Evaluation of anti-proliferative and pro-apoptotic property of 7,3′-dihydroxyflavone against MCF-7 cell line using in-vitro, in-silico and gene expression analysis

 

Y. Jennifer Monica1, R. Kavitha2, Karthik3

13rdyear Post Graduate, Department of Pharmacology, Third floor, medical College block, SRMC and RI, Sri Ramachandra Institute of Higher Education and Research, No.1 Ramachandra Nagar Porur, Chennai - 600 116 Tamil Nadu, India.

2Professor and Head of The Department,  Department of Pharmacology, Third floor, medical College block, SRMC and RI, Sri Ramachandra Institute of Higher Education and Research, No.1 Ramachandra Nagar Porur, Chennai - 600 116 Tamil Nadu, India.

3Associate Professor, Department of Pharmacology, Third floor, medical College block, SRMC and RI, Sri Ramachandra Institute of Higher Education and Research, No.1 Ramachandra Nagar Porur, Chennai - 600 116 Tamil Nadu, India.

*Corresponding Author E-mail: jennifermonica945@gmail.com

 

ABSTRACT:

With approximately ten million deaths from cancer in 2020, cancer is a major cause of death worldwide. In 2020, around 25% of newly diagnosed cancer patients will have a breast cancer, which has the highest incidence rates. Furthermore, one-third of female cancer patients under 60 have breast cancer, and the disease accounts for 18% of cancer-related deaths.In- vitro, in- silico and gene expression studies were done  to assess the 7,3′-dihydroxyflavone's anti-proliferative and pro-apoptotic activity. First, we used the MTT assay to show that 7,3′-dihydroxyflavone had anti-proliferative properties. Second, we assessed the apoptotic activity, which was verified by the Propidium Iodide Assay and Annexin V, which showed cells at every stage of the process. Third, we investigated the gene expression level of p53, p21, and Nfkb by RT-PCR analysis. Fourth, we studied the binding affinity of 7,3′-dihydroxyflavone against p53, p21, and Nfkb by cellular docking and protein-ligand interaction studies. Molecular docking of 7,3′- dihydroxyflavone were performed against p53, p21, and Nkfb receptors by using AutoDock Vina. An ADMET predictions was performed to inspect the chance of 7,3′- dihydroxyflavone being anti-cancer drug.

 

KEYWORDS: 7,3'-dihydroxyflavone, MTT, p53, p21, Nfkb, RT-PCR, Molecular docking, ADMET.

 

 


 

INTRODUCTION: 

Cancer is a non- communicable diseases that are uncontrolled cell proliferation and overexcited cell division and differentiation1. The search for effective cancer treatments is a complex undertaking requiring a thorough comprehension of the complicated cellular mechanisms that underlies the aetiology and course of the illness.

 

In order to increase the survival rates of those afflicted by the disease, scientists are always working to develop safer and more effective medicines for cancer therapy, this is a dynamic and active area of scientific research2. Based on mRNA gene expression levels, breast cancer may be categorised into molecular subtypes that provide insights into new treatment modalities and patient stratifications that influence breast cancer patients' care. Treatment for breast cancer is complex and involves several different treatments to be carried out in different sequences, such as hormone therapy, biological therapies, radiation therapy, chemotherapy, and surgery3. In 2020,the worldwide mortality-to-incidence ratio for breast cancer was 0.30, indicating a favourable 5-year survival rate. The comparative survival rates for localised and regional breast cancer were 76.3% and 47.4%. respectively, in less developed nations like India 4,5. The toxicity of agents towards normal cells and tissues, along with the emergence of treatment resistance, lead to side effects that hinder the efficacy of anti-cancer therapies that target molecules6.

 

Pharmacokinetics and getting enough active medication into cancer cells in vivo are the  two most difficult aspects7. New anticancer drugs with unique mechanisms of action are constantly needed, as is the discovery of fresh potential cellular targets and effective treatment plans that target only cancer cells8. In this context, we predicted in our work that 7,3′- dihydroxyflavone would function as an anti-proliferative drug in breast cancertreatment with the ability to interact with Nfkb, p21, and p53. Better alignment to the allosteric area and a better fit within the binding pockets of p53, p21, and Nfkb were expected outcomes of the greater conformational flexibility provided by the amide bond, leading to increased binding affinity.

 

METHODOLOGY:

Cell Culture:

Cell Culture The cells were cultured in tissue flasks using a 1:1 combination of MEM (Minimum Essential media) and Ham's F12 media, enriched with foetal bovine serum and antibiotic, at a temperature of 37℃. Upon reaching a confluence density of 70 to 80%, the cells were trypsinized and seeded to a 96-well plate at a density of 1×106 cells per well. The plate was then incubated at 37℃ for 48 hours in a 5% CO2 incubator9.

 

MTT Assay for evaluation of Cell Proliferative Activity:

Dimethyl Sulfoxide (DMSO) was used to dissolve the chemical 7,3'-Dihydroxyflavone, whichwas subsequently added to the wellsat concentrations of 31.25μg/mL, 62.50μg/mL, 125μg/mL, and 500 μg/mL.Following which these samples were  placed in an incubator for a duration of 24hours.The untreated (Control) and treated cells exhibited morphological changes, which were identified and photographed using a digital inverted microscope at a magnification of 40X.Thereafter Phosphate Buffer Saline (PBS) was used to wash the cells. at a pH of 7.4, and then 20 microliters of MTT solution (5 milligrammes per millilitre in PBS) were introduced into each well. The resultant mixture was subjected to incubation for a duration of 3 hours. After a 3 hour incubation period, the formazan crystals produced by metabolically active cells were allowed to air-dry in a dark place for 30 minutes. Following that,in 100 microliters of DMSO  the crystals were dissolvedand the UV absorbance was quantified at a wavelength of 570nm using an UV-VIS Spectrophotometer. The authors10.

 

Annexin V-FITC/PI Apoptotic Assay for Measurement of Apoptosis:

Necrotic and Apoptotic cells were differentiated on the basis of Annexin VFluorescein Isothiocyanate and Propidium Iodide (Annexin V-FITC/PI) doublestaining. In order to estimate various phases of apoptosis, with the inhibitory concentration (IC50) of 7,3’-Dihydroxyflavone the  MCF-7 cells were treated and cultured for 24hours. Untreated cells cultured in the presence of 1% DMSO (v/v) were taken as the negative control. Following treatment, after rinsing the cells with 1X binding buffer, they were treated for 10 to 15 minutes at room temperature in a dark environment with 10 μL of Propidium Iodide (50 μg/mL in PBS) and 5 μL of Annexin V-FITC. Finally, the cells were visualised using a fluorescent microscope.11,12.

 

Gene Expression analysis of p21, p53, NFkB:

Assessment of the p21, p53, and NFkB gene expression, was done using real time PCR. Trizol Reagent (Sigma), a standardised procedure was used to separate the entire RNA. Using a PrimeScript first strand cDNA synthesis kit, 2μg of RNA were utilised for reverse transcription-based cDNA synthesis. (TakaRa, Japan). The targeted genes were amplified using specific primers. SYBR green dye and all of the necessary PCR components are included in the GoTaq® qPCR Master Mix (Promega), which was used to run the PCR reaction. Utilising a Biorad CFX96 PCR instrument, real-time PCR was carried out. The outcomes were analysed using the comparative CT technique, and Schmittgen and Livak's (2008)  2−∆∆CT method was used to calculate the fold change13.

 

Molecular Docking:

Using AutoDock Vina 4.2 software, molecular docking studies were carried out for the 7,3'-dihydroxyflavone inside the active sites of p53 (PDB: 3DCY), p21 (PDB: 2AST), and nuclear factor kappa B (PDB: 1NFK). It was used to conduct a molecular docking study of 7,3'-dihydroxyflavone on the active site of human p21, p53, and NFkB14,15. The threedimensional crystal Nucleotide kappa B (PDB: 1NFK), p53 (PDB: 3DCY), and p21 (2AST) structures have been retrieved from http://www.rcsb.org, the Protein Data Bank. Using the Dock Prep tool of the UCSF Chimaera programme16, the structures of three proteins were generated to eliminate heteroatoms and water molecules all of the receptor structure, insertion of polar hydrogens, and repair of missing side chain atoms. Using Open Babel, the 7,3'-dihydroxyflavone SDF format was changed to the pdb format17. Using MGLTools 1.1.6, the receptor and ligands wereconverted to pdbqt format. The coordinates of the ligand in the PDB record served as the centre of the docking search space, which had values of X=20, Y=20, and Z=20. The maximum binding affinity with receptors was determined by considering the binding pose that adopts the lowest energy conformation. We were able to locate the protein binding site by molecular visualising the 7,3′- dihydroxyflavone in its lowest energy conformations. The software called Discovery Studio 2021 was used to analyze the protein-ligand interaction18.

 

In-Silico Drug-likeness and ADMET Study:

Using the pkCSM online server, the drug-likeness and ADMET analysis were carried out.(https://biosig.lab.uq.edu.au/pkcsm/) (19).

 

RESULTS:

The findings indicate that the 7,3′-dihydroxyflavone inhibited the proliferation of cancerous cells in MCF7 cell line. The outcomes obtained from the MTT assay of 7,3′-dihydroxyflavone against MCF-7cell line breast of cancer to validate the anti-cancer activity of 7,3′-dihydroxyflavone at various concentration are shown in Table 1 and Doxorubicin was used a positive control.

 

Table No.1: Summary of Anti-Proliferative Effect by MTT Assay

Concentrations (μg/mL)

Average of Absorbance

Cell Viability (%)

Percentage of Inhibition

Control (No drug treatment)

0.753

100

0

31.25

0.691

91.766

8.234

62.5

0.571

75.830

24.170

125

0.306

40.637

59.363

250

0.190

25.232

74.768

500

0.090

11.886

88.114


 

Figure No.1: Anti-Proliferative Activity of 7,3 Dihydroxyflavone

 

Figure No.2: IC50 graph of 7,3 Dihydroxyflavone


 

Figure No. 3: Cytotoxicity effect of 7,3 Dihydroxyflavone

 

Annexin V-FITC/PI Apoptotic Assay for Measurement of Apoptosis:

PI staining and annexin V-FITC analysis of apoptosis.The fluorophores PI and annexin V were used to investigate the various stages of apoptosis.The test item at the IC50 levels effectively stimulates the translocation of phosphatidylserine (annexin V+), according to the results of the annexin V-FITC staining procedure. Green FITC stain,

 indicating the presence of early apoptosis in MCF-7 cells after 24hours21. Exposure of phosphatidylserine to the cell's outer membrane is one of the distinguishing characteristics of apoptosis.22. The exposed test item cells exhibited the adequate level of PS externalisation, or green fluorescence, on the plasmamembrane, necessary for the initiation of apoptotic processes.23. Most cells showed positive responses to both PI and annexin V (annexin V+/PI+), suggesting the existence of late-apoptotic cells A high percentage of PI and annexin V staining were seen on the test plate, indicating mid-phase apoptosis(24).A minor percentage of the treated plate's cells also showed good annexin-positive and strong PI-positive fractions, which were raised to intense red nuclear staining without any green stain, signifying the late stage of apoptosis. Annexin V and PI (annexin V-/PI-) did not stain control MCF-7 cells, indicating that these cells were alive and had not undergone apoptosis. Thus, it was evident from the results that the test chemical, 7,3-Dihydroxyflavone, caused MCF-7 cells to undergo apoptosis.

 


Figure No.4: Compound at IC50 successfully induced apoptosis.Phosphatidylserine translocation (labeled by FITC/Annexin V+ green stain). Loss of membrane integrity (labeled by red fluorescent stain (PI+) of nuclear and chromosome) thus indicating ,induction of apoptosis in MCF-7 cells at 24 hours. (A) Control MCF-7 cells were neither stained by both PI nor Annexin V (Annexin V-/PI-). The cells did not undergo apoptosis and were viable. (B) High percentage of Annexin V, shown by FITC green stain, indicating early apoptosis (C) High fraction of PI accompanying by Annexin v staining and indicating the Mid-phase apoptosis, and (D) Strong PI-positivity , increased further to a strong red staining in the nuclear area, signifying the last stage of apoptosis. These findings clearly imply that 7,3′-dihydroxyflavone,promotes apoptosis at all stages in MCF-7 cells.

 


Gene Expression analysis of p21, p53, NFkB:

Effects on mRNA expression of p21, p53, NFkB on MCF7. The administration of 7,3′-dihydroxyflavone up-regulated the mRNA levels of p21, p53, whereas downregulated NFkB. Moreover, we found that there are significant differences in the gene expression levels (Figure 3). The verification of p21, p53, and NFkB mRNA expression in MCF7 cells by RT-qPCR among control groups (doxorubicin, negative control). The standard error of the mean (SEM) is shown by error bars. Gene products for NFkB, p21, and p53 have been connected to pathways leading to programmed cell death. We have examined the expression of p53, p21, and NFkB in a number of 1 1 1 4 31 32 39 human breast cancer cell lines. We discovered a negative connection between NFkB, p21, and p53 expression. According to our findings, in breast cancer cell lines, 7,3′- dihydroxyflavone downregulated NFkB and elevated the p53 and p21 genes.

 

(A)

(B)

(C)

Figure No.5: Gene Expression Results. (A) Effect of 7,3′-dihydroxyflavone (187.801 μg/ml) on p53 gene expression in breast cancer cell line (MCF-7). (B) Effect of 7,3′-dihydroxyflavone (187.801 μg/ml) on p21 gene expression in breast cancer cell line (MCF-7). (C) Effect of 7,3′-dihydroxyflavone (187.801 μg/ml) on p21 gene expression in breast cancer cell line (MCF-7). The results are given as a fold change from the control after the target gene expression is normalised to GAPDH mRNA expression. The mean + SEM of three separate observations is shown by each bar. ‘*’ indicates the statistical significance, at the p<0.05 between the drug treatment and control groups.

Molecular Docking:

Crystallographic structures of human p53 (PDB: 3DCY), p21 (PDB: 2AST), and nuclear factor kappa B (PDB: 1NFK) were retrieved from the Protein Data Bank in order to determine the molecular mechanism involved in the anti-proliferative action of the 7,3′-dihydroxyflavone. Molecular docking requires a certain level of binding affinity between the ligand and protein. Molecular docking was performed by computing the affinities between 7,3′-dihydroxyflavone and protein targets. We assessed the 7,3′-dihydroxyflavone's binding affinities and modalities of binding to each protein using the open-source AutoDock Vina programme, which optimises docking by utilising a scoring function Lamarckian Genetic algorithm. The conformation binding mode of the lowest Gibbs free energy of binding (ΔG) was used to evaluate the interactions between the ligand and the protein.

 

The amino acids in p21 involved in this interaction are ARG 2167 and MET 3058 also by hydrogen bonding and hydrophobic interactions for 7,3′- dihydroxyflavone (Figure 6B). The amino acids in Nfkb involved in this interaction are GLY 166, LEU 176, GLN 177, and ARG 228 by hydrogen bonding and hydrophobic interaction for 7,3′-dihydroxyflavone (Figure 6C). The catalytic core of protein targets, which includes amino acids p53, p21, and Nfkb, may account for the inhibitory impact of 7,3′-dihydroxyflavone.

 

Figure No.6: Ribbon representation of the binding mode of 7,3′-dihydroxyflavone to human p53. The protein is represented as rainbow ribbons. The ligand, 7,3′-dihydroxyflavone is was represented in stick model.

 

Figure No.7: Ribbon representation of the binding mode of 7,3′-dihydroxyflavone to human p21. The protein is represented as rainbow ribbons. The ligand, 7,3′-dihydroxyflavone is was represented in stick model.

 

Figure No.8: Ribbon representation of the binding mode of 7,3′-dihydroxyflavone to human Nfkb. The protein is represented as rainbow ribbons. The ligand, 7,3′-dihydroxyflavone is was represented in stick model.

 

(A)                                                   (B)

(C)

Figure No.9: 2D Interaction Plot of Docked Poses. (A) p53, (B) p21, and (C) Nfkb.The structure and interactions of 7,3′-dihydroxyflavone with Nfkb, p21, and p53 active sites. The primary source of binding energy to the 7,3′-dihydroxyflavone's binding affinity for p53, p21, and Nfkb was shown to be this substructure. For hydrogen bonds, interactions are displayed in green, for π-alkyl in pink, and for cation-π in orange.

 

Table No.2: Binding energies from in silico docking towards human p53, p21, and Nfkb

Receptors

Minimum Binding Energies (Kcal/mol)

p53

-7.0

p21

-7.4

Nfkb

-6.4

 

Drug-likeness and ADMET Study:

When it comes to potential drug like candidates, 7,3′-dihydroxyflavone conforms to Lipinski's "Rule of Five," also known as Pfizer's rule of five.(Table 3). These guidelines enable us to assess 7,3′-dihydroxyflavone's potential as a medication.The majority of oral active drugs with excellent bioavailability are said to meet all four requirements:  number of hydrogen bond acceptors ≥ 10, number of hydrogen bond acceptors ≥ 500 Da, limited lipophilicity represented as logP ≥ 5, and number of hydrogen bonds.

 

Table No.3: Drug-likeness Parameters

Descriptor

Value

Molecular Weight

254.241

LogP

2.8712

No. of Rotatable Bonds

1

No. of Acceptors

4

No. of Hydrogen Bond Donors

2

Surface Area

107.725

 

When using a hit-to-lead strategy, pharmacokinetics (PK) characteristics are crucial in the hit compound selection process. Therefore, we used the pkCSM programme to examine these properties in silico. The results of this evaluation revealed that 7,3′- dihydroxyflavone has 94.22% of bioavailability, -3.605 mol/L (water solubility), skin permeability of -2.751 log Kp, binding with P-glycoprotein substrate, no interaction with P-glycoprotein I and II inhibitor, In theview of distribution, Volume of distribution (VDss) (human) (0.063 L/kg), Fraction unbound (human) (0.146 Fu), BBB permeability of -0.057 log BB, and CNS permeability of-1.927 log PS. The metabolism prediction showing inhibition towards CYP1A2 inhibitior, CYP2C19 inhibitior, CYP2C9 inhibitior, no inhibition towards CYP2D6 substrate,CYP3A4 substrate, CYP2D6 inhibitior, CYP3A4 inhibitior. The excretion process shows 0.242ml/min/kg of total clearance, and no interaction with Renal OCT2 substrate. In terms of toxicity, it shows no toxicity towards AMES, hERG I inhibitor, hERG II inhibitor, Hepatotoxicity, Skin Sensitisation, T.Pyriformis toxicity (0.654μL), Minnow toxicity (1.178mM). The Maximum tolerated dose (human) (0.175 log mg/kg/day), Oral Rat Acute Toxicity (LD50) (2.208mol/kg), and Oral Rat Chronic Toxicity (1.231 log mg/kg_bw/day) was also calculated. The drug-likeness prediction of 7,3′-dihydroxyflavone was summarized in Table 3.

 

DISCUSSION:

Worldwide, over a million women receive a breast cancer diagnosis each year, and more than half of them will pass away as a result of the illness. The most frequent cancer and the main reason why women die from cancer is breast cancer. The general prognosis for breast cancer has improved due to widespread screening programmes and the introduction of novel therapies. For women with late-stage or metastatic breast cancer, the average annual survival rate is still rather poor. Five years after diagnosis, just 35 percent of women with advanced breast cancer still live. When breast cancer is detected at a late stage, when there is little chance of recovery, one-third of women receive this diagnosis 32. In both industrialised and developing nations, breast cancer is regarded as one of the worst illnesses. To design and create more selective, effective, and low-toxicity therapeutic medicines for the treatment of cancer, ongoing research is necessary34. The increased ability to diagnose and treat patients has resulted in a steady rise in survival rates. Many physical and psychological changes are brought about by the diagnosis and treatment of cancer31. The survival rates of breast cancer in India are low because the detection takes place late. The only way to change these numbers is by increasing awareness35,36,37,38,39,40.

 

A collection of cells that proliferate uncontrollably and grow malignantly is called cancer. Currently, chemotherapy, radiation therapy, gene therapy, immunotherapy, molecularly targeted therapy, and surgery are the main forms of cancer treatment. However, some tumour characteristics have thus far hampered the therapeutic effectiveness of these treatments. Unusual signalling pathway activation contributes to tumour pathogenesis and is essential for the development, advancement, and recurrence of malignancies. Cancer patients now have better results thanks to targeted medicines that target oncogenic signalling effectors25. It is believed that 50% of cancers especially breast cancer are caused by gene alteration of cell cycle regulating genes, which continue to be the most frequently associated genes in many common human cancers.Higher eukaryotes have developed many checkpoint mechanisms to detect and respond to cellular disruption.Modifications to checkpoint mechanisms compromise the integrity of the genome, promote the development of cancer, and significantly reduce the efficacy of anticancer treatments.26. The tumour suppressor protein p53 activates several genes that limit development or cause apoptosis, which controls the DNA damage-induced checkpoint.The 165 amino acid protein known as p21 mediates the p53-dependent G1 growth arrest. Additionally, earlier studies have shown that the p21 protein acts as a growth suppressor of cancers by inducing cell cycle arrest in response to a variety of stimuli.


Superior modulator of many tumour suppressor pathways, facilitating anti-proliferative effects not mediated by the p53 tumour suppressor pathway27. According to recent studies, p21 may sometimes promote cellular proliferation and carcinogenicity. Although p21 is typically dysregulated in human cancers, its expression suggests that it could still be functional. Depending on the cellular milieu, either as an oncogene or a tumour suppressor.The simple hypothesis that p21 serves as a tumour suppressor has been called into question by the finding that it may exhibit carcinogenic properties. Several human cancers overexpress p21, including those of the breast, prostate, cervical, and squamous cell carcinomas. The grade, invasiveness, and aggressiveness of the tumour are often positively correlated with this overexpression, which is a poor prognostic indicator28. An essential signalling system involved in the aetiology and management of cancer is NFκB. Studies on the NFκB-signaling pathway for targeted cancer therapy have revealed excessive activation of this pathway in a variety of tumour         tissues 29.

 

In a related work, Mohammed et al. (2023) designed a novel medication by molecular docking with ERa and applying the Structure Based medication Design (SBDD) technique to predict the drug's ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) profile. The software utilised for result analysis and prediction were BIOVIA Discovery Studio Visualizer 2020, SwissADME, and GOLD (Genetic Optimisation for Ligand Docking)30. In an another study, the author assessed the cytotoxic activity of sulfonamide derivatives against a panel of cell lines representing breast cancer, lung cancer, and NCI-H23, NCI-H522, as well as one normal cell line, HEK-293T (a human embryonic kidney cell line); the results were compared with those of the standard medication, doxorubicin33.

 

CONCLUSION:

In conclusion, we established the anti-cancer properties of 7,3′-dihydroxyflavone. The compound 7,3′-dihydroxyflavone were assessed in vitro using the human cancer cell line MCF7. The other experimental molecular studies were validated by the molecular docking research. 7,3′-dihydroxyflavones have the potential to be powerful anticancer candidates since they can upregulate the expression of p53 and p21 and downregulate NFkB.

 

CONFLICT OF INTEREST:

The authors have no conflicts of interest regarding this investigation.

 

REFERENCES:

1.      Madaminov A, Khasanov A, Khatamov S, Abdurakhmonov O, Amonov A, Shukurov Z, Khudayorov M, Bekmirzaev R, Nishonboev L. Cancer-dysregulation of the cell cycle and transduction of cascade signals. Romanian Journal of Rhinology. 2021; Jul; 11(43): 90-100.

2.      Krzyszczyk P, Acevedo A, Davidoff EJ, Timmins LM, Marrero-Berrios I, Patel M, White C, Lowe C, Sherba JJ, Hartmanshenn C, O'Neill KM, Balter ML, Fritz ZR, Androulakis IP, Schloss RS, Yarmush ML. The growing role of precision and personalized medicine for cancer treatment. Technology (Singap World Sci). 2018; Sep-Dec; 6(3-4): 79-100. doi: 10.1142/S2339547818300020. Epub 2019 Jan 11. PMID: 30713991; PMCID: PMC6352312.

3.      Smolarz B, Nowak AZ, Romanowicz H. Breast Cancer-Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature). Cancers (Basel). 2022; May 23; 14(10): 2569. doi: 10.3390/cancers14102569. PMID: 35626173; PMCID: PMC9139759.

4.      Luo C, Li N, Lu B, Cai J, Lu M, Zhang Y, Chen H, Dai M. Global and regional trends in incidence and mortality of female breast cancer and associated factors at national level in 2000 to 2019. Chin Med J (Engl). 2022; Jan 5; 135(1): 42-51. doi: 10.1097/CM9.0000000000001814. PMID: 34593698; PMCID: PMC8850868.

5.      Sharma R. Global, regional, national burden of breast cancer in 185 countries: evidence from GLOBOCAN 2018. Breast Cancer Res Treat. 2021; Jun; 187(2): 557-567. doi: 10.1007/s10549-020-06083-6. Epub 2021 Jan 30. PMID: 33515396.

6.      Nikolaou M, Pavlopoulou A, Georgakilas AG, Kyrodimos E. The challenge of drug resistance in cancer treatment: a current overview. Clin Exp Metastasis. 2018; Apr; 35(4): 309-318. doi: 10.1007/s10585-018-9903-0. Epub 2018 May 24. PMID: 29799080.

7.      Ruiz-Garcia A, Bermejo M, Moss A, Casabo VG. Pharmacokinetics in drug discovery. J Pharm Sci. 2008; Feb; 97(2): 654-90. doi: 10.1002/jps.21009. PMID: 17630642.

8.      Cui W, Aouidate A, Wang S, Yu Q, Li Y, Yuan S. Discovering Anti-Cancer Drugs via Computational Methods. Front Pharmacol. 2020; May 20; 11: 733. doi: 10.3389/fphar.2020.00733. PMID: 32508653; PMCID: PMC7251168.

9.      Mukherjee S, Malik P, Mukherjee TK. Common Reagents and Medium for Mammalian Cell Culture. InPractical Approach to Mammalian Cell and Organ Culture 2023; Jan 20: 137-185). Singapore: Springer Nature Singapore.

10.   Bahuguna A, Khan I, Bajpai VK, Kang SC. MTT assay to evaluate the cytotoxic potential of a drug.  Bangladesh Journal of Pharmacology. 2017; Apr 8; 12(2): 115-8.

11.   Chen S, Cheng AC, Wang MS, Peng X. Detection of apoptosis induced by new type gosling viral enteritis virus in vitro through fluorescein annexin V-FITC/PI double labeling. World J Gastroenterol. 2008 Apr 14; 14(14): 2174-8. doi: 10.3748/wjg.14.2174. PMID: 18407590; PMCID: PMC2703841.

12.   Hwang SY, Cho SH, Lee BH, Song YJ, Lee EK. Cellular imaging assay for early evaluation of an apoptosis inducer. Apoptosis. 2011; Oct; 16(10): 1068-75. doi: 10.1007/s10495-011-0636-7. PMID: 21830083.

13.   Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008; 3(6): 1101-8. doi: 10.1038/nprot.2008.73. PMID: 18546601.

14.   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. 2010; Jan 30; 31(2): 455-61. doi: 10.1002/jcc.21334. PMID: 19499576; PMCID: PMC3041641.

15.   Butt SS, Badshah Y, Shabbir M, Rafiq M. Molecular Docking Using Chimera and Autodock Vina Software for Nonbioinformaticians. JMIR Bioinform Biotechnol. 2020; Jun 19; 1(1): e14232. doi: 10.2196/14232. PMID: 38943236.

16.   Huang CC, Meng EC, Morris JH, Pettersen EF, Ferrin TE. Enhancing UCSF Chimera through web services. Nucleic Acids Res. 2014; Jul; 42(Web Server issue):W478-84. doi: 10.1093/nar/gku377. Epub 2014 May 26. PMID: 24861624; PMCID: PMC4086125.

17.   O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: An open chemical toolbox. J Cheminform. 2011; Oct 7; 3: 33. doi: 10.1186/1758-2946-3-33. PMID: 21982300; PMCID: PMC3198950.

18.   Jejurikar BL, Rohane SH. Drug designing in discovery studio.

19.   Pires DE, Blundell TL, Ascher DB. pkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph-Based Signatures. J Med Chem. 2015; May 14; 58(9): 4066-72. doi: 10.1021/acs.jmedchem.5b00104. Epub 2015 Apr 22. PMID: 25860834; PMCID: PMC4434528.

20.   Mariño G, Kroemer G. Mechanisms of apoptotic phosphatidylserine exposure. Cell Res. 2013; Nov; 23(11): 1247-8. doi: 10.1038/cr.2013.115. Epub 2013 Aug 27. PMID: 23979019; PMCID: PMC3817543.

21.   Palma PF, Baggio GL, Spada C, Silva RD, Ferreira SI, Treitinger A. Evaluation of annexin V and Calcein-AM as markers of mononuclear cell apoptosis during human immunodeficiency virus infection. Braz J Infect Dis. 2008; Apr; 12(2): 108-14. doi: 10.1590/s1413-86702008000200003. PMID: 18641846.

22.   Schlegel RA, Williamson P. Phosphatidylserine, a death knell. Cell Death Differ. 2001; Jun; 8(6): 551-63. doi: 10.1038/sj.cdd.4400817. PMID: 11536005.

23.   Kim YE, Chen J, Langen R, Chan JR. Monitoring apoptosis and neuronal degeneration by real-time detection of phosphatidylserine externalization using a polarity-sensitive indicator of viability and apoptosis. Nat Protoc. 2010; Aug; 5(8): 1396-405. doi: 10.1038/nprot.2010.101. Epub 2010; Jul 8. PMID: 20671723; PMCID: PMC4340478.

24.   Lugli E, Troiano L, Ferraresi R, Roat E, Prada N, Nasi M, Pinti M, Cooper EL, Cossarizza A. Characterization of cells with different mitochondrial membrane potential during apoptosis. Cytometry A. 2005; Nov; 68(1): 28-35. doi: 10.1002/cyto.a.20188. PMID: 16184612.

25.   Baudino TA. Targeted Cancer Therapy: The Next Generation of Cancer Treatment. Curr Drug Discov Technol. 2015; 12(1): 3-20. doi: 10.2174/1570163812666150602144310. PMID: 26033233.

26.   Engeland K. Cell cycle regulation: p53-p21-RB signaling. Cell Death Differ. 2022; May; 29(5): 946-960. doi: 10.1038/s41418-022-00988-z. Epub 2022 Mar 31. PMID: 35361964; PMCID: PMC9090780.

27.   Rane CK, Minden A. P21 activated kinase signaling in cancer. Semin Cancer Biol. 2019; Feb; 54: 40-49. doi: 10.1016/j.semcancer.2018.01.006. Epub 2018 Jan 9. PMID: 29330094.

28.   Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009; Jun; 9(6): 400-14. doi: 10.1038/nrc2657. PMID: 19440234; PMCID: PMC2722839.

29.   Xia L, Tan S, Zhou Y, Lin J, Wang H, Oyang L, Tian Y, Liu L, Su M, Wang H, Cao D, Liao Q. Role of the NFκB-signaling pathway in cancer. Onco Targets Ther. 2018; Apr 11; 11: 2063-2073. doi: 10.2147/OTT.S161109. PMID: 29695914; PMCID: PMC5905465.

30.   Mohammed F. Al-Owaidi, Monther F. Al Ameri. Docking Study, ADMET Profiling of the Designed 3-Chlorobenzo[b]thiophene-2-Carbonyl Chloride Derivatives: Promising Anti-Breast Cancer Activity. Research Journal of Pharmacy and Technology. 2023; 16(2): 901-7.

31.   Sunita Srivastava, Anil Kumar. Breast Cancer Survivorship among Indian Women: An Overview. Asian Journal of Nursing Education and Research. 2022; 12(3): 262-6.

32.   V. N. Dange, S. J. Shid, C.S. Magdum, S.K. Mohite. A Review on Breast cancer: An Overview. Asian J. Pharm. Res. 2017; 7(1): 49-51.

33.   Sarav A. Desai , Prakash S. Sukhramani, Maulik P. Suthar, Vipul P. Patel. Biological Cytotoxicity Evaluation of Sulfonamide Derivatives as Anti-Lung and Anti-Breast Cancer Activity. Asian J. Research Chem. 2011; 4(4): April, 671-677.

34.   Balgovind, Omprakash Goshain. Benzimidazole: A Promising target for the development of novel drug molecules against breast cancer. Asian Journal of Research in Chemistry. 2023; 16(4): 303-0.

35.   Arti Tiwari, Maharaj Singh, Bharati Sahu. Risk Factors for Breast Cancer. International Journal of Nursing Education and Research. 2022; 10(3): 276-2.

36.   GK Sudhakar, Vasudev Pai, Arvind Pai. An overview on current Strategies in Breast Cancer Therapy. Research J. Pharmacology and Pharmacodynamics. 2013; 5(6): 353-355.

37.   M Lavanya, Asish Bhaumik, A Gopi Reddy, Ch Manasa, B Kalyani, S Sushmitha. Evaluation of Anticancer Activity of Ethanolic and Ethylacetoacetate Extracts of Sweet Cherry Against Human Breast Cancer Cell Line MCF-7. Research J. Pharmacology and Pharmacodynamics. 2016; 8(2): 65-70.

38.   Pushpa A. Karale, Mahesh A. Karale, Mokshada C. Utikar. Advanced Molecular Targeted Therapy in Breast Cancer. Res. J. Pharmacology and Pharmacodynamics. 2018; 10(1): 29-37.

39.   N. Habeela Jainab,, M. K. Mohan Maruga Raja. In Silico Molecular Docking Studies on the Chemical Constituents of Clerodendrum phlomidis for its Cytotoxic Potential against Breast Cancer Markers. Research J. Pharm. and Tech. 2018; 11(4): 1612-1618.

40.   Fadilah Fadilah, Lowilius Wiyono, Brenda Cristie Edina, Risya Amelia Rahmawati, Linda Erlina, Aryo Tedjo, Rafika Indah Paramita. In Silico Study and In Vitro test of Extract Kaempferia pandurata Roxb. as Anti ER (+) Breast Cancer Cell Line MCF-7. Research J. Pharm. and Tech. 2019; 12(5): 2391-2395.

 

 

 

 

Received on 08.03.2024      Revised on 19.07.2024

Accepted on 27.09.2024      Published on 28.01.2025

Available online from February 27, 2025

Research J. Pharmacy and Technology. 2025;18(2):481-488.

DOI: 10.52711/0974-360X.2025.00073

© RJPT All right reserved

 

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License.