Skip to main content

Synthesis, anticancer evaluation, molecular docking and ADME study of novel pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidines as potential tropomyosin receptor kinase A (TrKA) inhibitors

Abstract

The starting compound 3-amino-1,7-dihydro-4H-pyrazolo[4,3-c]pyridine-4,6(5H)-dione (1) is reacted with each of diketone and β-ketoester, forming pyridopyrazolo[1,5-a]pyrimidines 4a,b and 14a,b, respectively. The compounds 4 and 14 reacted with each of aromatic aldehyde and arenediazonium salt to give the respective arylidenes and arylhydrazo derivatives, respectively. The structure of the new products was established using spectroscopic techniques. The cytotoxic activity of selected targets was tested in vitro against three cancer cell lines MCF7, HepG2 and HCT116. The data obtained from enzymatic assays of TrKA indicated that compounds 7b and 16c have the strongest inhibitory effects on TrKA with IC50 = 0.064 ± 0.0037 μg/ml and IC50 = 0.047 ± 0.0027 μg/ml, respectively, compared to the standard drug Larotrectinib with IC50 = 0.034 ± 0.0021 μg/ml for the HepG2 cancer cell line. In cell cycle analysis, compounds 7b, 15b, 16a and 16c caused the greatest arrest in cell cycle at the G2/M phase. In addition, compound 15b has a higher apoptosis-inducing effect (36.72%) than compounds 7b (34.70%), 16a (21.14) and 16c (26.54%). Compounds 7b, 16a and 16c were shown fit tightly into the active site of the TrKA kinase crystal structure (PDB: 5H3Q). Also, ADME study was performed on some selected potent anticancer compounds described in this study.

Highlights

  • A series of new pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine derivatives were synthesized.

  • The anticancer activity of the new compounds were tested in vitro.

  • Compounds 7b and 16c showed broad spectrum potent anticancer activity.

  • Compound 15b induced cell cycle arrest at G2/M phase in HepG-2 cell line.

  • Compound 7c, the most promising agent, can be absorbed very easily by the gastrointestinal tract with potential BBB permeability.

Peer Review reports

Introduction

Cancer is the growth of cells in certain parts of the body that grow out of control and can invade other tissues. Cancer is the second leading cause of death worldwide and chemotherapy, radiotherapy, and/or surgery are the most common cancer treatment techniques. Over the past decade, much research has focused on finding new therapies that reduce the side effects of conventional treatments.

The identification of gene fusions in certain cancers has provided a practical target for expanding therapeutic options and advancing precision medicine. These genetic abnormalities lead to the expression of constitutively active fusion proteins that are carcinogenic drivers [1]. Gene fusions are a type of mutation that commonly occurs in many types of cancer. They often result from chromosomal rearrangement that cause migration of coding or regulatory regions between genes. The tropomyosin tyrosine receptor kinase (TrK) family is of interest because the NTRK genes encoding have been implicated in gene fusions identified in a variety of adult and pediatric tumors. Three members of TrKA, encode transmembrane proteins NTRK1, TrKB (NTRK2) and TrKC (NTRK3) [2, 3]. As shown in Fig. 1, Trks are activated by the a family of nerve growth factors including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), Neurotrophin-4 (NT-4) and Neurotrophin-3 (NT-3) [3].

Fig. 1
figure 1

The three types of tropomyosin receptor kinases (Trks)

Larotrectinib is an inhibitor of the tropomyosin receptor kinases TrkA, TrkB, and TrkC (approved by the FDA in 2018). It has been indicated in adults and adolescents with solid tumors harboring NTRK gene fusions without a known acquired resistance mutations, in case of metastases or undergoing surgery. Resection can cause serious complications. Figure 2 shows another multitarget type-I kinase inhibitor with a pyrazole ring, such as entrectinib [4,5,6,7]. Despite the high response rates achieved with first-generation TrK inhibitors, drug resistance still exists, ultimately leading to treatment failure [8, 9]. Additionally, TrKA is the most commonly identified oncogene, found in several tumor types at a rate of approximately 7.4% (4% for TRKB and 3.4% for TRKA) [10, 11].

Fig. 2
figure 2

Larotrectinib and entrectinib as type-I multi-target kinase inhibitors

Furthermore, TrKA has been shown to mediate the stimulation of early tumor growth [12]. Therefore, inhibiting TrKA signaling is an attractive clinical approach for cancer therapy. Therefore, it is highly desirable to obtain new selective Trk inhibitors with different chemical scaffolds as new anti-neuroblastoma (NB) agents. Previously, two TrKA inhibitors were approved by the U.S. Food and Drug Administration (FDA). Larotrectinib was approved for solid tumors with NTRK gene fusions in November 2018 [13], with very low IC50 value for the Trk family (IC50 = 2–20 nM), and significant activity outside this kinase family [14]. Entrectinib was approved in August 2019 for NTRK gene fusion-positive or ROS1-positive solid tumors [15]. According to the classification of Shokat et al. [16] all are classified as type I kinase inhibitors.

Additionally, some pyrazolo[1,5-a]pyrimidine derivatives such as I, II and III showed good activity against HCT116, HeLa and HepG2 cell lines, respectively [17,18,19]. Moreover, the standard drug dinaciclib IV acts as a potent and selective cyclin-dependent kinase (CDK) inhibitor (Fig. 3) [20, 21].

Fig. 3
figure 3

Some pyrazolo[1,5-a]pyrimidines such as I–III with a standard drug dinaciclib as anticancer agents

Based on our research program to synthesize several bioactive heterocyclic compounds [22,23,24,25,26,27,28,29,30,31,32,33], we followed our previous work [24], which showed that some pyrazolopyridine derivatives have good cytotoxic activity against the MCF7 and HepG2 cell lines, respectively. Therefore, we synthesized other series of some novel heterocycles containing pyrazolo[1,5-a] pyrimidine hybrid with pyridine moiety to evaluate their anticancer activity against the three cell lines MCF7, HepG2 and HCT116. Moreover, evaluation of these compounds against TrKA enzyme was done (Fig. 4).

Fig. 4
figure 4

Our target compounds as anticancer agents and TrKA inhibitors compared to larotrectinib

Experimental

Materials and methods

The melting points are uncorrected and measured on an Electrothermal instrument (9100). Infrared spectra were recorded on a Perkin Elmer 1430 spectrophotometer (KBr pellet). On a Varian Gemini NMR spectrometer using tetramethylsilane as the internal reference and the results are expected as δ value, the 1H NMR and.13C NMR spectra were recorded at deuterated dimethylsulfoxide at 300 and 75 MHz. Mass spectra were performed on a Shimadzu GCMS-QP 1000 Ex mass spectrometer at 70 eV. Elemental analysis was performed at the Center for Microanalyses of Cairo University, Giza, Egypt. Enzyme, cell cycle and apoptosis inhibition were performed at VACSERA, Cairo, Egypt. Compound 1 was prepared according to the previous literature [34]

General procedure of synthesis of 4a,b

In 15 ml of DMF, a mixture of compound 1 (0.01 mol) and diketones 2a,b (0.01 mol) containing few drops of piperidine was heated under reflux for 10 h. The resulting solid was filtered, washed with ethanol and recrystallized from DMF.

2,4-Dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-diones (4a)

Brown crystals, yield 86%, m.p. 330 °C, νmax/cm−1 (KBr) 3187 (NH), 1702 (CO); 1H NMR (DMSO-d6) δ = 2.58 (s, 3H, CH3), 2.70 (s, 3H, CH3), 4.05 (s, 2H, CH2), 7.16 (s, 1H, pyrimidine-H), 10.82 (s, 1H, NH); 13 C NMR (DMSO-d6) δ = 14.55, 16.74, 50.60, 72.32, 79.24, 79.75, 114.41, 117.11, 143.55, 154.16, 167.77; m/z 230 = (M+, 67.6%), 214 (2.78%), 201 (3.19%), 187 (85.1%), 159 (18.79%), 132 (9.1%), 113 (23.6%), 101 (18.87%), 87 (22.2%), 78 (13.4%), 59 (100%), 52 (6.85%); Anal. Calcd for C11H10N4O2: C, 57.39; H, 4.38; N, 24.34. Found: C, 57.53; H, 4.55; N, 24.12%.

2,4-Diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (4b)

Brown crystals, yield 76%, m.p. 300 °C, νmax/cm−1 (KBr) 3181 (NH), 1693 (CO); 1H NMR (DMSO-d6) δ = 4.04 (s, 2H, CH2), 7.55–7.59 (m, 7H, Ar–H), 7.61 (s, 1H, Ar), 7.98 (m, 2H, Ar–H), 8.11 (m, 1H, CH), 11.0 (s, 1H, NH); m/z 354 = (M+, 100%), 311 (93.1%), 282 (8.1%), 255 (5.2%), 204 (18.8%), 189 (9.3%), 155 (13.3%), 127 (17.5%), 102 (57.1%), 77 (28.2%), 64 (8.0%), 51 (11.4%); Anal. Calcd for C21H14N4O2: C, 71.18; H, 3.98; N, 15.81. Found: C, 71.33; H, 3.79; N, 15.57%.

General procedure of synthesis of 7a–t

A mixture of compounds 4a (or 4b) (0.01 mol) and the appropriate aldehyde 6aj (0.01 mol) in DMF (15 ml) with few drops of piperidine was refluxed for 5 h. The reaction mixture was cooled at room temperature, the solid so formed was collected by filtration and recrystallized from DMF.

7-Benzylidene-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7a)

Brown crystals, yield 63%, m.p. 240 °C, νmax/cm−1 (KBr) 3181 (NH), 1698 (CO); 1H NMR (DMSO-d6) δ = 2.54 (s, 3H, CH3), 2.62 (s, 3H, CH3), 7.14 (s, 1H, CH), 7.48 (m, 3H, Ar–H), 8.15 (s, 1H, CH), 8.31–8.33 (s, 2H, Ar), 11.0 (s, 1H, NH); 13C NMR (DMSO-d6) δ = 17.02, 24.90, 98.38, 112.48, 118.74, 128.34, 131.67, 132.94, 133.95, 145.40, 146.33, 146.94, 150.60, 159.70, 163.62, 166.00; Anal. Calcd for C18H14N4O2: C, 67.92; H, 4.43; N, 17.60. Found: C, 67.75; H, 4.54; N, 17.36%.

7-(4-Methoxyphenyl-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10-(7H,9H)-dione (7b)

Brown crystals, yield 70%, m.p. 280 °C, νmax/cm−1 (KBr) 3211 (NH), 1697 (CO); 1H NMR (DMSO-d6) δ = 2.55 (s, 3H, CH3), 2.69 (s, 3H, CH3), 3.68 (s, 3H, OCH3), 7.0 (d, J = 6 Hz, 2H, Ar–H), 7.12 (s, 1H, Ar–H), 8.10 (s, 1H, CH), 8.49 (d, J = 6 Hz, 2H, Ar–H), 10.93 (s, 1H, NH); Anal. Calcd for C19H16N4O3: C, 65.51; H, 4.63; N, 16.08. Found: C, 65.38; H, 4.74; N, 16.34%.

7-(4-Chlorophenyl-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7c)

Yellow crystals, yield 71%, m.p. 260 °C, νmax/cm−1 (KBr) 3211 (NH), 1697 (CO); 1H NMR (DMSO-d6) δ = 2.46 (s, 3H, CH3), 2.71 (s, 3H, CH3), 7.18 (s, 1H, CH), 7.46–7.63 (m, 4H, Ar–H). 8.42 (s, 1H, CH), 11.02 (s, 1H, NH); Anal. Calcd for C18H13ClN4O2: C, 61.28; H, 3.71; Cl, 10.05; N, 15.88. Found: C, 61.38; H, 3.59; N, 15.65%.

7-(2-Hydroxybenzylidene)-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7d)

Brown crystals, yield 89%, m.p > 360 °C, νmax/cm−1 (KBr) 3417 (OH), 3269 (NH), 1691(CO);1H NMR (DMSO-d6) δ = 2.58 (s, 3H, CH3), 2.63 (s, 3H, CH3), 6.75–7.44 (m, 5H, Ar–H),8.47 (s, 1H, CH), 10.41 (s, 1H, OH), 11.07 (s, 1H, NH); Anal. Calcd for C18H14N4O3: C, 64.67; H, 4.22; N, 16.76. Found: C, 64.51; H, 4.35; N, 16.54%.

7-(2,5-Dimethoxyphenyl-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7e)

Yellow crystals, yield 90%, m.p. 275 °C, νmax/cm−1 (KBr) 3195 (NH), 170 (CO); 1H NMR (DMSO-d6) δ = 2.41 (s, 3H, CH3), 2.68 (s, 3H, CH3), 3.36 (s, 3H, OCH3), 3.81 (s, 3H, OCH3), 6.71–7.19 (m, 4H, Ar–H), 8.32 (s, 1H, CH), 10.89 (s, 1H, NH); Anal. Calcd for C20H18N4O4: C, 63.49; H, 4.80; N, 14.81. Found: C, 63.31; H, 4.64; N, 14.54%.

2,4-Dimethyl-7-(3,4,5-trimethoxybenzylidene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7f)

Yellow crystals, yield 82%, m.p.240 °C, νmax/cm−1 (KBr) 3211 (NH), 1701 (CO); 13C NMR (DMSO-d6) δ = 17.38, 24.77, 56.87, 60.72, 98.47, 111.55, 112.47, 117.18, 129.30, 141.25, 146.45, 150.88, 152.68, 159.58, 163.50, 166.22; Anal. Calcd for C21H20N4O5: C, 61.76; H, 4.94; N, 13.72. Found: C, 61.65; H, 4.78; N, 13.51%.

7-(Benzo[d][1,3]dioxol-5-ylmethylene)-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]-pyrimidine-8,10(7H,9H)-dione (7g)

Yellow crystals, yield 65%, m.p. 300 °C, νmax/cm−1 (KBr) 3169 (NH), 1682(CO);1H NMR (DMSO-d6) δ = 2.57 (s, 3H, CH3), 2.67 (s, 3H, CH3), 6.14 (s, 2H, CH2), 6.98 (d, 1H, J = 8.1 Hz, Ar–H), 7.14 (s, 1H, CH), 7.69 (d, 1H, J = 7.8 Hz, Ar–H), 8.03 (s, 1H, Ar–H)8.66 (s, 1H, CH), 10.94 (s, 1H, NH); Anal. Calcd for C19H14N4O4: C, 62.98; H, 3.89; N, 15.46. Found: C, 62.81; H, 3.70; N, 15.68%.

7-(Furan-2-ylmethylene)-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7h)

Brown crystals, yield 61%, m.p 305 °C, νmax/cm−1 (KBr) 3217 (NH), 1696 (CO);1H NMR (DMSO-d6) δ = 2.60 (s, 3H, CH3), 2.81 (s, 3H, CH3), 6.88(t, 1H, J = 3.6 Hz, Ar–H), 7.22 (s, 1H, CH),7.94 (d, 1H, J = 5.4 Hz, Ar–H), 8.14 (s, 1H, CH), 8.70(d, 1H, J = 3.64 Hz, Ar–H), 11.04 (s, 1H, NH); Anal. Calcd for C16H12N4O2: C, 62.33; H, 3.92; N, 18.17. Found: C, 62.51; H, 3.73; N, 18.40%.

2,4-Dimethyl-7-(thiophen-2-ylmethylene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7i)

Brown crystals, yield 85%, m.p. 290 °C, νmax/cm−1 (KBr) 3174 (NH), 1683 (CO);1H NMR (DMSO-d6) δ = 2.62 (s, 3H, CH3), 2.91 (s, 3H, CH3), 7.24 (s, 1H, CH),7.33 (t, 1H, J = 9 Hz, Ar–H), 8.15(d, 1H, J = 5.1 Hz, Ar–H), 8.21(d, 1H, J = 3.3 Hz, Ar–H), 8.42 (s, 1H, CH), 11.01 (s, 1H, NH); Anal. Calcd for C16H12N4O2S: C, 59.25; H, 3.73; N, 17.27; S, 9.88.Found: C, 59.43; H, 3.54; N, 17.49; S, 9.67%.

2,4-Dimethyl-7-(pyridin-4-ylmethylene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7j)

Brown crystals, yield 81%, m.p. 270 °C, νmax/cm−1 (KBr) 3435 (NH), 1695(CO);1H NMR (DMSO-d6) δ = 2.49 (s, 3H, CH3), 2.60 (s, 3H, CH3), 7.24 (s, 1H, CH), 8.03–81.2 (m, 3H, Ar–H), 8.68–8.71 (m, 2H, Ar–H), 11.23 (s, 1H, NH); Anal. Calcd for C17H13N5O2: C, 63.94; H, 4.10; N, 21.93. Found: C, 63.81; H, 4.27; N, 21.69%.

7-Benzylidene-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7k)

Brown crystals, yield 67%, m.p. 275 °C, νmax/cm−1 (KBr) 3231 (NH), 1691 (CO); 1H NMR (DMSO-d6) δ = 7.35 (m, 1H, Ar–H). 7.55–7.91 (m, 4H, Ar–H), 8.03–8.15 (m, 11H, Ar–H), 8.36 (s, 1H, CH), 11.21 (s, 1H, NH); 13C NMR (DMSO-d6) δ = 99.43, 108.54, 118.74, 128.33, 128.66, 128.95, 129.47, 130.52, 130.69, 131.51, 131.79, 131.89, 132.76, 134.05, 136.35, 145.73, 147.61, 147.75, 151.63, 159.30, 159.70, 166.05; Anal. Calcd for C28H18N4O2: C, 76.01; H, 4.10; N, 12.66. Found: C, 76.14; H, 4.26; N, 12.43%.

7-(4-Methoxybenzylidene-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7l)

Yellow crystals, yield 64%, m.p. 315 °C, νmax/cm−1 (KBr) 3216 (NH), 1697 (CO); 1H NMR (DMSO-d6) δ = 3.84 (s, 3H, OCH3), 7.13–7.21 (m, 6H, Ar–H). 7.52–7.92 (m, 7H, Ar–H), − 8.03–8.21 (m, 3H, Ar–H and CH), 11.09 (s, 1H, NH); Anal. Calcd for C29H20N4O3: C, 73.72; H, 4.27; N, 11.86. Found: C, 73.54; H, 4.40; N, 11.63%.

7-(4-Chlorobenzylidene)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7m)

Brown crystals, yield 63%, m.p. 320 °C, νmax/cm−1 (KBr) 3209 (NH), 1687 (CO); 1H NMR (DMSO-d6) δ = 7.39 (d, 2H, J = 8.7 Hz, CH), 7.60–7.66 (m, 7H, Ar–H and CH), 8.08 (d, 2H, J = 8.4 Hz, CH), 8.12 (s, 1H,CH), 8.19–8.21 (m, 2H, Ar–H), 8.42–8.45 (m, 2H, Ar–H), 11.25 (s, 1H, NH);Anal. Calcd for C28H17ClN4O2: C, 70.52; H, 3.59; Cl, 7.43; N, 11.75. Found: C, 70.37; H, 3.43; Cl, 7.62; N, 11.53%.

7-(2-Hydroxybenzylidene)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7n)

Brown crystals, yield 75%, m.p. 275 °C, νmax/cm−1 (KBr) 3426 (OH), 3176 (NH), 1684 (CO); 1H NMR (DMSO-d6) δ = 6.77 (t, 1H, J = 7.5 Hz, CH), 6.92 (d, 1H, J = 7.8 Hz, CH), 7.32 (t, 1H, J = 7.2 Hz, CH), 7.57–7.68 (m, 7H, Ar–H), 8.08 (s, 1H,CH), 8.15 (d, 2H, J = 7.5 Hz, CH), 8.40–8.56 (m, 3H, Ar–H), 10.32 (s, 1H, OH), 11.13 (s, 1H, NH); Anal. Calcd for C28H18N4O3: C, 73.35; H, 3.96; N, 12.22. Found: C, 73.52; H, 3.77; N, 12.46%.

7-(2,5-Dimethoxybenzylidene)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7o)

Brown crystals, yield 66%, m.p. 275 °C, νmax/cm−1 (KBr) 3269 (NH), 1700 (CO);1680 (CO); 1H NMR (DMSO-d6) δ = 3.39 (s, 3H, OCH3), 3.78 (s, 3H, OCH3), 6.99 (d, 1H, J = 6.9 Hz, CH), 7.06 (d, 1H, J = 6.6 Hz, CH), 7.40 (t, 2H, J = 7.5 Hz, CH), 7.56–7.57 (m, 4H, Ar–H), 7.64 (s, 1H, CH), 8.04–8.07 (m, 3H, Ar–H and CH),8.32 (s, 1H, CH), 8.37–8.39 (m, 2H, Ar),11.17 (s, 1H, NH); Anal. Calcd for C30H22N4O4: C, 71.70; H, 4.41; N, 11.15. Found: C, 71.55; H, 4.59; N, 11.39%.

2,4-Diphenyl-7-(3,4,5-trimethoxybenzylidene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]-pyrimidine-8,10(7H,9H)-dione (7p)

Brown crystals, yield 65%, m.p. 315 °C, νmax/cm−1 (KBr) 3186 (NH), 1705 (CO);1677 (CO); 1H NMR (DMSO-d6) δ = 3.40 (s, 6H, 2OCH3), 3.76 (s, 3H, OCH3), 7.43–7.66 (m, 8H, Ar–H),8.03 (s, 1H, CH), 8.04–8.08 (m, 2H, Ar–H), 8.18 (s, 1H, CH), 8.37–8.39 (m, 2H, Ar–H), 11.16 (s, 1H, NH); Anal. Calcd for C31H24N4O5: C, 69.92; H, 4.54; N, 10.52. Found: C, 69.79; H, 4.69; N, 10.76%.

7-(Benzo[d][1,3]dioxol-5-ylmethylene)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]-pyrimidine-8,10(7H,9H)-dione (7q)

Brown crystals, yield 89%, m.p. 335 °C, νmax/cm−1 (KBr) 3227 (NH), 1688 (CO);13C NMR (DMSO-d6) δ = 99.24, 102.36, 108.56, 112.18, 116.01, 128.15, 128.29, 129.15, 129.45, 130.21, 130.69, 130.75, 131.76, 131.86, 136.33, 145.96, 147.68, 150.72, 151.96, 159.22, 159.69, 166.37; Anal. Calcd for C29H18N4O4: C, 71.60; H, 3.73; N, 11.52. Found: C, 71.76; H, 3.64; N, 11.73%.

7-(Furan-2-ylmethylene)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7r)

Brown crystals, yield 70%, m.p. 335 °C, νmax/cm−1 (KBr) 3219 (NH), 1684 (CO); 1H NMR (DMSO-d6) δ = 6.65 (d, 1H, J = 5.7 Hz, CH), 7.61–7.63 (m, 3H, Ar–H), 7.72–7.76 (m, 3H, Ar–H), 8.02 (s, 1H,CH), 8.10–8.19 (m, 4H, Ar–H), 8.40–8.46 (m, 3H, Ar–H), 11.19 (s, 1H, NH); Anal. Calcd for C26H16N4O3: C, 72.22; H, 3.73; N, 12.96.Found: C, 72.39; H, 3.59; N, 12.73%.

2,4-Diphenyl-7-(thiophen-2-ylmethylene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7s)

Brown crystals, yield 61%, m.p. 315 °C, νmax/cm−1 (KBr) 3219 (NH), 1684 (CO); 1H NMR (DMSO-d6) δ = 7.18 (d.d, 1H, J = 4.92 Hz, CH), 7.57–7.60 (m, 3H, Ar–H), 7.69–7.76 (m, 3H, Ar–H), 7.96 (d, 1H, J = 5 Hz, CH), 8.07 (s, 1H,CH), 8.15 (dd, 2H, J = 7.92 Hz, CH),8.35–8.36 (m, 2H, Ar–H), 8.40–8.43 (m, 2H, Ar–H), 11.15 (s, 1H, NH); Anal. Calcd for C26H16N4O2S: C, 69.63; H, 3.60; N, 12.49; S, 7.15. Found: C, 69.44; H, 3.76; N, 12.74; S, 7.34%.

2,4-Diphenyl-7-(pyridin-4-ylmethylene)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (7t)

Brown crystals, yield 68%, m.p. 340 °C, νmax/cm−1 (KBr) 3238 (NH), 1696 (CO); 1H NMR (DMSO-d6) δ = 7.57–7.60 (m, 5H, Ar–H), 7.66 (d, 1H, J = 7.36 Hz, CH), 7.88 (d, 2H, J = 5.56 Hz, CH), 8.01 (d, 2H, J = 7.44 Hz, CH), 8.10 (s, 1H, CH), 8.13 (s, 1H, CH), 8.39–8.41 (m, 2H, Ar–H),8.61 (d, 2H, J = 5.76 Hz, CH), 11.35 (s, 1H, NH); Anal. Calcd for C27H17N5O2: C, 73.13; H, 3.86; N, 15.79. Found: C, 73.32; H, 3.72; N, 15.55%.

General procedure of synthesis of compounds 9a-d and 11a-d

Method A: Compound 4a or 4b (0.01 mol) and sodium acetate (0.01 mol) were stirred in DMF (5 ml) under cooling in an ice-bath (0–5 °C). To the resulting cold solution is added portionwise a cold solution of the appropriate arenediazonium chlorides 8ad. The mixture was stirred again under cooling conditions for 3 h., the resulting solid was filtered, washed with water and recrystallized from DMF.

Method B: A mixture of 3-amino-7-(2-arylhydrazono)-1,7-dihydro-4H-pyrazolo[4,3-c]pyridine-4,6-diones 10ad (0.01 mol) and each of acetylacetone 3a or dibenzoyl methane 3b was refluxed in DMF (10 ml) in the presence of piperidine for 7 h. The solid that formed was filtered and recrystallized from DMF.

2,4-Dimethyl-7-(2-phenylhydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (9a)

Brown crystals, yield 65% (A), 60% (B), m.p.300 °C, νmax/cm−1 (KBr) 3140 (NH), 1675 (CO); 1H NMR (DMSO-d6) δ = 2.63 (s, 3H, CH3), 2.80 (s, 3H, CH3), 7.30 (s, 1H, CH), 7.42–7.54 (m, 5H, Ar–H), 11.01 (s, 1H, NH), 12.43 (s, 1H, NH); Anal. Calcd for C17H14N6O2: C, 61.07; H, 4.22; N, 25.14. Found: C, 61.23; H, 4.41; N, 25.36%.

2,4-Dimethyl-7-(2-(p-tolyl)hydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (9b)

Yellow crystals, yield 66% (A), 61% (B), m.p. 285 °C, νmax/cm−1 (KBr) 3187 (NH), 1687 (CO); 1H NMR (DMSO-d6) δ = 2.30 (s, 3H, CH3), 2.62 (s, 3H, CH3), 2.87 (s, 3H, CH3), 7.19–7.42 (m, 5H, Ar–H), 10.94 (s, 1H, NH), 12.40 (s, 1H, NH); m/z 348 = (M+, 100%), 319 (5.2%), 304 (8.5%), 257 (3.7%), 229 (12.4%), 199 (99.1%), 174 (35.1%), 158 (23.5%), 105 (28.3%), 91 (67.6%), 77 (42.3%), 65 (38.7%); Anal. Calcd for C18H16N6O2: C, 62.06; H, 4.63; N, 24.12. Found: C, 62.25; H, 4.49; N, 24.35%.

7-(2-(4-Methoxyphenyl)hydrazono)-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyri-midine-8,10(7H,9H)-dione (9c)

Brown crystals, yield 72% (A), 68% (B), m.p. 305 °C, νmax/cm−1 (KBr) 3156 (NH), 1679 (CO); 1H NMR (DMSO-d6) δ = 2.44 (s, 3H, CH3), 2.71 (s, 3H, CH3), 3.76 (s, 3H, OCH3), 7.22–7.63 (m, 5H, Ar–H), 10.82 (s, 1H, NH), 12.11 (s, 1H, NH); m/z 364 = (M+, 2.75%), 230 (84.97%), 187 (100%), 158 (16.1%), 132 (5.2%), 108 (9.3%), 78 (6.8%), 65 (5.4%); Anal. Calcd for C18H16N6O3: C, 59.34; H, 4.43; N, 23.07. Found: C, 59.47; H, 4.27; N, 23.30%.

7-(2-(4-Chlorophenyl)hydrazono)-2,4-dimethylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (9d)

Brown crystals, yield 67% (A), 62% (B), m.p. 300 °C, νmax/cm−1 (KBr) 3183 (NH), 1676(CO); 1H NMR (DMSO-d6) δ = 2.56 (s, 3H, CH3), 2.76 (s, 3H, CH3), 7.25 (s, 1H, CH), 7.41–7.51 (m, 4H, Ar–H), 10.96 (s, 1H, NH), 12.26 (s, 1H, NH); Anal. Calcd for C17H13ClN6O2: C, 55.37; H, 3.55; Cl, 9.61; N, 22.79. Found: C, 55.51; H, 3.38; Cl, 9.67; N, 22.58%.

2,4-Diphenyl-7-(2-phenylhydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (11a)

Brown crystals, yield 70% (A), 68% (B), m.p. 315 °C, νmax/cm−1 (KBr) 3369 (NH), 1689(CO); 1H NMR (DMSO-d6) δ = 7.21 (d, 2H, J = 7.2 Hz, CH), 7.42–7.46 (m, 3H, Ar–H), 7.60–7.80 (m, 6H, Ar–H),8.20 (s, 1H, CH), 8.29–8.43 (m, 4H, Ar–H),11.09 (s, 1H, NH), 12.36 (s, 1H, NH); Anal. Calcd for C27H18N6O2: C, 70.73; H, 3.96; N, 18.33. Found: C, 70.55; H, 3.80; N, 18.55%.

2,4-Diphenyl-7-(2-(p-tolyl)hydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (11b)

Yellow crystals, yield 66% (A), 67% (B), m.p. 320 °C, νmax/cm−1 (KBr) 3189 (NH), 1701 (CO);1H NMR (DMSO-d6) δ = 2.25 (s, 3H, CH3), 7.03 (d, 2H, J = 8.4 Hz, CH), 7.15 (d, 2H, J = 8.1 Hz, CH), 7.56–7.58 (m, 3H, Ar–H), 7.75–7.77 (m, 3H, Ar–H),8.13 (s, 1H, Ar–H),8.25–8.27 (m, 2H, Ar–H),8.38–8.41 (m, 2H, Ar–H),11.01 (s, 1H, NH), 12.27 (s, 1H, NH); Anal. Calcd for C28H20N6O2: C, 71.18; H, 4.27; N, 17.79. Found: C, 71.34; H, 4.46; N, 17.54%.

7-(2-(4-Methoxyphenyl)hydrazono)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyri-midine-8,10(7H,9H)-dione (11c)

Brown crystals, yield 60% (A), 62% (B), m.p. 325 °C, νmax/cm−1 (KBr) 3265 (NH), 1692 (CO); 1H NMR (DMSO-d6) δ = 3.81 (s, 3H, OCH3), 7.14–7.39 (m, 4H, Ar–H), 7.53–7.72 (m, 6H, Ar–H), 8.09 (s, 1H,CH), 8.21–8.39 (m, 4H, Ar–H), 11.07 (s, 1H, NH), 12.35 (s, 1H, NH); Anal. Calcd for C28H20N6O3: C, 68.84; H, 4.13; N, 17.20. Found: C, 68.69; H, 4.29; N, 17.43%.

7-(2-(4-Chlorophenyl)hydrazono)-2,4-diphenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (11d)

Brown crystals, yield 72% (A), 68% (B), m.p. 335 °C, νmax/cm−1 (KBr) 3148 (NH), 1686 (CO); 1H NMR (DMSO-d6) δ = 7.13–7.32 (m, 4H, Ar–H), 7.59–7.85 (m, 6H, Ar–H), 8.13(s, 1H,CH), 8.27–8.45 (m, 4H, Ar–H), 11.13 (s, 1H, NH), 12.42 (s, 1H, NH); m/z 492 = (M+, 30.2%), 452 (1.8%), 423 (1.3%), 396 (2.0%), 367 (3.6%), 346 (17.6%), 323 (24.6%), 304 (28.7%), 282 (17.0%), 231 (16.5%), 204 (21.5%), 165 (22.8%), 129 (55.3%), 111 (97.9%), 99 (48.5%), 77 (100%), 43 (85.1%);Anal. Calcd for C27H17ClN6O2: C, 65.79; H, 3.48; Cl, 7.19; N, 17.05. Found: C, 65.62; H, 3.32; Cl, 7.41; N, 17.28%.

General procedure for synthesis of compounds 14a,b

A mixture of compound 1 (0.01 mol) and β-ketoesters 12a,b (0.01 mol) was refluxed in glacial acetic acid (20 ml) for 9 h. The resulting solid was collected by filtration and recrystallized from DMF.

4-Hydroxy-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (14a)

Brown crystals, yield 65%, m.p. > 360 °C, νmax/cm−1 (KBr) 3430 (OH), 3187 (NH), 1689 (CO); 1H NMR (DMSO-d6) δ = 2.37 (s, 3H, CH3), 3.95 (s, 2H, CH2), 5.86 (s, 1H, CH), 10.96 (s, 1H, NH), 12.80 (s, 1H, OH); m/z 232 = (M+, 100%), 214 (29.3%), 204 (5.7%), 189 (5.1%), 175 (2.1%), 159 (9.0%), 148 (5.2%), 133 (40.2%), 120 (4.1%), 105 (11.2%), 92 (7.4%), 78 (9.2%), 65 (29.2%), 52 (7.9%);Anal. Calcd for C10H8N4O3: C, 51.73; H, 3.47; N, 24.13.Found: C, 51.57; H, 3.28; N, 24.36%.

4-Hydroxy-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (14b)

Brown crystals, yield 65%, m.p. 310 °C, νmax/cm−1 (KBr) 3404 (OH),3189 (NH), 1688 (CO); 1H NMR (DMSO-d6) δ = 3.83 (s, 1H, OH), 4.00 (s, 2H, CH2), 6.22 (s, 1H, CH), 7.51–7.59 (m, 3H, Ar–H), 7.74–7.77 (m, 2H, Ar–H), 10.97 (s, 1H, NH); m/z 294 = (M+, 100%), 276 (22.2%), 265 (1.8%), 251 (4.6%), 232 (2.9), 220 (3.9%), 195 (21.1%), 166 (29.4%), 140 (6.4%), 129 (20.2%), 123 (26.0%), 102 (66.3%), 92 (4.7%), 76 (17.8%), 66 (28.8%), 51 (13.0%);Anal. Calcd for C15H10N4O3: C, 61.22; H, 3.43; N, 19.04. Found: C, 61.37; H, 3.26; N, 19.26%.

General procedure for synthesis of compounds 15af

Refluxing of a mixture of compounds 14a,b (0.01 mol) and aldehydes 6ac (0.01 mol) in DMF (20 ml) in a few drops of piperidine for 10 h. The solid that formed was filtered and crystallized from DMF.

7-Benzylidene-4-hydroxy-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15a)

Brown crystals, yield 79%, m.p. 305 °C, νmax/cm−1 (KBr) 3419 (OH), 3203 (NH), 1704 (CO); 13C NMR (DMSO-d6) δ = 19.38, 95.29, 100.20, 118.19, 128.71, 131.98, 133.48, 133.81, 141.91, 145.54, 148.10, 152.58, 155.27, 160.24, 166.42; Anal. Calcd for C17H12N4O3: C, 63.75; H, 3.78; N, 17.49.Found: C, 63.66; H, 3.65; N, 17.71%.

4-Hydroxy-7-(4-methoxybenzylidene)-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15b)

Brown crystals, yield 84%, m.p. 345 °C, νmax/cm−1 (KBr) 3423 (OH), 3195 (NH), 1708 (CO); 1H NMR (DMSO-d6) δ = 2.41 (s, 3H, CH3), 3.89 (s, 3H, OCH3), 5.95 (s, 1H, CH), 7.07 (d, 2H, J = 8.8 Hz, Ar–H), 8.19 (s, 1H, CH), 8.75 (d, 2H, J = 8.8 Hz, Ar–H), 11.21 (s, 1H, NH), 12.75 (s, 1H, OH); Anal. Calcd for C18H14N4O4: C, 61.71; H, 4.03; N, 15.99.Found: C, 61.71; H, 4.03; N, 15.99%.

7-(4-Chlorobenzylidene)-4-hydroxy-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15c)

Brown crystals, yield 65%, m.p. 330 °C, νmax/cm−1 (KBr) 3434 (OH), 3188 (NH), 1713 (CO);1H NMR (DMSO) δ = 2.39 (s, 3H, CH3), 5.93 (s, 1H, Ar–H), 7.53 (dd, 2H, J = 8.7 Hz, Ar–H), 8.16 (s, 1H, CH), 8.52 (d, 2H, J = 7.8 Hz, Ar–H), 11.29 (s, 1H, NH), 12.76 (s, 1H, OH); Anal. Calcd for C17H11ClN4O3: C, 57.56; H, 3.13; Cl, 9.99; N, 15.79.Found: C, 57.40; H, 3.32; Cl, 9.82; N, 15.57%.

7-Benzylidene-4-hydroxy-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15d)

Brown crystals, yield 69%, m.p. 300 °C, νmax/cm−1 (KBr) 3387 (OH), 3179 (NH), 1689 (CO); 1H NMR (DMSO-d6) δ = 3.37 (s, 1H, OH), 6.24 (s, 1H, CH), 7.23–7.56 (m, 6H, Ar), 7.61–7.67 (m, 4H, Ar), 8.11 (s, 1H, CH), 11.33 (s, 1H, NH); Anal. Calcd for C22H14N4O3: C, 69.10; H, 3.69; N, 14.65.Found: C, 69.26; H, 3.56; N, 14.44%.

4-Hydroxy-7-(4-methoxybenzylidene)-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15e)

Brown crystals, yield 63%, m.p. 300 °C, νmax/cm−1 (KBr) 3401 (OH), 3197 (NH), 1705 (CO); 1H NMR (DMSO-d6) δ = 3.39 (s, 1H, OH), 3.76 (s, 3H, OCH3), 6.23 (s, 1H, CH), 7.11–7.42 (m, 5H, Ar–H), 7.51–7.59 (m, 4H, Ar–H), 8.06 (s, 1H, CH), 11.41 (s, 1H, NH); 13 C NMR (DMSO-d6) δ = 56.06, 79.24, 79.50, 79.79, 99.59, 114.25, 114.42, 116.33, 127.0, 128.72, 129.05, 135.72, 136.75, 139.50, 142.46, 145.64, 145.83, 160.0, 161.24, 162.09, 166.81, 167.19, 170.02; Anal. Calcd for C23H16N4O4: C, 66.99; H, 3.91; N, 13.59.Found: C, 66.82; H, 3.79; N, 13.83%.

7-(4-Chlorobenzylidene)-4-hydroxy-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (15f)

Brown crystals, yield 72%, m.p. 330 °C, νmax/cm−1 (KBr) 3412 (OH), 3197 (NH), 1704 (CO); 1H NMR (DMSO-d6) δ = 3.34 (s, 1H, OH), 6.28 (s, 1H, CH), 7.54–7.60 (m, 5H, Ar–H), 7.76–7.79 (m, 2H, Ar–H), 8.17 (s, 1H, CH), 8.52 (d, 2H, J = 8.4 Hz, Ar–H), 11.30 (s, 1H, NH); Anal. Calcd for C22H13ClN4O3: C, 63.39; H, 3.14; Cl, 8.50; N, 13.44. Found: C, 63.53; H, 3.31; Cl, 8.33; N, 13.67%.

General procedure of synthesis of compounds 16a–h

A cold solution of arenediazonium chlorides 8ad was added drop wise in an ice-bath (0–5 °C) to a mixture of compound 4a (0.01 mol) and sodium acetate (0.01 mol) in DMF (5 ml), after stirring for 3 h. After forming, the resultant solid was filtrated, washed with water and recrystallized from DMF.

4-Hydroxy-2-methyl-7-(2-phenylhydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimid-ine-8,10(7H,9H)-dione (16a)

Brown crystals, yield 69%, m.p. 336 °C, νmax/cm−1 (KBr) 3411 (OH), 3201 (NH), 1702 (CO); 1H NMR (DMSO-d6) δ = 2.33 (s, 3H, CH3), 5.93 (s, 1H, CH), 7.11–7.56 (m, 5H, Ar–H), 11.08 (s, 1H, NH), 12.41 (s, 1H, NH), 12.94 (s, 1H, OH); m/z 336 = (M+, 46.3%), 307 (3.1%), 298 (15.8%), 270 (15.8%), 259 (7.2%), 232 (9.1%), 203 (14.0%), 176 (62.4%), 133 (14.3%), 121 (15.9%), 105 (20.3%), 91 (43.2%), 77 (100%), 44 (50.2%); Anal. Calcd for C16H12N6O3: C, 57.14; H, 3.60; N, 24.99. Found: C, 57.33; H, 3.47; N, 24.73%.

4-Hydroxy-2-methyl-7-(2-(p-tolyl)hydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16b)

Brown crystals, yield 71%, m.p. 330 °C, νmax/cm−1 (KBr) 3441 (OH), 3183 (NH), 1694 (CO); 1H NMR (DMSO-d6) δ = 2.34 (s, 3H, CH3), 2.43 (s, 3H, CH3), 5.79–5.90 (m, 2H, Ar–H), 7.19–7.35 (m, 3H, Ar–H), 11.05 (s, 1H, NH), 12.33 (s, 1H, NH), 12.78 (s, 1H, OH); Anal. Calcd for C17H14N6O3: C, 58.28; H, 4.03; N, 23.99. Found: C, 58.44; H, 4.21; N, 23.74%.

4-Hydroxy-7-(2-(4-methoxyphenyl)hydrazono)-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16c)

Brown crystals, yield 67%, m.p. 321 °C, νmax/cm−1 (KBr) 3426 (OH), 3195 (NH), 1702 (CO); 1H NMR (DMSO-d6) δ = 2.41 (s, 3H, CH3), 3.76 (s, 3H, OCH3), 5.91 (s, 1H, CH), 7.43–7.66 (m, 4H, Ar–H), 11.19 (s, 1H, NH), 12.37 (s, 1H, NH), 12.87 (s, 1H, OH); m/z 366 (M+, 49.7%), 338 (4.6%), 300 (6.2%), 298 (97.6%), 270 (100%), 242 (35.7%), 232 (17.5%), 201 (16.9%), 176 (22.3%), 159 (15.3%), 133 (18.1%), 121 (79.4%), 102 (42.8%), 77 (72.8%), 67 (95.2%), 51 (28.5%); Anal. Calcd for C17H14N6O4: C, 55.74; H, 3.85; N, 22.94. Found: C, 55.87; H, 3.69; N, 22.73%.

7-(2-(4-Chlorophenyl)hydrazono)-4-hydroxy-2-methylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16d)

Brown crystals, yield 65%, m.p. 310 °C, νmax/cm−1 (KBr) 3436 (OH), 3181 (NH), 1689 (CO); 1H NMR (DMSO-d6) δ = 2.39 (s, 3H, CH3), 5.98 (s, 1H, CH), 7.36 (d, 2H, J = 8.7 Hz, Ar–H), 7.46 (d, 2H, J = 8.7 Hz, Ar–H), 11.15 (s, 1H, NH), 12.38 (s, 1H, NH), 12.95 (s, 1H, OH); Anal. Calcd for C16H11ClN6O3: C, 51.83; H, 2.99; Cl, 9.56; N, 22.67. Found: C, 51.69; H, 2.82; Cl, 9.71; N, 22.43%.

4-Hydroxy-2-phenyl-7-(2-phenylhydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16e)

Brown crystals, yield 61%, m.p. 300 °C, νmax/cm−1 (KBr) 3411 (OH), 3173 (NH), 1706 (CO); 1H NMR (DMSO-d6) δ = 3.51 (s, 1H, OH), 6.35 (s, 1H, Ar–H), 7.21–7.33 (m, 5H, Ar–H), 7.44–7.60 (m, 3H, Ar–H), 7.75–7.80 (m, 2H, Ar–H), 11.09 (s, 1H, NH), 12.54 (s, 1H, NH); Anal. Calcd for C21H14N6O3: C, 63.31; H, 3.54; N, 21.10. Found: C, 63.44; H, 3.38; N, 21.36%.

4-Hydroxy-2-phenyl-7-(2-(p-tolyl)hydrazono)pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16f)

Brown crystals, yield 63%, m.p. 320 °C, νmax/cm−1 (KBr) 3437 (OH), 3186 (NH), 1685 (CO); 1H NMR (DMSO) δ = 2.31 (s, 3H, CH3), 3.39 (s, 1H, OH), 6.35 (s, 1H, Ar), 7.21–7.33 (m, 4H, Ar), 7.44–7.60 (m, 3H, Ar), 7.75–7.80 (m, 2H, Ar), 11.09 (s, 1H, NH), 12.54 (s, 1H, NH); Anal. Calcd for C22H16N6O3: C, 64.07; H, 3.91; N, 20.38. Found: C, 64.25; H, 3.77; N, 20.62%.

4-Hydroxy-7-(2-(4-methoxyphenyl)hydrazono)-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16g)

Brown crystals, yield 64%, m.p. 323 °C, νmax/cm−1 (KBr) 3418 (OH), 3205 (NH), 1697 (CO); 1H NMR (DMSO-d6) δ = 3.48 (s, 1H, OH), 3.81 (s, 3H, OCH3), 6.35 (s, 1H, Ar–H), 7.21–7.33 (m, 4H, Ar–H), 7.44–7.60 (m, 3H, Ar–H), 7.75–7.80 (m, 2H, Ar–H), 11.09 (s, 1H, NH), 12.54 (s, 1H, NH); 13C NMR (DMSO-d6) δ = 55.73, 79.22, 79.75, 99.42, 100.0, 115.53, 117.03, 117.64, 128.72, 129.0, 129.33, 131.40, 136.17, 147.60, 155.80, 157.0, 159.40, 161.89, 162.20, 169.83, 170.91, 171.27; Anal. Calcd for C22H16N6O4: C, 61.68; H, 3.76; N, 19.62. Found: C, 61.47; H, 3.62; N, 19.89%.

7-(2-(4-Chlorophenyl)hydrazono)-4-hydroxy-2-phenylpyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine-8,10(7H,9H)-dione (16h)

Brown crystals, yield 76%, m.p. 330 °C, νmax/cm−1 (KBr) 3429 (OH), 3199 (NH), 1709 (CO); 1H NMR (DMSO-d6) δ = 3.52 (s, 1H, OH), 6.35 (s, 1H, Ar–H), 7.21–7.33 (m, 4H, Ar–H), 7.44–7.60 (m, 3H, Ar–H), 7.75–7.80 (m, 2H, Ar), 11.09 (s, 1H, NH), 12.54 (s, 1H, NH); Anal. Calcd for C21H13ClN6O3: C, 58.28; H, 3.03; Cl, 8.19; N, 19.42. Found: C, 58.15; H, 3.22; Cl, 8.41; N, 19.19%

Biological investigation

Materials and methods

Cell line

The three cell lines MCF7, HePG2 and HTC 116 were obtained from ATCC via Holding company for biological products and vaccines (VACSERA), Cairo, Egypt. Doxorubicin was used as a standard anticancer drug for comparison.

Chemical reagents

The reagents are RPMI-1640 medium, MTT and DMSO (sigma co., St. Louis, USA), Fetal Bovine serum (GIBCO, UK).

MTT assay

Determination of the inhibitory effects of compounds on cell growth was performed through the MTT assay [35, 36]. This colorimetric assay is based on the conversion of the yellow tetrazolium bromide (MTT) to a purple formazan derivative by mitochondrial succinate dehydrogenase in viable cells. The protocol was discussed in details in Additional file 1.

Tropomyosin receptor kinase A (TrKA) inhibitory assay

The TrkA assay Kit is designed to measure TrkA activity for screening and profiling applications using Kinase-Glo® MAX as a detection reagent. The TrkA Assay Kit comes in a convenient 96-well format, with enough purified recombinant TrkA enzyme, TrkA substrate, ATP and kinase assay buffer for 100 enzyme reactions. The method was discussed in details in the ESI.

In-vitro cell cycle analysis

HepG-2 cells are pre-cultured in 25 cm2 cell culture flask. RPMI-1640 medium was used. Tested compounds 7b, 9c, 15b, 16a and 16c were used in the cell treatment at their IC50 by dissolving them in the required medium separately. The procedure was discussed in details in the ESI.

Annexin V-FITC apoptosis assay

HepG-2 cells were harvested and incubated with compounds 7b, 15b, 16a and 16c separately for 48 h. Then, the cells were collected and washed with PBS two successive times followed by centrifugation. After that, the cells were treated with Annexin V-FITC and propidium iodide (PI) using the apoptosis detection kit (BD Biosciences and Annexin V-FITC and PI binding were analyzed by a flow cytometer.

Molecular docking study

Molecular docking study was performed using program “Molecular Operating Environment (MOE) 2009. The protein structure was downloaded from the PDB data bank (http://www.rcsb.org/PDB codes: 5H3Q). The steps were discussed in details in the ESI.

In silico ADME studies

Physicochemical characteristics of 4a, 7ac, 9c, 15b, 16a, and 16c were detected through Swiss Target Predication methodology [37, 38].

Results and discussion

Chemistry

Reactivity of 3-amino-1,7-dihydro-4H-pyrazolo[4,3-c]pyridine-4,6(5H)-dione 1 as a precursor of some heterocycles of interesting biological activity [24, 39] encouraging us to continue our research on the synthesis of new compounds as a potential anticancer agents. Thus, condensation of compound 1 with each of acetylacetone 2a and dibenzoylmethane 2b, respectively, in N,N-dimethylformamide with a few drops of piperidine afforded products 4a, b. The structures of 4a, b were proven by spectroscopic techniques (Scheme 1). The IR spectrum of compound 4a shows absorption bands at 3187 and 1702 cm−1 assigned to the NH and CO groups, respectively. Its 1H NMR spectrum revealed three singlet signals assigned to the two methyl and methylene protons at δ = 2.58, 2.70 and 4.05 ppm, respectively, in addition to a singlet signal at δ = 7.16 ppm for pyrimidine proton. In addition, the D2O exchangeable signal appeared at δ = 10.82 ppm corresponding to the NH proton. Furthermore, the mass spectrum of 4a displayed a molecular ion peak at m/z = 230 (M+, 67.6%), consistent with the molecular formula C11H10N4O2 (Scheme 2).

Scheme 1
scheme 1

Synthesis of pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidines 4a,b

Scheme 2
scheme 2

Synthesis of compounds 7at

The mechanism of the formation of compounds 4a, b was suggested to proceed through nucleophilic attack of the exocyclic amino group in compound 1 on the ketonic function of acetylacetone 2a, followed by intramolecular cyclization with elimination of water from the intermediate 3 to produce. The other pathway that leads to formation of compounds 5a, b was excluded as shown in Scheme 1.

Each of compounds 4a (and 4b) was condensed with the appropriate aromatic aldehyde 6aj in refluxing DMF in presence of traces of piperidine to yield the respective arylmethylene derivatives 7at. The structures of 7at were supported by spectroscopic techniques. Compound 7a exhibits absorption bands in its IR chart at ν 3181 and 1698 cm−1 assigned to the NH and CO groups, respectively. Its 13C NMR exhibited characteristic signals at 17.02 (CH3), 128.34 (CH=C), 163.62 (CO) and 166.00 (CO), in addition to the expected signals (Scheme 2).

Further coupling of compound 4a with arenadiazonium salts 8ad in DMF containing sodium acetate at 0–5 °C afforded the corresponding arylhydrazono derivatives 9ad (Scheme 3). The resulting structures were established by elemental analysis and spectroscopic data. For example, the IR spectrum of 9b is characterized by the presence of absorption bands at 3187 and 1687 cm−1 due to the NH and CO groups, respectively. Also, in 1H NMR spectrum appeared three singlet signals at δ = 2.30, 2.62 and 2.87 ppm due to three methyl groups, as well as two other singlet signals that can be exchanged with D2O at δ = 10.94 and 12.40 ppm due to two NH protons. The mass spectrum showed a molecular ion peak at m/z = 348 (M+, 100%), corresponding to the molecular formula C18H16N6O2.. Compounds 9ad were also obtained by an alternative chemical route by condensing aryl hydrazo derivatives 10ad [24] with acetylacetone 3a under reflux conditions in DMF using piperidine as basic medium (Scheme 3).

Scheme 3
scheme 3

Synthetic route of arylhydrazonopyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine diones

Similarly, 4b coupled with arenadiazonium salts 8ad under the same reaction conditions to produce arylhydrazono derivatives 11ad (Scheme 4). The structures generated are supported by spectroscopic data (see exp.). Compounds 11ad were also obtained by condensation of each of compounds 10ad with dibenzoylmethane 3b, as shown in Scheme 4.

Scheme 4
scheme 4

Synthesis of arylhydrazono derivatives 11ad

On the other hand, cyclocondensation of compound 1 with β-Ketoesters 12a,b in glacial acetic acid upon reflux led to the formation of products 14a,b (Scheme 5), which were confirmed by spectroscopic tools. The IR spectrum of compound 14a was characterized by the presence of absorption bands at 3430, 3187 and 1689 cm−1 assigned to the OH, NH and CO groups, respectively. The 1H NMR spectrum also revealed a singlet signals assigned to methyl, methylene, pyrimidine-H, NH and OH protons at δ = 2.37, 3.95, 5.86, 10.96, and 12.80 ppm, respectively. The mass spectrum also exhibited a molecular ion peak at m/z = 232 = (M+, 100%), confirming that the molecular formula C10H8N4O3. The mechanism of formation of 14 is thought to occur initially by nucleophilic attack of the exocyclic amino group on 1 into the ketonic function of β-ketoesters 12a,b leading to the elimination of water molecule, followed by the intramolecular cyclization, followed by elimination of the ethanol molecule to obtain the enol structure 14 instead of the keto form 13 as shown in Scheme 5.

Scheme 5
scheme 5

Synthetic route for 4-hydroxy-2-substituted pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]-pyrimidine-8,10-diones 14a,b

Condensation of each of 14a,b with the suitable aromatic aldehyde 6ac in DMF under reflux conditions using a few drops of piperidine yielded the respective arylmethylene derivatives 15af (Scheme 6). The IR spectrum of 15a presented absorption bands at 3419, 3203 and 1704 cm−1 assigned to OH, NH and CO groups, respectively. Its 13C NMR chart revealed the characteristic signals at 19.38 (CH3), 152.58 (CO), 155.27 (CO), 160.24 (C=N) and 166.42 (=C–OH) in addition to other signals assigned for aromatic carbons (see exp.).

Scheme 6
scheme 6

Synthesis of arylmethylene derivatives 15af

The coupling reaction of compounds 14a,b with arenediazonium chlorides 8ad in DMF containing sodium acetate at 0–5 °C yielded the corresponding arylhydrazono derivatives 16ah. The structure of 16ah was determined by elemental analysis and spectral data. The IR spectrum of compound 16c was characterized by the presence of absorption bands at 3426, 3195 and 1702 cm−1 owing to the OH, NH and CO groups, respectively. Moreover, 1H NMR chart of compound 16c appeared two singlets at δ = 2.41 and 3.76 ppm due to methyl and methoxy protons along with three other singlet signals exchangeable with D2O in the region 11.19, 12.37 and 12.87 ppm due to three protons of 2NH and OH. The mass spectrum also showed a molecular ion peak at m/z = 366 (M+, 49.7%), which confirmed its molecular formula C17H14N6O4 (Scheme 7).

Scheme 7
scheme 7

Synthesis of arylhydrazono derivatives 16ah

Biological activity

Anticancer activity

Compounds 1, 4a,b, 7ac, 7k, l, 9ac, 14a, 15b, 16ac were selected to be investigated against three human cancer cell lines MCF7, HepG2 and HCT116 cell lines using MTT assay using doxorubicin as the standard drug. Each point is the mean ± SD (standard deviation) of three independent experiments performed in triplicate, using the prism software program (integrated Graphpad software, version 3). Cytotoxicity was assessed at concentrations of 5, 10 and 20 µg/l and the IC50 values of the tested compounds compared to the reference drug were evaluated as shown in Table 1, 2 and 3. In addition, the percentage of the viable cells was measured and compared with the control (Figs. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20). From the results presented in Table 1, compounds 7b and 16c strongest cytotoxic activity against MCF7 with IC50 = 3.864 and 3.805 µg/l, respectively, among the tested compounds compared to the doxorubicin (IC50 = 2.527 µg/l). Other compounds tested showed moderate to weak cytotoxic activity.

Table 1 The IC50 values (the drug concentrations that inhibited 50% of cell proliferation) of the compounds on MCF7 cell line
Table 2 The IC50 values (the drug concentrations that inhibited 50% of cell proliferation) of the compounds on HepG2 cell line
Table 3 The IC50 values (the drug concentrations that inhibited 50% of cell proliferation) of the compounds on HCT-116 cell line
Fig. 5
figure 5

IC50 values of 1

Fig. 6
figure 6

IC50 values of 4a

Fig. 7
figure 7

IC50 values of 4b

Fig. 8
figure 8

IC50 values of 7a

Fig. 9
figure 9

IC50 values of 7b

Fig. 10
figure 10

IC50 values of 7c

Fig. 11
figure 11

IC50 values of 7k

Fig. 12
figure 12

IC50 values of 7l

Fig. 13
figure 13

IC50 values of 9a

Fig. 14
figure 14

IC50 values of 9b

Fig. 15
figure 15

IC50 values of 9c

Fig. 16
figure 16

IC50 values of 14a

Fig. 17
figure 17

IC50 values of 15b

Fig. 18
figure 18

IC50 values of 16a

Fig. 19
figure 19

IC50 values of 16b

Fig. 20
figure 20

IC50 values of 16c

Furthermore, from screening the cytotoxic activity of the tested compounds against HepG2 cell line, we can infer that, compounds 7b, 15b, 16a and 16c showed higher potency against the HepG-2 cell line with IC50 = 4.250, 4.641, 3.555 and 3.427 µg/l, respectively compared to the reference drug (IC50 = 4.749 µg/l). The remaining tested compounds showed moderate to weak activity (Table 2).

Based on the results of the cytotoxic activity of the tested compounds against HCT116 (Table 3), compound 7b exhibited a higher cytotoxic activity against the HCT116 (IC50 = 2.487 µg/l) compared to the doxorubicin (IC50 = 3.641 µg/l). Additionally, compounds 7c, 16a and 16c exhibited high anticancer activity against HCT116 with IC50 values of 4.072, 4.369 and 4.503 µg/l, respectively. The other rested compounds showed moderate to low activity.

Structure–activity relationship (SAR)

The results obtained from the anticancer activity of some newly prepared compounds show the all tested compounds have antitumor activity against all the three cell lines (MCF7, HepG2 and HCT116) (Fig. 21).

Fig. 21
figure 21

Structure activity relationship (SAR) of some synthesized compounds

Initially, parent compound 1 exhibited a moderate cytotoxicity (IC50 = 7.818 µg/l) against the HepG2 cell line compared to doxorubicin (IC50 = 4.749 µg/l). When compound 1 was converted into a tricyclic ring system containing a pyrimidine ring as in compounds 4a,b, the anticancer activity varies depending on the nature of the substituents present on the pyrimidine ring. Therefore, when the substituent of compound 4 is a methyl group like 4a, the anticancer activity gradually increases towards HCT116 with an IC50 of 3.966 µg/l, equivalent to doxorubicin (IC50 = 3.641 µg/l). When adding another aryl group to 2nd position of compounds 4a,b, the anticancer activity changes depending on the position of the substituents on the aryl group. Thus, the anticancer activity does not change when the aryl group is a phenyl ring. However, when the methoxy group was introduced as a donor group in the aryl moiety as in 7b, the anticancer activity increased towards MCF7 (IC50 = 3.864 µg/l), HepG2 (IC50 = 4.250 µg/l) and HCT116 (IC50 = 2.487 µg/l) as shown in Tables 1, 2 and 3. On the other hand, when compounds 7a,b contain a chlorine atom on the aryl group as in the case of 7c, the anticancer activity was reduced in all three cell lines tested. Coupling of compounds 4a,b with arenediazonium salts afforded arylhydrazo derivatives 9ad which, have different cytotoxicities depending on the nature of the substituent on the arylhydrazo moiety. Thus, when the arylhydrazo was a phenyl or tolyl group, it had low anticancer activity against all three cell lines, whereas for the aryl moieties containing a methoxy group as donating group like 9c, the anticancer activity increased especially in the HCT116 cell line with IC50 = 3.778 µg/l. Furthermore, when compound 1 was condensed with a β-ketoester to form a tricyclic ring system containing a hydroxyl group as in 14a,b, it appeared to have weak anticancer activity. However, when compounds 14a,b have a methoxy group as in 15b, the anticancer activity was increased against HepG2 with IC50 = 4.641 µg/l compared to doxorubicin (IC50 = 4.749 µg/l). Also, when compounds 14a,b were coupled with arenediazonium salts to afford arylhydrazo derivatives 16ac, the anticancer activity was increased in the case of the arylhydrazo group with the methoxy group, as in the case of 16c (Tables 1, 2, and 3).

Enzyme inhibition assay

Compounds 7b, 9c, 15b, 16a and 16c with the strongest anticancer activity were tested for tropomyosin kinase A receptor inhibitory activity by a kinase assay technique utilizing Larotrectinib as a positive control. The data listed in Table 4 and Fig. 22 demonstrate that compound 16c has the strongest inhibitory effect among the tested compounds against to tropomyosin receptor kinase A (TrKA) with IC50 = 0.047 ± 0.0027 μg/ml compared to Larotrectinib with IC50 = 0.034 ± 0.0021 μg/ml using the HepG2 cancer cell line. While, compounds 7b and 16a have moderate activity anti-TrKA with IC50 = 0.064 ± 0.0037 and 0.072 ± 0.0042 μg/ml, respectively. In addition, compounds 9c and 15b have weak activity against TrKA with IC50 = 0.158 ± 0.0092 and 0.101 ± 0.0059 μg/ml, respectively. Therefore, compound 16c can cause cancer cell line death by inhibiting the enzyme tropomyosin receptor kinase A, possibly because it contains a methoxy group as donating group.

Table 4 Inhibitory activity of compounds 7b, 9c, 15b, 16a and 16c against tropomyosin receptor kinase A in vitro using kinase assay technique
Fig. 22
figure 22

Enzyme inhibition of tested compounds

Cell cycle analysis

For cell cycle analysis, stained DNA from HepG2 cancer cells was treated with compounds 7b, 15b, 16a and 16c that induce cancer cell death by inhibiting TrKA. From the results in Table 5, it can be seen that the proportion of cells at phase in the pre-G1 of compounds 7b, 15b, 16a and 16c increased the proportion of cells at phase in the G2/M by about 4, 4, 2.5 and 3 folds, respectively. Additionally, HepG2 cells were arrested in the cell cycle at G2/M phase by compounds 7b and 15b among the tested compounds 16a and 16c (Table 5 and Figs. 23, 24, 25, 26, 27, 28).

Table 5 Cell cycle analysis in HepG2 using compounds 7b, 15b, 16a and 16c
Fig. 23
figure 23

Cell cycle analysis of compounds 7b, 15b, 16a and 16c. Compounds 7b, 15b, 16a and 16c increased the ratio of cells at phases in the G2/M by about 4, 4, 2.5 and 3 times, respectively, and HepG2 cells were arrested in the cell cycle at G2/M phase by compounds 7b and 15b

Fig. 24
figure 24

Cell cycle of control HepG-2

Fig. 25
figure 25

Cell cycle of compound 7b

Fig. 26
figure 26

Cell cycle of compound 15b

Fig. 27
figure 27

Cell cycle of compound 16a

Fig. 28
figure 28

Cell cycle of compound 16c

Detection of apoptosis assay

Early and late apoptosis was determined after treatment of HepG2 cells with compounds 7b, 15b, 16a and 16c compared with untreated control cells. The late apoptosis rate increased by about 13, 20, 4 and 3 times, respectively, showing a higher efficiency than the early apoptosis ratio 5, 2, 8 and 6 times, respectively. Total apoptosis from treatment of HepG2 cells with compound 15b showed the higher apoptotic induction efficiency compared with other tested compounds 7b, 16a and 16c (Table 6 and Fig. 29).

Table 6 The effect of compounds 7b, 15b, 16a and 16c on HepG2 cell lines
Fig. 29
figure 29

Apoptosis and necrosis of tested compounds 7b, 15b, 16a, 16c with control

Molecular docking study

The most potent inhibitory compounds 7b, 16a and 16c as well as the standard drug Larotrectinib against TrKA were docked with the crystal structure of tropomyosin receptor kinase A (TrKA) (PDB: 5H3Q, Fig. 30) used the molecular operating environment docking (MOE) 2009 to find the exact binding pattern to the receptor. From the present studies, it was found that all the anchored compounds exhibited good binding energies ranging from − 7.3801 to − 6.5837 kcal mol−1 and displayed good fitness with the active site of the 5H3Q protein. Thus, the standard drug Larotrectinib exhibits two hydrogen bond interactions with bond length 2.99Ǻ and 3.06 Ǻ with amino acid residues Lys 544 and Asp 668, respectively and binding energy (S) = − 7.1325 kcal mol−1 (Fig. 31). Compound 7b appears to have a hydrogen bond interaction with a bond length 2.98 Ǻ between the carbonyl function of the pyridine moiety and the amino acid residue Lys 544 as well as a cation-cation interaction between the 4-methoxyphenyl group and His 489 with S = − 7.3801 kcal mol−1 (Fig. 32). On the other hand, compound 16a exhibits a binding energy of S = − 7.0296 kcal mol−1 and appears two hydrogen bond interactions with bond length equal to 3.12 and 3.46 Ǻ between the two carbonyl groups of each of pyridine and pyrimidine rings, respectively and the amino acid residues Lys 544 and Arg 673 (Fig. 33). Additionally, compound 16c the most potent inhibitory activity against TrKA exhibits two hydrogen bond interactions, one between the carbonyl group of the pyrindine ring and Met 507 with bond length of 3.52 Ǻ and the other between the carbonyl group of the pyrimidine ring and Asp 596 with bond length equal to 3.17 Ǻ, as well as a cation-cation bond interaction between pyrazole ring and Val 524 with S = − 7.4667 kcal mol−1 (Fig. 34). All data presented from the molecular docking study for larotrectinib, 7b, 16a, and 16c are listed in Table 7.

Fig. 30
figure 30

Interaction of 5H3Q with the active site in 2D and 3D

Fig. 31
figure 31

Interaction of larotrectinib with the active site of 5H3Q in 2D and 3D

Fig. 32
figure 32

Interaction of 7b with the active site of 5H3Q in 2D and 3D

Fig. 33
figure 33

Interaction of 16a with the active site of 5H3Q in 2D and 3D

Fig. 34
figure 34

Interaction of 16c with the active site of 5H3Q in 2D and 3D

Table 7 Interactions of compounds 7b, 16a, 16c, and Larotrectinib with 5H3Q enzyme

In silico ADME studies

In silico prediction of potential pharmacokinetic properties absorption, distribution, metabolism and excretion toxicity (ADME/T) properties calculated using Swiss ADME (http://www.swissadme.ch/) online tools are presented in Table 8. Some physical properties such as absorption, distribution, metabolism, excretion and toxicity are important for any oral drug. Lipinski`s rule of five (RO5), posits that the lipophilicity and solubility are more essential properties than other properties, and rule states that most “drug-like” molecules have log P ≤ 5, molecular weight ≤ 500, number of hydrogen bond acceptors ≤ 10, and number of hydrogen bond donors ≤ 5. Compounds that violate more than one of these rules may have bioavailability problems. According to this rule, the compounds 4a, 7ac, 9c, 15b, 16a, 16b and 16c have violated all parameters of Lipinski’s rule of five. The results listed in Table 9 show that all the compounds have TPSA values and compounds 4a, and 7ac have optimal topological polar surface area (TPSA) of 76.36, 76.36, 85.59, and 76.36 Ǻ2, respectively. This means that compounds 4a, 7a, 7b and 7c are better able of permeate cell membranes and adhere to RO5 and are well absorbed through the gastrointestinal tract. In silico predictions of toxicological properties were determined using the Osiris property explorer program (http://www.prperty explorer-cheminfo.org) online tools is presented in Table 9. In the toxicological profile of the compounds, 9c and 16c may exhibit medium tumorigenicity, but compounds 7b, 9c, 15b, 16a and 16c are high risk in the reproductive system is expected. Additionally, all compounds have no irritant effects. Compounds 4a, 7a, and 7c did not cause the toxicity problems mentioned in the software used in this study. All of the compounds studied have positive drug-likeness values, meaning that they all contained fragments commonly found in commercial drugs (Table 9).

Table 8 Important pharmacokinetic parameters for bioavailability of compounds 4a, 7a, 7b, 7c, 9c, 15b, 16a, 16b and 16c
Table 9 Important toxicity predication of compounds 4a, 7a, 7b, 7c, 9c, 15b, 16a, 16b and 16c

Swissadme helps us provide information on poorly and highly absorbed drugs to model passive intestinal absorption through the human intestinal tract. Graphical prediction of intestinal absorption and blood–brain barrier permeation of the most potent anticancer activity compounds 4a, 7a, 7b, 7c, 9c, 15b, 16a, 16b and 16c against MCF7, HepG2 and HCT116 are shown in Fig. 35. Boiled egg diagram showing the bioavailability property space for wlog P and TPSA [white area means that intestinal absorption; The yellow area means it has entered the brain well, the intestinal are well absorbed; and the gray area means the intestinal have poor absorption]. This provides a simple visual cue to profile new compounds for their oral absorption. All compounds studied were found in the white region. Additionally, PGP+ (substrate) and PGP (non-substrate) are denoted by blue and red dots for molecules predicated to be CNS efflux or not efflux by P-glycoprotein, respectively. Therefore, all the studied compounds 4a, 7a, 7b, 7c, 9c, 15b, 16a, 16b and 16c are not substrates of P-gp (PGP-), hence, we can say that these compounds have good bioavailability. In this series, compound 7c gave BBB and a low TPSA of 76.36. This suggests that the molecule can be absorbed very easily through the gastrointestinal tract and preferentially acts as a hydrophobic agent and can be easily transported across the blood–brain barrier.

Fig. 35
figure 35

Boiled-egg depicts gastrointestinal absorption and brain penetration of compounds 4a, 7ac, 9c, 15b and 16ac

Conclusion

In this study, a novel series of pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidine derivatives were synthesized. The anticancer activity of these compounds was tested on MCF7, HepG2 and HCT116 cell lines in comparison to doxorubicin. The results showed that some of the synthesized compounds have significant cytotoxic activity. Compound 7b exhibited high and broad spectrum anticancer activity against all cell lines tested. TrKA inhibition assays on 7b, 9c, 15b, 16a and 16c showed a decrease in TrKA expression with IC50 values below 0.2 μg/ml. The most potent anticancer targets were examined for their effects on cell cycle distribution and apoptosis induction. The results revealed that 7b and 15b induced arrest at the G2/M phase of the cell cycle in HepG2 cells among the other tested compounds. Furthermore, docking studies revealed that 7b, 16a and 16c bind with high affinity to the active site of TrKA. In addition, compounds 7b, 15b, 16a and 16c appear to be well absorbed from the gastrointestinal tract. These results suggest that these compounds may be a promising tools for the production of more potent anticancer agents.

Availability of data and materials

The datasets used and/or analyzed during the present study available from the electronic additional file.

References

  1. Amatu A, Sartore-Bianchi A, Bencardino K, Pizzutilo GE, Tosi F, Siena S. Tropomyosin receptor kinase (TRK) biology and the role of NTRK gene fusions in cancer. Ann Oncol. 2019;30:viii5–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hechtman JF. NTRK insights: best practices for pathologists. Mod Pathol. 2022;35:298–305.

    Article  PubMed  Google Scholar 

  3. Jin W. Cancers. Roles of TrkC signaling in the regulation of tumorigenicity and metastasis of cancer. Cancers. 2020;12:147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yakes FM, Chen J, Tan J, Yamaguchi K, Shi Y, Yu P, Qian F, Chu F, Bentzien F, Cancilla B, Orf J, You A, Laird AD, Engst S, Lee L, Lesch J, Chou YC, Joly AH. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther. 2011;10:2298–308.

    Article  CAS  PubMed  Google Scholar 

  5. Patwardhan PP, Ivy KS, Musi E, de Stanchina E, Schwartz GK. Significant blockade of multiple receptor tyrosine kinases by MGCD516 (Sitravatinib), a novel small molecule inhibitor, shows potent anti-tumor activity in preclinical models of sarcoma. Oncotarget. 2016;7:4093–109.

    Article  PubMed  Google Scholar 

  6. Smith BD, Kaufman MD, Leary CB, Turner BA, Wise SC, Ahn YM, Booth RJ, Caldwell TM, Ensinger CL, Hood MM, Lu WP, Patt TW, Patt WC, Rutkoski TJ, Samarakoon T, Telikepalli H, Vogeti L, Vogeti S, Yates KM, Chun L, Stewart LJ, Clare M, Flynn DL. Altiratinib inhibits tumor growth, invasion, angiogenesis, and microenvironment-mediated drug resistance via balanced inhibition of MET, TIE2, and VEGFR2. Mol Cancer Ther. 2015;14:2023–34.

    Article  CAS  PubMed  Google Scholar 

  7. Smith BD, Leary CB, Turner BA, Kaufman MD, Wise SC, Rendueles MERG, Fagin JA, Flynn DL. Altiratinib is a potent inhibitor of TRK kinases and is efficacious in TRK-fusion driven cancer models. Cancer Res. 2015;75:790.

    Article  Google Scholar 

  8. Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy Nat. Rev Clin Oncol. 2018;75:731–47.

    Article  Google Scholar 

  9. Drilon A, Nagasubramanian R, Blake JF, Ku N, Tuch BB, Ebata K, Smith S, Lauriault V, Kolakowski GR, Brandhuber BJ, Larsen PD, Bouhana KS, Winski SL, Hamor R, Wu WI, Parker A, Morales TH, Sullivan FX, DeWolf WE, Wollenberg LA, Gordon PR, Douglas-Lindsay DN, Scaltriti M, Benayed R, Raj S, Hanusch B, Schram AM, Jonsson P, Berger MF, Hechtman JF, Taylor BS, Andrews S, Rothenberg SM, Hyman DM. A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion–positive solid tumors. Cancer Discov. 2017;7:963–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yan W, Lakkaniga NR, Carlomagno F, Santoro M, McDonald NQ, Lv F, Gunaganti N, Frett B, Li HY. Insights into current tropomyosin receptor kinase (TRK) inhibitors: development and clinical application. J Med Chem. 2019;62:1731–60.

    Article  CAS  PubMed  Google Scholar 

  11. Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 2015;5:25–34.

    Article  CAS  PubMed  Google Scholar 

  12. Ricciuti B, Genova C, Crino L, Libra M, Leonardi GC. Antitumor activity of larotrectinib in tumors harboring NTRK gene fusions: a short review on the current evidence. Onco Targets and Therapy. 2019;12:3171–9.

    Article  CAS  Google Scholar 

  13. FDA.gov (2018). FDA approves larotrectinib for solid tumors with NTRK gene fusions. http://www.fda.gov/drug/fda-approves-larotrectinib-solid-tumors-ntrk-gene-fusions.

  14. Berger S, Martens UM, Bochum S. In small molecules in oncology. Berlin: Springer; 2019. p. 142–50.

    Google Scholar 

  15. FDA.gov (2019). FDA approves entrectinib for NTRK solid tumors and ROS-1 NSCLC. http://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsxlc.

  16. Dar AC, Shokat KM. The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annu Rev Biochem. 2011;80:769–95.

    Article  CAS  PubMed  Google Scholar 

  17. Gouda MA, Berghot MA, Shoeib AI, Khalil AM. Synthesis and antimicrobial of new anthraquinone derivatives incorporating pyrazole moiety. Eur J Med Chem. 2010;45:1843–8.

    Article  CAS  PubMed  Google Scholar 

  18. Gopalsamy A, Yang H, Ellingboe JW, Tsou HR, Zhang N, Honores E, Powell D, Miranda M, McGinnis JP, Rabindran SK. Pyrazolo[1,5-a]pyrimidin-7-yl phenyl amides as novel antiproliferative agents: Exploration of core and headpiece structure–activity relationships. Bioorg Med Chem Lett. 2005;15:1591–4.

    Article  CAS  PubMed  Google Scholar 

  19. Ahmed OM, Mohamed MA, Ahmed RR, Ahmed SA. Synthesis and antitumor activities of some new pyridines and pyrazolo[1,5-a]pyrimidines. Eur J Med Chem. 2009;44:3519–23.

    Article  CAS  PubMed  Google Scholar 

  20. Hassan AS, Hafez TS, Osman SAM, Ali MM. Synthesis and in vitro cytotoxic activity of novel pyrazolo[1,5-a]pyrimidines and related Schiff bases Turk. J Chem. 2015;39:1102–13.

    CAS  Google Scholar 

  21. Paruch K, Dwyer MP, Alvarez C, Brown C, Chen TY, Doll RJ, Keertikar K, Knutson C, McKittrick B, Rivera J, Rossman R, Tucker G, Fischmann T, Hruza A, Madison V, Nomeir AA, Wang Y, Kirschmeier P, Lees E, Parry D, Sqambellone N, Seqhezzi W, Schultz L, Shanahan F, Wiswell D, Xu X, Zhou Q, James RA, Paradkar VM, Park H, Rokosz LR, Stauffer TM, Guzi T. Discovery of dinaciclib (SCH 727965): a potent and selective inhibitor of cyclin-dependent kinases. ACS Med Chem Lett. 2010;1:204–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Metwally NH, Badawy MA, Okpy DS. Synthesis and anticancer activity of some new thiopyrano[2,3-d]thiazoles incorporating pyrazole moiety Chem. Pharm Bull. 2015;63:495–503.

    Article  Google Scholar 

  23. Metwally NH, Abdelrazek FM, Eldaly SM. Synthesis and anticancer activity of some new heterocyclic compounds based on 1-cyanoacetyl-3,5-dimethylpyrazole Res. Chem Intermed. 2016;42:1071–89.

    Article  CAS  Google Scholar 

  24. Metwally NH, Dee EA. Synthesis, assessment on human breast, liver and colon cell lines and molecular modeling study using novel pyrazolo[4,3-c]pyridine derivatives. Bioorg Chem. 2018;77:203–14.

    Article  CAS  PubMed  Google Scholar 

  25. Metwally NH, Abdelrazek FM, Eldaly SM. Synthesis, molecular docking, and biological evaluation of some novel bis-heterocyclic compounds based N, N-([1,1`biphenyldiyl])bis(2-cyanoacetamide) as potential anticancer agents. J Heter Chem. 2018;55:2668–82.

    Article  CAS  Google Scholar 

  26. Metwally NH, Radwan IT, El-Serwy WS, Mohamed MA. Design, synthesis, DNA assessment and molecular docking study of novel 2-(pyridine-2-ylimino)thiazolidin-4-one derivatives as potent antifungal agents. Bioorg Chem. 2019;84:456–67.

    Article  CAS  PubMed  Google Scholar 

  27. Metwally NH, Mohamed SM, Ragb EA. Design, synthesis, anticancer evaluation, molecular docking and cell cycleanalysis of 3-methyl-4,7-dihydropyrazolo[1,5-a]pyrimidine derivatives as potent histone lysine demethylases (KDM) inhibitors and apoptosis inducers. Bioorg Chem. 2019;88: 102929.

    Article  CAS  PubMed  Google Scholar 

  28. Metwally NH, Mohamed MS. New imidazolone derivatives comprising a benzoate or sulfonamide moiety as anti-inflammatory and antibacterial inhibitors: design, synthesis, selective COX-2. DHFR and molecular-modeling study Bioorg Chem. 2019;99: 103438.

    PubMed  Google Scholar 

  29. Metwally NH, Abdallah SO, Abdel Mohsen MM. Design, green one-pot synthesis and molecular docking study of novel N, N-bis(cyanoacetyl)hydrazines and bis-coumarins as effective inhibitors of DNA gyrase and topoisomerase IV. Bioorg Chem. 2020;97: 103672.

    Article  CAS  PubMed  Google Scholar 

  30. Metwally NH, Mohamed MS, Deeb EA. Synthesis, anticancer evaluation, DK2 inhibition, and apoptotic activity assessment with molecular docking modeling of new class of pyrazolo[1,5-a]pyrimidines. Res Chem Intermed. 2021;47:5027–60.

    Article  CAS  Google Scholar 

  31. Metwally NH, Abd-Elmoety AS. Novel fluorinated pyrazolo[1,5-a]pyrimidines: In a way from synthesis and docking studies to biological evaluation. J Mol Struct. 2022;1257:132590.

    Article  CAS  Google Scholar 

  32. Metwally NH, Badawy MA, Okpy DS. Synthesis, biological evaluation of novel thiopyrano[2,3-d]thiazoles incorporating arylsulfonate moiety as potential inhibitors of tubulin polymerization, and molecular modeling studiesJ Mol. Struct. 2022;1258: 132848.

    Google Scholar 

  33. Metwally NH, El-Desoky EA. Novel thiopyrano[2,3-d]thiazole-pyrazole hybrids as potential nonsulfonamide human carbonic anhydrase IX and XII inhibitors: design, synthesis, and biochemical studies. ACS Omega, 2023 in press.

  34. Sato T. Reaction of hydrazine hydrate and phenylhydrazine with malononitrile. J Org Chem. 1959;24:963–6.

    Article  CAS  Google Scholar 

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

    CAS  Google Scholar 

  36. Denizot F, Lang R, Immunol J. Rapid colorimetric assay for cell growth and survival. Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. Methods. 1986;22:271–7.

    Google Scholar 

  37. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches toestimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 1997;23:3–25.

    Article  CAS  Google Scholar 

  38. Daina A, Zoete V. A BOILED-Egg to predict gastrointestinal absorption and brain penetration of small molecules. Chem Med Chem. 2016;11:1117–21.

    Article  CAS  PubMed  Google Scholar 

  39. Metwally NH, Deeb EA. Aminopyrazolo[4,3-c]pyridine-4,6-dione as a precursor for novel pyrazolo[4,5,1-ij][1,6]naphtyridines and pyrido[4`,3`:3,4]pyrazolo[1,5-a]. Synth Commun. 2018;48:1614–28.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB). No Funding.

Author information

Authors and Affiliations

Authors

Contributions

Nadia Hanafy Metwally has generally supervised the work and has provides the conceptions, follow the data interpretations, original manuscript writing and reviewing process handling. Emad Abdullah Deeb has performed the experimental of chemistry work, carried out the analysis and manuscript writing. Ibrahim Walid Hasani has collected the data of anticancer work and data interpretation.

Corresponding author

Correspondence to Nadia Hanafy Metwally.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Figure S1.

Mass spectrum of compound 2. Figure S2. IR spectrum of compound 2. Figure S3. 1H NMR spectrum of compound 2. Figure S4. Mass spectrum of compound 4a. Figure S5. IR spectrum of compound 4a. Figure S6. 1H NMR spectrum of compound 4a. Figure S7. 13C NMR spectrum of compound 4a. Figure S8. Mass spectrum of compound 4b. Figure S9. 1H NMR spectrum of compound 4b. Figure S10. 1H NMR spectrum of compound 7a. Figure S11. 13C NMR spectrum of compound 7a. Figure S12. 1H NMR spectrum of compound 7b. Figure S13. 1H NMR spectrum of compound 7f. Figure S14. 13C NMR spectrum of compound 7f. Figure S15. IR spectrum of compound 7g. Figure S16. 1H NMR spectrum of compound 7g. Figure S17. 1H NMR spectrum of compound 7h. Figure S18. IR spectrum of compound 7i. Figure S19. 1HNMR spectrum of compound 7i. Figure S20. IR spectrum of compound 7j. Figure S21. 1H NMR spectrum of compound 7k. Figure S22. 13C NMR spectrum of compound 7k. Figure S23. 1HNMR spectrum of compound 7m. Figure S24. IR spectrum of compound 7n. Figure S25. 1HNMR spectrum of compound 7n. Figure S26. IR spectrum of compound 7o. Figure S27. 1HNMR spectrum of compound 7o. Figure S28. IR spectrum of compound 7p. Figure S29. 1HNMR spectrum of compound 7p. Figure S30. 13C NMR spectrum of compound 7q. Figure S31. 1H NMR spectrum of compound 7r. Figure S32. 1H NMR spectrum of compound 7s. Figure S33. 1H NMR spectrum of compound 7t. Figure S34. IR spectrum of compound 9a. Figure S35. IR spectrum of compound 9b. Figure S36. Mass spectrum of compound 9b. Figure S37. MS spectrum of compound 9c. Figure S38. IR spectrum of compound 9d. Figure S39. IR spectrum of compound 11a. Figure S40. 1H NMR spectrum of compound 11a. Figure S41. 1H NMR spectrum of compound 11b. Figure S42. 1H NMR spectrum of compound 11c. Figure S43. Mass spectrum of compound 11d. Figure S44. Mass spectrum of compound 14a. Figure S45. IR spectrum of compound 14a. Figure S46. Mass spectrum of compound 14b. Figure S47. IR spectrum of compound 14b. Figure S48. 1H NMR spectrum of compound 14b. Figure S49. 1H NMR spectrum of compound 14b (D2O). Figure S50. 13C NMR spectrum of compound 15a. Figure S51. 1H NMR spectrum of compound 15b. Figure S52. IR spectrum of compound 15c. Figure S53. 1H NMR spectrum of compound 15c. Figure S54. 13C NMR spectrum of compound 15e. Figure S55. 1H NMR spectrum of compound 15f. Figure S56. MS spectrum of compound 16a. Figure S57. IR spectrum of compound 16b. Figure S58. 1H NMR spectrum of compound 16b. Figure S59. MS spectrum of compound 16c. Figure S60. IR spectrum of compound 16d. Figure S61. 1H NMR spectrum of compound 16d. Figure S62. IR spectrum of compound 16f. Figure S63. 13C NMR spectrum of compound 16g. Biological methods for MTT assay, Tropomyosm receptor kinase A inhibitory assay and Annexin-VFITC apoptosis assay were discussed in details in ESI.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Metwally, N.H., Deeb, E.A. & Hasani, I.W. Synthesis, anticancer evaluation, molecular docking and ADME study of novel pyrido[4ʹ,3ʹ:3,4]pyrazolo[1,5-a]pyrimidines as potential tropomyosin receptor kinase A (TrKA) inhibitors. BMC Chemistry 18, 68 (2024). https://doi.org/10.1186/s13065-024-01166-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13065-024-01166-7

Keywords