Research Article
Design, Synthesis and Biological Evaluation of Novel C5-Modified Pyrimidine Acyclic Nucleosides
Nikolaos Kollatos, Christos Mitsos, Roula Karetsou, Styliani Danai Antonaki, Aggeliki Panagiotopoulou, Niki Tzioumaki, Stella Manta, Dominique Schols and Dimitri Komiotis*
Corresponding Author: Dr. Dimitri Komiotis, University of Thessaly, Department of Biochemistry and Biotechnology, Laboratory of Bioorganic Chemistry, Viopolis, 41500 Larissa, Greece
Received: August 30, 2019; Revised: January 09, 2020; Accepted: September 09, 2019
Citation: Kollatos N, Mitsos C, Karetsou R, Antonaki SD, Panagiotopoulou A, et al. (2020) Design, Synthesis and Biological Evaluation of Novel C5-Modified Pyrimidine Acyclic Nucleosides. J Pharm Drug Res, 3(1): 257-267.
Copyrights: ©2020 Kollatos N, Mitsos C, Karetsou R, Antonaki SD, Panagiotopoulou A, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Share :
  • 842

    Views & Citations
  • 10

    Likes & Shares

We report the synthesis of novel C5-arylalkynyl acyclic pyrimidine analogues with α, β-unsaturated carbonyl structures, at the acyclic portion, using a double Wittig reaction. The newly synthesized compounds were tested for their cytostatic activity against a broad panel of cancer cells. The antiviral assays showed that (E)-ethyl 4-(((E)-4-ethoxy-1-((2-fluorophenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13a) had a noticeable activity against TK- VZV strain (EC50 20 μΜ, MCC 100 μΜ).

 

Keywords: C5-pyrimidine acyclic nucleosides, Cytotoxic antiviral activity

 

Abbreviations: 13C NMR: Carbon-13 Nuclear Magnetic Resonance Spectroscopy; 1H NMR: Proton Magnetic Resonance Spectroscopy; AcOH: Acetic Acid; CD3OD: Deuterated Methanol; CDCl3: Deuterated Chloroform; CH2Cl2: Dichloromethane; CH3CN: Acetonitrile; CuI: Copper(I) Iodide; DMF: Dimethyl Formamide; DMSO-d6: Deuterated Dimethyl Sulfoxide; Et3N: Triethylamine; EtOAc: Ethyl Acetate; HMDS: Hexamethyldisilazane; MeOH: Methanol; MW: Microwave; Na2SO4: Sodium Sulfate; NaHCO3: Sodium Bicarbonate; Pd(PPh3)4: Tetrakis(triphenylphosphine)palladium(0); TLC: Thin Layer Chromatography; TMS: Tetramethylsilane; TMSOTf: Trimethylsilyl Trifluoromethanesulfonate; UV-Vis: Ultraviolet-Visible Spectroscopy

INTRODUCTION

The synthesis of acyclic nucleosides such as Acyclovir (ACV) [1] and the discovery of its important antiviral properties have opened a new era in antiviral therapy, the study of acyclic nucleosides with excellent biological properties against a broad band of RNA viruses. Acyclovir is highly active against Herpes Simplex Virus (HSV), where it prevents virus replication by interacting with the DNA polymerase after metabolizing to its active triphosphate structure by disrupting DNA synthesis of the virus.

Among the numerous modified acyclic nucleosides with interesting biological properties [2-4], C5 modified acyclic nucleosides of uracil have been investigated as anticancer and antiviral agents [5-8]. Specifically Carmofur is used to treat colorectal cancer [9] and inhibits acidic ceramidase (ASAH1) [10] which plays an important role in the occurrence of metastatic breast cancer [11], while 1-[4-hydroxy-3-(hydroxymethyl)-1-butyl]-5-(1-azido-2-chloroethyl)uracil proved to be effective in vitro against Duck hepatitis B virus (EC50 0.31-1.55 µM) and Cytomegalovirus (EC50 3.1 µM) [12]. Furthermore, C5-arylalkynyl nucleosides of uracil showed antiviral activity, against Coxsackie virus B4, respiratory syncytial virus and yellow fever virus [13-16] and cytotoxic activity against murine leukemia, human T-lymphocyte, cervix carcinoma and hepatocellular carcinoma cells [17].

Considering the biological importance of acyclic nucleosides and the significant antiviral activity demonstrated by C5 modified uracil 3΄-deoxy and 3΄-deoxy-3΄-C-methyl ones [16], we have decided to synthesize novel acyclic uracil nucleosides with α, β-unsaturated carbonyl structures incorporated into the acyclic fragment. The α, β-unsaturated carbonyl structures have been shown to be vital for the biological activity exhibited by both plant products and synthetic molecules [18-23], whereas introduction of a double bond into the acyclic portion of modified nucleosides appears to increase their activity [24,25]. Thus, we performed the synthesis and biological evaluation of C5-arylalkynyl uracil acyclic nucleosides bearing α, β-unsaturated carbonyl structures at the acyclic portion of the novel synthesized analogues.

EXPERIMENTAL SECTION

General methods

Melting points were recorded in a Mel-Temp apparatus and are uncorrected. Thin Layer Chromatography (TLC) was performed on Merck pre-coated 60F254 plates. Reactions were monitored by TLC on silica gel, with detection by UV light (254 nm) or by charring with sulfuric acid. Flash column chromatography was performed using silica gel (240-400 mesh, Merck). 1H and 13C NMR spectra were obtained at ambient temperature using a Bruker 300 spectrometer at 300 and 75.5 MHz, respectively using chloroform-d (CDCl3), dimethylsulfoxide-d6 (DMSO-d6) or methanol-d4 (CD3OD) with internal tetramethylsilane (TMS). Chemical shifts (δ) are given in ppm measured downfield from TMS and spin-spin coupling constants in Hz. Mass spectra were obtained on a ThermoQuestFinnigan AQA Mass Spectrometer (electrospray ionization). All microwave irradiation experiments were carried out in a dedicated CEM-Explorer and CEM Discover monomode microwave apparatus, operating at a frequency of 2.45 GHz, with continuous irradiation power, from 0 to 300 W with utilization of the standard absorbance level of 300 W maximum powers. The reactions were carried out in 10 mL glass tubes, sealed with a Teflon septum and placed in the microwave cavity. Initially, microwave irradiation of requisite Watts was used, and the temperature was ramped from room temperature to the desired temperature. Once this was reached the reaction mixture was held at this temperature for the required time. The reaction mixture was continuously stirred during the reaction. The temperature was measured with an IR sensor on the outer surface of the process vial. After the irradiation period, gas jet cooling rapidly cooled the reaction vessel to ambient temperature. Dichloromethane was distilled from phosphorous pentoxide and stored over 4 Å molecular sieves. Acetonitrile and toluene were distilled from calcium hydride and stored over 3 Å molecular sieves. Diethylether (Et2O) was freshly distilled under nitrogen from sodium/benzophenone before use. Pyridine was stored over potassium hydroxide pellets, N,N-Dimethylformamide (DMF) was stored over 3 Å molecular sieves. All reactions sensitive to oxygen or moisture were carried out under an Argon atmosphere.

Anti-proliferative assays

Compounds 4-9, 12-14, were evaluated for their cytostatic activity against the human cells: pancreatic adenocarcinoma (Capan-1), chronic myeloid leukemia (Hap-1), colorectal carcinoma (HCT-116), lung carcinoma (NCI-H460), acute lymphoblastic leukemia (DND-41), acute myeloid leukemia (HL-60), chronic myeloid leukemia (K-562) and non-Hodgkin lymphoma (Z-138). All assays were performed in 96-well microtiter plates. To each well (5-7.5) × 104 tumor cells were added,along withand varying concentrations of the test compounds ranging from 250, 50, 10, 2, 0.4 to 0.08 µM. The tumor cells were then allowed to proliferate at 37°C in a humidified CO2-controlled atmosphere. To obtain optimal growth curves, 2 days of the: pancreatic adenocarcinoma (Capan-1), chronic myeloid leukemia (Hap-1), colorectal carcinoma (HCT-116), lung carcinoma (NCI-H460), acute lymphoblastic leukemia (DND-41), acute myeloid leukemia (HL-60) and 3 days for the chronic myeloid leukemia (K-562) and non-Hodgkin lymphoma (Z-138), were required. At the end of the incubation period, the cells were counted in a Coulter counter. The IC50 (50% inhibitory concentration) was defined as the concentration of the compound that inhibited cell proliferation by 50%.

Antiviral activity assays

The antiviral tests were based on inhibition of the virus-induced cytopathicity in Human Embryonic Lung (HEL) (Varicella-Zoster Virus (VZV)) cell cultures. Confluent cell cultures in microtiter 96-well plates were inoculated with 100 cell culture inhibitory dose-50 (CCID50) of virus (1 CCID50 being the virus dose to infect 50% of the cell cultures). After a 1 h virus adsorption period, the residual virus was removed, and the cell cultures were incubated in the presence of varying concentrations (200, 40, 8, … µM) of the test compounds. Viral cytopathicity was recorded as soon as it reached completion in the control virus-infected cell cultures, which were not treated with the test compounds.

Synthesis of C5 alkynyl 1-(1-((1,3-dihydropropan-2-yl)oxy)-2-hydroxyethyl)-5-iodouracil (7, 9)1-(5΄-O-Trityl-ribofuranozyl)-5-iodouracil (2)

A solution of trityl chloride (2.8 g, 9.90 mmol) in anhydrous dichloromethane (19 mL) was added drop wise to a solution of 1 (3.0 g, 8.26 mmol) in anhydrous pyridine (35 mL) at 0°C. Following the addition, the mixture was allowed to warm slowly to room temperature and set aside for 12 h. Methanol (7 mL) and ethyl acetate (50 mL) were added and the mixture was washed successively with saturated aqueous NaHCO3 (200 mL) and H2O (100 mL). The organic phase was dried over MgSO4 and evaporated under reduced pressure. The resulting residue was crystallized from chloroform to give 6 (2.3 g; 82%). m.p. 162-163°C; [α]D22=-8 (c 0.10, CHCl3); Rf=0.27 (EtOAc/Hexane, 7:3); λmax 383 nm (ε 21460); 1H-NMR (300 MHz, CD3OD) δ 8.14 (s, 1H, H-6), 7.49-7.24 (m, 15H, Tr), 5.90 (d, 1H, J1΄,2΄=4.8 Hz, H-1΄), 5.35-4.31 (m, 2H, H-2΄, H-4΄), 4.10 (t, 1H, J=2.7, J=1.2 Hz, H-3΄), 3.41 (dd, 1H, J4΄-5a΄=3.2 Hz, J5a΄-5b΄=10.8 Hz, H-5b΄), 4.55 (dd, 1H, J4΄-5b΄=2.1 Hz, H-5a΄); 13C NMR (75.5 MHz, CDCl3) δ 161.80, 150.83, 144.19, 143.65, 129.71, 128.34, 126.28, 97.67, 94.83, 86.93, 73.22, 70.11, 68.42, 63.92; Anal. Calcd. for C9H11IN2O6: C, 59.41; H, 4.11; N, 4.57% found: C, 59.61; H, 4.41; N, 4.97%; Mass (M+H)+: 612.08.

1-(2-Hydroxy-1-(1-hydroxy-3-(trityloxy)propan-2-yl)oxy)ethyl)-5-iodouracil (3)

Compound 2 (1.1 g, 1.8 mmol) was added to a stirred solution of NaIO4 (425 mg, 1.98 mmol) in H2O (19 mL) and MeOH (19 mL) leading to immediate precipitation of NaIO3. After 1 h at room temperature, any residual periodate was destroyed with a drop of ethylene glycol. The reaction mixture was stirred for 1 h at room temperature with NaBH4 (500 mg, 18.5 mmol), neutralized with aqueous NaHCO3 and then extracted with ethyl acetate (EtOAc) (4 × 300 mL). The organic layer was washed with NaHSO4, dried over anhydrous Na2SO4, evaporated to dryness and purified by column chromatography with EtOAc/Hexane (7:3) to give compound 3 (650 mg, 86%) as a syrup. [α]D22+13 (c 0.10, MeOH); Rf=0.35 (EtOAc/Hexane, 7:3); λmax 305 nm (ε21400); 1HNMR (300 MHz, CDCl3) δ 9.97 (brs, 1H, NH), 7.84 (s, 1H, H-6), 7.39-7.25 (m, 15H,Tr), 5.99 (t, 1H, J1΄,2a΄=5.2 Hz, J1΄,2b΄=5.0 Hz, H-1΄), 3.87-3.72 (m, 7H, H-2΄, H-3΄, H-4΄, OH, OH), 3.26 (dd, 1H, J4΄-5a΄=3.7 Hz, J5a΄-5b΄=10.8 Hz, H-5a΄), 3.16 (dd, 1H, J4΄-5b΄=6.0 Hz, H-5b΄); 13C NMR (75.5 MHz, CDCl3) δ 161.80, 150.83, 144.19, 143.65, 129.71, 128.34, 126.28, 96.25, 94.83, 83.63, 68.42, 63.92, 61.81, 61.13; Anal. Calcd for C28H27IN2O6: C, 54.73; H, 4.43; N, 4.56. Found: C, 54.33; H, 4.83; N, 4.96; ESI-MS (m/z): 614.09 (M+H+).

1-(1-((1,3-Dihydroxypropan-2-yl)oxy)-2-hydroxyethyl)-5-iodouracil (4)

To a solution containing 3 (1 g, 1.62 mmol) is added 1: 1 mixture of formic acid (HCOOH)/diethyl ether (Et2O) (29 ml) and reflux for 30 min. The mixture is then neutralized with solid sodium bicarbonate (NaHCO3) and extracted sequentially with sodium chloride (NaCl) and water (H2O), dried over magnesium sulphate (MgSO4), evaporated to dryness and purified by column chromatography with CH2Cl2/MeOH (9:1) to give compound 4 (554 mg, 92%) as a syrup. [α]D22+9 (c 0.14, MeOH); Rf=0.25 (CH2Cl2/MeOH 9:1); λmax 286 nm (ε18592); 1HNMR (300 MHz, CD3OD) δ 8.12 (s, 1H, H-6), 7.55-7.11 (m, 4H, Bz), 8.99 (t, 1H, J=4.9 Hz, J=4.9 Hz N-CH-C), 3.80-3.51 (m, 7H, 3x -CH2OH, C-CH-C); 13C NMR (75.5 MHz, CD3OD) δ 161.30, 150.83, 144.19, 96.25, 94.83, 80.63, 68.22, 60.92, 58.32; Anal. Calcd for C9H13IN2O6: C, 29.05; H, 3.52; N, 7.53. Found: C, 29.45; H, 3.92; N, 7.93; ESI-MS (m/z): 372.98 (M+H+).

2-(2-Acetoxy-1-(5-iodouracil)ethoxy)propan-1,3-diyldiacetylo (5)

To a solution of 4 (84 mg, 0.23 mmol) added dry pyridine (2 ml) and acetic anhydride (1 ml). The reaction was carried out at room temperature for 1 h, then was quenched with MeOH at 0°C and was concentrated in vacuum. The residue was diluted with ethyl acetate (EtOAc), washed with saturated NaHSO4, NaHCO3 and H2O. The organic extract was dried over anhydrous Na2SO4, filtered and evaporated to dryness to give compound 5 (80 mg, 95%) as a white crystal. M.P. 215-217°C; [α]D22+12 (c 0.25, CHCl3); Rf=0.22 (EtOAc/Hexane, 2:8); λmax 286 nm (ε 16459); 1HNMR (300 MHz, CDCl3) δ 8.47 (brs, 1H, NH), 7.85 (s, 1H, H-6), 6.13 (t, 1H, J=5.1 Hz, J=5.3 Hz N-CH-C), 4.46-3.95 (m, 7H, 3x-CH2OAc, C-CH-C), 2.13, 2.09, 2.07 (3s, 9H, 3x-OAc);13CNMR (75.5 MHz, CDCl3) δ 170.48, 170.44. 169.98, 159.46, 150.24, 144.14, 81.34, 75.76, 68.85, 63.36, 63.05, 62.50, 21.06, 20.67, 20.49; Anal. Calcd for C15H19IN2O9: C, 36.16; H, 3.84, N 5.62. Found: C, 36.56; H, 4.04, N 5.92; ESI-MS (m/z): 416.99 (M+H+).

General procedure for the preparation of the C5-arylalkynyl uracil acyclic nucleosides-6,8

Mixtures of the appropriate alkynes (0.72 mmol), Pd (PPh3)4 (28 mg, 0.02 mmol), CuI (5.3 mg, 0.02 mmol), triethylamine (116 μl, 0.34 mmol) and 2-(2-Acetoxy-1-(5-iodouracil)ethoxy)propan-1,3-diyldiacetylo (5) (100 mg, 0.24 mmol) in 1.0 mL of anhydrous DMF, were irradiated in a microwave apparatus (200 W maximum power) for 5 min at 50°С. The reaction mixture was concentrated under reduced pressure and the crude residue was purified by flash chromatography on silica gel. The purified material was dried in vacuo to afford the corresponding derivatives 6, 8 in 78-81% yields.

2-(2-Acetoxy-1-(5-((2-fluorophenyl)ethynyl)-uracil)ethoxy)propan-1,3-diyl diacetyl (6a)

75 mg, 78% as white foam; [α]D22=+12 (c 0.15, CHCl3); Rf=0.24 (EtOAc/Hexane 2:8); λmax 286 nm (ε17254); 1HNMR (300 MHz, CDCl3) δ 8.70 (brs, 1H, NH), 7.80 (s, 1H, H-6), 7.52 (t, 1H, J=7.5 Hz, J=7.1 Hz, Bz), 7.35 (dd, 1H, J=7.2 Hz, J=13.8 Hz, Bz), 7.14-7.07 (m, 2H, Bz), 6.20 (t, 1H, J=5.3 Hz, J=5.6 Hz N-CH-C), 4.46-3.95 (m, 7H, 3x-CH2OAc, C-CH-C), 2.13, 2.09, 2.08 (3s, 9H, 3xOAc); 13CNMR (75.5 MHz, CDCl3) δ 170.60, 170.48. 170.00, 160.49, 149.61, 142.08, 133.56, 130.74, 130.62, 124.09, 124.03, 115.70, 115.37, 100.69, 81.30, 75.55, 63.39, 63.03, 62.44, 21.06, 20.67, 20.50; Anal. Calcd. for C23H23FN2O9: C, 56.33; H, 4.73; N, 5.71%; Found: C, 56.73; H, 4.33; N, 5.91%; ESI-MS (m/z): Mass (M+H)+: 491.14.

2-(2-Acetoxy-1-(5-((2-chlorophenyl)ethynyl)-uracil)ethoxy)propan-1,3-diyldiacetyl (6b)

80 mg, 79% as white foam; [α]D22=+15 (c 0.19, CHCl3); Rf=0.26 (EtOAc/Hexane 2:8); λmax 286 nm (ε 17845); 1HNMR (300 MHz, CDCl3) δ 8.56 (brs, 1H, NH), 7.80 (s, 1H, H-6), 7.55 (t, 1H, J=7.5 Hz, J=7.1 Hz, Bz), 7.41 (dd, 1H, J=7.2 Hz, J=13.8 Hz, Bz), 7.30-7.22 (m, 2H, Bz), 6.19 (t, 1H, J=5.2 Hz, J=5.5 Hz N-CH-C), 4.47-3.93 (m, 7H, 3x-CH2OAc, C-CH-C), 2.14, 2.09 (2s, 9H, 3xOAc); 13CNMR (75.5 MHz, CDCl3) δ 170.53, 170.48. 170.00, 160.33, 149.55, 142.09, 135.76, 133.45, 129.90, 129.33, 126.54, 122.19, 100.73, 91.03, 84.66, 81.36, 75.70, 63.38, 63.05, 62.47, 20.78, 20.67, 20.51; Anal. Calcd. for C23H23ClN2O9: C, 54.50; H, 4.57; N, 5.53%; Found: C, 54.10; H, 4.27; N, 5.93%; ESI-MS (m/z): Mass (M+H)+: 507.11.

2-(2-Acetoxy-1-(5-((1,4-dimethylphenyl)ethynyl)-uracily)ethoxy)propan-1,3-diyldiacetyl (6c)

80 mg, 79% as white solid; M.P. 251-253°C; [α]D22=+12 (c 0.22, CHCl3); Rf=0.30 (EtOAc/Hexane 2:8); λmax 286 nm (ε 14523); 1HNMR (300 MHz, CDCl3) δ 8.28 (brs, 1H, NH), 7.71 (s, 1H, H-6), 7.22 (t, 1H, J=4.5 Hz, J=5.9 Hz, Bz), 7.11 (d, 1H, J=4.5 Hz, Bz ), 7.06 (d, 1H, J=7.2 Hz, Bz), 6.19 (t, 1H, J=5.6 Hz, J=5.6 Hz N-CH-C), 4.47-3.96 (m, 7H, 3x-CH2OAc, C-CH-C), 2.44, 2.29 (2s, 6H, 2x CH3)2.14, 2.08, 2.07 (3s, 9H, 3xOAc); 13CNMR (75.5 MHz, CDCl3) δ 170.53, 170.48. 170.00, 160.33, 149.55, 142.09, 135.76, 133.45, 129.90, 129.33, 126.54, 122.19, 100.73, 91.03, 84.66, 81.36, 75.70, 63.38, 63.05, 62.47, 20.74, 20.70, 20.62, 20.54, 20.21; Anal. Calcd. for C25H28N2O9: C, 59.59; H, 5.64; N, 5.50%; Found: C, 59.59; H, 5.24; N, 5.20%; ESI-MS (m/z): Mass (M+H)+: 501.18.

2-(2-Acetoxy-1-(5-((6-methoxynapthlene)ethynyl-uracil)ethoxy)propan-1,3-diyldiacetyl (8)

83 mg, 81% as white solid; M.P. 286-289°C; [α]D22=-2 (c 0.16, CHCl3); Rf=0.30 (EtOAc/Hexane 2:8); λmax 286 nm (ε 16895); 1HNMR (300 MHz, CDCl3) δ 8.43 (brs, 1H, NH), 7.96 (s, 1H, H-6), 7.77 (s, 1H, napthalene), 7.70 (t, 2H, J=7.7 Hz, J=8.2 Hz, napthalene), 7.49 (d, 1H, J=8.7 Hz, napthalene), 7.16 (d, 1H, J=8.7 Hz, napthalene), 7.11 (s, 1H, napthalene), 6.20 (t, 1H, J=5.4 Hz, J=5.6 Hz N-CH-C), 4.48-3.97 (m, 7H, 3x-CH2OAc, C-CH-C), 3.93 (s, 3H, OCH3), 2.14, 2.11, 2.09 (3s, 9H, 3xOAc); 13CNMR (75.5 MHz, CDCl3) δ 170.53, 170.48. 170.00, 160.33, 158.92, 149.55, 142.09, 135.76, 133.45, 129.90, 129.33, 126.54, 122.19, 119.89, 117.50, 105.96, 100.73, 91.03, 84.66, 81.36, 75.70, 63.38, 63.05, 62.47, 55.41, 20.74, 20.70, 20.62; Anal. Calcd. for C28H28N2O10: C, 60.87; H, 5.11; N, 5.07%; Found: C, 60.47; H, 5.31; N, 5.27%; ESI-MS (m/z): Mass (M+H)+: 553.17.

General procedure for the preparation of the unprotected C5-arylalkynyl uracil acyclic nucleosides-7,9

The protected nucleosides 6, 8 (0.12 mmol), were treated with methanolic ammonia (saturated at 0°C, 6.7 mL). The solution was stirred overnight at room temperature and then evaporated under reduced pressure. The residue obtained was purified by flash column chromatography to afford the unprotected derivatives 7, 9 in 65-85% yields, as white solids.

1-(1-((1,3-Dihydroxypropan-2-yl)oxy)-2-hydroxyethyl)-5-[(2-fluorophenyl) ethynyl] uracil (7a)

80 mg, 85% as white solid; M.P. 189-192°C; [α]D22=+8 (c 0.24, MeOH); Rf=0.19 (CH2Cl2/MeOH 9:1); λmax 286 nm (ε 13564); 1HNMR (300 MHz, CD3OD) δ 8.12 (s, 1H, H-6), 7.55-7.11 (m, 4H, Bz), 8.99 (t, 1H, J=4.9 Hz, J=4.9 Hz N-CH-C), 3.80-3.51 (m, 7H, 3x-CH2OH, C-CH-C); 13CNMR (75.5 MHz, CD3OD) δ 162.79, 150.64, 144.70, 133.25, 130.25, 130.12, 123.99, 123.93, 115.24, 114.90, 98.71, 85.62, 84.36, 81.11, 62.21, 61.13, 60.69; Anal. Calcd. for C17H17FN2O6: C, 56.04; H, 4.70; N, 7.69%; Found: C, 56.44; H, 4.30; N, 8.09%; ESI-MS (m/z): Mass (M+H)+: 365.11.

1-(1-((1,3-Dihydroxypropan-2-yl)oxy)-2-hydroxyethyl)-5-[(2-chlorophenyl) ethynyl] uracil (7b)

84 mg, 85% as white solid; M.P. 195-197°C; [α]D22=+9 (c 0.23, MeOH); Rf=0.22 (CH2Cl2/MeOH 9:1); λmax 286 nm (ε 15732); 1HNMR (300 MHz, CD3OD) δ 8.12 (s, 1H, H-6), 7.59-7.27 (m, 4H, Bz), 5.99 (t, 1H, J1΄-2a΄=4.9 Hz, J1΄-2b΄=4.9 Hz N-CH-C), 3.79-3.53 (m, 7H, 3x-CH2OH, C-CH-C); 13CNMR (75.5 MHz, CD3OD) δ 164.16, 152.08, 146.15, 136.60, 134.53, 130.85, 127.90, 124.21, 100.19, 90.49, 87.23, 85.88, 82.58, 63.70, 62.81, 62.20; Anal. Calcd. for C17H17ClN2O6: C, 53.62; H, 4.50; N, 7.36%; Found: C, 54.02; H, 4.30; N, 7.76%; ESI-MS (m/z): Mass (M+H)+: 381.08.

1-(1-((1,3-Dihydroxypropan-2-yl)oxy)-2-hydroxyethyl)-5-[(1,4-dimethylphenyl) ethynyl] uracil (7c)

88 mg, 85% as white solid; M.P. 220-221°C; [α]D22=+12 (c 0.18, MeOH); Rf=0.28 (CH2Cl2/MeOH 9:1); λmax 282 nm (ε 18563); 1HNMR (300 MHz, CD3OD) δ 8.07 (s, 1H, H-6), 7.25 (s, 1H, ArH), 7.10 (d, 1H, J=7.6, Bz), 7.04 (d, 1H, Bz), 5.99 (t, 1H, J=4.9 Hz, J=4.9 Hz N-CH-C), 3.80-3.52 (m, 7H, 3x-CH2OH, C-CH-C), 2.40, 2.26 (2s, 6H, 2xCH3); 13CNMR (75.5 MHz, CD3OD) δ 164.16, 152.08, 146.15, 136.60, 134.53, 130.85, 127.90, 124.21, 100.19, 90.49, 87.23, 85.88, 82.58, 63.70, 62.81, 62.20, 19.37, 18.94.; Anal. Calcd. for C19H22N2O6: C, 60.95; H, 5.92; N, 7.48%; Found: C, 60.55; H, 5.62; N, 7.08%; ESI-MS (m/z): Mass (M+H)+: 375.15.

1-(1-((1,3-Dihydroxypropan-2-yl)oxy)-2-hydroxyethyl)-5-[(6-methoxynapthalene) ethynyl] uracil (9)

88 mg, 65% as white solid; M.P. 162-163°C; [α]D22=+15 (c 0.17, MeOH); Rf=0.15 (CH2Cl2/MeOH 9:1); λmax 286 nm (ε 19654); 1HNMR (300 MHz, CD3OD) δ 8.12 (s, 1H, H-6), 7.94-7.12 (m, 6H, naphthalene), 5.99 (t, 1H, J=5.15 Hz, J=5.13 Hz N-CH-C), 3.90 (s, 3H, OCH3), 3.82-3.53 (m, 7H, 3x-CH2OH, C-CH-C); 13CNMR (75.5 MHz, CD3OD) δ 164.57, 160.07, 152.13, 145.48, 135.85, 132.14, 130. 38, 129.95, 129.69, 128.09, 120.60, 119.17, 106.90, 100.77, 94.59, 85.79, 82.55, 81.41, 63.69, 62.82, 62.16, 55.89; Anal. Calcd. for C22H22N2O7: C, 61.97; H, 5.20; N, 6.57%; Found: C, 62.27; H, 5.60; N, 6.97%; ESI-MS (m/z): Mass (M+H)+: 427.14.

Synthesis of (E)-ethyl 4-(((E)-4-ethoxy-1-(5-iodo-uracil)-4-oxobout-2-en-1-yl)oxy)-5-(trityloxy)pent-2-enoate (11)

A solution of sodium periodate (0.23 g, 1.09 mmol) in water (5 mL) was slowly added to a cooled stirred solution of the protected nucleoside 2 (0.612 g, 1 mmol) in methanol (10 mL). The reaction mixture was stirred at room temperature for 1 h and then filtered to remove the salts. The solution was diluted with ethyl acetate (15 mL), washed with a saturated solution of NaCl (2 × 15 mL) and dried over MgSO4. The solvent was removed by evaporation under reduced pressure to yield a white powder of the corresponding dialdehyde derivative. The crude dialdehyde was dissolved in freshly distilled tetrahydrofuran (5 mL) and (ethoxycarbonylmethylene) triphenylphosphorane (0.871 g, 2.5 mmol) added and the mixture was heated at 40°C for 1 h under nitrogen. The reaction mixture washed twice with a saturated solution of ammonium chloride (2 × 30 mL), dried over MgSO4, evaporated to a yellow syrup and purified by column chromatography (ethyl acetate/hexane, 2:8) to afford the protected nucleoside 11 (0.435 g, 53%) as a foam. [α]D22 +22 (c 0.25, CHCl3); Rf=0.52 (EtOAc/Hexane, 2:8); λmax 286 nm (ε 24538); 1HNMR (300 MHz, CDCl3) δ 8.62 (s, 1H, ΝΗ), 7.61 (s, 1H, H-6), 7.41-7.25 (m, 15H, trityl), 6.72 (dd, 1H, J=3.5 Hz, J=15.6 Hz, Η-3), 6.69 (dd, 1H, J=6.7 Hz, J=15.8 Hz, Η-2), 6.35 (dd, 1H, J=6.7 Hz, J=15.8 Hz, Η-2΄), 6.29 (t, 1H, J=3.4 Hz, Η-1), 6.11 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 4.28 (dd, 2H, J=6.1 Hz, J=13.3 Hz, CH2), 4.21 (dd, 2H, J=7.1 Hz, J=14.2 Hz, CH2), 4.03-3.99 (m, 1H, H-4), 3.38 (dd, 1H, J=7.4 Hz, J=11.0 Hz, -Η-5), 3.23 (dd, 1H, J=3.4 Hz, Η-5΄), 1.35 (t, 3H, J=7.2 Hz, CH3), 1.29 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.32, 164.96, 159.38, 150.07, 144.07, 143.36, 141.21, 139.76, 128.64, 128.47, 128.05, 127.96, 127.90, 127.30, 127.23, 127.14, 126.10, 125.27, 87.38, 79.79, 77.24, 70.25, 65.56, 61.26, 60.90, 14.15; Anal. Calcd for C36H35ΙN2O8: C, 57.61; H, 4.70, N 3.73. Found: C, 58.01; H, 5.10, N 4.13; ESI-MS (m/z): 751.14 (M+H+).

Synthesis of (E)-ethyl 4-(((E)-4-ethoxy-1-(5-iodo-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydroxypent-2-enoate (12)

Compound 12 was synthesized from 11 by a similar procedure to that described for the preparation of 4. The crude product was purified by flash column chromatography (EtOAc/Hexane, 3:7) to give analogue 12 (88 mg, 95%) as a white solid. M.P. 182-184°C; [a]D22=23 (c 0.31, CHCl3); Rf=0.35 (EtOAc/ Hexane, 3:7); λmax 286 nm (ε 24167)); 1HNMR (300 MHz, CDCl3) δ 8.80 (s, 1H, ΝΗ), 7.71 (s, 1H, H-6), 6.79 (dd, 1H, J=6.7 Hz, J=15.3 Hz, Η-2 ), 6.75 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-3), 6.38 (t, 1H, J=3.3 Hz,Η-1), 6.35 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 6.11 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-2΄), 4.32 (s, 1H, OH), 4.28-4.18 (m, 4H, 2xCH2, H-4), 3.74 (dd, 1H, J=2.9 Hz, J=11.9 Hz, -Η-5), 3.67 (dd, 1H, J=7.5 Hz, Η-5΄), 1.34 (t, 3H, J=7.2 Hz, CH3), 1.31 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.48, 165.01, 159.96, 150.61, 144.68, 144.04, 139.63, 126.23, 125.15, 80.59, 79.07, 69.76, 64.25, 61.29, 60.98, 14.14; Anal. Calcd.for C17H21ΙN2O8: C, 40.17; H, 4.16, N 5.51. Found: C, 40.57; H, 4.36, N 5.81; ESI-MS (m/z): 509.03 (M+H+).

General procedure for the preparation of the C5-arylalkynyl (E)-ethyl 4-(((E)-4-ethoxy-1-(5-iodo-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydroxypent-2-enoate (13, 14)

Mixtures of the appropriate alkynes (0.72 mmol), Pd (PPh3)4 (28 mg, 0.02 mmol), CuI (5.3 mg, 0.02 mmol), triethylamine (116 μl, 0.34 mmol) and (E)-ethyl 4-(((E)-4-ethoxy-1-(5-iodo-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydroxypent-2-enoate (12) (100 mg, 0.20 mmol) in 1.0 mL of anhydrous DMF, were irradiated in a microwave apparatus (200 W maximum power) for 5 min at 50°С. The reaction mixture was concentrated under reduced pressure and the crude residue was purified by flash chromatography on silica gel. The purified material was dried in vacuo to afford the corresponding derivatives 13, 14 in 69-82% yields.

(E)-ethyl 4-(((E)-4-ethoxy-1-((2-fluorophenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13a)

75 mg, 74% as white foam; [α]D22=+21 (c 0.14, CHCl3); Rf=0.36 (EtOAc/Hexane3:7); λmax 286 nm (ε 19547); 1HNMR (300 MHz, CDCl3) δ 8.83 (s, 1H, ΝΗ), 7.66 (s, 1H, H-6), 7.51 (dd, 1H, J=8.2 Hz, J=7.4 Hz, Bz), 7.32 (dd, 1H, J=8.2 Hz, J=7.4 Hz, Bz), 7.01 (dd, 2H, J=6.7 Hz, J=15.3 Hz, Bz), 6.81 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-3), 6.77(dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-2), 6.46 (t, 1H, J=3.3 Hz, Η-1), 6.39 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 6.13 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-2΄), 4.36 (s, 1H, OH), 4.28-4.20 (m, 5H, 2xCH2, H-4), 3.74 (dd, 1H, J=2.9 Hz, J=11.9 Hz , Η-5), 3.67 (dd, 1H, J=7.5 Hz, Η-5΄), 1.33 (t, 3H, J=7.2 Hz, CH3), 1.30 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.48, 165.01, 160.07, 159.96, 150.61, 144.68, 144.04, 139.63, 133.11, 126.23, 126.08, 125.15, 123.09, 115.01, 109.32, 93.25, 86.43, 80.59, 79.07, 69.76, 64.25, 61.29, 60.98, 14.14; Anal. Calcd. for C25H25FN2O8: C, 60.00; H, 5.03; N, 5.60%; Found: C, 60.20; H, 5.13; N, 5.90%; ESI-MS (m/z): Mass (M+H)+: 501.16.

(E)-ethyl 4-(((E)-4-ethoxy-1-((2-chlorophenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13b)

75 mg, 78% as white foam; [α]D22=+19 (c 0.15, CHCl3); Rf=0.42 (EtOAc/ Hexane 3:7); λmax 286 nm (ε 21547); 1HNMR (300 MHz, CDCl3) δ8.70 (s, 1H, ΝΗ), 7.66 (s, 1H, H-6), 7.56 (dd, 1H, J=8.2 Hz, J=7.4 Hz, Bz), 7.40 (dd, 1H, J=8.2 Hz, J=7.4 Hz, Bz), 7.21 (dd, 2H, J=6.7 Hz, J=15.3 Hz, Bz), 6.80 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-3), 6.78 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-2), 6.45 (t, 1H, J=3.3 Hz, Η-1), 6.39 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 6.14 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-2΄), 4.34 (s, 1H, OH), 4.28-4.21 (m, 5H, 2xCH2, H-4), 3.74 (dd, 1H, J=2.9 Hz, J=11.9 Hz, H-5), 3.67 (dd, 1H, J=7.5 Hz,H-5΄), 1.34 (t, 3H, J=7.2 Hz, CH3), 1.31 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.48, 165.01, 160.07, 159.96, 150.61, 144.68, 144.04, 139.63, 133.11, 126.23, 126.08, 125.15, 123.09, 115.01, 109.32, 93.25, 86.43, 80.59, 79.07, 69.76, 64.25, 61.29, 60.98, 14.14; Anal. Calcd. for C25H25ClN2O8: C, 58.09; H, 4.87; N, 5.42%; Found: C, 58.49; H, 4.47; N, 5.82%; ESI-MS (m/z): Mass (M+H)+: 517.13.

(E)-ethyl4-(((E)-4-ethoxy-1-((1,4-dimethylphenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13c)

79 mg, 82% as white foam; [α]D22=+24 (c 0.18, CHCl3); Rf=0.42 (EtOAc/Hexane 3:7); λmax 286 nm (ε 21574); 1HNMR (300 MHz, CDCl3) δ 8.83 (s, 1H, ΝΗ), 7.57 (s, 1H, H-6), 7.27 (s, 1H, Bz), 7.07 (d, 1H, J=7.7 Hz, Bz), 7.02 (d, 1H, J=7.7 Hz, Bz), 6.80 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-3), 6.77 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-2), 6.45 (t, 1H, J=3.3 Hz, Η-1), 6.39 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 6.14 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-2΄), 4.35 (s, 1H, OH), 4.26-4.20 (m, 5H, 2xCH2, H-4), 3.73 (dd, 1H, J=2.9 Hz, J=11.9 Hz Η-5), 3.67 (dd, 1H, J=7.5 Hz,Η-5΄), 2.42, 2.27 (2s, 6H, 2xCH3), 1.32 (t, 3H, J=7.2 Hz, CH3), 1.29 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.48, 165.01, 160.07, 159.96, 150.61, 144.68, 144.04, 139.63, 133.11, 126.23, 126.08, 125.15, 123.09, 115.01, 109.32, 93.25, 86.43, 80.59, 79.07, 69.76, 64.25, 61.29, 60.98, 20.75, 20.22, 14.34, 14.14; Anal. Calcd. for C27H30N2O8: C, 63.52; H, 5.92; N, 5.49%; Found: C, 63.92; H, 5.62; N, 5.89%; ESI-MS (m/z): Mass (M+H)+: 511.20.

(E)-ethyl4-(((E)-4-ethoxy-1-((6-methoxynaphtalene)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (14)

68 mg, 69% as white solid; M.P. 192-193°C; [α]D22=+24 (c 0.10, CHCl3); Rf=0.28 (EtOAc/Hexane 3:7); λmax 286 nm (ε 26413); 1HNMR (300 MHz, CDCl3) δ 8.83 (s, 1H, ΝΗ), 7.57 (s, 1H, H-6), 7.27 (s, 1H, Bz), 7.07 (d, 1H, J=7.7 Hz, Bz), 7.02 (d, 1H, J=7.7 Hz, Bz), 6.80 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-3), 6.77 (dd, 1H, J=3.9 Hz, J=15.8 Hz, Η-2), 6.45 (t, 1H, J=3.3 Hz, Η-1), 6.39 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-3΄), 6.14 (dd, 1H, J=1.2 Hz, J=15.8 Hz, Η-2΄), 4.00 (s, 3H, OCH3), 4.35 (s, 1H, OH), 4.26-4.20 (m, 5H, 2xCH2, H-4), 3.73 (dd, 1H, J=2.9 Hz, J=11.9 Hz , Η-5), 3.67 (dd, 1H, J=7.5 Hz, Η-5΄), 1.32 (t, 3H, J=7.2 Hz, CH3), 1.29 (t, 3H, J=7.2 Hz, CH3); 13CNMR (75.5 MHz, CDCl3) δ 165.48, 165.01, 160.07, 159.96, 150.61, 144.68, 144.04, 139.63, 133.11, 129.64, 129.16, 126.23, 126.08, 125.15, 123.09, 115.01, 109.32, 105. 93, 93.25, 86.43, 80.59, 79.07, 69.76, 64.25, 61.29, 60.98, 55.82, 14.28; Anal. Calcd. for C30H30N2O9: C, 64.05; H, 5.38; N, 4.98%; Found: C, 64.25; H, 5.58; N, 4.78%; ESI-MS (m/z): Mass (M+H)+: 563.20.

RESULTS AND DISCUSSION

Chemistry

Our first synthetic efforts focused on the preparation of the C5-substituted uracil 1-(1-((1,3-dihydroxyprapan-2-yl)oxy)-2-hydroxyethyl) (Figure 1). Uridine 1 was treated in pyridine with an excess of trityl chloride (TrCl), in the presence of a catalytic amount of 4,4-dimethylaminopyridine (DMAP) to give the corresponding trityl derivative 2 in 82% yield [26]. The oxidative cleavage of the cis-diol in the 2΄,3΄-position of compound 2 followed by borohydride reduction of the resulting aldehyde (one pot), furnished the corresponding acyclic nucleoside 3. Deprotection of the 5΄-O-trityl 3 by treatment with formic acid (HCOOH) in diethylether (Et2O) gave the acyclic nucleoside of iodouracil 4 [27]. The next step of the synthesis involves the acetylation of the hydroxyl groups of the nucleoside 4 using acetic anhydride in the presence of pyridine led to acetylated derivative 5. In order to extract more detailed structure-activity relationships, diverse alkyne substituents R were selected, which included a phenyl ring substituted with halogens (6a, R=2-fluoro, 6b, R=2-chloro) or methyl groups (6c, R=2,5-dimethyl) and a polycyclic aromatic hydrocarbon substituted with a methoxyl group (8, R=6-methoxynaphthalene). In a typical experiment, the acetylated acyclic nucleoside of 5-iodouracil (5) was mixed with N,N-dimethylformamide (DMF), the appropriate alkyne, triethylamine (base), copper(I) iodide (CuI) (co-catalyst) and tetrakis(triphenylphosphine) palladium (0) (Pd(PPh3)4) (catalyst) and were irradiated at 50°C for 5 min. After removing all the volatile materials in vacuo, the solid obtained was purified by flash chromatography to provide the C5-alkynyl acyclic nucleosides 6, 8, which upon treatment with saturated methanolic ammonia afforded the unprotected derivatives 7 and 9, in good yields (65-85%).

The next objective was to synthesize C5 modified acyclic nucleosides of uracil, wherein the acyclic moiety would have been introduced into α, β-unsaturated carbonyl structures that have been shown to confer particular biological activity on the molecules carrying them. The synthesis of the C5-substituted uracil α, β-unsaturated carbonyl acyclic nucleosides is outlined in Figure 2. The oxidative cleavage of the cis-diol in the 2΄,3΄-position of compound 2 was achieved with sodium periodate in a mixture of methanol/water. The dialdehyde were unstable, hence after isolation they were directly subjected to a double Wittig olefination, using (ethoxycarbonylmethylene) triphenylphosphorane in tetrahydrofurane (THF) at 40°C for 1 h afforded compound 11in 58% yield. It was of particular note, that under such reaction conditions no diastereoisomeric by products observed, by 1ΗNMR and COSY spectroscopy, indicating that complete chiral integrity was retained at carbon atoms C1 and C4 of 11, the iodouracil base retains the β configuration and finally the protons of the two double bonds are in trans position as we observe large coupling constants (15.8 Hz) [28-30]. Deprotection of the 5΄-O-trityl 11 by treatment with formic acid (HCOOH) in diethylether (Et2O) gave the acyclic nucleoside of iodouracil 12. Using the same one-pot Sonogashira protocol as previously discussed, the novel aryl alkynyl acyclic nucleosides 13 and 14 were obtained, in satisfying yields (69-82%).

Biological evaluation

Compounds 4-9, 12-14, were evaluated for their cytostatic activity against the human cells: pancreatic adenocarcinoma (Capan-1), chronic myeloid leukemia (Hap-1), colorectal carcinoma (HCT-116), lung carcinoma (NCI-H460), acute lymphoblastic leukemia (DND-41), acute myeloid leukemia (HL-60), chronic myeloid leukemia (K-562) and non-Hodgkin lymphoma (Z-138) and their antiviral activity varicella-zoster virus (VZV) in human embryonic lung (HEL) cell cultures.The results of cytotoxic and antiviral activity are shown in Tables 1 and 2, respectively. Unfortunately none of the tested compound showed any significant cytostatic activity at a broad panel of cancer cell lines. The antiviral assays showed that (E)-ethyl 4-(((E)-4-ethoxy-1-((2-fluorophenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13a) had a noticeable activity against TK- VZV strain (EC50 20 μΜ, MCC 100 μΜ).

CONCLUSION

In the present study, we report the synthesis of novel C5-arylalkynyl pyrimidine acyclic nucleosides, by developing highly efficient synthetic routes. Nucleoside (E)-ethyl 4-(((E)-4-ethoxy-1-((2-fluorophenyl)ethynyl-uracil)-4-oxobout-2-en-1-yl)oxy)-5-hydropent-2-enoate (13a) had a noticeable activity against TK- VZV strain (EC50 20 μΜ, MCC 100 μΜ) (Table 2).

CONFLICT OF INTEREST

The authors confirm that this article content has no conflict of interest.

1.       Elion GB, Rideout JL, De Miranda P, Collins P, Bauer DJ (1975) Biological activities of some purine arabinosides. Ann N Y Acad Sci 255: 468-480.

2.       Ferrero M, Gotor V (2000) Biocatalytic selective modifications of conventional nucleosides, carbocyclic nucleosides and C-Nucleosides. Chem Rev 100: 4319-4348.

3.        Galmarini CM, Mackey JR, Dumontet C (2002) Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol. 3: 415-424.

4.       Raić-Malić S, Meščić A (2015) Recent trends in 1, 2, 3-triazolo-nucleosides as promising anti-infective and anticancer agents. Curr Med Chem 22: 1462-1499.

5.       Pałasz A, Ciez D (2015) In search of uracil derivatives as bioactive agents. Uracils and fused uracils: Synthesis, biological activity and applications. Eur J Med Chem 97: 582-611.

6.       Sari O, Roy V, Balzarini J, Snoeck R, Andrei G, et al. (2012) Synthesis and antiviral evaluation of C5-substituted-(1,3-diyne)-2′-deoxyuridines. Eur J Med Chem 53: 220-228.

7.       Montagu A, Roy V, Balzarini J, Snoeck R, Andrei G, et al. (2011) Synthesis of new C5-(1-substituted-1,2,3-triazol-4 or 5-yl)-2′-deoxyuridines and their antiviral evaluation. Eur J Med Chem 46: 778-786.

8.       Lee YS, Park SM, Kim HM, Park SK, Lee K, et al, (2009) C5-Modified nucleosides exhibiting anticancer activity. Bioorg Med Chem Lett 19: 4688-4691.

9.       Realini N, Solorzano C, Pagliuca C, Pizzirani D, Armirotti A, et al. (2013) Discovery of highly potent acid ceramidase inhibitors with in vitro tumor chemosensitizing activity. Sci Rep 3: 1035-1041.

10.    Pizzirani D, Bach A, Realini N, Armirotti A, Mengatto L, et al. (2015) Benzoxazolone carboxamides: Potent and systemically active inhibitors of intracellular acid ceramidase. Angew Chem Int Ed 54: 485-489.

11.    Lucki NC, Sewer MB (2012) Genistein stimulates MCF-7 breast cancer cell growth by inducing acid ceramidase (ASAH1) gene expression. J Biol Chem 286: 19399.

12.    Flowers M, Fabrias G, Delgado A, Casas J, Abad JL, et al. (2012) C6-Ceramide and targeted inhibition of acid ceramidase induce synergistic decreases in breast cancer cell growth. Breast Cancer Res Treat 133: 447-458.

13.     Kumar R, Sharma N, Nath M, Saffran HA, Tyrrell DL (2001) Synthesis and antiviral activity of novel acyclic nucleoside analogues of 5-(1-Azido-2-haloethyl) uracils. J Med Chem 44: 4225-4229.

14.    Dimopoulou A, Manta S, Kiritsis C, Gkaragkouni DN, Papasotiriou I, et al. (2013) Rapid microwave-enhanced synthesis of C5-alkynyl pyranonucleosides as novel cytotoxic antitumor agents. Bioorg Med Chem Lett 23: 1330-1333.

15.    Dimopoulou A, Kollatos N, Manta S, Panagiotopoulou A, Karastergiou A, et al.(2017) Facile microwave-assisted synthesis of various C5-modified pyrimidine pyranonucleosides as potential cytotoxic antitumor agents. Curr Microwave Chem 4: 324-338.

16.    Kollatos N, Mitsos C, Manta S, Tzioumaki N, Giannakas C, et al. (2019) Design, synthesis and biological evaluation of novel C5-modified pyrimidine ribofuranonucleosides as potential antitumor or/and antiviral agents. Med Chem. 

17.    Gazivoda T, Raic´-Malic´ S, Krištafor V, Makuc D, Plavec J, et al. (2008) Synthesis, cytostatic and anti-HIV evaluations of the new unsaturated acyclic C-5 pyrimidine nucleoside analogues. Bioorg Med Chem 16: 5624.

18.    Tzioumaki N, Tsoukala E, Manta S, Agelis G, Balzarini J, et al. (2009) Synthesis, antiviral and cytostatic evaluation of unsaturated exomethylene and keto D-lyxopyranonucleoside analogues. Arch Pharm Chem Life Sci 342: 353-360.

19.    Tzioumaki N, Tsoukala E, Manta S, Kiritsis C, Balzarini J, et al. (2011) Efficient synthesis of exomethylene- and keto-exomethylene-D-glucopyranosyl nucleoside analogs as potential cytotoxic agents. Carbohydr Res 346: 328-333.

20.    Agelis G, Tzioumaki N, Tselios T, Botic´ T, Cencic A, et al. (2008) Synthesis and molecular modeling of unsaturated exomethylene pyranonucleoside analogues with antitumor and antiviral activities. Eur J  Med Chem 43: 1366-1385.

21.    Rüngeler P, Castro V, Mora G, Gören N, Vichnewski W, et al. (1999) Inhibition of transcription factor NF-κB by sesquiterpene lactones: A proposed molecular mechanism of action. Biorg Med Chem 7: 2343-2352.

22.    Bazzaro M, Anchoori RK, Mudiam MKR, Issaenko O, Kumar S, et al. (2011) α, β-unsaturated carbonyl system of chalcone-based derivatives is responsible for broad inhibition of proteasomal activity and preferential killing of human papilloma virus (HPV) positive cervical cancer cells. J Med Chem 54: 449-456.

23.    Arshad L, Jantan I, Haque A (2017) Immunosuppressive effects of natural α, β-unsaturated carbonyl-based compounds and their analogs and derivatives, on immune cells: A review. Front Pharmacol 8: 1-21.

24.    Wu Y, Hong JH (2005) Synthesis and anti-HIV activity of novel phenyl branched cyclopropyl nucleosides Il. Farmaco 60: 739-744.

25.    Haines DR, Tseng CKH, Marquez VE (1987) Synthesis and biological activity of unsaturated carboacyclic purine nucleoside analogs. J Med Chem 30: 943-947.

26.    Matsuda A, Okajima H, Masuda A, Kakefuda A, Yoshimura Y, et al. (1992) Nucleosides and nucleotides. 104. Radical and palladium-catalyzed deoxygenation of the allylic alcohol systems in the sugar moiety of pyrimidine nucleosides. Nucleosides Nucleotides 104: 197-226.

27.     Komiotis D, Agelis G, Manta S, Tzioumaki N, Tsoukala E (2006) A facile, one‐step conversion of 6‐O‐trityl and 6‐O‐TBDMS monosaccharides into the corresponding formate esters. J Carbohydr Chem 25: 441-450.

28.    Aksel A, Bothner BY (1965) Geminal and vicinal proton-proton coupling constants in organic compounds. Adv Magn Reson 1: 195-316.

29.     Robins MJ, Barr PJ, (1983) Nucleic acid related compounds. 39. Efficient conversion of 5-iodo to 5-alkynyl and derived 5-substituted uracil bases and nucleosides. J Org Chem 48: 1854-1862.

30.    Tong GL, Lee WW, Goodman L (1967) Synthesis of some 3΄-O-Methyl purine ribonucleosides. J Org Chem 32: 1984-1986.