Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

http://pubs.acs.

org/journal/acsodf Article

Conventional and Microwave-Assisted Synthesis, Antitubercular


Activity, and Molecular Docking Studies of Pyrazole and Oxadiazole
Hybrids
Nisheeth C. Desai,* Kandarp Bhatt, Jahnvi Monapara, Unnat Pandit, and Vijay M. Khedkar
Cite This: ACS Omega 2021, 6, 28270−28284 Read Online

ACCESS Metrics & More Article Recommendations *


sı Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Microwave-assisted organic reaction enhancement (MORE)


has become more important in synthetic organic chemistry for efficient
resource utilization. In this study, we synthesized bioactive compounds
Downloaded via 175.100.128.2 on March 10, 2022 at 05:49:41 (UTC).

using both traditional and microwave methods. Microwave-assisted


synthesis takes less time and produces higher yields and quality than
conventional approaches. We reported the synthesis of N′-(1-(2-(3-(4-
chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-
yl)ethylidene) substituted hydrazides (4a−t). We also tested them against
two strains: M. tuberculosis H37Ra and M. bovis BCG. Against M. tuberculosis
H37Ra, the compounds 4e, 4h, 4k, 4p, and 4s were the most effective.
Compounds 4f, 4g, and 4s showed significant activity against M. bovis BCG.
The structures of newly synthesized molecules were determined using
spectral methods. Furthermore, molecular docking investigations into the
active site of mycobacterial InhA yielded well-clustered solutions for these
compounds’ binding modalities producing a binding affinity in the range of −10.366 to −8.037. Theoretical results were in good
accord with the observed experimental values. The docking score of compound 4e was −10.366, and the Glide energy was −66.459
kcal/mol.

■ INTRODUCTION
In the recent past, 10 million new cases of tuberculosis were
products.27 Microwave-assisted synthesis takes less time and
produces higher yields of derivatives.
Strategically constructed innovative antitubercular drugs can
reported worldwide.1 Despite the fact that medications to treat
exemplify two or more active scaffolds to create a single
tuberculosis are available in the market, bacteria started molecule with improved biological potency in the hybrid
developing resistance toward drugs like isoniazid and strategy to integrate physiologically active heterocycles. The
rifampicin. Due to this, there is a continuous need to search novel pyrazole and oxadiazole hybrids described in this paper
for improved medicines. Pyrazole is an interesting heterocycle could be a valuable addition for the development of new
to investigate. It has also gotten a lot of interest because it is antitubercular agents with a novel mechanism of action. We
being studied for a broad-spectrum biological property.2−14 created scaffolds 4a−t by incorporating the pharmacophore
The 1,3,4-oxadiazole is being studied because of its potentially feature of thioacetazone (antitubercular antibiotic), furamizole
relevant pharmacological activity.15−26 (antibiotic), and lonazolac (anti-inflammatory) in pursuit of
To improve antitubercular activity, an efficient synthetic novel heterocycles with potency to deal with tuberculosis
protocol for combining the pyrazole moiety with oxadiazole resistance. The structure−activity relationship (SAR) of the
has been established. Figure 1 depicts commercially accessible various pharmacophores was taken into account when
pharmaceuticals with pyrazole and 1,3,4-oxadiazole motifs. We designing the molecule (Figure 1). By changing the side
present the synthesis of certain novel structural hybrids of chain of thioacetazone and altering the dihydrooxadiazole ring
pyrazole and 1,3,4-oxadiazole pharmacophores in a single of furamizole, the novel scaffolds 4a−t were created. The
molecular framework to investigate their antitubercular efficacy
in vitro. Scheme 1 depicts the traditional and microwave- Received: August 15, 2021
assisted synthesis pathways for named compounds. Accepted: September 24, 2021
Microwave-assisted organic reaction enhancement (MORE) Published: October 11, 2021
has gained popularity because microwave-assisted procedures
offer the advantage of maximizing resource utilization and
providing a pure product with fewer undesirable side
© 2021 The Authors. Published by
American Chemical Society https://doi.org/10.1021/acsomega.1c04411
28270 ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Figure 1. Design concept based on commercially available drugs comprising the heterocycles pyrazole and oxadiazole.

Scheme 1. Synthetic Pathway of Titled Compounds 4a-t

inclusion of an active pharmacophore was linked to the substituted phenyl rings containing distinct electron-donating
introduction of a 1,3,4-substituted pyrazole ring from and electron-withdrawing groups.
lonazolac. The phenyl ring of thioacetazone was replaced by We have reported several 1-(2-(1H-indol-3-yl)-5-(pyridin-4-
a five-membered heterocyclic ring (dihydro-1,3,4-oxadiazole), yl)-1,3,4-oxadiazol-3(2H)-yl)-3-(aryl)prop-2-en-1-ones28 and
and the side chain of thioacetazone was terminated by investigated their antitubercular efficacy in vitro. The results
28271 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Figure 2. Comparison and structural modifications in our previous study based on oxadiazole hybrids with the reported work.

revealed that the compounds tested have antitubercular 1-phenyl-1H-pyrazole−4-carbaldehyde)benzohydrazide (2),


activity. The antitubercular potency of 1-(2-(1H-indol-3-yl)- was prepared by dissolving 3-(4-chlorophenyl)-1-phenyl-1H-
5-(pyridin-4-yl)-1,3,4-oxadiazol-3(2H)-yl)-3-(2-nitrophenyl)- pyrazole-4-carbaldehyde (1) and benzohydrazide in 1,4-
prop-2-en-1-one with MIC and IC50 values against M. bovis dioxane and refluxing for 4 h. The generation of chalcone
BCG in the active state was 1.03 and 0.48 μg/mL, respectively, derivative (2) is explained by the nucleophilic attack of
among the studied substances. The active molecule had a nitrogen on the carbonyl carbon of aldehyde followed by
docking score of −8.267 and a Glide energy of −54.856 kcal/ proton transfer, protonation, removal of water molecule, and
mol when it docked into the active site of the mycobacterial deprotonation. The intermediate 1-(2-(3-(4-chlorophenyl)-1-
enoyl reductase (InhA) enzyme. These findings piqued our phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
interest in developing new hybrids based on the oxadiazole ethenone (3) is produced by refluxing the chalcone derivative
pharmacophore. Herein, we reported the synthesis of N′-(1-(2- (2) in acetic anhydride at 140 °C for 5 h. A well-dissolved
(3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl- mixture of intermediate (3), appropriate benzohydrazides, and
1,3,4-oxadiazol-3(2H)-yl)ethylidene) substituted hydrazides a pinch of anhydrous ZnCl2 in ethanol is refluxed for 7−9 h to
(4a−t) and in vitro antitubercular efficacy. The structural yield N′-(1-(2-(3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-
changes, such as replacing the indole moiety with a substituted 5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethylidene) substituted hy-
pyrazole ring, increased the pharmacological potency. The drazides (4a−t). Microwave-assisted synthesis of the identical
Schiff base linker was used to replace the chalcone linker compounds reduces reaction time from 7−9 h to 9−10 min,
between the nitrophenyl ring and the oxadiazole ring. At three resulting in a 79−92% improvement in product yield (see
different concentrations, the synthesized compounds showed Table 1). In the experimental part, the details of microwave
substantial inhibition. In the active state, compound 4e had synthesis are described.
excellent antitubercular activity against M. tuberculosis H37Ra, The final structures of N′-(1-(2-(3-(4-chlorophenyl)-1-
with MIC and IC50 values of 0.92 and 0.52 μg/mL, phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
respectively. The docking score of compound 4e was ethylidene) substituted hydrazides (4a−t) were confirmed by
−10.366, and the Glide energy was −66.459 kcal/mol. Figure different spectroscopic analysis data. Compound 4c exhibited a
2 shows the structural alterations, as well as a comparative distinct absorption band at 1691 cm−1, which corresponded to
study of activity and molecular docking research.


the presence of a carbonyl group in the structure. The Schiff
base’s N−H stretching (secondary amide stretching) and O−
RESULTS AND DISCUSSION H stretching of the −OH group linked to the phenyl ring
Chemistry. The traditional and microwave-assisted syn- caused different vibrations at 3367 and 3447 cm−1. The
thesis pathways for novel N′-(1-(2-(3-(4-chlorophenyl)-1- aromatic ring’s C−H stretching and the −CHCH− group
phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- produced vibrations at 3066 and 3024 cm−1, respectively. The
ethylidene) substituted hydrazides (4a−t) are shown in C−O−C stretching of the oxadiazole nucleus and C−Cl
Scheme 1. The chalcone derivative, N′-(3-(4-chlorophenyl)- stretching caused absorptions at 1232 and 747 cm−1. A singlet
28272 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Table 1. Optimization of Reaction Conditions for the hydroxyl proton, respectively. The presence of the −CONH−
Conventional and Microwave Methods group in the molecule’s arrangement was demonstrated by a
singlet peak at 12.30 ppm.
conventional method microwave method
The methyl carbon of the methyl group produced a signal at
yield reaction time yield reaction time 12.9 ppm in 13C NMR, while the carbon connected to the
entry -R (%) (min) (%) (min)
−CH3 group produced a signal at 147.3 ppm. Two signals
4a -C6H5 77 420 92 10 appeared due to carbons of the oxadiazole ring at 157.6 and
4b -2-OH-C6H4 65 450 80 9 78.4 ppm, proving the presence of the oxadiazole ring, while
4c -3-OH-C6H4 69 450 83 8
three signals appeared due to carbons of the pyrazole ring at
4d -4-OH-C6H4 61 450 80 9
117.9, 123.1, and 149.4 ppm, proving the presence of the
4e -4-NO2-C6H4 58 420 79 8
pyrazole ring in the structure of compound 4c. At 163.7 ppm,
4f -3-CH3-C6H4 73 450 90 9
the carbonyl carbon of the amide group produced a distinct
4g -4-CH3-C6H4 71 450 90 8
signal. In mass spectra, m/z = 577.17 corresponds to the
4h -4-OCH3-C6H4 62 450 85 9
molecular weight suggested. The details of spectral information
4i -3-Cl-C6H4 68 480 88 10
are given in the Supplementary Information.
4j -4-Cl-C6H4 74 480 92 10
4k -CH2-C6H5 57 450 79 9
Biological Evaluation. Antimycobacterial Activity. In
4l -CH2NHCO- 60 540 81 10
both active and dormant modes, the newly synthesized
C6H5 compounds demonstrated good effectiveness against M. bovis
4m -CH2-O-C6H4-2- 64 540 85 10 BCG and M. tuberculosis H37Ra. The antitubercular action of
CH3 the pyrazole and oxadiazole hybrids 4e, 4h, 4k, 4p, and 4s
4n -CH2-O-C6H4-3- 63 540 85 9 against M. tuberculosis H37Ra was described. The compounds
CH3
4e, 4k, and 4p had excellent MICs against M. tuberculosis
4o -CH2-O-C6H4-4- 70 540 90 9
CH3 H37Ra in an active state, ranging from 0.92 to 2.56 μg/mL.
4p -CH2-O-C6H4-2- 61 480 83 10 Compound 4e was shown to be the most effective against
NO2 active M. tuberculosis H37Ra, with an active MIC of 0.92 μg/
4q -CH2-O-C6H4-3- 55 480 80 9 mL and a dormant MIC of 1.26 μg/mL. In active and dormant
NO2
stages, compound 4k had MICs of 1.31 and 1.53 μg/mL,
4r -CH2-O-C6H4-4- 66 480 85 9
NO2 respectively, against M. tuberculosis H37Ra. Compound 4p was
4s -CH2-O-C6H4-2- 73 450 91 8 around 2 to 3 times less active against M. tuberculosis H37Ra
Cl than the most active compound 4e (active state MIC: 2.56 μg/
4t -CH2-O-C6H4-4- 69 450 87 8 mL, dormant state MIC: 2.32 μg/mL). In a dormant
Cl condition, compound 4s had an excellent MIC value of 1.03
μg/mL against M. tuberculosis H37Ra. Compounds 4e, 4h, 4k,
signal at 5.69 ppm in the 1H NMR spectra of compound 4c 4p, and 4s had MICs ranging from 1.03 to 2.32 μg/mL against
was attributable to the oxadiazole ring’s C2−H, while peaks at M. tuberculosis H37Ra in a dormant condition. Compounds 4f
2.17 and 5.30 ppm were three −CH3 group protons and one (active state MIC: 2.71 μg/mL, dormant state MIC: 2.47 μg/

Table 2. Results of Primary Screening against M. tuberculosis H37Ra and M. bovis BCG of N′-(1-(2-(3-(4-Bromophenyl)-1-
phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethylidene)-(aryl)-benzohydrazides

at dormant M. tuberculosis H37Ra at dormant M. bovis BCG


entry -R 30 μg/mL 10 μg/mL 3 μg/mL 30 μg/mL 10 μg/mL 3 μg/mL
4a -C6H5 61.72 51.64 32.34 40.34 34.70 20.43
4b -2-OH-C6H4 54.14 60.21 21.99 100.53 100.80 99.85
4c -3-OH-C6H4 37.94 46.17 14.61 0.29 6.92 7.51
4d -4-OH-C6H4 98.45 94.86 14.86 99.55 100.57 99.63
4e -4-NO2-C6H4 95.09 92.48 11.22 99.55 100.38 38.49
4f -3-CH3-C6H4 98.47 85.31 25.24 99.24 100.48 98.79
4g -4-CH3-C6H4 7.98 19.22 3.22 96.14 98.65 99.47
4h -4-OCH3-C6H4 95.44 91.47 86.22 97.25 99.82 100.25
4i -3-Cl-C6H4 46.72 26.60 18.61 99.03 81.67 15.34
4j -4-Cl-C6H4 28.24 3.92 35.08 75.22 19.21 20.53
4k -CH2-C6H5 98.74 93.71 96.13 97.77 89.29 56.75
4l -CH2NHCO-C6H5 3.09 −17.07 35.30 58.41 21.15 8.60
4m -CH2-O-C6H4-2-CH3 38.95 7.09 33.03 37.25 37.51 33.01
4n -CH2-O-C6H4-3-CH3 −22.83 −35.88 3.56 71.56 4.17 −18.98
4o -CH2-O-C6H4-4-CH3 85.53 78.95 0.75 81.96 48.21 −1.06
4p -CH2-O-C6H4-2-NO2 95.19 90.23 94.58 98.81 94.62 91.41
4q -H2-O-C6H4-3-NO2 34.13 31.12 13.95 7.79 0.04 −7.87
4r -CH2-O-C6H4-4-NO2 54.11 40.25 7.88 33.24 26.42 20.26
4s -CH2-O-C6H4-2-Cl 93.68 98.02 93.52 82.60 86.27 83.22
4t -CH2-O-C6H4-4-Cl 11.41 −1.79 −4.38 9.62 25.12 13.69

28273 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Table 3. Antitubercular Screening Results of the Tested Compounds against Mycobacterium tuberculosis H37Ra and
Mycobacterium bovis BCG Strainsa,b

28274 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Table 3. continued

a
MIC = minimum inhibitory concentration in μg/mL. bIC50 = 50% inhibitory concentration in μg/mL. cRifampicin = reference for Mycobacterium
tuberculosis H37Ra and Mycobacterium bovis BCG strains. dIsoniazid = reference for Mycobacterium tuberculosis H37Ra and Mycobacterium bovis BCG
strains.

mL) and 4g (active state MIC: 2.19 μg/mL, dormant state compound 4s had an excellent MIC value of 0.79 μg/mL. The
MIC: 2.12 μg/mL) were found to have excellent MIC values antitubercular action was greatly impacted by the substitution
for M. bovis BCG. In the active state of M. bovis BCG, pattern at the phenyl ring, as shown in Tables 2 and 3. The
28275 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Figure 3. Primary and secondary antitubercular screening of pyrazole−oxadiazole hybrids (4a−t) revealed a structure−activity relationship.

antitubercular activity against M. tuberculosis H37Ra was action against the M. tb. strain. It inhibited the M. tb. strain by
affected by electron-withdrawing groups like −NO2 and −Cl, 95.44% in the primary screening, with MIC values of 19.42 and
but the antitubercular activity against M. bovis BCG was 1.12 μg/mL in active and dormant states, respectively.
affected by electron-donating groups like −CH3 and −OCH3. The antitubercular activity of both the screening, the
The antitubercular activity of titled compounds 4a−t was electron-donating −CH3 group at the third and fourth
determined using a Nitrate reductase assay and an XTT positions had showed excellent activity against the M. bovis
Reduction Menadione assay, respectively, against M. tuber- strain. In primary screening, a 3-CH3 derivative of the
culosis H37Ra and M. bovis BCG. The primary screening was benzohydrazide motif showed 99.24% inhibition and a MIC
done at doses of 30, 10, and 3 μg/mL at first. For the current of 2.71 μg/mL in the active state and 2.47 μg/mL in the
study, we chose drugs that showed 90% bacilli inhibition at or dormant state against the M. bovis strain. In the active and
below 30 μg/mL for the dose−response curve. We chose the dormant states, the 4-CH3 derivative of the benzohydrazide
drugs rifampicin and isoniazid as a reference, both of which are scaffold inhibited M. bovis with a MIC of 2.19 and 2.12 μg/mL,
commercially accessible for clinical use.29 Tables 2 and 3 respectively. In both screenings, the SAR study of benzohy-
summarize the findings. drazide derivatives revealed that electron-donating tendencies
Structure−Activity Relationship. We created some novel were more prevalent than electron-withdrawing tendencies.
pyrazole−oxadiazole hybrids as possible antitubercular agents The functional groups like −CH3 and −OCH3 at various
in this study. The current study’s findings revealed some positions showed increased effectiveness against M. tb. and M.
encouraging results in terms of the structure−activity relation- bovis strains. Electron-withdrawing functional groups in
ship. To investigate the impact of functional groups on acetohydrazide derivatives exhibited good efficacy against the
antitubercular assessment, we used a variety of electron- M. tb. strain. In primary screening, the −NO2 group at the
withdrawing and electron-donating functional groups. Func- second position showed 95.19% inhibition against the M. tb.
tional groups have played a key role in improving strain, with MIC values of 2.56 μg/mL in the active state and
antitubercular activity in both primary and secondary screen- 2.32 μg/mL in the dormant state. The electron-withdrawing
ing. In primary and secondary screening, the electron- −Cl group in the second position showed 93.68% inhibition in
withdrawing −NO 2 group at the fourth position in primary and secondary screening against the M. tb. strain,
benzohydrazide derivatives exhibited significant antitubercular having MIC values of 24.43 and 1.03 μg/mL in active and
action. In initial screening, it displayed 95.09% inhibition dormant states, respectively. The electron-withdrawing −NO2
against the M. tb. strain, with MIC values of 0.92 and 1.26 μg/ and −Cl groups at the second position are more prominent
mL in active and dormant states, respectively. In primary and than electron-donating groups at various positions, according
secondary screening, the electron-donating −OCH3 group, to an evaluation of acetohydrazide derivatives. The cumulative
which was placed at the fourth position, showed significant impacts of active motifs of pyrazole−oxadiazole hybrids (4a−
28276 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

t) on the structure−activity relationship are demonstrated in

Ser94 (2.41),
H-bonding
Figure 3.

(2.57),

(2.77)

(1.98)

(2.30)

(2.14)

(1.99)
Ala22

Ser20

Thr196
Lys165

Lys165

Lys165
Molecular Docking Study. Molecular docking is now a
well-established computational technique for predicting the
predominant binding mode(s) of a ligand within a protein

(2.135)

(2.362)

(2.644)
stacking
(biological target) of a given three-dimensional structure, and

Phe149

Phe149
π−π

Phe97
it has become a vital aspect of drug development research. It
gives researchers information on binding affinities, binding

(−1.122), Thr196 (−1.626),


(−1.711), Lys165 (−2.343),

(−1.199), Lys165 (−2.278),

(−1.879), Ser209 (−1.298),


mechanisms, and types of interactions that guide protein−

(−1.008), Ser20 (−1.905)

(−2.797), Ser20 (−1.246)

Lys165 (−2.62), Met147


electrostatic (kcal/mol)
ligand complexation, allowing them to develop and examine

Ile219 (−1.587), Lys165


Ile210 (−1.968), Ser209

Ile219 (−1.134), Ser209

Ile219 (−1.949), Ile210


115 (−1.104), Gly96
261 (−0.972), Met199
115 (−1.527), Ala22
the potential of existing lead compounds and their equivalents.

169 (−1.101), 148


In the absence of resources to perform enzymatic assays, in
silico computational chemistry techniques such as molecular

(−1.019)

(−1.514)

(−0.907)
docking have become critical for identifying the targets for
various molecules and their associated thermodynamic
interactions with the target enzyme that govern pathogen
inhibition. Mycobacterial enoyl reductase is said to be

(−1.928), Tyr158 (−1.92), Phe149 (−2.145), Met147 (−1.768), Met103 (−2.843), 99 (−1.067), Met98 (−1.018), Met97 (−1.186), Gly96
(−1.343), Ala191 (−1.968), Lys165 (−1.685), Met161 (−2.058), Tyr158 (−7.209), Ala157 (−1.567), Met155 (−1.093), Phe149 (−4.211),
Met147 (−2.55), Met103 (−4.69), Met98 (−1.443), Met97 (−1.903), Gly96 (−1.844), Ile95 (−0. 956), Ser94 (−2.361), Ile21 (−2.832),

Ala198 (−2.206), 197 (−1.785), Thr196 (−2.607), Lys165 (−1.098), Met161 (−2.168), Phe149 (−0.964), 148 (−1.325), Met147 (−2.952),

Ile215 (−2.153), Ile202 (−2.073), Met199 (−2.55), Ala198 (−2.256), Thr196 (−3.008), Ile194 (−2.197), Pro193 (−1.352), 192 (−0.964),
122 (−1.526), Met103 (−1.463), 100 (−1.971), 99 (−1.184), Met98 (−1.394), Met97 (−1.587), Gly96 (−2.034), Ile95 (−2.61), Ser94
inhibited by the oxadiazole scaffold (InhA).30−32 As a result,

Ala191 (−1.812), Met161 (−3.095), Tyr158 (−4.064), Phe149 (−4.414), 148 (−1.567), Met147 (−3.112), Met103 (−3.963), Met98
(−1.636), Tyr158 (−2.63), Ala157 (−0.931), Phe149 (−3.592), 148 (−1.487), Met147 (−2.197), Met103 (−3.31), Met98 (−1.465),
Ile215 (−2.438), Leu207 (−1.711), Ile202 (−1.788), Met199 (−2.915), Ala198 (−2.376), Thr196 (−3.184), Ile194 (−2.484), Pro193

Ile215 (−1.36), Leu207 (−1.033), Ile202 (−2.438), Met199 (−2.054), Ala198 (−2.077), Thr196 (−1.48), Lys165 (−1.782), Met161
the 3D structure of M. tuberculosis mycobacterial enoyl

Ile215 (−1.248), Leu207 (−1.001), Ile202 (−1.748), Met199 (−1.831), Ala198 (−2.032), 197 (−0.967), Thr196 (−1.522), Met161
reductase (InhA) complexed with its inhibitor was chosen as
the target protein for the molecular docking study for the

Table 4. Results of the Per-Residue Interaction Analysis for the Pyrazole Clubbed Oxadiazole Hybrid Heterocycles
active compounds (4e, 4f, 4g, 4p, and 4s) in the series to

per-residue interaction energy analysis

Met97 (−1.211), Gly96 (−2.566), Ile95 (−1.145), Ser94 (−1.875), Ile21 (−1.679), Ser20 (−1.193), Ile16 (−0.987)
rationalize the promising antitubercular results and investigate
the possible interactions of the studied compounds. The enoyl-
ACP (CoA) reductase (FabI/ENR/InhA) is a key enzyme in

(−2.111), 41 (−2.473), Ile21 (−2.046), Ser20 (−1.798), Ser19 (−1.078), Thr17 (−1.89), Ile16 (−2.208)
the type II fatty acid biosynthesis pathway in mycobacteria,
and its inhibition stunts the growth of a fast-growing model
organism. Mycobacterium smegmatis has been demonstrated to
block mycolic acid formation and promote cell lysis,33 making

(−1.447), Met97 (−1.818), Gly96 (−2.095), Ile21 (−2.087), Ser20 (−1.446), Ile16 (−1.076)
(−2.182), Ile95 (−1.439), Ser94 (−1.943), Ile21 (−1.881), Ser20 (−1.187), Ile16 (−1.326)
it a suitable target for medication development against
tuberculosis.
The outcome of the molecular docking study revealed that
van der Waals (kcal/mol)

all the studied compounds (4e, 4f, 4g, 4p, and 4s) were
successfully docked, making a snug fit into the active site of
InhA at coordinates very close to that of the native ligand in
the crystal structure with varying degrees of affinities. Their
complexes with protein structure were stabilized by several
bonded and nonbonded interactions with the amino acids
surrounding the active site. The findings of the molecular
docking investigation are described in Table 4, which shows
that there is a substantial association between docking scores
and biological activity, with active molecules producing higher
scores and those with low activity producing lower docking
scores. Their binding energies were likewise found to be
negative (−66.459 to −55.157 kcal/mol), indicating that they
Ser20 (−1.992), Gly14 (−1.127)

could be used to rationally design compounds that target InhA.


Furthermore, the thermodynamic elements influencing the
binding of these compounds to InhA were investigated in-
depth by analyzing per-residue interactions between the
enzyme and these ligands. However, for the sake of brevity,
we have discussed these details for compound 4e, which is the
most active, while we summarized for other molecules in Table
4.
The best docked configuration of the most active oxadiazole
derivative 4e into the active site of mycobacterial enoyl
−66.459

−55.157

−58.133

−62.547

−64.465
binding
energy

reductase (InhA) (Figure 4) revealed that the inhibitor snugly


fits the active site, making multiple intimate contacts through
bonded and nonbonded interactions. This molecule has the
highest affinity in the series (docking score of −10.366 and
−10.366

−8.037

−8.159

−8.909

−9.972
docking
score

binding energy of −66.459 kcal/mol), which can be explained


in terms of unique bonded and nonbonded per-residue
interactions with the active site residues. The visual inspection
entry

4p
4g
4e

4s
4f

of the bound pose of 4e revealed that the compound is


28277 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Figure 4. Binding mode of compound 4e into the active site of mycobacterial enoyl reductase (InhA) (on the right side: the π−π stacking
interaction is represented using green lines, while pink lines represent the hydrogen bonding interactions).

stabilized within the active site through an extensive network of


favorable van der Waals contacts with Ile215 (−2.438 kcal/
■ CONCLUSIONS
The findings of the investigation suggested that the titled
mol), Ile202 (−1.788 kcal/mol), Met199 (−2.915 kcal/mol), compounds will be important in the development of
Ala198 (−2.376 kcal/mol), Thr196 (−3.184 kcal/mol), Ile194 antitubercular drugs in the future. Microwave-assisted syn-
(−2.484 kcal/mol), Pro193 (−1.343 kcal/mol), Ala191 thesis and the hybrid molecule idea were used to produce and
(−1.968 kcal/mol), Tyr158 (−7.209 kcal/mol), Ala157 develop the active chemicals. Our findings showed that the
(−1.567 kcal/mol), and Phe149 (−4.211 kcal/mol) through chemical transformation took place in a relatively short period,
the N′-(1-(2-(3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)- the purity of the compounds was higher, and the yields were
5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethylidene)-4-nitrobenzo- higher than the traditional approach. As a result, the
hydrazidebenzohydrazide component of 4e got engaged in a pharmaceutical industry will rely heavily on this technique.
similar network of van der Waals interactions with Leu207 Our previously published work was used to describe the
(−1.711 kcal/mol), Lys165 (−1.685 kcal/mol), Met161 compounds (Figure 2). The fact that the molecule with the
(−2.058 kcal/mol), Met155 (−1.093 kcal/mol), Met147 nitro group at the para position has outstanding activity and a
(−2.55 kcal/mol), Met103 (−4.69 kcal/mol), Met98 high docking score motivated us to develop new hybrids based
(−1.443 kcal/mol), Met97 (−1.903 kcal/mol), Gly96 on the oxadiazole core structure and explore their
(−1.844 kcal/mol), Ile95 (−0.956 kcal/mol), Ser94 (−2.361 antitubercular activity. The study found that compounds
kcal/mol), Ile21 (−2.832 kcal/mol), Ser20 (−1.992 kcal/mol), with electron-withdrawing functional groups had excellent
and Gly14 (−1.127 kcal/mol) residues lining the active site of MICs against M. tuberculosis H37Ra, whereas compounds with
InhA. Significant electrostatic interactions with Ile210 (−1.968 electron-donating functional groups have antitubercular action
kcal/mol), Ser209 (−1.711 kcal/mol), Lys165 (−2.343 kcal/ against M. bovis BCG but no activity against M. tuberculosis
mol), 115 (−1.527 kcal/mol), Ala22 (−1.008 kcal/mol), and H37Ra. The pyrazole−oxadiazole hybrids 4e, 4h, 4p, and 4s
Ser20 (−1.905 kcal/mol) residues in the active site are also were found to be most effective against M. tuberculosis H37Ra,
responsible for 4e’s increased binding affinity. Furthermore, at while 4f and 4g have substantial activity against M. bovis BCG.
bonding distances of 2.41, 2.25, and 2.77, respectively, a strong Molecular docking studies were used to gain an insight into the
hydrogen bonding interaction was detected between the nitro mechanism of action, which suggested that these molecules
group and the Ser94, Ala22, and Ser20 residues. A significant possess significant binding affinity toward the critical target
π−π stacking was also observed between the Phe149 (2.135) mycobacterium InhA. These findings revealed important
and the p-chlorophenyl ring of 4e. Such hydrogen bonding information about possible binding interactions, implying
interactions and the pi−pi stacking serve as an ″anchor″ that this class of compounds can be further selected to build
guiding the 3D space position of the ligand in the active site InhA specific inhibitors. Iterative synthesis and computer
and facilitate the steric and electrostatic interactions adding to modeling are being used to make a variety of structural
the stability of the enzyme-inhibitor complex. The other changes, and the results will be shared as soon as possible.


oxadiazole analogues (4f, 4g, 4p, and 4s) were also seen to
possess an optimum binding affinity for the enzyme engaging
in a similar network of interactions with the active side residues EXPERIMENTAL SECTION
but quantitatively decreasing gradually with their observed Materials and Methods. Melting points were determined
antitubercular activity. Overall, the per-residue interaction on an electrothermal melting point apparatus and are reported
analysis revealed that the primary driving forces for mechanical uncorrected. The completion of the reaction and purity of all
interlocking of these ligands are the steric complementarity compounds were checked on aluminum-coated TLC plates 60
between these ligands and the active site of InhA that is F254 (E. Merck) using chloroform/methanol (9.5:0.5 v/v) as
reflected in the relatively higher number of favorable van der the mobile phase and visualized under ultraviolet (UV) light or
Waals interactions compared to other components contribu- iodine vapor. Elemental analysis (% C, H, N) was carried out
ting to the overall binding affinity. by a Perkin-Elmer 2400 CHN analyzer. IR spectra of all
28278 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

compounds were taken on a Perkin-Elmer FT-IR spectropho- min at a maximum power of 250 W at 30 s intervals. The
tometer in KBr. 1H NMR and 13C NMR spectra were recorded product was extracted from methanol when the reaction
on Bruker (400 MHz) and (100 MHz) spectrometers, mixture was cooled. A crude product was obtained by diluting
respectively, using DMSO-d6 as a solvent and TMS as an methanolic extract with ice-cold water, which was then filtered,
internal standard. Chemical shifts are reported in parts per washed, and recrystallized from methanol to yield compound
million (ppm). A mass spectrum was scanned on a Shimadzu 3, resulting in an 88% yield.
LC−MS 2010 spectrophotometer. Anhydrous reactions were Preparation of N′-(1-(2-(3-(4-Chlorophenyl)-1-phenyl-1H-
carried out in oven-dried glassware in a nitrogen atmosphere. pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethylidene)
Microwave-assisted synthesis was carried out using Synthos Substituted Hydrazides (4a−t). 1-(2-(3-(4-Chlorophenyl)-1-
3000.34 phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
General Procedure for the Preparation of N′-(1-(2-(3- ethanone (3, 0.01 mol) and appropriate benzohydrazides (0.01
(4-Chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl- mol) were dissolved in pure alcohol (99.9%), and a pinch of
1,3,4-oxadiazol-3(2H)-yl)ethylidene) Substituted Hydra- anhydrous ZnCl2 was added. Under microwave irradiation, the
zides (4a-t) by the Conventional Method. Preparation of reaction mixture was magnetically agitated for 8−10 min with a
N′-((3-(4-Chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)- maximum power of 300 W intermittently at 30 s intervals. The
methylene) Benzohydrazide (2). 3-(4-Chlorophenyl)-1-phe- reaction mixture was then quenched, and the resulting
nyl-1H-pyrazole-4-carbaldehyde (1, 0.01 mol) and benzohy- precipitates were filtered, washed, and recrystallized from
drazide (0.01 mol) were dissolved in 1,4-dioxane (20 mL) and methanol to yield compounds 4a−t.
refluxed for 4 h. The crystals developed as soon as the product Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
was cooled, and the product was crystallized from methanol to nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
obtain compound 2. Yield 63%, mp 187−189 °C. ethylidene)benzohydrazide (4a). Yield 77%, mp 229−232
Preparation of 1-(2-(3-(4-Chlorophenyl)-1-phenyl-1H-pyr- °C; IR (KBr) λmax: 3365 (N−H stretching, secondary amide),
azol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethanone (3). 3066, 3024 (C−H stretching, aromatic ring), 1691 (CO
Acetic anhydride (0.03 mol) was added to compound N′- stretching), 1548, 1488 (CC, CN stretching, aromatic
((3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)methylene)- ring), 1232 (C−O−C stretching, oxadiazole ring), 747 (C−Cl
benzohydrazide (2, 0.01 mol) and refluxed at 140 °C for 5 h. stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.16 (s,
The resulting mixture was transferred to crushed ice once the 3H, −CH3), 5.65 (s, 1H, C2−H, oxadiazole ring), 6.88−8.04
reaction mixture reached the room temperature. The (m, 20H, Ar−H and C5−H pyrazole), 12.36 (s, 1H,
precipitates were filtered and crystallized with methanol after −CONH); 13C NMR (DMSO-d6, 100 MHz): 12.5, 78.2,
being oven-dried, yielding compound 3. Yield 73%, mp 180− 117.6, 119.4 (2), 123.4, 125.3 (2), 126.1, 127.3 (2), 128.7 (4),
182 °C. Anal. calcd for C25H19ClN4O2: C-67.79, H-4.32, N- 128.8 (2), 129.3 (4), 131.2, 131.7, 132.1, 132.5, 132.6, 134.8,
12.62; found: C-67.86, H-4.4, N-12.6%. 139.4, 147.3, 149.2, 157.6, 163.8; LCMS: m/z = 561.2 (M+);
Preparation of N′-(1-(2-(3-(4-Chlorophenyl)-1-phenyl-1H- anal. calcd for C32H25ClN6O2: C, 68.51; H, 4.49; N, 14.98;
pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethylidene) found: C, 68.62; H, 4.55; N, 15.00%.
Substituted Hydrazides (4a-t). An equimolar mixture of Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
intermediate compound 1-(2-(3-(4-chlorophenyl)-1-phenyl- nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethanone ethylidene)-2-hydroxybenzohydrazide (4b). Yield 65%, mp
(3, 0.01 mol) and appropriate benzohydrazides (0.01 mol) was 227−230 °C; IR (KBr) λmax: 3465 (O−H stretching, Ar−OH),
dissolved in ethanol (95%) (20 mL), and a pinch of anhydrous 3370 (N−H stretching, secondary amide), 3068, 3028 (C−H
ZnCl2 was added to the mixture. The reaction mixture was stretching, aromatic ring), 1696 (CO stretching), 1550,
then refluxed for 7−9 h at 80 °C. The crystals formed after 1492 (CC, CN stretching, aromatic ring), 1200 (C−O−
cooling the reaction mixture were filtered and recrystallized C stretching, oxadiazole ring), 740 (C−Cl stretching) cm−1;
from methanol to give compound 4a−t.
1
H NMR (DMSO-d6, 400 MHz): 2.19 (s, 3H, −CH3), 5.35 (s,
General Procedure for the Preparation of N′-(1-(2-(3- 1H, −OH), 5.69 (s, 1H, C2−H oxadiazole ring), 6.92−8.09
(4-Chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl- (m, 19H, Ar−H and C5−H pyrazole), 12.35 (s, 1H,
1,3,4-oxadiazol-3(2H)-yl)ethylidene) Substituted Hydra- −CONH); 13C NMR (DMSO-d6, 100 MHz): 12.8, 78.6,
zides (4a−t) by Microwave-Assisted Synthesis. Prepara- 117.2, 117.3 (2), 119.3 (2), 119.4, 121.5, 123.4, 126.4 (2),
tion of N′-((3-(4-Chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)- 128.6, 128.9 (2), 129.1, 129.4 (4), 129.5, 129.7, 131.0, 131.2,
methylene) Benzohydrazide (2). Compounds 3-(4-chloro- 133.6, 134.8, 139.2, 147.2, 149.3, 150.2, 159.3, 163.7; LCMS:
phenyl)-1-phenyl-1H-pyrazole-4-carbaldehyde (1, 0.01 mol), m/z = 577.6 (M+); anal. calcd for C32H25ClN6O3: C, 66.61; H,
benzohydrazide (0.01 mol), and 1,4-dioxane (5 mL) were 4.37; N, 14.56; found: C, 66.63; H, 4.50; N, 14.70%.
swirled together and microwave irradiated for 30 s. This Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
reaction mixture was heated for a further 5 min. When the nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
reaction was finished, the contents were placed in crushed ice ethylidene)-3-hydroxybenzohydrazide (4c). Yield 69%, mp
with water and recrystallized in methanol to form compound 2, 209−211 °C; IR (KBr) λmax: 3447 (O−H stretching, Ar−OH),
resulting in an 82% yield. 3367 (N−H stretching, secondary amide), 3066, 3024 (C−H
Preparation of 1-(2-(3-(4-Chlorophenyl)-1-phenyl-1H-pyr- stretching, aromatic ring), 1691 (CO stretching), 1548,
azol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)ethanone (3). 1488 (CC, CN stretching, aromatic ring), 1232 (C−O−
This silica gel (1 g) was added to a combination of compound C stretching, oxadiazole ring), 747 (C−Cl stretching) cm−1;
N′-((3-(4-chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)- 1
H NMR (DMSO-d6, 400 MHz): 2.17 (s, 3H, −CH3), 5.30 (s,
methylene)benzohydrazide (2, 0.01 mol) and acetic anhydride 1H, −OH), 5.69 (s, 1H, C2−H oxadiazole ring), 6.90−8.05
(3 mL) at room temperature. Under microwave irradiation, the (m, 19H, Ar−H and C5−H pyrazole), 12.30 (s, 1H,
absorbed material was air-dried and magnetically agitated for 5 −CONH); 13C NMR (DMSO-d6, 100 MHz): 12.9, 78.4,
28279 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

115.3, 117.9, 119.4, 119.6 (2), 120.4, 123.1, 125.4 (2), 126.3, stretching), 1546, 1440 (CC, CN stretching, aromatic
128.4 (2), 128.8 (2), 129.3 (4), 130.5, 131.5, 131.9, 134.1, ring), 1240 (C−O−C stretching, oxadiazole ring), 701 (C−Cl
135.9, 139.2, 147.3, 149.4, 157.6, 158.2, 158.3, 163.7; LCMS: stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.08 (s,
m/z = 577.1 (M+); anal. calcd for C32H25ClN6O3: C, 66.61; H, 3H, −CH3), 2.34 (s, 3H, −CH3), 5.68 (s, 1H, C2−H
4.37; N, 14.56; found: C, 66.70; H, 4.50; N, 14.65%. oxadiazole ring), 6.80−7.99 (m, 19H, Ar−H and C5−H
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- pyrazole), 12.35 (s, 1H, −CONH); 13C NMR (DMSO-d6, 100
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- MHz): 12.4, 21.8, 78.1, 117.3, 119.3 (2), 123.7, 125.3 (2),
ethylidene)-4-hydroxybenzohydrazide (4d). Yield 65%, mp 126.8, 127.4 (2), 128.6 (2), 128.9 (2), 129.3 (4), 129.4 (2),
241−244 °C; IR (KBr) λmax: 3434 (O−H stretching, Ar−OH), 129.8, 131.5, 131.8, 132.8, 134.8, 139.4, 147.2, 149.7, 151.1,
3380 (N−H stretching, secondary amide), 3070, 3012 (C−H 157.3, 163.7. LCMS: m/z = 575.3 (M+); anal. calcd for
stretching, aromatic ring), 1612 (CO stretching), 1509, C33H27ClN6O2: C, 68.92; H, 4.73; N, 14.61; found: C, 68.96;
1412 (CC, CN stretching, aromatic ring), 1232 (C−O− H, 4.63; N, 14.70%.
C stretching, oxadiazole ring), 730 (C−Cl stretching) cm−1; Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
1
H NMR (DMSO-d6, 400 MHz): 2.15 (s, 3H, −CH3), 5.32 (s, nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
1H, −OH), 5.66 (s, 1H, C2−H oxadiazole ring), 6.90−8.09 ethylidene)-4-methoxybenzohydrazide (4h). Yield 62%, mp
(m, 19H, Ar−H and C5−H pyrazole), 12.36 (s, 1H, 248−251 °C; IR (KBr) λmax: 3320 (N−H stretching,
−CONH); 13C NMR (DMSO-d6, 100 MHz): 12.3, 78.1, secondary amide), 3014, 3020 (C−H stretching, aromatic
116.3 (2), 117.6, 119.4 (2), 123.7, 125.1, 125.6 (2), 126.4, ring), 2850 (C−H stretching, −OCH3),2824, 1605(CO
128.0, 128.1 (2), 128.4 (2), 128.6, 129.3 (4), 131.0, 131.7, stretching), 1554, 1428 (CC, CN stretching, aromatic
132.4, 134.8, 139.4, 147.1, 149.4, 157.4, 161.7, 163.7; LCMS: ring), 1212 (C−O−C stretching, oxadiazole ring), 758 (C−Cl
m/z = 577.2 (M+); anal. calcd for C32H25ClN6O3: C, 66.61; H, stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.05 (s,
4.37; N, 14.56; found: C, 66.62; H, 4.39; N, 14.60%. 3H, −CH3), 3.80 (s, 3H, −OCH3), 5.72 (s, 1H, C2−H
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- oxadiazole ring), 6.80−7.80 (m, 19H, Ar−H and C5−H
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- pyrazole), 12.33 (s, 1H, −CONH); 13C NMR (DMSO-d6, 100
ethylidene)-4-nitrobenzohydrazide (4e). Yield 58%, mp 271− MHz): 12.8, 55.6, 78.4, 114.7 (2), 117.7, 119.4 (2), 123.0,
273 °C; IR (KBr) λmax: 3388 (N−H stretching, secondary 125.4 (2), 126.7, 128.1, 128.4 (2), 128.7 (2), 128.8 (2), 129.4
amide), 3077, 3013 (C−H stretching, aromatic ring), 1530, (4), 131.2, 131.7, 132.0, 134.7, 139.4, 147.4, 149.4, 157.1,
1610 (CO stretching), 1530 (NO stretching, Ar−NO2), 163.7, 164.0; LCMS: m/z = 591.2 (M+); anal. calcd for
1500, 1429 (CC, CN stretching, aromatic ring), 1200 C33H27ClN6O3: C, 67.06; H, 4.60; N, 14.22; found: C, 67.09;
(C−O−C stretching, oxadiazole ring), 700 (C−Cl stretching) H, 4.70; N, 14.26%.
cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.12 (s, 3H, −CH3), Characterization of 3-Chloro-N′-(1-(2-(3-(4-chlorophen-
5.60 (s, 1H, C2−H oxadiazole ring), 6.85−8.04 (m, 19H, Ar− yl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-
H and C5−H pyrazole), 12.32 (s, 1H, −CONH); 13C NMR 3(2H)-yl)ethylidene)benzohydrazide (4i). Yield 68%, mp
(DMSO-d6, 100 MHz): 12.0, 78.7, 117.4, 119.7 (2), 123.5, 183−185 °C; IR (KBr) λmax: 3338 (N−H stretching,
124.4 (2), 125.3 (2), 126.7, 128.3 (2), 128.4 (2), 129.3 (4), secondary amide), 3023, 3020 (C−H stretching, aromatic
129.4 (2), 131.4, 134.4, 138.3, 138.4, 139.4, 147.2, 149.5, ring), 1609 (CO stretching), 1562, 1430 (CC, CN
150.7, 151.0, 157.5, 163.7; LCMS: m/z = 605.2 (M+); anal. stretching, aromatic ring), 1219 (C−O−C stretching,
calcd for C32H24ClN7O4: C, 63.42; H, 3.99; N, 16.18; found: oxadiazole ring), 775, 758 (C−Cl stretching) cm−1; 1H
C, 63.55; H, 4.10; N, 16.20%. NMR (DMSO-d6, 400 MHz): 2.06 (s, 3H, −CH3), 5.71 (s,
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- 1H, C2−H oxadiazole ring), 6.82−7.99 (m, 19H, Ar−H and
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- C5−H pyrazole), 12.34 (s, 1H, −CONH); 13C NMR (DMSO-
ethylidene)-3-methylbenzohydrazide (4f). Yield 73%, mp d6, 100 MHz): 12.4, 78.1, 117.4, 119.7 (2), 120.1, 123.1, 125.4
245−247 °C; IR (KBr) λmax: 3344 (N−H stretching, (2), 125.7, 126.7, 127.4, 128.1 (2), 128.4 (2), 129.1 (4), 130.7,
secondary amide), 3066, 3002 (C−H stretching, aromatic 131.5, 132.4, 132.7, 134.7, 134.8, 135.7, 139.4, 147.1, 149.2,
ring), 2930 (C−H stretching, −CH3 group), 1619 (CO 157.5, 163.4; LCMS: m/z = 594.1 (M+); anal. calcd for
stretching), 1503, 1412 (CC, CN stretching, aromatic C32H24Cl2N6O2: C, 64.54; H, 4.06; N, 14.11; found: C, 64.70;
ring), 1214 (C−O−C stretching, oxadiazole ring), 705 (C−Cl H, 4.10; N, 14.20%.
stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.11 (s, Characterization of 4-Chloro-N′-(1-(2-(3-(4-chlorophen-
3H, −CH3), 2.34 (s, 3H, −CH3), 5.61 (s, 1H, C2−H yl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-
oxadiazole ring), 6.82−8.02 (m, 19H, Ar−H and C5−H 3(2H)-yl)ethylidene)benzohydrazide (4j). Yield 74%, mp
pyrazole), 12.31 (s, 1H, −CONH); 13C NMR (DMSO-d6, 100 191−193 °C; IR (KBr) λmax: 3336 (N−H stretching,
MHz): 12.1, 21.7, 78.1, 117.5, 119.5 (2), 123.7, 124.7, 125.0, secondary amide), 3014, 3001 (C−H stretching, aromatic
125.1 (2), 126.7, 128.1 (2), 128.4 (2), 129.4 (4), 131.5, 131.8, ring), 1606 (CO stretching), 1534, 1429 (CC, CN
131.9, 132.2, 132.4, 134.4, 134.7, 138.3, 139.2, 147.3, 149.4, stretching, aromatic ring), 1212 (C−O−C stretching,
157.9, 163.5; LCMS: m/z = 575.7 (M+); anal. calcd for oxadiazole ring), 770, 754 (C−Cl stretching) cm−1; 1H
C33H27ClN6O2: C, 68.92; H, 4.73; N, 14.61; found: C, 68.96; NMR (DMSO-d6, 400 MHz): 2.06 (s, 3H, −CH3), 5.73 (s,
H, 4.80; N, 14.69%. 1H, C2−H oxadiazole ring), 6.82−7.96 (m, 19H, Ar−H and
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- C5−H pyrazole), 12.33 (s, 1H, −CONH); 13C NMR (DMSO-
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- d6, 100 MHz): 12.0, 78.3, 117.3, 119.2 (2), 120.3, 123.7, 125.4
ethylidene)-4-methylbenzohydrazide (4g). Yield 71%, mp (2), 126.1, 128.0 (2), 128.1 (4), 129.6 (4), 130.3 (2), 130.8,
233−236 °C; IR (KBr) λmax: 3312 (N−H stretching, 131.7, 132.7, 134.4, 137.1, 139.2, 147.1, 149.3, 157.3, 163.9;
secondary amide), 3016, 3020 (C−H stretching, aromatic LCMS: m/z = 595.2 (M+); anal. calcd for C32H24Cl2N6O2: C,
ring), 2924 (C−H stretching, −CH3 group), 1600 (CO 64.54; H, 4.06; N, 14.11; found: C, 64.60; H, 4.10; N, 14.15%.
28280 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- 2.03 (s, 3H, −CH3), 2.19 (s, 3H, CH3), 4.73 (s, 2H, −OCH2),
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- 5.71 (s, 1H, C2−H oxadiazole ring), 6.78−7.93 (m, 19H, Ar−
ethylidene)-2-phenylacetohydrazide (4k). Yield 57%, mp H and C5−H pyrazole), 12.39 (s, 1H, −CONH); 13C NMR
244−246 °C; IR (KBr) λmax: 3333 (N−H stretching, (DMSO-d6, 100 MHz): 12.1, 21.1, 69.3, 78.2, 111.6 (2), 113.4,
secondary amide), 3014, 3020 (C−H stretching, aromatic 117.3, 119.4 (2), 121.6, 123.5, 125.2 (2), 126.8, 128.3 (2),
ring), 2855 (C−H stretching, −CH2−), 1595 (CO 128.7 (2), 129.2, 129.4 (4), 131.5, 132.2, 134.7, 139.4, 139.5,
stretching), 1554, 1428 (CC, CN stretching, aromatic 147.2, 149.3, 157.2, 160.1, 171.1; LCMS: m/z = 605.4 (M+);
ring), 1218 (C−O−C stretching, oxadiazole ring), 758 (C−Cl anal. calcd for C34H29ClN6O3: C, 67.49; H, 4.57; N, 15.86;
stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): 2.00 (s, found: C, 67.58; H, 4.63; N, 15.96%.
3H, −CH3), 3.81 (s, 2H, −CH2), 5.70 (s, 1H, C2−H Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
oxadiazole ring), 7.26−7.62 (m, 20H, Ar−H and C5−H nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
pyrazole), 12.36 (s, 1H, −CONH); 13C NMR (DMSO-d6, 100 ethylidene)-2-(p-tolyloxy)acetohydrazide (4o). Yield 70%,
MHz): 12.9, 16.5, 42.7, 79.2, 117.3, 119.7 (2), 123.8, 125.7 mp 239−241 °C; IR (KBr) λmax: 3014, 3045, 3035 (C−H
(2), 126.7, 127.4, 128.2 (2), 128.4 (2), 129.1 (2), 129.3 (4), stretching, aromatic ring), 2920 (C−H stretching, −CH3
129.6 (2), 130.7, 131.7, 134.7, 135.7, 139.8, 147.0, 149.4, group), 2813 (C−H stretching, −CH2−), 1697, 1647 (C
150.4, 171.9; LCMS: m/z = 575.1 (M+); anal. calcd for O stretching), 1530, 1416 (CC, CN stretching, aromatic
C33H27ClN6O2: C, 68.52; H, 4.73; N, 14.61; found: C, 68.60; ring), 1336, 1216 (C−O−C stretching, oxadiazole ring), 763
H, 4.75; N, 14.70%. (C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz):
Characterization of N-(2-(2-(1-(2-(3-(4-Chlorophenyl)-1- 2.09 (s, 3H, −CH3), 2.20 (s, 3H, CH3), 4.79 (s, 2H, −OCH2),
phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- 5.75 (s, 1H, C2−H oxadiazole ring), 6.87−7.95 (m, 19H, Ar−
ethylidene)hydrazinyl)-2-oxoethyl)benzamide (4l). Yield H and C5−H pyrazole),.42 (s, 1H, −CONH); 13C NMR
60%, mp 275−278 °C; IR (KBr) λmax: 3012, 3020, 3030 (DMSO-d6, 100 MHz): 12.8, 21.7, 69.6, 78.1, 114.3 (2), 117.5,
(C−H stretching, aromatic ring), 2867 (C−H stretching, 119.1 (2), 123.8, 125.9 (2), 126.3, 128.6 (2), 128.8 (2), 129.4
−CH2−), 1700, 1632 (CO stretching), 1550, 1430 (CC, (4), 130.5 (2), 130.8, 131.4, 131.8, 132.5, 134.7, 139.4, 147.3,
CN stretching, aromatic ring), 1210 (C−O−C stretching, 149.3, 155.3, 157.3, 171.3; LCMS: m/z = 605.4 (M+); anal.
oxadiazole ring), 715 (C−Cl stretching) cm−1; 1H NMR calcd for C34H29ClN6O3: C, 67.49; H, 4.83; N, 13.89; found:
(DMSO-d6, 400 MHz): 2.02 (s, 3H, −CH3), 2.23 (s, 2H, C, 67.53; H, 4.90; N, 13.96%.
−CH2), 5.69 (s, 1H, C2−H oxadiazole ring), 6.99−7.99 (m, Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
19H, Ar−H and C5−H pyrazole), 7.51 (s, 1H, −NHCH2), nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
7.56 (s, 1H, NH), 12.33 (s, 1H, −CONH); 13C NMR ethylidene)-2-(2-nitrophenoxy)acetohydrazide (4p). Yield
(DMSO-d6, 100 MHz): 12.4, 45.7, 78.1, 117.5, 119.2 (2), 61%, mp 272−275 °C; IR (KBr) λmax: 3036, 3012, 3060
123.7, 125.7 (2), 126.3, 127.4 (2), 128.5 (4), 128.7 (2), 129.4 (C−H stretching, aromatic ring), 2860 (C−H stretching,
(4), 131.5, 131.7, 132.4, 132.7, 134.7, 134.8, 139.4, 147.4, −CH2−), 1612, 1623 (CO stretching), 1533 (NO
149.1, 157.4, 167.2, 171.2; LCMS: m/z = 618.2 (M+); anal. stretching, Ar−NO2), 1545, 1420 (CC, CN stretching,
calcd for C34H28ClN7O3: C, 66.07; H, 4.57; N, 15.86; found: aromatic ring), 1220 (C−O−C stretching, oxadiazole ring),
C, 66.10; H, 4.75; N, 15.96%. 723 (C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- MHz): 2.12 (s, 3H, −CH3), 4.72 (s, 2H, −OCH2), 5.75 (s,
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- 1H, C2−H oxadiazole ring), 6.94−7.98 (m, 19H, Ar−H and
ethylidene)-2-(o-tolyloxy)acetohydrazide (4m). Yield 64%, C5−H pyrazole), 12.42 (s, 1H, −CONH); 13C NMR (DMSO-
mp 226−228 °C; IR (KBr) λmax: 3009, 3035, 3025 (C−H d6, 100 MHz): 12.4, 68.1, 78.0, 115.0, 117.6, 119.4 (2), 121.9,
stretching, aromatic ring), 2930 (C−H stretching, −CH3 123.4, 125.9 (2), 126.7, 127.1, 128.3 (2), 128.7 (2), 129.4 (4),
group), 2860 (C−H stretching, −CH2−), 1735, 1635 (C 131.2, 131.4, 131.9, 132.2, 134.0, 139.5, 140.7, 147.1, 149.3,
O stretching), 1555, 1435 (CC, CN stretching, aromatic 152.4, 157.2, 171.6; LCMS: m/z = 636.2 (M+); anal. calcd for
ring), 1325, 1200 (C−O−C stretching, oxadiazole ring), 720 C33H26ClN7O5: C, 62.31; H, 4.12; N, 15.41; found: C, 62.36;
(C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): H, 4.05; N, 15.60%.
2.01 (s, 3H, −CH3), 2.16 (s, 3H, CH3), 4.66 (s, 2H, −OCH2), Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe-
5.69 (s, 1H, C2−H oxadiazole ring), 6.84−7.99 (m, 19H, Ar− nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)-
H and C5−H pyrazole), 12.33 (s, 1H, −CONH); 13C NMR ethylidene)-2-(3-nitrophenoxy)acetohydrazide (4q). Yield
(DMSO-d6, 100 MHz): 12.0, 15.2, 69.4, 78.1, 112.4, 117.3, 55%, mp 183−185 °C; IR (KBr) λmax: 3066, 3085, 3012
119.4 (2), 120.7, 123.1, 125.1 (2), 126.2, 126.4, 126.6, 128.3 (C−H stretching, aromatic ring), 2865 (C−H stretching,
(2), 128.4 (2), 129.3 (4), 131.4, 131.6, 131.7, 132.4, 134.0, −CH2−), 1620, 1612 (CO stretching), 1520 (NO
139.1, 147.2, 149.5, 157.1, 158.4, 171.3; LCMS: m/z = 605.3 stretching, Ar−NO2), 1515, 1425 (CC, CN stretching,
(M+); anal. calcd for C34H29ClN6O3: C, 67.49; H, 4.57; N, aromatic ring), 1225 (C−O−C stretching, oxadiazole ring),
15.86; found: C, 67.56; H, 4.63; N, 15.90%. 712 (C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400
Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- MHz): 2.18 (s, 3H, −CH3), 4.65 (s, 2H, −OCH2), 5.70 (s,
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- 1H, C2−H oxadiazole ring), 7.38−7.99 (m, 19H, Ar−H and
ethylidene)-2-(m-tolyloxy)acetohydrazide (4n). Yield 63%, C5−H pyrazole), 12.44 (s, 1H, −CONH); 13C NMR (DMSO-
mp 212−215 °C; IR (KBr) λmax: 3012, 3040, 3030 (C−H d6, 100 MHz): 12.3, 69.2, 78.1, 111.0, 116.3, 117.1, 119.8 (2),
stretching, aromatic ring), 2925 (C−H stretching, −CH3 120.1, 123.8, 125.6 (2), 126.3, 128.3 (2), 128.8 (2), 129.6 (4),
group), 2800 (C−H stretching, −CH2−), 1695, 1640 (C 130.7, 131.4, 131.9, 132.4, 134.6, 139.1, 147.1, 149.3, 151.1,
O stretching), 1528, 1418 (CC, CN stretching, aromatic 157.9, 161.9, 171.3; LCMS: m/z = 635.6 (M+); anal. calcd for
ring), 1302, 1215 (C−O−C stretching, oxadiazole ring), 730 C33H26ClN7O5: C, 62.31; H, 4.12; N, 15.41; found: C, 62.36;
(C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400 MHz): H, 4.20; N, 15.50%.
28281 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Characterization of N′-(1-(2-(3-(4-Chlorophenyl)-1-phe- suitable for use in molecular docking studies, first, the
nyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-oxadiazol-3(2H)-yl)- optimization and minimization of the enzyme-inhibitor
ethylidene)-2-(4-nitrophenoxy)acetohydrazide (4r). Yield complex were performed using the protein preparation wizard
66%, mp 207−210 °C; IR (KBr) λmax: 3061, 3080, 3014 tool applying the OPLS-2005 force field. This involved
(C−H stretching, aromatic ring), 2800 (C−H stretching, eliminating the crystallographically observed water molecules
−CH2−), 1610, 1623 (CO stretching), 1525 (NO (water without H bonds) since no water molecule was found
stretching, Ar−NO2), 1517, 1410 (CC, CN stretching, to be conserved in the ligand−protein interaction, adding
aromatic ring), 1220 (C−O−C stretching, oxadiazole ring), hydrogen atoms corresponding to pH 7.0 considering the
725 (C−Cl stretching) cm−1; 1H NMR (DMSO-d6, 400 appropriate ionization states for the acidic as well as basic
MHz): 2.14 (s, 3H, −CH3), 4.63 (s, 2H, −OCH2), 5.72 (s, amino acid residues assigning the correct bond orders.
1H, C2−H oxadiazole ring), 7.25−8.15 (m, 19H, Ar−H and Following the assignment of charge and protonation state,
C5−H pyrazole), 12.47 (s, 1H, −CONH); 13C NMR (DMSO- finally, energy minimization was performed using the OPLS-
d6, 100 MHz): 12.7, 69.3, 78.4, 115.0 (2), 117.9, 119.3 (2), 2005 force field until the average root-mean-square deviation
123.0, 125.1 (2), 125.6 (2), 126.7, 128.0 (2), 128.6 (2), 129.4 (RMSD) of the nonhydrogen atoms reached 0.3 Å to relieve
(4), 131.0, 131.5, 132.7, 134.7, 139.5, 140.6, 147.8, 149.7, the steric clashes among the residues due to the addition of
157.6, 164.6, 171.9; LCMS: m/z = 636.1 (M+); anal. calcd for hydrogen atoms.
C33H26ClN7O5: C, 62.41; H, 4.12; N, 15.41; found: C, 62.50; On the other hand, the initial 3D structures of the
H, 4.23; N, 15.50%. oxadiazole derivatives were sketched using the build panel in
Characterization of 2-(2-Chlorophenoxy)-N′-(1-(2-(3-(4- Maestro and subsequently optimized through the LigPrep
chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-ox- module that performs addition of hydrogen atoms; adjustment
adiazol-3(2H)-yl)ethylidene)acetohydrazide (4s). Yield 73%, of realistic bond lengths as well as angles; and correction of
mp 269−272 °C; IR (KBr) λmax: 3061, 3080, 3014 (C−H chiralities, ionization states, tautomers, stereochemistries, and
stretching, aromatic ring), 2800 (C−H stretching, −CH2−), ring conformations. Partial atomic charges were ascribed using
1610, 1623 (CO stretching), 1525, 1410 (CC, CN the OPLS-2005 force field, and possible ionization states were
stretching, aromatic ring), 1220 (C−O−C stretching, generated for each ligand at a pH of 7.0. Finally, the geometries
oxadiazole ring), 740, 725 (C−Cl stretching) cm−1; 1H of these compounds were optimized by energy minimization
NMR (DMSO-d6, 400 MHz): 2.18 (s, 3H, −CH3), 4.69 (s, until a gradient of 0.01 kcal/mol/Å was reached.
2H, −OCH2), 5.71 (s, 1H, C2−H oxadiazole ring), 6.95−7.98 After ensuring that the ligand and protein structures were in
(m, 19H, Ar−H and C5−H pyrazole), 12.52 (s, 1H, the correct form, the shape and properties of the binding site of
−CONH); 13C NMR (DMSO-d6, 100 MHz): 12.0, 68.9, the InhA enzyme were defined and set up for docking using the
78.0, 112.7, 117.3, 119.2 (2), 122.8, 122.9, 123.0, 125.3 (2), receptor grid generation panel. Since the inhibitor’s coordinates
126.7, 127.3, 128.6 (2), 128.8 (2), 129.5 (4), 131.0, 131.8, in the complex are known, the active site was defined by a 10 ×
132.0, 132.7, 134.9, 139.3, 147.9, 149.4, 154.3, 157.6, 171.6; 10 × 10 Å box centered on the native ligand in the crystal
LCMS: m/z = 625.3 (M+); anal. calcd for C33H26Cl2N6O3: C, complex. The receptor grid generation program uses two cubical
63.37; H, 4.19; N, 13.44; found: C, 63.40; H, 4.20; N, 13.55%. boxes with a common centroid for calculations: a larger
Characterization of 2-(4-Chlorophenoxy)-N′-(1-(2-(3-(4- enclosing and a smaller binding box. With the non-covalently
chlorophenyl)-1-phenyl-1H-pyrazol-4-yl)-5-phenyl-1,3,4-ox- bound cocrystallized ligand, the grid file was created using this
adiazol-3(2H)-yl)ethylidene)acetohydrazide (4t). Yield 69%, native ligand at the center of the two boxes which were kept
mp 280−282 °C; IR (KBr) λmax: 3051, 3088, 3012 (C−H sufficiently large in order to allow the ligands to explore a large
stretching, aromatic ring), 2850 (C−H stretching, −CH2−), portion of the enzyme structure in search of the best binding
1612, 1623 (CO stretching), 1512, 1413 (CC, CN conformation.
stretching, aromatic ring), 1224 (C−O−C stretching, These optimized protein and ligand geometries were used as
oxadiazole ring), 750, 725 (C−Cl stretching) cm−1; 1H input for the binding affinity study. Glide is an interactive
NMR (DMSO-d6, 400 MHz): 2.23 (s, 3H, −CH3), 4.72 (s, molecular graphics program for studying ligand−receptor
2H, −OCH2), 5.70 (s, 1H, C2−H oxadiazole ring), 6.90−7.98 interactions and identifying the probable binding site for the
(m, 19H, Ar−H and C5−H pyrazole), 12.58 (s, 1H, biomolecules. The final scoring and ranking of the docked
−CONH); 13C NMR (DMSO-d6, 100 MHz): 12.8, 69.5, poses were carried out using Schrödinger’s proprietary extra
78.4, 117.0, 117.6 (2), 119.7 (2), 123.0, 125.8 (2), 126.0, precision (i.e., with GlideXP) scoring function. The scoring
126.9, 128.7 (2), 128.9 (2), 129.3 (4), 130.7 (2), 131.7, 131.9, function is equipped with force-field-based parameters that
132.7, 134.9, 139.6, 147.8, 149.0, 156.9, 157.6, 171.4; LCMS: account for contributions from steric and electrostatic
m/z = 625.4 (M+); anal. calcd for C33H26Cl2N6O3: C, 63.37; interaction energies, solvation and repulsive interactions,
H, 4.19; N, 13.44; found: C, 63.50; H, 4.30; N, 13.55%. hydrophobic interactions, hydrogen bonding, and metal−
Methodology for Molecular Docking. The molecular ligand interactions, all incorporated in the empirical energy
docking study was performed with the Glide (Grid-Based functions. Maestro’s Pose Viewer utility was used to visualize
Ligand Docking with Energetics)35−37 module incorporated in and analyze the output file generated in terms of the docking
the Schrödinger molecular modeling software (Schrödinger, poses of the ligands for the key elements of interaction with the
Inc., USA, 2016) to gain an insight into the binding modes of enzyme.


the oxadiazole derivatives (4a−t) into the active site of the
mycobacterial enoyl reductase (InhA) enzyme. With this ASSOCIATED CONTENT
purpose, the crystal structure of the mycobacterial enoyl
reductase (InhA) complexed with its inhibitor was retrieved *
sı Supporting Information

from the protein data bank (PDB) (PDB code: 4TZK).38 The Supporting Information is available free of charge at
Since the protein structure file obtained from the PDB was not https://pubs.acs.org/doi/10.1021/acsomega.1c04411.
28282 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

Additional figures illustrating the characterization of Evaluation of New Pyrazole Derivatives as Cell Cycle Inhibitors.
synthetics by 1H NMR and 13C NMR, mass, and IR Arab. J. Chem. 2019, 12, 816−824.
(5) Mehranpour, A. M.; Hashemnia, S.; Abbasi, S. Synthesis of New
spectra (PDF) Pyrazole Derivatives Using Vinamidinium Salts. Org. Chem. Res. 2017,


3, 187−190.
AUTHOR INFORMATION (6) Naim, M. J.; Alam, O.; Nawaz, F.; Alam, M. J.; Alam, P. Current
Status of Pyrazole and its Biological Activities. J. Pharm. Bioallied. Sci.
Corresponding Author 2016, 8, 2−17.
Nisheeth C. Desai − Division of Medicinal Chemistry, (7) Prabhudeva, M. G.; Vivek, H. K.; Kumar, K. A. Synthesis of
Department of Chemistry, Mahatma Gandhi Campus, Novel Pyrazole Carboxamides using Reusable Catalyst as Antimicro-
Maharaja Krishnakumarsinhji Bhavnagar University, bial Agents and Molecular Docking Studies. Chem. Data Collect. 2019,
Bhavnagar, Gujarat 364002, India; orcid.org/0000- 20, 100193.
0001-6864-8661; Email: dnisheeth@rediffmail.com (8) Takate, S. J.; Shinde, A. D.; Karale, B. K.; Akolkar, H.; Nawale,
L.; Sarkar, D.; Mhaske, P. C. Thiazolyl-Pyrazole Derivatives as
Authors Potential Antimycobacterial Agents. Bioorg. Med. Chem. Lett. 2019,
Kandarp Bhatt − Division of Medicinal Chemistry, 29, 1199−1202.
(9) Gedawy, E. M.; Kassab, A. E.; El Kerdawy, A. M. Design,
Department of Chemistry, Mahatma Gandhi Campus,
Synthesis and Biological Evaluation of Novel Pyrazole Sulfonamide
Maharaja Krishnakumarsinhji Bhavnagar University, Derivatives as Dual COX-2/5-LOX Inhibitors. Eur. J. Med. Chem.
Bhavnagar, Gujarat 364002, India 2020, 189, 112066.
Jahnvi Monapara − Division of Medicinal Chemistry, (10) Yamali, C.; Gul, H. I.; Ece, A.; Taslimi, P.; Gulcin, I. Synthesis,
Department of Chemistry, Mahatma Gandhi Campus, Molecular Modeling, and Biological Evaluation of 4-[5-aryl-3-
Maharaja Krishnakumarsinhji Bhavnagar University, (thiophen-2-yl)-4, 5-dihydro-1h-pyrazol-1-yl] benzenesulfonamides
Bhavnagar, Gujarat 364002, India Toward Acetylcholinesterase, Carbonic Anhydrase I and II Enzymes.
Unnat Pandit − Special Centre for Systems Medicine, Chem. Biol. Drug Des. 2017, 91, 854−866.
Jawaharlal Nehru University, New Delhi 110067, India; (11) Abadi, A. H.; Eissa, A. A. H.; Hassan, G. S. Synthesis of Novel
orcid.org/0000-0001-5526-9814 1, 3, 4-Trisubstituted Pyrazole Derivatives and Their Evaluation as
Vijay M. Khedkar − Department of Pharmaceutical Antitumor and Antiangiogenic Agents. Chem. Pharm. Bull. 2003, 51,
838−844.
Chemistry, School of Pharmacy, Vishwakarma University,
(12) Ohno, R.; Watanabe, A.; Matsukawa, T.; Ueda, T.; Sakurai, H.;
Pune, Maharashtra 411048, India Hori, M.; Hirai, K. Synthesis and Herbicidal Activity of New Pyrazole-
Complete contact information is available at: 4-Carboxamide Derivatives. J. Pestic. Sci. 2004, 29, 15−26.
https://pubs.acs.org/10.1021/acsomega.1c04411 (13) Meng, F.-J.; Sun, T.; Dong, W.-Z.; Li, M.-H.; Tuo, Z.-Z.
Discovery of Novel Pyrazole Derivatives as Potent Neuraminidase
Notes Inhibitors against Influenza H1N1 Virus. Arch. Pharm. 2016, 349,
168−174.
The authors declare no competing financial interest.


(14) Zhao, P. L.; Wang, L.; Zhu, X. L.; Huang, X.; Zhan, C. G.; Wu,
J. W.; Yang, G. F. Subnanomolar Inhibitor of Cytochrome Bc1
ACKNOWLEDGMENTS Complex Designed by Optimizing Interaction with Conformationally
Prof. Nisheeth C. Desai is thankful to the University Grant Flexible Residues. J. Am. Chem. Soc. 2010, 132, 185−194.
Commission, New Delhi, for the financial support under BSR (15) Shaikh, A.; Meshram, J. Novel 1,3,4-Oxadiazole Derivatives of
Dihydropyrimidinones: Synthesis, Anti-Inflammatory, Anthelmintic,
Faculty-Fellowship 2019 (F18-1/2011 (BSR)). Jahnvi D.
and Antibacterial Activity Evaluation. J. Heterocycl. Chem. 2016, 53,
Monapara is grateful to the Department of Science and 1176−1182.
Technology, INSPIRE PROGRAM, for the award of INSPIRE (16) Modi, V.; Modi, P. Oxadiazole: Synthesis, Characterization and
Fellowship (IF180817). The authors are thankful to the Biological Activities. J. Saudi Chem. Soc. 2012, 16, 327−332.
National Repository of Small Molecules (NRM) facility, CSIR- (17) Bhat, K.; Sufeera, K.; Chaitanya, S. Synthesis, Characterization
National Chemical Laboratory (CSIR-NCL), Pune, for and Biological Activity Studies of 1,3,4-Oxadiazole Analogs. J. Young
performing the antitubercular activity. The authors are also Pharm. 2011, 3, 310−314.
thankful to Schrödinger Inc. for providing the demo license of (18) Banerjee, A. G.; Das, N.; Shengule, S. A.; Srivastava, R. S.;
Schrödinger Suite that has tremendously helped in the Shrivastava, S. K. Synthesis, Characterization, Evaluation and
computational study. The authors are also thankful to Priyanka Molecular Dynamics Studies of 5, 6-Diphenyl-1,2,4-triazin-3(2H)-
Desai, founder of iScribblers, for the linguistic editing of the one Derivatives Bearing 5-Substituted 1,3,4-Oxadiazole as Potential
manuscript. Anti-Inflammatory and Analgesic Agents. Eur. J. Med. Chem. 2015,


101, 81−95.
(19) Sreenivasulu, R.; Tej, M. B.; Jadav, S. S.; Sujitha, P.; Kumar, C.
REFERENCES G.; Raju, R. R. Synthesis, Anticancer Evaluation and Molecular
(1) World Health Organization. Tuberculosis Fact Sheet. https:// Docking Studies of 2,5-Bis(indolyl)-1,3,4-oxadiazoles. J. Mol. Struct.
www.who.int/news-room/fact-sheets/detail/tuberculosis. Accessed 13 2020, 1208, 127875.
April 2020. (20) Rathore, A.; Sudhakar, R.; Ahsan, M. J.; Ali, A.; Subbarao, N.;
(2) Rao, H. S. P.; Adigopula, L. N.; Ramadas, K. One-Pot Synthesis Jadav, S. S.; Umar, S.; Yar, M. S. In vivo Anti-Inflammatory Activity
of Densely Substituted Pyrazolo[3,4-b]-4,7-dihydropyridines. ACS and Docking Study of Newly Synthesized Benzimidazole Derivatives
Comb. Sci. 2017, 19, 279−285. Bearing Oxadiazole and Morpholine Rings. Bioorg. Chem. 2017, 70,
(3) El-Borai, M. A.; Rizk, H. F.; Sadek, M. R.; El-Keiy, M. M. An 107−117.
Eco-Friendly Synthesis of Heterocyclic Moieties Condensed with (21) Shaykoon, M. S.; Marzouk, A. A.; Soltan, O. M.; Wanas, A. S.;
Pyrazole System under Green Conditions and Their Biological Radwan, M. M.; Gouda, A. M.; Youssif, B. G. M.; Abdel-Aziz, M.
Activity. Green Sustainable Chem. 2016, 06, 88−100. Design, Synthesis and Antitrypanosomal Activity of Heteroaryl-Based
(4) Nitulescu, G. M.; Matei, L.; Aldea, I. M.; Draghici, C.; Olaru, O. 1,2,4-Triazole and 1,3,4-Oxadiazole Derivatives. Bioorg. Chem. 2020,
T.; Bleotu, C. Ultrasound-Assisted Synthesis and Anticancer 100, 103933.

28283 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284
ACS Omega http://pubs.acs.org/journal/acsodf Article

(22) Ambhore, A. N.; Kamble, S. S.; Kadam, S. N.; Kamble, R. D.; Substituted Dihydrothiophene Ureidoformamides at Room Temper-
Hebade, M. J.; Hese, S. V.; Gaikwad, M. V.; Meshram, R. J.; Gacche, ature Using Diisopropyl Ethyl Ammonium Acetate: A Green
R. N.; Dawane, B. S. Design, Synthesis and in silico Study of Pyridine Perspective. ACS Omega 2020, 5, 29055−29067.
Based 1,3,4-Oxadiazole Embedded Hydrazinecarbothioamide Deriv- (38) http://www.rcsb.org/pdb/. (Accessed on August 1st, 2021)
atives as Potent Anti-Tubercular Agent. Comput. Biol. Chem. 2019, 80,
54−65.
(23) Desai, N. C.; Trivedi, A.; Somani, H.; Jadeja, K. A.; Vaja, D.;
Nawale, L.; Khedkar, V. M.; Sarkar, D. Synthesis, Biological
Evaluation, and Molecular Docking Study of Pyridine Clubbed
1,3,4-Oxadiazoles as Potential Antituberculars. Synth. Comm. 2018,
48, 524−540.
(24) Khan, A.; Sarkar, D. A Simple Whole Cell Based High
Throughput Screening Protocol Using Mycobacterium Bovis BCG for
Inhibitors Against Dormant and Active Tubercle bacilli. J. Microbiol.
Methods 2008, 73, 62−68.
(25) Chen, Q.; Zhu, X. L.; Jiang, L. L.; Liu, Z. M.; Yang, G. F.
Synthesis, Antifungal Activity and CoMFA Analysis of Novel 1,2,4-
Triazolo[1,5-a]Pyrimidine Derivatives. Eur. J. Med. Chem. 2008, 43,
595−603.
(26) Zhang, M. Z.; Mulholland, N.; Beattie, D.; Irwin, D.; Gu, Y. C.;
Chen, Q.; Yang, G. F.; Clough, J. Synthesis and Antifungal Activity of
3-(1,3,4-Oxadiazol-5-Yl)-Indoles and 3-(1,3,4-Oxadiazol-5-Yl)Methyl-
Indoles. Eur. J. Med. Chem. 2013, 63, 22−32.
(27) Pathak, G.; Das, D.; Rokhum, S. L. A Microwave-Assisted
Highly Practical Chemoselective Esterification and Amidation of
Carboxylic Acids. RSC Adv. 2016, 6, 93729−93740.
(28) Desai, N. C.; Somani, H.; Trivedi, A.; Bhatt, K.; Nawale, L.;
Khedkar, V. M.; Jha, P. C.; Sarkar, D. Synthesis, Biological Evaluation
and Molecular Docking Study of Some Novel Indole and Pyridine
Based 1, 3, 4-Oxadiazole Derivatives as Potential Antitubercular
Agents. Bioorg. Med. Chem. Lett. 2016, 26, 1776−1783.
(29) Leung, A. K.; White, E. L.; Ross, L. J.; Reynolds, R. C.; DeVito,
J. A.; Borhani, D. W. Structure of Mycobacterium tuberculosis FtsZ
Reveals Unexpected, G Protein-Like Conformational Switches. J. Mol.
Biol. 2004, 342, 953−970.
(30) Asgaonkar, K. D.; Mote, G. D.; Chitre, T. S. QSAR and
Molecular Docking Studies of Oxadiazole-Ligated Pyrrole Derivatives
as Enoyl-ACP (CoA) Reductase Inhibitors. Sci. Pharm. 2014, 82, 71−
85.
(31) Kini, S. G.; Bhat, A. R.; Bryant, B.; Williamson, J. S.; Dayan, F.
E. Synthesis, Antitubercular Activity and Docking Study of Novel
Cyclic Azole Substituted Diphenyl Ether Derivatives. Eur. J. Med.
Chem. 2009, 44, 492−500.
(32) Mathew, B.; Suresh, J.; Mathew, G. E.; Sonia, G.; Krishnan, G.
K. Design, Synthesis, Toxicity Estimation and Molecular Docking
Studies of N-(furan-2-yl)-1-(5-substituted) phenyl-1,3,4-oxadiazol-2-
yl methanimine as Antitubercular Agents. Indian J. Pharm. Sci. 2014,
76, 401−406.
(33) Vilchèze, C.; Morbidoni, H. R.; Weisbrod, T. R.; Iwamoto, H.;
Kuo, M.; Sacchettini, J. C.; Jacobs, W. R. Inactivation of the inhA-
Encoded Fatty Acid Synthase II (FASII) Enoyl-Acyl Carrier Protein
Reductase Induces Accumulation of the FASI End Products and Cell
Lysis of Mycobacterium smegmatis. J. Bacteriol. 2000, 182, 4059−4067.
(34) Desai, N. C.; Kotadiya, G. M. Microwave-Assisted Synthesis of
Benzimidazole bearing 1, 3, 4-Oxadiazole Derivatives: Screening for
Their in vitro Antimicrobial Activity. Med. Chem. Res. 2014, 23, 4021−
4033.
(35) Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.;
Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.;
Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A New
Approach for Rapid, Accurate Docking and Scoring. 1. Method and
Assessment of Docking Accuracy. J. Med. Chem. 2004, 47, 1739−
1749.
(36) Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.;
Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: A New Approach for
Rapid, Accurate Docking and Scoring. 2. Enrichment Factors in
Database Screening. J. Med. Chem. 2004, 47, 1750−1759.
(37) Jadhav, C. K.; Nipate, A. S.; Chate, A. V.; Dofe, V. S.;
Sangshetti, J. N.; Khedkar, V. M.; Gill, C. H. Rapid Construction of

28284 https://doi.org/10.1021/acsomega.1c04411
ACS Omega 2021, 6, 28270−28284

You might also like