5-O-Acetylpinostrobin derivatives as Estrogen-α inhibitors: Molecular docking and Pharmacokinetic analysis

 

Anita Puspa Widiyana1,2, Tri Widiandani3*, Siswandono Siswodihardjo3,4

1Doctoral Program of Pharmaceutical Sciences, Faculty of Pharmacy,

Universitas Airlangga, Surabaya 60155, Indonesia.

2Department of Pharmacy, Faculty of Medicine,

Universitas Islam Malang, Malang 65144, Indonesia.

3Department of Pharmaceutical Sciences, Faculty of Pharmacy,

Universitas Airlangga, Surabaya 60155, Indonesia.

4Department of Pharmacy, Faculty of Pharmacy,

Institut Ilmu Kesehatan Bhakti Wiyata, Kediri 64100, Indonesia.

*Corresponding Author E-mail: tri-w@ff.unair.ac.id

 

ABSTRACT:

Estrogen receptor alpha (ER-α) is an attractive and important target in breast cancer tissue development and a trigger for carcinogenesis. More than 70–75% of most breast cancers are ER-α. However, due to problems with ER-α inhibitors as major challenges related to intrinsic resistance, recurrent metastasis, additive effects, and estradiol antagonists, it’s necessary to identify new effective and selective inhibitor compounds. The aim of this study was to predict the anticancer activity, pharmacokinetics, and toxicity of twenty 5-O-acetylpinostrobin derivatives. The method used for activity prediction was the molecular docking approach with the Molegro Virtual Docker. While pharmacokinetics and toxicity (ADMET) studies with pkCSM online. The results of the molecular docking study showed a rerank score value of twenty 5-O-acetylpinostrobin derivatives of -113.8310 to -71.7388 kcal/mol. Eight compounds (AP-10-12, 14-16, and 19-20) had a smaller rerank score than pinostrobin and native ligand. The similarity of amino acid residue interactions such as a donor hydrogen bond between the oxygen atom of the chroman-4-on ring and Arg 394 as well as a steric interaction between Glu 353 and the C-aromatic of the chroman-4-on ring. The results of pharmacokinetics and toxicity studies showed that all compounds derived from 5-O-acetylpinostrobin have good absorption, distribution, metabolism, and excretion, as well as low toxicity. The conclusion of this study was that compound codes AP-10, 11, 14, 15, 19, and 20 have greater predicted breast anticancer activity, better pharmacokinetics, and lower toxicity than pinostrobin and 4-hydroxytamoxifen. Therefore, these six compounds have the potential to be developed further.

 

KEYWORDS: ADMET, 5-O-acetylpinostrobin, Breast cancer, Estrogen receptor alpha, Molecular docking.

 

 


 

INTRODUCTION: 

The cancer mortality rate is one of the main causes and a serious concern for the world community1,2,3. Breast cancer ranks second in the world, and the increase in incidence is getting higher every year4,5. Based on data from the International Agency for Research on Cancer (WHO) in 2020, 15.5% of deaths occurred in women6.

 

The onset of breast cancer originates from cells in the ductal and lobular layers, as well as some tissues in small amounts7,8. Overexpression of estrogen receptor α (ERα) causes 70–70% of breast cancers. Estrogen binds to ERα, which forms an active receptor complex, causing cell proliferation9,10. In general, breast cancer treatment can be achieved through the processes of radiotherapy, surgery, hormone administration, and chemotherapy11,12. The various side effects of breast cancer therapy and the limitations of anti-breast cancer drugs create challenges and urgent needs in the development of drugs that are more selective and have high activity as therapy13. The development of bioactive compounds derived from herbal plants can be one solution to obtaining safer active compounds14.

 

Nowadays, many structural modifications of pinostrobin (P) have been developed as a marker compound of Boesenbergia pandurata rhizome. The activities of pinostrobin include anti-cancer, antibacterial, anti-inflammatory, antioxidant, antiviral, and others. The development of pinostrobin has increased over the decades as an anti-cancer drug candidate15. However, the research progress remained slow, and the breast cancer activity was low when compared to currently available drugs16. The existence of structural modifications aimed at improving the lipophilic, electronic, and steric properties of pinostrobin, which increase its interaction with the target receptor. Several studies have proved that the addition of prenyl groups to pinostrobin can inhibit MCF-7 cancer cells better than pinostrobin17. Modification of pinostrobin in the hydroxyl group with the acyl group was predicted to have activity as a breast anticancer through HER-2 inhibition18. Meanwhile, there has been no research related to the potential modification of pinostrobin hydroxyl groups with acyl groups in producing 5-O-acetylpinostrobin derivatives. Therefore, computational chemistry strategies that reduce the cost and time of laboratory activities are highly attractive to be carried out in obtaining new compounds that are strong and selective19

 

Computational chemistry models are one of the most efficient and powerful methods for studying the prediction of activity, pharmacokinetics, and toxicity20,21. Activity prediction by molecular docking to predict the accuracy of the ligand with the active side of the target protein by indicating the quantitative prediction of the rerank score between the ligand-receptor complex22. Increasing drug failures in phase 1 clinical trials make pharmacokinetic and toxicity studies important23,24. The goal is to provide a selection of new compounds with good pharmacokinetic properties that are safer at the onset of drug discovery25. In this study, computational chemistry studies, including molecular docking, pharmacokinetic prediction, and toxicity, were performed. Therefore, the aim of this study was to evaluate the predicted activity, pharmacokinetics, and toxicity of twenty potential 5-O-acetylpinostrobin derivatives as ERα inhibitors.

MATERIALS AND METHODS:

Materials:

The tools used were a Fujitsu AH554 computer, NVIDIA®, Core(TM) i7, CPU @ 2.20 GHz, and 16 GB of RAM. The software used included Molegro Virtual Docker (MVD) v. 6, ChemBio Draw 2D, and 3D v. 20.1.1. The websites used were the pkCSM web (https://biosig.lab.uq.edu.au/pkcsm/) and Smile Translator (https://cactus.nci.nih.gov/translate/). The research materials consisted of twenty 5-O-acetylpinostrobin derivatives such as 5-O-4-(dimethylamino) benzoylpinostrobin (AP-1), 5-O-benzoylpinostrobin (AP-2), 5-O-cyclohexanecarbonylpinostrobin (AP-3), 5-O-2-phenylacetylylpinostrobin (AP-4), 5-O-cyclopentanecarbonylpinostrobin (AP-5), 5-O-3-phenylpropanoylpinostrobin (AP-6), 5-O-cyclobutanecarbonylpinostrobin (AP-7), 5-O-2-(methylsulfonyl) acetylpinostrobin (AP-8), 5-O-cyclopropanecarbonylpinostrobin (AP-9), 5-O-pivalylpinostrobin (AP-10), 5-O-2-(methylthio) acetylpinostrobin (AP-11), 5-O-2,2-dichloroacetylpinostrobin (AP-12), 5-O-isobutonylpinostrobin (AP-13), 5-O-propionylpinostrobin (AP-14), 5-O-3,3,3-trifluoropropanoylpinostrobin (AP-15), 5-O-formylpinostrobin (AP-16), 5-O-dimethylcarbamylpinostrobin (AP-17), 5-O-methoxyformylpinostrobin (AP-18), 5-O-2,2,2-trifluoroacetylpinostrobin (AP-19), and 5-O-acetylpinostrobin (AP-20).

 

2D and 3D structure preparation:

Each of the twenty 5-O-acetylpinostrobin compounds were drawn with the ChemBio Draw 2D program and saved in .cdxml format. The 2D structures were copied into the ChemBio Draw 3D program, and energy was minimized with MMFF94 minimization. The structures were saved in .mol2 and .sdf formats26,27.

 

Receptor preparation:

The receptor used is ERα with PDB code ID: 3ERT, which was obtained from the RSCB Protein Data Bank website (http://rcsb.org) and downloaded in .pdb format28.

 

The validation of docking methods:

The Molegro Virtual Docker (MVD) program was used to validate docking the native ligand (4-hydroxytamoxifen) with the 3ERT. The setup was completed by removing water, and then cavity detection was performed as the binding site for the native ligand. Cavity 1 (volume 365.056), grid resolution of 0.30 Å, and binding sites of x: 31.83, y: -1.71, and z: 25.20. Setting the algorithm, namely the number of runs of 10 with the parameters of the maximum iterations of 1500, the maximum population size of 50, and pose generation of 100.00. In addition, simplex evolution was used at max steps 300 and distance factor 1.0029. The Mean Square Deviation (RMSD) score was used as a parameter to assess the validation process. RMSD states the average difference of atomic position from ligand replication with crystallography results. RMSD results < 2 A indicate the position of replication docking results closer to the ligand crystallography30.

 

Molecular Docking:

Molecular docking on twenty 5-O-acetylpinostrobin derivatives was performed in the same manner as the validation process. The results obtained were the rerank score and ligand-receptor amino acid interactions. Amino acid interactions were recorded based on two types of interactions, namely hydrogen bonds and steric interactions. The rerank score was a linear combination of E-inter (steric, Van der Waals, hydrogen bonding, and electrostatic) between ligand and protein and E-intra. (torsion, sp2-sp2, hydrogen bonding, Van der Waals, and electrostatic) of the ligand that has been measured based on predetermined coefficients31.

 

Determination of pharmacokinetics and toxicity parameters:

The 3D structures of twenty 5-O-acetylpinostrobin derivatives saved in .sdf format were translated into .smile format through the website https://cactus.nci.nih.gov/translate/. All compounds in .smile format were entered into the pkCSM website32. The parameters determined include absorption (Caco-2 permeability, %HIA), distribution (VDss, BBB permeability), metabolism (CYP3A4 and CYP2D6 inhibitor), excretion (Cltot, Renal OCT-2 substrate), and toxicity (LD50, hepatotoxicity).

 

RESULT:

The validation results of the docking method:

The validation results obtained an RMSD value of 0.829 A (<2 A). A comparison between the structure of 4-hydroxytamoxifen (4-OHT) as a native ligand for re-docking results with ERα can be seen in Figure 1. The amino acid interaction of 4-OHT before and after docking are shown in Figure 2.

 

Figure 1. Redocking result of 4-OHT with RMSD 0.829 Å

 

Figure 2. Interaction of amino acids with 4-OHT before docking (blue) and after docking (yellow)

 

Molecular docking results:

Twenty 5-O-acetylpinostrobin derivatives had rerank scores ranging from -113.8310 to -71.7388 kcal/mol. Eight compounds (AP-10-12, 14-16, and 19-20) had a smaller rerank score than pinostrobin and native ligand. The amino acid interaction with the ligand was divided into two categories: hydrogen bond and steric interaction. The rerank score and amino acid interactions are shown in Table 1.


 

Table 1. Rerank scores and amino acid interactions of 5-O-acetylpinostrobin derivatives

No. AP

Amino acid interaction

Rerank score

(kcal/mol)

No. AP

Amino acid interaction

Rerank score

(kcal/mol)

Hydrogen bond

Steric interaction

Hydrogen bond

Steric interaction

1

Thr 347

Asp 351, Leu 354, and Leu 536

-97.2996

11

Leu 327, Arg 394, and Lys 449

Glu 353, Pro 325, Pro 324, and Arg 394

-100.0770

2

Cys 530

Lys 529, Val 533, and Trp 383

-71.7388

12

Leu 327 and Lys 449

Arg 394, Glu 353, Leu 327, and Pro 324

-96.6048

3

Thr 347

Met 343, Leu 346, Leu 387, Trp 383, and Ala 350

-87.4017

13

-

Ala 350, Leu 384, and Leu 349

-77.7985

4

Cys 530

Met 528 and Val 533

-95.2128

14

Leu 327, Lys 449, and Arg 394

Ile 326, Glu 353, Pro 324, and Arg 394

-105.5050

5

Thr 347

Met 421, Thr 347, Leu 384, and Leu 349

-96.5739

15

Leu 327, Lys 449, and Arg 394

Arg 394, Ile 326, Glu 353, Pro 325, Pro 324, and Met 357

-113.8310

6

Leu 536 and Cys 530

Lys 529

-86.7957

16

Lys 449 and Arg 394

Pro 325, Glu 353, His 356, Pro 324, and Gly 390

-88.7383

7

Leu 327 and Lys 449

Arg 394, Glu 353, Pro 324, and Met 357

-95.2809

17

Thr 347

Leu 384 and Leu 349

-81.2168

8

Lys 449, Arg 394, and Leu 327

Pro 324, Glu 353, Leu 327, Pro 325, Ile 386, Gly 390, Lys 449, and Met 357

-83.5465

18

Arg 394 and Lys 449

Pro 325, Pro 324, and Gly 390

-100.0020

9

Lys 449, Arg 394, and Leu 327

Ile 326, Arg 394, Glu 353, Pro 325, Gly 390, Ile 386, Lys 449, and Met 357

-88.1631

19

Leu 327, Arg 394, and Lys 449

Arg 394, Glu 353, Pro 325, Pro 324, and Leu 287

-107.7860

10

Leu 327, Arg 394, and Lys 449

Glu 353, Met 357, Pro 324, Arg 394, and Ile 326

-100.9870

20

Leu 327, Arg 394, and Lys 449

Arg 394, Leu 327, Ile 326, Glu 353, and Pro 324

-99.2747

 

 

 

 

P

Lys 449

His 356, Pro 324, Lys 449, Ile 386, Gly 390, Glu 323, and Glu 353

-81.2292

 

 

 

 

4-OHT

Glu 353 and Arg 394

Glu 353

-91.3077

 


Pharmacokinetics and toxicity results:

The results of pharmacokinetic studies consist of absorption (Caco-2 permeability, %HIA), distribution (VDss, BBB permeability), metabolism (CYP3A4 and CYP 2D6 inhibitors), excretion (Cltot, Renal OCT-2 substrate), and toxicity (LD50, hepatotoxicity) as shown in Table 2. Caco2 permeability score of 1.156 to 1.511 and %HIA of 94.006 to 97.720%. VDss was -0.891 to -0.412 log L/kg and BBB permeability was -1.084 to -0.087. All compounds were not CYP2D6 inhibitors, and there were ten compounds that were not CYP3A4 inhibitors. Cltot of 0.136 to 1.049 mL/min/kg and not as Renal OCT2 substrate. LD50 of 813.669 to 1,169.305 g/kgBW.


 

Table 2. Pharmacokinetics and toxicity prediction results for 5-O-acetylpinostrobin derivatives

No. AP

Absorption

Distribution

Metabolism

Excretion

Toxicity

Caco2 permeability

%HIA

VDss

 

BBB permeability

CYP2D6 inhibitior

CYP3A4 inhibitior

Cltot

 

Renal OCT2 substrate

LD50

Hepatotoxicity

 

1

1.318

97.498

-0.801

-0.189

No

Yes

0.453

No

1169.305

Yes

2

1.184

97.101

-0.882

-0.142

No

No

0.567

No

1050.288

No

3

1.243

95.984

-0.638

-0.229

No

Yes

1.049

No

838.490

No

4

1.216

97.471

-0.891

-0.154

No

Yes

0.332

No

1038.635

Yes

5

1.224

96.274

-0.600

-0.234

No

Yes

0.246

No

828.087

No

6

1.231

97.067

-0.867

-0.203

No

Yes

0.410

No

1058.444

Yes

7

1.223

97.003

-0.554

-0.220

No

Yes

0.243

No

825.650

No

8

1.511

97.720

-0.794

-1.084

No

No

0.602

No

1047.080

Yes

No. AP

Absorption

Distribution

Metabolism

Excretion

Toxicity

Caco2 permeability

%HIA

VDss

 

BBB permeability

CYP2D6 inhibitior

CYP3A4 inhibitior

Cltot

 

Renal OCT2 substrate

LD50

Hepatotoxicity

 

9

1.213

96.972

-0.513

-0.248

No

Yes

0.322

No

813.669

No

10

1.259

96.724

-0.698

-0.160

No

Yes

0.239

No

835.321

No

11

1.156

96.627

-0.445

-0.190

No

No

0.496

No

951.605

No

12

1.164

94.990

-0.520

-0.174

No

No

0.342

No

906.517

No

13

1.477

97.007

-0.608

-0.189

No

Yes

0.314

No

809.764

No

14

1.406

96.889

-0.412

-0.199

No

No

0.408

No

829.908

No

15

1.173

94.042

-0.556

-0.158

No

Yes

0.136

No

1145.144

No

16

1.348

97.244

-0.479

-0.148

No

No

0.185

No

753.182

No

17

1.267

94.006

-0.486

-0.087

No

No

0.508

No

869.785

Yes

18

1.404

95.883

-0.735

-0.675

No

No

0.296

No

887.777

No

19

1.438

94.198

-0.572

-0.110

No

No

0.099

No

1134.766

No

20

1.379

96.877

-0.482

-0.385

No

No

0.386

No

827.960

No

P

1.303

94.104

-0.187

0.229

No

No

0.206

No

801.779

No

4-OHT

1.120

95.378

0.108

-0.288

Yes

Yes

0.441

No

637.591

No

Note: % HIA: Human Intestinal Absorption (%), VDss: Steady State of Volume Distribution (log L/kg)), BBB: Blood Brain Barrier (log BB), CYP2D6: Cytochrome P2D6, CYP3A4: Cytochrome P3A4, Cltot: total clearance (mL/min/kg), LD50: Oral rat acute toxicity (g/kgBW).

 


DISCUSSION:

Drug development using molecular docking, pharmacokinetic prediction, and toxicity studies of twenty 5-O-acetylpinostrobin derivatives is a first step in the discovery of breast cancer drug candidates. Derivatives were obtained by substituting OH-pinostrobin with acyl groups. Twenty acyl groups improve the lipophilic (π), electronic (δ*), and steric (Es) properties based on the Topliss tree33. Molecular docking was performed to determine the binding affinity and ligand-protein interaction34. The selected target receptor is ERα (PDB ID: 3ERT), which is based on X-ray high resolution of 1.98 Å (< 2 Å), the conditions under which the crystal structure of the protein was obtained, widely used in international journals, and bound with 4-hydroxytamoxifen as a native ligand for ERα35,36. Before molecular docking, the validation of the docking method was measured by the RMSD score. The RMSD score shows the level of difference from the atomic position before and after the docking proces37. Since the RMSD threshold is less than 2 Å, the average value of the three replications of the re-docking process is valid38,39. The RMSD score of re-docking in the research was 0.829 Å. The interaction of amino acid residues with 4-OHT before and after docking was similar. The interaction before docking consists of two donor hydrogen bond interactions (Arg 394, Glu 353) and an electrostatic interaction (Asp 351). Docking consists of three donor hydrogen bond interactions (Arg 394, Glu 353, and Thr 347) and an electrostatic interaction (Asp 351). The molecular docking value obtained was the rerank score, which was the value of binding energy needed to form a bond between the ligand and the receptor40,41. Eight compounds (AP-10-12, 14-16, and 19-20) had a smaller rerank score than pinostrobin and native ligand. The similarity of amino acid residue interactions, such as a donor hydrogen bond between the oxygen atom of the chroman-4-on ring and Arg 394, as well as a steric interaction between Glu 353 and the C-aromatic of the chroman-4-on ring.

 

For the pharmacokinetic prediction of twenty 5-O-acetylpinostrobin derivatives, the selected absorption parameters were Caco2 permeability and %HIA. All derivatives showed Caco2 permeability values > 0.90 (1.156–1.511), and AP 1–20 had higher values than 4-OHT (1.120). Caco2 permeability is the ability of drug absorption using an in vitro model of the human intestinal mucosa42,43. %HIA in all compounds showed values above 30% (94.006–97.720%); compounds AP-1 to AP-14, AP-16, and AP-18 to AP-20 had higher %HIA values than pinostrobin (94.104%) and 4-OHT (95.378%). %HIA is used to predict the proportion of compounds absorbed by the human small intestine because the intestine is the main organ for oral route drug absorption43. The results of the analysis of Caco2 permeability and %HIA parameters revealed that all compounds had good absorption.

 

The selected distribution parameters were VDss and BBB permeability. All derivative compounds had log VDss values in the low category, < -0.15 (-0.891 to -0.412). All compounds had lower VDss values compared to pinostrobin (-0.187). VDss, or steady-state volume of distribution, is the volume required for the total dose of a drug to be evenly distributed to produce the same concentration as in blood plasma44. The BBB permeability values of all derivatives were < -1, which meant they had a low distribution in the brain. BBB permeability indicates the ability of a drug to enter the brain, this is considered to help reduce side effects and toxicity or to increase the effectiveness of drugs whose pharmacological activity is in the brain44,45. The analysis of VDss and BBB permeability parameters were analyzed, and all compounds had a good distribution.

 

The selected metabolism parameters are inhibition of the cytochromes CYP2D6 and CYP3A4. All compounds were shown not to be CYP2D6 inhibitors, and compounds AP-2, 8, 11, 12, 14, and AP-16 to AP-20 were not CYP3A4 inhibitors. According to Cronin-Fenton and Lash (2011), CYP2D6 and CYP3A4 show a relationship in their binding affinity to selective esterogen receptors (ER), the strength of ER affinity predicts the anti-cancer response46. Analysis of CYP2D6 and CYP3A4 inhibitor parameters showed that all compounds had good metabolism.

 

Excretion parameters consist of Cltot and Renal OCT2 substrate. Cltot indicates an estimate of how much of the compound is cleared by the liver and kidneys47. The Cltot of all derivatives was <5 mL/min/kg, which means the value is low. All compounds showed that none were Renal OCT2 substrates. The results of the Cltot and Renal OCT2 substrate parameter analyses showed that all compounds had good excretion.

 

Toxicity parameters consist of LD50 and hepatotoxicity, which are the two toxicity parameters. LD50, or lethal dose, is used to assess the relative toxicity of a compound. All compounds have an LD50 > 5000 mg/kg, which means they belong to class VI, which is not toxic. The LD50 of all compounds shows that compound code AP-16 has the lowest LD50, while AP-1 has the highest. The higher the LD50, the greater the dose limit to reach the lethal dose48. Compounds that showed no hepatotoxicity were AP-2, 3, 5, 7, 9-16, 19, and AP-20. Hepatotoxicity has been linked with liver safety. The results of the analysis of LD50 and hepatotoxicity parameters revealed that all compounds had low toxicity, even though compounds AP-1, 4, 6, 8, and AP-17 were in the hepatotoxic category.

 

The six compounds (AP-10, 11, 14, 15, 19, and 20) were selected based on these three predictions (molecular docking, pharmacokinetics, and toxicity prediction). Based on molecular docking results, the six selected compounds have the smallest rerank score compared to pinostrobin and 4-hydroxytamoxifen, as well as a similarity of amino acid residues with 4-hydroxytamoxifen. Based on the results of pharmacokinetic prediction, the six compounds have good absorption (higher value of Caco-2 permeability and % HIA), distribution (lower value of VDss and BBB permeability), metabolism (not to be CYP3A4 and partly as CYP 2D6 inhibitors), and excretion (low Cltot, not to be Renal OCT-2 substrate) compared to pinostrobin and 4-hydroxytamoxifen. Based on the toxicity prediction results, the six compounds had low toxicity and were not hepatotoxic.

 

CONCLUSION:

Molecular docking, pharmacokinetics, and toxicity prediction were used as the basis of drug development. Based on three predictions, it concluded that compounds AP-10, 11, 14, 15, 19, and 20 had greater predicted anti-breastcancer activity, better pharmacokinetics, and lower toxicity than pinostrobin and 4-hydroxytamoxifen. Therefore, these six compounds have the potential to be developed further.

 

CONFLICT OF INTEREST:

There was no conflict of interest from any of the authors in this manuscript.

 

ACKNOWLEDGMENTS:

The authors would like to thank the Faculty of Medicine at Universitas Islam Malang for providing support and access to the lab.

 

REFERENCES:

1.      Waill AE, Mohamed AM, Walid F, Mahmoud E, Ibrahim M.N, Ghoson MD. Molecular identification, metabolites profiling, anti-breast cancer, anti-colorectal cancer, and antioxidant potentials of Streptomyces zaomyceticus AA1 isolated from a remote bat cave in Egypt. Research J. Pharm. and Tech. 2020; 13(7): 3072-3080. doi: 10.5958/0974-360X.2020.00545.4

2.      Bharti A, Dheeraj A. In vivo and in vitro investigation of cytotoxic and antitumor activities of polyphenolic leaf extract of Hibiscus sabdariffa against, breast cancer cell lines. Research J. Pharm. and Tech. 2019; 13(2): 615-620. doi: 10.5958/0974-360X.2020.00116.X

3.      Krishnaveni K, Rosmi J, Sumitha SK, Teena J, Shanmuga SR, Sambathkumar R. A study on socio demographic and associated risk factors for cancer patients in private cancer hospital, Bangalore, India. Research J. Pharm. and Tech. 2018; 11(2): 677-680. doi: 10.5958/0974-360X.2018.00127.0

4.      Nahla EE, Enas AE, Naglaa FK, Hend MS, Eman GH. New shining stars in the sky of breast cancer diagnosis and prognosis: A Review. Research J. Pharm. and Tech. 2022; 15(8): 3808-3. doi: 10.52711/0974-360X.2022.00639

5.      Viani A, Andi AM. Combination effects of Abelmoschus manihot (L.) Medik of n-hexane extracts and doxorubicin in breast cancer 4T1 cells line. Research J. Pharm. and Tech. 2022; 15(2): 639-2. doi: 10.52711/0974-360X.2022.00105

6.      Jainab NH, Mohan Maruga Raja MK. 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. doi: 10.5958/0974-360X.2018.00300.1

7.      Suresh R, D, Benitojohnson, C, Maheswari C, Venkatnarayanan R, Manavalan, R. Chemo preventive activity of Triumfetta rhomboidea in 7, 12-dimethylbenz (A) anthracene Induced breast cancer in Sprague –Dawley rat model. Research J. Pharm. and Tech. 2017; 10(3): 687-692. doi: 10.5958/0974-360X.2017.00128.7

8.      Sara JK, AbdulSahib KA, Abdul-Ameer NA. Compartment between breast cancer patient undergoing chemotherapy and healthy women by cytokinesis-block micronucleus testand nuclear division index. Research J. Pharm. and Tech. 2018; 11(2): 705-710. doi: 10.5958/0974-360X.2018.00133.6

9.      Feng Y, Spezia M, Huang S, Yuan C, Zeng Z, Zhang L, Ji X, Liu W, Huang B, Luo W, Liu B, Lei Y, Du S, Vuppalapati A, Luu HH, Haydon RC, He TC, Ren G. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes and Diseases. 2018; 5(2): 77–106. doi:10.1016/j.gendis.2018.05.001

10.   Mohend AN, Hadi MA, Alaa SA. Comparative study between paired primary and relapsed breast cancer patients based on clinicopathological features and molecular subtypes of breast cancer in babylon province. Research J. Pharm. and Tech. 2018; 11(6): 2365-2371. doi: 10.5958/0974-360X.2018.00439.0

11.   Jayashree V, Malarkodi V. Breast cancer and various prognostic biomarkers for the diagnosis of the disease: A Review. Research J. Pharm. and Tech. 2017; 10(9): 3211-3216. doi: 10.5958/0974-360X.2017.00570.4

12.   Hanaa HA, Hadeer AA, Ghada HE, Hebatallah GH, Emad FE. Quercetin offers chemopreventive potential against breast cancer by targeting a network of signalling pathways. Research J. Pharm. and Tech. 2021; 14(5): 2829-2839. doi: 10.52711/0974-360X.2021.00499

13.   Shanthi D, R. Saravanan. Evaluation of cytotoxicity of normal Vero and anticancer activity of human breast cancer cell lines by aqueous unripe fruit extract of Solanum torvum. Research J. Pharm. and Tech. 2021; 14(7): 3504-8. doi: 10.52711/0974-360X.2021.00607

14.   Aguslina K, Siswandono S, I Ketut S. Synthesis and cytotoxic activity of N-(4-bromo)-benzoyl-N’phenylthiourea and 4-(tert-butyl)-N-benzoylurea on primary cells of HER2-positive breast cancer. Research J. Pharm. and Tech. 2021; 14(3):1195-1200. doi: 10.5958/0974-360X.2021.00213.4

15.   Wiyono L, Edina BC, Rahmawanti RA, Azizah NN, Paramita RI, Purwaningsih EH, Fadilah F. Isolation, synthesis nanoparticle, and in-vitro test of pinostrobin from Kaempferia pandurata on MCF-7 and MDAMB-231 breast cancer cell. Research J. Pharm. and Tech. 2020; 13(6): 2797-2801. doi: 10.5958/0974-360X.2020.00497.7

16.   Praditapuspa EN, Siswandono, Widiandani T. In silico analysis of pinostrobin derivatives from boesenbergia pandurata on ErbB4 kinase target and QSPR linear models to predict drug clearance for searching anti-breast cancer drug candidates. Pharmacognosy Journal. 2021; 13(5): 1143–1149. https://doi.org/10.5530/pj.2021.13.147

17.   Poerwono H, Sasaki S, Hattori Y, Higashiyama K. Efficient microwave-assisted prenylation of pinostrobin and biological evaluation of its derivatives as antitumor agents. Bioorganic and Medicinal Chemistry Letters. 2010; 20: 2086–2089. doi:10.1016/j.bmcl.2010.02.068.

18.   Agustin SL, Widiandani T, Hardjono S, Purwanto BT. QSAR of acyl pinostrobin derivatives as Anti-breast cancer against HER-2 receptor and their ADMET properties based on in silico Study. Research Journal of Pharmacy and Technology. 2022; 15(10):4641-8. doi: 10.52711/0974-360X.2022.00779

19.   Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-aided ligand discovery for estrogen receptor alpha. Int J Mol Sci. 2020; 21(12): 1–49. doi: 10.3390/ijms21124193

20.   Dara S, Dhamercherla S, Jadav SS, Babu CM, Ahsan MJ. Machine learning in drug discovery: A Review. Artificial Intelligence Review. 2022; 55(3): 1947–1999. doi 10.1007/s10462-021-10058-4

21.   Chinchole PP, Wankhede SB. Comparative in silico drug likeness and in vitro study of some schiff’s bases as potent COX-II inhibitors. Research J. Pharm. and Tech. 2019; 12(10): 4973-4980. doi: 10.5958/0974-360X.2019.00862.X

22.   Ferreira LG, Dos Santos RN, Oliva G, Andricopulo AD. Molecular docking and structure-based drug design strategies. Molecules. 2015; 20(7); 13384–13421. doi:10.3390/molecules200713384

23.   Marimuthu SCV, Murugesan J, Babkiewicz E, Maszczyk P, Sankaranarayanan M, Thangamariappan E. Pharmacoinformatics-based approach for uncovering the quorum-quenching activity of phytocompounds against the oral pathogen, streptococcus mutans. Molecules. 2023; 28(14): 1–20. doi: 10.3390/molecules28145514.

24.   Karavadi B, Shivashankari S. Computational study on receptors related to aggressive conduct. Research J. Pharm. and Tech. 2018; 11(5): 1729-1733. doi: 10.5958/0974-360X.2018.00321.9

25.   Beck TC, Springs K, Morningstar JE, Mills C, Stoddard A, Guo L, et al. Application of pharmacokinetic prediction platforms in the design of optimized anti-cancer drugs. Molecules. 2022; 27(12): 1–10. doi: 10.3390/molecules27123678

26.   Widiyana AP, Putra GS, Muchlashi LA, Sulistyowaty MI, Budiati T. Design and molecular docking studies of quinazoline derivatives as antiproliferation. JFKI. 2016; 3(1): 32–8. doi: 10.20473/jfiki.v3i22016.44-48

27.   Aiyalu R, Lakshmanan K, Balasubramanian HB, Ramasamy A. Screening of iridoids and seco-iridoids in gentianaceae family against liver corrective targets using molecular docking studies. Research J. Pharm. and Tech. 2019; 12(12): 5735-5738. doi: 10.5958/0974-360X.2019.00992.2

28.   Baul HS, Rajiniraja M. Favorable binding of quercetin to α-synuclein as potential target in parkinson disease: An insilico approach. Research J. Pharm. and Tech. 2018; 11(1): 203-206 doi: 10.5958/0974-360X.2018.00038.0

29.   Puspaningtyas AR. Docking molekul dengan metoda molegro virtual docker dari ekstrak ir psidium guajava, linn dan citrus sinensis, peels sebagai inhibitor pada tirosinase untuk pemutih kulit. JKTI. 2013; 15(1); 31-39.

30.   El Aissouq A, Toufik H, Stitou M, Ouammou A, Lamchouri F. In silico design of novel tetra-substituted pyridinylimidazoles derivatives as c-Jun N-terminal kinase-3 inhibitors, using 2D/3D-QSAR studies, molecular docking and ADMET prediction. Int J Pept Res Ther. 2020; 26(3): 1335–51. doi: 10.1007/s10989-019-09939-8

31.   Singh SP, Deb CR, Ahmed SU, Saratchandra Y, Konwar BK. Molecular docking simulation analysis of the interaction of dietary flavonols with heat shock protein 90. J Biomed Res. 2016; 30(1): 67–74. doi: 10.7555/JBR.30.20130158

32.   Abdullahi SH, Uzairu A, Shallangwa GA, Uba S, Umar AB. Pharmacokinetic profiling of quinazoline-4(3H)-one analogs as EGFR inhibitors: 3D-QSAR modeling, molecular docking studies and the design of therapeutic agents. J Taibah Univ Med Sci. 2023; 18(5): 1018–29. doi: 10.1016/j.jtumed.2023.02.015

33.   Siswandono and Soekardjo B. Kimia Medisinal Ed 2. Ailangga University Press, Surabaya. 2008.

34.   Reddy NN, Hung S, Swamy MK, Sanjeev A, Reddy PM. Synthesis and rational design of new appended 1,2,3-triazole-uracil ensembles as promising anti-tumor agents via in silico VEGFR-2 Transferase inhibition. Molecules. 2021; 26(7): 1952. doi: 10.3390/molecules26071952

35.   Hosfield DJ, Weber S, Li S, Sauvage M, Joiner CF., Hancock GR, Sullivan EA, Ndukwe E, Han R, Cush S, Lainé M, Mader SC. Greene GL., Fanning SW. Stereospecific lasofoxifene derivatives reveal the interplay between estrogen receptor alpha stability and antagonistic activity in ESR1 mutant breast cancer cells. eLife. 2022; 16(11): e72512. doi: 10.7554/eLife.72512

36.   Maximov PY, Abderrahman B, Fanning SW, Sengupta S, Fan P, Curpan RF., Maria D, Rincon, Q, Rajan SS, Greene GL, Jordan, VC. Endoxifen, 4-hydroxytamoxifen and an estrogenic derivative modulate estrogen receptor complex mediated apoptosis in breast cancers. Mol Pharmacol. 2018; 94(2): 812–822. doi: 10.1124/mol.117.111385

37.   Ibrahim ZY, Uzairu A, Shallangwa GA, Abechi SE, Isyaku S. Virtual screening and molecular dynamic simulations of the antimalarial derivatives of 2-anilino 4-amino substituted quinazolines docked against a Pf-DHODH protein target. Egypt J Med Hum Genet. 2022; 23(1): 119. doi: 10.1186/s43042-022-00329-2

38.   Fadhil Pratama MR, Poerwono H, Siswodihardjo S. Introducing a two‐dimensional graph of docking score difference vs. similarity of ligand‐receptor interactions. Indones J Biotechnol. 2021; 26(1): 54–60. doi: 10.22146/ijbiotech.62194

39.   Khdar ZA, Sliman F, Kousara M. Design and in-silico ADMET analysis of new benzopyrane-derived PIM-1 inhibitors. Research J. Pharm. and Tech. 2019; 12(11): 5413-5423. doi: 10.5958/0974-360X.2019.00939.9

40.   Illian DN, Widiyana AP, Hasana AR, Maysarah H, Al Mustaniroh SS, Basyuni M. In silico approach: Prediction of ADMET, molecular docking, and QSPR of secondary metabolites in Mangroves. J Appl Pharm Sci. 2022; 12(11): 021–029. doi: 10.7324/JAPS.2022.121103

41.   Malik A, Malik N, Dhiman P, Khatkar A, Kakkar S. Molecular docking, synthesis, α-amylase inhibition, urease inhibition and antioxidant evaluation of 4-hydroxy-3-methoxy benzoic acid Research J. Pharm. and Tech. 2019; 12(12): 5653-5663. derivatives. doi: 10.5958/0974-360X.2019.00978.8

42.   Mandal M, Sahoo SK, Patra P, Mallik S, Zhao Z. In silico ranking of phenolics for therapeutic effectiveness on cancer stem cells. BMC Bioinformatics. 2020; 21(21): 1–18. doi: 10.1186/s12859-020-03849-z

43.   Bitew M, Desalegn T, Demissie TB, Belayneh A, Endale M, Eswaramoorthy R. Pharmacokinetics and drug-likeness of antidiabetic flavonoids: Molecular docking and DFT study. PLoS One . 2021; 16(12): e0260853. doi:10.1371/journal.pone.0260853

44.   Taupiqurrohman O, Rezaldi F, Amalia D, Suryani Y. Anticancer potency of dimethyl 2-(2-hydroxy-2-methoxypropilidine) malonate in Kombucha. J Biodjati. 2022; 7(1): 86–94. doi: 10.15575/biodjati.v7i1.14634

45.   Hardjono S, Widiandani T, Purwanto BT, Nasyanka AL. Molecular docking of N-benzoyl-N'-(4-fluorophenyl) thiourea derivatives as anticancer drug candidate and their ADMET prediction. Research J. Pharm. and Tech. 2019; 12(5): 2160-2166. doi: 10.5958/0974-360X.2019.00359.7

46.   Cronin-Fenton DP, Lash TL. Clinical epidemiology and pharmacology of CYP2D6 inhibition related to breast cancer outcomes. Expert Rev Clin Pharmacol. 2011; 4(3): 363–77. doi:10.1586/ecp.11.18.

47.   Lombardo F, Obach RS, Varma M V., Stringer R, Berellini G. Clearance mechanism assignment and total clearance prediction in human based upon in silico models. J Med Chem. 2014; 57(10): 4397–405. doi.org/10.1021/jm500436v

48.   Gadaleta D, Vuković K, Toma C, Lavado GJ, Karmaus AL, Mansouri K. SAR and QSAR modeling of a large collection of LD50 rat acute oral toxicity data. J Cheminform. 2019; 11(1): 1–16. doi: 10.1186/s13321-019-0383-2

 

 

 

Received on 23.02.2024      Revised on 17.05.2024

Accepted on 16.07.2024      Published on 24.12.2024

Available online from December 27, 2024

Research J. Pharmacy and Technology. 2024;17(12):5996-6002.

DOI: 10.52711/0974-360X.2024.00910

© RJPT All right reserved