Next Article in Journal
Five New Meroterpenoids from the Fruiting Bodies of the Basidiomycete Clitocybe clavipes with Cytotoxic Activity
Previous Article in Journal
The Synthesis and Properties of Liquid Crystalline Polyurethanes, Chemically Modified by Polyhedral Oligomericsilsesquioxanes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Isoxazolidine and γ-Lactam Analogues of Homonucleosides

1
Bioorganic Chemistry Laboratory, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
2
Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(22), 4014; https://doi.org/10.3390/molecules24224014
Submission received: 2 October 2019 / Revised: 23 October 2019 / Accepted: 5 November 2019 / Published: 6 November 2019
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Homonucleoside analogues cis-16 and trans-17 having a (5-methoxycarbonyl)isoxazolidine framework were synthesized via the 1,3-dipolar cycloaddition of nucleobase-derived nitrones with methyl acrylate. Hydrogenolysis of the isoxazolidines containing thymine, dihydrouracil, theophylline and adenine moieties efficiently led to the formation of the respective γ-lactam analogues. γ-Lactam analogues having 5-bromouracil and 5-chlorouracil fragments were synthesized by treatment of uracil-containing γ-lactams with NBS and NCS. Isoxazolidine and γ-lactam analogues of homonucleosides obtained herein were evaluated for activity against a broad range of DNA and RNA viruses. None of the compounds that were tested exhibited antiviral or cytotoxic activity at concentrations up to 100 µM. The cytostatic activities of all compounds toward nine cancerous cell lines was tested. γ-Lactams trans-15e (Cl-Ura) and cis-15h (Theo) appeared the most active toward pancreatic adenocarcinoma cells (Capan-1), showing IC50 values 21.5 and 18.2 µM, respectively. Isoxazolidine cis-15e (Cl-Ura) inhibited the proliferation of colorectal carcinoma (HCT-116).

Graphical Abstract

1. Introduction

Nucleoside analogues belong to a group of important antiviral and antitumor drugs [1,2,3,4]. However, resistance to drugs and their toxicity are considered major factors limiting the effectiveness of therapies. Structural modifications of available drugs from a class of nucleoside analogues, including sugar and/or nucleobase residues, resulted in the discovery of a variety of therapeutically-useful antiviral [5] (e.g., azidothymidine (AZT) [6], carbovir [7], lobucavir [8], dioxolane T [9] and lamivudine [10]) and anticancer [11,12] (e.g., cladribine [13], gemcitabine [14], azacitidine [15] and cytarabine [14,16], clofarabine [17]) agents (Figure 1).
On the other hand, the introduction of a methylene bridge between a nucleobase and the sugar partly led to the formation of homonucleosides, which are known for their resistance to hydrolytic or enzymatic cleavage, as well as for the rotational freedom when compared with the natural nucleosides. Furthermore, it was shown that homonucleosides are substrates for cellular kinases and are able to pair with other nucleosides by Watson–Crick interactions without appreciable modifications of the structure of DNA (or RNA) molecules [18]. 1′-Homonucleosides containing adenine 1 or guanine 2 were found to be active against herpes simplex virus (minimum inhibitory concentration (MIC) = 5–20 µg/mL) and vaccinia virus [19,20,21], while compound 3 (IC50 = 25.2 µg/mL) showed activity against the influenza type virus AH1N1 (Figure 2) [22]. Homonucleoside analogue of 2′-deoxyuridine 4 has been described as a selective inhibitor of viral uracil-DNA glycosylase (UDG), while exerting only small effect on the human enzymes [23].
Incorporation of the isoxazolidine ring into a nucleoside framework as a sugar part replacer was first proposed by Tronchet and co-workers [24,25] with the intention to study antiviral [20,26,27,28,29,30,31], antibacterial [32] and antifungal [33] activities of the compounds. Other research groups then intensively explored this field further. Thus, a compound 5 [34] and phosphonylated derivatives 6 [35] and 7 [36,37,38] inhibited the reverse transcriptase activity of avian Moloney virus (AMV) and human immunodeficiency virus (HIV) at a level comparable with that of tenofovir (1 nM) and 10-fold higher than that of AZT (10 nM). Moreover, their cytotoxicity was significantly lower (50% cytotoxic concentration (CC50) > 500 µM) in comparison with that of AZT (CC50 = 12.14 µM) (Figure 3). In order to obtain compounds with a sufficient stability for hydrolytic or enzymatic cleavage, analogues of 1′-homonucleosides 811 (Figure 3) were also synthesized [39,40,41,42,43].
In the isoxazolidine nucleoside/nucleotide analogues described so far, a nucleobase is attached to C5; therefore, the 1,3-dipolar cycloaddition of nitrones and N-vinyl or N-allyl nucleobases has been found a convenient method for their preparation [44]. Recently, we designed isoxazolidine analogues of 1′-homonucleos(t)ides 12 (Figure 3), in which nucleobases were attached to C3 to underscore the fundamental difference to the already-described ones [45,46]. In the synthesis of homonucleosides 12, the 1,3-dipolar cycloaddition was also employed, but in that case, nucleobase-derived nitrones and selected alkenes were applied [45,46]. Since no significant activity of compounds 12 toward the viruses tested was noticed, we wondered whether the replacement of an isoxazolidine moiety with a γ-lactam ring would lead to an improvement of antiviral properties of newly-designed compounds 15. The idea of incorporation of the γ-lactam moiety has been supported by the presence of this subunit in many natural products (Figure 4), which showed antiviral [47,48], cytotoxic [49,50,51,52,53], anti-inflammatory [53,54,55] and antimicrobial [56,57] properties, among others. For this reason the assumption that homonucleoside analogues 15 (Scheme 1) having the γ-lactam moiety could possess interesting biological properties as antiviral or cytotoxic is justified [58].

2. Results and Discussion

2.1. Chemistry

The synthetic strategy for γ-lactam homonucleosides 15 relies on the 1,3-dipolar cycloaddition of nucleobase-derived nitrones 13 with methyl acrylate, followed by the hydrogenolysis of the N–O bond in an isoxazolidine scaffold, which was accompanied by the intramolecular cyclization to transform cycloadducts 14 into compounds 15 (Scheme 1).
Nucleobase-derived nitrones 13 were synthesized from N-(2-oxoethyl)nucleobases and N-methylhydroxylamine, as described previously [45,46]. The 1,3-dipolar cycloadditions of the nucleobase-derived nitrones 13 to methyl acrylate were carried out at 60 °C in methanol for 24 h (Scheme 2, Table 1) and afforded mixtures of diastereoisomeric isoxazolidines cis-14 and trans-14. The cis/trans ratios of the isoxazolidines were calculated from the 1H-NMR spectra of the crude reaction mixtures by the comparison of diagnostic resonances of the H2C-4 protons in the isoxazolidine ring, as well as the signals of the respective protons of nucleobase moieties. In some cases, differences in resonances of the CH3-N protons for diastereoisomeric isoxazolidines cis-14 and trans-14 were also diagnostic. The crude reaction mixtures of the diastereoisomers were subjected to column chromatography; however, only pure isomeric isoxazolidines cis-14a and trans-14a were isolated (Table 1, entry a). In the other cases the respective fractions enriched in the isomers cis-14b14i or trans-14b14i were collected. The subsequent separation of diastereoisomeric mixture of isoxazolidines cis-14b14h or trans-14b14h on an HPLC column allowed us to obtain pure isomers cis-14b14h and trans-14b14h (Table 1, entry b–h), and the amounts, however small, were sufficient to perform a full characterization of the newly-synthesized compounds and their further biological screening. Unfortunately, the attempts at separating diastereoisomeric isoxazolidines cis-14i and trans-14i appeared fruitless, even with HPLC.
The relative configurations of isoxazolidines cis-14a and trans-14a have already been established [45]. As we have recently described, (5-methoxycarbonyl)isoxazolidine cis-14a was reduced with sodium borohydride in ethanol to the known (5-hydroxymethyl)isoxazolidine cis-16 [45] (Scheme 3). During this transformation, the structure of the isoxazolidine skeleton remains unchanged, and therefore, a relative configuration of the isoxazolidine cis-14a was unambiguously established. Consequently, the isomeric (5-methoxycarbonyl)isoxazolidine was identified as trans-14a. Since similar spectral patterns for the respective isoxazolidine protons (H3, H4ɑ, H4β and H5) were observed in series of cis-14b14i and trans-14b14i when compared to those observed for cycloadducts cis-14a and trans-14a, their configurations were analogously assigned.
A detailed conformational analysis for isoxazolidines cis-14a14i and trans-14a14i was performed based on HCCH vicinal coupling constants extracted from the 1H-NMR spectra. The vicinal couplings for cis-14a14f and cis-14h14i (J(H3-H4ɑ) = 7.6–8.4 Hz, J(H3-H4β) = 2.0–3.3 Hz, J(H4ɑ-H5) = 9.7–9.9 Hz, J(H4β-H5) = 5.0–5.3 Hz) suggest the 2E conformation of the isoxazolidine ring in which the sterically bulky nucleobase-CH2 units are located pseudoequatorially (Figure 5, structure 17). The coupling constant values extracted for trans-14a14f and trans-14h14i (J(H3-H4ɑ) = 7.4–7.8 Hz, J(H3-H4β) = 2.0–3.4 Hz, J(H4ɑ-H5) = 7.4–7.5 Hz, J(H4β-H5) = 8.7–9.0 Hz) bring to mind the 2E conformation of the isoxazolidine ring in which the nucleobase-CH2 substituents again adopt pseudoequatorial positions (Figure 5, structure 18). Surprisingly, the preferred conformations for dihydrouacil-containing isoxazolidines cis-14g and trans-14g could not be assigned, and sets of couplings indicate the existence of conformational equilibrium of isoxazolidine ring.
For the either cis or trans isoxazolidines containing uracil or substituted uracil residues 14a14f, we also noticed that diastereotopic hydrogen atoms in CH2–base units which resonate at a lower field (4.05–3.89 ppm) showed significantly smaller vicinal coupling constants to H–C3 (3.3–3.9 Hz) than those associated with higher field signals (3.59–3.32 ppm) when values of 9.1–9.7 Hz were observed. One may conclude that high-field hydrogens are antiperiplanar to H–C3, while low-field ones are oriented gauche, as depicted by the Newman projection 19 (Figure 5). Although real reasons for the restricted rotation around C3–CH2Base bond remain obscure to us at the moment, they may simply reflect repulsive interactions of sterically bulky CH2–base groups with Me–N and Hβ–C4–Hα fragments.
Finally, attempts at transformation of isoxazolidine cycloadducts cis-14/trans-14 into γ-lactam derivatives trans-15/cis-15 were made, following the procedure applied previously for transformation of cis-14a into trans-15g [45]. It was expected that since hydrogenation of the N–O bond in the isoxazolidine scaffold releases the amino group, the subsequent intramolecular cyclization to γ-lactams trans-15/cis-15 should occur spontaneously (Scheme 4). Consequently, the isoxazolidine cis-14 would provide trans-15, while isoxazolidine trans-14 should be a substrate for the synthesis of cis-configured γ-lactam cis-15. With that in mind, we concluded that not only can pure diastereoisomers cis-14 and trans-14 be used for the synthesis of trans-15/cis-15, but so can mixtures of the respective isomers cis-14/trans-14, although significantly enriched in one diastereoisomer to maintain control on the stereochemistry of the products formed.
As we recently reported [45], the hydrogenation of uracil-containing isoxazolidine homonucleoside cis-14a (B = Ura) proceeded smoothly and the intramolecular cyclization of intermediary γ-aminocarboxylic ester to a γ-lactam was accompanied by hydrogenation of the uracil residue to produce the lactam trans-15g (B = diH-Ura) in a good yield (80%) instead of the expected compound trans-15a (B = Ura) (Table 2, entry a). Similarly, cis-15g (B = diH-Ura) was obtained from trans-14a (B = Ura) (85%).
On the other hand, when a 80:20 mixture of thymine-containing isoxazolidines cis-14b and trans-14b (B = Thy) was subjected to hydrogenation, the respective 80:20 mixture of γ-lactam trans-15b and cis-15b (B = Thy) was obtained. From this mixture pure diastereoisomers were isolated after column chromatography followed by HPLC (Table 2, entry b).
During the hydrogenation of 5-halogenated uracil derivatives cis-14/trans-14 (B = X-Ura), we faced the same problem as in case of uracil derivatives. Moreover, halogen atoms were removed from nucleobase skeletons. For example, hydrogenation of a 60:40 mixture of cis-14d and trans-14d isoxazolidines (B = Br-Ura) led to the formation of an inseparable mixture of trans-15a, cis-15a and trans-15g and cis-15g (53:35:8:2) (Table 2, entry d). Analogously, a mixture of trans-15a, cis-15a, trans-15g and cis-15g (4:2:74:20) was obtained from the 73:27 mixture of isoxazolidines cis-14e and trans-14e (B = Cl-Ura) (Table 2, entry e). Similarly, hydrogenation of a cis-14c (B = F-Ura) gave dihydrouracil-containing γ-lactam trans-15g (B = diH-Ura) (Table 2, entry c).
Furthermore, the respective mixtures of isoxazolidines cis-14g/trans-14g, cis-14h/trans-14h and cis-14i/trans-14i were successfully hydrogenated to provide γ-lactams containing dihydrouracil (trans-15g/cis-15g), theophylline (trans-15h/cis-15h) and adenine (trans-15i/cis-15i) as nucleobases (Table 2, entry g–i).
To supplement a library of designed γ-lactam homonucleosides trans-15/cis-15 with analogues having bromine (trans-15d/cis-15d) and chlorine (trans-15e/cis-15e) atoms, the halogenation of uracil derivatives was proposed [59,60,61]. Treatment of the above-mentioned (Table 1, entry d), inseparable mixture of γ-lactams trans-15a, cis-15a, trans-15g and trans-15g (53:35:8:2) with N-bromosuccinimide (NBS) [59] led to the formation of the mixture of γ-lactams trans-15d, cis-15d, together with unreacted trans-15g and cis-15g (50:37:9:4) (Scheme 5). The purification of this mixture on a silica gel column followed by HPLC allowed us to isolate trans-15d and cis-15d in 30% and 9.3% yields, respectively. Similarly, the reaction of an analogous mixture of trans-15a, cis-15a, trans-15g and cis-15g with N-chlorosuccinimide (NCS) [60] gave the respective mixture of compounds trans-15e, cis-15e, trans-15g and cis-15g (55:34:8:3), from which trans-15e and cis-15e were obtained in 6% and 4.2% yields, respectively (Scheme 5).

2.2. Antiviral and Cytostatic Evaluation

2.1.1. Antiviral Activity

All isoxazolidines 14 and γ-lactams 15 obtained were evaluated for their activities against a wide variety of DNA and RNA viruses using the following cell-based assays: (a) human embryonic lung (HEL) cell cultures: herpes simplex virus-1 (KOS strain), herpes simplex virus-2 (G strain), vaccinia virus, thymidine kinase deficient (acyclovir-resistant) herpes simplex virus-1 (TK KOS ACVr), adenovirus-2, human coronavirus (229E strain), cytomegalovirus (AD-169 and Davis strains) and varicella-zoster virus (TK+ VZV and TK VZV strains); (b) HeLa cell cultures: vesicular stomatitis virus, Coxsackie virus B4 and respiratory syncytial virus; (c) Vero cell cultures: para-influenza-3 virus, reovirus-1, Sindbis virus, Coxsackie virus B4, Punta Toro virus and yellow fever virus; and (d) MDCK cell cultures: influenza A virus (H1N1 and H3N2 subtypes) and influenza B virus. Ganciclovir, cidofovir, acyclovir, brivudin, zalcitabine, zanamivir, alovudine, mycophenolic acid, amantadine, rimantadine, ribavirin, dextran sulfate (molecular weight 10000, DS-10000) and Urtica dioica agglutinin (UDA) were used as the reference compounds. The antiviral activity was expressed as the EC50: the compound concentration required to reduce virus plaque formation (VZV) by 50% or to reduce virus-induced cytopathicity by 50% (other viruses). The cytotoxicity of the compounds tested toward the uninfected host cells was defined as the minimum cytotoxic concentration (MCC) that causes a microscopically detectable alteration of normal cell morphology. The 50% cytotoxic concentration (CC50), causing a 50% decrease in cell viability was determined using a colorimetric MTS assay system. None of the tested compounds showed appreciable antiviral activity toward any of the tested DNA and RNA viruses at the concentration up to 100 μM. At the same time, none of them affected the cell morphologies of used uninfected host cells.

2.1.2. Cytostatic Activity

The 50% cytostatic inhibitory concentration (IC50) causing a 50% decrease in cell proliferation was determined for all isolated isoxazolidines 14 and γ-lactams 15 toward nine cancerous cell lines (Capan-1 (pancreatic adenocarcinoma), Hap1 (chronic myeloid leukemia), HCT-116 (colorectal carcinoma), NCI-H460 (lung carcinoma), DND-41 (acute lymphoblastic leukemia), HL-60 (acute myeloid leukemia), K-562 (chronic myeloid leukemia), MM.1S (multiple myeloma) and Z-138 (non-Hodgkin lymphoma)) and normal retina (non-cancerous) cells (hTERT RPE-1). Docetaxel, etoposide and stauroporine were used as the reference compounds.
All of the compounds tested were not toxic to non-cancerous retina cells (hTERT RPE-1) at concentrations up to 100 μM. They were also not active against DND-41, HL-60, K-562, MM.1S or Z-138 cancer cells. All isoxazolidines 14 and γ-lactams 15 we tested, except cis-14c (B = F-Ura), exhibited moderate activity against pancreatic adenocarcinoma cells (Capan-1) (IC50 = 18.2 to 95 µM), and among them, γ-lactams trans-15e (Cl-Ura) and cis-15h (Theo) were the most active, with IC50 values of 21.5 and 18.2 µM, respectively (Table 3). In most cases, both cis-configured isoxazolidines 14 and cis-configured γ-lactams 15 were slightly less active when compared to the respective trans-isomers (e.g., cis-14bf versus trans-14bf, cis-14hi versus trans-14hI and cis-15b versus trans-15b, cis-15d vs. trans-15d); however, that correlation did to apply for uracil, dihydrouracil and theophiline-derivatives 14a, 15g and 15h. Moreover, several isoxazolidines 14 and γ-lactams 15 inhibited the proliferation of Hap1 (IC50 = 40.9 to 58.5 µM), HCT-116 (IC50 = 26.7 to 68,4 µM) and NCI-H460 cells (IC50 = 28.6 to 62.3 µM) (Table 3).

3. Experimental Section

3.1. General

1H-NMR spectra were taken in CDCl3, CD3OD, D2O and DMSO on the following spectrometers: Varian Gemini 2000BB (200 MHz), Varian Mercury-300 and Bruker Avance III spectrometers (600 MHz) with TMS as internal standard. 13C-NMR spectra were recorded in CDCl3, CD3OD, D2O and DMSO on the Bruker Avance III spectrometer (at 150 MHz, Bruker Instruments, Karlsruhe, Germany) and a Varian Mercury-300 machine (Varian NMR Instrument, Palo Alto, CA, USA) at 75 MHz. IR spectra were measured on a Bruker Alpha-T FT-IR spectrometer (Bruker Optik GmbH, Ettlingen, Germany). Melting points were determined on Boetius apparatus and are reported uncorrected. Elemental analyses were performed by the Microanalytical Laboratory of our faculty on a Perkin–Elmer PE 2400 CHNS analyzer (Perkin-Elmer Corp., Norwalk, CT, USA). The following adsorbents were used: column chromatography, Merck silica gel 60 (70–230 mesh), analytical TLC, Merck TLC plastic sheets and silica gel 60 F254. The preparative HPLC experiment was performed on a Waters chromatograph equipped with a Waters 2545 binary gradient module (Waters Corporation, Milford, MA, USA) and a Waters 2998 photodiode array detector (190–600 nm).
1H- and 13C-NMR spectra of all new synthesized compounds are provided in Supplementary Materials.

3.2. General Procedure for the Synthesis of Isoxazolidines cis-14 and trans-14

To a solution of a nitrone (1.00 mmol) in methanol (9 mL), methyl acrylate (10.0 mL) was added. The mixture was stirred at 60 °C for 24 h. The solvent was removed in vacuo and crude products were purified on silica gel columns using chloroform-methanol (100:1, 50:1, 20:1, 10:1, v/v) as eluents. The respective fractions were subjected to HPLC on a X Bridge Prep, C18, 5 µm, OBD, 19 × 100 mm column using water/methanol (90:10, 85:15, v/v) to afford pure isoxazolidines.
Methyl cis-3-{[2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]methyl}-2-methylisoxazolidine-5-carboxylate (cis-14a). Yield: 26%; a colorless amorphous solid; m.p. = 107–108 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3478, 3196, 3097, 3054, 2958, 1684, 1457, 1376, 1252, 1214, 1078, 811; 1H-NMR (200 MHz, CDCl3) δ: 9.84 (br s, 1H, NH), 7.37 (d, 1H, 3J = 7.8 Hz), 5.67 (d, 1H, 3J = 7.8 Hz), 4.74 (dd, 1H, 3J(H5–H4α) = 9.9 Hz, 3J(H5–H4β) = 5.1 Hz, HC5), 3.93 (dd, 1H, 2J(HCH) = 12.9 Hz, 3J(HCC–H3) = 3.3 Hz, HCH-Ura), 3.78 (s, 3H, C(O)OCH3), 3.55 (dd, 1H, 2J(HCH) = 12.9 Hz, 3J(HCC–H3) = 9.6 Hz, HCH-Ura), 3.47 (dddd, 1H, 3J(H3–CCH) = 9.6 Hz, 3J(H3–H4α) = 7.8 Hz, 3J(H3–CCH) = 3.3 Hz, 3J(H3–H4β) = 2.4 Hz, HC3), 2.86 (ddd, 1H, 2J(H4α–H4β) = 13.5 Hz, 3J(H4α–H5) = 9.9 Hz, 3J(H4α–H3) = 7.8 Hz, HαC4), 2.62 (s, 3H, CH3N), 2.17 (ddd, 1H, 2J(H4β–H4α) = 13.5 Hz, 3J(H4β–H5) = 5.1 Hz, 3J(H4β–H3) = 2.4 Hz, HβC4); 13C-NMR (75 MHz, CDCl3) δ: 171.45 (C(O)OCH3), 164.29 (C4′), 151.34 (C2′), 146.82 (C5′), 101.45 (C6′), 74.52 (C5), 65.48 (C3), 52.90 (C(O)OCH3), 50.82 (CH2-Ura), 44.59 (CH3N), 34.95 (C4). Anal. Calcd for C11H15N3O5: C, 49.07; H, 5.62; N, 15.61. Found: C, 48.99; H, 5.82; N, 15.80.
Methyl trans-3-{[2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]methyl}-2-methylisoxazolidine-5-carboxylate (trans-14a). Yield: 19%; a colorless amorphous solid; m.p. = 151–152 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3192, 3045, 2961, 2830, 1730, 1704, 1675, 1451, 1212, 1088, 1039, 914; 1H-NMR (600 MHz, CDCl3) δ: 9.47 (br s, 1H, NH), 7.30 (d, 1H, 3J = 7.9 Hz), 5.68 (d, 1H, 3J = 7.9 Hz), 4.54 (dd, 1H, 3J(H5–H4β) = 9.0 Hz, 3J(H5–H4α) = 7.4 Hz, HC5), 4.02 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.8 Hz, HCH-Ura), 3.79 (s, 3H, C(O)OCH3), 3.49 (dddd, 1H, 3J(H3–CCH) = 9.3 Hz, 3J(H3–H4α) = 7.4 Hz, 3J(H3–CCH) = 3.8 Hz, 3J(H3–H4β) = 2.4 Hz HC3), 3.36 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.3 Hz, HCH-Ura), 2.81 (ddd, 1H, 2J(H4α–H4β) = 13.3 Hz, 3J(H4α–H5) = 7.4 Hz, 3J(H4α–H3) = 7.4 Hz, HαC4), 2.68 (s, 3H, CH3N), 2.38 (ddd, 1H, 2J(Hβ4–Hα4) = 13.3 Hz, 3J(H4β–H5) = 9.0 Hz, 3J(H4β–H3) = 2.4 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.33 (C(O)OCH3), 163.84 (C4′), 151.14 (C2′), 146.27 (C5′), 101.50 (C6′), 76.28 (C5), 65.73 (C3), 52.58 (C(O)OCH3), 49.97 (CH2-Ura), 45.66 (CH3N), 34.64 (C4). Anal. Calcd for C11H15N3O5 × 0.25 H2O: C, 48.26; H, 5.71; N, 15.35. Found: C, 48.10; H, 5.53; N, 15.02.
Methyl cis-2-methyl-3-{[5-methyl-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]methyl}isoxazolidine-5-carboxylate (cis-14b). Yield: 7.4%; a colorless amorphous solid; m.p. = 73–74 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3168, 3037, 2955, 2893, 2830, 1746, 1701, 1469, 1437, 1214, 1074, 891; 1H-NMR (200 MHz, CDCl3) δ: 8.63 (br s, 1H, NH), 7.15 (q, 1H, 4J = 1.1 Hz), 3.82 (dd, 1H, 3J(H5–H4α) = 9.8 Hz, 3J(H5–H4β) = 5.2 Hz, HC5), 3.82 (dd, 1H, 2J(HCH) = 12.9 Hz, 3J(HCC–H3) = 3.3 Hz, HCH–Thy), 3.79 (s, 3H, C(O)OCH3), 3.55–3.45 (m, 2H, HCH–Thy, HC3), 2.86 (ddd, 1H, 2J(H4α–H4β) = 13.6 Hz, 3J(H4α–H5) = 9.8 Hz, 3J(H4α–H3) = 7.6 Hz, HαC4), 2.61 (s, 3H, CH3N), 2.17 (ddd, 1H, 2J(H4β–H4α) = 13.6 Hz, 3J(H4β–H5) = 5.2 Hz, 3J(H4β–H3) = 2.1 Hz, HβC4), 1.91 (d, 3H, 4J = 1.1 Hz, CH3); 13C-NMR (150 MHz, CDCl3) δ: 171.45 (C(O)OCH3), 164.16 (C4′), 150.97 (C2′), 142.62 (C5′), 109.69 (C6′), 74.37 (C5), 65.47 (C3), 52.63 (C(O)OCH3), 50.76 (CH2–Thy), 44.49 (CH3N), 34.83 (C4), 12.83 (CH3). Anal. Calcd for C12H17N3O5: C, 50.88; H, 6.05; N, 14.83. Found: C, 50.83; H, 5.86; N, 14.86.
Methyl trans-2-methyl-3-{[5-methyl-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]methyl}isoxazolidine-5-carboxylate (trans-14b). Yield: 17%; a colorless amorphous solid; m.p. = 139–140 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3472, 3175, 3038, 2958, 2929, 2855, 2822, 1745, 1688, 1468, 1436, 1202, 1058; 1H-NMR (600 MHz, CDCl3) δ: 8.59 (br s, 1H, NH), 7.13 (s, 1H), 4.56 (dd, 1H, 3J(H5–H4β) = 8.9 Hz, 3J(H5–H4α) = 7.4 Hz, HC5), 3.98 (dd, 1H, 2J(HCH) = 13.8 Hz, 3J(HCC–H3) = 3.9 Hz, HCH–Thy), 3.81 (s, 3H, C(O)OCH3), 3.50 (dddd, 1H, 3J(H3–CCH) = 9.1 Hz, 3J(H3–H4α) = 7.4 Hz, 3J(H3–CCH) = 3.9 Hz, 3J(H3–H4β) = 2.6 Hz, HC3), 3.36 (dd, 1H, 2J(HCH) = 13.8 Hz, 3J(HCC–H3) = 9.1 Hz, HCH–Thy), 2.82 (ddd, 1H, 2J(H4α–H4β) = 13.5 Hz, 3J(H4α–H5) = 7.4 Hz, 3J(H4α–H3) = 7.4 Hz, HαC4), 2.70 (s, 3H, CH3N), 2.39 (ddd, 1H, 2J(H4β–H4α) = 13.5 Hz, 3J(H4β–H5) = 8.9 Hz, 3J(H4β–H3) = 2.6 Hz, HβC4), 1.94 (s, 3H, CH3); 13C-NMR (150 MHz, CDCl3) δ: 172.32 (C(O)OCH3), 164.01 (C4′), 150.92 (C2′), 142.15 (C5′), 109.93 (C6′), 76.24 (C5), 65.81 (C3), 52.59 (C(O)OCH3), 50.00 (CH2–Thy), 45.72 (CH3N), 34.73 (C4), 12.21 (CH3). Anal. Calcd for C12H17N3O5: C, 50.88; H, 6.05; N, 14.83. Found: C, 50.94; H, 6.03; N, 14.95.
Methyl cis-3-{[5-fluoro-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (cis-14c). Yield: 38%; a colorless amorphous solid; m.p. = 139–140 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3441, 3166, 3055, 3028, 2958, 2923, 2850, 1725, 1691, 1658, 1470, 1436, 1242, 1074; 1H-NMR (600 MHz, CDCl3) δ: 9.81 (br s, 1H, NH), 7.43 (d, 1H, 3J = 5.8 Hz), 4.75 (dd, 1H, 3J(H5–H4α) = 9.9 Hz, 3J(H5–H4β) = 5.1 Hz, HC5), 3.93 (dd, 1H, 2J(HCH) = 13.5 Hz, 3J(HCC–H3) = 3.3 Hz, HCH–FUra), 3.80 (s, 3H, C(O)OCH3), 3.55 (dd, 1H, 2J(HCH) = 13.5 Hz, 3J(HCC–H3) = 9.5 Hz, HCH–FUra), 3.48 (dddd, 1H, 3J(H3–CCH) = 9.5 Hz, 3J(H3–H4α) = 7.9 Hz, 3J(H3–CCH) = 3.3 Hz, 3J(H3–H4β) = 2.0 Hz, HC3), 2.87 (ddd, 1H, 2J(H4α–H4β) = 13.4 Hz, 3J(H4α–H5) = 9.9 Hz, 3J(H4α–H3) = 7.9 Hz, HαC4), 2.64 (s, 3H, CH3N), 2.19 (ddd, 1H, 2J(H4β–H4α) = 13.4 Hz, 3J(H4β–H5) = 5.1 Hz, 3J(H4β–H3) = 2.0 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.42 (C(O)OCH3), 157.48 (d, 2J(CCF) = 25.8 Hz, C4′), 149.96 (C2′), 139.83 (d, 1J(CF) = 234.2 Hz, C5′), 131.13 (d, 2J(CCF) = 32.8 Hz, C6′), 74.41 (C5), 65.37 (C3), 52.68 (C(O)OCH3), 50.57 (CH2–FUra), 44.27 (CH3N), 34.61 (C4). Anal. Calcd for C11H14FN3O5: C, 46.00; H, 4.91; N, 14.63. Found: C, 46.36; H, 4.62; N, 14.55.
Methyl trans-3-{[5-fluoro-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (trans-14c). Yield: 31%; a colorless amorphous solid; m.p. = 179–180 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3470, 3167, 3029, 2960, 2920, 2836, 1744, 1722, 1693, 1658, 1382, 1240, 1206, 1101, 907; 1H-NMR (600 MHz, CDCl3) δ: 8.29 (br s, 1H, NH), 7.47 (d, 1H, 3J = 5.7 Hz), 4.56 (dd, 1H, 3J(H5–H4β) = 9.0 Hz, 3J(H5–H4α) = 7.5 Hz, HC5), 4.03 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.6 Hz, HCH–FUra), 3.83 (s, 3H, C(O)OCH3), 3.52 (dddd, 1H, 3J(H3–CCH) = 9.5 Hz, 3J(H3–H4α) = 7.5 Hz, 3J(H3–CCH) = 3.6 Hz, 3J(H3–H4β) = 2.2 Hz, HC3), 3.31 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.5 Hz, HCH–FUra), 2.86 (ddd, 1H, 2J(H4α–H4β) = 13.4 Hz, 3J(H4α–H5) = 7.5 Hz, 3J(H4α–H3) = 7.5 Hz, HαC4), 2.71 (s, 3H, CH3N), 2.39 (ddd, 1H, 2J(H4β–H4α) = 13.4 Hz, 3J(H4β–H5) = 9.0 Hz, 3J(H4β–H3) = 2.2 Hz, HβC4); signals of trans-14c were extracted from the 13C-NMR spectrum of a 60:40 mixture of trans-14c and cis-14c); 13C-NMR (150 MHz, CDCl3) δ: 172.29 (C(O)OCH3), 157.09 (d, 2J(CCF) = 26.2 Hz, C4′), 149.58 (C2′), 139.84 (d, 1J(CF) = 235.6 Hz, C5′), 130.71 (d, 2J(CCF) = 33.0 Hz, C6′), 76.35 (C5), 65.75 (C3), 52.69 (C(O)OCH3), 49.96 (CH2–FUra), 45.61 (CH3N), 34.42 (C4). Anal. Calcd for C11H14FN3O5 × 0.25 H2O: C, 45.29; H, 5.01; N, 14.40. Found: C, 45.55; H, 4.99; N, 14.12.
Methyl cis-3-{[5-bromo-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (cis-14d). Yield: 3.5%; a colorless amorphous solid; m.p. = 62–64 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3156, 3094, 3007, 2959, 2922, 2876, 2831, 1759, 1710, 1679, 1461, 1285, 1044, 924; 1H-NMR (600 MHz, CDCl3) δ: 8.67 (br s, 1H, NH), 7.74 (s, 1H), 4.76 (dd, 1H, 3J(H5–H4α) = 9.9 Hz, 3J(H5–H4β) = 5.0 Hz, HC5), 3.95 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.8 Hz, HCH-BrUra), 3.83 (s, 3H, C(O)OCH3), 3.59 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.6 Hz, HCH–BrUra), 3.47 (dddd, 1H, 3J(H3–CCH) = 9.6 Hz, 3J(H3–H4α) = 8.2 Hz, 3J(H3–CCH) = 3.8 Hz, 3J(H3–H4β) = 2.1 Hz, HC3), 2.88 (ddd, 1H, 2J(H4α–H4β) = 13.4 Hz, 3J(H4α–H5) = 9.9 Hz, 3J(H4α–H3) = 8.2 Hz, HαC4), 2.66 (s, 3H, CH3N), 2.20 (ddd, 1H, 2J(H4β–H4α) = 13.4 Hz, 3J(H4β–H5) = 5.0 Hz, 3J(H4β–H3) = 2.1 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.33 (C(O)OCH3), 159.27 (C4′), 150.21 (C2′), 146.01 (C5′), 95.52 (C6′), 74.44 (C5), 65.29 (C3), 52.74 (C(O)OCH3), 50.73 (CH2–BrUra), 44.33 (CH3N), 34.52 (C4). Anal. Calcd for C11H14BrN3O5: C, 37.95; H, 4.05; N, 12.07. Found: C, 37.95; H, 3.97; N, 11.90.
Methyl trans-3-{[5-bromo-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (trans-14d). Yield: 15%; a colorless amorphous solid; m.p. = 185–186 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3416, 3156, 3093, 3004, 2959, 2924, 2878, 2833, 1711, 1681, 1614, 1511, 1463, 1285, 1255, 1032; 1H-NMR (600 MHz, CDCl3) δ: 8.32 (br s, 1H, NH), 7.67 (s, 1H), 4.57 (dd, 1H, 3J(H4β–H5) = 9.0 Hz, 3J(H4α–H5) = 7.4 Hz, HC5), 4.04 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.7 Hz, HCH–BrUra), 3.83 (s, 3H, C(O)OCH3), 3.50 (dddd, 1H, 3J(HCC–H3) = 9.7 Hz, 3J(H4α–H3) = 7.4 Hz, 3J(HCC–H3) = 3.7 Hz, 3J(H4β–H3) = 2.0 Hz, HC3), 3.33 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.7 Hz, HCH–BrUra), 2.87 (ddd, 1H, 2J(H4α–H4β) = 13.5 Hz, 3J(H4α–H5) = 7.4 Hz, 3J(H4α–H3) = 7.4 Hz, HαC4), 2.70 (s, 3H, CH3N), 2.38 (ddd, 1H, 2J(H4β–H4α) = 13.5 Hz, 3J(H4β–H5) = 9.0 Hz, 3J(H4β–H3) = 2.0 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.28 (C(O)OCH3), 159.10 (C4′), 150.07 (C2′), 145.66 (C5′), 95.68 (C6′), 76.34 (C5), 65.61 (C3), 52.67 (C(O)OCH3), 50.10 (CH2–BrUra), 45.65 (CH3N), 34.33 (C4). Anal. Calcd for C11H14BrN3O5: C, 37.95; H, 4.05; N, 12.07. Found: C, 38.03; H, 3.91; N, 11.78.
Methyl cis-3-{[5-chloro-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (cis-14e). Yield: 5.7%; a colorless amorphous solid; m.p. = 129–131 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3473, 3161, 3036, 2926, 2824, 1748, 1717, 1666, 1627, 1442, 1424, 1342, 1215, 1179, 1049, 1007, 902, 719; 1H-NMR (600 MHz, CDCl3) δ: 9.32 (br s, 1H, NH), 7.63 (s, 1H), 4.76 (dd, 1H, 3J(H5–H4α) = 9.9 Hz, 3J(H5–H4β) = 5.0 Hz, HC5), 3.95 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.8 Hz, HCH–ClUra), 3.82 (s, 3H, C(O)OCH3), 3.58 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.5 Hz, HCH–ClUra), 3.48 (dddd, 1H, 3J(H3–CCH) = 9.5 Hz, 3J(H3–H4α) = 8.2 Hz, 3J(H3–CCH) = 3.8 Hz, 3J(H3–H4β) = 2.2 Hz, HC3), 2.88 (ddd, 1H, 2J(H4α–H4β) = 13.5 Hz, 3J(H4α–H5) = 9.9 Hz, 3J(H4α–H3) = 8.2 Hz, HαC4), 2.65 (s, 3H, CH3N), 2.20 (ddd, 1H, 2J(H4β–H4α) = 13.5 Hz, 3J(H4β–H5) = 5.0 Hz, 3J(H4β–H3) = 2.2 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.37 (C(O)OCH3), 159.55 (C4′), 150.34 (C2′), 143.53 (C5′), 107.94 (C6′), 74.43 (C5), 65.28 (C3), 52.69 (C(O)OCH3), 50.71 (CH2–ClUra), 44.30 (CH3N), 34.57 (C4). Anal. Calcd for C11H14ClN3O5: C, 43.50; H, 4.65; N, 13.84; Found: C, 43.67; H, 4.75; N, 14.00.
Methyl trans-3-{[5-chloro-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (trans-14e). Yield: 5.0%; a colorless amorphous solid; m.p. = 165–167 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3453, 3165, 3029, 2952, 2843, 1756, 1697, 1678, 1628, 1468, 1432, 1365, 1209, 1181, 1056, 1028, 903, 752; 1H-NMR (600 MHz, CDCl3) δ: 8.34 (br s, 1H, NH), 7.57 (s, 1H), 4.56 (dd, 1H, 3J(H5–H4β) = 9.0 Hz, 3J(H5–H4α) = 7.5 Hz, HC5), 4.05 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.7 Hz, HCH–ClUra), 3.83 (s, 3H, C(O)OCH3), 3.50 (dddd, 1H, 3J(H3–CCH) = 9.7 Hz, 3J(H3–H4α) = 7.5 Hz, 3J(H3–CCH) = 3.7 Hz, 3J(H3–H4β) = 2.0 Hz, HC3), 3.32 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.7 Hz, HCH–ClUra), 2.87 (ddd, 1H, 2J(H4α–H4β) = 13.5 Hz, 3J(H4α–H5) = 7.5 Hz, 3J(H4α–H3) = 7.5 Hz, HαC4), 2.70 (s, 3H, CH3N), 2.38 (ddd, 1H, 2J(H4β–H4α) = 13.5 Hz, 3J(H4β–H5) = 9.0 Hz, 3J(H4β–H3) = 2.0 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.26 (C(O)OCH3), 158.89 (C4′), 149.74 (C2′), 143.14 (C5′), 108.06 (C6′), 76.36 (C5), 65.62 (C3), 52.65 (C(O)OCH3), 50.11 (CH2–ClUra), 45.64 (CH3N), 34.34 (C4). Anal. Calcd for C11H14ClN3O5: C, 43.50; H, 4.65; N, 13.84; Found: C, 43.66; H, 4.80; N, 13.90.
Methyl cis-3-{[5-iodo-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (cis-14f). Yield: 2.7%; a colorless amorphous solid; m.p. = 189–191 °C (crystallized from ethyl acetate–hexane). IR (KBr, cm−1) νmax: 3407, 3144, 3089, 3002, 2918, 2828, 1759, 1709, 1667, 1606, 1427, 1335, 1246, 1209, 1035, 886, 621; 1H-NMR (600 MHz, CDCl3) δ: 7.79 (s, 1H), 4.76 (dd, 1H, 3J(H5–H4α) = 9.8 Hz, 3J(H5–H4β) = 5.3 Hz, HC5), 3.92 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.8 Hz, HCH–IUra), 3.83 (s, 3H, C(O)OCH3), 3.59 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.6 Hz, HCH–IUra), 3.46 (dddd, 1H, 3J(H3–CCH) = 9.6 Hz, 3J(H3–H4α) = 8.4 Hz, 3J(H3–CCH) = 3.8 Hz, 3J(H3–H4β) = 2.2 Hz, HC3), 2.87 (ddd, 1H, 2J(H4α–H4β) = 13.6 Hz, 3J(H4α–H5) = 9.8 Hz, 3J(H4α–H3) = 8.4 Hz, HαC4), 2.64 (s, 3H, CH3N), 2.19 (ddd, 1H, 2J(H4β–H4α) = 13.6 Hz, 3J(H4β–H5) = 5.3 Hz, 3J(H4β–H3) = 2.2 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.32 (C(O)OCH3), 160.27 (C4′), 150.94 (C2′), 150.51 (C6′), 74.43 (C5), 66.62 (C3), 65.33 (C5′), 52.76 (C(O)OCH3), 50.73 (CH2–IUra), 44.36 (CH3N), 34.53 (C4). Anal. Calcd for C11H14IN3O5: C, 33.44; H, 3.57; N, 10.63. Found: C, 33.34; H, 3.62; N, 10.51.
Methyl trans-3-{[5-iodo-2,4-dioxo-3,4–dihydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (tran-14f). Yield: 5.3%; a colorless amorphous solid; m.p. = 214–216 °C (crystallized from chloroform–methanol). IR (KBr, cm−1) νmax: 3453, 3148, 3002, 2962, 2829, 1680, 1607, 1460, 1431, 1298, 1253, 1080, 895, 816, 721, 622; 1H-NMR (600 MHz, CDCl3) δ: 7.75 (s, 1H), 4.56 (dd, 1H, 3J(H5–H4β) = 9.0 Hz, 3J(H5–H4α) = 7.5 Hz, HC5), 4.03 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 3.7 Hz, HCH–IUra), 3.83 (s, 3H, C(O)OCH3), 3.55 (dddd, 1H, 3J(H3–CCH) = 9.7 Hz, 3J(H3–H4α) = 7.5 Hz, 3J(H3–CCH) = 3.7 Hz, 3J(H3–H4β) = 2.0 Hz, HC3), 3.34 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H3) = 9.7 Hz, HCH–IUra), 2.86 (ddd, 1H, 2J(H4α–H4β) = 13.4 Hz, 3J(H4α–H5) = 7.5 Hz, 3J(H4α–H3) = 7.5 Hz, HαC4), 2.70 (s, 3H, CH3N), 2.37 (ddd, 1H, 2J(H4β–H4α) = 13.4 Hz, 3J(H4β–H5) = 9.0 Hz, 3J(H4β–H3) = 2.0 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.28 (C(O)OCH3), 160.10 (C4′), 150.62 (C2′), 150.38 (C6′), 76.32 (C5), 66.78 (C3), 65.63 (C5′), 52.67 (C(O)OCH3), 50.05 (CH2–IUra), 45.66 (CH3N), 34.34 (C4). Anal. Calcd for C11H14IN3O5: C, 33.44; H, 3.57; N, 10.63. Found: C, 33.57; H, 3.45; N, 10.76.
Methyl cis-3-{[2,4-dioxotetrahydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (cis-14g). Yield: 6.4%; a colorless amorphous solid; m.p. = 102–104 °C (crystallized from ethyl acetate–hexane). IR (KBr, cm−1) νmax: 3366, 3243, 3110, 2957, 2921, 1721, 1675, 1486, 1440, 1384, 1331, 1228, 1205, 1152, 1060, 801; 1H-NMR (600 MHz, CDCl3) δ: 5.61 (br s, 1H, NH), 4.61 (dd, 1H, 3J(H5–H4α) = 9.5 Hz, 3J(H5–H4β) = 5.4 Hz, HC5), 3.96 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 4.8 Hz, HCH–dihydroUra), 3.87 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 6.9 Hz, HCH–dihydroUra), 3.80 (s, 3H, C(O)OCH3), 3.41 (td, 2H, 3J = 7.0 Hz, 2J = 2.7 Hz, CH2CH2N), 3.10–3.00 (br m, 1H, HC3), 2.76–2.73 (m, 5H, CH3N and C(O)CH2CH2N), 2.67 (ddd, 1H, 2J(H4α–H4β) = 12.9 Hz, 3J(H4α–H5) = 9.5 Hz, 3J(H4α–H3) = 7.8 Hz, HαC4), 2.45 (ddd, 1H, 2J(H4β–H4α) = 12.9 Hz, 3J(H4β–H3) = 6.6 Hz, 3J(H4β–H5) = 5.4 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.25 (C(O)OCH3), 169.61 (C4′), 154.65 (C2′), 74.19 (C5), 66.42 (C3), 52.36 (C(O)OCH3), 44.31 (CH2–dihydroUra), 40.97 (CH3N), 36.71 (C6′), 35.33 (C4), 31.74 (C5′). Anal. Calcd for C11H17N3O5: C, 48.70; H, 6.32; N, 15.49. Found: C, 48.91; H, 6.29; N, 15.33.
Methyl trans-3-{[2,4-dioxotetrahydropyrimidin-1(2H)-yl]metyl}-2-methylisoxazolidine-5-carboxylate (trans-14g). Yield: 8.1%; a colorless amorphous solid; m.p. = 73–75 °C (crystallized from ethyl acetate–hexane). IR (KBr, cm−1) νmax: 3369, 3241, 3113, 2959, 2923, 1724, 1679, 1486, 1440, 1387, 1228, 1154, 1098, 1010, 763; 1H-NMR (600 MHz, CDCl3) δ: 5.71 (br s, 1H, NH), 4.62 (dd, 1H, 3J(H5–H4β) = 8.6 Hz, 3J(H5–H4α) = 6.9 Hz, HC5), 3.91 (dd, 1H, 2J(HCH) = 13.5 Hz, 3J(HCC–H3) = 5.9 Hz, HCH–dihydroUra), 3.89 (dd, 1H, 2J(HCH) = 13.5 Hz, 3J(HCC–H3) = 7.3 Hz, HCH–dihydroUra), 3.78 (s, 3H, C(O)OCH3), 3.44 (td, 2H, 3J = 6.8 Hz, 2J = 2.8 Hz), 3.32 (dddd, 1H, 3J(H3–CCH) = 7.3 Hz, 3J(H3–H4α) = 6.9 Hz, 3J(H3–CCH) = 5.9 Hz, 3J(H3–H4β) = 4.9 Hz, HC3), 2.80 (s, 3H, CH3N), 2.77 (t, 2H, 3J = 6.8 Hz), 2.55 (ddd, 1H, 2J(H4α–H4β) = 12.9 Hz, 3J(H4α–H5) = 6.9 Hz, 3J(H4α–H3) = 6.9 Hz, HαC4), 2.45 (ddd, 1H, 2J(H4β–H4α) = 12.9 Hz, 3J(H4β–H5) = 8.6 Hz, 3J(H4β–H3) = 4.9 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.54 (C(O)OCH3), 169.65 (C4′), 154.71 (C2′), 75.71 (C5), 65.62 (C3), 52.31 (C(O)OCH3), 45.31 (CH2–dihydroUra), 41.19 (CH3N), 36.14 (C6′), 35.39 (C4), 31.64 (C5′). Anal. Calcd for C11H17N3O5: C, 48.70; H, 6.32; N, 15.49. Found: C, 49.05; H, 6.46; N, 15.58.
Methyl cis-3-[(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-9H-purin-9-yl)methyl]-2-methylisoxazolidine-5-carboxylate (cis-14h). Yield: 23%; a colorless amorphous solid; m.p. = 139–141 °C (crystallized from chloroform–diethyl ether). IR (KBr, cm−1) νmax: 2999, 2941, 1738, 1702, 1660,1546, 1455, 1409, 1373, 1286, 1184, 1091, 724; 1H-NMR (600 MHz, CDCl3) δ: 7.71 (s, 1H, HC8′), 4.75 (dd, 1H, 3J(H5–H4α) = 9.7 Hz, 3J(H5–H4β) = 5.1 Hz, HC5), 4.38 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 4.7 Hz, HCH–The), 4.29 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 8.8 Hz, HCH–The), 3.83 (s, 3H, NCH3), 3.62 (s, 3H, C(O)OCH3), 3.60 (dddd, 1H, 3J(H3–CCH) = 8.8 Hz, 3J(H3–H4α) = 8.3 Hz, 3J(H3–CCH) = 4.7 Hz, 3J(H3–H4β) = 3.0 Hz, HC3), 3.44 (s, 3H, NCH3), 2.86 (ddd, 1H, 2J(H4α–H4β) = 13.3 Hz, 3J(H4α–H5) = 9.7 Hz, 3J(H4α–H3) = 8.3 Hz, HαC4), 2.59 (s, 3H, CH3N), 2.25 (ddd, 1H, 2J(H4β–H4α) = 13.3 Hz, 3J(H4β–H5) = 5.1 Hz, 3J(H4β–H3) = 3.0 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.47 (C(O)OCH3), 155.44 (C2′), 151.64 (C6′), 149.16 (C5′), 142.72 (C8′), 106.43 (C4′), 74.46 (C5), 66.68 (C3), 52.65 (C(O)OCH3), 49.11 (CH2–The), 44.52 (CH3N), 34.77 (C4), 29.81 (NCH3), 27.96 (NCH3). Anal. Calcd for C14H19N5O5: C, 49.85; H, 5.68; N, 20.76. Found: C, 49.75; H, 5.58; N, 20.64.
Methyl trans-3-[(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-9H-purin-9-yl)methyl]-2-methylisoxazolidine-5-carboxylate (trans-14h). Yield: 13%; a colorless amorphous solid; m.p. = 91–92 °C (crystallized from chloroform–diethyl ether). IR (KBr, cm−1) νmax: 2953, 2884, 1738, 1697, 1649, 1545, 1430, 1406, 1286, 1203, 1024, 973, 745; 1H-NMR (200 MHz, CDCl3) δ: 7.66 (s, 1H, HC8′), 4.54 (dd, 1H, 3J(H5–H4β) = 8.9 Hz, 3J(H5–H4α) = 7.5 Hz, HC5), 4.43 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 4.3 Hz, HCH–The), 4.12 (dd, 1H, 2J(HCH) = 13.7 Hz, 3J(HCC–H3) = 8.7 Hz, HCH–The), 3.78 (s, 3H, NCH3), 3.60 (s, 3H, C(O)OCH3), 3.55 (dddd, 1H, 3J(H3–CCH) = 8.7 Hz, 3J(H3–H4α) = 7.5 Hz, 3J(H3–CCH) = 4.3 Hz, 3J(H3–H4β) = 3.2 Hz, HC3), 3.42 (s, 3H, NCH3), 2.80 (ddd, 1H, 2J(H4α–H4β) = 13.3 Hz, 3J(H4α–H5) = 7.5 Hz, 3J(H4α–H3) = 7.5 Hz, HαC4), 2.62 (s, 3H, CH3N), 2.39 (ddd, 1H, 2J(H4β–H4α) = 13.3 Hz, 3J(H4β–H5) = 8.9 Hz, 3J(H4β–H3) = 3.2 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.11 (C(O)OCH3), 155.44 (C2′), 151.60 (C6′), 149.02 (C5′), 142.51 (C8′), 106.46 (C4′), 76.08 (C5), 66.70 (C3), 52.55 (C(O)OCH3), 48.55 (CH2–The), 45.65 (CH3N), 34.99 (C4), 29.80 (NCH3), 27.96 (NCH3). Anal. Calcd for C14H19N5O5: C, 49.85; H, 5.68; N, 20.76. Found: C, 49.60; H, 5.71; N, 20.65.
Methyl cis-3-[(6-amino-9H-purin-9-yl)methyl]-2-methylisoxazolidine-5-carboxylate (cis-14i). NMR signals of cis-14i were extracted from the spectra of a 67:33 mixture of cis-14i and trans-14i. 1H-NMR (300 MHz, CDCl3) δ: 8.34 (s, 1H, HC2′), 7.97 (s, 1H, HC8′), 5.96 (br s, 2H, NH2), 4.72 (dd, 1H, 3J(H5–H4α) = 9.7 Hz, 3J(H5–H4β) = 5.1 Hz, HC5), 4.26–4.23 (m, 2H, CH2–Ade), 3.78 (s, 3H, C(O)OCH3), 3.54–3.47 (m, 1H, HC3), 2.83 (ddd, 1H, 2J(H4α–H4β) = 13.4 Hz, 3J(H4α–H5) = 9.7 Hz, 3J(H4α–H3) = 8.2 Hz, HαC4), 2.58 (s, 3H, CH3N), 2.25 (ddd, 1H, 2J(H4β–H4α) = 13.4 Hz, 3J(H4β–H5) = 5.1 Hz, 3J(H4β–H3) = 3.3 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 172.09 (C(O)OCH3), 155.49 (C2′), 152.83 (C6′), 150.05 (C4′), 141.72 (C8′), 119.33 (C5′), 76.04 (C5), 66.34 (C3), 52.52 (C(O)OCH3), 45.54 (CH2–Ade), 45.36 (CH3N), 35.16 (C4). Anal. Calcd for C12H16N6O3: C, 49.31; H, 5.52; N, 28.75. Found: C, 49.22; H, 5.71; N, 28.52 (obtained on a 67:33 mixture of cis-14i and trans-14i).
Methyl trans-3-[(6-amino-9H-purin-9-yl)methyl]-2-methylisoxazolidine-5-carboxylate (trans-14i). NMR signals of trans-14i were extracted from the spectra of a 68:32 mixture of cis-14i and trans-14i 1H-NMR (300 MHz, CDCl3) δ: 8.35 (s, 1H, HC2′), 7.95 (s, 1H, HC8′), 5.94 (br s, 2H, NH2), 4.49 (dd, 1H, 3J(H4β–H5) = 8.7 Hz, 3J(H4α–H5) = 7.5 Hz, HC5), 4.28 (dd, 1H, 2J(HCH) = 14.1 Hz, 3J(HCC–H3) = 4.5 Hz, HCH–Ade), 4.14 (dd, 1H, 2J(HCH) = 14.1 Hz, 3J(HCC–H3) = 8.0 Hz, HCH–Ade), 3.77 (s, 3H, C(O)OCH3), 3.50 (dddd, 1H, 3J(HCC–H3) = 8.0 Hz, 3J(H4α–H3) = 7.8 Hz, 3J(HCC–H3) = 4.5 Hz, 3J(H4β–H3) = 3.4 Hz, HC3), 2.77 (ddd, 1H, 2J(H4α–H4β) = 13.2 Hz, 3J(H4α–H5) = 7.5 Hz, 3J(H4α–H3) = 7.8 Hz, HαC4), 2.64 (s, 3H, CH3N), 2.30 (ddd, 1H, 2J(H4β–H4α) = 13.2 Hz, 3J(H4β–H5) = 8.7 Hz, 3J(H4β–H3) = 3.4 Hz, HβC4); 13C-NMR (150 MHz, CDCl3) δ: 171.39 (C(O)OCH3), 155.42 (C2′), 152.65 (C6′), 149.98 (C4′), 142.01 (C8′), 119.44 (C5′), 74.48 (C5), 66.11 (C3), 52.62 (C(O)OCH3), 45.88 (CH2–Ade), 44.41 (CH3N), 34.99 (C4). Anal. Calcd for C12H16N6O3: C, 49.31; H, 5.52; N, 28.75. Found: C, 49.17; H, 5.24; N, 28.39 (obtained on a 67:33 mixture of cis-14i and trans-14i).

3.3. General Procedure for the Synthesis of γ-Lactams trans-15 and cis-15

A solution of the respective isoxazolidines cis-14 and trans-14 (1.00 mmol) in methanol (3 mL) was stirred under atmospheric pressure of hydrogen over Pd/C (0.05 mmol) with Pd(OH)2/C (0.05 mmol) at room temperature for 1–2 days. The suspension was filtered through a layer of Celite. The solvent was removed in vacuo and crude products were chromatographed on silica gel columns with chloroform:methanol mixtures (50:1, 20:1 and 10:1, v/v) and then on HPLC using a X Bridge Prep, C8, 5 µm, OBD, 19 × 100 mm column and a water:methanol mixture (98:2, v/v) as an eluent to give γ-lactams trans-15 and cis-15.
trans-1-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]-5-methylpyrimidine-2,4(1H,3H)-dione (trans-15b). Yield: 29%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3391, 3171, 3042, 2938, 2813, 2518, 1686, 1472, 1323, 1093, 944; 1H-NMR (600 MHz, CD3OD) δ: 7.38 (q, 1H, 4J = 1.0 Hz), 4.29 (dd, 1H, 3J(H4–H3β) = 9.0 Hz, 3J(H4–H3α) = 8.1 Hz, HC4), 4.01 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 4.6 Hz, HCH–Thy), 3.92 (dddd, 1H, 3J(H2–H3β) = 9.0 Hz, 3J(H2–CCH) = 5.9 Hz, 3J(H2–CCH) = 4.6 Hz, 3J(H2–H3α) = 1.0 Hz, HC2), 3.84 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 5.9 Hz, HCH–Thy), 2.92 (s, 3H, CH3N), 2.46 (ddd, 1H, 2J(H3α–H3β) = 13.2 Hz, 3J(H3α–H4) = 8.1 Hz, 3J(H3α–H2) = 1.0 Hz, HαC3), 2.02 (ddd, 1H, 2J(H3β–H3α) = 13.2 Hz, 3J(H3β–H4) = 9.0 Hz, 3J(H3β–H2) = 9.0 Hz, HβC3), 1.90 (d, 3H, 4J = 1.0 Hz, CH3); 13C-NMR (150 MHz, D2O) δ: 176.25 (C(O)), 166.91 (C4′), 152.55 (C2′), 142.71 (C5′), 111.53 (C6′), 68.26 (C4), 56.84 (CH2–Thy), 49.78 (C2), 31.81 (C3), 29.19 (CH3N), 11.32 (CH3). Anal. Calcd for C11H15N3O4: C, 52.17; H, 5.97; N, 16.59. Found: C, 52.25; H, 5.67; N, 16.59.
cis-1-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]-5-methylpyrimidine-2,4(1H,3H)-dione (cis-15b). Yield: 6.0%; a colorless amorphous solid; m.p. > 230 °C (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3552, 3454, 3389, 3315, 3175, 3042, 2952, 2930, 2832, 1692, 1661, 1469, 1127; 1H-NMR (600 MHz, D2O) δ: 7.36 (q, 1H, 4J = 1.1 Hz), 4.35 (dd, 1H, 3J(H4–H3α) = 8.3 Hz, 3J(H4–H3β) = 6.1 Hz, HC4), 4.19 (dd, 1H, 2J(HCH) = 14.4 Hz, 3J(HCC–H2) = 4.9 Hz, HCH–Thy), 3.94 (dddd, 1H, 3J(H2–H3α) = 7.6 Hz, 3J(H2–CCH) = 5.9 Hz, 3J(H2–H3β) = 5.8 Hz, 3J(H2–CCH) = 4.9 Hz, HC2), 3.90 (dd, 1H, 2J(HCH) = 14.4 Hz, 3J(HCC–H2) = 5.9 Hz, HCH–Thy), 2.88 (s, 3H, CH3N), 2.56 (ddd, 1H, 2J(H3α–H3β) = 13.7 Hz, 3J(H3α–H4) = 8.3 Hz, 3J(H3α–H2) = 7.6 Hz, HαC3), 1.86 (q, 3H, 4J = 1.1 Hz, CH3), 1.68 (ddd, 1H, 2J(H3β–H3α) = 13.7 Hz, 3J(H3β–H4) = 6.1 Hz, 3J(H3β–H2) = 5.8 Hz, HβC3); 13C-NMR (150 MHz, D2O) δ: 176.48 (C(O)), 167.35 (C4′), 152.94 (C2′), 143.01 (C5′), 111.23 (C6′), 69.11 (C4), 56.82 (CH2–Thy), 50.26 (C2), 31.06 (C3), 28.82 (CH3N), 11.38 (CH3). Anal. Calcd for C11H15N3O4: C, 52.17; H, 5.97; N, 16.59. Found: C, 52.39; H, 5.55; N, 16.60.
trans-1-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)metyl]dihydropyrimidine-2,4(1H,3H)-dione (trans-15g). Yield: 4.0%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3226, 3097, 3048, 2959, 2873, 1708, 1670, 1488, 1462, 1255, 1140, 1048. 1H-NMR (600 MHz, CD3OD) δ: 4.54 (br s, 1H, NH), 4.42 (dd, 1H, 3J(H4–H3β) = 8.7 Hz, 3J(H4–H3α) = 8.1 Hz, HC4), 3.85 (dddd, 1H, 3J(H2–H3β) = 8.7 Hz, 3J(H2–CCH) = 6.5 Hz, 3J(H2–CCH) = 4.8 Hz, 3J(H2–H3α) = 1.6 Hz, HC2), 3.71 (dd, 1H, 2J(HCH) = 14.1 Hz, 3J(HCC–H2) = 4.8 Hz, HCH–dihydroUra), 3.58–3.49 (m, 2H), 3.46 (dd, 1H, 2J(HCH) = 14.1 Hz, 3J(HCC–H2) = 6.5 Hz, HCH–dihydroUra), 2.94 (s, 3H, CH3N), 2.72–2.63 (m, 2H), 2.43 (ddd, 1H, 2J(H3α–H3β) = 13.1 Hz, 3J(H3α–H4) = 8.1 Hz, 3J(H3α–H2) = 1.6 Hz, HαC3), 2.02 (ddd, 1H, 2J(H3β–H3α) = 13.1 Hz, 3J(H3β–H4) = 8.7 Hz, 3J(H3β–H2) = 8.7 Hz, HβC3); 13C-NMR (150 MHz, CD3OD) δ: 175.44 (C(O)), 171.24 (C4′), 154.34 (C2′), 68.17 (C4), 56.28 (CH2–dihydroUra), 48.60 (C2), 43.19 (C6′), 32.44 (C5′), 30.50 (CH3N), 27.81 (C3). Anal. Calcd for C10H15N3O4: C, 49.79; H, 6.27; N, 17.42. Found: C, 49.80; H, 6.26; N, 17.40.
cis-1-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)metyl]dihydropyrimidine-2,4(1H,3H)-dione (cis-15g). Yield: 23%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3298, 3243, 3047, 2959, 2822, 1674, 1461, 1239, 1062. 1H-NMR (600 MHz, CD3OD) δ: 4.63 (br s, 1H, NH), 4.26 (dd, 1H, 3J(H4–H3α) = 8.2 Hz, 3J(H4–H3β) = 7.0 Hz, HC4), 3.93 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 4.6 Hz, HCH–dihydroUra), 3.79–3.75 (m, 1H, HC2), 3.56–3.53 (m, 2H), 3.45 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 5.9 Hz, HCH–dihydroUra), 2.93 (s, 3H, CH3N), 2.70 (td, 2H, 3J = 6.6 Hz, 2J = 2.9 Hz), 2.56 (ddd, 1H, 2J(H3α–H3β) = 13.4 Hz, 3J(H3α–H4) = 8.2 Hz, 3J(H3α–H2) = 7.5 Hz, HαC3), 1.73 (ddd, 1H, 2J(H3β–H3α) = 13.4 Hz, 3J(H3β–H4) = 7.0 Hz, 3J(H3β–H2) = 6.5 Hz, HβC3); 13C-NMR (150 MHz, CD3OD) δ: 175.79 (C(O)), 171.29 (C4′), 154.26 (C2′), 68.99 (C4), 55.97 (CH2–dihydroUra), 49.14 (C2), 43.16 (C6′), 31.93 (C5′), 30.38 (CH3N), 27.50 (C3). Anal. Calcd for C10H15N3O4: C, 49.79; H, 6.27; N, 17.42. Found: C, 49.68; H, 6.17; N, 17.30.
trans-9-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]-1,3-dimethyl-3,9-dihydro-1H-purine-2,6-dione (trans-15h). Yield: 34%; a colorless amorphous solid; m.p. > 230 °C (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3345, 3103, 2923, 2853, 1709, 1680, 1665, 1546, 1469, 1407,1316, 1237, 1085, 761. 1H-NMR (600 MHz, CDCl3) δ: 7.52 (s, 1H), 4.67 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 4.8 Hz, HCH–The), 4.21 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 6.4 Hz, HCH–The), 4.16 (dd, 1H, 3J(H4–H3α) = 8.6 Hz, 3J(H4–H3β) = 8.6 Hz, HC4), 4.07–4.04 (m, 1H, HC2), 3.62 (s, 3H, CH3), 3.45 (s, 3H, CH3), 2.94 (s, 3H, CH3N), 2.38 (ddd, 1H, 2J(H3α–H3β) = 13.4 Hz, 3J(H3α–H4) = 8.6 Hz, 3J(H3α–H2) = 8.0 Hz, HαC3), 2.14 (ddd, 1H, 2J(H3β–H3α) = 13.4 Hz, 3J(H3β–H2) = 9.0 Hz, 3J(H3β–H4) = 8.6 Hz, HβC3); 13C-NMR (150 MHz, CDCl3) δ: 174.89 (C(O)), 155.35 (C2′), 151.48 (C6′), 149.23 (C4′), 141.20 (C8′), 106.92 (C5′), 67.79 (C4), 57.33 (CH2–The), 48.56 (C2), 31.70 (CH3), 29.89 (CH3N), 29.07 (C3), 28.09 (CH3). Anal. Calcd for C13H17N5O4 × 0.2 H2O: C, 50.22; H, 5.64; N, 22.53. Found: C, 50.06; H, 5.46; N, 22.40.
cis-9-[(4-Hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]-1,3-dimethyl-3,9–dihydro-1H-purine-2,6-dione (cis-15h). Yield: 26%; a colorless amorphous solid; m.p. > 230 °C (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3284, 3112, 2999, 2961, 2920, 1694, 1679, 1654, 1457, 1296, 1238, 1085, 749. 1H-NMR (600 MHz, CDCl3) δ: 7.65 (s, 1H), 4.83 (dd, 1H, 2J(HCH) = 13.4 Hz, 3J(HCC–H2) = 4.8 Hz, HCH–The), 4.31 (dd, 1H, 3J(H4–H3α) = 7.6 Hz, 3J(H4–H3β) = 4.0 Hz, HC4), 4.23 (dd, 1H, 2J(HCH) = 13.4 Hz, 3J(HCC–H2) = 7.9 Hz, HCH–The), 3.94 (dddd, 1H, 3J(H2–CCH) = 7.9 Hz, 3J(H2–H3α) = 7.6 Hz, 3J(H2–H3β) = 4.0 Hz, 3J(H2–CCH) = 4.8 Hz, HC2), 3.62 (s, 3H, CH3), 3.44 (s, 3H, CH3), 2.96 (s, 3H, CH3N), 2.38 (ddd, 1H, 2J(H3α–H3β) = 13.8 Hz, 3J(H3α–H4) = 7.6 Hz, 3J(H3α–H2) = 7.6 Hz, HαC3), 1.76 (ddd, 1H, 2J(H3β–H3α) = 13.8 Hz, 3J(H3β–H4) = 4.0 Hz, 3J(H3β–H2) = 4.0 Hz, HβC3); 13C-NMR (150 MHz, CDCl3) δ: 175.09 (C(O)), 155.33 (C2′), 151.55 (C6′), 149.36 (C4′), 141.63 (C8′), 106.83 (C5′), 69.26 (C4), 57.75 (CH2–The), 50.21 (C2), 31.31 (CH3), 29.89 (CH3N), 28.82 (C3), 28.06 (CH3). Anal. Calcd for C13H17N5O4: C, 50.81; H, 5.58; N, 22.79. Found: C, 50.73; H, 5.67; N, 22.96.
trans-5-[(6-Amino-9H-purin-9-yl)methyl]-3-hydroxy-1-methylpyrrolidin-2-one (trans-15i). Yield: 28%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from water). IR (KBr, cm−1) νmax: 3319, 2955, 2923, 2853, 1687, 1603, 1483, 1326, 1244, 1132, 1000, 797. 1H-NMR (300 MHz, DMSO) δ: 8.17 (s, 1H, HC2′), 8.04 (s, 1H, HC8′), 7.23 (br s, 2H, NH2), 5.35 (d, 1H, J = 5.6 Hz, OH), 4.37 (dAB, 1H, 2JAB = 14.6 Hz, 3J(HCC–H2) = 3.8 Hz, HCH–Ade), 4.32 (dAB, 1H, 2JAB = 14.6 Hz, 3J(HCC–H2) = 4.5 Hz, HCH–Ade), 3.96–3.94 (m, 1H), 3.26–3.22 (m, 1H), 2.82 (s, 3H, CH3N), 2.30–2.23 (dd, 1H, 2J(H3β–H3α) = 13.1 Hz, 3J = 8.3 Hz, HαC3), 1.82 (ddd, 1H, 2J(H3β–H3α) = 13.1 Hz, 3J = 9.1 Hz, 3J = 9.0 Hz, HβC3); 13C-NMR (150 MHz, DMSO) δ: 174.34 (C(O)), 156.52 (C2′), 153.15 (C6′), 150.41 (C4′), 141.54 (C8′), 118.89 (C5′), 67.09 (C4), 56.52 (CH2–Ade), 44.56 (C2), 32.58 (C3), 28.52 (CH3N). Anal. Calcd for C11H14N6O2: C, 50.38; H, 5.38; N, 32.04. Found: C, 50.30; H, 5.20; N, 32.34.
cis-5-[(6-Amino-9H-purin-9-yl)methyl]-3-hydroxy-1-methylpyrrolidin-2-one (cis-15i). A colorless oil. IR (KBr, cm−1) νmax: 3469, 3341, 3173, 3071, 2959, 2922, 2852, 1745, 1601, 1511, 1473, 1303, 1270, 1038. NMR signals of cis-15i were extracted from the spectra of a 32:78 mixture of trans-15i and cis-15i 1H-NMR (300 MHz, DMSO-d6) δ: 8.14 (s, 1H, HC2′), 8.11 (s, 1H, HC8′), 7.20 (br s, 2H, NH2), 5.51 (d, 1H, J = 5.3 Hz, OH), 4.50 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 4.7 Hz, HCH–Ade), 4.29 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 6.1 Hz, HCH–Ade), 4.05–4.01 (m, 1H), 3.90–3.85 (m, 1H), 2.77 (s, 3H, CH3N), 2.27–2.23 (m, 1H, HαC3), 1.45 (dt, 1H 2J = 14.2 Hz, 3J = 7.0 Hz, HβC3); 13C-NMR (150 MHz, DMSO) δ: 175.09 (C(O)), 156.47 (C2′), 153.04 (C6′), 150.42 (C4′), 141.53 (C8′), 119.02 (C5′), 68.61 (C4), 55.78 (CH2–Ade), 45.84 (C2), 32.34 (C3), 28.57 (CH3N). Anal. Calcd for C11H14N6O2: C, 50.38; H, 5.38; N, 32.04. Found: C, 50.23; H, 5.08; N, 31.95 (obtained on a 40:60 mixture of trans-15i and cis-15i).

3.4. Synthesis of γ-Lactams cis-15d and trans-15d

A solution of a 53:35:8:2 mixture of γ-lactams trans-15a, cis-15a, trans-15g and trans-15g (0.130 g, 0.54 mmol) and N-bromosuccinimide (0.106 g, 0.594 mmol) in DMF (4 mL) was stirred at room temperature for 24 h. The solvent was removed in vacuo and the crude product was purified on a silica gel column using chloroform-methanol (100:1, v/v) as an eluent. The respective fractions were subjected to chromatography on a X Bridge Prep, C8, 5 µm, OBD, 19 × 100 mm column using water/methanol (98:2, v/v) to give pure γ-lactams trans-15d (0.051 g, 30%) and cis-15d (0.016 g, 9.3%).
trans-5-Bromo-1-[(4-hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]pyrimidine-2,4(1H,3H)-dione (trans-15d). Yield: 30%; a colorless amorphous solid; m.p. = 199–200 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3381, 3123, 3022, 2908, 2818, 1681, 1502, 1461, 1212 1017, 939. 1H-NMR (600 MHz, D2O) δ: 8.00 (s, 1H), 4.41 (dd, 1H, 3J(H4–H3β) = 8.9 Hz, 3J(H4–H3α) = 8.7 Hz, HC4), 4.03 (dd, 1H, 2J(HCH) = 13.4 Hz, 3J(HCC–H2) = 4.9 Hz, HCH–BrUra), 4.00 (dddd, 1H, 3J(H2–H3β) = 8.9 Hz, 3J(H2–CCH) = 5.0 Hz, 3J(H2–CCH) = 4.9Hz, 3J(H2–H3α) = 5.7 Hz, HC2), 3.92(dd, 1H, 2J(HCH) = 13.4 Hz, 3J(HCC–H2) = 5.0 Hz, HCH–BrUra), 2.88 (s, 3H, CH3N), 2.46 (ddd, 1H, 2J(H3α–H3β) = 13.5 Hz, 3J(H3α–H4) = 8.7 Hz, 3J(H3α–H2) = 5.7 Hz, HαC3), 2.07 (ddd, 1H, 2J(H3β–H3α) = 13.5 Hz, 3J(H3β–H4) = 8.9 Hz, 3J(H3β–H2) = 8.9 Hz, HβC3); 13C-NMR (150 MHz, CD3OD) δ: 175.66 (C(O)), 160.46 (C4′), 151.08 (C2′), 145.19 (C5′), 95.73 (C6′), 68.04 (C4), 56.46 (CH2–BrUra), 49.47 (C2), 32.13 (C3), 28.26 (CH3N). Anal. Calcd for C10H12BrN3O4: C, 37.76; H, 3.80; N, 13.21. Found: C, 37.99; H, 3.85; N, 13.42.
cis-5-Bromo-1-[(4-hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]pyrimidine-2,4(1H,3H)-dione (cis-15d). Yield: 9.3%; a colorless amorphous solid; m.p. = 209–210 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3550, 3452, 3170, 3040, 2960, 2853, 2780, 1678, 1629, 1460, 1344, 1133. 1H-NMR (600 MHz, D2O) δ: 7.90 (s, 1H), 4.34 (dd, 1H, 3J(H4–H3α) = 8.1 Hz, 3J(H4–H3β) = 6.1 Hz, HC4), 4.21 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 4.9 Hz, HCH–BrUra), 3.94 (dddd, 1H, 3J(H2–CCH) = 9.8 Hz, 3J(H2–H3α) = 7.8 Hz, 3J(H2–H3β) = 7.1 Hz, 3J(H2–CCH) = 4.9 Hz, HC2), 3.87 (dd, 1H, 2J(HCH) = 14.3 Hz, 3J(HCC–H2) = 9.8 Hz, HCH–BrUra), 2.88 (s, 3H, CH3N), 2.56 (ddd, 1H, 2J(H3α–H3β) = 13.7 Hz, 3J(H3α–H4) = 8.1 Hz, 3J(H3α–H2) = 7.8 Hz, HαC3), 1.68 (ddd, 1H, 2J(H3β–H3α) = 13.7 Hz, 3J(H3β–H2) = 7.1 Hz, 3J(H3β–H4) = 6.1 Hz, HβC3). 13C-NMR signals of cis-15d were extracted from the spectra of a 53:47 mixture of cis-15d and trans-15d 13C-NMR (150 MHz, CD3OD) δ: 175.44 (C(O)), 160.57 (C4′), 151.16 (C2′), 145.36 (C5′), 95.52 (C6′), 67.79 (C4), 56.14 (CH2–BrUra), 50.47 (C2), 31.48 (C3), 27.71 (CH3N). Anal. Calcd for C10H12BrN3O4: C, 37.76; H, 3.80; N, 13.21. Found: C, 38.00; H, 3.98; N, 13.50.

3.5. Synthesis of γ-Lactams cis-15e and trans-15e

A solution of a 53:35:8:2 mixture of γ-lactams trans-15a, cis-15a, trans-15g and trans-15g (0.136 g, 0.61 mmol) and N-chlorosuccinimide (0.163 g, 1.22 mmol) in freshly distilled pyridine (12 mL) was stirred at 100 °C for 1 h. The solvent was removed in vacuo and the crude product was purified on a silica gel column using chloroform-methanol (100:1, v/v) as an eluent. The respective fractions were subjected to chromatography on an X Bridge Prep, C8, 5 µm, OBD, 19 × 100 mm column using water/methanol (98:2, v/v) to give pure γ-lactams trans-15e (0.01 g, 6%) and cis-15d (0.007 g, 4.2%).
trans-5-Chloro-1-[(4-hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]pyrimidine-2,4(1H,3H)-dione (trans-15e). Yield: 4.2%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3404, 3160, 3035, 2957, 2824, 1749, 1716, 1665, 1627, 1442, 1342, 1214, 1069, 878, 750. 1H-NMR (600 MHz, CD3OD) δ: 7.90 (s, 1H), 4.33 (dd, 1H, 3J(H4–H3β) = 8.9 Hz, 3J(H4–H3α) = 8.1 Hz, HC4), 4.03 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 4.6 Hz, HCH–ClUra), 4.00 (dddd, 1H, 3J(H2–H3β) = 8.9 Hz, 3J(H2–CCH) = 6.2 Hz, 3J(H2–CCH) = 4.6 Hz, 3J(H2–H3α) = 0.8 Hz, HC2), 3.86 (dd, 1H, 2J(HCH) = 13.9 Hz, 3J(HCC–H2) = 6.2 Hz, HCH–ClUra), 2.93 (s, 3H, CH3N), 2.41 (ddd, 1H, 2J(H3α–H3β) = 13.3 Hz, 3J(H3α–H4) = 8.1 Hz, 3J(H3α–H2) = 0.8 Hz, HαC3), 2.02 (ddd, 1H, 2J(H3β–H3α) = 13.3 Hz, 3J(H3β–H4) = 8.9 Hz, 3J(H3β–H2) = 8.9 Hz, HβC3). 13C-NMR (150 MHz, CD3OD) δ: 175.47 (C(O)), 163.12 (C4′), 153.08 (C2′), 142.11 (C5′), 108.39 (C6′), 67.83 (C4), 56.23 (CH2–ClUra), 49.65 (C2), 32.02 (C3), 28.03 (CH3N). Anal. Calcd for C10H12ClN3O4: C, 43.89; H, 4.42; N, 15.35. Found: C, 43.78; H, 4.40; N, 15.37
cis-5-Chloro-1-[(4-hydroxy-1-methyl-5-oxopyrrolidin-2-yl)methyl]pyrimidine-2,4(1H,3H)-dione (cis-15e). Yield: 6.0%; a colorless amorphous solid; m.p. > 230 °C with decomposition (crystallized from methanol–chloroform). IR (KBr, cm−1) νmax: 3393, 3328, 3162, 3030, 2952, 2843, 1756, 1676, 1627, 1468, 1431, 1335, 1196, 903, 781. 1H-NMR (600 MHz, CD3OD) δ: 7.85 (s, 1H), 4.25–4.21 (m, 2H, HC4, HCH–ClUra), 3.90–3.86 (m, 1H, HC2), 3.84 (dd, 1H, 2J(HCH) = 13.4Hz, 3J(HCC–H2) = 6.5 Hz, HCH–ClUra), 2.92 (s, 3H, CH3N), 2.48 (ddd, 1H, 2J(H3α–H3β) = 13.6 Hz, 3J = 7.8 Hz, 3J = 7.6 Hz, HαC3), 1.75 (ddd, 1H, 2J(H3β–H3α) = 13.6 Hz, 3J = 5.6 Hz, 3J = 5.3 Hz, HβC3). 13C-NMR (150 MHz, CD3OD) δ: 175.69 (C(O)), 160.42 (C4′), 153.48 (C2′), 142.36 (C5′), 108.24 (C6′), 68.93 (C4), 56.27 (CH2–BrUra), 50.81 (C2), 31.58 (C3), 27.72 (CH3N). Anal. Calcd for C10H12ClN3O4: C, 43.89; H, 4.42; N, 15.35. Found: C, 43.80; H, 4.37; N, 15.30.

3.6. Antiviral Activity Assays

The compounds were evaluated against different herpesviruses, including herpes simplex virus type 1 (HSV-1) strain KOS, thymidine kinase-deficient (TK) HSV-1 KOS strain resistant to ACV (ACVr), herpes simplex virus type 2 (HSV-2) strain G, adeno virus-2, human coronavirus, varicella-zoster virus (VZV) TK+ strain Oka, TK VZV strains 07-1, human cytomegalovirus (HCMV) strains AD-169 and Davis; para-influenza-3 virus, reovirus-1, Sindbis virus, Coxsackie virus B4, Punta Toro virus, respiratory syncytial virus (RSV), vesicular stomatitis virus, yellow fever virus, influenza A virus subtypes H1N1 (A/PR/8) and H3N2 (A/HK/7/87); and influenza B virus (B/HK/5/72). The antiviral assays were based on inhibition of virus-induced cytopathicity or plaque formation (for VZV) in human embryonic lung (HEL) fibroblasts, African green monkey kidney cells (Vero), human epithelial cervix carcinoma cells (HeLa), or Madin Darby canine kidney cells (MDCK). Confluent cell cultures in microtiter 96-well plates were inoculated with 100 CCID50 of virus (1 CCID50 being the virus dose to infect 50% of the cell cultures) or with 20 plaque forming units (PFU) (for VZV) and the cell cultures were incubated in the presence of varying concentrations of the test compounds. Viral cytopathicity or plaque formation (VZV) was recorded as soon as it reached completion in the control virus-infected cell cultures that were not treated with the test compounds. Antiviral activity was expressed as the EC50 or compound concentration required to reduce virus-induced cytopathicity or viral plaque formation by 50%. Cytotoxicity of the test compounds was expressed as the minimum cytotoxic concentration (MCC) or the compound concentration that caused a microscopically detectable alteration of cell morphology.

3.7. Cytostatic Activity against Immortalized Cell Lines

All assays were performed in 96-well microtiter plates. To each well was added (5−7.5) × 104 tumor cells and a given amount of the test compound. The cells were allowed to proliferate at 37 °C in a humidified, CO2-controlled atmosphere. 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%.

4. Conclusions

A series of isoxazolidine analogues of homonucleosides cis-14 and trans-14 was synthesized by 1,3-dipolar cycloaddition of nucleobase-derived nitrones 13 and methyl acrylate. Based on 1H-NMR vicinal coupling constants the preferred 2E conformations were established for the isoxazolidine ring in both cis-14 and trans-14. Hydrogenolytic transformation of isoxazolidines cis-14 and trans-14 into γ-lactams trans-15 and cis-15 was then performed to produce the respective γ-lactams containing thymine (trans-15b/cis-15b), dihydrouracil (trans-15g/cis-15g), theophylline (trans-15h/cis-15h) and adenine (trans-15i/cis-15i) as nucleobases by the application of the established protocol.
Since, during the hydrogenation of 5-halogenated uracil-containing isoxazolidine homonucleosides, the halogen atom was removed from a nucleobase skeleton, 5-bromouracil and 5-chlorouracil derivatives trans-15d/cis-15d (Br-Ura) and trans-15e/cis-15e (Cl-Ura) were obtained via the treatment of uracil-containing γ-lactams with NBS and NCS, respectively.
All obtained isoxazolidine and γ-lactam homonucleosides were evaluated against a broad-spectrum of DNA and RNA viruses and appeared inactive at concentrations up to 100 µM. Antiproliferative properties of the obtained isoxazolidines and γ-lactams were evaluated on nine cancerous cell lines and several of them exhibited moderate inhibitory activity, and at the same time they were inactive toward normal retina cells.

Supplementary Materials

The Supplementary Materials are available online.

Author Contributions

The research group from the Medical University of Lodz (D.G.P., I.E.G., and J.G.) conceived the research project, participated in all steps of the research, interpreted the results, discussed the experimental data and prepared the manuscript. The research group from KU Leuven (D.S., R.S. and G.A.) conducted the biological assays and provided the experimental procedures and results. All authors commented on and approved the manuscript.

Funding

This work was funded by the National Science Centre (grant UMO-2013/11/N/NZ7/00723—synthetic part of the project) and KU Leuven (GOA 15/19 TBA—biological screening).

Acknowledgments

Special thanks are forwarded to the Rega Institute collaborators: Leentje Persoons, Ellen De Waegenaere, Bianca Stals, Kirsten Lepaige, Neils Willems and Nathalie Van Winkel.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chu, C.K. (Ed.) Antiviral Nucleosides: Chiral Synthesis and Chemotheraphy; Elsevier: Amsterdam, The Netherlands, 2003. [Google Scholar]
  2. De Clercq, E. New developments in anti-HIV chemotherapy. Biochim. Biophys. Acta 2002, 1587, 258–275. [Google Scholar] [CrossRef] [Green Version]
  3. Jordheim, L.P.; Durantel, D.; Zoulim, F.; Dumontet, C. Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat. Rev. Drug Discov. 2013, 12, 447–464. [Google Scholar] [CrossRef] [PubMed]
  4. De Clercq, E. Highlights in antiviral drug research: Antivirals at the horizon. Med. Res. Rev. 2013, 33, 1215–1248. [Google Scholar] [CrossRef] [PubMed]
  5. De Clercq, E. A 40-year journey in search of selective antiviral chemotherapy. Annu. Rev. Pharmacol. Toxicol. 2011, 51, 1–24. [Google Scholar] [CrossRef] [PubMed]
  6. De Clercq, E. HIV resistance to reverse transcriptase inhibitors. Biochem. Pharmacol. 1994, 47, 155–169. [Google Scholar] [CrossRef]
  7. Vince, R.; Hua, M.; Brownell, J.; Daluge, S.; Lee, F.; Shannon, W.M.; Lavelle, G.C.; Qualls, J.; Weislow, O.S.; Kiser, R.; et al. Potent and selective activity of a new carbocyclic nucleoside analog (Carbovir: NSC 614846) against human immunodeficiency virus In vitro. Biochem. Biophys. Res. Commun. 1988, 156, 1046–1053. [Google Scholar] [CrossRef] [Green Version]
  8. Tenney, D.J.; Yamanaka, G.; Voss, S.M.; Cianci, C.W.; Tuomari, A.V.; Sheaffer, A.K.; Alam, M.; Colonno, R.J. Lobucavir is phosphorylated in human cytomegalovirus-infected and -uninfected cells and inhibits the viral DNA polymerase. Antimicrob. Agents Chemother. 1997, 41, 2680–2685. [Google Scholar] [CrossRef] [Green Version]
  9. Norbeck, D.W.; Spanton, S.; Broder, S.; Mitsuya, H. (±)-Dioxolane-T ((±)-1-[(2β,4β)-2-(hydroxymethyl)-4-dioxolanyl]thymine): A new 2′,3′-dideoxynucleoside prototype with in vitro activity against HIV. Tetrahedron Lett. 1989, 30, 6263–6266. [Google Scholar] [CrossRef]
  10. Balzarini, J.; Naesens, L.; De Clercq, E. New antivirals—Mechanism of action and resistance development. Curr. Opin. Microbiol. 1998, 1, 535–546. [Google Scholar] [CrossRef]
  11. Galmarini, C.M.; Jordheim, L.; Dumontet, C. Pyrimidine nucleoside analogs in cancer treatment. Expert Rev. Anticancer Ther. 2003, 3, 717–728. [Google Scholar] [CrossRef]
  12. Ewald, B.; Sampath, D.; Plunkett, W. Nucleoside analogs: Molecular mechanisms signaling cell death. Oncogene 2008, 27, 6522–6537. [Google Scholar] [CrossRef] [PubMed]
  13. Deeks, E.D. Cladribine Tablets: A Review in Relapsing MS. CNS Drugs 2018, 32, 785–796. [Google Scholar] [CrossRef] [PubMed]
  14. Noble, S.; Goa, K.L. Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lung cancer and pancreatic cancer. Drugs 1997, 54, 447–472. [Google Scholar] [CrossRef]
  15. Scott, L.J. Azacitidine: A Review in Myelodysplastic Syndromes and Acute Myeloid Leukaemia. Drugs 2016, 76, 889–900. [Google Scholar] [CrossRef] [PubMed]
  16. Reese, N.D.; Schiller, G.J. High-Dose Cytarabine (HD araC) in the Treatment of Leukemias: A Review. Curr. Hematol. Malig. Rep. 2013, 8, 141–148. [Google Scholar] [CrossRef] [PubMed]
  17. Bonate, P.L.; Arthaud, L.; Cantrell, W.R., Jr.; Stephenson, K.; Secrist III, J.A.; Weitman, S. Discovery and development of clofarabine: A nucleoside analogue for treating cancer. Nat. Rev. Drug Discov. 2006, 5, 855–863. [Google Scholar] [CrossRef] [PubMed]
  18. Wróblewski, A.E.; Głowacka, I.E.; Piotrowska, D.G. 1′-Homonucleosides and their structural analogues: A review. Eur. J. Med. Chem. 2016, 118, 121–142. [Google Scholar] [CrossRef]
  19. Hossain, N.; Hendrix, C.; Lescrinier, E.; Van Aerschot, A.; Busson, R.; De Clercq, E.; Herdewijn, P. Homo-N-nucleosides: Incorporation into oligonucleotides and antiviral activity. Bioorg. Med. Chem. Lett. 1996, 6, 1465–1468. [Google Scholar] [CrossRef]
  20. De Clercq, E.; Descamps, J.; Verhelst, G.; Walker, R.T.; Jones, A.S.; Torrence, P.F.; Shugar, D. Comparative efficacy of antiherpes drugs against different strains of herpes simplex virus. J. Infect. Dis. 1980, 141, 563–574. [Google Scholar] [CrossRef]
  21. De Clercq, E.; Hols, A.; Rosenberg, I.; Sakuma, T.; Balzarini, J.; Maudgal, P.C.A. A novel selective broad-spectrum anti-DNA virus agent. Nature 1986, 323, 464–467. [Google Scholar] [CrossRef]
  22. Saladino, R.; Neri, V.; Checconi, P.; Celestino, I.; Nencioni, L.; Palmara, A.T.; Crucianelli, M. Synthesis of 2′-deoxy-1′-homo-N-nucleosides with anti-influenza activity by catalytic methyltrioxorhenium (MTO)/H2O2 oxyfunctionalization. Chem. Eur. J. 2013, 19, 2392–2404. [Google Scholar] [CrossRef] [PubMed]
  23. Sekino, Y.; Bruner, S.D.; Verdine, G.L. Selective inhibition of herpes simplex virus type-1 uracil-DNA glycosylase by designed substrate analogs. J. Biol. Chem. 2000, 275, 36506–36508. [Google Scholar] [CrossRef] [PubMed]
  24. Tronchet, J.M.J.; Winter-Mihaly, E.; Habashi, F.; Schwarzenbach, D.; Likic, U.; Geoffroy, M. Dérivés de désoxy-hydroxylamino-sucres et radicaux libres diglycosylnitroxydes correspondants. Communication préliminaire. Helv. Chem. Acta 1981, 64, 610–616. [Google Scholar] [CrossRef]
  25. Tronchet, J.M.J.; Iznaden, M.; Barbalat-Rey, F.; Dhimane, H.; Ricca, A.; Balzarini, J.; De Clercq, E. Isoxazolidine analogs of nucleosides. Eur. J. Med. Chem. 1992, 27, 555–560. [Google Scholar] [CrossRef]
  26. Loh, B.; Vozzolo, L.; Mok, B.J.; Lee, C.C.; Fitzmaurice, R.J.; Caddick, S.; Fassati, A. Inhibition of HIV-1 replication by isoxazolidine and isoxazole sulfonamides. Chem. Biol. Drug Des. 2010, 75, 461–474. [Google Scholar] [CrossRef]
  27. Hirano, K.; Kubota, T.; Tsuda, M.; Mikami, Y.; Kobayashi, J. Pyrinodemins B-D, potent cytotoxic bis-pyridine alkaloids from marine sponge amphimedon sp. Chem. Pharm. Bull. 2000, 48, 974–977. [Google Scholar] [CrossRef]
  28. Rescifina, A.; Chiacchio, M.A.; Corsaro, A.; De Clerq, E.; Iannazzo, D.; Mastino, A.; Piperno, A.; Romeo, G.; Romeo, R.; Valveri, V. Synthesis and Biological Activity of Isoxazolidinyl Polycyclic Aromatic Hydrocarbons:  Potential DNA Intercalators. J. Med. Chem. 2006, 49, 709–715. [Google Scholar] [CrossRef]
  29. Lee, L.W.; Taylor, C.C.E.; Desaulniers, J.P.; Zhang, M.; Hojfeldt, J.W.; Pan, Q.; Mapp, A.K. Inhibition of ErbB2(Her2) expression with small molecule transcription factor mimics. Bioorg. Med. Chem. Lett. 2009, 19, 6233–6236. [Google Scholar] [CrossRef] [Green Version]
  30. Rescifina, A.; Chiacchio, U.; Corsaro, A.; Piperno, A.; Romeo, R. Isoxazolidinyl polycyclic aromatic hydrocarbons as DNA-intercalating antitumor agents. Eur. J. Med. Chem. 2011, 46, 129–136. [Google Scholar] [CrossRef]
  31. Piotrowska, D.G.; Cieślak, M.; Królewska, K.; Wróblewski, A.E. Design, synthesis and cytotoxicity of a new series of isoxazolidine based nucleoside analogues. Arch. Pharm. Chem. Life Sci. 2011, 344, 301–310. [Google Scholar] [CrossRef]
  32. Damodiran, M.; Sivakumar, P.M.; Senthil Kumar, R.; Muraligharan, D.; Phani Kumar, B.V.; Doble, M.; Perumal, P.T. Antibacterial activity, quantitative structure–activity relationship and diastereoselective synthesis of isoxazolidine derivatives via 1,3-dipolar cycloaddition of D-glucose derived nitrone with olefin. Chem. Biol. Drug Des. 2009, 74, 494–506. [Google Scholar] [CrossRef] [PubMed]
  33. Sadashiva, M.P.; Mallesha, H.; Murthy, K.K.; Rangappa, K.S. Enhancement in antimicrobial activity of 2-(phenyl)-3-(2-butyl-4-chloro-1H-imidazolyl)-5-butylate isoxazolidine. Bioorg. Med. Chem. Lett. 2005, 15, 1811–1814. [Google Scholar] [CrossRef] [PubMed]
  34. Chiacchio, U.; Corsaro, A.; Iannazzo, D.; Piperno, A.; Pistara, V.; Rescifina, A.; Romeo, R.; Valveri, V.; Mastino, A.; Romeo, G. Enantioselective Syntheses and Cytotoxicity of N,O-Nucleosides. J. Med. Chem. 2003, 46, 3696–3702. [Google Scholar] [CrossRef] [PubMed]
  35. Piperno, A.; Giofre, S.V.; Iannazzo, D.; Romeo, R.; Romeo, G.; Chiacchio, U.; Rescifina, A.; Piotrowska, D.G. Synthesis of C-4′Truncated Phosphonated Carbocyclic 2′-Oxa-3′-azanucleosides as Antiviral Agents. J. Org. Chem. 2010, 75, 2798–2805. [Google Scholar] [CrossRef]
  36. Chiacchio, U.; Balestrieri, E.; Macchi, B.; Iannazzo, D.; Piperno, A.; Rescifina, A.; Romeo, R.; Saglimbeni, M.; Sciortino, M.T.; Valveri, V.; et al. Synthesis of Phosphonated Carbocyclic 2′-Oxa-aza-nucleosides: Novel Inhibitors of Reverse Transcriptase. J. Med. Chem. 2005, 48, 1389–1394. [Google Scholar] [CrossRef]
  37. Chiacchio, U.; Rescifina, A.; Iannazzo, D.; Piperno, A.; Romeo, R.; Borrello, L.; Sciortino, M.T.; Balestrieri, E.; Macchi, B.; Mastino, A.; et al. Phosphonated Carbocyclic 2′-Oxa-3′-azanucleosides as New Antiretroviral Agents. J. Med. Chem. 2007, 50, 3747–3750. [Google Scholar] [CrossRef]
  38. Balestrieri, E.; Matteucci, C.; Ascolani, A.; Piperno, A.; Romeo, R.; Romeo, G.; Chiacchio, U.; Mastino, A.; Macchi, B. Effect of phosphonated carbocyclic 2′-oxa-3′-aza-nucleoside on human T-cell leukemia virus type 1 infection in vitro. Antimicrob. Agents Chemother. 2008, 52, 54–64. [Google Scholar] [CrossRef]
  39. Chiacchio, U.; Corsaro, A.; Iannazzo, D.; Piperno, A.; Pistara, V.; Rescifina, A.; Romeo, R.; Sindona, G.; Romeo, G. Diastereo- and enantioselective synthesis of N,O-nucleosides. Tetrahedron Asymmetry 2003, 14, 2717–2723. [Google Scholar] [CrossRef]
  40. Chiacchio, U.; Saita, M.G.; Crispino, L.; Gumina, G.; Mangiafico, S.; Pistara, V.; Romeo, G.; Piperno, A.; De Clercq, E. Enantioselective synthesis of homocarbocyclic-2′-oxo-3′-azanucleosides. Tetrahedron 2006, 62, 1171–1181. [Google Scholar] [CrossRef]
  41. Chiacchio, U.; Genovese, F.; Iannazzo, D.; Librando, V.; Merino, P.; Rescifina, A.; Romeo, R.; Procopio, A.; Romeo, G. Diastereoselective synthesis of homo-N,O-nucleosides. Tetrahedron 2004, 60, 441–448. [Google Scholar] [CrossRef]
  42. Mannucci, V.; Cordero, F.M.; Piperno, A.; Romeo, G.; Brandi, A. Diastereoselective synthesis of a collection of new homonucleoside mimetics containing pyrrolo[1,2-b]isoxazoline and pyrrolidine rings. Tetrahedron Asymmetry 2008, 19, 1204–1209. [Google Scholar] [CrossRef]
  43. Romeo, G.; Giofre, S.A.; Piperno, A.; Romeo, R.; Chiacchio, M.A. Synthesis of N,O-homonucleosides with high conformational freedom. Arkivoc 2009, 8, 168–176. [Google Scholar]
  44. Pan, S.; Amankulor, N.M.; Zhao, K. Syntheses of isoxazolinyl and isoxazolidinyl nucleoside analogues. Tetrahedron 1998, 54, 6587–6604. [Google Scholar] [CrossRef]
  45. Gotkowska, J.; Balzarini, J.; Piotrowska, D.G. Synthesis of novel isoxazolidine analogues of homonucleosides. Tetrahedron Lett. 2012, 53, 7097–7100. [Google Scholar] [CrossRef]
  46. Piotrowska, D.G.; Balzarini, J.; Andrei, G.; Schols, D.; Snoeck, R.; Wróblewski, A.E.; Gotkowska, J. Novel isoxazolidine analogues of homonucleosides and homonucleotides. Tetrahedron 2016, 72, 8294–8308. [Google Scholar] [CrossRef]
  47. Takahashi, K.; Kawabata, M.; Uemura, D.; Iwadare, S.; Mitomo, R.; Nakano, F.; Matsuzaki, A. Structure of neooxazolomycin, an antitumor antibiotic. Tetrahedron Lett. 1985, 26, 1077–1078. [Google Scholar] [CrossRef]
  48. Kawazu, K.; Kanzaki, H.; Kawabata, G.; Kawai, S.; Kobayashi, A. Oxazolomycin Esters, Specific Inhibitors of Plant Transformation. Biosci. Biotechnol. Biochem. 1992, 56, 1382–1385. [Google Scholar] [CrossRef] [Green Version]
  49. Andrés, J.M.; de Elena, N.; Pedrosa, R.; Pérez-Encabo, A. Stereoselective synthesis of (5S,6S)- and (5S,6R)-aza-muricatacin from an L-glutamic acid derivative. Tetrahedron Asymmetry 2001, 12, 1503–1509. [Google Scholar] [CrossRef]
  50. Rieser, M.J.K.; John, F.; Wood, K.V.; McLaughlin, J.L. Muricatacin: A simple biologically active acetogenin derivative from the seeds of annona muricata (annonaceae). Tetrahedron Lett. 1991, 32, 1137–1140. [Google Scholar] [CrossRef]
  51. Baussanne, I.; Schwardt, O.; Royer, J.; Pichon, M.; Figadere, B.; Cavé, A. Synthesis of aza-muricatacin: An analogue of the bioactive muricatacin an acetogenin of Annonaceae. Tetrahedron Lett. 1997, 38, 2259–2262. [Google Scholar] [CrossRef]
  52. Cavé, A.; Chaboche, C.; Figadère, B.; Harmange, J.-C.; Laurens, A.; Peyrat, J.F.; Pichon, M.; Szlosek, M.; Cotte-Lafitte, J.; Quéro, A.M. Study of the structure-activity relationships of the acetogenin of annonaceae, muricatacin and analogues. Eur. J. Med. Chem. 1997, 32, 617–623. [Google Scholar] [CrossRef]
  53. Janecka, A.; Wyrębska, A.; Gach, K.; Fichna, J.; Janecki, T. Natural and synthetic α-methylenelactones and α-methylenelactams with anticancer potential. Drug Discov. Today 2012, 17, 561–572. [Google Scholar] [CrossRef] [PubMed]
  54. Baures, P.W.; Eggleston, D.S.; Erhard, K.F.; Cieslinski, L.B.; Torphy, T.J.; Christensen, S.B. Crystal structure, absolute configuration, and phosphodiesterase inhibitory activity of (+)-1-(4-bromobenzyl)-4-[(3-cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidinone. J. Med. Chem. 1993, 36, 3274–3277. [Google Scholar] [CrossRef] [PubMed]
  55. Akah, P.A.; Nwambie, A.I. Evaluation of Nigerian traditional medicines: 1. Plants used for rheumatic (inflammatory) disorders. J. Ethnopharmacol. 1994, 42, 179–182. [Google Scholar] [CrossRef]
  56. Barrett, A.G.M.; Head, J.; Smith, M.L.; Stock, N.S.; White, A.J.P.; Williams, D.J. Fleming-Tamao Oxidation and Masked Hydroxyl Functionality: Total Synthesis of (+)-Pramanicin and Structural Elucidation of the Antifungal Natural Product (-)-Pramanicin. J. Org. Chem. 1999, 64, 6005–6018. [Google Scholar] [CrossRef]
  57. Scgwartz, R.E.; Helms, G.L.; Bolessa, E.A.; Wilson, K.E.; Giacobbe, R.A.; Tkacz, J.S.; Bills, G.F.; Liesch, J.M.; Zink, D.L.; Curotto, J.E.; et al. Pramanicin, a novel antimicrobial agent from a fungal fermentation. Tetrahedron 1994, 50, 1675–1686. [Google Scholar] [CrossRef]
  58. Caruano, J.; Muccioli, G.G.; Robiette, R. Biologically active γ-lactams: Synthesis and natural sources. Org. Biomol. Chem. 2016, 14, 10134–10156. [Google Scholar] [CrossRef] [PubMed]
  59. Srivastav, N.C.; Shakya, N.; Bhavanam, S.; Agrawal, A.; Tse, C.; Desroches, N.; Kunimoto, D.Y.; Kumar, R. Antimycobacterial activities of 5-alkyl (or halo)-3′-substituted pyrimidine nucleoside analogs. Bioorg. Med. Chem. Lett. 2012, 22, 1091–1094. [Google Scholar] [CrossRef]
  60. Lazrek, H.B.; Taourirte, M.; Oulih, T.; Barascut, J.L.; Imbach, J.L.; Pannecouque, C.; Witrouw, M.; De Clercq, E. Synthesis and anti-HIV activity of new modified 1,2,3-triazole acyclonucleosides. Nucleosides Nucleotides Nucleic Acids 2001, 20, 1949–1960. [Google Scholar] [CrossRef]
  61. Strittmatter, T.; Aschenbrenner, J.; Hardt, N.; Marx, A. Synthesis of 4′-C-alkylated-5-iodo-2′-deoxypyrimidine nucleosides. Arkivoc 2013, 2, 46–59. [Google Scholar]
Sample Availability: Samples are not avaliable.
Figure 1. Examples of biologically-active nucleoside analogues.
Figure 1. Examples of biologically-active nucleoside analogues.
Molecules 24 04014 g001
Figure 2. Examples of biologically active 1′-homonucleosides.
Figure 2. Examples of biologically active 1′-homonucleosides.
Molecules 24 04014 g002
Figure 3. Examples of isoxazolidine nucleoside/nucleotide analogues.
Figure 3. Examples of isoxazolidine nucleoside/nucleotide analogues.
Molecules 24 04014 g003
Figure 4. Examples of some bioactive natural molecules containing a γ-lactam core.
Figure 4. Examples of some bioactive natural molecules containing a γ-lactam core.
Molecules 24 04014 g004
Scheme 1. Retrosynthesis of γ-lactam analogues of 1′-homonucleosides 15.
Scheme 1. Retrosynthesis of γ-lactam analogues of 1′-homonucleosides 15.
Molecules 24 04014 sch001
Scheme 2. Synthesis of isoxazolidine analogues of 1′-homonucleosides trans-14 and cis-14.
Scheme 2. Synthesis of isoxazolidine analogues of 1′-homonucleosides trans-14 and cis-14.
Molecules 24 04014 sch002
Scheme 3. Synthesis of the isoxazolidine cis-16 [45].
Scheme 3. Synthesis of the isoxazolidine cis-16 [45].
Molecules 24 04014 sch003
Figure 5. Preferred conformations 17 and 18 of isoxazolidines cis-14a14i and trans-14a14i, respectively, and the Newman projection 19.
Figure 5. Preferred conformations 17 and 18 of isoxazolidines cis-14a14i and trans-14a14i, respectively, and the Newman projection 19.
Molecules 24 04014 g005
Scheme 4. Synthesis of γ-lactam analogues of 1′-homonucleosides trans-15 and cis-15.
Scheme 4. Synthesis of γ-lactam analogues of 1′-homonucleosides trans-15 and cis-15.
Molecules 24 04014 sch004
Scheme 5. Synthesis of γ-lactams trans-15d and cis-15d (X = Br), and trans-15e and cis-15e (X = Cl).
Scheme 5. Synthesis of γ-lactams trans-15d and cis-15d (X = Br), and trans-15e and cis-15e (X = Cl).
Molecules 24 04014 sch005
Table 1. Isoxazolidines trans-14 and cis-14 obtained according to Scheme 2.
Table 1. Isoxazolidines trans-14 and cis-14 obtained according to Scheme 2.
EntryNucleobase BRatio of cis-14:trans-14Yield (%)
a Molecules 24 04014 i00150:50 [45]cis-14a − 26 a;
cis-14a + trans-14a − 50 b;
trans-14a − 19 a
b Molecules 24 04014 i00251:49cis-14b − 7.4 a;
cis-14b + trans-14b − 66 b;
trans-14b − 17 a
c Molecules 24 04014 i00350:50cis-14c − 38 a;
cis-14c + trans-14c − 4.4 b;
trans-14c − 31 a
d Molecules 24 04014 i00450:50cis-14d − 3.5 a;
cis-14d + trans-14d − 63 b;
trans-14d − 15 a
e Molecules 24 04014 i00550:50cis-14e − 5.7 a;
cis-14e + trans-14e − 75 b;
trans-14e − 5.0 a
f Molecules 24 04014 i00650:50cis-14f − 2.7 a;
cis-14f + trans-14f − 75 b;
trans-14f − 5.3 a
g Molecules 24 04014 i00746:54cis-14g − 6.4 a;
cis-14g + trans-14g − 57 b;
trans-14g − 8.1 a
h Molecules 24 04014 i00850:50cis-14h − 23 a;
cis-14h + trans-14h − 62 b;
trans-14h − 13 a
i Molecules 24 04014 i00949:51cis-14i + trans-14i − 64 b
a Yield of a pure diastereoisomer; b yield of a pure mixture of diastereoisomers;
Table 2. γ-Lactams trans-15 and cis-15 obtained according to Scheme 4.
Table 2. γ-Lactams trans-15 and cis-15 obtained according to Scheme 4.
EntryNucleobase (B)SubstratesProductsYields [%]
aUracis-14atrans-15g [45]80 a
trans-14acis-15g85 a
bThycis-14b:trans-14b
(80:20)
trans-15b:cis-15b
(80:20)
trans-15b − 29 a
cis-15b + trans-15b – 24 b
cis-15b – 6.0 a
cF-Uracis-14ctrans-15g70 a
dBr-Uracis-14d:trans-14d
(60:40)
trans-15a:cis-15a:trans-15g:cis-15g
(53:35:8:2)
Inseparable (77%)
eCl-Uracis-14e:trans-14e
(77:23)
trans-15a:cis-15a:trans-15g:cis-15g
(4:2:20:74 c)
Inseparable (80%)
fI-Uracis-14f:trans-14fn.a.c
gDihydro-Uracis-14g:trans-14g
(86:14)
trans-15g:cis-15g
(81:19)
trans-15g – 4.0 a
cis-15g + trans-15g – 47 b
cis-15g – 23 a
hTheophyllinecis-14htrans-15htrans-15h – 41 a
cis-14h:trans-14h
(77:23)
trans-15h:cis-15h
(80:20)
trans-15h – 34 a
cis-15h + trans-15h – 10 b
cis-15h – 26 a
iAdecis-14i:trans-14i
(85:15)
trans-15i:cis-15i
87:13
trans-15i – 28 a
cis-15i + trans-15i – 7.3 b
cis-14i:trans-14i
(42:58)
trans-15i:cis-15i
46:54
cis-15i + trans-15i – 55 b
a Yield of a pure diastereoisomer; b yield of a pure mixture of diastereoisomers; c not applied: since the dehalogenation of 5-halogenated uracil derivatives 14c, 14d and 14e was observed, the hydrogenation of 5-iodouracil derivatives cis-14f/trans-14f was not performed.
Table 3. Inhibitory effect of isoxazolidines 14 and γ-lactams 15 against the proliferation of cancerous cells.
Table 3. Inhibitory effect of isoxazolidines 14 and γ-lactams 15 against the proliferation of cancerous cells.
BaseCompoundIC50 (µM)
hTERT RPE-1Capan-1Hap1HCT-116NCI-H460
Retina (Non Cancerous)Pancreatic AdenocarcinomaChronic Myeloid LeukemiaColorectal CarcinomaLung Carcinoma
Uracis-14a>10054.8>100>100>100
Uratrans-14a>10062.6>100>10090.3
Thycis-14b>10055.263.0>10060.3
Thytrans-14b>10051.3>100>100>100
F-Uracis-14c>100>100>100>100>100
F-Uratrans-14c>10049.0>100>100>100
Br-Uracis-14d>10095.0>10086.462.3
Br-Uratrans-14d>10067.8>100>10076.1
Cl-Uracis-14e>10051.244.226.748.8
Cl-Uratrans-14e>10038.542.743.541.9
I-Uracis-14f97.142.040.957.944.6
I-Uratrans-14f>10041.447.053.541.1
dihydro-Uratrans-14g>10038.358.161.048.6
Theocis-14h>10062.245.149.033.2
Theotrans-14h>10043.253.660.951.2
Adecis-14i:trans-14i
(99:1)
>10073.5>100>100>100
Adecis-14i:trans-14i
(36:64)
>10058.1>10068.485.7
Thycis-15b:trans-15b
(64:36)
>10058.0>100>10050.0
Thytrans-15b>10049.286.553.355.6
Br-Uracis-15d:trans-15d
(53:47)
>10052.149.990.049.4
Br-Uratrans-15d>10024.858.5>10042.1
Cl-Uracis-15e>10028.051.558.528.6
Cl-Uratrans-15e>10018.254.5>10047.7
dihydro-Uracis-15g>10041.356.3>10042.7
dihydro-Uratrans-15g>10046.749.4>10069.3
Theocis-15h>10021.550.243.542.5
Theotrans-15h>10047.2>10045.749.8
Adetrans-15i>10053.747.865.754.7
DocetaxelDocetaxel25.00.951.190.250.89
EtoposideEtoposide0.230.150.041.031.35
StauroporineStauroporine0.250.663.550.0911.50

Share and Cite

MDPI and ACS Style

Piotrowska, D.G.; Głowacka, I.E.; Schols, D.; Snoeck, R.; Andrei, G.; Gotkowska, J. Novel Isoxazolidine and γ-Lactam Analogues of Homonucleosides. Molecules 2019, 24, 4014. https://doi.org/10.3390/molecules24224014

AMA Style

Piotrowska DG, Głowacka IE, Schols D, Snoeck R, Andrei G, Gotkowska J. Novel Isoxazolidine and γ-Lactam Analogues of Homonucleosides. Molecules. 2019; 24(22):4014. https://doi.org/10.3390/molecules24224014

Chicago/Turabian Style

Piotrowska, Dorota G., Iwona E. Głowacka, Dominique Schols, Robert Snoeck, Graciela Andrei, and Joanna Gotkowska. 2019. "Novel Isoxazolidine and γ-Lactam Analogues of Homonucleosides" Molecules 24, no. 22: 4014. https://doi.org/10.3390/molecules24224014

Article Metrics

Back to TopTop