Bioorganic & Medicinal Chemistry: A B C D e F F G G H B

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Bioorganic & Medicinal Chemistry 27 (2019) 115148

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


journal homepage: www.elsevier.com/locate/bmc

Design, synthesis, in vitro, and in silico studies of novel diarylimidazole-1,2, T


3-triazole hybrids as potent α-glucosidase inhibitors
Mina Saeedia,b, Maryam Mohammadi-Khanaposhtanic, Mohammad Sadegh Asgarid,
Nafiseh Eghbalnejade, Somaye Imanparastf, Mohammad Ali Faramarzif, Bagher Larijanig,
Mohammad Mahdavig, , Tahmineh Akbarzadehh,b,
⁎ ⁎

a
Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
b
Persian Medicine and Pharmacy Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
c
Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
d
School of Chemistry, College of Science, University of Tehran, Tehran, Iran
e
Faculty of Pharmacy, International Campus (TUMS-IC), Tehran University of Medical Sciences, Tehran, Iran
f
Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
g
Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
h
Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran

ARTICLE INFO ABSTRACT

This paper is dedicated to our unique teacher in In this work, new derivatives of diarylimidazole-1,2,3-triazole 7a-p were designed, synthesized, and evaluated
chemistry and medicinal chemistry, Professor for their in vitro α-glucosidase inhibitory activity. All compounds showed potent inhibitory activity in the range
Abbas Shafiee (1937–2016) of IC50 = 90.4–246.7 µM comparing with acarbose as the standard drug (IC50 = 750.0 µM). Among the syn-
thesized compounds, compounds 7b, 7c, and 7e were approximately 8 times more potent than acarbose. The
Keywords: kinetic study of those compounds indicated that they acted as the competitive inhibitors of α-glucosidase.
Diarylimidazole Molecular docking studies were also carried out for compounds 7b, 7c, and 7e using modeled α-glucosidase to
Docking study
find the interaction modes responsible for the desired inhibitory activity.
α-Glucosidase inhibitors
Kinetic study
1,2,3-triazole

1. Introduction Imidazole and its derivatives are one of the most important nitrogen
containing heterocyclic scaffolds in medicinal chemistry11 possessing
α-Glucosidase is an intestinal enzyme which catalyzes the breakage versatile biological properties including anticancer, analgesic, anti-
of the 1,4-α glycosidic bonds of polysaccharides and some disaccharides tubercular, anti-inflammatory, anti-Alzheimer’s, antiviral, and antic-
converting them into the absorbable glucose.1 This enzyme is involved onvulsant activities.12–18 Also, recent studies have confirmed α-gluco-
in various carbohydrate-related diseases such as diabetes, viral infec- sidase inhibitory activity of imidazole derivatives,19–21 e.g. 2-(4,5-
tions, cancer, and pompe disease.2–5 In this regard, various α-glucosi- diphenyl-1H-imidazol-2-yl)phenol A showed good α-glucosidase in-
dase inhibitors such as acarbose (Glucobay), miglitol (Glyset), and vo- hibitory activity (IC50 = 74.3 μM) comparing with acarbose
glibose (Volix, Basen) are currently prescribed for the treatment of (IC50 = 38.2 μM).19 1,2,3-Triazole and its derivatives have been also
patients with non-insulin-dependent (type 2) diabetes. The above found as privileged scaffolds for drug discovery developments due to
mentioned α-glucosidase inhibitors contain sugar moieties and their stable metabolism, less adverse, high selectivity, and user-friendly
synthesis need complicated multistep procedures.6 On the other hand, synthesis via click reaction.22 In recent years, several hybrid derivatives
administration of these inhibitors can bring undesirable side effects containing 1,2,3-triazole ring23–26 such as chalcone-1,2,3-triazole B
including serious gastrointestinal disorders such as diarrhea and flatu- (IC50 = 67.8 μM comparing with acarbose IC50 = 23.9 μM) possessing
lence.7 Thus, the development of efficient small molecules possessing α-glucosidase inhibitory activity have been reported.23
potent α-glucosidase inhibitory activity has recently attracted a great Molecular hybridization has been a strong tool for design and synthesis
attention.8–10 of new bioactive compounds. It is expected that combination of various


Corresponding authors at: Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran (T. Akbarzadeh).
E-mail addresses: momahdavi@tums.ac.ir (M. Mahdavi), akbarzad@tums.ac.ir (T. Akbarzadeh).

https://doi.org/10.1016/j.bmc.2019.115148
Received 21 May 2019; Received in revised form 8 September 2019; Accepted 1 October 2019
Available online 15 October 2019
0968-0896/ © 2019 Elsevier Ltd. All rights reserved.
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

Fig. 1. Design of diarylimidazole-1,2,3-triazole hybrids.

pharmacophores with different mechanisms of action can be helpful for derivative 1, ammonium acetate 2, and 4-(prop-2-ynyloxy) benzaldehyde
inducing favorite biological activity. In this respect, in view of α-glucosi- derivative 3 in acetic acid was heated at reflux for 1 h to afford compound
dase inhibitory activity of compounds A and B, a novel series of diaryli- 4. Next, 1,2,3-triazole scaffold was constructed via click reaction and for
midazole-1,2,3-triazole hybrids were designed and synthesized as potent this purpose, various benzyl halides 5 reacted with sodium azide in the
α-glucosidase inhibitors (Fig. 1). Apart from in vitro assessment of target mixture of H2O and t-BuOH (1:1) in the presence of Et3N at room tem-
compounds, their possible mode of interaction with α-glucosidase was perature to produce benzyl azides 6 in situ. Then, the mixture of compound
investigated through kinetic and docking studies. 4, sodium ascorbate, and CuSO4·5H2O was added to the freshly prepared
azides 6 and the reaction was continued at room temperature for 24–48 h
2. Results and discussion to give the target compounds 7a-p.

2.1. Chemistry 2.2. In vitro α-glucosidase inhibitory activity

The synthetic route for the preparation of diarylimidazole-1,2,3-tria- In vitro α-glucosidase inhibitory activity of synthesized diarylimi-
zoles 7a-p has been depicted in Scheme 1. Initially, a mixture of benzil dazole-1,2,3-triazole hybrids was assayed according to the literature27

Scheme 1. Reagents and conditions for the synthesis of compounds 7a-p: (a) AcOH, reflux, 1 h and (b) CuSO4·5H2O, sodium ascorbate, rt, 24–48 h.

2
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

Table 1 benzyl ring deteriorated inhibitory activity in such a manner that


In vitro α-glucosidase inhibitory activity of compounds 7. compound 7c showed much better activity, approximately two times,
than compounds 7d and 7i.
Further investigation comes back to compounds 7j-l possessing two
4-methoxyaryl groups connected to imidazole moiety. Comparing the
corresponding inhibitory activity with those analogues (compounds 7a,
7c, and 7f, respectively) revealed that better activity was only obtained
by compound 7j (IC50 = 129.5 µM) comparing with compound 7a
(IC50 = 199.0 µM). In the case of halogenated compounds 7k
Entry Compound 7 R1 R2 R3 IC50 (µM)a
(IC50 = 179.0 µM) and 7l (IC50 = 143.6 µM), introduction of methoxy
1 7a H H 2-CH3 199.0 ± 0.4 groups did not induce higher activity.
2 7b H H 3,5-diCH3 90.4 ± 0.5 The last part of discussion is related to the series of compounds 7m-
3 7c H H 4-F 100.0 ± 0.5 p which possessed methoxy group connected to the phenoxy moiety. In
4 7d H H 4-Cl 246.7 ± 0.7
this respect, compound 7o containing 4-chlorobenzyl group connected
5 7e H H 2,3-diCl 97.7 ± 0.7
6 7f H H 3,4-diCl 106.0 ± 0.8 1,2,3-triazole ring showed the best activity (IC50 = 150.2 µM).
7 7g H H 2-Br 118.7 ± 0.6 Replacement of Cl by F, derivatives 7m (IC50 = 162.9 µM) and 7n
8 7h H H 3-Br 135.4 ± 0.8 (IC50 = 188.5 µM) at the 2- or 4- position led to lower inhibitory ac-
9 7i H H 4-Br 204.3 ± 0.6 tivity. It seems that the presence of 4-chlorobenzyl moiety was effective
10 7j OCH3 H 2-CH3 129.5 ± 1.2
in this series of compounds. Finally, the presence of an electron-with-
11 7k OCH3 H 4-F 179.0 ± 1.0
12 7l OCH3 H 3,4-diCl 143.6 ± 1.0 drawing group (NO2) not only did not improve inhibitory activity but
13 7m H OCH3 2-F 162.9 ± 1.0 also decreased activity against α-glucosidase (IC50 = 201.6 µM).
14 7n H OCH3 4-F 188.5 ± 1.0 It can be concluded that the substituents on the benzyl moiety
15 7o H OCH3 4-Cl 150.2 ± 0.9
connected to the 1,2,3-triazole ring played a remarkable role in in vitro
16 7p H OCH3 4-NO2 201.6 ± 1.1
17 Acarboseb 750.0 ± 1.5
α-glucosidase inhibitory activity and the efficacy of methoxy groups
either on the aryl groups connected to imidazole moiety or phenoxy
a
Data are expressed as mean ± SE (three independent experiments). link is generally depended on those substituents. In this regard, α-glu-
b
IC50 values for α-glucosidase inhibitory activity of 1-deoxynojirimycin and cosidase inhibitory activity of compounds 7c, 7k, and 7n possessing 4-
miglitol have been reported as IC50 = 192.0 μM28 and 59.76 μg/mL, respec- fluorobenzyl group revealed that the introduction of methoxy groups
tively in the literature.29 into the aryl or linker moieties led to lower activity (7c > 7k > 7n).
Similarly, in the case of compounds 7f and 7l possessing 3,4-di-
comparing with acarbose as the reference drug (Table 1). To obtain the chlorobenzyl group, the absence of methoxy group led to better activity
best insight into the role of the presence and position of substituents on (7f > 7l). In different manner, compound 7a showed lower activity
the aryl ring connected to imidazole and 1,2,3-triazole moieties as well than compound 7j. Both compounds contained 2-methylbenzyl moiety
as phenoxy linker in the inhibitory activity, a wide variety of com- but the absence and presence of methoxy depicted different activity
pounds 7 were synthesized. In this regard, synthesized compounds were comparing with compounds 7f/7l and compounds 7c/7k/7n. Also, in
considered in three categories of 7a-i (Table 1, R1 = H and R2 = H), 7j- the case of compounds 7d and 7o having 4-chlorobenzyl group, the
l (Table 1, R1 = OCH3 and R2 = H), and 7m-p (Table 1, R1 = H and presence of methoxy group was found to be effective for inducing de-
R2 = OCH3). sired inhibitory activity.
It is worth mentioning that all compounds 7 demonstrated much
better inhibitory activity at least 3 times than acarbose. The best ac- 2.3. Kinetic study
tivity was obtained by compound 7b (IC50 = 90.4 µM) which was ap-
proximately 8 times more potent than acarbose (IC50 = 750.0 µM). It The kinetic analysis of α-glucosidase inhibition by compounds 7b,
contained two phenyl groups connected to imidazole ring, phenoxy 7c, and 7e was performed according to our previous report.27 The
linker with no substituent, and two methyl groups at the 3- and 5- mechanism of inhibition and Ki values for those compounds were de-
positions of benzyl moiety connected to 1,2,3-triazole ring. Changing termined by Lineweaver–Burk plots and secondary re-plot of these
the position and number of methyl group led to the reduction of activity plots, respectively (Figs. 2–4).
of the analogue of 7b, compound 7a (IC50 = 199.0 µM) possessing 2- As can be seen in Figs. 2a–4a, with increasing the concentration of
methylbenzyl group connected to 1,2,3-triazole moiety. In the first the inhibitor, the values of Vmax remained constant and the values of Km
category of compounds 7a-i, counterparts of compound 7b, compounds increased. It indicated that compounds 7b, 7c, and 7e acted as com-
7c-i possessing halobenzyl connected to 1,2,3-triazole ring showed α- petitive inhibitors of α-glucosidase binding to the enzyme active site in
glucosidase inhibitory activity generally depending on the number and competition with substrate. Furthermore, the plot of Km versus different
position of halogens. In this series of compounds (7c-i), compound 7e concentrations of inhibitors gave an estimate of the inhibition constant,
containing two Cl groups at the 2- and 3- positions of aryl ring showed Ki values of 87.0, 109.0, and 100.4 µM for compounds 7b, 7c, and 7e,
very good activity against α-glucosidase (IC50 = 97.7 µM). respectively.
Interestingly, changing the position of chlorine groups to 3- and 4- led
to a slight decrease in the inhibitory activity of compound 7f 2.4. Docking study
(IC50 = 106.0 µM). It should be noted that compound 7d possessing a
chlorine group at the 4- position of aryl ring showed lower activity than Molecular docking simulations were applied to distinguish interac-
compounds 7e and 7f. As can be seen in Table 1, the presence of tions between the synthesized compounds and α-glucosidase. Due to
fluorobenzyl moiety led to a good in vitro α-glucosidase inhibitory ac- the lack of crystallographic structure of α-glucosidase from S. cerevisiae,
tivity as compound 7c was found to be a relatively potent inhibitor homology model of this enzyme was built according to our pervious
(IC50 = 100.0 µM). Finally, compounds 7g-i containing bromobenzyl study.27 The superposed structure of acarbose and the most potent
moiety depicted versatile inhibitory activity affected by the position of compound 7b in the active site of α-glucosidase is shown in Fig. 5 (left).
bromine. The order of activity for compounds 7g-i, was found to be Acarbose formed hydrogen bonding interactions with residues Asn241,
7g > 7h > 7i (IC50s = 118.7, 135.4, and 204.3 µM, respectively). It is Pro309, Thr301, Glu304, Thr307, Ser308, Arg312, Gln322, and a hy-
clear that the presence of halogen except fluorine at the 4-position of drophobic interaction with His279 (Fig. 5, right).27

3
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

a) b)
80
8
Concentration of Inhibitor
0 µM

1/V(∆ PNP 405 nm/min) -1


40 µM 60
6
65 µM
90 µM

Km
40 4

20 2

0
0
-100 -80 -60 -40 -20 0 20 40 60 80 100
-2 -1 0 1 2
-1
1/[S](PNPG, mM ) [I], µM
-2

Fig. 2. (a) Lineweaver–Burk plots for the inhibition of α-glucosidase by compound 7b. (b) The secondary plot between Km and various concentrations of compound
7b (b).

Detailed binding modes of compounds 7b, 7c, and 7e were also Compound 7e unlike compounds 7b and 7c, established no hy-
shown in Figs. 6–8. As can be seen in those figures, the following in- drogen bonding interaction with the active site of enzyme (Figs. 6–8).
teractions were mutual for three studied compounds: (1) hydrophobic 2,3-Dichlorobenzyl moiety of compound 7e formed interactions with
interactions of Val316 and Ala326 with phenyl groups attached to the Asp408, Phe157, and Phe158 (Fig. 8).
imidazole moiety, (2) a π-anion interaction between Glu304 and phe- The binding energy values for compounds 7b (−11.17 kcal/mol),
noxy moiety, (3) hydrophobic interactions of Pro309 with imidazole 7c (−11.22 kcal/mol), and 7e (−11.46 kcal/mol) revealed that they
and phenoxy moieties, and (4) hydrophobic interactions of Arg312 with bound more readily to the active site than the standard drug acarbose
1,2,3-triazole ring and benzyl group. (−4.04 kcal/mol)27 which was in good agreement with those results
The most potent compound 7b made further interactions as de- obtained in in vitro assay.
scribed above. Two hydrogen bonding interactions were formed be-
tween NH of imidazole moiety and Thr307 as well as nitrogen of 1,2,3-
triazole ring and Arg312. Also, two hydrophobic interactions were 3. Conclusion
distinguished between 3,5-dimethylbenzyl moiety and Phe300 and
Arg439 (Fig. 6). As can be seen in the Figs. 6 and 7, comparing the In conclusion, we synthesized a new series of diarylimidazole-1,2,3-
interaction mode of compound 7b with that of compound 7c revealed triazole derivatives 7a-p as potent α-glucosidase inhibitors comparing
that compound 7c formed only a hydrogen bond interaction with the with acarbose as the reference drug. Among them, compounds 7b, 7c,
active site (between NH of imidazole moiety and Thr307) while com- and 7e with inhibitory activity approximately 8 times more potent than
pound 7b established two hydrogen bonding interactions. On the other acarbose were found to be the most active compounds. Also, their ki-
hand, 1,2,3-triazole ring of the compound 7c formed additional inter- netic studies indicated competitive inhibitory activity toward α-gluco-
actions with His239 (a hydrophobic interaction, a π-π interaction, and a sidase. It is worth mentioning that docking study of compounds 7b, 7c,
π-cation interaction). Also, 4-fluorobenzyl moiety of compound 7c in- and 7e revealed that they well fitted in the active site of α-glucosidase
teracted with Asp408 via a π-anion interaction (Fig. 7). with the binding energies less than acarbose.

a) b)
7

Concentration of Inhibitor 30
6
-1

0 µM
1/V(∆ PNP 405 nm, min)

40 µM
70 µM 5
20
100 µM
4
Km

10 3

0 1
-1.5 -1.0 -0.5 0.0 0.5 1.0 1.5
0
1/[S](PNPG, mM-1 )
-150 -100 -50 0 50 100 150
-10
-1 [I], µM

Fig. 3. (a) Lineweaver–Burk plots for the inhibition of α-glucosidase by compound 7c. (b) The secondary plot between Km and various concentrations of compound
7c.

4
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

a) b)

50 7

Concentration of Inhibitor

-1
6

1/V(∆ PNP 405 nm, min)


40
0 µM 5
37 µM
67 µM 30
97 µM 4

Km
20 3

2
10

1
0
-1.5 -1.0 -0.5 0.0 0.5 1.0 1.5 0
-150 -100 -50 0 50 100 150
-10 1/[S](PNPG, mM-1 )
[I], µM

Fig. 4. (a) Lineweaver–Burk plots for the inhibition of α-glucosidase by compound 7e. (b) The secondary plot between Km and various concentrations of compound
7e.

Fig. 5. Acarbose (cyan) and the most potent compounds 7b (pink) superimposed in the active site pocket of modeled α-glucosidase. (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of this article.)

4. Experimental 4.2. General procedure for the synthesis of diarylimidazole-1,2,3-triazole


hybrids 7a-p
Melting points of synthesized compounds 7a-p were determined on
a Kofler hot stage apparatus. 1H and 13C NMR spectra were determined A mixture of benzyl halid derivative 5 (1.1 mmol) and sodium azide
on a Bruker and Varian FT-500, using TMS as an internal standard. IR (0.9 mmol) in the presence of Et3N (1.3 mmol) in water/t-BuOH (8 mL,
spectra were recorded using KBr disks on a Nicolet Magna FTIR 550 1:1) was stirred at room temperature for 1 h. Subsequently, the mixture
spectrophotometer. Elemental analysis was carried out with an of compound 4 (1 mmol), CuSO4·5H2O (7 mol%), and catalytic amounts
Elemental Analyzer system GmbH VarioEL CHN mode. Compound 3 of sodium ascorbate was added to the freshly prepared benzyl azide
was prepared according to the procedure described in the literature.30 derivative 6 and stirred at room temperature for 24–48 h. Upon com-
pletion of the reaction (monitored by TLC), the reaction mixture was
4.1. General procedure for the preparation of 4,5-diaryl-2-(4-(prop-2- poured into crushed ice. Then, the precipitate was filtered off, washed
ynyloxy)phenyl)-1H-imidazole derivatives 4 with cold water, and purified by recrystallization from ethanol to give
the corresponding derivatives 7a-p.
A mixture of benzil derivative 1 (1 mmol), ammonium acetate 2
(10 mmol), and 4-(prop-2-ynyloxy) benzaldehyde derivative 3 4.2.1. 4-((4-(4,5-Diphenyl-1H-imidazol-2-yl)phenoxy)methyl)-1-(2-
(1 mmol) in acetic acid was heated at reflux for 1 h. Upon completion of methylbenzyl)-1H-1,2,3-triazole 7a
the reaction (checked by TLC), the reaction mixture was poured into White powder, Yield: 75%, mp = 203–205 °C. IR (KBr): 3418, 3062,
water, the precipitate was filtered off, and washed with water to afford 1610 cm−1. 1H NMR (500 MHz, CDCl3): δ = 2.25 (s, 3H, CH3), 5.11 (s,
compound 4. 2H, CH2), 5.50 (s, 2H, CH2), 6.93 (d, J = 8.5 Hz, 2H, H2, H6), 7.13 (d,

5
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

Fig. 6. Docked conformer of compound 7b in the active site of α-glucosidase.

J = 7.5 Hz, 1H, H3′), 7.19–7.29 (m, 9H, H4′, H5′, H6′, Ph), 7.39 (s, 1H, 126.3, 126.6, 127.0, 127.5, 128.0, 128.2, 128.5, 135.6, 136.7, 137.8,
triazole), 7.42–7.62 (m, 4H, Ph), 7.78 (d, J = 8.5 Hz, 2H, H3, H5), 9.91 142.8, 145.4, 158.1. Anal Calcd for C33H29N5O: C, 77.47; H, 5.71; N,
(s, 1H, NH). 13C NMR (125 MHz, CDCl3): δ = 20.7, 52.4, 62.0, 115.1, 13.69. Found C, 77.20; H, 5.85; N, 13.51.
122.5, 123.5, 126.7, 126.9, 127.9, 128.2, 128.8, 129.2, 129.5, 131.1,
132.3, 134.9, 136.9, 138.0, 138.1, 144.1, 146.1, 158.7. Anal Calcd for
C32H27N5O: C, 77.24; H, 5.47; N, 14.07. Found C, 77.50; H, 5.21; N, 4.2.3. 4-((4-(4,5-Diphenyl-1H-imidazol-2-yl)phenoxy)methyl)-1-(4-
14.25. fluorobenzyl)-1H-1,2,3-triazole 7c
White powder, Yield: 65%, mp = 180–182 °C. IR (KBr): 3415, 3065,
1615 cm−1. 1H NMR (500 MHz, CDCl3): δ = 5.21 (s, 2H, CH2), 5.53 (s,
4.2.2. 1-(3,5-Dimethylbenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)
2H, CH2), 7.02–7.06 (m, 4H, H2, H6, H3′, H5′), 7.30–7.68 (m, 14H,
phenoxy)methyl)-1H-1,2,3-triazole 7b
2 × Ph, H3, H5, H2′, H6′), 8.31 (s, 1H, triazole), 11.96 (s, 1H, NH). 13C
White powder, Yield: 75%, mp = 203–205 °C. IR (KBr): 3415, 3065,
NMR (125 MHz, CDCl3): δ = 52.8, 61.5, 114.4, 115.4 (JC-F = 22.5 Hz),
1615 cm−1. 1H NMR (500 MHz, CDCl3): δ = 2.24 (s, 6H, 2 × CH3),
122.5, 123.8, 124.0, 125.4, 126.7, 127.5, 127.7, 127.9, 129.4, 135.9,
5.20 (s, 2H, CH2), 5.52 (s, 2H, CH2), 6.93–6.96 (m, 3H, H2′, H4′, H6′),
139.2, 142.5, 144.3, 158.0, 161.1 (JC-F = 251.2 Hz). Anal Calcd for
7.13 (d, J = 8.5 Hz, 2H, H2, H6), 7.21–7.55 (m, 10H, 2 × Ph), 8.01 (d,
C31H24FN5O: C, 74.24; H, 4.82; N, 13.96. Found C, 74.55; H, 4.62; N,
J = 8.5 Hz, 2H, H3, H5), 8.26 (s, 1H, triazole), 12.50 (s, 1H, NH). 13C
14.20.
NMR (125 MHz, CDCl3): 20.7, 52.8, 61.1, 114.8, 123.4, 124.5, 125.6,

Fig. 7. Docked conformer of compound 7c in the active site of α-glucosidase.

6
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

Fig. 8. Docked conformer of compound 7e in the active site of α-glucosidase.

4.2.4. 1-(4-Chlorobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)phenoxy) 8.01 (d, J = 8.5 Hz, 2H, H3, H5), 8.27 (s, 1H, triazole), 12.49 (s, 1H,
methyl)-1H-1,2,3-triazole 7d NH). 13C NMR (125 MHz, DMSO‑d6): δ = 52.9, 61.1, 114.9, 122.8,
Cream powder, Yield: 83%, mp = 206–208 °C. IR (KBr): 3420, 123.4, 125.0, 126.3, 126.6, 127.0, 127.5, 128.0, 128.2, 128.3, 128.5,
2855, 1675, 1580 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 5.22 (s, 130.3, 130.4, 132.8, 136.7, 142.7, 145.8, 158.1. Anal Calcd for
2H, CH2), 5.64 (s, 2H, CH2), 7.15 (d, J = 8.8 Hz, 2H, H2, H6), C31H24BrN5O: C, 66.20; H, 4.30; N, 12.45. Found C, 66.54; H, 4.44; N,
7.22–7.50 (m, 10H, 2 × Ph), 7.52 (d, J = 8.0 Hz, 2H, H2′, H6′), 7.56 (d, 12.25.
J = 8.0 Hz, 2H, H3′, H5′), 8.02 (d, J = 8.8 Hz, 2H, H3, H5), 8.34 (s, 1H,
triazole), 12.55 (s, 1H, NH). 13C NMR (125 MHz, DMSO‑d6) δ = 52.9, 4.2.8. 1-(3-Bromobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)phenoxy)
61.6, 115.4, 123.9, 125.2, 126.9, 127.1, 127.5, 128.0, 128.1, 128.6, methyl)-1H-1,2,3-triazole 7h
128.8, 129.1, 129.2, 130.4, 131.7, 133.3, 135.4, 135.7, 137.2, 143.4, Cream powder, Yield: 91%, mp = 223–225 °C. IR (KBr): 3425,
146.0, 158.6. Anal Calcd for C31H24ClN5O: C, 71.88; H, 4.67; N, 13.52. 2955, 2825, 1670, 1585 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 5.23
Found C, 71.70; H, 4.52; N, 13.38. (s, 2H, CH2), 5.65 (s, 2H, CH2), 7.17 (d, J = 8.7 Hz, 2H, H2, H6),
7.22–7.58 (m, 14H, 2 × Ph, H2′, H4′, H5′, H6′), 8.06 (d, J = 8.5 Hz,
4.2.5. 1-(2,3-Dichlorobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl) 2H, H3, H5), 8.37 (s, 1H, triazole), 12.56 (s, 1H, NH). 13C NMR
phenoxy)methyl)-1H-1,2,3-triazole 7e (125 MHz, DMSO‑d6): δ = 61.2, 52.0, 114.9, 121.9, 123.5, 124.9,
White powder, Yield: 80%, mp = 191–193 °C. IR (KBr): 3416, 3060, 125.1, 126.7, 127.1, 127.4, 128.2, 128.3, 130.7, 130.9, 131.1, 135.3,
1609 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 5.22 (s, 2H, CH2), 5.79 136.8, 138.6, 143.0, 145.6, 158.2. Anal Calcd for C31H24BrN5O: C,
(s, 2H, CH2), 7.14 (d, J = 8.5 Hz, 2H, H2, H6), 7.18 (d, J = 7.5 Hz, 1H, 66.20; H, 4.30; N, 12.45. Found C, 66.34; H, 4.46; N, 12.68.
H6′), 7.21 (t, J = 7.5 Hz, 1H, H5′), 7.28–7.65 (m, 10H, 2 × Ph), 7.66
(d, J = 7.5 Hz, 1H, H4′), 8.01 (d, J = 8.5 Hz, 2H, H3, H5), 8.30 (s, 1H, 4.2.9. 1-(4-Bromobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)phenoxy)
triazole), 12.49 (s, 1H, imidazole). 13C NMR (125 MHz, DMSO‑d6): methyl)-1H-1,2,3-triazole 7i
δ = 51.1, 61.1, 114.9, 125.1, 126.3, 126.6, 127.0, 127.5, 128.0, 128.2, Cream powder, Yield: 87%, mp = 218–219 °C. IR (KBr): 3425,
128.5, 128.9, 130.5, 131.1, 132.2, 135.2, 135.7, 136.7, 142.8, 145.6, 2855, 1675, 1565 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 5.23 (s,
158.1. Anal Calcd for C31H23Cl2N5O: C, 67.40; H, 4.20; N, 12.68. Found 2H, CH2), 5.63 (s, 2H, CH2), 7.16 (d, J = 8.4 Hz, 2H, H2, H6), 7.23 (d,
C, 67.15; H, 4.43; N, 12.41. J = 7.5 Hz, 2H, H2′, H6′), 7.29–7.61 (m, 12H, 2 × Ph, H3′, H5′), 8.04
(d, J = 8.3 Hz, 2H, H3, H5), 8.34 (s, 1H, triazole), 12.58 (s, 1H, NH).
13
4.2.6. 1-(3,4-Dichlorobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl) C NMR (125 MHz, DMSO): δ = 52.1, 61.2, 114.9, 121.4, 124.8,
phenoxy)methyl)-1H-1,2,3-triazole 7f 126.4, 126.7, 127.1, 127.6, 128.1, 128.3, 128.6, 130.2, 131.7, 135.4,
White powder, Yield: 80%, mp = 198–200 °C. IR (KBr): 3426, 3055, 136.8, 143.0, 145.6, 158.2. Anal Calcd for C31H24BrN5O: C, 66.20; H,
1607 cm−1. 1H NMR (CDCl3, 500 MHz): δ = 5.14 (s, 2H, CH2), 5.42 (s, 4.30; N, 12.45. Found C, 66.34; H, 4.52; N, 12.62.
2H, CH2), 6.93 (d, J = 8.5 Hz, 2H, H2, H6), 7.05 (d, J = 8.0 Hz, 1H,
H5′), 7.33 (s, 1H, H2′), 7.41 (d, J = 8.0 Hz, 1H, H6′), 7.28–7.52 (m, 4.2.10. 4-((4-(4,5-bis(4-Methoxyphenyl)-1H-imidazol-2-yl)phenoxy)
10H, 2 × Ph), 7.55 (s, 1H, triazole), 7.78 (d, J = 8.5 Hz, 2H, H3, H5). methyl)-1-(2-methylbenzyl)-1H-1,2,3-triazole 7j
13
C NMR (125 MHz, CDCl3): δ = 52.9, 62.0, 115.2, 122.8, 122.9, 123.6, White powder, Yield: 70%, mp = 102–105 °C. IR (KBr): 3430, 2934,
126.9, 127.2, 127.8, 128.5, 128.9, 129.9, 131.0, 131.2, 131.5, 134.5, 1612 cm−1. 1H NMR (500 MHz, CDCl3): δ = 2.26 (s, 3H, CH3), 3.80 (s,
135.0, 141.0, 144.7, 146.0, 158.6. Anal Calcd for C31H23Cl2N5O: C, 6H, 2 × OMe), 5.14 (s, 2H, CH2), 5.51 (s, 2H, CH2), 6.83 (d, J = 8.0 Hz,
67.40; H, 4.20; N, 12.68. Found C, 67.58; H, 4.40; N, 12.80. 4H, 2 × 4-methoxyaryl), 6.94 (d, J = 8.5 Hz, 2H, H2, H6), 7.14 (d,
J = 7.0 Hz, 1H, H6′), 7.19–7.28 (m, 2H, H4′, H5′), 7.41–7.44 (m, 5H,
4.2.7. 1-(2-Bromobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)phenoxy) 2 × 4-methoxyaryl, H3′), 7.78 (d, J = 8.5 Hz, 2H, H3, H5), 7.98 (s, 1H,
methyl)-1H-1,2,3-triazole 7g triazole). 13C NMR (125 MHz, CDCl3): δ = 20.5, 52.4, 55.3, 62.1, 114.0,
White powder, Yield: 80%, mp = 205–207 °C. IR (KBr): 3416, 3060, 115.1, 122.5, 123.7, 126.7, 126.8, 129.1, 129.2, 129.5, 129.8, 130.0,
1609 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 5.22 (s, 2H, CH2), 5.71 131.1, 132.5, 135.8, 144.0, 145.5, 146.5, 158.6, 158.9. Anal Calcd for
(s, 2H, CH2), 7.14 (d, J = 8.5 Hz, 2H, H2, H6), 7.17–7.23 (m, 3H, H4′, C34H31N5O2: C, 73.23; H, 5.60; N, 12.56. Found C, 73.54; H, 5.41; N,
H5′, H6′), 7.27–7.54 (m, 10H, 2 × Ph), 7.69 (d, J = 7.5 Hz, 1H, H3′), 12.34.

7
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

4.2.11. 4-((4-(4,5-bis(4-Methoxyphenyl)-1H-imidazol-2-yl)phenoxy) 128.6, 128.8, 128.9, 129.1, 129.2, 130.4, 131.7, 133.3, 135.4, 135.7,
methyl)-1-(4-fluorobenzyl)-1H-1,2,3-triazole7k 137.2, 143.4, 146.0, 148.0, 149.6. Anal Calcd for C32H26ClN5O2: C,
White powder, Yield: 75%, mp = 170–172 °C. IR (KBr): 3425, 3037, 70.13; H, 4.78; N, 12.78. Found C, 70.38; H, 4.59; N, 12.62.
1608 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.75 (s, 3H, OMe), 3.80
(s, 3H, OMe), 5.20 (s, 2H, CH2), 5.61 (s, 2H, CH2), 6.86 (d, J = 8.5 Hz, 4.2.16. 4-((4-(4,5-Diphenyl-1H-imidazol-2-yl)-2-methoxyphenoxy)
2H, 4-methoxyaryl), 6.99 (d, J = 8.5 Hz, 2H, 4-methoxyaryl), 7.11 (d, methyl)-1-(4-nitrobenzyl)-1H-1,2,3-triazole 7p
J = 8.5 Hz, 2H, H2, H6), 7.21 (t, J = 8.5 Hz, 2H, H3′, H5′), 7.39–7.45 Cream powder, Yield: 95%, mp = 233–235 °C. IR (KBr): 3450,
(m, 6H, 2 × 4-methoxyaryl, H2′, H6′), 7.97 (d, J = 8.5 Hz, 2H, H3, H5), 2955, 2850, 1690, 1585 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.84
8.29 (s, 1H, triazole), 12.30 (s, 1H, NH). 13C NMR (125 MHz, (s, 3H, OCH3), 5.22 (s, 2H, CH2), 5.83 (s, 2H, CH2), 7.23–7.57 (m, 13H,
DMSO‑d6): δ = 57.0, 59.9, 60.1, 18.5, 119.0, 120.5 (JC-F = 21.2 Hz), 2 × Ph, H6, H2′, H6′), 7.68 (d, J = 8.5 Hz, 1H, H5), 7.71 (s, 1H, H3),
126.0, 128.0, 128.6, 129.5, 131.0, 131.4, 133.0, 134.5, 135.2, 141.2, 8.26 (d, J = 8.3 Hz, 2H, H3′, H5′), 8.40 (s, 1H, triazole), 12.60 (s, 1H,
148.0, 149.7, 156.0, 157.3, 161.9 (JC-F = 245.0 Hz). Anal Calcd for NH). 13C NMR (125 MHz, DMSO): δ = 52.1, 55.7, 61.9, 109.2, 114.0,
C33H28FN5O2: C, 70.58; H, 5.03; N, 12.47. Found C, 70.74; H, 4.86; N, 118.0, 123.9, 124.0, 124.1, 125.5, 126.6, 127.2, 127.9, 128.3, 128.6,
12.21. 128.8, 129.2, 131.3, 135.4, 136.9, 143.3, 143.5, 145.8, 147.4, 147.8,
149.3. Anal Calcd for C32H26N6O4: C, 68.81; H, 4.69; N, 15.05. Found
4.2.12. 4-((4-(4,5-bis(4-Methoxyphenyl)-1H-imidazol-2-yl)phenoxy) C, 68.66; H, 4.58; N, 14.94.
methyl)-1-(3,4-dichlorobenzyl)-1H-1,2,3-triazole 7l
White powder, Yield: 70%, mp = 102–104 °C. IR (KBr): 3430, 2999, 4.3. In vitro α-glucosidase inhibition assay
1613 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.75 (s, 3H, OMe), 3.80
(s, 3H, OMe), 5.21 (s, 2H, CH2), 5.65 (s, 2H, CH2), 6.87–6.99 (m, 5H, A mixture containing phosphate buffer saline (135 µL, pH 6.8,
2 × 4-methoxyaryl, H6′), 7.12 (d, J = 7.0 Hz, 2H, H2, H6), 7.30 (d, 50 mM), test compound (20 µL) dissolved in DMSO (10% final con-
J = 7.0 Hz, 1H, H5′), 7.41–7.45 (m, 4H, 2 × 4-methoxyaryl), 7.64 (s, centration), and solution of α-glucosidase (Saccharomyces cerevisiae,
1H, H2′), 7.97 (d, J = 7.0 Hz, 2H, H3, H5), 8.34 (s, 1H, triazole), 12.33 EC3.2.1.20, 20 U/mg, 20 µL) was pre incubated at 37 °C for 10 min in a
(s, 1H, NH). 13C NMR (125 MHz, DMSO‑d6): δ = 51.4, 55.0, 61.1, 96-well plate. In order to initiate the enzyme-catalyzed reaction, 25 µL
113.5, 114.0, 123.6, 124.7, 126.5, 128.1, 128.3, 129.5, 130.0, 130.9, of substrate (p-nitrophenyl-α-D-glucopyranoside, 4 mM) was added to
132.3, 133.5, 136.8, 137.8, 140.5, 140.8, 143.0, 149.9, 157.9. Anal each well and the absorbance change was recorded at 400 nm within
Calcd for C33H27Cl2N5O2: C, 64.71; H, 4.44; N, 11.43. Found C, 64.51; 20 min at 37 °C. The percentage of inhibition for tested compounds 7a-
H, 4.67; N, 11.25. p, control, and acarbose as positive control was calculated using the
following formula:
4.2.13. 4-((4-(4,5-Diphenyl-1H-imidazol-2-yl)-2-methoxyphenoxy) % Inhibition = [(Abs control − Abs sample)/Abs control] × 100.
methyl)-1-(2-fluorobenzyl)-1H-1,2,3-triazole 7m IC50 values were calculated from the nonlinear regression curve using
Cream powder, Yield: 88%, mp = 214–216 °C. IR (KBr): 3350, the Logit method.
2950, 2825, 1680, 1575 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.82
(s, 3H, OCH3), 5.17 (s, 2H, OCH2), 5.70 (s, 2H, OCH2), 7.20–7.67 (m, 4.4. Kinetic study
15H, 2 × Ph, H6, H3′, H4′, H5′, H6′), 7.65 (d, J = 8.0 Hz, 1H, H5), 7.67
(s, 1H, H3), 8.29 (s, 1H, triazole), 12.56 (s, 1H, NH). 13C NMR The α-glucosidase solution (1 U/mL, 20 μL) was incubated with
(125 MHz, DMSO): δ = 61.6, 55.5, 46.9, 109.0, 113.7, 115.6 (d, JC- different concentrations of 0, 40, 65, and 90 μM of the most potent
F = 20.0 Hz), 117.7, 122.8 (d, JC-F = 13.7 Hz), 123.8, 124.8 (d, JC- compound 7b (20 μL) for 15 min at 30 °C. Then, the enzymatic reaction
F = 3.7 Hz), 126.4, 127.0, 127.7, 128.1, 128.6 (d, JC-F = 30.0 Hz), was initiated by adding various concentrations of p-nitrophenyl gluco-
130.8, 131.2, 135.2, 136.8, 142.8, 145.6, 147.6, 149.1, 161.1 (d, JC- pyranoside as substrate (1–4 mM). Finally, change in the absorbance
F = 245.0 Hz). Anal Calcd for C32H26FN5O2: C, 72.30; H, 4.93; N, 13.17. was recorded at 405 nm for 20 min using spectrophotometer (Gen5,
Found C, 72.33; H, 4.88; N, 13.14. Power wave xs2, BioTek, America).
A Lineweaver–Burk plot was generated to identify the type of in-
4.2.14. 4-((4-(4,5-Diphenyl-1H-imidazol-2-yl)-2-methoxyphenoxy) hibition and the Michaelis–Menten constant (Km) value was determined
methyl)-1-(4-fluorobenzyl)-1H-1,2,3-triazole 7n from plot between reciprocal of the substrate concentration (1/[S]) and
Cream powder, Yield: 86%, mp = 247–249 °C. IR (KBr): 3420, reciprocal of enzyme rate (1/V) over various inhibitor concentrations.
2965, 2850, 1690, 1585 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.83 Experimental inhibitor constant (Ki) value was constructed by sec-
(s, 3H, OCH3), 5.18 (s, 2H, OCH2), 5.62 (s, 2H, OCH2), 7.22–7.69 (m, ondary plots of the inhibitor concentration [I] versus Km.
13H, 2 × Ph, H6, H3′, H5′), 7.53 (dd, J = 8.0, 5.0 Hz, 2H, H2′, H6′), Also, the same procedure was conducted for compounds 7c and 7e
7.66 (d, J = 8.0 Hz, 1H, H5), 7.69 (s, 1H, H3), 8.31 (s, 1H, triazole), with different concentrations of 0, 40, 70, and 100 µM for compound 7c
12.58 (s, 1H, NH). 13C NMR (125 MHz, DMSO): δ = 52.0, 61.7, 55.5, and 0, 37, 67, and 97 µM for compound 7e.
109.0, 113.8, 115.6 (d, JC-F = 21.2 Hz), 117.8, 123.8, 124.8, 126.4,
127.1, 127.7, 128.2, 128.4, 128.7, 130.3, 130.4, 131.3, 132.3, 135.3, 4.5. Docking study
136.8, 142.9, 145.6, 147.6, 149.1, 161.9 (d, JC-F = 242.5 Hz). Anal
Calcd for C32H26FN5O2: C, 72.30; H, 4.93; N, 13.17. Found C, 72.12; H, Docking studies of the most potent compounds 7b, 7c, and 7e were
4.72; N, 13.42 performed exactly based on our previous report.27

4.2.15. 1-(4-Chlorobenzyl)-4-((4-(4,5-diphenyl-1H-imidazol-2-yl)-2- Declaration of Competing Interest


methoxyphenoxy)methyl)-1H-1,2,3-triazole 7o
Cream powder, Yield: 84%, mp = 228–230 °C. IR (KBr): 3420, There is no conflict of interest.
2855, 1670, 1589 cm−1. 1H NMR (500 MHz, DMSO‑d6): δ = 3.83 (s,
3H, OCH3), 5.19 (s, 2H, OCH2), 5.64 (s, 2H, OCH2), 7.21–7.47 (m, 11H, Acknowledgements
2 × Ph, H6), 7.51 (d, J = 8.0 Hz, 2H, H2′, H6′), 7.56 (d, J = 8.0 Hz, 2H,
H3′, H5′), 7.65 (d, J = 8.0 Hz, 1H, H5), 7.69 (s, 1H, H3), 8.32 (s, 1H, This work was supported by Research Council of Tehran University
triazole), 12.57 (s, 1H, NH). 13C NMR (125 MHz, DMSO): δ = 52.5, of Medical Sciences and International Campus (TUMS-IC) with project
56.0, 62.2, 109.4, 114.3, 118.20, 124.3, 125.4, 126.9, 127.5, 128.1, No. 94-01-169-28719.

8
M. Saeedi, et al. Bioorganic & Medicinal Chemistry 27 (2019) 115148

Appendix A. Supplementary material 15. Almirante L, Polo L, Mugnaini A, et al. Murmann W. Derivatives of imidazole. I.
Synthesis and reactions of imidazo[1,2-a]pyridines with analgesic, antiin-
flammatory, antipyretic, and anticonvulsant activity. J Med Chem. 1965;8:305–312.
Supplementary data to this article can be found online at https:// 16. Sameem B, Saeedi M, Mahdavi M, Shafiee A. A review on tacrine-based scaffolds as
doi.org/10.1016/j.bmc.2019.115148. multi-target drugs (MTDLs) for Alzheimer's disease. Eur J Med Chem.
2017;128:332–345.
17. Sharma D, Narasimhan B, Kumar P, et al. Synthesis, antimicrobial and antiviral
References evaluation of substituted imidazole derivatives. Eur J Med Chem.
2009;44:2347–2353.
1. Scheen AJ. Is there a role for α-glucosidase inhibitors in the prevention of type 2 18. Karakurt A, Dalkara S, Özalp M, Özbey S, Kendi E, Stables JP. Synthesis of some 1-(2-
diabetes mellitus? Drugs. 2003;63:933–951. naphthyl)-2-(imidazole-1-yl)ethanone oxime and oxime ether derivatives and their
2. Baron AD. Postprandial hyperglycaemia and α-glucosidase inhibitors. Diabetes Res anticonvulsant and antimicrobial activities. Eur J Med Chem. 2001;36:421–433.
Clin Pract. 1998;40:S51–S55. 19. Yar M, Bajda M, Shahzad S, et al. Organocatalyzed solvent free an efficient novel
3. Howe JD, Smith N, Lee MR, et al. Novel imino sugar α-glucosidase inhibitors as synthesis of 2,4,5-trisubstituted imidazoles for α-glucosidase inhibition to treat
antiviral compounds. Bioorg Med Chem. 2013;21:4831–4838. diabetes. Bioorg Chem. 2015;58:65–71.
4. Pili R, Chang J, Partis RA, Mueller RA, Chrest FJ, Passaniti A. The α-glucosidase I 20. Chaudhry F, Choudhry S, Huma R, et al. Hetarylcoumarins: synthesis and biological
inhibitor castanospermine alters endothelial cell glycosylation, prevents angiogen- evaluation as potent α-glucosidase inhibitors. Bioorg Chem. 2017;73:1–9.
esis, and inhibits tumor growth. Cancer Res. 1995;55:2920–2926. 21. Chaudhry F, Naureen S, Huma R, et al. In search of new α-glucosidase inhibitors:
5. Shimada Y, Nishimura E, Hoshina H, et al. Proteasome inhibitor bortezomib en- Imidazolylpyrazole derivatives. Bioorg Chem. 2017;71:102–109.
hances the activity of multiple mutant forms of lysosomal α-glucosidase in pompe 22. Agalave SG, Maujan SR, Pore VS. Click chemistry: 1,2,3-triazoles as pharmacophores.
disease. JIMD Rep. 2015;18:33–39. Chem Asian J. 2011;6:2696–2718.
6. Park SH, Kim JY, Kim JS, Jung C, Song DK, Ham WH. 1,3-Oxazine as a chiral building 23. Chinthala Y, Thakur S, Tirunagari S, et al. docking and ADMET studies of novel
block used in the total synthesis of (+)-1-deoxynojirimycin and (2R,5R)-dihydrox- chalcone triazoles for anti-cancer and anti-diabetic activity. Eur J Med Chem.
ymethyl-(3R,4R)-dihydroxypyrrolidine. Tetrahedron Asymmetry. 2015;26:657–661. 2015;93:564–573.
7. Hollander P. Safety profile of acarbose, an α-glucosidase inhibitor. Drugs. 24. da Rocha DR, Santos WC, Lima ES, Ferreira VF. Synthesis of 1,2,3-triazole glyco-
1992;44:47–53. conjugates as inhibitors of α-glucosidases. Carbohydr Res. 2012;350:14–19.
8. Wardrop DJ, Waidyarachchi SL. Synthesis and biological activity of naturally oc- 25. Wang G, Peng Z, Wang J, Li J, Li X. Synthesis and biological evaluation of novel
curring α-glucosidase inhibitors. Nat Prod Rep. 2010;27:1431–1468. 2,4,5-triarylimidazole-1,2,3-triazole derivatives via click chemistry as α-glucosidase
9. Xie W, Tanabe G, Xu J, et al. Research progress of synthesis and structure-activity inhibitors. Bioorg Med Chem Lett. 2016;26:5719–5723.
relationship studies on sulfonium-type α-glucosidase inhibitors isolated from Salacia 26. Saeedi M, Mohammadi-Khanaposhtani M, Pourrabia P, et al. Design and synthesis of
genus plants. Mini-Rev Org Chem. 2013;10:141–159. novel quinazolinone-1,2,3-triazole hybrids as new anti-diabetic agents: In vitro α-
10. Saeedi M, Hadjiakhondi A, Nabavi SM, Manayi A. Heterocyclic compounds: effective glucosidase inhibition, kinetic, and docking study. Bioog Chem. 2019;83:161–169.
α-amylase and α-glucosidase inhibitors. Curr Top Med Chem. 2017;17:428–440. 27. Adib M, Peytam F, Rahmanian-Jazi M, et al. New 6-amino-pyrido[2,3-d]pyrimidine-
11. Zhang L, Peng XM, Damu GL, Geng RX, Zhou CH. Comprehensive review in current 2,4-diones as novel agents to treat type 2 diabetes: a simple and efficient synthesis, α-
developments of imidazole-based medicinal chemistry. Med Res Rev. glucosidase inhibition, molecular modeling and kinetic study. Eur J Med Chem.
2014;34:340–437. 2018;155:353–363.
12. Wang XQ, Liu LX, Li Y, et al. Design, synthesis and biological evaluation of novel 28. Zhang XQ, Mou XF, Mao N, et al. Design, semisynthesis, α-glucosidase inhibitory,
hybrid compounds of imidazole scaffold-based 2-benzylbenzofuran as potent antic- cytotoxic, and antibacterial activities of p-terphenyl derivatives. Eur J Med Chem.
ancer agents. Eur J Med Chem. 2013;62:111–121. 2018;146:232–244.
13. Nath M, Saini PK, Kumar A. New di- and triorganotin (IV) complexes of tripodal 29. Mauldina MG, Sauriasari R, Elya B. α-Glucosidase inhibitory activity from ethyl
Schiff base ligand containing three imidazole arms: Synthesis, structural character- acetate extract of Antidesma bunius (L.) Spreng stem bark containing triterpenoids.
ization, anti-inflammatory activity and thermal studies. J Organomet Chem. Pharmacogn Mag. 2017;13:590–594.
2010;695:1353–1362. 30. Mahdavi M, Ashtari A, Khoshneviszadeh M, et al. Synthesis of new benzimidazole-
14. Pandey J, Tiwari VK, Verma SS, et al. Synthesis and antitubercular screening of 1,2,3-triazole hybrids as tyrosinase inhibitors. Chem Biodiversity. 2018;15:e1800120.
imidazole derivatives. Eur J Med Chem. 2009;44:3350–3355.

You might also like