Skip to main content

Synthesis and anticancer evaluation of some novel pyrimido[5,4-e][1,2,4]triazines and pyrazolo[3,4-d]pyrimidine using DMF-DMA as methylating and cyclizing agent

Abstract

Background

Described a series of main target compounds pyrimido[5,4-e][1,2,4]triazines is obtained via condensation of 6-hydrazinyluracil with different aromatic aldehydes to give the hydrazones followed by nitrosation with HNO2 then intramolecular cyclization. On the other hand, pyrazolopyrimidines can be obtained by the reaction of hydrazones with dimethylformamide-dimethylacetal (DMF-DMA), DMF-DMA in the presence of DMF or by refluxing the hydrazinyluracil with DMF-DMA in the presence of DMF directly. The newly synthesized compounds are evaluated in vitro for their anticancer activity against human lung carcinoma (A549).

Results

A newly substituted compounds of benzaldehyde-pyrimidin-4-yl)hydrazones (5af), pyrimido[5,4-e][1,2,4]triazines 6ae, arylethylidenehydrazinylpyrimidine 7a,b and pyrazolopyrimidines 9,11 are screened for cytotoxic activity against human lung carcinoma (A549) cell line. They exhibited a good yield. Compound 6b shows the highest effect with IC50 value 3.6 μM, followed by compounds 9, 5a, 8, 5e, 6e, 5b, 5f, 7a, 5c, 6c, 7b, 6a, 11, 5d and 6d.

Conclusion

A simple and efficient route is used for the synthesis of pyrimido[5,4-e][1,2,4]triazines and pyrazolopyrimidines. The synthesized compounds are screened for antitumor activity.

Background

Triazine is analogues of six membered benzene ring via replacing the three carbon atoms with nitrogens. 1,2,4-triazine and their fused ring structures with one or more heterocycles represent an important class of nitrogen heterocycles compounds. It possess the motif part of naturally and synthetic pharmaceutical products [1,2,3,4,5,6,7,8,9]. They exhibit a broad spectrum biological effects [10] with antibacterial [11, 12], antitumor [13, 14], anticonvulsant [15], anti-inflammatory [16], and antiviral properties [17]. 6-Azacytosine and 6-azauracil are used as effective antiviral and antitumor activities [18,19,20,21]. The tirapazamine (TPZ) is efficacious in the treatment of different human cancer cells via inducing DNA damage in poorly oxygenated tumor cells [22].

The pyrimidotriazine antibiotics represent a wide spectrum of both antimicrobial and antitumor activities [23]. Pyrimido[5,4-e][1,2,4]triazine constitutes the essential active ingredient of the antibiotics like fervenulin (which is formed from actinomyces), xanthothricin, and reumycin [2, 3]. Reumycin [24] is isolated from actinomyces rectus bruneus and used as an antitumor antibiotic for treating brain tumors. Other hetero annelated 1,2,4-triazines have clinical antiviral effect against influenza A and B viruses [4], anti-HIV and anticancer activity [5, 6]. They also show antimicrobial, antifungal effects and cytotoxicity to MCF-7 cells [7, 8]. Fervenulin (planomycin), and its tautomeric isomer toxoflavin (panthothricin) {1.6-dimethylpyrimido[5,4-e][1,2,4]triazine-5,7(1H,6H)-dione} reveal a wide spectrum antibacterial, antifungal, herbicidal and anticancer activities [25,26,27].

Pyrazolopyrimidines constitute the core of many drugs with wide variety of applications in the field of medicine. They are bioactive isomeric purine analogues and have a significant activity as antimetabolites in purine biochemical reactions [28,29,30,31]. They have diverse pharmacological effect as tuberculostatic [32], antimicrobial activities [33], neuroleptic [34], CNS depressant [32], antihypertensive [35] and antileishmanial [36].

In this regard, our strategy is directed towards the synthesis of new pyrimido[5,4-e][1,2,4]triazine and pyrazolopyrimidine compounds because they have found wide applications in pharmaceutical fields. The structure of newly synthesized pyrimidotriazines and pyrazolopyrimidines are proven on the basis of their 1H-NMR, mass spectral data, IR and elemental analysis.

Result and discussion

Chemistry

In continuation to our research, the importance of fervenulin and its diverse pharmacological activity on the medical field, especially as antitumors, we became interested in the prospect of developing our strategies to synthesize new fervenulin analogues of pyrimidotriazine and pyrazolopyrimidine derivatives using 6-hydrazinyl-1-propyluracil (4) as a core for construction. This substrate is prepared via simple hydrolysis of 2,4,6-trichloropyrimidine [37] followed by N-1 selective alkylation using propyl iodide in DMSO in the presence of potassium carbonate as a basic medium [38] then hydrazinolysis of 6-chloro-1-propyluracil (3) with hydrazine hydrate [38,39,40]. Condensation of substrate 4 with different aromatic aldehydes in ethanol at room temperature for 1 h leads to the formation of hydrazones 5af in a good yield (Scheme 1).

Scheme 1
scheme 1

Reaction of 6-hydrazinyluracil with different aromatic aldehydes and formation of pyrimidotriazines. a = NaOH/H2O/Reflux; b = PrI/K2CO3/DMSO; c = NH2NH2.H2O/rt; d = ArCHO/EtOH/rt; e = NaNO2/AcOH/Reflux

The IR spectra of hydrazones displayed the N–H stretching bands at 3271–3122 cm−1. The stretching band of the two C=O groups (Amide I) is displayed within the range 1740–1625 cm−1. Compound 5d showed O–H stretching bands at 3560 cm−1 while the nitro group in compound 5e shows strong asymmetric and symmetric NO2 stretching bands at 1514 and 1337 cm−1, respectively. The 1H-NMR spectra supported the previous observation from the IR spectra, where N3–H and C6–NH is highly deshielded. They appeared around δ 10.75–10.05 ppm, while the α-CH of hydrazone appeared at the range δ 8.48–8.24 ppm. The C5-H was the most shielded as expected around δ 5.46–5.30 ppm. The downfield shift of the α-carbon of hydrazone appeared around δ 145 ppm in the 13C-NMR spectra.

Pyrimidotriazines 6ae is isolated by the nitrosation of hydrazone compounds 5ae at C-5 with in situ prepared nitrous acid. The inseparable 5-nitroso-derivatives undergoes cyclization via the nucleophilic attack of the electron rich α-carbon of the hydrazones on the nitroso group to form hydroxylamine intermediates, which are converted into the target pyrimidotriazines 6ae by protonation of the N-hydroxyl group followed by the elimination of H3O+ (Scheme 2). The IR spectra of 6ae displayed broad absorption bands of NH stretching in the region of 3180–3135 cm−1. The two bands of C=O groups gave rise in the region of 1725–1670 cm−1.

Scheme 2
scheme 2

The plausible reaction mechanism formation of compounds 5af and 6ae

Moreover, the cyclization of the hydrazone series are confirmed in 1H-NMR spectra through the disappearance of both the α-CH hydrazone at δ 8.48–8.24 ppm and the C5-H of uracil at δ 5.46–5.30 ppm.

Another condensation reaction is obtained via condensation of 4 with different acetophenones by stirring at room temperature for 3–4 h (Scheme 3). The IR spectra of 7a,b displayed stretching bands at the range of 3188–3154 cm−1 due to N–H absorption and characteristic bands at the range 1715–1691 cm−1 due to absorption of C=O groups. The mass spectra of these compounds show the expected molecular ions, whereas their 1H-NMR spectra exhibited two signals at δ 11.11–11.06 ppm and at δ 9.13–8.96 ppm ascribed for N3–H and C6–NH protons respectively. The singlet signals of the methyl group protons at the α-carbon appeared at δ 2.42–2.37 ppm while for the CH-5 position appeared at δ 5.48–5.39 ppm. 13C-NMR confirmed the structure of 7a,b where the key signals at δ 78.8–79.8 ppm and δ 14.3–14.2 ppm are assigned to sp2 carbon at position 5 and sp3 carbon attached to the α-carbon respectively.

Scheme 3
scheme 3

Synthesis of pyrazolopyrimidines. a = EtOH/rt; b = DMF-DMA/Reflux/1 h; c = DMF-DMA/DMF/Reflux/15 min; d = DMF-DMA/DMF/Reflux/1 h; e = DMF-DMA/Reflux/12 h

The target compound 9 is prepared by refluxing of 7a with DMF-DMA for 12 h or DMF-DMA in presence of DMF as a solvent for 1 h (Scheme 3). DMF-DMA is a convenient electrophile to introduce one-carbon units. The reaction proceeds via nucleophilic attack of C-5 to electrophilic carbon center of acetal in DMF-DMA followed by intramolecular cyclization and elimination of dimethylamine. A subsequent methylation of NH-5 is observed which arises from O–CH3 group of the acetal not N–CH3 as illustrated in Scheme 4. The plausible mechanism is proved by isolation of the intermediate 8. This intermediate is easily identified in IR, Mass, 1H-NMR and 13C-NMR spectra. A broad stretching absorption band of NH appears at the region of 3136 cm−1 of the intermediate 8 in IR spectra and disappears in the target compound 9. Furthermore, 1H-NMR showed the disappearance of CH-5 proton at δ 5.48–5.39 ppm and the appearance of a singlet signal at δ 8.08 ppm characteristic for CH–N proton, a singlet signal of α-C–CH3 at δ 2.39 ppm and two characteristic N–CH3 group at δ 3.19, 3.05 ppm of compound 8 and disappears in compound 9 due to the elimination of dimethylamine (Scheme 4).

Scheme 4
scheme 4

The plausible reaction mechanism formation of compounds 7a,b and the intermediate 8 and 9

Whereas, the alkylation on N-5 in compound 9 is proven without doubt by the disappearance of a singlet signal of NH-5 proton at δ 10.13 ppm and the appearance of the sp3 singlet signal at δ 3.23 ppm characteristic of CH3 appears in 1H-NMR and a signal at δ 27.6 ppm in 13C-NMR. In addition, the 1H-NMR shows a singlet signal of CH-3 at δ 8.60 ppm and two doublet signals at δ 5.71–5.61 ppm corresponding to the two protons of methylene group which indicates that they are not magnetically equivalent. 13C-NMR shows the appearance of signals at δ 134.2 and 101.7 ppm characteristic for C-3 and methylene carbon atom respectively.

Treatment of 4 with DMF-DMA in the presence of DMF by refluxing for 1 h yielded compound 11 (Scheme 3). IR spectra shows characteristic absorption band at 1751, 1698 cm−1 for C=O groups. 1H-NMR spectrum displays three singlet signals at δ 8.41, 3.88 and 3.19 ppm for CH-3, N(2)–CH3 and N(5)–CH3 respectively. On the other hand, 13C-NMR showed C–N(2) at δ 40.4 ppm and C–N(5) at δ 27.5 ppm which confirms the alkylation of N(5) with DMF-DMA. The plausible mechanism for this reaction is shown in (Scheme 5).

Scheme 5
scheme 5

The plausible reaction mechanism formation of compound 11

Biological investigation

Cytotoxic activity

The in vitro growth inhibitory rates against human lung carcinoma (A549) cell line and effective antitumor doses (as measured by IC50) of the synthesized compounds are investigated in comparison with the well-known anticancer standard drugs toxoflavin and 5-fluorouracil, using crystal violet colorimetric viability assay. Data generated are used to plot dose response curves and presented in Table 1 and Fig. 1. The results reveal that all the tested compounds show high variation in the inhibitory growth rates and activities to the tumor cell line in a concentration dependent manner as shown in (Table 1).

Table 1 The IC50 values represent the compound concentration (μM) required to inhibit A549 tumor cell proliferation by 50%
Fig. 1
figure 1

Growth inhibition curves showing A549 cell line treated with the tested compounds at different concentrations compared with reference drugs 5-flourouracil and toxoflavin

From the results in Fig. 1, it is clear that all the tested compounds are found to be very active at 500 μM against human lung carcinoma (A549) cell line after treatment for 72 h with inhibition ratio values between 60 and 97%. The difference between inhibitory activities of all compounds with different concentrations is statistically significant (p  < 0.001).

The highest activity against human lung carcinoma (A549) cell line is measured for compound 6b with IC50 value 3.6 μM, followed by compounds 9, 5a, 8, 5e, 6e, 5b, 5f, 7a, 5c, 6c, 7b, 6a, 11, 5d and 6d with IC50 values of 26.3, 26.8, 28.4, 49.3, 53.8, 54.7, 60.2, 60.5, 74.3, 81.5, 104.6, 107.1, 123, 238.7, and 379.4 μM, compared with reference drugs 5-fluorouracil (10.5 μM) and toxoflavin (0.7 μM).

Methods

Instruments

All melting points were determined with an electrothermal melting-temperature II apparatus and are uncorrected. Element analyses are performed at the regional center for mycology and biotechnology at Al-Azhar University. The infrared (IR) spectra are recorded using potassium bromide disc technique on Nikolet IR 200 FT IR. Mass spectra are recorded on a DI-50 unit of Shimadzu GC/MS-QP 5050A at the regional center for mycology and biotechnology at Al-Azhar University. 1H-NMR and 13C-NMR spectra are determined on Bruker 400 MHz spectrometer using DMSO-d6 as a solvent, applied nucleic acid research center, Zagazig University, Egypt. All reactions are monitored by TLC using precoated plastic sheets silica gel (Merck 60 F254). Spots are visualized by irradiation with UV light (254 nm). The used solvent system is chloroform: methanol (9:1) and ethyl acetate: toluene (1:1).

Synthesis

6-Chlorouracil (2) was prepared according to the reported method [37].

6-Chloro-1-propyluracil (3) was prepared according to the reported method [38].

6-Hydrazinyl-1-propyluracil (4) [38,39,40].

4-Substituted benzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazones (5af)

A mixture of 6-hydrazinyl-1-propyluracil (4) (2.17 mmol) and appropriate benzaldehydes (2.17 mmol) in ethanol (25 mL) is stirred at room temperature for 1 h. The formed precipitate is collected by filtration, washed with ethanol and crystallized from ethanol.

Benzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazone (5a)

Yield: 83%; m.p. = 218–219 °C; IR (KBr) νmax (cm−1): 3224 (NH), 3045 (CH arom.), 2969, 2908 (CH aliph.), 1739, 1647 (C=O), 1550 (C=N), 1516 (C=C); 1H-NMR (DMSO-d6): 10.69 (s, 1H, NH), 10.38 (s, 1H, NH), 8.39 (s, 1H, CH), 7.73–7.71(d, 2H, J = 7.6 Hz, Harom), 7.45–7.42 (m, 3H, arom.), 5.38 (s, 1H, CH-5), 3.89–3.85 (t, 2H, CH2), 1.60–1.55 (m, 2H, CH2), 0.91–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.4, 152.4, 151.0, 146.5, 134.0, 130.0, 128.9, 126.9, 77.2, 42.2, 21.0, 10.7 ppm; MS: m/z (%) = M+, 272 (83), 243 (61), 216 (36), 153 (36), 145 (31), 144 (25), 110 (27), 106 (58), 104 (100), 103 (22), 90 (38), 89 (33), 77 (52); Anal. calcd. for C14H16N4O2 (272.30): C, 61.75; H, 5.92; N, 20.58. Found: C, 61.86; H, 5.97; N, 20.73.

4-Chlorobenzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazone (5b)

Yield: 85%; m.p. = 238–239 °C; IR (KBr) νmax (cm−1): 3224 (NH), 3056 (CH arom.), 2936 (CH aliph.), 1700, 1630 (C=O), 1594 (C=N), 1558 (C=C), 870 (p-substituted phenyl); 1H-NMR (DMSO-d6): 10.68 (s, 1H, NH), 10.42 (s, 1H, NH), 8.37 (s, 1H, CH), 7.75–7.73 (d, 2H, J = 8.4 Hz, Harom), 7.51–7.49 (d, 2H, J = 8.4 Hz, Harom), 5.39 (s, 1H, CH-5), 3.89–3.85 (t, 2H, CH2), 1.60–1.54 (m, 2H, CH2), 0.90–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.4, 152.4, 151.0, 145.1, 134.4, 133.0, 129.0, 128.5, 77.4, 42.2, 21.0, 10.7 ppm; MS: m/z (%) = M + 2, 308 (31), M+, 306 (100), 279 (30), 277 (96), 252 (22), 250 (64), 179 (31), 178 (27), 154 (12), 153 (52), 152 (39), 142 (23), 140 (82), 139 (33), 138 (86), 136 (63), 127 (27), 125 (17), 124 (25), 113 (22), 111 (48), 110 (37); Anal. calcd. for C14H15ClN4O2 (306.74): C, 54.82; H, 4.93; N, 18.26. Found: C, 55.04; H, 5.01; N, 18.43.

4-Bromobenzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazone (5c)

Yield: 84%; m.p. = 242–243 °C; IR (KBr) νmax (cm−1): 3122 (NH), 3028 (CH arom.), 2971 (CH aliph.), 1740, 1647 (C=O), 1548 (C=N), 1515 (C=C), 869 (p-substituted phenyl); 1H-NMR (DMSO-d6): 10.68 (s, 1H, NH), 10.43 (s, 1H, NH), 8.36 (s, 1H, CH), 7.70–7.63 (m, 4H, arom.), 5.39 (s,1H, CH-5), 3.89–3.85 (t, 2H, CH2),1.60–1.54 (m, 2H, CH2), 0.90–0.84 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.5, 152.4, 151.0, 145.2, 133.3, 131.9, 128.7, 123.2, 77.4, 42.2, 21.0, 10.7 ppm; MS: m/z (%) = M + 2, 353 (6), M+, 351 (12), 350 (77), 323 (23), 321 (68), 296 (61), 294 (100), 186 (50), 183 (48), 182 (47), 181 (44), 168 (30), 157 (36), 154 (20), 253 (27), 152 (56), 144 (41), 140 (33), 110 (31), 102 (23), 89 (47), 76 (41); Anal. calcd. for C14H15BrN4O2 (351.20): C, 47.88; H, 4.3; N, 15.95. Found: C, 48.02; H, 4.28; N, 16.02.

4-Hydroxybenzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazone (5d)

Yield: 78%; m.p. = 213–214 °C; IR (KBr) νmax (cm−1): 3560 (OH), 3271 (NH), 3020 (CH arom.), 2968 (CH aliph.), 1705, 1625 (C=O), 1581 (C=N), 1512 (C=C), 834 (p-substituted phenyl); 1H-NMR (DMSO-d6): 10.58 (s, 1H, NH), 10.15 (s, 1H, NH), 9.90 (s, 1H, OH), 8.28 (s, 1H, CH), 7.56–7.53 (d, 2H, J = 8.4 Hz, Harom), 6.84–6.82 (d, 2H, J = 8.4 Hz, Harom), 5.32 (s, 1H, CH-5), 3.87 - 3.85 (t, 2H, CH2),1.59–1.57 (m, 2H, CH2), 0.90–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.5, 159.4, 152.5, 151.1, 147.0, 128.7, 125.1, 115.8, 76.6, 42.1, 21.0, 10.7 ppm; MS: m/z (%) = M+, 288 (40), 259 (21), 232 (31), 161 (72), 160 (48), 146 (24), 122 (61), 121 (26), 120 (88), 119 (100), 106 (30), 1105 (20); Anal. calcd. for C14H16N4O3 (288.30): C, 58.32; H, 5.59; N, 19.43. Found: C, 58.50; H, 5.67; N, 19.61.

4-Nitrobenzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazone (5e)

Yield: 91%; m.p. = 228–230 °C; IR (KBr) νmax (cm−1): 3150 (NH), 3022 (CH arom.), 2968 (CH aliph.), 1740, 1692 (C=O), 1593 (C=N), 1561 (C=C), 1514 (NO2asymstr), 1337 (NO2symstr), 843 (p-substituted phenyl); 1H-NMR (DMSO-d6): 10.75 (s, 1H, NH), 10.68 (s, 1H, NH), 8.48 (s, 1H, CH), 8.28–8.26 (d, 2H, J = 7.2 Hz, Harom), 7.99–7.97 (d, 2H, J = 7.2 Hz, Harom), 5.46 (s, 1H, CH-5), 3.90–3.87 (t, 2H, CH2), 1.61–1.55 (m, 2H, CH2), 0.91–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.4, 152.3, 151.0, 147.7, 143.7, 140.3, 127.8, 124.1, 78.1, 42.3, 21.1, 10.7 ppm; MS: m/z (%) = M+, 317 (39), 288 (100), 261 (83), 190 (38), 168 (22), 153 (91), 152 (54), 151 (34), 149 (33), 127 (41), 110 (48), 103 (34), 89 (60), 84 (31), 76 (33), 68 (50); Anal. calcd. for C14H15N5O4 (317.30): C, 52.99; H, 4.76; N, 22.07. Found: C, 53.15; H, 4.83; N, 22.24.

4-(Dimethylamino)benzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydro-pyrimidin-4-yl)hydrazone (5f)

Yield: 76%; m.p. = 234–235 °C; IR (KBr) νmax (cm−1): 3220 (NH), 3045 (CH arom.), 2958, 2869 (CH aliph.), 1729, 1693 (C=O), 1593 (C=N), 1519 (C=C), 855 (p-substituted phenyl); 1H-NMR (DMSO-d6): 10.54 (s, 1H, NH), 10.05 (s, 1H, NH), 8.24 (s, 1H, CH), 7.53–7.51 (d, 2H, J = 8.8 Hz, Harom), 6.76–6.74 (d, 2H, J = 8.8 Hz, Harom), 5.30 (s, 1H, CH-5), 3.87–3.83 (t, 2H, CH2), 2.97 (s, 6H, 2 CH3), 1.59–1.54 (m, 2H, CH2), 0.90–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.4, 158.1, 154.0, 151.9, 147.6, 128.2, 121.3, 111.8, 76.3, 42.0, 41.1, 21.0, 10.7 ppm; MS: m/z (%) = M+, 315 (100), 314 (10), 259 (8), 202 (11), 188 (21), 173 (46), 148 (49), 147 (61), 146 (50), 145 (43), 133 (39), 132 (30); Anal. calcd. for C16H21N5O2 (315.37): C, 60.94; H, 6.71; N, 22.21. Found: C, 61.23; H, 6.83; N, 22.37.

3-Aryl-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-diones (6ae)

A solution of 4-substituted benzaldehyde(2,6-dioxo-3-propyl-1,2,3,6-tetrahydropyrimidin-4-yl)hydrazones (5ae) (0.98 mmol) in glacial acetic acid (4 mL) is treated with sodium nitrite (1.16 mmol) by heating under reflux for 3–4 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL); the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:2) to afford compounds 6ae.

3-Phenyl-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-dione (6a)

Yield: 71%; m.p. = 290–291 °C; IR (KBr) νmax (cm−1): 3173 (NH), 3024 (CH arom.), 2968, 2840 (CH aliph.), 1716, 1670 (C=O), 1565 (C=N), 1535 (C=C); 1H-NMR (DMSO-d6): 12.25 (s, 1H, NH), 8.42–8.40 (d, 2H, J = 5.2 Hz, Harom), 7.62–7.61 (m, 3H, arom.), 3.24–3.21 (t, 2H, CH2), 1.68–1.63 (m, 2H, CH2), 0.98–0.94 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 160.4, 154.7, 150.8, 149.4, 146.2, 134.2, 131.3, 129.3, 127.1, 42.4, 20.6, 11.0 ppm; MS: m/z (%) = M+, 283 (16), 255 (20), 254 (38), 213 (17), 171 (13), 105 (100), 104 (13), 103 (10), 77 (29); Anal. calcd. for C14H13N5O2 (283.28): C, 59.36; H, 4.63; N, 24.72. Found: C, 59.58; H, 4.71; N, 24.89.

3-(4-Chlorophenyl)-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-dione (6b)

Yield: 78%; m.p. = 258–260 °C; IR (KBr) νmax (cm−1): 3135 (NH), 3034 (CH arom.), 2972, 2839 (CH aliph.), 1725, 1672 (C=O), 1586 (C=N), 1553 (C=C), 841 (p-substituted phenyl); 1H-NMR (DMSO-d6): 12.32 (s, 1H, NH), 8.42–8.40 (d, 2H, J = 8.4 Hz, Harom), 7.69–7.67 (d, 2H, J = 8.4 Hz, Harom), 4.25–4.21 (t, 2H, CH2), 1.76–1.71 (m, 2H, CH2), 0.98–0.94 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 160.2, 158.1, 150.8, 149.3, 136.2, 134.4, 134.0, 129.3, 128.6, 42.9, 20.3, 11.0 ppm; MS: m/z (%) = M + 2, 319 (6), M+, 317 (17), 290 (24), 289 (26), 288 (64), 249 (14), 247 (40), 141 (31), 139 (100),; Anal. calcd. for C14H12ClN5O2 (317.73): C, 52.92; H, 3.81; N, 22.04. Found: C, 53.14; H, 3.87; N, 22.27.

3-(4-Bromophenyl)-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-dione (6c)

Yield: 76%; m.p. = 250–252 °C; IR (KBr) νmax (cm−1): 3180 (NH), 3084 (CH arom.), 2809 (CH aliph.), 1712, 1675 (C=O), 1579 (C=N), 1545 (C=C) 845 (p-substituted phenyl); 1H-NMR (DMSO-d6): 12.22 (s, 1H, NH), 8.35–8.33 (d, 2H, J = 8.8 Hz, Harom), 7.84–7.82 (d, 2H, J = 8.8 Hz, Harom), 4.25–4.21 (t, 2H, CH2), 1.77–1.71 (m, 2H, CH2), 0.98–0.94 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 160.0, 158.2, 150.8, 149.1, 134.5, 132.3, 131.3, 129.0, 125.2, 42.9, 20.3, 11.0 ppm; MS: m/z (%) = M + 2, 364 (2), M+, 362 (3), 258 (29), 257 (15), 256 (15), 254 (24), 222 (100), 202 (31), 188 (20), 187 (22), 164 (22), 163 (21), 121 (24), 69 (64), 44 (78), 40 (53); Anal. calcd. for C14H12BrN5O2 (362.18): C, 46.43; H, 3.34; N, 19.34. Found: C, 46.71; H, 3.39; N, 19.51.

3-(4-Hydroxyphenyl)-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-dione (6d)

Yield: 82%; m.p. = 236–238 °C; IR (KBr) νmax (cm−1): 3170 (NH), 3026 (CH arom.), 2833 (CH aliph.), 1722, 1677 (C=O), 1558 (C=N), 1537 (C=C), 846 (p-substituted phenyl); 1H-NMR (DMSO-d6): 12.22 (s, 1H, NH), 8.24–8.22 (d, 2H, J = 8.4 Hz, Harom), 6.74–6.72 (d, 2H, J = 8.4 Hz, Harom), 3.79–3.76 (t, 2H, CH2), 1.69–1.63 (m, 2H, CH2), 0.89–0.85 (t, 3H, CH3); MS: m/z (%) = M+, 299 (17), 271 (22), 256 (20), 223 (34), 222 (40), 221 (100), 105 (60), 100 (94), 98 (76), 84 (33), 83 (29), 77 (29), 69 (31), 57 (26); Anal. calcd. for C14H13N5O3 (299.28): C, 56.18; H, 4.38; N, 23.40. Found: C, 56.34; H, 4.47; N, 23.62.

3-(4-Nitrophenyl)-8-propylpyrimido[5,4-e][1,2,4]triazine-5,7(6H,8H)-dione (6c)

Yield: 70%; m.p. = 267–268 °C; IR (KBr) νmax (cm−1): 3165 (NH), 3067 (CH arom.), 2974, 2811 (CH aliph.), 1720,1702 (C=O), 1606 (C = N), 1559 (C=C), 1518 (NO2asymstr),1346 (NO2symstr), 844 (p-substituted phenyl); 1H-NMR (DMSO-d6): 12.31 (s, 1H, NH), 8.66–8.62 (d, 2H, J = 9.2 Hz, Harom), 8.46–8.43 (d, 2H, J = 9.2 Hz, Harom), 4.25–4.21 (t, 2H, CH2), 1.75–1.70 (m, 2H, CH2), 0.98–0.94 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 162.0, 156.9, 151,4, 151.2, 148.9, 140.4, 134.2, 128.2, 124.4, 42.8, 20.4, 11.1 ppm; MS: m/z (%) = M+, 328 (11), 300 (18), 299 (63), 258 (32), 244 (21), 151 (42), 150 (100), 104 (18), 76 (20), 65 (21), 43 (23); Anal. calcd. for C14H13N5O3 (328.28): C, 51.22; H, 3.68; N, 25.60. Found: C, 51.37; H, 3.65; N, 25.81.

6-[2-(1-Arylethylidene)hydrazino]-1-propylpyrimidine-2,4(1H,3H)-diones (7a,b)

A mixture of 6-hydrazinyl-1-propyluracil (4) (2.72 mmol) and appropriate acetophenones (2.72 mmol) in ethanol (30 mL) is stirred at room temperature for 3–4 h. The formed precipitate is collected by filtration, washed with ethanol and crystallized from ethanol.

6-[2-(1-Phenylethylidene)hydrazino]-1-propylpyrimidine-2,4(1H,3H)-dione (7a)

Yield: 83%; m.p. = 207–208 °C; IR (KBr) νmax (cm−1): 3154 (NH), 3052 (CH arom.), 2963, 2870 (CH aliph.), 1715, 1691 (C=O), 1602 (C=N), 1499 (C=C); 1H-NMR (DMSO-d6): 11.06 (s, 1H, NH), 8.96 (s, 1H, NH), 7.90–7.82 (dd, 2H, J = 9.2 Hz, Harom), 7.44–7.43 (m, 3H, arom.), 5.39 (s, 1H, CH-5), 3.92–3.90 (t, 2H, CH2), 2.37 (s, 3H, α-CH3), 1.68–1.63 (m, 2H, CH2), 0.93–0.87 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 166.5, 162.5, 161.1, 151.2, 137.6, 129.6, 128.5, 126.5, 78.8, 43.3, 20.2, 14.3, 11.3 ppm; MS: m/z (%) = M+, 286 (44), 271 (44), 257 (21), 167 (36), 159 (64), 158 (64), 144 (45), 131 (40), 124 (20), 120 (100), 118 (48), 104 (72), 103 (38), 96 (21), 78 (30), 77 (96); Anal. calcd. for C15H18N4O2 (286.32): C, 62.92; H, 6.34; N, 19.57. Found: C, 63.14; H, 6.39; N, 19.71.

6-[2-{1-[4-(Dimethylamino)phenyl]ethylidene}hydrazino]-1-propylpyrimidine-2,4 (1H,3H)-dione (7b)

Yield: 79%; m.p. = 258–260 °C; IR (KBr) νmax (cm−1): 3188 (NH), 3069 (CH arom.), 2961 (CH aliph.), 1704 (C=O), 1593 (C=N), 1504 (C=C), 858 (p-substituted phenyl); 1H-NMR (DMSO-d6): 11.11 (s, 1H, NH), 9.13 (s, 1H, NH), 8.29–8.26 (d, 2H, J = 8.8 Hz, Harom), 8.16–8.13 (d, 1H, J = 8.8 Hz, Harom), 8.10–8.08 (d, 1H, J = 8.8 Hz, Harom), 5.48 (s, 1H, CH-5), 4.02–3.94 (t, 2H, CH2), 3.29 (s, 6H, 2CH3), 2.42 (s, 3H, α-CH3), 1.68–1.64 (m, 2H, CH2), 0.94–0.89 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 166.5, 162.5, 159.7, 151.1, 150.9, 144.0, 127.3, 123.6, 79.8, 43.4, 42.4, 20.2, 14.2, 11.3 ppm; MS: m/z (%) = M+, 329 (2), 302 (100), 289 (8), 274 (21), 203 (34), 149 (21), 124 (12), 117 (55), 96 (21); Anal. calcd. for C17H23N5O2 (329.39): C, 61.99; H, 7.04; N, 21.26. Found: C, 62.12; H, 7.18; N, 21.49.

5-[(Dimethylamino)methylene]-1-propylpyrimidine-2,4,6(1H,3H,5H)-trione 6-{[1-phenylethylidene]hydrazone} (8)

Method A

A solution of 6-[2-(1-phenylethylidene)hydrazino]-1-propylpyrimidine-2,4(1H,3H)-dione (7a) (0.7 mmol) in dimethylformamide-dimethylacetal (4 mL) is heated under reflux for 1 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL); the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 8.

Method B

A solution of 6-[2-(1-phenylethylidene)hydrazino]-1-propylpyrimidine-2,4(1H,3H)-dione (7a) (0.7 mmol) in dimethylformamide-dimethylacetal (1.5 mL) and DMF (1.5 mL) is heated under reflux for 15 min. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL); the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 8.

Yield: method A 90%, method B 94%; m.p. = 220–221 °C; IR (KBr) νmax (cm−1): 3136 (NH), 3030 (CH arom.), 2952, 2866 (CH aliph.), 1690, 1656 (C=O), 1562 (C=N), 1512 (C=C); 1H-NMR (DMSO-d6): 10.13 (s, 1H, NH), 8.08 (s, 1H, CH), 7.73–7.71 (d, 2H, J = 7.6 Hz, Harom), 7.45–7.36 (m, 3H, arom.), 3.97–3.94 (t, 2H, CH2), 3.19 (s, 3H, N-CH3), 3.05 (s, 3H, N-CH3), 2.39 (s, 3H, CH3), 1.69–1.64 (m, 2H, CH2), 0.92–0.88 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 163.8, 159.3, 155.0, 153.6, 151.0, 138.5, 128.6, 128.3, 125.4, 83.0, 46.2, 42.9, 42.7, 20.5, 14.0, 11.3 ppm; MS: m/z (%) = M+, 341 (100), 325 (35), 297 (63), 296 (49), 160 (37), 123 (30), 103 (42), 91 (24), 77 (42), 42 (24); Anal. calcd. for C18H23N5O2 (341.40): C, 63.32; H, 6.79; N, 20.51. Found: C, 62.95; H, 7.34; N, 20.39.

5-Methyl-2-(1-phenylvinyl)-7-propyl-2H-pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione (9)

Method A

A solution of 6-[2-(1-phenylethylidene) hydrazino]-1-propyl-pyrimidine-2,4(1H,3H)-dione (7a) (1.05 mmol) in dimethylformamide-dimethyl acetal (1.5 mL) and DMF (1.5 mL) is heated under reflux for 1 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL), the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 9.

Method B

A mixture of 6-[2-(1-phenylethylidene)hydrazino]-1-propyl- pyrimidine-2,4(1H,3H)-dione (7a) (1.05 mmol) and dimethylformamide-dimethylacetal (3 mL) is heated under reflux for 12 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL), the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 9.

Method C

A mixture of 5-[(dimethylamino)methylene]-1-propylpyrimidine-2,4,6(1H,3H,5H)-trione 6-{[1-phenylethylidene]hydrazone} (8) (1.17 mmol) and DMF-DMA (3 mL) is heated under reflux for 1 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (15 mL), the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 9.

Yield: method A 82%, method B 74%, method C 92%; m.p. = 148–149 °C; IR (KBr) νmax (cm−1): 3095 (CH arom.) 2950, 2875 (CH aliph.), 1760, 1701 (C=O), 1591 (C=N), 1546 (C=C); 1H-NMR (DMSO-d6): 8.60 (s, 1H, CH-3), 7.44–7.36 (m, 5H, arom.), 5.71–5.61 (dd, 2H, =CH2), 3.87–3.83 (t, 2H, CH2), 3.23 (s, 3H, CH3), 1.69–1.64 (m, 2H, CH2), 0.86–0.83 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 157.8, 151.0, 150.3, 144.6, 134.2, 131.8, 129.5, 128.5, 127.3, 109.0, 101.7, 44.7, 27.6, 20.2, 11.0 ppm; MS: m/z (%) = M+, 310 (100), 268 (75), 267 (54), 224 (65), 122 (64), 103 (67),77 (34); Anal. calcd. for C17H18N4O2 (310.35): C, 65.79; H, 5.85; N, 18.05. Found: C, 66.01; H, 5.89; N, 18.24.

2,5-Dimethyl-7-propyl-2H-pyrazolo[3,4-d]pyrimidine-4,6(5H,7H)-dione (11)

A solution of 6-hydrazinyl-1-propyluracil (4) (1.63 mmol) in dimethylformamide-dimethylacetal (1.5 mL) and DMF (1.5 mL) is heated under reflux for 1 h. The reaction mixture is evaporated under reduced pressure. The residue is treated with ethanol (10 mL), the formed precipitate is filtered, washed with ethanol, and crystallized from DMF/ethanol (1:3) to afford compound 11.

Yield: 88%; m.p. = 167–169 °C; IR (KBr) νmax (cm−1): 3094 (CH arom.), 2951, 2877 (CH aliph.), 1751, 1698 (C=O), 1586 (C=N), 1544 (C=C); 1H-NMR (DMSO-d6): 8.41 (s, 1H, CH-3), 3.88 (s, 1H, N(2)-CH3), 3.87–3.83 (t, 2H, CH2), 3.19 (s, 3H, N(5)-CH3), 1.70–1.65 (m, 2H, CH2), 0.90–0.86 (t, 3H, CH3); 13C-NMR (DMSO-d6): δ = 157.9, 151.0, 149.8, 131.8, 100.1, 44.7, 40.4, 27.5, 20.3, 11.0 ppm; MS: m/z (%) = M+, 222 (30), 180 (52), 135 (100), 123 (28), 42 (15); Anal. calcd. for C10H14N4O2 (222.24): C, 54.04; H, 6.35; N, 25.21. Found: C, 54.13; H, 6.43; N, 25.45.

Biological investigation

Evaluation of the antitumor activity

Mammalian cell lines

The cell line that used in this study was human lung carcinoma cell line (A549 cells) is obtained from tissue culture Unit, VACSERA, Cairo, Egypt.

The mammalian cells are propagated in Dulbecco’s modified Eagle’s [41] medium (DMEM) or RPMI-1640 depending on the type of cell line supplemented with 10% heat-inactivated fetal bovine serum, 1% l-glutamine, HEPES buffer and 50 µg/mL gentamycin. All cells are maintained at 37 °C in a humidified atmosphere with 5% CO2 and are subcultured two times a week along experimentation.

i-Antitumor activity evaluation using viability assay

Antitumor activity assay is carried out according to the method described literature [42]. All the experiments concerning the cytotoxicity evaluation are performed and analyzed by tissue culture unit at the regional center for mycology and biotechnology RCMB, Al-Azhar University, Cairo, Egypt.

Procedure

The A549 tumor cells are seeded in 96-well plate in 100 µL of growth medium at a cell concentration of 1 × 104 cells/well. After 24 h of seeding, the monolayers are then washed with sterile phosphate buffered saline (0.01 M pH 7.2) and simultaneously the cells are treated with 100 µL from different dilutions of the test sample in fresh maintenance medium and incubated at 37 °C. Different two-fold dilutions of the tested compound (started from 500 to 0.25 µM) are added to confluent cell monolayers dispensed into 96-well, flat-bottomed microtiter plates (Falcon, NJ, USA) using a multichannel pipette. The microtiter plates are incubated at 37 °C in a humidified incubator with 5% CO2 for a period of 72 h. Untreated cells are served as controls. Three independent experiments are performed each containing six replicates for each concentration of the tested samples. The cytotoxic effects of the tested compounds are then measured using crystal violet staining viability assay. Briefly, after 72 h of treatment, the medium is removed, 100 μL of 0.5% of crystal violet in 50% methanol is added to each well and incubated for 20 min at room temperature and subsequently excess dye is washed out gently by distilled water. The plate is allowed to dry then the viable crystal violet-stained cells are lysed using 33% glacial acetic acid solution. Absorbance at 570 nm is then measured in each well using microplate reader (Sunrise, TECAN, Inc, USA). Toxoflavin and 5-fluorouracil are used as positive control. The absorbance is proportional to the number of surviving cells in the culture plate.

Conclusions

A series of newly synthesized compounds of substituted benzaldehyde-pyrimidin-4-yl)hydrazones (5af), pyrimido[5,4-e][1,2,4]triazines 6ae, arylethylidenehydrazinylpyrimidines 7a,b and pyrazolopyrimidines 9,11 are prepared via a simple method starting from the substrate 6-hydrazinyl-1-propyluracil (4). The synthesized compounds exhibited good cytotoxic activity against human lung carcinoma (A549) cell line and the highest effect is measured for compound 6b with IC50 value 3.6 μM, followed by compounds 9, 5a, 8, 5e, 6e, 5b, 5f, 7a, 5c, 6c, 7b, 6a, 11, 5d and 6d with IC50 values of 26.3, 26.8, 28.4, 49.3, 53.8, 54.7, 60.2, 60.5, 74.3, 81.5, 104.6, 107.1, 123, 238.7, and 379.4 μM, compared with reference drug 5-fluorouracil (10.5 μM).

References

  1. Izmest’ev AN, Gazieva GA, Sigay NV, Serkov SA, Karnoukhova VA, Kachala VV, Shashkov AS, Zanin IE, Kravchenko AN, Makhova NN (2016) An effective one-pot access to polynuclear dispiroheterocyclic structures comprising pyrrolidinyloxindole and imidazothiazolotriazine moieties via a 1,3-dipolar cycloaddition strategy, Beilstein. J Org Chem 12:2240-2249

    Google Scholar 

  2. Ruanpanun P, Laatsch H, Tangchitsomkid N, Lumyong S (2011) Nematicidal activity of fervenulin isolated from a nematicidal actinomycete, Streptomyces sp. CMU-MH021, on Meloidogyne incognita. World J Microbiol Biotechnol 27:1373–1380

    Article  CAS  PubMed  Google Scholar 

  3. Nagamatsu T, Yamasaki H, Hirota T, Yamato M, Kido Y, Shibata M, Yoneda F (1993) Syntheses of 3-substituted 1-methyl-6-phenylpyrimido[5,4-e][1,2,4]triazine-5,7(1H,6H)-diones (6-phenyl analogs of toxoflavin) and their 4-oxides, and evaluation of antimicrobial activity of toxoflavins and their analogs. Chem Pharm Bull 41:362–368

    Article  CAS  PubMed  Google Scholar 

  4. Kiselev OI, Deyeva EG, Melnicova TI, Kozeletskaia KN, Kiselev AS, Rusinov VL, Charushin VN, Chupahin ON (2012) A new antiviral drug triazavirin: results of phase II clinical trial. Vopr Virusol 57:9–12

    CAS  PubMed  Google Scholar 

  5. Abdel-Rahman RM, Seada M, Fawzy M, El-Baz I (1994) Synthesis of some new 1,6-dihydro-3-substituted 6-spiro-(9′-fluorene)-1,2,4-triazin-5-(4H)-ones as potential anti HIV and anticancer drugs. Pharmazie 49:729–733

    CAS  PubMed  Google Scholar 

  6. Abdel-Rahman RM, Seada M, Fawzy M, El-Baz I (1994) Synthesis of some new 1,6-dihydro-3-substituted-6-spiro-(9′-fluorene)-1,2,4-triazin-5-(4H)-on es as potential anti HIV and anticancer drugs. Boll Chim Farm 133:381–388

    CAS  PubMed  Google Scholar 

  7. Abd El-Moneim M, Hasanen JA, El-Deen IM, Abd El-Fattah W (2015) Synthesis of fused 1,2,4-triazines as potential antimicrobial and antitumor agents. Res Chem Intermed 41:3543–3561

    Article  CAS  Google Scholar 

  8. El-Nassan HB (2012) Synthesis, antitumor activity and SAR study of novel [1,2,4]triazino[4,5-a]benzimidazole derivatives. Eur J Med Chem 53:22–27

    Article  CAS  PubMed  Google Scholar 

  9. Abraham S, Hadd MJ, Tran L, Vickers T, Sindac J, Milanov ZV, Holladay MW, Bhagwat SS, Hua H, Ford Pulido JM, Cramer MD, Gitnick D, James J, Dao A, Belli B, Armstrong RC, Treiber DK, Liu G (2011) Novel series of pyrrolotriazine analogs as highly potent pan-Aurora kinase inhibitors. Bioorg Med Chem Lett 21:5296–5300

    Article  CAS  PubMed  Google Scholar 

  10. Crespin L, Biancalana L, Morack T, Blakemore DC, Ley SV (2017) One-pot acid-catalyzed ring opening/cyclization/oxidation of aziridines with N-tosylhydrazones: access to 1,2,4-triazines. Org Lett 19:1084–1087

    Article  CAS  PubMed  Google Scholar 

  11. Sztanke K, Pasternak K, Rajtar B, Sztanke M, Majek M, Polz-Dacewicz M (2007) Identification of antibacterial and antiviral activities of novel fused 1,2,4-triazine esters. Bioorg Med Chem. 15:5480–5486

    Article  CAS  PubMed  Google Scholar 

  12. Culakova H, Dzugasova V, Gbelska Y, Subik J (2013) Antibacterial activity of CTBT (7-chlorotetrazolo[5,1-c]benzo[1,2,4]triazine) generating reactive oxygen species. Microbiol Res 168:147–152

    Article  CAS  PubMed  Google Scholar 

  13. Yurttaş L, Demirayak S, Ilgın S, Atlı O (2014) In vitro antitumor activity evaluation of some 1,2,4-triazine derivatives bearing piperazine amide moiety against breast cancer cells. Bioorg Med Chem 22:6313–6323

    Article  CAS  PubMed  Google Scholar 

  14. Karczmarzyk Z, Wysocki W, Urbaczy-Lipowsa Z, Kalici P, Bielawsa A, Bielawsi K, Ławeca J (2015) Synthetic approaches for sulfur derivatives containing 1,2,4-triazine moiety: their activity for in vitro screening towards two human cancer cell lines. Chem Pharm Bull. 63:531–537

    Article  PubMed  Google Scholar 

  15. Iranneja H, Nadri H, Naderi N, Rezaeian SN, Zafari N, Foroumadi A (2015) Anticonvulsant activity of 1,2,4-triazine derivatives with pyridyl side chain: synthesis, biological, and computational study. Med Chem Res 24(6):2505–2513

    Article  CAS  Google Scholar 

  16. Khoshneviszadeh M, Ghahremani MH, Foroumadi A, Miri R, Firuzi O, Madadkar-Sobhani A, Edraki N, Parsa M, Shafiee A (2013) Design, synthesis and biological evaluation of novel anti-cytokine 1,2,4-triazine derivatives. Bioorg Med Chem 21:6708–6717

    Article  CAS  PubMed  Google Scholar 

  17. Rusinov VL, Egorov IN, Chupakhin ON, Belanov EF, Bormotov NI, Serova OA (2012) Synthesis and antiviral activity of 1,2,4-triazine derivatives. Pharm Chem J 45:655–659

    Article  CAS  Google Scholar 

  18. Sidwell RW, Dixon GJ, Sellers SM, Schabel FM (1968) In vivo antiviral properties of biologically active compounds. Appl Microbiol 16(2):370–392

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Falke D, Rada B (1970) 6-Azauridine as an inhibitor of the synthesis of Herpesvirus hominis. Actavirologica 14(2):115–123

    CAS  Google Scholar 

  20. Creasey WA, Fink ME, Handschumacker RE, Calabresi P (1963) Clinical and pharmacological studies with 2′,3′,5′-triacetyl-6-azauridine. Cancer Res 23:444–453

    CAS  PubMed  Google Scholar 

  21. Walters TR, Aur RJA, Hernandez K, Vietti T, Pinkel D (1972) 6-Azauridine in combination chemotherapy of childhood acute myelocytic leukemia. Cancer 29(4):1057–1060

    Article  CAS  PubMed  Google Scholar 

  22. Sarkar U, Glaser R, Parsons ZD, Barnes CL, Gates KS (2010) Synthesis, crystal structure, and rotational energy profile of 3-cyclopropyl-1,2,4-benzotriazine 1,4-di-N-oxide. J Chem Crystallogr 40(7):624–629

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kumar R, Singh AD, Singh J, Singh H, Roy RK, Chaudhary A (2014) 1,2,3-Triazine scaffold as a potent biologically active moiety: a mini review. Mini Rev Med Chem 14(1):72–83

    Article  CAS  PubMed  Google Scholar 

  24. Bulychev Yu, Yakunlna N, Shorshnev S, Yeslpov S (1996) Poster sessions: 22. Drug design, synthesis, acquisition and structure-activity relationships “synthesis and cytotoxic activity of alkyl and hydroxyalkyl derivatives of reumycin”. Ann Oncol 7:105–113

    Google Scholar 

  25. Ludovic V, Groleau M, Dekimpe V, Deziel E (2007) Burkholderia diversity and versatility: an inventory of the extracellular products. J Microbiol Biotechnol 17(9):1407–1429

    Google Scholar 

  26. Goh KC, Wang H, Yu N, Zhou Y, Zheng Y, Lim Z, Sangthongpitag K, Fang L, Du M, Wang X (2004) PLK1 as a potential drug target in cancer therapy. Drug Dev Res 62:349–361

    Article  CAS  Google Scholar 

  27. Nagamatsu T (2001) Syntheses, transformation, and biological activities of 7-azapteridine antibiotics: toxoflavin, fervenulin, reumycin and their analogs. Recent Res Dev Org Bioorg Chem 4:97–121

    CAS  Google Scholar 

  28. Aly AA, Gad El-Karim IA (2011) facile synthesis of new pyrazolopyrimidine derivatives of potential biosignificant interest. J Korean Chem Soc 55(5):781–786

    Article  CAS  Google Scholar 

  29. Tollefson MB, Acker BA, Jacobsen EJ, Hughes RO, Walker JK, Fox DNA, Palmer MJ, Freeman SK, Yu Y, Bond BR (2010) 1-(2-Ethoxyethyl)-1H-pyrazolo[4,3-d]pyrimidines as potent phosphodiesterase 5 (PDE5) inhibitors. Bioorg Med Chem Lett 20(10):3120–3124

    Article  CAS  PubMed  Google Scholar 

  30. Kumar R, Joshi YC (2010) Synthesis, spectral studies and biological activity of novel 1H-1,4-diazepine derivatives. Indian J Chem Sect B 49B(1):84–88

    CAS  Google Scholar 

  31. Ivachtchenko AV, Dmitriev DE, Golovina ES, Dubrovskaya ES, Kadieva MG, Koryakova AG, Kysil VM, Mitkin OD, Tkachenko SE, Okun IM, Vorobiov AA (2010) Synthesis of cycloalkane-annelated 3-phenylsulfonyl-pyrazolo[1,5-a]pyrimidines and their evaluation as 5-HT6 receptor antagonists. Bioorg Med Chem Lett 20(7):2133–2136

    Article  CAS  PubMed  Google Scholar 

  32. Bakavoli M, Bagherzadeh G, Vaseghifar M, Shiri A, Pordel M, Mashreghi M, Pordeli P, Araghi M (2010) Molecular iodine promoted synthesis of new pyrazolo[3,4-d]pyrimidine derivatives as potential antibacterial agents. Eur J Med Chem 45(2):647–650

    Article  CAS  PubMed  Google Scholar 

  33. Curran KJ, Verheijen JC, Kaplan J, Richard DJ, Toral-Barza L, Hollander I, Lucas J, Ayral-Kaloustian S, Yu K, Zask A (2010) Pyrazolopyrimidines as highly potent and selective, ATP-competitive inhibitors of the mammalian target of rapamycin (mTOR): optimization of the 1-substituent. Bioorg Med Chem Lett 20(4):1440–1444

    Article  CAS  PubMed  Google Scholar 

  34. Kim I, Song JH, Park CM, Jeong JW, Kim HR, Ha JR, No Z, Hyun YL, Cho YS, Kang NS, Jeon DJ (2010) Design, synthesis, and evaluation of 2-aryl-7-(3′,4′-dialkoxyphenyl)-pyrazolo[1,5-a]pyrimidines as novel PDE-4 inhibitors. Bioorg Med Chem Lett 20(3):922–926

    Article  CAS  PubMed  Google Scholar 

  35. Ali HI, Fujita T, Akaho E, Nagamatsu T (2010) A comparative study of AutoDock and PMF scoring performances, and SAR of 2-substituted pyrazolotriazolopyrimidines and 4-substituted pyrazolopyrimidines as potent xanthine oxidase inhibitors. J Comput Aided Mol Des 24(1):57–75

    Article  CAS  PubMed  Google Scholar 

  36. Schenone S, Brullo C, Bruno O, Bondavalli F, Mosti L, Maga G, Crespan E, Carraro F, Manetti F, Tintori C, Botta M (2008) Synthesis, biological evaluation and docking studies of 4-amino substituted 1H-pyrazolo[3,4-d]pyrimidines. Eur J Med Chem 43(12):2665–2676

    Article  CAS  PubMed  Google Scholar 

  37. Cresswell RM, Wood HCS (1960) The biosynthesis of pteridines. Part I. The synthesis of riboflavin. J Chem Soc. 4768–4775

  38. El-Kalyoubi S, Agili F (2016) A novel synthesis of fused uracils: indenopyrimidopyridazines, pyrimidopyridazines, and pyrazolopyrimidines for antimicrobial and antitumor evalution. Molecules. https://doi.org/10.3390/molecules21121714

    Article  PubMed  Google Scholar 

  39. Furukawa Y, Maki Y, (1986) 3-Aminopyrazolo[3,4-d]pyrimidine derivatives and production thereof. US Patent 4,603,203

  40. Youssif S, Assy M (1996) Fervenulin, 4-deazafervenulin and 5-deaza-alloxazine analogues: Synthesis and Antimicribial activity. J Chem Res 442:2546

    Google Scholar 

  41. Halawa AH, Elaasser MM, El Kerdawy AM, Abd El-Hady AMAI, Emam HA, El-Agrody AM (2017) Anticancer activities, molecular docking and structure–activity relationship of novel synthesized 4H-chromene, and 5H-chromeno[2,3-d]pyrimidine candidates. Med Chem Res 26(10):2624–2638

    Article  CAS  Google Scholar 

  42. Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65:55–63

    Article  CAS  Google Scholar 

Download references

Authors’ contributions

SAEK formulated the research idea, conceived and prepared the manuscript; SAEK performed the experiments and analyzed the data; SAEK wrote the paper. The author read and approved the final manuscript.

Acknowledgements

The Author wishes to thank Dr. Mahmoud Elaasser for carrying out and drafting the biological activity of this work; who is an associate professor at the Regional Center for Mycology and Biotechnology at Al-Azhar University, Cairo, Egypt.

Competing interests

The author declares no competing interests.

Ethics approval and consent to participate

Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Samar A. El-Kalyoubi.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

El-Kalyoubi, S.A. Synthesis and anticancer evaluation of some novel pyrimido[5,4-e][1,2,4]triazines and pyrazolo[3,4-d]pyrimidine using DMF-DMA as methylating and cyclizing agent. Chemistry Central Journal 12, 64 (2018). https://doi.org/10.1186/s13065-018-0424-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13065-018-0424-3

Keywords