ArticlePDF Available

Discovery of novel 2,6-disubstituted pyridazinone derivatives as acetylcholinesterase inhibitors

Authors:

Abstract and Figures

2,6-Disubstituted pyridazinone 4 was identified by HTS as a novel acetylcholinesterase (AChE) inhibitor. Under SAR development, compound 17e stood out as displaying high AChE inhibitory activity and AChE/butyrylcholinesterase (BuChE) selectivity in vitro. Docking studies revealed that 17e might interact with the catalytic active site (CAS) and the peripheral anionic site (PAS) simultaneously. Based on this novel binding information, 6-ortho-tolylamino and N-ethyl-N-isopropylacetamide substituted piperidine were disclosed as new PAS and CAS binders.
Content may be subject to copyright.
Original article
Discovery of novel 2,6-disubstituted pyridazinone derivatives
as acetylcholinesterase inhibitors
Weiqiang Xing
a
,
1
,YanFu
b
,
1
, Zhangxing Shi
a
, Dong Lu
a
, Haiyan Zhang
b
,
*
, Youhong Hu
a
,
*
a
State Key Laboratory of Drug Research, Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences,
555 Zu Chong Zhi Road, Shanghai 201203, China
b
State Key Laboratory of Drug Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences,
555 Zu Chong Zhi Road, Shanghai 201203, China
article info
Article history:
Received 3 November 2012
Received in revised form
24 January 2013
Accepted 27 January 2013
Available online 8 February 2013
Keywords:
Pyridazinone
Acetylcholinesterase
Alzheimers disease
Docking study
abstract
2,6-Disubstituted pyridazinone 4was identied by HTS as a novel acetylcholinesterase (AChE) inhibitor.
Under SAR development, compound 17 e stood out as displaying high AChE inhibitory activity and AChE/
butyrylcholinesterase (BuChE) selectivity in vitro. Docking studies revealed that 17e might interact with
the catalytic active site (CAS) and the peripheral anionic site (PAS) simultaneously. Based on this novel
binding information, 6-ortho-tolylamino and N-ethyl-N-isopropylacetamide substituted piperidine were
disclosed as new PAS and CAS binders.
Ó2013 Elsevier Masson SAS. All rights reserved.
1. Introduction
Alzheimers disease (AD) is a complex neurodegenerative dis-
order of the central nervous system. It is estimated that nearly 36
million people worldwide are now suffering from AD, and the
gure would be increased to about 66 million by 2030 if no
breakthroughs were made. Acetylcholinesterase (AChE), a serine
protease, is responsible for acetylcholine hydrolysis and plays a
fundamental role in impulse transmission by terminating the
action of the neurotransmitter acetylcholine at the cholinergic
synapses and neuromuscular junction [1]. Among the various
approaches for treating AD, inhibition of AChE is still prevailing in
treating or alleviating the symptoms of AD. Tacrine (1,Fig. 1), a
nonselective AChE/butyrylcholinesterase (BuChE) inhibitor, was
the rst drug approved by FDA in 1993. Other selective inhibitors,
such as donepezil (2,Fig.1), also reached the market sequentially. In
recent years, novel AChE inhibitors are continuingly discovered
from natural resources or by synthetic approaches, bearing such as
berberine [2e6], coumarin [7,8], benzofuran [9],
b
-carboline [10],
quinoline [11e13], benzophenone [14,15], triazin [16], ferulic acid
[17] and naphthyridine [18] frameworks as the primary pharma-
cophoric scaffolds.
The most interesting structural character of AChE protein is the
presence of a deep narrow gorge, at the bottom of which a triad lies
in the catalytic anionic site (CAS). Besides, the peripheral anionic
site (PAS) lies at the entrance of the gorge as a regulatorysite. Based
on these ndings, Pang et al. rst reported a tacrine dimer 3(Fig. 1)
as a bivalent AChE inhibitor which interacts with CAS and PAS
simultaneously [19]. This compound displayed much higher AChE
inhibitory activity and specicity over BuChE than the parent
compound 1. Through this strategy, the second generation AChE
inhibitors are developed by utilizing one or two known scaffolds of
AChE inhibitor and most of them possess elevated potency and
improved AChE/BuChE selectivity prole [20e28].
Through an in vitro HTS campaign with diverse compounds
libraries inhouse, pyridazinone derivative 4(Fig. 1) stood out as a
potential AChE inhibitor with an IC
50
of 1.66
m
M. To our best
knowledge, only a few compounds bearing this scaffold were
reported with a liner pyridazine-donepezil hybrid 5(Fig. 1) in the
literature [29e31], which behaved as a dual site binding inhibitor.
By optimizing the 3,6-substitutions on the pyridazine ring, high
AChE afnity of the compound was achieved. However, this type of
molecules displayed low AChE/BuChE selectivity, which might lead
*Corresponding authors.
E-mail addresses: hzhang@mail.shcnc.ac.cn (H. Zhang), yhhu@mail.shcnc.ac.cn
(Y. Hu).
1
Authors equally contributed to this work.
Contents lists available at SciVerse ScienceDirect
European Journal of Medicinal Chemistry
journal homepage: http://www.elsevier.com/locate/ejmech
0223-5234/$ esee front matter Ó2013 Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.ejmech.2013.01.056
European Journal of Medicinal Chemistry 63 (2013) 95e103
to undesirable peripheral side effects. As the compound 4bears
different substituents on pyridazinone ring from 5a/5b and does
not share any structural similarity with donepezil, we realized that
the SAR of 4might not parallel with that of 5a/5b or donepezil, thus
leading this de novo pyridazinone molecule to be worth of opti-
mization for nding diverse AChE inhibitors with high potency and
AChE/BuChE selectivity.
2. Results and discussion
2.1. Chemistry
The general synthetic route to build the focused AChE-targeted
inhibitor library is illustrated in Scheme 1. Compound 9could be
obtained by a sequential process involving alkylation of corre-
sponding pyridazinone (6)[32e34] and followed by reduction of
the nitro group under Fe/HOAc in reuxing EtOH. The nal products
were obtained by 9condensing the substituted piperidines in the
presence of triphosgene.
2.2. SAR study and docking study
Structural inspections of the compounds in our HTS library
revealed that the terminal 4-diethyl amide piperidine in 4may be
essential for activity. Firstly, we commenced with our work by
changing the substituents on the pyridazinone ring (Table 1).
Replacing the phenyl in 4by a single hydrogen (10) caused a slight
loss in activity, which indicated that the phenyl might function as a
hydrophobic pharmacophore. As the AChE binding pocket is a
narrow hydrophobic gorge, a small hydrophobic methyl group was
added to the phenyl (11aec) for modulating the binding afnity.
Compound 11c,ameta-methyl substituted pattern, displayed a
slight enhancement in activity with an IC
50
of 0.746
m
M. Besides,
other meta-halo substituted analogs (11d ee) remained biological
activity at enzymatic level. When the conjugated system in 4was
extended by replacing the phenyl with naphthyl (12aeb), or by
adding a cyano group on meta position (12c), the potency of com-
pounds was evidently enhanced. The most promising compound
12b displayed a potent activity with an IC
50
of 0.188
m
M, which was
about 9 folds more potent than the no-conjugation extended
compound 4.
In order to get a comprehensive understanding of the SAR,
docking study was performed. Crystal complex (code: 1EVE) of
donepezil and AChE protein was downloaded from Protein Data
Bank, in which the positively charged piperidine nitrogen in
donepezil achieves a caution-
p
interaction with Phe330 and the
terminal benzyl ring involves in
p
e
p
stacking with aromatic Trp84.
Docking results showed that 12b may function as a bi-functional
AChE inhibitor with novel binding conformation, which the naph-
thyl occupies in the PAS pocket and the 4-diethyl amide piperidine
is located at the CAS near the bottom of the gorge. The carbonyl
oxygen on pyridazinone ring possibly takes part in hydrogen
bonding interaction with Phe288 and the 2-benzyl onpyridazinone
involves in
p
e
p
stacking with Tyr334 (Fig. 2). No direct hydrogen
bond interaction was observed in the urea motif of 12b, however,
just as donepezil complex, a water bridged hydrogen bonding
might occur in biological circumstances.
Fig. 1. Representative AChE inhibitors.
Scheme 1. Synthesis of analogues
a
.
Table 1
AChE inhibitory activity of 6-aryl substituted pyridazinones.
No. R IC
50
(
m
M) No. R IC
50
(
m
M)
41.66 11d 0.623
10 H 4.90 11e 0.648
11a 2.61 12a 0.462
11b 2.01 12b 0.188
11c 0.746 12c 0.220
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e10396
As shown in Fig. 3, docking results revealed that the naphthyl for
PAS binder in 12b not only acts as a hydrophobic group involving in
the van der Waals interactions with the protein, but also functions
as a
p
e
p
interaction element, which mainly works by the extended
aromatic ring B. Considering the hydrophobic effects induced by
the phenyl A in 12b and the methyl in 11c , a novel 6-aniline pyr-
idazinone scaffold was designed, in which the ring B is remained as
a
p
e
p
interaction motif and ring A is broken by a single nitrogen
linker. In this new scaffold,
p
e
p
stacking efciency of ring B may
also be elevated due to the increased rotation freedom.
Based on above analyses, 6-substituted aniline pyridazinones
were synthesized. As shown in Table 2, when the naphthyl
(hydrophobic-
p
motif) in 12b was replaced by a single aniline
(nitrogen-
p
motif, 13), the AChE inhibitory activity of 13 decreased
drastically (about 17 folds). However, the activity of 14 was restored
when the aniline was changed to more hydrophobic naphthalen-1-
amine, which indicated that the hydrophobic substitutions on the
aniline ring played a great part in binding afnity. Subsequently,
other analogs with different substituted anilines were synthesized
(15aef). The activity of compounds was decreased in the presence
of para substitutions (15c,f). The ortho substitutions signicantly
increased the binding afnity probably due to a similar hydro-
phobic effect induced by ring A in 12b. In addition, a prevailing
Magic Methyleffect emerged in these pyridazinone compounds
[35], a single 2-methyl aniline derivative (15a) showed the prom-
inent inhibitory potency with an IC
50
of 0.049
m
M. It was about 34
and 4-fold more potent than 4and 12b, respectively. Further
enlarging the steric hindrance to dimethyl, ethyl or isopropyl (16ae
c) decreased the activity.
Docking studies showed that 15a displays a similar binding
mode as 12b (Fig. 4A). Overlapping compounds 15a,4and 12b
(Fig. 4B) revealed that changing the phenyl in 4with naphthyl (12b)
or adding a conjugated group (12c) made it possible for
p
e
p
stacking with Trp279, which led to an increase in activity. In the
case of 15a , the 2-methyl aniline does act as a bi-functional group:
the aromatic phenyl stacks with Trp279 while the terminal methyl
t within the PAS hydrophobic pocket perfectly.
After the modication of the active compounds for PAS binding
site, we optimized the CAS binding motifs to synthesize compounds
17a ef(Table 3). Cyclic analog 17a with the terminal morpholine
motif lost the activity totally. Among the acyclic amide series (17b e
f), the inhibitory activity of the compounds strongly paralleled with
the hydrophobic effect of the substitutions. Among them, the N-
ethyl-N-isopropyl amide derivative 17e showed slightly more
potent than 15a . This phenomenon is consistent with the hydro-
phobic environment of the CAS binding pocket.
2.3. Selectivity prole for AChE/BuChE
Moreover, the most potent and representative compounds were
selected for evaluation of AChE/BuChE selectivity prole. As shown
in Table 4, compared with the reported 3-amino-6-aryl pyridazine
series [29,30], 2,6-disubstituted pyridazin-3-one series displayed
about 200-fold or higher AChE/BuChE selectivity. For this scaffold,
AChE inhibitory activity of the compounds could be enhanced by
modifying PAS binding motif without affecting the BuChE afnity
by comparing 4with 12b,or13 with 15 a. In the catalytic binding
pocket, increasing the hydrophobic groups of the amide (17d ee)
lowered the IC
50
s for AChE accompanying with slight increasing
for the binding afnities of BuChE, However, the selectivity proles
were still maintained.
Fig. 2. Model of compound 12a bound to AChE. The naphthyl and 4-substituted
piperidine interact with PAS and CAS respectively.
Fig. 3. Designing strategy for aniline series.
Table 2
AChE inhibitory activity of 6-aniline substituted pyridazinones.
No. Ar IC
50
(
m
M) No. Ar IC
50
(
m
M)
13 3.28 15d 0.119
14 0.126 15e 0.135
15a 0.049 15f 4.84
15b 1.42 16a 0.607
15c 19.8 16b 0.188
16c 1.50
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103 97
3. Conclusion
In this report, a series of novel 2,6-disubstituted pyridazinone
derivatives for AChE inhibition was optimized. Under the SAR
development, high AChE afnity of the compounds was achieved
by optimizing different substituents on the pyridazinone ring,
without sacricing the AChE/BuChE selectivity prole.
Docking study revealed that these pyridazinones might behave
as dual binding site inhibitors with novel binding conformation.
Utilizing this information, a delicate SAR study was performed at
PAS binding site and 6-ortho-tolylamino substitution was opti-
mized for involving in both
p
e
p
stacking and Magic Methyl
hydrophobic interaction. Moreover, rather than the prevailing N-
benzyl piperidine reported in donepezil, a novel hydrophobic 4-
substituted piperidine motif for catalytic pocket binder was dis-
closed, which also provided a new platform for designing AChE
inhibitor. Further biological evaluations of these pyridazinone ser-
ies are ongoing.
4. Experimental section
4.1. General information
Reagents were puried prior to use unless otherwise stated.
Column chromatography was carried out on silica gel (200e
300 mesh).
1
H NMR and
13
C NMR spectral data were recorded in
DMSO-d
6
,CD
3
OD or acetone-d
6
on Varian Mercury 500, 400 or 300
NMR spectrometer and Chemical shifts (
d
) were reported in parts
per million (ppm), and the signals were described as brs (broad
singlet), d (doublet), dd (doublet of doublet), m (multiple), q
(quarter), s (singlet), and t (triplet). Coupling constants (Jvalues)
were given in Hz. Low-resolution mass spectra (ESI) was obtained
using Agilent HPLC-MS (1260-6120B) and high-resolution mass
spectra (ESI) were obtained using Waters Q-Tof Ultima apparatus.
4.2. General procedures
To a solution of pyridazinone derivative [32e34] (0.5 mmol) in
DMF (10 mL) was added 1-(chloromethyl) 3-nitrobenzene
(0.52 mmol) and Cs
2
CO
3
(0.55 mmol), the resulting reaction mix-
ture was stirred at 40e50
C until no starting materials was
detected by TLC (about 3 h). The solvent was removed under
reduced pressure and the residue was dissolved in EtOAc (30 mL),
washed with brine (3 10 mL). The organic layer was dried over
anhydrous Na
2
SO
4
and concentrated in vacuo. The crude product
was dissolved in 95% ethanol (50 mL) containing 10 mmol acetic
acid. Iron powder (2 mmol) was added and the resulting mixture
Fig. 4. (A) Model of compound 15 a bound to AChE. The ortho-tolylamino and
4-substituted piperidine interact with PAS and CAS, respectively. (B) Overlap of 4
(magenta), 12b (yellow), and 15a (green). The naphthyl in 12b and ortho-methyl amine
in 15a function as
p
e
p
stacking element and involve in PAS van der Waals interaction.
Table 3
SAR study at catalytic binding site.
No. R IC
50
(
m
M)
17a NA
17b 7.60
17c 0.473
17d 0.046
15a 0.050
17e 0.028
17f 0.116
Table 4
Selectivity prole of representative compounds for AChE versus BuChE.
No. IC
50
(
m
M) Ratio of IC
50
(BuChE/AChE)
AChE BuChE
41.66 >40 >24
12b 0.188 >40 >212
13 3.28 >40 >12
15a 0.049 >40 >816
17c 0.473 >40 >84
17d 0.046 10.0 217
17e 0.028 5.50 196
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e10398
was stirred for 5 h. After cooled to room temperature, the reaction
mixture was ltered through celite and the lter cake was washed
with 95% ethanol (3 15 mL). The combined ethanol layers were
evaporated in vacuo and the residue was re-dissolved in ethyl
acetate (30 mL). The organic layer was washed with brine
(3 10 mL) and 2 M NaOH (10 mL) sequentially. The organic layer
was dried over anhydrous Na
2
SO
4,
evaporated in vacuo to afford the
crude 2-aminobenzyl-6-substituted-pyridazin-3(2H)-ones, which
were used without further purication.
To a stirred solution of 2-aminobenzyl-6-substituted-pyridazin-
3(2H)-one and triphosgene (1 mmol) in dry dichloromethane
(5 mL) was added triethylamine (2 mmol) under nitrogen atmos-
phere. A solution of the corresponding piperidine (1 mmol) in
dichloromethane (5 mL) was added 5e10 min later and the mixture
was stirred at room temperature overnight. The reaction mixture
was diluted with dichloromethane (15 mL) and washed with water
(3 20 mL). The organic phases were separated, dried over anhy-
drous Na
2
SO
4
and concentrated in vacuo. The residue was puried
by column chromatography to afford the products.
4.2.1. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-phenylpyridazin-1(6H)-yl)
methyl)phenyl)piperidine-1,4-dicarboxamide (4)
Yield: 81%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.03e7.87 (m, 4H),
7.61e7.38 (m, 5H), 7.19 (t, J¼7.9 Hz, 1H), 7.04 (d, J¼7.7 Hz, 1H), 6.99
(d, J¼9.9 Hz,1H), 5.32 (s, 2H), 4.30e4.15 (m, 2H), 3.47e3.37 (m, 2H),
3.32 (d, J¼6.8 Hz, 2H), 2.99e2.74 (m, 3H), 1.68 (s, 4H), 1.18 (t,
J¼7.0 Hz, 3H), 1.03 (t, J¼6.9 Hz, 3H);
13
C NMR (125 MHz,MeOD-d
4
)
d
174.67,160.25, 156.20, 145.43, 139.91, 136.53, 134.31, 131.02, 129.41,
129.22, 128.51, 128.35, 125.74, 122.58, 120.41, 120.20, 55.33 (eCH
2
e
Phe), 43.31 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.84 (eCON(CH
2
CH
3
)
2
),
40.29 (eCON(CH
2
CH
3
)
2
), 38.32 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.39
(eNHCON(CH
2
CH
2
)
2
CHCOe), 13.85 (eCON(CH
2
CH
3
)
2
), 11.90 (e
CON(CH
2
CH
3
)
2
); HRMS(ESI): m/z[M þH]
þ
488; HRMS(ESI) calcd
for C
28
H
33
N
5
O
3
Na [M þNa]
þ
510.2481, found 510.2492.
4.2.2. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxopyridazin-1(6H)-yl)methyl)
phenyl)piperidine-1,4-dicarboxamide (10)
Yield: 58%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.06 (s, 1H), 7.82
(dd, J¼3.6, 1.4 Hz, 1H), 7.53 (d, J¼8.2 Hz,1H), 7.47 (s,1H), 7.33(dd,
J¼9.5, 3.8 Hz, 1H), 7.16 (t, J¼7.9 Hz, 1H), 6.94 (d, J¼7.7 Hz, 1H),
6.87 (dd, J¼9.5, 1.6 Hz, 1H), 5.21 (s, 2H), 4.29e4.15 (m, 2H), 3.43
(q, J¼7.1 Hz, 2H), 3.32 (q, J¼7.0 Hz, 2H), 2.98e2.77 (m, 3H), 1.73e
1.59 (m, 4H), 1.18 (t, J¼7.1 Hz, 3H), 1.03 (t, J¼7.0 Hz, 3H);
13
C
NMR (125 MHz, MeOD-d
4
)
d
174.73, 161.04, 156.17, 139.92, 137.38,
136.56,132.36, 129.08, 128.25, 122.32, 120.22,120.04, 54.88 (eCH
2
e
Phe), 43.29 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.81 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.28 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.39 (eNHCON(CH
2
CH
2
)
2
CHCOe), 13.73 (eCON(CH
2
CH
3
)
2
), 11.78
(eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z412 [M þH]
þ
; HRMS(ESI) calcd
for C
22
H
30
N
5
O
3
[M þH]
þ
412.2349, found 412.2331.
4.2.3. Diethyl-N
1
-(3-((6-oxo-3-(o-tolyl)pyridazin-1(6H)-yl)methyl)
phenyl)piperidine-1,4-dicarboxamide (11a)
Yield: 69%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.01 (s, 1H), 7.53 (t,
J¼8.3 Hz, 3H), 7.41 (d, J¼7.1 Hz, 1H), 7.35e7.23 (m, 3H), 7.17 (t,
J¼7.7 Hz, 1H), 7.03e6.92 (m, 2H), 5.27 (s, 2H), 4.31e4.16 (m, 2H),
3.42 (q, J¼7.0 Hz, 2H), 3.34e3.26 (m, 2H), 2.87 (ddd, J¼21.2, 14.9,
7.1 Hz, 3H), 2.32 (s, 3H), 1.75e1.61 (m, 4H),1.17 (t, J¼7.1 Hz, 3H),1.02
(t, J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.72, 159.99,
156.17, 147.71, 139.97, 136.68, 135.94, 134.90, 134.25, 130.60, 128.91,
128.80, 128.71,128.29, 125.72, 122.49,120.40, 120.06, 55.02 (eCH
2
e
Phe), 43.30 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.29 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.39
(eNHCON(CH
2
CH
2
)
2
CHCOe), 19.20 (eNHePheCH
3
), 13.73 (e
CON(CH
2
CH
3
)
2
), 11.79 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
502; HRMS(ESI) calcd for C
29
H
35
N
5
O
3
Na [M þNa]
þ
524.2638, found
524.2657.
4.2.4. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(p-tolyl)pyridazin-1(6H)-yl)
methyl)phenyl)piperidine-1,4-dicarboxamide(11 b )
Yield: 61%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.02 (s, 1H), 7.90 (d,
J¼9.7 Hz, 1H), 7.78 (d, J¼8.3 Hz, 2H), 7.53 (d, J¼10.8 Hz, 2H), 7.27
(d, J¼8.0 Hz, 2H), 7.17 (t, J¼7.8 Hz, 1H), 7.02 (d, J¼7.4 Hz, 1H), 6.95
(d, J¼9.7 Hz, 1H), 5.29 (s, 2H), 4.30e4.15 (m, 2H), 3.42 (q, J¼7.2 Hz,
2H), 3.31 (q, J¼7.0 Hz, 2H), 2.96e2.73 (m, 3H), 2.35 (s, 3H),1.71e1.58
(m, 4H), 1.17 (t, J¼7.1 Hz, 3H), 1.02 (t, J¼7.0 Hz, 3H);
13
C NMR
(126 MHz, MeOD-d
4
)
d
174.72, 160.21, 156.21, 145.44, 139.94, 139.53,
136.66, 131.53, 130.93,129.26, 129.11, 128.29, 125.62, 122.50, 120.31,
120.13, 55.18 (eCH
2
ePhe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe),
41.79 (eCON(CH
2
CH
3
)
2
), 40.24 (eCON(CH
2
CH
3
)
2
), 38.28 (eNHCON
(CH
2
CH
2
)
2
CHCOe), 28.37 (eNHCON(CH
2
CH
2
)
2
CHCOe), 19.80 (e
NHePheCH
3
), 13.71 (eCON(CH
2
CH
3
)
2
), 11.77 (eCON(CH
2
CH
3
)
2
);
LCMS(ESI) m/z[M þH]
þ
502; HRMS(ESI) calcd for C
29
H
35
N
5
O
3
Na
[M þNa]
þ
524.2638, found 524.2622.
4.2.5. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(m-tolyl)pyridazin-1(6H)-yl)
methyl)phenyl)piperidine-1,4-dicarboxamide (11 c )
Yield: 73%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.08 (s, 1H), 7.88 (d,
J¼9.8 Hz, 1H), 7.72 (s, 1H), 7.65 (d, J¼7.8 Hz, 1H), 7.61 (s, 1H), 7.51
(d, J¼8.8 Hz, 1H), 7.32 (t, J¼7.7 Hz,1H), 7.22 (d, J¼7.6 Hz, 1H), 7.17
(t, J¼7.9 Hz,1H), 7.01 (d, J¼7.5 Hz,1H), 6.94 (d, J¼9.7 Hz, 1H), 5.29
(s, 2H), 4.28e4.15 (m, 2H), 3.47e3.28 (m, 4H), 3.00e2.71 (m, 3H),
2.37 (s, 3H), 1.75e1.55 (m, 4H), 1.22e1.10 (t, J¼7.0 Hz, 3H), 1.01 (t,
J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.66, 160.15,
156.08, 145.40, 140.02, 138.35, 136.63, 134.19, 131.01, 129.91, 129.23,
128.40,128.33, 126.24,122.88, 122.43,120.22, 120.00, 55.24 (eCH
2
e
Phe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.24 (eCON(CH
2
CH
3
)
2
), 38.25 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.39 (eNHCON(CH
2
CH
2
)
2
CHCOe), 20.13 (eNHePheCH
3
e), 13.79
(eCON(CH
2
CH
3
)
2
), 11.86 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z
[M þH]
þ
502; HRMS(ESI) calcd for C
29
H
35
N
5
O
3
Na [M þNa]
þ
524.2638, found 524.2637.
4.2.6. N
1
-(3-((3-(3-Bromophenyl)-6-oxopyridazin-1(6H)-yl)
methyl)phenyl)-N
4
,N
4
-diethylpiperidine-1,4-dicarboxamide (11d)
Yield: 86%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.08 (s, 1H), 8.03e
7.93 (m, 2H), 7.90 (d, J¼8.0 Hz, 1H), 7.60 (s, 2H), 7.51 (d, J¼8.1 Hz,
1H), 7.42 (t, J¼7.7 Hz, 1H), 7.18 (t, J¼7.7 Hz, 1H), 7.00 (t, J¼9.3 Hz,
2H), 5.31 (s, 2H), 4.28e4.15 (m, 2H), 3.42 (d, J¼7.0 Hz, 2H), 3.31 (d,
J¼7.2 Hz, 2H), 3.01e2.76 (m, 3H), 1.67 (s, 4H), 1.17 (t, J¼6.8 Hz, 3H),
1.02 (t, J¼6.9 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.72,
160.14, 156.16, 143.77, 139.99, 136.47, 132.03,130.76, 130.25, 129.50,
128.54, 128.33, 124.49, 122.54, 120.30, 120.12, 55.28 (eCH
2
ePhe),
43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
), 40.25
(eCON(CH
2
CH
3
)
2
), 38.27 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.38 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 13.72 (eCON(CH
2
CH
3
)
2
), 11.78 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
566; HRMS(ESI) calcd
for C
28
H
32
N
5
O
3
NaBr [M þNa]
þ
588.1586, found 588.1569.
4.2.7. N
1
-(3-((3-(3-Chlorophenyl)-6-oxopyridazin-1(6H)-yl)
methyl)phenyl)-N
4
,N
4
-diethylpiperidine-1,4-dicarboxamide (11e)
Yield: 92%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.06 (s, 1H), 7.96 (d,
J¼9.8 Hz, 2H), 7.85 (d, J¼7.1 Hz, 1H), 7.63 (s, 1H), 7.55e7.43 (m, 3H),
7.18 (t, J¼7.8 Hz, 1H), 7.02 (t, J¼8.1 Hz, 2H), 5.32 (s, 2H), 4.30e4.19
(m, 2H), 3.50e3.25 (m, 4H), 3.01e2.75 (m, 3H), 1.78e1.60 (m, 4H),
1.18 (dd, J¼8.3, 5.8 Hz, 3H), 1.03 (t, J¼7.0 Hz, 3H);
13
C NMR
(125 MHz, MeOD-d
4
)
d
174.72, 160.16, 156.18, 143.90, 139.98, 136.47,
136.29, 134.54, 130.79, 130.03, 129.50, 129.05, 128.32, 125.61,
124.07, 122.55, 120.33, 120.13, 55.28 (eCH
2
ePhe), 43.27 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 41.79 (eCON(CH
2
CH
3
)
2
), 40.24 (e
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103 99
CON(CH
2
CH
3
)
2
), 38.27 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.37 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 13.71 (eCON(CH
2
CH
3
)
2
), 11.77 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
522; HRMS(ESI) calcd
for C
28
H
32
N
5
O
3
NaCl [M þNa]
þ
544.2091, found 544.2084.
4.2.8. N
4
,N
4
-Diethyl-N
1
-(3-((3-(naphthalen-2-yl)-6-oxopyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (12a)
Yield: 88%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.39 (s, 1H), 8.25e
7.70 (m, 6H), 7.68 (s, 1H), 7.53 (d, J¼6.3 Hz, 3H), 7.19 (t, J¼7.8 Hz,
1H), 7.06 (d, J¼7.6 Hz, 1H), 6.98 (d, J¼9.8 Hz, 1H), 5.33 (s, 2H),
4.30e4.15 (m, 2H), 3.44e3.29 (m, 4H), 2.98e2.73 (m, 3H), 1.68 (m,
4H), 1.23e1.09 (t, J¼7.0 Hz, 3H), 1.02 (t, J¼7.0 Hz, 3H);
13
C NMR
(125 MHz, MeOD-d
4
)
d
174.69, 160.16, 156.18, 145.07, 140.00, 136.63,
133.66, 133.14, 131.52, 130.89,129.24, 128.33, 128.22, 128.20,127.23,
126.61, 126.21,125.33, 122.82, 122.57, 120.33, 120.09, 55.24 (eCH
2
e
Phe), 43.27 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.78 (eCON(CH
2
CH
3
)
2
),
40.23 (eCON(CH
2
CH
3
)
2
), 38.25 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.36 (eNHCON(CH
2
CH
2
)
2
CHCOe), 13.70 (eCON(CH
2
CH
3
)
2
), 11.77
(eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
538; HRMS(ESI) calcd
for C
32
H
35
N
5
O
3
Na [M þNa]
þ
560.2638, found 560.2643.
4.2.9. N
4
,N
4
-Diethyl-N
1
-(3-((3-(naphthalen-1-yl)-6-oxopyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (12b)
Yield: 76%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.19e7.87 (m, 4H),
7.7 2e7.46 (m, 7H), 7.22 (t, J¼7.8 Hz, 1H), 7.03 (q, J¼8.7 Hz, 2H), 5.34
(s, 2H), 4.30e4.16 (m, 2H), 3.47e3.27 (m, 4H), 3.15e2.25 (m, 3H),
1.80e1.50(m, 4H), 1.23e1.12 (t, J¼7.0 Hz, 3H), 1.03 (t, J¼7.0 Hz, 3H);
13
C NMR (100 MHz, MeOD-d
4
)
d
174.74, 160.15, 156.21,
147.00,140.08, 136.73,134.90, 133.93,132.83,130.49,129.56,129.22,
128.46, 128.20,127.13, 126.62, 125.93,124.87,124.70, 122.86, 120.70,
120.26, 55.04 (eCH
2
ePhe), 43.35 (eNHCON(CH
2
CH
2
)
2
CHCOe),
41.88 (eCON(CH
2
CH
3
)
2
), 40.34 (eCON(CH
2
CH
3
)
2
), 38.36 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 28.46 (eNHCON(CH
2
CH
2
)
2
CHCOe),
13.86 (eCON(CH
2
CH
3
)
2
), 11.91 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z
[M þH]
þ
538; HRMS(ESI) calcd for C
32
H
35
N
5
O
3
Na [M þNa]
þ
560.2638, found 560.2631.
4.2.10. N
1
-(3-((3-(3-Cyanophenyl)-6-oxopyridazin-1(6H)-yl)
methyl)phenyl)-N
4
,N
4
-diethyl piperidine-1,4-dicarboxamide (12c)
Yield: 79%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.31 (s, 1H), 8.23 (d,
J¼7.8Hz, 1H), 8.05 (s, 1H), 8.01 (s,1H), 7.82 (d, J¼7.6 Hz, 1H), 7.70e
7.65 (m, 2H), 7.48 (d, J¼7.9 Hz, 1H), 7.19 (t, J¼7.8 Hz, 1H), 7.05 (d,
J¼7.7Hz, 1H), 7.00 (d, J¼9.8 Hz, 1H), 4.30e4.17 (m, 2H), 3.43e3.25
(m, 4H), 3.00e2.80 (m, 3H),1.70e1.60 (m, 4H), 1.12 (t, J¼7.0 Hz, 3H),
1.03(t, J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.71,160.04,
156.10, 143.14, 140.06, 136.39, 135.65, 132.42, 130.56, 130.07, 129.64,
129.27, 128.36, 122.58, 120.29, 120.01, 117.87, 112.71, 55.26 (eCH
2
e
Phe), 43.29 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.27 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.39
(eNHCON(CH
2
CH
2
)
2
CHCOe), 13.74 (eCON(CH
2
CH
3
)
2
), 11.80 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
513; HRMS(ESI) calcd for
C
29
H
32
N
6
O
3
Na [M þNa]
þ
535.2434, found 535.2439.
4.2.11. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(phenylamino)pyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (13 )
Yield: 64%:
1
H NMR (300 MHz, CD
3
OD-d
4
)
d
7.47 (d, J¼8.2 Hz,
2H), 7.41 (s, 1H), 7.33 (d, J¼8.7 Hz, 1H), 7.25e7.07 (m, 4H), 7.08 (d,
J¼7.6 Hz,1H), 6.92 (t, J¼7.8 Hz, 2H), 5.20 (s, 2H), 4.30e4.15 (m, 2H),
3.50e3.30 (m, 4H), 3.05e2.72 (m, 3H), 1.80e1.64 (m, 4H), 1.23 (t,
J¼7.0 Hz, 3H),1.10 (t, J¼7.1 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.74, 158.48, 156.24, 145.41, 140.36, 139.85, 137.09, 130.15, 128.22,
128.17, 122.72, 121.03,120.72, 120.05, 117.66, 101.92, 54.02 (eCH
2
e
Phe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.79 (eCON(CH
2
CH
3
)
2
),
40.24 (eCON(CH
2
CH
3
)
2
), 38.30 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.38
(eNHCON(CH
2
CH
2
)
2
CHCOe), 13.69 (eCON(CH
2
CH
3
)
2
), 11.75 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
503; HRMS(ESI) calcd
for C
28
H
34
N
6
O
3
Na [M þNa]
þ
525.2590, found 525.2618.
4.2.12. N
4
,N
4
-Diethyl-N
1
-(3-((3-(naphthalen-1-ylamino)-6-
oxopyridazin-1(6H)-yl)methyl)phenyl) piperidine-1,4-
dicarboxamide (14)
Yield: 81%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.23 (d, J¼8.8 Hz,
1H), 8.16 (s, 1H), 8.03 (s,1H), 7.99e7.91 (m, 1H), 7.88 (d, J¼4.7 Hz,
1H), 7.63e7.38 (m, 7H), 7.18 (t, J¼7.8 Hz,1H), 6.99 (d, J¼7.8 Hz, 1H),
6.87 (d, J¼9.8 Hz, 1H), 5.10 (s, 2H), 4.30e4.15 (m, 2H), 3.48e3.39
(m, 2H), 3.39e3.25 (m, 2H), 3.00e2.18 (m, 3H), 1.79e1.62 (m, 4H),
1.19 (t, J¼6.8 Hz, 3H), 1.04 (t, J¼7.1 Hz, 3H);
13
C NMR (125 MHz,
MeOD-d
4
)
d
174.84, 158.86, 156.34, 146.75, 139.88, 137.14, 135.47,
134.45, 130.31, 128.29, 128.10, 128.03, 127.27, 125.56, 125.40,125.33,
123.40, 122.83, 121.69, 120.85, 120.16, 117.56, 54.11 (eCH
2
ePhe),
43.39 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.91 (eCON(CH
2
CH
3
)
2
), 40.36
(eCON(CH
2
CH
3
)
2
), 38.41 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.51 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 13.82 (eCON(CH
2
CH
3
)
2
), 11.89 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
553; HRMS(ESI) calcd
for C
32
H
36
N
6
O
3
Na [M þNa]
þ
575.2747, found 575.2739.
4.2.13. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(o-tolylamino)pyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (15a)
Yield: 80%:
1
H NMR (300 MHz, DMSO-d
6
)
d
8.51 (s, 1H), 7.93 (s,
1H), 7.60 (d, J¼9.0 Hz, 1H), 7.38 (m, 2H), 7.31e7.05 (m, 3H), 6.96e
6.79 (m, 3H), 4.20e4.12 (m 2H), 3.40e3.24 (m, 4H), 2.94e2.69 (m,
3H), 2.16 (s, 3H), 1.67e1.44 (m, 4H), 1.13 (t, J¼7.0 Hz, 3H), 0.99 (t,
J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.73, 158.67,
156.21, 146.39, 139.77, 137.96, 137.05, 130.17, 130.02, 129.93, 128.15,
127.87, 125.84, 123.29, 122.65, 121.85, 120.75, 120.04, 53.87 (eCH
2
e
Phe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.30 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.40
(eNHCON(CH
2
CH
2
)
2
CHCOe), 16.77 (eNHePheCH
3
), 13.71 (e
CON(CH
2
CH
3
)
2
), 11.77 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
517; HRMS(ESI) calcd for C
29
H
36
N
6
O
3
Na [M þNa]
þ
539.2747, found
539.2742.
4.2.14. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(m-tolylamino)pyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (15b)
Yield: 78%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.15 (s, 1H), 8.01 (s,
1H), 7.56 (s, 2H), 7.40 (s,1H), 7.34 (d, J¼7.9 Hz,1H), 7.26e7.10 (m,3H),
7.07 (d, J¼7.5 Hz, 1H), 6.83 (d, J¼9.8 Hz,1H), 6.74 (d, J¼6.8 Hz,1H),
5.15 (s, 2H), 4.30e4.20 (m, 2H), 3.44 (t, J¼7.0 Hz, 2H), 3.34 (q,
J¼7.0Hz, 2H), 3.00e2.92 (m, 3H), 2.28 (s, 3H),1.75e1.64(m, 4H), 1.20
(d, J¼7.0 Hz, 3H), 1.05 (t, J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-
d
4
)
d
174.66, 158.38, 156.14, 145.39, 140.29, 139.92, 138.00,
137.23, 129.97, 128.30, 128.19, 128.14, 122.79, 121.82, 120.80, 120.09,
118.22, 114.84, 53.96 (eCH
2
ePhe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.81 (eCON(CH
2
CH
3
)
2
), 40.25 (eCON(CH
2
CH
3
)
2
), 38.26
(eNHCON(CH
2
CH
2
)
2
CHCOe), 28.41 (eNHCON(CH
2
CH
2
)
2
CH COe),
20.41 (eNHePheCH
3
), 13.80 (eCON(CH
2
CH
3
)
2
), 11.87 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
517; HRMS(ESI) calcd for
C
29
H
36
N
6
O
3
Na [M þNa]
þ
593.2747, found 593.2744.
4.2.15. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-(p-tolylamino)pyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide (15c)
Yield: 82%:
1
H NMR (300 MHz, DMSO-d
6
)
d
8.88 (s, 1H), 8.49 (s,
1H), 7.45 (s, 1H), 7.40e7.32 (m, 3H), 7.20e7.13 (m, 2H), 7.02 (d,
J¼8.5 Hz, 2H), 6.88e6.78 (m, 2H), 4.15e4.05 (m, 2H), 3.30e3.19 (m,
4H), 2.90e2.70 (m, 3H), 2.19 (s, 3H), 1.65e1.40 (m, 4H), 1.11 (t,
J¼7.1 Hz, 3H), 0.97 (t, J¼7.0 Hz, 3H);
13
C NMR (100 MHz, MeOD-d
4
)
d
174.79,158.51, 156.32, 145.63,139.90, 137.90, 137.17,130.55,130.06,
128.74, 128.28, 128.22, 122.79, 120.78, 120.11, 117.87, 54.12 (eCH
2
e
Phe), 43.36 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.86 (eCON(CH
2
CH
3
)
2
),
40.31(eCON(CH
2
CH
3
)
2
), 38.36 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.46
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103100
(eNHCON(CH
2
CH
2
)
2
CHCOe), 19.40 (eNHePheCH
3
), 13.77 (e
CON(CH
2
CH
3
)
2
), 11.83 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
517; HRMS(ESI) calcd for C
29
H
36
N
6
O
3
Na [M þNa]
þ
593.2747, found
593.2745.
4.2.16. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-((2-(triuoromethyl)phenyl)
amino)pyridazin-1(6H)-yl)methyl)phenyl)piperidine-1,4-
dicarboxamide (15d)
Yield: 75%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.06 (s, 1H), 7.91 (d,
J¼8.4 Hz, 1H), 7.64 (d, J¼7.9 Hz, 1H), 7.61e7.53 (m, 2H), 7.50 (d,
J¼8.1 Hz, 1H), 7.39 (d, J¼9.8 Hz, 2H), 7.25e7.10 (m, 2H), 6.97 (d,
J¼7.6 Hz, 1H), 6.88 (d, J¼9.8 Hz, 1H), 5.07 (s, 2H), 4.30e4.15 (m,
2H), 3.50e3.40 (m, 2H), 3.40e3.25 (m, 2H), 2.90e2.77 (m, 3H),
1.78e1.63 (m, 4H), 1.23e1.14 (t, J¼7.0 Hz, 3H), 1.04 (t, J¼7.0 Hz,
3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.68, 158.74, 156.12, 145.73,
139.89,137.65,136.99, 132.35,130.67,128.23, 127.90, 125.92, 125.88,
125.10, 123.35, 122.67, 121.71, 121.48, 120.76, 120.03, 53.98 (eCH
2
e
Phe), 43.30 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.82 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.29 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.44 (eNHCON(CH
2
CH
2
)
2
CHCOe), 13.80 (eCON(CH
2
CH
3
)
2
), 11.86
(eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
571; HRMS(ESI) calcd
for C
29
H
33
N
6
O
3
NaF
3
[M þNa]
þ
593.2464, found 593.2485.
4.2.17. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-((3-(triuoromethyl)phenyl)
amino)pyridazin-1(6H)-yl) methyl)phenyl)piperidine-1,4-
dicarboxamide (15e)
Yield: 74%:
1
H NMR (400 MHz, CD
3
OD)
d
7.93 (s, 1H), 7.57 (d,
J¼9.2 Hz,1H), 7.46 (s, 1H), 7.32 (t, J¼8.0 Hz, 2H), 7.21 (t, J¼7.8 Hz,
1H), 7.12 (d, J¼7.7 Hz,1H), 7.09 (d, J¼7.6 Hz,1H), 7.00 (d, J¼9.7 Hz,
1H), 6.76(d, J¼9.7 Hz, 1H), 5.12 (s,2H), 4.18 (d, J¼13.2 Hz, 2H), 3.45e
3.34 (m, 2H), 3.30 (t, J¼7.1 Hz, 2H), 2.99e2.82 (m, 2H), 2.82e2.73 (m,
1H), 1.69 (dd, J¼19.6, 8.4 Hz, 4H), 1.17 (t, J¼7.1 Hz, 3H), 1.05 (t,
J¼7.1 Hz, 3H);
13
C NMR (125MHz, MeOD-d
4
)
d
174.63,158.28,156.16,
144.83, 141.02, 139.97, 137.01, 130.45, 129.02, 128.36, 127.95, 125.36,
123.20, 123.03, 121.08, 120.67, 120.38, 117.01, 113.70, 54.15 (eCH
2
e
Phe), 43.31 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.24 (eCON(CH
2
CH
3
)
2
), 38.26 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.40
(eNHCON(CH
2
CH
2
)
2
CHCOe), 13.80 (eCON(CH
2
CH
3
)
2
), 11.87 (e
CON(CH
2
CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
571; HRMS(ESI) calcd for
C
29
H
33
N
6
O
3
NaF
3
[M þNa]
þ
593.2464, found 593.2449.
4.2.18. N
4
,N
4
-Diethyl-N
1
-(3-((6-oxo-3-((4-(triuoromethyl)phenyl)
amino)pyridazin-1(6H)-yl) methyl)phenyl)piperidine-1,4-
dicarboxamide (15f)
Yield: 58%:
1
H NMR (300 MHz, CD
3
OD-d
4
)
d
7.62 (d, J¼8.6 Hz,
2H), 7.54e7.45 (m, 3H), 7.35e7.29 (m, 1H), 7.26 (t, J¼7.7 Hz,1H), 7.18
(d, J¼9.7 Hz,1H), 7.09 (d, J¼7.2 Hz,1H), 6.94 (d, J¼9.7 Hz,1H), 4.25e
4.15 (m, 2H), 3.50e3.31 (m, 4H), 3.32e2.78 (m, 3H), 1.77e1.66 (m,
3H), 1.23 (t, J¼7.1 Hz, 3H),1.09 (t, J¼7.1 Hz, 3H);
13
C NMR (126 MHz,
DMSO-d
6
)
d
173.40, 157.48, 155.21, 155.14, 144.53, 143.87, 143.78,
141.31, 141.21, 137.65, 131.83, 128.75, 128.37, 128.30, 126.40,
126.14, 123.99, 122.11, 120.87, 120.61, 119.99, 119.89, 119.01, 118.91,
117.30, 117.24, 53.73 (eCH
2
ePhe), 43.71 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.48 (eCON(CH
2
CH
3
)
2
), 39.76 (eCON(CH
2
CH
3
)
2
), 37.86
(eNHCON(CH
2
CH
2
)
2
CHCOe), 28.93 (eNHCON(CH
2
CH
2
)
2
CHCOe),
15.34 (eCON(CH
2
CH
3
)
2
), 13.39 (eCON(CH
2
CH
3
)
2
); LCMS(ESI) m/z
[M þH]
þ
571; HRMS(ESI) calcd for C
29
H
33
N
6
O
3
NaF
3
[M þNa]
þ
593.2464, found 593.2482.
4.2.19. N
1
-(3-((3-((2,6-Dimethylphenyl)amino)-6-oxopyridazin-
1(6H)-yl)methyl)phenyl)-N
4
,N
4
-diethylpiperidine-1,4-
dicarboxamide (16a)
Yield: 76%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.01 (s,1H), 7.51 (d,
J¼9.1 Hz, 1H), 7.31 (d, J¼7.0 Hz, 2H), 7.16e7.02 (m, 5H), 6.79 (d,
J¼9.7 Hz, 2H), 4.92 (s, 2H), 4.3e4.15 (m, 2H), 3.51e3.31 (m, 4H),
3.10e2.25 (m, 3H), 2.14 (s, 6H),1.77e1.63 (m, 4H), 1.19 (t, J¼7.0 Hz,
3H), 1.04 (t, J¼7.0 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.74,
158.58, 156.17, 147.34, 139.60, 136.96, 135.97, 135.60, 130.43, 128.02,
127.62, 126.56, 125.95, 122.47, 120.84, 120.01, 53.60 (eCH
2
ePhe),
43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.81 (eCON(CH
2
CH
3
)
2
), 40.25
(eCON(CH
2
CH
3
)
2
), 38.32 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.43 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 17.07, 13.72 (eCON(CH
2
CH
3
)
2
), 11.78 (e
CON(CH
2
CH
3
)
2
); HRMS(ESI) m/z[M þH]
þ
531; HRMS(ESI) calcd
for C
30
H
38
N
6
O
3
Na [M þNa]
þ
553.2903, found 553.2876.
4.2.20. N
4
,N
4
-Diethyl-N
1
-(3-((3-((2-ethylphenyl)amino)-6-
oxopyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-
dicarboxamide (16b)
Yield: 70%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.07 (s, 1H), 7.68 (d,
J¼8.0 Hz, 1H), 7.52 (d, J¼8.8 Hz, 2H), 7.36 (s, 1H), 7.26 (t, J¼6.6 Hz,
1H), 7.21e7.08 (m, 3H), 6.99 (m, 2H), 6.85e6.76 (m, 1H), 5.06 (s, 2H),
4.24 (m, 2H), 3.50e3.28 (m, 4H), 3.10e2.28 (m, 3H), 2.66 (q,
J¼7.5 Hz, 2H), 1.78e1.62 (m, 4H), 1.24e1.09 (m, 6H), 1.15e1.10 (m,
3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.73, 158.68, 156.21,
146.84, 139.75, 137.22, 137.06, 136.55, 130.19, 128.25, 128.13,
127.68, 125.83, 123.96, 123.11, 122.64, 120.75, 120.04, 53.86 (eCH
2
e
Phe), 43.28 (eNHCON(CH
2
CH
2
)
2
CHCOe), 41.80 (eCON(CH
2
CH
3
)
2
),
40.25 (eCON(CH
2
CH
3
)
2
), 38.31 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.41 (eNHCON(CH
2
CH
2
)
2
CHCOe), 23.76 (eNHePheCH
2
CH
3
),
13.71 (eCON(CH
2
CH
3
)
2
), 13.16 (eNHePheCH
2
CH
3
), 11.78 (e
CON(CH
2
CH
3
)
2
); HRMS(ESI) m/z[M þH]
þ
531; HRMS(ESI) calcd
for C
30
H
38
N
6
O
3
Na [M þNa]
þ
553.2903, found 553.2903.
4.2.21. N
4
,N
4
-Diethyl-N
1
-(3-((3-((2-isopropylphenyl)amino)-6-
oxopyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-
dicarboxamide (16c)
Yield: 89%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.10 (s, 1H), 7.49 (m,
4H), 7.27 (d, J¼7.4 Hz, 1H) , 7.2 2 (d, J¼9.8 Hz, 1H), 7.18e7.01 (m, 3H),
6.91 (d, J¼7.5 Hz, 1H), 6.79 (d, J¼9.8 Hz,1H), 5.03 (s, 2H), 4.30e4.15
(m,2H),3.50e3.25(m,5H),2.99e2.73 (m, 3H), 1.75e1.61 (m, 4H) ,
1.2 5e1.05 (m, 9H), 1.02 (t, J¼7.0 Hz , 3H);
13
C NMR (125 MHz, MeOD-
d
4
)
d
174.73, 158.71, 156.21, 147.50, 142.44, 139.71, 137.06, 136.38,
130.20, 128.11, 127.46, 125.67, 125.31, 124.83, 124.61, 122.46, 120.59,
120.00, 53.92 (eCH
2
ePhe), 43.27 (eNHCON(CH
2
CH
2
)
2
CHCOe),
41.80 (eCON(CH
2
CH
3
)
2
), 40.24 (eCON(CH
2
CH
3
)
2
), 38.30 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 28.40 (eNHCON(CH
2
CH
2
)
2
CHCOe), 27.21
(PheCH(CH
3
)
2
e), 22.22 (PheCH(CH
3
)
2
e), 13.71 (eCON(CH
2
CH
3
)
2
),
11.77 (eCON(CH
2
CH
3
)
2
); HRMS(ESI) m/z[M þH]
þ
545; HRMS(ESI)
calcd for C
31
H
40
N
6
O
3
Na [M þH]
þ
567.3060, found 567.3058.
4.2.22. 4-(Morpholine-4-carbonyl)-N-(3-((6-oxo-3-(o-tolylamino)
pyridazin-1(6H)-yl)methyl) phenyl)piperidine-1-carboxamide (17a)
Yield: 89%:
1
H NMR (300 MHz, acetone-d
6
)
d
7.65 (s,1H), 7.37 (d,
J¼7.7 Hz, 1H), 7.20e7.15 (d, J¼9.2 Hz, 2H), 6.92 (d, J¼9.6 Hz, 2H),
6.85e6.70 (m, 3H), 6.64e6.51 (m, 2H), 6.46 (d, J¼9.5 Hz,1H), 3.90e
3.75 (m, 2H), 3.30e3.20 (s, 4H), 3.19e3.08 (m, 2H), 2.57e2.48 (m,
5H), 1.88 (s, 3H), 1.75e1.60 (m, 4H);
13
C NMR (126 MHz, MeOD-d
4
)
d
174.05, 158.78, 156.32, 146.49, 139.86, 138.07, 137.17, 130.29, 130.13,
130.03,128.28, 127.99, 125.95, 123.39,122.78, 121.95, 120.89, 120.16,
66.61 (CON(CH
2
CH
2
)
2
O), 66.41(CON(CH
2
CH
2
)
2
O), 53.99 (eCH
2
e
Phe), 45.83 (CON(CH
2
CH
2
)
2
O), 43.35 (eNHCON(CH
2
CH
2
)
2
CHCOe),
42.05 (CON(CH
2
CH
2
)
2
O), 37.75 (eNHCON(CH
2
CH
2
)
2
CHCOe),
28.21 (eNHCON(CH
2
CH
2
)
2
CHCOe); LCMS(ESI) m/z[M þH]
þ
531;
HRMS(ESI) calcd for C
29
H
34
N
6
O
4
Na [M þNa]
þ
553.2539, found
553.2514.
4.2.23. N
4
,N
4
-Dimethyl-N
1
-(3-((6-oxo-3-(o-tolylamino)pyridazin-
1(6H)-yl)methyl)phenyl) piperidine-1,4-dicarboxamide(17b)
Yield: 93%:
1
H NMR (300 MHz, acetone-d
6
)
d
8.04 (s, 1H), 7.72 (d,
J¼8.7 Hz, 1H), 7.51 (m, 2H), 7.33 (s, 1H), 7.28 (d, J¼9.8 Hz, 1H),
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103 101
7.21e7.09 (m, 3H), 7.00e6.70 (m, 2H), 6.82 (d, J¼9.8 Hz, 1H), 5.07
(s, 2H), 4.30e4.15 (m, 2H), 3.10 (s, 3H), 2.92e2.80 (m, 3H), 2.86 (s,
3H), 2.24 (s, 3H), 1.76e1.54 (m, 4H);
13
C NMR (125 MHz, MeOD-d
4
)
d
175.45, 158.78, 156.32, 146.49, 139.89, 138.07, 137.17, 130.29, 130.14,
130.04,128.27,128.00, 125.95,123.40, 122.75, 121.96,120.88,120.14,
53.98 (eCH
2
ePhe), 43.40 (eNHCON(CH
2
CH
2
)
2
CHCOe), 38.25 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 36.19 (eCON(CH
3
)
2
), 34.65 (e
CON(CH
3
)
2
), 28.01 (eNHCON(CH
2
CH
2
)
2
CHCOe), 16.90; LCMS(ESI)
m/z[M þH]
þ
489; HRMS(ESI) calcd for C
27
H
32
N
6
O
3
Na [M þNa]
þ
511.2434, found 511.2453.
4.2.24. N
4
-Ethyl-N
4
-methyl-N
1
-(3-((6-oxo-3-(o-tolylamino)
pyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-
dicarboxamide(17c )
Yield: 75%:
1
H NMR (300 MHz, CD
3
OD)
d
7.48 (d, J¼8.0 Hz, 1H),
7.31 (m, 2H), 7.22 (t, J¼7.9 Hz, 2H), 7.18e7.05 (m, 2H), 6.96 (t,
J¼7.8 Hz, 2H), 6.91e6.82 (m, 1H), 5.09 (s, 2H), 4.25e4.10 (m, 2H),
3.52e3.27 (m, 2H), 3.08 (s, 1.5H), 3.03e2.78 (m, 4.5H), 2.18 (s, 3H),
1.80e1.58 (m, 4H),1.21 (t, J¼7.1 Hz,1.5H),1.08 (t, J¼7.1 H z, 1. 5H) ;
13
C
NMR (125 MHz, MeOD-d
4
)
d
175.27, 174.89, 158.78, 156.32, 146.48,
139.89, 138.08, 137.18, 130.29, 130.14, 130.01, 128.27, 127.98,
125.96, 123.39, 122.76, 121.94, 120.86, 120.15, 53.98 (eCH
2
ePhe),
44.14 (eCON(CH
3
)CH
2
CH
3
), 43.41 (eNHCON(CH
2
CH
2
)
2
CHCOe),
43.38 (eNHCON(CH
2
CH
2
)
2
CHCOe), 42.53 (eCON(CH
3
)CH
2
CH
3
),
38.45 (eNHCON(CH
2
CH
2
)
2
CHCOe), 38.20 (eNHCON(CH
2
CH
2
)
2
CHCOe), 33.79 (eCON(CH
3
)CH
2
CH
3
), 32.17 (eCON(CH
3
)CH
2
CH
3
),
28.54 (eNHCON(CH
2
CH
2
)
2
CHCOe), 27.95 (eNHCON(CH
2
CH
2
)
2
CHCOe), 16.89, 13.02 (eCON(CH
3
)CH
2
CH
3
), 11.04 (eCON(CH
3
)
CH
2
CH
3
); LCMS(ESI) m/z[M þH]
þ
503; HRMS(ESI) calcd for
C
28
H
34
N
6
O
3
Na [M þNa]
þ
525.2590, found 525.2600.
4.2.25. N
4
-Isopropyl-N
4
-methyl-N
1
-(3-((6-oxo-3-(o-tolylamino)
pyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-dicarboxamide
(17d )
Yield: 68%:
1
H NMR (300 MHz, CD
3
OD)
d
7.49 (d, J¼8.0 Hz, 1H),
7.33e7.27 (m, 2H), 7.27e7.19 (m, 2H), 7.19e7.06 (m, 2H), 6.97 (t,
J¼7.7 Hz, 2H), 6.88 (d, J¼9.7 Hz, 1H), 5.10 (s, 2H), 4.80e4.71 (m,
0.5H), 4.39e4.24 (m, 0.5H), 4.25e4.12 (m, 2H), 3.04e2.81 (m, 4.5H),
2.77 (s, 1.5H), 2.19 (s, 3H),1.82e1.59 (m, 4H), 1.24 (d, J¼6.6 Hz, 3H),
1.10 (d, J¼6.8 Hz, 3H);
13
C NMR (125 MHz, MeOD-d
4
)
d
174.92,
174.88, 158.78, 156.33, 146.49, 139.88, 138.07, 137.17, 130.28,
130.13, 130.04, 128.26, 127.97, 125.95, 123.40, 122.76, 121.97,
120.87, 120.16, 53.97 (eCH
2
ePhe), 44.38 (eCON(CH
3
)CH(CH
3
)
2
),
43.44 (eNHCON(CH
2
CH
2
)
2
CHCOe), 43.40 (eNHCON (CH
2
CH
2
)
2
CH
COe), 39.00 (eNHCON(CH
2
CH
2
)
2
CHCOe), 38.43 (eNHCON(CH
2
CH
2
)
2
CHCOe), 28.65 (eNHCON(CH
2
CH
2
)
2
CHCOe), 27.99 (eNHCON
(CH
2
CH
2
)
2
CHCOe), 27.24 (eCO N(CH
3
)CH(CH
3
)
2
), 25.43 (eCON
(CH
3
)CH(CH
3
)
2
), 19.49 (eCON(CH
3
)CH(CH
3
)
2
), 18.10 (eCON(CH
3
)
CH(CH
3
)
2
), 16.88 (eNHePheCH
3
); LCMS(ESI) m/z[M þH]
þ
517;
HRMS(ESI) calcd for C
29
H
36
N
6
O
3
Na [M þNa]
þ
539.2747, found
539.2733.
4.2.26. N
4
-Ethyl-N
4
-isopropyl-N
1
-(3-((6-oxo-3-(o-tolylamino)
pyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-
dicarboxamide(17e )
Yield: 55%:
1
H NMR (300 MHz, CD
3
OD)
d
7.49 (d, J¼8.0 Hz, 1H),
7.33 (d, J¼7.8 Hz, 2H), 7.29e7.19 (m, 2H), 7.19e7.05 (m, 2H), 6.98 (d,
J¼7.6 Hz, 2H), 6.88 (d, J¼9.7 Hz, 1H), 5.10 (s, 2H), 4.65e4.50 (m,
0.5H), 4.30e4.12 (m, 2.5H), 3.43e3.20 (m, 2H), 3.10e2.75 (m, 3H),
2.19 (s, 3H), 1.90e1.75 (m, 4H),1.28e1.08 (m, 9H);
13
C NMR (125MHz,
MeOD-d
4
)
d
175.20, 174.34, 158.63, 156.15, 146.35, 139.79, 137.97,
137.03, 130.16, 130.06, 129.80, 128.19, 127.93, 125.87, 123.24, 122.62,
121.74, 120.81, 120.03, 53.98 (eCH
2
ePhe), 45.73 (eCON(CH
3
)
CH(CH
3
)
2
), 43.32 (eNHCON(CH
2
CH
2
)
2
CHCOe), 42.02 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 39.25 (eNHCON(CH
2
CH
2
)
2
CHCOe),
38.43 (eNHCON(CH
2
CH
2
)
2
CHCOe), 37.02 (eCON(CH
2
CH
3
)
CH(CH
3
)
2
), 35.27 (eCON(CH
2
CH
3
)CH(CH
3
)
2
), 28.52 (eNHCO
N(CH
2
CH
2
)
2
CHCOe), 28.50 (eNHCON(CH
2
CH
2
)
2
CHCOe), 20.27 (e
CON(CH
2
CH
3
)CH(CH
3
)
2
), 19.13 (eCON(CH
2
CH
3
)CH(CH
3
)
2
), 16.87 (e
NHePheCH
3
), 16.17 (eCON(CH
2
CH
3
)CH(CH
3
)
2
), 13.71 (e
CON(CH
2
CH
3
)CH(CH
3
)
2
); LCMS(ESI) m/z[M þH]
þ
531; HRMS(ESI)
calcd for C
30
H
38
N
6
O
3
Na [M þNa]
þ
553.2903, found 553.2926.
4.2.27. N
4
,N
4
-Diisopropyl-N
1
-(3-((6-oxo-3-(o-tolylamino)
pyridazin-1(6H)-yl)methyl) phenyl)piperidine-1,4-dicarboxamide
(17f )
Yield: 62%:
1
H NMR (300 MHz, acetone-d
6
)
d
7.97 (s,1H), 7.77e7.66
(m,1H), 7.51 (m, 2H), 7.34e7.21(m,2H),7.21e7.10 (m, 3H), 6.99e6.70
(m, 2H), 6.81 (d, J¼9.8 Hz, 1H), 5.06 (s, 2H),4.25e4.11(m,4H),3.55(s,
1H), 3.01e2.74 (m, 3H), 2.23 (s, 3H), 1.74e1.62 (m, 4H), 1. 36e1.19 ( m,
12H) ;
13
C NMR (125 MHz, MeOD-d
4
)
d
158.80, 156.36, 146.51,
139.88, 138.07, 137.17, 130.29, 130.12, 130.08, 128.25, 127.97, 125.94,
123.41, 122.76, 122.00, 120.85, 120.16, 53.97 (eCH
2
ePhe), 45.77 (e
CON(CH(CH
3
)
2
)
2
), 43.43 (eNHCON(CH
2
CH
2
)
2
CHCOe), 39.84 (e
NHCON(CH
2
CH
2
)
2
CHCOe), 28.56 (eNHCON(CH
2
CH
2
)
2
CHCOe),
19.52 (eCON(CH(CH
3
)
2
)
2
), 16.86; LCMS(ESI) m/z[M þH]
þ
545;
HRMS(ESI) calcd for C
31
H
40
N
6
O
3
Na [M þNa]
þ
567.3060, found
567.3073.
5. Materials and methods
5.1. Biological assays
Cholinesterase (ChE) activity was evaluated using modied
Ellmans spectrophotometric method [36]. Individual compounds
were dissolved in dimethyl sulfoxide (DMSO) to a concentration of
10 mM and diluted to appropriate concentrations in double-
distilled water. For in vitro tests of inhibition, 4 mL reaction mix-
tures consisting 0.1 mL of test compound, selective substrate
[0.6 mL acetylthiocholine iodide for AChE (2 mM) or 0.8 mL S-
butyrylthiocholine iodide for BuChE (2 mM)], 1 mL of phosphate
buffered solution (0.1 mM), and 0.1 mL of enzyme (homogenate of
rat cerebral cortex for AChE, ratserum for BuChE) were incubated at
37
C for 8 min, and the reaction was terminated with 1 mL of 3%
(w/v) sodium dodecylsulfate (SDS). Finally,1 mL of 0.2% (w/v) 5,5
0
-
Dithiobis (2-nitrobenzoic acid) (DTNB) was added, and the yellow
anion, 5-thio-2-nitrobenzate, was measured at 440 nm. All samples
were assayed in duplicate.
5.2. Docking
The crystal structure of donepezileAChE complex (code ID:
1EVE) was downloaded from the Protein Data Bank. Further
preparation of the protein included deleting the waters, addition of
hydrogen atoms and applying CHARMm forceeld. The 3D Struc-
tures of 4,12b and 15a were built and performed geometry opti-
mization by molecular mechanics.
Docking studies were performed using the CDOCKER protocol of
Discovery Studio 2.1 program. The binding sphere was generated at
the center of the donepezil with a radius of 11.5 Å. Then the ligand
donepezil was deleted and the docking results were obtained using
the default settings except the value of random conformations was
set to 300 and the value of top hits was set to 20. The docking
results were analyzed using Discovery Studio 3.5 Visualizer.
Acknowledgments
This work was supported by grant from National Science &
Technology Major Project Key New Drug Creation and Manu-
facturing Program(2012ZX09301001-001).
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103102
Appendix A. Supplementary data
Supplementary data related to this article can be found at http://
dx.doi.org/10.1016/j.ejmech.2013.01.056.
References
[1] G. Zimmerman, H. Soreq, Termination and beyond: acetylcholinesterase as a
modulator of synaptic transmission, Cell. Tissue Res. 326 (2006) 655e669.
[2] H.T. Xiao, J. Peng, Y. Liang, J. Yang, X. Bai, X.Y. Hao, F.-M. Yang, Q.Y. Sun,
Acetylcholinesterase inhibitors from Corydalis yanhusuo, Nat. Prod. Res. 25
(2011) 1418e1422.
[3] S.F. Tsai, S.S. Lee, Characterization of acetylcholinesterase inhibitory con-
stituents from annona glabra assisted by HPLC microfractionation, J. Nat. Prod.
73 (2010) 1632e1635.
[4] L. Huang, Z. Luo, F. He, A. Shi, F. Qin, X. Li, Berberine derivatives, with sub-
stituted amino groups linked at the 9-position, as inhibitors of acetylcholi-
nesterase/butyrylcholinesterase, Bioorg. Med. Chem. Lett. 20 (2010) 6649e
6652.
[5] L. Huang, A. Shi, F. He, X. Li, Synthesis, biological evaluation, and molecular
modeling of berberine derivatives as potent acetylcholinesterase inhibitors,
Bioorg. Med. Chem. 18 (2010) 1244e1251.
[6] W.J. Shan, L. Huang, Q. Zhou, F.C. Meng, X.-S. Li, Synthesis, biological evalua-
tion of 9-N-substituted berberine derivatives as multi-functional agents of
antioxidant, inhibitors of acetylcholinesterase, butyrylcholinesterase and
amyloid-
b
aggregation, Eur. J. Med. Chem. 46 (2011) 5885e5893.
[7] L. Piazzi, A. Cavalli, F. Colizzi, F. Belluti, M. Bartolini, F. Mancini, M. Recanatini,
V. Andrisano, A. Rampa, Multi-target-directed coumarin derivatives: hAChE
and BACE1 inhibitors as potential anti-Alzheimer compounds, Bioorg. Med.
Chem. Lett. 18 (2008) 423e426.
[8] X. Zhou, X.B. Wang, T. Wang, L.Y. Kong, Design, synthesis, and acetylcholi-
nesterase inhibitory activity of novel coumarin analogues, Bioorg. Med. Chem.
16 (2008) 8011e8021.
[9] J. Liu, V. Dumontet, A.L. Simonin, B.I. Iorga, V. Guerineau, M. Litaudon,
V.H. Nguyen, F. Gueritte, Benzofurans from styrax agrestis as acetylcholines-
terase inhibitors: structureeactivity relationships and molecular modeling
studies, J. Nat. Prod. 74 (2011) 2081e2088.
[10] Y. Rook, K.U. Schmidtke, F. Gaube, D. Schepmann, B. Wuensch, J. Heilmann,
J. Lehmann, T. Winckler, Bivalent
b
-carbolines as potential multitarget anti-
Alzheimer agents, J. Med. Chem. 53 (2010) 3611e3617.
[11] H. Zheng, M.B.H. Youdim, M. Fridkin, Selective acetylcholinesterase inhibitor
activated by acetylcholinesterase releases an active chelator with neuro-
rescuing and anti-amyloid activities, ACS Chem. Neurosci. 1 (2010) 737e746.
[12] H. Zheng, M.B.H. Youdim, M. Fridkin, Site-activated multifunctional chelator
with acetylcholinesterase and neuroprotectiveeneurorestorative moieties for
Alzheimers therapy, J. Med. Chem. 52 (2009) 4095e4098.
[13] C. Martins, M.C. Carreiras, R. León, C. de los Ríos, M. Bartolini, V. Andrisano,
I. Iriepa, I. Moraleda, E. Gálvez, M. García, J. Egea, A. Samadi, M. Chioua, J. Marco-
Contelles, Synthesis and biological assessment of diversely substituted furo
[2,3-b]quinolin-4-amine and pyrrolo[2,3-b]quinolin-4-amine derivatives, as
novel tacrine analogues, Eur. J. Med. Chem. 46 (2011) 6119e6130.
[14] F. Belluti, L. Piazzi, A. Bisi, S. Gobbi, M. Bartolini, A. Cavalli, P. Valenti, A. Rampa,
Design, synthesis, and evaluation of benzophenone derivatives as novel ace-
tylcholinesterase inhibitors, Eur. J. Med. Chem. 44 (2009) 1341e1348.
[15] F. Belluti, M. Bartolini, G. Bottegoni, A. Bisi, A. Cavalli, V. Andrisano, A. Rampa,
Benzophenone-based derivatives: a novel series of potent and selective dual
inhibitors of acetylcholinesterase and acetylcholinesterase-induced beta-
amyloid aggregation, Eur. J. Med. Chem. 46 (2011) 1682e1693.
[16] M. Catto, A.A. Berezin, D. Lo Re, G. Loizou, M. Demetriades, A. De Stradis,
F. Campagna, P.A. Koutentis, A. Carotti, Design, synthesis and biological
evaluation of benzo[e][1,2,4]triazin-7(1H)-one and [1,2,4]-triazino[5,6,1-jk]
carbazol-6-one derivatives as dual inhibitors of beta-amyloid aggregation and
acetyl/butyryl cholinesterase, Eur. J. Med. Chem. 58 (2012) 84e97.
[17] Y. Chen, J. Su, L. Fang, M. Liu, S. Peng, H. Liao, J. Lehmann, Y. Zhang, Tacrine-
ferulic acid-nitric oxide (NO) donor trihybrids as potent, multifunctional acetyl-
and butyrylcholinesterase inhibitors, J. Med. Chem. 55 (2012) 4309e4321.
[18] A. Samadi, C. de los Ríos, I. Bolea, M. Chioua, I. Iriepa, I. Moraleda, M. Bartolini,
V. Andrisano, E. Gálvez, C. Valderas, M. Unzeta, J. Marco-Contelles, Multi-
potent MAO and cholinesterase inhibitors for the treatment of Alzheimers
disease: synthesis, pharmacological analysis and molecular modeling of het-
erocyclic substituted alkyl and cycloalkyl propargyl amine, Eur. J. Med. Chem.
52 (2012) 251e262.
[19] Y.P. Pang, P. Quiram, T. Jelacic, F. Hong, S. Brimijoin, Highly potent, selective,
and low cost bis-tetrahydroaminacrine inhibitors of acetylcholinesterase,
J. Biol. Chem. 271 (1996) 23646e23649.
[20] F. Leonetti, M. Catto, O. Nicolotti, L. Pisani, A. Cappa, A. Stefanachi, A. Carotti,
Homo- and hetero-bivalent edrophonium-like ammonium salts as highly
potent, dual binding site AChE inhibitors, Bioorg. Med. Chem. 16 (2008)
7450e7456.
[21] X. He, S. Feng, Z.F. Wang, Y. Shi, S. Zheng, Y. Xia, H. Jiang, X.-C. Tang, D. Bai,
Study on dual-site inhibitors of acetylcholinesterase: highly potent deriva-
tives of bis- and bifunctional huperzine B, Bioorg. Med. Chem. 15 (2007)
1394e1408.
[22] S. Gemma, E. Gabellieri, P. Huleatt, C. Fattorusso, M. Borriello, B. Catalanotti,
S. Butini, A.M. De, E. Novellino, V. Nacci, T. Belinskaya, A. Saxena, G. Campiani,
Discovery of huperzine a-tacrine hybrids as potent inhibitors of human
cholinesterases targeting their midgorge recognition sites, J. Med. Chem. 49
(2006) 3421e3425.
[23] S. Feng, Z. Wang, X. He, S. Zheng, Y. Xia, H. Jiang, X. Tang, D. Bai, Bis-huperzine
B: highly potent and selective acetylcholinesterase inhibitors, J. Med. Chem.
48 (2005) 655e657.
[24] P.R. Carlier, Y.F. Han, E.S.H. Chow, C.P.L. Li, H. Wang, T.X. Lieu, H.S. Wong,
Y.P. Pang, Evaluation of short-tether bis-THA acetylcholinesterase inhibitors.
A further test of the dual binding site hypothesis, Bioorg. Med. Chem. 7 (1999)
351e357.
[25] A. Badia, J.E. Banos, P. Camps, J. Contreras, D.M. Gorbig, D. Munoz-Torrero,
M. Simon, N.M. Vivas, Synthesis and evaluation of tacrine-huperzine A hybrids
as acetylcholinesterase inhibitors of potential interest for the treatment of
Alzheimers disease, Bioorg. Med. Chem. 6 (1998) 427e440.
[26] S. Hamulakova, L. Janovec, M. Hrabinova, P. Kristian, K. Kuca, M. Banasova,
J. Imrich, Synthesis, design and biological evaluation of novel highly potent
tacrine congeners for the treatment of Alzheimers disease, Eur. J. Med. Chem.
55 (2012) 23e31.
[27] A. Samadi, M. Estrada, C. Pérez, M.I. Rodríguez-Franco, I. Iriepa, I. Moraleda,
M. Chioua, J. Marco-Contelles, Pyridonepezils, new dual AChE inhibitors as
potential drugs for the treatment of Alzheimers disease: synthesis, bio-
logical assessment, and molecular modeling, Eur. J. Med. Chem. 57 (2012)
296e301.
[28] H. Tang, L.Z. Zhao, H.T. Zhao, S.L. Huang, S.M. Zhong, J.K. Qin, Z.F. Chen,
Z.S. Huang, H. Liang, Hybrids of oxoisoaporphine-tacrine congeners: novel
acetylcholinesterase and acetylcholinesterase-induced
b
-amyloid aggregation
inhibitors, Eur. J. Med. Chem. 46 (2011) 4970e4979.
[29] J.M. Contreras, I. Parrot, W. Sippl, Y.M. Rival, C.G. Wermuth, Design, synthesis,
and structureeactivity relationships of a series of 3-[2-(1-benzylpiperidin-4-
yl)ethylamino] pyridazine derivatives as acetylcholinesterase inhibitors,
J. Med. Chem. 44 (2001) 2707e2718.
[30] J.M. Contreras, Y.M. Rival, S. Chayer, J.J. Bourguignon, C.G. Wermuth, Ami-
nopyridazines as acetylcholinesterase inhibitors, J. Med. Chem. 42 (1999)
730e741.
[31] S. Utku, M. Gokce, I. Orhan, M.F. Sahin, Synthesis of novel 6-substituted-
3(2H)-pyridazinone-2-acetyl-2-(substituted/-nonsubstituted benzal)hydra-
zone derivatives and acetylcholinesrase and butyrylcholinesterase inhibitory
activities in vitro, Arzneimittelforschung 61 (2011) 1e7.
[32] K. Abouzid, M. Abdel Hakeem, O. Khalil, Y. Maklad, Pyridazinone derivatives:
design, synthesis, and in vitro vasorelaxant activity, Bioorg. Med. Chem. 16
(2008) 382e389.
[33] W.J. Coates, A. McKillop, One-pot preparation of 6-substituted 3(2H)-pyr-
idazinones from ketones, Synthesis (1993) 334e342.
[34] S. Lin, Z. Liu, Y. Hu, Microwave-enhanced efcient synthesis of diversied 3,6-
disubstituted pyridazines, J. Comb. Chem. 9 (2007) 742e744.
[35] C.S. Leung, S.S.F. Leung, J. Tirado-Rives, W.L. Jorgensen, Methyl effects on
protein-ligand binding, J. Med. Chem. 55 (2012) 4489e4500.
[36] G.L. Ellman, K.D. Courtney, V. Andres Jr., R.M. Featherstone, A new and rapid
colorimetric determination of acetylcholinesterase activity, Biochem. Phar-
macol. 7 (1961) 88e95.
W. Xing et al. / European Journal of Medicinal Chemistry 63 (2013) 95e103 103
... Applications of substituted pyridazines as antidepressant, antihypertensive, anticonvulsant, antibacterial, diuretic, anti-HIV, anticancer, antiinflammatory, analgesic, cardiovascular, and neuroprotective agents are available in the literature [3][4][5][6][7][8][9][10][11][12][13]. Some examples for medical use of pyridazine derivatives are zardaverine and imazodane as cardiotonic PDE III inhibitors and emorfazone as an analgesic agent ( [14]. ...
... . Xing et al. synthesized 2,6-disubstituted pyridazinone analogs and investigated their acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitor activities. Among them, compound 4 was reported to be highly potent and selective against the cited enzymes (Figure 1)[14]. ...
... J Med Chem 2005;48(6 ):1919-29. Structure of the drugs bearing pyridazinone skeleton and compound 4[14]. ...
Article
Background: The pyridazinone nucleus has been incorporated into a wide variety of therapeutically interesting molecules to transform them into better drugs. Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are known to be serine hydrolase enzymes responsible for the hydrolysis of acetylcholine (ACh). Inhibition of cholinesterases is an effective method to curb Alzheimer's disease. Here, we prepared 12 new 6-substituted-3(2H)-pyridazinone-2-acetyl-2-(nonsubstituted/4-substituted benzenesulfonohydrazide) derivatives and evaluated their inhibitory effects on AChE/BChE in pursuit of potent dual inhibitors for Alzheirmer's Disease. We also tried to get insights into binding interactions of the synthesized compounds in the active site of both enzymes by using molecular docking approach. Method: We obtained our compounds by the reaction of various substituted/nonsubstituted benzenesulfonic acid derivatives with 6-substitutedphenyl-3(2H)-pyridazinone-2-yl acetohydrazide and determined their anticholinesterase activities according to the Ellman's method. Molecular docking studies were done using Glide and the results were evaluated on Maestro (Schrödinger, LLC, New York, NY, 2019). Results: The title compounds showed moderate inhibition at 100 μg/ml against both enzymes, yet with better activity against BChE. Compound VI2a emerged as a dual inhibitor with 25.02% and 51.70% inhibition against AChE and BChE, respectively. Conclusion: This study supports that novel pyridazinone derivates may be used for the development of new BChE inhibitory agents. It was less potent than the reference drugs, yet promising for further modifications as a lead. The ability of the compounds to adopt energetically more favourable conformations and to engage in more key interactions in the ECBChE active gorge explains their better activity profile against ECBChE.
... Pyridazinone derivatives like 2,6-disubstituted-pyridazinones, [1] 3(2H)pyridazinones [2] have appeared as potent Alzheimer's disease (AD) agents. Research on acetylcholinesterase of many structures for example liquiritigenin, deoxyvasicinone hybrids etc. different from pyridazinones against Alzheimer's is ongoing. ...
Article
Full-text available
In this study, a series of N‐substituted‐(p‐tolyl)pyridazin‐3(2H)‐one derivatives were synthesized and evaluated for their AChE inhibitory activity. The chemical structures of novel compounds 5(a–m) were confirmed by ¹H‐NMR, ¹³C‐NMR, IR and HRMS analysis. In order to eliminate the symptomatic effects of Alzheimer's disease, the proposed compounds were evaluated by acetylcholinesterase inhibition activity study in accordance with the cholinergic hypothesis. The results revealed that the N‐substituted‐(p‐tolyl)pyridazin‐3(2H)‐one derivatives inhibited the enzymes significantly. Ki values for acetylcholinesterase in the range of 0.56±0.15–4.12±1.42 μM. Compound 5 h demonstrated the greatest in AChE activity compared with tacrine (0.56±0.15 μM). Molecular docking studies were performed for all compounds that compared tacrine in AChE activity in‐vitro. As a result of molecular docking studies (ΔGBind, docking score, XP Gscore, Glide energy, Glide emodel), 5 f, 5 g and 5 h compounds showed good inhibitory properties in the AChE active site as in silico.
... Acetylcholinesterase (AChE) activity was evaluated according to previously reported protocol [16]. Briefly, the frozen cortex tissues (approximately 20 mg) were homogenized in cold 75 mM sodium phosphate buffer (pH 7.4) with 0.4 mM iso-OMPA. ...
Article
Full-text available
28-O-caffeoyl betulin (B-CA) has been demonstrated to reduce the cerebral infarct volume caused by transient middle cerebral artery occlusion (MCAO) injury. B-CA is a novel derivative of naturally occurring caffeoyl triterpene with little information associated with its pharmacological target(s). To date no data is available regarding the effect of B-CA on brain metabolism. In the present study, a ¹H-NMR-based metabolomics approach was applied to investigate the therapeutic effects of B-CA on brain metabolism following MCAO in rats. Global metabolic profiles of the cortex in acute period (9 h after focal ischemia onset) after MCAO were compared between the groups (sham; MCAO + vehicle; MCAO + B-CA). MCAO induced several changes in the ipsilateral cortex of ischemic rats, which consequently led to the neuronal damage featured with the downregulation of NAA, including energy metabolism dysfunctions, oxidative stress, and neurotransmitter metabolism. Treatment with B-CA showed statistically significant rescue effects on the ischemic cortex of MCAO rats. Specifically, treatment with B-CA ameliorated the energy metabolism dysfunctions (back-regulating the levels of succinate, lactate, BCAAs, and carnitine), oxidative stress (upregulating the level of glutathione), and neurotransmitter metabolism disturbances (back-regulating the levels of γ-aminobutyric acid and acetylcholine) associated with the progression of ischemic stroke. With the administration of B-CA, the levels of three phospholipid related metabolites (O-phosphocholine, O-phosphoethanolamine, sn-glycero-3-phosphocholine) and NAA improved significantly. Overall, our findings suggest that treatment with B-CA may provide neuroprotection by augmenting the metabolic changes observed in the cortex following MCAO in rats.
... anticancer and cytotoxic activity [4][5][6][7][8][9][10][11], antiulcer activity [12], analgesic and anti-inflammatory activity [6,[13][14][15][16][17][18][19][20][21][22], vasorelaxant and platelet antiaggregatory activity [23][24][25][26][27]. Moreover, some derivatives of pyridazinone act as potent carbonic anhydrase [11,28] or acetylcholinesterase [29] inhibitors, histamine H 3 receptor antagonists [30,31] or anticonvulsant agents [32]. ...
Article
Novel Mannich base analogues of pyrrolo[3,4-d]pyridazinone 7a,b-13a,b are designed and synthesized as potential anti-inflammatory agents. The title compounds are obtained via convenient one-pot synthesis with good yields. Their structures and properties are described by spectroscopic techniques and elemental analyses. The aim of this study is to evaluate the inhibitory activity of the new derivatives against both cyclooxygenase isoforms COX1 and COX2 as well as their cytotoxicity. The results clearly indicate that the tested compounds 7a,b-13a,b are not toxic, all show better affinity towards isoform COX-2, and some of them act as selective COX-2 inhibitors. Moreover, every examined derivative of pyrrolo[3,4-d]pyridazinone demonstrates better inhibitory activity towards COX-2 and a superior COX-2/COX-1 selectivity ratio compared to the reference drug meloxicam. Molecular docking studies confirm that compounds 7a,b-13a,b preferably bind COX-2 and all of them bind to the active site of cyclooxygenase in a way very similar to meloxicam. Subsequently, taking into account that inflammation is strongly correlated with oxidative stress and both of these processes can potentiate each other, synthesized Mannich bases are evaluated for potential antioxidant activity. Most of the investigated derivatives reduce induced oxidative and nitrosative stress. Moreover, compounds 7a,b, 8a, 10a,b, 11b, 12a,b-13a,b protect chromatin from oxidative stress and decrease the number of DNA strand breaks caused by intracellular growth of free radicals. Finally, a study of the binding mechanism between compounds 7a,b-13a,b and bovine serum albumin (BSA) was carried out. According to spectroscopic and molecular docking studies, all examined derivatives interact with BSA, which suggests their potential long half-life in vivo.
... Thus, cholinesterase inhibitors are used in the treatment of various neurodegenerative disorders, which occur as a result of reduced cortical and hippocampal levels of ACh. AD, is one of these disorders as it is a complex neurodegenerative disorder characterized by synapse dysfunction, neuronal death, loss of memory and learning ability [1,2,[7][8][9]. Various acetylcholinesterase inhibitors such as tacrine, donepezil, rivastigmine, and galantamine ( Fig. 1) are currently used for the treatment of symptomatic AD [10][11][12][13][14]. ...
Article
Full-text available
Background Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are known serine hydrolase enzymes responsible for the hydrolysis of acetylcholine (ACh), which is a significant neurotransmitter for regulation of cognition in animals. Inhibition of cholinesterases is an effective method to curb Alzheimer’s disease, a progressive and fatal neurological disorder. Docking studies were performed for the compounds F18 and F111 also interaction modes using AChE and BChE enzymes active sites were determined. Objective In this study, 30 new hydrazone derivatives were synthesized. Then, we evaluated their anticholinesterase activity of compounds. We also tried to get insights into binding interactions of the synthesized compounds in the active site of both enzymes by using molecular docking approach. Method The compounds were synthesized by the reaction of various substituted/nonsubstituted benzaldehydes with 6-(substitute/nonsubstituephenyl)-3(2H)-pyridazinone-2-yl propiyohydrazide. Anticholinesterase activity of the compounds were determined according to the Ellman’s method. Molecular docking studies were done by using the ADT package version 1.5.6rc3. RMSD values were obtained using Lamarckian Genetic Algorithm and scoring function of AutoDock 4.2 release 4.2.5.1 software. Results The activities of the compounds were compared with galantamine as cholinesterase enzyme inhibitor, and some of the compounds showed higher BChE inhibitory activity than galantamine. Compound F111 was shown best BChE inhibitor affect in 50 µM dose, 89.43% inhibition of BChE (IC50=4.27±0.36 µM). Conclusion This study supports that novel hydrazone derivates may be used for the development of new BChE inhibitory agents.
Article
Pyridazinone analogs possess diverse types of pharmacological activities, such as anticancer, antimicrobial, anticonvulsant, analgesic, anti-inflammatory, antioxidant, antihypertensive, antisecretory, antiulcer, and other useful pharmacological activities. They also possess cyclooxygenase (COX) inhibitors, dipeptidyl peptidase inhibitors, phosphodiesterase inhibitors, glutamate transporter activators, adenosine receptor antagonists, serotonin receptors antagonists, lipooxygenase, cholinesterase, vasodilator, and anesthetics. Pyridazine rings are the essential structure for some marketed drugs, such as pimobendan, levosimendan as a cardiotonic drug, and emorfozan as an analgesic and anti-inflammatory (Non-steroidal anti-inflammatory drug) agent. So, researchers all over the world have paid attention to synthesizing various pyridazinone compounds mainly due to the ease of design and synthesis of different analogs and variables in the pharmacological responses. This review article focuses on the pharmacological activities of different pyridazine analogs.
Article
A phosphoric acid-catalyzed regio- and enantioselective N2-propargylation of pyridazinones has been achieved, providing chiral N2-alkylated pyridazinones in moderate to good yields (up to 93%) with good enantioselectivities (up to 96%...
Article
A novel series of benzamide derivatives were successively designed and synthesized prepared from the pyridazinone scaffold. Among them, (S)-17b, demonstrated potent inhibitory activity in vitro toward human class I HDAC isoforms and human myelodysplastic syndrome (SKM-1) cell line. Also, (S)-17b strongly increased the intracellular level of acetyl-histone H3 and P21 simultaneously and effectively induced G1 cell cycle arrest and apoptosis. Through oral dosing in SKM-1 xenograft models, (S)-17b exhibited excellent in vivo antitumor activity. In addition, compound (S)-17b showed better antitumor efficacy on mouse models with intact immune system than those with thymus deficiencies. Furthermore, this compound displayed a favorable pharmacokinetic profile in ICR mice and SD rat, respectively, minimal metabolic property differences among hepatocytes from five species, and a low inhibition upon the human ether-a-go-go (hERG) channel with an IC50 value of 34.6 μΜ. This novel compound (S)-17b may serve as a new drug candidate for further investigation.
Article
Full-text available
Pyridazinones are important nitrogen rich heterocyclic core that has been driving the substanial medicinal interest due to their wide range of biological activities. This privileged scaffold forms a central core in numerous potent compounds, which results in the development of novel anticancer drugs with fruitful biological activities. The current review article summarizes the progressive development of novel pyridazinone derivatives that are targets for numerous receptors such as C‐met kinase inhibitors, PARP inhibitor, Tubulin polymerization inhibitors, Dihydro folate reductase inhibitors, B‐Raf inhibitors, Bruton tyrosine kinase inhibitors, FER tyrosine kinase inhibitors, fibroblast growth factor receptors (FGFRs). It features the various simple techniques for the synthesis of pyridazinones and also highlights the mechanistic insights into anticancer properties of pyridazinone derivatives
Article
New ten compounds bearing pyridazinone ring (5a-5j) were designed and synthesized as acetylcholinesterase inhibitors. The new derivatives were acquired via the reaction of propionohydrazides with substituted/nonsubstituted sulphonylchlorides. The structures of the synthesized compounds were explained using FT-IR, ¹H-NMR, ¹³C-NMR, elemental analysis and HRMS spectra. The inhibition profiles of the synthesized compounds on AChE were researched by comparing their IC50 and KI values. According to the activity studies, all the compounds showed significant inhibitory activity against AChE relative to the reference compound Tacrine. The compound 5g showed the best acetylcholinesterase inhibitory effect with a KI value of 11.61 ± 0.77 nM. For all compounds, the parameters of the interaction points on the receptor side were determined on the ligand basis with the 4D-QSAR model. The synthesized pyridazinone derivatives, 5(a-j), were screened for their acetylcholinesterase inhibitory potential, and the results determined that among the series, compounds 5g, 5f and 5j showed the best inhibition, respectively. For anti-Alzheimer activities, 5g, 5f and 5j compounds were performed in silico studies to understand the binding site, binding energy properties in molecular docking.
Article
A one-pot process for the preparation of 6-phenyl-3(2H)-pyridazinone from acetophenone and glyoxylic acid has been investigated and shown to have wide utility in the preparation of 6- and 5,6-substituted 3(2H)-pyridazinones. Limitations to the process encountered with 2'-hydroxyacetophenone and with basic hetero-aromatic ketones have been overcome, and the processes described offer the rapid and efficient synthesis of many 6-substituted pyridazinones from readily available ketones.
Article
Alzheimer's disease (AD) onset and progression are associated with the dysregulation of multiple and complex physiological processes and a successful therapeutic approach should therefore address more than one target. Two new chemical entities, the easily accessible heterocyclic scaffolds 1,3-diphenylbenzo[e][1,2,4]triazin-7(1H)-one (benzotriazinone I) and 2-phenyl-6H-[1,2,4]triazino[5,6,1-jk]carbazol-6-one (triazafluoranthenone II), were explored for their multitarget-directed inhibition of beta-amyloid (Aβ) fibrillization and acetyl- (AChE) and/or butyryl- (BChE) cholinesterase, three valuable targets for AD therapy. Introduction of appropriate amine substituents at positions 6 and 5 on scaffold I and II, respectively, allowed the preparation of a series of compounds that were tested as Aβ(1-40) aggregation and cholinesterase inhibitors. Potent inhibitors of Aβ self-aggregation were discovered and among them benzotriazinone 7 exhibited an outstanding IC(50) equal to 0.37 μM. Compounds bearing a basic amine linked to the heterocyclic scaffold through a linear alkyl chain of varying length also afforded good ChE inhibitors. In particular, benzotriazinone 24 and triazafluoranthenone 38 were endowed with an interesting multiple activity, the former displaying IC(50) values of 1.4, 1.5 and 1.9 μM on Aβ aggregation and AChE and BChE inhibition, respectively, and the latter showing IC(50) values of 1.4 and an outstanding 0.025 μM in the Aβ aggregation and BChE inhibition, respectively. Benzotriazinone 24 and triazafluoranthenone 29, selected owing to their suitable aqueous solubility and Aβ aggregation inhibition, were submitted to a time course kinetic assay followed with thioflavin T (ThT) spectrofluorimetry, circular dichroism (CD) and transmission electron microscopy (TEM). Experimental data indicated that 24 acted at a low concentration ratio (10 μM 24vs. 50 μM Aβ), stabilizing the unstructured Aβ peptide and inhibiting fibrillogenesis, and that 29 also acted as fibrillization inhibitor, but likely enhancing and stabilizing the β-sheet arrangement of Aβ to yield protofibrillar species as detected by TEM.
Article
New tacrine derivatives 5a-d, 6a-d with piperazino-ethyl spacer linked with corresponding secondary amines and tacrine homodimer 8 were synthesized and tested as cholinesterase inhibitors on human acetylcholinesterase (hAChE) and human plasmatic butyrylcholinesterase (hBChE). In most cases the majority of synthesized derivatives exhibit a high AChE and BChE inhibitory activity with IC(50) values in the low-nanomolar range, being clearly more potent than the reference standard tacrine (9-amino-1,2,3,4-tetrahydroacridine, 1) and 7-MEOTA (7-methoxy-9-amino-1,2,3,4-tetrahydroacridine). Among them, inhibitors 8 and 5c, showed a strong inhibitory activity against hAChE, with an IC(50) value of 4.49 nM and 4.97, nM resp., and a high selectivity to hAChE. The compound 5d acted as the most potent inhibitor against hBChE with an IC(50) value of 33.7 nM and exhibited also a good selectivity towards hBChE. The dissociation constants K(i) of the selected inhibitors were compared with their IC(50) values. Molecular modeling studies were performed to predict the binding modes between individual derivatives and hAChE/hBChE.
Article
The finding that acetylcholinesterase (AChE) colocalizes with β-amyloid (Aβ) and promotes and accelerates Aβ aggregation has renewed an intense interest in developing new multifunctional AChE inhibitors as potential disease-modifying drugs for Alzheimer's therapy. To this end, we have developed a new class of selective AChE inhibitors with site-activated chelating activity. The identified lead, HLA20A, exhibits little affinity for metal (Fe, Cu, and Zn) ions but can be activated following inhibition of AChE to liberate an active chelator, HLA20. HLA20 has been shown to possess neuroprotective and neurorescuing activities in vitro and in vivo with the ability to lower amyloid precursor holoprotein (APP) expression and Aβ generation and inhibit Aβ aggregation induced by metal (Fe, Cu, and Zn) ion. HLA20A inhibited AChE in a time and concentration dependent manner with an HLA20A-AChE complex constant (K(i)) of 9.66 × 10(-6) M, a carbamylation rate (k(+2)) of 0.14 min(-1), and a second-order rate (k(i)) of 1.45 × 10 (4) M(-1) min(-1), comparable to those of rivastigmine. HLA20A showed little iron-binding capacity and activity against iron-induced lipid peroxidation (LPO) at concentrations of 1-50 μM, while HLA20 exhibited high potency in iron-binding and in inhibiting iron-induced LPO. At a concentration of 10 μM, HLA20A showed some activity against monoamine oxidase (MAO)-A and -B when tested in rat brain homogenates. Defined restrictively by Lipinski's rules, both HLA20A and HLA20 satisfied drug-like criteria and possible oral and brain permeability, but HLA20A was more lipophilic and considerably less toxic in human SHSY5Y neuroblastoma cells at high concentrations (25 or 50 μM). Together our data suggest that HLA20A may represent a promising lead for further development for Alzheimer's disease therapy.
Article
In search of multifunctional cholinesterase inhibitors as potential anti-Alzheimer drug candidates, tacrine-ferulic acid-NO donor trihybrids were synthesized and tested for their cholinesterase inhibitory activities, release of nitric oxide, vasodilator properties, cognition improving potency, and hepatotoxicity. All of the novel target compounds show higher in vitro cholinesterase inhibitory activity than tacrine. Three selected compounds (3a, 3f, and 3k) produce moderate vasorelaxation in vitro, which correlates with the release of nitric oxide. Compared to its non-nitrate dihybrid analogue (3u), the trihybrid 3f exhibits better performance in improving the scopolamine-induced cognition impairment (mice) and, furthermore, less hepatotoxicity than tacrine.
Article
The synthesis, pharmacological evaluation and molecular modeling of heterocyclic substituted alkyl and cycloalkyl propargyl amines 1-7 of type I, and 9-12 of type II, designed as multipotent inhibitors able to simultaneously inhibit monoamine oxidases (MAO-A/B) as well as cholinesterase (AChE/BuChE) enzymes, as potential drugs for the treatment of Alzheimer's disease, are described. Indole derivatives 1-7 of type I are well known MAO inhibitors whose capacity to inhibit AChE and BuChE was here investigated for the first time. As a result, compound 7 was identified as a MAO-B inhibitor (IC(50) = 31 ± 2 nM) and a moderately selective eqBuChE inhibitor (IC(50) = 4.7 ± 0.2 μM). Conversely, the new and readily available 5-amino-7-(prop-2-yn-1-yl)-6,7,8,9-tetrahydropyrido[2,3-b][1,6]naphthyridine derivatives 9-13 of type II are poor MAO inhibitors, but showed AChE selective inhibition, compound 12 being the most attractive as it acts as a non-competitive inhibitor on EeAChE (IC(50) = 25 ± 3 nM, K(i) = 65 nM). The ability of this compound to interact with the AChE peripheral binding site was confirmed by kinetic studies and by molecular modeling investigation. Studies on human ChEs confirmed that 12 is a selective AChE inhibitor with inhibitory potency in the submicromolar range. Moreover, in agreement with its mode of action, 12 was shown to be able to inhibit Aβ aggregation induced by hAChE by 30.6%.
Article
The effects of addition of a methyl group to a lead compound on biological activity are examined. A literature analysis of >2000 cases reveals that an activity boost of a factor of 10 or more is found with an 8% frequency, and a 100-fold boost is a 1 in 200 event. Four cases in the latter category are analyzed in depth to elucidate any unusual aspects of the protein-ligand binding, distribution of water molecules, and changes in conformational energetics. The analyses include Monte Carlo/free-energy perturbation (MC/FEP) calculations for methyl replacements in inhibitor series for p38α MAP kinase, ACK1, PTP1B, and thrombin. Methyl substitutions ortho to an aryl ring can be particularly effective at improving activity by inducing a propitious conformational change. The greatest improvements in activity arise from coupling the conformational gain with the burial of the methyl group in a hydrophobic region of the protein.
Article
A photometric method for determining acetylcholinesterase activity of tissue extracts, homogenates, cell suspensions, etc., has been described. The enzyme activity is measured by following the increase of yellow color produced from thiocholine when it reacts with dithiobisnitrobenzoate ion. It is based on coupling of these reactions: The latter reaction is rapid and the assay is sensitive (i.e. a 10 μ1 sample of blood is adequate). The use of a recorder has been most helpful, but is not essential. The method has been used to study the enzyme in human erythrocytes and homogenates of rat brain, kidney, lungs, liver and muscle tissue. Kinetic constants determined by this system for erythrocyte eholinesterase are presented. The data obtained with acetylthiocholine as substrate are similar to those with acetylcholine.
Article
A series of 9-N-substituted berberine derivatives were synthesized and biologically evaluated as antioxidant and inhibitors of acetylcholinesterase (AChE), butyrylcholinesterase and amyloid-β aggregation. Most of these compounds exhibited very good antioxidant activities, inhibitive activities of AChE and amyloid-β aggregation. Among them, compound 8d, (o-methylphenethyl)amino linked at the 9-position of berberine, was found to be a good antioxidant (with 4.05 μM of Trolox equivalents), potent inhibitor of AChE (an IC(50) value of 0.027 μM), and high active inhibitor of amyloid-β aggregation (an IC(50) value of 2.73 μM).