Evaluation of 3-Ethyl-5-fluoro-2-phenylimino-thiazolidin-4-one derivatives: Molecular docking against kinase protein and ADME studies

 

Shreyash D. Kadam1, Denni Mammen1*, Laxmikant B. Nikam2, Rahul R. Bagul2, Ajit Borhade2

1School of Science, Navrachana University, Vasana-Bhayli Road, Bhayli, Vadodara, Gujarat, India, 391410.

2 Gujarat Fluorochemicals Limited, Ranjitnagar, Panchamahal, Gujarat-389380, India.

*Corresponding Author E-mail: drdenni.mammen@gmail.com

 

ABSTRACT:

A number of new compounds have been synthesized by the authors containing fluorinated thiazolidin-4-one ring. With the aim to assess the anti-cancer potential of all the synthesized derivatives,theywere computationally tested against 1T46 C-Kit Tyrosine Kinase protein. Almost all of the evaluated derivatives showed decent affinity towards the protein, with favourable binding poses through hydrogen bonding, halogen binding and pi-sigma bonding. The amino acid lysine at position 623 in the protein chain exhibited hydrogen bond formation with each compound, along with other amino acids. Furthermore, the in silico ADME predictions suggest that the majority of the synthesized compounds exhibit favourable drug-like characteristics, with low potential for adverse effects and toxicity. The molecules possessing oxygen-containing functionalities such as –NO2, -OCF3, -OCF2CF2H and –OH have been shown to be able to cross the Human Intestinal lining. The fluorine-containing moieties such as difluoro, trifluoro, -CF3, chloro-fluoro, and difluorobenzylamino were predicted in order to cross BBB (Blood-Brain-Barrier). Current study has revealed that the synthesized compounds show promising anticancer potential.

 

KEYWORDS: Fluorine, Thiazolidinone, In-silico, Docking, Autodock, Vina, Discovery Studio, Ligand, Protein, Receptor, Kinase, ADME, BBB, HIA.

 

 


INTRODUCTION: 

In the current era, various diseases are emerging day by day and their treatment is a challenging issue for the world. Past few years have witnessed significant advancement in computational technology which provided one of the best gift to scientific researchers specially in the field of medicinal chemistry which contributed a lot to drug discovery and given rise to many new possibilities i.e. the ability to accurately predict the biological activity of compounds to stimulate new possibilities for the advancement of novel drugs design.

 

The major advantages of in-silico study is that it is fast, reliable, productive, cost-effective, also it saves actual resources i.e. just conducting an in-vitro study randomly which end up getting poor results, moreover docking

 

which is a sub-category of in-silico study helps us to predict exact binding sites and select exact target protein by which we can determine precise biological activity. In short, docking and ADME1–3 studies assist in study molecular biology based on structure, and drug design using computational tools. Docking analysis4–7 is an eye-catching technique used to identify drug-biomolecule interactions for the rational drug design8 and themain objective of analysing the docking between ligand and protein is to predict the accurate binding modes of a ligand with the 3D protein structure from structure databases such as Protein Data Bank (PDB). There are various types of molecular docking interactions9 such as drug-protein or ligand-protein, protein-protein, protein-nucleic acid, enzyme-substrate, and drug-nucleic acid interactions. Thus we can define molecular docking technique as a computational technique which includes a proper protocol by which we can predict the conformation or binding sites of a receptor-ligand complex, where the receptor is usually a protein molecule and the ligand is a synthesized small molecule. If everything works perfectly then it results in formation of stable protein-ligand adduct10that are vital to execute their biological functions and depending upon selected target and ligand binding properties, it predicts the three-dimensional structure of complex and provides binding affinity11.

 

The drug preparation also involves evaluation of absorption, distribution, metabolism and excretion i.e. ADME12–17 properties and at the preliminary stages of drug discovery if these properties are known so it decreases chances of failures in pharmacokinetics properties in later phases of development due to which drug moiety will reach the market more efficiently. Moreover, for a synthesized compound to function as an effective drug, it must not only reach its target within the body at the appropriate concentration but also remain in that location in a bioactive state for a sufficient duration to facilitate the intended biological processes. Therefore in this study, we have also made use of the Swiss ADME web tool1–3 for analyzing pharmacokinetics, physicochemical properties, drug-likeness and medicinal-chemistry friendliness, in addition to which we obtained unique boiled egg diagram which was helpful for understanding HIA and BBB pattern on scale of WLOGP vs TPSA. Hence, by using this computational method we can perform virtual screening and select the most interesting and promising molecules from the synthesized moieties and proceed them for their further modifications or for in-vitro studies.

 

Thiazolidinone18–26 also known as “wonder nucleus”27 are the oxo derivatives of thiazolidine group that is pertaining to a significant class of heterocyclic compounds characterized by the presence of sulfur and nitrogen atoms, along with the carbonyl group situated at the fourth position within a five-membered ring. 28–34. Also importance of fluorine35 is well known and it was observed that small organic molecules which act as pharmaceutical drugs mostly consist of at least one fluorine atom or a fluorinated functional group like CF3, Ar-F in its structure. It was reported in many papers that around 20% of the marketed drugs were fluorinated organic compounds. Numerous of these compounds have fluorinated aniline as a precursor for its synthesis.

 

In the present work we have undergone in silico analysis36 of novel 3-Ethyl-5-fluoro-2-phenylimino-thiazolidin-4-one (2-19) derivatives which were synthesized using different Fluorinated anilines against 1T46 target protein Figure. 137–39. In addition to this, we also have selected doxorubicin Figure 1 as standard due to the fact that further we are going to continue this study for in-vitro anticancer40,41 evaluation in which we are going to take doxorubicin as our standard42. So in order to gain full details of the drug we had performed docking and ADME analysis for doxorubicin40as well.

 

Shown below Figure 1 are the synthesized derivatives on which in silico studies have been performed against kinase protein 1T46.


 

Figure 1. C-Kit tyrosine kinase Protein 43 1T46 used as targeting macromolecule for molecular docking, doxorubicin (1) anti-cancer standard and synthesized novel fluorinated thiazolidin-4-one derivatives(2-19).


 

MATERIALS AND METHODS:

In silico activity:

1) Docking Study:

In this research, we assessed the affinity and binding modes of the studied molecules with respect to the target protein 38,39. Initially, the crystal structures of the target proteins (see Figure 1) had their water molecules removed, leaving behind only the main-chain amino acids crucial for binding. The co-crystallized ligands served as reference points for identifying the binding pockets, after which they were removed. Subsequently, polar hydrogen atoms were added to the protein structures for protonation. The structures of the investigated compounds (Figure 1) were drawn using ChemDraw Ultra 7.0 and BIOVIA Discovery Studio Visualizer 2021, and these structures were saved in PDB format. Subsequently, the saved files were imported into MGL AutoDock Tools software for protein preparation. The protein was selected as the macromolecule and then saved in PDBQT format. A configuration file was generated, containing information such as the receptor name, ligand name, output file name, and the X, Y, Z coordinates of the grid box, along with the size dimensions of the grid box (X, Y, Z). The ligand was prepared, and any available rotatable bonds were added. The next step involved the use of the Command Prompt to execute the AutoDock Vina software for each target receptor by ligand, by entering the necessary codes or commands. In each case, the algorithm generated 9 docked structural poses, along with data on affinity and RMSD. Out of which one best pose was selected and its interactions were shown in Figures 2 and 3 and Tables 1, 2 and 3.

 

2) ADME Study:

ADME analysis was performed via using Swiss ADME site 38,39. Chem Draw Ultra 7.0 software was employed to create the structures of the compounds. These structures were then converted into SMILES notation using the Swiss ADME tool, which also provided estimations of various physicochemical parameters and pharmacokinetic characteristics. Additionally, the BOILED EGG method was employed to predict the lipophilic nature and polarity characteristics of the small molecules has been shown in Figure 4.

 

RESULTSAND DISCUSSION:

In-silico activity:

1) Docking studies:

Docking studies38,39 lend a hand in analysing and getting the information regarding the appropriate orientation of synthesized organic ligand within the active site also known as “pocket site” of the large molecule i.e. protein. Here the binding pockets were selected on the foundation of pre-attached inhibitor inside the crystal structure of respective proteins. The promising compound was selected on the basis of some guidelines such as docking energy at 0.00 RMSD value, binding modes and molecular interactions at the active site of the macromolecule. The synthesized ligands have been docked against C-KIT Tyrosine Kinase protein as depicted in Figure 1. The resulting docking scores and their interactions with amino acids are depicted in the Table 1.

 

The results show that substituted fluorinated thiazolidin-4-one derivatives (2-19) exhibit diverse bonding interactions like H-bonding, Pi-Sigma, and halogen interactions towards 1T46Kinase. The compounds displayeddecentaffinity values shown within the range of -7.7 to -9.2 kcal/mol with favorable binding poses. The amino acid LYS623 from kinase displayed hydrogen bonding with almost all the compounds such as 3, 4, 5, 6, 7, 8, 10, 11, 12, 14, 15, 17, 18 and 19. Apart from lysine various amino acids such as ASP810, GLY812, CYS673, THR670, HIS790, and GLY676 were also involved in H bonding with most compounds. Among 18 ligands themoiety13demonstrated strong interactions towards kinase protein, with active site amino acids with best binding energy -8.8 kcal/mol, while the molecule 19 similarly showed a valueof -9.2 kcal/mol.


 

Table 1. Docking scores and molecular interactions of the molecules with C-KIT Tyrosine Kinase protein

Compounds No.

Docking Energy

RMS

No of Interactions

Interaction Residues

No of H Bonds

Bond Length (Ĺ)

Pi-Sigma

Halogen

H-Bonding

Doxorubicin

-7.7

0.00

9

VAL643

-

ASP810

2

2.62003

GLY812

2.95235

2

-7.7

0.00

5

THR670, UNK0

-

-

0

-

3

-8.2

0.00

8

VAL603, LEU799

GLU640, GLU671

LYS623

1

2.97711

4

-8.2

0.00

10

THR670, UNK0

ALA621, GLU640

LYS623

1

2.65556

5

-8.3

0.00

9

THR670, UNK0 

GLU640

LYS623

1

2.81076

6

-8.5

0.00

9

THR670, UNK0

-

ASP810

2

2.3832

LYS623

3.35007

7

-8.5

0.00

11

THR670, UNK0

ALA621, GLU640

LYS623

2

2.62329

ASP810

2.64758

8

-8.6

0.00

12

THR670, UNK0

GLU640

LYS623

1

2.77714

9

-7.8

0.00

6

THR670, UNK0

-

-

0

-

10

 -8.5

0.00

7

LEU595, LEU799, UNK0

VAL668

LYS623

1

2.89271

11

-8.5

0.00

8

LEU595, LEU799, LEU799

VAL668, GLU671

LYS623

1

2.85801

12

-7.9

0.00

14

THR670, UNK0

ALA621, VAL668

LYS623

2

2.51838

THR670

1.55935

13

-8.8

0.00

12

THR670, UNK0

GLU640

CYS673

2

2.86699

ASP810

2.44389

14

-8.5

0.00

12

UNK0

GLU640, ASP810

LYS623

3

2.54003

LYS623

2.62268

LYS623

3.05381

15

-7.8

0.00

23

LEU644

GLU640, GLU640, ILE808, CYS809, ASP810

LYS623

5

2.83288

ASP810

2.3607

ASP810

2.38726

HIS790

3.31246

UNK0

3.40309

16

-8.3

0.00

10

THR670, UNK1

-

-

-

-

17

-7.7

0.00

7

LEU799

GLU640

LYS623

2

2.82539

GLY676

3.6142

18

-8.0

0.00

7

THR670, UNK0

-

LYS623

1

2.76003

19

-9.2

0.00

20

UNK0

GLU640, VAL668

LYS623

3

2.7977

LYS623

2.86177

CYS673

2.65578

 

Table 2. Total number of favorable interactions: 13

Sr No.

Name

Colour

Distance

Category

Types Of Bonds

Bond From

Bonds

Bond To

Bonds

1

A:LYS623:HZ3 - :UNK0:O11

 

2.7977

Hydrogen Bond

Conventional Hydrogen Bond

A: LYS623: HZ3

H-Donor

: UNK0: O11

H-Acceptor

2

A:LYS623:HZ3 - :UNK0:F22

 

2.86177

Hydrogen Bond; Halogen

Conventional Hydrogen Bond; Halogen(Fluorine)

A:LYS623:

HZ3

H-Donor; Halogen Acceptor

:UNK0:F22

H-Acceptor; Halogen

3

A:CYS673:HN - :UNK0:S10

 

2.65578

Hydrogen Bond

Conventional Hydrogen Bond

A:CYS673:HN

H-Donor

:UNK0:S10

H-Acceptor

4

A:GLU640:OE2 - : UNK0:F22

 

3.1512

Halogen

Halogen (Fluorine)

A:GLU640:

OE2

Halogen Acceptor

:UNK0:F22

Halogen

5

A:VAL668:O - :UNK0:F20

 

3.44341

Halogen

Halogen (Fluorine)

A:VAL668:O

Halogen Acceptor

:UNK0:F20

Halogen

6

:UNK0:C16 - A:PHE811

 

3.46679

Hydrophobic

Pi-Sigma

:UNK0:C16

C-H

A:PHE811

Pi-Orbitals

7

:UNK0:C16 - :UNK0

 

3.61395

Hydrophobic

Pi-Sigma

:UNK0:C16

C-H

:UNK0

Pi-Orbitals

8

:UNK0:C19 - A:LYS623

 

4.01386

Hydrophobic

Alkyl

:UNK0:C19

Alkyl

A:LYS623

Alkyl

9

:UNK0:C19 - A:LEU644

 

5.34396

Hydrophobic

Alkyl

:UNK0:C19

Alkyl

A:LEU644

Alkyl

10

:UNK0:C19 - A:VAL668

 

4.00351

Hydrophobic

Alkyl

:UNK0:C19

Alkyl

A:VAL668

Alkyl

11

:UNK0 - A:VAL603

 

4.87354

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:VAL603

Alkyl

12

:UNK0 - A:ALA621

 

4.72358

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:ALA621

Alkyl

13

:UNK0 - A:VAL654

 

4.83263

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:VAL654

Alkyl

 

Figure 2. The 2D and 3D representation of molecular docking interactions for 3-Ethyl-5-fluoro-2-(4-trifluoromethoxy-phenylimino)-thiazolidin-4-one19, against 1T46


The two best moieties observed in this docking study having the lowest affinity value are displayed below in which 3-Ethyl-5-fluoro-2-(4-trifluoromethoxy-phenylimino)-thiazolidin-4-one 19 and 3-Ethyl-5-fluoro-2-(3-trifluoromethyl-phenylimino)-thiazolidin-4-one 13 as a ligand performed molecular docking against Tyrosine Kinase 1T46. Both these molecules showed best and most favourable interactions among all synthesized molecules due to the trifluoro functionalities. Other molecules with different functionalities showed differing interactions with values just slightly lower than these two molecules. Nine different poses were observed during the docking process, and the pose with the least affinity was chosen as the most favourable docking pose. This selected pose was then used to investigate ligand interactions. The interactions were visualized using both 2D diagrams and 3D representations.

 

3-Ethyl-5-fluoro-2-(4-trifluoromethoxy-phenylimino)-thiazolidin-4-one 19 docking analysis outputs against 1T46 kinase macromolecule

Within the specific molecule, a total of 13 favourable interactions were observed as the ligand binds to the specific sites in the selected pose. Table 2 provides details about the bonds between the ligand and amino acids, including information such as origin, type, and length of the bonds.The ligand showed three types of interactions with the amino acids at specific positions in the protein chain. These include hydrogen bonds, halogen bonds and hydrophobic interaction.

 


 

3-Ethyl-5-fluoro-2-(3-trifluoromethyl-phenylimino)-thiazolidin-4-one(13) docking analysis outputs against 1T46 kinase macromolecule

Table 3: Total number of favorable interactions: 12

Sr No.

Name

Color

Distance

Category

Types of Bonds

From

Bonds

To

Bonds

1

A:CYS673:HN -:UNK0:S10

 

2.86699

Hydrogen Bond

Conventional Hydrogen Bond

A:CYS673:HN

H-Donor

:UNK0:S10

H-Acceptor

2

A:ASP810:HN - :UNK0:F19

 

2.44389

Hydrogen Bond; Halogen

Conventional Hydrogen Bond; Halogen (Fluorine)

A:ASP810:HN

H-Donor; Halogen Acceptor

:UNK0:F19

H-Acceptor; Halogen

3

A:GLU640:OE1 -:UNK0:F21

 

2.85466

Halogen

Halogen (Fluorine)

A:GLU640:OE1

Halogen Acceptor

:UNK0:F21

Halogen

4

A:THR670:CG2 -:UNK0

 

3.84625

Hydrophobic

Pi-Sigma

A:THR670:CG2

C-H

:UNK0

Pi-Orbitals

5

:UNK0:C15 - A:PHE811

 

3.50772

Hydrophobic

Pi-Sigma

:UNK0: C15

C-H

A:PHE811

Pi-Orbitals

6

:UNK0:C15 - :UNK0

 

3.77157

Hydrophobic

Pi-Sigma

:UNK0: C15

C-H

:UNK0

Pi-Orbitals

7

:UNK0:C18 - A:LEU644

 

4.88556

Hydrophobic

Alkyl

:UNK0: C18

Alkyl

A:LEU644

Alkyl

8

:UNK0:C18 - A:VAL654

 

4.2237

Hydrophobic

Alkyl

:UNK0: C18

Alkyl

A:VAL654

Alkyl

9

:UNK0 - A:VAL603

 

4.73897

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:VAL603

Alkyl

10

:UNK0 - A:ALA621

 

4.68568

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:ALA621

Alkyl

11

:UNK0 - A:LYS623

 

5.1237

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:LYS623

Alkyl

12

:UNK0 - A:VAL654

 

5.18833

Hydrophobic

Pi-Alkyl

:UNK0

Pi-Orbitals

A:VAL654

Alkyl

 

Figure 3. The 2D and 3D representation of molecular docking interactions for 3-Ethyl-5-fluoro-2-(3-trifluoromethyl-phenylimino)-thiazolidin-4-one(13), against 1T46


In the specific molecule, a total of 12 favorable interactions were identified as the ligand bound to the designated pocket site in the chosen pose. The ligand showed hydrogen bonds, halogen bonds and hydrophobic interaction with the protein.

 

2) ADME Study:

From the results of ADME analysis38,39 we got to know about various properties of all 18 synthesized fluorinated structures of thiazolidine-4-ones. Basically, molecules within the white portion of the boiled egg diagram Figure 4 are having capability of being absorbed through the intestine i.e. HIA. The molecules depicted in the yellow portion are the ones which have the capability to cross the blood brain barrier i.e. BBB. During the analysis it was observed that the standard anticancer drug doxorubicin was out of the scanning range of the ADME analysis. So, it was positioned in neither yellow nor white portion of the diagram. This is because doxorubicin has low bioavailability when ingested orally due to which it is administered in form of an injectable.

 

Figure 4. ADME boiled egg diagram for the synthesized molecules

 

According to the analysis, we concluded thata total 16fluorinated moietiesi.e. 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 18 and 19 show promise of being able to cross the lining of human intestine. Interestingly among these 18moieties just molecule 16 which is having nitro functionalitywas not able to enter blood brain barrier and molecule 18 was on the border line andexhibits the potential of the ideal molecule that could cross the BBB barrier due to the existence of hydroxyl functionality rest all other 14molecules displayed good permeability, and capable of crossing the BBB barrier.

 

The white portion of the boiled egg diagram shows the physico-chemical space where molecules have the greatest probability of being absorbed by the human intestine. The lining of the intestines is made up of epithelial cells, which are tightly packed and covered by mucus. These cells facilitate the uptake of water and other polar substances. The derivatives containing oxygen-containing polar groups such as –NO2, -OH, -OCF3 and -OCF2CF2H are observed to be able to cross this membrane as predicted by the diagram. The yellow portion in the diagram indicated the similar space where molecules having the highest probability to cross the BBB have been predicted. This layer is made up of endothelial cells which are tightly bound to allow selective permeation of molecule to the brain cells from the bloodstream. The derivatives having relatively less polar functional groups seem to facilitate the crossing of the barrier due to the presence of lipids in the membrane. It has been observed that hydrophobic drugs cross the BBBbarrier more easily than hydrophilic drugs.

 

The derivatives having mono –CF3, difluoro, trifluoro, amino, benzyl amino, chloro-fluoro and difluoro benzyl amino groups seem to show this property. The presences of fluorine-containing functional groups seem to increase the lipophilic character of the molecule as observed in the figure.


 

Table 3. Physico-chemical Properties

Molecule No

Physico-chemical Properties

Formula

Molecular Weight (MW)

Number of H-bond acceptors

Number of H-bond donors

Molar Refractivity

Topological Surface Area (TPSA)

Molecule 1

C27H29NO11

543.52

12

6

132.66

206.07

Molecule 2

C11H11FN2OS

238.28

3

0

67.42

57.97

Molecule 3

C11H9F3N2OS

274.26

5

0

67.33

57.97

Molecule 4

C11H9F3N2OS

274.26

5

0

67.33

57.97

Molecule 5

C11H9F3N2OS

274.26

5

0

67.33

57.97

Molecule 6

C11H9F3N2OS

274.26

5

0

67.33

57.97

Molecule 7

C11H8F4N2OS

292.25

6

0

67.29

57.97

Molecule 8

C11H9ClF2N2OS

290.72

4

0

72.39

57.97

Molecule 9

C11H10BrFN2OS

317.18

3

0

75.12

57.97

Molecule 10

C12H13FN2OS

252.31

3

0

71.67

57.97

Molecule 11

C12H11F3N2OS

288.29

5

0

71.58

57.97

Molecule 12

C12H10F4N2OS

306.28

6

0

72.42

57.97

Molecule 13

C12H10F4N2OS

306.28

6

0

72.42

57.97

Molecule 14

C12H10F4N2OS

306.28

6

0

72.42

57.97

Molecule 15

C13H9F7N2OS

374.28

9

0

77.42

57.97

Molecule 16

C11H10FN3O3S

283.28

5

0

76.24

103.79

Molecule 17

C13H9Cl2F5N2O2S

423.19

8

0

88.98

67.2

Molecule 18

C11H11FN2O2S

254.28

4

1

69.44

78.2

Molecule 19

C12H10F4N2O2S

322.28

7

0

74.1

67.2

 

Table 4. Lipophilic nature

Molecule No

Lipophilic nature

iLOGP

XLOGP3

WLOGP

MLOGP

Silicos-IT Log P

Consensus Log P

Molecule 1

2.58

1.27

-0.32

-2.1

1.17

0.52

Molecule 2

2.32

2.93

2.6

1.63

2.6

2.42

Molecule 3

2.44

3.14

3.72

2.44

3.44

3.04

Molecule 4

2.38

3.14

3.72

2.44

3.44

3.02

Molecule 5

2.32

3.14

3.72

2.44

3.44

3.01

Molecule 6

2.44

3.14

3.72

2.44

3.44

3.04

Molecule 7

2.32

3.24

4.28

2.84

3.88

3.31

Molecule 8

2.59

3.66

3.82

2.58

3.66

3.26

Molecule 9

2.55

3.63

3.37

2.31

3.27

3.03

Molecule 10

2.24

2.87

2.32

1.64

2.95

2.4

Molecule 11

2.46

3.07

3.44

2.44

3.8

3.04

Molecule 12

2.4

3.82

4.78

2.57

3.65

3.44

Molecule 13

2.55

3.82

4.78

2.57

3.65

3.47

Molecule 14

2.59

3.82

4.78

2.57

3.65

3.48

Molecule 15

2.89

4.7

6.95

3.47

4.76

4.55

Molecule 16

1.97

2.76

2.51

0.61

0.45

1.66

Molecule 17

3.05

5.78

6.83

3.12

5.05

4.77

Molecule 18

2.03

2.58

2.31

1.05

2.12

2.02

Molecule 19

2.72

4.12

4.76

1.72

3.21

3.31

 

Table 5. Solubility in water

Molecule

Solubility in water

ESOL Log S

ESOL Solubility (mg/mL)

ESOL Solubility (mol/L)

ESOL Class

Ali Log S

Solubility (mg/mL)

Molecule 1

-3.91

6.72E-02

1.24E-04

Soluble

-5.2

3.46E-03

Molecule 2

-3.31

1.17E-01

4.91E-04

Soluble

-3.81

3.70E-02

Molecule 3

-3.63

6.38E-02

2.33E-04

Soluble

-4.03

2.58E-02

Molecule 4

-3.63

6.38E-02

2.33E-04

Soluble

-4.03

2.58E-02

Molecule 5

-3.63

6.38E-02

2.33E-04

Soluble

-4.03

2.58E-02

Molecule 6

-3.63

6.38E-02

2.33E-04

Soluble

-4.03

2.58E-02

Molecule 7

-3.79

4.69E-02

1.60E-04

Soluble

-4.13

2.16E-02

Molecule 8

-4.06

2.52E-02

8.65E-05

Moderately soluble

-4.57

7.89E-03

Molecule 9

-4.22

1.90E-02

5.99E-05

Moderately soluble

-4.54

9.24E-03

Molecule 10

-3.28

1.34E-01

5.30E-04

Soluble

-3.75

4.52E-02

Molecule 11

-3.6

7.29E-02

2.53E-04

Soluble

-3.95

3.20E-02

Molecule 12

-4.17

2.07E-02

6.77E-05

Moderately soluble

-4.73

5.67E-03

Molecule 13

-4.17

2.07E-02

6.77E-05

Moderately soluble

-4.73

5.67E-03

Molecule 14

-4.17

2.07E-02

6.77E-05

Moderately soluble

-4.73

5.67E-03

Molecule 15

-5.04

3.39E-03

9.07E-06

Moderately soluble

-5.65

8.46E-04

Molecule 16

-3.37

1.21E-01

4.26E-04

Soluble

-4.59

7.20E-03

Molecule 17

-5.95

4.72E-04

1.11E-06

Moderately soluble

-6.96

4.64E-05

Molecule 18

-3.17

1.71E-01

6.74E-04

Soluble

-3.87

3.42E-02

Molecule 19

-4.38

1.34E-02

4.16E-05

Moderately soluble

-5.24

1.86E-03

 

Table 5. Cont…….

Molecule

Solubility in water

Solubility (mol/L)

Class

Silicos-IT LogSw

Silicos-IT Solubility (mg/mL)

Silicos-IT Solubility (mol/L)

Silicos-IT class

Molecule 1

6.36E-06

Moderately soluble

-3.46

1.87E-01

3.44E-04

Soluble

Molecule 2

1.55E-04

Soluble

-3.44

8.57E-02

3.60E-04

Soluble

Molecule 3

9.40E-05

Moderately soluble

-4

2.77E-02

1.01E-04

Soluble

Molecule 4

9.40E-05

Moderately soluble

-4

2.77E-02

1.01E-04

Soluble

Molecule 5

9.40E-05

Moderately soluble

-4

2.77E-02

1.01E-04

Soluble

Molecule 6

9.40E-05

Moderately soluble

-4

2.77E-02

1.01E-04

Soluble

Molecule 7

7.40E-05

Moderately soluble

-4.27

1.58E-02

5.40E-05

Moderately soluble

Molecule 8

2.71E-05

Moderately soluble

-4.33

1.37E-02

4.72E-05

Moderately soluble

Molecule 9

2.91E-05

Moderately soluble

-4.27

1.70E-02

5.37E-05

Moderately soluble

Molecule 10

1.79E-04

Soluble

-3.85

3.59E-02

1.42E-04

Soluble

Molecule 11

1.11E-04

Soluble

-4.39

1.16E-02

4.03E-05

Moderately soluble

Molecule 12

1.85E-05

Moderately soluble

-4.31

1.50E-02

4.90E-05

Moderately soluble

Molecule 13

1.85E-05

Moderately soluble

-4.31

1.50E-02

4.90E-05

Moderately soluble

Molecule 14

1.85E-05

Moderately soluble

-4.31

1.50E-02

4.90E-05

Moderately soluble

Molecule 15

2.26E-06

Moderately soluble

-5.15

2.63E-03

7.03E-06

Moderately soluble

Molecule 16

2.54E-05

Moderately soluble

-2.81

4.35E-01

1.53E-03

Soluble

Molecule 17

1.10E-07

Poorly soluble

-5.53

1.26E-03

2.99E-06

Moderately soluble

Molecule 18

1.35E-04

Soluble

-2.87

3.45E-01

1.36E-03

Soluble

Molecule 19

5.78E-06

Moderately soluble

-4.04

2.93E-02

9.08E-05

Moderately soluble

 

 

Table 6. Pharmacokinetics

Molecule No

Pharmacokinetics

GI absorption

BBB permeant

Pgp substrate

CYP1A2 inhibitor

CYP2C19 inhibitor

CYP2C9 inhibitor

CYP2D6 inhibitor

CYP3A4 inhibitor

log Kp (cm/s)

Molecule 1

Low

No

Yes

No

No

No

No

No

-8.71

Molecule 2

High

Yes

No

Yes

Yes

No

No

No

-5.67

Molecule 3

High

Yes

No

Yes

Yes

No

No

No

-5.74

Molecule 4

High

Yes

No

Yes

Yes

No

No

No

-5.74

Molecule 5

High

Yes

No

Yes

Yes

No

No

No

-5.74

Molecule 6

High

Yes

No

Yes

Yes

No

No

No

-5.74

Molecule 7

High

Yes

No

No

Yes

No

No

No

-5.78

Molecule 8

High

Yes

No

Yes

Yes

Yes

No

No

-5.47

Molecule 9

High

Yes

No

Yes

Yes

Yes

No

No

-5.66

Molecule 10

High

Yes

No

Yes

Yes

No

No

No

-5.8

Molecule 11

High

Yes

No

Yes

Yes

No

No

No

-5.88

Molecule 12

High

Yes

No

Yes

Yes

Yes

No

No

-5.46

Molecule 13

High

Yes

No

Yes

Yes

Yes

No

No

-5.46

Molecule 14

High

Yes

No

Yes

Yes

Yes

No

No

-5.46

Molecule 15

Low

No

No

No

Yes

Yes

No

No

-5.25

Molecule 16

High

No

No

No

Yes

No

No

No

-6.07

Molecule 17

Low

No

No

No

Yes

Yes

No

Yes

-4.78

Molecule 18

High

No

No

Yes

Yes

No

No

No

-6.02

Molecule 19

High

Yes

No

Yes

Yes

Yes

No

No

-5.34

 

Table 7. Drugsimilarity

Molecule No

Drugsimilarity

Lipinski violations

Ghose violations

Veber violations

Egan violations

Muegge violations

Bioavailability Score

Molecule 1

3

2

1

1

3

0.17

Molecule 2

0

0

0

0

0

0.55

Molecule 3

0

0

0

0

0

0.55

Molecule 4

0

0

0

0

0

0.55

Molecule 5

0

0

0

0

0

0.55

Molecule 6

0

0

0

0

0

0.55

Molecule 7

0

0

0

0

0

0.55

Molecule 8

0

0

0

0

0

0.55

Molecule 9

0

0

0

0

0

0.55

Molecule 10

0

0

0

0

0

0.55

Molecule 11

0

0

0

0

0

0.55

Molecule 12

0

0

0

0

0

0.55

Molecule 13

0

0

0

0

0

0.55

Molecule 14

0

0

0

0

0

0.55

Molecule 15

0

1

0

1

0

0.55

Molecule 16

0

0

0

0

0

0.55

Molecule 17

0

1

0

1

1

0.55

Molecule 18

0

0

0

0

0

0.55

Molecule 19

0

0

0

0

0

0.55

 


Table 8. Medicinal Chemistry

Molecule No

Medicinal Chemistry

PAINS alerts

Brenk alerts

Leadlikeness violations

Synthetic Accessibility

Molecule 1

1

1

1

5.81

Molecule 2

0

1

1

3.81

Molecule 3

0

1

0

3.83

Molecule 4

0

1

0

3.82

Molecule 5

0

1

0

3.77

Molecule 6

0

1

0

3.77

Molecule 7

0

2

0

3.82

Molecule 8

0

1

1

3.74

Molecule 9

0

1

1

3.79

Molecule 10

0

1

0

3.78

Molecule 11

0

1

0

3.76

Molecule 12

0

1

1

3.8

Molecule 13

0

1

1

3.78

Molecule 14

0

1

1

3.76

Molecule 15

0

1

2

3.91

Molecule 16

0

3

0

3.87

Molecule 17

0

2

2

3.9

Molecule 18

0

1

0

3.76

Molecule 19

0

1

1

3.81

 

CONCLUSION:

In this work, 18 novel fluorinated molecules i.e. 3-Ethyl-5-fluoro-2-phenylimino-thiazolidin-4-one derivatives were evaluated for their in-silico analysis against C-KIT Tyrosine Kinase Protein 1T46. Molecular docking analysis revealed that compounds 13 and 19 were found to be potent moieties. The ADME study revealed that most of the compounds exhibit low adverse effects. These initial findings serve as a lead for the development of more potent and selectively targeted hybrid anticancer drugs.

 

CONFLICT OF INTEREST:

The authors declare that there are no conflict of interests regarding the publication of this article.

 

REFERENCES:

1.      Daina, A.; Michielin, O.; Zoete, V. ILOGP: A Simple, Robust, and Efficient Description of n-Octanol/Water Partition Coefficient for Drug Design Using the GB/SA Approach. J. Chem. Inf. Model. 2014; 54(12): 3284–3301. https://doi.org/https://doi.org/10.1021/ci500467k.

2.      Daina, A.; Zoete, V. A BOILED-Egg To Predict Gastrointestinal Absorption and Brain Penetration of Small Molecules. ChemMedChem. 2016; 11(11): 1117–1121. https://doi.org/10.1002/cmdc.201600182.

3.      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. 2017; 7 (42717): 1–13. https://doi.org/10.1038/srep42717.

4.      Lengauer, T.; Rarey, M. Computational Methods for Biomolecular Docking. Curr. Opin. Struct. Biol. 1996; 6(3): 402–406. https://doi.org/10.1016/S0959-440X(96)80061-3.

5.      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 Derivatives. Res. J. Pharm. Technol. 2019; 12(12): 5653–5663. https://doi.org/10.5958/0974-360X.2019.00978.8.

6.      Girija, K.; Jamuna, B. Design and Synthesis of Some Novel Schiff’s Base Aryl Imidazole Derivatives, Characterization, Docking and Study of Their Anti-Microbial Activity. Res. J. Pharm. Technol. 2015; 8(4): 407–415. https://doi.org/10.5958/0974-360X.2015.00069.4.

7.      Subramnian, G.; Rajagopal, K.; Sherin, F. Molecular Docking Studies, in Silico ADMET Screening of Some Novel Thiazolidine Substituted Oxadiazoles as Sirtuin 3 Activators Targeting Parkinson’s Disease. Res. J. Pharm. Technol. 2020; 13(6): 2708–2714. https://doi.org/10.5958/0974-360X.2020.00482.5.

8.      Meng, X.-Y.; Zhang, H.-X.; Mezei, M.; Cui, M. Molecular Docking: A Powerful Approach for Structure-Based Drug Discovery. Curr Comput Aided Drug Des. 2011; 7(2): 146–157.

9.      R. D. Taylor, P.J. Jewsbury, J. W. E. A Review of Protein-Small Molecule Docking Methods. J. Comput. Aided. Mol. Des. 2002; 16: 151–166.

10.   Yunta, M. J. R. Docking and Ligand Binding Affinity: Uses and Pitfalls. Am. J. Model. Optim. 2016; 4(3): 74–114. https://doi.org/10.12691/ajmo-4-3-2.

11.   Pantsar, T.; Poso, A. Binding Affinity via Docking: Fact and Fiction. Molecules. 2018; 23(8). https://doi.org/10.3390/molecules23081899.

12.   Butina, D.; Segall, M. D.; Frankcombe, K. Predicting ADME Properties in Silico: Methods and Models. Drug Discov. Today. 2002; 7(11): 83–88. https://doi.org/10.1016/S1359-6446(02)02288-2.

13.   Szakács, G.; Váradi, A.; Özvegy-Laczka, C.; Sarkadi, B. The Role of ABC Transporters in Drug Absorption, Distribution, Metabolism, Excretion and Toxicity (ADME-Tox). Drug Discov. Today. 2008; 13(9): 379–393. https://doi.org/10.1016/j.drudis.2007.12.010.

14.   Yu, H.; Adedoyin, A. ADME-Tox in Drug Discovery: Integration of Experimental and Computational Technologies. Drug Discov. Today. 2003; 8 (18): 852–861. https://doi.org/10.1016/S1359-6446(03)02828-9.

15.   Selick, H. E.; Beresford, A. P.; Tarbit, M. H. The Emerging Importance of Predictive ADME Simulation in Drug Discovery. Drug Discov. Today. 2002; 7(2): 109–116. https://doi.org/10.1016/S1359-6446(01)02100-6.

16.   Di, L. Strategic Approaches to Optimizing Peptide ADME Properties. AAPS J. 2015; 17(1): 134–143. https://doi.org/10.1208/s12248-014-9687-3.

17.   Ekins, S.; Waller, C. L.; Swaan, P. W.; Cruciani, G.; Wrighton, S. A.; Wikel, J. H. Progress in Predicting Human ADME Parameters in Silico. J. Pharmacol. Toxicol. Methods. 2000; 44(1): 251–272. https://doi.org/10.1016/S1056-8719(00)00109-X.

18.   Oubella, A.; El Mansouri, A. E.; Fawzi, M.; Bimoussa, A.; Laamari, Y.; Auhmani, A.; Morjani, H.; Robert, A.; Riahi, A.; Youssef Ait Itto, M. Thiazolidinone-Linked1,2,3-Triazoles with Monoterpenic Skeleton as New Potential Anticancer Agents: Design, Synthesis and Molecular Docking Studies. Bioorg. Chem. 2021; 115: 105184. https://doi.org/10.1016/j.bioorg.2021.105184.

19.   Ansari, M. F.; Siddiqui, S. M.; Ahmad, K.; Avecilla, F.; Dharavath, S.; Gourinath, S.; Azam, A. Synthesis, Antiamoebic and Molecular Docking Studies of Furan-Thiazolidinone Hybrids. Eur. J. Med. Chem. 2016; 124: 393–406. https://doi.org/10.1016/j.ejmech.2016.08.053.

20.   Rahim, F.; Taha, M.; Ullah, H.; Wadood, A.; Selvaraj, M.; Rab, A.; Sajid, M.; Shah, S. A. A.; Uddin, N.; Gollapalli, M. Synthesis of New Arylhydrazide Bearing Schiff Bases/Thiazolidinone: α-Amylase, Urease Activities and Their Molecular Docking Studies. Bioorg. Chem. 2019; 91: 103112. https://doi.org/10.1016/j.bioorg.2019.103112.

21.   Genc Bilgicli, H.; Taslimi, P.; Akyuz, B.; Tuzun, B.; Gulcin, İ. Synthesis, Characterization, Biological Evaluation, and Molecular Docking Studies of Some Piperonyl-Based 4-Thiazolidinone Derivatives. Arch. Pharm. (Weinheim). 2020; 353(1): 1–9. https://doi.org/10.1002/ardp.201900304.

22.   Ahmed, S. A.; Odde, S.; Daga, P. R.; Bowling, J. J.; Mesbah, M. K.; Youssef, D. T.; Khalifa, S. I.; Doerksen, R. J.; Hamann, M. T. Latrunculin with a Highly Oxidized Thiazolidinone Ring: Structure Assignment and Actin Docking. Org. Lett. 2007; 9(23): 4773–4776. https://doi.org/10.1021/ol7020675.

23.   Adnan, A. M. A.; Mahdi, M. F.; Khan, A. K. New 2-Methyl Benzimidazole Derivatives Bearing 4-Thiazolidinone Heterocyclic Rings: Synthesis, Preliminary Pharmacological Assessment and Docking Studies. Res. J. Pharm. Technol. 2021; 14(3): 1515–1520. https://doi.org/10.5958/0974-360X.2021.00269.9.

24.   Kotte, D.; Gullapelli, K.; Gavaji, B.; Merugu, R.; Maroju, R.; Patwari, M. An Efficient Synthesis, Anti Inflammatory Activity and Molecular Docking Studies of New Triazinanes and Iminothiazolidinones. Res. J. Pharm. Technol. 2020; 13(10): 4743. https://doi.org/10.5958/0974-360x.2020.00836.7.

25.   Puttaraj, C.; Bhalgat, C. M.; Chitale, S. K.; Ramesh, B. Synthesis and Biological Activities of Some Novel Heterocyclic Compounds Containing Thiazolidinone Derivatives. Res. J. Pharm. Technol. 2011; 4(6): 972–975.

26.   Govindarao, K.; Srinivasan, N.; Suresh, R. Synthesis, Characterization and Antimicrobial Evaluation of Novel Schiff Bases of Aryl Amines Based 2-Azetidinones and 4-Thiazolidinones. Res. J. Pharm. Technol. 2020; 13(1): 168. https://doi.org/10.5958/0974-360x.2020.00034.7.

27.   Mulay, A.; Ghodke, M.; Nikalje, A. P. G. Exploring Potential of 4-Thiazolidinone. Int. J. Pharm. Pharm. Sci. 2009; 1(1). https://doi.org/10.1002/chin.201035248.

28.   Kadam, S. D.; Mammen, D.; Kadam, D. S.; Patil, S. G.; Bagul, R. R.; Doshi, A.; Patel, F. Synthesis of Novel Fluorinated 5-Benzylidine-3-Ethyl-2-(2,3,4-Trifluorophenylimino)Thiazolidin-4-One Derivatives Using Knoevenagel Reaction and Evaluation of Their in Vitro Antimicrobial Potentials. Asian J. Chem. 2023; 35(8): 1884–1890. https://doi.org/https://doi.org/10.14233/ajchem.2023.28052.

29.   Szychowski, K. A.; Leja, M. L.; Kaminskyy, D. V.; Binduga, U. E.; Pinyazhko, O. R.; Lesyk, R. B.; Gmiński, J. Study of Novel Anticancer 4-Thiazolidinone Derivatives. Chem. Biol. Interact. 2017; 262: 46–56. https://doi.org/10.1016/j.cbi.2016.12.008.

30.   Senkardes, S.; Kucukguzel, S. Recent Progress on Synthesis and Anticancer Activity of 4-Thiazolidinone. Mini. Rev. Org. Chem. 2016; 13(5): 377–388. https://doi.org/10.2174/1570193x13666160826154159.

31.   Kobylinska, L. I.; Boiko, N. M.; Panchuk, R. R.; Grytsyna, I. I.; Klyuchivska, O. Y.; Biletska, L. P.; Lesyk, R. B.; Zimenkovsky, B. S.; Stoika, R. S. Putative Anticancer Potential of Novel 4-Thiazolidinone Derivatives: Cytotoxicity toward Rat C6 Glioma in Vitro and Correlation of General Toxicity with the Balance of Free Radical Oxidation in Rats. Croat. Med. J. 2016; 57(2): 151–164. https://doi.org/10.3325/cmj.2016.57.151.

32.   Ture, A.; Ergül, M.; Ergül, M.; Altun, A.; Küçükgüzel, I. Design, Synthesis, and Anticancer Activity of Novel 4-Thiazolidinone-Phenylaminopyrimidine Hybrids. Mol. Divers. 2020. https://doi.org/10.1007/s11030-020-10087-1.

33.   Kulabaş, N.; Ozakpinar, O. B.; Özsavcı, D.; Leyssen, P.; Neyts, J.; Küçükgüzel, İ. Synthesis, Characterization and Biological Evaluation of Thioureas, Acylthioureas and 4-Thiazolidinones as Anticancer and Antiviral Agents. Marmara Pharm. J. 2017; 21(2): 371–384. https://doi.org/10.12991/marupj.300913.

34.   Senkardes, S.; Kucukguzel, S. G. Recent Progress on Synthesis and Anticancer Activity of 4-Thiazolidinone. 2021; No. 13(5).

35.   Mahmood, F. F. Optimization Geometry of Benzamide and Di-Fluorine Benzamide Molecules. Res. J. Pharm. Technol. 2018; 11(9): 3978–3982. https://doi.org/10.5958/0974-360X.2018.00731.X.

36.   Nair, N. P.; Joy, J.; Kumar, S. S.; Sathianarayanan, S.; Manakadan, A. A.; Saranya, T. S. In- Silico Docking Studies of Coumarin Derivatives as Caspase 8 and PDE4 Antagonist. Res. J. Pharm. Technol. 2016; 9(12): 2199–2204. https://doi.org/10.5958/0974-360X.2016.00445.5.

37.   Kadam, D.; Patil, S.; Kadam, S.; Doshi, A.; Patel, F. Synthesis of Novel 5-Arylidine-3-Ethyl-2-(2, 4, 5-Trifluorophenylimino)-Thiazolidin-One Derivatives Using Ultrasonic Knoevengel Conditions and Evaluation of Its Antimicrobial Activity. JETIR 2022; 9(8): 691–698. https://doi.org/http://doi.one/10.1729/Journal.31452.

38.   Kadam, S. D.; Mammen, D.; Kadam, D. S.; Patil, S. G. In Silico Molecular Docking against C-KIT Tyrosine Kinase and ADME Studies of 3-Ethyl-2-(2,3,4-Trifluoro-Phenylimino)-Thiazolidin-4-One Derivatives. Asian J. Res. Chem. 2023; 16(1): 13–22. https://doi.org/http://dx.doi.org/10.52711/0974-4150.2023.00010.

39.   Kadam, D. S.; Patil, G. S.; Mammen, D.; Kadam, S. D.; More, V. In Silico Molecular Docking Againstc- KIT Tyrosine Kinase and ADME Studies of 4- Thiazolidinone Derivatives. J. Appl. Organomet. Chem. 2023; 3(1): 13–27. https://doi.org/https://doi.org/10.22034/jaoc.2023.355363.1058.

40.   Shaty, M. H.; Al-Ezzi, M. I.; Arif, I. S.; Basil, D. Effect of Metformin on Inflammatory Markers Involved in Cardiotoxicity Induced by Doxorubicin. Res. J. Pharm. Technol. 2019; 12(12): 5815–5821. https://doi.org/10.5958/0974-360X.2019.01007.2.

41.   Parameswari, P.; Devika, R. In Silico Molecular Docking Studies of Quercetin Compound against Anti-Inflammatory and Anticancer Proteins. Res. J. Pharm. Technol. 2019; 12(11): 5305–5309. https://doi.org/10.5958/0974-360X.2019.00919.3.

42.   Nikhila, G.; Naveen, P.; Udayakumar, D.; Manjunatha, K. Indole-3-Carbinol and 1,3,4-Oxadiazole Hybrids: Synthesis and Study of Anti-Proliferative and Anti-Microbial Activity. Aust. J. Chem. 2015; 68: 1603–1613. https://doi.org/https://doi.org/10.1071/CH15116.

43.   Mol, C. D.; Dougan, D. R.; Schneider, T. R.; Skene, R. J.; Kraus, M. L.; Scheibe, D. N.; Snell, G. P.; Zou, H.; Sang, B. C.; Wilson, K. P. Structural Basis for the Autoinhibition and STI-571 Inhibition of c-Kit Tyrosine Kinase. J. Biol. Chem. 2004; 279(30): 31655–31663. https://doi.org/10.1074/jbc.M403319200.

 

 

 

 

Received on 25.11.2023            Modified on 11.03.2024

Accepted on 15.05.2024           © RJPT All right reserved

Research J. Pharm. and Tech 2024; 17(9):4559-4568.

DOI: 10.52711/0974-360X.2024.00704