You are on page 1of 18

Journal of Molecular Structure 1292 (2023) 136184

Contents lists available at ScienceDirect

Journal of Molecular Structure


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

Exploration of quinoxaline-benzimidazole hybrids as apoptosis-inducing


agents and tubulin polymerisation inhibitors
Ojaswitha Ommi a, Shrilekha Chilvery b, Priyanka Sudhir Dhopat a, Anamika Sharma b,
Harshada Anil Bhalerao c, Srinivas Reddy Dannaram c, Srinivas Nanduri a, Rajesh Sonti c, *,
Chandraiah Godugu b, *, Venkata Madhavi Yaddanapudi a, *
a
Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
b
Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
c
Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India

A R T I C L E I N F O A B S T R A C T

Keywords: We herein report the design and synthesis of a new class of quinoxaline-benzimidazole hybrids based on the
Quinoxaline molecular hybridization concept and evaluation of their in vitro cytotoxicity profile against human cancer cell
Benzimidazole lines, i.e., colorectal, lung, skin, and mouse melanoma cell line. Among all the screened compounds, compound
Tubulin polymerisation
5l (2-(1-(3, 4-dichlorobenzyl)-1H-benzo[d]imidazol-2-yl)quinoxaline) exhibited potent cytotoxicity against lung
Cytotoxicity
Apoptosis
cancer cell line (A549) with an IC50 of 4.37 ± 0.09 µM with a cytospecificity towards cancer cells. Further, AO/
Molecular docking EB, DAPI, DCFDA, and JC-1 staining techniques confirmed that 5l induced apoptosis in the A549 cell line. In
addition, 5l in annexin V-FITC/PI assay induced early apoptosis. The clonogenic assay revealed that 5l inhibited
colony formation in a dose-dependent manner. Cell cycle distribution analysis unveiled that 5l caused the arrest
of G2/M phase of cell cycle. Moreover, 5l inhibited tubulin polymerization with an IC50 value of < 2.19 µM.
Docking studies revealed that 5l has a prominent binding affinity towards colchicine binding site of α/β- tubulin
with good protein-ligand interactions with a binding energy of -45.139 kcal/mol. In silico ADME/T prediction
studies disclosed the favourable drug-like properties of 5l.

1. Introduction inhibitors with diverse heterocyclic backbones, mainly those targeting


colchicine binding site, have attracted much attention because of their
Cancer prevails as the primary cause of death globally, amounting to least susceptibility to p-gp mediated drug resistance [11–16].
nearly 10 million deaths in 2020 and an estimated 1958,310 new cancer Quinoxalines are proven heterocyclic rings that demonstrated anti­
cases and 609,820 cancer deaths to occur in 2023 [1–3]. Furthermore, in cancer potential through various mechanisms (kinase, tubulin poly­
2040, an increase in cancer cases of ~29.5 million per year and cancer merisation, and topoisomerase inhibitors) [17–21] Some of the
related deaths to 16.4 million is predicted [3]. Despite chemotherapy quinoxaline based tubulin polymerisation inhibitors (A-C) are given in
being the most offered treatment option, it suffers complications such as Fig. 1 [18,22–25]. Also, discrete pharmacological activities of qui­
off-target side effects and drug resistance [4–6]. Therefore, developing noxalines are well documented [26–29]. Furthermore, quinoxaline
newer antineoplastic agents with improved potency, high target speci­ moiety is part of several FDA-approved drugs and a few ongoing clinical
ficity, and least resistance is required [7,8]. Microtubules are regarded trials with diverse therapeutic activities [30]. On the other hand,
as one of the critical targets among the several cancer-inhibiting targets benzimidazole, a bioisostere of purine, has attained “privileged scaffold”
since they are crucial for cell division and functionality. Microtubules status owing to its presence in many breakthrough drugs with unique
are polymers comprised of heterodimers of α- and β- tubulins in a pharmacological profile [31–36]. There have been tremendous reports
head-to-tail fashion [9]. They maintain dynamic equilibrium through manifesting the anticancer profile of benzimidazole hybrids [36–40].
continuous polymerization and depolymerization, and any disruption in Nocodazole D [41,42], Denibulin E [43,44], and BAL27862 F [45]
this process leads to cell apoptosis [10]. Tubulin polymerization (Fig. 1) are among the several benzimidazole-based anticancer drugs

* Corresponding authors.
E-mail addresses: rajesh.sonti@niperhyd.ac.in (R. Sonti), chandra.niperhyd@gov.in (C. Godugu), yvmadhavi.niperhyd@gov.in (V.M. Yaddanapudi).

https://doi.org/10.1016/j.molstruc.2023.136184
Received 15 May 2023; Received in revised form 5 July 2023; Accepted 9 July 2023
Available online 10 July 2023
0022-2860/© 2023 Elsevier B.V. All rights reserved.
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

that act via tubulin polymerization inhibition [39]. Molecules bearing benzyl, triazole, and amino alcohol) with a primary goal of finding
N-benzylated benzimidazoles have been explored extensively for their potent molecules with an improved therapeutic profile. Molecular hy­
anticancer properties. Telmisartan G, a well-established antihyperten­ bridization involves the amalgamation of two or more pharmacologi­
sive drug containing an N-benzylated benzimidazole core, is reported to cally essential subunits into a single architecture with improved activity
exhibit anticancer activity [46]. Tsung-Chieh Shih et al., demonstrated than the parent pharmacophores [45].
the synergistic activity of LLS30 H(N-benzylated benzimidazole deriv­
ative) in combination with docetaxel and its ability to inhibit the pro­ 2. Results and discussion
gression of prostate cancer cells in vivo [47,48]. On the other hand,
Triazole, a bioisostere of amide and many others, offers metabolic sta­ 2.1. Chemistry
bility and H-bond forming capacity to the molecule and is known to
exhibit anticancer activity on different cancer cell lines when linked to A library of N-substituted 2-(1H-benzo[d]imidazol-2-yl)quinoxaline
benzimidazoles [49–52]. Goud et al., reported the anticancer potential derivatives comprising 32 compounds was synthesized as outlined in
of Benzimidazole-triazole hybrids, and the potent compound I has Scheme 1. Condensation between o-phenylenediamine 1 and pyr­
shown significant growth inhibition against lung cancer (A549) cells uvaldehyde(methylglyoxal) in water gave the intermediate methyl­
with an IC50 value of 0.63 ± 0.21 µM [53]. β-amino alcohols are one of quinoxaline 2 with 85% yield [66,67]. Oxidation of intermediate 2 using
the important structural components in many potential therapeutics (e. selenium dioxide in the presence of a catalytic amount of tert‑butyl
g., Ethambutol, Phenylephrine, and Propranolol, to name a few). They hydroperoxide resulted in quinoxaline-2-aldehyde 3 in excellent yields
are known to exhibit various biological activites such as antimicrobial [67]. Condensation of thus obtained aldehyde 3, with o-phenylenedi­
[54,55], antimalarial [56,57], antihypertensive [58,59], anticancer amine using sodium metabisulphite via the formation of the
[60–64] and as α- and β-adrenergic agonists [65]. Baker, J.R., and group aldehyde-bisulfite adduct, afforded the key intermediate 4 in good yield
reported anticancer properties of aminoalchol acrylonitriles, among [68,69]. Synthesis of N-benzylated derivatives 5a-q was achieved by
which compound J showed selectivity towards lung cancer cell line treating 4 with various substituted benzyl halides using sodium hy­
(Fig. 1) [64]. droxide as a base in DMF at room temperature with excellent yields of
Thus, considering the potential antiproliferative effects of quinoxa­ 90–95%. N-propargylated intermediate 6 was prepared by the reaction
line and benzimidazole moieties as tubulin polymerization inhibitors, of 4 with propargyl bromide employing sodium hydroxide as the base in
we designed quinoxaline-benzimidazole hybrids based on the molecular acetone upon heating [70,71]. A series of aromatic azides were syn­
hybridization concept (Fig. 2) by incorporating 3 different fragments (N- thesized from the respective amines following reported procedures using

Fig. 1. Representative examples of quinoxaline and benzimidazole containing tubulin polymerization inhibitors (A–F) and reported anticancer agents containing N-
benzyl (G, H), triazole (I), and amino alcohol (J) motifs.

2
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 2. Illustration of design rationale: Representative examples of quinoxaline (A–C) and benzimidazole (D, E) containing tubulin polymerization inhibitors and
design of quinoxaline-benzimidazole hybrids (5a-q, 8a-l, and 9a-f) via molecular hybridization approach.

sodium nitrite in aqueous HCl to give diazonium salt, which is later the D4 proton gave all the scalar coupled proton network of the D ring.
displaced by sodium azide [72]. Cycloaddition of the alkyne component Further, 2D 1H–13C HSQC provided the aryl carbon assignments of the A,
6 with different aromatic azides in the presence of copper sulfate and B and D rings. The 1D 13C spectra of the analogous compounds are
sodium ascorbate in DMF-water solvent mixture at room temperature assigned easily by stacking the spectra and identifying merged signals
yielded the respective triazole derivatives 8a-i in 85–90% yields [70]. through compound 4, 5 h and 5l (Fig. 3).
The reaction of intermediate 4 with epichlorohydrin using sodium hy­
droxide as a base in acetone upon heating accomplished intermediate 7 2.3. Pharmacology
with 82% yield [73]. Further, the ring opening of epoxide with various
aliphatic amines in DMF under microwave irradiation resulted in the 2.3.1. In vitro cytotoxicity study
formation of amino alcohol derivatives, 9a-f, in 60–80% yields [74,75]. The synthesized N-substituted 2-(1H-benzo[d]imidazol-2-yl)qui­
noxaline derivatives 5a-q, 8a-i, and 9a-f along with the standard
2.2. NMR study colchicine were evaluated for their in vitro cytotoxicity potential against
a panel of human cancer cell lines such as HCT116 (Human Colorectal
Structures of the key intermediate (4, 6, and 7) and final compounds Carcinoma), A549 (Human Lung Adenocarcinoma), SK-ML (Human
(5a-q, 8a-i and 9a-g) were confirmed by 1H, 13C NMR and HRMS. The Melanoma) along with one mouse cancer cell line B16F10 (Murine
aromatic carbons (C–H) of the representative compound i.e., 4 (inter­ Melanoma) by employing MTT (3-(4,5-dimethylthiazol-2-yl)− 2,5-
mediate) are specifically and unambiguously assigned. On compound 4, diphenyltetrazolium bromide) assay. The IC50 values from the in vitro
we have performed selective 1D ROESY by irradiating the NH (C1) cytotoxicity study are presented in Table 1.
resonance that resulted in an ROE to the D4. A 1D selective TOCSY on Detailed investigation of these results revealed that N-benzylated

3
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Scheme 1. Synthetic route to quinoxaline-benzimidazole hybrids 5a-q, 8a-i and 9a-f.

series 5a-q exhibited more promising results than the triazole 8a-i and significant activity in the SK-ML cell line with an IC50 value of 7.61 ±
amino alcohol 9a-f derivatives. Dihalo benzyl derivatives 5f, 5 g, and 5l 0.50 µM. These observations are summarized in Fig. 4.
displayed IC50 of < 50 µM against all tested cancer cell lines, particularly Several reports have described the cytotoxic properties of quinoxa­
compound 5l showed the highest cytotoxicity of IC50 4.37 ± 0.09 µM line, benzimidazole-based compounds. In our study, a combination of
against the A549 cell line. Also, compound 5l exhibited cytotoxicity quinoxaline and N-benzylated benzimidazole resulted in compounds
against two other tested cancer cell lines, HCT116 and SK-ML, with IC50 with good cytotoxicity. 3,4-diCl phenyl substitution has been found in
values of 7.13 ± 0.19 µM and 6.25 ± 0.59 µM, respectively. Interest­ some anticancer agents such as H (LLS30) [47,48] and J [64] (Fig. 1),
ingly, the IC50 value of 5l against two regular human cell lines, HEK-293 wherein 3,4-dihalobenzyl substitution has shown excellent cytotoxic
(Human Embryonic Kidney) and HaCaT (Human Keratinocyte) was activity, which can be correlated to our lead compound 5l. Based on the
found to be 16.98 ± 0.41 µM and 16.52 ± 0.23 µM, respectively. On the observed cytotoxicity results, 5l was chosen for further studies in the
A549 cell line, the IC50 value was 4.37 ± 0.09, indicating cytospecificity A549 cell line.
of compound 5l towards cancer cell lines.
The results disclose that most benzyl derivatives with halogens 2.3.2. Determination of apoptosis
(electron-withdrawing groups) demonstrated better activity than those The compound 5l was further evaluated for apoptosis induction
substituted with electron-donating groups (Me, OMe, iPr) or without any ability by performing various morphological and quantitative assays on
substitution. Compound 5k with trifluoromethyl group expressed se­ the A549 cell line.
lective cytotoxicity against two cancer cell lines HCT116 (IC50: 11.94 ±
0.76 µM), A549 (IC50: 10.32 ± 1.41 µM). Compounds 5c (NO2), 5j (CN) Phase contrast microscopy. Initially, A549 cells were treated with
displayed poor or no activity. Notably, triazole 8a-i and amino alcohol different concentrations of compound 5l (2.19, 4.37, and 8.74 µM) for
derivatives 9b-e exhibited moderate to low cytotoxicity against the 48 h and observed for morphological changes under a phase contrast
tested cancer cell lines. However, compound 9a retained its cytotoxicity microscope. Compound 5l displayed characteristic apoptotic features
with < 20 µM in all the tested cancer cell lines. Compound 9f showed like cell shrinkage, disruption of the cell membrane, and decrease in the

4
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 3. (a) 1D 1H 500 MHz NMR spectra in DMSO‑d6; (i) 1H NMR spectrum of compound 4. (ii) 1H NMR selective TOCSY spectrum illustrating the irradiation of the
D4 peak of compound 4. (iii) 1H NMR selective ROESY spectrum illustrating the irradiation of C1 peak of compound 4. (b) 2D 1H − 13C HSQC spectrum of the
aromatic region of compound-4. (c) 1D 13C overlay spectra of compound 4, 5H and 5 L illustrating the assignments of aryl carbons.

number of viable cells in a dose-dependent fashion, of which images concentrations of compound 5l and studied for apoptosis using DCFDA
were captured and presented in Fig. 5. stain. As evident from the results (Fig. 8), the intensity of green fluo­
rescence is directly proportional to the compound’s concentration,
Acridine orange/Ethidium bromide (AO/EB) staining. AO/EB assay is a implying that dose-dependent induction of apoptosis in the A549 cell
dual staining technique to differentiate between live, apoptotic, and line by compound 5l.
necrotic cells. Acridine orange enters cells with the intact cell membrane
(live and early apoptotic cells) and stains green. In contrast, ethidium 2.3.3. Flow cytometry analysis
bromide stains the cells whose cell membrane integrity is vanished (late
apoptotic and necrotic cells) as orange or red [75]. Results from this Effect on mitochondrial membrane potential(DΨ m). Mitochondrial mem­
assay revealed that control cells exhibited normal morphology and brane potential (DΨm) is the driving force for the synthesis of ATP in
appeared green in color. Nevertheless, cells treated with compound 5l mitochondria and for the transport of charged compounds that are
displayed apoptotic features like cell shrinkage, membrane blebbing, crucial for cell viability. Hence, maintenance of stability DΨm is a pre­
chromatin condensation, and the red appearance of late apoptotic and requisite for healthy cell functionality [78]. Loss of DΨm leads to
necrotic cells (Fig. 6). increased ROS levels and oxidative stress in mitochondria, eventually
leading to cell death, which occurs during apoptosis. JC-1 dye stains
4,6-diamino-2-phenylindole (DAPI) staining. DAPI is a fluorescent dye normal polarized mitochondria possessing J-aggregates red in color,
that binds to AT-rich DNA regions and visualizes chromatin condensa­ whereas depolarized mitochondria with J-monomers of apoptotic cells
tion or nuclear damage. Upon staining by DAPI, normal cells appear appear green. The effect of compound 5l at different concentrations
light blue, whereas apoptotic cells develop bright blue color [76]. In the (2.19, 4.37, and 8.74 µM) on DΨm in A549 cells was investigated using
current study, horse-shoe-shaped, bright, fragmented nuclei formation membrane-permeate JC-1 dye. The results revealed a significant in­
can be seen in compound 5l-treated A549 cells, indicating apoptosis; crease in green fluorescence (depolarised cells-J monomers) in a
however, control cells showed intact nuclei (Fig. 7). dose-dependent manner from 2.19 to 8.74 µM, indicating apoptosis
associated with mitochondria (Fig. 9).
2′,7′-Dichlorofluorescein diacetate (DCFDA) staining. DCFDA stain de­
termines the compound’s ability to generate intracellular reactive oxy­ Annexin V-FITC/Propidium iodide dual staining. Annexin V-FITC/Propi­
gen species (ROS), thereby inducing apoptosis. DCFDA oxidizes to 2′,7′- dium iodide (PI) assay is used for the quantification of apoptosis and also
Dichlorofluorescein in the presence of reactive oxygen species and to identify the apoptosis phase. Annexin V binds to externalized phos­
shows green fluorescence [77]. A549 cells were treated with varied phatidyl serine in apoptotic cells with intact cell membranes and stains

5
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Table 1 them positively. On the contrary, PIbinds to apoptotic and necrotic cells
In-vitro cytotoxicity data (IC50 values in µM)a for the synthesized quinoxaline- whose membrane integrity is lost. A549 cells were treated with varied
benzimidazole hybrids 5a-q, 8a-i, and 9a-f against different cancer cell lines. concentrations (2.19, 4.37, and 8.74 µM) of compound 5l and stained
Compound HCT116b A549c SK-MLd B16F10e with Annexin V-FITC/PI. The cells at different stages were represented
5a >50 32.09±6.01 >50 >50
on four quadrant graph, such as live cells (LL; FITC-/PI-), early apoptotic
5b 27.44±4.12 23.27±1.97 >50 36.53±1.06 cells (LR; FITC+/PI-), late apoptotic cells (UR; FITC+/PI+), and necrotic
5c >50 >50 >50 >50 cells (UL; FITC-/PI +). Results from Fig. 10 indicate early apoptosis, late
5d 31.7 ± 3.61 29.38±0.91 44.86±2.19 31.08±7.67 apoptosis, and necrosis induction at concentrations 2.19 and 4.37 µM by
5e 42.85±6.14
>50 >50 >50
compound 5l. Compound 5l promotes significant early apoptosis in
5f 27.10±2.17 18.69±0.08 18.75±1.31 34.78±2.20
5g 12.75±0.56 23.77±2.56 24.42±0.18 28.60±0.93 A549 cells at 2.19 µM. However, the increased cocnentration led to only
5h 16.97±0.96 >50 >50 >50 a slight increase in early apoptosis.
5i >50 24.11±0.29 >50 >50
5j 35.10±0.53 >50 >50 >50
Cell cycle analysis. Cell cycle analysis was performed on A549 cells after
5k 11.94±0.76 10.32±1.41 >50 >50
5l 7.13±0.19 4.37±0.09 6.25±0.59 16.64±1.51 treatment with compound 5l (2.19, 4.37, and 8.74 µM) for 48 h using
5m 20.95±0.70 >50 >50 20.17±0.18 flow cytometry to determine the phase of cell cycle arrest. Results from
5n 35.94±1.77 >50 >50 >50 Fig. 11 showed that exponential increase in cells at the G2/M phase after
5o 14.70±0.28 23.50±0.35 >50 17.13±0.11 treatment with compound 5l (69.2% at 2.19 µM and 90.9% at 4.37 µM)
5p 23.24±0.93 21.64±0.49
compared to the untreated control group (57.8%), indicating G2/M
>50 >50
5q >50 >50 >50 >50
8a >50 18.76±0.63 >50 33.85±2.76 phase of cell cycle arrest.
8b >50 38.88±3.93 >50 25.77±0.53
8c >50 22.61±0.36 47.6 ± 3.74 37.65±5.09 2.3.4. Clonogenic assay
8d 12.78±0.77 22.11±0.09 27.45±0.03 23.03±0.38
Clonogenic assay reveals the proliferative ability of a single cell to
8e >50 33.02±2.62 37.58±3.79 24.45±1.17
8f 40.86±2.85 >50 >50 >50 form colonies. Herein, the media containing A549 cells was treated with
8g 16.31±0.42 >50 >50 49.33±8.22 different concentrations of compound 5l (2.19, 4.37, and 8.74 µM) and
8h 25.90±7.26 23.07±1.33 50.39±2.98 >50 observed for the formation of colonies after one week. The cells were
8i >50 43.56±4.97 >50 >50 stained with crystal violet for the visualization of colonies. The results
9a 15.99±0.88 11.80±0.02 11.66±0.01 8.73±0.19
showed an apparent reduction in the number of colonies after one week
9b >50 >50 >50 >50
9c >50 >50 >50 >50 in a concentration-dependent manner (Fig. 12).
9d >50 >50 >50 >50
9e >50 >50 30.43±4.97 39.01±5.30 2.3.5. Effect on tubulin polymerization
9f 11.28±0.00 7.61±0.50 18.2 ± 0.23
>50
Compound 5l induced cell cycle arrest at the G2/M phase, typically
Colchicinef 0.55±1.86 1.03±1.37 11.08±1.77 1.70±0.55
associated with tubulin polymerization inhibition. Moreover, it is clear
a
50% inhibition concentration determined after 48 h of compound treatment; from the literature that quinoxaline and benzimidazole-based de­
Data indicate the mean values ± SEM of three independent experiments. rivatives show anticancer activity via tubulin polymerization inhibition.
b
Colorectal carcinoma,. Therefore, the potent compound 5l was evaluated for in vitro enzyme
c
Lung adenocarcinoma,.
d assay to determine the possible mechanism of action and to observe
Melanoma,.
e effects on cellular microtubules. The assay was performed by monitoring
Mouse melanoma, and.
f
Reference compound. changes in excitation wavelength (350 nm) at 37 οC for 1 h. The assay
was carried out at different concentrations (2.19, 4.37, and 8.74 µM) by
taking colchicine (1.04 µM) and paclitaxel (3 µM) as positive and
negative controls, respectively. Interestingly, compound 5l efficiently

Fig. 4. Structure-activity relationships (SAR) developed based on cytotoxic evaluation of compounds.

6
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 5. Morphological changes observed in A549 cells treated with compound 5l at three different concentrations (2.19, 4.37, and 8.74 µM) in comparison with the
untreated control and standard colchicine (1.03 µM) for 48 h.

Fig. 6. AO/EB dual staining in A549 cells treated with compound 5l at three different concentrations (2.19, 4.37, and 8.74 µM) compared with the untreated control
and standard colchicine (1.03 µM) for 48 h. Compound 5l treated cells exhibited apoptotic body formation, membrane blebbing, and cell shrinkage.

inhibited tubulin polymerization (50% inhibition at all the tested con­ displayed a hydrogen bond interaction with the ASN101. The phenyl
centrations) with an IC50 value of <2.19 µM in comparison to untreated ring of the quinoxaline showed π-cation interaction with the LYS254. In
control, DMSO (Fig. 13). Further, we also performed an immunofluo­ addition, several hydrophobic interactions were noticed between com­
rescence assay to investigate the effect of 5l on tubulin expression at pound 5l and the active site residues THR179, ALA180, LYS352,
IC50 concentration compared with colchicine and paclitaxel as stan­ ALA250, LYS254, LEU248, and ALA354. These interactions stabilize the
dards. As shown in Fig. 14, tubulin expression was suppressed by 5l and binding of the potent compound 5l to the colchicine binding pocket of
colchicine compared to the control, whereas the opposite trend was tubulin, thereby supporting the in vitro enzyme inhibition assay
observed with the paclitaxel. These results suggested the inhibition of (Fig. 15).
tubulin polymerization on treatment with 5l, supporting the in vitro
enzyme assay results. 2.5. In-silico ADME/T and prime MM/GBSA binding energy studies

2.4. Molecular docking QikProp program of Schrödinger software was used to study the
drug-likeness of the most potent compound 5l in terms of ADME/T
The most potent compound, 5l was docked with tubulin-colchicine- properties, and the results are given in Table 2. In silico studies revealed
soblidotin: stathmin-like domain complex (PDB ID: 3E22) [51,79,80] to that compound 5l follows Lipinski’s rule of five, and its physicochemical
determine the binding mode and type of interactions with the tubulin descriptor values are in the recommended range [82]. Further,
using Schrödinger Release, 2021 [81]. The nitrogen atom at the 4th MM-GBSA (Molecular mechanics generalized born surface area) bind­
position of the quinoxaline ring acted as a hydrogen bond acceptor and ing free energy calculations confirmed that compound 5l has excellent

7
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 7. Nuclear morphological changes in A549 cell line after DAPI staining. A549 cells were treated with compound 5l at three different concentrations (2.19, 4.37,
and 8.74 µM) compared to the untreated control and standard colchicine (1.03 µM) for 48 h. White arrows represent membrane blebbing and fragmentation of nuclei,
and red arrows represent horse-shoe-shaped nuclei.

Fig. 8. DCFDA staining in A549 cells treated with compound 5l at three different concentrations (2.19, 4.37, and 8.74 µM) compared with the untreated control and
standard colchicine (1.03 µM) for 48 h. Compound 5l treated cells showed a dose-dependent increase in the green fluorescence compared to the control indicating
ROS-mediated apoptosis induction.

binding capability with a binding energy value of − 45.139 kcal/mol. polymerization. Molecular modeling studies confirmed the binding of
compound 5l at the colchicine binding site of tubulin with a binding
3. Conclusion energy of − 45.139 kcal/mol. Hence, 5l generated out of molecular
hybridistion technique with excellent anticancer activity could be
In conclusion, a new series of quinoxaline-benzimidazole hybrids considered for further development in the discovery of potent anti-
were designed, synthesized, and evaluated for their cytotoxicity against cancer drugs.
human colorectal, lung, and skin cancer cell lines and one mouse mel­
anoma cell line by using an MTT assay. Among these hybrids, compound 4. Experimental section
5l exhibited the highest cytotoxicity on all the tested human cancer cell
lines. Compound 5l exhibited potent cytotoxicity on the A549 cell line All the chemicals, reagents, and solvents were obtained from com­
(IC50: 4.37±0.09 µM) and four-fold selectivity upon regular human cell mercial providers and were used as such without any further purifica­
lines (IC50 in HEK-293 and HaCaT was found to be 16.98±0.41 µM and tion. Reactions were monitored using TLC-MERCK pre-coated silica gel
16.52±0.23 µM, respectively). Different staining techniques (AO/EB, 60-F254 (0.5 mm) aluminum plates under UV light. Compounds were
DAPI, and DCFDA) demonstrated induction of apoptosis by compound purified by column chromatography using silica of 60–120 mesh. 1H and
13
5l in A549 cell lines with various apoptotic features like alteration in cell C NMR spectra were recorded on Bruker Avance 500 MHz spectrom­
size/shape, formation of horse-shoe shaped nuclei, as well as frag­ eter using tetramethyl silane (TMS) as the internal standard and by
mented bright nuclei. Further, flow cytometric analysis revealed that making samples in CDCl3 or DMSO‑d6. Chemical shifts are reported in
compound 5l triggered ROS generation, disrupting mitochondrial ppm and referenced to TMS (δ 0.00 for 1H NMR and 13C NMR) or sol­
membrane potential and the G2/M phase of cell cycle arrest. The vents used for NMR recording CDCl3 (δ 7.26 for 1H NMR and 77.2 13C
enzyme-based assay revealed that compound 5l inhibited tubulin NMR) or DMSO‑d6 (δ 2.50 for 1H NMR and 39.5 for 13C NMR). Spin

8
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 9. Effect on mitochondrial membrane potential. JC-1 staining in A549 cells treated with compound 5l at three different concentrations (2.19, 4.37, and 8.74 µM)
in comparison with the untreated control and standard colchicine (1.03 µM) for 48 h. Compared to the control, a dose-dependent increase in the green fluorescence
(J-monomers) and a decrease in the red fluorescence (J-aggregates) of compound 5l was observed.

Fig. 10. (A) Effect of compound 5l on apoptosis at three different concentrations (2.19, 4.37, and 8.74 µM) compared with the untreated control and standard for 48
h using annexin V-FITC/PI dual staining technique. (B) Statistical representation of percentage live, apoptotic and necrotic cells post-treatment with compound 5l,
colchicine, and control. Data was represented as the Mean ± SEM (n = 3) and analysed by one-way ANOVA following Tukey’s multiple comparison test. ***P <
0.001; **p<0.001; *p<0.05 vs. Control.

multiplicities for 1H NMR are described as s (singlet), brs (broad singlet), 4.1. Chemistry
d (doublet), dd (double doublet), t (triplet), and m (multiplet). Coupling
constant values are reported in hertz (Hz). For fluorine-containing 4.1.1. General procedure for the synthesis of intermediate, 2-(1H-Benzo[d]
compounds, the carbon-fluorine coupling is denoted as JCF. Pulse pro­ imidazol-2-yl)quinoxaline, 3
grams from BRUKER standard library were used to record the experi­ To a solution of o-phenylene diamine, 1 (1 equiv.) in water was
ments. All processing and analysis for compounds 4, 5H and 5 L were added pyruvaldehyde (1 equiv.) at room temperature and stirred for 30
done using Bruker TopSpin 3.6.3 software. HRMS was recorded on an min. After completion (checked by TLC), the reaction mixture was
Agilent quadrupole-time-of-flight (QTOF) mass spectrometer 6540 se­ extracted with EtOAc. Combined organic layers were washed with brine,
ries instrument and was performed in electrospray ionization (ESI) dried over Na2SO4, and finally evaporated to give methyl quinoxaline 2
techniques at 70 eV. The melting point was taken using the Stuart R as a brown oil (85%) which is used as such in the next step without any
SMP30 apparatus. Microwave reactions were conducted in Monowave further purification. Crude methyl quinoxaline was dissolved in dioxane,
300 single-mode microwave reactor (make-Anton Paar GmbH). Micro­ and selenium dioxide (1.5 equiv.) was added, followed by a catalytic
wave reactions were performed in SiC10 Silicon Carbide reaction vessels amount of TBHP. The reaction mixture was refluxed for 1.5 h. Thereafter
(10 mL) or reusable Pyrex vials (30 mL). the solvent was evaporated from the reaction mixture, and ethyl acetate
was added and subjected to celite filtration. Obtained filtrate was then

9
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 11. (A) Effect of different concentrations of compound 5l (2.19, 4.37, and 8.74 µM) on A549 cell cycle progression in comparison with the untreated control,
standard, and colchicine treated cells. (B) Graphical representation of quantification of cells at different phases of cell cycle. Data was represented as the Mean ± SEM
(n = 3) and analysed by one-way ANOVA following Tukey’s multiple comparison test. ***P < 0.00 vs. Control.

Fig. 12. (A) Effect of compound 5l (2.19, 4.37, and 8.74 µM) on colony forming ability of the A549 cell line. (B) & (C) Statistical representation of the effect of the
compound 5l on colony forming ability of the A549 cell line. Data was represented as the Mean ± SEM (n = 3) and analysed by one-way ANOVA following Tukey’s
multiple comparison test. ***P < 0.001; *p<0.05 vs. Control.

washed with brine, dried over Na2SO4, and concentrated under a vac­ collected using vacuum filtration and recrystallized using methanol to
uum. The residue was further purified by column chromatography to give the intermediate 4 in 85% yield.
give the intermediate aldehyde 3 in 90% yield. A mixture of aldehyde 3 Light brown solid; yield 85%; mp: 191–192 ◦ C; 1H NMR (500 MHz,
(1 equiv.), o-phenylene diamine (1 equiv.), and Na2S2O5 (1 equiv.) in DMSO‑d6) δ 13.46 (s, 1H), 9.82 (s, 1H), 8.23 – 8.16 (m, 2H), 7.98 – 7.91
DMF was stirred at rt for 30 mins. Upon completion, the reaction (m, 2H), 7.82 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 7.9 Hz, 1H), 7.34 (t, J =
mixture was poured onto crushed ice, and the resulting precipitate was 7.0 Hz, 1H), 7.29 (t, J = 7.0 Hz, 1H); 13C NMR (125 MHz, DMSO‑d6) δ

10
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Fig. 13. (A) Effect of compound 5l (2.19, 4.37, and 8.74 µM) on the tubulin polymerization. Colchicine and paclitaxel were used as positive and negativecontrols,
respectively. (B) Percentage inhibition of tubulin by compound 5l at various concentrations.

Fig. 14. Microscopic images of immunofluorescence-labeled tubulin in A549 cells treated with compound 5l at IC50 concentration (4.37 µM) showed decrease in
protein expression as colchicine compared to paclitaxel. Tubulin was stained red and nuclei was stained blue.

Fig. 15. (A) 3D interaction of compound 5l at the colchicine binding site of tubulin protein (PDB ID: 3E22); The yellow dotted line indicates H-bonding, and the
green dotted line indicates π-cation interaction. (B) 2D interaction of compound 5l, π-cation interaction is represented as red arrow, and hydrogen bonding is shown
with magneta arrow.

148.72, 144.02, 143.93, 143.55, 141.92, 140.99, 135.14, 131.14, followed by the addition of NaOH pellet(1 equiv.). The reaction mixture
130.74, 129.20, 128.87, 124.17, 122.43, 119.83, 112.35;HRMS-QTOF was stirred at rt for 5 h or till the TLC showed the disappearance of
(ESI): m/z calcd. for [M + H]+ C15H11N4 247.0983; found 247.0972. starting material. Afterward, the reaction mixture was poured onto
crushed ice. Thus the formed precipitate was isolated by vacuum
4.1.2. General procedure for the synthesis of 2-(1-(Substituted benzyl)− filtration and subsequently purified by column chromatography eluting
1H-benzo[d]imidazol-2-yl)quinoxalines, 5a-q at EtOAc: Hexane 3: 7.
To a solution of 2-(1H-benzo[d]imidazol-2-yl)quinoxaline, 4 (1
equiv.) in DMF was added substituted benzyl chloride (1 equiv.)

11
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

Table 2 (125 MHz, DMSO‑d6) δ 147.35, 146.04, 145.01, 142.77, 141.59,


QikProp prediction of drug-likeness and physicochemical properties of potent 140.55, 137.79, 137.30, 131.93(2C), 131.63, 131.59, 129.71, 129.50
compound 5l. (2C), 129.48, 125.13, 123.76, 120.81, 120.75, 111.83, 48.43; HRMS-
S. Descriptors Recommended Compound QTOF (ESI): m/z calcd. for [M + H]+ C22H16BrN4 415.0558; found
No. range 5l 415.0547
1. Molecular weight 130–725 405.285
2. H-bond donors 0–6 0 2-(1-(3-Chlorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5e). Yel­
3. H-bond acceptors 2–20 3.5 low solid; yield 92%; mp: 195–197 ◦ C; 1H NMR (500 MHz, CDCl3) δ
4. QP logP (O/W) (predicted octanol/ − 2.0 to 6.5 6.265
water partition coefficient)
10.05 (s, 1H), 8.18 – 8.14 (m, 1H), 8.02 – 7.97 (m, 2H), 7.83 – 7.76 (m,
5. SASA (Total solvent accessible surface 300–1000 698.987 2H), 7.46 – 7.37 (m, 3H), 7.30 (s, 1H), 7.23 – 7.18 (m, 2H), 7.09 (d, J =
area in Å2) 6.5 Hz, 1H), 6.25 (s, 2H); 13C NMR (125 MHz, CDCl3) δ 147.15, 145.96,
6. CNS − 2 to +2 1 144.43, 142.41, 141.87, 140.76, 139.19, 136.81, 134.68, 130.79,
7. QP log HERG (predicted IC50 value for <− 5 − 7.27
130.61, 130.07, 129.49, 129.37, 127.85, 126.98, 125.04, 124.80,
blockage of HERG K+ channels.)
8. QP log BB (predicted brain/blood − 3 to 1.2 0.277 123.78, 120.73, 110.52, 48.81; HRMS-QTOF (ESI): m/z calcd. for [M +
partition coefficient) H]+ C22H16ClN4 371.1063; found 371.1048
9. QP Caco (predicted apparent Caco-2 <25 poor, >500 3736.537
cell permeability in nm s− 1) great
2-(1-(2,4-Dichlorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5f).
10. Dipole moment 1 – 12.5 3.247
11. QP logKhsa (prediction of binding to − 1.5 to 1.5 1.126 Beige solid; yield 95%; mp: 180–182 ◦ C; 1H NMR (500 MHz, CDCl3) δ
human serum albumin) 10.05 (s, 1H), 8.13 (dd, J = 8.1, 1.4 Hz, 1H), 8.02 (dd, J = 6.8, 1.6 Hz,
12. QP Polrz (predicted polarizability in 13–70 45.83 1H), 7.92 – 7.88 (m, 1H), 7.81 – 7.73 (m, 2H), 7.51 (d, J = 2.1 Hz, 1H),
cubic angstroms) 7.45 – 7.37 (m, 3H), 6.98 (dd, J = 8.4, 2.1 Hz, 1H), 6.49 (d, J = 8.4 Hz,
13. QP logkp (predicted skin permeability) − 8 to − 1 − 0.446
14. Human oral absorption 1–3 1
1H), 6.28 (s, 2H); 13C NMR (125 MHz, CDCl3) δ 147.09, 145.69, 143.99,
15. Rule of five 4 1 142.08, 141.88, 140.79, 136.67, 133.76, 133.52, 132.63, 130.93,
16. TPSA/ PSA(Van der Waals surface area 7.0–200.0 36.582 130.67, 129.56, 129.41, 129.36, 127.80, 127.61, 125.31, 124.09,
of polar nitrogen and oxygen atoms 120.76, 110.22, 46.89; HRMS-QTOF (ESI): m/z calcd. for [M + H]+
and carbonyl carbon atoms.
C22H15Cl2N4 405.0673; found 405.0656
17. Number of rotatable bonds 0–15 2
18. Binding energy (-kcal/mol) − 45.139
2-(1-(2-Chloro-4-fluorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5
g). Orange solid; yield 92%; mp: 184–186 ◦ C; 1H NMR (500 MHz,
2-(1-(4-Isopropylbenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5a). CDCl3) δ 10.04 (s, 1H), 8.15 – 8.12 (m, 1H), 8.02 – 7.99 (m, 1H), 7.92 –
Off-white solid; yield 90%; mp: 175–177 ◦ C; 1H NMR (500 MHz, CDCl3) 7.89 (m, 1H), 7.80 – 7.73 (m, 2H), 7.45 – 7.41 (m, 1H), 7.45 – 7.41 (m,
δ 10.03 (s, 1H), 8.18 – 8.14 (m, 1H), 8.07 – 8.03 (m, 1H), 7.98 – 7.95 (m, 1H), 7.41 – 7.38 (m, 2H), 7.24 (dd, J = 8.3, 2.6 Hz, 1H), 6.73 (td, J = 8.4,
1H), 7.83 – 7.76 (m, 2H), 7.51 – 7.46 (m, 1H), 7.40 – 7.35 (m, 2H), 7.18 2.6 Hz, 1H), 6.55 (dd, J = 8.7, 5.9 Hz, 1H), 6.28 (s, 2H); 13C NMR (125
(d, J = 8.2 Hz, 2H), 7.11 (d, J = 8.2 Hz, 2H), 6.28 (s, 2H), 2.88 – 2.78 (m, MHz, CDCl3) δ 161.66 (d, J = 249.6 Hz), 147.12, 145.74, 144.03,
1H), 1.18 (s, 3H), 1.16 (s, 4H); 13C NMR (125 MHz, DMSO‑d6) δ 147.94, 142.06, 141.90, 140.82, 136.70, 132.62, 132.54, 130.88, 130.73 (d, J =
147.36, 146.13, 145.27, 142.87, 141.63, 140.59, 137.38, 135.52, 3.5 Hz), 130.62, 129.48 (d, J = 13.9 Hz), 128.06 (d, J = 8.8 Hz), 125.26,
131.53(2C), 129.70, 129.49, 127.37(2C), 126.92(2C), 124.95, 123.58, 124.05, 120.72, 116.99 (d, J = 25.0 Hz), 114.52 (d, J = 21.2 Hz),
120.69, 112.02, 48.60, 33.42, 24.15; HRMS-QTOF (ESI): m/z calcd. for 110.29, 46.75; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C22H15ClFN4
[M + H]+ C25H23N4 379.1922; found 379.1925. 389.0969; found 389.0958

2-(1-(4-Fluorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5b). Off- 2-(1-Benzyl-1H-benzo[d]imidazol-2-yl)quinoxaline (5 h). Yellow solid;


white solid; yield 92%; mp: 209–211 ◦ C; 1H NMR (500 MHz, CDCl3) δ yield 90%; mp: 195–197 ◦ C; 1H NMR (500 MHz, CDCl3) δ 10.09 (s, 1H),
10.04 (s, 1H), 8.19 – 8.15 (m, 1H), 8.02 – 7.96 (m, 2H), 7.83 – 7.77 (m, 8.20 – 8.17 (m, 1H), 8.06 – 8.00 (m, 2H), 7.85 – 7.78 (m, 2H), 7.51 –
2H), 7.47 – 7.43 (m, 1H), 7.41 – 7.38 (m, 2H), 7.26 – 7.22 (m, 3H), 6.96 7.48 (m, 1H), 7.45 – 7.39 (m, 2H), 7.31 – 7.29 (m, 1H), 7.28 – 7.22 (m,
(t, J = 8.7 Hz, 2H), 6.25 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 161.79 4H), 6.33 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 146.86, 145.58,
(d, J = 243.3 Hz), 147.29, 146.08, 145.07, 142.77, 141.57, 140.54, 144.64, 142.28, 141.06, 140.05, 137.79, 136.93, 131.09, 131.07,
137.27, 134.47(d, J = 2.8 Hz), 131.62, 131.59, 129.72, 129.47(d, J = 129.21, 128.99, 128.54(2C), 127.25, 126.77(2C), 124.54, 123.17,
2.8 Hz, 2C), 129.39, 125.08, 123.72, 120.72, 115.83 (d, J = 21.5 Hz, 120.21, 111.49, 48.41; HRMS-QTOF (ESI): m/z calcd. for [M + H]+
2C), 111.92, 48.21; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C22H17N4 337.1453; found 337.1448
C22H16FN4 355.1359; found 355.1363
2-(1-(4-(Tert‑butyl)benzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5i).
2-(1-(4-Nitrobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5c). Yel­ Off-white solid; yield 93%; mp: 220–223 ◦ C; 1H NMR (500 MHz, CDCl3)
low solid; yield 90%; mp: 192–194 ◦ C; 1H NMR (500 MHz, CDCl3) δ δ 10.00 (s, 1H), 8.17 – 8.13 (m, 1H), 8.07 – 8.03 (m, 1H), 7.96 – 7.91 (m,
10.05 (s, 1H), 8.16 (d, J = 8.7 Hz, 3H), 8.01 (d, J = 7.1 Hz, 1H), 7.88 (dd, 1H), 7.81 – 7.75 (m, 2H), 7.49 – 7.45 (m, 1H), 7.38 – 7.33 (m, 2H), 7.26
J = 8.1, 1.4 Hz, 1H), 7.83 – 7.75 (m, 2H), 7.46 – 7.39 (m, 4H), 7.39 (s, (d, J = 8.2 Hz, 2H), 7.19 (d, J = 8.4 Hz, 2H), 6.27 (s, 2H), 1.23 (s, 9H);
1H), 6.37 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 147.41, 147.13, 13
C NMR (125 MHz, DMSO‑d6) δ 150.21, 147.28, 146.15, 145.28,
146.36, 145.96, 144.81, 142.74, 141.56, 140.49, 137.34, 131.67, 142.82, 141.61, 140.57, 137.34, 135.16, 131.59(2C), 129.75, 129.50,
131.59, 129.69, 129.46, 128.28(2C), 125.30, 124.27(2C), 123.92, 127.15(2C), 125.81(2C), 124.98, 123.62, 120.69, 112.09, 48.45, 34.60,
120.84, 111.70, 48.77; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ 31.48; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C26H25N4 393.2079;
C22H16N5O2382.1304; found 382.1307 found 393.2067

2-(1-(4-Bromobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5d). 4-((2-(Quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl)methyl)benzonitrile


White solid; yield 93%; mp: 245–247 ◦ C; 1H NMR (500 MHz, CDCl3) δ (5j). Cream solid; yield 95%; mp: 232–234 ◦ C; 1H NMR (500 MHz,
10.02 (s, 1H), 8.17 – 8.14 (m, 1H), 7.98 – 7.96 (m, 2H), 7.82 – 7.74 (m, DMSO‑d6) δ 9.88 (s, 1H), 8.18 – 8.13 (m, 1H), 8.05 – 8.00 (m, 1H), 7.95
2H), 7.43 – 7.36 (m, 5H), 7.12 (d, J = 8.5 Hz, 2H), 6.22 (s, 2H); 13C NMR – 7.89 (m, 3H), 7.76 (t, J = 7.2 Hz, 3H), 7.46 – 7.38 (m, 4H), 6.40 (s, 2H);

12
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

13
C NMR (125 MHz, DMSO‑d6) δ 147.39, 145.98, 144.86, 144.25, 3H), 7.77 (d, J = 7.8 Hz, 1H), 7.45 – 7.36 (m, 2H), 7.26 (dd, J = 9.1, 3.1
142.72, 141.56, 140.48, 137.34, 133.03(2C), 131.66, 131.59, 129.67, Hz, 2H), 7.02 – 6.97 (m, 1H), 6.84 (t, J = 7.7 Hz, 1H), 6.35 (s, 2H); 13C
129.47, 128.05(2C), 125.26, 123.89, 120.81, 119.12, 111.72, 110.46, NMR (125 MHz, DMSO‑d6) δ 160.17 (d, J = 244.5 Hz), 147.45, 145.92,
48.86; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C23H16N5 362.1405; 144.99, 142.65, 141.52, 140.51, 137.49, 131.62(2C), 129.75 (d, J = 8.2
found 362.1416 Hz), 129.48 (d, J = 3.6 Hz), 128.46 (d, J = 4.1 Hz) 125.39 (d, J = 14.2
Hz), 125.18, 125.12, 125.10, 123.78, 120.77, 115.83 (d, J = 21.0 Hz),
2-(1-(4-(Trifluoromethyl)benzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline 111.66, 43.32; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C22H16FN4
(5k). Off-white solid; yield 90%; mp: 197–199 ◦ C; 1H NMR (500 MHz, 355.1359; found 355.1353
DMSO‑d6) δ 9.89 (s, 1H), 8.20 – 8.12 (m, 1H), 8.07 – 8.00 (m, 1H), 7.92
(d, J = 5.5 Hz, 3H), 7.75 (d, J = 7.7 Hz, 1H), 7.67 (d, J = 8.1 Hz, 2H), 2-(1-(3-Methylbenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5q).
7.48 (d, J = 8.0 Hz, 2H), 7.45 – 7.36 (m, 2H), 6.41 (s, 2H); 13C NMR Beige solid; yield 92%; mp: 173–175 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ
(125 MHz, DMSO‑d6) δ 147.38, 146.01, 144.93, 143.21, 142.76, 9.87 (s, 1H), 8.18 – 8.10 (m, 2H), 7.95 – 7.91 (m, 2H), 7.89 (d, J = 7.4
141.57, 140.50, 137.34, 131.62, 131.60 129.66, 129.47, 128.31 (d, J = Hz, 1H), 7.74 (d, J = 8.2 Hz, 1H), 7.42 – 7.34 (m, 1H), 7.13 (dd, J = 9.7,
31.7 Hz), 127.90, 125.97 (q, J = 3.8 Hz, 4C), 125.22, 123.83, 120.79, 5.3 Hz, 2H), 7.00 (d, J = 7.9 Hz, 2H), 6.29 (s, 2H), 2.18 (s, 3H); 13C NMR
111.76, 48.70; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C23H16F3N4 (125 MHz, DMSO‑d6) δ 147.41, 146.09, 145.16, 142.78, 141.56,
405.1327; found 405.1315 140.56, 138.18, 138.12, 137.43, 131.58(2C), 129.68, 129.49, 128.93,
128.41, 127.89, 125.00, 124.31, 123.63, 120.69, 111.99, 48.88, 21.42;
2-(1-(3,4-Dichlorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5l). HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C23H19N4 351.1609; found
Cream solid; yield 92%; mp: 214–216 ◦ C; 1H NMR (500 MHz, DMSO‑d6) 351.1600
δ 9.88 (s, 1H), 8.19 – 8.15 (m, 1H), 8.12 – 8.07 (m, 1H), 7.96 – 7.93 (m,
1H), 7.91 (d, J = 7.6 Hz, 1H), 7.78 (d, J = 7.8 Hz, 1H), 7.69 (d, J = 2.0 4.1.3. 2-(1-(Prop-2-yn-1-yl)− 1H-benzo[d]imidazol-2-yl)quinoxaline, 6
Hz, 1H), 7.54 (d, J = 8.4 Hz, 1H), 7.46 – 7.37 (m, 2H), 7.19 (dd, J = 8.4, A stirred solution of intermediate 3 (1 equiv.), NaOH (1 equiv.) and
2.0 Hz, 1H), 6.31 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 146.89, propargyl bromide (1.2 equiv.) in acetone were refluxed for 2 h. On
145.61, 144.44, 142.26, 141.13, 140.04, 139.14, 136.77, 131.21, completion, acetone was evaporated from the reaction mixture, to
131.15, 131.09, 130.84, 129.86, 129.19, 129.12, 129.04, 127.10, which ice-cold water was added later. The solid intermediate was
124.80, 123.42, 120.34, 111.31, 47.51; HRMS-QTOF (ESI): m/z calcd. filtered, extensively washed with ice-cold water, and dried under vac­
for [M + H]+ C22H15Cl2N4 405.0673; found 405.0651 uum which is then subjected to column chromartography to obtain
propargylated intermediate 6 as a white solid (65% yield).
2-(1-(4-Methoxybenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5 m). White solid; yield 65%; mp: 160–163 ◦ C; 1H NMR (500 MHz,
Off-white solid; yield 90%; mp: 187–189 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 9.90 (s, 1H), 8.34 – 8.29 (m, 1H), 8.27 – 8.24 (m, 1H), 8.08
DMSO‑d6) δ 9.87 (s, 1H), 8.20 – 8.14 (m, 2H), 7.97 – 7.92 (m, 2H), 7.87 – 8.00 (m, 2H), 7.94 (d, J = 8.0 Hz, 1H), 7.90 (d, J = 8.1 Hz, 1H), 7.53 (t,
(d, J = 8.1 Hz, 1H), 7.78 (d, J = 7.8 Hz, 1H), 7.42 – 7.33 (m, 1H), 7.25 (d, J = 8.1 Hz, 1H), 7.45 (t, J = 8.0 Hz, 1H), 6.05 (d, J = 2.4 Hz, 2H); 13C
J = 8.8 Hz, 1H), 6.81 (d, J = 8.8 Hz, 1H), 6.27 (s, 2H), 3.64 (s, 3H); 13C NMR (125 MHz, DMSO‑d6) δ 146.77, 145.91, 144.98, 142.63, 141.67,
NMR (125 MHz, DMSO‑d6) δ 158.96, 147.24, 146.11, 145.20, 142.81, 140.59, 136.61, 131.74, 131.67, 129.90, 129.52, 125.10, 123.86,
141.56, 140.55, 137.28, 131.54(2C), 130.08, 129.71, 129.47, 128.78 120.72, 111.83, 79.49, 75.79, 35.74; HRMS-QTOF (ESI): m/z calcd. for
(2C), 124.91, 123.57, 120.66, 114.40(2C), 112.03, 55.42, 48.25; HRMS- [M + H]+ C18H13N4 285.1140; found 285.1106
QTOF (ESI): m/z calcd. for [M + H]+ C23H19N4O 367.1558; found
367.1543 4.1.4. General procedure for the synthesis of 2-(1-((1-(Substituted
phenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d]imidazol-2-yl)
2-(1-(4-Methylbenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5n). Off- quinoxaline, 8a-h
white solid; yield 91%; mp: 177–179 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 2-(1-(prop‑2-yn-1-yl)− 1H-benzo[d]imidazol-2-yl)quinoxaline 6 (1
9.87 (s, 1H), 8.19 – 8.11 (m, 2H), 7.96 – 7.91 (m, 2H), 7.88 (d, J = 7.4 equiv.), and appropriate azide (1 equiv.) were dissolved in a mixture of
Hz, 1H), 7.75 (d, J = 7.6 Hz, 1H), 7.42 – 7.34 (m, 1H), 7.16 (d, J = 8.1 DMF: H2O (10: 0.5) followed by addition of CuSO4⋅5H2O (0.2 equiv.)
Hz, 2H), 7.06 (d, J = 8.0 Hz, 2H), 6.30 (s, 2H), 2.19 (s, 3H); 13C NMR and sodium ascorbate (0.4 equiv.). The resulting reaction mixture was
(125 MHz, DMSO‑d6) δ 147.33, 146.09, 145.19, 142.78, 141.56, stirred for 15 h at rt. After completion, the reaction mixture was poured
140.57, 137.39, 136.93, 135.21, 131.60(2C), 129.73, 129.57(2C), onto crushed ice and filtered using vacuum filtration. Obtained solids
129.49, 127.27(2C), 124.98, 123.62, 120.68, 112.02, 48.63, 21.04; were further purified by column chromatography eluting at EtOAc:
HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C23H19N4 351.1609; found Hexane 4: 6 to afford the target derivatives 8a-h in 85–90% yields.
351.1593
2-(1-((1-(3,4,5-Trimethoxyphenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-
2-(1-(3-Fluorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5o). Off- benzo[d]imidazol-2-yl)quinoxaline (8a). Off-white solid; yield 85%; mp:
white solid; yield 92%; mp: 252–254 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 212–214 ◦ C; 1H NMR (500 MHz, CDCl3) δ 10.08 (s, 1H), 8.22 – 8.18 (m,
9.88 (s, 1H), 8.19 – 8.15 (m, 1H), 8.12 – 8.08 (m, 1H), 7.96 – 7.92 (m, 1H), 8.12 (dd, J = 6.6, 2.9 Hz, 1H), 7.96 (d, J = 7.9 Hz, 1H), 7.91 (s, 1H),
2H), 7.91 (d, J = 7.6 Hz, 1H), 7.77 (d, J = 7.6 Hz, 1H), 7.45 – 7.36 (m, 7.87 – 7.82 (m, 2H), 7.80 (d, J = 8.3 Hz, 1H), 7.46 (t, J = 7.2 Hz, 1H),
1H), 7.32 (td, J = 8.1, 6.2 Hz, 1H), 7.17 (d, J = 10.1 Hz, 1H), 7.07 – 7.02 7.41 (t, J = 7.1 Hz, 1H), 6.81 (s, 2H), 6.45 (s, 2H), 3.83 (s, 4H); 13C NMR
(m, 2H), 6.35 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 162.66 (d, J = (125 MHz, DMSO‑d6) δ 153.86(2C), 147.30, 146.08, 145.21, 144.84,
243.5 Hz),147.35, 146.08, 145.02, 142.73, 141.57, 141.28, 141.23, 142.75, 141.62, 140.65, 137.90, 137.10, 132.83, 131.61, 131.55,
140.52, 137.32, 131.62 (d, J = 5.1 Hz), 131.09 (d, J = 8.3 Hz), 129.67, 129.93, 129.48, 124.96, 123.69, 122.35, 120.62, 112.06, 98.85(2C),
129.49, 125.16, 123.79, 123.20 (d, J = 2.5 Hz), 120.75, 114.58 (d, J = 60.62(2C), 56.72, 41.36; HRMS-QTOF (ESI): m/z calcd. for [M + H]+
20.9 Hz), 114.21 (d, J = 22.1 Hz), 111.86, 48.49; HRMS-QTOF (ESI): m/ C27H24N7O3 494.1940; found 494.1932
z calcd. for [M + H]+ C22H16FN4 355.1359; found 355.1342
2-(1-((1-(4-Chlorophenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d]
2-(1-(2-Fluorobenzyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (5p). Yel­ imidazol-2-yl)quinoxaline (8b). Light-brown solid; yield 88%; mp:
low solid; yield 90%; mp: 188–190 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 243–245 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 9.90 (s, 1H), 8.81 (s, 1H),
9.85 (s, 1H), 8.17 – 8.12 (m, 1H), 8.02 – 7.98 (m, 1H), 7.94 – 7.89 (m, 8.29 – 8.24 (m, 1H), 8.22 – 8.16 (m, 1H), 7.98 – 7.93 (m, 2H), 7.89 (dd,

13
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

J = 12.9, 8.1 Hz, 1H), 7.84 (d, J = 8.9 Hz, 1H), 7.59 (d, J = 8.9 Hz, 1H), 123.72, 122.49, 121.72, 120.61, 120.15 (q, J = 5.4 Hz), 118.97, 112.04,
7.44 (t, J = 8.1 Hz, 1H), 7.37 (t, J = 8.0 Hz, 1H), 6.48 (s, 2H); 13C NMR 41.33; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C25H16BrF3N7
(125 MHz, DMSO‑d6) δ 147.20, 146.08, 145.23, 145.23, 142.75, 550.0602; found 550.0606
141.64, 140.69, 137.07, 135.73, 133.44, 131.60, 131.55, 130.20(2C),
129.98, 129.46, 124.97, 123.71, 122.33(2C), 122.14, 120.59, 112.09, 2-(1-((1-(3,5-Difluorophenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo
41.25; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C24H17ClN7 [d]imidazol-2-yl)quinoxaline (8 h)*. Yellow solid; yield 88%; mp:
438.1234; found 438.1257 232–234 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 9.88 (s, 1H), 8.82 (s, 1H),
8.25 – 8.20 (m, 1H), 8.18 – 8.14 (m, 1H), 7.95 – 7.90 (m, 2H), 7.87 (d, J
2-(1-((1-(4-Fluorophenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d] = 8.2 Hz, 2H), 7.67 (dd, J = 8.0, 2.0 Hz, 1H), 7.43 (t, J = 8.1 Hz, 1H),
imidazol-2-yl)quinoxaline (8c). Cream solid; yield 90%; mp: 209–211 ◦ C; 7.39 – 7.31 (m, 2H), 6.46 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ
1
H NMR (500 MHz, DMSO‑d6) δ 9.90 (s, 1H), 8.77 (s, 1H), 8.29 – 8.25 163.20 (d, J = 246.9 Hz), 163.08 (d, J = 246.9 Hz), 147.11, 146.03,
(m, 1H), 8.21 – 8.17 (m, 1H), 7.98 – 7.93 (m, 2H), 7.89 (dd, J = 16.8, 145.57, 145.15, 142.75, 141.62, 140.65, 138.65 (q, J = 13.2 Hz),
8.1 Hz, 2H), 7.86 – 7.82 (m, 1H), 7.44 (t, J = 8.2 Hz, 1H), 7.41 – 7.35 (m, 137.03, 131.54, 131.48, 130.00, 129.42, 124.96, 123.69, 122.31,
3H), 6.47 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 162.07 (d, J = 245.4 120.60, 112.00, 104.53, 104.32 (d, J = 8.2 Hz, 2C), 41.33; HRMS-QTOF
Hz), 147.12, 146.08, 145.21, 145.07, 142.74, 141.62, 140.66, 137.05, (ESI): m/z calcd. for [M + H]+ C24H16F2N7 440.1435; found 440.1445.
133.49, 131.51 (d, J = 3.8 Hz, 2C), 129.98, 129.44, 124.92, 123.66,
122.98 (d, J = 8.8 Hz, 2C), 122.31, 120.58, 117.05 (d, J = 23.3 Hz, 2C), 2-(1-((1-(3,4-Dimethoxyphenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-
112.10, 41.23; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C24H17FN7 benzo[d]imidazol-2-yl)quinoxaline (8i). Cream solid; yield 88%; mp:
422.1529; found 422.1557. 234–236 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 9.89 (s, 1H), 8.80 (s, 1H),
8.23 (d, J = 35.6 Hz, 2H), 7.92 (d, J = 33.6 Hz, 4H), 7.41 (d, J = 30.4 Hz,
2-(1-((1-(4-Nitrophenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d]imi­ 2H), 6.97 (s, 2H), 6.56 (s, 1H), 6.46 (s, 2H), 3.77 (s, 6H); 13C NMR (125
dazol-2-yl)quinoxaline (8d). Pale yellow solid; yield 86%; mp: 262–264 MHz, DMSO‑d6) δ 161.56(2C), 147.22, 146.09, 145.23, 144.95, 142.75,

C; 1H NMR (500 MHz, DMSO‑d6) δ 9.65 (s, 1H), 8.72 (s, 1H), 8.12 (d, J 141.62, 140.66, 138.44, 137.09, 131.57, 131.52, 129.94, 129.47,
= 9.1 Hz, 2H), 8.00 (dd, J = 6.4, 3.2 Hz, 1H), 7.94 (dd, J = 6.4, 3.2 Hz, 124.94, 123.67, 122.20, 120.60, 112.08, 100.62, 98.96(2C), 56.14(2C),
1H), 7.88 (d, J = 9.1 Hz, 2H), 7.74 – 7.67 (m, 2H), 7.64 (t, J = 8.5 Hz, 41.32; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C26H22N7O2
2H), 7.20 (t, J = 7.5 Hz, 1H), 7.13 (t, J = 7.5 Hz, 1H), 6.25 (s, 2H); 13C 464.1835; found 464.1836
NMR (125 MHz, DMSO‑d6) δ 147.19, 147.16, 146.02, 145.77, 145.15,
142.69, 141.61, 141.13, 140.67, 137.02, 131.67, 131.61, 129.98, 4.1.5. General procedure for the synthesis of 2-(1-(Oxiran-2-ylmethyl)−
129.44, 125.91(2C), 125.05, 123.79, 122.46, 121.18(2C), 120.59, 1H-benzo[d]imidazol-2-yl)quinoxaline, 7
112.03, 41.23; HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C24H17N8O2 A mixture of 2-(1H-benzo[d]imidazol-2-yl)quinoxaline 4 (1 equiv.),
449.1474; found 449.1469 epichlorohydrin (2 equiv.), and sodium hydroxide (1 equiv.) in acetone
was refluxed for 30 min. Upon completion, the solvent was evaporated
2-(1-((1-(4-Methoxyphenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d] from the reaction mixture to which ice-cold water was added and
imidazol-2-yl)quinoxaline (8e). Off-white solid; yield 85%; mp: 214–216 filtered using vacuum filtration to obtain the epoxy intermediate 7 as a

C; 1H NMR (500 MHz, DMSO‑d6) δ 9.89 (s, 1H), 8.68 (s, 1H), 8.30 – yellow solid in 82% yield.
8.25 (m, 1H), 8.21 – 8.17 (m, 1H), 7.98 – 7.93 (m, 2H), 7.91 (d, J = 8.1 Cream solid; yield 82%; mp: 155–157 ◦ C; 1H NMR (500 MHz,
Hz, 1H), 7.87 (d, J = 8.0 Hz, 1H), 7.69 (d, J = 9.1 Hz, 1H), 7.44 (t, J = DMSO‑d6) δ 9.83 (s, 1H), 8.24 – 8.16 (m, 2H), 7.99 – 7.93 (m, 2H), 7.85
7.6 Hz, 1H), 7.37 (t, J = 7.2 Hz, 1H), 7.05 (d, J = 9.1 Hz, 1H), 6.45 (s, (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.1 Hz, 1H), 7.42 (t, J = 7.6 Hz, 1H),
2H), 3.78 (s, 3H); 13C NMR (125 MHz, DMSO‑d6) δ 159.71, 147.16, 7.36 (t, J = 7.1 Hz, 1H), 5.44 (dd, J = 15.0, 3.3 Hz, 1H), 4.92 (dd, J =
146.12, 145.27, 144.75, 142.74, 141.63, 140.68, 137.07, 131.56, 15.0, 5.8 Hz, 1H), 3.64 – 3.60 (m, 1H), 2.81 (dd, J = 5.0, 4.1 Hz, 1H),
131.54, 130.35, 129.98, 129.47, 124.91, 123.66, 122.28(2C), 122.04, 2.70 (dd, J = 5.0, 2.6 Hz, 1H); 13C NMR (125 MHz, DMSO‑d6) δ 147.70,
120.57, 115.22(2C), 112.16, 55.99, 41.25; HRMS-QTOF (ESI): m/z 146.16, 145.23, 142.63, 141.61, 140.62, 137.64, 131.56, 131.52,
calcd. for [M + H]+ C25H20N7O 434.1729; found 434.1755 129.89, 129.48, 124.80, 123.55, 120.48, 112.21, 51.34, 47.47, 45.49;
HRMS-QTOF (ESI): m/z calcd. for [M + H]+ C18H15N4O 303.1245;
2-(1-((1-(3-Chlorophenyl)− 1H-1,2,3-triazol-4-yl)methyl)− 1H-benzo[d] found 303.1248
imidazol-2-yl)quinoxaline (8f). yellow solid; yield 87%; mp: 197–199 ◦ C;
1
H NMR (500 MHz, DMSO‑d6) δ 9.89 (s, 1H), 8.84 (s, 1H), 8.27 – 8.22 4.1.6. General procedure for the synthesis of 1-(Substituted amino)− 3-(2-
(m, 1H), 8.20 – 8.16 (m, 1H), 7.96 – 7.94 (m, 1H), 7.93 (dd, J = 4.1, 2.1 (quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl) propan-2-ol, 9a-f
Hz, 2H), 7.90 – 7.86 (m, 2H), 7.82 (d, J = 11.1 Hz, 1H), 7.54 (t, J = 8.1 A mixture of epoxide (1 equiv.) and an excess of aliphatic amines (up
Hz, 1H), 7.51 – 7.47 (m, 1H), 7.44 (t, J = 8.1 Hz, 1H), 7.37 (t, J = 8.1 Hz, to 3 equiv.) in DMF were stirred in a closed microwave vessel at 180 ◦ C
1H), 6.47 (s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 147.11, 146.05, for 20 mins. After completion, the reaction mixture was cooled to room
145.29, 145.18, 142.75, 141.61, 140.65, 137.95, 137.04, 134.53, temperature, to which water was added and extracted with ethyl ace­
131.91, 131.51, 131.45, 129.97, 129.42, 128.90, 124.93, 123.65, tate. The organic layer was washed with brine, dried over Na2SO4, and
122.20, 120.59, 120.32, 119.15, 112.03, 41.31; HRMS-QTOF (ESI): m/z concentrated under reduced pressure. The residue was then purified by
calcd. for [M + H]+ C24H17ClN7 438.1234; found 438.1247. column chromatography using methanol and DCM (2: 8) as the eluent to
give the target derivatives 9a-g in 60 - 80% yields.
2-(1-((1-(4-Bromo-3-(trifluoromethyl)phenyl)− 1H-1,2,3-triazol-4-yl)
methyl)− 1H-benzo[d]imidazol-2-yl)quinoxaline (8 g). Brown solid; yield 1-Morpholino-3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl)propan-
85%; mp: 164–166 ◦ C; 1H NMR (500 MHz, DMSO‑d6) δ 9.89 (s, 1H), 2-ol (9a). Light yellow solid; yield 80%; mp: 239–241 ◦ C; 1H NMR (500
8.94 (s, 1H), 8.24 (dd, J = 6.2, 3.4 Hz, 1H), 8.21 (s, 1H), 8.18 (dd, J = MHz, DMSO‑d6) δ 9.80 (s, 1H), 8.19 (d, J = 8.1 Hz, 2H), 8.01 – 7.93 (m,
6.2, 3.4 Hz, 1H), 8.09 – 8.02 (m, 2H), 7.94 (dd, J = 6.3, 3.4 Hz, 2H), 7.88 2H), 7.83 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 8.1 Hz, 1H), 7.39 (t, J = 7.6
(d, J = 8.2 Hz, 2H), 7.44 (t, J = 7.6 Hz, 1H), 7.37 (t, J = 7.6 Hz, 1H), 6.48 Hz, 1H), 7.33 (t, J = 7.5 Hz, 1H), 5.13 (dd, J = 14.2, 3.7 Hz, 1H), 4.91 (d,
(s, 2H); 13C NMR (125 MHz, DMSO‑d6) δ 147.17, 146.05, 145.56, J = 5.3 Hz, 1H), 4.83 (dd, J = 14.1, 8.3 Hz, 1H), 4.22 – 4.15 (m, 1H),
145.17, 142.74, 141.63, 140.66, 137.02, 136.32, 131.61, 131.52, 3.46 – 3.41 (m, 3H), 2.49 – 2.40 (m, 2H), 2.32 (s, 4H); 13C NMR (125
130.04 (d, J = 31.2 Hz), 130.00, 129.45, 125.87, 124.99, 123.90, MHz, DMSO‑d6) δ 148.10, 146.36, 145.79, 142.71, 141.50, 140.63,

14
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

138.03, 131.52, 131.47, 129.70, 129.52, 124.32, 123.13, 120.29, 129.46, 124.69, 123.48, 120.51, 112.43, 66.99, 50.18, 49.55, 47.53,
112.69, 67.71, 66.50(2C), 63.16, 54.47(2C), 50.25; HRMS-QTOF (ESI): 31.57, 28.85(2C), 26.34, 25.67, 22.50, 14.40; HRMS-QTOF (ESI): m/z
m/z calcd. for [M + H]+ C22H24N5O2 390.1930; found 390.1939 calcd. for [M + H]+ C26H34N5O 432.2763; found 432.2771

1-(Piperazin-1-yl)− 3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl) 4.2. Pharmacology


propan-2-ol (9b). Off-white solid; yield 80%; mp: 170–172 ◦ C; 1H NMR
(500 MHz, DMSO‑d6) δ 9.81 (s, 1H), 8.22 – 8.17 (m, 2H), 8.01 – 7.95 (m, 4.2.1. Cell culture
2H), 7.94 (s, 1H), 7.83 (d, J = 8.0 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.40 The HCT116 (Human Colorectal Carcinoma), A549 (Human Lung
(t, J = 8.1 Hz, 1H), 7.33 (t, J = 8.1 Hz, 1H), 5.12 (dd, J = 14.2, 3.9 Hz, Adenocarcinoma), SK-ML (Human Melanoma), B16F10 (Murine Mela­
1H), 4.93 (d, J = 5.3 Hz, 1H), 4.84 (dd, J = 14.1, 8.4 Hz, 1H), 4.25 – 4.17 noma), HEK-293 (Human Embryonic Kidney), and HaCaT (Human
(m, 1H), 3.29 – 3.19 (m, 4H), 2.56 – 2.53 (m, 1H), 2.49 – 2.45 (m, 1H), Keratinocyte) cell lines, were obtained from National centre for Cell
2.36 (t, J = 4.9 Hz, 2H), 2.31 (t, J = 5.1 Hz, 2H); 13C NMR (125 MHz, Science, Pune, India. Cells were maintained in appropriate media
DMSO‑d6) δ 147.62, 145.90, 145.30, 142.23, 141.04, 140.17, 137.57, enriched with 10% fetal bovine serum (FBS) and stabilized with 1%
131.09, 131.02, 129.25, 129.06, 123.89, 122.69, 119.84, 112.23, 67.49, antibiotic-antimycotic solution (Sigma-Aldrich). Cells were incubated at
62.05, 53.95, 52.81, 49.79, 44.65, 38.85; HRMS-QTOF (ESI): m/z calcd. 37 ◦ C and subcultured after attaining 80–90% confluency using 0.25%
for [M + H]+ C22H25N6O 389.2089; found 389.2087 trypsin/1 mM EDTA solution for further passage. 10 mM stock solution
was prepared and diluted with respective media to obtain the required
1-(Piperidin-1-yl)− 3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl) concentrations for both compounds and standard by dissolving in
propan-2-ol (9c). Cream solid; yield 75%; mp: 194–195 ◦ C; 1H NMR DMSO.
(500 MHz, DMSO‑d6) δ 9.80 (s, 1H), 8.21 – 8.18 (m, 2H), 8.00 – 7.93 (m,
2H), 7.83 (d, J = 7.9 Hz, 1H), 7.77 (d, J = 8.1 Hz, 1H), 7.42 – 7.37 (m, 4.2.2. MTT assay
1H), 7.35 – 7.30 (m, 1H), 5.14 (dd, J = 14.2, 3.9 Hz, 1H), 4.78 (dd, J = MTT assay is a colorimetric, sensitive, and reliable study used to
14.1, 8.3 Hz, 2H), 4.15 (s, 1H), 2.45 – 2.35 (m, 2H), 2.28 (s, 4H), 1.40 – determine cell viability. Metabolically active cells reduce MTT 3-(4,5-
1.34 (m, 4H), 1.33 – 1.28 (m, 2H); 13C NMR (125 MHz, DMSO‑d6) δ dimethylthiazol-2-yl)− 2,5-diphenyl tetrazolium bromide) into insol­
148.16, 146.38, 145.84, 142.74, 141.52, 140.65, 138.06, 131.45, uble formazan, which is measured as the level of absorbance denoting
131.43, 129.67, 129.52, 124.27, 123.10, 120.29, 112.63, 67.83, 63.70, the viability of cells. Initially, cells were seeded in 96-well plates with
55.27(2C), 50.43, 25.95(2C), 24.37; HRMS-QTOF (ESI): m/z calcd. for the medium at a density of 1000–4000 per well in 100 µl and incubated
[M + H]+ C23H26N5O 388.2137; found 388.2176 overnight to grow and adhere. Then, the old media was replaced with
fresh media with different compound concentrations and incubated for
1-(Propylamino)− 3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl) 48 h. The treatment media was removed, and 100 µl of MTT (0.5 mg/
propan-2-ol (9d). White solid; yield 70%; mp: 224–226 ◦ C; 1H NMR mL) was added and incubated at 37 ◦ C for 4 h. Then the supernatant was
(500 MHz, DMSO‑d6) δ 9.87 (s, 1H), 8.49 (s, 1H), 8.35 – 8.31 (m, 1H), removed, and formazan crystals formed were solubilized by the addition
8.22 – 8.18 (m, 1H), 8.02 – 7.95 (m, 2H), 7.86 (dd, J = 16.8, 8.1 Hz, 2H), of 100 µL of DMSO. The quantity of formazan was measured using a
7.46 – 7.41 (m, 1H), 7.39 – 7.35 (m, 1H), 5.91 (d, J = 5.4 Hz, 1H), 5.09 multimode plate reader (Perkin Elmer, USA) at 570 nM wavelength.
(dd, J = 14.2, 4.3 Hz, 1H), 4.90 (dd, J = 14.2, 8.1 Hz, 1H), 4.50 (s, 1H),
3.26 (dd, J = 12.6, 2.4 Hz, 1H), 3.12 (dd, J = 12.6, 10.1 Hz, 1H), 2.88 (t, 4.2.3. Phase contrast microscopy
J = 7.9 Hz, 1H), 1.66 – 1.55 (m, 2H), 0.87 (t, J = 7.4 Hz, 3H); 13C NMR A549 cells were plated at a concentration of 40,000–50,000 cells/
(125 MHz, DMSO‑d6) δ 147.78, 146.28, 145.34, 142.72, 141.58, well in a 24-well plate, and different concentrations of compound 5l
140.60, 137.93, 131.60, 131.43, 130.13, 129.45, 124.67, 123.47, were added. Treated plates were incubated in a CO2 incubator for 48 h.
120.52, 112.40, 66.99, 50.22, 49.55, 49.19, 19.27, 11.32; HRMS-QTOF Later, cells were visualized using a microscope (Nikon, Inc. Japan)
(ESI): m/z calcd. for [M + H]+ C21H24N5O 362.1980; found 362.2023 under a bright field and captured the observed morphological changes at
20X magnification.
1-(Pentylamino)− 3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl)
4.2.4. Acridine orange/ethidium bromide (AO/EB) staining
propan-2-ol(9e). White solid; yield 60%; mp: 180–182 ◦ C; 1H NMR (500
MHz, DMSO‑d6) δ 9.81 (s, 1H), 8.24 – 8.18 (m, 2H), 7.99 – 7.93 (m, 2H), A549 cells were plated at a concentration of 40,000–50,000cells/
well in a 24-well plate, and different concentrations of compound 5l
7.82 (dd, J = 18.1, 8.0 Hz, 2H), 7.42 – 7.37 (m, 1H), 7.36 – 7.32 (m, 1H),
5.10 (dd, J = 14.1, 4.0 Hz, 1H), 4.83 (dd, J = 14.1, 8.2 Hz, 1H), 4.16 – were added. Treated plates were incubated in a CO2 incubator for 48 h,
followed by the addition of 10 µL of Acridine Orange and Ethidium
4.11 (m, 1H), 2.71 (dd, J = 11.9, 4.6 Hz, 1H), 2.62 (dd, J = 11.9, 6.7 Hz,
1H), 2.49 – 2.46 (m, 2H), 1.90 (s, 2H), 1.40 – 1.34 (m, 2H), 1.26 – 1.20 Bromide of 10 μg/ml each was added respectively. Cells were visualized
using a fluorescence microscope (Nikon, Inc. Japan with excitation (488
(m, 4H), 0.84 (t, J = 7.0 Hz, 3H); 13C NMR (125 MHz, DMSO‑d6) δ
148.01, 146.30, 145.64, 142.70, 141.52, 140.66, 137.98, 131.46, nm) and emission (550 nm) and images were captured at 20X
magnification.
131.40, 129.80, 129.48, 124.36, 123.19, 120.33, 112.63, 69.87, 53.65,
50.05, 49.82, 29.45, 22.51, 21.66, 14.39; HRMS-QTOF (ESI): m/z calcd.
4.2.5. 4,6-diamino-2-phenylindole (DAPI) staining
for [M + H]+ C23H28N5O 390.2293; found 390.2344
A549 cells were plated at a concentration of 40,000–50,000 cells/
well in a 24-well plate, and different concentrations of compound 5l
1-(Octylamino)− 3-(2-(quinoxalin-2-yl)− 1H-benzo[d]imidazol-1-yl)
were added. Treated plates were incubated in a CO2 incubator for 48 h,
propan-2-ol (9f). White solid; yield 60%; mp: 193–195 ◦ C; 1H NMR (500
followed by washing with PBS and fixed with 4% paraformaldehyde for
MHz, DMSO‑d6) δ 9.87 (s, 1H), 8.45 (s, 1H), 8.35 – 8.32 (m, 1H), 8.23 –
15 min. After the removal of Paraformaldehyde, cells were per­
8.18 (m, 1H), 8.01 – 7.95 (m, 2H), 7.86 (dd, J = 13.7, 8.1 Hz, 2H), 7.47 –
meabilized with 0.1% Triton X for 15 min and stained with DAPI(10 µg/
7.42 (m, 1H), 7.39 – 7.35 (m, 1H), 5.87 (s, 1H), 5.09 (dd, J = 14.2, 4.2
mL). Later, the staining solution was removed, and the wells were
Hz, 1H), 4.89 (dd, J = 14.2, 8.0 Hz, 1H), 4.49 (s, 1H), 3.25 (dd, J = 12.6,
washed with PBS. Cells were visualized using a fluorescence microscope
2.3 Hz, 1H), 3.11 (dd, J = 12.6, 10.1 Hz, 1H), 2.88 (t, J = 8.0 Hz, 2H),
(Nikon, Inc. Japan) with excitation at 359 nm and emission at 461 nm
1.65 – 1.50 (m, 2H), 1.25 (dd, J = 14.9, 7.8 Hz, 4H), 1.22 (s, 6H), 0.85 (t,
using a DAPI filter at 20X magnifications.
J = 6.9 Hz, 3H); 13C NMR (125 MHz, DMSO‑d6) δ 147.75, 146.27,
145.34, 142.69, 141.56, 140.59, 137.92, 131.62, 131.42, 130.14,

15
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

4.2.6. 2′,7′-Dichlorofluorescein diacetate (DCFDA) staining along with the 5l


A549 cells were plated at a concentration of 40,000–50,000cells/
well in a 24-well plate, and different concentrations of compound 5l 5. Molecular docking
were added. Treated plates were incubated in a CO2 incubator for 48 h
and stained with DCFDA (10 µM) for 30 min at 37 ◦ C. Later, the staining Molecular docking study was performed using the glide docking
solution was removed, and wells were washed with PBS to remove the module of the Schrödinger suite release 2021 [81]. The crystal co­
excess dye. Further, cells were visualized using a fluorescence micro­ ordinates of α,β-tubulin subunits (PDB: 3E22) were obtained from the
scope (Nikon, Inc. Japan) with excitation at 498 nm and emission at 530 RCSB. The protein was prepared using the Protein Preparation Wizard of
nm and captured images at 20X magnifications. the Schrödinger suite by adding hydrogens, bond orders were assigned,
and water molecules were removed within 5 Å around the co-crystal.
4.2.7. Effect on mitochondrial membrane potential(DΨ m) The protein structure was minimized using the OPLS-5 force field. The
A549 cells were plated at 40,000–50,000 cells/well in a 24-well grid for docking was generated by taking the centroid of the co-crystal
plate, and different concentrations of compound 5l were added. and validated by re-docking of co-crystal to an RMSD of 0.92. Com­
Treated plates were incubated in a CO2 incubator for 48 h and stained pound 5l was drawn and prepared using LigPrep, and was docked using
with 5 μg/mL of 5,5′,6,6′-Tetrachloro-1,1′,3,3′ tetraethyl benzimidazole the GLIDE module in the standard precision mode.
carbocyanine iodide (JC1 dye) for 30 min at 37 ◦ C. Later, the staining
solution was removed, and the wells were washed with PBS to remove Author contributions
the excess dye.. Further, fluorescence images were captured using a
fluorescence microscope at 20X magnification (Nikon, Inc. Japan). Ojaswitha Ommi: conceptualization, design, optimization, synthesis,
data analysis & interpretation and writing – original draft.
4.2.8. Annexin V-FITC/Propidium iodide dual staining Shrilekha chilvery: biological studies
A549 cells were plated at a concentration of 1 × 106 cells/well in a 6- Priyanka Sudhir Dhopat: Synthesis
well plate, and different concentrations of compound 5l were added. Anamika sharma: biological studies
Treated plates were incubated in a CO2 incubator for 48 h, and cells were Harshada Anil Bhalerao and Srinivas Reddy Dannaram: NMR and
trypsinized using trypsin-EDTA, followed by twice washing with ice- mass studies
cold PBS. Cells were centrifuged to obtain a pellet. Then the superna­ Srinivas Nanduri: Writing-review & editing
tant was removed from each tube, and the cells pellet was resuspended Rajesh Sonti: Supervision, writing – review & editing
in a 1 X binding buffer containing 5 μL and 1 μL of FITC-conjugated Chandraiah Godugu: Supervision, Resources
annexin V and PI, respectively. The tubes were incubated for 15 min Venkata Madhavi Yaddanapudi: Project administrator,
in the dark as per the manufacturer’s protocol of the Apoptosis Detection Supervision, Writing-review & editing
Kit (Invitrogen). Later, 400 μL of 1x binding buffer was added, and cells
were kept on ice for flow cytometric analysis using a flow cytometer (BD Declaration of Competing Interest
FACSVerseTM, USA).
The authors declare that they have no known competing financial
4.2.9. Cell cycle analysis interests or personal relationships that could have appeared to influence
A549 cells were plated at a concentration of 1 × 106 cells/well in a 6- the work reported in this paper.
well plate, and different concentrations of compound 5l were added.
Treated plates were incubated in a CO2 incubator for 48 h, and cells were Data availability
trypsinized using trypsin-EDTA, followed by twice washing with ice-
cold PBS. Cells were centrifuged and then fixed in 70% ethanol and No data was used for the research described in the article.
stored at 4 ◦ C overnight. Fixed cells were further centrifuged, and the
cells pellet was washed with cell cycle buffer before being stained with
Enzyme A and nuclear dye for 30 min at 37 ◦ C in the dark. Further, flow Supplementary materials
cytometric analysis was performed using a flow cytometer (BD FACS­
VerseTM, USA). Supplementary material associated with this article can be found, in
the online version, at doi:10.1016/j.molstruc.2023.136184.
4.2.10. Effect on tubulin polymerization
To examine the effect of compound 5l on tubulin inhibition, a References
fluorescence-based in vitro tubulin polymerization assay was executed
based on the manufacturer’s protocol (Cytoskeleton, Inc. BK011). [1] H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray,
Colchicine was used as a standard in the assay at 1.03 µM final con­ Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality
worldwide for 36 cancers in 185 countries, CA Cancer J. Clin. 71 (3) (2021)
centration, and 2.19, 4.37, and 8.74 µM concentrations of compound 5l 209–249.
were used for the assay. The 50% tubulin polymerization inhibitory [2] R.L. Siegel, K.D. Miller, N.S. Wagle, A. Jemal, Cancer statistics, 2023, CA Cancer J.
concentration of 5l was calculated based on the assay result. Clin. 73 (1) (2023) 17–48.
[3] M.R. Elmorsy, S.E. Mahmoud, A.A. Fadda, E. Abdel-Latif, M.A. Abdelmoaz,
Synthesis, biological evaluation and molecular docking of new triphenylamine-
4.2.11. Immunofluorescence assay linked pyridine, thiazole and pyrazole analogues as anticancer agents, BMC Chem.
A549 cells were treated with compound 5l (4.37 µM) in a 4-cham­ 16 (1) (2022) 88.
[4] F.U. Vaidya, A. Sufiyan Chhipa, V. Mishra, V.K. Gupta, S.G. Rawat, A. Kumar,
bered slide for 48 h. cells were washed in phosphate-buffered saline,
C. Pathak, Molecular and cellular paradigms of multidrug resistance in cancer,
fixed with 4% paraformaldehyde, and permeabilized using 0.1% triton- Cancer Rep. 5 (12) (2022) e1291.
X. After blocking with 0.3% bovine serum albumin, the primary anti­ [5] J. Tímár, A. Uhlyarik, On-target side effects of targeted therapeutics of cancer,
body for beta-tubulin (1:200; Cell Signal Technology) and fluorescence- Pathol. Oncol. Res. POR 28 (2022), 1610694.
[6] P. Musyuni, J. Bai, A. Sheikh, K. Vasanthan, G. Jain, M. Abourehab, V. Lather,
labeled-secondary antibody was subsequently added to the cells. Cell G. Aggarwal, P. Kesharwani, D. Pandita, Precision medicine: ray of hope in
nuclei were stained by DAPI. Images were acquired with a Leica Laser overcoming cancer multidrug resistance, Drug Resist. Updates 65 (2022), 100889.
Scanning Spectral Confocal Microscope (Leica, Germany) with overlays [7] L. Zhong, Y. Li, L. Xiong, W. Wang, M. Wu, T. Yuan, W. Yang, C. Tian, Z. Miao,
T. Wang, S. Yang, Small molecules in targeted cancer therapy: advances,
from separate images of tubulin (red) and nuclei (blue). For comparison, challenges, and future perspectives, Signal Transduct. Target. Ther. 6 (1) (2021)
we also included standards Colchicine (1.04 µM) and paclitaxel (3 µM) 201.

16
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

[8] P.L. Bedard, D.M. Hyman, M.S. Davids, L.L. Siu, Small molecules, big impact: 20 [33] I. Beltran-Hortelano, V. Alcolea, M. Font, S. Pérez-Silanes, The role of imidazole
years of targeted therapy in oncology, Lancet 395 (10229) (2020) 1078–1088 and benzimidazole heterocycles in Chagas disease: a review, Eur. J. Med. Chem.
(London, England). 206 (2020), 112692.
[9] N.B. Gudimchuk, J.R. McIntosh, Regulation of microtubule dynamics, mechanics [34] H.E. Hashem, Y. El Bakri, An overview on novel synthetic approaches and
and function through the growing tip, Nat. Rev. Mol. Cell Biol. 22 (12) (2021) medicinal applications of benzimidazole compounds, Arab. J. Chem. 14 (11)
777–795. (2021), 103418.
[10] A.L. Parker, M. Kavallaris, J.A. McCarroll, Microtubules and their role in cellular [35] B. Pathare, T. Bansode, Review- biological active benzimidazole derivatives,
stress in cancer, Front. Oncol. 4 (2014). Results Chem. 3 (2021), 100200.
[11] Y. Lu, J. Chen, M. Xiao, W. Li, D.D. Miller, An overview of tubulin inhibitors that [36] L.S. Feng, W.Q. Su, J.B. Cheng, T. Xiao, H.Z. Li, D.A. Chen, Z.L. Zhang,
interact with the colchicine binding site, Pharm. Res. 29 (11) (2012) 2943–2971. Benzimidazole hybrids as anticancer drugs: an updated review on anticancer
[12] E.C. McLoughlin, N.M. O’Boyle, Colchicine-binding site inhibitors from chemistry properties, structure-activity relationship, and mechanisms of action (2019-2021),
to clinic: a review, Pharmaceuticals 13 (1) (2020) 8. Arch. Pharm. 355 (6) (2022), e2200051 (Weinheim).
[13] C.M. Lin, H.H. Ho, G.R. Pettit, E. Hamel, Antimitotic natural products [37] Y.T. Lee, Y.J. Tan, C.E. Oon, Benzimidazole and its derivatives as cancer
combretastatin A-4 and combretastatin A-2: studies on the mechanism of their therapeutics: the potential role from traditional to precision medicine, Acta Pharm.
inhibition of the binding of colchicine to tubulin, Biochemistry 28 (17) (1989) Sin. B 13 (2) (2023) 478–497.
6984–6991. [38] K. Laxmikeshav, P. Sharma, M. Palepu, P. Sharma, A. Mahale, J. George,
[14] D.S. Lawrence, J.E. Copper, C.D. Smith, Structure-activity studies of substituted R. Phanindranath, M.P. Dandekar, O.P. Kulkarni, N. Nagesh, N. Shankaraiah,
quinoxalinones as multiple-drug-resistance antagonists, J. Med. Chem. 44 (4) Benzimidazole based bis-carboxamide derivatives as promising cytotoxic agents:
(2001) 594–601. design, synthesis, in silico and tubulin polymerization inhibition, J. Mol. Struct.
[15] P. Diana, A. Martorana, P. Barraja, A. Montalbano, G. Dattolo, G. Cirrincione, 1271 (2023), 134078.
F. Dall’acqua, A. Salvador, D. Vedaldi, G. Basso, G. Viola, Isoindolo[2,1-a] [39] K. Laxmikeshav, A. Himaja, N. Shankaraiah, Exploration of benzimidazoles as
quinoxaline derivatives, novel potent antitumor agents with dual inhibition of potential microtubule modulators: an insight in the synthetic and therapeutic
tubulin polymerization and topoisomerase I, J. Med. Chem. 51 (8) (2008) evolution, J. Mol. Struct. 1253 (2022), 132251.
2387–2399. [40] C.F. Torres, E.M. Garcia-Rubino, C. Lozano-Lopez, F.D. Kawano, L.V. Eifler-Lima, L.
[16] V. Spanò, M. Pennati, B. Parrino, A. Carbone, A. Montalbano, V. Cilibrasi, V. Zuco, V.G. Poser, M.J. Campos, Imidazoles and benzimidazoles as tubulin-modulators for
A. Lopergolo, D. Cominetti, P. Diana, G. Cirrincione, P. Barraja, N. Zaffaroni, anti-cancer therapy, Curr. Med. Chem. 22 (11) (2015) 1312–1323.
Preclinical activity of new [1,2]oxazolo[5,4-e]isoindole derivatives in diffuse [41] S.M. Attia, Molecular cytogenetic evaluation of the mechanism of genotoxic
malignant peritoneal mesothelioma, J. Med. Chem. 59 (15) (2016) 7223–7238. potential of amsacrine and nocodazole in mouse bone marrow cells, J. Appl.
[17] M. Montana, F. Mathias, T. Terme, P. Vanelle, Antitumoral activity of quinoxaline Toxicol. JAT 33 (6) (2013) 426–433.
derivatives: a systematic review, Eur. J. Med. Chem. 163 (2019) 136–147. [42] R.J. Vasquez, B. Howell, A.M. Yvon, P. Wadsworth, L. Cassimeris, Nanomolar
[18] M. Alswah, A.H. Bayoumi, K. Elgamal, A. Elmorsy, S. Ihmaid, H.E.A. Ahmed, concentrations of nocodazole alter microtubule dynamic instability in vivo and in
Design, synthesis and cytotoxic evaluation of novel chalcone derivatives bearing vitro, Mol. Biol. Cell 8 (6) (1997) 973–985.
triazolo[4,3-a]-quinoxaline moieties as potent anticancer agents with dual EGFR [43] A.D. Ricart, E.A. Ashton, M.M. Cooney, J. Sarantopoulos, J.M. Brell, M.A. Feldman,
kinase and tubulin polymerization inhibitory effects, 2017. (Accessed 1 23). K.E. Ruby, K. Matsuda, M.S. Munsey, G. Medina, A. Zambito, A.W. Tolcher, S.
[19] A. Courbet, N. Bec, C. Constant, C. Larroque, M. Pugniere, S. El Messaoudi, C. Remick, A phase I study of MN-029 (denibulin), a novel vascular-disrupting
Z. Zghaib, S. Khier, C. Deleuze-Masquefa, F. Gattacceca, Imidazoquinoxaline agent, in patients with advanced solid tumors, Cancer Chemother. Pharmacol. 68
anticancer derivatives and imiquimod interact with tubulin: characterization of (4) (2011) 959–970.
molecular microtubule inhibiting mechanisms in correlation with cytotoxicity, [44] W. Shi, D.W. Siemann, Preclinical studies of the novel vascular disrupting agent
PLoS ONE 12 (8) (2017), e0182022. MN-029, Anticancer Res. 25 (6b) (2005) 3899–3904.
[20] P. Diana, A. Martorana, P. Barraja, A. Montalbano, G. Dattolo, G. Cirrincione, [45] C. Viegas-Junior, A. Danuello, V. da Silva Bolzani, E.J. Barreiro, C.A. Fraga,
F. Dall’Acqua, A. Salvador, D. Vedaldi, G. Basso, G. Viola, Isoindolo[2,1-a] Molecular hybridization: a useful tool in the design of new drug prototypes, Curr.
quinoxaline derivatives, novel potent antitumor agents with dual inhibition of Med. Chem. 14 (17) (2007) 1829–1852.
tubulin polymerization and topoisomerase I, J. Med. Chem. 51 (8) (2008) [46] M. Matsuyama, K. Funao, K. Kuratsukuri, T. Tanaka, Y. Kawahito, H. Sano,
2387–2399. J. Chargui, J.L. Touraine, N. Yoshimura, R. Yoshimura, Telmisartan inhibits human
[21] Z. Zghaib, J.F. Guichou, J. Vappiani, N. Bec, K. Hadj-Kaddour, L.A. Vincent, urological cancer cell growth through early apoptosis, Exp. Ther. Med. 1 (2) (2010)
S. Paniagua-Gayraud, C. Larroque, G. Moarbess, P. Cuq, I. Kassab, C. Deleuze- 301–306.
Masquéfa, M. Diab-Assaf, P.A. Bonnet, New imidazoquinoxaline derivatives: [47] T.C. Shih, R. Liu, C.T. Wu, X. Li, W. Xiao, X. Deng, S. Kiss, T. Wang, X.J. Chen,
synthesis, biological evaluation on melanoma, effect on tubulin polymerization and R. Carney, H.J. Kung, Y. Duan, P.M. Ghosh, K.S. Lam, Targeting Galectin-1 impairs
structure-activity relationships, Bioorg. Med. Chem. 24 (11) (2016) 2433–2440. castration-resistant prostate cancer progression and invasion, Clin. Cancer Res. 24
[22] T. Liang, X. Zhou, L. Lu, H. Dong, Y. Zhang, Y. Xu, J. Qi, Y. Zhang, J. Wang, (17) (2018) 4319–4331.
Structure-activity relationships and antiproliferative effects of 1,2,3,4-4H-qui­ [48] T.C. Shih, R. Liu, G. Fung, G. Bhardwaj, P.M. Ghosh, K.S. Lam, A novel galectin-1
noxaline derivatives as tubulin polymerization inhibitors, Bioorg. Chem. 110 inhibitor discovered through one-bead two-compound library potentiates the
(2021), 104793. antitumor effects of paclitaxel in vivo, Mol. Cancer Ther. 16 (7) (2017) 1212–1223.
[23] J. Qi, J. Huang, X. Zhou, W. Luo, J. Xie, L. Niu, Z. Yan, Y. Luo, Y. Men, Y. Chen, [49] T.G. Kraljević, A. Harej, M. Sedić, S.K. Pavelić, V. Stepanić, D. Drenjančević,
Y. Zhang, J. Wang, Synthesis and biological evaluation of quinoxaline derivatives J. Talapko, S. Raić-Malić, Synthesis, in vitro anticancer and antibacterial activities
as tubulin polymerization inhibitors that elevate intracellular ROS and triggers and in silico studies of new 4-substituted 1,2,3-triazole-coumarin hybrids, Eur. J.
apoptosis via mitochondrial pathway, Chem. Biol. Drug Des. 93 (4) (2019) Med. Chem. 124 (2016) 794–808.
617–627. [50] K. El Bourakadi, M.E.M. Mekhzoum, C. Saby, H. Morjani, H. Chakchak,
[24] J. Qi, H. Dong, J. Huang, S. Zhang, L. Niu, Y. Zhang, J. Wang, Synthesis and N. Merghoub, A.E.K. Qaiss, R. Bouhfid, Synthesis, characterization and in vitro
biological evaluation of N-substituted 3-oxo-1,2,3,4-tetrahydro-quinoxaline-6-car­ anticancer activity of thiabendazole-derived 1,2,3-triazole derivatives, New J.
boxylic acid derivatives as tubulin polymerization inhibitors, Eur. J. Med. Chem. Chem. 44 (28) (2020) 12099–12106.
143 (2018) 8–20. [51] K. Laxmikeshav, M. Sayali, G. Devabattula, D.G. Valapil, A. Mahale, P. Sharma,
[25] Z. Zghaib, J.F. Guichou, J. Vappiani, N. Bec, K. Hadj-Kaddour, L.A. Vincent, J. George, R. Phanindranath, C. Godugu, O.P. Kulkarni, N. Nagesh, N. Shankaraiah,
S. Paniagua-Gayraud, C. Larroque, G. Moarbess, P. Cuq, I. Kassab, C. Deleuze- Triazolo-linked benzimidazoles as tubulin polymerization inhibitors and DNA
Masquéfa, M. Diab-Assaf, P.A. Bonnet, New imidazoquinoxaline derivatives: intercalators: design, synthesis, cytotoxicity, and docking studies, Arch. Pharm.
synthesis, biological evaluation on melanoma, effect on tubulin polymerization and 356 (5) (2023), e2200449.
structure–activity relationships, Bioorg. Med. Chem. 24 (11) (2016) 2433–2440. [52] A. Djemoui, A. Naouri, M.R. Ouahrani, D. Djemoui, S. Lahcene, M.B. Lahrech,
[26] J.A. Pereira, A.M. Pessoa, M.N.D.S. Cordeiro, R. Fernandes, C. Prudêncio, J. L. Boukenna, H.M.T. Albuquerque, L. Saher, D.H.A. Rocha, F.L. Monteiro, L.
P. Noronha, M. Vieira, Quinoxaline, its derivatives and applications: a State of the A. Helguero, K. Bachari, O. Talhi, A.M.S. Silva, A step-by-step synthesis of triazole-
Art review, Eur. J. Med. Chem. 97 (2015) 664–672. benzimidazole-chalcone hybrids: anticancer activity in human cells+, J. Mol.
[27] X. Jiang, K. Wu, R. Bai, P. Zhang, Y. Zhang, Functionalized quinoxalinones as Struct. 1204 (2020), 127487.
privileged structures with broad-ranging pharmacological activities, Eur. J. Med. [53] N. Sridhar Goud, V. Pooladanda, K. Muni Chandra, P.S. Lakshmi Soukya, R. Alvala,
Chem. 229 (2022), 114085. P. Kumar, C. Nagaraj, R. Dawn Bharath, I.A. Qureshi, C. Godugu, M. Alvala, Novel
[28] S. Tariq, K. Somakala, M. Amir, Quinoxaline: an insight into the recent benzimidazole-triazole hybrids as apoptosis inducing agents in lung cancer: design,
pharmacological advances, Eur. J. Med. Chem. 143 (2018) 542–557. synthesis, 18F-radiolabeling & galectin-1 inhibition studies, Bioorg. Chem. 102
[29] M. Montana, V. Montero, O. Khoumeri, P. Vanelle, Quinoxaline derivatives as (2020), 104125.
antiviral agents: a systematic review, Molecules 25 (12) (2020) 2784. [54] J.R. Baker, P.J. Cossar, M.A.T. Blaskovich, A.G. Elliott, J. Zuegg, M.A. Cooper, P.
[30] S.K. Suthar, N.S. Chundawat, G.P. Singh, J.M. Padrón, Y.K. Jhala, Quinoxaline: a J. Lewis, A. McCluskey, Amino alcohols as potential antibiotic and antifungal
comprehension of current pharmacological advancement in medicinal chemistry, leads, Molecules 27 (7) (2022) (Basel, Switzerland).
Eur. J. Med. Chem. Rep. 5 (2022), 100040. [55] K. Chennakesava Rao, Y. Arun, K. Easwaramoorthi, C. Balachandran, T. Prakasam,
[31] G.F. Zha, H.D. Preetham, S. Rangappa, K.S. Sharath Kumar, Y.R. Girish, K. T. Eswara Yuvaraj, P.T. Perumal, Synthesis, antimicrobial and molecular docking
P. Rakesh, M. Ashrafizadeh, A. Zarrabi, K.S. Rangappa, Benzimidazole analogues as studies of enantiomerically pure N-alkylated β-amino alcohols from
efficient arsenals in war against methicillin-resistance staphylococcus aureus phenylpropanolamines, Bioorg. Med. Chem. Lett. 24 (14) (2014) 3057–3063.
(MRSA) and its SAR studies, Bioorg. Chem. 115 (2021), 105175. [56] A. Dassonville-Klimpt, J. Schneider, C. Damiani, C. Tisnerat, A. Cohen, N. Azas,
[32] A.C. G, R. Gondru, Y. Li, J. Banothu, Coumarin–benzimidazole hybrids: a review of M. Marchivie, J. Guillon, C. Mullié, P. Agnamey, A. Totet, J. Dormoi, N. Taudon,
developments in medicinal chemistry, Eur. J. Med. Chem. 227 (2022), 113921. B. Pradines, P. Sonnet, Design, synthesis, and characterization of novel

17
O. Ommi et al. Journal of Molecular Structure 1292 (2023) 136184

aminoalcohol quinolines with strong in vitro antimalarial activity, Eur. J. Med. [70] L. Deswal, V. Verma, D. Kumar, C.P. Kaushik, A. Kumar, Y. Deswal, S. Punia,
Chem. 228 (2022), 113981. Synthesis and antidiabetic evaluation of benzimidazole-tethered 1,2,3-triazoles,
[57] M. Quiliano, A. Mendoza, K.Y. Fong, A. Pabón, N.E. Goldfarb, I. Fabing, Arch. Pharm. 353 (9) (2020), e2000090.
A. Vettorazzi, A. López de Cerain, B.M. Dunn, G. Garavito, D.W. Wright, E. Deharo, [71] Ö.D. Can, D. Osmaniye, Ü. Demir Özkay, B.N. Sağlık, S. Levent, S. Ilgın, M. Baysal,
S. Pérez-Silanes, I. Aldana, S. Galiano, Exploring the scope of new arylamino Y. Özkay, Z.A. Kaplancıklı, MAO enzymes inhibitory activity of new benzimidazole
alcohol derivatives: synthesis, antimalarial evaluation, toxicological studies, and derivatives including hydrazone and propargyl side chains, Eur. J. Med. Chem. 131
target exploration, Int. J. Parasitol. Drugs Drug Resist. 6 (3) (2016) 184–198. (2017) 92–106.
[58] O. Goshain, B. Ahmed, Antihypertensive activity, toxicity and molecular docking [72] S.K. Sahoo, M.N. Ahmad, G. Kaul, S. Nanduri, A. Dasgupta, S. Chopra, V.
study of newly synthesized xanthon derivatives (xanthonoxypropanolamine), PLoS M. Yaddanapudi, Synthesis and evaluation of triazole congeners of nitro-
ONE 14 (8) (2019), e0220920. benzothiazinones potentially active against drug resistant Mycobacterium
[59] J.P. Bonte, M.C. Piancastelli, I. Lesieur, J.C. Lamar, M. Beaughard, G. Dureng, tuberculosis demonstrating bactericidal efficacy, RSC Med. Chem. 13 (5) (2022)
Amino ketone and amino alcohol derivatives of benzoxazolinone: synthesis, 585–593.
adrenergic and antihypertensive properties, Eur. J. Med. Chem. 25 (4) (1990) [73] S. Satish, A. Srivastava, P. Yadav, S. Varshney, R. Choudhary, V.M. Balaramnavar,
361–368. T. Narender, A.N. Gaikwad, Aegeline inspired synthesis of novel amino alcohol and
[60] P. Mahendran, A. Jeya Rajendran, C. Balachandran, A. Stalin, S. Rangan, thiazolidinedione hybrids with antiadipogenic activity in 3T3-L1 cells, Eur. J. Med.
L. Kothandapani, K. Chennakesava Rao, S. Awale, B.N. Hiteshkumar, Synthesis of Chem. 143 (2018) 780–791.
novel β-amino alcohols from phenylacetylcarbinol: cytotoxicity activity against [74] S. Kumar, C. Upadhyay, M. Bansal, M. Grishina, B.S. Chhikara, V. Potemkin,
A549 cells and molecular docking, Res. Chem. Intermed. 44 (1) (2018) 535–552. B. Rathi, Poonam, Experimental and computational studies of microwave-assisted,
[61] C. Teixeira, R.B. Pereira, N.F.S. Pinto, C.M.M. Coelho, M.J.G. Fernandes, A. facile ring opening of epoxide with less reactive aromatic amines in nitromethane,
G. Fortes, M.S.T. Gonçalves, D.M. Pereira, Eugenol β-amino/β-alkoxy alcohols with ACS Omega 5 (30) (2020) 18746–18757.
selective anticancer activity, Int. J. Mol. Sci. 23 (7) (2022) 3759. [75] K. Liu, P.C. Liu, R. Liu, X. Wu, Dual AO/EB staining to detect apoptosis in
[62] Z. Chen, T. Yang, W. Wang, J. Yao, S. Han, Y. Tao, R. Wang, L. Duan, Synthesis and osteosarcoma cells compared with flow cytometry, Med. Sci. Monit. Basic Res. 21
biological evaluation of carbazole aminoalcohols as antitumor agents, (2015) 15–20.
ChemistrySelect 3 (44) (2018) 12630–12638. [76] B.I. Tarnowski, F.G. Spinale, J.H. Nicholson, DAPI as a useful stain for nuclear
[63] W. Wang, X. Sun, D. Sun, S. Li, Y. Yu, T. Yang, J. Yao, Z. Chen, L. Duan, Carbazole quantitation, Biotech. Histochem. 66 (6) (1991) 296–302.
aminoalcohols induce antiproliferation and apoptosis of human tumor cells by [77] E. Eruslanov, S. Kusmartsev, Identification of ROS using oxidized DCFDA and flow-
inhibiting topoisomeraseI, ChemMedChem 11 (24) (2016) 2675–2681. cytometry, Methods Mol. Biol. 594 (2010) 57–72 (Clifton, N.J.).
[64] J.R. Baker, C.C. Russell, J. Gilbert, A. McCluskey, J.A. Sakoff, Amino alcohol [78] L.D. Zorova, V.A. Popkov, E.Y. Plotnikov, D.N. Silachev, I.B. Pevzner, S.
acrylonitriles as broad spectrum and tumour selective cytotoxic agents, RSC Med. S. Jankauskas, V.A. Babenko, S.D. Zorov, A.V. Balakireva, M. Juhaszova, S.
Chem. 12 (6) (2021) 929–942. J. Sollott, D.B. Zorov, Mitochondrial membrane potential, Anal. Biochem. 552
[65] M.M. Weinberger, Use of ephedrine in bronchodilator therapy, Pediatr. Clin. N. (2018) 50–59.
Am. 22 (1) (1975) 121–127. [79] A. Cormier, M. Marchand, R.B.G. Ravelli, M. Knossow, B. Gigant, Structural insight
[66] C. Delpivo, G. Micheletti, C. Boga, A. Green, Synthesis of quinoxalines and 2,3- into the inhibition of tubulin by vinca domain peptide ligands, EMBO Rep. 9 (11)
dihydropyrazines, Synthesis 45 (2013) 1546–1552. (2008) 1101–1106.
[67] Y. Goto, Y. Tagawa, K. Yamashita, Y. Higuchi, Improved oxidation of active methyl [80] M. Alswah, A.H. Bayoumi, K. Elgamal, A. Elmorsy, S. Ihmaid, H.E.A. Ahmed,
group of N-heteroaromatic compounds by selenium dioxide in the presence of Design, synthesis and cytotoxic evaluation of novel chalcone derivatives bearing
tertbutyl hydroperoxide, Heterocycles 60 (2003) 953–957. triazolo[4,3-a]-quinoxaline moieties as potent anticancer agents with dual EGFR
[68] K. Matsumura, M. Ono, M. Yoshimura, H. Kimura, H. Watanabe, Y. Okamoto, kinase and tubulin polymerization inhibitory effects, Molecules 23 (1) (2017) 48.
M. Ihara, R. Takahashi, H. Saji, Synthesis and biological evaluation of novel styryl [81] Schrödinger Release, 1: Glide, Schrödinger, LLC, 3, Schrödinger Release, New
benzimidazole derivatives as probes for imaging of neurofibrillary tangles in York, NY, 2021, 2021.
Alzheimer’s disease, Bioorg. Med. Chem. 21 (11) (2013) 3356–3362. [82] Schrödinger, QikProp 4.4 User Manual, Schrödinger, 2015. http://gohom.win
[69] M. Kumar, S.K. Pandey, N. Chaudhary, A. Mishra, D. Gupta, Highly efficient /ManualHom/Schrodinger/Schrodinger_2015-2_docs/qikprop/qikprop_user_man
method for the synthesis of substituted benzimidazoles using sodium metabisulfite ual.pdf.
adsorbed on silica gel, Results Chem. 4 (2022), 100403.

18

You might also like