ISSN : 0970 - 020X, ONLINE ISSN : 2231-5039
     FacebookTwitterLinkedinMendeley

Synthesis and Cyclooxygenase-2 Inhibitory Activity Evaluation of Some Pyridazine Derivatives

Mohd Imran1*, Abida Ash Mohd1, Naira Nayeem1, Nawaf M. Al-Otaibi2, Malik Homoud3, Muhannad Thafi Alshammari 3

1Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.

2Department of Clinical Pharmacy, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.

3College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.

Corresponding Author E-mail: mohammad.Baks@nbu.edu.sa

DOI : http://dx.doi.org/10.13005/ojc/390504

Article Publishing History
Article Received on : 04 Sep 2023
Article Accepted on : 21 Oct 2023
Article Published : 31 Oct 2023
Article Metrics
Article Review Details
Reviewed by: Dr. Dattatraya N. Pansare
Second Review by: Dr. Jatin Mehta
Final Approval by: Dr.S. Geetha
ABSTRACT:

This work aimed to discover safe and effective pyridazine-based cyclooxygenase-2 (COX-2) inhibitors. Thirty-three pyridazine-based compounds (compounds 1 to 33) were designed. The in silico studies were conducted to predict their toxicity, docking scores (DS), pharmacokinetic parameters, and drug-likeliness properties compared to celecoxib. Based on the safety and efficacy data obtained by in silico studies, four compounds (7, 12, 16, and 24) were synthesized, and the spectral analysis confirmed their chemical structures. Additionally, the in vitro COX-2 inhibitory activity of these four compounds was evaluated. Eleven compounds were predicted as non-toxic compounds. The DS of four compounds, 7 (DS = -9.72 kcal/mol), 12 (DS = -10.48 kcal/mol), 16 (DS = -9.71 kcal/mol), and 24 (DS = -9.46 kcal/mol), was better than celecoxib (DS = -9.15). These compounds (7, 12, 16, and 24) also demonstrated better oral absorption (83.53% each) than celecoxib (79.20%) in addition to their promising drug-likeliness properties. The compounds 7 (101.23%; p < 0.05), 12 (109.56%; p < 0.05), 16 (108.25%; p < 0.05), and 24 (103.90%; p < 0.05) also exhibited superior COX-2 inhibition to celecoxib (100%; p < 0.05). Compounds 7, 12, 16, and 24 are useful lead compounds in developing drugs for various diseases in which high levels of COX-2 are implicated.

KEYWORDS:

Benzothiazole; COX-2 inhibition; in silico studies; Pyridazine; synthesis

Download this article as: 

Copy the following to cite this article:

Imran M, Mohd A. A, Nayeem N, Al-Otaibi N. M, Homoud M, Alshammari M. T. Synthesis and Cyclooxygenase-2 Inhibitory Activity Evaluation of Some Pyridazine Derivatives. Orient J Chem 2023;39(5).


Copy the following to cite this URL:

Imran M, Mohd A. A, Nayeem N, Al-Otaibi N. M, Homoud M, Alshammari M. T. Synthesis and Cyclooxygenase-2 Inhibitory Activity Evaluation of Some Pyridazine Derivatives. Orient J Chem 2023;39(5). Available from: https://bit.ly/3Qmzb8m


Introduction

Cyclooxygenase-2 (COX-2), an inducible enzyme, is produced during the inflammation of the body cells1. The untreated inflammation may progress to many diseases, including disability-causing diseases (different types of arthritis and Alzheimer’s disease), cancer, pancreatitis, hepatitis, atherosclerosis, CNS diseases (epilepsy and depression), asthma, irritable bowel disease, and kidney injury2. The commonly used non-steroidal anti-inflammatory drugs (NSAID) block COX-1 and COX-2 enzymes. COX-1, a constitutive cell enzyme as opposed to COX-2, supports the maintenance of kidney functions, platelet aggregation, and gastric mucosa activities. The adverse effects of NSAIDs (gastric ulcers and kidney malfunctions) are mostly caused by COX-1 inhibition1-3. As a result, the COX-2 enzyme inhibitors (celecoxib and rofecoxib) were created4. However, celecoxib and rofecoxib have recently demonstrated cardiac toxicity effects5. These effects make it imperative to develop better COX-2 inhibitors.

Pyridazine-based compounds demonstrate diverse biological activities6-12, including COX-2 inhibitory activity1,2. Emorfazone (Pentoil) (Figure 1) is a clinically used pyridazine-based anti-inflammatory agent and is claimed to lack ulcerogenic effects associated with traditional NSAIDs13.

Figure 1: Chemical structure of emorfazone.

Click here to View Figure

The Zomipirac-based pyridazine derivatives have also displayed potent COX-2 inhibition2. The potential for multiple structural modifications in the pyridazine ring and literature confirming the COX-2 inhibitory activity of the pyridazine nucleus makes the pyridazine-based compounds a suitable framework for creating non-ulcerogenic COX-2 inhibitors14. Accordingly, this study was planned to discover safe and effective COX-2 inhibitors.

Experimental

General

Sigma Aldrich (USA) provided the analytical-grade chemicals utilized in this study. The Gallenkamp melting point apparatus, Shimadzu 440 spectrophotometer (for generating FTIR data), Varian Gemini 125/500 MHz spectrophotometer (for recording 13C-NMR and 1H-NMR data, respectively), and a 70 eV GCMS/QP 1000 Ex mass spectrophotometer (for obtaining the mass spectra) were used to obtain the spectral data of the synthesized compounds.

Design of the compounds

Thirty-three compounds were designed using ChemDraw (version 21) software (Figure 2). The reaction between the intermediates disclosed in the United States Patent Number US4052395A and the commercially available 2-hydrazineylbenzo[d]thiazole served as the basis for designing these compounds 15.

Figure 2: Chemical structures of the designed compounds (DC).

Click here to View Figure

Toxicity prediction

All thirty-three DCs (compounds 1 to 33) were assessed for their toxicity properties employing the ProTox-II web server 16. The Mol-Files of the DC were created with the ChemDraw software. These Mol-Files were opened with the notepad, and the contents were copied and pasted into the ProTox-II web server. The start button was pressed to obtain the toxicity data of the compounds (Table 1).

Molecular docking

The Molecular Operating Environment software (MOE) (2019.0102 version, Chemical Computing Group Inc., Canada) was utilized for this study. The COX-2 protein (PDB ID: 5-KIR) was utilized for this purpose, employing celecoxib as a standard 1,2,13. The 5-KIR protein was uploaded into the software, and the Quickprep button was pressed to obtain the purified and ready-to-use 5-KIR protein for docking. The MDB files of compounds (7, 8, 12, 14, 16, 17, 19, 20, 21, 22, 24, and celecoxib) were created. The docking was started utilizing ready-to-use 5-KIR proteins and the MDB files of the compounds, and the docking scores (DS in kcal/mol) and root mean square deviation (RMSD) of each docked compound were noted (Table 2).

Prediction of the pharmacokinetic parameters

The pharmacokinetic parameters were predicted by Swiss-ADME software 16,17. The Mole-Files of the compounds were inserted in the software with the import button, the run button was pressed, and the data was recorded (Table 2). The % absorption of the compounds was also calculated utilizing the following formula 3.

%Absorption = 109 – (0.345 x TPSA)

Synthesis of compounds 7, 12, 16, and 24

An equimolar mixture of 4-(3,5-dichloro-4-propylphenyl)-4-oxobutanoic acid (0.01 mole) and 2-hydrazineylbenzo[d]thiazole (0.01 mole) was refluxed in ethanol (50 ml) in the presence of a catalytic amount of sodium acetate for two hours. A precipitate was obtained, which was filtered and recrystallized from ethanol to obtain compound 7.  This method was also used to synthesize compounds 12, 16, and 24 by replacing 4-(3,5-dichloro-4-propylphenyl)-4-oxobutanoic acid with 4-(4-butyl-3,5-dichlorophenyl)-4-oxobutanoic acid, 4-oxo-4-(3,4,5-trichlorophenyl)butanoic acid, and 4-(3,4-dichlorophenyl)-4-oxobutanoic acid, respectively (Scheme 1). Table 3 provides the physical and spectral information for compounds 7, 12, 16, and 24.

Scheme 1: Synthesis of compounds 7, 12, 16, and 24

Click here to View Scheme

In vitro COX-2 inhibitory activity

The COX-1/COX-2 inhibitory activity of the compounds 7, 12, 16, 24, celecoxib, and indomethacin was carried out by the 10-fold dilution method (1-10−4 µg/mL) employing Cayman’s human COX-1/COX-2 kit (560131, Ann Arbor, MI, USA) 1-3. The reagent preparation and experiment execution followed the supplier’s instructions. The previous publication also provides a brief procedure of this experiment and calculations of the IC50 values for COX-1/COX-2 inhibition by regression analysis 1-3. The selectivity index (SI = IC50 for COX-1/IC50 for COX-2) was also calculated with the obtained data (Table 4).

Statistical analysis

The experimental data were statistically analyzed using the SPSS software (version 20, Chicago, IL, USA). Results are statistically significant if p < 0.05 (N = 3; Mean ± SD) is achieved.

Results

The toxicity study of the thirty-three DC was assessed with the ProTox II software16. The results revealed that nineteen compounds were hepatotoxic, six showed carcinogenic properties, and three demonstrated both hepatotoxic and carcinogenic behavior. Overall, twenty-two DCs displayed either hepatotoxic or carcinogenic properties. Eleven DCs were found to be non-toxic (Table 1).

Table 1: Predicted toxicity data of the non-toxic DC

Compound

Oral LD50 (mg/kg)

Oral toxicity class

Hepatotoxicity

Carcinogenicity

Immunotoxicity

Mutagenicity

Cytotoxicity

Celecoxib

1400

4

No

Yes

No

No

No

7

1000

4

No

No

No

No

No

8

1000

4

No

No

No

No

No

12

1000

4

No

No

No

No

No

14

1000

4

No

No

No

No

No

16

1000

4

No

No

No

No

No

17

1000

4

No

No

No

No

No

19

1000

4

No

No

No

No

No

20

1000

4

No

No

No

No

No

21

1000

4

No

No

No

No

No

22

1000

4

No

No

No

No

No

24

1000

4

No

No

No

No

No

The eleven compounds were selected for their docking and Swiss-ADME analyses (Table 2)1,2,13,17. The molecular docking study of eleven compounds for COX-2 protein (PDB ID: 5-KIR) revealed that compounds 7 (DS = -9.72 kcal/mol), 12 (DS = -10.48 kcal/mol), 16 (DS = -9.71 kcal/mol), and 24 (DS = -9.46 kcal/mol) demonstrated better DS than celecoxib (DS = -9.15). Other compounds exhibited low docking scores concerning celecoxib. The RMSD values of the compounds were < 1.5, representing good binding of the compounds with the target site16. Compounds 7, 12, 16, and 24 had better absorption (83.53% each) than celecoxib (79.20%). The compounds 14 and 17 also had better absorption than celecoxib but a lower docking score than celecoxib. Other compounds had less absorption and DS than celecoxib. None of the compounds was permeant to the blood-brain barrier, except compound 24. None of the compounds was an inhibitor of CYP2D6 or a substrate for P-gp. Compounds 19, 20, 21 and 22 tended to inhibit CYP3A4. All the compounds qualified the Lipinski rule of drug-likeliness.

Table 2: The DS, pharmacokinetic parameters, and drug-likeness properties of the compounds

Compounds

DS (kcal/mol)

RMSD (Å)

Log P (o/w)

TPSA (Å2)

Pharmacokinetics

Drug-likeness

(Lipinski rule)

Calculated % absorption

GI absorption

BBB permeant

P-gp substrate

CYP2D6 inhibitor

CYP3A4 inhibitor

Celecoxib

-9.15

0.88

3.40

86.36

High

No

No

No

No

Yes

79.20

7

-9.72

1.19

5.36

73.80

High

No

No

No

No

Yes

83.53

8

-7.62

1.44

4.32

94.03

High

No

No

No

No

Yes

76.55

12

-10.48

1.19

5.70

73.80

High

No

No

No

No

Yes

83.53

14

-7.10

0.59

4.46

73.80

High

No

No

No

No

Yes

83.53

16

-9.71

1.07

4.87

73.80

High

No

No

No

No

Yes

83.53

17

-7.96

0.98

4.46

73.80

High

No

No

No

No

Yes

83.53

19

-7.77

1.12

3.82

94.03

High

No

No

No

Yes

Yes

76.55

20

-7.26

0.82

3.98

94.03

High

No

No

No

Yes

Yes

76.55

21

-6.65

1.26

3.52

94.03

High

No

No

No

Yes

Yes

76.55

22

-7.24

0.77

3.46

94.03

High

No

No

No

Yes

Yes

76.55

24

-9.46

1.06

4.38

73.80

High

Yes

No

No

No

Yes

83.53

TPSA = Topological polar surface area.

Based on the predicted toxicity, molecular docking, and the Swiss-ADME data, compounds 7, 12, 16, and 24 were selected for their synthesis (Scheme 1). The compounds’ spectral data corresponded to the assigned structures of 7, 12, 16, and 24 (Table 3).

Table 3: Characterization data of compounds 7, 12, 16, and 24

Compound

(MF; MW; M.P.; Rf values*; FTIR in KBr, ν in cm-1)

1H-NMR

(DMSO-d6, 500 MHz, δ in ppm)

13C-NMR

(DMSO-d6, 125 MHz, δ in ppm)

Mass (m/z)

7

(C20H17Cl2N3OS; 418; 188-190oC; 0.73; 1660 (C=O),

1570 (C=N), 1522 (C=C) and 1113 (C–S))

0.95 (t, 3H, -CH3), 1.62 (m, 2H, -CH2-CH3), 2.42 (t, 2H, methylene of C4-pyridazine), 2.62 (t, 2H, –CH2-CH2-CH3), 2.90 (t, 2H, methylene of C5-pyridazine), 7.50-7.52 (dd, 2H, Ar-H), 7.75 (s, 2H, Ar-H), 8.14-8.17 (dd, 2H, Ar-H)

11.6 (-CH3), 21.0 (-CH2-CH3), 22.3 (C4, pyridazine), 25.6 (-CH2-CH2-CH3), 30.3 (C5, pyridazine), 117.2, 120.7, 123.4, 124.2, 126.3 (2C), 129.7, 133.0, 133.8 (2C), 140.8, 145.4, 152.1, 167.0 (C=O, pyridazine), 173.4 (C2, benzothiazole)

418 (M+, 100%), 419 (M++1) 420 (M++2), 284, 231, 188, 135

12

(C21H19Cl2N3OS; 432; 176-178oC; 0.71; 1661 (C=O), 1571 (C=N), 1521 (C=C), 1111 (C–S))

0.88 (t, 3H, -CH3), 1.31 (m, 2H, –CH2-CH3), 1.51 (m, 2H, –CH2-CH2-CH3), 2.42 (t, 2H, methylene of C4-pyridazine), 2.61 (t, 2H, –CH2-CH2-CH2-CH3), 2.90 (t, 2H, methylene of C5-pyridazine), 7.50-7.53 (dd, 2H, Ar-H), 7.76 (s, 2H, Ar-H), 8.12-8.17 (dd, 2H, Ar-H)

12.1 (-CH3), 20.3 (-CH2-CH3), 22.4 (C5, pyridazine), 23.2 (-CH2CH2-CH2-CH3), 28.3 (-CH2-CH2-CH2-CH3), 31.4 (C4, pyridazine), 111.2, 120.7, 123.4, 124.2, 126.0 (2C), 129.8, 133.0, 133.9 (2C), 141.4, 145.4, 152.1, 167.0 (C=O, pyridazine), 173.4 (C2, benzothiazole)

432 (M+, 100%), 433 (M++1), 434 431 (M++2), 284, 231, 202, 135

16

(C17H10Cl3N3OS; 410; 192-194oC; 0.74; 1665 (C=O), 1573 (C=N), 1525 (C=C), 1115 (C–S))

2.42 (t, 2H, methylene of C4-pyridazine), 2.91 (t, 2H, methylene of C5-pyridazine), 7.72 (s, 2H, Ar-H), 7.50-7.53 (dd, 2H, Ar-H), 8.12-8.16 (dd, 2H, Ar-H)

22.3 (C5, pyridazine), 30.3 (C4, pyridazine), 117.2, 120.7, 123.4, 124.2, 127.6 (2C), 129.7, 133.8, 134.5, 141.4 (2C), 145.4, 152.1, 167.0 (C=O, pyridazine), 173.4 (C2, benzothiazole)

410 (M+, 100%), 411 (M++1), 412 (M++1), 284, 231, 179, 135

24

(C17H11Cl2N3OS; 376; 181-183oC; 0.78; 1663 (C=O), 1572 (C=N), 1522 (C=C), 1114 (C–S))

2.41 (t, 2H, methylene of C4-pyridazine), 2.90 (t, 2H, methylene of C5-pyridazine), 7.50-7.52 (dd, 2H, Ar-H), 7.67 (d, 1H, Ar-H), 7.84-7.87 (dd, 2H, Ar-H), 8.12-8.16 (dd, 2H, Ar-H)

22.3 (C5, pyridazine), 30.3 (C4, pyridazine), 117.2, 120.7, 123.4, 124.2, 125.2, 129.2 (2C), 129.7, 132.4 (2C), 134.6, 145.4, 152.1, 167.0 (C=O, pyridazine), 173.4 (C2, benzothiazole)

376 (M+, 100%), 377 (M++1) 378 (M++2), 284, 231, 145, 135

MF: Molecular formula; MW: Molecular weight; M.P.: Melting point; *Rf values in a benzene and acetone mixture (8:2).

Compounds 7, 12, 16, and 24 were evaluated for their COX-1 and COX-2 inhibition against indomethacin (non-specific COX inhibitor) and celecoxib (COX-2 inhibitor) (Table 4)1-3. The results of Table 4 indicate that compounds 7, 12, 16, and 24 were more effective COX-2 inhibitors than celecoxib but less effective COX-1 inhibitors than indomethacin (Figure 3).

Table 4: In vitro COX-1/COX-2 inhibitory activity of 7, 12, 16, and 24

Compounds

COX-1

(IC50, nM*)

%COX-1 inhibition

COX-2

(IC50, nM*)

%COX-2 inhibition

SI

%SI

7

360.5 ± 0.40

59.36

17.88 ± 0.18

101.23

20.16

109.62

12

365.30 ± 0.28

58.58

16.52 ± 0.25

109.56

22.11

120.22

16

380.10 ± 0.15

56.30

16.72 ± 0.38

108.25

22.73

123.59

24

376.35 ± 0.40

56.86

17.42 ± 0.50

103.90

21.60

117.45

Celecoxib

333.0 ± 0.22

64.26

18.10 ± 0.51

100

18.39

100

Indomethacin

214.0 ± 0.14

100

69.10 ± 0,32

26.19

3.09

16.80

*p < 0.05; SI = Selectivity index.

Figure 3: COX-2 inhibitory activity of 7, 12, 16, 24,  celecoxib, and indomethacin

Click here to View Figure

Discussion

This study focused on discovering effective and safe pyridazine-based COX-2 inhibitors for treating disorders linked to elevated COX-2 levels. We designed thirty-three pyridazine-based compounds and performed various experiments (in silico studies, molecular docking, synthesis, and COX-2 inhibition activity). The in silico toxicity data of the DC revealed similar LD50 (1000 mg/kg) and toxicity class (class 4) (Table 1). This is due to their structural similarities18. It is also imperative to note that celecoxib displayed carcinogenic behavior, but eleven compounds were non-toxic (Table 1). The eleven non-toxic compounds were subjected to their molecular docking study. In the molecular docking study, the larger negative value for the DS and an RMSD < 1.5 specifies a compound’s greater affinity and good binding of the compounds with COX-2, respectively1,2. The docking study revealed that compounds 7 (DS = -9.72 kcal/mol), 12 (DS = -10.48 kcal/mol), 16 (DS = -9.71 kcal/mol), and 24 (DS = -9.46 kcal/mol) were more potent than celecoxib (DS = -9.15 kcal/mol) in inhibiting the COX-2 enzyme (Table 2). Accordingly, compounds 7, 12, 16, and 24 were synthesized, and their chemical structures were confirmed by spectral analysis (Table 3).

The in vitro COX-2 inhibitory activity of compounds 7 (R = R2 = Cl; R1 = n-propyl; COX-2 inhibition = 101.23%), 12 (R = R2 = Cl; R1 = n-butyl; COX-2 inhibition = 109.56%), 16 (R = R1 = R2 = Cl; COX-2 inhibition = 108.25%), and 24 (R = R1 = Cl; R2 = H; COX-2 inhibition = 103.90%) was better than celecoxib (COX-2 inhibition = 100%) (Table 4). This phenomenon aligned with the molecular docking data (Table 2). It is known that the n-butyl group (compound 12) is more lipophilic than the n-propyl group (compound 7), and trichloro substituents (compound 16) provide higher lipophilicity to compounds than dichloro substituents (compound 24)19,20,21. The benzothiazole ring is a lipophilic ring, which may also contribute to the lipophilic character of DC22. This understanding is also evident from the Log P data (an indicator of the compound’s lipophilicity) that compounds 7, 12, 16, and 24 have greater lipophilicity than celecoxib (Table 1). This effect reveals that higher lipophilicity is required for DC’s potent COX-2 inhibitory activity. This understanding also aligns with previous studies suggesting that lipophilic compounds are better COX-2 inhibitors23.

The data of Table 2 also reflects that compounds 7, 12, 16, and 24 were not an inhibitor of CYP2D6/CYP3A4 or a substrate for P-gp, implying that these compounds may not pose metabolism-related drug interactions with other medicines24. These compounds also qualified Lipinski’s rule of drug-likeliness, suggesting the possibility of their development as a drug molecule1,13,17. The compounds 7, 12, 16, and 24 inhibit the COX-1 enzyme to a lesser extent than celecoxib and indomethacin (Table 4), signifying their better safety profile concerning the ulcerogenic effects associated with traditional NSAIDs1,2. Compounds 7, 12, 16, and 24 may be useful as lead compounds in developing drugs for various diseases (including some disability-causing diseases) in which high levels of COX-2 are implicated. Except for compound 24, none of the compounds could pass through the blood-brain barrier (Table 2). This fact implies compound 24 and its derivative may be more useful in developing anti-inflammatory agents for certain CNS diseases like epilepsy, Alzheimer’s disease, and depression (Figure 4).

Figure 4: Diseases instigated by elevated COX-2 levels 2

Click here to View Figure

Many pyridazine-based pharmacodynamic agents have been developed to treat various diseases like hypertension (indolidan and bemoradan), congestive heart failure (levosimendan and pimobendane), depression (minaprine), PDE3-associated illness (imazodan and zardaverine) and cancer (Olaparib)2,7. Accordingly, compounds 7, 12, 16, and 24 may further be assessed to check their efficacy against these diseases. The authors believe additional research is necessary to confirm these theories about compounds 7, 12, 16, and 24. Further, the chemical structures of our designed compounds can be altered easily and serve as a new template for developing safe, effective, and potent COX-2 inhibitors.

Conclusion

Four compounds (7, 12, 16, and 24) displayed in silico study-based non-toxic properties, appreciable pharmacokinetic parameters, and drug-likeliness assets. These compounds were also more effective than celecoxib at blocking COX-2 activity. The chemical structures of the DC can be altered easily and serve as a new template for developing safe, effective, and potent COX-2 inhibitors. Accordingly, compounds 7, 12, 16, and 24 and their derivatives may be useful in developing drugs against diseases demonstrating high levels of COX-2 enzyme. However, more research is advised to confirm these potential implications for our molecules.

Acknowledgment

The authors gratefully acknowledge the approval and the support of this research study by the grant no. PHAR-2023-12-2272 from the Deanship of Scientific Research at Northern Border University, Arar, K.S.A.

Conflict of Interest

No conflict of interest is associated with this work.

References

  1. Khan, A.; Diwan, A.; Thabet, H.K.; Imran, M. Drug Dev. Res. 2020, 81(5), 573-584.
    CrossRef
  2. Khan, A.; Diwan, A.; Thabet, H.K.; Imran, M.; Bakht, M.A. Molecules 2020, 25(9), 2002.
    CrossRef
  3. Imran, M.  Indian J. Het. Chem. 2020, 30(2), 291-295.
  4. Ahmadi, M.; Bekeschus, S.; Weltmann, K.D.; von Woedtke, T.; Wende, K. RSC Med. Chem. 2022, 13(5), 471-496.
    CrossRef
  5. Fanelli, A.; Ghisi, D.; Aprile, P.L.; Lapi, F. Ther. Adv. Drug Saf. 2017, 8(6), 173-182.
    CrossRef
  6. Imran, M.; Nayeem, N. Orient J. Chem. 2016, 32(1), 267-274.
    CrossRef
  7. Imran, M.; Abida. Trop. J. Pharm. Res. 2016, 15(7), 1579-1590.
    CrossRef
  8. Asif, M.; Imran, M. Anal. Chem. Lett. 2020, 10(3), 414-427.
    CrossRef
  9. Imran, M.; Asif, M. Russ. J. Bioorg. Chem. 2020, 46, 726-744.
    CrossRef
  10. Imran, M.; Asif, M. Russ. J. Bioorg. Chem. 2020, 46, 745-767.
    CrossRef
  11. Alghamdi, S.; Imran, M.; Kamal, M.; Asif, M. Pharm. Chem. J. 2021, 55, 915-919.
    CrossRef
  12. Alghamdi, S.; Imran, M., Kamal, M.; Asif, M. Pharm. Chem. J.2022,55, 1367-1371
    CrossRef
  13. Imran, M. Molbank 2020, M1155.
    CrossRef
  14. Singh, J.; Sharma, D.; Bansal, R. Future Med. Chem. 2017, 9(1), 95-127.
    CrossRef
  15. Jojima, T.; Takahi, Y. United States Patent Number US4052395A, October 4, 1977 (Available from: https://patents.google.com/patent/ US4052395A/en?oq=US4052395A).
  16. Imran, M.; Mohd, A.A.; Nayeem, N.; Alaqel, S.I. Trop. J. Pharm. Res. 2023, 22(6), 1263-1269.
  17. Daina, A.; Michielin, O.; Zoete, V. Sci. Rep. 2017, 7, 42717.
    CrossRef
  18. Yang, C.; Rathman, J.F.; Mostrag, A.; Ribeiro, J.V.; Hobocienski, B.; Magdziarz, T.; Kulkarni, S.; Barton-Maclaren, T. Chem. Res. Toxicol. 2023, 36(7), 1081-1106.
    CrossRef
  19. Ginex, T.; Vazquez, J.; Gilbert, E.; Herrero, E.; Luque, F.J. Future Med. Chem. 2019, 11(10), 1177-1193.
    CrossRef
  20. Bayliss, M.K.; Butler, J.; Feldman, P.L.; Green, D.V.; Leeson, P.D.; Palovich, M.R.; Taylor, A.J. Drug Discov. Today. 2016, 21(10), 1719-1727.
    CrossRef
  21. Tsopelas, F.; Giaginis, C.; Tsantili-Kakoulidou, A. Expert Opin. Drug Discov. 2017, 12(9), 885-896.
    CrossRef
  22. Asiri, Y.I.; Alsayari, A.; Muhsinah, A.B.; Mabkhot, Y.N.; Hassan, M.Z. J. Pharm. Pharmacol. 2020, 72(11), 1459-1480.
    CrossRef
  23. Ribeiro, D.; Proença, C.; Varela, C.; Janela, J.; Tavares da Silva, E.J.; Fernandes, E.; Roleira, F.M.F. Bioorg. Chem. 2019, 91, 103179.
    CrossRef
  24. Garnett, W.R. Pharmacotherapy 2001, 21(10), 1223-1232.
    CrossRef


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

About The Author