Next Article in Journal
Fluorination of Naturally Occurring N6-Benzyladenosine Remarkably Increased Its Antiviral Activity and Selectivity
Next Article in Special Issue
Tetrabutylammonium Iodide–Promoted Thiolation of Oxindoles Using Sulfonyl Chlorides as Sulfenylation Reagents
Previous Article in Journal
High-Resolution Inhibition Profiling Combined with HPLC-HRMS-SPE-NMR for Identification of PTP1B Inhibitors from Vietnamese Plants
Previous Article in Special Issue
Study on the Alkylation Reactions of N(7)-Unsubstituted 1,3-Diazaoxindoles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Antitumor Evaluation and Molecular Docking of New Morpholine Based Heterocycles

by
Zeinab A. Muhammad
1,
Mastoura M. Edrees
1,2,
Rasha A. M. Faty
3,
Sobhi M. Gomha
3,*,
Seham S. Alterary
4 and
Yahia N. Mabkhot
4,*
1
Department of Organic Chemistry, National Organization for Drug Control and Research (NODCAR), Giza 12311, Egypt
2
Department of Chemistry, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
3
Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt
4
Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Submission received: 7 June 2017 / Revised: 8 July 2017 / Accepted: 17 July 2017 / Published: 20 July 2017
(This article belongs to the Collection Heterocyclic Compounds)

Abstract

:
A series of new morpholinylchalcones was prepared and then used as building blocks for constructing a series of 7-morpholino-2-thioxo-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-ones via their reaction with 6-aminothiouracil. The latter thiones reacted with the appropriate hydrazonoyl chloride to give the corresponding pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones. The assigned structures for all the newly synthesized compounds were confirmed on the basis of elemental analyses and spectral data and the mechanisms of their formation were also discussed. Most of the synthesized compounds were tested for in vitro activity against human lung cancer (A-549) and human hepatocellular carcinoma (HepG-2) cell lines compared with the employed standard antitumor drug (cisplatin) and the results revealed that compounds 8, 4e and 7b have promising activities against the A-549 cell line (IC50 values of 2.78 ± 0.86 μg/mL, 5.37 ± 0.95 μg/mL and 5.70 ± 0.91 μg/mL, respectively) while compound 7b has promising activity against the HepG-2 cell lines (IC50 = 3.54 ± 1.11 μg/mL). Moreover, computational studies using MOE 2014.09 software supported the biological activity results.

1. Introduction

In recent years, interest in the chemistry of hydrazonoyl halides has been renewed because of the development of novel synthetic routes and their use as synthons for compounds that find many applications [1,2,3,4,5,6,7,8,9,10]. The literature reveals that the presence of a morpholine ring on a heterocyclic system contributes to enhanced pharmacological activities in many cases [11,12,13,14,15,16]. In addition, many pyridopyrimidine derivatives have a variety of effects of chemical and biological significance such as antimicrobial, analgesic, anti-allergic, antitumor, antihypertensive, antileishmanial, antifolate, anti-inflammatory, ant-tuberculostatic, anticonvulsant, diuretic, potassium sparing, and anti-aggressive activities [17,18,19,20,21,22,23,24,25,26]. In view of all these reports and in continuation of our previous work on the synthesis of bioactive heterocyclic compounds [27,28,29,30,31,32,33], herein, we were interested in synthesizing a new series of morpholinylchalcones and utilizing them as precursors for the synthesis of pyridopyrimidines and triazolopyridopyrimidines to evaluate their anticancer activities.

2. Results and Discussion

2.1. Chemistry

The required starting compounds, namely 1-morpholino-1-(2-phenylhydrazono)propan-2-one (2a) and 1-morpholino-1-(2-(p-tolyl)hydrazono)propan-2-one (2b) were prepared by a previously reported method (Scheme 1) [34]. The morpholinohydrazonopropanone derivatives 2a,b were next used as starting compounds for preparation of a number of novel chalcone derivatives. Thus stirring a mixture of 1-morpholino-1-(2-arylhydrazono)propan-2-ones 2a,b and the appropriate benzaldehyde derivatives 3ac in glacial acetic acid in the presence of a catalytic amount of concentrated H2SO4, gave the morpholinylchalcone derivatives 4af in good yield (Scheme 1). The assigned structures of the products 4af was confirmed based on both elemental analyses and spectral data (IR, 1H-NMR and MS). The IR spectra of compounds 4af revealed in each case two absorption bands in the regions υ 3250–3236, 1683–1676 cm−1 attributed to the NH and C=O groups. The 1H-NMR spectra of compounds 4af showed in each case three signals assigned for the CH=CH and NH in addition to the expected signals for the morpholine and aromatic protons (see Experimental). For example, the 1H-NMR spectrum of compound 4a taken as a typical example of the products 4, revealed three signals at δ = 7.45 (d, J = 8 Hz, 1H, CH=CH), 7.79 (d, J = 8 Hz, 1H, CH=CH), 10.58 (brs, 1H, NH) and morpholine protons ppm in addition to the characteristic signals of the aromatic protons. The 13C-NMR spectrum of compound 4a showed three signals at δ = 25.76, 68.04, 188.22 ppm assignable for the morpholine-C and the carbonyl-C, in addition to twelve aromatic and olifinic carbons. Moreover, the mass spectrum of 4a revealed a molecular ion peak at m/z = 335 which is consistent with its expected molecular weight.
The structures assigned for products 4 were further evidenced via alternative method. Thus, condensation of 2-oxo-N-phenylpropanehydrazonoyl chloride (1) with benzaldehyde 3a in glacial acetic acid in the presence of catalytic amount of concentrated H2SO4, afforded 2-oxo-N,4-diphenylbut-3-enehydrazonoyl chloride (5). Refluxing an equimolar amounts of 5 and morpholine in ethanol for 3 h, gave a product identical in all respects (m.p., mixed m.p. and IR spectra) with compound 4a obtained from the 2a + 3a reaction (Scheme 1).
The morpholinylchalcone derivatives 4ae were then used for the preparation of novel series of 2-thioxo-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-one derivatives bearing morpholine moieties. Thus, reaction of 6-amino-2-thioxo-2,3-dihydropyrimidin-4(1H)-one (6) and the appropriate chalcones 4ae in ethanol under reflux for 8–12 h led to the formation of one isolable product as evidenced by TLC analysis of the crude product (Scheme 2). The structures of the products was identified to be 7ae based on elemental analysis and spectral data. For example, the IR spectrum of 7ae revealed three absorption bands near υmax 3125, 3243, 3447 cm−1 due to the three NH groups, and another absorption band near υmax 1679 cm−1 attributed to the carbonyl group. The 1H-NMR spectra displayed three singlet signals near δ 9.67, 10.65 and 11.17 ppm attributed to the three NH protons (disappeared by d2o), in addition to the expected signals due to the morpholine and aryl protons. Also, the 13C-NMR spectra showed the expected number of aliphatic and aromatic signals. The mass spectra of the products 7ae revealed in each case a molecular ion peak m/z at the expected molecular weight calculated for each compound (see Experimental).
For a much more rigorous identification of the structures of compounds 7ae, a comparison with authentic material prepared from the reaction between thione 8 (obtained from reaction of chalcone 5 and compound 6) with morpholine was achieved, the product obtained from this reaction was identical to that from the reaction of 4a and 6 (Scheme 2).
On the other hand, compound 7d was reacted with hydrazonoyl halides 1a,b (9a,b) in dioxane in the presence of triethylamine to give 1,5-dihydropyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidine derivatives 10ad, respectively (Scheme 3). Spectroscopic data as well as elemental analyses of the obtained products were in complete agreement with the assigned structures 10ad (see Experimental part).
An alternative synthetic route for the pyridotriazolopyrimidine derivatives 10ad was executed whereby compound 4d was refluxed with 3-acetyl-7-amino-1-phenyl-[1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-one (11) in ethanol afforded the respective authentic sample 10a (Scheme 3).

2.2. Cytotoxic Activity

The in vitro growth inhibitory activity of the newly synthesized compounds 4a,c,e,f, 5, 7ac, 8 and 10ad was investigated against two carcinoma cell lines, a human lung cancer cell line (A-549) and a human hepatocellular carcinoma cell line (HepG-2), in comparison with a well-known anticancer standard drug (cisplatin) under the same conditions using a colorimetric MTT assay. Data generated were used to plot a dose response curve of which the concentration of test compounds required to kill 50% of cell population (IC50) was determined. The results are depicted in Table 1 revealed that the descending order of activity of the newly synthesized compounds towards the lung carcinoma cell line (A-549) were as follows: 8 > 4e > 7b > 7c > 4f > 7a > 5 > 10d > 10c > 4a > 4c > 10a > 10b. The descending order of activity of the newly synthesized compounds towards the human hepatocellular carcinoma cell line (HepG-2) were as follows: 7b > 7c > 10d > 4f > 5 > 7a > 10c > 4e > 10b > 4c > 4a > 10a > 8.

Examination of the SAR Leads to the Following Conclusions

  • The results revealed that all the tested compounds showed inhibitory activity to the tumor cell lines in a concentration dependent manner.
  • The activities of the synthesized compounds depend on the structural skeleton and electronic environment of the molecules.
  • Compounds 8, 4e and 7b were the most active (IC50 values of 2.78 ± 0.86, 5.37 ± 0.95 and 5.70 ± 0.91 μg/mL, respectively) against the lung carcinoma cell line (A-549), compared with cisplatin reference drug with IC50 value of 0.95 ± 0.90 μg/mL (Figure 1), while the remaining compounds have moderate inhibitory activity (IC50 = 6.79 ± 1.11 − 26.8 ± 0.75 µg/mL).
  • Compounds 7b, 7c, 10d and 4f were the most active (IC50 value of 3.54 ± 1.11, 8.42 ± 1.15, 8.72 ± 0.89 and 9.78 ± 0.78 μg/mL, respectively) against the human hepatocellular carcinoma cell line (HepG-2), compared with the reference drug cisplatin with an IC50 value of 1.40 ± 1.1μg/mL (Figure 1). The other compounds have moderate inhibitory activity (IC50 = 12.4 ± 0.98 − 29.9 ± 0.93 µg/mL).
  • Among the morpholinylchalcone derivatives, the dimethylchalcone 4e is the most active one against the A549 (IC50 = 5.37 ± 0.95 μg/mL) line, while the methylchlorochalcone 4f is the most active one against the HepG-2 cell line (A549) (IC50 = 9.78 ± 0.78 μg/mL).
  • For pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones 10ad: Compounds 10c and 10d (substituted with COOEt group at position 3) have more in vitro inhibitory activity than compounds 10a and 10b (substituted with a COCH3 group at position 3). Also compound 10d is more active than 10c where the p-substitution with a methyl group increases the activity via its +I effect.

2.3. Molecular Docking

A major problem in chemotherapy is resistance to the chemotherapeutic agents. There are generally two major forms of resistance encountered in the clinic. One is intrinsic resistance, which is a property of the tumor cells and is not triggered by drug exposure. The other is known as acquired resistance, which occurs following exposure to the drug(s). Anti-folates such as methotrexate (MTX) and fluoropyrimidines such as 5-fluorouracil (5-FU) have been used in the clinic for the management of childhood acute lymphoblastic leukemias (ALL) and colorectal cancer, respectively, with modest success. Among the several enzymes that participate in the synthesis of nucleic acid precursors, DHFR is an important target for several human dis-eases, namely, protozoal, bacterial and fungal infections, psoriasis, autoimmune diseases and neoplastic diseases. Traditionally, several DHFR inhibitors are reported as potential drug candidates in various diseases.
In the late 1950s DHFR was discovered as a ubiquitous enzyme with respect to drug design due to its central role in the synthesis of DNA. Most eukaryotic organisms synthesize the essential metabolite thymidylate via the thymidylate cycle, which consists of three enzymes: serine hydroxymethyl transferase; thymidylate synthase (TS) and the much promising DHFR. It reduces the NADPH-dependent 7,8-DHF to 5,6,7,8-THF utilizing NADPH2+ as cofactor. This THF acts in the conversion of deoxyuridylate (dUMP) to deoxythymidylate (dTMP) by thymidylate synthetase. Inhibition of TS or of DHFR leads to thymineless death, which has found clinical utility as antitmalarial, antiprotozoal and antimicrobial agents.
The MOE 2014.010 package software was used to analyze all docking poses and binding energies between compound 7b and the enzyme dihydrofolate reductase (DHFR) to evaluate the affinity of 7b according to its binding energy with the enzyme.
From Figure 2 and Figure 3 which represent all the binding energies of the two compounds.it is clear that the total binding energy of compound 7b equals −1.6 E (Kcal/mol), showing good affinity with the DHFR enzyme by forming four pi-hydrogen interactions with binding energy −1.4 E (kcal/mol) , one hydrogen acceptor interaction with binding energy −0.2 E (Kcal/mol) and one pi-pi interaction with almost zero binding energy.on the other hand compound comp showing affinity to the DHFR enzyme by −1.3 E (Kcal/mol) by making one hydrogen donor interaction with −0.6 E (Kcal/mol) and tow pi-hydrogen interactions with −0.7 E (Kcal/mol).

Bioactivity and ADME Toxicity

Due to its impact on society, the design of new drugs has the potential to interest a wide audience, and provides a rare opportunity to introduce several concepts in chemistry and biochemistry. Drug design can be seen as a multi-objective cyclic optimization process. Indeed, it is important to develop the understanding not only that a drug is generally an effective ligand for a protein of therapeutic interest, but also that these molecules need to have drug-like properties. Computer-aided drug design and bioinformatics approaches play a fundamental role in addressing these different challenges. Basically, drug design consists of the conception of molecules that are complementary to the protein target in terms of 3D-shape and charge distribution, to optimize molecular recognition and binding. On the contrary, ligand based approaches rely on the knowledge implicitly contained in the chemical structure or physical properties of other molecules that bind to the biological target of interest.
Molecular properties in relation to lipophilicity, drug likeness, or pharmacokinetics (PK), for example. These molecular properties are fundamental in drug design. Indeed, although a high affinity for the protein target is essential, it is not sufficient for the designed small molecule to become a drug: to obtain a therapeutic effect, the molecule needs to reach its target in the body, and stay there long enough for the expected biological events to occur. Therefore, to support efficiently the design of new drugs, it is important to predict their PK behaviors with computer-aided approaches.
A computational study was also carried out including prediction of pharmacokinetic properties, toxicity and bioactivity studies. In Table 2 Molecular properties were calculated on the basis of Lipinski’s rule and its components, Furthermore, TPSA values of the tested compounds, the prediction of bioactivity scores of the compounds were recorded by calculating the activity scores of GPCR, ion channel modulator, kinase inhibitor, nuclear receptor ligand, protease inhibitor and enzyme inhibitor.
Physicochemical properties, with an emphasis on lipophilicity computed by a variety of methodologies to enable a consensus approach by calculating the molecular weight and TPSA of the tested compounds. Druglikeness, estimated by simple rules to evaluate oral bioavailability through evaluating the compounds according to Lipinski rule five and also Pharmacokinetics, which predicts several ADME behaviors (e.g., substrate of P-glycoprotein, cytochromes P450, gastrointestinal absorption, brain blood barrier) by binary classification models relying on physicochemical descriptors. While medicinal chemistry that gives a score for synthetic accessibility, leadlikeness and pan-assay interference structure of molecules together with structural alerts for problematic fragments.
The docking study was performed using the MOE 2014.010 software. The crystal structure of the enzyme dihydrofolate reductase (DHFR, PDB ID (3NU0)) was downloaded out from Protein Data Bank website. Regularization and optimization for protein and ligand were performed. Determination of the essential amino acids in the binding site was carried out and compared with that presented in the literature. The performance of the docking method was evaluated by re-docking the crystal ligand into the assigned active dihydrofolate reductase (DHFR) enzyme to determine a RMSD value. Interactive docking to the selected active site was carried out for all the conformers of interesting compounds. Each docked compound was assigned a score according to its fit in the ligand binding pocket (LBP) and its binding mode.

3. Experimental

3.1. General Information

Melting points were measured on an Electrothermal IA 9000 series digital melting point apparatus (Bibby Sci. Lim. Stone, Staffordshire, UK). IR spectra were measured on Shimadzu FTIR 8101 PC infrared spectrophotometers (Shimadzu, Tokyo, Japan) in potassium bromide discs. NMR spectra were measured on a Varian Mercury VX-300 NMR spectrometer (Varian, Inc., Karlsruhe, Germany) operating at 300 MHz (1H-NMR) and run in deuterated dimethylsulfoxide (DMSO-d6). Chemical shifts were related to that of the solvent. Mass spectra were recorded on a Shimadzu GCMS-QP1000 EX mass spectrometer at 70 eV. Elemental analyses were measured by using an Elementar Vario LIII CHNS analyzer (GmbH & Co.KG, Hanau, Germany). Antitumor activity of the products was measured at the Regional Center for Mycology and Biotechnology at Al-Azhar University, Cairo, Egypt. Hydrazonoyl halides were prepared following a literature method [35].

3.1.1. Synthesis of Chalcones 4af

A mixture of 1-morpholino-1-(2-arylhydrazono)propan-2-ones 2a,b (10 mmol) and the appropriate benzaldehyde derivatives 3ac (10 mmol) in glacial acetic acid (20 mL) containing a few drops of concentrated H2SO4 was stirred at room temperature for 4 h (monitored by TLC). The formed precipitate after cooling was isolated by filtration, washed with ethanol, dried and recrystallized from ethanol to give products 4af.
1-Morpholino-4-phenyl-1-(2-phenylhydrazono)but-3-en-2-one (4a), Yellow solid, (80% yield), m.p. 203–205 °C; IR (KBr) υmax 1606 (C=N), 1678 (C=O), 2985, 2919 (C–H), 3243 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.30–2.34 (m, 4H, 2CH2), 3.65 (m, 4H, 2CH2), 6.57–7.33 (m, 10H, Ar–H), 7.45 (d, J = 8 Hz, 1H, CH=CH), 7.79 (d, J = 8 Hz, 1H, CH=CH), 10.58 (brs, 1H, NH); 13C-NMR (DMSO-d6): δ 25.76, 68.04 (CH2), 112.76, 114.82, 115.30, 122.76, 124.07, 129.49, 129.78, 129.89, 130.15, 132.25, 140.67 (Ar–C and C=N), 188.22 (C=O); MS m/z (%) 335 (M+, 5), 277 (20), 213 (24), 198 (62), 159 (47), 141 (38), 99 (43), 80 (42), 67 (48), 43 (100). Anal. Calcd. for C20H21N3O2 (335.40): C, 71.62; H, 6.31; N, 12.53. Found: C, 71.95; H, 6.11; N, 12.13%.
1-Morpholino-1-(2-phenylhydrazono)-4-(p-tolyl)but-3-en-2-one (4b), Yellow solid, (75% yield), m.p. 200–202 °C; IR (KBr) υmax 1599 (C=N), 1680 (C=O), 3021, 2949 (C–H2), 1H-NMR (DMSO-d6) δ 7.05–7.60 (m, 9H, Ar-H), 7.87 (d, J = 8 Hz, 1H, CH=CH), 8.37 (d, J = 8 Hz, 1H, CH=CH), 10.59 (brs, 1H, NH); MS m/z (%) 349 (M+, 6), 340 (12), 268 (28), 236 (11), 164 (6), 123 (7), 108 (19), 92 (72), 76 (55), 68 (22), 65 (65), 43 (100). Anal. Calcd. for C21H23N3O2 (349.43): C, 72.18; H, 6.63; N, 12.03. Found: C, 72.51; H, 6.40; N, 11.80%.
4-(4-Chlorophenyl)-1-morpholino-1-(2-phenylhydrazono)but-3-en-2-one (4c), Yellow solid, (85% yield), m.p. 192–194 °C; IR (KBr) υmax 1599 (C=N), 1680 (C=O), 3019, 2950 (C–H), 3245 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.20–2.35 (m, 4H, 2CH2), 3.54 (m, 4H, 2CH2), 7.09–7.59 (m, 9H, Ar–H), 7.84 (d, J = 8 Hz, 1H, CH=CH), 8.37 (d, J = 8 Hz, 1H, CH=CH), 10.75 (brs, 1H, NH); MS m/z (%) 371 (M+ + 2, 1), 369 (M+, 4), 196 (3), 165 (4), 123 (5), 111 (8), 97 (11), 83 (20), 77 (20), 69 (42), 57 (47), 43 (100), 41 (25). Anal. Calcd. for C20H20N3O2Cl (369.84): C, 64.95; H, 5.45; N, 11.36. Found: C, 63.78; H, 5.05; N, 11.02%.
1-Morpholino-4-phenyl-1-(2-(p-tolyl)hydrazono)but-3-en-2-one (4d), Yellow solid, (70% yield), m.p. 188–190 °C; IR (KBr) υmax 1599 (C=N), 1683 (C=O), 3021, 2948 (C-H), 3236 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.18–2.47 (m, 4H, 2CH2), 2.53 (s, 3H, CH3), 3.54 (m, 4H, 2CH2), 7.06–7.60 (m, 9H, Ar–H), 7.86 (d, J = 8 Hz, 1H, CH=CH), 8.38 (d, J = 8 Hz, 1H, CH=CH), 10.45 (brs, 1H, NH); 13C-NMR (DMSO-d6): δ 21.83 (CH3), 26.76, 69.38, (CH2), 111.76, 114.82, 115.30, 123.76, 124.07, 129.39, 129.78, 130.89, 131.15, 132.25, 140.67 (Ar–C and C=N), 188.22 (C=O); MS m/z (%) 349 (M+, 6), 340 (12), 268 (28), 236 (11), 164 (6), 123 (7), 108 (19), 92 (72), 76 (55), 68 (22), 65 (65), 43 (100). Anal. Calcd. for C21H23N3O2 (349.43): C, 72.18; H, 6.63; N, 12.03. Found: C, 72.55; H, 5.05; N, 12.44%.
1-Morpholino-4-(p-tolyl)-1-(2-(p-tolyl)hydrazono)but-3-en-2-one (4e), Yellow solid, (70% yield), m.p. 203–205 °C; IR (KBr) υmax 1608 (C=N), 1678 (C=O), 2985, 2918 (C–H), 3250 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.13–2.41 (m, 4H, 2CH2), 2.45 (s, 3H, CH3), 2.58 (s, 3H, CH3), 3.54 (m, 4H, 2CH2), 6.91–7.72 (m, 8H, Ar–H), 7.82 (d, J = 8 Hz, 1H, CH=CH), 8.40 (d, J = 8 Hz, 1H, CH=CH), 10.80 (brs, 1H, NH); MS m/z (%) 363 (M+, 2), 321 (2), 291 (3), 275 (4), 233 (4), 208 (5), 178 (3), 119 (40), 106 (45), 91 (100), 77 (37), 65 (49), 43 (75). Anal. Calcd. for C22H25N3O2 (363.45): C, 72.70; H, 6.93; N, 11.56. Found: C, 73.02; H, 6.55; N, 11.16%.
4-(4-Chlorophenyl)-1-morpholino-1-(2-(p-tolyl)hydrazono)but-3-en-2-one (4f), Yellow solid, (80% yield), m.p. 206–208 °C; IR (KBr) υmax 1613 (C=N), 1676 (C=O), 2974, 2940 (C–H), 3243 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.24 (s, 3H, CH3), 2.42–2.47 (m, 4H, 2CH2), 3.74 (m, 4H, 2CH2), 7.12–7.53 (m, 8H, Ar–H), 7.79 (d, J = 8 Hz, 1H, CH=CH), 7.91 (d, J = 8 Hz, 1H, CH=CH), 10.58 (brs, 1H, NH); MS m/z (%) 385 (M+ + 2, 1), 383 (M+, 3), 313 (3), 262 (3), 210 (4), 165 (4), 139 (24), 121 (10), 106 (27), 91 (36), 77 (42), 69 (58), 57 (62), 43 (100). Anal. Calcd. for C21H22N3O2Cl (383.87): C, 65.71; H, 5.78; N, 6.94. Found: C, 65.95; H, 5.55; N, 6.53%.

3.1.2. Synthesis of 2-Oxo-N,4-diphenylbut-3-enehydrazonoyl Chloride (5)

A mixture of 2-oxo-N-phenylpropanehydrazonoyl chloride 1a (0.98 g, 5 m mol) and the benzaldehyde 3a (0.53 g, 5 mmol) in glacial acetic acid (20 mL) containing drops of concentrated H2SO4 was stirred at room temperature for 4 h (monitored by TLC). The formed precipitate after cooling was isolated by filtration, washed with ethanol, dried and recrystallized from ethanol to give product 5 as yellow solid, (70% yield), m.p. 209–211 °C; IR (KBr) υmax 1601 (C=N), 1680 (C=O), 2950, 3018, 3055, (C–H), 3245 (NH) cm−1; 1H-NMR (DMSO-d6) δ 6.99 (d, J = 6 Hz, 1H, CH=CH–CO), 7.01–7.44 (m, 10H, Ar–H), 7.33 (d, J = 6 Hz, 1H, CH=CH–CO), 10.64 (brs, 1H, NH); MS m/z (%) 286 (M+ + 2, 2), 284 (M+, 7), 262 (9), 239 (15), 137 (12), 123 (11), 109 (31), 97 (22), 81 (50), 69 (64), 57 (63), 43 (100). Anal. Calcd. for C16H13ClN2O (284.74): C, 67.49; H, 4.60; N, 9.84. Found: C, 67.82; H, 4.20; N, 9.55%.

3.1.3. Alternative Synthesis of 4a

Equimolar amounts of 5 (0.284 g, l mmol) and morpholine (0.87 g, 1 mmol) in ethanol (15 mL) was refluxed for 3 h, gave product identical in all respects (m.p., mixed m.p. and IR spectra) with compound 4a which obtained from reaction of 2a + 3a.

3.1.4. Synthesis of Thiones 7ae

A mixture of 6-amino-2-thioxo-2,3-dihydropyrimidin-4(1H)-one (6) (0.143 g, 1 mmol) and the appropriate chalcones 4ae (1 mmol) in ethanol (20 mL) was refluxed for 8–12 h (monitored by TLC). The formed precipitate after cooling was isolated by filtration, washed with ethanol, dried and recrystallized from ethanol to give products 7ae.
7-(Morpholino(2-phenylhydrazono)methyl)-5-phenyl-2-thioxo-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-one (7a), Yellow solid, (70% yield), m.p. 150–152 °C; IR (KBr) υmax 1613 (C=N), 1679 (C=O), 2919, 2983, 3030 (C–H), 3125, 3243, 3447 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 2.30–2.42 (m, 4H, 2CH2), 3.01–308 (m, 4H, 2CH2), 7.01–7.94 (m, 11H, Ar–H), 9.67, 10.65, 11.17 (3brs, 3H, 3NH); 13C-NMR (DMSO-d6): δ 25.80, 78.62 (CH2), 112.96, 115.30, 119.41, 123.23, 123.27, 125.07, 128.83, 129.74, 131.13, 132.11, 132.96, 138.02, 142.96, 148.36, 154.78 (Ar–C and C=N), 162.08 (C=O), 175.01 (C=S); MS m/z (%) 458 (M+, 3), 313 (5), 236 (6), 192 (13), 152 (4), 129 (9), 121 (10), 107 (13), 98 (20), 97 (33), 81 (37), 71 (50), 57 (79), 43 (100). Anal. Calcd. for C24H22N6O2S (458.54): C, 62.87; H, 4.84; N, 18.33. Found: C, 63.17; H, 4.71; N, 18.16%.
7-(Morpholino(2-phenylhydrazono)methyl)-2-thioxo-5-(p-tolyl)-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-one (7b), Yellow solid, (70% yield), m.p. 158–160 °C; IR (KBr) υmax 1608 (C=N), 1679 (C=O), 2926, 3057 (C–H), 3175, 3247, 3425 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 2.24 (s, 3H, CH3), 2.30 (m, 4H, 2CH2), 3.31 (m, 4H, 2CH2), 7.12–7.33 (m, 10H, Ar–H), 10.57, 11.48, 11.58 (3brs, 3H, 3NH); MS m/z (%) 472 (M+, 1), 368 (9), 313 (7), 237 (7), 178 (6), 143 (54), 115 (16), 98 (22), 83 (24), 69 (44), 43 (100). Anal. Calcd. for C25H24N6O2S (472.57): C, 63.54; H, 5.12; N, 17.78. Found: C, 63.68; H, 4.05; N, 17.59%.
5-(4-Chlorophenyl)-7-(morpholino(2-phenylhydrazono)methyl)-2-thioxo-2,3-dihydropyrido [2,3-d]pyrimidin-4(1H)-one (7c), Yellow solid, (70% yield), m.p. 149–151 °C; IR (KBr) υmax 1608 (C=N), 1695 (C=O), 2954, 3038 (C–H), 3172, 3397, 3412 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 2.36 (m, 4H, 2CH2), 3.38 (m, 4H, 2CH2), 6.99–7.44 (m, 10H, Ar–H), 10.64, 11.49, 11.59 (3brs, 3H, 3NH); MS m/z (%) 494 (M+, 12), 492 (39), 390 (38), 202 (39), 161 (23), 127 (100), 84 (80), 77 (82). Anal. Calcd. for C24H21ClN6O2 (492.98): C, 58.47; H, 4.29; N, 17.05. Found: C, 58.78; H, 4.01; N, 16.70%.
7-(Morpholino(2-(p-tolyl)hydrazono)methyl)-5-phenyl-2-thioxo-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-one (7d), Yellow solid, (70% yield), m.p. 183–185 °C; IR (KBr) υmax 1600 (C=N), 1637 (C=O), 2950, 3058 (C–H), 3246, 3330, 3426 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 2.23 (m, 4H, 2CH2), 2.47 (s, 3H, CH3), 3.35 (m, 4H, 2CH2), 6.95–7.42 (m, 10H, Ar–H), 10.64, 11.49, 11.59 (3brs, 3H, 3NH); MS m/z (%) 472 (M+, 10), 368 (9), 313 (7), 237 (7), 178 (6), 143 (54), 115 (16), 98 (22), 83 (24), 69 (44), 55 (45), 43 (100). Anal. Calcd. for C25H24N6O2S (472.56): C, 63.54; H, 5.12; N, 17.78. Found: C, 63.78; H, 4.05; N, 17.19%.
5-(4-Chlorophenyl)-7-(morpholino(2-(p-tolyl)hydrazono)methyl)-2-thioxo-2,3-dihydropyrido[2,3-d]-pyrimidin-4(1H)-one (7e), Yellow solid, (70% yield), m.p. 160–162 °C; IR (KBr) υmax 1601 (C=N), 1676 (C=O), 2972, 3031 (C–H), 3243, 3326, 3425 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 2.24 (s, 3H, CH3), 2.46 (m, 4H, 2CH2), 3.32 (m, 4H, 2CH2), 7.12–7.33 (m, 9H, Ar–H), 10.57, 11.49, 11.59 (3brs, 3H, 3NH); 13C-NMR (DMSO-d6): δ 20.80 (CH3), 25.76, 78.62 (CH2), 111.93, 115.30, 122.76, 125.37, 130.15, 132.25, 135.47, 140.67, 142.01, 145.96, 147.13, 148.22, 152.84, 154.77 (Ar–C and C=N), 162.07 (C=O), 175.02 (C=S); MS m/z (%) 509 (M+ + 2, 5), 507 (M+, 14), 409 (11), 304 (38), 272 (24), 240 (42), 208 (20), 195 (17), 174 (39), 163 (23), 131 (28), 79 (100). Anal. Calcd. for C25H23ClN6O2S (507.01): C, 59.22; H, 4.57; N, 16.58. Found: C, 59.25; H, 4.05; N, 16.21%.

3.1.5. Alternative Synthesis of 7a

Equimolar amounts of 8 (0.407 g, l mmol) and morpholine (0.87 g, 1 mmol) in ethanol (15 mL) was refluxed for 3 h, gave product identical in all respects (m.p., mixed m.p. and IR spectra) with compound 7a which obtained from reaction of 4a + 6.
Synthesis of 4-oxo-N,5-diphenyl-2-thioxo-1,2,3,4-tetrahydropyrido[2,3-d]pyrimidine-7-carbohydrazonoyl chloride (8), A mixture of 5 (0.568 g, 2 mmol) and 6-amino-2-thioxo-2,3-dihydropyrimidin-4(1H)-one (6) (0.286 g, 2 mmol) in ethanol (20 mL) was refluxed for 6 h (monitored by TLC). The formed precipitate after cooling was isolated by filtration, washed with ethanol, dried and recrystallized from ethanol to give product 8 as yellow solid, Yellow solid, (70% yield), m.p. 203–205 °C; IR (KBr) υmax 1588 (C=N), 1641 (C=O), 2973, 3097, 3199 (C–H), 3242, 3316, 3424 (3NH) cm−1; 1H-NMR (DMSO-d6) δ 6.35–7.43 (m, 11H, Ar–H), 10.64, 11.49, 11.59 (3brs, 3H, 3NH); MS m/z (%) 409 (M+ + 2, 4), 407 (M+, 10), 392 (25), 370 (10), 344 (26), 332 (15), 310 (100), 283 (33), 268 (74), 108 (10), 92 (39), 77 (24), 56 (16). Anal. Calcd. for C20H14ClN5OS (407.88): C, 58.90; H, 3.46; N, 17.17. Found: C, 59.24; H, 3.15; N, 16.89%.

3.1.6. Synthesis of Pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones 10ad

To a mixture of equimolar amounts of thione 7d (0.472 g, 1 mmol) and the appropriate hydrazonoyl halides 1a,b (9a,b) (1 mmol of each) in dioxane (20 mL) was added triethylamine (0.1 mL, 1 mmol). The reaction mixture was refluxed till all of the starting materials have disappeared and hydrogen sulfide gas ceased to evolve (6–12 h monitored by TLC). The solvent was evaporated and the residue was treated with methanol. The solid that formed was filtered off and crystallized from the appropriate solvent to give compounds 10ad.
3-Acetyl-8-(morpholino(2-(p-tolyl)hydrazono)methyl)-1,6-diphenylpyrido[2,3-d][1,2,4]triazolo[4,3-a]-pyrimidin-5(1H)-one (10a), Browen solid, (70% yield), m.p. 230–232 °C; IR (KBr) υmax 1594 (C=N), 1654, 1675 (2C=O), 2933, 3042 (C–H), 3429 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.24 (s, 3H, CH3), 2.42 (m, 4H, 2CH2), 2.61 (s, 3H, CH3), 3.54 (m, 4H, 2CH2), 6.59–7.93 (m, 15H, Ar–H), 11.22 (brs, 1H, NH); 13C-NMR (DMSO-d6): δ 20.51, 25.58 (CH3), 26.03, 66.80 (CH2), 113.17, 114.84, 115.36, 120.58, 122.46, 124.03, 125.64, 125.77, 129.43, 129.61, 129.81, 129.91, 130.01, 130.30, 130.33, 130.46, 130.78, 132.16, 134.83, 140.43 (Ar–C and C=N), 168.19, 196.22 (2C=O); MS m/z (%) 598 (M+, 3), 353 (12), 247 (20), 196 (15), 121 (15), 108 (27), 105 (24), 92 (49), 77 (47), 69 (21), 65 (25), 43 (100). Anal. Calcd. for C34H30N8O3 (598): C, 68.21; H, 5.05; N, 18.72. Found: C, 68.55; H, 4.85; N, 18.41%.
3-Acetyl-8-(morpholino(2-(p-tolyl)hydrazono)methyl)-6-phenyl-1-(p-tolyl)pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-one (10b), Brown solid, (70% yield), m.p. 238–240 °C; IR (KBr) υmax 1596 (C=N), 1653, 1682 (2C=O), 2970, 3027 (C–H), 3434 (NH) cm−1; 1H-NMR (DMSO-d6) δ 2.24 (s, 3H, CH3), 2.37 (m, 4H, 2CH2), 2.40 (s, 3H, CH3), 2.63 (s, 3H, CH3), 3.41 (m, 4H, 2CH2), 7.03–7.76 (m, 14H, Ar–H), 11.17 (brs, 1H, NH); MS m/z (%) 612 (M+, 4), 519 (4), 494 (7), 460 (13), 443 (63), 429 (34), 400 (30), 384 (32), 337 (12), 315 (18), 291 (16), 216 (30), 201 (62), 187 (53), 126 (43), 68 (100). Anal. Calcd. for C35H32N8O3 (612.68): C, 68.61; H, 5.26; N, 18.29. Found: C, 68.93; H, 5.01; N, 17.89%.
Ethyl 8-(morpholino(2-(p-tolyl)hydrazono)methyl)-5-oxo-1,6-diphenyl-1,5-dihydropyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidine-3-carboxylate (10c), Brown solid, (70% yield), m.p. 244–245 °C; IR (KBr) υmax 1597 (C=N), 1658, 1712 (2C=O), 2979, 3061 (C–H), 3427 (NH) cm−1; 1H-NMR (DMSO-d6) δ 1.26 (t, J = 7.2 Hz, 3H, CH3CH2), 2.31–2.43 (m, 4H, 2CH2), 2.59 (s, 3H, CH3), 3.54–360 (m, 4H, 2CH2), 4.24 (q J = 7.2 Hz, 2H, CH3CH2), 6.97–7.97 (m, 15H, Ar–H), 10.52 (brs, 1H, NH); 13C-NMR (DMSO-d6): δ 14.56, 25.61 (2CH3), 27.43, 62.62, 69.48 (3CH2), 115.01, 115.05, 115.36, 120.40, 122.65, 122.87, 126.44, 129.11, 129.37, 129.41, 129.68, 129.75, 129.78, 129.85, 129.94, 130.01, 130.36, 130.41, 138.74, 143.12 (Ar–C and C=N), 162.52, 174.52 (2C=O); MS m/z (%) 628 (M+, 7), 511 (18), 472 (9), 397 (6), 209 (14), 161 (23), 127 (10), 119 (26), 104 (88), 84 (100), 77 (69). Anal. Calcd. for C35H32N8O4 (628.68): C, 66.87; H, 5.13; N, 17.82. Found: C, 67.21; H, 4.80; N, 17.55%.
Ethyl 8-(morpholino(2-(p-tolyl)hydrazono)methyl)-5-oxo-6-phenyl-1-(p-tolyl)-1,5-dihydropyrido [2,3-d][1,2,4]triazolo[4,3-a]pyrimidine-3-carboxylate (10d), Brown solid, (70% yield), m.p. 256–258 °C; IR (KBr) υmax 1591 (C=N)1663,1717 (2C=O), 2979, 3028 (C–H), 3436 (NH) cm−1; 1H-NMR (DMSO-d6) δ 1.21 (t, J = 7.2 Hz, 3H, CH3CH2), 2.24 (s, 3H, CH3), 2.37 (s, 3H, CH3), 2.48 (m, 4H, 2CH2), 4.25 (q, J = 7.2 Hz, 2H, CH3CH2), 3.40 (m, 4H, 2CH2), 6.95–7.98 (m, 14H, Ar–H), 11.18 (brs, 1H, NH); MS m/z (%) 642 (M+, 12), 602 (21), 511 (18), 472 (9), 397 (6), 209 (14), 161 (23), 127 (10), 119 (26), 104 (88), 84 (100), 77 (69). Anal. Calcd. for C36H34N8O4 (642.71): C, 67.28; H, 5.33; N, 17.43. Found: C, 67.59; H, 5.02; N, 17.09%.

3.1.7. Alternate Synthesis of 10a

A mixture of acetyltriazolopyrimidine 11 (0.269 g, l mmol) and chalcone 4d (0.349 g, 1 mmol) in ethanol (15 mL) was refluxed for 6 h, gave product identical in all respects (m.p., mixed m.p. and IR spectra) with compound 10a which obtained from reaction of 7d + 1a.

3.2. Anticancer Activity

The cytotoxic evaluation of the synthesized compounds was carried out at the Regional Center for Mycology and Biotechnology at Al-Azhar University, Cairo, Egypt according to the reported method [36,37].

3.3. Molecular Modeling

Docking studies were performed using the MOE 2014.09 software. Regularization and optimization for protein and ligand were performed. Each docked compound was assigned a score according to its fit in the ligand binding pocket (LBP) and its binding mode [38,39,40].

4. Conclusions

In our present work, we present an efficient synthesis of novel morpholinylchalcones, which have not been hitherto reported. These chalcones were used as building blocks for constructing a series of pyridopyrimidinethiones and pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones. The structures of the newly synthesized compounds were established on the basis of spectroscopic evidences and their synthesis by alternative methods. The in vitro growth inhibitory activity of the synthesized compounds against hepatocellular carcinoma (HepG-2) and human lung cancer (A-549) cell lines were investigated in comparison with cisplatin as reference drug using MTT assays and the results revealed promising activities of four compounds. Moreover, computational studies using MOE 2014.09 software supported the biological activity results.

Supplementary Materials

Supplementary materials are available online.

Acknowledgments

The authors extend their sincere appreciation to the Deanship of Scientific Research at the King Saud University for its funding this Prolific Research group (PRG-1437-29).

Author Contributions

S.M.G. and M.M.E. designed research; S.M.G., Z.A.M., R.A.M.F., S.S.A. and Y.N.M. performed research, analyzed the data, wrote the paper and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abbas, I.M.; Gomha, S.M.; Elaasser, M.M.; Bauomi, M.A. Synthesis and biological evaluation of new pyridines containing imidazole moiety as antimicrobial and anticancer agents. Turk. J. Chem. 2015, 39, 334–346. [Google Scholar] [CrossRef]
  2. Abdallah, M.A.; Riyadh, S.M.; Abbas, I.M.; Gomha, S.M. Synthesis and biological activities of 7-arylazo-7H-pyrazolo[5,1-c][1,2,4]triazolo-6(5H)-ones and 7-arylhydrazono-7H-[1,2,4]triazolo[3,4-b][1,3,4] thiadiazines. J. Chin. Chem. Soc. 2005, 52, 987–994. [Google Scholar] [CrossRef]
  3. Gomha, S.M.; Abdel-Aziz, H.M.; Khalil, K.D. Synthesis and SAR study of the novel thiadiazole-imidazole derivatives as new anti-cancer agents. Chem. Pharm. Bull. 2016, 64, 1356–1363. [Google Scholar] [CrossRef] [PubMed]
  4. Gomha, S.M.; Abdel-Aziz, H.A. Synthesis of new heterocycles derived from 3-(3-methyl-1H-indol-2-yl)-3-oxopropanenitrile as potent antifungal agents. Bull. Korean Chem. Soc. 2012, 33, 2985–2990. [Google Scholar] [CrossRef]
  5. Gomha, S.M.; Abdel-Aziz, H.A. Synthesis of new functionalized derivatives of indolo[2,3-e][1,2,4]-triazolo-[4,5-b]-1,2,4-triazine. J. Serb. Chem. Soc. 2013, 78, 1119–1125. [Google Scholar] [CrossRef]
  6. Gomha, S.M. A facile one-pot synthesis of 6,7,8,9-tetrahydrobenzo[4,5]thieno[2,3-d]-1,2,4-triazolo[4,5-a]pyrimidin-5-ones. Monatsh. Chem. 2009, 140, 213–220. [Google Scholar] [CrossRef]
  7. Gomha, S.M.; Riyadh, S.M. Synthesis of triazolo[4,3-b][1,2,4,5]tetrazines and triazolo[3,4-b][1,3,4]thiadiazines using chitosan as ecofriendly catalyst under microwave irradiation. Arkivoc 2009, 11, 58–68. [Google Scholar]
  8. Abbas, I.M.; Riyadh, S.M.; Abdallah, M.A.; Gomha, S.M. A novel route to tetracyclic fused tetrazines and thiadiazines. J. Heterocycl. Chem. 2006, 43, 935–942. [Google Scholar] [CrossRef]
  9. Gomha, S.M.; Badrey, G.; Abdalla, M.M.; Arafa, R.K. Novel Anti-HIV-1 NNRTIs Based on a Pyrazolo[4,3-d]isoxazole Backbone Scaffold: Design, synthesis and exploration of molecular basis of action. MedChemComm 2014, 5, 1685–1692. [Google Scholar] [CrossRef]
  10. Gomha, S.M.; Abdelrazek, F.M.; Abdulla, M.M. Synthesis of new functionalised derivatives of [1,2,4]triazolo[4,3-a]pyrimidine and pyrimido[2,1-b][1,3,5]thiadiazine as aromatase inhibitors. J. Chem. Res. 2015, 39, 425–429. [Google Scholar]
  11. Plech, T.; Wujec, M.; Siwek, A.; Osikowska, U.; Malm, A. Synthesis and antimicrobial activity of thiosemicarbazides, s-triazoles and their Mannich bases bearing 3-chlorophenyl moiety. Eur. J. Med. Chem. 2011, 46, 241–248. [Google Scholar] [CrossRef] [PubMed]
  12. Bayrak, H.; Demirbas, H.; Karaoglu, S.A.; Demirbas, N. Synthesis of some new 1,2,4-triazoles, their Mannich and Schiff bases and evaluation of their antimicrobial activities. Eur. J. Med. Chem. 2009, 44, 1057–1066. [Google Scholar] [CrossRef] [PubMed]
  13. Alberto, I.; Juan, R.; Marcela, R.; Carlos, E.; Jairo, Q.; Rodrigo, A.; Manuel, N.; Justo, C.; María, V.R.; Susana, A.Z.; et al. Synthesis, antifungal and antitumor activity of novel (Z)-5-hetarylmethylidene-1,3-thiazol-4-ones and (Z)-5-ethylidene-1,3-thiazol-4-ones. Molecules 2013, 18, 5482–5497. [Google Scholar]
  14. Brown, G.R.; Foubister, A.J.; Forster, G.; Stribling, D. (S)-3-[(Benzyloxy)methyl]morpholine Hydrochloride: A Nonstimulant Appetite Suppressant without Conventional Neurotransmitter Releasing Properties. J. Med. Chem. 1986, 29, 1288–1290. [Google Scholar] [CrossRef] [PubMed]
  15. Ihmaid, S.; Al-Rawi, J.; Bradley, C.; Angove, M.J.; Robertson, M.N.; Clark, R.L. Synthesis, structural elucidation, DNA-PK inhibition, homology modelling and anti-platelet activity of morpholino-substituted-1,3-naphth-oxazines. Bioorg. Med. Chem. 2011, 19, 3983–3994. [Google Scholar] [CrossRef] [PubMed]
  16. Sunny, M.; Shabana, I.K.; Diwan, S.R. 4-Aminoquinoline-triazine-based hybrids with improved in vitro antimalarial activity against CQ-sensitive and CQ-resistant strains of plasmodium falciparum. Chem. Biol. Drug Des. 2013, 81, 625–630. [Google Scholar]
  17. Cordeu, L.; Cubedo, E.; Bandres, E.; Rebollo, A.; Saenz, X.; Chozas, H.; Dominguez, M.V.; Echeverria, M.; Mendivil, B.; Sanmartin, C.; et al. Biological profile of new apoptotic agents based on 2,4-pyrido[2,3-d]pyrimidine derivatives. Bioorg. Med. Chem. 2007, 15, 1659–1669. [Google Scholar] [CrossRef] [PubMed]
  18. Font, M.; Gonzalez, A.; Palop, J.A.; Sanmartin, C. New insights into the structural requirements for pro-apoptotic agents based on 2,4-diaminoquinazoline, 2,4-diaminopyrido[2,3-d]pyrimidine and 2,4-diaminopyrimidine derivatives. Eur. J. Med. Chem. 2011, 46, 3887–3899. [Google Scholar] [CrossRef] [PubMed]
  19. Dorsey, J.F.; Jove, R.; Kraker, A.J.; Wu, J. The pyrido[2,3-d]pyrimidine derivative PD180970 inhibits p210Bcr-Abl tyrosine kinase and induces apoptosis of K562 leukemic cells. Cancer Res. 2000, 60, 3127–3131. [Google Scholar] [PubMed]
  20. Deyanov, A.B.; Niyazov, R.K.; Nazmetdivov, F.Y.; Syropyatov, B.Y.; Kolla, V.E.; Konshin, M.E. Synthesis and biological activity of amides and nitriles of 2-arylami-no-5-carboxy(carbethoxy)-6-methylnicotinic acids and 1-aryl-6-carbethoxy-7-methyl-4-oxo-1,4-dihydro- pyrido[2,3-d]pyrimidines. Pharm. Chem. J. 1991, 25, 248–250. [Google Scholar] [CrossRef]
  21. Grivsky, E.M.; Lee, S.; Sigel, C.W.; Duch, D.S.; Nichol, C.A. Synthesis and antitumor activity of 2,4-diamino-6-(2,5-dimethoxybenzyl)-5-methylpyrido[2,3-d]pyrimidine. J. Med. Chem. 1980, 23, 327–329. [Google Scholar] [CrossRef] [PubMed]
  22. Thompson, A.M.; Bridges, A.J.; Fry, D.W.; Kraker, A.J.; Denny, W.A. Tyrosine kinase inhibitors.7.7-ami-no-4-(phenylamino)-and7-amino-4-[(phenylmethyl)amino]pyrido[4,3-d]pyrimidines: A new class of inhibitors of the tyrosine kinase activity of the epidermal growth factor receptor. J. Med. Chem. 1995, 38, 3780–3788. [Google Scholar] [CrossRef] [PubMed]
  23. Donkor, I.O.; Klein, C.L.; Liang, L.; Zhu, N.; Bradley, E.; Clark, A.M. Synthesis and antimicrobial activity of some 6,7-annulated pyrido[2,3-d]pyrimidines. J. Pharm. Sci. 1995, 84, 661–664. [Google Scholar] [CrossRef] [PubMed]
  24. Pastor, A.; Alajarin, R.; Vaquero, J.J.; Alvarez-Builla, J.; Casa-Juana, M.F.; Sunkel, C.; Priego, J.G.; Fonseca, I.; Sanz-Aparicio, J. Synthesis and structure of new pyrido[2,3-d]pyrimidine derivatives with calcium channel antagonist activity. Tetrahedron 1994, 50, 8085–8098. [Google Scholar] [CrossRef]
  25. Agarwal, A.; Ashutosh, R.; Goyal, N.; Chauhan, P.M.S.; Gupta, S. Dihydropyrido[2,3-d]pyrimidines as a new class of antileishmanial agents. Bioorg. Med. Chem. 2005, 13, 6678–6684. [Google Scholar] [CrossRef] [PubMed]
  26. Monge, A.; Merino, V.M.; Sanmartin, C.; Fernandez, F.J.; Ochoa, M.C.; Berllver, C.; Artigas, P.; Alvarez, E.F. 2-Arylamino-4-oxo-3,4-dihydropyrido-[2,3-d]pyrimidines: Synthesis and diuretic activity. Eur. J. Med. Chem. 1989, 24, 24–29. [Google Scholar] [CrossRef]
  27. Gomha, S.M.; Ahmed, S.M.; Abdelhamid, A.O. Synthesis and cytotoxicity evaluation of some novel thiazoles, thiadiazoles, and pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-one incorporating triazole moiety. Molecules 2015, 20, 1357–1376. [Google Scholar] [CrossRef] [PubMed]
  28. Gomha, S.M.; Khalil, K.D. A convenient ultrasound-promoted synthesis and cytotoxic activity of some new thiazole derivatives bearing a coumarin nucleus. Molecules 2012, 17, 9335–9347. [Google Scholar] [CrossRef] [PubMed]
  29. Gomha, S.M.; Abbas, I.M.; Elaasser, M.M.; Mabrouk, B.K.A. Synthesis, molecular docking and pharmacological study of pyrimido-thiadiazinones and its bis-derivatives. Lett. Drug Des. Discov. 2017, 14, 434–443. [Google Scholar] [CrossRef]
  30. Gomha, S.M.; Eldebss, T.M.A.; Abdulla, M.M.; Mayhoub, A.S. Diphenylpyrroles: Novel p53 Activators. Eur. J. Med. Chem. 2014, 82, 472–479. [Google Scholar] [CrossRef] [PubMed]
  31. Gomha, S.M.; Dawood, K.M. Synthesis of novel indolizine, pyrrolo[1,2-a]quinoline, and 4,5-dihydrothiophene derivatives via nitrogen ylides and their antimicrobial evaluation. J. Chem. Res. 2014, 38, 515–519. [Google Scholar] [CrossRef]
  32. Gomha, S.M.; Eldebss, T.M.A.; Badrey, M.G.; Abdulla, M.M.; Mayhoub, A.S. Novel 4-heteroaryl-antipyrines as DPP-IV Inhibitors. Chem. Biol. Drug Des. 2015, 86, 1292–1303. [Google Scholar] [CrossRef] [PubMed]
  33. Gomha, S.M.; Edrees, M.M.; Altalbawy, F.M.A. Synthesis and characterization of some new bis-pyrazolyl-thiazoles incorporating the thiophene moiety as potent anti-tumor agents. Int. J. Mol. Sci. 2016, 17, 1499. [Google Scholar] [CrossRef] [PubMed]
  34. Hassaneen, H.M.; Abdelhamid, A.O.; Shawali, A.S.; Pagni, R. A Study of the effect of nitro group in the synthesis of pyrazoles and thiadiazolines from hydrazidoyl halides. Heterocycles 1982, 19, 319–326. [Google Scholar]
  35. Shawali, S.; Gomha, S.M. A New entry for short and regioselective synthesis of [1,2,4]triazolo[4,3-b][1,2,4]-triazin-7(1H)-one. Adv. Synth. Catal. 2000, 342, 599–604. [Google Scholar] [CrossRef]
  36. Gomha, S.M.; Riyadh, S.M.; Mahmmoud, E.A.; Elaasser, M.M. Synthesis and anticancer activities of thiazoles, 1,3-thiazines, and thiazolidine using chitosan-grafted-poly(vinylpyridine) as basic catalyst. Heterocycles 2015, 91, 1227–1243. [Google Scholar]
  37. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  38. Sharma, M.; Chauhan, P.M.S. Dihydrofolate reductase as a therapeutic target for infectious diseases: Opportunities and challenges. Future Med. Chem. 2012, 4, 1335–1365. [Google Scholar] [CrossRef] [PubMed]
  39. Daina, A.; Zoete, V. A boiled-egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem 2016, 11, 1117–1121. [Google Scholar] [CrossRef] [PubMed]
  40. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2001, 46, 3–25. [Google Scholar] [CrossRef]
Sample Availability: Samples of the compounds 4a,c,e,f, 5, 7ac, 8 and 10ad are available from the authors.
Scheme 1. Synthesis of morpholinylchalcones 4af.
Scheme 1. Synthesis of morpholinylchalcones 4af.
Molecules 22 01211 sch001
Scheme 2. Synthesis of pyridopyrimidinethiones 7ae.
Scheme 2. Synthesis of pyridopyrimidinethiones 7ae.
Molecules 22 01211 sch002
Scheme 3. Synthesis of pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones 10ad.
Scheme 3. Synthesis of pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-ones 10ad.
Molecules 22 01211 sch003
Figure 1. The most active compounds compared to cisplatin.
Figure 1. The most active compounds compared to cisplatin.
Molecules 22 01211 g001
Figure 2. Hhydrogen bonding between the compound 7b and the DHFR enzyme pocket amino acids.
Figure 2. Hhydrogen bonding between the compound 7b and the DHFR enzyme pocket amino acids.
Molecules 22 01211 g002
Figure 3. The interesting compound 7b fitted into the enzyme pocket by interacting with amino acids found in the pocket.
Figure 3. The interesting compound 7b fitted into the enzyme pocket by interacting with amino acids found in the pocket.
Molecules 22 01211 g003
Table 1. The in vitro inhibitory activity of tested compounds against tumor cell lines expressed as IC50 values (μg/mL) ± standard deviation from three replicates.
Table 1. The in vitro inhibitory activity of tested compounds against tumor cell lines expressed as IC50 values (μg/mL) ± standard deviation from three replicates.
Tested CompoundsTumor Cell LinesTested CompoundsTumor Cell Lines
A-549HepG2A-549HepG2
4a16.3 ± 1.3121.1 ± 0.917c6.79 ± 1.118.42 ± 1.15
4c24.0 ± 1.2120.0 ± 1.2382.78 ± 0.8629.9 ± 0.93
4e5.37 ± 0.9515.68 ± 1.1210a24.47 ± 1.2327.68 ± 1.31
4f7.38 ± 0.829.78 ± 0.7810b26.8 ± 0.7517.7 ± 0.73
510.3 ± 0.9112.4 ± 0.9810c15.2 ± 1.4214.9 ± 1.14
7a9.41 ± 0.7913.9 ± 0.7710d12.2 ± 0.888.72 ± 0.89
7b5.7 ± 0.913.54 ± 1.11Cisplatin0.95 ± 0.91.4 ± 1.1
Table 2. Bioactivity and ADME toxicity.
Table 2. Bioactivity and ADME toxicity.
Compound8
Molecular weight472.56 g/mol
Num. rotatable bonds5
Num. H-bond acceptors4
Num. H-bond donors3
TPSA130.49 Å2
GI absorptionHigh
BBB permeantNo
P-gp substrateYes
CYP1A2 inhibitorNo
Log Kp (skin permeation)−6.43 cm/s
LipinskiYes; 0 violation
PAINS0 alert
LeadlikenessNo; 2 violations: MW >350, XLOGP3 >3.5
Synthetic accessibility3.76
Topological polar surface area (TPSA), gastrointestinal absorption (GI absorption), blood brain barrier (BBB) permeant, P-glycoprotein substrate (P-gp substrate), cytochrome P50 1A2 inhibitor (CYP1A2 inhibitor), pan-assay interference structure (PAINS).

Share and Cite

MDPI and ACS Style

Muhammad, Z.A.; Edrees, M.M.; Faty, R.A.M.; Gomha, S.M.; Alterary, S.S.; Mabkhot, Y.N. Synthesis, Antitumor Evaluation and Molecular Docking of New Morpholine Based Heterocycles. Molecules 2017, 22, 1211. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22071211

AMA Style

Muhammad ZA, Edrees MM, Faty RAM, Gomha SM, Alterary SS, Mabkhot YN. Synthesis, Antitumor Evaluation and Molecular Docking of New Morpholine Based Heterocycles. Molecules. 2017; 22(7):1211. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22071211

Chicago/Turabian Style

Muhammad, Zeinab A., Mastoura M. Edrees, Rasha A. M. Faty, Sobhi M. Gomha, Seham S. Alterary, and Yahia N. Mabkhot. 2017. "Synthesis, Antitumor Evaluation and Molecular Docking of New Morpholine Based Heterocycles" Molecules 22, no. 7: 1211. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22071211

Article Metrics

Back to TopTop