Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Journal of Molecular Structure 1246 (2021) 131183

Contents lists available at ScienceDirect

Journal of Molecular Structure


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

Design, one-pot synthesis, molecular docking study, and antibacterial


evaluation of novel 2H-chromene based imidazo[1,2-a]pyridine
derivatives as potent peptide deformylase inhibitors
Nilima Priyadarsini Mishra a, Seetaram Mohapatra a,∗, Chita Ranjan Sahoo b,
Bishnu Prasad Raiguru a, Sabita Nayak a, Subhrakant Jena c, Rabindra Nath Padhy b,∗
a
Organic Synthesis Laboratory, Department of Chemistry, Ravenshaw University, Cuttack, Odisha 753003, India
b
Central Research Laboratory, IMS & Sum Hospital, Siksha ‘O’ Anusandhan University, Bhubaneswar, Odisha 52003, India
c
National Institute of Science Education and Research, Bhubaneswar, Odisha 752 050, India

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

Article history: An efficient, environmentally friendly, one-pot three-component synthesis of a series of 2H-chromene-
Received 27 February 2021 based imidazo[1,2-a]pyridines had been designed and were synthesized. This protocol was developed by
Revised 19 July 2021
the reaction of substituted 2H-chromene aldehydes and 2-aminopyridine in a mixture of nitroalkane and
Accepted 24 July 2021
DMF under microwave irradiation at 60W, 100°C in 15 min with the presence of FeCl3 as the catalyst. The
Available online 28 July 2021
products were obtained in excellent yields with high functional group tolerance. All these compounds had
Keywords: been investigated further in vitro for evaluation of antibacterial potency by agar-well diffusion method
2H-chromene against human pathogenic Gram-positive and Gram-negative bacteria, with the determination of min-
Imidazo[1,2-α ]pyridines imum inhibitory concentration (MIC) values. Indeed, compound 13i strongly inhibited peptide deformy-
Antibacterial lase (MIC = 16 μg/ml) in the Gram-negative Escherichia coli, in silico. From structure-activity relationships
Peptide deformylase inhibitor based on the biological and molecular docking results and the potent antibacterial activities, it could be
Molecular docking study
stated that the selected synthetic compounds could be used as potent drugable antibacterial agents.
© 2021 Elsevier B.V. All rights reserved.

1. Introduction Moreover, any viral infection drastically degrades the immunity-


strength of a body, resulting in the consequent opportunistic bac-
People of all age groups get decimated each year in each geo- terial infections from the environment or the dormant resident
graphic zone from bacteria as balanced parasites, e.g., suppurative pathogenic bacteria of the body, as in the COVID-19 pandemic
infections from the Gram-positive bacterium Staphylococcus aureus pneumonia-causing Klebsiella pneumoniae devastating human, for
[1] and staggering tuberculosis and leprosy bacilli [2]; or from instance [6]. The most unfortunate situational tragedy in the hu-
destructive parasites, namely, diarrhea/ dysentery from enterococci man body is that one virulent bacterium causing an infection in-
and Vibrio cholera with instantly triggering of a life-threatening sit- fects several innards sometimes with ‘toxic shock syndrome’ tak-
uation at a blistering pace, despite the use of a plethora of emu- ing the total grip of the body till the body is decimated. Thus,
lating drugs for common and uncommon infectious diseases [3]. whether for a patient complaining of an infectious urinary/ respira-
Moreover, mortality of children under the age of 5 in low-income tory/wound ailment or if a surgeon after a surgical episode prefers
and middle-income countries in South-East Asia, tribal and rural to prescribe an antibiotic of a higher generation pre-emptively
India, sub-Saharan Africa, Latin America has been always a major (called as empiric therapy in ‘antimicrobial stewardship program’)
concern of UNICEF and WHO respiratory/chest/urinary tract infec- to be sure that the infection terminates forthwith [7]. The addi-
tions are the most commonplace infections causing morbidity and tion of newer antibacterials and antibiotics have negative conse-
premature mortality [4,5]. Adults often fall into the vicious cycle, quences in that, a fraction of bacteria can develop resistance to
“Poverty begets illness, in turn, illness begets poverty”, in several the administered drug and escape to the human environment [8].
developing countries. Intrinsically, drug resistance characters are exchanged among total
bacterial flora in the body as well as, in water/air environments
and are spread as multidrug-resistant (MDR) bacteria, both in hos-

Corresponding authors. pital and community settings [9–13]. From the use of antibiotics
E-mail addresses: seetaram.mohapatra@gmail.com (S. Mohapatra),
of higher generations, the health domain is impacted gigantically;
rnpadhy54@gmail.com (R.N. Padhy).

https://doi.org/10.1016/j.molstruc.2021.131183
0022-2860/© 2021 Elsevier B.V. All rights reserved.
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

consequently, structural modifications of obsolete drugs and intro- the concerned parent compound. Encouraged by the remark-
duction of newer safer drugable molecules to pharmaceutics have able antibacterial properties of both the pharmacophores, some
been the present trend for control, as examples seen often and 2H-chromene and imidazo[1,2-a]pyridine derivatives as hybrid
with other chemicals of curio [14]. Progressively evolving infec- molecules and planned and were designed for study the antibac-
tions with MDR bacteria must be in control with an iron hand with terial activity against the bacteria S. aureus, Streptococcus pyogenes
some eclectic approach(s). Thus it would be appropriated to ver- and E. coli, Klebsiella oxytoca isolated from clinical samples of
ify 2H-chromene-based imidazo[1,2-a]pyridine derivatives as pep- patients in Sum hospital, as sizes of zones of inhibition seen in
tide deformylase (PDF) inhibitors since these could be potent in agar-well diffusion method in vitro, along with the determina-
the control of human pathogenic bacteria [15]. tion of minimum inhibitory concentration (MIC) values of the
Currently, bacterial PDF is a suitable target and has received at- compounds.
tention for its exploration as a ’possible novel antibiotic agent’ [16].
In bacterial cells, PDF is involved in the post-translational modifi- 2. Result and discussion
cation of emerging polypeptides, and in this way, it removes the
formyl group of N-formyl methionine of the nascent protein to af- 2.1. Chemistry
ford a mature protein for use in bacterial protein synthesis [17]. In
mammalian cells, PDF was less active in comparison to its bacterial The 2H-chromene-3-carbaldehyde derivatives 11(a–i) and 2-
counterpart, and also mammalian cytosolic protein synthesis does phenyl-2H-chromene-3-carbaldehyde derivatives 11’(a–h) had been
not produce N-formylated polypeptides [18]. On the other hand, used as the starting materials. Initially, the 2H-chromene-3-
PDF is a striking and unique target for treating resistant bacteria carbaldehydes 11(a–i) were synthesized by the reaction of sali-
as it produced the mature protein in bacterial cells [19]. cylaldehydes 9 with acrolein 10 using K2 CO3 in dioxane under
From the last decade, various types of PDF inhibitors had been a reflux condition for 2h to afford 2H-chromene-3-carbaldehyde
developed by several pharmaceutical companies and academia derivatives 11(a–i) in good to excellent yields. On the other hand,
such as peptidic and pseudopeptidic analogs [20]. Among differ- the 2-phenyl-2H-chromene-3-carbaldehydes 11’(a–h) were synthe-
ent pseudopeptidic analogs “actinonin’’, a naturally occurring an- sized by following oxa-Michael aldol reaction of salicylaldehydes 9
tibiotic was reported as the first PDF inhibitor in 1962, which had a with cinnamaldehyde 10’ in the presence of pyrrolidine in DMSO
modest antibacterial activity against both Gram-positive and Gram- at a room temperature for 12 h with a good to excellent yields
negative human pathogenic bacteria [17]. However, BB83698 and (Scheme 1) [52–54].
LBM415 were the first two PDF inhibitors, which underwent hu- After the successful synthesis of starting materials, 2H-
man clinical trials [21,22]. The non-peptidic PDF inhibitors were chromene-3-carbaldehydes 11(a–i) and 2-phenyl-2H-chromene-3-
not much explored, as those are expected to be less suscepti- carbaldehydes 11’(a–h), the synthesis of the 2H-chromene based
ble to degradation [23]. By Merck research groups, biaryl acid imidazo[1,2-a] pyridine derivatives 13(a–q) via, one-pot aza-Henry
analogs were developed and evaluated as a PDF inhibitor against reaction was explored. The individual reactions of substituted 2H-
the Gram-negative bacterium Escherichia coli PDF enzyme [24]. chromene aldehydes 11(a–i) and 11’(a–h) with 2-aminopyridine 12
A report of β - sulfonyl and β -sulfinylhydroxamic acid deriva- and nitromethane in the presence of different catalysts and sol-
tives as PDF inhibitors against E. coli and another Gram-negative vents to establish the feasibility of the strategy and to optimize
Moraxella catarrhalis focused on the synthesis of various hetero- the reaction conditions were pursued. Initially, the reaction was
cyclic compounds as PDF inhibitors had focused [25]. Further- studied in the conventional heating at 40 to 120 °C for 4 to 6 h,
more, 5-Bromo-1H-indole-3-acetohydroxamic acid was reported by but the desired product was not obtained. Later on, the same re-
another group as a PDF inhibitor [26]. The 2,5-dihydropyrrole action in the ‘microwave irradiation method’ at 40 to 120 °C in
formyl hydroxyamino derivatives were also reported as PDF in- 30–60W by varying catalyst loading and solvents was carried out.
hibitors [23]. Furthermore, benzofuran-4,5-diones was developed However, the reaction of 2H-chromene-3-carbaldehydes 11a and 2-
as non-hydroxamic acid and non-peptidomimetic PDF inhibitor amino pyridine 12 in the presence of catalyst FeCl3 (10 mol%) in
[27]. Concomitantly, the antibacterial activities of oxazolidine LBM- DMF and nitromethane binary solvent system at 40 °C and 30W in
415 analogs as PDF inhibitors were reported [28]. A promising 10 min, no product was formed. Again the reaction was tried at a
target formyl hydroxyamino derivatives as a potent PDF inhibitor slightly higher temperature i.e., 80 °C in a similar reaction condi-
against drug-resistant bacteria were also developed [29]. Con- tion which provided slightly better yield (45%) of the product [55].
sequently, biphenyl tetrazole-thiazolidinediones were regarded as We studied the reaction in different solvents, the CH3 CN, toluene,
novel bacterial PDF inhibitors [30]. THF, and DMSO (entries1–5, Table 1); but those attempts provided
As per the literature survey, it was found that there are poor yields of products in comparison with, the use of a mixture
different drug molecules used as PDF inhibitors that contain of nitromethane and DMF (entry 6, Table 1). After the standard-
O/S/N-heterocycles as the core structure; nevertheless, nitrogen- ization of solvent, further, the reaction by increasing the temper-
containing imidazopyridine being an important heterocycle are not ature, power, and time were studied. It was noticed that increase
explored yet as PDF inhibitors. Indeed, imidazopyridine is a bio- in temperature, microwave power, and time increased the yield of
logically active compound and exhibits a wide range of biologi- the reaction; while better effectiveness was achieved with an ex-
cal activities such as antibacterial, antifungal, antiviral, antitumor, cellent yield up to 88% when the reaction proceeded at 100 °C and
and anti-inflammatory activities [31–35]. Other than this, the natu- 60W for 15 min in the presence of FeCl3 (30 mol%); thus, an excel-
rally occurring small pharmacophore 2H-chromene and its deriva- lent yield of product was obtained (entry 10, Table 1). Furthermore,
tives are also known to possess a large spectrum of activities such an increase in temperature, the yield was decreased (entry 11–12,
as antiallergic, antitumor, antiviral, antioxidant, anti-inflammatory, Table 1). Moreover, when the reaction was carried out in a neat
antibacterial, and anticancer [36–43]. The high lipophilicity of 2H- condition without any solvent, comparatively a very poor yield
chromene derivatives allows a better cell permeability across the of the target product had resulted. However, the common Lewis
bacterial cell membrane [44]. acids like, AlCl3 , ZnCl2 and CuCl were not effective for this reaction
Furthermore, some imidazo[1,2-a]pyridine derivatives and (entry 15–17, Table 1). The optimal reaction conditions were ob-
chromene derivatives have shown potent antibacterial activities tained using 2H-chromene-3-carbaldehydes 11a and 2-amino pyri-
which are shown (Fig 1) [45–51]. In present circumstances, hybrid dine 12, in the presence of the catalyst FeCl3 (30 mol%) in DMF
molecules get more attention as these show more potencies than and nitromethane binary solvent system at 100 °C, with 60W in

2
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Fig 1. Imidazopyridine (1–6), 2H-chromene based antibacterial agents (7,8), and designed 2H-chromene based imidazo[1,2-a]pyridine derivatives as synthetic targets.

Table 1
Optimization of the reaction conditionsa .

Entry Catalyst(mol%) Solvent Temp(°C) Watt Time(min) yieldb

1 FeCl3 (10%) DMF 40 30 10 n.r


2 FeCl3 (10%) DMF 80 30 10 45%
3 FeCl3 (10%) Toluene 80 30 10 20%
4 FeCl3 (10%) CH3 CN 80 30 10 34%
5 FeCl3 (10%) THF 80 30 10 16%
6 FeCl3 (10%) DMSO 80 30 10 28%
7 FeCl3 (10%) DMF 90 30 10 55%
8 FeCl3 (20%) DMF 90 60 10 63%
9 FeCl3 (30%) DMF 90 60 15 69%
10 FeCl3 (30%) DMF 100 60 15 88%
11 FeCl3 (30%) DMF 110 60 15 74%
12 FeCl3 (30%) DMF 120 60 20 62%
13 FeCl3 (30%) - 100 60 15 14%
14 FeCl3 (30%) - 110 60 20 19%
15 AlCl3 (30%) DMF 100 60 15 5%
16 ZnCl3 (30%) DMF 100 60 15 10%
17 CuCl(30%) DMF 100 60 15 7%
18 - DMF 100 60 15 n.r
a
Reaction conditions: Substituted 2H-chromene aldehydes (1.0 mmol), 2-
aminopyridine (1.2 mmol), FeCl3 (0.3 mmol) in a mixture of nitroalkane and
DMF (1:1) at 100°C in 60W for 15min.
b
Isolated yields, obtained by column chromatography.

15min. The completion of the reaction was monitored by TLC and tried in 2H-chromene-3-carbaldehydes 11(a–i) and 2-phenyl-2H-
the reaction mixture was extracted with a mixture of ethyl ac- chromene-3-carbaldehydes 11’(a–h) separately (Scheme 2). Both
etate and water. The organic layer was separated and was made electron-rich and electron-deficient substituents on 2H-chromene-
to dry over anhydrous Na2 SO4; further, the products were evapo- 3-carbaldehydes reacted efficiently with 2-aminopyridine to afford
rated under reduced pressure. The crude reaction mixture was pu- the desired products 13(a–q) with comparatively good to excel-
rified by column chromatography. The structures of 2H-chromene- lent yields. The methoxy and ethoxy groups on 2H-chromene-
based imidazo[1,2-a]pyridine derivatives 13a were established by 3-carbaldehydes and 2-phenyl-2H-chromene-3-carbaldehydes gave
1 H NMR, 13 C NMR, and mass spectroscopy. the corresponding products, namely 13(b–d), and 13(k–n) in good
With the optimized reaction conditions in hand, to broaden the to high yields (80–86%). Slightly decrease in the yield at 73-
scope of this protocol, the reaction by varying substituents were 78% was noticed when halogenated and naphthyl 2H-chromene-

3
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

8.0 and,-8.0 kCal/mol were noted. Furthermore, we have stud-


ied the antibacterial activity of all synthesized compounds 13(a-q)
against Gram-ve and Gram+ve bacterium and antibacterial results
revealed that these compounds were active against E. coli, K. oxy-
toca, S. aureus, and S. pyogenes (Table 3). Among all, compounds
13e, 13g, 13h, 13i, and 13n had better antibacterial activity. Partic-
ularly, the compound 13i was potentially active against E. coli, with
18 mm, as the zone of inhibition in the agar well diffusion method,
and 16 μg/mL was the MIC value, comparable to that of the stan-
dard drug Ciprofloxacin MIC value 28 μg/mL for each. Indeed, the
compounds containing the naphthyl group had significant antibac-
terial activity [56,57].

2.2.2. Structure-activity relationships (SAR studies)


In compounds, 13o, 13p, 13q, 13k,13l and 13m (2-phenyl
chromene based imidazo[1,2-a]pyridine) in presence of halogens
and methoxy group in different positions of the benzene ring of
benzopyran had less binding interactions with peptide deformy-
lase of both Gram-negative and Gram-positive bacteria like E. coli
and S. aureus; but the presence of the ethoxy group in 13n caused
increased binding interactions, so that 13n had a better antibac-
Scheme 1. Synthesis of 2H-chromene-3-carbaldehydes 11(a–i) and 2-Phenyl-2H- terial activity towards both human pathogens E. coli and S. au-
chromene-3- carbaldehydes 11’(a–h). reus with MIC values, 20 and 24 μg/mL, respectively. Surprisingly,
in compounds, 13e, 13g, 13h, and 13i the phenyl group is absent
in the 2-position of benzopyran), but with the presence of halo-
3-carbaldehydes and 2-phenyl-2H-chromene-3-carbaldehydes were gens and naphthyl substituents in different positions of the ben-
used as a starting material. However, in proceeding with the re- zene ring of benzopyran had a comparatively more binding inter-
action with simple 2-phenyl-2H-chromene-3-carbaldehydes 11’a, a action with peptide deformylase of both Gram-negative and Gram-
better yield (87%) of 13j was obtained. Structures of the synthe- positive bacteria such as E. coli and S. aureus. Moreover, the pres-
sized compounds, 13(a–q) were confirmed by 1 H NMR, 13 C NMR, ence of methoxy and bromo groups in the benzene ring of ben-
and mass spectroscopy (Table 2). zopyran showed comparatively a lesser binding interaction with
The probable mechanism of the reaction is presented in the peptide deformylase. Compounds 13e, 13g, 13h, and 13i, how-
Scheme 3. as, the reaction of 2-aminopyridine 12 and chromene ever, had better antibacterial activities towards both E. coli and S.
aldehyde 11 forms imine A, which underwent aza-Henry reaction aureus with higher MIC values. But at the naphthyl group at 5, 6
by CH3 NO2 and FeCl3 to produce intermediate B. Thereafter, tau- positions of the benzene ring of 2H-chromene having imidazo[1,2-
tomerization of the intermediate B gave intermediate C, which un- a]pyridine, the 13i caused a higher antibacterial activity against E.
derwent an intramolecular cyclization to give intermediate D. Fi- coli, and with the MIC value 16 μg/mL.
nally, the 2H-chromene based imidazo[1,2-a]pyridine derivative 13
was resulted by the subsequent eliminations of both water and
2.3. Computational studies
HNO [55].
To comprehend the mechanism of inhibition PDF enzyme,
2.2. Biological evaluation molecular docking studies were performed between the newly
synthesized 2H-chromene based imidazo[1,2-α ]pyridine deriva-
2.2.1. Antibacterial study tives, 13(a–q), against the PDF enzyme of E. coli (PDBID:1DFF), as
The binding affinity of the synthesized compounds 13(a–q) was well as, of S. aureus (PDBID:1LMH) [58]. The molecular docking
studied in silico with PDF. The results suggested that the com- studies signified the binding modes of action and the structural
pounds 13e, 13g, 13h, 13i, and 13n with good binding affinity val- optimizations of the synthesized compounds. The docking scores
ues as docking scores of 8.1 kCal/mol, -8.3 kCal/mol, -8.2 kCal/mol, of the newly synthesized compounds, 13(a–q) ranged from, -7.3
-8.7 kCal/mol, and -8.3 kCal/mol, respectively in case of E. coli; to -8.7 kCal/mol in the case of E. coli (PDBID:1DFF), and -6.2 to
while in case of S. aureus, docking scores of -6.6, -7.9, -7.8, - -8.0 kCal/mol in the case of S. aureus (PDBID:1LMH). The docking

Scheme 2. Synthesis of 2H-chromene based imidazo[1,2-a]pyridine derivatives 13(a–q)

4
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Table 2
Substrate scope for the synthesis of 2H-chromene based imidazo[1,2-α ]pyridine deriva-
tives 13(a–q) by the microwave method.

Serial no Compound Time (min) Yield(%)

1 15 88

2 15 85

3 15 84

4 15 82

5 15 78

6 15 77

7 15 73

8 15 75

(continued on next page)

5
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Table 2 (continued)

Serial no Compound Time (min) Yield(%)

9 15 74

10 15 87

11 15 86

12 15 85

13 15 83

14 15 80

15 15 79

(continued on next page)

6
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Table 2 (continued)

Serial no Compound Time (min) Yield(%)

16 15 77

17 15 72

Scheme 3. Proposed reaction mechanism of 2H-chromene based imidazo[1,2-a]pyridine.

scores and binding interactions are presented (SI-Table 1). Found interactions. The binding interaction of the most active PDF in-
from docking studies of compounds 13e, 13g, 13h, 13i, and 13n hibitor 13i is presented (Fig 2). The compound 13i held into the
that those possess some higher negative docking score values, viz., active pockets of PDF enzymes of both E. coli and S. aureus formed
-8.1 kCal/mol, -8.3 kCal/mol, -8.2 kCal/mol, -8.7 kCal/mol, and - several hydrophobic and Van der Waals interactions with amino
8.3 kCal/mol, respectively in case of E. coli; while, in case of S. acid residues, such as His132, Cys129, Ile44, Gly89, Arg97, Leu91of
aureus, docking scores were lesser, -6.6 kCal/mol, -7.9 kCal/mol, E. coli, with Glu185, Val59, His154, Val151 of S. aureus, respec-
-7.8 kCal/mol, -8.0 kCal/mol and,-8.0 kCal/mol for the respective tively with binding score -8.7 kCal/mol and -8.0 kCal/mol (Table 4).
five compounds, in succession. The binding energy values of all From the 3D-interaction of the synthesized 2H-chromene-based
compounds signify that compounds were held in the active pocket imidazo[1,2-α ]pyridine derivatives; it was found that the com-
of PDF of the target bacteria, E. coli, and S. aureus. The inter- pound 13i containing the naphthyl group was important for the
actions of compounds with PDF occur through hydrogen bond- PDF-inhibitibitory activity and consequently possesses a potent an-
ing, alkyl, π -π interaction, π - donor hydrogen bonding and π -σ tibacterial activity.

7
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Fig. 2. Structure-activity relationship of chromene based imidazopyridines.

Fig. 3. Docking study of compound 13i with E. coli and binding interaction with PDF (PDBID:1DFF).

8
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

Table 3
Antimicrobial activities of synthesized 2H-chromene based imidazo[1,2-a]pyridine derivatives.

Compound E. coli K. oxytoca S. aureus S. pyogenes


name
ZI (mm) MIC (μg/mL) ZI (mm) MIC (μg/mL) ZI (mm) MIC (μg/mL) ZI (mm) MIC (μg/mL)

13a 12 36 12 36 13 32 13 36
13b 13 32 12 36 13 32 13 36
13c 12 36 13 36 12 36 14 32
13d 14 28 13 36 13 36 13 32
13e 16 24 14 32 15 24 14 28
13f 14 28 13 36 13 32 13 32
13g 17 20 14 32 17 20 15 28
13h 16 24 14 28 16 24 14 24
13i 18 16 15 24 17 20 16 28
13j 14 28 12 32 14 28 13 32
13k 13 28 11 36 13 32 12 32
13l 14 24 12 36 13 32 12 36
13m 13 28 11 36 13 32 13 32
13n 17 20 15 28 16 24 14 28
13o 14 28 11 36 12 32 13 32
13p 13 32 11 36 12 32 13 32
13q 13 32 11 36 12 32 13 32
Std 17 28 18 24 28 28 18 20

ZI: Zone of inhibition; MIC: minimum inhibitory concentration; Std: Standard, Ciprofloxacin.

Fig. 4. Docking study of compound 13i with S. aureus and binding interaction with PDF (PDBID:1LMH).

Table 4
Docking score (kCal/mol) and binding interactions of compound 13i with peptide deformylase.

Testedcompound Docking score (kCal/mol) and binding interactions with peptide deformylase

E. coli (PDBID:1DFF) S. aureus (PDBID:1LMH)


13i -8.7 -8.0
HIS132, CYS129, ILE44, GLY89, ARG97, LEU91 GLU185, VAL59, HIS154, VAL151

3. Conclusion ghoulish bacterial pathogens, S. aureus, S. pyogenes, E. coli, and K.


oxytoca as used in vitro for this study. Moreover, the compounds
Herein, a simple and novel one-pot, microwave-assisted syn- 13e, 13g, 13h, 13i, and 13n could be suggested for further studies
thesis of 2H-chromene based imidazo[1,2-a]pyridines are reported, in pharmacological drug development attempts.
and evaluated for antibacterial activities along with the MIC values
of the compounds, against human pathogenic representative two 4. Experimental
Gram-positive and Gram-negative bacteria. The results indicated
that these selected during the synthesis of these compounds could 4.1. Instrumentation
be suggested as potent compounds with antibacterial potency for
two reasons namely, (1) molecular docking results of these com- All reagents and solvents were purchased from commercial sup-
pounds and PDF individually landed at encouraging docking score pliers. 2-aminopyridine, nitromethane, FeCl3 , and all solvents were
results, (2) the in vitro antibacterial screening by agar-well diffu- purchased from Sigma-Aldrich. Substituted 2H-chromene aldehy-
sion method followed by MIC assay had too encouraging outcomes. des were synthesized in the laboratory. Progress of the reaction
Thus, it would not be out of place to state that the clinical frater- was monitored by TLC on silica gel 60 F254 -coated TLC plates
nity is in search of such novel synthetic potent molecules for the (Merck KGaA, Darmstadt, Germany) and was visualized by Short-

9
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

UV light at 254 nm. The proton nuclear magnetic resonance (1 H 4.2.4.


and 13 C NMR) spectra were recorded on a JEOL 400 MHz NMR 4.2.4.2-(8-ethoxy-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13d)
spectrometer. Chemical shifts (δ ) are reported in parts per million, Brick red solid; m.p. 197–199 °C,1 H NMR(400 MHz, CDCl3 , δ
downfield from the internal standard (TMS, δ = 0.00 ppm) rela- ppm) δ 1.47 (t, J = 6.8 Hz, 3H), 4.13 (q, J = 6.8 Hz, 2H), 5.24 (s,
tive to residual CDCl3 (1 H: δ = 7.26 ppm, 13 C: δ = 77.00 ppm) 2H), 6.77–6.87 (m, 4H), 7.16–7.20 (m, 1H), 7.27 (s, 1H), 7.58–7.60
as an internal reference. Coupling constants (J) were reported in (m, 2H), 8.08 (d, J = 6.8 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ
Hertz (Hz). Peak multiplicity was indicated as follows: s-singlet, d- ppm): 14.9, 64.5, 66.2, 108.5, 112.5, 113.5, 117.3, 119.6, 120.1, 121.2,
doublet, t-triplet, q-quartet, m-multiplet, and dd-doublet of a dou- 124.6, 125.3, 125.5, 125.6, 142.1, 142.7, 145.7, 146.8. ESI-HRMS:
blet. Melting points were recorded with an electrothermal digi- m/z [M + H]+ calcd for C18 H16 N2 O2 : 293.1212; found: 293.1286.
tal melting point apparatus measured on a Stuart SPM10 melt- Calcd%:C, 73.95; H, 5.52; N, 9.58; Found% C, 73.94; H, 5.50; N, 9.58.
ing point instrument. High-resolution mass spectra (HRMS) were
recorded at IIT, Madras as well as, NISER, Bhubaneswar. CHN anal- 4.2.5.
ysis were recorded at central laboratory-OUAT, Bhubaneswar. The 4.2.5.2-(6-chloro-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13e)
final product was synthesized using a CEM microwave synthesizer Brown solid; m.p. 204–208 °C; 1 H NMR(400 MHz, CDCl3 , δ
(model No-908010). ppm) 5.21 (s, 2H), 6.77–6.81 (m, 2H), 7.05-7.08 (s, 2H), 7.19–7.23
(m, 2H), 7.58–7.62 (m, 2H), 8.09 (d, J = 7.2 Hz, 1H); 13 C NMR
(100 MHz, CDCl3 , δ ppm): 66.2, 108.8,112.7, 116.8, 117.4, 118.9,
4.2. General procedure for the synthesis of compound 13(a–q) 124.2, 125.5, 125.6, 126.3, 126.5, 126.9, 128.5, 141.7, 145.8, 152.1.
ESI-HRMS: m/z [M + H]+ calcd for C16 H11 ClN2 O: 283.0560; found:
A mixture of substituted 2H-chromene aldehydes(1.0mmol), 2- 283.0634. Calcd%: C, 67.97; H, 3.92; N, 9.91; Found% C, 67.95; H,
aminopyridine (1.2 mmol), and FeCl3 (0.3mmol) in a mixture of ni- 3.91; N, 9.90.
troalkane and DMF (1:1) was taken in a clean dry microwave tube
and was placed under microwave irradiation at 60W,100 °C for 4.2.6.
15 min. The progress of the reaction was monitored by TLC. Af- 4.2.6.2-(6-bromo-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13f)
ter completion of the reaction, the reaction mixture was diluted Brown solid; m.p. 207–209 °C; 1 H NMR (400 MHz, CDCl3 , δ
with ethyl acetate and the product was extracted with water. The ppm) 5.20 (s, 2H), 6.73 (d, J = 7.6 Hz, 1H), 6.78–6.81 (m, 1H), 7.17–
organic layer was separated, dried over anhydrous Na2 SO4, and 7.26 (m, 4H),7.58–7.61 (m, 2H), 8.08 (d, J = 6.4 Hz, 1H); 13 C NMR
evaporated under reduced pressure. The crude reaction mixture (100 MHz, CDCl3 , δ ppm): 66.2, 108.8, 112.7, 113.6, 117.2, 117.4,
was purified by column chromatography to furnish the desired 2H- 118.7, 124.7, 125.5, 125.6, 126.8, 129.3, 131.4, 141.7, 145.7, 152.6.
chromene based imidazo[1,2-a]pyridines 13(a–q) with good to high ESI-HRMS: m/z [M + H]+ calcd for C16 H11 BrN2 O: 327.0055; found:
yield (72–88%). 327.0131. Calcd%: C, 58.74; H, 3.39; N, 8.56; Found% C, 58.71; H,
3.36; N, 8.55.

4.2.1. 4.2.1.2-(2H-chromen-3-yl)imidazo[1,2-a]pyridine(13a)
4.2.7.
Brown solid; m.p. 198–200 °C; 1 H NMR(400 MHz, CDCl3 , δ
4.2.7.2-(6,8-dibromo-2H-chromen-3-yl)imidazo[1,2-a]pyridine
ppm) δ 5.20 (s, 2H), 6.76–6.79 (m, 1H), 6.85 (d, 1H, J = 8Hz),
(13g)
6.91–6.94 (m, 1H), 7.12–7.13 (m, 2H), 7.15–7.21 (m, 1H), 7.27 (s,
Deep red solid; m.p. 211–213 °C; 1 H NMR(400 MHz, CDCl3 , δ
1H), 7.59–7.60 (m, 2H), 8.08 (d, 1H, J = 6.8 Hz); 13 C NMR (100
ppm) 5.32 (s, 2H), 6.79–6.82 (m, 1H), 7.16 (d, J = 2.4 Hz, 1H), 7.17–
MHz, CDCl3 , δ ppm): 66.1, 108.4, 112.5, 115.5, 117.3, 120.1, 121.6,
7.18 (m, 1H), 7.20-7.24 (m, 1H), 7.46 (d, J = 2.0 Hz, 1H), 7.58–7.61
122.8, 125.3, 125.6, 127.2, 129.0, 142.2, 145.7, 153.6 . ESI-HRMS:
(m, 2H), 8.09–8.11 (m, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm):
m/z [M + H]+ calcd for C16 H12 N2 O: 249.0950; found: 249.0872.
66.9, 109.0, 110.3, 112.8, 113.5, 117.5, 118.2, 125.5, 125.7, 125.8,
Calcd%: C, 77.40; H, 4.87; N, 11.28; Found% C, 77.38; H, 4.85; N,
127.4, 128.6, 134.0, 141.1, 145.8, 149.5. ESI-HRMS: m/z [M + H]+
11.25.
calcd for C16 H10 Br2 N2 O: 406.9139; found: 406.9210. Calcd%: C,
47.33; H, 2.48; N, 6.90; Found% C, 47.30; H, 2.46; N, 6.88.
4.2.2.
4.2.2.2-(6-methoxy-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13b) 4.2.8.
Brick red solid; m.p. 196-201 °C; 1 H NMR(400 MHz, CDCl3 , δ 4.2.8.2-(6,8-dichloro-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13h)
ppm) δ 3.78 (s, 3H), 5.14 (s, 2H), 6.68–6.70 (m, 2H), 6.78–6.81 (m, Brown solid; m.p. 207–208 °C; 1 H NMR (400 MHz, CDCl3 , δ
2H), 7.17–7.21 (m, 1H), 7.24–7.26 (m, 1H), 7.57–7.60 (m, 2H), 8.08 ppm) 5.30 (s, 2H), 6.79–6.83 (m, 1H), 6.99 (d, J =2Hz, 1H), 7.16–7.19
(d, J = 6.4 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): 55.7, 66.1, (m, 2H), 7.22–7.24 (m, 1H), 7.59–7.61 (m, 2H), 8.10 (d, J = 7.2 Hz,
108.6, 112.2, 112.5, 114.0, 116.0, 117.3, 120.2, 123.6, 125.4, 125.6, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): 66.7, 109.0, 112.8, 117.4,
126.6, 142.1, 145.7, 147.6, 154.3. ESI-HRMS: m/z [M + H]+ calcd for 118.2, 121.2, 125.0, 125.6, 125.7, 126.1, 127.4, 128.6, 141.0, 145.8,
C17 H14 N2 O2 : 279.1055; found:279.1262. Calcd%: C, 73.37; H, 5.07; 147.9. ESI-HRMS: m/z [M + H]+ calcd for C16 H10 Cl2 N2 O: 317.0170;
N, 10.07; Found% C, 73.33; H, 5.06; N, 10.05. found: 317.0257. Calcd%: C, 60.59; H, 3.18; N, 8.83; Found% C,
60.58; H, 3.15; N, 8.82.

4.2.3. 4.2.9.
4.2.3.2-(7-methoxy-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13c) 4.2.9.2-(3H-benzo[f]chromen-2-yl)imidazo[1,2-a]pyridine(13i)
Deep brown solid; m.p. 194-198 °C, 1 H NMR(400 MHz, CDCl3 , Brown solid; m.p. 209–211 °C; 1 H NMR (400 MHz, CDCl3 , δ
δ ppm) δ 3.79 (s, 3H), 5.17 (s, 2H), 6.45–6.49 (m, 2H), 6.75–6.77 ppm) 5.27 (s, 2H), 6.80-6.83 (m, 1H), 7.14 (d, J = 8.8 Hz, 1H),
(m, 1H), 7.03 (d, J = 7.6 Hz, 1H), 7.16–7.22 (m, 2H), 7.55–7.59 (m, 7.19–7.23 (m, 1H), 7.36–7.39 (m, 1H), 7.49-7.53 (m, 1H), 7.63–7.68
2H), 8.07 (d, J = 6.8 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): (m, 3H), 7.76 (d, J = 8.4 Hz, 1H), 7.99 (s, 1H), 8.11 (d, J = 6.4
55.4, 66.2, 101.5, 107.4, 107.9, 112.4, 116.0, 117.1, 120.0, 122.5, 125.2, Hz, 1H), 8.20 (d, J = 8.0 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ
125.5, 127.9, 142.5, 145.6, 154.9, 160.7. ESI-HRMS: m/z [M + H]+ ppm): 66.1, 108.6, 112.5, 116.0, 116.4, 117.3, 117.3, 121.9,123.8, 123.9,
calcd for C17 H14 N2 O2 : 279.1055; found:279.1113. Calcd%: C, 73.37; 125.4, 125.6, 126.6,128.5,129.3, 129.6, 130.2, 142.6, 145.8, 152.0.
H, 5.07; N, 10.07; Found% C, 73.36; H, 5.06; N, 10.07. ESI-HRMS: m/z [M + H]+ calcd for C20 H14 N2 O: 299.1106; found:

10
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

299.1178. Calcd%: C, 80.52; H, 4.73; N, 9.39; Found% C, 80.50; H, 4.2.15. 4.2.15.2-(6-bromo-2-phenyl-2H-chromen-3-yl)imidazo[1,2-


4.70; N, 9.38 . a]pyridine(13o)
Brown solid; m.p. 195–197 °C; 1 H NMR (400 MHz, CDCl3 , δ
4.2.10. ppm) 6.31 (s, 1H), 6.64 (d, J = 8.4 Hz, 1H), 6.71-6.74 (m, 1H),
4.2.10.2-(2-phenyl-2H-chromen-3-yl)imidazo[1,2-a]pyridine(13j) 7.13–7.19 (m, 2H), 7.27–7.28 (m, 4H), 7.34 (s, 1H), 7.46–7.48 (m,
Brown solid; m.p. 198-200 °C; 1 H NMR (400 MHz, CDCl3 , δ 2H), 7.56–7.58 (m, 2H), 7.95 (d, J = 6.8 Hz, 1H); 13 C NMR (100
ppm) 6.31 (s, 1H), 6.69-6.72 (m, 1H), 6.76 (d, J = 8 Hz,1H), 6.86– MHz, CDCl3 , δ ppm): 76.7, 109.6, 112.5, 113.5, 117.3, 118.1, 119.6,
6.90 (m, 1H), 7.05–7.09 (m, 1H), 7.14-7.21 (m, 2H), 7.25–7.30 (m, 124.5, 125.5, 125.6, 127.9, 127.9, 128.2, 128.7, 128.7, 128.9, 129.3,
4H), 7.49–7.50 (m, 2H), 7.56 (d, J = 9.2 Hz, 1H), 7.65 (s, 1H), 7.92 131.8, 138.3, 142.1, 145.6, 150.8. ESI-HRMS: m/z [M+H]+ calcd
(d, J = 6.4 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): 77.3, 109.2, for C22 H15 BrN2 O: 403.0368; found: 403.0203. Calcd%: C, 65.52; H,
112.3, 116.3, 117.2, 120.9, 121.4, 122.4, 125.3, 125.6, 126.9, 127.1, 3.75; N, 6.95; Found% C, 65.50; H, 3.72; N, 6.94.
127.9, 128.6, 128.7, 129.3, 138.8, 142.6, 145.5, 151.7. ESI-HRMS: m/z
[M + H]+ calcd for C22 H16 N2 O: 325.1263; found: 325.1339. Calcd%: 4.2.16. 4.2.16.2-(6-chloro-2-phenyl-2H-chromen-3-yl)imidazo[1,2-
C, 81.46; H, 4.97; N, 8.64; Found% C, 81.44; H,4.95; N, 8.61. a]pyridine(13p)
Brown solid; m.p. 197-199o C; 1 H NMR(400 MHz, CDCl3 , δ ppm)
4.2.11. 4.2.11.2-(8-methoxy-2-phenyl-2H-chromen-3-yl)imidazo[1,2- 6.31 (s, 1H), 6.68-6.74 (m, 2H), 7.01 (dd, J = 2.8 Hz, J = 8.8 Hz,
a]pyridine(13k) 1H), 7.15–7.19 (m, 2H), 7.27–7.29 (m, 3H), 7.34 (s, 1H), 7.45–7.49
Brown solid; m.p. 198–201o C; 1 H NMR (400 MHz, CDCl3 , δ (m, 2H), 7.56–7.58 (m, 2H), 7.95 (d, J = 6.8 Hz, 1H); 13 C NMR (100
ppm) 3.78 (s, 3H), 6.38 (s, 1H), 6.71–6.75 (m, 2H), 6.81–6.85 (m, MHz, CDCl3 , δ ppm): 76.7, 109.6, 112.4, 117.3, 117.7, 119.7, 123.9,
2H), 7.13–7.17 (m, 1H), 7.24–7.28 (m, 3H), 7.35 (s, 1H), 7.53-7.58 125.5, 125.6, 126.1, 126.4, 127.9, 127.9, 128.3, 128.7, 128.8, 128.8,
(m, 3H), 7.65 (s, 1H), 7.95 (d, J = 7.2 Hz, 1H); 13 C NMR (100 128.9, 138.3, 142.1, 145.5, 150.2. ESI-HRMS: m/z [M + H]+ calcd
MHz, CDCl3 , δ ppm): 56.4, 76.8, 109.4, 112.4, 112.8, 117.3, 119.7, for C22 H15 ClN2 O: 359.0873; found: 359.0979. Calcd%: C, 73.64; H,
120.9, 121.2, 123.4, 125.5, 125.7, 127.2, 128.0, 128.6, 128.7, 138.8, 4.21; N, 7.81; Found% C, 73.61; H, 4.19; N, 7.79.
140.8, 142.7, 145.7, 148.3. ESI-HRMS: m/z [M + H]+ calcd for
C23 H18 N2 O2 : 355.1368; found: 355.1420. Calcd%: C, 77.95; H, 5.12;
4.2.17. 4.2.17.2-(8-bromo-6-chloro-2-phenyl-2H-chromen-3-
N, 7.90; Found% C, 77.90; H, 5.06; N, 7.85.
yl)imidazo[1,2-a]pyridine(13q)
Brown solid; m.p. 201–204 °C; 1 H NMR (400 MHz, CDCl3 , δ
4.2.12. 4.2.12.2-(6-methoxy-2-phenyl-2H-chromen-3-
ppm) 6.45 (s, 1H), 6.74–6.77 (m, 1H), 7.09 (d, J= 2 Hz,1H), 7.18–7.22
yl)imidazo[1,2-a]pyridine(13l)
(m, 2H),7.27–7.28 (m, 3H), 7.41 (s, 1H), 7.49–7.51 (m, 2H), 7.55 (s,
Brown solid; m.p. 197–199o C; 1 H NMR (400 MHz, CDCl3 , δ
1H), 7.59 (d, J = 9.2 Hz, 1H), 7.98 (d, J = 6.8 Hz, 1H); 13 C NMR (100
ppm) 3.76 (s, 3H), 6.25 (s, 1H), 6.61–6.65 (m, 1H), 6.68–6.73 (m,
MHz, CDCl3 , δ ppm): 77.3, 109.9, 110.9, 112.6, 117.4, 119.4, 125.1,
2H), 6.76 (d, J = 2.8 Hz, 1H), 7.13–7.17 (m, 1H), 7.24–7.29 (m, 3H),
125.6, 125.7, 125.8, 126.6, 127.8, 127.8, 128.6, 128.6, 128.9, 129.2,
7.33 (s, 1H), 7.47–7.49 (m, 2H), 7.56 (d, J = 9.2 Hz, 1H), 7.62(s,
131.5, 137.7, 141.9, 145.7, 147.4. Calcd%: C, 60.37; H, 3.22; N, 6.40;
1H), 7.94 (d, J = 6.0 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm):
Found% C, 60.35; H, 3.20; N, 6.38.
55.6, 76.7, 109.3, 112.0, 112.3, 114.5, 116.9, 117.2, 121.0, 123.2, 125.4,
125.6, 127.8, 127.9, 128.5, 128.6, 138.6, 142.6, 145.5, 145.6, 154.1.
ESI-HRMS: m/z [M + H]+ calcd for C23 H18 N2 O2 : 355.1368; found: 4.3. Antibacterial assay
355.1457. Calcd%: C, 77.95; H, 5.12; N, 7.90; Found% C, 77.92; H,
5.11; N, 7.88. 2H-chromene based imidazo[1,2-a]pyridines derivatives were
studied in vitro for antibacterial assay against Gram -ve (E. coli and
4.2.13. 4.2.13.2-(7-methoxy-2-phenyl-2H-chromen-3- K. oxytoca) and Gram +ve (S. aureus and S. pyogene) bacteria for
yl)imidazo[1,2-a]pyridine(13m) the zone sizes of inhibitions and MIC assays. Preliminarily, these
Brown solid; m.p. 196–198 °C; 1 H NMR (400 MHz, CDCl3 , δ cited bacterial strains were procured from CRL, Institute of Medi-
ppm) 3.73 (s, 3H), 6.28 (s, 1H), 6.35 (d, J = 2.4 Hz, 1H), 6.45 cal Science, and SUM Hospital. Then, bacterial strains were inocu-
(dd, J = 2.4 Hz, J = 8.0 Hz, 1H), 6.69 (t, J = 7.2 Hz, 1H), 7.10– lated followed by incubated for 24–48 h. By the by, sterile media
7.15 (m, 2H), 7.27–7.29 (m, 4H), 7.49–7.56 (m, 3H), 7.61 (s, 1H), was allowed to aliquot each Petri plates further solidifying, con-
7.93 (d, J = 6.8 Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): comitantly each well was loaded with test samples at 60μl. There-
55.3, 76.7, 102.1, 107.4, 108.7, 112.1, 115.7, 117.1, 120.7, 124.0, 125.2, after plates were incubated for 24h at 37 ˚C to the estimation of
125.5, 127.8, 127.9, 128.6, 128.7, 138.9, 142.9, 145.5, 153.0, 160.9. the zone of inhibition. Moreover, MIC assay was assessed previ-
ESI-HRMS: m/z [M + H]+ calcd for C23 H18 N2 O2 : 355.1368; found: ously reported [53], 96-well plate with various concentrations at
355.1417. Calcd%: C, 77.95; H, 5.12; N, 7.90; Found% C, 77.93; H, 20 0,10 0,75,50,40,36,32,28,24,20,16,12,8,4 and 2μg/ml. These plates
5.09; N, 7.88. were allowed to aliquot nutrient broth, bacterial suspension, and
various concentrated test samples. Consequently, individual wells
were treated with TTC dye for monitoring bacterial growth inhibi-
4.2.14. 4.2.14.2-(8-ethoxy-2-phenyl-2H-chromen-3-yl)imidazo[1,2-
tion [54].
a]pyridine(13n)
Brown solid; m.p. 199–201 °C; 1 H NMR (400 MHz, CDCl3 , δ
ppm) δ 1.32 (t, J = 6.8 Hz, 3H), 3.98 (q, J = 6.8 Hz, 2H), 6.37 (s, 1H), 4.4. Molecular docking study
6.70–6.75 (m, 2H), 6.79–6.86 (m, 2H), 7.14-7.18 (m, 1H), 7.24–7.25
(m, 3H), 7.36 (s, 1H), 7.53–7.58 (m, 3H), 7.65 (s, 1H), 7.96 (d, J = 6.8 Firstly, 2H-chromene based imidazo [1,2-α ]pyridines derivatives
Hz, 1H); 13 C NMR (100 MHz, CDCl3 , δ ppm): 14.8, 65.0, 76.7, 109.2, 13(a–q) were drawn virtually by using ChemDraw-v12.0 and each
112.3, 114.9, 117.2, 119.8, 120.9, 121.1, 123.7, 125.4, 125.6, 127.1, structure was correctly 2D-geometric optimization with clean-up
127.9, 127.9, 128.4, 128.4, 128.5, 138.8, 141.4, 142.8, 145.6, 147.4. errors. Then, each structure was 3D optimized and energy min-
ESI-HRMS: m/z [M + H]+ calcd for C24 H20 N2 O2 : 369.1525; found: imization by ChemSketch-v2015 and open Babel-v2.4. Concomi-
369.1601. Calcd%: C, 78.24; H, 5.47; N, 7.60; Found% C, 78.22; H, tantly, the crystal structure of bacterial protein-peptide deformy-
5.45; N, 7.59. lase (PDBID 1DEF and 1LMH) was retrieved from the protein data

11
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

bank, then removal of heteroatoms and missing side-chains. More- [12] J.M. Clements, A.P. Ayscough, K. Keavey, S.P. East, Peptide deformylase in-
over, a molecular docking study was performed using Autodock- hibitors, potential for a new class of broad-spectrum antibacterials, Curr. Med.
Chem. Anti Infect. Agents 1 (2002) 239–249.
v4.2 of this prepared ligand (derivatives) and proteins by integu- [13] J.M. Martin, M. Green, K.A. Barbadora, E.R. Wald, Erythromycin-resistant group
ment of Lamarckian algorithm with each run for 270 0 0 genera- a streptococci in school children in Pittsburgh, N. Engl. J. Med. 346 (2002)
tions. All the confirmed docking runs were analyzed and the gen- 1200–1206.
[14] J.N. Sangshetti, F.A.K. Khan, D.B. Shinde, Peptide deformylase: a new target in
erated complex was analyzed by BIOVIA DS-v4.5 for intermolecular antibacterial, antimalarial and anticancer drug discovery, Curr. Med. Chem. 22
residue interactions [55]. (2016) 214–236.
[15] C.R. Sahoo, S.K. Paidesetty, B. Dehury, R.N. Padhy, Molecular dynamics and
Declaration of Competing Interest computational study of mannich-based coumarin derivatives: potent tyrosine
kinase inhibitor, J. Biomol. Struct. Dyn. 18 (2020) 5419–5428.
[16] K.T. Nguyen, C.C X.Hu, R. Chakrabarti, M. X.Zhu, D. Pei, Characterization of a
The authors declare the following financial interests/personal human peptide deformylase: implications for antibacterial drug design, Bio-
relationships which may be considered as potential competing in- chemistry 42 (2003) 9952–9958.
[17] Z. Yuan, R.J. White, The evolution of peptide deformylase as a target: contri-
terests: bution of biochemistry, genetics and genomics, Biochem. Pharmacol. 71 (2006)
1042–1047.
CRediT authorship contribution statement [18] Z. Yuan, J. Trias, R.J. White, Deformylase as a novel antibacterial target, Drug.
Discov. Today 6 (2001) 954–961.
[19] K. Aubart, M. Zalacain, Peptide deformylase inhibitors, Prog. Med. Chem. 44
Nilima Priyadarsini Mishra: Conceptualization, Methodology, (2006) 109–143.
Writing – original draft. Seetaram Mohapatra: Conceptualiza- [20] K.B. Alexander, Y. Qingfeng, Y.Q. Steven, M.K. Haldar, D.K. Srivastava, Solven-
tion, Supervision, Project administration. Chita Ranjan Sahoo: Re- t-assisted slow conversion of a dithiazole derivative produces a competitive
inhibitor of peptide deformylase, Biochim. Biophys. Acta 1804 (2010) 704–713.
sources, Software. Bishnu Prasad Raiguru: Data curation. Sabita [21] D. Z.Chen, D. V.Patel, C. J.Hackbarth, Z.Y W.Wang, Actinonin, a naturally oc-
Nayak: Writing – review & editing. Subhrakant Jena: Formal anal- curring antibacterial agent, is a potent deformylase inhibitor, Biochemistry 39
ysis. Rabindra Nath Padhy: Conceptualization. (20 0 0) 1256–1262.
[22] D.C R.Jain, R. J.White, D. V.Patel, Z. Yuan, Bacterial peptide deformylase in-
hibitors: a new class of antibacterial agents, Curr. Med. Chem. 12 (2005)
Acknowledgments 1607–1621.
[23] K.T.N X.Hu, V. C.Jiang, H.E.M D.Lofland, D. Pei, Macrocyclic inhibitors for pep-
The author NPM acknowledges INSPIRE program tide deformylase: a structure−activity relationship study of the ring size, J.
Med. Chem. 47 (2004) 4941–4949.
(DST/INSPIRE/03/2015/004518, New Delhi) for providing finan- [24] F.A.K. Khan, K.S. Jadhav, R.H. Patil, D.B. Shinde, R.B. Arote, J.N. Sangshetti,
cial supports in the form of INSPIRE FELLOWSHIP. The author SRM Biphenyltetrazole-thiazolidinediones as novel bacterial peptide deformylase
is grateful to the CSIR, New Delhi, for the financial support No- inhibitors: Synthesis, biological evaluations and molecular docking study,
Biomed. Pharmacother. 83 (2016) 1146–1153.
02(0381)/19/EMR-II, for the work. [25] M.K Gupta, P. Mishra, P. Prathipati, A.K. Saxena, 2D-QSAR in hydroxamic acid
derivatives as peptide deformylase inhibitors and antibacterial agents, Bioorg.
Supplementary materials Med. Chem. 10 (2002) 3713–3716.
[26] S. Petit, Y. Duroc, V. Larue, C.L C.Giglione, C. Soulama, A. Denis, T.M F.Dardel,
I. Artaud, Structure-activity relationship analysis of the peptide deformylase
Supplementary material associated with this article can be inhibitor 5-bromo-1H-indole-3-acetohydroxamic acid, Chem. Med. Chem 4
found, in the online version, at doi:10.1016/j.molstruc.2021.131183. (2009) 261–275.
[27] W. Shi, Y. Duan, Y. Qian, M. Li, L. Yang, W. Hu, Design, synthesis, and antibac-
References terial activity of 2,5-dihydropyrrole formyl hydroxyamino derivatives as novel
peptide deformylase inhibitors, Bioorg. Med. Chem. Lett. 20 (2010) 3592–3595.
[1] C.R. Sahoo, J. Sahoo, M. Mahapatra, D. Lenka, P.K. Sahu, B. Dehury, R.N. Padhy, [28] D.S C.Antczak, M.G.F B.Bassit, Y. Li, D.A.S E.Stanchina, H. Djaballah, Identifi-
S.K. Paidesetty, Coumarin derivatives as promising antibacterial agent, Arab. J. cation of benzofuran-4,5-diones as novel and selective non-hydroxamic acid,
Chem. 14 (2021) 102922. non-peptidomimetic based inhibitors of human peptide deformylase, Bioorg.
[2] S.S. Swain, S.K. Paidesetty, B. Dehury, M. Das, S.C. Vedithi, R.N. Padhy, Med. Chem. Lett. 21 (2011) 4528–4532.
Computer-aided synthesis of dapsone-phytochemical conjugates against [29] L. Yu, W. Zhou, Z. Wang, Synthesis and in vitro antibacterial activity of oxazo-
dapsone-resistant mycobacterium leprae, Sci. Rep. 10 (2020) 6839, doi:10. lidine LBM-415 analogs as peptide deformylase inhibitors, Bioorg. Med. Chem.
1038/s41598- 020- 63913- 9. Lett. 21 (2011) 1541–1544.
[3] S. Rath, S.R. Das, R.N. Padhy, Surveillance of bacteria pseudomonas aeruginosa [30] R.A.Al-Q T.H.Al-Tel, Postgroebke–blackburn multicomponent protocol: synthe-
and MRSA associated with chronic suppurative otitis mediaVigilância de bac- sis of new polyfunctionalimidazo[1,2-a]pyridine and imidazo[1,2-a]pyrimidine
térias pseudomonas aeruginosae MRSA associadas à otite média crônica supu- derivatives as potential antimicrobial agents, Eur. J. Med. Chem. 45 (12) (2010)
rativa, Braz. J. Otorhinolaryngol. 83 (2017) 201–206. 5848–5855.
[4] M.P. Mishra, N.K. Debata, R.N. Padhy, Surveillance of multidrug-resistant [31] G.L.T.Z. Z.A.Kaplancikli, G.R A.O.Zdemi˙r, Synthesis and anticandidal activity
uropathogenic bacteria in hospitalized patients in Indian, Asian Pac. J. Trop. of some imidazopyridine derivatives, J. Enzym. Inhib. Med.Ch. 23 (6) (2008)
Biomed. 3 (2013) 315–324. 866–870.
[5] J.R. Goldsteina, R.D. Lee, in: Demographic Perspectives on the Mortality of [32] M. Lhassani, O. Chavignon, J.M. Chezal, J.C. Teulade, J.P. Chapat, R. Snoeck,
COVID-19 and other Epidemics, 117, PNAS, 2020, pp. 22035–22041. G. Andrei, J. Balzarini, E. De Clercq, A. Gueiffier, Synthesis and antiviral activity
[6] K. Arteaga-Livias, K. Pinzas-Acosta, L. Perez-Abad, V. Panduro-Correa, of imidazo[1,2-a] pyridines, Eur. J. Med. Chem. 34 (1999) 271–274.
A.A. Rabaan, S. Pecho-Silva, B. Dámaso-Mata, A multidrug-resistant Klebsiella [33] M. Hranjec, I. Piantanida, M. Kralj, L. Suman, K. Pavelıc´, G. Karmin-
pneumoniae outbreak in a Peruvian hospital: Another threat from the COVID- ski-Zamola, Novel amidino-substituted thienyl- and furylvinyl benzimidazole:
19 pandemic, Infect. Control Hosp. Epidemiol. (2021), doi:10.1017/ice.2020. derivatives and their photochemical conversion into corresponding diazacy-
1401. clopenta[c]fluorenes. synthesis, interactions with DNA and RNA, and antitumor
[7] S. Doron, L.E. Davidson, A. Stewardship, Antimicrobial Stewardship, Mayo Clin. evaluation, J. Med. Chem. 51 (2008) 4899–4910.
Proc. 86 (2011) 1113–1123. [34] C. Hamdouchi, J. de Blas, M. del Prado, J. Gruber, B.A. Heinz, L. Vance,
[8] L. Wang, C. Hu, L. Shao, The antimicrobial activity of nanoparticles: present 2-Amino-3-substituted-6-[(E)-1-phenyl-2-(N-methylcarbamoyl)vinyl]imidazo
situation and prospects for the future, Int. J. Nanomed. 12 (2017) 1227–1249. [1,2-a]pyridines as a novel class of inhibitors of human rhinovirus: stereospe-
[9] K.D. Vihta, N. Stoesser, M.J. Llewelyn, T.P. Quan, T. Davies, N.J Fawcett, L. Dunn, cific synthesis and antiviral activity, J. Med. Chem. 42 (1999) 50–59.
K. Jeffery, C.C. Butler, G. Hayward, M. Andersson, M. Morgan, S. Oakley, A. Ma- [35] F. Borges, F. Roleira, N. Milhazes, L. Santana, E. Uriarte, Simple coumarins and
son, S. Hopkins, D.H. Wyllie, D.W. Crook, M.H. Wilcox, A.P. Johnson, T.E.A. Peto, analogues in medicinal chemistry: occurrence, synthesis and biological activity,
A.S. Walker. Trends Over time in Escherichia Coli Bloodstream Infections, Uri- Curr. Med. Chem. 12 (2005) 887.
nary Tract Infections, and Antibiotic Susceptibilities in Oxfordshire, UK, 1998– [36] S. Jain, P.K. Paliwal, G.N. Babu, A. Bhatewara, DABCO promoted one-pot synthe-
2016: a study of electronic health records. Lancelet, (10) (2018) 1138-1149. sis of dihydropyrano(c)chromene and pyrano[2,3-d]pyrimidine derivatives and
[10] P. Calı, L. Narum, S. Mukhija, A. Hjelmencrantz, Isoxazole-3-hydroxamic their biological activities, J. Saudi Chem. Soc. 18 (2014) 535–540.
acid derivatives as peptide deformylase inhibitors and potential antibacterial [37] B.S. Kuarm, Y.T. Reddy, J.V. Madhav, P.A. Crooks, B. Rajitha, 3-[Benzimidazo-
agents, Bioorg. Med. Chem. Lett. 14 (2004) 5997–6000. and 3-[benzothiadiazole imidazo-(1,2-c) quinazolin-5-yl]-2H-chromene-2-ones
[11] S. Yang, W. Shi, D. Xing, Z. Zhao, L.Y F.Lv, Y. Yang, W. Hu, Synthesis, antibac- as potent antimicrobial agents, Bioorg. Med. Chem. Lett. 21 (2011) 524–527.
terial activity, and biological evaluation of formyl hydroxyamino derivatives [38] J. Mori, M. Iwashima, M. Takeuchi, H. Saito, A synthetic study on antiviral and
as novel potent peptide deformylase inhibitors against drug-resistant bacteria, antioxidative chromene derivative, Chem. Pharm. Bull. 54 (2006) 1–6, doi:10.
Eur. J. Med. Chem. 86 (2014) 133–152. 1248/cpb.54.391.

12
N.P. Mishra, S. Mohapatra, C.R. Sahoo et al. Journal of Molecular Structure 1246 (2021) 131183

[39] P. Sivaguru, R. Sandhiya, M. Adhiyaman, A. Lalitha, Synthesis and antioxidant chromene-fused pyrrole derivatives through multicomponent reaction and
properties of novel 2H-chromene-3-carboxylate and 3-acetyl-2H-chromene evaluation of antibacterial activity with molecular docking investigation, J.
derivatives, Tetrahedron Lett 57 (2016) 2496–2501. Heterocycl. Chem. 57 (2) (2020) 575–589.
[40] S.T. Chung, W.H. Huang, C.K. Huang, F.C. Liu, R.Y. Huang, C.C. Wu, [50] N. Baral, S. Mohapatra, B.P. Raiguru, N.P. Mishra, P. Panda, S. Nayak, S.K. Pandey,
A.R. Lee, Synthesis and anti-inflammatory activities of 4H-chromene P.S. Kumar, C.R. Sahoo, Microwave-assisted rapid and efficient synthesis of new
and chromeno[2,3-b]pyridine derivatives, Res. Chem. Intermed. 42 (2016) series of chromene-based 1,2,4-oxadiazole derivatives and evaluation of an-
1195–1215. tibacterial activity with molecular docking investigation, J. Heterocycl. Chem.
[41] M. Mashhadinezhad, M. Mamaghani, M. Rassa, F. Shirini, A facile green syn- 56 (2) (2019) 552–565.
thesis of chromene derivatives as antioxidant and antibacterial agents through [51] C.R. Sahoo, R. Patro, J. Sahoo, R.N. Padhy, S.K. Paidesetty, Design, molecular
a modified natural soil, ChemistrySelect 4 (2019) 4920–4932. docking of synthesized schiff-based thiazole/pyridine derivatives as potent an-
[42] I. Zghab, M.B.M B.Trimeche, M. Hassine, D. Touboul, H.B. Jannet, Regiospe- tibacterial inhibitor, Indian Drugs 56 (11) (2019) 20–25.
cific synthesis, antibacterial and anticoagulant activities of novel isoxazoline [52] P. Panda, S. Nayak, S.K. Sahoo, S. Mohapatra, D. Nayak, R. Pradhan,
chromene derivatives, Arab. J. Chem. 10 (2017) S2651–S2658. C.N. Kundu, Diastereoselective synthesis of novel spiroindanone fused
[43] A.H. Halawa, M.M. Elaasser, A.M. El Kerdawy, A.M.A.I. Abd El-Hady, pyrano[3,2-c]chromene derivatives following hetero-diels-alder reaction and in
H.A. Emam, A.M. El-Agrody, Anticancer activities, molecular docking and vitro anticancer studies, RSC Adv. 8 (2018) 16802–16814.
structure–activity relationship of novel synthesized 4H-chromene, and [53] S. Nayak, S. Chakroborty, S. Bhakta, P. Panda, S. Mohapa-
5H-chromeno[2,3-d]pyrimidine candidates, Med. Chem. Res. 26 (2017) tra, S. Kumar, P.K. Jena, C. Purohit, Design and synthesis of
2624–2638. (E)-4-(2-Phenyl-2H-chromen-3-yl)but-3-en-2-ones and evaluation of their
[44] S. Kumar, N. Sharma, I.K. Maurya, A.K. Bhasin, N. Wangoo, P. Brandão, in vitro antimicrobial activity, Lett. Org. Chem. 12 (2015) 352–358.
K.K.B V.Félix, R.K. Sharma, Facile synthesis, structural evaluation, antimicrobial [54] J. Zhang, C. Lou, Z. Hu, M. Yan, Organocatalytic conjugate addition of nitroalka-
activity and synergistic effects of novel imidazo[1,2-a]pyridine based organose- nes to 2H-chromene-3-carbaldehydes: synthesis of highly functionalized chro-
lenium compounds, Eur. J. Med. Chem. 123 (2016) 916–924. maffin derivatives, Arkivoc (2009) 362–375.
[45] Y.D. Bodke, S.A. Biradar, R. Kenchappa, Catalyst free synthesis of 2-(3, [55] S. Santra, S. Mitra, A.K. Bagdi, A. Majee, A. Hajra, Iron(III)-catalyzed three-com-
4-dichlorophenyl)-1, 8a-dihydroimidazo [1, 2-a] pyridine derivatives as potent ponent domino strategy for the synthesis of imidazo[1,2-a]pyridines, Tetrahe-
antimicrobial agents, J. Chem. Chem. Sci. 6 (4) (2016) 269–276. dron Lett. 55 (2014) 5151–5155.
[46] G. Rajitha, V. Ravibabu, G. Ramesh, B. Rajitha, Synthesis and antimicro- [56] A.K. Bishoyi, C.R. Sahoo, A.P. Sahoo, R.N. Padhy, Bio-synthesis of silver nanopar-
bial activity of novel imidazo[1,2-a]pyridinopyrimidine-2,4,6(1H,3H,5H)-triones ticles with the brackish water blue-green alga Oscillatoria princeps and an-
and thioxopyrimidine-4,6(1H,5H)diones, Res. Chem. Intermed. 42 (2016) tibacterial assessment, Appl. Nanosci. 11 (2021) 389–398.
1989–1998. [57] C.P. Mandhata, C.R. Sahoo, C.S. Mahanta, R.N. Padhy, Isolation, biosynthe-
[47] S. Kuthyala, M.K Shankar, G.K Nagaraja, S.C.X.R. Synthesis, Hirshfeld and an- sis and antimicrobial activity of gold nanoparticles produced with ex-
timicrobial evaluation of some new imidazopyridine nucleus incorporated with tracts of Anabaena spiroides, Bioproc. Biosyst. Eng. (2021), doi:10.1007/
oxadiazole scaffold, ChemistrySelect 3 (2018) 12894–12899. s00449- 021- 02544- 4.
[48] A.K.J N.Devi, V. Singh, Assessment of novel pyrazolopyridinone fused imida- [58] C.R. Sahoo, S. Moharana, C.P. Mandhata, A.K. Bishoyi, S.K. Paidesetty,
zopyridines as potential antimicrobial agents, Karbala. Int. J. Mod. Sci. 4 (1) R.N. Padhy, Biogenic silver nanoparticle synthesis with cyanobacterium
(2018) 164–170. chroococcus minutes isolated from Baliharchandi sea- mouth, Odisha, and in
[49] N. Baral, D.R. Mishra, N.P. Mishra, S. Mohapatra, B.P. Raiguru, P. Panda, S. Nayak, vitro antibacterial activity, Saud. J. Biol. Sci. 27 (6) (2020) 1580–1586.
M. Nayak, P.S. Kumar, Microwave-assisted rapid and efficient synthesis of

13

You might also like