Next Article in Journal
A Novel Synthesis of the Oxazolidinone Antithrombotic Agent Rivaroxaban
Previous Article in Journal
Pharmacological Effects of “Jutsu” (Atractylodis rhizome and Atractylodis lanceae rhizome) on 1-(2,5-Dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-Induced Head Twitch Response in Mice (I)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of New Quinoxalines Containing an Oxirane Ring by the TDAE Strategy and in Vitro Evaluation in Neuroblastoma Cell Lines

1
Aix-Marseille Université, CNRS, Institut de Chimie Radicalaire ICR, UMR 7273, Laboratoire de Pharmaco-Chimie Radicalaire, Marseille 13385, France
2
Aix-Marseille Université, INSERM, CRO2, UMR_S911, Marseille 13385, France
3
AP-HM, Hôpital Timone, Pharmacie, Marseille 13005, France
*
Author to whom correspondence should be addressed.
Molecules 2014, 19(9), 14987-14998; https://doi.org/10.3390/molecules190914987
Submission received: 25 July 2014 / Revised: 9 September 2014 / Accepted: 11 September 2014 / Published: 18 September 2014
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Neuroblastoma is an aggressive pediatric malignancy with significant chemotherapeutic resistance. In order to obtain new compounds active on neuroblastoma cell lines, we investigated the reactivity of carbanion formed via TDAE in quinoxaline series. The new synthesized compounds were tested for their anti-proliferative activity on two neuroblastoma cell lines, and seven oxirane derivatives obtained interesting activities.

1. Introduction

Neuroblastoma is a neuroendocrine tumor that remains a major therapeutic challenge in pediatric oncology. Treating it requires aggressive multimodal therapy. As response rates to chemotherapy are low, surgery remains the only effective treatment. However, since many tumors have metastasized at the time of diagnosis, long-term survival rates for children with advanced disease are poor, and undesirable side effects frequently occur. Accordingly, there is a substantial need for new therapeutic options.
The quinoxaline derivatives show very interesting biological properties (anti-viral [1], anti-cancer [2], anti-leishmanial [3]) and are far from having revealed their full potential in medicinal chemistry [4]. Many drug candidates bearing quinoxaline core structures are in clinical trials in anti-viral [1], anti-cancer [2], anti-bacterial and CNS (central nervous system) therapeutic areas. Among them, XK469 and chloroquinoxaline sulfonamide (CQS) are known as anti-neoplastic quinoxaline topoisomerase II inhibitors [5,6,7] (see Scheme 1). XK469, or (±)-2-[4-(7-chloro-2-quinoxaliny)oxy] phenoxy propionic acid, is an analog of the herbicide, Assure®, synthesized by DuPont Company, which possesses anti-tumor activity.
Scheme 1. Structures of XK469 and chloroquinoxaline sulfonamide (CQS).
Scheme 1. Structures of XK469 and chloroquinoxaline sulfonamide (CQS).
Molecules 19 14987 g001
The quinoxaline anti-tumor agent, XK469, inhibited neuroblastoma tumor growth in a 14-year-old with relapsed neuroblastoma, who experienced disease stabilization for 14 months [8].
Tetrakis(dimethylamino)ethylene (TDAE) is a reducing agent, which reacts with halogenated derivatives to generate, under mild conditions, an anion via two sequential transfers of one electron [9,10,11,12]. We have shown that, from o- or p-nitrobenzyl chloride, TDAE can generate a nitrobenzyl carbanion, which is able to react with various electrophiles, such as aromatic aldehydes [13], ketones, α-keto-esters, α-ketolactams and diethyl ketomalonate [14,15,16].
In continuation of our research program centered on the design and synthesis of novel bioactive molecules [17,18,19,20,21], we decided to explore the anti-proliferative potential of quinoxaline compounds.

2. Results and Discussion

2.1. Chemistry

In the present study, 13 quinoxalines were evaluated against human neuroblastoma (SK-N-SH and IMR-32—SK-N-SH and IMR-32 are not acronyms but the appellations of cell lines) cell lines, 12 of which were prepared in our lab (clinical compound XK469 was purchased). Apart from molecules 8, 15, 16, all synthesized molecules bear an oxirane moiety. Compounds synthesized after molecules 3b and 4a were observed to have an interesting activity against SK-N-SH and IMR-32 cell lines. Thus, a structural homogeneous quinoxaline series was obtained, presenting a structural analogy with XK469.
The 2-(dibromomethyl)quinoxaline 2 was synthesized in one step from 2-methylquinoxaline 1, as previously described (Scheme 2) [22].
Scheme 2. Synthesis of 2-(dibromomethyl)quinoxaline 2.
Scheme 2. Synthesis of 2-(dibromomethyl)quinoxaline 2.
Molecules 19 14987 g002
Oxiranes 34ab resulted from the reaction of 2-(dibromomethyl)quinoxaline with aromatic aldehydes in the presence of TDAE (Scheme 3) [22].
Scheme 3. Reactivity of aromatic aldehydes in the presence of TDAE.
Scheme 3. Reactivity of aromatic aldehydes in the presence of TDAE.
Molecules 19 14987 g003
Our next step was to test whether the nature of the aldehyde changed the biological effect on neuroblastoma cell lines. Thus, we investigated the reaction of 2-(dibromomethyl)quinoxaline 2 with three equivalents of aldehydes 56 in the presence of TDAE at −20 °C for 1 h, followed by 2 h at room temperature. Only trans-oxirane 7 and unexpected 1-(quinoxalin-2-ylmethyl)-1H-pyrrole-2-carbaldehyde 8 were isolated (Scheme 4).
Scheme 4. Reactivity of heterocyclic aldehydes in the presence of TDAE.
Scheme 4. Reactivity of heterocyclic aldehydes in the presence of TDAE.
Molecules 19 14987 g004
The α-bromo carbanion formed by the action of the TDAE could react as a base with the 1H-pyrrole-2-carbaldehyde, leading to the corresponding pyrrolic anion and to the 2-bromomethyl-quinoxaline. The formation of Compound 8 could thus result from a nucleophilic substitution between these two species (Scheme 5).
To study the effect of steric hindrance, we investigated the reaction of 2-(dibromomethyl)quinoxaline 2 with two isatin derivatives, 910, in the presence of TDAE under classical TDAE conditions. These reactions led to a mixture of like/unlike-isomers of corresponding oxiranes 910ab in good yields, as shown in Scheme 6.
Scheme 5. Reactivity of 1H-pyrrole-2-carbaldehyde in the presence of TDAE.
Scheme 5. Reactivity of 1H-pyrrole-2-carbaldehyde in the presence of TDAE.
Molecules 19 14987 g005
Scheme 6. Reactivity of various isatin derivatives in the presence of TDAE.
Scheme 6. Reactivity of various isatin derivatives in the presence of TDAE.
Molecules 19 14987 g006
Diastereoisomers were separable, and their configuration was identified by NMR-analysis from the γ-left effect, as previously described [23]. Finally, the impact of the oxirane group on biological activity was studied. Thus, the reaction observed with Compound 6 was extended to other pyrrole derivatives (Scheme 7).
Scheme 7. Reactivity of various carbonyl pyrrole in the presence of TDAE.
Scheme 7. Reactivity of various carbonyl pyrrole in the presence of TDAE.
Molecules 19 14987 g007

2.2. In Vitro Anti-Proliferative Activity of Quinoxalines Derivatives against Cancer Cell Lines

Quinoxalines 3ab, 4ab, 7, 8, 9ab, 10ab, 15, 16 and clinical compound XK469 were evaluated for their in vitro anti-proliferative activity on a panel of SK-N-SH and IMR-32 cancer cell lines, using the conventional microculture tetrazolium reduction assay [24,25,26].
Results of these evaluations are summarized in Table 1.
Table 1. Anti-proliferative activity of quinoxaline derivatives.
Table 1. Anti-proliferative activity of quinoxaline derivatives.
StructureSK-N-SH Cell LineIMR-32 Cell Line
IC50 ± SD (μM)IC50 ± SD (μM)
XK469 Molecules 19 14987 i0014.6 ± 1.013.0 ± 2.9
3a
cis-isomer
Molecules 19 14987 i00293.0 ± 10.564.5 ± 9.7
3b
trans-isomer
Molecules 19 14987 i00216.0 ± 1.214.7 ± 1.4
4a
cis-isomer
Molecules 19 14987 i00332.5 ± 1.925.9 ± 2.4
4b
trans-isomer
Molecules 19 14987 i003> 10045.9 ± 7.2
7
trans-isomer
Molecules 19 14987 i0043.9 ± 0.25.0 ± 0.9
8 Molecules 19 14987 i005>100>100
9a
like-isomer
Molecules 19 14987 i00625.0 ± 3.115.4 ± 4.8
9b
unlike-isomer
Molecules 19 14987 i00727.2 ± 6.414.0 ± 2.8
10a
like-isomer
Molecules 19 14987 i00821.8 ± 1.57.3 ±1.6
10b
unlike-isomer
Molecules 19 14987 i00918.9 ± 4.19.7 ± 2.7
15 Molecules 19 14987 i010>100>100
16 Molecules 19 14987 i011>100>100
The IC50 (concentration that inhibits 50% of cell proliferation) values show clearly that the presence of an oxirane nucleus is necessary to obtain anti-proliferative activity on the two tested neuroblastoma cell lines. The reaction between the 2-(dibromomethyl)quinoxaline 2 and the pyrrole derivatives led to three compounds, 8, 15, 16, for which no activity was observed on either of the cell lines at the maximal tested dose. All oxirane derivatives displayed substantial anti-proliferative activity towards neuroblastoma cell lines. These results confirm that the oxirane group played a key role in the anti-proliferative activity [27,28,29,30].
Compound 7 demonstrated a better activity than XK469, used as the reference drug, on SK-N-SH cells (IC50 = 3.9 ± 0.2 μM vs. 4.6 ± 1.0 μM, respectively) and IMR-32 cells (IC50 = 5.0 ± 0.9 μM vs. 13.0 ± 2.9 μM, respectively).
This first study on only four couples of stereoisomers does not allow us to end on the importance of the stereochemistry on the activity. The synthesis and the evaluation of new couples of stereoisomers with an oxirane group can allow one to appreciate the importance of the stereochemistry.
This initial structure activity relationship (SAR) study will be pursued. The nature of the heterocyclic aldehyde and the use of azaheterocycles other than quinoxaline are currently under investigation.

3. Experimental Section

3.1. General

Melting points were determined on a Büchi B-540 and are uncorrected. Elemental analyses were carried out on an Interscience Flash EA 1112 series (Thermo Finnigan, San Jose, CA, USA) elemental analyzer at the Spectropole, Faculté des Sciences, site de Saint-Jérôme. Both 1H- and 13C-NMR spectra were determined on a Bruker Avance 200 spectrometer (operating at 200 MHz for 1H and 50 MHz for 13C). 1H and 13C-NMR shifts (δ) were reported in parts per million (ppm) with respect to CDCl3 7.26 ppm for 1H and 77.0 ppm for 13C and DMSO-d6 2.50 for 1H and 39.7 ppm for 13C. Multiplicities were represented by s (singlet), d (doublet), t (triplet), q (quartet) and m (multiplet). The adsorbent used for column chromatography was: silica gel 60 (Merck, Darmstadt, Germany, 230–400 mesh). Thin-layer chromatography was performed with Merck 60F-254 silica gel (0.25 mm layer thickness) in an appropriate solvent.
The general procedure for the reaction of 2-(dibromomethyl)-quinoxaline 2 with various aldehydes 36 or isatin and pyrrolic derivatives 913, using TDAE:
To a two-necked flask equipped with a silica-gel drying tube and a nitrogen inlet was added, under nitrogen at –20 °C, 10 mL of anhydrous DMF solution of 2-(dibromomethyl)-quinoxaline 2 (0.45 g, 1.5 mmol) and various aldehydes 36 or isatin and pyrrolic derivatives 913, (4.5 mmol, 3 equivalents). The solution was stirred and maintained at this temperature for 30 min, and then the TDAE (0.3 g, 1.5 mmol) was added dropwise (via a syringe). A red color immediately developed with the formation of a white fine precipitate. The solution was vigorously stirred at −20 °C for 1 h and then warmed up to room temperature for 2 h. After this time, TLC analysis (dichloromethane) clearly showed that Compound 2 was totally consumed. The orange-red turbid solution was filtered and hydrolyzed with 80 mL of H2O. The aqueous solution was extracted with chloroform (3 × 40 mL), and the combined organic layers were washed with H2O (3 × 40 mL) and dried over MgSO4. Evaporation of the solvent left an orange solid as a crude product. Purification by silica gel chromatography (5/5 dichloromethane/ethyl acetate) and recrystallization from ethanol gave the corresponding derivatives.
4-(3-quinolanin-2-yl-oxiranyl)benzonitrile 4a, pink solid, m.p. 161.5 °C; 1H-NMR (CDCl3): δ 4.24 (d, J = 4.3 Hz, 1H, CH); 4.33 (d, J = 4.3 Hz, 1H, CH); 7.52 (d, J = 8.6 Hz, 2H, 2×CH); 7.72 (d, J = 8.6 Hz, 2H, 2×CH); 7.84 (m, 2H, 2×CH); 8.14 (m, 2H, 2×CH); 8.88 (s, 1H, CH); 13C-NMR (CDCl3): δ 59.2 (2×CH), 112.2 (C), 118.2 (C); 127.4 (2×CH); 128.8 (CH), 129.4 (CH); 130.2 (CH); 130.6 (CH); 132.0 (2×CH); 138.5 (C); 141.5 (C); 142.0 (CH); 142.8 (C); 151.0 (C). Elemental analysis calculated: C, 74.71; H, 4.06; N, 15.38; elemental analysis found: C, 74.47; H, 4.22; N, 15.05.
4-(3-quinolanin-2-yl-oxiranyl)benzonitrile 4b, pink solid, m.p. 178.5 °C 1H-NMR (CDCl3): δ 4.61 (d, J = 1.9 Hz, 1H, CH); 4.71 (d, J = 1.9 Hz, 1H, CH); 7.48 (m, 2H, 2×CH); 7.74 (m, 4H, 4×CH); 8.01 (m, 2H, 2×CH); 8.66 (s, 1H, CH); 13C-NMR (CDCl3): δ 60.8 (CH); 62.1 (CH); 112.7 (C); 118.4 (C); 126.4 (2×CH); 129.2 (CH); 129.5 (CH); 130.4 (CH); 130.8 (CH); 132.6 (2×CH); 141.3 (C); 141.9 (C); 142.1 (CH); 142.7 (C); 150.3 (C). Elemental analysis calculated: C, 74.71; H, 4.06; N, 15.38; elemental analysis found: C, 74.29; H, 4.23; N, 15.07.
2-[3-(5-Nitrofuran-2-yl)oxiran-2-yl]quinoxaline 7, maroon solid, m.p. 171 °C 1H-NMR (CDCl3): δ 4.44 (d, J = 1.9 Hz, 1H, CH); 4.74 (d, J = 1.9 Hz, 1H, CH); 6.77 (d, J = 3.7 Hz, 1H, CH); 7.34 (d, J = 3.7 Hz, 1H, CH); 7.81 (m, 2H, 2×CH); 8.12 (m, 2H, 2×CH); 8.89 (s, 1H, CH); 13C-NMR (CDCl3): δ 54.0 (CH); 59.4 (CH); 112.2 (CH); 112.5 (CH); 129.2 (CH); 129.5 (CH); 130.6 (CH); 130.9 (CH); 141.9 (CH); 142.4 (C); 142.8 (C);149.1 (C); 152.5 (C). Elemental analysis calculated: C, 59.37; H, 3.20; N, 14.84; elemental analysis found: C, 59.75; H, 3.23; N, 14.52.
1-(quinoxalin-2-ylmethyl)-1H-pyrrole-2-carbaldehyde 8, black solid, m.p. 275 °C 1H-NMR (DMSO): δ 5.88 (s, 2H, CH2); 6.32 (m, 1H, CH); 7.01 (m, 1H, CH); 7.18 (s, 1H, CH); 7.72 (m, 2H, 2×CH); 8.04 (m, 2H, 2×CH); 8.63 (s, 1H, CH); 9.55 (s, 1H, CH); 13C-NMR (DMSO): δ 54.1 (CH2); 110.7 (CH); 125.1 (CH); 129.1 (CH); 129.2 (CH); 130.0 (C); 130.2 (CH); 131.4 (C); 132.0 (CH); 141.7 (C); 142.0 (C); 144.0 (CH); 151.8 (C); 179.7 (CH=O). Elemental analysis calculated: C, 70.87; H, 4.67; N, 17.71; elemental analysis found: C, 70.90; H, 4.81; N, 17.71.
3'-(quinoxalin-2-yl)spiro[indoline-3,2'-oxiran]-2-on 9a, orange powder, m.p. 197 °C 1H-NMR (DMSO): δ 5.32 (s, 1H); 6.94 (m, 1H); 7.10 (m, 1H); 7.50 (m, 3H) 7.84 (m, 2H); 8.00 (m, 1H); 9.15 (s, 1H); 10.82 (s, 1H); 13C-NMR (DMSO): δ 62.21 (C); 64.53 (CH); 112.67 (CH); 122.62 (C); 123.24 (CH); 125.17 (CH); 129.04 (CH); 129.50 (CH); 130.88 (CH); 130.98 (CH); 138.85 (CH); 141.09 (C); 141.73 (C); 143.80 (C); 144.72 (CH); 151.19 (C); 171.07 (C=O). Elemental analysis calculated: C, 70.58; H, 3.83; N, 14.52; elemental analysis found: C, 70.58; H, 4.01; N, 14.24.
3'-(quinoxalin-2-yl)spiro[indoline-3,2'-oxiran]-2-one 9b, orange powder, m.p. 212 °C 1H-NMR (DMSO): δ 5.00 (s, 1H); 6.64 (m, 2H); 6.92 (m, 1H); 7.21 (m, 1H); 7.93 (m, 2H); 8.14 (m, 2H); 9.19 (s, 1H); 11.00 (s, 1H); 13C-NMR (DMSO): δ 62.65 (C); 63.75 (CH); 111.38 (CH); 120.53 (C); 122.25 (CH); 123.98 (CH); 129.39 (CH); 129.63 (CH); 131.25 (CH) 131.28 (CH); 131.55 (CH); 141.28 (C); 142.02 (C); 144.52 (C); 145.19 (CH); 149.32 (C); 171.91 (C=O). Elemental analysis calculated: C, 70.58; H, 3.83; N, 14.52; elemental analysis found: C, 69.94; H, 3.99; N, 14.12.
5-nitro-3'-(quinoxalin-2-yl)spiro[indoline-3,2'-oxiran]-2-one 10a, beige powder, m.p. 250 °C 1H-NMR (DMSO): δ 5.63 (s, 1H); 7.13 (m, 1H); 7.15 (m, 2H); 8.01 (m, 1H); 8.11 (m, 1H); 8.35 (m, 2H); 9.16 (s, 1H); 11.50 (s, 1H); 13C-NMR (DMSO): δ 61.61 (C); 64.66 (CH); 111.52 (CH); 119.63 (CH); 123.87 (C); 128.19 (C); 129.11 (CH); 129.52 (CH); 131.10 (CH); 141.13 (C); 141.82 (C); 143.17 (C); 144.64 (CH); 148.75 (CH); 149.92 (CH); 171.53 (C=O). Elemental analysis calculated: C, 61.08; H, 3.02; N, 16.76; elemental analysis found: C, 61.68; H, 3.06; N, 16.76.
5-nitro-3'-(quinoxalin-2-yl)spiro[indoline-3,2'-oxiran]-2-one 10b, beige powder, m.p. 230 °C 1H-NMR (DMSO): δ 5.13 (s, 1H); 7.08 (m, 1H); 7.95 (m, 2H); 8.20 (m, 4H); 9.22 (s, 1H); 11.63 (s, 1H); 13C-NMR (DMSO): δ 61.92 (C); 64.12 (CH); 111.21 (CH); 120.88 (CH); 121.95 (C); 127.82 (C); 129.21 (CH); 129.67 (CH); 131.37 (CH); 140.99 (C); 142.20 (C); 142.47 (C); 146.77 (CH); 148.98 (CH); 150.79 (CH); 156.47 (C-NO2); 172.58 (C=O). Elemental analysis calculated: C, 61.08; H, 3.02; N, 16.76; elemental analysis found: C, 61.08; H, 3.54; N, 16.54.
1-(quinoxalin-2-ylmethyl)-1H-indole-3-carbaldehyde 15, black powder, m.p. 116 °C 1H-NMR (DMSO): δ 5.95 (s, 2H); 7.24 (s, 2H); 7.60 (m, 1H); 7.82 (m, 2H); 7.94 (m, 2H); 8.10 (m, 1H); 8.54 (s, 1H); 8.97 (s, 1H); 9.97 (s, 1H); 13C-NMR (DMSO): δ 50.45 (CH2); 111.71 (CH); 118.20 (C); 121.61 (CH); 123.15 (CH); 124.26 (CH); 125.24 (C); 129.31 (CH) 129.43 (CH); 130.79 (CH); 131.19 (CH); 137.77 (C); 141.52 (C); 141.81 (C); 142.15 (CH); 145.25 (CH); 152.21 (C); 185.41 (C=O). Elemental analysis calculated: C, 75.25; H, 4.56; N, 14.63; elemental analysis found: C, 74.98; H, 4.68; N, 14.62.
1-(1-(quinoxalin-2-ylmethyl)-1H-pyrrol-2-yl)ethanone 16, black powder, m.p. 72 °C 1H-NMR (DMSO): δ 2.28 (s, 3H); 5.83 (s, 2H); 7.20 (m, 1H); 7.41 (m, 1H); 7.81 (m, 2H); 7.95 (m, 1H); 8.04 (m, 1H); 8.50 (s, 1H); 13C-NMR (DMSO): δ 27.27 (CH3); 52.79 (CH2); 109.11 (CH); 121.43 (CH); 129.13 (CH); 129.35 (CH); 130.24 (CH); 130.27 (C); 130.91 (CH) ; 132.79 (CH); 141.45 (C); 141.48 (C); 144.57 (CH); 154.12 (C); 188.22 (C=O). Elemental analysis calculated: C, 71.70; H, 5.21; N, 16.72; elemental analysis found: C, 71.14; H, 5.17; N, 16.58.

3.2. In Vitro Biological Evaluation

Drugs: Stock solutions of quinoxaline were prepared in dimethylsulfoxide (Sigma). Stock solutions were aliquoted and stored at −20 °C. For culture and experiments in living cells, drugs were freshly diluted at appropriate concentrations in culture medium.
Cell culture: Neuroblastoma cancer cells, namely SK-N-SH and IMR-32 cells, were routinely maintained in standard RPMI 1640 (l-glutamine +) culture medium (Invitrogen) supplemented with 10% fetal bovine serum (Lonza) and 1% penicillin and streptomycin (Invitrogen). All cells were routinely maintained at 37 °C and 5% CO2.
Growth experiments: Exponentially growing cells (37,500 cells/cm²) were seeded in 96-well plates for 24 h for SK-N-SH cells and for 72 h for IMR-32 cells and then incubated with the drugs for 72 h. The number of viable cells was estimated using the colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma) assay according to our previous work [17]. After drug treatment, the medium was replaced by fresh medium containing MTT (0.5 mg/mL), and cells were incubated at 37 °C for 2 h. Then, the MTT solution was removed, and DMSO was used to dilute the formazan crystals formed in the surviving cells. Finally, absorbance was measured at 600 nm with a Multiskan (Ascent) plate reader. Inhibiting concentrations were graphically determined. IC50 is defined as the concentration that inhibits 50% of cell proliferation. At least three independent experiments (in triplicate) were performed, and data were expressed as the mean ± SD.

4. Conclusion

Having previously demonstrated how the TDAE strategy can be used to obtain a series of highly functionalized quinoxalinic oxiranes, we present herein an original reactivity initiated by TDAE with compounds possessing labile hydrogen. Biological evaluation of these synthesized compounds revealed promising anti-proliferative activity toward SK-N-SH and IMR-32 cell lines for most of the compounds bearing an oxirane group. Biological results showed that Compound 7 is more active than XK-469, a quinoxaline derivative involved in clinical trials, confirming that the oxirane group plays a key role in the anti-proliferative activity.
Further investigation on SAR and the mechanism of the anti-proliferative activity of these compounds will be carried out in next works. Moreover, due to promising results observed with Compound 7, it will be tested as potential anti-tumor drug on other cell lines.

Acknowledgments

This work was supported by the Centre National de la Recherche Scientifique. We express our thanks to Vincent Remusat for 1H and 13C NMR spectra recording.

Author Contributions

M.M., T.T. and P.V. conceived of and designed the study. M.M. and O.K. designed the experiments and interpreted the results. F.C and M.-A.E. conducted and interpreted the biological tests.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harmenberg, J.; Akesson-Johansson, A.; Graslund, A.; Malmfors, T.; Bergman, J.; Wahren, B.; Akerfeldt, S.; Lundblad, L.; Cox, S. The mechanism of action of the anti-herpes virus compound 2,3-dimethyl-6(2-dimethylaminoethyl)-6H-indolo-(2,3-b)quinoxaline. Antiviral Res. 1991, 15, 193–204. [Google Scholar]
  2. Naylor, M.A.; Stephen, M.A.; Nolan, J.; Sutton, B.; Tocher, J.H.; Fielden, E.M.; Adams, J.E.; Strafford, I. Heterocyclic mono-N-oxides with potential applications as bioreductive anti-tumour drugs: Part 1. 8-Alkylamino-substituted phenylimidazo [1,2-a] quinoxalines. Anticancer Drug Des. 1993, 8, 439–461. [Google Scholar]
  3. Hui, X.; Desrivot, J.; Bories, C.; Loiseau, P.M.; Franck, X.; Hocquemiller, R.; Figadere, B. Synthesis and antiprotozoal activity of some new synthetic substituted quinoxalines. Bioorg. Med. Chem. Lett. 2006, 16, 815–820. [Google Scholar]
  4. Cheeseman, G.W.H.; Cookson, R.F. The Chemistry of Heterocyclic Compounds; Weissberger, A., Taylor, E.C., Eds.; John Wiley and Sons: New York, NY, USA, 1979; Volume 35, p. 1. [Google Scholar]
  5. Corbett, T.H.; Lorusso, P.M.; Demchick, L.; Simpson, C.; Pugh, S.; White, K.; Kushner, J.; Polin, L.; Meyer, J.; Czarnecki, J.; et al. Preclinical antitumor efficacy of analogs of XK469: Sodium-(2-[4-(7-chloro-2-quinoxalinyloxy)phenoxy]propionate. Investig. New Drugs 1998, 16, 129–139. [Google Scholar]
  6. Lorusso, P.M.; Parchment, R.; Demchik, L.; Knight, J.; Polin, L.; Dzubow, J.; Behrens, C.; Harrison, B.; Trainor, G.; Corbett, T.H. Preclinical antitumor activity of XK469 (NSC 656889). Investig. New Drugs 1998, 16, 287–296. [Google Scholar]
  7. Rigas, J.R.; Miller, V.A.; Tong, W.P.; Roistacher, N.; Kris, M.G.; Orazem, J.P.; Young, C.W.; Warrell, R.P., Jr. Clinical and pharmacology study of chloroquinoxaline sulfonamide given on a weekly schedule. Cancer Chemother. Pharmacol. 1995, 35, 483–488. [Google Scholar] [CrossRef]
  8. Kakodkar, N.C.; Peddinti, R.; Kletzel, M.; Tian, Y.; Guerrero, L.J.; Undevia, S.D.; Geary, D.; Chlenski, A.; Yang, Q.; Salwen, H.R.; et al. The quinoxaline anti-tumor agent (R+)XK469 inhibits neuroblastoma tumor growth. Pediatr. Blood Cancer 2011, 56, 164–467. [Google Scholar]
  9. Takechi, N.; Ait-Mohand, S.; Médebielle, M.; Dolbier, W.R., Jr. Nucleophilic trifluoromethylation of acyl chlorides using the trifluoromethyl iodide/TDAE reagent. Tetrahedron Lett. 2002, 43, 4317–4319. [Google Scholar]
  10. Ait-Mohand, S.; Takechi, N.; Médebielle, M.; Dolbier, W.R., Jr. Nucleophilic trifluoromethylation using trifluoromethyl iodide. A new and simple alternative for the trifluoromethylation of aldehydes and ketones. Org. Lett. 2001, 3, 4271–4273. [Google Scholar]
  11. Médebielle, M.; Keirouz, R.; Okada, E.; Ashida, T. Tetrakis(dimethylamino)ethylene (TDAE) mediated addition of heterocyclic difluoromethyl anions to heteroaryl aldehydes. A facile synthetic method fornew gem-difluorinated alcohols derived from 4-bromo-1-naphthylamine and 8-quinolylamine. Synlett 2001, 2001, 821–823. [Google Scholar]
  12. Dolbier, W.R., Jr.; Médebielle, M.; Ait-Mohand, S. Single electron transfer approaches to the practical synthesis of aromaticand heterocyclic-CF2H derivatives. Tetrahedron Lett. 2001, 42, 4811–4814. [Google Scholar]
  13. Giuglio-Tonolo, G.; Terme, T.; Médebielle, M.; Vanelle, P. Original reaction of p-nitrobenzylchloride with aldehydes using tetrakis(dimethylamino)ethylene (TDAE). Tetrahedron Lett. 2003, 44, 6433–6435. [Google Scholar]
  14. Giuglio-Tonolo, G.; Terme, T.; Médebielle, M.; Vanelle, P. Nitrobenzylation of α-carbonyl ester derivatives using TDAE approach. Tetrahedron Lett. 2004, 45, 5121–5124. [Google Scholar]
  15. Amiri-Attou, O.; Terme, T.; Vanelle, P. Functionalization of 6-nitrobenzo[1,3]dioxole with carbonyl compounds via TDAE methodology. Molecules 2005, 10, 545–551. [Google Scholar]
  16. Montana, M.; Crozet, M.D.; Castera-Ducros, C.; Terme, T.; Vanelle, P. Rapid synthesis of new azaheterocyclic hydroxy-malonate derivatives using TDAE approach. Heterocycles 2008, 75, 925–932. [Google Scholar]
  17. Crozet, M.P.; Giraud, L.; Sabuco, J.F.; Vanelle, P.; Barreau, M. SRN1 reactions of a tetra-substituted-1,4-benzoquinone. Tetrahedron Lett. 1991, 32, 4125–4128. [Google Scholar]
  18. Delmas, F.; Gasquet, M.; Timon-David, P.; Madadi, N.; Vanelle, P.; Vaille, A.; Maldonado, J. Synthesis and in vitro anti-protozoan activity of new 5-nitrothiophene oxime ether derivatives. Eur. J. Med. Chem. 1993, 28, 23–27. [Google Scholar]
  19. Crozet, M.P.; Gellis, A.; Pasquier, C.; Vanelle, P.; Aune, J.P. Electron transfer reactivity in 5-nitrouracile series. Tetrahedron Lett. 1995, 36, 525–528. [Google Scholar]
  20. Curti, C.; Laget, M.; Ortiz Carle, A.; Gellis, A.; Vanelle, P. Rapid synthesis of sulfone derivatives as potential anti-infectious agents. Eur. J. Med. Chem. 2007, 42, 880–884. [Google Scholar]
  21. Crozet, M.D.; Botta, C.; Gasquet, M.; Curti, C.; Remusat, V.; Hutter, S.; Chapelle, O.; Azas, N.; de Méo, M.; Vanelle, P. Lowering of 5-nitroimidazole’s mutagenicity: Towards optimal antiparasitic pharmacophore. Eur. J. Med. Chem. 2009, 44, 653–659. [Google Scholar]
  22. Montana, M.; Terme, T.; Vanelle, P. Original synthesis of oxiranes via TDAE methodology: Reaction of 2,2-dibromomethylquinoxaline with aromatic aldehydes. Tetrahedron Lett. 2005, 46, 8373–8376. [Google Scholar]
  23. Montana, M.; Terme, T.; Vanelle, P. Original synthesis of alpha-chloroketones in azaheterocyclic series using TDAE approach. Tetrahedron Lett. 2006, 47, 6573–6576. [Google Scholar]
  24. Alley, M.C.; Scudiero, D.A.; Monks, A.; Hursey, M.L.; Czerwinski, M.J.; Fine, D.L.; Abbott, B.J.; Mayo, J.G.; Shoemaker, R.H.; Boyd, M.R. Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay. Cancer Res. 1988, 48, 589–601. [Google Scholar]
  25. Van de Loosdrecht, A.A.; Beelen, R.H.; Ossenkoppele, G.J.; Broekhoven, M.G.; Langenhuijsen, M.M. A tetrazolium-based colorimetric MTT assay to quantitate human monocyte mediated cytotoxicity against leukemic cells from cell lines and patients with acute myeloid leukemia. J. Immunol. Methods 1994, 174, 311–320. [Google Scholar]
  26. Estève, M.A.; Carré, M.; Bourgarel-Rey, V.; Kruczynski, A.; Raspaglio, G.; Ferlini, C.; Braguer, D. Bcl-2 down-regulation and tubulin subtype composition are involved in resistance of ovarian cancer cells to vinflunine. Mol. Cancer Ther. 2006, 5, 2824–2833. [Google Scholar]
  27. House, H.O. Modern Synthetic Reactions, 2nd ed.; W.A. Benjamin, Inc.: New York, NY, USA, 1972; p. 459. [Google Scholar]
  28. Eilon, G.F.; Gu, J.; Slater, L.M.; Hara, K.; Jacobs, J.W. Tumor apoptosis induced by epoxide-containing piperazines, a new class of anti-cancer agents. Cancer Chemother. Pharmacol. 2000, 45, 183–191. [Google Scholar]
  29. Han, H.; Zhao, Y.; Cuthbertson, T.; Hartman, R.F.; Rose, S.D. Cell cycle arrest and apoptosis induction by an anticancer chalcone epoxide. Arch. Pharm. 2010, 343, 429–439. [Google Scholar]
  30. Gheeya, J.; Johansson, P.; Cgen, Q.R.; Dexheimer, T.; Metaféria, B.; Song, Y.K.; Wei, J.S.; He, J.; Pommier, Y.; Khan, J. Expression profiling identifies epoxy anthraquinone derivative as a DNA topoisomerase inhibitor. Cancer Lett. 2010, 293, 124–131. [Google Scholar]
  • Sample Availability: Samples of the compounds are available from authors.

Share and Cite

MDPI and ACS Style

Montana, M.; Correard, F.; Khoumeri, O.; Esteve, M.-A.; Terme, T.; Vanelle, P. Synthesis of New Quinoxalines Containing an Oxirane Ring by the TDAE Strategy and in Vitro Evaluation in Neuroblastoma Cell Lines. Molecules 2014, 19, 14987-14998. https://doi.org/10.3390/molecules190914987

AMA Style

Montana M, Correard F, Khoumeri O, Esteve M-A, Terme T, Vanelle P. Synthesis of New Quinoxalines Containing an Oxirane Ring by the TDAE Strategy and in Vitro Evaluation in Neuroblastoma Cell Lines. Molecules. 2014; 19(9):14987-14998. https://doi.org/10.3390/molecules190914987

Chicago/Turabian Style

Montana, Marc, Florian Correard, Omar Khoumeri, Marie-Anne Esteve, Thierry Terme, and Patrice Vanelle. 2014. "Synthesis of New Quinoxalines Containing an Oxirane Ring by the TDAE Strategy and in Vitro Evaluation in Neuroblastoma Cell Lines" Molecules 19, no. 9: 14987-14998. https://doi.org/10.3390/molecules190914987

Article Metrics

Back to TopTop