Next Article in Journal
Theoretical Investigation on Nearsightedness of Finite Model and Molecular Systems Based on Linear Response Function Analysis
Previous Article in Journal
Protoberberine Isoquinoline Alkaloids from Arcangelisia gusanlung
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of the New Ring System Bispyrido[4',3':4,5]pyrrolo [1,2-a:1',2'-d]pyrazine and Its Deaza Analogue

Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
*
Author to whom correspondence should be addressed.
Molecules 2014, 19(9), 13342-13357; https://doi.org/10.3390/molecules190913342
Submission received: 24 July 2014 / Revised: 18 August 2014 / Accepted: 20 August 2014 / Published: 29 August 2014
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Derivatives of the new ring systems bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione and its deaza analogue pyrido[4'',3'':4',5']pyrrolo-[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione were conveniently synthesized through a four-step sequence. Symmetrical derivatives of the former ring system were obtained through self condensation. On the other hand, condensation of 6-azaindole carboxylic acid with indole 2-carboxylic acid afforded the deaza analogue ring system. Derivatives of the title ring system were tested by the National Cancer Institute (Bethesda, MD, USA) and four of them exhibited modest activity against MCF7 (a breast cancer cell line) and/or UO-31 (a renal cancer cell line).

1. Introduction

Piperazine-2,5-diones represent a very interesting class of compounds because this heterocyclic system is found in many unique natural products [1]. In recent years there has been a growing awareness of the diversity and biological roles played by many diketopiperazines among the over one-hundred found in Nature. Many derivatives have antiviral (e.g., the gliotoxins and sporidesmins), phytotoxic (e.g., cyclo(Pro-Tyr)) and antibiotic (e.g., bicyclomycin) properties, whereas other compounds show antineoplastic activity, in particular phenylahistin (1, Figure 1), a fungal metabolite isolated from culture broths of Aspergillus ustus NFC-F038, which is a result of a condensation between l-phenylalanine and an isoprenylated dehydrohistidine residue with a quaternary carbon at C-5 of the imidazole ring [2].
Figure 1. Chemical structures of diketopiperazine derivatives 16.
Figure 1. Chemical structures of diketopiperazine derivatives 16.
Molecules 19 13342 g001
It is a colchicine-like microtubule binding agent endowed with cytotoxic activity against a wide variety of tumor cell lines [2,3,4], since it is able to competitively inhibit the binding site of colchicine to tubuline [3]. Phenylahistin derivatives were synthesized [5] with the aim of finding new antineoplastic derivatives, but also to understand the structural features necessary for the anti-microtubule activity. One of the most interesting compounds was revealed to be plinabulin (2, Figure 1) [6] a potent microtubule-targeting agent; it showed cytotoxic activity (IC50 = 15 nM) against human colon adenocarcinoma HT-29 cell line and it is currently in phase II clinical trials [7]. SAR studies revealed that the hydrogen bond between N8-H and N3 is crucial, allowing the formation of a rigid uniplanar pseudo-three-ring structure necessary for the binding to the microtubules.
Considering also that some properly decorated 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-diones 3 that are indolo-diketopiperazines showed cytotoxic activity in the µM range against L1210 cell line [8,9,10] and, in particular, that 2,9-dimethoxy derivative gave complete inhibition of erythrocyte differentiation, whether spontaneous or induced by haemin, in leukemia K562 cell line at 50 µM, we decided to further explore the biological potential of these compounds. Considering the experience acquired in the course of our research on polycyclic nitrogen systems bearing pyrrole [11,12,13], indole [14,15,16,17,18], isoindole [19,20,21,22] and indazole [23] moieties with antitumor activity, we have decided to synthetize diaza- and aza-analogues of the ring system 3 bearing two (compounds 4, 5) or one (compound 6) nitrogen atoms in the aromatic moiety in order to verify the antineoplastic properties of this new heterocyclic ring system.
Considering that the new compounds have the diketopiperazine core, capable of a colchicine-like microtubule binding, molecular docking studies were performed in order to investigate the potential binding ability of compounds 46 on tubulin. For this purpose, all compounds were docked in two different tubulin crystal structures (PDB ID code: 1SA0 [24] and 3HKD [25]) that represent two potential binding mode for colchicine site ligands.
In the 1SA0 crystal structure, colchicine, a tubulin assembly inhibitor, is the co-crystallized ligand and its binding site is located at the α,β interface of tubulin subunits [24]. In the crystal structure 3HKD, TN-16, a pyrrolidine-2,4-dione derivative, is the co-crystallized ligand. It inhibits microtubule assembly by competing with colchicine for tubulin binding [25,26]. The TN-16 binding pocket is located on the interface between the α and β subunits of the tubulin dimer and slightly extended out of the β subunit [25,27]. The X-ray crystal structures were prepared using Protein Preparation Wizard. Docking was carried out using Glide software SP mode default parameters [28].
An evaluation of the docking score results indicated that compounds 46 showed the best Glide docking score values in 3HKD (Glide score values between −9.739 and −8.927), compared to those obtained in the 1SA0 structure (Glide score values between −6.888 and −4.832) in which they did not show a good superimposition to colchicine. The only exception was for compound 4d, that was not docked by Glide in 3HKD (Table 1).
Table 1. Derivatives 4ad, 5ae and 6ad docking scores for 3HKD and 1SA0.
Table 1. Derivatives 4ad, 5ae and 6ad docking scores for 3HKD and 1SA0.
Compound3HKD1SA0
4a−8.927−6.643
4b−9.562−6.675
4c−9.354−6.007
4dnd−6.455
5a−9.289−6.661
5b−9.641−6.700
5c−9.203−4.832
5d−9.739−6.477
5e−9.653−6.122
6a−9.299−6.705
6b−9.648−6.150
6c−9.690−6.380
6d−9.718−6.888
nd: Not determined.
Analyzing the binding mode of the planned compounds in 3HKD, they showed H-bond interactions between the Glu 200 residue and one of the two carbonyl groups, interacting with the binding site in a way similar to the native ligand TN-16 (Figure 2). Although all compounds showed similar docking score values (Table 1), unsubstituted compounds 4a and 6a showed lower docking score values than the corresponding substituted derivatives. Generally the presence of a methoxy group in one of the two indole or aza-indole moieties seems to stabilize the tubulin-ligand complex through hydrophobic interactions with the Val 238 residue. On the basis of the docking studies we planned the synthesis of derivatives 46 in order to verify whether they were endowed with interesting biological properties.
Figure 2. Wall eyed superimposition of compounds 4ad, 5ae and 6ad with TN-16 (red).
Figure 2. Wall eyed superimposition of compounds 4ad, 5ae and 6ad with TN-16 (red).
Molecules 19 13342 g002

2. Results and Discussion

The key intermediates of the synthetic pathway for the pentacyclic new ring systems are 1H-pyrrolo[2,3-c]pyridine-2-carboxylic acids 10ad (Scheme 1). Commercially available pyridines 7a,b were reacted with diethyl oxalate using potassium ethoxide as the base to give the corresponding derivatives 8a,b in 50 and 45% yield, respectively [29]; pyridines 7c,d were synthetized from the suitable 2-chloro derivatives through nucleophilic substitution with sodium methoxide [30,31]. The so-obtained methoxypyridines were reacted with diethyl oxalate using t-BuOK as the base allowing the isolation of compounds 8c [30] and 8d in good yields (72%–75%). The latter compound was isolated as the enolic tautomer. Derivatives 8a,b were reduced with iron in saturated aqueous NH4Cl and THF to avoid halogen displacement. On the other hand compounds 8c,d were dissolved in EtOH and hydrogenated over 10% Pd on charcoal. After an appropriate work-up of the reaction mixture, derivatives 9ad were obtained in good yields (60%–85%). Carboxylic acid derivatives 10ad were obtained in excellent yields (71%–95%) through alkaline hydrolysis of the corresponding ethyl esters.
Derivatives 10ad were cyclized at room temperature in anhydrous THF with 4-dimethylaminopyridine (DMAP) and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI) as activating agents to give the new pentacyclic ring systems. Symmetrical derivatives 4ad were obtained by self-condensation of the corresponding 6-aza-indole carboxylic acids 10ad (Table 2).
Scheme 1. Synthesis of derivatives 4ad, 5ae and 6ad.
Scheme 1. Synthesis of derivatives 4ad, 5ae and 6ad.
Molecules 19 13342 g003
Reagents and conditions: (i) Diethyl oxalate, potassium ethoxide in diethyl ether and EtOH, rt, 15–72 h (8a,b) or t-BuOK, in diethyl ether and ethanol, reflux, 4 h then 24 h rt (8c,d); (ii) Fe, saturated aqueous NH4Cl, THF, EtOH, reflux, 2 h (9a,b) or H2/Pd-C, EtOH (9c,d); (iii) NaOH 2M, EtOH, reflux 1–2 h; (iv) DMAP, EDCI, THF, rt, 48 h; (v) Indole-2-carboxylic acid, DMAP, EDCI, THF, rt, 48 h.
Table 2. Derivatives 4ad, 5ae and 6ad.
Table 2. Derivatives 4ad, 5ae and 6ad.
CompoundR1R2R3R4Yields(%)
4aHHHH25
4bClHHCl30
4cOCH3HHOCH320
4dHOCH3OCH3H28
5aHHOCH3H40
5bOCH3HOCH3H55
5cHHHOCH342
5dClHOCH3H44
5eClHHOCH345
6aHH--33
6bClH--65
6cOCH3H--65
6dHOCH3--30
For the synthesis of the asymmetrical compounds 5ae, the activation of the proper acid 10ad with EDCI was followed by the addition of the suitable carboxylic acid and a further addition of EDCI in order to allow the intramolecular cyclization. In particular, 5ae were obtained from the condensation of 10a with 10d; 10c with 10d; 10a with 10c; 10b with 10d, and 10b with 10c, respectively (Table 2). The reaction mixture was particularly difficult to purify because of the presence not only of the asymmetrical desired derivatives 5ae, but also of 4%–6% of the symmetrical ones 4ad as byproducts of the reaction.
Moreover, through the synthetic pathway previously described it was possible to synthesize the deaza-analogues 6ad (Table 2), from the reaction between derivatives 10ad and commercially available indole-2-carboxylic acid (Scheme 1). Also in this case, not only the desired compounds 6ad were isolated from the reaction mixture, but also the symmetrical ones 4ad (3%–6%) as byproducts of the reaction together with 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-dione deriving from the indole-2-carboxylic acid self-condensation (7%–9%).
All the synthesized derivatives of the new ring system 6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione 4ad, 5ae and their deaza-analogues 6ad, were submitted to the National Cancer Institute (Bethesda, MD, USA) for screening. All derivatives were prescreened according to the NCI protocol at 10−5 M dose on the full panel of 60 human cancer cell lines derived from nine human cancer cell types that have been grouped in disease sub-panels including leukemia, non-small-cell lung, colon, central nervous system, melanoma, ovarian, renal, prostate and breast tumour cell lines.[32]
None of the prescreened derivatives were selected for the five dose screening (NCI-60 DTP Human Tumor Cell Line Screen), since only derivatives 5a and 6a, 6c and 6d showed moderate antineoplastic activity at micromolar concentrations. In particular derivative 5a exhibited modest activity against the UO-31 renal cancer sub-panel cell line with a growth inhibitory percentage of 47.0; unsubstituted deaza analogue 6a and 9-methoxy substituted derivative 6c were shown to be selective against the MCF7 breast cancer cell line with growth inhibitory percentages of 50.6 and 39.5, respectively. More interesting results were obtained from the 11-methoxy substituted compound 6d which was shown to be selective against both the UO-31 renal cancer sub-panel and the MCF7 breast cancer sub-panel cell lines with growth inhibitory percentages of 46.6 and 50.9, respectively.

3. Experimental Section

3.1. Chemistry

Anhydrous organic solvents were prepared by the appropriate procedures prior to use. The other organic solvents were reagent grade and used as received. Analytical TLC was performed on Merck Kieselgel 60-F254 plates. Column chromatography was performed with Merck silica gel 230–400 mesh ASTM or with a Büchi Sepacor prepacked cartridge system chromatography module.
All melting points were taken on a Buchi-Tottoli capillary apparatus and are uncorrected; IR spectra were determined in CHBr3, with a Shimadzu FT/IR 8400S spectrophotometer; 1H- and 13C-NMR spectra were measured in DMSO-d6 or CDCl3 solutions, at 200 and 50.3 MHz, respectively, using a Bruker Avance II series 200 MHz spectrometer. Elemental analyses (C, H, N) were within 0.4% of the theoretical values and were recorded with a VARIO EL III elemental analyzer.

3.1.1. General Procedure for the Preparation of 2-Methoxy-pyridines 7c,d

These compounds were synthesized according to the previously described procedure [30,31].
2-Methoxy-4-methyl 5-nitropyridine (7c). This compound was obtained in 95% yield. Analytical and spectroscopic data are in accordance to those reported in literature [30].
2-Methoxy-4-methyl 3-nitropyridine (7d). This compound was obtained in 80% yield. Analytical and spectroscopic data are in accordance to those reported in literature [31].

3.1.2. General Procedure for the Preparation of Ethyl-3-(nitropyridin-4-yl)-2-oxopropanoates 8a,b

These compounds were synthesized according to the previously described procedure [29].
Ethyl-3-(3-nitropyridin-4-yl)-2-oxopropanoate (8a). This compound was obtained in 50% yield. Analytical and spectroscopic data are in accordance to those reported in literature [29].
Ethyl-3-(2-chloro-5-nitropyridin-4-yl)-2-oxopropanoate (8b). This compound was obtained in 45% yield. Analytical and spectroscopic data are in accordance to those reported in literature [29].

3.1.3. General Procedure for the Preparation of Ethyl-3-(nitropyridin-4-yl)-2-oxopropanoates 8c,d

To a stirred solution of t-BuOK (2.4 mmol) in anhydrous EtOH (1 mL) and diethyl ether (10 mL) diethyl oxalate (2.4 mmol, 0.3 mL) was added under a nitrogen atmosphere. The reaction mixture was kept at room temperature for 15 min, then a solution of the suitable pyridine 7c,d (2.4 mmol) was added and the reaction mixture was refluxed for 4 h and stirred at room temperature 24 h. The orange residue thus obtained was shaken in diethyl ether, filtered off and air dried. Water (9.2 mL) was added and acetic acid was added until pH 4.0. The desired product was filtered off, and dried in the desiccator to afford the desired products as cream solids.
Ethyl 3-(2-methoxy-5-nitropyridin-4-yl)-2-oxopropanoate (8c). This compound was obtained in 72% yield. Analytical and spectroscopic data are in accordance to those reported in literature [30,33].
Ethyl 3-(2-methoxy-3-nitropyridin-4-yl)-2-oxopropanoate (8d). Title compound 8d was isolated as the enolic tautomer. Rf = 0.33 (CH2Cl2); mp 78.4–79.6 °C; yield 75%; IR: 3426 (OH), 1706 (CO) cm1; 1H-NMR (DMSO-d6) δ: 1.28 (3H, t, J = 6.0 Hz, CH3), 3.97 (3H, s, OCH3), 4.28 (2H, q, J = 6.0 Hz, CH2), 6.00 (1H, s, CH), 7.86 (1H, d, J = 6.00 Hz, H-5), 8.34 (1H, d, J = 6.0 Hz, H-6), 11.30 (1H, bs, OH). 13C-NMR (DMSO-d6) δ: 13.9 (q), 54.4 (q), 62.2 (t), 97.2 (d), 116.4 (d), 132.7 (s), 136.3 (s), 148.3 (d), 148.8 (s), 154.3 (s), 163.0 (s). Anal. Calcd for C11H12N2O6 (268.22): C, 49.26; H, 4.51; N, 10.44. Found: C, 49.21; H, 4.75; N, 10.16.

3.1.4. General Procedure for the Preparation of Ethyl 1H-pyrrolo[2,3-c]pyridine-2-carboxylates 9a,b

These compounds were synthesized according to the previously described procedure [29,34].
Ethyl 1H-pyrrolo[2,3-c]pyridine-2-carboxylate (9a). This compound was obtained in 60% yield. Analytical and spectroscopic data are in accordance to those reported in literature [29].
Ethyl 5-chloro-1H-pyrrolo[2,3-c]pyridine-2-carboxylate (9b). This compound was obtained in 60% yield. Analytical and spectroscopic data are in accordance to those reported in literature [34].

3.1.5. General Procedure for the Preparation of Ethyl 1H-pyrrolo[2,3-c]pyridine-2-carboxylates 9c,d

Derivatives 8c,d (2.9 mmol) were dissolved in EtOH (40 mL) and hydrogenated over 10% Pd on charcoal. The catalyst was removed by filtration under argon and the solvent was evaporated in vacuo.
Ethyl 5-methoxy-1H-pyrrolo[2,3-c]pyridine-2-carboxylate (9c). This compound was obtained in 85% yield. Analytical and spectroscopic data are in accordance to those reported in literature [33].
Ethyl 7-methoxy-1H-pyrrolo[2,3-c]pyridine-2-carboxylate (9d). Title compound 9d was purified by flash-chromatography using CH2Cl2/ethyl acetate 96:4. Rf = 0.63 (CH2Cl2/ethyl acetate 95:5) as a white powder; mp 134.1–135.0 °C; yield 75%; IR: 3435 (NH), 1708 (CO) cm1; 1H-NMR (CDCl3) δ: 1.41 (3H, t, J = 6.0 Hz, CH3), 4.09 (3H, s, OCH3), 4.43 (2H, q, J = 6.0 Hz, CH2), 7.13–7.17 (2H, m, H-3, H-4), 7.77 (1H, d, J = 6.0 Hz, H-5), 9.61 (1H, bs, NH). 13C-NMR (CDCl3) δ: 14.3 (q), 53.3 (q), 61.4 (t), 107.7 (d), 110.8 (d), 122.3 (s), 129.3 (s), 133.0 (s), 135.9 (d), 151.9 (s), 161.4 (s). Anal. Calcd for C11H12N2O3 (220.22): C, 59.99; H, 5.49; N, 12.72. Found: C, 60.14; H, 5.66; N, 12.57.

3.1.6. General Procedure for the Preparation of 1H-pyrrolo[2,3-c]pyridine-2-carboxylic Acids 10ad

To a stirred solution of 9ad (1.3 mmol) in EtOH (12 mL) 2M NaOH was added (1.7 mmol, 1.1 mL). The reaction mixture was heated under reflux for 1h (10a) or 2h (10b) and the solvent was evaporated. Water (10 mL) was added and acetic acid was added until pH 4.0. The desired product was filtered off, dried into the desiccators to afford the desired product.
1H-Pyrrolo[2,3-c]pyridine-2-carboxylic acid (10a). This compound was obtained in 95% yield. Analytical and spectroscopic data are in accordance to those reported in literature [29].
5-Chloro-1H-pyrrolo[2,3-c]pyridine-2-carboxylic acid (10b). This compound was obtained in 71% yield. Analytical and spectroscopic data are in accordance to those reported in literature [29].
5-Methoxy-1H-pyrrolo[2,3-c]pyridine-2-carboxylic acid (10c). This compound was obtained in 80% yield. Analytical and spectroscopic data are in accordance to those reported in literature [35].
7-Methoxy-1H-pyrrolo[2,3-c]pyridine-2-carboxylic acid (10d). This compound was obtained after 1 h reflux as a white powder. Rf = 0.40 (CH2Cl2/MeOH 9:1); mp 269.3–271.1 °C; yield 82%; IR: 3550 (NH), 3311 (OH), 1718 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 4.02 (3H, s, OCH3), 7.07 (1H, s, H-3), 7.21 (1H, d, J = 6.0 Hz , H-4), 7.68 (1H, d, J = 6.0 Hz, H-5), 9.61(1H, bs, NH), 12.30 (1H, bs, OH). 13C-NMR (DMSO-d6) δ: 52.7 (q), 106.8 (d), 110.5 (d), 122.0 (s), 131.5 (s), 132.6 (s), 134.8 (d), 151.6 (s), 162.3 (s). Anal. Calcd for C9H8N2O3(192.17): C, 56.25; H, 4.20; N, 14.58. Found: C, 56.29; H, 4.24; N, 14.37.

3.1.7. General Procedure for the Preparation of 6H,13H-Bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-diones 4ad

To a stirred solution of 10ad (2.3 mmol) in anhydrous THF (20 mL) dimethylaminopyridine (DMAP, 2.3 mmol) was added, followed by EDCI (4.8 mmol) addition after 10 min; the reaction mixture was stirred for 48h at room temperature. The solid was collected by filtration and recrystallizated from CH2Cl2 and MeOH, affording the desired products as yellow solids. Compounds 4ad were characterized only by 1H-NMR spectroscopy. The poor solubility of the title compounds prevented the 13C-NMR spectra from being recorded.
6H,13H-Bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (4a). Rf = 0.34 (CH2Cl2/MeOH 95:5); mp 352.3–353.9 °C; yield 25%; IR: 1722 (CO) cm1; 1H-NMR (DMSO-d6) δ: 7.91 (2H, d, J = 6.0 Hz, H-4 and H-11), 7.92 (2H, s, H-5 and H-12), 8.60 (2H, d, J = 6.0 Hz, H-3 and H-10), 9.71 (2H, s, H-1 and H-8). Anal. Calcd for C16H8N4O2 (288.26): C, 66.67; H, 2.80; N, 19.44. Found: C, 66.62; H, 2.84; N, 19.39.
3,10-Dichloro-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (4b). Rf = 0.60 (CH2Cl2/MeOH 98:2); mp 380.6–381.9 °C; yield 30%; IR: 1716 (CO) cm1; 1H-NMR (DMSO-d6) δ: 7.88 (2H, s, H-4 and H-11), 8.05 (2H, s, H-5 and H-12), 9.47 (2H, s, H-1 and H-8). Anal. Calcd for C16H6Cl2N4O2 (357.15): C, 53.81; H, 1.69; N, 15.69. Found: C, 53.89; H, 1.78; N, 15.97.
3,10-Dimethoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (4c). Rf = 0.57 (CH2Cl2/MeOH 98:2); mp 343.0–344.2 °C; yield 20%; IR: 1710 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 3.95 (3H, s, OCH3), 7.26 (2H, s, H-4 and H-11), 7.73 (2H, s, H-5 and H-12), 9.27 (2H, s, H-1 and H-8). Anal. Calcd for C18H12N4O4 (348.31): C, 62.07; H, 3.47; N, 16.09. Found: C, 61.92; H, 3.53; N, 15.95.
1,8-Dimethoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (4d). Rf = 0.45 (CH2Cl2/MeOH 98:2); mp 380.6–381.9 °C; yield 28%; IR: 1723 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 4.06 (3H, s, OCH3), 7.44 (2H, d, J = 6.0 Hz, H-4 and H-11), 7.79 (2H, s, H-5 and H-12), 8.12 (2H, d, J = 6.0 Hz, H-3 and H-10). Anal. Calcd for C18H12N4O4 (348.31): C, 62.07; H, 3.47; N, 16.09. Found: C, 61.83; H, 3.66; N, 16.05.

3.1.8. General Procedure for the Preparation of 6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-diones 5ae

To a stirred solution of 10ad (2.3 mmol) in anhydrous THF (20 mL) dimethylaminopyridine (DMAP, 2.3 mmol) was added, followed by EDCI (1.2 mmol) after 10 min; the reaction mixture was stirred at room temperature for 1h. The suitable acid 10ad (1.0 mmol) and EDCI (1.2 mmol) were added and the reaction mixture was stirred for 48 h. The solid was collected by filtration, purified by flash chromatography using CH2Cl2/MeOH 98:2 and recrystallized from CH2Cl2 and MeOH, affording the desired product as a yellow solid. Compounds 5ae were characterized only by 1H-NMR spectroscopy. The poor solubility of the title compounds prevented 13C-NMR spectra from being recorded.
8-Methoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (5a). This product was obtained by reaction of 10a with 10d. Rf = 0.46 (CH2Cl2/MeOH 98:2); mp 328.4–329.0 °C; yield 40%; IR: 1712 (CO), 1694 (CO) cm1; 1H-NMR (DMSO-d6) δ: 4.06 (3H, s, OCH3), 7.45 (1H, d, J = 6.0 Hz, H-11), 7.82 (1H, s, H-12), 7.89–7.92 (2H, m, H-5 and H-4), 8.14 (1H, d, J = 6.0 Hz, H-10), 8.60 (1H, d, J = 4.0 Hz, H-3), 9.67 (1H, s H-1). Anal. Calcd for C17H10N4O3 (318.29): C, 64.15; H, 3.17; N, 17.60. Found: C, 63.87; H, 3.13; N, 17.75. From this reaction derivatives 4a (yield 4%) and 4d (yield 6%) were also isolated.
1,10-Dimethoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (5b). This product was obtained by reaction of 10c with 10d. Rf = 0.34 (CH2Cl2/MeOH 95:5); mp 309.1–309.4 °C; yield 55%; IR: 1712 (CO), 1689 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 3.94 (3H, s, OCH3), 4.05 (3H, s, OCH3), 7.25 (1H, s, H-12), 7.42 (1H, d, J = 4.0 Hz, H-4), 7.79 (1H, s, H-11), 7.84 (1H, s, H-5), 8.12 (1H, d, J = 4.0 Hz, H-3), 9.24 (1H, s, H-8). Anal. Calcd for C18H12N4O4 (348.31): C, 62.07; H, 3.47; N, 16.09. Found: C, 62.20; H, 3.42; N, 16.25. From this reaction derivatives 4c (yield 5%) and 4d (yield 6%) were also isolated.
3-Methoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (5c). This product was obtained by reaction of 10a with 10c. Rf = 0.37 (CH2Cl2/MeOH 98:2); mp 271.1–271.8 °C; yield 42%; IR: 1718 (CO), 1707 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 3.95 (3H, s, OCH3), 7.26 (1H, s, H-12), 7.78 (1H, s, H-5), 7.87 (1H, s, H-4), 7.90 (1H, d, J = 6.0 Hz, H-11), 7.59 (1H, d, J = 6.0 Hz, H-10), 9.28 (1H, s, H-8), 9.68 (1H, s, H-1). Anal. Calcd for C17H10N4O3 (318.29): C, 64.15; H, 3.17; N, 17.60. Found: C, 64.06; H, 3.08; N, 17.89. From this reaction derivatives 4a (yield 4%) and 4c (yield 5%) were also isolated.
10-Chloro-1-methoxy-6H,13H-bispyrido[4',3':4,5]pyrrolo-[1,2-a:1',2'-d]pyrazine-6,13-dione (5d). This product was obtained by reaction of 10b with 10d. Rf = 0.47 (CH2Cl2/MeOH 98:2); mp 292.2–293.0 °C; yield 44%; IR: 1712 (CO), 1690 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 4.06 (3H, s, OCH3), 7.45 (1H, d J = 6.0 Hz, H-4), 7.75 (1H, s, H-5), 7.92 (1H, s, H-12), 8.05 (1H, s, H-11), 8.14 (1H, d, J = 6.0 Hz, H-3), 9.44 (1H, s, H-8). Anal. Calcd for C17H9ClN4O3 (352.73): C, 57.89; H, 2.57; N, 15.88. Found: C, 57.60; H, 2.48; N, 15.96. From this reaction derivatives 4b (yield 6%) and 4d (yield 5%) were also isolated.
3-Chloro-10-methoxy-6H,13H-bispyrido[4',3':4,5]-pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione (5e). This product was obtained by reaction of 10b with 10c. Rf = 0.56 (CH2Cl2/MeOH 98:2); mp 312.0–312.5 °C; yield 45%; IR: 1720 (CO), 1705 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 3.96 (3H, s, OCH3), 7.26 (1H, s, H-11), 7.80 (1H, s, H-12), 7.82 (1H, s, H-5), 8.04 (1H, s, H-4), 9.27 (1H, s, H-8), 9.46 (1H, s, H-1). Anal. Calcd for C17H9ClN4O3 (352.73): C, 57.89; H, 2.57; N, 15.88. Found: C, 57.80; H, 2.49; N, 16.16. From this reaction derivatives 4b (yield 5%) and 4c (yield 5%) were also isolated.

3.1.9. General Procedure for the Preparation of 6H,13H-Pyrido[4'',3'':4',5']pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-diones 6ad

To a stirred solution of the suitable 10ad (1.2 mmol) in anhydrous THF (20 mL) dimethylaminopyridine (DMAP) (1.2 mmol) was added, followed by EDCI (1.2 mmol) after 10 min; the reaction mixture was stirred at room temperature for 1h. Indole 2-carboxylic acid (1.0 mmol) and EDCI (1.2 mmol) were added and the reaction mixture was stirred for 48 h. The solid was collected by filtration, purified by flash chromatography using using CH2Cl2/MeOH 98:2 and recrystallized with CH2Cl2 and MeOH, affording the desired products as yellow solid. Compounds 6ad were characterized only by 1H-NMR spectroscopy. The poor solubility of the title compounds prevented 13C-NMR spectra from being recorded.
6H,13H-Pyrido[4'',3'':4',5']pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione (6a). Rf = 0.28 (CH2Cl2/MeOH 98:2); mp 347.4–347.8 °C; yield 33%; IR: 1701 (broad, CO) cm−1;1H-NMR (DMSO-d6) δ: 7.48 (1H, td, J = 6.0 2.0 Hz, H-9), 7.67 (1H, td, J = 6.0 2.0 Hz, H-10), 7.85 (1H, s, H-12), 7.88–7.93 (3H, m, H-4, H-5 and H-8), 8.48 (1H, d, J = 6.0 Hz, H-11), 8.58 (1H, d, J = 6.0 Hz, H-3), 9.71 (1H, s, H-1). Anal. Calcd for C17H9N3O2 (287.27): C, 71.08; H, 3.16; N, 14.63. Found: C, 71.29; H, 3.29; N, 14.84. From this reaction derivatives 4a (yield 5%) and 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-dione (yield 8%) whose analytical and spectroscopic data are in accordance to those reported in literature were also isolated [36].
3-Chloro-6H,13H-pyrido[4'',3'':4',5']pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione (6b). Rf = 0.63 (CH2Cl2/MeOH 98:2); mp 306.3–306.7 °C; yield 65%; IR: 1700 (broad, CO) cm−1; 1H-NMR (DMSO-d6) δ: 7.49 (1H, t, J = 8.0 Hz, H-9), 7.67 (1H, t, J = 8.0 Hz, H-10), 7.77 (1H, s, H-12), 7.92 (1H, d, J = 8.0 Hz, H-8), 7.95 (1H, s, H-5), 8.02 (1H, s, H-4), 8.48 (1H, d, J = 8.0 Hz, H-11), 9.48 (1H, s, H-1). Anal. Calcd for C17H8ClN3O2 (321.72): C, 63.47; H, 2.51; N, 13.06. Found: C, 63.68; H, 2.46; N, 13.30. From this reaction were also isolated derivatives 4b (yield 3%) and 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-dione (yield 7%) whose analytical and spectroscopic data are in accordance to those reported in literature [36].
3-Methoxy-6H,13H-pyrido[4'',3'':4',5']pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione (6c). Rf = 0.65 (CH2Cl2/MeOH 98:2); mp 279.0–279.4 °C; yield 65%; IR: 1727 (CO), 1702 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 3.95 (3H, s, OCH3), 7.24 (1H, s, H-4), 7.47 (1H, t, J = 8.0 Hz, H-9), 7.65 (1H, t, J = 8.0 Hz, H-10), 7.71 (1H, s, H-12), 7.87 (1H, s, H-5), 7.91 (1H, d, J = 8.0 Hz, H-8), 8.46 (1H, d, J = 8.0 Hz, H-11), 9.29 (1H, s, H-1). Anal. Calcd for C18H11N3O3 (317.30): C, 68.14; H, 3.49; N, 13.24. Found: C, 68.09; H, 3.70; N, 13.13. From this reaction were also isolated derivatives 4c (yield 4%) and 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-dione (yield 7%) whose analytical and spectroscopic data are in accordance to those reported in literature [36].
1-Methoxy-6H,13H-pyrido[4'',3'':4',5']pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione (6d). Rf = 0.63 (CH2Cl2/MeOH 98:2); mp 283.8–283.9 °C; yield 30%; IR: 1712 (CO), 1690 (CO) cm−1; 1H-NMR (DMSO-d6) δ: 4.05 (3H, s, OCH3), 7.41–7.50 (2H, m, H-4 and H-9), 7.64 (1H, t, J = 8.0 Hz, H-10), 7.81 (2H, s, H-5 and H-12), 7.90 (1H, d, J = 8.0 Hz, H-8), 8.10 (1H, d, J = 6.0 Hz, H-3), 8.43 (1H, d, J = 8.0 Hz, H-11). Anal. Calcd for C18H11N3O3 (317.30): C, 68.14; H, 3.49; N, 13.24. Found: C, 68.39; H, 3.45; N, 12.95. From this reaction were also isolated derivatives 4d (yield 6%) and 6H,13H-pyrazino[1,2-a:4,5-a']diindole-6,13-dione (yield 9%) whose analytical and spectroscopic data are in accordance to those reported in literature [36].

3.2. Docking

Docking studies were performed for all designed compounds by Glide 5.9 (Schrödinger Inc., New York, NY, USA, 2013). The X-ray crystallographic structures of tubulin (PDB code 3HKD [24] and 1SA0 [23]) were downloaded from Protein Data Bank [37]. For Glide docking studies, the stathmin-like domain and chains B, C were removed. The proteins were minimized by Protein Preparation Wizard. Partial atomic charges were assigned according to the OPLS_2005 force field. A radius of 20 Å was selected for active site cavity during receptor grid generation with the center defined by the co-crystallized ligand TN-16 and colchicine. All compounds used in the docking study with Glide were built within Maestro by using the build module of Schrödinger Inc. (2013). Docking calculations were performed using standard mode of Glide Program. To validate the Glide docking protocol, TN-16 was redocked into the binding site. The docking structure was compared to the crystal structure showing that this protocol successfully reproduces the crystal TN-16 tubulin complex.

3.3. Biology

Methodology of the in Vitro Cancer Screen

In vitro cancer screens were done according to the NCI protocol at 10−5 M dose on the full panel of 60 human cancer cell lines derived from nine human cancer cell types that have been grouped in disease sub-panels including leukemia (CCRF-CEM, HL-60(TB), K-562, MOLT-4, RPMI-8226, SR), non-small-cell lung (A549/ATCC, EKVX, HOP-62, HOP-92, NCI-H226, NCI-H23, NCI-H322M, NCI-H460, NCI-H522), colon (COLO 205, HCC-2998, HCT-116, HCT-15, HT29, KM12, SW-620), central nervous system (SF-268, SF-295, SF-539, SNB-19, SNB-75, U251), melanoma (LOX IMVI, MALME-3M, M14, MDA-MB-435, SK-MEL-2, SK-MEL-28, SK-MEL-5, UACC-257, UACC-62), ovarian (IGROV1, OVCAR-3, OVCAR-4, OVCAR-5, OVCAR-8, NCI/ADR-RES, SK-OV-3), renal (786-0, A498, ACHN, CAKI-1, RXF 393, SN12C, TK-10, UO-31), prostate (PC-3, DU-145) and breast tumour (MCF7, MDA-MB-231/ATCC, HS 578T, BT-549, T-47D, MDA-MB-468) cell lines [32].
The human tumor cell lines of the cancer screening panel are grown in RPMI 1640 medium containing 5% fetal bovine serum and 2 mM l-glutamine. For a typical screening experiment, cells are inoculated into 96 well microtiter plates in 100 µL at plating densities ranging from 5000 to 40,000 cells/well depending on the doubling time of individual cell lines. After cell inoculation, the microtiter plates are incubated at 37 °C, 5% CO2, 95% air and 100% relative humidity for 24 h prior to addition of experimental drugs. After 24 h, two plates of each cell line are fixed in situ with TCA, to represent a measurement of the cell population for each cell line at the time of drug addition (Tz). Experimental drugs are solubilized in dimethyl sulfoxide at 400-fold the desired final maximum test concentration and stored frozen prior to use. At the time of drug addition, an aliquot of frozen concentrate is thawed and diluted to twice the desired final maximum test concentration with complete medium containing 50 µg/mL gentamicin. Aliquots of 100 µL of drug are added to the appropriate microtiter wells already containing 100 µL of medium, resulting in the required final drug concentration. Following drug addition, the plates are incubated for an additional 48 h at 37 °C, 5% CO2, 95% air, and 100% relative humidity. For adherent cells, the assay is terminated by the addition of cold TCA. Cells are fixed in situ by the gentle addition of 50 µL of cold 50% (w/v) TCA (final concentration, 10% TCA) and incubated for 60 min at 4 °C. The supernatant is discarded, and the plates are washed five times with tap water and air dried. Sulforhodamine B (SRB) solution (100 µL) at 0.4% (w/v) in 1% acetic acid is added to each well, and plates are incubated for 10 min at room temperature. After staining, unbound dye is removed by washing five times with 1% acetic acid and the plates are air dried. Bound stain is subsequently solubilized with 10 mM trizma base, and the absorbance is read on an automated plate reader at a wavelength of 515 nm. For suspension cells, the methodology is the same except that the assay is terminated by fixing settled cells at the bottom of the wells by gently adding 50 µL of 80% TCA (final concentration, 16% TCA). Using the seven absorbance measurements [time zero, (Tz), control growth, (C), and test growth in the presence of drug (Ti)], the percentage growth is calculated. Percentage growth inhibition is calculated as:
[(Ti - Tz)/(C − Tz)] × 100 for concentrations for which Ti ≥ Tz
[(Ti - Tz)/Tz] × 100 for concentrations for which Ti ˂ Tz
For further information to see NCI website [38].

4. Conclusions

In conclusion, we have reported the synthesis of derivatives of the new ring systems 6H,13H-bispyrido[4',3':4,5]pyrrolo[1,2-a:1',2'-d]pyrazine-6,13-dione 4, 5 and 6H,13H-pyrido[4'',3'':4',5']-pyrrolo[1',2':4,5]pyrazino[1,2-a]indole-6,13-dione 6 using a simple and versatile synthetic pathway. All derivatives were prescreened according to the NCI protocol at 10−5 M dose on the full panel of 60 human cancer cell lines derived from nine human cancer cell types. Only derivatives 5a and 6a, 6c and 6d showed a moderate antineoplastic activity at micromolar concentration. In particular derivative 5a exhibited modest activity against the UO-31 renal cancer sub-panel cell line; deaza analogue 6a and the 9-methoxy substituted derivative 6c were shown to be selective against the MCF7 breast cancer cell line. More interesting results were obtained from the 11-methoxy substituted compound 6d which showed selectivity against both the UO-31 renal cancer sub-panel and the MCF7 breast cancer sub-panel cell lines. Unfortunately the moderate activity showed by derivatives 5a and 6a, 6c and 6d against a limited number of cell lines could not allow a reliable SAR evaluation. However, the antiproliferative activity shown by derivatives 5a and 6a, 6c and 6d, although modest, encourages further studies directed toward the synthesis of new compounds with an improved growth inhibitory effect.

Acknowledgments

This work was financially supported by Ministero dell’Istruzione dell’Università e della Ricerca. We thank the National Cancer Institute (Bethesda, MD) for the antitumour tests reported in this paper.

Author Contributions

Girolamo Cirrincione, Patrizia Diana, Alessandra Montalbano and Paola Barraja designed research; Barbara Parrino performed docking studies and analyzed the data, Alessandra Montalbano, Anna Carbone and Virginia Spanò performed research and analyzed the data; Girolamo Cirrincione, Patrizia Diana, Alessandra Montalbano, Barbara Parrino and Paola Barraja wrote the paper. All authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hilton, S.; Rossiter, S. Three heterocyclic ring fused (5-6-5). In Comprehensive Heterocyclic Chemistry III; Jones, K., Ed.; Elsevier Ltd.: Oxford, UK, 2008; Volume 12, pp. 747–751. [Google Scholar]
  2. Kanoh, K.; Kohno, S.; Asari, T.; Harada, T.; Katada, J.; Muramatsu, M.; Kawashima, H.; Sekiya, H.; Uno, I. (−)-Phenylahistin: A new mammalian cell cycle inhibitor produced by Aspergillus ustus. Bioorg. Med. Chem. Lett. 1997, 7, 2847–2852. [Google Scholar] [CrossRef]
  3. Kanoh, K.; Kohno, S.; Katada, J.; Takahashi, J.; Uno, I. (−)-Phenylahistin arrests cells in mitosis by inhibiting tubulin polymerization. J. Antibiot. 1999, 52, 134–141. [Google Scholar] [CrossRef]
  4. Kanoh, K.; Kohno, S.; Katada, J.; Hayashi, Y.; Muramatsu, M.; Uno, I. Antitumor activity of phenylahistin in vitro and in vivo. Biosci. Biotechnol. Biochem. 1999, 63, 1130–1133. [Google Scholar] [CrossRef]
  5. Kanoh, K.; Kohno, S.; Katada, J.; Takahashi, J.; Uno, I.; Hayashi, Y. Synthesis and biological activities of phenylahistin derivatives. Bioorg. Med. Chem. 1999, 7, 1451–1457. [Google Scholar] [CrossRef]
  6. Yamazaki, Y.; Sumikura, M.; Hidaka, K.; Yasui, H.; Kiso, Y.; Yakushiji, F.; Hayashi, Y. Anti-microtubule “plinabulin” chemical probe KPU-244-B3 labeled both α- and β-tubulin. Bioorg. Med. Chem. 2010, 18, 3169–3174. [Google Scholar] [CrossRef]
  7. Yamazaki, Y.; Tanaka, K.; Nicholson, B.; Deyanat-Yazdi, G.; Potts, B.; Yoshida, T.; Oda, A.; Kitagawa, T.; Orikasa, S.; Kiso, Y.; et al. Synthesis and structure-activity relationship study of antimicrotubule agents Phenylahistin derivatives with a didehydropiperazine-2,5-dione structure. J. Med. Chem. 2012, 55, 1056–1071. [Google Scholar] [CrossRef]
  8. Boger, D.L.; Fink, B.E.; Hedrick, M.P. A new class of highly cytotoxic diketopiperazines. Bioorg. Med. Chem. Lett. 2000, 10, 1019–1020. [Google Scholar] [CrossRef]
  9. Paleni, D. Preparation of Dioxopyrazinodiindoles as Neoplasm Inhibitors. FR 2650590, 8 February 1991. [Google Scholar]
  10. Paleni, D. Preparation and Formulation of Dioxopyrazinodiindoles as Cell Differentiation Inhibitors. DE 4102921, 6 August 1992. [Google Scholar]
  11. Barraja, P.; Caracausi, L.; Diana, P.; Carbone, A.; Montalbano, A.; Cirrincione, G.; Brun, P.; Palù, G.; Castagliuolo, I.; Dall’Acqua, F.; et al. Synthesis of pyrrolo[3,2-h]quinolinones with good photochemotherapeutic activity and no DNA damage. Bioorg. Med. Chem. 2010, 18, 4830–4843. [Google Scholar] [CrossRef]
  12. Barraja, P.; Caracausi, L.; Diana, P.; Montalbano, A.; Carbone, A.; Salvador, A.; Brun, P.; Castagliuolo, I.; Tisi, S.; Dall’Acqua, F.; et al. Pyrrolo[3,2-h]quinazolines as photochemotherapeutic agents. ChemMedChem 2011, 6, 1238–1248. [Google Scholar] [CrossRef]
  13. Carbone, A.; Parrino, B.; Barraja, P.; Spanò, V.; Cirrincione, G.; Diana, P.; Maier, A.; Kelter, G.; Fiebig, H.-H. Synthesis and antiproliferative activity of 2,5-bis(3'-indolyl)pyrroles, analogues of the marine alkaloid nortopsentin. Mar. Drugs 2013, 11, 643–654. [Google Scholar] [CrossRef]
  14. Barraja, P.; Caracausi, L.; Diana, P.; Spanò, V.; Montalbano, A.; Carbone, A.; Parrino, B.; Cirrincione, G. Synthesis and antiproliferative activity of the ring system [1,2]oxazolo[4,5-g]indole. ChemMedChem 2012, 7, 1901–1904. [Google Scholar] [CrossRef]
  15. Diana, P.; Carbone, A.; Barraja, P.; Montalbano, A.; Parrino, B.; Lopergolo, A.; Pennati, M.; Zaffaroni, N; Cirrincione, G. Synthesis and antitumor activity of 3-(2-phenyl-1,3-thiazol-4-yl)-1H-indoles and 3-(2-phenyl-1,3-thiazol-4-yl)-1H-7-azaindoles. ChemMedChem 2011, 6, 1300–1309. [Google Scholar] [CrossRef]
  16. Carbone, A.; Pennati, M.; Parrino, B.; Lopergolo, A.; Barraja, P.; Montalbano, A.; Spanò, V.; Sbarra, S.; Doldi, V.; de Cesare, M.; et al. Novel 1H-pyrrolo[2,3-b]pyridine derivatives nortopsentin analogues: Synthesis and antitumor activity in peritoneal mesothelioma experimental models. J. Med. Chem. 2013, 56, 7060–7072. [Google Scholar] [CrossRef]
  17. Carbone, A.; Pennati, M.; Barraja, P.; Montalbano, A.; Parrino, B.; Spanò, V.; Lopergolo, A.; Sbarra, S.; Doldi, V.; Zaffaroni, N.; et al. Synthesis and antiproliferative activity of substituted 3-[2-(1H-indol-3-yl)-1,3-thiazol-4-yl]-1H-pyrrolo[3,2-b]pyridines, marine alkaloid nortopsentin analogues. Curr. Med. Chem. 2014, 21, 1654–1666. [Google Scholar] [CrossRef]
  18. Barraja, P.; Diana, P.; Montalbano, A.; Dattolo, G.; Cirrincione, G.; Viola, G.; Vedaldi, D.; Dall’Acqua, F. Pyrrolo[2,3-h]quinolinones: A new ring system with potent photoantiproliferative activity. Bioorg. Med. Chem. 2006, 14, 8712–8728. [Google Scholar] [CrossRef]
  19. Barraja, P.; Spanò, V.; Diana, P.; Carbone, A.; Cirrincione, G.; Vedaldi, D.; Salvador, A.; Viola, G.; Dall’Acqua, F. Pyrano[2,3-e]isoindol-2-ones, a new angelicin heteroanalogues. Bioorg. Med. Chem. Lett. 2009, 19, 1711–1714. [Google Scholar] [CrossRef]
  20. Barraja, P.; Spanò, V.; Diana, P.; Carbone, A.; Cirrincione, G. Synthesis of the new ring system 6,8-dihydro-5H-pyrrolo[3,4-h]quinazoline. Tetrahedron Lett. 2009, 50, 5389–5391. [Google Scholar]
  21. Barraja, P.; Diana, P.; Montalbano, A.; Carbone, A.; Viola, G.; Basso, G.; Salvador, A.; Vedaldi, D.; Dall’Acqua, F.; Cirrincione, G. Pyrrolo[3,4-h]quinolinones a new class of photochemotherapeutic agents. Bioorg. Med. Chem. 2011, 19, 2326–2341. [Google Scholar] [CrossRef]
  22. Spanò, V.; Montalbano, A.; Carbone, A.; Parrino, B.; Diana, P.; Cirrincione, G.; Castagliuolo, I.; Brun, P.; Issinger, O.-G.; Tisi, S.; et al. Synthesis of a new class of pyrrolo[3,4-h]quinazolines with antimitotic activity. Eur. J. Med. Chem. 2014, 74, 340–357. [Google Scholar] [CrossRef]
  23. Barraja, P.; Spanò, V.; Giallombardo, D.; Diana, P.; Montalbano, A.; Carbone, A.; Parrino, B.; Cirrincione, G. Synthesis of [1,2]oxazolo[5,4-e]indazoles as antitumour agents. Tetrahedron 2013, 69, 6474–6477. [Google Scholar] [CrossRef]
  24. Ravelli, R.B.G.; Gigant, B.; Curmi, P.A.; Jourdain, I.; Lachkar, S.; Sobel, A.; Knossow, M. Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature 2004, 428, 198–202. [Google Scholar]
  25. Dorléans, A.; Gigant, B.; Ravelli, R.B.G.; Mailliet, P.; Mikol, V.; Knossow, M. Variations in the colchicine-binding domain provide insight into the structural switch of tubulin. Proc. Natl. Acad. Sci. USA 2009, 106, 13775–13779. [Google Scholar]
  26. Arai, T. Inhibition of microtubule assembly in vitro by TN-16, a synthetic antitumor drug. FEBS Lett. 1983, 155, 273–276. [Google Scholar] [CrossRef]
  27. Barbier, P.; Dorléans, A.; Devred, F; Sanz, L.; Allegro, D.; Alfonso, C.; Knossow, M.; Peyrot, V.; Andreu, J.M. Stathmin and interfacial microtubule inhibitors recognize a naturally curved conformation of tubulin dimmers. J. Biol. Chem. 2010, 285, 31672–31681. [Google Scholar] [CrossRef]
  28. Glide, version 5.9; Schrödinger, LLC: New York, NY, USA, 2013.
  29. Bradley, S.E.; Krulle, T.M.; Murray, P.J.; Procter, M.J.; Rowley, R.J.; Sambrook, S.C.P.; Thomas, G.H. Preparation of Pyrrolopyridine-2-Carboxylic Acid Amide as Inhibitors of Glycogen Phosphorylase. WO 2004104001, 2 December 2004. [Google Scholar]
  30. Casara, P.; le Diguarher, T.; Durand, D.; Geneste, O.; Hickman, J. New Tricyclic Derivatives, Their Preparation as Pro-Apoptotic and Antitumor Agents and Their Pharmaceutical Compositions Containing Them. WO 2010007248, 21 January 2010. [Google Scholar]
  31. Evans, G.B.; Furneaux, R.H.; Hutchison, T.L.; Kezar, H.S.; Morris, P.E., Jr.; Schramm, V.L.; Tyler, P.C. Addition of lithiated 9-deazapurine derivatives to a carbohydrate cyclic imine: Convergent synthesis of the aza-C-nucleoside immucillins. J. Org. Chem. 2001, 66, 5723–5730. [Google Scholar] [CrossRef]
  32. Monks, A.; Scudiero, D.; Skehan, P.; Shoemaker, R.; Paull, K.; Vistica, D.; Hose, C.; Langley, J.; Cronise, P.; Vaigro-Wolff, A.; et al. Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J. Natl. Cancer Inst. 1991, 83, 757–766. [Google Scholar] [CrossRef]
  33. Frydman, B.; Despuy, M.E.; Rapoport, H. Pyrroles from azaindoles. A synthesis of porphobilinogen. J. Am. Chem. Soc. 1965, 3530–3531. [Google Scholar]
  34. Dubois, L.; Evanno, Y.; Malanda, A. Preparation Of 1-(Arylalkyl)-1H-Pyrrolopyridine-2-Carboxamide Derivatives as VR1 Type Capsaicin Receptor Antagonists. WO 2007010138, 25 January 2007. [Google Scholar]
  35. Frydman, B.; Reil, S.J.; Boned, J.; Rapoport, H. Synthesis of substituted 4- and 6-azaindoles. J. Org. Chem. 1968, 33, 3762–3766. [Google Scholar] [CrossRef]
  36. Qiao, G.G.; Meutermans, W.; Wong, M.W.; Traeubel, M.; Wentrup, C. (Cyanovinyl)ketenes from azafulvenones. An apparent retro-Wolff rearrangement. J. Am. Chem. Soc. 1996, 118, 3852–3861. [Google Scholar]
  37. Protein Data Bank. Available online: http://www.rcsb.org/pdb (accessed on 28 August 2014).
  38. NCI-60 DTP Human tumor cell line screen. Available online: http://dtp.nci.nih.gov/branches/btb/ivclsp.html (accessed on 20 July 2012).
  • Sample Availability: Not available.

Share and Cite

MDPI and ACS Style

Parrino, B.; Spanò, V.; Carbone, A.; Barraja, P.; Diana, P.; Cirrincione, G.; Montalbano, A. Synthesis of the New Ring System Bispyrido[4',3':4,5]pyrrolo [1,2-a:1',2'-d]pyrazine and Its Deaza Analogue. Molecules 2014, 19, 13342-13357. https://doi.org/10.3390/molecules190913342

AMA Style

Parrino B, Spanò V, Carbone A, Barraja P, Diana P, Cirrincione G, Montalbano A. Synthesis of the New Ring System Bispyrido[4',3':4,5]pyrrolo [1,2-a:1',2'-d]pyrazine and Its Deaza Analogue. Molecules. 2014; 19(9):13342-13357. https://doi.org/10.3390/molecules190913342

Chicago/Turabian Style

Parrino, Barbara, Virginia Spanò, Anna Carbone, Paola Barraja, Patrizia Diana, Girolamo Cirrincione, and Alessandra Montalbano. 2014. "Synthesis of the New Ring System Bispyrido[4',3':4,5]pyrrolo [1,2-a:1',2'-d]pyrazine and Its Deaza Analogue" Molecules 19, no. 9: 13342-13357. https://doi.org/10.3390/molecules190913342

Article Metrics

Back to TopTop