1,385
Views
11
CrossRef citations to date
0
Altmetric
Research Article

Synthesis of new pyridothienopyrimidinone derivatives as Pim-1 inhibitors

, &
Pages 457-467 | Received 09 Jul 2016, Accepted 08 Nov 2016, Published online: 18 Jan 2017

Abstract

Four series of pyridothienopyrimidin-4-one derivatives were designed and prepared to improve the pim-1 inhibitory activity of the previously reported thieno[2,3-b]pyridines. Significant improvement in the pim-1 inhibition and cytotoxic activity was achieved using structure rigidification strategy via ring closure. Six compounds (6c, 7a, 7c, 7d, 8b and 9) showed highly potent pim-1 inhibitory activity with IC50 of 4.62, 1.18, 1.38, 1.97, 8.83 and 4.18 μM, respectively. Four other compounds (6b, 6d, 7b and 8a) showed moderate pim-1 inhibition. The most active compounds were tested for their cytotoxic activity on three cell lines [MCF7, HCT116 and PC3]. Compounds 7a [the 2-(2-chlorophenyl)-2,3-dihydro derivative] and 7d [the 2-(2-(trifluoromethyl)-phenyl)-2,3-dihydro derivative] displayed the most potent cytotoxic effect on the three cell lines tested consistent with their highest estimated pim-1 IC50 values.

Introduction

The Provirus Integration in Maloney (Pim) kinases represent a family of constitutively active serine/threonine kinases and include three subtypes (pim-1, pim-2 and pim-3). Pim kinases regulate many biological processes such as cell cycle, cell proliferation, apoptosis and drug resistanceCitation1–4. Being expressed in many types of solid and hematological cancers and almost absent in benign lesions, pim kinases proved to be a successful anti-cancer drug target of low toxicityCitation5–12.

Most of the research published on pim inhibitors focused on pim-1 inhibitors while pim-2 is more difficult to be targeted due to its low Km for ATP (100-fold lower than that of Pim-1 and Pim-3)Citation13–16.

Recently reported manuscripts indicated that pim-1 kinase plays a significant role in stem cell proliferation, self-renewal and expansionCitation17. These facts encourage the use Pim-1 inhibitors as a promising targeted therapy in cancer stem cellsCitation5,Citation17.

Indeed, some of the pim-1 inhibitors have entered phase I clinical trials such as the thiazolidin-2,4-dione derivative AZD1208 (I)Citation1,Citation18 and the benzonaphthyridine derivative CX-4595 (II)Citation1,Citation19,Citation20. While others such as the imidazo[1,2-b]pyridazine derivative SGI-9481 (also known as TP-3654, III)Citation1 and the highly potent pyridine derivative LGB321 (IV)Citation1,Citation21,Citation22 are under preclinical studies ().

Figure 1. Pim-1 inhibitors under clinical and preclinical studies.

Figure 1. Pim-1 inhibitors under clinical and preclinical studies.

The most important feature of pim-1 kinase active site is the presence of proline base at positions 123 and 125 within the hinge region. This extends the hinge region length and moves it 4 Ao to the left and thus prevents the formation of the second H-bond between the hinge backbone and the adenine moiety of ATPCitation23.

Almost all the reported pim kinase inhibitors are ATP-competitive and can be classified into ATP-mimetics and non-ATP mimetics. The ATP-mimetics bind directly to the hinge region usually via H-bonding with the backbone oxygen of Glu121 and exhibited great enzyme potency but with limited or poor selectivity over other kinasesCitation1. On the other hand, the non-ATP mimetics bind to the ATP active site in a manner different from ATP and most of them form H-bond with Lys67Citation1. Generally, they interact with the portion of the active site opposite to the hinge region and this portion differs significantly between kinases. Thus, non-ATP mimietics tend to be more selective to pim-1 enzyme and meanwhile exhibited great potency to the enzyme. Compounds IIV () are all non-ATP mimietics.

In an attempt to prepare potent pim-1 inhibitors that can be used as anticancer agents, we had recently reported the pim-1 inhibitory activity of thieno[2,3-b]pyridine derivatives V ()Citation24 as bioisosteres to benzofuran-2-carboxylic acidsCitation25. However, their pim-1 inhibitions as well as their cytotoxic activities were only moderate to poor. This literature recorded some of the efforts done by our lab to improve the pim-1 kinase inhibitory activity and the cytotoxic activity of thieno[2,3-b]pyridine scaffold.

Figure 2. Designing pyridothienopyrimidinones as Pim-1 inhibitors.

Figure 2. Designing pyridothienopyrimidinones as Pim-1 inhibitors.

Upon designing thieno[2,3-b]pyridine derivatives as isosteres to benzofuran-2-carboxylic acids, we assumed that the amidic C=O can form H-bond with Lys67. Therefore, the poor activity achieved by these derivatives might originate from the flexibility of the amidic bond resulting in improper orientation of the carbonyl group towards Lys67. Accordingly, structure rigidification via ring closure of the 3-amino-thieno[2,3-b]pyridine-2-carboxamide core into pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one scaffold might orient the carbonyl group properly towards Lys67.

Searching the literature revealed no published data on pyridothienopyrimidinones as pim-1 kinase inhibitors. However, their bioisosteres benzothienopyrimidin-4-ones VI and VIICitation14,Citation26,Citation27 and benzofuropyrimidin-4-ones VIII and IXCitation28,Citation29 had been reported earlier and exhibited potent and selective pim kinase inhibition (). The X-ray crystallography of representatives of both series bound to pim-1 showed similar binding mode in the ATP-binding siteCitation27,Citation29. In both series, the 4-carbonyl group formed H-bond to Lys67, while the group at the 8-position (Br or aryl) occupied the hydrophobic area against the hinge region. The substituent at position 2 was exposed to the solvent and thus can accommodate various groups, however, the best results were achieved with ortho-substituted phenyl or aliphatic amines like dimethylaminomethylene groupCitation27,Citation29. As such, both series might act as non-ATP mimetics.

Figure 3. Examples of benzothienopyrimidinones and benzofuropyrimidinones as Pim-1 inhibitors.

Figure 3. Examples of benzothienopyrimidinones and benzofuropyrimidinones as Pim-1 inhibitors.

Encouraged by all the after mentioned findings, we thought of examining the pyridothienopyrimidinone scaffold as a novel class of pim-1 inhibitors. Here in, the bromine atom at position 8 was kept constant to ensure hydrophobic interaction within the pim-1 ATP active site. The carbonyl group at position 4 was also kept constant to ensure binding to Lys67. However, different substitutions were introduced at position 2 in order to establish a SAR for this new scaffold. First, the unsubstituted derivative 5 was prepared. Then, two series of 2-substituted phenyl compounds and two series of alkyl substituted derivatives were prepared. The ortho-substituted phenyl series (6ad) was prepared to mimic compound VIII. The ortho-substitution used were chloro and flouro as halogens, OH as electron donating group and CF3 as bulky, hydrophobic and electron withdrawing group. In the second series (7ad), the 2,3-dihydropyrimidinones, bearing the same 2-aryl groups as 6ad, were prepared to test whether aromaticity of the pyrimidinone ring was essential for enzyme activity or not. In the third series (8ac), different alkyl groups with different chain lengths were prepared. In the last series (911), carbonyl containing alkyl groups were prepared (Schemes 1–3). Docking study of these compounds in the pim-1 active site indicated the required binding mode with high energy scores (data not shown).

Scheme 1. Synthesis of the starting compounds 1–5.

Scheme 1. Synthesis of the starting compounds 1–5.

Scheme 2. Synthesis of the target compounds 6–8.

Scheme 2. Synthesis of the target compounds 6–8.

Scheme 3. Synthesis of the target compounds 9–11.

Scheme 3. Synthesis of the target compounds 9–11.

All the synthesized compounds were examined for their pim-1 enzyme inhibitory activity and the most active compounds were further tested for their anti-proliferative activity using three different cell lines. Up to our knowledge, this is the first published work describing the pim-1 inhibitory activity of pyridothienopyrimidinone derivatives.

Experimental

General notes

Griffin apparatus was used to determine the melting points and they were uncorrected. Shimadzu IR 435 spectrophotometer recorded the IR spectra and the values were represented in cm−1. Bruker 400 MHz and 100 MHz spectrophotometer recorded 1H NMR and 13C NMR spectra, respectively. TMS was used as an internal standard and chemical shifts were recorded in ppm on δ scale. Both IR and NMR spectra were carried out at Faculty of Pharmacy, Cairo University, Cairo, Egypt. The electron impact (EI) mass spectra were recorded on Thermo Scientific ISQLT single quadrapole mass spectrometer. Both mass spectra and elemental analyses were carried out at the regional center for mycology and biotechnology, Al-Azhar University, Cairo, Egypt. Analytical thin layer chromatography (TLC) on silica gel plates containing UV indicator was employed routinely to follow the course of the reactions and to check the purity of the products. All reagents and solvents were purified and dried by standard techniques.

5-Bromo-4,6-dimethyl-2-thioxo-1,2-dihydropyridine-3-carbonitrile (1), 5-bromo-2-[(cyanomethyl)sulfanyl]-4,6-dimethylpyridine-3-carbonitrile (2), 3-amino-5-bromo-4,6-dimethylthieno[2,3-b]pyridine-2-carbonitrile (3) and 8-bromo-7,9-dimethylpyrido[3′,2′:4,5]thieno[3,2-d]pyrimidin-4(3H)-one (5) were prepared according to the published methodsCitation30.

3-Amino-5-bromo-4,6-dimethylthieno[2,3-b]pyridine-2-carboxamide (4)

A mixture of 3-amino-5-bromo-4,6-dimethylthieno[2,3-b]pyridine-2-carbonitrile (3) (1 g, 3.5 mmol) and 5% alc. KOH solution (1 g KOH in 20 mL ethanol) was heated under reflux for 3 h. The reaction was cooled and diluted with water (100 mL). The solid was filtered, dried and crystallized from ethanol.

Yield: 88%; mp: 260–261 °CCitation31; IR (cm−1): 3464, 3417, 3317, 3267 (two NH2), 2924, 2854 (CH-aliphatic), 1666 (C = O); 1H NMR (DMSO-d6) δ ppm 2.68 (s, 3H, CH3), 2.86 (s, 3H, CH3), 6.89 (s, 2H, NH2, D2O exchangeable), 7.25 (s, 2H, NH2, D2O exchangeable); Anal. Calcd for C10H10BrN3OS: C, 40.01; H, 3.36; N, 14.00. Found: C, 40.23; H, 3.40; N, 14.21.

General procedure for the synthesis of 8-bromo-2-(2-substituted phenyl)-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-ones 6a-d

A mixture of 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 (0.6 g, 2 mmol), the appropriate 2-substituted benzaldehyde (2 mmol) and conc. HCl (0.5 mL) in DMF (5 mL) was heated under reflux for 12 h. The reaction was cooled, and the solid formed was filtered, dried and crystallized from the suitable solvent.

8-Bromo-2-(2-chlorophenyl)-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (6a)

Crystallized from DMF. Yield: 69%; mp: >300 °C; IR (cm−1): 3421 (NH), 2962, 2924 (CH-aliphatic), 1662 (C = O); 1H NMR (DMSO-d6) δ ppm 2.74 (s, 3H, CH3), 3.00 (s, 3H, CH3), 7.53–7.78 (m, 4H, Ar-H), 13.36 (s, 1H, NH, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 19.7, 26.8 (CH3), 122.9, 125.7, 127.8, 130.4, 131.8, 132.1, 132.5, 133.3, 144.2, 146.8, 151.6, 154.2, 158.4, 159.8 (aromatic carbons), 161.8 (C = O); Anal. Calcd for C17H11BrClN3OS: C, 48.53; H, 2.64; N, 9.99. Found: C, 48.70; H, 2.67; N, 10.14.

8-Bromo-2-(2-fluorophenyl)-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (6b)

Crystallized from DMF. Yield: 53%; mp: 295–296 °C; IR (cm−1): 3402, (NH), 2962, 2920 (CH-aliphatic), 1670 (C = O); 1H NMR (DMSO-d6) δ ppm 2.76 (s, 3H, CH3), 3.07 (s, 3H, CH3), 7.41–7.90 (m, 4H, Ar-H), 13.26 (s, 1H, NH, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 19.0, 23.1 (CH3), 117.5, 123.3, 128.8, 129.2, 129.9, 132.4, 134.4, 137.7, 140.2, 143.8, 154.8, 155.6, 156.6, 164.4 (aromatic carbons), 170.9 (C = O); MS m/z: 405 [(M + 2)+, 17.34%], 403 [M+, 21.19%], 43 [100%]; Anal. Calcd for C17H11BrFN3OS: C, 50.51; H, 2.74; N, 10.39. Found: C, 50.76; H, 2.81; N, 10.53.

8-Bromo-2-(2-hydroxyphenyl)-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (6c)

Crystallized from acetic acid. Yield: 57%; mp: 190–191 °C; IR (cm−1): 3394, 3309 (NH/OH), 2924, 2854 (CH-aliphatic), 1662 (C = O); 1H NMR (DMSO-d6) δ ppm 2.84 (s, 3H, CH3), 2.98 (s, 3H, CH3), 6.80–7.44 (m, 4H, Ar-H), 8.72 (s, 1H, OH, D2O exchangeable), 12.82 (s, 1H, NH, D2O exchangeable); Anal. Calcd for C17H12BrN3O2S: C, 50.76; H, 3.01; N, 10.45. Found: C, 50.98; H, 3.04; N, 10.67.

8-Bromo-7,9-dimethyl-2-[2-(trifluoromethyl)phenyl]pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (6d)

Crystallized from DMF. Yield: 61%; mp: 255–256 °C; IR (cm−1): 3464 (NH), 2954, 2924 (CH-aliphatic), 1670 (C = O); 1H NMR (DMSO-d6) δ ppm 2.66 (s, 3H, CH3), 2.84 (s, 3H, CH3), 7.81–7.97 (m, 4H, Ar-H), 13.45 (s, 1H, NH, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 20.1, 26.8 (CH3), 121.1, 122.1, 125.0, 131.3, 132.8, 144.4, 146.6, 147.9, 151.2, 154.5, 156.9, 157.2, 158.3, 159.8, 162.7 (CF3 and aromatic carbons), 167.4 (C = O); MS m/z: 455 [(M + 2)+, 14.02%], 453 [M+, 17.20%], 77 [100%]; Anal. Calcd for C18H11BrF3N3OS: C, 47.59; H, 2.44; N, 9.25. Found: C, 47.67; H, 2.48; N, 9.44.

General procedure for the synthesis of 8-bromo-2-(2-substituted phenyl)-7,9-dimethyl-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-ones 7a–d

A mixture of 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 (0.6 g, 2 mmol) and the appropriate 2-substituted benzaldehyde (2 mmol) in glacial acetic acid (5 mL) was heated under reflux for 10 h. The reaction was cooled, and the solid formed was filtered, dried and crystallized from acetic acid.

8-Bromo-2-(2-chlorophenyl)-7,9-dimethyl-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-one (7a)

Yield: 78%; mp: >300 °C; IR (cm−1): 3433, 3275 (NH), 2947, 2897 (CH-aliphatic), 1643 (C = O); 1H NMR (DMSO-d6) δ ppm 2.67 (s, 3H, CH3), 2.77 (s, 3H, CH3), 6.11–6.14 (dd, 1H, CH-2, J= 7.2 Hz, J= 3.2 Hz), 7.18 (d, 1H, NH, J= 7.2 Hz, D2O exchangeable), 7.36–7.54 (m, 4H, Ar-H), 8.34 (d, 1H, NH, J= 3.2 Hz, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 19.7, 26.7 (CH3), 64.3 (CH-2), 110.5, 121.4, 124.5, 127.4, 128.5, 130.5, 132.4, 132.4, 137.8, 144.2, 144.7, 157.4, 159.5 (aromatic carbons), 161.8 (C = O); Anal. Calcd for C17H13BrClN3OS: C, 48.30; H, 3.10; N, 9.94. Found: C, 48.53; H, 3.07; N, 10.19.

8-Bromo-2-(2-fluorophenyl)-7,9-dimethyl-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-one (7b)

Yield: 78%; mp: 292–293 °C; IR (cm−1): 3410, 3275 (NH), 2954, 2908 (CH-aliphatic), 1635 (C = O); 1H NMR (DMSO-d6) δ ppm 2.66 (s, 3H, CH3), 2.76 (s, 3H, CH3), 6.13–6.16 (dd, 1H, CH-2, J= 6.4 Hz, J= 2.8 Hz), 7.11 (d, 1H, NH, J= 6.8 Hz, D2O exchangeable), 7.17–7.48 (m, 4H, Ar-H), 8.36 (d, 1H, NH, J= 2.8 Hz, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 19.8, 26.7 (CH3), 61.8 (CH-2), 110.4, 116.1, 116.3, 121.4, 124.5, 128.0, 128.5, 130.9, 144.4, 144.7, 157.3, 159.1, 159.5 (aromatic carbons), 161.7 (C = O); Anal. Calcd for C17H13BrFN3OS: C, 50.26; H, 3.23; N, 10.34. Found: C, 50.49; H, 3.29; N, 10.50.

8-Bromo-2-(2-hydroxyphenyl)-7,9-dimethyl-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-one (7c)

Yield: 74%; mp: >300 °C; IR (cm−1): 3421, 3390, 3309 (NH/OH), 2924, 2854 (CH-aliphatic), 1662 (C = O); 1H NMR (DMSO-d6) δ ppm 2.62 (s, 3H, CH3), 2.73 (s, 3H, CH3), 6.06–6.08 (dd, 1H, CH-2, J= 6.0 Hz, J= 2.8 Hz), 6.77 (d, 1H, NH, J= 6.4 Hz, D2O exchangeable), 6.80–7.41 (m, 4H, Ar-H), 7.99 (d, 1H, NH, J= 2.8 Hz, D2O exchangeable), 9.99 (s, 1H, OH, D2O exchangeable); MS m/z: 403 [(M + 2-H2)+, 19.99%], 401 [(M-H2)+, 16.19%], 93[C6H4OH+, 25.25%], 91 [100%]; Anal. Calcd for C17H14BrN3O2S: C, 50.50; H, 3.49; N, 10.39. Found: C, 50.78; H, 3.55; N, 10.56.

8-Bromo-7,9-dimethyl-2-[2-(trifluoromethyl)phenyl]-2,3-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-one (7d)

Yield: 31%; mp: 267–268 °C; IR (cm−1): 3394, 3286 (NH), 2974, 2920 (CH-aliphatic), 1658 (C = O); 1H NMR (DMSO-d6) δ ppm 2.66 (s, 3H, CH3), 2.71 (s, 3H, CH3), 6.16 (d, 1H, CH-2, J= 6.4 Hz), 6.87 (d, 1H, NH, J= 7.2 Hz, D2O exchangeable), 7.60–7.94 (m, 4H, Ar-H), 8.37 (s, 1H, NH, D2O exchangeable); MS m/z: 457 [(M + 2)+, 10.14%], 455 [M+, 17.43%], 403 [100%]; Anal. Calcd for C18H13BrF3N3OS: C, 47.38; H, 2.87; N, 9.21. Found: C, 47.52; H, 2.91; N, 9.44.

General procedure for the synthesis of 8-bromo-7,9-dimethyl-2-alkyl-pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-ones 8a–c

A mixture of 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 (0.6 g, 2 mmol) and the appropriate acid anhydride (5 mL) was heated under reflux for 5 h. The reaction was cooled, poured onto ice-cold water (50 mL) and left overnight. The solid formed was filtered, dried and crystallized from the suitable solvent.

8-Bromo-2,7,9-trimethylpyrido[3',2':4,5]thieno[3,2–d]pyrimidin-4(3H)-one (8a)

Crystallized from acetic acid. Yield: 72%; mp: >300 °C; IR (cm−1): 3417 (NH), 2924, 2870 (CH-aliphatic), 1670 (C = O); 1H NMR (DMSO-d6) δ ppm 2.46 (s, 3H, CH3), 2.72 (s, 3H, CH3), 3.01 (s, 3H, CH3), 12.95 (s, 1H, NH, D2O exchangeable); MS m/z: 325 [(M + 2)+, 94.00%], 323 [M+, 100%]; Anal. Calcd for C12H10BrN3OS: C, 44.46; H, 3.11; N, 12.96. Found: C, 44.70; H, 3.17; N, 13.15.

8-Bromo-7,9-dimethyl-2-(trifluoromethyl)pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (8b)

Crystallized from acetic acid. Yield: 61%; mp: 253–254 °C; IR (cm−1): 3197 (NH), 2924, 2900 (CH-aliphatic), 1662 (C = O); 1H NMR (DMSO-d6) δ ppm 2.78 (s, 3H, CH3), 3.06 (s, 3H, CH3), 12.88 (s, 1H, NH, D2O exchangeable); 13C NMR (DMSO-d6) δ ppm 18.9, 26.6 (CH3), 117.9, 120.7, 121.0, 123.0, 124.6, 146.7, 159.3, 159.9, 162.2 (CF3 and aromatic carbons), 172.4 (C = O); MS m/z: 379 [(M + 2)+, 7.71%], 377 [M+, 3.94%], 42 [100%]; Anal. Calcd for C12H7BrF3N3OS: C, 38.11; H, 1.87; N, 11.11. Found: C, 38.24; H, 1.90; N, 11.23.

8-Bromo-2-ethyl-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (8c)

Crystallized from DMF. Yield: 73%; mp: >300 °C; IR (cm−1): 3421 (NH), 2981, 2920 (CH-aliphatic), 1670 (C = O); 1H NMR (DMSO-d6) δ ppm 1.26 (t, 3H, CH3CH2, J= 7.6 Hz), 2.71 (s, 3H, CH3), 2.71–2.75 (q, 2H, CH3CH2, J= 7.6 Hz), 2.95 (s, 3H, CH3), 12.78 (s, 1H, NH, D2O exchangeable); Anal. Calcd for C13H12BrN3OS: C, 46.16; H, 3.58; N, 12.42. Found: C, 46.42; H, 3.63; N, 12.66.

8-Bromo-7,9-dimethyl-2-(2-oxopropyl)pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-one (9)

A mixture of 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 (0.6 g, 2 mmol) and ethyl acetoacetate (6 mL) was heated under reflux for 8 h. The reaction was cooled and the solid formed was filtered, washed with ethanol (2 × 25 mL), dried and crystallized from acetic acid.

Yield: 42%; mp: 266–267 °C; IR (cm−1): 3410 (NH), 2924, 2819 (CH-aliphatic), 1685, 1639 (C = O); 1H NMR (DMSO-d6) δ ppm 2.28 (s, 3H, CH3), 2.67 (s, 3H, CH3), 2.93 (s, 3H, CH3), 3.96 (s, 2H, CH2), 12.84 (s, 1H, NH, D2O exchangeable); MS m/z: 367 [(M + 2)+, 6.95%], 365 [M+, 5.19%], 43 [CH3CO+, 100%]; Anal. Calcd for C14H12BrN3O2S: C, 45.91; H, 3.30; N, 11.47. Found: C, 46.09; H, 3.37; N, 11.59.

Ethyl (8-bromo-7,9-dimethyl-4-oxo-3,4-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-2-yl)acetate (10)

A mixture of 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 (0.6 g, 2 mmol) and diethyl malonate (6 mL) was heated under reflux for 8 h. The reaction was cooled and the solid was filtered, washed with ethanol (2 × 25 mL), dried and crystallized from acetic acid.

Yield: 84%; mp: 277–278 °C; IR (cm−1): 3294 (NH), 2924, 2854 (CH-aliphatic), 1747, 1658 (C = O); 1H NMR (DMSO-d6) δ ppm 1.21 (t, 3H, CH3CH2, J= 7.2 Hz), 2.68 (s, 3H, CH3), 2.92 (s, 3H, CH3), 3.86 (s, 2H, CH2), 4.14–4.19 (q, 2H, CH3CH2, J= 7.2 Hz), 13.04 (s, 1H, NH, D2O exchangeable); MS m/z: 397 [(M + 2)+, 100%], 395 [M+, 93.05%], 352 [(M + 2- C2H5O)+, 4.65%], 350 [(M- C2H5O)+, 5.05%]; Anal. Calcd for C15H14BrN3O3S: C, 45.47; H, 3.56; N, 10.60. Found: C, 45.80; H, 3.61; N, 10.84.

(8-Bromo-7,9-dimethyl-4-oxo-3,4-dihydropyrido[3',2':4,5]thieno[3,2-d]pyrimidin-2-yl)acetic acid (11)

A mixture of the acetate ester 10 (0.4 g, 1 mmol) and 10% alc. KOH solution (1 g KOH in 10 mL ethanol) was heated under reflux for 4 h. The reaction was cooled, diluted with water (25 mL) and acidified with conc. HCl. The precipitate was filtered, dried and crystallized from DMF.

Yield: 65%; mp: >300 °C; IR (cm−1): 3433, 3248 (NH/OH), 2924, 2854 (CH-aliphatic), 1720, 1666 (C = O); 1H NMR (CF3COOD, DMSO-d6) δ ppm 2.66 (s, 3H, CH3), 2.93 (s, 3H, CH3), 3.75 (s, 2H, CH2); Anal. Calcd for C13H10BrN3O3S: C, 42.41; H, 2.74; N, 11.41. Found: C, 42.69; H, 2.72; N, 11.56.

Pim-1 kinase inhibitory activity

Materials and methods

The kinase inhibitory activity of the synthesized compounds was determined using the Kinexus compound profiling service, Canada. All the compounds were tested for their inhibitory activity against pim-1 kinase at 50 μM. The kinase used was cloned, expressed and purified using proprietary methods. Quality control testing is routinely performed to ensure compliance to acceptable standards. 33P-ATP was purchased from PerkinElmer. All other materials were of standard laboratory grade.

Pim-1 kinase protein assay

The protein kinase target profiling was executed via employing a radioisotope assay format. All the assays were performed in a prepared radioactive working area. The protein kinase profiling assays were performed at room temperature for 20–30 min in a final volume of 25 μL according to the following assay reaction components: Component 1; 5 μl of diluted active protein kinase (∼10–50 nM final concentration in the assay). Component 2; 5 μl of stock solution of substrate. Component 3; 5 μl of kinase assay buffer. Component 4; 5 μl of the test compound, Staurosporine at 1 μM or 10% DMSO. Component 5; 5 μl of 33P-ATP (250 μM stock solution, 0.8 μCi).

The assay was initiated by the addition of 33P-ATP, followed by incubation at ambient temperature for 30 min. The assay was then terminated by spotting 10 μL of the reaction mixture onto a Multiscreen phosphocellulose P81 plate. The Multiscreen phosphocellulose P81 plate was washed three times for approximately 15 min each in a 1% phosphoric acid solution. The radioactivity on the P81 plate was counted in the presence of scintillation fluid in a Trilux scintillation counter. Blank control was set up that included all the assay components except the addition of the appropriate substrate (replaced with equal volume of assay dilution buffer). The corrected activity for protein kinase target was determined by removing the blank control value. The results were displayed in terms of percent inhibition and IC50 for the most active compounds. and showed the obtained results.

Figure 4. Percent inhibition of the test compounds and staurosporine (Sta) against pim-1 kinase.

Figure 4. Percent inhibition of the test compounds and staurosporine (Sta) against pim-1 kinase.

Table 1. Results of pim-1 kinase inhibition achieved by the test compounds at 50 μM.

In vitro cytotoxic activity

Cell culture

Cancer cells from different cancer cell lines were purchased from American type Cell Culture collection (ATCC, Manassas, VA). The cell lines used in this study were human breast adenocarcinoma (MCF7), human colon adenocarcinoma (HCT116) and human prostate cancer cells (PC3). The cell lines were grown on the appropriate growth medium Dulbecco's modified Eagle's medium (DMEM) or Roswell Park Memorial Institute medium (RPMI 1640) supplemented with 100 mg/mL of streptomycin, 100 units/mL of penicillin and 10% of heat-inactivated fetal bovine serum in a humidified, 5% (v/v) CO2 atmosphere at 37 °C.

Cytotoxicity assay by 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT)

Exponentially growing cells from different cancer cell lines were trypsinized, counted and seeded at the appropriate densities (2000–10 000 cells/0.33 cm2 well) into 96-well microtiter plates. The cells were incubated in a humidified atmosphere at 37°C for 24 h. Then, the cells were exposed to different concentrations of compounds 6c, 7a, 7c, 7d, 8b and 9 (0.1, 10, 100 and 1000 μM) for 72 h. The viability of the treated cells was determined using MTT technique. The media were removed; cells were incubated with 200 μL of 5% MTT solution/well (Sigma Aldrich, St. Louis, MO) and were allowed to metabolize the dye into colored-insoluble formazan crystals for 2 h. The remaining MTT solution was discarded from the wells and the formazan crystals were dissolved in 200 μL/well-acidified isopropanol for 30 min, covered with aluminum foil and with continuous shaking using a MaxQ 2000 plate shaker (Thermo Fisher Scientific Inc, MI) at room temperature. The absorbance was measured at 570 nm using a Stat FaxR 4200 plate reader (Awareness Technology, Inc., Palm City, FL). The cell viability were expressed as percentage of control and the concentration that induces 50% of maximum inhibition of cell proliferation (IC50) was determined for each compound using Graph Pad Prism version 5 software (Graph Pad software Inc, CA)Citation32,Citation33. The results are shown in and represented graphically in .

Figure 5. IC50 in μM of compounds 6c, 7a, 7c, 7d, 8b and 9 on three cell lines.

Figure 5. IC50 in μM of compounds 6c, 7a, 7c, 7d, 8b and 9 on three cell lines.

Table 2. Results of in vitro cytotoxic screening of compounds 6c, 7a, 7c, 7d, 8b and 9 on three cell lines.

Results and discussion

Chemistry

The synthesis of the target compounds was outlined in Schemes 1–3.

The synthesis of the starting compound 4 was accomplished via alkaline hydrolysis of 3-amino-5-bromo-4,6-dimethylthieno[2,3-b]pyridine-2-carbonitrile (3). The latter compound was prepared as reported by Madkour et al.Citation30. The formation of compound 4 was confirmed by IR spectroscopy that showed the disappearance of the characteristic CN band and the appearance of C = O band at 1666 cm−1 and two NH2 forked bands at 3464–3267 cm−1. The 1H NMR spectrum of the carboxamide derivative 4 showed two exchangeable NH2 signals at δ 6.89 and δ 7.25 ppm. It is noteworthy that compound 4 was previously reported as unexpected product during acid catalyzed hydrolysis of N2,N2-di(5-methyl-2-furylmethyl)-3-amino-4,6-dimethylthieno[2,3-b]pyridine-2-carboxamideCitation31.

Reacting the 3-aminothieno[2,3-b]pyridine-2-carboxamide 4 with 2-substituted benzaldehydes in DMF and few drops of conc. HCl afforded 8-bromo-2-(2-substituted phenyl)-7,9-dimethylpyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(3H)-ones 6ad. The IR spectra of compounds 6ad showed one NH band at 3464–3309 cm−1 together with C = O band at 1670–1662 cm−1. Their 1H NMR spectra revealed the presence of one exchangeable singlet signal at δ 12.82–13.45 ppm corresponding to NH proton as well as the aromatic signals at δ 7–8 ppm. The 13C NMR spectra of 6a, 6b and 6d showed a signal at δ 161–170 ppm corresponding to C = O carbon.

On the other hand, reacting compound 4 with 2-substituted benzaldehydes in acetic acid gave 2,3-dihydropyridothienopyrimidin-4-ones 7ad. The formation of the dihydro products was confirmed by the spectral data. Thus, the IR spectra of compounds 7ad showed two NH bands at 3433–3275 cm−1 and one C = O band at 1662–1635 cm−1. A bright evidence was obtained from the 1H NMR spectra of compounds 7ad that revealed the presence of two exchangeable doublet signals at δ 6.77–7.18 ppm and δ 7.99–8.37 ppm corresponding to two NH protons as well as a doublet of doublets signal at δ 6.06–6.16 ppm corresponding to CH-2 proton. Moreover, the 13C NMR spectra of compounds 7a,b showed a signal at δ 61.8–64.8 ppm corresponding to CH-2 carbon together with the signals of the introduced aromatic carbons and the C = O carbons.

Reacting the starting compound 4 with different acid anhydrides afforded 2-methyl, 2-trifluoromethyl and 2-ethylpyridothienopyrimidinones 8ac. The IR spectroscopy confirmed the structures of 8ac through the appearance of NH band at 3421–3197 cm−1 and C = O band at 1662–1670 cm−1. Meanwhile, their 1H NMR spectra showed an NH exchangeable singlet signal at δ 12.78–12.95 ppm. The 13C NMR spectrum of compound 8b showed a signal at δ 172.4 ppm corresponding to C = O carbon.

The reaction of the amino amide derivative 4 and ethyl acetoacetate or diethyl malonate gave the 2-(2-oxopropyl) derivative 9 and the acetate derivative 10, respectively. The IR spectrum of both compounds showed two carbonyl bands at 1685 and 1639 cm−1 in case of compound 9 and 1747, 1658 cm−1 in case of compound 10. The 1H NMR spectrum of compound 9 revealed the appearance of two singlet signals at δ 2.28 ppm and δ 3.96 ppm corresponding to the 2-oxopropyl protons as well as an exchangeable singlet signal at δ 12.84 ppm indicating the cyclization of the pyrimidine ring. Whilst, the 1H NMR spectrum of compound 10 showed a singlet signal at δ 3.86 ppm assigned to the methylene proton beside triplet and quartet signals assigned to the ethyl protons.

Alkaline hydrolysis of the ester group of compound 10 using 10% alc. KOH resulted in the formation of the acetic acid derivative 11. Its IR spectrum indicated the absence of the ester C = O at 1747 cm−1 and the appearance of acidic C = O at 1720 cm−1. While, its 1H NMR spectrum revealed the absence of the characteristic triplet and quartet signals of the ester group.

Pim-1 kinase inhibitory activity

All the compounds were tested for their ability to inhibit pim-1 kinase at 50 μM using the Kinexus compound profiling service, Canada and the results (in terms of percentage inhibition and IC50 for the active compounds) were displayed in and represented graphically in .

The results indicated that six compounds exhibited highly potent pim-1 inhibitory activity in the range of 81–98%. These compounds were 6c (85%), 7a (81%), 7c (93%), 7d (98%), 8b (96%) and 9 (89%). Consequently, their IC50 were determined as: 6c (4.62 μM), 7a (1.18 μM), 7c (1.38 μM), 7d (1.97 μM), 8b (8.83 μM) and 9 (4.18 μM).

Besides, four compounds showed moderate inhibitory activity in the range of 51–66% [compounds 6b (61%), 6d (54%), 7b (66%) and 8a (51%)]. The rest of the synthesized compounds displayed poor enzyme inhibitory activity. These results were far better than those obtained with thieno[2,3-b]pyridine derivativesCitation24.

SAR study of the pyrido[3',2':4,5]thieno[3,2-d]pyrimidin-4(1H)-ones as pim-1 inhibitors indicated the following points:

  • The 2-unsubstituted derivative 5 showed poor enzyme inhibitory activity (37%).

  • Regarding the 2-(2-substituted phenyl) series (6ad), it was found that the 2-chlorophenyl derivative 6a gave poor inhibition of pim-1 kinase (36%) with IC50 >100 μM. However, other members of the same series showed moderate to potent activity (54–85%). The 2-hydroxyphenyl derivative 6c showed the highest pim-1 inhibitory activity in this series (85% inhibition of pim-1 kinase with IC50 4.62 μM).

  • The 2-(2-substituted phenyl)-2,3-dihydro series (7ad) afforded the most potent pim-1 inhibitors in this study. Members belonging to this series showed pim-1 inhibition in the range of 66-98% with IC50 values in the range of 1.18-1.97 μM. The 2-chlorophenyl derivative 7a showed the highest pim-1 kinase inhibition with IC50 of 1.18 μM (compare with 6a). Thus, it seemed that aromaticity of the pyrimidine ring was not essential for pim-1 inhibition. Further study of the effect of substitution at meta or para positions of the phenyl ring is still needed

  • The 2-alkyl derivatives 8ac exhibited great variability in their activities as pim-1 inhibitors. Thus, while the 2-methyl derivative 8a showed moderate pim-1 inhibition (51%), its replacement with 2-triflouromethyl group in 8b enhanced the activity significantly (96% inhibition and IC50 of 8.83 μM). On the other hand, increasing the chain length into 2-ethyl group (compound 8c) reduced the enzyme inhibition greatly (23%).

  • Regarding the carbonyl containing alkyl series 911, it was found that the oxopropyl derivative 9 showed potent pim-1 inhibitory activity (89% with IC50 of 4.18 μM). Nevertheless, the ethyl acetate derivative 10 and its acid derivative 11 gave poor pim-1 inhibition.

In vitro cytotoxic activity

The most active pim-1 inhibitors in this study work, namely, compounds 6c, 7a, 7c, 7d, 8b and 9 were screened for their cytotoxic activity against three cell lines using MTT methodCitation32,Citation33. The cell lines examined were the human breast adenocarcinoma (MCF7), the human colon adenocarcinoma (HCT116) and the human prostate cancer cells (PC3). The results in terms of IC50 in μM are given in and represented graphically in .

From the results, it can be concluded that MCF7 and HCT116 cell lines were more sensitive to the action of the compounds than PC3 cell line.

Compounds 7a [the 2-(2-chlorophenyl)-2,3-dihydro derivative] and 7d [the 2-(2-(trifluoromethyl)-phenyl)-2,3-dihydro derivative] displayed the most potent cytotoxic effect on the three cell lines tested with IC50 values between 18 and 38 μM. These results were consistent with their high kinase IC50 values. Whilst, compound 8b [the 2-(trifluoromethyl) derivative] showed potent cytotoxic activity on MCF7 and HCT116 cell lines and moderate cytotoxic effect on PC3 cell line.

Both compounds 6c and 7c exhibited moderate cytotoxic effect on all the cell lines tested, whilst compound 9 displayed weak cytotoxic activity on the three cell lines.

Again, the results obtained here were better than those obtained with thieno[2,3-b]pyridine derivatives.

Conclusion

Structure rigidification via ring closure proved to be a successful strategy to improve the pim-1 inhibitory activity as well as the cytotoxic activity of thieno[2,3-b]pyridines. In the present work, four series of pyridothienopyrimidin-4-one derivatives were designed and prepared as pim-1 inhibitors. While only one thieno[2,3-b]pyridine derivative displayed potent pim-1 inhibition with IC50 of 12.71 μM, six pyridothienopyrimidin-4-ones (6c, 7a, 7c, 7d, 8b and 9) showed highly potent pim-1 inhibitory activity with IC50 of 4.62, 1.18, 1.38, 1.97, 8.83 and 4.18 μM, respectively. SAR study of pyridothienopyrimidin-4-ones indicated that the 2-(2-substituted phenyl)-2,3-dihydro series 7ad afforded the most potent pim-1 inhibitors. The most active compounds were tested for their cytotoxic activity on three cell lines [MCF7, HCT116 and PC3]. Compounds 7a [the 2-(2-chlorophenyl)-2,3-dihydro derivative] and 7d [the 2-(2-(trifluoromethyl)-phenyl)-2,3-dihydro derivative] exhibited the most potent cytotoxic activity on the three cell lines tested. A significant improvement of the cytotoxic activity was also noticed relative to the precursors thieno[2,3-b]pyridine derivatives. The results of the cytotoxicity were in good agreement with the pim-1 IC50 values. Further work on pyridothienopyrimidin-4-ones is still needed to obtain more potent pim-1 inhibitors and to improve the physicochemical properties of these derivatives.

Acknowledgements

The authors are grateful to Kinexus lab, Canada, for performing the kinase inhibitory assays.

Disclosure statement

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this article.

References

  • Le BT, Kumarasiri M, Adams JRJ, et al. Targeting Pim kinases for cancer treatment: opportunities and challenges. Future Med Chem 2015;7:35–53.
  • Narlik-Grassow M, Blanco-Aparicio C, Carnero A. The PIM family of serine/threonine kinases in cancer. Med Res Rev 2014;34:136–59.
  • Nawijn MC, Alendar A, Berns A. For better or for worse: the role of Pim oncogenes in tumorigenesis. Nat Rev Cancer 2011;11:23–34.
  • Cuypers HT, Selten G, Quint W, et al. Murine leukemia virus-induced T-cell lymphomagenesis: integration of proviruses in a distinct chromosomal region. Cell 1984;37:141–50.
  • Tursynbay Y, Zhang J, Li Z, et al. Pim-1 kinase as cancer drug target: an update (Review). Biomed Rep 2016;4:140–6.
  • Keane NA, Reidy M, Natoni A, et al. Targeting the Pim kinases in multiple myeloma. Blood Cancer J 2015;5:e325.
  • Foulks JM, Carpenter KJ, Luo B, et al. A small-molecule inhibitor of pim kinases as a potential treatment for urothelial carcinomas. Neoplasia 2014;16:403–12.
  • Decker S, Finter J, Forde A, et al. PIM kinases are essential for chronic lymphocytic leukemia cell survival (PIM2/3) and CXCR4-mediated microenvironmental interactions (PIM1). Mol Cancer Ther 2014;13:1231–45.
  • Lu J, Zavorotinskaya T, Dai Y, et al. Pim2 is required for maintaining multiple myeloma cell growth through modulating TSC2 phosphorylation. Blood 2013;122:1610–20.
  • Drygin D, Haddach M, Pierre F, Ryckman DM. Potential use of selective and nonselective pim kinase inhibitors for cancer therapy. J Med Chem 2012;55:8199–208.
  • Guo S, Mao X, Chen J, et al. Overexpression of Pim-1 in bladder cancer. J Exp Clin Cancer Res 2010;29:161–7.
  • Brault L, Gasser C, Bracher F, et al. PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica 2010;95:1004–15.
  • Merke AL, Meggers E, Ocker M. PIM1 kinase as a target for cancer therapy. Expert Opin Investig Drugs 2012;21:425–36.
  • Morwick T. Pim kinase inhibitors: a survey of the patent literature. Expert Opin Ther Patents 2010;20:193–212.
  • Arunesh GM, Shanthi E, Krishna MH, et al. Small molecule inhibitors of PIM1 kinase: July 2009 to February 2013 patent update. Expert Opin Ther Patents 2014;24:5–17.
  • Qian K, Wang L, Cywin CL, et al. Hit to lead account of the discovery of a new class of inhibitors of Pim kinases and crystallographic studies revealing an unusual kinase binding mode. J Med Chem 2009;52:1814–27.
  • Xie Y, Bayakhmetov S. PIM1 kinase as a promise of targeted therapy in prostate cancer stem cells (Review). Mol Clin Oncol 2016;4:13–17.
  • Keeton EK, McEachern K, Dillman KS, et al. AZD1208, a potent and selective pan-Pim kinase inhibitor, demonstrates efficacy in preclinical models of acute myeloid leukemia. Blood 2014;123:905–13.
  • Siddiqui-Jain A, Drygin D, Streiner N, et al. CX-4945, an orally bioavailable selective inhibitor of protein kinase CK2, inhibits prosurvival and angiogenic signaling and exhibits antitumor efficacy. Cancer Res 2010;70:10288–98.
  • Pierre F, Stefan E, Nédellec AS, et al. 7-(4H-1,2,4-Triazol-3-yl)benzo[c][2,6]naphthyridines: a novel class of Pim kinase inhibitors with potent cell antiproliferative activity. Bioorg Med Chem Lett 2011;21:6687–92.
  • Garcia PD, Langowski JL, Wang Y, et al. Pan-PIM kinase inhibition provides a novel therapy for treating hematological cancers. Clin Cancer Res 2014;20:1834–45.
  • Burger MT, Han W, Lan J, et al. Structure guided optimization, in vitro, in vivo activity, and activity of Pan-PIM kinase inhibitors. ACS Med Chem Lett 2013;4:1193–7.
  • Qian KC, Wang L, Hickey ER, et al. Structural basis of constitutive activity and a unique nucleotide binding mode of human Pim-1 Kinase. J Biol Chem 2005;280:6130–7.
  • Naguib BH, El-Nassan HB. Synthesis of new thieno[2,3-b]pyridine derivatives as pim-1 inhibitors. J Enzyme Inhib Med Chem 2016;31:1718–25.
  • Xiang Y, Hirth B, Asmussen G, et al. The discovery of novel benzofuran-2-carboxylic acids as potent Pim-1 inhibitors. Bioorg Med Chem Lett 2011;21:3050–6.
  • Abbott Laboratories. Pim kinase inhibitors as cancer chemotherapeutics. WO2008082839; 2008.
  • Tao Z-F, Hasvold L, Leverson JD, et al. Discovery of 3H-Benzo[4,5]thieno[3,2-d]pyrimidine-4-ones as potent, highly selective, and orally bioavailable inhibitors of the human protooncogene proviral insertion site in moloney murine leukemia virus (PIM) kinases. J Med Chem 2009;52:6621–36.
  • Exelixis, Inc. Benzofuropyrimidinones as protein kinase inhibitors. WO2009086264; 2009.
  • Tsuhako AL, Brown DS, Koltun ES, et al. The design, synthesis, and biological evaluation of PIM kinase inhibitors. Bioorg Med Chem Lett 2012;22:3732–8.
  • Madkour HMF, Afify AAE, Abdalha AA, et al. Synthetic utility of enaminonitrile moiety in heterocyclic synthesis: synthesis of some new thienopyrimidines. Phosphorus Sulfur Silicon Relat Elem 2009;184:719–32.
  • Stroganova TA, Vasilin VK, Zelenskaya EA, et al. Some transformations of tertiary N-furfurylamides of aromatic and heteroaromatic carboxylic acids under acidic conditions. Synthesis 2008;19:3088–98.
  • Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55–63.
  • Scudiere DA, Shoemaker RH, Paul KD, et al. Evaluation of a soluble Tetrazolium/Formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Res 1988;48:4827–33.