIPT 5
Results of in-vitro COX inhibition assay of synthesized compounds 4-14 have been shown in
107
table-1. SAR investigation revealed that all derivatives have shown selective activity against
108
COX-2 isoform and these compounds appeared inactive for COX-1 isoform at the concentration
109
of 10 µM. A compound containing unsubstituted phenyl ring (compound 4) showed the medium
110
class of inhibitory activity towards COX-2 (IC50 = 12.80 µM). Installation of an electron
111
donating group like methyl group on the phenyl ring (MTL-1) profoundly increased inhibitory
112
activity towards COX-2 and shown an IC50 value 2.15µM. Further, substitution with dimethyl
113
group on the phenyl ring (MTL-2) also showed similar type of inhibition. Next, the 4- methoxy
114
group substitution on the phenyl ring (Compound 7) decreased the inhibitory activity against
115
COX-2 as compared to methyl substitution and this derivative displayed an IC50 of 7.15 µM.
116
Substitution with a strong electronegative groups like fluoro (Compound 8) provided medium
117
range of inhibition (IC50 = 12.49 µM). However, di-fluoro substitution (Compound 9) yielded a
118
more potent inhibitor of COX-2 (IC50 = 6.42 µM) as compared to mono fluoro substitution.
119
Chloro substitution (Compound 10) on phenyl ring provided active inhibitor for COX-2 isoform
120
with an IC50 of 5.32 µM. However, 2, 3 dichlorobenzene (compound 11), benzyl (compound 12),
121
phenylethyl (compound 13) and ethyl derivative (compound 14) also did not show satisfactory
122
inhibitory activity against COX-2.
123
Thus, the SAR study clearly indicates that an electron donating group containing phenyl
124
derivatives appear to be potent derivatives and especially 4-methyl substitution on the phenyl
125
ring (MTL-1) appeared worthy, which displayed most potent inhibitor in the entire series.
126 127
Table1 In vitro inhibition data of compounds 4-14 against COX-1 and COX-2
EP
TE D
M AN U
SC
RI PT
106
AC C
Compounds 4 5 (MTL-1) 6 (MTL-2) 7 8 9 10 11 12 13
% inhibition (10 µM) COX-1 COX-2 b n.a 50 n.ab 75 b n.a 64 n.ab 62 b n.a 54 n.ab 62 b n.a 70 n.ab 46 b n.a 21 n.ab 22
IC50(µM)a COX-2 12.80 2.15 3.14 7.15 12.49 6.42 5.32 n.dc n.dc n.dc 5
ACCEPTED MANUSCRIPT 6
14 Etoricoxibd[15]
n.ab -
n.dc 1.1
32 -
128 129 130 131 132 133 134 135
2.3. Subcutaneous Pentylenetetrazole (sc-PTZ) seizure test
136
Pentyleneterazole (PTZ) is a well-known chemo-convulsant and binds to the gamma-amino
137
butyric acid ‘A’ subtype (GABA-A) receptor in the brain [16]. The sc-PTZ seizure test is well
138
documented test to identify the anticonvulsant potential of drug candidates in pre-clinical stage
139
of anti-epileptic drug discovery. Most potent COX-2 inhibitors MTL-1 and MTL-2, along with
140
two standards Etoricoxib (selective COX-2 inhibitor) and Sodium Valproate (potent anti-
141
epileptic drug) were evaluated in sc-PTZ induced acute epilepsy mice model (Table 2). Results
142
have shown that MTL-1 prominently inhibited sc-PTZ induced convulsions at the dose of 30
143
mg/kg and displayed 83% protection in both time intervals (0.5 and 4h). Additionally, MTL-1
144
showed similar protection activity at a dose of 300 mg/kg. However, at the dose of 100 mg/kg
145
protection percentage slightly reduced at 0.5h time interval. Another COX-2 inhibitor, MTL-2
146
did not displayed promising seizure protection against sc-PTZ induced seizures and only at high
147
dose (300 mg/kg) it showed 33 and 50% protection at 0.5 and 4h time intervals, respectively.
148
Standard COX-2 inhibitor Etoricoxib showed satisfactory protection from seizure and displayed
149
67 % protection at both time intervals. However, standard anti-epileptic drug sodium valproate at
150
300 mg/kg has shown 100 and 50% protection at 0.5 and 4h time intervals, correspondingly.
151
Thus, these observations point out that MTL-1 exhibited excellent protection in sc-PTZ induced
152
seizure model as compared to standard drug Etoricoxib. In quantitative anticonvulsant studies,
153
MTL-1 has also shown an ED50 value of 11.32 mg/kg in this model.
155
EP
TE D
M AN U
SC
RI PT
IC50 values, means of three independent experiments, represent the concentration required produce 50% enzyme inhibition. b Not active, no inhibition was found at 10 µM of test compound. c Not determined. d value taken from reference 15.
AC C
154
a
156 157
6
ACCEPTED MANUSCRIPT 7
158
Table 2: In-vivo anticonvulsant screening of MTL-1, MTL-2, Etoricoxib and Sodium Valproate
159
4hb
Compounds
161
(Dose in mg/kg)
P/Ta
% protection
162
MTL-1(30)
5/6
83
5/6
163
MTL-1(100)
4/6
67
5/6
MTL-1 (300)
5/6
83
5/6
MTL-2 (30)
1/6
17
MTL-2 (100)
2/6
33
MTL-2 (300)
2/6
Etoricoxib (1)
3/6
Etoricoxib (5)
4/6
Etoricoxib (10)
3/6
P/Ta
a
6/6
83
1/6
17
2/6
33
33
3/6
50
50
3/6
50
67
4/6
67
50
3/6
50
100
3/6
50
M AN U
TE D
Sodium Valporate (300)
83
P/T denotes number of protected animals verses total number of animals tested. Time intervals after drug administration.
EP
b
2.4. Time course anti-convulsant study To investigate longer duration of action, a time course study of MTL-1 has been performed at the
AC C
165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187
83
SC
164
% protection
RI PT
0.5hb
160
188
dose of 30mg/kg in sc-PTZ induced seizure model and seizure score was documented up to 6h
189
after drug administration (Table. 3). Results indicated that MTL-1 has efficient capability to
190
protect seizures with longer duration of action. MTL-1 has shown 83-67 % seizure protection up
191
to 2h of drug administration. Further, the efficacy of MTL-1 was enhanced and percentage
192
protection reached up to 83 % after 3-4 h of drug treatment. It was observed that MTL-1 restored
193
protection up to 67 % after 6h of drug administration, which indicates its longer duration of 7
ACCEPTED MANUSCRIPT 8
action against seizure episodes. Hence, with this investigation MTL-1 appeared as potent anti-
195
convulsant agent that endowed with longer duration action.
196
Table. 3 Time course evaluation of MTL-1 in sc-PTZ seizure test with six Swiss Albino mice
197
each time
Percentage seizure protection 83 67 67 83 83 67
SC
0.5 1 2 3 4 6
Protected/ Number of animals used 5/6 4/6 4/6 5/6 5/6 4/6
M AN U
Time interval (h)
RI PT
194
198 199 200 201 202
2.5. Evaluation of MTL-1 in PTZ induced chronic model of epilepsy (kindling model)
203
epileptogenic processes in the brain. PTZ is a potent chemo-convulsant and it increases activities
204
in major epileptogenic centers of the forebrain like the amygdala and the piriform cortex [17].
205
Kindling is described by repetitive administration of an initial sub-convulsive electrical or
206
chemical stimulus, resulting in progressive multiplication of seizure activity, which culminates in
207
generalized seizures that resembling human epileptic syndrome [18]. In the present study, PTZ
208
was used to induce kindling in adult male Wistar rats to visualize anti-epileptogenic effect of
209
compound MTL-1. A study conducted by Katyal J. et al. have shown that Etoricoxib (ETX)
210
exerted anti-epileptogenic effect at low dose in Pentylenetetrazole-kindled rats [19]. Etoricoxib is a
211
selective COX-2 inhibitor having anti-epileptiogenic potency; therefore ETX was chosen as a
212
reference drug to compare the potency of test compound MTL-1. A total number of four groups
213
(control; PTZ; PTZ+ ETX and PTZ+ MTL-1) were constituted. Chronic treatment for a period of
214
28 days with sub-convulsive dose of PTZ (40 mg/kg) induced successful kindling in rats of all
215
groups. Seizures were visualized manually by keeping the rats in individual cages after injecting
216
PTZ for the period of 45 minutes. The resultant behavioral seizure activity was categorized
217
according to revised Racine scale. Control group animals received only vehicle throughout the
218
study and did not show any seizure like activity. The animals of PTZ group have initially
aThe
test compound MTL-1 was administered i.p. to Swiss Albino mice (6 animals in each group).
AC C
EP
TE D
Animal kindling models are the most commonly used models which are designed to study
8
ACCEPTED MANUSCRIPT 9
developed behavioral seizure patterns which were later recognized as secondary generalized
220
seizures similar to the chronic epileptic disorder.
221
It was clearly witnessed that animals belongs to PTZ group developed generalized tonic-clonic
222
seizures gradually and significant difference (p<0.0001) was seen, when compared to control
223
group animals (Fig. 2). In contrast, pretreatment with ETX in PTZ+ ETX group showed decreased
224
(p<0.05) mean kindling score in comparison with PTZ group. Mortality was not observed in any
225
group except PTZ group, in which two animals died due to severe tonic-clonic seizures followed
226
by high pitch vocalization. However, more noticeable anti-seizure effect was shown by animals
227
treated with test compound MTL-1 at the dose of 30 mg/kg. Our results have shown that MTL-1
228
treated rats displayed a seizure score 1.5±0.98, 1.75±1.11, 1.79±0.93 and 2.12±1.23 at first,
229
second, third and fourth week respectively as compared to ETX treated rats which were shown a
230
seizure score 2.33±1.43, 2.42±1.11, 2.79±1.38 and 3.12±1.33 at first, second, third and fourth
231
week, respectively.
232
However, PTZ alone treated rats showed 2.70±1.27, 3.79±1.0, 4.54±0.83 and 5.08±0.58 seizure
233
scores at first, second, third and fourth week respectively. Thus, these results clearly indicate that
234
MTL-1 excellently reduced seizure scores as compared to ETX and PTZ alone treated rats which
235
indicate strong anti-epileptic effect of MTL-1. Overall kindling results displayed that animals
236
treated with compound MTL-1 pointedly (p<0.0001) attenuated seizure severity when compared to
237
PTZ-kindled animals.
238
2.6. Impact of MTL-1 on cognition deficit kindled rats
239
Epilepsy affects the person in several aspects of life, including physical, mental and cognitive
240
functions. Cognition deficit includes loss of intelligence, thinking, remembering and
241
understanding [20]. According to a survey conducted by International Bureau of Epilepsy stated
242
that 44% of patients with epilepsy complained of difficult learning, and 45% of slowness in
243
thinking ability [21]. Therefore, anticonvulsant agents which have capability to improve
244
cognition impairment along with anti-seizure activity may perform excellent management in
245
epilepsy. Cognition enhancement potential of MTL-1 has been assessed in PTZ induced chronic
246
model of epilepsy by performing well established social recognition, object recognition and
247
light-dark test.
248
2.6.1. Social recognition test
AC C
EP
TE D
M AN U
SC
RI PT
219
9
ACCEPTED MANUSCRIPT 10
A well recognized social recognition test has been accomplished to confirm cognition enhancing
250
ability of MTL-1 in PTZ kindled rats [22]. Our results indicate that MTL-1 significantly reduced
251
the T2 (second interaction trial) /T1 (first interaction time) ratio as compared to the vehicle
252
which reflects the strengthening of short-term memory by MTL-1 (Fig. 3). The ratio of T2/T1 in
253
MTL-1 group was 0.561± 0.139 as compared to PTZ group which has displayed T2/T1= 0.928 ±
254
0.234. Thus, MTL-1 successfully ameliorated cognition deficit induced by chronic PTZ
255
treatment, which evidently supports cognition enhancer potential of potent anticonvulsant agent
256
MTL-1.
257
2.6.2. Object recognition test
258
An object recognition test is the most commonly used behavioral test and reflected as the crucial
259
assessment to observe cognitive enhancer ability of pharmacological drugs [23]. In this behavioral
260
test MTL-1 treated group also appeared as satisfactory cognition enhancer agents along with its
261
anti-seizure effect (Fig. 4A and B). Animals treated with MTL-1 showed less percentage
262
exploration preference for a similar object (3.9%) while, this percentage was increased for novel
263
object (5.7%) as similar to control group animals which indicate MTL-1 significantly improved
264
cognition impairment induced by PTZ.
265
2.6.3. Light-dark test
266
The light-dark test is widely used in pharmacology to assess cognition deficit/anxiety like behavior
267
in animals and based on the native aversion of rodents to brightly illuminated areas [24]. The
268
results of this experiment again advocate cognition boosting aptitude of MTL-1 and MTL-1 treated
269
animals spent significantly higher time in light as compared to PTZ kindled animals (Fig. 5).
270
Results showed that percentage time spent in light by MTL-1 treated group was 13.3 ± 4.6 s as
271
compared to PTZ treated group which was 3.05± 1.25 s (Fig. 5A). The percentage time spent in the
272
dark by MTL-1 treated group was 86.6 ± 4.6 s as compared to PTZ treated group and vehicle
273
group which were shown 96.9± 1.25 and 80.6 ± 2.5 s respectively (Fig. 5B.).
AC C
EP
TE D
M AN U
SC
RI PT
249
274 275
2.7. Molecular Docking study
10
ACCEPTED MANUSCRIPT 11
To determine the molecular interaction of MTL-1, a docking analysis was performed with the
277
coordination of COX-2 (PDB ID: 6COX) using AutoDock 4.2. Results showed that MTL-1 is
278
preferentially accommodated at the cyclooxygenase active site of COX-2 (Fig. 6) and Arg120,
279
Ser353 and Tyr324 are involved in H-bond interaction with MTL-1. We observed that
280
methylsulfonyl benzene moiety of MTL-1 is deep merged in the lower hydrophobic channel,
281
surrounding with residues Val116, Leu117, Leu352, Tyr355, Leu359, Tyr385 and Ala527
282
whereas, methyl phenyl of MTL-1 is protruded towards the upper hydrophobic domain of the
283
cavity and is engaged in hydrophobic interaction with residues: Leu352, Trp387, Ala516, Ile517
284
and Phe518.
285
2.8. Molecular dynamic simulation study
286
To analyze the structural stability and molecular interaction of COX2-MTL-1 complex, a MD
287
simulation was carried out using biomolecular simulation package AMBER14 for the period of
288
250 ns at 300K. The dynamical change of all Cα backbone RMSD of COX-2 and COX-2-CX
289
complex is shown in Fig. 7A. The RMSD plot shows initial drop down in trajectory during 0-25
290
ns in water, however, change in RMSD of 1.5 Å is seen for 25-75 ns, which suggested the initial
291
perturbation in structure. The trajectory remains stable during 75-210 ns and small drift of 0.5 Å
292
is observed at 225 ns, although, it is settled quickly with the end of simulation at 250 ns, which
293
suggested the stable conformation of COX-2 in water at 300K. The RMSD trajectory of COX-2-
294
MTL-1complex shows relatively stable conformation as compared to protein in unbind state.
295
With the initial perturbation trajectory attained the equilibrium and remain stable ~85 ns of
296
simulation. We find a drift of 1.0 Å at 90 ns, which is settled after 100 ns, and slow but
297
continuous dropdown in trajectory is observed till the simulation is finished at 250 ns. The initial
298
change in confirmation suggested the spatial fitting of ligand at the active site. The stable RMSD
299
during 100-250 ns reveals the stable interaction of ligand at binding pocket of COX-2.The
300
average RMSD value 2.29±0.36 and 2.55±0.33 of COX-2 in free and bind state also indicated
301
the higher conformational stability of MTL-1 at the active site of COX-2 enzyme.
302
2.7.1. Radius of gyration (Rg) determination
303
Radius of gyration (Rg) is another important and valuable parameter to determine the structural
304
stability and compactness of protein. Herein, to analyze the structural integrity of protein-ligand
AC C
EP
TE D
M AN U
SC
RI PT
276
11
ACCEPTED MANUSCRIPT 12
complex we computed the time evolution plot of Rg backbone, as shown in Fig. 7B. As depicted
306
from Fig. 6B, the structural entity of COX-2 is remained stable during the simulation of 250 ns
307
with the average Rg value 24.27±0.15 Å. We observed the initial change of 0.5 Å in Rg value of
308
COX-2-MTL-1 during 0-50 ns of simulation, which is settled at 75 ns and equilibrium, is
309
maintained till the simulation is finished at 250 ns. The average Rg value 24.46±0.16 Å of
310
complex suggested that secondary and tertiary conformation of COX-2 is retained during the
311
simulation, whereas, initial rise in Rg suggested the preferential binding of ligand at the active
312
site of COX-2.
313
2.7.2. RMSF determination
314
To determine the dynamic progression of COX-2 and COX-2-MTL-1, we also analyzed the
315
time–average RMSF plot of all C αatoms. As shown in Fig. 8, the average fluctuation of residues
316
having < 4-6Å, are belonging to stable conformation (α-helices and β-sheets), however, loop
317
region showed average fluctuation <6.0 Å. The structure of COX-2 consists of 40% α-helices
318
and 5% β-sheets, and loops are imbedded between the regular secondary conformations. The
319
RMSF plot of protein-ligand complex showed the least average fluctuation of residues are
320
belonging to α-helix-2 (His90), α-helix-4 (Val116, Ser119, Arg120), α-helix-7 (Asp190,
321
Gln192),α-helix-11 (His351, Leu352, Ser353),α-helix-13(Phe381, Leu384),α-helix-22 (Gly526,
322
Ala527, Ser530) and
323
suggested the coordination with ligand at the active site of COX-2.
324
2.7.3. Interaction energy (MM-PBSA) observation
325
To estimate the binding free energy of COX-2-MTL-1 complex, we used the MM-PBSA.py, a
326
compatible program with AMBER to compute various components of binding energy i.e. the
327
electrostatic, van der Walls, polar and non-polar solvation energy. Figure 9 shows that the
328
average values are converged in MMPBSA calculations. Although, we carried out MM-PBSA
329
calculations for the entire 250 ns, however, observing the convergence, results of last 50 ns of
330
simulation are shown here having binding free energy -45.02± 2.67 (Fig.9).
331
2.8. Sub-acute toxicity study
332
Toxicity of new chemical entity is considered as a major obstacle in path of drug discovery and
333
development [24]. Many of potent pre-clinically active pharmacological drugs could not approve
TE D
M AN U
SC
RI PT
305
Tyr385, Trp387, Ala516, Phe518) which
AC C
EP
residues loops (Tyr355,
12
ACCEPTED MANUSCRIPT 13
for clinical use due to their toxic nature [25]. Various reports indicate that COX-2 inhibitors
335
produce cardiovascular toxicity which might be dependent on the dose as well as on the duration
336
of treatment [26]. Cardiovascular toxicity associated with COX-2 treatment is due to an inequity
337
between COX-2 derived vasoprotective prostacyclin (PGI2) in the endothelium and COX-1
338
induced thrombotic thromboxane A2 (TXA2) in platelets [27]. It is also documented that prolong
339
use of COX-2 inhibitor showed gastrointestinal and renal toxicity [28].
340
Therefore, a sub-acute toxicity study has been performed to evaluate safety nature of novel
341
chemical compound MTL-1 in normal, healthy Wistar rats at the dose of 200 mg/kg/bwt. During
342
the whole experimental period, no sign of visible toxicity was observed. Hematological analysis
343
results have indicated that treatment with MTL-1 did not alter crucial hematological parameters
344
such as RBC, WBC and platelet counts, which indicated the nontoxic nature of MTL-1upon 14
345
days treatment (Table. 4). Further, liver function associated biomarkers were also quantified and
346
results displayed that treatment with MTL-1 did not significantly modify serum glutamate
347
oxaloacetatetransaminase (SGOT), serum glutamate pyruvate transaminase (SGPT), alkaline
348
phosphatase (ALP), total protein, and total bilirubin as compared to non treated control animals.
349
Thus, MTL-1 did not exert any remarkable toxicity to liver upon chronic treatment (Table. 5).
350
Furthermore, renal toxicity related biomarkers such as uric acid, creatinine, and urea were also
351
deliberated after 14 days administration of MTL-1. Obtained data indicated that MTL-1 did not
352
produce significant renal toxicity upon 14 days oral administration (Table 5).
353
Overall, these preliminary toxicological evaluations have proven MTL-1 as a safe COX-2
354
inhibitor which also having valuable anticonvulsant activity and cognition boosting capability.
355 356
Table 4: Hematological parameters after oral administration of vehicle and MTL-1 for 14 days in rats.
AC C
EP
TE D
M AN U
SC
RI PT
334
Hematological parameters with units Hb(g/dl) TLC (thou/mm3) Neutrophil %
Control (Vehicle, po) ± S.D 12.87±0.6
MTL-1 (200 mg/kg, po) ± S.D 14.15±0.07
7.23±1
6.25±0.64
17.33±3.21
21.5±2.1
13
ACCEPTED MANUSCRIPT 14
77.33±2.08
73±4.24
Eosinophil %
2.67±0.58
2.5±0.7
Monocyte %
2.67±0.58
2.5±0.7
Basophils %
0±0
RBC (mill/mm3)
8.19±0.64
Platelet count (thou/mm3)
1119±142.9
0.5±0.7
7.34±0.65
990±56.57
SC
357
RI PT
Lymphocyte %
Table 5: Liver and renal function test after oral administration of vehicle and MTL-1for
359
14 days in rats. Biochemical parameters (liver and kidney) with units Total Bilirubin SGOT
Alkaline phosphate
Control (Vehicle, po) ± S.D 0.27 ± 0.068
MTL-1 (200 mg/kg, po) ±S.D 0.4±0
77.7 ± 1.05
76.65±2.33
64.3±3.5
59.6±11.45
TE D
SGPT
M AN U
358
115.9±14.5
123.5±2.12
6.7±0.26
6.55±0.21
40.83±1.16
31.45±2.62
0.63±0.05
0.9±0
3.13±0.4
1.45±0.35
Calcium
10.67±0.61
9.65±0.07
Phosphorous
7.83±0.29
7.2±0.14
144.87±0.75
145.05±0.21
5.14±0.05
4.9±0.14
107.13±0.85
104±5.66
Total protein
Creatinine
AC C
Uric acid
EP
Blood urea
Sodium
Potassium Chloride
14
ACCEPTED MANUSCRIPT 15
3. Conclusion
361
Herein, we report discovery of a novel series of methylsulfonyl benzene derivatives as potent
362
and selective COX-2 inhibitors, which endowed with admirable anti-convulsant effect. Our in-
363
vitro evaluation provided MTL-1 and MTL-2 as potent and selective COX-2 inhibitors in the
364
entire series. MTL-1 displayed excellent protection against sc-PTZ induced seizure in acute
365
models of epilepsy. Additionally, in time course study MTL-1 showed a long duration of action
366
and displayed 67% seizure protection after 6h of drug administration. Further, MTL-1 also
367
showed satisfactory protection in PTZ induced chronic model of epilepsy as compared to
368
standard COX-2 inhibitor ETX and successfully improved cognition impairment in PTZ kindled
369
rats. In sub-acute toxicity study MTL-1 did not exert any significant toxicity, as compared to
370
control group animals. Furthermore, in-silico docking and MD simulation studies revealed that
371
MTL-1 nicely fitted in active site of COX-2 with appropriate H-bond, hydrophobic and polar
372
interaction. Thus, MLT-1 became visible as a potent COX-2 inhibitor, which also showed
373
excellent anti-convulsant and cognition boosting potential without exerting any significant
374
toxicity.
375
4. Experimental section
376
4.1. Chemistry
377
Chemicals and reagents were purchased from Sigma Aldrich (USA), S.D Fine Chemicals (India),
378
Merck (Germany) and TCI (Japan). Melting points were determined with open capillaries by
379
using model KSPII, KRUSS, (Germany). The nuclear magnetic resonance (NMR) spectra were
380
obtained on high resolution Jeol-400 MHz NMR spectrophotometer (USA) DMSO-d6 using
381
tetramethylsilane (TMS) as the internal reference. Chemical shifts (δ) were expressed in parts per
382
million relative to TMS, and the following abbreviations were used to describe the peak patterns
383
when appropriate: s, (singlet); d, (doublet); t, (triplet); m, (multiplet) and brs (broad singlet). The
384
coupling constant (J) values are given in hertz (Hz). Mass spectra were recorded on an Agilent
385
6310 Ion trap LC/MS and elemental analysis (C, H and N) was performed on Elementar
386
analysensysteme.
387
4.1.1 Synthesis of phenyl (4-(methylsulfonyl) phenyl) carbamate (2)
388
An equimolar (1mM) amount of 4-(methylsulfonyl) aniline (1mM), 1 and phenyl chloroformate
389
were stirred in tetrahydrofuran (THF) with a catalytic amount of pyridine for 14h at RT.
AC C
EP
TE D
M AN U
SC
RI PT
360
15
ACCEPTED MANUSCRIPT 16
Reaction mixture was poured in chilled water and appeared precipitate was filtered to achieve
391
carbamate derivative 2.
392
Phenyl (4-(methylsulfonyl) phenyl) carbamate (2)
393
Crystalline white solid, yield: 92%; mp: 158-1600C; 1H NMR(400 MHz, DMSO-d6): δ 3.15 (s,
394
1H, CH3), 7.23-7.28 (m, 3H, Ar-H), 7.41-7.45 (m, 2H, Ar-H), 7.71 (d, 2H, Ar-H, J= 9.1Hz), 7.87
395
(d, 2H, Ar-H, J= 9.1Hz), 10.74 (s, 1H, NH); LC–MS: m/z 291(M+1).
RI PT
390
396
4.1.2 Synthesis of N-(4-(methylsulfonyl)phenyl)hydrazinecarboxamide (3)
398
A mixture of carbamate derivative 2 and hydrazine hydrate was heated at temperature 100° C in
399
1,4-dioxane for 6h. Reaction mixture was poured in chilled water and product extracted with
400
ethyl acetate twice. Combined organic layer was treated with anhydrous sodium sulfate and
401
evaporated with reduced pressure to yield carboxamide derivative 3.
M AN U
SC
397
402
N-(4-(methylsulfonyl)phenyl)hydrazinecarboxamide (3)
404
Crystalline white solid, yield: 87%; mp: 180-1820C; 1H NMR (400 MHz, DMSO-d6): δ 3.13 (s,
405
1H, CH3), 4.42 (brs, 2H, NH2), 7.72- 7.78 (m, 5H, Ar-H+ NH), 9.13 (s, 1H, NH); LC–MS: m/z
406
229(M+1)
407
4.1.3
408
(alkyl/arylcarbamothioyl)hydrazinecarboxamide (4-14)
409
An equimolar ratio of carboxamide derivative 3 and various substituted isothiocyanates were
410
refluxed (900C) in mixture of dried acetonitrile and ethanol for 8-10h. After completion of
411
reaction solvent was evaporated and the reaction mixture was diluted with ethyl acetate. The
412
organic layer was washed with brine solution twice, dried with anhydrous sodium sulfate and
413
followed by evaporation, provided crude desired products in good yield. Crude products were
414
purified by column chromatography using chloroform: methanol (96:4) as eluent.
for
synthesis
of
N-(4-(methylsulfonyl)phenyl)-2-
EP
procedure
AC C
415
General
TE D
403
416
4.1.3.1 N-(4-(methylsulfonyl) phenyl)-2-(phenylcarbamothioyl) hydrazinecarboxamide (4)
417
White solid; yield 70%; mp: 170-1720C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13 (s, 3H, CH3),
418
7.13(t, 1H, J= 7.3Hz, Ar), 7.50(d, 2H, J= 8. 0Hz, Ar); 7.74(d, 2H, J= 8.2Hz, Ar), 7.81(d, 2H, J= 16
ACCEPTED MANUSCRIPT 17
13
8.72 Hz, Ar), 8.49(s, 1H, NH), 9.39(s,1H, NH), 9.53(s,1H, NH),9.86(s, 1H, NH);
C NMR
420
(DMSO-d6, 100 MHz): δ 43.9, 118.0, 124.9, 128.0, 128.2, 132.2, 139.1, 144.5,154.6, 181.7; LC-
421
MS: 365 (M+1); Anal.Calcd for C15H16N4O3S2: C, 49.43; H, 4.43; N, 15.37; Found: C, 49.68; H,
422
4.63; N,15.15.
423
4.1.3. 2 N-(4-(methylsulfonyl) phenyl)-2-(p-tolylcarbamothioyl) hydrazinecarboxamide (5)
424
White solid; yield 75%; mp: 182-1830C; 1H NMR (DMSO-d6, 400 MHz): δ 2.25(s, 3H, CH3),
425
3.12(s, 3H, CH3), 7.10(d, 2H, J= 8.2Hz, Ar), 7.34(d, 2H, J= 8.68Hz, Ar), 7.72(d, 2H, J= 8.6Hz,
426
Ar), 7.79(d, 2H, J= 9.1Hz), 8.4(s, 1H, NH), 9.36(s, 1H, NH), 9.44(s, 1H, NH), 9.70(s, 1H, NH)
427
; 13C NMR (DMSO-d6, 100 MHz): δ 20.5, 43.9, 117.9, 125.2, 128.1,128.5, 133.1, 134.0, 136.6,
428
144.5, 154.6, 181.8; LC-MS: 379 (M+1); Anal.Calcd. for C16H18N4O3S2: C, 50.78; H, 4.79; N,
429
14.80; Found: C, 51.01; H, 4.59; N, 15.04.
M AN U
SC
RI PT
419
430 431
4.1.3.32-((2,6-dimethylphenyl)carbamothioyl)-N-(4-(methylsulfonyl)phenyl)hydrazine
433
carboxamide (6)
434
White solid; yield 72%; mp: 185-1570C; 1H NMR (DMSO-d6, 400 MHz): δ 2.13(s, 6H, 2xCH3),
435
3.12(s, 3H, CH3), 7.01-7.05(m, 3H, Ar), 7.68-7.80(m, 4H, Ar), 8.53(s, 1H, NH), 9.25(s, 1H,
436
NH),9.40(s, 2H, 2xNH);
437
128.2, 133.1, 136.4, 136.9, 144.6, 154.4, 156.8, 181.9; LC-MS: 393 (M+1); Anal.Calcd for
438
C17H20N4O3S2: C, 52.02; H, 5.14; N, 14.27; Found: C,50.26; H,4.91; N,14.06.
439
4.1.3.4.2-((4-methoxyphenyl) carbamothioyl)-N-(4-(methylsulfonyl)phenyl)
440
hydrazinecarboxamide (7)
441
White solid; yield 68%; mp: 176-1770C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3),
442
3.68(s, 3H, OCH3), 6.83(d, 2H, J= 9.16Hz, 2H), 7.38(d, 2H, J= 9.16 Hz, Ar), 7.74-7.80(m, 4H,
443
Ar), 7.95(s, 1H, NH), 8.21(s, 1H, NH), 8.65(1H, NH), 9.31(s, 1H, NH);
444
100 MHz): δ 43.9, 55.2, 113.2, 118.0, 126.7, 128.2, 131.9, 133.1, 144.5, 154.7, 156.6, 182.2;
445
LC-MS: 395 (M+1); Anal.Calcd for C16H18N4O4S2: C, 48.72; H, 4.60; N, 14.20; Found: C,48.91;
446
H, 4.76; N,14.02.
447
4.1.3.5 2-((4-fluorophenyl) carbamothioyl)-N-(4-(methylsulfonyl)phenyl)hydrazinecarboxamide
448
(8)
TE D
432
C NMR (DMSO-d6, 100 MHz): δ 17.9, 43.9, 117.9, 126.7, 127.4,
AC C
EP
13
13
C NMR (DMSO-d6,
17
ACCEPTED MANUSCRIPT 18
449
White solid; yield 74%; mp:172-1740C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3),
450
7.37(d, 2H, J= 8.2Hz, Ar), 7.54(d, 2H, J= 8.7Hz, Ar), 7.73(d, 2H, J= 8.6Hz, Ar), 7.80(d, 2H, J=
451
9.1Hz, Ar), 8.49(s, 1H, NH), 9.40(s, 1H, NH), 9.62(s, 1H, NH), 9.92(s,1H, NH);
452
(DMSO-d6, 100 MHz): δ 43.9, 114.5, 114.8, 128.1, 133.2, 135.4, 144.4, 156.6, 132.0; LC-MS:
453
383 (M+1); Anal.Calcd for C15H15FN4O3S2: C, 47.11; H, 3.95; N, 14.65; Found: C,47.37;
454
H,3.83; N,14.41.
455
4.1.3.62-((2, 6-difluorophenyl)carbamothioyl)-N-(4-(methylsulfonyl)phenyl)
456
hydrazinecarboxamide (9)
457
White solid; yield 65%; mp: 166-1680C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3),
458
7.11(t, 2H, J= 8.26Hz, Ar), 7.32-7.39(m,1H, Ar), 7.71-7.81(m, 4H, Ar), 8.65(s, 1H, NH),
459
9.34(s,1H, NH), 9.47(s,1H,NH), 9.84(s,1H,NH); 13C NMR (DMSO-d6, 100 MHz): δ 43.8, 111.7,
460
117.8, 128.2, 128.8, 133.2, 144.3, 154.3, 157.5, 160.1,162.0,183.3; LC-MS: 401 (M+1);
461
Anal.Calcd for C15H14F2N4O3S2: C, 44.99; H, 3.52; F, 9.49; N, 13.99; Found: C,44.81; H,3.73;
462
N,14.18.
463
4.1.3.7 2-((4-chlorophenyl) carbamothioyl)-N-(4-(methylsulfonyl)phenyl)hydrazinecarboxamide
464
(10)
465
White solid; yield 67%; mp: 172-1730C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3),
466
7.29(s, 2H, Ar), 7.53(s, 2H, Ar), 7.77-7.79(m, 4H, Ar), 8.13(s, 1H, NH), 8.26(s, 1H, NH),8.99(s,
467
1H, NH), 9.34(s,1H, NH); LC-MS: 399 (M+1); Anal.Calcd for C15H15ClN4O3S2: C, 45.17; H,
468
3.79; N, 14.05; Found: C,45.43; H,4.01; N,13.81.
469
4.1.3.8 2-((2, 3-dichlorophenyl)carbamothioyl)-N-(4(methylsulfonyl)phenyl)
470
hydrazinecarboxamide (11)
471
White solid; yield 71%; mp: 288-2890C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3,
472
7.35(t,1H, J= 7.9Hz, Ar), 7.45(s, 1H, Ar), 7.52(d, J= 8.2Hz, Ar), 7.73(d, 2H, J= 8.68Hz, Ar),
473
7.80(d, 2H, J= 9.1Hz, Ar), 8.64(s, 1H, NH), 9.38(s, 1H, NH), 9.80(s, 2H, 2xNH);
474
(DMSO-d6, 100 MHz): δ 43.9, 118.0, 127.5, 128.2, 129.5, 131.6, 133.3, 144.4, 154.5, 182.4; LC-
475
MS: 432 (M+1); Anal.Calcd for C15H14Cl2N4O3S2: C, 41.58; H, 3.26; N, 12.93; Found: C,41.84;
476
H,3.09; N,12.68.
477
4.1.3.9 2-(benzylcarbamothioyl)-N-(4-(methylsulfonyl) phenyl)hydrazinecarboxamide (12)
C NMR
AC C
EP
TE D
M AN U
SC
RI PT
13
13
C NMR
18
ACCEPTED MANUSCRIPT 19
White solid; yield 75%; mp: 176-1770C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3), ,
479
4.72(d, 2H, CH2), 7.20-7.29(m, 5H, Ar), 7.72(d, 2H, J= 8.72Hz, Ar), 7.79(d, 2H, J= 8.72Hz),
480
8.40(s, 1H, NH), 8.69(s, 1H, NH), 9.27(s, 1H, NH), 9.30(s, 1H, NH); 13C NMR (DMSO-d6, 100
481
MHz): δ 43.8, 46.7, 117.9, 126.3, 126.9, 127.5, 127.8, 128.4, 133.1, 139.2, 144.5, 154.6, 182.8;
482
LC-MS: 379 (M+1); Anal.Calcd for C16H18N4O3S2: C, 50.78; H, 4.79; N, 14.80; Found: C,51.02;
483
H,4.97; N,14.54.
484
4.1.3.10 N-(4-(methylsulfonyl)phenyl)-2-(phenethylcarbamothioyl)hydrazinecarboxamide (13)
485
White solid; yield 79%; mp: 177-1790C; 1H NMR (DMSO-d6, 400 MHz): δ 3.13(s, 3H, CH3),
486
2.82(t, 2H, J= 7.8Hz, CH2), 3.64(q, 2H, J= 7.4Hz, CH2), 7.17-7.25(m, 5H, Ar), 7.73(d, 2H, J=
487
8.72Hz, Ar), 7.80(d, 2H, J= 8.7Hz, Ar), 8.22(s, 1H, NH), 8.32(s, 1H, NH), 9.18(s, 1H, NH),
488
9.28(s, 1H, NH); 13C NMR (DMSO-d6, 100 MHz): δ 34.8, 43.9, 45.2, 118.0, 126.1, 128.1, 128.3,
489
128.6, 133.2, 139.3, 144.4, 154.7, 182.1; LC-MS: 393 (M+1); Anal.Calcd for C17H20N4O3S2: C,
490
52.02; H, 5.14; N, 14.27; Found: C,52.30; H,4.99; N,14.51.
491
4.1.3.11 2-(ethylcarbamothioyl)-N-(4-(methylsulfonyl)phenyl)hydrazinecarboxamide(14)
492
White solid; yield 64%; mp: 176-1780C; 1H NMR (DMSO-d6, 400 MHz): δ 1.05(t, 3H, J=
493
7.0Hz, CH3), 3.12(s, 3H, CH3), 3.46(q, 2H, J= 6.9Hz, CH2),7.71(d, 2H, J= 9.1Hz, Ar), 7.78(d,
494
2H, J= 8.7Hz, Ar), 8.12(s, 1H, NH), 8.28(s, 1H, NH), 9.06(s, 1H, NH), 9.25(s, 1H, NH);
495
NMR (DMSO-d6, 100 MHz): δ 14.4, 38.5, 43.9, 118.0,128.1, 133.1, 144.4, 154.7, 181.9; LC-
496
MS: 317 (M+1); Anal.Calcd for C11H16N4O3S2: C, 41.76; H, 5.10; N, 17.71; Found: C,41.99;
497
H,5.29; N,17.51.
498
4.2. In-vitro COX-2/COX-1 inhibition assay
499
COX-2/ COX-1 inhibition assay has been performed according to manufacturer’s instructions
500
[29]. Briefly, stock solutions of test compounds were dissolved in a minimum volume of DMSO.
501
Provided 3ml assay buffer was diluted with 27ml of HPLC grade water and this final buffer
502
(100mM Tris HCl, PH-8.0) was used in the whole assay. Whole assay was performed in 96 well
503
plate and 150µl of assay buffer, 10 µl of heme, 10 µl of enzyme, and 10 µl of solvent were added
504
in 100% initial activity well. Further, 160 µl of assay buffer, 10 µl of heme and 10 µl of solvent
505
were added in background well. Furthermore, 150 µl of assay buffer, 10 µl of heme, 10 µl of
506
enzyme and various concentrations of inhibitor (10 µl) were added in inhibitor well. Reaction
507
mixture was incubated for five minutes at room temperature followed by addition of ADPH (10
13
C
AC C
EP
TE D
M AN U
SC
RI PT
478
19
ACCEPTED MANUSCRIPT 20
µl). Reaction was initiating by quick addition of Arachidonic acid solution (10 µl) to inhibitor
509
well and control well. After providing two minutes incubation, absorbance of plate was taken at
510
excitation wavelength 535 and an emission wavelength 590 nm. Percentage inhibition was
511
determined by subtracting each inhibitor sample value from 100% initial activity sample, divided
512
by 100% initial activity and multiply by 100. IC50 was calculated by percentage inhibition with
513
Microsoft excel.
514
4.3. Pharmacology
515
Adult Swiss Albino male mice (25–30 g) and male Wistar rats (200-250 g) were used as
516
experimental animals. All animals were kept under standard animal laboratory conditions at Dr.
517
B.R Ambedkar Center for Biomedical Research, University of Delhi, India. Each animal was
518
allowed for free access to food and water, except during the experimental period. Subcutaneous
519
PTZ and PTZ-induced kindling tests were performed according to the standard protocol
520
illustrated by Antiepileptic Drug Development (ADD) program of the National Institute of
521
Health (NIH, USA). All the experimental protocols were prior approved by the Institutional
522
Animal Ethics Committee (IAEC) for animal care.
523
Chemical and drugs
524
Pentylenetetrazole was procured from Sigma Aldrich (St. Louis, USA); sodium valproate
525
(Unimed
526
pharmacological assays, the test compound MTL-1 was freshly suspended in a 1% gum acacia in
527
sterile saline solution. The vehicle represents 1% gum acacia in sterile saline. Fresh
528
pentylenetetrazole was used and dissolved in normal sterile saline to such concentrations that
529
requisite doses were administered in a volume of 10 ml/kg (mice or rats) body weight. Sodium
530
valproate and etoricoxib at desired doses were also dissolved in sterile saline before use.
531
4.3.1. sc-PTZ test
532
Pentylenetetrazole (PTZ) was administrated subcutaneously (sc) at the convulsive dose of 85
533
mg/kg (CD97). The test compound MTL-1 was administered intraperitoneally (i.p.) at the doses
534
of 30, 100 and 300 mg/kg in mice and after 0.5 h and 4 h, PTZ was injected. Standard COX-2
535
inhibitor etoricoxib was tested on three doses (1, 5 and 10 mg/kg, i.p.) while, standard AED
536
sodium valproate was tested at 300 mg/kg, i.p. at both time intervals. The mice were placed in a
TE D
Ltd,
India)
and
etoricoxib
(Themis
Medicare,
India).
For
AC C
EP
Technologies
M AN U
SC
RI PT
508
20
ACCEPTED MANUSCRIPT 21
537
clear rectangular plastic cage, permitting a full view of the animal’s seizure episodes and
538
observations were carried out for 30 min. The episode of clonic seizures, tonic seizures was
539
documented carefully according to our previous method [30-33].
RI PT
540
4.3.2 Time course study
542
The in-vivo time course study was performed in mice to judge long duration action of MTL-1.
543
The test compound MTL-1 was administered i.p. at the fixed dose of 30 mg/kg, and the
544
percentage of seizure protection was examined at varying time intervals such as 0.5, 1, 2, 3, 4,
545
and 6 h by administering convulsive dose of PTZ (85 mg/kg, sc) [30].
SC
541
546
4.3.3 Anticonvulsant Quantification Studies (ED50 Determination)
548
The median effective dose (ED50) of MTL-1 was evaluated by sc-PTZ test using mice and was
549
administered i.p. to each animal at the varied doses till three-five points were established between
550
the dose level of 0% protection and 100% protection. The ED50 and the 95% confidence interval
551
were deliberated by the Graphpad prism 5 [31].
553
4.3.3. Induction of kindling by PTZ: In-vivo Epileptiogenesis model
554
All animals were randomly divided into four groups (n=8 in each group), which are as follows:
555
1: Control group (vehicle daily, i.p. for 28 days).
556 557
2: PTZ group (PTZ at 40 mg/kg, i.p. on alternate days from 1st to 28th day + vehicle daily, i.p. for 28 days).
558 559
3: PTZ + ETX group (PTZ at 40 mg/kg, i.p. on alternate days from 1st to 28th day + etoricoxib at 5 mg/kg daily, i.p. for 28 days).
560 561
4: PTZ + MTL-1 group (PTZ at 40 mg/kg, i.p. on alternate days from 1st to 28th day + MTL-1 at 30 mg/kg daily, i.p. for 28 days).
AC C
EP
TE D
552
M AN U
547
562 563
Immediately, after the PTZ injection, the animals were observed for the incidence of convulsive
564
behavior. The resultant behavioral seizure activity was categorized according to revised Racine
565
scale: 21
ACCEPTED MANUSCRIPT 22
Stage 1- Sudden behavioral arrest and/or motionless staring;
567
Stage 2-Facial jerking with muzzle or muzzle and eye;
568
Stage 3- Neck jerks;
569
Stage 4-Clonic seizure in a sitting position;
570
Stage 5- Convulsions including clonic and/or tonic–clonic seizures while lying on the belly and/or
571
pure tonic seizures;
572
Stage 6- Convulsions including clonic and/or tonic–clonic seizures while lying on the side /or wild
573
jumping.
574
Additionally, all tested animals were observed for next 45 min to check for lethality before
575
returning to the home cage. The animals were considered to be kindled after receiving 14th PTZ
576
injection and after having reached at least three consecutive stages 5 or 6 seizures. The collective
577
kindling score was (calculated by taking the average of the individual behavior, seizure pattern of
578
each mouse in a group and dividing them with the number of animals present in the corresponding
579
group) plotted against time duration of kindling period. On the test day, all groups except group I
580
was tested for PTZ challenge dose test consisting single injection of 35 mg/kg, i.p. This high dose
581
of PTZ shortens the threshold of seizure activity more and thus produces lethality and status
582
epileptics in kindled rats [32].
SC
M AN U
TE D
583
RI PT
566
4.3.4 Neurobehavioral Tests
585
The cognition assessment of MTL-1 was performed by various types of neurobehavioral tests,
586
including social recognition test, novel object recognition test and dark/light test. These
587
experiments were performed in developed kindled rats, which consisted of three groups consist
588
of six animals in each group. Group A (vehicle only), Group B (PTZ alone) and Group C (PTZ +
589
MTL-1). All the experiments were performed in an animal experimental room where the
590
temperature (24±2˚C) was maintained. The animal experimental room was kept silent during the
591
experiments. Experiments were performed during 9.00 h to 14.00 h.
AC C
EP
584
592 593
4.3.4.1 Social Recognition Test
594
The social recognition test is a widely accepted method to measure short term memory relies on
595
the olfactory cues. Adult (200-250 g) and juvenile rats (60-65 g) were used and this test was 22
ACCEPTED MANUSCRIPT 23
performed according to the method previously reported by Timmermann et al. with minor
597
modifications [34]. Briefly, animals were acclimatized in the animal house test room for 60
598
minutes before starting social recognition test. After acclimatization, each rat was placed alone in
599
separate cages for the 30 minute habituation period. After habituation, each rat was first placed
600
with a juvenile rat in the test cage for 3 minutes and first interaction time (T1) was noticed
601
manually by stop watch. It was entitled as a first interaction trial. After the completion of the first
602
interaction trial, juvenile rats were taken back in their original cages from test cages. Second
603
interaction trial (T2) was started after 2 h of first interaction trial in which each rat was again
604
interacted with same juvenile rat for 5 minutes and second interaction time T2 was noticed. For
605
the analysis point of view the ratio of T2/T1 was calculated for each rat and averaged was taken
606
for all groups. The lower ratio of T2/T1 is indicator of improved cognition [34].
607
M AN U
SC
RI PT
596
608 609 610
4.3.4.2 Novel Object Recognition Test
611
by Prickaerts et al., 1997 with certain modifications. This test consists of three distinct phases
612
named habituation, training and test. During the first phase, the rats were placed in the test
613
apparatus without any object for 2 days habituation, twice for 3 min in each day. Testing session
614
was commenced which consisted two trials T1 and T2, each with a duration of three minutes. In
615
T1 trial each animal was exposed with two similar objects and interaction time T1 was noted.
616
After 1 h interval period second session was started and one of the similar objects was replaced
617
with new one and interaction time T2 was noticed. Percent time exploration of both objects, i.e.
618
similar and novel by each individual animal was calculated. The exploration parameter was
619
defined by animal behavior to explore the object by directing the nose to the object at a distance
620
of no more than 2 cm and or touching the object with the nose. To avoid olfactory cues, the
621
objects were meticulously cleaned with 70% ethanol after each trial. Time spent exploring each
622
object was calculated by an observer blind and was expressed as a percentage of the total
623
exploration time [35].
AC C
EP
TE D
The Novel object recognition (NOR) test was performed in male Wistar rats as method described
624 625
4.3.4.3 Light-dark (LD) test 23
ACCEPTED MANUSCRIPT 24
The light-dark test was performed to understand the role of MTL-1 in the cognitive function
627
including anxiety- like behavior. The rats which spends less time in the light region of light-dark
628
box is considered as an indication of anxiety-like behavior. The experiment was performed with
629
minor alteration in the original protocol of Chen et. al [36]. The test was performed by the light-
630
dark apparatus made up of polycarbonate comprise of two compartments (30x 30x 30cm) light
631
compartment and dark compartment. Both compartments are connected to each other by black
632
plastic tunnel (30x 30x15 cm) and separated by black lid on the junction of both compartments.
633
Rats generally prefer to stay in the dark region due to nocturnal in nature, but more depressed
634
rats or cognition impaired rats also prefer dark region while rats having improved cognition
635
prefer light region of light-dark box. Light chamber was illuminated by a 60 W lamp. Before
636
starting experiment each animal of all groups were familiarized with light- dark box for 3
637
minutes 24h earlier and next day actual experiment was started. Animals of all groups were
638
placed in light-dark box for 3 minutes and the time spent in each compartment was noticed by
639
stop watch. The percentage time spent in each compartment was calculated for all three groups
640
[36].
641
4.3.5. Molecular Docking and MD simulation
642
The molecular docking of MTL-1was carried out with the X-ray coordinates of COX-2 (PDB ID:
643
6COX) using AutoDock 4.2. The protein and ligand files were prepared with MGL tools and
644
multiple docking was run as described earlier [37-40]. The Lamarckian genetic algorithm (LGA)
645
was applied to define the best docking conformations. As a result, 10 top-posed docking
646
conformations were obtained and the best docking pose in terms of binding free energy was
647
subjected to molecular dynamics (MD) simulation for 250 ns to determine the consistent
648
interactions using AMBER14 software. The t-leap module of AMBER was used to prepare the
649
system for protein and protein-ligand complex and protonation states of the protein residues were
650
defined with PROPKA [41]. The parameterization of protein and ligand was done with ff14SB
651
and GAFF force field, respectively [42] and, a partial atomic charge on the ligand was assigned
652
with AM1-BCC [43]. Each system was neutralized with the addition of Na+ ions. All simulations
653
were performed using explicit TIP3P water molecules padding around 10Å. Each protein-ligand
654
system was minimized in two steps using steepest descent, followed by conjugate gradient
655
minimization. In the first step, ions and solvent molecules were minimized by restraining the
AC C
EP
TE D
M AN U
SC
RI PT
626
24
ACCEPTED MANUSCRIPT 25
solute and in second step entire system was minimized removing the restraints. The system was
657
then gradually heated to 300 K in six steps. The simulations were equilibrated for 1ns prior to
658
production run and the production run of 250 ns was carried out for protein and protein-ligand
659
complex using NPT ensemble. The trajectory was calculated with the time step of 2fs and
660
SHAKE was used to constrain hydrogen bonds. Finally, the data was collected at every 10ps for
661
the analysis. The structural stability of the protein-ligand complex was assessed in terms of root
662
mean square deviation (RMSD) and radius of gyration (Rg) of backbone Cα atoms.
663
Statistical analysis
664
Statistical analysis was performed by one-way and two way of variance ANOVA followed by
665
post-hoc Tukey’s and bonferroni test, respectively. The p value less than 0.05 was regarded to be
666
statistically significant. Statistical analysis was done using the GraphPad Prism 51 software (La
667
Jolla, USA). Data were represented as mean ± standard deviation (Mean ± SD).
M AN U
SC
RI PT
656
668
Acknowledgment
670
Chandra Bhushan Mishra is thankful to the Department of health research for financial support,
671
Shikha Kumari is thankful to DBT for Post-doctoral fellowship, Amresh Prakash is thankful to
672
Department of Science and Technology (DST), Delhi, India for the young scientist award.
673
Rajesh Yadav is thankful to ACBR, University of Delhi for the financial support. Manisha
674
Tiwari is thankful to the University of Delhi for sanctioning research funds. The University
675
Science Instrumentation Center (USIC) is deeply acknowledged for providing NMR spectral
676
analysis of the synthesized compounds.
EP
References 1. S. L. Moshe, E. Perucca, P. Ryvlin, T.Tomson, Epilepsy: new advances, Lancet. 6736 (2014) 60456-6.
AC C
677 678 679 680 681 682 683 684 685 686
TE D
669
2. H. E. Scharfman, The neurobiology of epilepsy, Curr Neurol Neurosci Rep.7 (2007) 348-54. 3. A.T. Berg, S. F. Berkovic, M. J. Brodie, J. Buchhalter, J. H. Cross, W. Van Emde Boas, J. Engel, J. French, T. A Glauser, G.W Mathern, S. L Moshé, D. Nordli, P. Plouin, I. E. Scheffer, Revised terminology and concepts for organization of seizures and epilepsies:
25
ACCEPTED MANUSCRIPT 26
report of the ILAE Commission on Classification and Terminology, 2005-2009, Epilepsia. 51 (2010) 676-85.
RI PT
4. N. Marchi, T. Granata, D. Janigro, Inflammatory pathways of seizure disorders, Trends Neurosci. 37 (2014) 55-65. 5. S. Kumari, C. B. Mishra, M.Tiwari, Polypharmacological Drugs in the Treatment of Epilepsy: The Comprehensive Review of Marketed and New Emerging Molecules, Curr Pharm Des. 22 (2016) 3212-25. and
inflammation,
SC
6. E. Ricciotti, G. A. Fitz-Gerald, Prostaglandins ThrombVascBiol. 31 (2011) 986-1000.
Arterioscler
M AN U
7. A. Dey, X. Kang, J. Qiu, Y. Du, J. Jiang, Anti-Inflammatory Small Molecules To Treat Seizures and Epilepsy: From Bench to Bedside, Trends Pharmacol Sci. 37 (2016) 463-84. 8. A. M. Paoletti, S. Piccirilli, N. Costa, D. Rotiroti, G. Bagetta, G. Nistico, Systemic administration of Nψ-nitro-L-arginine methyl ester and indomethacin reduces the elevation of brain PGE2 content and prevents seizures and hippocampal damage evoked by Licl and tacrine in rat, Exp Neurol. 149 (1998) 349–55.
TE D
9. A. Dhir, P. S. Naidu, S. K. Kulkarni, Effect of cyclooxygenase inhibitors on pentylenetetrazol (PTZ)-induced convulsions: Possible mechanism of action, ProgNeuropsychopharmacolBiol Psychiatry. 30 (2006) 1478-85.
EP
10. A. Zarghi, S. Arfaei, Selective COX-2 Inhibitors: A Review of Their Structure-Activity Relationships, Iran J Pharm Res. 10 (2011) 655-83. 11. M. De-Bisschop, What are the risks of long-term NSAIDs and COX-2 inhibitors? JFamPract. 52 (2003) 199-200.
AC C
687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725
12. A. Zarghi, P. N. P. Rao and E. E. Knaus, Design and synthesis of new rofecoxib analogs as selective cyclooxygenase-2 (COX-2) inhibitors: replacement of the methanesulfonyl pharmacophore by a N-acetylsulfonamidobioisostere, J. Pharm. Pharm. Sci. 10 (2007) 15967. 13. A. Zarghi, S. Kakhgi, A. Hadipoor, B. Daraee, O.G Dadrass, M. Hedayati, Design andsynthesis of 1,3-diarylurea derivatives as selective cyclooxygenase (COX-2) inhibitorsBioorg Med Chem Lett. 18(2008)1336-9. 26
ACCEPTED MANUSCRIPT 27
RI PT
14. M. M Ghorab, M. S. A. El-Gaby, M. S. Alsaid, Y. A. M. M. Elshaier, A. M. Soliman, F. F. El-Senduny, F. A. Badria, A. Y. A Sherif, Novel Thiourea Derivatives Bearing Sulfonamide Moiety asAnticancer Agents Through COX-2 Inhibition. Anticancer Agents Med Chem. 17 (2017) 1411-1425
SC
15. D. Riendeau, M. D Percival, C. Brideau, S. Charleson, D. Dubé, D. Ethier, J. P. Falgueyret, R.W. Friesen, R. Gordon, G. Greig, J. Guay, J. Mancini, M. Ouellet, E.Wong, L. Xu, S. Boyce, D. Visco, Y. Girard, P. Prasit, R. Zamboni, I. W. Rodger, M. Gresser, A. W. FordHutchinson, R. N. Young, C. C. Chan. Etoricoxib (MK-0663): preclinical profile and comparison with other agents that selectively inhibit cyclooxygenase-2, JPharmacolExpTher. 296 (2001) 558-66.
M AN U
16. Q. R. Huang, L. C. Bell- Horner, I. D.Mohammad, F. D.Covey, J. A. Drewe, G. H. Dillon, Pentylenetetrazole-induced inhibitionof recombinant-aminobutyric acid type A (GABAA) receptors:mechanism and site of action, J. Pharmacol. Exp. Ther. 298 (2001) 986−995. 17. K. Gale, Subcortical structures and pathways involved in convulsive seizure generation. J Clin Neurophysiol. 9 (1992) 264-77.
TE D
18. N. Y. Lukomskaya, V. V. Lavrenteva, L. A. Starshinova, E. P. Zabkho, L. V. Gorbunova, , T. B. Tikhonova, V. E. Gmiro, L. G. Magazanik, Effects of ionotropic glutamate receptor channel blockers on the development of pentylenetetrazol kindling in mice, Neurosci Behav Physiol. 37 (2007) 75-81.
EP
19. J. Katyal, H. Kumar, Y. K. Gupta, Anticonvulsant activity of the cyclooxygenase-2 (COX-2) inhibitor etoricoxib in pentylenetetrazole-kindled rats is associated with memory impairment, Epilepsy Behav. 44 (2015) 98-103. 20. M. Jalava, M. Sillanpää, C. Camfield, P. Camfield, Social adjustment andcompetence 35 years after onset of childhood epilepsy: a prospective controlled study, Epilepsia. 38 (1997) 70815.
AC C
726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764
21. K. Perrine, B. P. Hermann, K. J. Meador, B. G. Vickrey, J.A. Cramer, R.D. Hays, O. Devinsky, The relationship of neuropsychological functioning to quality of life in epilepsy, Arch Neurol. 52 (1995) 997-1003. 22. M. Antunes, G. Biala, The novel object recognition memory: neurobiology, test procedure, and its modifications, Cogn. Process.13 (2012) 93-110. 27
ACCEPTED MANUSCRIPT 28
23. N. Kulesskaya, V. Voikar, Assessment of mouse anxiety-like behavior in thelight-dark box and open-field arena: role of equipment and procedure, Physiol. Behav. 133 (2014) 30-8.
RI PT
24. T. Burt, S. Nandal, Pharmacometabolomics in Early-Phase Clinical Development. ClinTransl Sci. 9 (2016) 128-38. 25. C. Katiyar, A. Gupta, S. Kanjilal, S. Katiyar, Drug discovery from plant sources: An integrated approach. Ayu. 33 (2012)10-9. 26. J.M. Ritter, I. Harding, J. B. Warren Precaution, cyclooxygenase inhibition and cardiovascular risk, Trends Pharmcol. Sci. 30 (2009) 503-508.
SC
765 766 767 768 769 770 771 772 773 774 775 776
27. G. A. FitzGerald, Coxibs and cardiovascular disease, N. Engl. J. Med. 351 (2004) 17091711.
779 780
28. A. Zarghi, S. Arfaei, Selective COX-2 Inhibitors: A Review of Their Structure-Activity Relationships, Iran J Pharm Res. 10 (2011) 655-83.
781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799
29. COX Inhibitor Screening Assay kit, Item No. 700100; Cayman Chemical Co.
M AN U
777 778
TE D
30. C. B. Mishra, S. Kumari, A. Angeli, S. M. Monti, M. Buonanno, M. Tiwari, C. T. Supuran, Discovery of Benzenesulfonamides with Potent Human Carbonic Anhydrase Inhibitory and Effective Anticonvulsant Action: Design, Synthesis, and PharmacologicalAssessment, J Med Chem. 60 (2017) 2456-2469.
EP
31. C. B. Mishra, S. Kumari, M. Tiwari, Design and synthesis of some new1-phenyl-3/4-[4(aryl/heteroaryl/alkyl-piperazine1-yl)-phenyl-ureas as potentanticonvulsant and antidepressant agents, Arch Pharm Res. 39 (2016) 603-17.
AC C
32. S. Kumari, C. B. Mishra, M. Tiwari, Design, synthesis and pharmacological evaluation of N[4-(4-(alkyl/aryl/heteroaryl)-piperazin-1-yl)-phenyl]-carbamicacid ethyl ester derivatives as novel anticonvulsant agents, Bioorg Med ChemLett. 25 (2015)1092-9. 33. S. Kumari, C. B. Mishra, M. Tiwari, Pharmacological evaluation of novel1-[4-(4benzo[1,3]dioxol-5-ylmethyl-piperazin-1-yl)-phenyl]-3-phenyl-urea aspotent anticonvulsant and antidepressant agent, Pharmacol Rep. 68 (2016) 250-8.
28
ACCEPTED MANUSCRIPT 29
34. D. B. Timmermann, K. Sandager-Nielsen, T. Dyhring, M. Smith, A. M. Jacobsen, E.
801
Nielsen, M. Grunnet, J. K.Christensen, D. Peters, K. Kohlhaas, G. M. lsen, P. K. Ahring,
802
Augmentation of cognitive function by NS9283, a stoichiometry-dependent positive allosteric
803
modulator of α2- and α4-containing nicotinic acetylcholine receptors, Br J Pharmacol 167,
804
(2012) 164-82.
805
35. J. Prickaerts, H. W. M. Steinbusch, J. F. M. Smits, J. de Vente, Possible role of nitric oxide-
806
cyclic GMP pathway in object recognition memory: effects of 7- nitroindazole and zaprinast,
807
Eur. J. Pharmacol. 337 (1997) 125-136.
SC
M AN U
36. Z. chen, H. Wei, A. Pertovaara, J. Wang, S. Carlson, Anxiety- and activity-related effects of paracetamol on healthy and neuropathic rats. Pharmacol Res Perspect 2018, e00367, 1-11. 37. S. Kumari, C. B. Mishra, D. Idrees, A. Prakash, R. Yadav, M.I. Hassan, M. Tiwari,Design, synthesis, in silico and biological evaluation of novel2-(4-(4-substituted piperazin-1yl)benzylidene)hydrazine carboxamides, MolDivers. 21 (2017) 163-174.
TE D
38. C. B. Mishra, S. Kumari, A. Manral, A. Prakash, V. Saini, A. M. Lynn, M. Tiwari, Design, synthesis, in-silico and biological evaluation of novel donepezil derivatives as multi-targetdirected ligands for the treatment of Alzheimer's disease, Eur J MedChem. 125 (2017) 736-750.
EP
39. C. B. Mishra, A. Manral, S. Kumari, V. Saini, M. Tiwari, Design, synthesis and evaluation of novel indandione derivatives as multifunctional agents with cholinesterase inhibition, anti-βamyloid aggregation, antioxidant and neuroprotection properties against Alzheimer's disease, Bioorg Med Chem. 24 (2016) 3829-41. 40. S. Kumari, D. Idrees, C. B. Mishra, A. Prakash, Wahiduzzaman, F. Ahmad, M. I. Hassan, M. Tiwari, Design and synthesis of a novel class of carbonic anhydrase-IX inhibitor 1-(3-(phenyl/4 fluorophenyl)-7-imino-3H-[1,2,3]triazolo[4,5d]pyrimidin6(7H)yl)urea, J Mol Graph Model. 64 (2016) 101-109.
AC C
808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835
RI PT
800
41. C. R. Søndergaard, M. H. M. Olsson, M. Rostkowski, J. H. Jensen, Improved treatment of ligands and coupling effects in empirical calculation and rationalization of pKa values, J. Chem. Theory Comput.7 (2011) 2284–2295. 42. J. Wang, R. M. Wolf, J.W. Caldwell, P. A. Kollman, D.A. Case, Development and testing of a general amber force field, J. Comput. Chem. 25 (2004) 1157–1174. 29
ACCEPTED MANUSCRIPT 30
836 837 838 839 840
43. A. Jakalian, D. B. Jack, C. I. Bayly, Fast efficient generation of high-quality atomic charges, AM1-BCC model: II. Parameterization and validation, J. Comput. Chem.23 (2002) 1623-41.
841 842 843 844 845 846 847
Fig. 2. The visual assessment of seizure scores in PTZ induced kindling in all groups for a period of 4 weeks. Group 1: Control group given vehicle only; Group 2: PTZ treated; Group 3: PTZ (40 mg/kg, i.p.) + Etoricoxib (ETX; 5 mg/kg, i.p.); Group 4: PTZ (40 mg/kg, i.p.) + Test compound MTL-1; 30 mg/kg, i.p.). The mean seizure scores expressed (±S.D) for each group. ***=P<0.0001 compared to control group; **= P<0.001 compared to control group; a= P<0.05 compared with PTZ group; ###=P<0.0001 compared to PTZ group. Repeated measures ANOVA followed by post hoc Tukey’s test.
848 849 850 851 852 853
Fig. 3. Graphical representation of the recognition ratio in the social recognition test performed on vehicle and PTZ-Kindled animals after administration of test compound MTL-1. The recognition ratios were calculated by dividing T2 (subsequent juvenile rat interaction time) by T1 (initial juvenile rat exploration time). Data are expressed as mean ± S.D (n=6 in each group; ** P<0.001 when group B (PTZ) was compared with group C (PTZ+ MTL-1). (One-way ANOVA followed by post hoc Tukey’s test).
854 855 856 857 858 859 860
Fig. 4. Graphical representation of the percent exploration preferences in novel object recognition test performed on vehicle and PTZ-Kindled animals after administration of test compound MTL-1. (A) denotes percentage exploration preference by animals in T1 trial with two identical objects A and A’ (B) denotes percentage exploration preference by animals in T2 trial with one similar object A and novel object B. Data are expressed as mean ± S.D for n=6 rats in each group. **P<0.01 within group A (vehicle).*P<0.05 within group C (PTZ+ MTL-1). Statistical analysis was carried out by Two-way ANOVA followed by bonferroni test.
861 862 863 864 865 866 867 868 869 870 871
Fig. 5. Effect of administration of test compound MTL-1in PTZ-kindled rats during the Light/Dark Test: (A) Denotes the percentage of time spent of test animals in light area. The significance levels of ***P<0.0001 was observed, when vehicle treated rats (Group A) were compared with PTZ-Kindled animals (Group B) and ###P<0.0001 when PTZ-Kindled animals (Group B) were compared with group C animals (PTZ+ MTL-1). * P<0.05 vehicle treated rats (Group A) versus group C (PTZ+ MTL-1). (B) Percentage time duration of test animals in dark area. Each column represents mean ± S.D (n=6 rats in each group). *** P<0.0001 Group A versus PTZ-Kindled animals (Group B) and ###P<0.0001 PTZ-Kindled animals (Group B) versus group C animals (PTZ+ MTL-1).*P<0.05 when PTZ-Kindled animals (Group B) were compared with group C animals (PTZ+ MTL-1). Statistical analysis was carried out by One-way ANOVA followed by post hoc Tukey’s test
AC C
EP
TE D
M AN U
SC
RI PT
Figure legends Fig. 1. Marketed COX-2 inhibitors and designed novel COX-2 inhibitors
872 30
ACCEPTED MANUSCRIPT 31
Fig. 6. Molecular docking of MTL-1: (A) Cartoon view of docked pose with COX-2 at PyMol. (B) COX-2 active site residues involve in interactions are shown with stick and Comp. MTL-1 is represented in ball-and-stick model. Residues Arg120, Ser353 and Leu352 are involved in Hbond interaction shown with dashed line in black.
877 878 879 880
Fig. 7. (A) RMSD plots of COX (black) and complex with MTL-1 (red) in water at 300 K during 250 ns MD simulation; (B) Time evolution of radius of gyration (Rg) values, all atoms Cα backbone of COX-2 (black) and COX-2- MTL-1 (red) in water at temperature 300K for 250 ns.
881 882 883 884
Fig. 8. RMSF plot of Cα atoms of COX-2 (black) and COX-2- MTL-1 (red) in water at temperature 300K for 250 ns.
AC C
EP
TE D
M AN U
Fig. 9 Convergence plot of COX-2 and MTL-1 complex.
SC
RI PT
873 874 875 876
31
SC
RI PT
ACCEPTED MANUSCRIPT
M AN U
Fig. 1
6
***
Control Group
***
2
**,a
**,a
PT Z Group PT Z + ET X Group
**,a
PT Z + MT L-1 Group
**,a
###
###
2
3
###
###
0
1
AC C
-2
Fig.2
TE D
4
EP
Seizure Severity Score
***
***
Week s of treatment
4
5
AC C EP TE D
Fig.3 ro u
G ro up
p
B
(P TZ
)
on ly )
Z+ M
TL
-1 )
SC (P T
ro u
(V eh ic le
G
A
C
p
M AN U
G
Recognition Ratio (T2/T1)
**
0.5
0.0
RI PT
ACCEPTED MANUSCRIPT
1.5
1.0
A
Fig.5
AC C
100
B M TL -1 )
G ro
up
C
(P T
p
Z+
G ro u
SC
(P TZ
)
eh )
M TL -1 )
RI PT B
(V
B
(P TZ +
0
2
C
***
(P TZ )
###
A
B
10
A
0
G ro up
20
* A'
ro up
25
up
Exploration Preference (%)
8
G
Fig. 4
on ly )
A
cl e
A
G ro
A'
(V eh i
M TL -1 )
A
B
M AN U
(P TZ +
(P TZ )
2
% Time spent in Dark period
C
B
(V eh )
Exploration Preference (%)
4
A
15
TE D )
5
TL -1
ro up
ro up
A
A'
M
G
G
G ro up
6
ro up
(P TZ +
)
y)
(P TZ
on l
% Time spent in Light period
8
G
C
B
cl e
p
(V eh i
A
G ro u
A
0
EP p
ro up
G ro u
G
ACCEPTED MANUSCRIPT
** *
6
4
A
B
***
A
150
###
*
50
0
B
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
A
AC C
EP
TE D
Fig.6
A
B
ACCEPTED MANUSCRIPT
B
M AN U
SC
RI PT
Fig. 7
EP AC C
Fig.9
TE D
Fig. 8
ACCEPTED MANUSCRIPT
1. MTL-1 has appeared as selective and potent COX-2 inhibitor in the entire series. 2. MTL-1 successfully protected animals from sc-PTZ induced seizure. 3. In chronic model of epilepsy, MTL-1 also showed excellent anti-epileptic activity.
RI PT
4. MTL-1 nicely interacted with the active site of COX-2 which was confirmed by molecular docking and MD simulation studies.
AC C
EP
TE D
M AN U
SC
5. It also improved cognition impairment in kindled rats and appeared non toxic in sub-acute toxicity study.