2 Future Medicinal Chemistry
2 Future Medicinal Chemistry
2 Future Medicinal Chemistry
Graphical abstract:
Carbohydrazide plays an important role
in anti-cancer activity
O
Significant loss in potency
Order of Reactivity
R 3-OMe > 2-Cl > 2-F > 4-substituted
First draft submitted: 12 November 2022; Accepted for publication: 17 March 2023; Published online:
12 May 2023
Protein kinases are intracellular enzymes that regulate the phosphorylation of proteins [1]. They are the largest
group of kinases that phosphorylate at serine, threonine and tyrosine residues, and are involved in several signal
transduction pathways [2]. There are two main types of protein kinases: serine/threonine kinases and tyrosine kinases.
The serine/threonine kinase AKT, which is also referred to as protein kinase B, is a key component of phosphatidyl-
10.4155/fmc-2022-0271
C 2023 Newlands Press Future Med. Chem. (Epub ahead of print) ISSN 1756-8919
Research Article Yadav, Kumar, Bajaj & Mayur
inositol-3-kinase (PI3K), which plays a crucial role in regulating cell growth, metabolism, proliferation, survival,
migration and angiogenesis [3,4]. It is activated by phosphorylation on residue Ser473 or Thr308 and it phosphorylates
a variety of downstream protein substrates [5]. The overexpression or hyperactivation of AKT has been observed
in many cancers and is associated with increased cancer cell survival and proliferation; thus, targeting AKT
could provide an attractive approach for the design and development of small molecules as promising anticancer
agents [5,6].
Acridone is an interesting tricyclic N-heterocycle containing a unique planar structure [7]; it can intercalate with
DNA base pairs [8] and acts as a substrate for multidrug resistance [9,10], efflux pumps like P-glycoprotein [11,12] and
breast cancer resistance protein [13,14]. It has been reported in the literature that this scaffold has the potential to
interact with different biological targets [15,16] such as DNA topoisomerase I/II [17,18], telomerase [19], dihydrofolate
reductase [20] and protein kinases [21], including cyclin-dependent kinases [22] and AKT [23,24]. Hence the acridone
scaffold has emerged as an important anticancer compound and drawn the attention of medicinal chemists aiming
to synthesize novel molecules [25].
Many research scientists have tried to design and synthesize acridone-based scaffolds which would have the ability
to display anticancer activity by interacting with multiple targets. Among them, some studies have discovered that
substituted acridones have the ability to inhibit AKT kinase. For example, Houghton et al. patented the 2-Br/Cl-
N 10 -substituted acridone compounds as AKT inhibitors and revealed that these molecules can inhibit AKT
phosphorylation and kinase activity. Among them, most of the N 10 -butyl-substituted 2-chloroacridones (1a–1e)
were found to be effective against AKT as compared with N 10 -butyl-substituted 2-bromoacridones (except 2a and
2b) at <5 μM concentration [23]. Murahari et al. identified and reported a series of acridone–pyrimidine hybrids as
potent AKT inhibitors. Their study found that these hybrid molecules (3a–3c) showed a good cytotoxicity profile
against A-549 (lung), MCF-7 and MDA-MB-231 (breast) cancer cell lines and exerted their anticancer activity
partly by DNA intercalation. Further, mechanistic studies revealed that the compounds have the potential to inhibit
AKT kinase activity and induce apoptosis. An in vitro AKT kinase assay identified compounds 3a–3c as potent
AKT inhibitors, with an IC50 of <10 nM [24].
There are several AKT inhibitors under clinical investigation, including the ATP-competitive pan-AKT inhibitor
AZD5363, AstraZeneca’s invention, which is also known as capivasertib (Figure 1) [26,27]. It inhibits all AKT isoforms
(AKT1/AKT2/AKT/3) with a potency of <10 nM, and inhibited phosphorylation of AKT substrates in cells
with a potency of approximately 0.3–0.8 μM [28,29]. Currently, this drug is in a phase III clinical trial in two
different combinations: one is with fulvestrant in metastatic hormone receptor-positive/HER2-negative breast
cancer (CAPItello-291) and the second is in combination with palbociclib + fulvestrant in hormone receptor-
positive/HER2-negative advanced breast cancer (CAPItello-292) [27,30].
Helwa et al. designed, synthesized and evaluated 6-morphilino-pyrimidine derivatives as PI3-K inhibitors. Here,
the 6-morphilino-pyrimidine scaffold was linked with aromatic rings through a hydrazinyl linker. All the synthesized
compounds were screened for their cytotoxicity against a panel of NCI-60 cells. Among them, compound 4 showed
the most potent antiproliferative activity against the leukemia SR cell line, with an IC50 of 0.76 μM. The PI3K
kinase inhibition assay found that compound 4 was found to be a promising inhibitor, with IC50 values of 11.73,
6.09 and 11.18 μM against (α), (β) and (γ) of PI3K class IA, respectively. The presence of a hydrazinyl linker and
m-substitution on the phenyl ring resulted in an increase in anticancer activity against a panel of cell lines [31].
Based on previous studies of acridone containing a carbohydrazide linker and the interesting activity of hydrazinyl
compounds, we have synthesized acridone-2-carbohydrazide derivatives and characterized and screened them for
in vitro anticancer activity against human cancer cell lines A-549 (lung), MCF-7 and MDA-MB-231 (breast) and
A-431 (skin). In silico studies were carried out and included molecular docking and prediction of absorption,
distribution, metabolism, elimination and toxicity (ADMET) properties by using computational applications. We
performed molecular docking studies to identify the orientation and binding interactions with essential amino acids
at the active site of AKT1. Acridones have been reported as DNA intercalators [8,32,33]; hence, to estimate the binding
strength of the compounds, we carried out absorption titration against calf thymus DNA (CT-DNA). Quantitative
ELISA analysis of phosphorylated-AKT Ser473 (p-AKT) and total AKT, cell cycle analysis and apoptosis were
carried out for determining the underlying molecular mechanism of the most active derivatives.
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
O
O O
Cl Br
N
N N H H R
N N
R R O N
H
N O
1a R = 4’-Cl O
2a R = 4’-N-Pyrrolidine 3a R = H
1b R = 4’-N-Diethylamine 2b R = 4’-N-Morpholine
1c R = 4’-N-Methypiperazine 3b R = 2-F
1d R = 4’-N-Piperidine 3c R = 4-OMe
1e R = 4’-N-(E-hydroxyethyl)piperazine
Reported AKT inhibitors
O
N
N
O OH
NH2 H
N N N Br
Aromatic Ring HN N
O O
N
H H N
N N N
Acridone ring O N Linker H
H Cl Aromatic Ring
N O O
Aromatic Ring Linker
Linker O Capivasertib
Reported AKT Inhibitor Reported PI3-K inhibitor
(AZD5363)
3c 4
O O
R1 Heterocyclic
N
N Mono-/di-/tri-substituted
H
Aromatic -CH3/-OCH3/-F/
N
H Ring -Cl/ -NO2/-Br/-OH
Figure 1. Structures of acridone derivatives as AKT inhibitors with their design approach. (A) Reported AKT
inhibitors. (B) Design approach of acridone-2-carbohydrazide derivatives.
were recorded on a Fourier transform infrared (FTIR, Spectrum RXI, Perkin Elmer Spectra, Waltham, MA, USA).
The mass spectra in ESI-MS mode were recorded with 6410 Triple Quad LC/MS (Agilent Technologies Inc, Santa
Clara, CA, USA) using methanol as solvent.
9(10H)-acridone-2-carbohydrazide (7)
To a stirred solution of compound 6 (1 g, 3.7 mmol) in dry ethanol, hydrazine hydrate (0.337 ml, 1.5 equiv, 6.73
mmol) was added and refluxing continued for 12 h. The reaction mixture was monitored by TLC and evaporated
under reduced pressure to remove the ethanol. The residue was diluted with cold water, filtered and used as such
for further process.
Biological activity
In vitro cytotoxicity testing by sulforhodamine B assay
All these compounds were subjected to initial screening to evaluate their in vitro antiproliferative activity. A single
dose (25 μM) of the test compounds was used against a panel of cell lines which included non-small-cell lung
cancer (A-549), skin cancer (A-431) and breast cancer cells (MDA-MB-231 and MCF-7) and normal murine
embryonic fibroblast cells (NIH/3T3). Cells were obtained from the National Centre for Cell Science (Pune,
India) and cultured using Dulbecco’s modified Eagle medium (DMEM) (Genetix-Biotech Asia, New Delhi, India)
supplemented with 10% fetal bovine serum (Himedia, Mumbai, India) and 1% penicillin–streptomycin (Genetix-
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
Biotech Asia, New Delhi, India). The data were represented as percentage growth inhibition (GI%) of treated
cells.
A cytotoxicity study of the synthesized compounds was carried out by sulforhodamine B (SRB) assay, using
doxorubicin as a positive control. Cells were plated onto a 96-well plate with a density of 5000–10,000 cells/well
in a complete growth medium and were incubated for 24 h. They were treated with the tested compounds at
different concentrations (0.1–100 μM) for 72 h and then proceeded to SRB assay [35]. For the assay, 20% TCA
(Trichloro-acetic acid) was used for the fixation of cells and then the cells were stored at 4◦ C for 1 h. SRB reagent
(0.4% in acetic acid) was added for staining purposes and then cells were incubated at RT in the dark for 30 min.
Acetic acid (1%) was utilized to wash the cells for the removal of excess unbound SRB. After drying for 4 h, Tris
base was added to each well to solubilize the bound SRB and the optical density (OD) was measured at 510 nm
using a microplate reader [36].
Apoptosis assay
The annexin V/PI double staining method was used to carry out the apoptosis assay with the help of flow
cytometry [44]. An annexin V–fluorescein isothiocyanate (FITC)/PI apoptosis detection kit (BD Biosciences,
Franklin lakes, NJ, USA) was used for the determination of apoptosis. MCF-7 cells were plated in a six-well plate
with a density of 2.5 × 105 cells/well and incubated for 24 h. Then cells were treated with the selected compounds
(8k, 8v and 9h) at 5 μM concentration for 24 h. Cells were trypsinized and washed with PBS, then resuspended in
1× binding buffer at a concentration of 1.0 × 106 cells/ml. From that, 100 μl of the test solution at a concentration
of 1.0 × 105 cells was treated with the mixture of annexin V–FITC (5 μl) and PI (5 μl) and incubated in the dark
for 10–15 min at RT. Finally, 400 μl of binding buffer was added and cells were resuspended again just before the
flow cytometry analysis, performed using a FACSCalibur™ flow cytometer (BD Biosciences) at IIT Powai [31,41].
[DNA] [DNA] 1
= +
εa − εf εb − εf Kb (εb − εf )
Where [DNA] = concentration of DNA base pairs; Kb = binding constant; and εa, εf, εb are the apparent
extinction coefficients for the complex with DNA, without DNA and fully bound with DNA, respectively. A slope
and an intercept were obtained from the plotted graph of [DNA]/(εa − εf ) versus [DNA] which were equal to 1/(εb
− εf ) and 1/Kb /(εb − εf ), respectively. The Kb was calculated from the ratio of the slope to the intercept [47,48].
Molecular docking
To identify existing binding interactions of the synthesized molecules within the binding site of the target protein,
Autodock Vina software was used [49]. x-ray crystallographic structures of AKT1 (Protein Data Bank [PDB] ID:
4GV1) and DNA (PDB ID: 2GB9) were retrieved from the PDB [50]. A docking study was performed as per the
previously reported method [51–53]. Preparatory steps were carried out before docking analysis using Auto Dock
Tools. The 2D and 3D structures of all the synthesized compounds were generated. Both ligands and receptor
were transformed to the proper format for docking. The molecular docking was performed to observe possible
protein–ligand interactions and binding affinity. The docking score for each compound structure was determined
in kcal/mol. Results were analyzed in BIOVIA/Discovery Studio 2017R2 (Biovia, CA, USA) for the visualization
and structure analysis of the docked complexes of AKT1 and DNA. 2D ‘Receptor–Ligand Interactions’ modules
were used to check various types of binding interactions [54,55].
Biological evaluation
In vitro cytotoxicity assessment
All synthesized compounds were subjected to in vitro antiproliferative assessment against various cancer cell lines
(MCF-7, MDA-MB-231, A-549 and A-431). Compounds were initially screened against the cells at a single dose
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
O O O O
OH (i) OC2H5
N N
H H
5 6
(ii)
O O O O
O OH
N R NH2 (iv) N R
(iii) N N
N
H H H
N N N
H H H
R = Aryl, Heteroaryl R = Aryl
(8a–8w) 7 (9a–9g)
Figure 2. Scheme for synthesis of target compounds (8a-8w) and (9a-9h) with reagents and conditions. (i)
SOCl2 ,60◦ C, 3h and ethanol, 80◦ C, 6–8 h; (ii) hydrazine hydrate, 80◦ C, 10 h; (iii) aldehydes, glacial acetic acid, ethanol,
80◦ C, 12–14 h; (iv) ketones, glacial acetic acid, ethanol, 80◦ C, 12–16 h.
of 25 μM concentration and the mean GI% of all compounds was calculated. All the synthesized compounds
(8a–8w and 9a–9h) showed moderate-to-excellent inhibitory activity against all tested cell lines except A-431 at
25 μM. A total of 20 compounds displayed >50% growth inhibition against the MCF-7 cell line (Figure 3). From
them, compounds 8k, 8l, 8o, 8q, 8t, 8u, 8v and 9b–9h exerted good-to-excellent GI% ranging from 63.18 to
80.89%. Similarly, tested compounds displayed moderate inhibitory activity against the MDA-MB-231 cell line.
Compounds 8b, 8f, 8h, 8s, 8u, 8v, 8w, 9b, 9c and 9h demonstrated >50% growth inhibition. For the A-549
cell line, only five compounds (8t, 8w, 9a, 9e, 9g and 9h) had moderate-to-good GI%, ranging from 50.37 to
78.88%. Regarding the breast cancer cell lines, six compounds possessed good inhibitory activity against MCF-7
whereas three compounds displayed good growth inhibition of the MDA-MB-231 cell line.
The cellular toxicity of the selected compounds was also evaluated using normal murine embryonic fibroblast
(NIH/3T3) cell lines at a final concentration of 50 μM to see whether the molecules’ antiproliferative effect had
specificity for cancer cells. The results indicated that these compounds had no significant effect on the growth of
normal cells.
Based on the results of the initial screening of synthesized derivatives, it was found that breast cancer cell lines
MCF-7 and MDA-MB-231 were the most sensitive to the tested compounds, and therefore they were selected for
in vitro antiproliferative activity assessment at different dose levels. The growth inhibitory activity of benzylidene
derivatives (8b, 8c, 8i–8l, 8n, 8o and 8q–8v) and phenylethylidene derivatives (9b–9h) of acridone carbohydrazide
against MCF-7, MDA-MB-231 and A-549 was evaluated by using SRB assay. The results were expressed as half
maximum growth inhibitory concentration (IC50 – the concentration of drug required to exhibit 50% inhibition
of the cell growth as compared with control) and are shown in Table 1.
In vitro cytotoxicity screening against MCF-7 In vitro cytotoxicity screening against MDA-MB-231
100 100
% growth inhibition (GI %)
8a
8b
8c
8d
8e
8f
8g
8h
8i
8j
8k
8l
8m
8n
8o
8p
8q
8r
8s
8t
8u
8v
8w
9a
9b
9c
9d
9e
9f
9g
9h
DOX
In vitro cytotoxicity screening against A-549 In vitro cytotoxicity screening against A-431
100
100
% growth inhibition (GI %)
8a
8b
8c
8d
8e
8f
8g
8h
8i
8j
8k
8l
8m
8n
8o
8p
8q
8r
8s
8t
8u
8v
8w
9a
9b
9c
9d
9e
9f
9g
9h
DOX
Toxicity testing against NIH/3T3
100
90 50 μM
80
% cell viability
70
60
50
40
30
20
10
0
Control
8b
8c
8i
8j
8k
8l
8n
8o
8q
8r
8s
8t
8u
8v
9b
9c
9d
9e
9f
9g
9h
Figure 3. In vitro cytotoxicity screening of acridone-2-carbohydrazide derivative against cancer cell lines at a single dose of 25 μM. (A)
MCF-7 (B) MDA-MB-231 (C) A-549 and (D) A-431. (E) Toxicity testing of selected compounds against normal cells NIH/3T3 at a single dose
of 50 μM.
DOX: Doxorubicin.
The in vitro cytotoxicity study revealed that the substituted phenylethylidene derivatives displayed significant
cytotoxicity against breast cancer cell lines as compared with the substituted benzylidene derivatives. In the first
series (acridone carbohydrazides with benzylidene substitution), most of the compounds showed moderate-to-
good cytotoxic activity against both breast cancer cell lines, indicating that compounds containing the acridone
carbohydrazide with pyridine heterocyclic structure were toxic to cancer cells. Furthermore, the cytotoxic effect
of a few compounds (8b, 8i, 8j, 8k, 8t, 8u and 8v) on MCF-7 cells was much greater than that on MDA-
MB-231 and A-431 cells, indicating that the compounds were selective for AKT kinase. Among them, the most
promising compound 8v showed excellent activity against MCF-7 and MDA-MB-231 cells (IC50 : 6.24 and 9.92
μM, respectively). Compounds 8k, 8t and 8i exhibited good antiproliferative activity against MCF-7 cells, with
IC50 values of 7.92, 8.39 and 10.55 μM respectively, while compounds 8b, 8c, 8h, 8j, 8l, 8n, 8q, 8r and 8u
showed moderate cytotoxicity against MCF-7 cells with IC50 values of 15–30 μM. Among the title compounds
from this series, only the dimethyl amino benzylidene derivative (8t; IC50 : 11.32 μM) and 4-fluoro benzylidene
derivative (8i; IC50 : 19.65 μM) displayed good activity against the A-549 cell line.
In the second series of phenylethylidene derivatives of acridone carbohydrazides, compounds 9b, 9c, 9d and 9h
showed moderate cytotoxicity against MCF-7 and MDA-MB-231 breast cancer cell lines at concentrations <20
μM. Compound 9c showed better activity against MDA-MB-231 (IC50 : 7.45 μM) than against MCF-7 (IC50 :
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
14.47 μM). Compounds having the phenyl ring substituted with 4-fluoro (9e) or 2,4-dichloro (9h) displayed
better activity against the MCF-7 cell line, whereas phenyl ring substituted with 4-methoxy (9c) showed excellent
activity against MDA-MB-231. Compounds containing an ethylidene acridone carbohydrazide moiety were found
to be more potent against MDA-MB-231 cells, with IC50 values of 7.45–23.46 μM. Among them, 4-fluoro
phenylethylidene derivative 9e (IC50 : 8.52 μM) exhibited better activity against A-549. In comparison, the title
compounds 8v and 9h showed appreciable cytotoxicity against MDA-MB-231 and MCF-7 cell lines.
From these data, the cell proliferation assay determines that acridone carbohydrazide with phenylethylidene
substitution is preferred over benzylidene substitution for anticancer activity. Mono substitution on the benzyli-
dene moiety at either the ortho or meta position showed improved cytotoxicity against cancer cells. Acridone
carbohydrazide containing 4-pyridine derivatives exhibited potential anticancer activity against breast cancer cells.
acridone derivatives showed better DNA-binding affinity compared with the substituted phenylethylidene acridone
compounds. Among these compounds of substituted benzylidene derivatives, the 3-methoxy derivative (8k) and
4-dimethyl amino derivative (8t) showed good intrinsic binding constants of 2.12 × 105 and 3.79 × 105 M-1 ,
respectively. From the series of substituted phenylethylidene acridone compounds, the 4-fluoro derivative (9e) and
2,4-dichloro derivative (9h) showed comparatively low DNA-binding affinity, with Kb values of 1.03 × 105 and
0.77 × 105 M-1 , respectively. From the data obtained from the in vitro cytotoxicity assay results, compounds
containing 3-methoxy and 4-dimethyl amino substitutions showed IC50 values <10 μM. This suggests that these
compounds might exert their cytotoxicity through intercalation. Surprisingly, the most active compounds (8v and
9h) showed a relatively lower binding affinity, with Kb values of 0.87 × 105 and 0.77 × 105 M-1 , respectively. These
acridone derivatives did not display any significant DNA intercalation, which might be because of steric hindrance
due to their bulky structure. These compounds might therefore have produced good cytotoxicity results in the
proliferation assay through another mechanism. Thus further studies are required to find the exact mechanism
behind the cytotoxicity of these types of acridone molecules.
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
50
Total-AKT (µg/ml)
Count
100
Comp-PE-A
Comp-PE-A
103 103
50 50
SubG1 G0/G1 S G2M SubG1 G0/G1 S G2M
4.01 62.5 20.8 11.0 3.01 67.3 17.7 11.4
0 0
Q3 Q4 Q3 Q4
0 0 -103 99.5 0.11 -103 84.3 0.45
0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K
10 1
10 2
10 10
3 4
10 5
102
10 3
10 4
105
PE-Texas red-A PE-Texas red-A 101
Comp-FITC-A Comp-FITC-A
8v treated 9h treated 8v treated 9h treated
Q1 Q2 Q1 Q2
10 5 105
104 104
Comp-PE-A
100
Comp-PE-A
100
Count
Count
103 103
50 50
SubG1 G0/G1 S G2M SubG1 G0/G1 S G2M 0 0
69.1 17.7 8.27 4.29 65.9 19.8
3.92 8.91
Q3 Q4 Q3 Q4
-10 3
77.4 1.10 -103 83.9 0.60
0 0
0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K 101 102 103 104 105 101 102 103 104 105
PE-Texas red-A PE-Texas red-A Comp-FITC-A
Comp-FITC-A
60
50
40
30
20
10
0
Control 8k 8v 9h
Figure 4. MCF-7 cells were treated with compounds at different concentrations (0, 1, 2.5 and 5 μM) for 24 h. (A–C) Quantitative ELISA
assay results of compounds 8k, 8v and 9h on (A) levels of p-AKT Ser473 , (B) levels of total AKT and (C) the activation of p-AKT Ser473 . A
significant difference in p-AKT Ser473 was seen between the control group and the 8v-treated group. Compounds inhibit cancer cell
growth by suppressing the activation of AKT signaling. (D) Effect of compounds 8k, 8v and 9h on cell cycle distribution of MCF-7 cells
using flow cytometry. Representation of typical cell cycle profiles. (E) Quantitative analysis of selected compounds on the cell cycle
distribution. (F) Apoptosis assay of compounds 8k, 8v and 9h using flow cytometry after staining with annexin V–fluorescein
isothiocyanate/propidium iodide. MCF-7 cells were treated with 8k, 8v and 9h for 24 h and the results are represented as scatter plots of
propidium iodide (y-axis) versus annexin V (x-axis).
*p < 0.05; **p < 0.01; ***p < 0.001. Error bars: mean ± SD of three independent experiments.
ns: No significant difference.
compounds causing cell cycle arrest, as represented in Figure 4. These compounds were found to have the ability
to inhibit proliferation through cell cycle arrest at G0/G1 phase, which will induce apoptotic cell death [41,42].
and 4 (apoptotic cells) (Figure 4), being the early and late apoptotic cells, respectively. The cells that are positive
for both PI and annexin V are expressed in quadrant 1 (necrotic cells), and those that are negative for both PI and
annexin V are expressed in quadrant 3 (living cells).
The obtained results were compared with the untreated MCF-7 cell line as control, and the values of total
apoptosis (early and late apoptotic cells, quadrant 2 + quadrant 4) were calculated. For compound 8v, the total
apoptotic cells significantly increased from 0.51 to 5.52%, which was higher than the percentages seen with 8k and
9h at corresponding concentrations. For compounds 8k and 9h, the total apoptotic cell percentages were 3.76 and
2.82%, respectively. From the data represented in Figure 4, it is indicated that these compounds (8k, 8v and 9h)
increased the percentage of annexin V-positive apoptotic cells in both the early and late stages. These compounds
also showed an increase in the percentage of necrotic cells, which might be due to external environmental factors
such as metabolic stresses including hypoxia. Among them, compound 8v displayed a significant (tenfold) increase
in apoptotic cells as compared with the control. This study revealed that 8v displayed cytotoxicity against the
MCF-7 cell line through the induction of apoptosis.
In silico studies
Molecular docking
To understand the binding pattern of the synthesized compounds, molecular docking studies were performed with
AKT (PDB ID: 4GV1) and DNA (PDB ID: 2B9). Protein–ligand complexes were analyzed by docking score and
their orientation in the active site of the protein structure using Autodock Vina software. The results from the
molecular docking study revealed that all the tested compounds showed good binding energy with AKT rather than
DNA. Values for AKT ranged from -9.0 to -10.7 kcal/mol, whereas compounds showed minimal binding energy
with DNA (ranging from -7.4 to -9.6 kcal/mol). The 2D docking interactions of the most potent compounds are
depicted in Figure 5A & B.
The docking scores and binding poses of all derivatives were compared with those of the reference compound
(3c). A lower binding energy (more negative) score indicates stronger and more favorable binding between the
protein and ligand. Seven compounds (8b, 8k, 8m, 8t, 9d, 9g and 9h) showed lower interaction energies compared
with the reference compound, which showed a binding affinity of -9.5 kcal/mol. It formed two conventional
hydrogen bond interactions and one hydrophobic interaction (π–π T-shaped) with amino acids Lys276 and
Phe161, respectively. It developed some hydrophobic interactions (π–anion and π–cation) with Glu191, Asp292
and Lys179. We found that compounds 9g and 9h exhibited -10.7 and -10.3 kcal/mol binding affinity with the
target enzyme, respectively. These molecules formed many hydrophobic interactions (π–sigma, π–sulfur, π–π T-
shaped and π–alkyl) with Val164, Met281, Phe161, Ala177, Ala230, Leu156 and Phe438. Additionally, compound
9g formed two conventional hydrogen bond interactions with Lys158 and Lys179.
As per the results, the most common hydrophobic interactions were observed between the tricyclic aromatic ring
of the acridone scaffold and amino acid residues such as Val164, Leu156, Ala177, Ala230 and Met281. Most of
the compounds displayed a common hydrogen bonding interaction with amino acid residue Lys158. p-substituted
phenylethylidene derivatives (9c, 9d, 9e and 9f) formed common π–anion interactions with Asp292 and Lys179
as well as π–alkyl interactions with Val164, Ala177 and Met227 amino acid residues. The same type of π–alkyl
interaction was observed with the p-substituted benzylidene derivatives (8d, 8f, 8m and 8t). Additionally, in
molecules 8k and 8m, the benzene ring substituted at the third position with the methoxy group exhibited π–π
T-shaped interactions. Furthermore, in molecules 8i and 8j, the benzene ring substituted at the second position
with –F and –Cl groups showed halogen interactions with Gly162 and a hydrogen bond interaction with Lys276,
respectively (Supplementary Table 2).
Molecular docking studies with DNA revealed that the acridone ring formed π–π T-shaped interactions with
nucleotide base pairs dG A:6, dC A:5 and dG B:2. Along with that, the nitrogen atom of acridone hydrazide
formed two hydrogen bonds, one with nucleotide base pair dG B:2 and one with nucleotide base pair dG A:2 of
DNA. Docking results of compounds were compared with those of doxorubicin, which showed binding energy of
-9.6 kcal/mol. It formed some conventional hydrogen bond interactions with nucleotide base pairs dA A:4, dA
B:4, dC A:5, dG B:2, dT B:3 and π–alkyl interactions with dA A:4. We found that only compounds 9e, 9f and
9g showed better intercalation, with comparable binding energies -9.2, -9.6 and -9.3 kcal/mol, respectively. These
compounds displayed one common hydrogen bond interaction with nucleotide base pair dG B:2. This suggests that
these molecules can intercalate with DNA, which might be partly responsible for their anticancer activity. However,
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
i MET ii ASP
TYR
A:229
A:227 A:292
ALA
THR GLU A:177
A:291 A:234
ALA VAL
PHE MET
A:177 VAL GLY A:164 THR
A:164 A:159 A:161 A:291 A:281
ASP
MET A:292
A:281
LYS PHE MET
A:179 A:161 A:227
GLU
TYR A:198
A:229
LEU
A:181
ALA LYS
LYS A:179 GLY
A:230
GLU A:163 A:157 THR
LEU GLY GLY
A:191 LEU A:211
A:156 A:157 A:162 LYS A:156
PHE LYS A:158
A:438 A:158 LEU PHE
THR GLY
A:181 A:438 ALA
A:195 A:159
ILE A:230
A:186
iii iv THR
A:195
LYS GLU
PHE A:179 A:191 GLU
LEU ASP
A:438 A:198 LEU
A:156 A:292
ASP LYS A:181
ALA ALA VAL A:179 GLU
A:292
A:230 A:177 A:164 PHE A:191
GLY A:161
TYR A:157
A:229 HIS
MET A:194
A:281
GLY
LEU LEU A:294
A:181 A:295 LYS
GLU A:276 GLY
ASP VAL
A:228 GLY A:159
A:274 A:164 A:162
PHE THR
GLY ASN A:161 A:160
MET A:159 A:279 LYS
A:227 THR LYS LYS A:158
A:291 A:158 THR A:163
A:312
v vi
DA
A:4
DA
DC DT B:4 DG DT
B:1 DG B:3 B:2 B:3
DC A:6
A:5 DG
B:2
DA
B:4
DG
A:6
DC
A:5
Figure 5. 2D representation of binding interactions of acridone molecules with AKT1 (PDB ID: 4GV1). (A) (i)
compound 9g, (ii) compound 9h, (iii) compound 8v and (iv) reference compound. (B) 2D representation of binding
interactions of acridone molecules with DNA (PDB ID: 2GB9), (v) compound 9f and (vi) doxorubicin. The dotted lines
in different colors reflect various types of interactions: hydrogen bonding (green), charge or polar interactions
(orange),π–alkyl/alkyl (light pink), π–sulfur (yellow), π–π T-shaped (pink) and π–σ (violet).
PDB: Protein Data Bank.
the most active compounds (8v and 9h) showed different binding patterns than doxorubicin, with less binding
energies (Supplementary Table 3). This indicates that these compounds might not act through DNA intercalation
but rely on a different mechanism for their good cytotoxicity.
Table 2. Data obtained from predicted absorption, distribution, metabolism, excretion and toxicity parameters.
Descriptors Compound ID
8b 8k 8t 8v 9c 9d 9e 9h
MW (g/mol) 367.4 371.39 447.48 342.35 385.42 369.42 373.38 424.28
No. of HBA 3 4 4 4 4 3 4 3
No. of HBD 2 2 2 2 2 2 2 2
TPSA 78.66 87.89 87.89 91.55 87.89 78.66 78.66 79
Consensus LogP 3.4 3.39 4.6 2.68 3.67 4.04 3.97 4.69
ESOL LogS -4.64 -4.7 -6.01 -3.97 -4.9 -5.14 -5.0 -6.02
Gastrointestinal High High High High High High High High
absorption
ESOL class MS MS PS Soluble MS MS MS PS
BBB permeation No No No No No No No No
BA score 0.55 0.55 0.55 0.55 0.55 0.55 0.55 0.55
Lipinski 0 0 0 0 0 0 0 0
violations
LD50 (mg/kg) 3192.04 1580.4 2228.4 1119.5 3108.5 2900.4 2103.4 2381.2
BA: Bioavailability; BBB: Blood–brain barrier; ESOL class: Solubility class in LogS scale; ESOL LogS: ESOL model logarithm of molar solubility in water; HBA: Hydrogen bond acceptor; HBD:
Hydrogen bond donor; LogP: Logarithm of the partition coefficient; MS: Moderately soluble; MW: Molecular weight; PS: Poorly soluble; TPSA: Topological polar surface area.
Lipinski rule to be developed as orally effective drugs [56]. Any violation of this rule might lead to problems with
pharmacokinetic properties.
As per the results shown in Table 2, all the subjected compounds displayed a molecular weight below 500 g/mol,
which indicates that they can be easily transported. The hydrogen bond acceptor (<10) and donor (<5) numbers
were found to be in the range of 3–4 and 0–2, respectively. Title compounds showed logP values in the range of
2.68–4.69 (values l<5 indicate good permeability). All the compounds showed high gastrointestinal absorption
and no blood–brain barrier permeation.
Results from in silico pharmacokinetic data revealed that all the compounds follow the Lipinski rule with no
violation. These compounds (8b, 8k, 8t, 8v, 9c–9e and 9h) have the desired pharmacokinetic properties and can
be considered as ‘drug-like’ for further development of a lead molecule.
Structure–activity relationship
After the results of the cell proliferation assay and AKT enzyme inhibition studies, compounds 8a–8w and 9a–9h,
bearing the same acridone carbohydrazide moiety but with different substituents on the phenyl ring, were subjected
to structure–activity relationship studies with a few assumptions being made, and the findings are summarized in
Figure 6.
The in vitro cytotoxicity evaluation of substituted acridone carbohydrazides revealed that phenylethylidene
derivatives were found to be more potent against MCF-7 and MDA-MB-231. The presence of electron-withdrawing
and -donating substituents at the o-, p- and m-positions of the phenyl ring plays a significant role in cytotoxicity. In
the series of benzylidene-substituted derivatives, the substitution of the methoxy group at the third position (8k)
of the phenyl ring improved the cytotoxicity against both the cell lines, whereas the di-substitution of the methoxy
group at the third and fourth positions (8m) remarkably reduced the cytotoxicity. However, the cytotoxicity of
the benzylidene acridone carbohydrazide moiety was significantly increased when it was substituted with an m/p-
methoxy group (8f and 8k), and was reduced with p-Cl (8h) and p-F (8d) groups. The presence of –Cl, –OH and
–F at the ortho position enhanced cytotoxicity against MCF-7 cells. When the aromatic ring of the benzylidene of
acridone carbohydrazide was replaced with a heterocyclic ring such as furan, the resultant compound (8w) showed
a significant loss in potency and cytotoxicity. The introduction of a pyridine ring instead of furan in the structure
resulted in compound 8v, which was found to be approximately fivefold more potent than 8w against breast cancer
cell lines and acted as the most potent AKT inhibitor.
In the case of phenylethylidene-substituted derivatives, di-substitution of a halo group (–Cl) at the second and
fourth positions (9h) improved the cytotoxicity against both cell lines as compared with the monosubstituted
compound (9f). Replacement of the benzylidene moiety of acridone carbohydrazide (8c, 8d, 8f, 8g, 8h) with
phenylethylidene (9b, 9c, 9d, 9e, 9f) produced IC50 values of <20 μM and resulted in a significant increase in
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
Order of Reactivity
R 3-OMe > 2-Cl > 2-F > 4-substituted
cytotoxicity, suggesting that the presence of a methyl group is needed to enhance the cytotoxic activity of the
compound. These results indicate that the electron-negativity, position and steric effect in the benzene ring may
change the cytotoxic profile of these compounds.
Conclusion
In this study, a series of compounds bearing acridone-2-carbohydrazide scaffolds were designed based on a rational
design approach, synthesized and evaluated against A-549, MCF-7, MDA-MB-231 and A-431 cancer cell lines.
An in vitro cytotoxicity assay revealed that compounds 8k, 8t, 8v and 9h displayed potent antiproliferative activity,
especially in MCF-7 cells, with IC50 values of 7.92, 8.39, 6.24 and 9.60 μM, respectively. Furthermore, these
compounds were found to be nontoxic to normal mouse embryonic fibroblast (NIH/3T3) cells. The in vitro AKT
kinase inhibition assay disclosed that compounds 8v and 9h exhibited strong growth inhibition at IC50 values of
1.75 and 2.40 μM, respectively. Quantitative ELISA showed that compounds 8k, 8v and 9h significantly reduced
the protein levels of p-AKT Ser473 (rather than total AKT) in a dose-dependent manner. All these results help
us conclude that inhibition of p-AKT Ser473 might be the main contributing factor toward the good cytotoxicity
exhibited by compounds 8v and 9h. Compound 8v was found to be a potential anticancer agent because of
selectivity to p-AKT Ser473 rather than total AKT.
Structure–activity relationship studies indicated that the presence of the carbohydrazide group and the hetero-
cyclic group played an important role in the antiproliferative activities of the tested compounds. The m-, o- and
p-substitutions on the phenyl ring of benzylidene and phenylethylidene derivatives of acridone carbohydrazides
gave high growth-inhibitory activity on breast cancer cell lines. Flow cytometry showed that the cell cycle was
arrested at the G0/G1 phase, which led to decreased DNA synthesis and thus induced apoptosis; hence the p-AKT
Ser473 inhibitory effect of selected derivatives might be correlated with changes in the cell cycle. According to
molecular docking studies, the compounds possessed the same type of binding interactions within the active site of
AKT. In silico studies predicted these compounds as promising and orally available compounds with low toxicity
profiles and suitable for further optimization as p-AKT Ser473 inhibitors in the treatment of breast cancer.
Future perspective
As AKT kinase plays a vital role in multiple cellular processes, it is a well-identified molecular target to design AKT
inhibitors as potential anticancer drug candidates. Our study demonstrates the acridone-2-carbohydrazide scaffold
as a lead molecule and opens possibilities for different structural modifications for designing newer anticancer
agents with fewer side effects. We also propose a future study of our compounds to be evaluated in the area of viral
infections and diseases such as malaria, tuberculosis and parasitic diseases, as acridones are reported to have a broad
spectrum of biological activities.
Summary points
• Synthesis and characterization and biological evaluation of acridone-2-carbohydrazide derivatives.
• Compounds 8k, 8v and 9h exhibited good cytotoxicity against MCF-7 cells (IC50 <10 μM).
• Compounds 8v and 9h exhibited promising AKT kinase inhibitory activity in MCF-7 cells (IC50 <2.5 μM).
• Quantitative p-AKT and total AKT revealed that compound 8v significantly reduced the levels of p-AKT Ser473
(rather than total AKT) in a dose-dependent manner.
• In a series of acridone-2-carbohydrazides, substituted phenylethylidene derivatives were found to be more
potent than substituted benzylidene derivatives.
• Molecular docking study of compounds showed good binding affinity to AKT kinase.
• Compounds 8k, 8v and 9h caused G0/G1-phase cell-cycle arrest and led to apoptosis.
• The study identifies compound 8v as the most potent agent to inhibit cancer cell growth via suppressing the
activation of AKT signaling through inhibition of p-AKT Ser473 .
Supplementary data
To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/
suppl/10.4155/fmc-2022-0271
Author contributions
T Yadav: study conduction, synthesis, data collection, analysis and interpretation of results, draft manuscript preparation. M Kumar:
methodology suggestions for biological studies, suggestions and supervision, writing (reviewing and editing, critical review of
manuscript). S Bajaj: molecular docking study and manuscript editing. Y Mayur: conceptual design, manuscript reviewing and
editing, approval of final version. All authors reviewed the results and approved the final version of the manuscript.
Acknowledgments
The authors are thankful to the Department of Health Research for providing fellowship. They would also like to acknowledge the
FACS Central Facility, Chemical Engineering Department, IIT Powai, Mumbai for cell cycle and cell apoptosis analysis of samples.
References
Papers of special note have been highlighted as: • of interest; •• of considerable interest
1. Theivendren P, Kunjiappan S, Hegde YM et al. Importance of protein kinase and its inhibitor: a review. In: Protein Kinases – Promising
Targets for Anticancer Drug Research. Singh R (Ed.). IntechOpen, London, UK (2021).
2. Giamas G, Stebbing J, Vorgias CE, Knippschild U. Protein kinases as targets for cancer treatment. Pharmacogenomics 8(8), 1005–1016
(2007).
3. Garcı́a-Aranda M, Redondo M. Protein kinase targets in breast cancer. Int. J. Mol. Sci. 18(12), 2543 (2017).
4. Martorana F, Motta G, Pavone G et al. AKT inhibitors: new weapons in the fight against breast cancer? Front. Pharmacol. 12, 662232
(2021).
• Demonstrates the importance of development of AKT kinase inhibitors as new anticancer drugs.
5. Song M, Bode AM, Dong Z, Lee MH. AKT as a therapeutic target for cancer. Cancer Res. 79(6), 1019–1031 (2019).
• Demonstrates the importance of AKT kinase as a validated target for cancer therapy.
6. Kumar A, Rajendran V, Sethumadhavan R, Purohit R. AKT kinase pathway: a leading target in cancer research. ScientificWorldJournal
2013, 756134 (2013).
7. Gensicka-Kowalewska M, Cholewiński G, Dzierzbicka K. Recent developments in the synthesis and biological activity of
acridine/acridone analogues. RSC Adv. 7(26), 15776–15804 (2017).
8. Mayur YC, Zaheeruddin, Peters GJ, Lemos C, Kathmann I, Prasad VVSR. Synthesis of 2-fluoro N10-substituted acridones and their
cytotoxicity studies in sensitive and resistant cancer cell lines and their DNA intercalation studies. Arch. Pharm. (Weinheim) 342(11),
640–650 (2009).
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group
Design, synthesis & evaluation of acridone-2-carbohydrazide derivatives as p-AKT Ser473 kinase inhibitors Research Article
9. Hegde R, Thimmaiah P, Yerigeri MC, Krishnegowda G, Thimmaiah KN, Houghton PJ. Anti-calmodulin acridone derivatives modulate
vinblastine resistance in multidrug resistant (MDR) cancer cells. Eur. J. Med. Chem. 39(2), 161–177 (2004).
10. Velingkar VS, Dandekar VD. Design, synthesis and evaluation of substituted
N-(3-arylpropyl)-9,10-dihydro-9-oxoacridine-4-carboxamides as potent MDR reversal agents in cancer. Chinese J. Chem. 29(3),
504–510 (2011).
11. Singh P, Kaur J, Kaur P, Kaur S. Search for MDR modulators: design, syntheses and evaluations of N-substituted acridones for
interactions with p-glycoprotein and Mg2+ . Bioorg. Med. Chem. 17(6), 2423–2427 (2009).
12. Kumar R, Kaur M, Bahia MS, Silakari O. Synthesis, cytotoxic study and docking based multidrug resistance modulator potential
analysis of 2-(9-oxoacridin-10(9H)-yl)-N-phenyl acetamides. Eur. J. Med. Chem. 80, 83–91 (2014).
13. Boumendjel A, Macalou S, Ahmed-Belkacem A, Blanc M, Di Pietro A. Acridone derivatives: design, synthesis, and inhibition of breast
cancer resistance protein ABCG2. Bioorg. Med. Chem. 15(8), 2892–2897 (2007).
14. Hyafil F, Vergely C, Du Vignaud P. In vitro and in vivo reversal of multidrug resistance by GF120918, an acridonecarboxamide
derivative. Cancer Res. 53(19), 4595–4602 (1993).
15. Waltemate J, Ivanov I, Ghasemi JB et al. 10-(4-Phenylpiperazine-1-carbonyl)acridin-9(10H)-ones and related compounds: synthesis,
antiproliferative activity and inhibition of tubulin polymerization. Bioorg. Med. Chem. Lett. 32, 127687 (2021).
16. Ma W, Bi J, Zhao C, Gao Y, Zhang G. Design, synthesis and biological evaluation of acridone glycosides as selective BChE inhibitors.
Carbohydr. Res. 491, 107977 (2020).
17. Krokidis MG, Molphy Z, Efthimiadou EK et al. Assessment of DNA topoisomerase i unwinding activity, radical scavenging capacity,
and inhibition of breast cancer cell viability of N-alkylacridones and N,N -dialkyl-9,9 -biacridylidenes. Biomolecules 9(5), 177(2019).
18. Kamata J, Okada T, Kotake Y et al. Synthesis and evaluation of novel pyrimido-acridone, -phenoxadine, and -carbazole as topoisomerase
II inhibitors. Chem. Pharm. Bull. (Tokyo) 52(9), 1071–1081 (2004).
19. Gao C, Li S, Lang X et al. Synthesis and evaluation of 10-(3,5-dimethoxy)benzyl-9(10H)-acridone derivatives as selective telomeric
G-quadruplex DNA ligands. Tetrahedron 68(38), 7920–7925 (2012).
20. Singh H, Kaur M, Kaur H et al. Structural tuning of acridones for developing anticancer agents targeting dihydrofolate reductase. Bioorg.
Med. Chem. Lett. 29(19), 126631 (2019).
•• Patent describing new acridone derivatives as AKT kinase inhibitors for development of new anticancer drugs.
21. Halbrook JW, Kesicki EA, Burgess LE, Schlachter ST, Eary CT, Schiro JG. WO2004085418A2 (2004).
22. Verdaguer E, Jordà EG, Canudas AM et al. 3-Amino thioacridone, a selective cyclin-dependent kinase 4 inhibitor, attenuates kainic
acid-induced apoptosis in neurons. Neuroscience 120(3), 599–603 (2003).
23. Houghton PJ, Thimmaiah KN, Easton JB. WO2006094207A2 (2006).
•• Paper describing new acridone derivatives as AKT kinase inhibitors for development of new anticancer drugs.
24. Murahari M, Prakash KV, Peters GJ, Mayur YC. Acridone–pyrimidine hybrids – design, synthesis, cytotoxicity studies in resistant and
sensitive cancer cells and molecular docking studies. Eur. J. Med. Chem. 139, 961–981 (2017).
25. Yadav TT, Murahari M, Peters GJ, Mayur YC. A comprehensive review on acridone based derivatives as future anti-cancer agents and
their structure activity relationships. Eur. J. Med. Chem. 239, 114527 (2022).
26. Nitulescu GM, Margina D, Juzenas P et al. Akt inhibitors in cancer treatment: the long journey from drug discovery to clinical use
(review). Int. J. Oncol. 48(3), 869–885 (2016).
27. Guo Y, Jin Y, Qu B, Zhang Y, Che J, Dong X. An updated patent review of Akt inhibitors (2016–present). Expert Opin. Ther. Pat.
31(9), 837–849 (2021).
28. Kalinsky K, Hong F, McCourt CK et al. Effect of capivasertib in patients with an AKT1 E17K-mutated tumor: NCI-MATCH
subprotocol EAY131-Y nonrandomized trial. JAMA Oncol. 7(2), 271–278 (2021).
29. Andrikopoulou A, Chatzinikolaou S, Panourgias E et al. The emerging role of capivasertib in breast cancer. Breast 63, 157–167 (2022).
30. Alves CL, Ehmsen S, Terp MG et al. Co-targeting CDK4/6 and AKT with endocrine therapy prevents progression in CDK4/6 inhibitor
and endocrine therapy-resistant breast cancer. Nat. Commun. 12(1), 5112 (2021).
31. Helwa AA, El-Dydamony NM, Radwan RA, Abdelraouf SM, Abdelnaby RM. Novel antiproliferative agents bearing
morpholinopyrimidine scaffold as PI3K inhibitors and apoptosis inducers; design, synthesis and molecular docking. Bioorg. Chem. 102,
104051 (2020).
32. Mayur Y, Ahmad O, Prasad VVSR, Purohit M, Srinivasulu N, Kumar SMS. Synthesis of 2-methyl N10-substituted acridones as selective
inhibitors of multidrug resistance (MDR) associated protein in cancer cells. Med. Chem. 4(5), 457–465 (2008).
33. Sathish NK, Prasad VVSR, Raghavendra NM, Kumar SMS, Mayur YC. Synthesis of novel 1,3-diacetoxy-acridones as cytotoxic agents
and their DNA-binding studies. Sci. Pharm. 77(1), 19–32 (2009).
34. Rajendra VVSR, Reddy GD, Kathmann I, Amareswararao M, Peters GJ. Nitric oxide releasing acridone carboxamide derivatives as
reverters of doxorubicin resistance in MCF7/Dx cancer cells. Bioorg. Chem. 64, 51–58 (2016).
35. Vichai V, Kirtikara K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 1(3), 1112–1116 (2006).
36. Orellana E, Kasinski A. Sulforhodamine B (SRB) assay in cell culture to investigate cell proliferation. Bio Protoc. 6(21), e1924 (2016).
37. Chuang CH, Cheng TC, Leu YL, Chuang KH, Tzou SC, Chen CS. Discovery of AKT kinase inhibitors through structure-based virtual
screening and their evaluation as potential anticancer agents. Int. J. Mol. Sci. 16(2), 3202–3212 (2015).
38. Cui Z, Li X, Li L et al. Design, synthesis and evaluation of acridine derivatives as multi-target Src and MEK kinase inhibitors for
anti-tumor treatment. Bioorg. Med. Chem. 24(2), 261–269 (2016).
39. Chen J, Wang Y, Zhao D et al. Chrysin serves as a novel inhibitor of DGKα/FAK interaction to suppress the malignancy of esophageal
squamous cell carcinoma (ESCC). Acta Pharm. Sin. B 11(1), 143–155 (2021).
40. Han Y, Tian Y, Wang R et al. Design, synthesis and biological evaluation of thieno[3,2-d]pyrimidine derivatives containing aroyl
hydrazone or aryl hydrazide moieties for PI3K and mTOR dual inhibition. Bioorg. Chem. 104, 104197 (2020).
41. Ding S, Dong X, Gao Z et al. Design, synthesis and biological evaluation of novel N-(3-amino-4-methoxyphenyl)acrylamide derivatives
as selective EGFRL858R/T790M kinase inhibitors. Bioorg. Chem. 118, 105471 (2022).
42. Pozarowski P, Darzynkiewicz Z. Analysis of cell cycle by flow cytometry. Methods Mol. Biol. 281, 301–311 (2004).
43. Darzynkiewicz Z, Bedner E, Smolewski P. Flow cytometry in analysis of cell cycle and apoptosis. Semin. Hematol. 38(2), 179–193
(2001).
44. Lakshmanan I, Batra S. Protocol for apoptosis assay by flow cytometry using annexin V staining method. Bio Protoc. 3(6), 374 (2013).
45. Thimmaiah K, Ugarkar AG, Martis EF, Shaikh MS, Coutinho EC, Yergeri MC. Drug–DNA interaction studies of acridone-based
derivatives. Nucleosides Nucleotides Nucleic Acids 34(5), 309–331 (2015).
46. David-Cordonnier M, Laine W, Lansiaux A et al. Covalent binding of antitumor benzoacronycines to double-stranded DNA induces
helix opening and the formation of single-stranded DNA: unique consequences of a novel DNA-bonding mechanism. Mol. Cancer Ther.
4(1), 71–80 (2005).
47. Sahu G, Tiekink ERT, Dinda R. Study of DNA interaction and cytotoxicity activity of oxidovanadium(V) complexes with ONO donor
schiff base ligands. Inorganics 9(9), 66 (2021).
48. Ganeshpandian M, Loganathan R, Suresh E, Riyasdeen A, Akbarsha MA, Palaniandavar M. New ruthenium(II) arene complexes of
anthracenyl-appended diazacycloalkanes: effect of ligand intercalation and hydrophobicity on DNA and protein binding and cleavage
and cytotoxicity. Dalt. Trans. 43(3), 1203–1219 (2014).
49. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization
and multithreading. J. Comput. Chem. 31(2), 455–461 (2010).
50. RCSB Protein Data Bank. Home page. www.rcsb.org/ (Accessed 30 January 2023).
51. Voura M, Khan P, Thysiadis S et al. Probing the inhibition of microtubule affinity regulating kinase 4 by N-substituted acridones. Sci.
Rep. 9(1), 1676 (2019).
52. Abbass EM, Khalil AK, Mohamed MM, Eissa IH, El-Naggar AM. Design, efficient synthesis, docking studies, and anticancer evaluation
of new quinoxalines as potential intercalative Topo II inhibitors and apoptosis inducers. Bioorg. Chem. 104, 104255 (2020).
53. El-Adl K, Ibrahim MK, Alesawy MSI, Eissa IH. [1,2,4]Triazolo[4,3-c]quinazoline and bis([1,2,4]triazolo)[4,3-a:4 ,3 -c]quinazoline
derived DNA intercalators: design, synthesis, in silico ADMET profile, molecular docking and anti-proliferative evaluation studies.
Bioorg. Med. Chem. 30, 115958 (2021).
54. Morris GM, Huey R, Lindstrom W et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J.
Comput. Chem. 30(16), 2785–2791 (2009).
55. Shntaif AH, Khan S, Tapadiya G et al. Rational drug design, synthesis, and biological evaluation of novel
N-(2-arylaminophenyl)-2,3-diphenylquinoxaline-6-sulfonamides as potential antimalarial, antifungal, and antibacterial agents. Digit.
Chin. Med. 4(4), 290–304 (2021).
56. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry
friendliness of small molecules. Sci. Rep. 7, 42717 (2017).
57. Zhu H, Martin TM, Ye L, Sedykh A, Young DM, Tropsha A. QSAR modeling of rat acute toxicity by oral exposure. Chem. Res. Toxicol.
22(12), 1913–1921 (2009).
10.4155/fmc-2022-0271 Future Med. Chem. (Epub ahead of print) future science group