Impact Factor: 5.6     h-index: 27

Document Type : Original Article

Authors

Department of Chemistry, Collage of Science, University of Baghdad, Baghdad, Iraq

Abstract

In this research study, the synthesis sequence of novel pyridazine and 1,2,4-Triazine derivatives was determined by reacting α-hydrazino-N-Carbazole acetamide (1) with acid anhydride derivatives used glacial acetic acid as a solvent to prepare compounds (2-9) pyridazine derivatives reacted compound (1) with phenyl isocyanate, phenyl thioisocyanate, and α-naphthyl isocyanate to use absolute ethanol as solvent to give compounds (10-12). Compounds (10-12) were condensed with (2N. NaOH) to give compounds (14-16) to denominate 1,2,4-Triazine derivatives. Reacted compound (1) with CS2/KOH used absolute ethanol as solvent to provide potassium salt (13). This salt reacts with 95% hydrazine hydrate to give compound (17). The newly synthesized compounds were tested against different microorganisms to evaluate their antimicrobial activities on bacterial strains, gram-positive bacteria, gram-negative bacteria, and fungal strains to identify the most efficient biologically active compounds.

Graphical Abstract

Synthesis and Study Impaction Antibacterial, Antifungal Activity Newly Pyridazine and 1,2,4-Triazine Derivatives

Keywords

Main Subjects

Introduction

Pyridazine is a heterocyclic organic compound with the molecular formula (CH)4N2 [1]. It contains a six-membered ring with two adjacent nitrogen atoms and is aromatic [2]. It is isomeric with two other (CH)4N2 rings, pyrimidine, and pyrazine. As one of the most critical N-heterocycles, diazine derivatives have attracted much attention nowadays due to their numerous advantages, such as ease of structural modification, [3-5] excellent optical [6,7], and electronic properties [8], and productive biological activities [9,10]. Among the three isomeric forms, the pyridazine units constitute critical components of biologically active structures [11]. However, their utility in creating optoelectronic functional materials has only recently received the attention of scientists in organic chemistry. Material community [12,13] can be classified into three different types according to the relative position of the nitrogen atoms: pyridazine (1,2-diazine), pyrimidine (1,3-diazine), and pyrazine (1,4-diazine) [11]. All the three isomers exhibit highly π-deficient character and thus can be used as the electron-withdrawing part in π-conjugated push−pull structures. In this way, diazine-based conjugated molecules with favorable luminescence induced by significant intramolecular charge transfer (ICT) estimate their potential applicability in OLED [14,15].

1,2,4-Triazine and its derivatives are an important class of heterocyclic compounds. They thus have been reported in a wide spectrum of biological activities [16], including antithrombotic [17], potential A-Glucosidase Inhibitors [16], anticancer [18], antiplatelet, thromboxane synthetase inhibition, 3 anti-inflammatories [19], antibacterial and antiviral activities [20]. 1,2,4-Triazine is one of the possible three isomers of the six-membered ring bearing three nitrogens. In the past, this system was also called as-triazines (asymmetric triazines) [18].

Materials and Methods

All starting chemical compounds were purchased from Fluka or Aldrich. Melting points (M.P) were determined using Gallenkamp and a Thomas capillary melting point apparatus in open glass capillaries. Uncorrected FTIR spectra of the KBr disc were acquired using a SHIMAZU FTIR-8400 Fourier transform infrared spectrophotometer. The total primary components and reagents were purified and readily available commercially. A 500 MHz spectrometer was used to acquire 1H-NMR and 13C-NMR spectra. Agilent Technologies model ultrashield nuclear magnetic resonance (NMR) spectra were recorded in dimethyl sulfoxide solvent (DMSO-d6), and the chemical shifts are given in (ppm) downfield using Tetramethylsilane (TMS) as a reference. Used to test against different microorganisms to evaluate their antimicrobial activities four bacterial strains, gram-positive bacteria (Bacillus cereus and Staphylococcus aureus), gram-negative bacteria (Escherichia coli and pseudomonas aeiuginosa), and two fungal strains (Rhizopuas Microrhizosporium and Candida albicans).

General preparation of pyridazine derivatives (2-9) [21]

To a 50 mL round bottom flask, add 0.002 mol hydrazide derivative (1) and 0.002 mol anhydride and refluxing for 6-8 hours. The solid product obtained (2-9) was cooled with crushed ice, filtered, and recrystallized using ethanol and dioxane.

Table 1 summarizes the physical properties and FTIR spectral data of the preparation of compounds (2-9).

Preparation of 2-(2-(9H-carbazol-9-yl)-2-oxoethyl)-N-phenyl hydrazine-1-carboxamide (10), 2-(2-(9H-carbazol-9-yl)-2-oxoethyl)-N-(naphthalen-1-yl) hydrazine-1-carboxamide (11), 2-(2-(9H-carbazol-9-yl)-2-oxoethyl)-N-phenyl hydrazine-1-carbothioamide (12) [22]

0.5 g of compound (1), 0.002 mol phenyl isocyanate, 0.002 mol of α-naphthyl isocyanate, and 0.02 mol phenylthioisocyanate in 10 mL absolute ethanol were mixed in a round bottom flask in reflux condition (5-6 h). It was filtered and dried before being recrystallized from acetone after cooling. The precipitate's physical properties and FTIR spectral data of the preparation of compounds are listed in Table 2.

Preparation of potassium 2-(2-(9H-carbazol-9-yl)-2-oxoethyl) hydrazine-1-carbodithioate (13) [23]

The mixture of 0.002 mol hydrazide derivative (1) in 0.002 mol ethanolic solution KOH dissolved in 12 mL absolute ethanol, and 0.002 mol of CS2 (lingeringly added to the solution). After filtration, the organic product washed and dry by ether. In the next step, salt (13), was used without further purification. The physical properties and FTIR spectral data of the compounds prepared are listed in Table 2.

Preparation of 5-(9H-carbazol-9-yl)-4-phenyl-1,4-dihydro-1, 2,4-triazin-3-ol (14); 5-(9H-carbazol-9-yl)-4-(naphthalen-1-yl)-1,4-dihydro-1,2,4-triazin-3-ol (15); 5-(9H-carbazol-9-yl)-4-phenyl-1,4-dihydro-1,2,4-triazine-3-thiol {41} [23]

In the one step, compounds (10-12) (0.0011 mol) were refluxed in 2N sodium hydroxide solution (10 mL) for 10-12 h. After cooling, the mixture and neutralized with (1:1) hydrochloric acid. The solid product was filtered and then recrystallized from ethanol. Physical properties of these compounds (14-16) are listed in Table 3.

Synthesis of 4-amino-5-(9H-carbazol-9-yl)-1,4-dihydro-1,2, 4-triazine-3-thiol (17) [23]

In round-bottomed flask 0.001 mol of potassium salt (13) were refluxed with 5 mL excess of 95% hydrazine hydrate until layover of the transmutation of hydrogen sulphide. During reflux, the color of the reaction mixture changed, resulting in a homogenous mixture, cooling the product obtained and acidified with hydrochloric acid 10% to yield pale yellow precipitate. It was purified via recrystallization from ethanol to get crystals. Physical properties and FTIR spectral data of the preparation of compound (17) are listed in Table 3.

Determination of Antibacterial Activity Susceptibility test [24]

Some synthesized compounds (2,5,6,7,8,10,12,14,16,17) were tested against four different microorganisms that were evaluated according to the well-diffusion method on bacterial strains: Two gram-positive Bacteria (Bacillus cereus and Staphylococcus aureus) and two gram-negative bacteria (Escherichia coli and pseudomonas aeiuginosa). The samples were dissolved in DMSO; the good diameter (5 mm) was cultured in a medium Muller Hinton ager at 37 °C for bacteria. The dishes were put in an incubator for 24 h. The disks’ surface was inoculated by 100 μL of both microorganism cultures tested.

Determination of antifungal activity susceptibility test [25]

Synthesized compounds (2,5,6,7,8,10,12,14,16,17) were tested against two fungal (Candida albicans and Rhizopus microrhizosporium). The sample was dissolved in DMSO, then cultured in two fungal medium Potato dextrose agar (PDA) at temperature 37 °C for 3-5 days.

Results and Discussion

The sequence of reactions to prepare novel pyridazine and 1,2,4-triazine derivatives was observed. This new synthesis of the compounds is demonstrated in Scheme 1.

Scheme 1: Synthesis of the compounds

The newly synthesized compounds were established by their melting point, yield, FT-IR, 1HNMR, and 13CNMR. 1,2-diazines derivatives compounds (2-9) were synthesized by the reaction of hydrazide derivative (1) with (succinic anhydride, glutaric anhydride, tetraphenyl phthalic anhydride, 3,3-teramethylene glutaric anhydride, 3-nitro phthalic anhydride, 2,3-dichloromaleic anhydride, phthalic anhydride, 1,2,4,5-benzenetetracarboxylic-1,2,4,5-di anhydride) respectively, in the presence of glacial acetic acid as a catalyst and as solvent.

The FTIR spectra of compounds (2-9) show the disappearance of the two absorbance bands for ʋ(NH2) at 3440 overlap cm-1 Asym., 3315 cm-1 sym. of hydrazide derivative (2) and show the appearance of bands at the range 3395-3360 cm-1 due to ʋ(N-H) group. Two cyclic carbonyl groups of compounds (2-9) appeared at 1751-1701 cm-1 and at 1706-1652 cm-1 for the ʋ(C=O) amide. All details of FTIR spectral data of compounds (2-9) are listed in Table 1.

1H-NMR spectrum of compound (6) showed a singlet signal at δ= 3.46 ppm due to (H2C-C=O) proton, multiplate signal at δ= 7.13-8.20 ppm due to aromatic rings (Ar-H) protons and a singlet signal at δ= 10.25 ppm for (N-N-H) as shown in Table 4.

1H-NMR spectrum of compound (7) showed a singlet signal at δ= 3.47 ppm due to (H2C-C=O) proton, multiplate signal at δ= 7.13-8.16 ppm due to aromatic rings (Ar-H) protons and a singlet signal at δ= 10.20 ppm for (N-N-H) as shown in Table 4 .13C-NMR spectral data of compounds (6-7) are listed in Table 5.

The hydrazide derivative (1) was converted to compounds (10-12) by reacting with phenyl isocyanate, α-naphthyl isocyanate, and phenyl isothiocyanate, respectively absolute ethanol. Compound (1) has been used for the synthesis of compound (13) via the reaction of hydrazide derivative (1) with CS2 in KOH/ethanolic to produce the dithiocarbazate salt (13).

FTIR spectra showed absorption bands at 3336, 3244, 3213, 3320 cm-1 for ʋ(N-H), 1704 overlap, 1683 overlap, 1699, 1670 cm-1 for ʋ(C=O) amide for compounds (10-13), with the appearance of a band at 1326, 1270 cm-1 for ʋ(C=S) of compounds (12,13) and disappearance of ʋ(NH2) for hydrazide derivative (1) at 3440 overlap cm-1 Asym., 3315 cm-1 sym. All details of FTIR spectral data of compounds (10-13) are listed in Table 2.

1H-NMR spectrum of compound (10) showed a singlet signal at δ= 3.40 ppm due to (H2C-C=O) proton, a singlet signal at δ= 4.19 ppm due to (HN-NH), multiplate signal at δ= 7.11-8.26 ppm due to aromatic rings (Ar-H) protons and a singlet signal at δ= 9.53 ppm for (O=C-NH) as shown in Table 6.

1H-NMR spectrum of compound (12) showed a singlet signal at δ= 3.42 ppm due to (H2C-C=O) proton, a singlet signal at δ= 5.66 ppm due to (HN-NH), multiplate signal at δ= 6.99-8.67 ppm due to aromatic rings (Ar-H) protons and a singlet signal at δ= 9.81 ppm for (S=C-NH) as shown in Table 6. 13C-NMR spectral data of compounds (10, 12) are listed in Table 7.

The reaction of compounds (10, 12) with NaOH (2N.) under refluxing condition affected intramolecular cyclization by losing H2O, giving the 1,2,4-triazine derivatives. These products of compounds (14, 16). Compound (13) has been used for the synthesis of compound (17), which was then cyclized via reflux with 95% hydrazine hydrate to produce 1,2,4-triazine derivative (17).

FTIR spectrum of compounds (14-17) showed absorption band at 3342, 3269, 3217, 3249 cm-1 due to ʋ(NH). While compound (17) appears two bands for ʋ(NH2) at (3500) cm-1 Asym., 3485 cm-1 sym. Compounds (14, 15) ʋ(OH) at 3440, 3430 cm-1. Compounds (16, 17) showed an absorption band at 2596, 2592 cm-1 due to ʋ(C=S). Compounds (14-17) absorption band at 1649, 1625, 1623, 1650 cm-1 to(C=N). All details of FTIR spectral data of compounds (14-17) were listed in Table 3.

1H-NMR spectrum of compound (14) showed a singlet signal at δ= 5.45 ppm due to (C=C-H) proton, multiplate signal at δ= 7.14-8.15 ppm due to aromatic rings (Ar-H) protons, a singlet signal at δ= 8.80 ppm for (N-NH) and a singlet signal at δ= 9.57 ppm for (-OH) as shown in Table 8.

1H-NMR spectrum of compound (16) showed a singlet signal at δ= 2.7 ppm for (-SH), a singlet signal at δ= 4.52 ppm due to (C=C-H) proton, multiplate signal at δ= 6.96-8.16 ppm due to aromatic rings (Ar-H) protons and a singlet signal at δ= 9.23 ppm for (N-NH) as shown in Table 8. 1H-NMR spectrum of compound (17) showed a singlet signal at δ=3.50ppm for (-SH), a singlet signal at δ= 5.40 ppm due to (C=C-H) proton, a singlet signal at δ= 5.405.40 ppm due to (C=C-H) proton, a singlet signal at δ= 5.70 ppm due to (NH2) proton, multiplate signal at δ=7.14-8.16 ppm due to aromatic rings (Ar-H) protons, and a singlet signal at δ=9.20 ppm for (N-NH) as shown in Table 8.13C-NMR spectral data of compounds (14, 16 and 17) are listed in Table 9.

Antibacterial activity

Well diffusion method was used to screen the antibacterial activity of some pyridazine and 1,2,4-Triazine derivatives compound against two gram-positive bacteria (Bacillus cereus and Staphylococcus aureus) and two gram-negative bacteria (Escherichia coli and pseudomonas aeiuginosa) by measuring the appeared inhibition zone in mm.

The results of antibacterial activity for the compounds (2, 10, 12, 16, 17) showed the higher antibacterial responses as follows: The starting material (compounds 2, 10, and 12) showed a vital inhibition zone with Escherichia coli (15 mm, 16 mm, and 18 mm, respectively). Compounds (2, 10 and 12) represent high antibacterial activities against Pseudomonas auroginosa (14 mm, 16 mm, and 17 mm). While Compounds (6, 7, 12, 14, and 16) exhibit significant inhibition responses Staphylococcus aureus (18 mm for both compounds 6, 7, 16 mm for both compounds 12, 16, and 15 mm). Only compound (17) shows a noticeable inhibition zone against Bacillus. Ceftrlaxone BP was used as a standard antibacterial drug, as shown in Table (10).

Antifungal activity

The synthesized compounds were tested on two strains of fungal (Rhizopuas Microrhizosporium and Candida albicans).

Compounds (2, 6, 7, 8, 10, 12, and 16) gave significantly higher effective responses with Rhizopuas Microrhizosporium, while compounds (5, 14, and 17) gave a low response. All compounds to give significant effective responses except compound (6) give the same response as the antibiotic, with Candida albicans. Flocazole was used as a standard antifungal drug, and the inhibition zones diameters for the tested compounds are illustrated in Table 10.

Conclusion

In the current study, the synthesis of a series of some novel pyridazine and 1,2,4-triazine derivatives from α-hydrazino-N-carbazole acetamide via reactive intermediates was presented. The microdilution susceptibility test in Muller-Hinton ager based on the well diffusion method was used to determine the antibacterial and antifungal activities of the newly synthesized compounds, which showed significant antimicrobial action.

 

Acknowledgments

I would like to thank all member staff of the department of chemistry, University of Baghdad.

 

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

 

Authors' contributions

All authors contributed toward data analysis, drafting and revising the paper and agreed to be responsible for all the aspects of this work.

Conflict of Interest

We have no conflicts of interest to disclose.

HOW TO CITE THIS ARTICLE

Mohammed Hasan Mohammed AL-Dahlaki, Suaad M.H. Al-Majidi, Synthesis and Study Impaction Antibacterial, Antifungal Activity Newly Pyridazine and 1,2,4-Triazine Derivatives. Chem. Methodol., 2022, 6(4) 269-279

DOI: 10.22034/chemm.2022.320935.1412

URL: http://www.chemmethod.com/article_144076.html

  1. Favre H.A., Powell W.H., RSC, 2013 [Google Scholar], [Publisher]
  2. GÜMÜŞ S., Turkish J. Chem., 2011, 35:803 [Crossref], [Google Scholar], [Publisher]
  3. Mathew T., Papp A.Á., Paknia F., Fustero S., Prakash G.S., Soc. Rev., 2017, 46:3060 [Crossref], [Google Scholar], [Publisher]
  4. Tang R., Zhang F., Fu Y., Xu Q., Wang X., Zhuang X., Wu D., Giannakopoulos A., Beljonne D., Feng X., Lett., 2014, 16:4726 [Crossref], [Google Scholar], [Publisher]
  5. Li J., Gao J., Xiong W.W., Zhang Q., Let., 2014, 55:4346 [Crossref], [Google Scholar], [Publisher]
  6. Itami K., Yamazaki D., Yoshida J.I., Amer. Chem. Soc., 2004, 126:15396 [Crossref], [Google Scholar], [Publisher]
  7. Malval J.P., Achelle S., Bodiou L., Spangenberg A., Gomez L.C., Soppera O., Robin-Le Guen F., Mater. Chem. C, 2014, 2:7869 [Crossref], [Google Scholar], [Publisher]
  8. Yasuda T., Sakai Y., Aramaki S., Yamamoto T., Mater, 2005, 17:6060 [Crossref], [Google Scholar], [Publisher]
  9. Laddha S.S., Bhatnagar S.P., Med. Chem., 2009, 17:6796 [Crossref], [Google Scholar], [Publisher]
  10. Xu L., Russu W.A., Med. Chem., 2013, 21:540 [Crossref], [Google Scholar], [Publisher]
  11. Li M., Yuan Y., Chen Y., ACS Appl. Mater Interfaces, 2018, 10:1237 [Crossref], [Google Scholar], [Publisher]
  12. Gao Z.Q., Mi B.X., Tam H.L., Cheah K.W., Chen C.H., Wong M.S., Lee S.T., Lee C.S., Mater., 2008, 20:774 [Crossref], [Google Scholar], [Publisher]
  13. Mi B.X., Wang P.F., Gao Z.Q., Lee C.S., Lee S.T., Hong H.L., Chen X.M., Wong M.S., Xia P.F., Cheah K.W., Mater., 2009, 21:339 [Crossref], [Google Scholar], [Publisher]
  14. Liu D., Zhang Z., Zhang H., Wang Y., Commun., 2013, 49:10001 [Crossref], [Google Scholar], [Publisher]
  15. Achelle S., Rodríguez-López J., Katan C., Robin-Le Guen F., Phys. Chem. C., 2016, 120:26986 [Crossref], [Google Scholar], [Publisher]
  16. Wang G., Peng Z., Wang J., Li X., Li J., J. Med. Chem., 2017, 125:423 [Crossref], [Google Scholar], [Publisher]
  17. Tamboli R.S., Giridhar R., Gandhi H.P., Kanhed A.M., Mande H.M., Yadav M.R., J. Enzy. Inhib. Med. Chem., 2016, 31:704 [Crossref], [Google Scholar], [Publisher]
  18. Cascioferro S., Parrino B., Spano V., Carbone A., Montalbano A., Barraja P., Diana P., Cirrincione G., J. Med. Chem., 2017, 142:328 [Crossref], [Google Scholar], [Publisher]
  19. Kar S., Ramamoorthy G., Sinha S., Ramanan M., Pola J.K., Golakoti N.R., Nanubolu J.B., Sahoo S.K., Dandamudi R.B., Doble M., New J. Chem., 2019, 43:9012 [Crossref], [Google Scholar], [Publisher]
  20. Tang X., Su S., Chen M., He J., Xia R., Guo T., Chen Y., Zhang C., Wang, J., Xue W., RSC Adv., 2019, 9:6011 [Crossref], [Google Scholar], [Publisher]
  21. Al-Adhami H.J., Al-Majidi S.M., Mathkor, T.H., Research Journal of Pharmacy and Technology 2020, 13:5317 [Crossref], [Google Scholar], [Publisher]
  22. Al-Majidi, S.M., Redhab, A., Zankoi Sulaimani, 2015, 17:33 [Crossref], [Google Scholar], [Publisher]
  23. Al-Khuzaie, M. G.; Al-Majidi, S. M., Iraqi J. Sci., 2014, 55:582 [Google Scholar]
  24. Singh S., Ahmad S., Mehta D., Alam S., Sci. Technol., 2020, 3:40 [Google Scholar]