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

Synthesis, Characterization, Biological Assay of New 5-(Pyridine-2-Yl)-1,3,4-Oxadiazol-2-Amine Derivatives and their Molecular Docking Studies

Prabhakar G 1,2, G V R Sai Madhukar1, 3 and Ramesh Domala 1*

1Department of Chemistry, Mahatma Gandhi University, Nalgonda 508254, Telangana state, India.

2B V Raju Institute of Technology, Narsapur, Medak 502313, Telangana state, India.

3Department of Chemistry, SRR Govt. Arts and Science College, Karimnagar-505001, India.

Corresponding Author E-mail: drdo.ramesh3@gmail.com

DOI : http://dx.doi.org/10.13005/ojc/400314

Article Publishing History
Article Received on : 04 Mar 2024
Article Accepted on : 04 May 2024
Article Published : 06 May 2024
Article Metrics
Article Review Details
Reviewed by: Dr. Noor ud Din Zargar
Second Review by: Dr. Niladry Ghosh
Final Approval by: Dr. Ioana Stanciu
ABSTRACT:

This study introduces a novel class of titled compounds, that are produced through a reaction of picolinohydrazide with various unsymmetrical anhydrides. The confirmation of the successful syntheses is affirmed through a detailed characterization protocol including Proton-NMR, 13C-NMR, FTIR, and Mass spectral studies. The resulting products, 2 as well as 3(a-e), underwent successive evaluations to determine their antibacterial capabilities on S. aureus and E. coli, and antifungal evaluation in relation to Candida albicans. Assessment of antimicrobial activity using the disc diffusion method and reference compounds revealed good efficacy in most of these synthesized compounds, revealing some that had very approving results. Computational docking showed that all new compounds exhibit good interactions. This study highlights the power of 5-(pyridine-2-yl)-1,3,4-oxadiazolo alkanamides against both anti-microbial strains.

KEYWORDS:

Biological Activity; Docking study; 1,3,4-oxadiazol-2-amine derivatives

Download this article as: 

Copy the following to cite this article:

Prabhakar G, Madhukar G. V. R. S, Domala R. Synthesis, Characterization, Biological Assay of New 5-(Pyridine-2-Yl)-1,3,4-Oxadiazol-2-Amine Derivatives and their Molecular Docking Studies. Orient J Chem 2024;40(3).


Copy the following to cite this URL:

Prabhakar G, Madhukar G. V. R. S, Domala R. Synthesis, Characterization, Biological Assay of New 5-(Pyridine-2-Yl)-1,3,4-Oxadiazol-2-Amine Derivatives and their Molecular Docking Studies. Orient J Chem 2024;40(3). Available from: https://bit.ly/3WzuNHY


Introduction

Since from the human evolution, microbial infections have become a challenging one in the medical treatment1, within the last few years, in addition to other complications faced during the treatment of infectious diseases today is antimicrobial resistance that has increased its prevalence almost exponentially. Abusing and overuse of antibiotics have sped up the creation of resistant bacteria strains by becoming ineffective treatments that were previously effective. The phenomenon has highlighted the need to develop innovative approaches for combating microbial diseases 2. The important classes refer to heterocyclic compounds, which have very diverse ring structures containing at least one atom such as nitrogen, sulphur or oxygen and show variety of biological activities3. However, in the process of antimicrobial drug formulation such substances present a promising potential to retard growth and increase the proliferation of various microbes. Scientists are currently investigating heterocyclic structure synthesis and modification in an attempt to improve their antimicrobial properties without adverse side effects.

Pyridine has a hetero aromatic ring with one nitrogen atom4. The analogues of pyridine have shown effective antimicrobial5-8, anticancer9-11, antiviral12, antidiabetic13, anti-inflammatory properties 14-15. Derivatives of oxadiazole are significant organic substances with a variety of uses. In the last ten years, 1,3,4 oxadiazoles, heterocycles with one oxygen and two nitrogens16, have clearly shown promising activity as potential antibacterial17-22, antifungal23-25, antiviral26, anticancer27-30, antidiabetic31-32 drugs. As to combat the challenges, in the present work we extended a new class of analogues using oxadiazoles and pyridine. The microbial assay and molecular binding studies of the prepared analogues were also presented.

Materilas and Methods

Each chemical reagent used in the current synthesis is bought from a commercial source and utilized as envisioned. Newly synthesized amide analogues and their chemical structures were confirmed using spectral techniques such as NMR, IR, and Mass interpretation. The NMR spectra of new samples were obtained under CDCl3 or DMSO-d6 solvent using a Bruker 300 Mz. The chemical shifts are presented in ppm with signals from TFAd: δ. 11.50 ppm for 1H and δ164.2 ppm for 13C NMR were driven further by the coupling constant (J). The infrared spectrum between 4000-500 cm-1 was recorded using the nicolate 380 FTIR spectrophotometer. Mass spectra obtained with a spectrophotometer Shimadzu LCMS 2010. In MEL-TEMP II, the melting points of prepared compounds were assessed. In addition, the obtained values are uncorrected.

General Procedure for 5-(Pyridine-2-yl)-1,3,4-oxadiazol-2-amine (2)

Picoline hydrazide (10 m.mol) (2), Cyanogen bromide (15 mmol) and ethanol were taken in RB flask. Refluxing the contents for a duration of 12 hours. After the stipulated time, the solution is cooled, and it is neutralized using an excess of NaHCO₃ solution. Further purification was done by recrystallization with ethanol, resulting in pure compound (2) in good yields.

Synthesis of Acylated N-(5-(Pyridine-2-yl)-1,3,4-Oxadiazol-2-yl) amines-(3a-e)

This produced compound 2 (1 g) was taken in 5 ml of symmetrical anhydride and refluxed gently in a short-air condenser for about 12 hours. Then the compound was cooled using little amount of water. Finally, purified by recrystallization from ethanol, resulting in the corresponding pure form of titled compounds in quantitative yield. Similarly, the remaining derivatives were prepared.

Table 1: The synthetic compounds Physical data.

 S. No

Compounds

Molecular

Formula

Molecular

Weight

M.P.

(0C)

Yield

(%)

1

2

C7H6N4O

162.15

197-199℃

71%

2

3a

C13H16N4O2

260.29

235-237℃

69%

3

3b

C14H18N4O

274.32

247-249℃

76%

4

3c

C15H20N4O2

288.34

143-145℃

81%

5

3d

C16H22N4O2

302.37

176-178℃

73%

6

3e

C17H24N4O2

316.4

210-212℃

75%

Characterization Data

Compound 2

IR (KBr, v cm-1): 3270.11, 3098.95 (-NH2-stretching), 1646.07 (C=N) 1045.62 (C-O-C, stretching, Oxadiazol ring); 1H-NMR (400M. Hz, δ ppm, dimethyl sulfoxide-d6) : 7.42-7.50 (br, 2H, -NH2, D2O Exchangeable), 7.51-7.52 (dd, 1H, C-H-stretching),7.94-7.99 (m, 2H), 8.67 (dd, Ar-H); 13C-NMR(400 M. Hz, dimethyl sulfoxide-d6) δ121.54, 125.37, 137.90, 143.62, 150.12, 157.62, 164.70; M.S: (m/z) 163.2 [M+1]+; Elemental Analysis: Found % (Calculated %): C, 51.85(51.89); H, 3.73 (3.79); N, 34.55 (34.59); O, 9.87 (9.92).

Compound 3a

IR (KBr, v cm-1): 3131.64 (-NH-stretch), 1H-NMR (400M. Hz, dimethyl sulfoxide-d6) : δ 0.82 (t, methyl-H), 1.32 (m, 6H), 1.61 (t, Methylene-H), 2.42 (br, 1H,-NH) , 7.60 (dd, 1H, Aromatic-H), 8.02 (m, 1H), 8.08 (m), 8.66(dd); M.S: m/z 261.4 [M +1]+;  Elemental Analysis: Found % (Calculated %): C, 59.99 (60.03); H, 6.20 (6.24); N, 21.52 (21.54); O, 12.29 (12.33).

Compound 3b

IR (KBr, v cm-1): 3329.43 (-NH-stretch),1H-NMR (400M. Hz, dimethyl sulfoxide-d6) : δ 00.85-.87 (t, 3H), 1.27 (m, 6H), 01.56 -01.59 (m, 2H), 2.30-2.34 (t, 2H-), 3.5 (br, 1H, -NH-),  7.53  (dd, 1H), 8.01 (m, 1H), 08.04-8.05 (m,1H), 8.71 (dd, 1H, -CH-); M.S: m/z 275.5 [M+1]+; Elemental Analysis: Found % (Calculated %): C, 61.30 (61.35); H, 6.61 (6.67); N, 20.42 (20.44); O, 11.66 (11.69).

Compound 3c

IR (KBr, v cm-1): 3039.40 (-NH-stretch),1H-NMR (400M. Hz, dimethyl sulfoxide-d6) : δ 00.85 (t, Methyl-H), 1.26 (m, 8H), 1.60 ( m, 2H), 2.40 ( t, 2H, -CH2), 07.60 (m, 1H), 8.01 (m, 1H), 8.10 (m, 1H), 8.74 (m, 1H, -CH-), 9.60 (1H, -NH-). M.S: m/z 289.5 [M+1]+ ; Elemental Analysis: Found % (Calculated %): C, 62.48 (62.52); H, 6.99 (7.04); N, 19.43 (19.47); O, 11.10 (11.14).

Compound 3d

IR (KBr, v cm−1): 3059.24 (-NH-stretch), 1727.57 (-C=O);  1642.89 (C=N) 1047.67 (C-O-C, stretching, Oxadiazol ring);  1H-NMR (400M.Hz,dimethyl sulfoxide-d6) : δ0.84 – 0.86 (t, Methyl-H), 1.25-1.28 (m, 10H) 01.58-01.61 (t,2H), 2.43-2.51 (m,2H), 07.60-07.62 (dd, 1H), 08.02 – 8.05 (m, 1H), 08.05-8.12 (m,1H), 8.7-8.76 (dd, 1H) 11.80 (br, 1H, -NH ), 13 C-NMR (400 M. Hz, dimethyl sulfoxide-d6): δ14.11, 22.27, 24.57, 24.65, 28.65, 28.74, 28.89, 31.42, 122.47, 126.23, 138.00, 142.90, 150.32, 158.23, 160,03, 171.03; M.S: (m/z) 303.5, [M+1]+;  Elemental Analysis: Found % (Calculated %) : C, 63.55 (63.59); H, 7.33 (7.36); N, 18.53 (18.57); O, 10.58 (10.63).

Compound 3e

IR (KBr, v cm-1): 3129.56 (-NH-stretch),1H-NMR (400 M. Hz, ppm, dimethyl sulfoxide-d6) : δ 0.86 (t, Methyl-H), 1.26 (m, 12H), 1.50 (m, methylene-H), 2.28 (t, 2H), 3.40 (br, 1H, -NH-). 7.52 ( m, 1H), 08.01-8.6 (m, 2H), 8.70 (m, Ar-H); M.S: m/z 317.6 [M+ 1]+; Elemental Analysis: Found % (Calculated %): C, 64.53 (64.59); H, 7.65 (7.68); N, 17.71 (17.75); O, 10.11 (10.15).

Biological activity

The potential antimicrobial activities of compound 2 and their novel various analogues 3 (a-e) were investigated. Four concentrations (25, 50, 75, and 100 µl) of the Agar well-diffusion method were used to assess the antibacterial activity. The activity index is calculated, and the zone of inhibition is calculated after 18 to 24 hours of incubation at 37°C. Using the well diffusion method, antifungal activity was investigated on Candida albicans at four concentrations (25-100 µl). Fungus spread on the prepared SDA culture plates, and after 48 hours of incubation, The activity index was computed.

Molecular Docking Study

The protein-ligand interactions of the prepared compounds 2, 3(a-e) were studied using Auto Dock Vina 4.233. The calculation of the gradient effectively gives the optimization algorithm a ”sense of direction” from a single evaluation. By using multithreading, The evaluation of the speed and accuracy of Vina during flexible redocking of the 190 receptor-ligand complexes making up the AutoDock 4 training set showed approximately two orders of magnitude improvement in speed and a simultaneous significantly better accuracy of the binding mode prediction.  Targeted proteins’ structures were obtained from the RCSB Protein Data Bank34. Utilizing BIOVIA-202035 and Auto Dock tools36, structural preparation and cleaning were completed. Improvement of ligand structure is made using Gaussian09 software package37.

Results and Discussion

Aiming at developing new drugs with promising antimicrobial activity, we synthesised a new amide derivatives of N-(5-(Pyridine-2-yl)-1,3,4-Oxadiazol-2-yl)amine with good pharmaceutical applications. The synthesized compounds label as 2 and 3(a-e). Different analytical methods such as 1H NMR, 13CNMR, mass spectrometry and FTIR were employed to structurally validate the synthesized compounds.

Scheme 1: Synthesis of compound 2 and 3 (a-e).

Click here to View Figure

The IR absorption values of the compound (2) structure analysis based on IR absorptions of the NH2 primary amine group have a stretching vibration at 3270.11 and 3098.95 cm−1. IR absorptions of the -C=N- (oxadiazol ring) and C-O-C functional groups vibration at 1646.07 and 1045.62 cm−1 respectively. The IR absorption values of the compound (3d) structure analysis based on IR absorptions of the NH-secondary amine group have a stretching vibration at 3059.24 cm−1. IR absorptions for -C=N (oxadiazol ring) and C-O-C functional groups vibration at 1642.89 and 1047.67 cm−1 respectively. 1H NMR Spectroscopy chemical shift values of compound (2) structure values are the aromatic protons were detected between 7.51 to 8.67 ppm, the -NH2 broad peak singlet located at 7.42–7.50 ppm (D2O exchangeable). 1H NMR Spectroscopy chemical shift values of the compound (3d) structure values are the aromatic protons were seen between 7.60 to 8.76 ppm, the -NH- broad peak singlet peak located at 11.80 ppm (D2O exchangeable), the methyl group of the side chain at 0.84-0.87 ppm, the -CH2– group attached to carbonyl at 2.43-2.51 ppm and the remaining twelve protons of methylene protons were observed at 1.26-1.61 ppm. 13C NMR spectroscopy of the compound (2) signals C-2 and C-5 carbons observed at 157.62 (C2-oxadiazol ring), 164.70 (C5– oxadiazol ring) ppm. Compound (3d) signals C-2 and C-5 carbons observed at 158.23 (C2– oxadiazol ring), 160.03 (C5– oxadiazol ring) ppm and signal of carbonyl carbon is seen at 171.03 ppm.

Anti-Microbial Activity Report of Synthesized Compounds

The anti-bacterial assay of new compounds noted as 3(a-e) was investigated with reference to ampicillin against bacterial stains such as S. aureus and E. coli. All the products have established promising results as mentioned in Table-2. The effectiveness of recently synthesized compounds was evaluated against Staphylococcus aureus and E. coli bacteria with standard reference (ampicillin) (Figure 1, Table 2). The compounds 3d, 3c and 3b showed better antibacterialstrengthat a 100 µg/ml concentration on S. aureus, with 17, 14 and 11 mm zones of inhibition. Compounds 3d and 3a showed good antibacterial strengthat 100 µg/ml zones of inhibition10 and 8 mm respectively on E. coli.

The antifungal activity of new compounds studied with Candida albicans using spread plate technique. The analysis of antifungal data (Figure 2, Table 2) revealed that all the synthesized compounds2, 3 (a-e) exhibited the most promising results against the tested fungus Candida albicans. The compound 3c has a zone of inhibition of 13mm, 15mm at 75 and 100µg concentrations. Whereas compound 3d has a zone of inhibition of 12mm and 17mm. compound 3e exhibits a zone of inhibition of 9mm and 17mm, compound 3b has a 12mm and 14mm zone of inhibition at concentrations of 75, 100 µg, which is more effective than the standard compound Fluconazole.

Table 2: Anti-Microbial activity against new compounds 2 & 3 (a-e)

Compounds

Concentration (µg) / Zone of Inhibition (mm)

Anti Bactrial Activity

 

Anti Fungal Activty

Staphylococcus aureus

E. Coli

Candida albicans

25

50

75

100

25

50

75

100

25µg

50µg

75µg

100µg

2

0

0

0

0

0

0

0

7

0

0

9

18

3a

0

0

5

8

0

0

5

8

0

0

10

14

3b

0

0

5

11

0

0

0

0

0

0

12

14

3c

0

6

9

14

0

0

0

0

0

0

13

15

3d

9

11

16

17

0

6

8

10

0

0

12

17

3e

0

0

6

6

0

0

5

5

0

0

9

17

Ampicillin

17

19

23

20

16

18

20

26

Fluconazole

0

0

8

15

 

Figure 1: anti-bacterial activity of compounds 2 & 3 (a-e).

Click here to View Figure

Figure 2: Compound 2 and 3(a-e) anti-fungal efficacy against Candida albicans

Click here to View Figure

Docking Study

The interaction behaviour and the binding energies of the 2 and 3(a-e) derivatives were examined using PDB ID: 5JZX, the family of Mur proteins catalyzes biosynthetic conversions of more than ten formation of the peptidoglycan layer on bacterial cell walls. UDP- N-acetylglucosamine-nolpyruvate reductase (MurB) further has a significant role to bind NADPH in protein. Fungal protein PDB ID: 4M8B Crystal structure of an isatin hydrolase bound to product analogue thioisatinate. The protein-ligand least energy docked poses show the hydrogen bonds that develop between the ligands and the designated proteins. (Shown in Figure 3 & 4 and Table 3 & 4). All compounds showed favourable docking energy within the limit of -6.0 to -8.73 kcal/mol, as denoted as Table-3 and Table-4. Among all the synthesized compounds 3e, 3d and 3c showed good binding energy against (PDB ID: 5JZX, 4M8D). Compounds 3e (-8.73 kcal/mol), 3d (-8.38 kcal/mol), 3c (-8.17 kcal/mol) were demonstrated better binding score.

Table 3: Compounds of 2 and 3 (a–e) binding energies using the PDB ID: 5JZX

S. No

Compounds

Binding Strength (K. Cal mol-1)

   

PDB ID: 5JZX

   

Binding energy

Hydrogen- Bonds

Bonding-Proteins

1

2

-6.31

5

SER130(2), SER70(2), ASN71

2

3a

-7.86

6

SER130, SER70(2), ALA67, GLY69(2)

3

3b

-7.87

5

SER70(2), GLY68, GLY69(2)

4

3c

-8.17

4

GLY69(2), SER70, ALA67

5

3d

-8.38

3

SER130, SER70, GLY68

6

3e

-8.73

4

SER130, PRO128, GLY68, ALA67

7

Amplcillin

-8.79

6

GLY69, PRO128, SER130(3), ALA67

8

Fluconazole

-5.48

3

VAL192, GLY69, ASN71

Table 4: Compounds of 2 and 3 (a–e) binding energies using the PDB ID: 4M8B

S. No

Compounds

Binding Strength (K. Cal mol-1)

   

PDB ID: 4M8B

   

Binding energy

Hydrogen- Bonds

Bonding-Proteins

1

2

-6.0

2

SER122, VALR178,

2

3a

-6.7

5

TYR144(3), LYS119, SER75,

3

3b

-7.0

2

ARG30(2)

4

3c

-7.2

3

ARG30(2), THR6

5

3d

-7.1

3

ARG30(2), THR6

6

3e

-7.2

3

ARG30(2), THR6

 

Figure 3: Indicating the binding poses and interactions of compound 2 and 3(a-e), internal standard to binding sites of target proteins.

Click here to View Figure

Figure 4: Indicating the binding poses and interactions of compound 2 and 3(a-e), to binding sites of target proteins.

Click here to View Figure

Conclusion

In conclusion, our successful and effective method has resulted in design of new analogues ranging from 3a–e, which were established by various spectroscopic analyses. The synthesized compounds were then subjected to assessment of their antimicrobial properties including antibacterial and anti-fungal activities. The results of newly synthesised derivatives were found promising. Notably, compounds 3d, 3c, and 3e were found with good antibacterial activity against S. auras and E. coli. All the new derivatives have shown excellent anti-fungal properties among which compound 3d, 3c and 3e have a zone greater than the reference compound against the Candida albicans fungal strain. Finally, this research highlights the antimicrobial potential of newly synthesized compounds, which may act as an option to fight with anti-microbial resistant. Among all the synthesized compounds 3e, 3d and 3c showed good binding energy against PDB ID 5JZX. As well as PDB ID 4M8B. Compounds 3c, 3d, and 3e demonstrated better binding score.

Acknowledgments

The Honourable Vice-Chancellor of M. G. University-Nalgonda and the Chairman B V Raju Institute of technology is acknowledged by the authors for providing facilities and continuing support.

Conflict of Interest

There is no conflict of interest.

References

  1. Annunziato, International journal of molecular sciences, 2019, 20(23), 5844.
    CrossRef
  2. Sharma A, Thakur N, Thakur A, Chauhan A, Babrah H. Cureus. 2023, 15(7): e42231.
    CrossRef
  3. Sharma, P. K., Amin, A., & Kumar, M. The Open Medicinal Chemistry Journal, 2020, 14(1), 49-64.
    CrossRef
  4. De S, Kumar S K A, Shah SK, Kazi S, Sarkar N, Banerjee S, Dey S., RSC Adv., 2022, 12, 15385-15406
    CrossRef
  5. Maghari, S., Ramezanpour, S., Darvish, F., Balalaie, S., Rominger, F., & Bijanzadeh, H. R. Tetrahedron, 2013, 69(8), 2075-2080.
    CrossRef
  6. Nadia Youssef Megally Abdo and Mona Monir Kamel, Chemical and pharmaceutical bulletin, 2015, 63(5), 369-376
    CrossRef
  7. Prabhu, Girish & Sureshbabu, Vommina. Tetrahedron Letters. 2012, 53, 4232–4234.
    CrossRef
  8. Niu, P., Kang, J., Tian, X., Song, L., Liu, H., Wu, J., Yu, W. and Chang, J. The Journal of organic chemistry, 2015, 80(2), 1018-1024
    CrossRef
  9. Wanale, Shivraj & Pachling, S.P., Poshatti, Shankar. Journal of Chemical and Pharmaceutical Research. 2012, 4, 2458-2462.
  10. Kutlu, E., Emen, F. M., Kismali, G., Kınaytürk, N. K., Kılıç, D., Karacolak, A. I., & Demirdogen, R. E. Journal of Molecular Structure, 2021, 1234, 130191.
    CrossRef
  11. Adimule, V., Yallur, B. C., Batakurki, S. Recent Developments in the Synthesis and Applications of Pyridines, Elsevier. 2023, 127-143.
    CrossRef
  12. Alizadeh SR, Ebrahimzadeh MA. Mini Reviews in Medicinal Chemistry, 2021, 21(17), 2584-2611.
    CrossRef
  13. Allaka, Tejeswara Rao, and Naresh Kumar Katari. Recent Developments in the Synthesis and Applications of Pyridines 2023, 605-625.
    CrossRef
  14. Ghattas, Abd-El-Badih AG, Ahmed Khodairy, Hassan M. Moustafa, Bahgat RM Hussein, Marwa M. Farghaly, and Moustafa O. Aboelez. Pharmaceutical Chemistry Journal, 2017, 51, 652-660.
    CrossRef
  15. Hajhashemi, Valiollah, Hoda Mojiri-Froshani, Lotfollah Saghaei, and Afshin Fassihi. Advanced Biomedical Research, 2014, 3(1), 134
    CrossRef
  16. Somani, Rakesh R., and Prabhakar Y. Shirodkar. “Oxadiazole: A biologically important heterocycle.” ChemInform, 2011, 42(10), 130-140
    CrossRef
  17. Bitla, S., Sagurthi, S. R., Dhanavath, R., Puchakayala, M. R., Birudaraju, S., Gayatri, A. A., & Atcha, K. R. Journal of Molecular Structure, 2020, 1220, 128705.
    CrossRef
  18. Gómez‐Saiz, P., García‐Tojal, J., Maestro, M.A., Mahía, J., Arnaiz, F.J., Lezama, L. and Rojo, T. European Journal of Inorganic Chemistry2003, 14, 2639-2650.
    CrossRef
  19. Vaishnav, y., Rajpurohit, s., Vyas, k. k. Oriental Journal of Chemistry, 2023, 39(4).
    CrossRef
  20. Long, B., Tian, B., Tang, Q., Hu, X., Han, L., Wang, Z., Gan, Z. Tetrahedron, 2021, 96, 132382.
    CrossRef
  21. Parameshwar, A., Selvam, V., Ghashang, M., Guhanathan, S. Oriental Journal of Chemistry, 2017, 33(3), 1502-12.
    CrossRef
  22. Saoud, S. A., Ali, K. F., & Shakir, R. M. Oriental Journal of Chemistry, 2017, 33(4), 1781.
    CrossRef
  23. Bitla, S., Sagurthi, S. R., Dhanavath, R., Puchakayala, M. R., Birudaraju, S., Gayatri, A. A.,  Atcha, K. R. Journal of Molecular Structure, 2020, 1220, 128705.
    CrossRef
  24. Prakash, O., Kumar, M., Kumar, R., Sharma, C., Aneja, K. R. European journal of medicinal chemistry, 2010, 45(9), 4252-4257.
    CrossRef
  25. Vaidya, A., Jain, S., Jain, P., Jain, P., Tiwari, N., Jain, R., K Agrawal, R. Mini Reviews in Medicinal Chemistry, 2016, 16(10), 825-845.
    CrossRef
  26. Saha, R., Tanwar, O., Marella, A., Mumtaz Alam, M., Akhter, M. Mini reviews in medicinal chemistry, 2013, 13(7), 1027-1046.
    CrossRef
  27. Nayak, S., Gaonkar, S. L., Musad, E. A., Dawsar, A. M. A. Journal of Saudi Chemical Society, 2021, 25(8), 101284.
    CrossRef
  28. Ladani, G. G., Patel, M. P. New J. Chem., 2015, 39, 9848–9857.
    CrossRef
  29. Kumar, Davinder, Navidha Aggarwal, Harsh Kumar, Garima Kapoor, Aakash Deep, Shabana Bibi, Aastha Sharma et al. Pharmaceuticals, 2023, 16, 6805.
    CrossRef
  30. Acar Çevik, U., Celik, I., Işık, A., Ahmad, I., Patel, H., Özkay, Y., Kaplancıklı, Z. A. Journal of Biomolecular Structure and Dynamics, 2023, 41(5), 1944-1958.
    CrossRef
  31. El Hassani, I. A., Brandán, S. A., Mortada, S., Arshad, S., Romano, E., Ramli, Y., Karrouchi, K. Journal of Molecular Structure, 2024, 1295, 136620.
    CrossRef
  32. Gilani, s. J., zaman, m. T., nagarajan, k., goel, r., & arora, a. International Bulletin of Drug Research, 2012, 2(1), 132-146.
    CrossRef
  33. El-Hachem, Nehme, Benjamin Haibe-Kains, Athar Khalil, Firas H. Kobeissy, and Georges Nemer. Neuroproteomics: Methods and Protocols, 2017, 391-403.
    CrossRef
  34. Rose, Peter W., Bojan Beran, Chunxiao Bi, Wolfgang F. Bluhm, Dimitris Dimitropoulos, David S. Goodsell, Andreas Prlić et al. Nucleic acids research, 2010, 39(suppl_1), D392-D401.
    CrossRef
  35. Afriza, Dhona, Fauzia Nilam Orienty, and Windi Putri Ayu. Journal of International Dental and Medical Research 2020, 13(4) 1286-1292.
  36. Md, K., Domala, R., Asian Journal of Chemistry, 2024, 36(3), 628–634.
    CrossRef
  37. Çapan, İ., Servi, S., Yıldırım, İ., Sert, Y. Chemistry Select, 2021, 6(23), 5838-5846.
    CrossRef

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

About The Author