Next Article in Journal
Correction: Hashemi et al. Effects of Abiotic Elicitors on Expression and Accumulation of Three Candidate Benzophenanthridine Alkaloids in Cultured Greater Celandine Cells. Molecules 2021, 26, 1395
Next Article in Special Issue
[1,5]-Hydride Shift Triggered N-Dealkylative Cyclization into 2-Oxo-1,2,3,4-tetrahydroquinoline-3-carboxylates via Boronate Complexes
Previous Article in Journal
Identification of Hair Growth Promoting Components in the Kernels of Prunus mira Koehne and Their Mechanism of Action
Previous Article in Special Issue
Recent Strategies in Transition-Metal-Catalyzed Sequential C–H Activation/Annulation for One-Step Construction of Functionalized Indazole Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

CK2 Inhibition and Antitumor Activity of 4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines

by
Daniil N. Lyapustin
1,
Svetlana K. Kotovskaya
1,
Ilya I. Butorin
1,
Evgeny N. Ulomsky
1,
Vladimir L. Rusinov
1,*,
Denis A. Babkov
2,
Alexander A. Pokhlebin
2,
Alexander A. Spasov
2,
Vsevolod V. Melekhin
1,3,
Maria D. Tokhtueva
1,
Anna V. Shcheglova
1,3 and
Oleg G. Makeev
3
1
Department of Organic and Biomolecular Chemistry, Ural Federal University, Mira St. 19, 620002 Ekaterinburg, Russia
2
Department of Pharmacology & Bioinformatics, Scientific Center for Innovative Drugs, Volgograd State Medical University, 400131 Volgograd, Russia
3
Department of Biology, Ural State Medical University, Repina 3, 620014 Yekaterinburg, Russia
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(16), 5239; https://doi.org/10.3390/molecules27165239
Submission received: 8 July 2022 / Revised: 4 August 2022 / Accepted: 12 August 2022 / Published: 17 August 2022

Abstract

:
Today, cancer is one of the most widespread and dangerous human diseases with a high mortality rate. Nevertheless, the search and application of new low-toxic and effective drugs, combined with the timely diagnosis of diseases, makes it possible to cure most types of tumors at an early stage. In this work, the range of new polysubstituted 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines was extended. The structure of all the obtained compounds was confirmed by the data of 1H, 13C NMR spectroscopy, IR spectroscopy, and elemental analysis. These compounds were evaluated against human recombinant CK2 using the ADP-GloTM assay. In addition, the IC50 parameters were calculated based on the results of the MTT test against glioblastoma (A-172), embryonic rhabdomyosarcoma (Rd), osteosarcoma (Hos), and human embryonic kidney (Hek-293) cells. Compounds 5f, 5h, and 5k showed a CK2 inhibitory activity close to the reference molecule (staurosporine). The most potential compound in the MTT test was 5m with an IC50 from 13 to 27 µM. Thus, our results demonstrate that 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines are promising for further investigation of their antitumor properties.

Graphical Abstract

1. Introduction

Cancer is one of the world′s leading causes of death, with an estimated number of 10 million deaths in 2020 [1]. However, many types of cancer are curable with early diagnosis and treatment. The establishment of alternative ways to treat tumor diseases allows for the use of new effective and low-toxic drugs in the early stages of the disease. One of the current trends is the inhibition of biological targets responsible for the growth, proliferation, and survival of tumor cells. From this point of view, type 2 casein kinase is a promising target for chemotherapy. The overexpression of casein kinase 2 (CK2) is closely associated with several cancers, including cancers of the head and neck, breast, kidney, lung, etc. [2,3,4,5,6,7,8,9], thus making CK2 a promising target for chemotherapy [10,11,12,13,14]. The ATP binding site of CK2 is smaller than that of most other kinases due to the presence of unique bulky residues, such as Val66 and Ile174, which create the prerequisites for the development of selective small molecule ATP-competitive inhibitors [15,16]. In the review article of CK2 and its inhibitors [17] by Iegre and colleagues, compounds of the azolo[1,5-a]pyrimidine series are noted as one of the most significant types of inhibitors over the past decade [18,19], along with azole derivatives(Figure 1) [20,21].
The antitumor activity of azolo[1,5-a]pyrimidines [22,23,24,25,26,27] has been related to the inhibition of cancer-associated kinases [28,29] (cyclin-dependent kinase 2 and phosphoinositide-3-kinase). However, recent Safari’s work demonstrates a positive trend in the cytotoxic effect of nitro-containing azolo[1,5-a]pyrimidines against human malignant melanoma cells (A375) and prostate cancer (PC3 cells, LNCaP cells) [30]. Examples of azolo[1,5-a]pyrimidines exhibiting antitumor activities are shown in Figure 2.
To continue our research on polysubstituted 6-nitroazolo[1,5-a]pyrimidines [31,32,33], we would like to present the synthesis of new compounds of this series, as well as their inhibitory activity against protein kinase CK2 and their cytotoxic effect against cultured tumor cells of human glioblastoma (A-172, ATCC CRL 1620), embryonic rhabdomyosarcoma (Rd, ATCC CRL 136), human osteosarcoma (Hos, ATCC CRL 1543), and human embryonic kidney (HEK-293).

2. Results and Discussion

2.1. Synthesis

In the present work, we studied compounds of the 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidine 5a-o and 6a-e series. These compounds were obtained by a multicomponent reaction between aminoazoles 1,2, 1-morpholino-2-nitroalkenes 3, and aldehydes 4 (Scheme 1). It was shown [31] that an initial reaction occurs between 1-morpholino-2-nitroalkenes 3 and aminoazoles 1,2, followed by heterocyclization to products 5, and the interaction of boron trifluoride etherate with the morpholinenitroalkene 3 leads to the formation of a corresponding alkyne and morpholinium tetrafluoroborate. The structure of all the obtained products 5,6 was confirmed by the data of 1H, 13C NMR spectroscopy, IR spectroscopy, and elemental analysis. The signals H-7 and C-7 are the characteristic for products 5,6 in the corresponding NMR spectra. It is interesting to note that in compounds 5a-d obtained from 3-aminopyrazole 3a, in the 1H spectra, the H-7 signal is in the region of 5.43–5.84 ppm, while in the 13C spectra, the characteristic C-7 signal is in the region of 34–40 ppm. In all other structures 5e-o, 6a-d, these signals are shifted to a weaker region of the spectrum in the region of 6.44–6.94 and 55–60 ppm, respectively (see Supplementary Materials). Apparently, the substituent and heteroatom in the azole ring affect the position of these signals.

2.2. CK2 Inhibition

Once in hand, target compounds were evaluated against human recombinant CK2 using the ADP-GloTM assay (Table 1). Initial screening at 50 μM revealed that compounds 5a, 5c, 5g, 5m, 5o, 6a, 6c, and 6d paradoxically enhance CK2 activity. Moderate inhibition was demonstrated by compounds 5f, 5h, 5k, 5l, and 6e. Derivatives 5f, 5h, and 5k were the most active inhibitors. One can notice that compounds bearing alkyl or alkylthio substituents at position C-2 and at position C-6 simultaneously tend to be more active, though the high structural similarity in this series does not allow us to define more comprehensive SAR. A dose–response study confirmed that compounds 5f, 5h, 5k, and 5l are micromolar CK2 inhibitors, while 6e has a low potency (Table 2). A hill coefficient around (−1) indicates that lead compounds as well as staurosporine behave like classical inhibitors that bind to a single kinase site.

2.3. Antitumor Activity

The IC50 parameters were calculated based on the results of the MTT test (Table 3). The values are defined in the range from 13 µM to >650 µM. It should be noted that the study of the 5a, 5b, and 5c cytotoxic effects was limited to exclude because of their low solubility.
Compounds 5j and 6a possessed the least pronounced cytotoxic effect on cells (in all cases IC50 > 0.4 mM), while the greatest decrease in the viability of tumor cells was noted with the addition of compounds 5m, 5o, 6c, and 6d. It is important to note that these azolopyrimidine compounds are characterized by a more pronounced suppression of the viability of tumor cells A-172, Rd, and Hos in comparison with the effect on human embryonic kidney cells Hek-293 (Figure 3 and Figure 4).
It was found that the IC50 for compounds 5m, 5o, 6c, and 6d in tumor cell lines studies, mostly, did not exceed 50 µM, whereas the cytotoxicity index on embryonic cells was higher than 169 µM (5m, 5o, and 6c). It should be noted that compounds 6d containing a triazole fragment with a CF3-substituent in the structure had similar micromolar IC50 values for kidney cells and tumor cells. The least cytotoxic effect on non-tumor cells was determined for compound 6c (227.50 µM). At the same time, our results indicate that compound 5m may have the most pronounced antitumor properties.
Meanwhile, the mechanisms for the suppression of cultured cells growth remain unclear and require further research. We can assume, that experimental data of cytotoxic action are not fully explained by the effect on CK2. On the one hand, it was noted that azolo[1,5-a]pyrimidines 5k, 5l, and 6e inhibit both the enzymatic activity of CK2 and the viability of tumor cells. On the other hand, compounds 5m, 5o, 6c, and 6d significantly inhibit the growth of neoplastic cells without affecting CK2. We can assume that the cytotoxic effect of the synthesized compounds may be due to the effect on other intracellular targets.

3. Materials and Methods

3.1. Chemical Experiment

Unless stated otherwise, all solvents and commercially available reactants/reagents were used as received. Non-commercial starting materials were prepared as described below or according to literature procedures. One-dimensional 1H and 13C NMR spectra, as well as two-dimensional 1H–13C HMBC experiments were acquired on a Bruker DRX-400 instrument (400 and 101 MHz, respectively) or a Bruker Avance NEO 600 instrument (600 and 151 MHz, respectively), equipped with a Prodigy broadband gradient cryoprobe, utilizing DMSO-d6 as solvent and TMS as internal standard. IR spectra were recorded on a Bruker Alpha FTIR spectrometer equipped with a ZnSe ATR accessory. Elemental analysis was performed on a PerkinElmer 2400 CHN analyzer. The reaction progress was controlled by TLC on Silufol UV-254 plates, eluent—EtOAc. Melting points were determined on a Stuart SMP3 apparatus at the heating rate of 7 °C/min. 1-Morpholino-2-nitroethylenes 3 were prepared according to a literature procedure [34].
4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines 5,6; General procedure 1.
A total of 3 Mmol (1.5 equiv., 0.37 mL) of BF3·Et2O was added to a suspension 2 mmol (1.0 equiv.) of corresponding aminoazole 1,2, 2 mmol (1.0 equiv.) of nitroalkene 3, and 2 mmol (1.0 equiv.) of aldehyde 4 in 5 mL n-BuOH. The reaction mixture was heated on oil bath at 120 °C for 2 h. The resulting solution was cooled to room temperature and stirred 15 min. The obtained precipitate was filtered off, washed with 15 mL of i-PrOH. The precipitate was suspended in 50 mL of water, stirred for 5 min, filtered off again, and washed with 15 mL of water.
4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines 5,6; General procedure 2.
A total of 3 Mmol (1.5 equiv., 0.37 mL) of BF3·Et2O was added to a suspension 2 mmol (1.0 equiv.) of corresponding aminoazole 1,2, 2 mmol (1.0 equiv.) of nitroalkene 3, and 2 mmol (1.0 equiv.) of aldehyde 4 in 5 mL n-BuOH. The reaction mixture was heated on oil bath at 120 °C for 2 h. After heating, the resulting solution was concentrated under reduced pressure. To the residue, 20 mL of 2M Na2CO3 and 50 mL of water was added and stirred for 20 min. Solution was extracted twice with 20 mL of EtOAc. To a water phase, 15 mL of hexane was added and mixture was neutralized by diluted HCl to pH 7. Resulting mixture was stirred for 30 min, filtered off, and washed with water.
6-Nitro-7-phenyl-4,7-dihydropyrazolo[1,5-a]pyrimidine (5a). The reaction was performed according to the general procedure 1 employing 0.166 g (2 mmol, 1 equiv.) of 3-aminopyrazole 1a, 0.316 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitroethylene 3a, and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. The product was recrystallized from DMF. The substance was dried over P2O5 at 170 °C. Yellow solid. Yield 0.387 g (80%). mp 295–297 °C. IR Spectrum,ν cm−1: 1528, 1417 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 5.43 (1H, s, H-7); 7.10–7.30 (5H, m, Ph); 7.41 (1H, s, H-2); 8.36 (1H, d, H-5, J = 6.1 Hz); 10.88 (1H, d, NH, J = 6.4 Hz); 12.45 (1H, s, H-3). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 38.2; 106.8; 124.3; 126.2; 126.5; 128.4; 127.3; 139.0; 144.2; 146.0. Anal. Calcd. for C12H10N4O2: C, 59.50; H, 4.16; N, 23.13. Found: C, 59.61; H, 4.20; N, 23.01.
7-(Anthracen-9-yl)-5-ethyl-7-nitro-4,7-dihydropyrazolo[1,5-a]pyrimidine (5b). The reaction was performed according to the general procedure 1 employing 0.166 g (2 mmol, 1 equiv.) of 3-aminopyrazole 1a, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitroethylene 3a and 0.412 g (2 mmol, 1 equiv.) of 9-anthracenecarbaldehyde 4b. The product was recrystallized from n-BuOH. The substance was dried over P2O5 at 170 °C. Yellow solid. Yield 0.385 g (53%). mp 215 °C with decomp. IR Spectrum, ν cm−1: 1518, 1277 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.38 (3H, t, CH2-CH3, J = 7.3 Hz); 2.73–2.84, 3.16–3.26 (2H, m, CH2-CH3); 8.40 (1H, s, H-2); 6.88 (1H, s, H-7); 7.08 (1H, s, H-9ʹ); 7.26 (1H, m, H-7ʹ); 7.35 (1H, m, H-2ʹ); 7.52 (2H, m, H-6ʹ); 7.60 (2H, m, H-3ʹ); 7.98 (1H, d, H-5ʹ, J = 8.4 Hz); 8.06 (1H, d, H-4ʹ, J = 8.4 Hz); 8.11 (1H, d, H-8ʹ, J = 9.1 Hz); 8.40 (1H, s, H-2); 8.71 (1H, d, H-1ʹ, J = 9.1 Hz); 13C {1H} NMR (101 MHz, DMSO-d6): δ = 13.5; 28.0; 34.8; 106.7; 124.6 (2C); 124.9; 125.0; 125.2; 125.5; 126.8; 126.9; 127.1; 128.7; 129.4; 129.9; 130.4; 131.5; 132.2; 137.5; 145.1; 155.6. Anal. Calcd. for C22H18N4O2: C, 71.34; H, 4.90; N, 15.13. Found: C, 71.52 H, 4.72; N, 14.99.
6-Nitro-5-methyl-7-(4ʹ-nitrophenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5c). A total of 3 Mmol (1.5 equiv., 0.37 mL) of BF3·Et2O was added to a suspension of 0.166 g (2 mmol, 1 equiv.) of 3-aminopyrazole 1a and 0.344 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitropropylene 3b in 5 mL n-BuOH. The reaction mixture was heated on oil bath at 80 °C for 15 min. After this, 0.302 g (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c was added to the obtained solution. The reaction mixture was heated on oil bath at 120 °C for 2 h. The resulting solution was cooled to room temperature and stirred for 15 min. The obtained precipitate was filtered off, washed with 15 mL of i-PrOH. The precipitate was suspended in 50 mL of water, stirred for 5 min, filtered off again and washed with 15 mL of water. To the residue, 20 mL of 2 M Na2CO3 and 50 mL of water were added and stirred for 20 min. The solution was extracted twice with 20 mL of EtOAc. To the water phase, 15 mL of hexane was added, and the mixture was neutralized by diluted HCl to pH 7. The resulting mixture was stirred overnight, filtered off, and washed with water. Yellow solid. Yield 0.355 g (59%). mp 198 °C with decomp. IR Spectrum, ν cm−1: 1535, 1352 (NO2); 1508, 1268 (NO2). 1H NMR (600 MHz, DMSO-d6): δ = 2.66 (3H, s, C-5-CH3); 5.64 (1H, s, H-5); 7.45 (1H, s, H-2); 7.50 (2H, d, H-2ʹ, J = 8.3 Hz); 8.12 (2H, d, H-3ʹ, J = 8.3 Hz); 10.95 (1H, s, NH); 12.49 (1H, s, H-3). 13C {1H} NMR (151 MHz, DMSO-d6): δ = 22.0; 39.8; 105.6; 121.8; 123.8; 127.0; 127.6; 144.2; 145.9; 152.5; 154.1. Anal. Calcd. for C13H11N5O4: C, 51.83; H, 3.68; N, 23.25 Found: C, 51.89; H, 3.73; N, 23.19.
6-Nitro-5-methyl-7-(thiophen-2ʹ-yl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5d). The reaction was performed according to the general procedure 1 employing 0.166 g (2 mmol, 1 equiv.) of 3-aminopyrazole 1a, 0.344 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitropropylene 2b and 0.184 mL (2 mmol, 1 equiv.) of thiophen-2-carbaldehyde 4f. Pale green solid. Yield 0.278 (53%). mp 210 °C with decomp. IR Spectrum, ν, cm−1: 1511, 1256 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 2.58 (3H, s, C-5-CH3); 5.84 (1H, s, H-7); 6.80–6.82 (1H, m, H-3ʹ); 6.83–6.87 (1H, m, H-4ʹ); 7.23 (1H, d, H-5ʹ, J = 5.0 Hz); 7.56 (1H, s, H-2); 10.85 (1H, s, NH); 12.49 (1H, s, H-3). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 21.8; 34.3; 106.2; 122.9; 123.1; 123.6; 126.5; 126.7; 144.5; 150.6; 151.1. Anal. Calcd. for C11H10N4O2S: C, 50.37; H, 3.84; N, 21.36. Found: C, 50.20; H, 3.99; N, 21.49.
3-Etoxycarbonyl-5-ethyl-6-nitro-7-phenyl-4,7-dihydropyrazolo[1,5-a]pyrimidine (5e). The reaction was performed according to the general procedure 1 employing 0.31 g (2 mmol, 1 equiv.) of 3-amino-4-etoxycarbonylpyrazole 1b, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.2 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. Orange yellow solid. Yield 0.424 g (62%). mp 127–129 °C. IR Spectrum, ν, cm−1: 1669 (C=O); 1584, 1289 (NO2). 1H NMR (600 MHz, DMSO-d6): δ = 1.32 (6H, t, CH2-CH3, J = 7.1 Hz); 3.19 (2H, q, C-5-CH2-CH3, J = 7.2 Hz); 4.18–4.38 (2H, m, C(O)-CH2-CH3); 6.55 (1H, s, H-7); 7.22–7.36 (5H, m, Ph); 7.66 (1H, s, H-2); 10.29 (1H, s, NH). 13C {1H} NMR (151 MHz, DMSO-d6): δ = 12.5; 14.3; 25.2; 59.4; 59.8; 97.6; 122.0; 127.1; 139.6; 128.4; 128.6; 137.4; 140.8; 152.7; 161.7. Anal. Calcd. for C17H18N4O4: C, 59.64; H, 5.30; N, 16.37. Found: C, 59.69; H, 5.32; N, 16.49.
3-Etoxycarbonyl-5-ethyl-6-nitro-7-(4ʹ-nitrophenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5f). The reaction was performed according to the general procedure 2 employing 0.31 g (2 mmol, 1 equiv.) of 3-amino-4-etoxycarbonylpyrazole 1b, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.302 g (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c. Pale yellow solid. Yield 0.425 g (55%). mp 156–158 °C. IR Spectrum, ν, cm−1: 1720 (C=O); 1579, 1519, 1350, 1307 (NO2). 1H NMR (600 MHz, DMSO-d6): δ = 1.34 (6H, m, CH2-CH3); 3.23 (3H, q, C-5-CH2-CH3); 4.30 (2H, q, C(O)-CH2-CH3); 6.72 (1H, s, H-7); 7.59 (2H, d, H-2ʹ, J = 8.4 Hz); 7.69 (1H, s, H-2); 8.19 (2H, d, H-3ʹ, J = 8.4 Hz); 10.48 (1H, s., NH). 13C {1H} NMR (151 MHz, DMSO-d6): δ = 12.5; 14.3; 25.2; 58.8; 59.9; 97.9; 121.3; 123.9; 128.6; 141.2; 146.4; 147.4; 153.5; 161.6. Anal. Calcd. for C17H17N5O6: C, 52.95; H, 4.57; N, 18.10. Found: C, 52.71; H, 4.42; N, 18.08.
3-Cyano-6-nitro-7-phenyl-4,7-dihydropyrazolo[1,5-a]pyrimidine (5g). The reaction was performed according to the general procedure 1 employing 0.216 g (2 mmol, 1 equiv.) of 3-amino-3-cyanopyrazole 1c, 0.316 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitroethylene 3c and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. The product was recrystallized from MeOH. Yellow solid. Yield 0.219 g (41%). mp 262 °C with decomp. IR Spectrum, ν, cm−1: 2232 (CN); 1593, 1333 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 6.62 (1H, s, H-7); 7.20–7.47 (5H, m, Ph); 7.95 (1H, s, H-2); 8.47 (1H, s, H-5); 12.43 (1H, br.s.; NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 59.6; 76.2; 112.6; 124.3; 127.5; 128.6; 128.7; 134.6; 138.8; 139.6; 143.1. Anal. Calcd. for C13H9N5O2: C, 58.43; H, 3.39; N, 26.21. Found: C, 58.49; H, 3.33; N, 26.29
3-Cyano-5-ehtyl-6-Nitro-7-(4ʹ-nitrophenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5h). The reaction was performed according to the general procedure 1 employing 0.216 g (2 mmol, 1 equiv.) of 3-amino-4-cyanopyrazole 1c, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.302 g (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c. The substance was dried over P2O5 at 170 °C. Yellow solid. Yield 0.394 g (58%). mp 230 °C with decomp. IR Spectrum, ν, cm−1: 2231 (CN); 1584, 1350 (NO2); 1520, 1315 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.32 (3H, t, CH2-CH3; J = Hz); 2.90–3.15 (2H, m, CH2-CH3); 6.76 (1H, s, H-7); 7.63 (2H, d, H-2ʹ, J = 8.3 Hz); 7.95 (1H, s, H-2); 8.17 (2H, d, H-3ʹ, J = 8.3 Hz); 12.07 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.4; 25.5; 59.1; 75.9; 112.7; 121.0; 123.8; 128.8; 139.3; 143.5; 146.1; 147.5; 153.0. Anal. Calcd. for C15H12N6O4: C, 52.94; H, 3.55; N, 24.70. Found: C, 52.89; H, 3.47; N, 24.69.
3-Cyano-5-ehtyl-6-Nitro-7-(4ʹ-metoxyphenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5i).
The reaction was performed according to the general procedure 1 employing 0.216 g (2 mmol, 1 equiv.) of 3-aminopyrazole 1c, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.24 mL (2 mmol, 1 equiv.) of 4-metoxybenzaldehyde 4c. The product was recrystallized from MeOH. Light-yellow solid. Yield 0.338 g (52%). mp 249 °C with decomp. IR Spectrum, ν, cm−1: 2229 (CN); 1577, 1307 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.10–1.48 (3H, m, CH2-CH3); 2.87–3.13 (2H, m, CH2-CH3); 3.71 (3H, s, O-CH3); 6.54 (1H, s, H-7); 6.79–6.94 (2H, m, H-3ʹ); 7.13–7.30 (2H, m, H-2ʹ); 7.92 (1H, s, H-2); 11.83 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.4; 25.4; 55.1; 59.2; 75.3; 112.9; 114.0; 122.0; 128.5; 131.5; 139.1; 143.1; 151.8; 159.3. Anal. Calcd. for C16H15N5O3: C, 59.07; H, 4.65; N, 21.53. Found: C, 59.17; H, 4.69; N, 21.44.
3-Cyano-5-Ethyl-6-nitro-7-(3ʹ-methoxy-4ʹ-hydroxyphenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5j). The reaction was performed according to the general procedure 1 employing 0.216 g (2 mmol, 1 equiv.) of 3-amino-4-cyanopyrazole 1c, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.304 g (2 mmol, 1 equiv.) of 3-metoxy-4-hydroxybenzaldehyde 4e. Pale yellow solid. Yield 0.355 g (52%). mp 241 °C with decomp. IR Spectrum, ν, cm−1: 3181 (OH); 2234 (CN); 1579, 1306 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.32 (3H, t, CH2-CH3, J = 7.3 Hz); 2.90–3.13 (2H, m, CH2-CH3); 3.74 (1H, s, O-CH3); 6.50 (1H, S, H-7); 6.62–6.69 (1H, m, H-5ʹ); 6.72 (1H, d, H-6ʹ, J = 8.1 Hz); 7.93 (1H, s, H-2); 9.13 (1H, s, OH); 11.79 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.5; 25.5; 55.7; 59.5; 75.3; 111.8; 112.9; 115.5; 119.5; 121.9; 130.2; 143.0; 147.0; 147.4; 151.7. Anal. Calcd. for C16H15N5O4: C, 56.30; H, 4.43; N, 20.52. Found: C, 56.39; H, 4.36; N, 20.44.
3-Cyano-5-ethyl-6-nitro-7-(thiophen-2-yl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5k). The reaction was performed according to the general procedure 1 employing 0.216 g (2 mmol, 1 equiv.) of 3-amino-4-cyanopyrazole 1c, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.18 mL (2 mmol, 1 equiv.) of thiophene-2-carbaldehyde 4f. Pale red solid. Yield 0.307 g (51%). mp 188–190 °C with decomp. IR Spectrum, ν, cm−1: 2232 (CN); 1576, 1300 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.30 (3H, t, CH2-CH3, J = 7.4 Hz); 2.84–3.14 (2H, m, CH2-CH3); 6.94 (1H, s, H-7); 6.89–6.98 (1H, s, H-3ʹ); 7.01–7.08 (1H, m, H-4ʹ); 7.41–7.49 (1H, d, H-5ʹ, J = 4.9 Hz); 8.00 (1H, s, H-2); 12.00 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.8; 25.9; 55.1; 76.1; 113.2; 122.1; 127.0; 127.1; 127.5; 139.6; 142.4; 143.8; 152.7. Anal. Calcd. for C13H11N5O2S: C, 51.82; H, 3.68; N, 23.24. Found: C, 51.69; H, 3.66; N, 23.36.
3-Cyano-2-methylthio-6-nitro-7-phenyl-4,7-dihydropyrazolo[1,5-a]pyrimidine (5l). The reaction was performed according to the general procedure 1 employing 0.308 g (2 mmol, 1 equiv.) of 3-amino-4-cyano-5-methylthiopyrazole 1d, 0.316 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitroethylene 3a and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. Yellow solid. Yield 0.319 g (51%). mp 219 °C with decomp. IR Spectrum, ν, cm−1: 2229 (CN); 1596, 1325 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 2.41 (3H, s, S-CH3); 6.56 (1H, s, H-7); 7.11–7.56 (5H, m, Ph); 8.45 (1H, s, H-2); 12.44 (1H, br. s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 13.8; 59.6; 76.1; 111.9; 124.6; 127.6; 128.6; 128.8; 134.3; 138.5; 140.8; 150.8. Anal. Calcd. for C14H11N5O2S: C, 53.67; H, 3.54; N, 22.35. Found: C, 53.75; H, 3.61; N, 22.19.
3-Cyano-5-methyl-2-methylthio-6-nitro-7-phenyl-4,7-dihydropyrazolo[1,5-a]pyrimidine (5m). The reaction was performed according to the general procedure 1 employing 0.308 g (2 mmol, 1 equiv.) of 3-amino-4-cyano-5-methylthiopyrazole 1d, 0.344 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitropropylene 3b and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. Sand color solid. Yield 0.294 g (45%). mp 251–253 °C with decomp. IR Spectrum, ν, cm−1: 2229 (CN); 1575, 1306 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 2.42 (3H, s, S-CH3); 2.67 (3H, s, C-5-CH3); 6.53 (1H, s, H-7); 7.27–7.47 (5H, m, Ph); 11.90 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 13.8; 19.6; 59.8; 75.3; 112.1; 122.6; 127.4 (2C); 128.6; 139.0; 140.4; 147.4; 150.9. Anal. Calcd. for C15H13N5O2S: C, 55.04; H, 4.00; N, 21.39. Found: C, 55.00; H, 4.04; N, 21.50.
5-Ehtyl-3-cyano-2-methylthio-6-nitro-7-(4ʹ-nitrophenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5n). The reaction was performed according to the general procedure 1 employing 0.308 g (2 mmol, 1 equiv.) of 3-amino-4-cyano-5-methylthiopyrazole 1d, 0.376 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.302 g (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c. The substance was dried over P2O5 at 170 °C. Yellow solid. Yield 0.363 g (47%). mp 220 °C with decomp. IR Spectrum, ν, cm−1: 2236 (CN); 1580, 1331 (NO2); 1524, 1350 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.30 (3H, t, CH2-CH3, J = 7.4 Hz); 2.40 (3H, s, S-CH3); 2.90–3.10 (2H, m, CH2-CH3); 6.71 (1H, s, H-7); 7.65 (2H, d, H-2ʹ, J = 8.3 Hz); 8.19 (2H, d, H-3ʹ, J = 8.3 Hz); 12.06 (1H, br.s.; NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.3; 13.7; 59.0; 75.7; 112.0; 121.4; 129.9; 128.9; 140.6; 145.8; 147.5; 151.4; 152.7. Anal. Calcd. for C16H14N6O4S: C, 49.74; H, 3.65; N, 21.75. Found: C, 49.69; H, 3.60; N, 21.81.
3-Cyano-2-methylthio-6-nitro-7-(4ʹ-nitrophenyl)-4,7-dihydropyrazolo[1,5-a]pyrimidine (5o). The reaction was performed according to the general procedure 2 employing 0.308 g (2 mmol, 1 equiv.) of 3-amino-4-cyano-5-methylthiopyrazole 1d, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c and 0.243 mL (2 mmol, 1 equiv.) of 4-metoxybenzaldehyde 4d. Yellow solid. Yield 0.378 g (51%). mp 183 °C with decomp. IR Spectrum, ν, cm−1: 2224 (CN); 1577, 1302 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.33 (3H, t, CH2-CH3, J = 7.3 Hz); 2.44 (1H, s, S-CH3); 3.01 (2H, q, CH2-CH3; J = 7.3 Hz); 3.75 (1H, s, O-CH3); 6.44 (1H, s, H-7); 6.84 (2H, d, H-3ʹ, J = 8.3 Hz); 7.19 (2H, d, H-2ʹ, J = 7.19 Hz); 11.68 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.8; 14.3; 25.9; 55.6; 59.7; 75.8; 112.6; 114.5; 122.8; 129.0; 131.7; 140.9; 151.3; 152.0; 159.9. Anal. Calcd. for C17H17N5O3S: C, 54.98; H, 4.61; N, 18.86. Found: C, 55.08; H, 4.59; N, 18.89.
6-Nitro-7-phenyl-4,7-dihydro-1,2,4-triazolo[1,5-a]pyrimidine (6a). The reaction was performed according to the general procedure 1 employing 0.168 g (2 mmol, 1 equiv.) of 3-amino-1,2,4-triazole 2a, 0.316 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitroethylene 3a and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. Yellow solid. Yield 0.253 g (52%). mp 269 °C with decomp. IR Spectrum, ν, cm−1: 1593, 1314 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 6.65 (1H, s, H-7); 7.15–7.60 (5H, m, Ph); 7.79 (1H, s, H-5); 8.54 (1H, s, H-2); 12.09 (1H, br. s., NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 59.5; 123.8; 127.4; 128.6; 128.6; 136.5; 38.8; 145.7; 151.0. Anal. Calcd. for C11H9N5O2: C, 54.32; H, 3.73; N, 28.79. Found: C, 54.21; H, 3.79; N, 28.69.
5-Methyl-2-methylthio-6-nitro-7-phenyl-4,7-dihydro-1,2,4-triazolo[1,5-a]pyrimidine (6b). The reaction was performed according to the general procedure 1 employing 0.260 g (2 mmol, 1 equiv.) of 3-amino-5-methylthio-1,2,4-triazole 2b, 0.344 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitropropylene 3b and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a. Pale yellow solid. Yield 0.327 g (54%). mp 274–276 °C. IR Spectrum, ν, cm−1: 1557, 1320 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 2.42(3H, s, S-CH3); 2.64 (3H, s, C-5-CH3); 6.56 (1H, s, H-7); 7.27–7.38 (5H, m, Ph); 11.90 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 13.5; 20.2; 59.8; 122.2; 127.4; 128.5; 128.6; 139.1; 146.2; 148.8; 160.0. Anal. Calcd. for C13H13N5O2S: C, 51.49; H, 4.41; N, 23.02. Found: C, 51.47; H, 4.32; N, 23.09.
5-Methyl-2-methylthio-6-nitro-7-(4ʹ-nitrophenyl)-4,7-dihydro-1,2,4-triazolo[1,5-a]pyrimidine (6c). The reaction was performed according to the general procedure 2 employing 0.260 g (2 mmol, 1 equiv.) of 3-amino-5-methylthio-1,2,4-triazole 2b, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 2b and 0.302 mL (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c. Yellow solid. Yield 0.384 g (53%). mp 239–241 °C. IR Spectrum, ν, cm−1: 1581, 1555, 1345, 1303 (NO2). 1H NMR (400 MHz, DMSO-d6): δ = 1.34 (3H, t, CH2-CH3, J = 7.3 Hz); 2.44 (3H, s, S-CH3); 2.91–3.11 (2H, m, CH2-CH3); 6.71 (1H, s, H-7); 7.64 (2H, d, H-2ʹ, J = 8.5 Hz); 8.22 (2H, d, H-3ʹ, J = 8.6 Hz); 12.00 (1H, s., NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.3; 13.5; 26.0; 59.0; 120.9; 123.84; 128.8; 146.0; 146.3; 147.5; 154.2; 160.5. Anal. Calcd. for C14H14N6O4S: C, 46.40; H, 3.89; N, 23.19. Found: C, 46.55; H, 3.77; N, 23.10.
5-Methyl-6-nitro-7-phenyl-2-trifluoromethyl-4,7-dihydro-1,2,4-triazolo[1,5-a]pyrimidine (6d).
To a suspension of 0.304 g (2 mmol, 1 equiv.) of 3-amino-5-trifluoromethyl-1,2,4-triazole 2c, 0.344 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitropropylene 3b and 0.20 mL (2 mmol, 1 equiv.) of benzaldehyde 4a in 5 mL n-BuOH 3 mmol (1.5 equiv., 0.37 mL) of BF3·Et2O was added. The reaction mixture was heated on oil bath at 120 °C for 2 h. The resulting solution was cooled to room temperature and evaporated. To residue 5 mL of n-heptane was added. The obtained suspension was stirred for 10 min, filtered off and washed with 20 mL of water. The product was recrystallized from i-PrOH-H2O 1/1. Yellow solid. Yield 0.273 g (42%); mp 233–235 °C. IR Spectrum, ν, cm−1: 1573, 1321 (NO2), 1133 (CF3). 1H NMR (400 MHz, DMSO-d6): δ = 2.66 (3H, s, C-5-CH3); 6.74 (1H, s, H-7); 7.31–7.45 (5H, m, Ph); 12.13 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 20.1; 60.4; 118.9 (q, J = 269.7 Hz); 122.5; 127.6; 128.7; 128.9; 138.4; 147.0; 148.8; 151.1 (q, J = 39.1 Hz). Anal. Calcd. for C13H10F3N5O2: C, 48.01; H, 3.10; N, 21.53. Found: C, 48.15; H, 3.24; N, 21.40.
5-Methyl-6-nitro-7-(4ʹ-nitrophenyl)-2-trifluoromethyl-4,7-dihydro-1,2,4-triazolo[1,5-a]pyrimidine (6e). 3 Mmol (1.5 equiv., 0.37 mL) of BF3·Et2O was added to a suspension of 0.304 g (2 mmol, 1 equiv.) of 3-amino-5-trifluoromethyl-1,2,4-triazole 2c, 0.372 g (2 mmol, 1 equiv.) of 1-morpholino-2-nitrobutylene 3c in 5 mL n-BuOH. The reaction mixture was heated on oil bath at 80 °C for 15 min. After this, 0.302 g (2 mmol, 1 equiv.) of 4-nitrobenzaldehyde 4c was added to the obtained solution. The reaction mixture was heated on oil bath at 120 °C for 2 h. The resulting solution was cooled to room temperature and evaporated. To residue, 5×3 mL of n-heptane was added, and the obtained mixture was decanted. The same procedure was carried out with water. The crude oil was dissolved in 5 mL of i-PrOH, and the obtained solution was left overnight. The obtained suspension was filtered off and recrystallized from i-PrOH/H2O 1/1. Pale yellow solid. Yield 0.322 g (42%). mp 228 °C with decomp. IR Spectrum, ν, cm−1: 1573, 1309 (NO2); 1152 (CF3). 1H NMR (400 MHz, DMSO-d6): δ = 1.32 (3H, t, CH2-CH3, J = 7.4 Hz); 2.85–3.11 (2H, m, CH2-CH3); 6.93 (1H, s, H-7); 7.75 (2H, d, H-2ʹ, J = 8.3 Hz); 8.21 (2H, d, H-3ʹ, J = 8.3 Hz); 12.29 (1H, s, NH). 13C {1H} NMR (101 MHz, DMSO-d6): δ = 12.2; 26.0; 59.6; 118.81 (q, J = 269.9 Hz); 121.3; 124.0; 129.2; 145.1; 147.2; 147.8; 151.5 (q, J = 39.2 Hz); 154.3. Anal. Calcd. for C14H11F3N6O4: C, 43.76; H, 2.89; N, 21.87. Found: C, 43.69; H, 2.80; N, 21.98.

3.2. Biological Experiments

3.2.1. CK2 Assay

Kinase activity was determined using the enzyme system CK2α1 (Promega V4482, Madison, WI, USA) and the ADP-GloTM kit (Promega V9101, Madison, WI, USA) in white 96-well plates (Nunc U96 Microwell 267350, Denmark). The reaction was carried out using 50 ng/well of N-GST labeled human recombinant CK2α1, 0.1 µg/µL bovine casein as a substrate, 10 μM ATP in 40 mM Tris buffer solution (pH 7.50) containing 20 mM MgCl2, 0.1 mg/mL of BSA, and 50 μM of DTT. Test compounds were added to 1.25% DMSO (final concentration 0.25%) and preincubated with kinase for 10 min. The reaction was carried out for 60 min at 25 °C in a thermostatically controlled PST-60HL shaker (Biosan. Beresfield, NSW, Latvia). ATP-dependent luminescence was measured at an integration time of 1000 ms using the Infinite M200 PRO microplate reader (Tecan. Austria). The ATP-competitive inhibitor Staurosporin (CAS 62996–74-1, Alfa Aesar J62837, 99+%) was used as a positive control. The experiments were performed in two parallels.

3.2.2. Cytotoxicity Study

Cell Culture

The studies were carried out on cultured cells of human glioblastoma (A-172, ATCC CRL 1620) [35], human osteosarcoma (Hos, ATCC CRL 1543) [36,37,38], human embryonic rhabdomyosarcoma (Rd, ATCC CRL 136) [39], and human embryonic kidney 293 cells (Hek-293, ATCC CRL 1573) [40] obtained from the Shared research facility “Vertebrate cell culture collection” (Institute of Cytology RAS, Saint-Petersburg, Russia). The cells were cultured using DMEM / F-12 medium containing 10% fetal bovine serum at 37°C, 5% CO2, and 98% humidity. Subculturing was performed using 0.25% trypsin solution when the culture reached ≥90% confluency.

Viability Assessment

The compounds were dissolved in DMSO. The solutions were diluted with DMEM/F-12 culture medium with 10% fetal bovine serum to the studied concentrations: 8, 16, 32, 64, 128, 256, 512, and 1024 µM. In all cases, the concentration of DMSO in the final solution did not exceed 1%. Cisplatin (cPt) was used as a positive control.
Cells were seeded in 96-well plates at a concentration of 4 × 103 cells per well. After 24 h, test compounds were added to the wells in a given concentration range. Then the cells were incubated for 72 h, after which a solution of MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) was added to the cultures at 20 µL (5 mg/mL) to the well. After 2.5 h, the medium was removed from the wells and 200 µL of a mixture of DMSO/i-PrOH 1/1 was added. Optical density was measured on a plate spectrophotometer at a wavelength of 570 nm.

Statistical Analysis

Statistical data processing was carried out in the RStudio program (Version 1.4.1106 © 2022–2021 RStudio, PBC, Boston, MA, USA) using the R package (version 4.1.2). The cytotoxicity index (IC50) was calculated by plotting dose–response curves using the “drc” package [41].

4. Conclusions

Thus, in this work we extended the library of the 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidine series, and also studied their antitumor properties. The inhibitory activity of these compounds against CK2 has been established, as well as their cytotoxic effect. Compounds of this series are comparable in inhibitory activity with the reference drug and exhibit a cytotoxic effect on tumor cells at micromolar concentrations. It is evident that the herein reported 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines have the potential to be studied as a new class of antitumor compounds.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/molecules27165239/s1, NMR Spectra of compounds 5,6, and biological experiments.

Author Contributions

Synthesis, D.N.L. and I.I.B.; methodology, D.N.L. and E.N.U.; writing, D.N.L., S.K.K. and V.L.R.; CK2 inhibition investigation, D.A.B., A.A.P. and A.A.S.; cytotoxicity study, V.V.M., M.D.T., A.V.S. and O.G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Ministry of Science and Higher Education of the Russian Federation, State Contract № FEUZ-2020–0058 (H687.42B.223/20).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within article.

Acknowledgments

The team of authors would like to thank the Laboratory for Comprehensive Research and Expert Evaluation of Organic Materials under the direction of O.S. Eltsov.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds 5,6 are available from the authors.

References

  1. Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today; International Agency for Research on Cancer: Lyon, France, 2020; Available online: https://gco.iarc.fr/today (accessed on 28 February 2021).
  2. Charoenrat, P.; Rusch, V.; Talbot, S.G.; Sarkaria, I.; Viale, A.; Socci, N.; Ngai, I.; Rao, P.; Singh, B. Casein Kinase II Alpha Subunit and C1-Inhibitor Are Independent Predictors of Outcome in Patients with Squamous Cell Carcinoma of the Lung. Clin. Cancer Res. 2004, 10, 5792–5803. [Google Scholar] [CrossRef] [PubMed]
  3. Ortega, C.E.; Seidner, Y.; Dominguez, I. Mining CK2 in Cancer. PLoS ONE 2014, 9, e115609. [Google Scholar] [CrossRef] [PubMed]
  4. Faust, R.A.; Gapany, M.; Tristani, P.; Davis, A.; Adams, G.L.; Ahmed, K. Elevated Protein Kinase CK2 Activity in Chromatin of Head and Neck Tumors: Association with Malignant Transformation. Cancer Lett. 1996, 101, 31–35. [Google Scholar] [CrossRef]
  5. Faust, R.A.; Niehans, G.; Gapany, M.; Hoistad, D.; Knapp, D.; Cherwitz, D.; Davis, A.; Adams, G.L.; Ahmed, K. Subcellular Immunolocalization of Protein Kinase CK2 in Normal and Carcinoma Cells. Int. J. Biochem. Cell Biol. 1999, 31, 941–949. [Google Scholar] [CrossRef]
  6. Gapany, M.; Faust, R.A.; Tawfic, S.; Davis, A.; Adams, G.L.; Ahmed, K. Association of Elevated Protein Kinase CK2 Activity with Aggressive Behavior of Squamous Cell Carcinoma of the Head and Neck. Mol. Med. 1995, 1, 659. [Google Scholar] [CrossRef]
  7. Kim, H.S.; Chang, Y.G.; Bae, H.J.; Eun, J.W.; Shen, Q.; Park, S.J.; Shin, W.C.; Lee, E.K.; Park, S.; Ahn, Y.M.; et al. Oncogenic Potential of CK2α and Its Regulatory Role in EGF-Induced HDAC2 Expression in Human Liver Cancer. FEBS J. 2014, 281, 851–861. [Google Scholar] [CrossRef]
  8. Rabjerg, M.; Bjerregaard, H.; Halekoh, U.; Jensen, B.L.; Walter, S.; Marcussen, N. Molecular Characterization of Clear Cell Renal Cell Carcinoma Identifies CSNK2A1, SPP1 and DEFB1 as Promising Novel Prognostic Markers. APMIS 2016, 124, 372–383. [Google Scholar] [CrossRef]
  9. Nitta, R.T.; Gholamin, S.; Feroze, A.H.; Agarwal, M.; Cheshier, S.H.; Mitra, S.S.; Li, G. Casein Kinase 2α Regulates Glioblastoma Brain Tumor-Initiating Cell Growth through the β-Catenin Pathway. Oncogene 2014, 34, 3688–3699. [Google Scholar] [CrossRef]
  10. Pagano, M.A.; Cesaro, L.; Meggio, F.; Pinna, L.A. Protein Kinase CK2: A Newcomer in the ‘Druggable Kinome’. Biochem. Soc. Trans. 2006, 34, 1303–1306. [Google Scholar] [CrossRef]
  11. Guerra, B.; Issinger, O.-G. Protein Kinase CK2 in Human Diseases. Curr. Med. Chem. 2008, 15, 1870–1886. [Google Scholar] [CrossRef]
  12. Silva-Pavez, E.; Tapia, J. Protein Kinase CK2 in Cancer Energetics. Front Oncol. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  13. Borgo, C.; D’Amore, C.; Sarno, S.; Salvi, M.; Ruzzene, M. Protein Kinase CK2: A Potential Therapeutic Target for Diverse Human Diseases. Curr. Signal Transduct. Ther. 2021, 6, 1–20. [Google Scholar] [CrossRef] [PubMed]
  14. Chua, M.M.J.; Lee, M.; Dominguez, I. Cancer-Type Dependent Expression of CK2 Transcripts. PLoS ONE 2017, 12, e0188854. [Google Scholar] [CrossRef]
  15. Pagano, M.A.; Bain, J.; Kazimierczuk, Z.; Sarno, S.; Ruzzene, M.; di Maira, G.; Elliott, M.; Orzeszko, A.; Cozza, G.; Meggio, F.; et al. The Selectivity of Inhibitors of Protein Kinase CK2: An Update. Biochem. J. 2008, 415, 353–365. [Google Scholar] [CrossRef] [PubMed]
  16. Trembley, J.H.; Chen, Z.; Unger, G.; Slaton, J.; Kren, B.T.; van Waes, C.; Ahmed, K. Emergence of Protein Kinase CK2 as a Key Target in Cancer Therapy. BioFactors. 2010, 36, 187–195. [Google Scholar] [CrossRef]
  17. Iegre, J.; Atkinson, E.L.; Brear, P.D.; Cooper, B.M.; Hyvönen, M.; Spring, D.R. Chemical Probes Targeting the Kinase CK2: A Journey Outside the Catalytic Box. Org. Biomol. Chem. 2021, 19, 4380–4396. [Google Scholar] [CrossRef]
  18. Dowling, J.E.; Alimzhanov, M.; Bao, L.; Chuaqui, C.; Denz, C.R.; Jenkins, E.; Larsen, N.A.; Lyne, P.D.; Pontz, T.; Ye, Q.; et al. Potent and Selective CK2 Kinase Inhibitors with Effects on Wnt Pathway Signaling in Vivo. ACS Med. Chem. Lett. 2016, 7, 300–305. [Google Scholar] [CrossRef] [PubMed]
  19. Wells, C.I.; Drewry, D.H.; Pickett, J.E.; Tjaden, A.; Krämer, A.; Müller, S.; Gyenis, L.; Menyhart, D.; Litchfield, D.W.; Knapp, S.; et al. Development of a Potent and Selective Chemical Probe for the Pleiotropic Kinase CK2. Cell Chem. Biol. 2021, 28, 546–558.e10. [Google Scholar] [CrossRef]
  20. Dalle Vedove, A.; Zonta, F.; Zanforlin, E.; Demitri, N.; Ribaudo, G.; Cazzanelli, G.; Ongaro, A.; Sarno, S.; Zagotto, G.; Battistutta, R.; et al. A Novel Class of Selective CK2 Inhibitors Targeting Its Open Hinge Conformation. Eur. J. Med. Chem. 2020, 195, 112267. [Google Scholar] [CrossRef]
  21. Oshima, T.; Niwa, Y.; Kuwata, K.; Srivastava, A.; Hyoda, T.; Tsuchiya, Y.; Kumagai, M.; Tsuyuguchi, M.; Tamaru, T.; Sugiyama, A.; et al. Cell-Based Screen Identifies a New Potent and Highly Selective CK2 Inhibitor for Modulation of Circadian Rhythms and Cancer Cell Growth. Sci. Adv. 2019, 5, 9060–9083. [Google Scholar] [CrossRef]
  22. Zhang, N.; Ayral-Kaloustian, S.; Nguyen, T.; Afragola, J.; Hernandez, R.; Lucas, J.; Gibbons, J.; Beyer, C. Synthesis and SAR of [1,2,4]Triazolo [1,5-a]Pyrimidines, a Class of Anticancer Agents with a Unique Mechanism of Tubulin Inhibition. J. Med. Chem. 2007, 50, 319–327. [Google Scholar] [CrossRef] [PubMed]
  23. Beyer, C.F.; Zhang, N.; Hernandez, R.; Vitale, D.; Lucas, J.; Nguyen, T.; Discafani, C.; Ayral-Kaloustian, S.; Gibbons, J.J. TTI-237: A Novel Microtubule-Active Compound with In Vivo Antitumor Activity. Cancer Res. 2008, 68, 2292–2300. [Google Scholar] [CrossRef] [PubMed]
  24. Wang-Gillam, A.; Arnold, S.M.; Bukowski, R.M.; Rothenberg, M.L.; Cooper, W.; Wang, K.K.; Gauthier, E.; Lockhart, A.C. A Phase I Dose Escalation Study of TTI-237 in Patients with Advanced Malignant Solid Tumors. Investig. New Drugs 2012, 30, 266–272. [Google Scholar] [CrossRef] [PubMed]
  25. Ayral-Kaloustian, S.; Zhang, N.; Beyer, C. Cevipabulin (TTI-237): Preclinical and Clinical Results for a Novel Antimicrotubule Agent. Methods Find. Exp. Clin. Pharmacol. 2009, 31, 443–447. [Google Scholar] [CrossRef] [PubMed]
  26. Sanchez, R.M.; Erhard, K.; Hardwicke, M.A.; Lin, H.; McSurdy-Freed, J.; Plant, R.; Raha, K.; Rominger, C.M.; Schaber, M.D.; Spengler, M.D.; et al. Synthesis and Structure–Activity Relationships of 1,2,4-Triazolo [1,5-a]Pyrimidin-7(3H)-Ones as Novel Series of Potent β Isoform Selective Phosphatidylinositol 3-Kinase Inhibitors. Bioorg. Med. Chem. Lett. 2012, 22, 3198–3202. [Google Scholar] [CrossRef]
  27. Hassan, A.Y.; Sarg, M.T.; Bayoumi, A.H.; El-Deeb, M.A. Synthesis and Anticancer Evaluation of Some Novel 5-Amino [1,2,4]Triazole Derivatives. J. Het. Chem. 2018, 55, 1450–1478. [Google Scholar] [CrossRef]
  28. Oukoloff, K.; Lucero, B.; Francisco, K.R.; Brunden, K.R.; Ballatore, C. 1,2,4-Triazolo [1,5-a]Pyrimidines in Drug Design. Eur. J. Med. Chem. 2019, 165, 332–346. [Google Scholar] [CrossRef]
  29. Pinheiro, S.; Pinheiro, E.M.C.; Muri, E.M.F.; Pessôa, J.C.; Cadorini, M.A.; Greco, S.J. Biological Activities of [1,2,4]Triazolo [1,5-a]Pyrimidines and Analogs. Med. Chem. Res. 2020, 29, 1751–1776. [Google Scholar] [CrossRef]
  30. Safari, F.; Bayat, M.; Nasri, S.; Karami, S. Synthesis and Evaluation of Anti-Tumor Activity of Novel Triazolo [1,5-a]Pyrimidine on Cancer Cells by Induction of Cellular Apoptosis and Inhibition of Epithelial-to-Mesenchymal Transition Process. Bioorg. Med. Chem. Lett. 2020, 30, 127111. [Google Scholar] [CrossRef]
  31. Lyapustin, D.N.; Ulomsky, E.N.; Zanakhov, T.O.; Rusinov, V.L. Three-Component Coupling of Aromatic Aldehydes, 1-Morpholino-2-Nitroalkenes, and 3-Aminoazoles via Boron Trifluoride Etherate Catalysis: Reaction Pathway and Features of the Formation of Intermediates. J. Org. Chem. 2019, 84, 15267–15275. [Google Scholar] [CrossRef]
  32. Lyapustin, D.N.; Ulomsky, E.N.; Rusinov, V.L. 6-Nitro-4,7-Dihydroazolo[1,5-a]Pyrimidines: An Alternative Mechanism of Formation and Studies of Alkylation. Chem. Het. Comp. 2020, 56, 1465–1472. [Google Scholar] [CrossRef]
  33. Lyapustin, D.N.; Ulomsky, E.N.; Balyakin, I.A.; Shchepochkin, A.v.; Rusinov, V.L.; Chupakhin, O.N. Oxidative Aromatization of 4,7-Dihydro-6-Nitroazolo[1,5-a]Pyrimidines: Synthetic Possibilities and Limitations, Mechanism of Destruction, and the Theoretical and Experimental Substantiation. Molecules 2021, 26, 4719. [Google Scholar] [CrossRef] [PubMed]
  34. Rusinov, V.L.; Drokin, R.A.; Tiufiakov, D.V.; Voinkov, E.K.; Ulomsky, E.N. Synthesis and Properties of the Salts of 1-Nitropropan-2-One and 1-Nitrobutan-2-One. Mendeleev Commun. 2020, 30, 177–179. [Google Scholar] [CrossRef]
  35. Giard, D.J.; Aaronson, S.A.; Todaro, G.J.; Arnstein, P.; Kersey, J.H.; Dosik, H.; Parks, W.P. In Vitro Cultivation of Human Tumors: Establishment of Cell Lines Derived From a Series of Solid Tumors. J. Natl. Cancer Inst. 1973, 51, 1417–1423. [Google Scholar] [CrossRef]
  36. Rhim, J.S.; Cho, H.Y.; Huebner, R.J. Non-Producer Human Cells Induced by Murine Sarcoma Virus. Int. J. Cancer 1975, 15, 23–29. [Google Scholar] [CrossRef]
  37. McAllister, R.M.; Gardner, M.B.; Greene, A.E.; Bradt, C.; Nichols, W.W.; Landing, B.H. Cultivation in vitro of cells derived from a human osteosarcoma. Cancer 1971, 27, 397–402. [Google Scholar] [CrossRef]
  38. Rhim, J.S.; Cho, H.Y.; Vernon, M.L.; Arnstein, P.; Huebner, R.J.; Gilden, R.v.; Nelson-Rees, W.A. Characterization of Non-Producer Human Cells Induced by Kirsten Sarcoma Virus. Int. J. Cancer 1975, 16, 840–849. [Google Scholar] [CrossRef]
  39. McAllister, R.M.; Melenyk, J.; Finklestein, J.Z.; Adams, E.C.; Gardner, M.B. Cultivation in Vitro of Cells Derived from a Human Rhabdomyosarcoma. Cancer 1969, 24, 520–526. [Google Scholar] [CrossRef]
  40. Graham, F.L.; Smiley, J.; Russell, W.C.; Nairn, R. Characteristics of a Human Cell Line Transformed by DNA from Human Adenovirus Type 5. J. Gen. Virol. 1977, 36, 59–72. [Google Scholar] [CrossRef]
  41. Ritz, C.; Baty, F.; Streibig, J.C.; Gerhard, D. Dose-Response Analysis Using R. PLoS ONE 2015, 10, e0146021. [Google Scholar] [CrossRef]
Figure 1. Most significant ATP-competitive CK2 inhibitors: A [19], B [20], C [21], D [18].
Figure 1. Most significant ATP-competitive CK2 inhibitors: A [19], B [20], C [21], D [18].
Molecules 27 05239 g001
Figure 2. Azolo[1,5-a]pyrimidines exhibiting antitumor activity: E [22,23,24,25], F [30], G [26], H [27].
Figure 2. Azolo[1,5-a]pyrimidines exhibiting antitumor activity: E [22,23,24,25], F [30], G [26], H [27].
Molecules 27 05239 g002
Scheme 1. Preparation of 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines 5,6.
Scheme 1. Preparation of 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines 5,6.
Molecules 27 05239 sch001
Figure 3. Cytotoxicity index IC50 ± SE for compounds 5m, 5o, 6c, 6d, and cPt in comparison.
Figure 3. Cytotoxicity index IC50 ± SE for compounds 5m, 5o, 6c, 6d, and cPt in comparison.
Molecules 27 05239 g003
Figure 4. Comparison of the selectivity ratio (normal/cancer cell).
Figure 4. Comparison of the selectivity ratio (normal/cancer cell).
Molecules 27 05239 g004
Table 1. Screening of the target compounds against CK2 activity.
Table 1. Screening of the target compounds against CK2 activity.
CompoundCK2 Inhibition at 50 μM, m ± SD (%)CompoundCK2 Inhibition at 50 μM, m ± SD (%)
5an.a.5k66.81 ± 7.97 **
5bn.a.5l48.35 ± 4.48 *
5cn.a.5mn.a.
5dn.a.5n19.80 ± 24.53
5en.a.5on.a.
5f53.81 ± 0.426an.a.
5gn.a.6bn.a.
5h54.80 ± 0.876cn.a.
5i3.66 ± 37.606dn.a.
5jn.a.6e39.82 ± 19.52
Staurosporine72.34 ± 6.39 **
n.a.-not active; * p < 0.05, ** p < 0.01-significance vs. DMSO-control. Kruskal–Wallis test.
Table 2. Inhibition of CK2 by the most active compounds.
Table 2. Inhibition of CK2 by the most active compounds.
CompoundCK2 IC50, μM95% C.I., μMHill Coefficient
5f52.8339.12–59.08−1.369
5h59.4732.99–66.81−1.566
5k52.2630.10–105.80−1.183
5l57.2057.14–57.26−2.947
6e>100--
Staurosporine69.8550.96–98.39−0.974
Table 3. Cytotoxicity index (IC50 ± SE) of 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines on glioblastoma (A-172), embryonic rhabdomyosarcoma (Rd), osteosarcoma (Hos), and human embryonic kidney (Hek-293) cells, µM.
Table 3. Cytotoxicity index (IC50 ± SE) of 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines on glioblastoma (A-172), embryonic rhabdomyosarcoma (Rd), osteosarcoma (Hos), and human embryonic kidney (Hek-293) cells, µM.
CompoundIC50, µM
Cells
A-172RdHosHek-293
5d28.91 ± 4.58105.54 ± 16.81103.53 ± 18.27543.74 ± 70.79
5e256.72 ± 12.2099.46 ± 4.43126.60 ± 7.17222.84 ± 6.33
5f212.36 ± 42.85121.97 ± 13.53170.14 ± 11.1635.86 ± 4.30
5g171.74 ± 8.63149.72 ± 11.01181.14 ± 7.47483.09 ± 37.61
5h323.41 ± 22.70378.59 ± 20.39234.97 ± 25.80153.47 ± 12.35
5i145.19 ± 8.96105.10 ± 10.7088.44 ± 5.8569.52 ± 8.69
5j566.09 ± 17.12673.44 ± 20.70522.38 ± 16.16581.39 ± 43.90
5k110.23 ± 2.9789.43 ± 10.4677.32 ± 3.06107.57 ± 11.10
5l77.79 ± 4.02124.66 ± 6.8092.91 ± 3.91162.30 ± 9.95
5m13.36 ± 0.9827.52 ± 2.7718.54 ± 1.79211.7 ± 10.77
5n119.75 ± 8.49151.44 ± 7.28112.26 ± 8.1978.70 ± 9.75
5o22.49 ± 2.9336.33 ± 3.4028.09 ± 3.91169.30 ± 10.88
6a434.08 ± 18.02448.21 ± 22.53542.85 ± 21.451131.79 ± 77.86
6b71.03 ± 2.07110.11 ± 4.9392.03 ± 3.08237.40 ± 11.99
6c41.27 ± 4.1071.15 ± 6.8440.88 ± 4.11227.50 ± 25.20
6d27.49 ± 1.6737.28 ± 3.7723.31 ± 1.8450.07 ± 4.43
6e82.44 ± 2.22192.69 ± 10.2793.72 ± 5.8954.16 ± 5.32
cPt3.64 ± 0.214.99 ± 0.312.36 ± 0.124.41 ± 0.24
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lyapustin, D.N.; Kotovskaya, S.K.; Butorin, I.I.; Ulomsky, E.N.; Rusinov, V.L.; Babkov, D.A.; Pokhlebin, A.A.; Spasov, A.A.; Melekhin, V.V.; Tokhtueva, M.D.; et al. CK2 Inhibition and Antitumor Activity of 4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines. Molecules 2022, 27, 5239. https://doi.org/10.3390/molecules27165239

AMA Style

Lyapustin DN, Kotovskaya SK, Butorin II, Ulomsky EN, Rusinov VL, Babkov DA, Pokhlebin AA, Spasov AA, Melekhin VV, Tokhtueva MD, et al. CK2 Inhibition and Antitumor Activity of 4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines. Molecules. 2022; 27(16):5239. https://doi.org/10.3390/molecules27165239

Chicago/Turabian Style

Lyapustin, Daniil N., Svetlana K. Kotovskaya, Ilya I. Butorin, Evgeny N. Ulomsky, Vladimir L. Rusinov, Denis A. Babkov, Alexander A. Pokhlebin, Alexander A. Spasov, Vsevolod V. Melekhin, Maria D. Tokhtueva, and et al. 2022. "CK2 Inhibition and Antitumor Activity of 4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines" Molecules 27, no. 16: 5239. https://doi.org/10.3390/molecules27165239

Article Metrics

Back to TopTop