Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 15.
Published in final edited form as: Eur J Med Chem. 2020 Dec 31;212:113125. doi: 10.1016/j.ejmech.2020.113125

Novel cyanothiouracil and cyanothiocytosine derivatives as concentration-dependent selective inhibitors of U87MG glioblastomas: Adenosine receptor binding and potent PDE4 inhibition

Zafer Sahin a,*, Sevde Nur Biltekin b, Leyla Yurttas c, Barkin Berk a, Yağmur Özhan d, Hande Sipahi d, Zhan-Guo Gao e, Kenneth A Jacobson e, Şeref Demirayak a
PMCID: PMC7880896  NIHMSID: NIHMS1658923  PMID: 33422981

Abstract

Thiouracil and thiocytosine are important heterocyclic pharmacophores having pharmacological diversity. Antitumor and antiviral activity is commonly associated with thiouracil and thiocytosine derivatives, which are well known fragments for adenosine receptor affinity with many associated pharmacological properties. In this respect, 33 novel compounds have been synthesized in two groups: 24 thiouracil derivatives (4a–x) and 9 thiocytosine derivatives (5a–i). Antitumor activity of all the compounds was determined in the U87 MG glioblastoma cell line. Compound 5e showed an anti-proliferative IC50 of 1.56 μM, which is slightly higher activity than cisplatin (1.67 μM). The 11 most active compounds showed no signficant binding to adenosine A1, A2A or A2B receptors at 1 μM. Brain tumors express high amounts of phosphodiesterases. Compounds were tested for PDE4 inhibition, and 5e and 5f showed the best potency (5e: 3.42 μM; 5f: 0.97 μM). Remakably, those compounds were also the most active against U87MG. However, the compounds lacked a cytotoxic effect on the HEK293 healthy cell line, which encourages further investigation.

Keywords: thiouracil, thiocytosine, adenosine, U87, antitumor, phosphosdiesterase, PDE, HEK293, MCF7

Graphical Abstract

graphic file with name nihms-1658923-f0001.jpg

1. Introduction

Purine and pyrimidine heterocycles are important chemical moieties that constitute structural components of nucleotides. Both of these bases themselves and their nucleoside phosphate structures have functions on various receptors and thus on physiological processes. Many of these substances themselves cannot be used as drugs, as they do not remain in circulation for a sufficient time. However, through development based on these structures, purine and pyrimidine receptors have been considered as drug targets [1]. Different pharmacological activities have been investigated on these chemical groups. Especially, effects of cyanothiouracil derivatives, such as anti-inflammatory [2], antidiabetic [3, 4], antitumor [513] and antifungal [14, 15] activities, have been discovered in some recent studies. Previously, thiouracil derivatives have been investigated on different cancer cell lines in vitro, but there is need to study their effects on brain tumor cells. As shown in Figure 1, cyanothiouracil derivatives compound I, II and III showed moderate inhibition on MCF7, HOP92 and HC116 cell lines. Purine and pyrimidine bases are found to be effective on phosphodiesterases, adenosine receptors and kinases. Glioblastomas expresses high amounts of these macromolecules, so purine and pyrimidine derivatives can be considered as a potential source of inhibitors of proliferation. These studies have encouraged efforts on cyanothiouracils and cyanothiocytosines. The 4-oxo forms are preferred over hydroxy forms when representing cyanothiouracils; whereas, amino forms are preferred over imino forms for cyanothiocytosines, with respect to tautomeric states described for 4-oxo and 4-amino pyrimidines [16, 17].

Figure 1.

Figure 1.

Synthesized compounds and pharmacologically active molecules bearing the same fragments.

Compounds similar to cyanothiocytosine, where the 4-oxo group of thiouracil is replaced with an amino group, have mainly been investigated for adenosine (A1, A2A, A2B, A3) receptor binding affinities and pharmacological activities in such conditions as cancer, heart failure, etc. [1, 1820]. Capadenoson (IV), a selective adenosine A1 agonist, was evaluated for heart failure but withdrawn from clinical trials due to side effects. LUF6941 (V) is a selective A1 agonist with enhanced residence time. VI is another selective A1 agonist with a binding Ki value of 1.9 nM. Furthermore, these derivatives have potential as antitumor agents, as they are bearing a pyrimidine core, which is commonly studied on different cell lines, but not extensively studied on brain tumor cells.

Adenosine receptor agonist and antagonist ligands typically contain a nitrogenous ring and an exocyclic amino group. This NH or NH2 group forms a hydrogen bond with an Asn amino acid residue in the binding site of adenosine receptors, as was shown for adenosine itself in an X-ray crystallograpic structure [21]. Also, most of the early generation of adenosine agonists contain a ribose sugar that will interact with a hydrophilic region of the binding site in the center of the transmembrane helical bundle. In addition, adenosine receptor agonists and partial agonists that do not contain ribose moieties (non-ribose nucleoside analogs) have been introduced in the literature in recent years [20, 22, 23]. Adenosine A1 and A3 agonists and A2A and A2B antagonists show antitumor activity, with respect to their secondary messengers. Selective inhibitors of each subtype have been investigated for their antitumor potential in vitro and/or in vivo.

Glioblastomas are responsible for almost 75% of aggressive malignant brain tumors. In the United States, approximately ten thousand people are diagnosed with malignant glioma annually, and survival of only 1 year is expected for 50% of these patients [24]. U87MG (Uppsala 87 Malignant Glioma) human glioma primary brain tumor cells are from an aggressive type of brain tumor and express high levels of adenosine receptors (especially A2 and A3), phosphodiesterase enzymes (especially subtype 4A) and kinases, and only a few drugs are being used to treat this tumors. Temozolomide is one of the primary options, which becomes ineffective upon the development of resistance, and U87 glioblastomas may also produce resistance against cisplatin and other effective drugs [2527]. Thus, combination therapy or discovery of novel potent molecules is required for an effective treatment [28, 29]. Kinases (mainly epidermal growth factor receptor, EGFR) and phosphodiesterases are potential targets for inhibition of U87MG cell line proliferation. Rolipram, a potent phosphodiesterase 4 inhibitor, potently inhibited U87 cell proliferation, but the mechanism of action is still unknown. It is still unclear whether the activity is related to phosphodiesterase or kinases [3032].

In this study, a series of novel cyanothiouracil and cyanothiocytosine derivatives (4a–x, 5a–i) (Table 1) were synthesized, characterized and evaluated for their antiproliferative potential against glioblastoma tumor (U87MG) and human embryonic kidney (HEK293) healthy cell lines. The 11 most potent compounds (4d, 4l, 5a–i) were subjected to adenosine A1, A2A and A2B receptor binding assays at a 1 μM concentration to determine if there is an interaction with these receptors. Antitumor activity could not be associated with adenosine receptors, and subsequently the active compounds were subjected to a phosphodiesterase 4 assay and an additional MTT assay in MCF7 cells, a common breast cancer cell line that does not express high levels of adenosine receptors or the same type of phosphodiesterases as U87MG cells [33]. Briefly, we evaluated in vitro whether the 4-oxo and/or 4-amino type novel non-nucleoside analogs display antitumor activity, and mechanism of action was investigated in PDE4 and adenosine receptor assays.

Table 1.

IC50 (mean ± SD) values of synthesized compounds and cisplatin on HEK293 and U87MG cells.

graphic file with name nihms-1658923-t0002.jpg
IC50 (μM)
Compound Ring R1 R2 U87MG HEK293
4a A H H 56.61 ± 0.02 93.54 ± 5.10
4b A 3-OMe H 59.72 ± 0.04 90.24 ± 3.63
4c A 4-OMe H 48.36 ± 0.01 76.24 ± 6.59
4d A 3,4-diOMe H 15.33 ± 0.03 91.77 ± 3.29
4e A 3-Cl H 28.21 ± 0.06 ND
4f A 4-Cl H 43.35 ± 0.05 ND
4g A 3,4-diCl H 65.04 ± 0.01 85,07 ± 0.33
4h A 3,4-OCH2O- H 33.69 ± 0.02 ND
4i A H 4-OMe 57.25 ± 0.02 ND
4j A 3-OMe 4-OMe 15.34 ± 0.06 95.39 ± 5.16
4k A 4-OMe 4-OMe 25.27 ± 0.03 ND
41 A 3,4-diOMe 4-OMe 12.47 ± 0.03 23.30 ± 3.14
4m A 3-Cl 4-OMe 78.90 ± 0.02 ND
4n A 4-Cl 4-OMe 45.93 ± 0.01 63.01 ± 0.46
4o A 3,4-diCl 4-OMe 68.47 ± 0.02 45.60 ± 2.51
4p A 3,4-OCH2O- 4-OMe 65.28 ± 0.02 ND
4q A H 4-Cl 27.57 ± 0.03 ND
4r A 3-OMe 4-Cl 40.97 ± 0.02 77.93 ± 0.41
4s A 4-OMe 4-Cl 35.49 ± 0.03 ND
4t A 3,4-diOMe 4-Cl 38.64 ± 0.05 ND
4u A 3-Cl 4-Cl 40.13 ± 0.05 30.05 ± 1.92
4v A 4-Cl 4-Cl 53.01 ± 0.01 75.07 ± 9.24
4w A 3,4-diCl 4-Cl 83.89 ± 0.01 46.89 ± 1.72
4x A 3,4-OCH2O- 4-Cl 55.53 ± 0.01 81.44 ± 4.41
5a B H H 28.95 ± 0.02 46.69 ± 4.65
5b B 4-OMe H 13.98 ± 0.03 ND
5c B 4-Cl H 10.80 ± 0.08 ND
5d B H 4-OMe 8.54 ± 0.04 ND
5e B 4-OMe 4-OMe 1.56 ± 0.09 ND
5f B 4-Cl 4-OMe 4.54 ± 0.06 39.16 ± 1.38
5g B H 4-Cl 11.42 ± 0.04 ND
5h B 4-OMe 4-Cl 10.57 ± 0.04 ND
5i B 4-Cl 4-Cl 3.42 ± 0.06 72.96 ± 2.33
cisplatin 1.67 ± 0.04 8.44 ± 0.86

(ND: Not determined. Means viability is higher than 50% at 100 μM)

2. Results

2.1. Chemistry

There are several structures that have characteristic peaks in the FT-IR spectra of the final compounds. A C≡N triple bond stretch band was observed in all compounds in the range of 2200–2210 cm−1. Bands belonging to the carbonyl group were observed in the 1600–1700 cm−1 range for all final compounds. In compounds with methoxy and methylenedioxy groups, characteristic C–O single bond stretch bands were observed between 1200–1300 cm−1. First aliphatic and then aromatic C–H stretch bands were observed in the range of 2800–3000 cm−1. The NH group of ring A, as seen in one tautomeric representation, could be observed in some of the IR spectra. The N–H stretch bands of the NH2 group in the group of compounds 5ai were observed around 3300 cm−1.

The hydrogens in the compounds can be seperated in 3 main groups as aliphatic, aromatic and those bound to heteroatoms. The first aliphatic group is a methyl group attached to the thiazole ring. Proton NMR peaks belonging to this group were observed in all compounds in the range of 2.6–2.7 ppm as singlets with a 3H integration. Other aliphatic hydrogens observed belong to the methylene (CH2) group in the neighborhood of sulfur and carbonyl. The hydrogens of this group were observed for all compounds as singlets with a 2H integration in the range of 4.45–4.7 ppm. The compounds 4bd, 4i, 4rt, 5b, 5d5f, and 5h carry a methoxy (OCH3) group. These methyl groups attached to oxygen were observed in the range of 3.6–3.85 ppm as a singlet with 3H integration (Figure 2). In the compounds 4d, 4l and 4t carrying 3,4-dimethoxyphenyl group, two of the methoxy groups overlapped, and a singlet was observed with 6H integration. CH2 hydrogens in compounds 4h, 4p and 4x carrying methylenedioxy groups were observed in the 5.85–5.92 ppm range with 2H integration.

Figure 2.

Figure 2.

1H NMR interpretation summary of the compounds

Aromatic hydrogens belonging to the A and B rings (Figure 2) were observed in the range of 6.7–8.0 ppm. While the peaks in the compounds with a methoxy (OCH3) group are approaching 6.7 ppm due to their electron donor property, in the compounds containing a Cl substituent approached 8 ppm due to the electron withdrawing effect. In 4-substituted phenyl derivatives, peaks were observed as doublet with 2H integration, representing 1–4 splitting, except for overlapping signals. Since the compounds 4f, 4n, 4q–x, 5c and 5f–i carry 4-Cl substituents, two peaks were observed as a doublet in the aromatic region and with 2H integration in these derivatives. In some compounds, peaks could not be differentiated due to overlaps, and multiplets were observed. NH2 or OH protons defined as X in the Figure, were not observed possibly due to the tautomeric structure. The aliphatic and aromatic hydrogen numbers were provided as observed for all compounds, and the structure of the compounds was assured.

For interpretation of 13C NMR spectral results, it is possible to group the carbon atoms in compounds into several groups. The first of these is the CH3 carbon in thiazole ring at position 4. The peak for this carbon was observed at 18.7 ppm in all compounds. It is expected that the methylene group between the sulfur and carbonyl group in the structure would be observed in the 38–40 ppm range. Since the spectra are taken in DMSO, the solvent peaks are also observed in this range in proximity to the CH2 carbon. The peak of OCH3 carbon was observed around 55 ppm in all compounds containing methoxy groups. In compounds containing more than one methoxy, these peaks coincided in some compounds, and it was possible to observe them separately in others. The carbon atom to which the cyano group is bound in the A ring, is expected and found in the range of 80–100 ppm. In compounds 4h, 4p and 4x containing methylenedioxy groups attached to the aromatic ring, the O-CH2-O carbon was observed in the range of 102–108 ppm. The carbon atom belonging to the CN group was observed at ~115 ppm. There are 4 aromatic rings in the compounds, two of which are phenyl and the other two are heteroaromatic rings. As expected, phenyl ring carbons were detected between 110 and 148 ppm. The carbon atoms of the heteroaromatic pyrimidine and thiazole rings were observed in the range of 150–170 ppm. These carbons can sometimes be observed overlapping due to their symmetrical state and the shading effect in three-dimensional positioning. Finally, the peak of the carbonyl group, one of the important determining groups of our compounds, was detectable in the range of 180–190 ppm. High resolution mass spectra gave the M+1 peaks accurately.

2.2. Biological Activity

2.2.1. U87MG and HEK293 cellular antitumor and cytotoxic activity measurement

The compounds’ antitumor and cytotoxic activity is summarized in Table 1. Accordingly, most of the compounds showed anti-proliferative activity on U87MG aggresive glioblastoma. Furthermore, almost all compounds seem did not attenuate the proliferation of healthy, noncancerous human embyonic kidney (HEK)293 cell line.

As seen in the effects of the compounds over a concentration range, the degree of inhibition increased with concentration (Table 2). At a concentration of 5 μM, compounds 5b–i were able to reduce viability by 70%. Compounds 5b–i at a concentration of 10 μM showed a modest increase compared to the 5 μM concentration, while only 4d, 4l and 4j from the 4a–x group caused <70% viability. Although compounds 4a–x and 5a at 50 μM showed a significantly increased antiproliferative effect, on average it remained above 45% (excluding 4d, 4l and 4j), while compounds between 5d–i reduced the viability to <20% on average. A significant increase in the efficacy of all compounds was observed at a 100 μM concentration, and compounds 4d, 5e, 5h and 5i produced an even greater effect than the standard cisplatin (Figure 3, Table 2).

Table 2.

% Viability values ± SD of synthesized compounds (5μM, 10μM, 50μM, 100μM) and cisplatin against U87MG tumor cells.

Compound No. 5 μM 10 μM 50 μM 100 μM
4a 92.10 ± 4.14 87.5 ± 2.12 77.5 ± 2.17 9.23 ± 1.09
4b 99.00 ± 3.33 90.2 ± 2.20 80.3 ± 4.10 21.8 ± 2.77
4c 97.30 ± 0.62 90.4 ± 0.77 52.1 ± 1.86 1.08 ± 0.45
4d 80.80 ± 6.19 69.8 ± 0.37 21.4 ± 3.52 6.48 ± 0.60
4e 86.90 ± 1.67 83.7 ± 3.58 50.6 ± 2.63 10.0 ± 1.79
4f 90.90 ± 2.11 85.2 ± 4.29 63.4 ± 3.37 22.5 ± 2.25
4g 98.60 ± 6.68 92.5 ± 2.32 75.3 ± 1.84 24.4 ± 2.11
4h 93.40 ± 4.44 84.2 ± 2.61 42.6 ± 2.39 18.7 ± 2.53
4i 99.00 ± 3.48 90.9 ± 0.84 72.5 ± 2.83 10.9 ± 4.02
4j 71.30 ± 1.10 66.7 ± 1.54 43.2 ± 1.18 12.4 ± 0.78
4k 97.00 ± 6.31 80.3 ± 0.96 32.6 ± 2.25 11.5 ± 3.86
4l 81.20 ± 3.45 57.6 ± 3.49 22.2 ± 2.57 5.39 ± 1.22
4m 95.7 ± 3.53 90.6 ± 3.17 83.4 ± 3.72 40.6 ± 3.08
4n 102.00 ± 9.35 92.7 ± 2.95 48.0 ± 2.64 2.85 ± 0.87
4o 101.00 ± 6.46 88.6 ± 1.97 78.4 ± 3.04 31.1 ± 2.80
4p 100.00 ± 4.02 88.3 ± 2.76 83.2 ± 3.65 22.9 ± 2.28
4q 88.9 ± 3.72 83.1 ± 2.62 41.6 ± 1.47 11.4 ± 1.58
4r 96.6 ± 4.09 89.7 ± 1.45 47.6 ± 1.30 8.54 ± 2.66
4s 92.1 ± 2.73 88.3 ± 3.64 46.3 ± 3.06 16.6 ± 1.40
4t 93.4 ± 0.80 83.3 ± 3.18 59.8 ± 3.56 18.7 ± 1.54
4u 80.2 ± 5.63 95.2 ± 3.05 52.6 ± 2.77 12.5 ± 2.26
4v 99.00 ± 4.97 98.4 ± 0.59 60.0 ± 1.71 12.8 ± 3.65
4w 101.00 ± 3.46 97.1 ± 1.02 93.9 ± 1.11 36.5 ± 3.66
4x 101.00 ± 3.76 95.0 ± 3.09 68.3 ± 2.86 9.91 ± 1.20
5a 103.00 ± 3.08 93.0 ± 2.95 26.3 ± 3.17 6.48 ± 1.70
5b 68.8 ± 3.46 60.8 ± 3.66 33.8 ± 2.51 17.2 ± 1.21
5c 62.9 ± 1.93 58.8 ± 1.24 44.3 ± 3.90 17.1 ± 2.84
5d 61.8 ± 3.38 57.8 ± 2.40 16.5 ± 1.27 5.25 ± 0.37
5e 35.6 ± 2.95 28.7 ± 2.26 13.0 ± 1.75 0.26 ± 0.45
5f 51.4 ± 1.25 44.0 ± 2.67 23.1 ± 3.02 3.90 ± 0.79
5g 71.1 ± 3.57 67.9 ± 2.28 15.2 ± 2.87 4.59 ± 1.06
5h 69.7 ± 0.78 64.5 ± 3.08 17.8 ± 2.40 0.79 ± 0.79
5i 46.1 ± 1.95 37.3 ± 3.52 15.5 ± 2.85 1.65 ± 0.87
Cisplatin 31.7 ± 1.53 18.2 ± 1.47 6.71 ± 1.64 2.25 ± 1.86
Figure 3.

Figure 3.

Effect of increasing concentrations of 5b–i on viability of U87MG cells.

In general, compounds 5b–i were significantly more active than the rest of the series (4a–x) except for 4d, 4j, and 4l. Unlike the 4a–x group, these compounds (5a–i) carry the amino group instead of the oxo at the 4th position of the pyrimidine ring. Compound 5a also contains an amino group at the 4 position, but its aromatic rings at both ends of the compound are unsubstituted. This compound showed reduced inhibition compared to 5b–i. Concentration dependent viability results for all synthesized molecules are summarized in Table 2.

With respect to the effects of the compounds on the HEK293 healthy cell line, 50% inhibition was not observed for 16 of the 33 synthesized compounds even at 100 μM. The IC50 values of the remaining molecules were quite high, that is, the cytotoxic effects of the molecules were very low. This situation is remarkable and important in terms of antitumor drug properties. Of the compounds, in particular 5e showed an IC50 of 1.56 μM against U87MG, while the IC50 of cisplatin was 1.67 μM (Table 1). Cisplatin also showed an IC50 of 8.44 μM on the healthy HEK293 cell line. Compound 5e had no cytotoxic effect even at high concentrations. Apart from that, while 5i: 3.42 μM, 5f: 4.54 μM and 5d had an IC50 of 8.54 μM against U87MG cells, these molecules did not show any cytotoxic effect in HEK293 cells. Although the compounds do not have significant effects in most of the series at 5 μM concentration, as seen in Figure 3, 5b–i compounds showed higher effects compared to the 4a–x group. Although there was no significant increase in the effect at 10 μM, the efficiency of compound 5e became evident and became closer to cisplatin. At a concentration of 50 μM, the active and non-active compounds were clearly delineated. Accordingly, 11 compounds, 4d, 4l, and 5a–i, were considered active and were tested for adenosine receptor binding affinity.

2.2.2. MCF7 cellular antitumor activity measurement

As shown in Table 3, the compounds displayed an antiproliferative effect on MCF7 cell line. Although the IC50 values were considerable, the anticancer activity was not high as U87MG cell line. Different cell lines have different levels of specific proteins such as enzymes and receptors that may affect proliferation. In general, cancer cell lines typically highly expresss proteins that are considered as cancer drug targets.

Table 3.

IC50 (mean ± SD) values of compounds and cisplatin on MCF7.

Compound IC50 (μM)
4d 63.71 ± 1.92
4l 24.02 ± 0.95
5a 30.14 ± 1.62
5b 27.15 ± 0.95
5c 20.17 ± 0.75
5d 72.12 ± 2.33
5e 24.31 ± 1.95
5f 11.25 ± 0.09
5g 25.22 ± 1.17
5h 10.09 ± 0.03
5i 12.52 ± 1.13
Cisplatin 2.51 ± 0.29

The MCF7 cell line is known to express kinases and phosphodiesterases, but the range of isoforms present may change from a cell line to the other. For example, brain tumors like glioblastomas usually expresses higher amounts of PDE4B and PDE7 than MCF7 cells. The lower anticancer activity observed is possibly due to this situation. The choice of a cell line to study after U87MG will require consideration of the mechanism of action. Among the compounds tested on MCF7 cells, 5f, 5h and 5i showed higher activity than the other 8 compounds.

2.2.3. Measurement of adenosine receptor binding affinity

The relationship of adenosine ligands with antitumor activity is mainly on glioblastomas. It is known that adenosine concentration (1.5 μM) in extracellular fluid of glioblastomas is lower than normal cells (3 μM). Antitumor effect studies are ongoing in all subtypes. The agonism of A3 receptors and the antagonism of A2A may provide antitumor activity, but the antitumor effect of A3 receptor agonists is most evident in in vivo models [34]. U87MG glioblastoma tumor cells were selected for the reasons described above to determine the pharmacological activity basis of the compounds. The degree of binding of compounds 4d, 4l, 5a–i to adenosine A1 and A2A receptor subtypes was measured. The most antitumor compound 5e additionally subjected to an A2B assay to probe the basis of its activity. According to the data given in Table 4, the values are not high enough to be considered receptor blockers, at least at a fixed concentration of 1 μM. Compounds 4d, 5a, 5e and 5g are the compounds showing the most binding inhibition (8.7–10%) at the A1 subtype. The highest degree of inhibition at the A2A receptor was observed for 4d and 4l, with 12% and 8.5% inhibition, respectively. Compound 5e showed 18% binding at 1 μM.

Table 4.

Inhibition of human A1 and A2A receptor radioligand binding by the synthesized substances (at 1 μM, expressed as percent of specific binding).

Cpd No. A1 [3H]DPCPX A2A [3H]ZM241385 Cpd No. A1 [3H]DPCPX A2A/A2B [3H]ZM241385
4d 10 12 5e 9.0 −18 / 18a
41 2.5 8.5 5f 1.1 −7.1
5a 9.5 6.5 5g 8.7 −1.4
5b −4.6 0.8 5h 1.2 −22
5c −23 −9.9 5i −3.7 1.8
5d −2.8 −2.6
a

Inhibition of A2B receptor binding by 10 μM 5e was 49%.

It seems that some of these values are negative values. In this radioligand binding method, negative (−) values are not meaningful and will be considered as zero. It was observed that compounds 4d, 4l and 5a showed weak binding at both A1 and A2A subtypes. Among the other compounds, 5g and 5h showed weak binding only on A1 receptors, 5e showed weak binding on A2A and A2B, and 5b and 5i were found to show only weak binding on A2A receptors.

Adenosine receptor binding was only evaluated at a concentration of 1 μM, and none of the compounds at this concentration showed high levels of binding. In the literature, adenosine ligands have been reported act at the nanomolar (nM) level. In addition, the A1 receptor activities of some compounds at a concentration of 10 μM were found to be similar [35, 36]. For a molecule to be considered an adenosine receptor ligand, some inhibition at the submicromolar level would be expected, so full concentration curves have not been determined. Therefore, the anticancer effect of the compounds is not associated with adenosine A1 or A2A receptor subtypes.

2.2.4. Measurement of PDE4B assay

Table 5 summarizes the IC50 values of the compounds tested for PDE4 inhibition. 3-Isobutyl-1-methylxanthine (IBMX), a general phosphodiesterase standard, had an IC50 value of 14.31 μM. Rolipram, a potent PDE4B inhibitor, showed a 0.32 μM IC50, which is consistent with the literature data. Compound 5e and 5f showed good potency on PDE4B, with 3.15 μM and 0.97 μM IC50 values respectively. These inhibitory potencies are considerably high, i.e. close to the most potent PDE4B inhibitor rolipram (Figure 4). There is a close correspondence between the most active compounds against U87MG cells and in the PDE4B assay. This parallelism is remarkable in the context of suggesting a basis of antitumor activity on U87MG.

Table 5.

IC50 (mean ± SD) values of compounds, IBMX and rolipram on PDE4B.

Cpd PDE4B IC50 (μM)
4d >100
41 17.11 ± 0.32
5a >100
5b 33.14 ± 0.13
5c 32.26 ± 0.37
5d 11.17 ± 0.12
5e 3.15 ± 0.12
5f 0.97 ± 0.35
5g 22.13 ± 0.56
5h 18.11 ± 0.34
5i 24.14 ± 2.15
Rolipram 0.32 ± 0.01
IBMX 14.31 ± 0.02
Figure 4.

Figure 4.

Logaritmic curves of active compounds on PDE assay

When comparing the chemical structures of the compounds with U87 antitumor activity, it seems that compounds 5a–i (thiocytosine derivatives) contain exocyclic amino groups in common. 4d, 4l and 4j were the only active compounds in the cyanothiouracil series 4a–x, and they have methoxyphenyl groups on the pyrimidine ring. Compound 4l and 4j also has a 4-methoxy group on the other phenyl ring attached to the thiazole. It is noteworthy that compound 5e, which is the most active in the 5a–i group, also carries 4-methoxyphenyl groups at both ends, parallel to the structures of 4d and 4l. In addition, compounds 4c, 4n, 5h, which act at 100 μM, also carry a methoxy group on one of the phenyl rings. The activity of structures bearing methylenedioxy and chlorine substituents remained relatively low. The IC50 value of temozolomide, the drug used most frequently against glioblastoma tumors, affected the U87MG cell line only in the range of 130–171 μM [37]. From this view, our compounds are important hits as they are potent and not cytotoxic.

Compounds with the highest antitumor activity did not show any parallel in adenosine A1 or A2A receptor results. The lack of correlation of adenosine receptor results and the antitumor effect precludes those receptors as the biochemical basis of the high antitumor effect. U87MG cells are in the class of glioblastomas, an important type of brain tumors. This group of tumors express phosphodiesterase 4 in particularly high amounts. Apart from that, it expresses kinase enzymes in high amounts as in all tumor types. MCF7 cells do not express PDE4 as much as U87MG. Considering the MCF7 cellular activity and phosphodiesterase enzyme results, the effect seems likely to occur through the phosphodiesterase enzyme. Compounds 5e, 5f and 5i that showed the highest effect in the U87MG cell line, were similarly the most effective compounds of the series in phosphodiesterase enzyme activity.

3. Conclusion

The importance of pyrimidine and thiouracil derivatives in drug development studies has been clearly demonstrated in recent studies. Developing resistance to temozolomide drug encourages the discovery of effective new drugs and the development of combination therapy for glioblastoma tumors [38, 39]. Thus, the current results are relevant to others working on similar chemotypes and/or pharmacological effects.

The studies conducted within the scope of this article have been important in terms of evaluating the thiouracil and thiocytosine derivatives together, investigating the biochemical bases of the antitumor effect and determining in vitro antitumor activity close to FDA approved drugs. In addition, further studies are planned to determine antiproliferative activity on other brain tumor cell lines, different healthy cell lines, the kinase phosphorylation levels in the presence of the active compounds, and subsequent in vivo experiments with a brain tumor model.

4. Experimental

4.1. Chemistry

All chemicals and biological materials were purchased from Sigma-Aldrich Chemical Corp and Honeywell Corp. All intermediate compounds were checked for their purities using thin-layer chromatography (TLC) with a Silica Gel 60 F254 TLC plate (Merck KGaA, Darmstadt, Germany). Melting points were measured by a Stuart Melting Point Apparatus SMP30 (Staffordshire, UK) and defined by recording the point where compound started to melt. Spectroscopic data were determined with the following instruments: Fourier transform IR (FT-IR), Perkin Elmer Spectrum Two FT-IR Spectrometer (Perkin Elmer, Inc, Waltham, Massachusetts, USA); 1H-NMR, Bruker 300- MHz UltraShield NMR Spectrophotometer (Bruker Corp, Billerica, Massachusetts, USA); and 13C-NMR, Bruker 75- MHz UltraShield NMR Spectrophotometer (Bruker Corp, Billerica, Massachusetts, USA) in DMSO-d6 using standard TMS. The high resolution mass spectra of the compounds were determined on a Shimadzu 8040 LC/MS/MS ITTOF system (Shimadzu, Tokyo, Japan) using a mass spectrometer with the electron spray method (ESI).

4.1.1. General synthesis of 1-(4-methyl-2-(4’-substitutedphenylthiazole-5-yl)ethane-1-one derivatives (Method A)

According to the Hantzsch method, 3-chloro-2,4-pentandione is added to the hot solution of aryl thioamide in absolute ethanol (Scheme 1) and boiled for 8 hours. After being controlled with TLC where the reaction is completed, it was left to cool, poured into cold water and neutralized by sodium acetate solution. After the precipiation was completed, it is taken and dried, crystallized from ethanol [40].

Scheme 1.

Scheme 1.

Synthesis of the compounds 1ac (i: aryl thioamide, 3-chloro-2,4-pentandione, EtOH, reflux 8 h, NaOAc; ii: product of i, CHCl3,0°C, Br2, r.t. 12 h, 30 min at 40°C

1-(4-Methyl-2-phenylthiazole-5-yl)ethane-1-one

Obtained according to Method A by the reaction of 3-chloro-2,4-pentanedione (110 mmol, 14.80 g) and thiobenzamide (100 mmol, 13.72 g). Yield: %90, m.p.: 68–69°C, m.p. reference: 68–70°C (Ethanol) [41].

1-(4-Methyl-2-(4’-methoxyphenyl)thiazole-5-yl)ethane-1-one

Obtained according to Method A by the reaction of 3-chloro-2,4-pentanedione (110 mmol, 14.80 g) and 4-methoxythiobenzamide (100 mmol, 16.72 g). Yield: 85%, m.p.: 88.8–90°C, m.p. reference: 89°C (Ethanol) [41].

1-(4-Methyl-2-(4’-chlorophenyl)thiazole-5-yl)ethane-1-one

Obtained according to Method A by the reaction of 3-chloro-2,4-pentanedione (110 mmol, 14.80 g) and 4-chlorothiobenzamide (100 mmol, 17.16 g). Yield: 90%, m.p.: 113–115°C, m.p. reference: 114–115°C (Ethanol) [42].

4.1.2. General synthesis of 2-bromo-1-(4-methyl-2-(4’-substitutedphenylthiazole-5-yl)ethane-1-one derivatives (1a–c)

The appropriate 1-(4-methyl-2-(4’substitutedphenylthiazole-5-yl)ethan-1-one derivative dissolved in chloroform and the reaction environment is brought to 0°C. After 10 minutes of stirring, bromine solution in chloroform added dropwise. Then it is stirred at room temperature for 12 hours. Then it is heated to 40°C and stirred for 30 minutes for 1b and 1c. After being controlled with TLC, where the reaction is completed, the saturated sodium bicarbonate solution is added and stirred for 10 minutes. Phases are separated and the aqueous phase is re-extracted with chloroform. After organic phase is dried using sodium sulphate, the solvent is evaporated at low pressure and then the product is obtained [43]. Since the reaction was not completed in the first trials, it was determined that there was a second bromine entry when it was heated for a long time. Bromine addition should be carried out at 0°C and the reaction should be controlled frequently, especially after heating. When the starting material was consumed the most and a second product started to form, the reaction was terminated and the product crystallized from ethanol.

2-Bromo-1- (4-methyl-2-phenyl-thiazol-5-yl) ethan-1-one (1a)

Obtained according to Method B by reaction of 1- (4-Methyl-2-phenylthiazol-5-yl) ethan-1-one (75 mmol, 16.30 g) and bromine (Br2) (75 mmol, 11.99 g). Yield: 76%, m.p.: 117.5–119°C, m.p. reference: 117–118°C (Ethanol) [44].

2-Bromo-1-(4-methyl-2- (4’-methoxyphenylthiazole-5-yl) ethan-1-one (1b)

By reaction of 1- (4-Methyl-2- (4’-methoxyphenylthiazol-5-yl) ethan-1-one (75 mmol, 18.55 g) and bromine (Br2) (75 mmol, 11.99 g), was obtained according to Method B. Yield: 80%, m.p.: 90.2–91°C, m.p. reference: 90–91°C (Ethanol) [45].

2-Bromo-1-(4-methyl-2- (4’-chlorophenylthiazole-5-yl) ethan-1-one (1c)

By reaction of 1-(4-methyl-2- (4’-chlorophenylthiazole-5-yl) ethan-1-one (75 mmol, 18.88 g) and bromine (Br2) (75 mmol, 11.99 g), was obtained according to Method B. Yield: 70%, m.p.: 117–118°C, m.p. reference: 117–118°C (ethanol) [45].

4.1.3. General synthesis of 2-mercapto-4-hydroxy-6-substitutedphenypyrimidine-5-carbonitrile derivatives (Method C)(2a–h)

In this method, ethyl cyanoacetate was used instead of methylene ketone in the Biginelli method. In addition, the reaction is not acid catalyzed, but base catalyzed (Scheme 2). Solvents such as dimethylformamide, acetone and acetonitrile are used as solvents [44]. Potassium carbonate is most commonly used as a base. The appropriate aldehyde, thiourea and ethyl cyanoacetate were dissolved in anhydrous ethanol and potassium carbonate was added. The reaction was cooled after boiling for 16 hours. The precipitated product was dissolved in 0.5 M NaOH solution. It was extracted 3 times and washed with ethyl acetate. It was then brought to pH 2 with the slow addition of a 1M HCl and the product precipitated. The product was crystallized from ethanol [2, 46, 47].

Scheme 2.

Scheme 2.

Synthesis of the compounds 2ah and 3ac (i: aldehyde, thiourea, K2CO3, ethyl cyanoacetate, EtOH, reflux 16h; ii: aldehyde, thiourea, K2CO3, malononitrile, EtOH, reflux 5h)

4-Hydroxy-2-mercapto-6-phenylpyrimidine-5-carbonitrile (2a)

By reaction of benzaldehyde (20 mmol, 2.12 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), was obtained according to Method C. Yield: %75, m.p.: 299–301°C, m.p. reference: 300–302°C (Acetic acid) [46].

4-hydroxy-2-mercapto-6-(3-methoxyphenyl)pyrimidine-5-carbonitrile (2b)

By reaction of 3-methoxybenzaldehyde (20 mmol, 2.72 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 78%, m.p.: 246–247°C, m.p. reference: 244°C (Ethanol) [48].

4-hydroxy-2-mercapto-6-(4-methoxyphenyl)pyrimidine-5-carbonitrile (2c)

By reaction of 4-methoxybenzaldehyde (20 mmol, 2.72 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 70%, m.p.: 280–282°C, m.p.: 281–283°C (Ethanol) [49].

4-(3,4-dimethoxyphenyl)-6-hydroxy-2-mercaptopyrimidine-5-carbonitrile (2d)

By reaction of 3,4-dimethoxybenzaldehyde (20 mmol, 3.32 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26g), obtained according to Method C. Yield: 80%, m.p.: 281–283°C, m.p. reference: 280–282°C (Acetic acid) [50].

4-(3-chlorophenyl)-6-hydroxy-2-mercaptopyrimidine-5-carbonitrile (2e)

By reaction of 3-chlorobenzaldehyde (20 mmol, 2.81 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 65%, m.p.: 227–228°C, m.p. reference: 229°C [51].

4-(4-chlorophenyl)-6-hydroxy-2-mercaptopyrimidine-5-carbonitrile (2f)

By reaction of 4-chlorobenzaldehyde (20 mmol, 2.81 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 85%, m.p.: 269.6–271°C, m.p. reference: 270–271°C (Acetic acid) [46]

4-(3,4-Dichlorophenyl)-6-hydroxy-2-mercaptopyrimidine-5-carbonitrile (2g)

By reaction of 3,4-dichlorobenzaldehyde (20 mmol, 3.50 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 77%, m.p.: 267–269°C, m.p. reference: 268°C [52].

4-(3,4-methylenedioxyphenyl)-6-hydroxy-2-mercaptopyrimidine-5-carbonitrile (2h)

By reaction of 3,4-methylenedioxybenzaldehyde (20 mmol, 3.00 g), thiourea (20 mmol, 1.5224 g), potassium carbonate (20 mmol, 2.76 g) and ethyl cyanoacetate (20 mmol, 2.26 g), obtained according to Method C. Yield: 87%, m.p.: 250.5–252°C, m.p. reference: 248–250°C (Ethanol) [50].

4.1.4. General synthesis of compounds 4a–x (Method D)

2a–h derivatives (100 mmol) and thiazolyl acetylbromide compounds (1a–c) (110 mmol) are dissolved in acetone and potassium carbonate (100 mmol) was added. The reaction was mixed for 12 hours at room temperature (Scheme 3). If it was not completed in this time, the reaction was boiled for 1 hour. After the reaction was completed, it was poured into cold water. The product was expected to be precipitated thoroughly. In the meantime, thorough solubility of potassium carbonate should be ensured in water. The precipitated product was filtered away. Compounds are crystallized from 80% DMSO (Dimethyl sulfoxide) - 20% water mixture. The resulted products are kept in vacuum survey seizing phosphorus pentaoxide (P2O5) at 45°C for overnight and ensured complete drying before analysis [15, 53].

Scheme 3.

Scheme 3.

Synthesis of the compounds 4a–x and 5a–i (i: 2a–h, 1a–c, acetone, K2CO3, r.t. 12h, reflux 1 h, i: 3a–c, 1a–c, acetone, K2CO3, r.t. 8 h, reflux 1 h).

2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-4-phenyl-1,6-dihydropyrimidine-5-carbonitrile (4a)

Yield: 17.21% m.p.: 210°C (decomposed). FT-IR υmax (cm−1): 3380.8 (NH), 3056–2914 (C–H), 2205 (C≡N), 1680.33 (C=O), 1555–1462 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 4.52 (2H, s, CH2), 7.29–7.37 (3H, m, Ar), 7.54 (3H, m, Ar), 7.65 (2H, d, J: 6.57 Hz, Ar), 7.98 (2H, d, J: 7.77 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.18 (CH2), 90.02 (C-CN), 119.72, 127.08, 128.37, 128.50, 129.89, 130.38, 130.77, 131.97, 132.58, 137.51, 159.44, 167.41, 169.03, 170.28, 188.04 (C=O). HRMS (M+H): For C23H16N4O2S2 Calcd: 445.0787, found: 445.0781.

4-(3-methoxyphenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4b)

Yield: 8.92% m.p.: 283°C (decomposed). FT-IR υmax (cm−1): 3397.8 (NH), 3056.5–2899 (C–H), 2206.6 (C≡N), 1675 (C=O), 1548–1446 (C=C, C=N), 1235.85 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.66 (3H, s, OCH3), 4.50 (2H, s, CH2), 6.94–6.95 (1H, m, Ar), 7.22–7.26 (3H,m, Ar), 7.50–7.56 (3H, m, Ar), 7.97–8.00 (2H, m, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 8.72 (CH3), 39.16 (CH2), 55.46 (OCH3), 90.04 (C-CN), 114.04, 115.79, 120.02, 120.77, 120.97, 126.54, 126.84, 127.05, 129.44, 129.68, 129.85, 130.64, 131.92, 132.62, 139.07, 159.26, 159.40, 167.10, 168.96, 188.16 (C=O). HRMS (M+H): For C24H18N4O3S2 calcd: 475.0893, found: 475.0890

4-(4-methoxyphenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4c)

Yield: 10.89% m.p.: 124.3°C. IR υmax (cm−1): 3394.5 (NH), 3000–2836 (C–H), 2206.6 (C≡N), 1671.6 (C=O), 1543.13–1413 (C=C, C=N), 1253.63 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.71 (3H, s, CH3), 3.67 (3H, s, OCH3), 4.45 (2H, s, CH2), 6.85 (2H, d, J: 8.62 Hz, Ar), 7.52–7.56 (3H, m, Ar), 7.65 (2H, d, J: 8.56 Hz, Ar), 7.98–8.01 (2H, m, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.76 (CH3), 39.16 (CH2), 55.53 (OCH3), 88.90 (C-CN), 113.63, 120.80, 127.05, 129.87, 130.01, 130.21, 130.86, 131.92, 132.62, 159.36, 160.81, 166.60, 168.92, 170.78, 170.85, 188.52 (C=O). HRMS (M+H): For C24H18N4O3S2 calcd: 475.0893, found: 475.0885

4-(3,4-dimethoxyphenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4d)

Yield: 39.17% m.p.: 244°C. FT-IR υmax (cm−1): 2977.8–2899 (C–H), 2206.6 (C≡N), 1658.5 (C=O), 1568.75–1416.32 (C=C, C=N), 1255.71 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.70 (6H, m, OCH3), 4.49 (2H, s, CH2), 6.88 (1H, d, J: 8.47 Hz, Ar), 7.32–7.38 (2H, m, Ar), 7.48–7.53 (3H, m, Ar), 7.96 (2H, d, J: 7.80 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.42 (CH2), 55.78 (OCH3), 55.84 (OCH3), 89.09 (C-CN), 111.24, 112.18, 120.85, 121.65, 127.02, 129.83, 130.21, 130.55, 131.91, 132.57, 148.42, 150.65, 159.50, 166.65, 168.98, 170.82, 170.99, 188.36 (C=O). HRMS (M+H): For C25H20N4O4S2 calcd: 505.0999, found: 505.0988

4-(3-chlorophenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4e)

Yield: 61.86% m.p.: 231°C. FT-IR υmax (cm−1): 3374.8 (NH), 2990.9–2902.3 (C–H), 2203.3 (C≡N), 1673.95 (C=O), 1560.23–1423.97 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 4.47 (2H, s, CH2), 7.37–7.41 (2H, m, Ar), 7.51–7.54 (3H, m, Ar), 7.55–7.64 (2H, m, Ar), 7.96–7.98 (2H, m, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.44 (CH2), 89.87 (C-CN), 120.09, 127.05, 128.12, 129.82, 129.96, 130.28, 131.89, 132.64, 133.34, 139.87, 159.39, 165.57, 169.00, 170.41, 171.35, 188.21 (C=O). HRMS (M+H): For C23H15ClN4O2S2 calcd: 479.0398, found: 479.0391.

4-(4-chlorophenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4f)

Yield: 70.36% m.p.: 314°C. FT-IR υmax (cm−1): 3371.6 (NH), 2987.6–2895.7 (C–H), 2209.8 (C≡N), 1658.5 (C=O), 1583–1417.96 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 4.43 (2H, s, CH2), 7.35 (2H, dd, J: 6.72 Hz, j: 1.86 Hz, Ar), 7.51–7.53 (3H, m, Ar), 7.65 (2H, dd, J: 6.70 Hz, j: 1.88 Hz, Ar), 7.93–7.96 (2H, m, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.72 (CH3), 39.37 (CH2), 89.75 (C-CN), 120.18, 127.02, 128.42, 129.83, 130.18, 130.76, 131.89, 132.54, 134.91, 136.58, 159.42, 166.14, 168.98, 170.64, 171.36, 188.45 (C=O). HRMS (M+H): For C23H15ClN4O2S2 calcd: 479.0386, found: 479.0386

4-(3,4-dichlorophenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4g)

Yield: 61.20% m.p.: 231.5°C. FT-IR υmax (cm−1): 3384.7 (NH), 2990.9–2902.3 (C–H), 2209.8 (C≡N), 1661.8 (C=O), 1567.83–1420.31 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.45 (2H, s, CH2), 7.48–7.56 (3H, m, Ar), 7.59 (1H, s, Ar), 7.66 (2H, dd, J: 8.4 Hz, j: 2.04 Hz, Ar), 7.75–7.80 (1H, m, Ar), 7.94–7.97 (2H, m, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.40 (CH2), 89.87 (C-CN), 119.94, 127.02, 128.46, 128.82, 129.80, 130.19, 130.53, 130.64, 130.70, 131.45, 131.87, 132.59, 132.89, 138.23, 159.36, 164.52, 168.99, 170.34, 171.50, 188.26 (C=O). HRMS (M+H): For C23H14Cl2N4O2S2 calcd: 513.0008, found: 513.0011

4-(benzo[d][1,3]dioxol-5-yl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4h)

Yield: 23.64% m.p.: 240.8°C. FT-IR υmax (cm−1): 3594.7 (NH), 2977.8–2915.4 (C–H), 2209.8 (C≡N), 1648.64 (C=O), 1503.41–1415.91 (C=C, C=N), 1254.60, 1242.78 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.67 (3H, s, CH3), 4.76 (2H, s, CH2), 5.85 (2H, s, CH2), 6.85 (1H, d, J: 8.23 Hz, Ar), 7.16, (1H, s, Ar), 7.36 (1H, dd, J: 8.19 Hz, j: 1.61 Hz, Ar), 7.52–7.59 (3H, m, Ar), 7.99 (2H, dd, J: 7.52 Hz, j: 1.58 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.74 (CH3), 39.18 (CH2), 102.22 (O-CH2-O), 92.01 (C-CN), 108.26, 108.83, 116.55, 124.29, 127.08, 129.20, 129.86, 130.48, 132.07, 132.52, 147.77, 150.55, 159.83, 161.94, 165.38, 166.59, 169.55, 186.51 (C=O). HRMS (M+H): C24H16N4O2S2 calcd: 489.0686, found: 489.0685

2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-4-phenyl-1,6-dihydropyrimidine-5-carbonitrile (4i)

Yield: 82.93% m.p.: 288°C. FT-IR υmax (cm−1): 3404.4 (NH), 3004–2872.7 (C–H), 2209.8 (C≡N), 1672.11 (C=O), 1553.10–1411.21 (C=C, C=N), 1265.51 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.84 (3H, s, CH3), 4.45 (2H, s, CH2), 7.08 (2H, d, J: 8.88 Hz, Ar), 7.32–7.35 (3H, m, Ar), 7.66 (2H, dd, J: 7.8 Hz, j: 1.65 Hz, Ar), 7.93 (2H, d, J: 8.83 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.77 (CH3), 39.15 (CH2), 55.95 (OCH3), 89.73 (C-CN), 115.23, 120.40, 125.35, 128.33, 128.45, 128.84, 129.77, 130.11, 137.91, 159.46, 162.34, 167.33, 169.02, 170.58, 171.07, 188.11 (C=O). HRMS (M+H): For C24H18N4O3S2 calcd: 475.0893, found: 475.0884

4-(3-methoxyphenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4j)

Yield: 36.75% m.p.: 212.5°C. FT-IR υmax (cm−1): 3322.3 (NH), 2977.8–2902.3 (C–H), 2213.1 (C≡N), 1670.63 (C=O), 1562.11–1442.88 (C=C, C=N), 1257.20, 1236.30 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.68 (3H, s, OCH3), 3.85 (3H, s, CH3), 4.46 (2H, s, CH2), 6.93–6.97 (1H, m, Ar), 7.08 (2H, dd, J: 6.84 Hz, j: 2.07 Hz, Ar), 7.22–7.26 (3H, m, Ar), 7.94 (2H, dd, J: 6.81 Hz, j: 2.08 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.16 (CH2), 55.45 (OCH3), 55.96 (OCH3), 89.74 (C-CN), 113.97, 115.22, 115.71, 120.41, 120.77, 125.39, 128.84, 129.42, 129.68, 139.31, 159.26, 159.43, 162.33, 167.04, 168.99, 170.57, 171.04, 188.08 (C=O). HRMS (M+H): For C25H20N4O4S2 calcd: 505.0999, found: 505.0985

4-(4-methoxyphenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4k)

Yield: 58.55% m.p.: 242.9°C. FT-IR υmax (cm−1): 3680 (NH), 2971.2–2840 (C–H), 2203.3 (C≡N), 1642.1 (C=O), 1606–1416.86 (C=C, C=N), 1288.33, 1242.77 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.69 (3H, s, OCH3), 3.84 (3H, s, CH3), 4.43 (2H, s, CH2), 6.85 (2H, d, J: 8.89 Hz, Ar), 7.07 (2H, d, J: 8.88 Hz, Ar), 7.66 (2H, d, J: 8.83 Hz, Ar), 7.93 (2H, d, J: 8.82 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.79 (CH3), 39.16 (CH2), 55.53 (OCH3), 56.00 (OCH3), 88.86 (C-CN), 113.65, 115.23, 120.82, 125.36, 128.82, 129.83, 130.03, 130.19, 159.45, 160.84, 162.35, 166.56, 169.00, 170.92, 188.28 (C=O). HRMS (M+H): For C25H20N4O4S2 calcd: 505.0999, found: 505.0986

4-(3,4-dimethoxyphenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4l)

Yield: 48.37% m.p. : 243.4°C. FT-IR υmax (cm−1): 3673.5 (NH), 2977.8–2836.6 (C–H), 2206.6 (C≡N), 1642.1 (C=O), 1606–1417.63 (C=C, C=N), 1255.24, 1228.20 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.71 (6H, s, OCH3), 3.84 (3H, s, CH3), 4.47 (2H, s, CH2), 6.90 (1H, d, J: 8.45 Hz, Ar), 7.06 (2H, d, J: 8.90 Hz, Ar), 7.35 (1H, d, J: 8.33 Hz, Ar), 7.37–7.38 (1H, m, Ar), 7.92 (2H, d, J: 8.87 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.77 (CH3), 39.15 (CH2), 55.77 (OCH3), 55.86 (OCH3), 55.94 (OCH3), 89.03 (C-CN), 111.26, 112.17, 115.21, 120.89, 121.64, 125.33, 128.81, 129.60, 130.26, 148.42, 150.63, 159.52, 162.35, 166.61, 169.04, 170.81, 170.90, 188.12 (C=O). HRMS (M+H): For C26H22N4O5S2 calcd: 535.1104, found: 535.1084

4-(3-chlorophenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4m)

Yield: 83.67% m.p.: 245°C. FT-IR υmax (cm−1): 3676.8, 3532.4 (NH), 2990.9–2908.8 (C–H), 2213.1 (C≡N), 1670.98 (C=O), 1606–1406.68 (C=C, C=N), 1259.41 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.84 (3H, s, CH3), 4.46 (2H, s, CH2), 7.07 (2H, brs, Ar), 7.42 (2H, brs, Ar), 7.65 (2H, brs, Ar), 7.92 (2H, brs, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.78 (CH3), 39.13 (CH2), 55.97 (OCH3), 89.90 (C-CN), 115.20, 119.95, 125.39, 127.06, 128.16, 128.85, 129.69, 130.05, 130.30, 133.36, 139.76, 159.44, 162.32, 165.56, 169.08, 170.29, 171.31, 187.88 (C=O). HRMS (M+H): For C24H17ClN4O3S2 calcd: 509.0503, found: 509.0493

4-(4-chlorophenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4n)

Yield: 51.98% m.p.: 278°C. FT-IR υmax (cm−1): 3371.6 (NH), 2971.2–2902.3 (C–H), 2213.1 (C≡N), 1652 (C=O), 1606–1411.64 (C=C, C=N), 1257.08 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.67 (3H, s, CH3), 3.83 (3H, s, CH3), 4.42 (2H, s, CH2), 7.07 (2H, d, J: 8.88 Hz, Ar), 7.38 (2H, d, J: 8.54 Hz, Ar), 7.67 (2H, d, J: 8.55 Hz, Ar), 7.91 (2H, d, J: 8.83 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.76 (CH3), 39.13 (CH2), 55.98 (OCH3), 89.69 (C-CN), 115.21, 120.23, 125.32, 128.44, 128.82, 129.79, 130.22, 134.88, 136.65, 159.46, 162.33, 166.09, 169.03, 170.52, 171.33, 188.19 (C=O). HRMS (M+H): For C24H17ClN4O3S2 calcd: 509.0503, found: 509.0488

4-(3,4-dichlorophenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4o)

Yield: 36.22% m.p.: 243°C. FT-IR υmax (cm−1): 3565.2, 3378.1 (NH), 2977.8–2836.6 (C–H), 2209.8 (C≡N), 1665 (C=O), 1556.59–1410.08 (C=C, C=N), 1262.69 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.84 (3H, s, CH3), 4.44 (2H, s, CH2), 7.06 (2H, d, J: 8.91 Hz, Ar), 7.60 (1H, d, J: 8.38 Hz, Ar), 7.68 (1H, dd, J: 8.4 Hz, j: 2.03 Hz, Ar), 7.81 (1H, d, J: 1.98 Hz, Ar), 7.91 (2H, dd, J: 6.81 Hz, j: 2.01 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.78 (CH3), 39.14 (CH2), 55.97 (OCH3), 89.82 (C-CN), 115.19, 119.97, 125.36, 128.49, 128.82, 129.73, 130.21, 130.66, 131.45, 132.87, 138.28, 159.40, 162.31, 164.48, 169.06, 170.26, 171.50, 188.00 (C=O). HRMS (M+H): For C24H16Cl2N4O3S2 calcd: 543.0114, found: 543.0101

4-(benzo[d][1,3]dioxol-5-yl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4p)

Yield: 44.09% m.p.: 216°C. FT-IR υmax (cm−1): 2899 (C–H), 2213.1 (C≡N), 1650 (C=O), 1643.63 (C=O), 1538.68–1429.88 (C=C, C=N), 1251.39 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.66 (3H, s, CH3), 3.85 (3H, s, OCH3), 4.59 (2H, s, CH2), 5.92 (2H, s, CH2), 6.86 (1H, d, J: 8.21 Hz, Ar), 7.08, (2H, d, J: 8.86 Hz, Ar), 7.17 (1H, d, J: 1.65 Hz, Ar), 7.32 (1H, dd, J: 8.18 Hz, j: 1.73 Hz, Ar), 7.93 (2H, d, J: 8.78 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.73 (CH3), 39.09 (CH2), 55.54 (OCH3), 89.82 (C-CN), 120.30, 128.33, 128.43, 128.78, 129.97, 130.12, 131.25, 131.41, 136.52, 137.83, 159.35, 167.38, 167.48, 170.22, 170.88, 188.35 (C=O). HRMS (M+H): For C25H18N4O5S2 calcd: 519.0791, found: 519.0779

2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-4-phenyl-1,6-dihydropyrimidine-5-carbonitrile (4q)

Yield: 53.49% m.p.: 160°C. FT-IR υmax (cm−1): 3365 (NH), 2964.6–2912.1 (C–H), 2206.6 (C≡N), 1670.81 (C=O), 1654.91 (C=O) 1535–1432.94 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 4.46 (2H, s, CH2), 7.28–7.38 (3H, m, Ar), 7.59–7.64 (4H, m, Ar), 8.01 (2H, d, J: 8.60 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.73 (CH3), 39.09 (CH2), 90.01 (C-CN), 120.30, 128.33, 128.43, 128.78, 129.97, 130.12, 131.25, 131.41, 136.52, 137.83, 159.35, 167.38, 167.48, 170.22, 170.88, 188.35 (C=O). HRMS (M+H): For C23H15ClN4O2S2 calcd: 479.0398, found: 479.0387

2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-4-(3-methoxyphenyl)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4r)

Yield: 21.41% m.p.: 195°C (decomposed). FT-IR υmax (cm−1): 3381.4 (NH), 2994.2–2908.8 (C–H), 2206.6 (C≡N), 1675.57 (C=O), 1551.61–1433.78 (C=C, C=N), 1231.89 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 3.67 (3H, s, OCH3), 4.46 (2H, s, CH2), 6.91–6.95 (1H, m, Ar), 7.18–7.25, (3H, m, Ar), 7.61 (2H, d, J: 8.53 Hz, Ar), 8.01 (2H, d, J: 8.52 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.67 (CH3), 39.72 (CH2), 55.47 (OCH3), 105.06 (C-CN), 113.97, 115.65, 120.30, 120.74, 128.77, 129.40, 129.93, 131.49, 136.48, 159.23, 159.33, 167.09, 188.35 (C=O). HRMS (M+H): For C24H17ClN4O3S2 calcd: 509.0503, found: 509.0494

2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-4-(4-methoxyphenyl)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4s)

Yield: 32.05% m.p.: 215°C. FT-IR υmax (cm−1): 3338.1 (NH), 2963.7–2838.9 (C–H), 2201.6 (C≡N), 1669.02 (C=O), 1537.92–1470 (C=C, C=N), 1253.44 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.66 (3H, s, OCH3), 4.49 (2H, s, CH2), 6.83 (2H, d, J: 8.83 Hz, Ar), 7.59–7.64 (4H, m, Ar), 8.00 (2H, d, J: 8.57 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.74 (CH3), 39.35 (CH2), 55.50 (OCH3), 89.56 (C-CN), 113.66, 128.76, 129.67, 129.96, 130.13, 131.32, 131.38, 136.54, 159.39, 161.02, 166.68, 167.58, 188.22 (C=O). HRMS (M+H): For C24H17ClN4O3S2 calcd: 509.0503, found: 509.0486

2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-4-(3,4-dimethoxyphenyl)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4t)

Yield: 48.59% m.p.: 243°C. FT-IR υmax (cm−1): 3568 (NH), 3091.8–2842.2 (C–H), 2205 (C≡N), 1653.76 (C=O), 1556.74–1433.31 (C=C, C=N), 1298.61, 1257.95 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.69 (6H, s, OCH3), 4.49 (2H, s, CH2), 6.89 (1H, d, J: 8.42 Hz, Ar), 7.29–7.34 (2H, m, Ar), 7.59 (2H, d, J: 8.55 Hz, Ar), 7.99 (2H, d, J: 8.55 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.70 (CH3), 39.72 (CH2), 55.80 (OCH3), 89.20 (C-CN), 111.24, 112.18, 120.67, 121.62, 128.75, 129.93, 130.18, 131.11, 131.42, 136.52, 148.41, 150.64, 159.37, 166.69, 167.52, 170.45, 188.35 (C=O). HRMS (M+H): For C25H19ClN4O4S2 calcd: 539.0609, found: 539.0612

4-(3-chlorophenyl)-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4u)

Yield: 50.71% m.p.: 245°C. FT-IR υmax (cm−1): 3397.3 (NH), 2980.1–2904.6 (C–H), 2214.8 (C≡N), 1671.69 (C=O), 1580.10–1434.03 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.50 (2H, s, CH2), 7.37–7.42 (2H, m, Ar), 7.58–7.66 (4H, m, Ar), 8.00 (2H, d, J: 8.60 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.74 (CH3), 39.08 (CH2), 85.43 (C-CN), 127.10, 128.16, 128.79, 129.94, 130.22, 130.34, 131.13, 131.44, 133.35, 136.52, 159.38, 165.65, 170.40, 188.42 (C=O). HRMS (M+H): For C23H14Cl2N4O2S2 calcd: 513.0008, found: 513.0010

4-(4-chlorophenyl)-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4v)

Yield: 70.51% m.p.: 287°C. FT-IR υmax (cm−1): 3370 (NH), 2990.9–2908.8 (C–H), 2209.8 (C≡N), 1668.95 (C=O), 1537.86–1396.57 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.47 (2H, s, CH2), 7.36 (2H, d, J: 8.59 Hz, Ar), 7.57–7.64 (4H, m, Ar), 7.98 (2H, d, J: 8.58 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.74 (CH3), 39.08 (CH2), 85.43 (C-CN), 127.10, 128.16, 128.79, 129.94, 130.22, 130.34, 131.12, 131.44, 133.35, 136.5158, 159.38, 165.65, 170.38, 188.36 (C=O). HRMS (M+H): For C23H14Cl2N4O2S2 calcd 513.0008, found: 513.0008

2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-4-(3,4-dichlorophenyl)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4w)

Yield: 68.84% m.p.: 258°C. FT-IR υmax (cm−1): 3370 (NH), 2990.9–2908.8 (C–H), 2213.1 (C≡N), 1665 (C=O), 1567.59–1432.79 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 4.44 (2H, s, CH2), 7.57–7.66 (4H, m, Ar), 7.77 (1H, d, J: 1.90 Hz, Ar), 7.98 (2H, dd, J: 6.6 Hz, j: 1.98 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.70 (CH3), 39.72 (CH2), 89.90 (C-CN), 119.90, 128.47, 128.74, 129.91, 130.17, 130.64, 131.26, 131.40, 131.44, 132.80, 136.48, 138.29, 159.24, 164.56, 167.48, 170.06, 171.37, 188.34 (C=O). HRMS (M+H): For C23H13Cl3N4O2S2 calcd: 548.0220, found: 548.0220

4-(benzo[d][1,3]dioxol-5-yl)-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-oxo-1,6-dihydropyrimidine-5-carbonitrile (4x)

Yield: 65.26% m.p.: 291.1°C. FT-IR υmax (cm−1): 3667 (NH), 2990.9–2905.9 (C–H), 2223 (C≡N), 1675 (C=O), 1642 (C=O), 1550–1373.04 (C=C, C=N), 1248.26 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.65 (3H, s, CH3), 4.74 (2H, s, CH2), 5.87 (2H, s, CH2), 6.85 (1H, d, J: 8.23 Hz, Ar), 7.13 (1H, d, J: 1.67 Hz, Ar), 7.35 (1H, dd, J: 8.26 Hz, j: 1.64 Hz, Ar), 7.60 (2H, d, J: 8.60 Hz, Ar), 7.98 (2H, d, J: 8.57 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.68 (CH3), 39.73 (CH2), 92.40 (C-CN), 102.26 (O-CH2-O), 108.27, 108.80, 116.48, 124.29, 128.77, 129.15, 129.93, 130.92, 131.33, 136.68, 147.76, 150.55, 159.81, 161.86, 165.30, 166.56, 168.11, 186.49 (C=O). HRMS (M+H): For C24H15ClN4O4S2 calcd: 523.0296, found: 523.0288

4.1.5. General synthesis of 4-amino-2-mercapto-6-substitutedphenylpyrimidine-5-carbonitrile derivatives (Method E) (3a–c)

Appropriate aldehyde, thiourea and malononitrile were dissolved in anhydrous ethanol and potassium carbonate was added. The reaction was controlled with TLC after boiling for 5 hours (ethyl acetate 1:2 petroleum ether). The precipitated product was washed with ethanol. The raw product was mixed with hot water and filtered. After the filtrate cools, it was acidified with acetic acid. It was taken and washed with water. Crystallized from ethanol.

4-amino-2-mercapto-6-phenylpyrimidine-5-carbonitrile (3a)

It was synthesized by Method E, using benzaldehyde (98.00 mmol, 10.4 g), thiourea (98.00 mmol, 7.46 g), potassium carbonate (98.00 mmol, 13.54 g) and malondinitrile (98.00 mmol, 6.47 g). Yield: 55%, m.p.: 255°C, No reference value

4-Amino-6-(4-methoxyphenyl)-2-mercaptopyrimidine-5-carbonitrile (3b)

It was synthesized by Method E, using 4-methoxybenzaldehyde (132.21 mmol, 18 g), thiourea (132.21 mmol, 10.06 g), potassium carbonate (132.21 mmol, 36.65 g) and malondinitrile (132.21 mmol, 8.73 g). Yield: 35%, m.p.: 261–262°C, m.p. reference: 258–260°C (N,N-DMF) [54].

4-Amino-6-(4-chlorophenyl)-2-mercaptopyrimidine-5-carbonitrile (3c)

It was synthesized by Method E, using 4-chlorobenzaldehyde (177.85 mmol, 25 g), thiourea (177.85 mmol, 13.54 g), potassium carbonate (177.85 mmol, 24.58 g) and malondinitrile (177.85 mmol, 11.75 mmol). Yield: 45%, m.p.: 268–269°C, m.p. reference: 268–270°C (1,4-dioxane) [54].

4.1.6. General synthesis of compounds 5a–i (Method F)

3a–c derivatives (50 mmol) and thiazolyl acetylbromide compounds 1a–c (55 mmol) are dissolved in acetone and then potassium carbonate (50 mmol) was added. The reaction was mixed for 12 hours at room temperature. If it was not completed in this time, the reaction was boiled for 1 hour. After the reaction was completed, it was poured into cold water. The product was expected to be precipitated thoroughly. In the meantime, thorough solubility of potassium carbonate should be ensured in water. The precipitated product was filtered away. Compounds are crystallized from ethanol twice. The resulted products are kept in vacuum survey seizing phosphorus pentaoxide (P2O5) at 45°C for overnight and ensured complete drying before analysis [15, 53]

4-amino-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)-6-phenylpyrimidine-5-carbonitrile (5a)

Yield: 41.04% m.p.: 196°C. FT-IR υmax (cm−1): 3676.8 (NH), 3299 (NH), 3145–2901.05 (C–H), 2216.4 (C≡N), 1638.8 (C=O), 1525.27–1393.71 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.64 (2H, s, CH2), 7.32–7.37 (2H, m, Ar), 7.42 (1H, d, J: 7.25 Hz, Ar), 7.54–7.58 (3H, m, Hz, Ar), 7.68 (2H, d, J: 7.07 Hz, Ar), 7.98 (2H, dd, J: 7.22 Hz, j: 1,78 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.79 (CH3), 39.17 (CH2), 83.37 (C-CN), 116.52, 127.09, 128.65, 128.79, 129.90, 130.81, 131.43, 132.03, 132.54, 136.11, 159.59, 163.73, 167.71, 169.2, 172.61, 187.31 (C=O). HRMS (M+H): For C23H17N5OS2 calcd: 444.0947, found: 444.0940

4-amino-6-(4-methoxyphenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)pyrimidine-5-carbonitrile (5b)

Yield: 23.52% m.p.: 218°C. FT-IR υmax (cm−1): 3689.9–3663.6 (NH2), 3378, 3309.2, (NH), 3168–2901.11 (C–H), 2213.1 (C≡N), 1671.6 (C=O), 1648.7 (C=O), 1541.13–1394.06 (C=C, C=N), 1251.39 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.67 (3H, s, CH3), 3.85 (3H, s, CH3), 4.62 (2H, s, CH2), 7.08 (2H, d, J: 8.01 Hz, Ar), 7.40 (3H, m, Ar), 7.69 (2H, d, J: 6.71 Hz, Ar), 7.94 (2H, d, J: 8.08 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.83 (CH3), 39.45 (CH2), 56.01 (OCH3), 85.08 (C-CN), 115.27, 116.54, 125.29, 128.66, 128.81, 128.88, 131.46, 136.12, 159.64, 162.43, 163.73, 167.68, 172.65, 187.07 (C=O). HRMS (M+H): For C24H19N5O2S2 calcd: 474.1053, found: 474.1047

4-amino-6-(4-chlorophenyl)-2-((2-(4-methyl-2-phenylthiazol-5-yl)-2-oxoethyl)thio)pyrimidine-5-carbonitrile (5c)

Yield: 28.20% m.p.: 231°C. FT-IR υmax (cm−1): 3397.3, 3325 (NH2), 3223.2 (NH), 2990–2894.7 (C–H), 2214.8 (C≡N), 1669.52 (C=O), 1525.40–1400 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.62 (2H, s, CH2), 7.34–7.43 (3H, m, Ar), 7.61 (2H, d, J: 8.62 Hz, Ar), 7.66 (2H, d, J: 7.06 Hz, Ar), 7.98 (2H, d, J: 8.61 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.17 (CH2), 83.39 (C-CN), 116.50, 128.65, 128.77, 129.97, 131.25, 131.36, 131.41, 136.10, 136.63, 159.57, 163.71, 167.73, 167.79, 172.57, 187.35 (C=O). HRMS (M+H): For C23H16ClN5OS2 calcd: 478.0534, found: 478.0534

4-amino-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-phenylpyrimidine-5-carbonitrile (5d)

Yield: 46.38% m.p.: 206°C. FT-IR υmax (cm−1): 3670.2 (NH2), 3407, 3225.6 (NH), 2987.53–2900 (C–H), 2209.8 (C≡N), 1675 (C=O), 1579.8–1315.72 (C=C, C=N), 1249.42 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3,64 (3H, s, OCH3), 4.62 (2H, s, CH2), 6.85 (2H, d, J: 8.89 Hz, Ar), 7.54–7.56 (3H, m, Ar), 7.68 (2H, d, J: 8.84 Hz, Ar), 7.99 (2H, dd, J: 7.35 Hz, j: 1.85 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.82 (CH3), 39.17 (CH2), 55.64 (OCH3), 82.38 (C-CN), 114.02, 116.87, 127.07, 128.20, 129.91, 130.62, 130.91, 132.04, 132.54, 159.58, 161.92, 163.88, 166.88, 169.26, 172.34, 187.48 (C=O). HRMS (M+H): For C24H19N5O2S2 calcd: 474.1053, found: 474.1050

4-amino-6-(4-methoxyphenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)pyrimidine-5-carbonitrile (5e)

Yield: 64.28% m.p.: 199.3°C. FT-IR υmax (cm−1): 3667 (NH2), 3476.6, 3235.5 (NH), 2987.51–2901.05 (C–H), 2206.6 (C≡N), 1665 (C=O), 1530.15–1393.63 (C=C, C=N), 1250.25 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.68 (3H, s, CH3), 3.67 (3H, s, OCH3), 3,85 (3H, s, OCH3), 4.60 (2H, s, CH2), 6.87 (2H, d, J: 8.96 Hz, Ar), 7.08 (2H, d, J: 8.93 Hz, Ar), 7.69 (2H, d, J: 8.90 Hz, Ar), 7.95 (2H, d, J: 8.86 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.86 (CH3), 39.17 (CH2), 55.67 (OCH3), 56.03 (OCH3), 82.34 (C-CN), 114.03, 115.27, 116.89, 125.29, 128.22, 128.87, 129.93, 130.64, 159.65, 161.94, 162.44, 163.88, 166.85, 169.33, 172.39, 187.23 (C=O). HRMS (M+H): For C25H21N5O3S2 calcd: 504.1159, found: 504.1150

4-amino-6-(4-chlorophenyl)-2-((2-(2-(4-methoxyphenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)pyrimidine-5-carbonitrile (5f)

Yield: 48.48% m.p.: 216.5°C. FT-IR υmax (cm−1): 3670.2 (NH2), 3328.9, 3181.2 (NH), 2987.54–2901.04 (C–H), 2209.8 (C≡N), 1658.5 (C=O), 1513.68–1371.62 (C=C, C=N), 1249.71 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.67 (3H, s, OCH3), 4.62 (2H, s, CH2), 6.85 (2H, d, J: 8.96 Hz, Ar), 7.60–7.68 (4H, m, Ar), 8.01 (2H, d, J: 8.60 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.78 (CH3), 39.17 (CH2), 55.66 (OCH3), 82.42 (C-CN), 114.02, 116.85, 128.21, 128.78, 129.98, 130.60, 131.37, 136.64, 159.53, 161.91, 163.86, 166.92, 167.81, 187.53 (C=O). HRMS (M+H): For C24H18ClN5O2S2 calcd: 508.0663, found: 508.0638

4-amino-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-phenylpyrimidine-5-carbonitrile (5g)

Yield: 88.79% m.p.: 228°C. FT-IR υmax (cm−1): 3420.3, 3334.9 (NH2), 3233 (NH), 3000–2898 (C–H), 2211.5 (C≡N), 1637.40 (C=O), 1521.65 –1493.19 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.62 (2H, s, CH2), 7.40 (2H, d, J: 8.60 Hz, Ar), 7.54–7.57 (3H, m, Ar), 7.67 (2H, d, J: 8.58 Hz, Ar), 7.98 (2H, dd, J: 7.64 Hz, j: 1.93 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.82 (CH3), 39.09 (CH2), 83.46 (C-CN), 116.35, 127.06, 128.76, 129.92, 130.60, 130.93, 132.06, 132.47, 134.85, 136.28, 159.53, 163.56, 166.62, 169.25, 172.70, 187.49 (C=O). HRMS (M+H): For C23H16ClN5OS2 calcd: 478.0558, found: 478.0573

4-amino-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)-6-(4-methoxyphenyl)pyrimidine-5-carbonitrile (5h)

Yield: 31.72% m.p.: 247°C. FT-IR υmax (cm−1): 3670.2(NH2), 3325.6, 3164.8 (NH), 2987.52–2900.47 (C–H), 2216.4 (C≡N), 1652 (C=O), 1518.50–1373.97 (C=C, C=N), 1248.10 (C–O). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.70 (3H, s, CH3), 3.65 (3H, s, OCH3), 4.61 (2H, s, CH2), 6.84 (2H, d, J: 8.91 Hz, Ar), 7.61 (2H, d, J: 8.61 Hz, Ar), 7.65 (2H, d, J: 8.88 Hz, Ar), 8.01 (2H, d, J: 8.61 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.81 (CH3), 39.09 (CH2), 55.61 (OCH3), 82.38 (C-CN), 113.99, 116.89, 128.18, 128.77, 129.99 130.60, 131.33, 131.42, 136.64, 159.51, 161.86, 163.83, 166.92, 167.81, 172.31, 187.57 (C=O). HRMS (M+H): For C24H18ClN5O2S2 calcd: 508.0663, found: 508.0664

4-amino-6-(4-chlorophenyl)-2-((2-(2-(4-chlorophenyl)-4-methylthiazol-5-yl)-2-oxoethyl)thio)pyrimidine-5-carbonitrile (5i)

Yield: 8.60% m.p.: 230°C. FT-IR υmax (cm−1): 3673.5 (NH2), 3430.6, 3335.5 (NH), 2987.50 – 2899 (C–H), 2219.7 (C≡N), 1661.8 (C=O), 1518.24–1368.60 (C=C, C=N). 1H NMR (300 MHz) DMSO-d6) δ (ppm): 2.69 (3H, s, CH3), 4.61 (2H, s, CH2), 7.40 (2H, d, J: 8.60 Hz, Ar), 7.61 (2H, d, J: 8.62 Hz, Ar), 7.65 (2H, d, J: 8.59 Hz, Ar), 7.99 (2H, d, J: 8.62 Hz, Ar). 13C NMR (75 MHz) DMSO-d6) δ (ppm): 18.75 (CH3), 39.17 (CH2), 87.84 (C-CN), 116.28, 128.76, 129.97, 130.56, 131.34, 134.86, 136.25, 136.62, 159.49, 163.58, 166.66, 167.79, 172.69, 187.52 (C=O). HRMS (M+H): For C23H15Cl2N5OS2 calcd: 512.0168, found: 512.0151.

4.2. Biological activity

4.2.1. Cellular U87MG antitumor MTT activity measurement

Subcultures of the cells

HEK293 cells were maintained in Dulbecco’s Modified Eagle medium (Gibco, UK) supplemented with 10% (v/v) fetal bovine serum (Gibco, UK), 10.000 U/mL penicillin, 10.000 μg/mL of streptomycin. The human glioblastoma cell lines U87MG were cultured in Dulbecco’s Modified Eagle’s medium/high glucose (Gibco, UK) supplemented with 10% heat inactivated fetal bovine serum (Gibco, UK), 1% (v/v) antibiotic-antimycotics solution (10.000 U/mL penicillin,10.000 μg/mL streptomycin and 0.25 μg/mL amphotericin B; Gibco, UK), and 1% (v/v) non-essential amino acids (Gibco, UK) at 37°C in a humidified 5% CO2 atmosphere. MCF7 cells were maintained in Dulbecco’s Modified Eagle medium- Nutrient Mixture F-12 (Gibco, UK) supplemented with 10% (v/v) fetal bovine serum (Gibco, UK), 10.000 U/mL penicillin, 10.000 μg/mL of streptomycin and %10 L-glutamine. The cells were passaged every 3 days [55].

Cytotoxic and anticancer activity

Cytotoxic activities of the tested compounds were determined by cell proliferation analysis using the standard 3- (4,5-dimethylthiazole-2 yl) −2,5-diphenyltetrazolium bromide (MTT) assay [5557]. In order to determine the toxic and proliferative effects of compounds, cells were seeded in 96-well plates add a density of 5.104 (HEK293 human embryonic kidney cell line (ATCC, USA), 4.104 (MCF7), 105 (U87MG) and per well 24 h prior to treatment. The compounds to be tested were first dissolved in DMSO (less than 1%) and diluted with DMEM medium for use in MTT assay. After 24 and 48 hours incubation times, the medium on the cells was removed, washed with D-PBS and 30 μL of MTT was added from stock solution (5 mg/mL, prepared in D-PBS). At the end of the 4 hour incubation period, 150 μL DMSO was added and the culture vessel was shaken at room temperature for homogeneity of the solution. The absorbance of the solution at a wavelength of 540 nm was measured in a microplate reader (EON, BioTek Instruments Inc.). Three independent experiments were performed at least [58, 59].

% Viability=(Absorbance experiment group)/(Absorbance control)×100%

4.2.2. Measuring adenosine receptor binding inhibition

Cell culture and membrane preparation

The degree of binding of the synthesized compounds to adenosine A1 and A2A receptor subtypes was tested. For this purpose, competitive attachment experiments NIH (American National Institute of Health) NIDDK unit. CHO cells (Chinese Hamster Ovary cells) producing recombinant hA1 (human adenosine A1) receptor and HEK293 (human embryonic kidney cell line) cells producing hA2AAR (human adenosine A2A) receptor, 10% fetal bovine serum in DMEM: F12 (1: 1) mixture, 100 U/mL penicillin, 100 μg/mL streptomycin, and 2 μmol/mL glutamine. Cells are seeded by trypsination. After homogenization and suspension, cells are centrifuged at 500 g for 10 min and the precipitate formed was suspended in 50 mM tris HCl. The suspension was homogenized for 10 seconds with an electrical homogenizer and immediately centrifuged at 20 kg for 20 minutes at 4°C. The resulting pellets are resuspended in 3 U/mL adenosine deaminase buffer and stored at −80°C until the time of use.

Measuring A1 and A2A adenosine receptor binding inhibition

50 μL each of 1 μM concentrations of test compounds and standard [3H]DPCPX radioligand (2 nM, PerkinElmer, Boston, MA) for hA1AR binding measurement with A1AR-expressing CHO cell membranes (40 μg/tube) 50 mM Tris- Incubate for 60 min at 25°C in a total volume of 200 μL in HCl buffer (pH: 7.4; MgCl2, 10mM) [60, 61].

For measurement of A2AAR and A2BAR binding, 50 μL each of 1 μM concentrations of test compounds and [3H]ZM241385 radioligand (15 nM, American Radiolabeled Chemicals, Inc., St. Louis, MO) with A2AR expressing HEK293 cell membranes (20 μg/tube). The mixture was incubated for 60 min at 25°C in a total volume of 200 μL in 50 mM Tris-HCl buffer (pH: 7.4; MgCl2, 10mM). The reaction was terminated by filtration through GF/B filters. Filters used for A1 and A2A measurement are placed in scintillation vials containing 5 mL of Hydrofluor scintillation buffer and counted with the help of a PerkinElmer Tricarb 2810TR Liquid Scintillation counter [62].

4.2.3. Phosphodiesterase enzyme (PDE) inhibition assay

Experiments were carried out following the instructions provided by the “PDE Activity Assay Kit” (ab139460, Abcam, Cambdrige, UK) using the modified method detailed by Araiz et al [63]. Stock solutions of the compounds were prepared with DMSO (Less than 1%). Recombinant human PDE4 was incubated with 0.5 μM 5′-cGMP and 10 μL 5′-nucleotidase under 30°C for 90 minutes. The kit based on the sequential hydrolysis of cyclic nucleotides by PDE4 and 5′-nucleotidase. The released phosphate by enzymatic cleavage was directly proportional to PDE4 activity. The reaction was terminated by incubation with termination buffer under room temperature for 20 minutes. OD 620nm were measured on a plate reader to determine the amount of GMP generated. For measurement of IC50, 6 concentrations of inhibitors were used under the substrate concentration the suitable enzyme concentration. The PDE4B and PDE4D inhibitor rolipram and PDE inhibitor IBMX were used as a positive control.

4.2.4. Statistical analysis.

Data are presented as mean±standard deviation. All experiments were performed at least in triplicates. Statistical comparisons were performed through the Student’s t-test or one-way ANOVA followed by Tukey test, for 2 or more experimental groups, respectively. Statistical significance was set at p < 0.05. Statistical analysis and artwork were performed using Graph Pad Prism 7.0d (Graph Pad Software, La Jolla, USA). Specific tests are detailed in the figure legends.

Supplementary Material

1
2
3

Figure 5.

Figure 5.

Graphical representation of IC50 values on all tested cell lines. An HEK293 bar is not given for 5d and 5e because the viability was more than 50%, even at 100 μM, and therefore IC50 values could not be calculated.

  • Novel 24 thiouracil and 9 thiocytidine derivative are synthesized

  • Especially thiocytidine derivatives showed potent activity on aggresive brain tumor cells

  • Compounds are not cytotoxic on HEK293 cell line

  • Antitumor potent molecules showed high inhibitory activity on PDE4

  • It is discussed that antitumor activity is associated with phosphodiesterase activity

Acknowledgements

This project is supported by NIDDK Intramural Research Program (ZIADK031117).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

There is not any conflict of interest by the authors.

PhD Pharm Zafer Sahin

References

  • [1].Jacobson KA, Jarvis MF, Williams M, Purine and pyrimidine (P2) receptors as drug targets, J. Med. Chem, 45 (2002) 4057–4093. 10.1021/jm020046y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Abbas SE, Awadallah FM, Ibrahin NA, Said EG, Kamel GM, New quinazolinone-pyrimidine hybrids: synthesis, anti-inflammatory, and ulcerogenicity studies, Eur. J. Med. Chem, 53 (2012) 141–149. 10.1016/j.ejmech.2012.03.050. [DOI] [PubMed] [Google Scholar]
  • [3].Sharma AK, Mahajan MP, Synthesis and [4+2] cycloaddition reactions of 4-(N-allyl-N-aryl)amino-1,3-diaza-1,3-butadienes with vinyl-, isopropenyl- and chloroketenes: Entry to novel pyrimidinone/fused pyrimidinone derivatives., Tetrahedron, 53 (1997) 13841–13854. 10.1016/S0040-4020(97)00873-9. [DOI] [Google Scholar]
  • [4].Patel A, Pasha Y, Modi A, Synthesis and biological evaluation of 4-aryl substituted −2-(5-carboxylicacid-1, 6-dihydro)-2-thiophenylethylene-6-oxopyrimidine as protein tyrosine phosphatase (PTP-1B) inhibitors, Int. J. Pharmtech. Res, 8 (2015) 136–143. ISSN: 0974–4304. [Google Scholar]
  • [5].Abbas SE, Aly EI, Awadallah FM, Mahmoud WR, 4-Substituted-1-phenyl-1H-pyrazolo[3,4-d]pyrimidine derivatives: design, synthesis, antitumor and EGFR tyrosine kinase inhibitory activity, Chem. Biol. Drug Des, 85 (2015) 608–622. 10.1111/cbdd.12451. [DOI] [PubMed] [Google Scholar]
  • [6].Awadallah FM, Piazza GA, Gary BD, Keeton AB, Canzoneri JC, Synthesis of some dihydropyrimidine-based compounds bearing pyrazoline moiety and evaluation of their antiproliferative activity, Eur. J. Med. Chem, 70 (2013) 273–279. 10.1016/j.ejmech.2013.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Fargualy AM, Habib NS, Ismail KA, Hassan AM, Sarg MT, Synthesis, biological evaluation and molecular docking studies of some pyrimidine derivatives, Eur. J. Med. Chem, 66 (2013) 276–295. 10.1016/j.ejmech.2013.05.028. [DOI] [PubMed] [Google Scholar]
  • [8].Ma LY, Zheng YC, Wang SQ, Wang B, Wang ZR, Pang LP, Zhang M, Wang JW, Ding L, Li J, Wang C, Hu B, Liu Y, Zhang XD, Wang JJ, Wang ZJ, Zhao W, Liu HM, Design, synthesis, and structure-activity relationship of novel LSD1 inhibitors based on pyrimidine-thiourea hybrids as potent, orally active antitumor agents, J. Med. Chem, 58 (2015) 1705–1716. 10.1021/acs.jmedchem.5b00037. [DOI] [PubMed] [Google Scholar]
  • [9].Ma LY, Pang LP, Wang B, Zhang M, Hu B, Xue DQ, Shao KP, Zhang BL, Liu Y, Zhang E, Liu HM, Design and synthesis of novel 1,2,3-triazole-pyrimidine hybrids as potential anticancer agents, Eur. J. Med. Chem, 86 (2014) 368–380. 10.1016/j.ejmech.2014.08.010. [DOI] [PubMed] [Google Scholar]
  • [10].Said MM, Taher AT, El-Nassan HB, El-Khouly EA, Synthesis of novel S-acyl and S-alkylpyrimidinone derivatives as potential cytotoxic agents., Res. Chem. Intermediat, 42 (2016) 6643–6662. 10.1007/s11164-016-2487-x. [DOI] [Google Scholar]
  • [11].Taher AT, Abou-Seri SM, Synthesis and bioactivity evaluation of new 6-aryl-5-cyano thiouracils as potential antimicrobial and anticancer agents, Molecules, 17 (2012) 9868–9886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Taher AT, Helwa AA, Synthesis, antitumor and antimicrobial testing of some new thiopyrimidine analogues, Chem. Pharm. Bull. (Tokyo), 60 (2012) 1305–1313. 10.3390/molecules17089868. [DOI] [PubMed] [Google Scholar]
  • [13].Abdel Gawad NM, Amin NH, Elsaadi MT, Mohamed FMM, Angeli A, De Luca V, Capasso C, Supuran CT, Synthesis of 4-(thiazol-2-ylamino)-benzenesulfonamides with carbonic anhydrase I, II and IX inhibitory activity and cytotoxic effects against breast cancer cell lines, Bioorg. Med. Chem, 24 (2016) 3043–3051. 10.1016/j.bmc.2016.05.016. [DOI] [PubMed] [Google Scholar]
  • [14].Mahran AM, Hassan NA, Osman DA, Ragab SS, Hassan AA, Synthesis and biological evaluation of novel pyrimidines derived from 6-aryl-5-cyano-2-thiouracil, Z. Naturforsch C. J. Biosci, 71 (2016) 133–140. 10.1515/znc-2015-0265. [DOI] [PubMed] [Google Scholar]
  • [15].Rami C, Patel L, Patel CN, Parmar JP, Synthesis, antifungal activity, and QSAR studies of 1,6-dihydropyrimidine derivatives, J. Pharm. Bioallied. Sci, 5 (2013) 277–289. 10.4103/0975-7406.120078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Stanovnik B, Tisler M, Katritzky AR, Denisko OV, Advances in Heterocyclic Chemistry, Elsevier, San Diego, 2006. [Google Scholar]
  • [17].Zieliński W, Kudelko A, Synthesis and basicity of 4-amino-2-phenylquinazolines., Monatsh. Chem, 131 (2000) 895–899. 10.1007/s007060070067. [DOI] [Google Scholar]
  • [18].Jacobson KA, Muller CE, Medicinal chemistry of adenosine, P2Y and P2X receptors, Neuropharmacology, 104 (2016) 31–49. 10.1016/j.neuropharm.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Jacobson KA, Tchilibon S, Joshi BV, Gao ZG, Chapter 13. A3 Adenosine Receptors, Annu. Rep. Med. Chem, 38 (2003) 121–130. 10.1016/S0065-7743(03)38014-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Louvel J, Guo D, Agliardi M, Mocking TA, Kars R, Pham TP, Xia L, de Vries H, Brussee J, Heitman LH, Ijzerman AP, Agonists for the adenosine A1 receptor with tunable residence time. A Case for nonribose 4-amino-6-aryl-5-cyano-2-thiopyrimidines, J. Med. Chem, 57 (2014) 3213–3222. 10.1021/jm401643m. [DOI] [PubMed] [Google Scholar]
  • [21].Lebon G, Warne T, Edwards PC, Bennett K, Langmead CJ, Leslie AG, Tate CG, Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature, 474(7352) (2011) 521–525. 10.1038/nature10136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Louvel J, Guo D, Soethoudt M, Mocking TA, Lenselink EB, Mulder-Krieger T, Heitman LH, AP IJ, Structure-kinetics relationships of Capadenoson derivatives as adenosine A1 receptor agonists, Eur. J. Med. Chem, 101 (2015) 681–691. 10.1016/j.ejmech.2015.07.023. [DOI] [PubMed] [Google Scholar]
  • [23].Lane JR, Klein Herenbrink C, van Westen GJ, Spoorendonk JA, Hoffmann C, AP IJ, A novel nonribose agonist, LUF5834, engages residues that are distinct from those of adenosine-like ligands to activate the adenosine A(2a) receptor, Mol. Pharmacol, 81 (2012) 475–487. 10.1124/mol.111.075937. [DOI] [PubMed] [Google Scholar]
  • [24].Zhu H, Li JT, Zheng F, Martin E, Kots AY, Krumenacker JS, Choi BK, McCutcheon IE, Weisbrodt N, Bogler O, Murad F, Bian K, Restoring soluble guanylyl cyclase expression and function blocks the aggressive course of glioma, Mol. Pharmacol, 80 (2011) 1076–1084. 10.1124/mol.111.073585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, Pekmezci M, Schwartzbaum JA, Turner MC, Walsh KM, Wrensch MR, Barnholtz-Sloan JS, The epidemiology of glioma in adults: a “state of the science” review, Neuro. Oncol, 16 (2014) 896–913. 10.1093/neuonc/nou087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Oh SJ, Yang JI, Kim O, Ahn EJ, Kang WD, Lee JH, Moon KS, Lee KH, Cho D, Human U87 glioblastoma cells with stemness features display enhanced sensitivity to natural killer cell cytotoxicity through altered expression of NKG2D ligand, Cancer Cell. Int, 17 (2017) 22 10.1186/s12935-017-0397-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].J. HS Lee Kyu Sang, Moon Seyoung, Choe Gheeyoung, Selection of a highly invasive cell population from a glioblastoma cell line and analysis of invasion-associated factors, Int. J. Clin. Exp. Pathol, 10 (2017) 6882–6892. ISSN:1936–2625/IJCEP0052436. [Google Scholar]
  • [28].Zeng D, Maa T, Wang U, Feoktistov I, Biaggioni I, Belardinelli L, Expression and function of A2B adenosine receptors in the U87MG tumor cells., Drug Dev. Res, 58 (2003) 405–411. 10.1002/ddr.10212. [DOI] [Google Scholar]
  • [29].Rocha R, Torres A, Ojeda K, Uribe D, Rocha D, Erices J, Niechi I, Ehrenfeld P, San Martin R, Quezada C, The Adenosine A(3) Receptor Regulates Differentiation of Glioblastoma Stem-Like Cells to Endothelial Cells under Hypoxia, Int. J. Mol. Sci, 19 (2018). 10.3390/ijms19041228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Caretta A, Mucignat-Caretta C, Protein kinase A in cancer, Cancers (Basel), 3 (2011) 913–926. 10.3390/cancers3010913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Cheng X, Phelps C, Taylor SS, Differential binding of cAMP-dependent protein kinase regulatory subunit isoforms Ialpha and IIbeta to the catalytic subunit, J. Biol. Chem, 276 (2001) 4102–4108. 10.1074/jbc.M006447200. [DOI] [PubMed] [Google Scholar]
  • [32].Ramezani S, Hadjighassem M, Vousooghi N, Parvaresh M, Arbabi F, Amini N, Joghataei MT, The Role of Protein Kinase B Signaling Pathway in Anti-Cancer Effect of Rolipram on Glioblastoma Multiforme: An In Vitro Study, Basic Clin. Neurosci, 8 (2017) 325–336. 10.18869/nirp.bcn.8.4.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Savai R, Pullamsetti SS, Banat GA, Weissmann N, Ghofrani HA, Grimminger F, Schermuly RT, Targeting cancer with phosphodiesterase inhibitors, Expert Opin. Investig. Drugs, 19 (2010) 117–131. 10.1517/13543780903485642. [DOI] [PubMed] [Google Scholar]
  • [34].Jacobson KA, Merighi S, Varani K, Borea PA, Baraldi S, Tabrizi MA, Romagnoli R, Baraldi PG, Ciancetta A, Tosh DK, Gao ZG, Gessi S. A3 adenosine receptors as modulators of inflammation: from medicinal chemistry to therapy. Med. Res. Rev, 38 (2018)1031–1072. 10.1002/med.21456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Carlin JL, Jain S, Gizewski E, Wan TC, Tosh DK, Xiao C, Auchampach JA, Jacobson KA, Gavrilova O, Reitman ML, Hypothermia in mouse is caused by adenosine A1 and A3 receptor agonists and AMP via three distinct mechanisms, Neuropharmacology, 114 (2017) 101–113. 10.1016/j.neuropharm.2016.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Romagnoli R, Baraldi PG, Lopez-Cara C, Cruz-Lopez O, Moorman AR, Massink A, AP IJ, Vincenzi F, Borea PA, Varani K, Synthesis and biological evaluation of a new series of 2-amino-3-aroyl thiophene derivatives as agonist allosteric modulators of the A1 adenosine receptor. A position-dependent effect study, Eur. J. Med. Chem, 101 (2015) 185–204. 10.1016/j.ejmech.2015.06.041. [DOI] [PubMed] [Google Scholar]
  • [37].Baer JC, Freeman AA, Newlands ES, Watson AJ, Rafferty JA, Margison GP, Depletion of O6-alkylguanine-DNA alkyltransferase correlates with potentiation of temozolomide and CCNU toxicity in human tumour cells, Br. J. Cancer, 67 (1993) 1299–1302. 10.1038/bjc.1993.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lee SY, Temozolomide resistance in glioblastoma multiforme, Genes Dis, 3 (2016) 198–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Lee CY, Strategies of temozolomide in future glioblastoma treatment, Onco. Targets Ther, 10 (2017) 265–270. 10.1016/j.gendis.2016.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Bondock S, Naser T, Ammar YA, Synthesis of some new 2-(3-pyridyl)-4,5-disubstituted thiazoles as potent antimicrobial agents, Eur. J. Med. Chem, 62 (2013) 270–279. 10.1016/j.ejmech.2012.12.050. [DOI] [PubMed] [Google Scholar]
  • [41].Kumar D, Kumar NM, Patel G, Gupta S, & Varma RS, A facile and eco-friendly synthesis of diarylthiazoles and diarylimidazoles in water, Tetrahedron Lett, 52 (2011) 1983–1986. 10.1016/j.tetlet.2011.02.069. [DOI] [Google Scholar]
  • [42].Mayhoub AS, Khaliq M, Kuhn RJ, Cushman M, Design, synthesis, and biological evaluation of thiazoles targeting flavivirus envelope proteins, J. Med. Chem, 54 (2011) 1704–1714. 10.1021/jm1013538. [DOI] [PubMed] [Google Scholar]
  • [43].Abhale YK, Deshmukh KK, Sasane AV, Chavan AP, & Mhaske PC, Fused Heterocycles: Synthesis and Antitubercular Activity of Novel 6-Substituted-2-(4-methyl-2-substituted phenylthiazol-5-yl)Himidazo[1,2-a]pyridine, Journal of Heterocyclic Chemistry, 53 (2015) 229–233. 10.1002/jhet.2409. [DOI] [Google Scholar]
  • [44].Okamiya J, The rates of the reaction of heterocyclic (thiophene and thiazole) bromoketones with thioamides., Nippon Kagaku Zasshi, 87 (1966) 594–600. [Google Scholar]
  • [45].Sawhney SN, Thiazole derivatives: Part I. Synthesis and antiinflammatory activity of some 2’-alkyl/aryl-2-aryl-4-methyl-4’,5-bithiazolyls and 2’-amino-substituted amino-2-aryl-4-methyl-4’,5-bithiazolyls., Indian J. Chem. B, 14 (1976) 552–555. [Google Scholar]
  • [46].Kambe S, Saito K, Kishi H, Sakurai A, Midorikawa H, A One-Step Synthesis of 4-Oxo-2-thioxopyrimidine Derivatives by the Ternary Condensation of Ethyl Cyanoacetate, Aldehydes, and Thiourea, Synthesis, (1979) 287–289. 10.1055/S-1979-28650. [DOI] [Google Scholar]
  • [47].Ding Y, Girardet JL, Smith KL, Larson G, Prigaro B, Wu JZ, Yao N, Parallel synthesis of 5-cyano-6-aryl-2-thiouracil derivatives as inhibitors for hepatitis C viral NS5B RNA-dependent RNA polymerase, Bioorg. Chem, 34 (2006) 26–38. 10.1016/j.bioorg.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • [48].Ram VJ, Chemotherapeutic agents. Part XX. Antileishmanial and immunoadjuvant activities of raonally designed thiopyrimidines., Indian J. Chem. B, 29 (1990) 1129–1133. [Google Scholar]
  • [49].Garcia N, Jose L, Lorente A, Soto JL, Synthesis of heterocyclic compounds. XXII. Preparation of 6-aryl-2-thioxopyrimidines, Heterocycles, 19 (1982) 305–311. [Google Scholar]
  • [50].Salem MAI, Madkour HMF, Marzouk MI, Azab ME, Mahmoud NFH, Pyrimidinthiones (Part I): Utility of 2-thioxopyrimidin-6-(1H)ones as ring transformer in the synthesis of fused bi- and tri-cyclic heterocyclic compounds and their potential biological activities., Phosphorus Sulfur Silicon Relat. Elem, 183 (2008) 2596–2614. 10.1080/10426500801967963. [DOI] [Google Scholar]
  • [51].Balalaie S, Bararjanian M, Rominger F, An efficient one-pot synthesis of 6-aryl-5-cyano-2-thiopyrimidinone derivatives and their piperidinium ionic forms, x-ray crystal structures., J.Het. Chem, 43 (2006) 821–826. 10.1002/jhet.5570430402. [DOI] [Google Scholar]
  • [52].Ram VJ, Chemotherapeutic Agents XVIII: Synthesis of π-Deficient Pyrimidines and Fused Pyrimidines as Leishmanicidal Agents., Arch. Pharm, 323 (1991) 895–899. 10.1002/ardp.2503241103. [DOI] [PubMed] [Google Scholar]
  • [53].Chen W, Huang YJ, Gundala SR, Yang H, Li M, Tai PC, Wang B, The first low microM SecA inhibitors, Bioorg. Med. Chem, 18 (2010) 1617–1625. 10.1016/j.bmc.2009.12.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].El-Agrody AM, Ali FM, Eid FA, El-Nassag MAA, El-Sherbeny G, Bedair AH, Synthesis and Antimicrobial Activity of Thioxopyrimidines and Related Derivatives, Phosphorus Sulfur Silicon Relat. Elem, 181 (2006) 839–864. 10.1080/10426500500272087. [DOI] [Google Scholar]
  • [55].Ertas M, Sahin Z, Bulbul EF, Bender C, Biltekin SN, Berk B, Yurttas L, Nalbur AM, Celik H, Demirayak S, Potent ribonucleotide reductase inhibitors: Thiazole-containing thiosemicarbazone derivatives, Arch. Pharm. (Weinheim), 352 (2019) e1900033 10.1002/ardp.201900033. [DOI] [PubMed] [Google Scholar]
  • [56].Sahin Z, Ertas M, Berk B, Biltekin SN, Yurttas L, Demirayak S, Studies on non-steroidal inhibitors of aromatase enzyme; 4-(aryl/heteroaryl)-2-(pyrimidin-2-yl)thiazole derivatives, Bioorg. Med. Chem, 26 (2018) 1986–1995. 10.1016/j.bmc.2018.02.048. [DOI] [PubMed] [Google Scholar]
  • [57].Ertas M, Sahin Z, Berk B, Yurttas L, Biltekin SN, Demirayak S, Pyridine-substituted thiazolylphenol derivatives: Synthesis, modeling studies, aromatase inhibition, and antiproliferative activity evaluation, Arch. Pharm. (Weinheim), 351 (2018) e1700272 10.1002/ardp.201700272. [DOI] [PubMed] [Google Scholar]
  • [58].Solinas M, Massi P, Cinquina V, Valenti M, Bolognini D, Gariboldi M, Monti E, Rubino T, Parolaro D, Cannabidiol, a non-psychoactive cannabinoid compound, inhibits proliferation and invasion in U87-MG and T98G glioma cells through a multitarget effect, PLoS One, 8 (2013) e76918 10.1371/journal.pone.0076918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Massi P, Vaccani A, Ceruti S, Colombo A, Abbracchio MP, Parolaro D, Antitumor effects of cannabidiol, a nonpsychoactive cannabinoid, on human glioma cell lines, J. Pharmacol. Exp. Ther, 308 (2004) 838–845. 10.1124/jpet.103.061002. [DOI] [PubMed] [Google Scholar]
  • [60].Jarvis MF, Schulz R, Hutchison AJ, Do UH, Sills MA, Williams M, [3H]CGS 21680, a selective A2 adenosine receptor agonist directly labels A2 receptors in rat brain, J. Pharmacol. Exp. Ther, 251 (1989) 888–893. [PubMed] [Google Scholar]
  • [61].Varano F, Catarzi D, Vincenzi F, Betti M, Falsini M, Ravani A, Borea PA, Colotta V, Varani K, Design, Synthesis, and Pharmacological Characterization of 2-(2-Furanyl)thiazolo[5,4-d]pyrimidine-5,7-diamine Derivatives: New Highly Potent A2A Adenosine Receptor Inverse Agonists with Antinociceptive Activity, J. Med. Chem, 59 (2016) 10564–10576. 10.1021/acs.jmedchem.6b01068. [DOI] [PubMed] [Google Scholar]
  • [62].Nayak A, Chandra G, Hwang I, Kim K, Hou X, Kim HO, Sahu PK, Roy KK, Yoo J, Lee Y, Cui M, Choi S, Moss SM, Phan K, Gao ZG, Ha H, Jacobson KA, Jeong LS, Synthesis and anti-renal fibrosis activity of conformationally locked truncated 2-hexynyl-N(6)-substituted-(N)-methanocarba-nucleosides as A3 adenosine receptor antagonists and partial agonists, J. Med. Chem, 57 (2014) 1344–1354. 10.1021/jm4015313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Araiz C, Yan A, Bettedi L, Samuelson I, Virtue S, McGavigan AK, Dani C, Vidal-Puig A, Foukas LC, Enhanced beta-adrenergic signalling underlies an age-dependent beneficial metabolic effect of PI3K p110alpha inactivation in adipose tissue, Nat. Commun, 10 (2019) 1546 10.1038/s41467-019-09514-1. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3

RESOURCES