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

Docking Studies of Potent Xanthine Oxidase Inhibitors - Molecules Patented and Published From 2011-2020

Tripti Arora* and Ketki Rani

SGT College of Pharmacy, SGT University, Gurugram, Haryana-122505, India.

Corresponding Author E-mail: tripti_pharmacy@sgtuniversity.org

DOI : http://dx.doi.org/10.13005/ojc/380406

Article Publishing History
Article Received on : 11 May 2022
Article Accepted on : 22 July 2022
Article Published : 27 Jul 2022
Article Metrics
Article Review Details
Reviewed by: Dr.G.Dayana Jeyaleela
Second Review by: Dr. G. K. Ayyadurai
Final Approval by: Dr. Ioana Stanciu
ABSTRACT:

Hyperuricemia is characterized by elevated serum uric (SUA) levels beyond 6.8mg/dl and is a major cause of gout. Clinically, the high uric acid levels are managed using oral xanthine oxidase inhibitors such as febuxostat; however, its long-term use affects liver functions and cannot be used with the person with compromised liver functions. Thus, searching for an alternate xanthine oxidase inhibitor devoid of side effects on the liver is of current interest. Several classes of XO inhibitors have been patented in recent years. Using a docking study, we investigated the binding mode of xanthine oxide inhibitors patented during 2011-2020.The study identified the crucial amino acid residues involved in drug-receptor interactions. The binding side residues and their role in stabilizing the drug-receptor complex is discussed in detail. The information gained from this study will help researchers to design potent and selective xanthine oxidase inhibitors to manage gout and other uric acid-related disorders effectively.

KEYWORDS:

Docking; Gout; Hyperuricemia; Uric acid; Xanthine oxidase inhibitors

Download this article as: 

Copy the following to cite this article:

Arora T, Rani K. Docking Studies of Potent Xanthine Oxidase Inhibitors - Molecules Patented and Published From 2011-2020. Orient J Chem 2022;38(4).


Copy the following to cite this URL:

Arora T, Rani K. Docking Studies of Potent Xanthine Oxidase Inhibitors - Molecules Patented and Published From 2011-2020. Orient J Chem 2022;38(4). Available from: https://bit.ly/3owmLgD


Introduction

Xanthine oxidase (XO) is a molybdoflavoprotein mainly found in milk, lung, heart, kidney, and vascular endothelium. XO is involved in purine metabolism, producing uric acid and reactive oxygen species leading to high serum uric acid levels, inflammation, and oxidative damage1-3. Xanthine oxidase inhibitors have been used to control elevated uric acid levels, known as hyperuricemia resulting in gout4,5. The first drug to inhibit XO and control hyperuricemia is allopurinol (purine-based) (Figure-1) approved by FDA in 1966; however, its associated adverse effects limit its clinical use. A High dose of allopurinol induces hypersensitivity reactions in combination with several other side effects6. Another non-purine XO inhibitor for treating hyperuricemia in patients with severe chronic renal impairment is Febuxostat (Figure-1), approved by the FDA7. After the augmented risk of issues related to heart with febuxostat, FDA, in the year 2019, issued a ‘Boxed warning’ even though the mechanism for cardiac toxicity is unclear. Post-FDA, WHO also issued a worldwide warning for the use of febuxostat only if any other drug cannot soothe a patient condition8,9. The recent patent study highlighted that a large number of efforts are made toward the discovery of non-purine and natural compounds. The literature has emphasized on highly potent molecules acting as XO inhibitors have nanomolar IC50 Values. However, they also show off-target activities associated with several adverse effects10. Structure-based drug design (SBDD) approach offers a valuable tool to design potent and target selective drug molecules, thus minimizing the side effects of the therapy. SBDD has been used to develop novel therapeutics for treating several disorders11. In the present study, we have carried out a docking study on some known XO inhibitors to understand their binding mechanism. The study helped us to propose new XO inhibitors. This will further enable researchers to develop potent and target-specific XO inhibitors. The material methods and results are presented here.

Figure 1: 2D-Structure of allopurinol and febuxostat.

Click here to View figure 

Materials and Methods

The study investigates the binding mode of potent XO inhibitors using a docking study. The database of XO inhibitors for docking study was developed considering the biological activity of XO inhibitors available from the public domain. The database was utilized in a systematic docking study, and amino acid residues involved in drug-receptor interactions were analyzed. The details of the material and methods are presented here.

Selection of Compounds for Docking studies

Compounds patented, published, or under clinical trials as potent xanthine oxidase inhibitors from 2011 to 2020 were taken for present research work10,12.Compounds were selected based on the IC50 value in the nanomolar range (Table 1). Two-dimensional (2D) files of molecules were transformed into three-dimensional (3D) format and optimized using the standard protocol in chem 3D software and MOE software.

Table 1: 2D-structure of xanthine oxidase inhibitors selected for docking studies.

Click here to View table

Development of docking model for structure-based virtual screening

The coordinates of bovine milk XO complexed with salicylic acid were retrieved from the protein data bank (PDB entry: 1fiq). XO is a metalloenzyme that contains molybdenum as a cofactor at the catalytic site. Here, molybdenum (VI) plays a crucial role in electron transfer reactions, consequently leading to substrate oxidation. In the present study, we have considered the catalytic site of XO as a target-binding site for inhibitors. Any compound that binds at the XO’s catalytic site may inhibit the natural substrate from interacting with the molybdenum cofactor and thus suppresses the XO activity [15]. Chemdraw software was used to draw ligands, and energy minimization of the ligand was done in the MOPAC module, using the AM1 method for closed-shell system, available in the CS Chem3D Ultra. The ligands were drawn in ChemDraw and subjected to energy minimization in the MOPAC module, using the AM1 procedure for closed-shell systems, implemented in the CS Chem3D Ultra.Using GOLD 5.3.0, ligands were docked into the catalytic site of xanthine oxidase15. Genetic algorithm-based ligand docking is enacted by Gold to enhance the contour of the ligand at the receptor-binding site. It employs the GoldScore fitness function to appraise the several conformations of ligand at the binding site. It consists of four components: protein-ligand hydrogen bond energy, protein-ligand van der Waals (vdw) energy, ligand internal vdw energy, and ligand torsional strain energy measured by the Goldscore fitness function 13. Docking was replicated ten times for each isomer, and the Goldscore fitness function was used to rank each pose. The highest scored conformations were carefully chosen for discussion.

Docking validation

TheDocking procedure was confirmed by reproducing the crystal structure conformation of the co-crystallized ligand using Pymol version 1.3. The validation was carried out by re-docking and calculating the RMSD value between the docked ligand and the reference ligand to evaluate the performance of the docking protocol. Protein-ligand docking is an instrument to envisage binding poses and evaluate the binding affinity.

Docking of selected compounds

 Docking studies for molecules with activity in a nanomolar range from patented and published molecules from 2011 to 2020 were carried out (Table 1). A Docking study was performed using GOLD 5.3.0 (Cambridge Crystallographic Data Center, Cambridge, UK).Based on binding affinity, Goldscore is used as a scoring function. The binding site was defined based on salicylic acid as a reference ligand. The cavity was set within 6 Å of the bound molecule affinity. Docking was replicated ten times for each isomer, and the Goldscore fitness function was used to rank each pose. Interactions with amino acids were checked using GOLD software images were prepared. Based on the interactions with amino acid residues Phe914, Phe1009, Arg880, Thr1010, docking results of hit compounds were assessed visually to select the best interaction pose.

Design of Derivatives and their docking study

Based on the docking score of compounds 8 and 23, two series of compounds A and B, were prepared for further docking study. Hydrophobic and hydrophilic groups were used to prepare derivatives. Docking was performed for the derivatives (Table 2 and Table 3).

Scheme 1

Click here to View scheme

Table 2: Structure of derivatives of scaffold 8 and their Goldscore.

S.No.

Compound

A

B

C

D

E

Goldscore

1

8a

-COOH

-CH3

H

H

-CH3

57.06

2

8b

-COOH

-CH3

H

H

-OH

60.20

3

8c

-COOH

-CH3

H

H

-NH2

59.56

4

8d

-COOH

-CH3

H

H

-Cl

59.22

5

8e

-COOH

-CH3

H

H

-F

57.02

6

8f

-COOH

-CH3

H

H

-NO2

73.45

7

8g

-COOH

-CH3

H

H

-NHCH3

56.61

8

8h

-COOH

-CH3

H

H

-OCH3

58.02

9

8i

-COOH

-CH3

H

H

-COCH3

59.09

10

8j

-COOH

-CH3

H

H

-COOH

66.86

11

8k

-COOH

-CH3

H

H

-OCOCH3

56.47

12

8l

-COOH

-CH3

H

H

-C2H5

56.20

13

8m

-COOH

-CH3

-CH3

H

H

61.25

14

8n

-COOH

-CH3

-OH

H

H

59.94

15

8o

-COOH

-CH3

-NH2

H

H

61.09

16

8p

-COOH

-CH3

-Cl

H

H

60.16

17

8q

-COOH

-CH3

-F

H

H

57.03

18

8r

-COOH

-CH3

-NO2

H

H

56.65

19

8s

-COOH

-CH3

-NHCH3

H

H

60.74

20

8t

-COOH

-CH3

-OCH3

H

H

59.35

21

8u

-COOH

-CH3

-COCH3

H

H

62.33

22

8v

-COOH

-CH3

-COOH

H

H

60.13

23

8w

-COOH

-CH3

-OCOCH3

H

H

66.64

24

8x

-COOH

-CH3

-C2H5

H

H

67.35

25

8y

-COOH

-CH3

H

-CH3

H

58.20

26

8z

-COOH

-CH3

H

-OH

H

59.54

27

8aa

-COOH

-CH3

H

-NH2

H

60.32

28

8ab

-COOH

-CH3

H

-Cl

H

58.20

29

8ac

-COOH

-CH3

H

-F

H

57.72

30

8ad

-COOH

-CH3

H

-NO2

H

49.43

31

8ae

-COOH

-CH3

H

-NHCH3

H

57.24

32

8af

-COOH

-CH3

H

-OCH3

H

61.59

33

8ag

-COOH

-CH3

H

-COCH3

H

61.29

34

8ah

-COOH

-CH3

H

-COOH

H

59.95

35

8ai

-COOH

-CH3

H

-OCOCH3

H

63.84

36

8aj

-COOH

-CH3

H

-C2H5

H

60.96

37

8ak

-COOH

-CH3

-OCH3

H

-NO2

76.11

38

8al

-COOH

-CH3

-OC2H5

H

-NO2

76.01

39

8am

-COOH

-CH3

c-propyl methyl

H

-NO2

68.92

40

8an

-COOH

-CH3

isopropyl

H

-NO2

65.44

41

8ao

-COOH

-CH3

isobutyl

H

-NO2

62.86

42

8ap

-COOH

-CH3

neopentyl

H

-NO2

62.82

43

8aq

-COOH

-CH3

-CH3

H

-NO2

69.66

44

8ar

-COOH

-CH3

-C2H5

H

-NO2

69.00

45

8as

-COOH

-CH3

C3H7

H

-NO2

71.91

46

8at

-COOH

-CH3

-CH2Cl

H

-NO2

69.90

47

8au

-COOH

-CH3

-CCl3

H

-NO2

63.61

Scheme 2

Click here to View scheme 

Table 3: Structure of derivatives of scaffold 23 and their Goldscore.

S. no.

Compound

A

B

C

D

E

Goldscore

1

23a

-OH

-H

-H

-H

-H

69.73

2

23b

-F

-H

-H

-H

-H

68.49

3

23c

-Cl

-H

-H

-H

-H

68.30

4

23d

-H

-OH

-H

-H

-H

69.30

5

23e

-H

-NH2

-H

-H

-H

68.59

6

23f

-H

a*

-H

-H

-H

60.57

7

23g

-H

-H

CH2OH

-H

-H

69.58

8

23h

-H

-H

-NH2

-H

-H

71.50

9

23i

-H

-H

-SH

-H

-H

72.39

10

23j

-H

-H

-H

-CF3

-H

69.41

11

23k

-H

-H

-H

-CH3

-H

68.10

12

23l

-H

-H

-H

-CCl3

-H

72.74

13

23m

-H

-H

-H

-OCH3

-H

71.48

14

23n

-H

-H

-H

-C2H5

-H

70.85

15

23o

-H

-H

-H

-H

-CF3

70.75

16

23p

-H

-H

-H

-H

-CH3

70.07

17

23q

-H

-H

-H

-H

-CH2F

70.10

18

23r

-H

-H

-H

-H

-CCl3

73.97

19

23s

-H

-H

-H

-H

isopropyl

74.01

20

23t

-H

-H

-H

-H

-C2H5

71.31

21

23u

-H

a**

-H

-H

-H

69.72

a*– C replaced with N in the ring without double bond, a**– C replaced with N in the ring with double bond.

Result and Discussion

Docking validation

Docking validation was done using Pymol 1.3 by giving the function “align docked ligand, reference ligand”. The RMSD was obtained as 0.00 Å as shown in figure 2.

Figure 2: Overlay of co-crystallized and docked conformation of salicylic acid. (Blue: co-crystallized conformation; Green: docked conformation).

Click here to View figure 

Molecular Docking

The docking score of the compounds, along with the docked conformation and interactions with the amino acids, are shown in Table 4. Interaction Diagrams of the suitable docking compounds are shown in figure 3. It was observed that most of the compounds had shown interactions with the Phe914 and Arg880, which play an essential role in xanthine oxidase inhibition.

Table 4: Docking score of high-ranked xanthine inhibitors (Table 1) and amino acid residues involved in D-R interactions.

S. no.

Compound

Docked Conformation

Gold score

Amino acid residues involved in D-R interactions

1

4

4

75.84

Phe914, Arg880, Asn768

2

40

3

75.39

Phe914, Arg880

3

37

2

74.81

Phe914, Arg880

4

1

9

74.68

Phe914, Arg880, Phe1009

5

3

3

74.58

Phe914, Arg880, Asn768

6

28

3

73.83

Phe914, Arg880, Glu1261

7

38

2

73.46

Phe914, Arg880, Thr1010

8

39

2

72.91

Phe914, Arg880

9

12

2

72.81

Phe914, Arg880, Thr1010, Glu802

10

27

1

72.6

Phe914, Arg880, Glu1261

11

25

2

72.5

Phe914, Arg880, Glu1261

12

24

2

72.32

Phe914, Arg880, Thr1010

13

26

2

70.64

Phe914, Arg880, Thr1010

14

21

10

70.53

Phe914, Arg880, Thr1010

15

19

3

70.51

Phe914

16

15

2

70.12

Phe914, Arg880

17

46

1

70.11

Phe914, Arg880

18

2

1

69.89

Phe914, Arg880

19

14

1

69.84

Phe914, Arg880

20

6

3

69.51

Phe914, Arg880, Glu1261

21

32

4

69.42

Asn768, Arg880

22

14

2

69.39

Phe914, Arg880

23

29

3

69.06

Phe914, Arg880

24

34

1

68.99

Phe914, Arg880, Thr1010, Asn768

25

18

1

68.86

Phe914, Glu1261, Ser876

26

8

2

68.76

Phe914, Arg880, Thr1010

27

23

1

68.63

Phe914, Arg880, Thr1010

28

9

5

67.62

Phe914, Arg880

29

5

4

67.52

Glu1261

 

Design of Derivatives and their docking study

The docking score of the promising derivative compounds, along with the docked conformation and interactions with the amino acids, are shown in Table 5. Interaction Diagrams of the suitable docked compounds are shown in Figures 3a and 3b. It was observed that the docking score of some of the derivatives increased by 5-10% in comparison to the parent compound.

Table 5: Docking score of high-ranked xanthine inhibitors (Table 2 and 3) and amino acid residues involved in D-R interactions.

S. no.

Compound

Docked Conformation

Gold score

Amino acid residues involved in D-R interactions

1

8f

3

73.45

Phe914, Glu802, Ala1072, MTE1333

2

8ak

1

76.11

Phe914, Glu802, Ala1079, Glu1261, MTE1333

3

8al

1

76.01

Phe914, Glu802, Ala1079, MTE1333

4

8as

3

71.91

Phe914, Glu802, Ala1079, Phe911, MTE1333

5

23r

1

73.7

Phe914, Arg880

6

23s

10

74.01

Phe914, Arg880

 

Figure 3a: Docked conformation and binding interactions of compounds 8 (a), 12 (b), 23(c), and 24(d) with amino acid residues.

Click here to View figure

Figure 3b: Docked conformation and binding interactions of compounds 27(a), 28(b), 34 (c) and 37(d) with amino acid residues.

Click here to View figure 

Conclusion

The present study was initiated to learn about docking studies and the interactions with amino acids required for xanthine oxidase inhibition for published and patented molecules from 2011-2020. The docking study of approximately 46 molecules with IC50 in the nanomolar range was performed. The docking studies showed interactions with amino acids such as Phe914, Phe1009, and Arg880. Molecules 8 and 23 can be potent molecules for xanthine oxidase inhibition based on their docking studies and interaction affinity with amino acids. Based on this, the derivatization of these two molecules was performed. It was observed that derivatized molecules i.e. 8f, 8ak, 8al, 8as, 23r and 23s, have shown around 10% increase in the gold score function compared to their parent molecules. The study provides a suitable docking model for researchers to design and develop potent and selective XO inhibitors.

Acknowledgment

The authors thank Dr. Manish Kumar Gupta, HOD, Department of Pharmaceutical chemistry & QA, and Prof. (Dr) Vijay Bhalla, Dean, SGT College of Pharmacy, SGT University, Gurugram-122505, for providing facilities for carrying out current Research work.

Conflict of Interest

The authors declare that there is no conflict of interest.

Funding Sources

There is no funding Source.

References

  1. Stockert, S. L.; Shinde, S. S.; Anderson, R.F.; Hille, R. J. Am. Chem. Soc. 2002,124(49),14554–14555.
  2.  Borges, F.; Fernandes, E.; Roleira, F. Curr. Med. Chem. 2002,9(2),195–217.
  3.  Hille, R. Eur. J. Inorg. Chem. 2006,10,1913–1926.
  4.  Rott, K. T.; Agudelo, C. A. J. Am. Med. Assoc. 2003,289(21),2857–2860.
  5.  Terkeltaub, R. A. N. Engl. J. Med. 2003,349(17),1647–1655.
  6. Stamp, L. K.; Taylor, W. J.; Jones, P. B.; Dockerty, J. L.; Drake, J.; Framptom, C.; Dalbeth, N. Arthritis Rheum. 2012,64(8),2529-2536.
  7. The Pharmaceutical Benefits Scheme. Available from: www.pbs.gov.au/medicine/item/10445R
  8. Choi, H.; Neogi, T.; Stamp, L.; Dalbeth, N.; Terkeltaub, R. Arthritis Rheum. 2018,70(11),1702-1709.
  9. FDA: https://www.fda.gov/drugs/drug-safety-and-availability/fdaadds-boxed-warning-increased-risk-death-gout-medicine-uloric-febuxostat (Accessed on 25-April-2020).
  10. Singh, J. V.; Bedi, P. M. S.; Singh, S.; Sharma, S. Expert Opinion on Therapeutic Patents.2020,30(10),769-780.
  11. Ruyck, J.D.; Brysbaert,G.; Blossey, R.; Lensink, M.F. Adv Appl Bioinform Chem. 2016,28(9),1-11.
  12.  Ojha, R.; Singh, J.; Ojha, A.; Singh, H.; Sharma, S.; Nepali, K. Expert Opinion on Therapeutic Patents.2016,27(3),311-345.
  13. Nepali, K.; Singh, G.; Turan, A.; Agarwal, A.; Sapra, S.; Kumar, R.; Banerjee, U. C.; Verma, P.K.; Satti, N. K.; Gupta, M. K.; Suri, O. P.; Dhar, K. L. Bioorganic & Medicinal Chemistry.2011,19(6),1950–1958.
  14. Enroth, C.; Eger, B. T.; Okamoto, K.; Nishino, T.; Pai, E. F. Biochemistry.2000, 97 (20),10723-10728.

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

About The Author