Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 May 23.
Published in final edited form as: J Med Chem. 2013 May 1;56(10):3806–3819. doi: 10.1021/jm301689x

Synthesis of New Quinolinequinone Derivatives and Preliminary Exploration of their Cytotoxic Properties

Charles M Keyari 1, Alison K Kearns 1, Nathan S Duncan 1, Emily A Eickholt 1, Geoffrey Abbott 1, Howard D Beall 1,*, Philippe Diaz 1,*
PMCID: PMC3752426  NIHMSID: NIHMS475173  PMID: 23574193

Abstract

A series of 7-amino- and 7-acetamidoquinoline-5,8-diones with aryl substituents at the 2-position were synthesized, characterized and evaluated as potential NAD(P)H:quinone oxidoreductase (NQO1)-directed antitumor agents. The synthesis of lavendamycin analogs is illustrated. Metabolism studies demonstrated that 7-amino-analogues were generally better substrates for NQO1 than 7-amido-analogues as were compounds with smaller heteroaromatic substituents at the C-2 position. Surprisingly, only two compounds, 7-acetamido-2-(8’-quinolinyl)quinoline-5,8-dione (11) and 7-amino-2-(2-pyridinyl)quinoline-5,8-dione (23) showed selective cytotoxicity toward the NQO1-expressing MDA468-NQ16 breast cancer cells versus the NQO1-null MDA468-WT cells. For all other compounds, NQO1 protected against quinoline-5,8-dione cytotoxicity. Compound 22 showed a potent activity against human breast cancer cells expressing or not expressing NQO1 with IC50 values of respectively 190 nM and 140 nM and a low NQO1 mediated reduction rate, which suggests that the mode of action of 22 differs from lavendamycin and involves an unidentified target(s).

Keywords: Lavendamycin, Suzuki coupling, microwave irradiation, palladium (0) catalysis, quinolinequinones, NQO1, antitumor, cytotoxicity

INTRODUCTION

Lavendamycin (Figure 1) is a quinolinequinone antibiotic with antitumor activity first isolated from Streptomyces lavendulae by Balitz et al in 1982.12 It is structurally related to Streptonigrin which was first isolated from Streptomyces flocculus.34 Streptonigrin is known for its potent cytotoxic properties, antitumor activity, and in vitro and in vivo antiviral properties and potent, broad spectrum antimicrobial properties. Although lavendamycin is not suitable for clinical use due to its toxicity, its analogs are less toxic and hence have potential as antitumor agents.5 Recent findings611 suggest that some indolequinones and quinolinequinones are excellent substrates for the quinone reductase enzyme, NAD(P)H:quinone oxidoreductase 1 (NQO1), and are selectively cytotoxic to cancer cell lines that overexpress NQO1. NQO1 is a ubiquitous flavoenzyme that catalyzes the 2-electron reduction of quinones to hydroquinones, and it is highly expressed in many solid tumors.12 This forms the basis for the synthesis of novel quinolinequinones structurally related to lavendamycin as potential NQO1-directed antitumor agents.

Figure 1.

Figure 1

Natural quinolinequinone antibiotics

Behforouz et al. (1996)13 first demonstrated that 7-aminoquinoline-5,8-diones can be efficiently prepared from commercially available 8-hydroxy-2-methylquinoline. Fryatt and co-workers7 also showed that by starting with 6-methoxyquinoline, 6-methoxy-2-chloroquinoline-5,8-dione was prepared and subsequent palladium(0) catalyzed reaction with boronic acids gave novel quinoline quinones under reflux for 24 hours. Further, in 2004,14 arylboronic acids were shown to be more reactive than their counterparts, the arylpinacolboronate esters when reacted with indole bromides in Suzuki couplings under reflux. The lower reactivity was attributed to steric factors in the arylpinacolboronate esters. Also, 3-aryl-indazoles have been synthesized by the reaction of haloindazoles (3-bromoindazole and 3-iodoindazole) with aryl boronic acids under Pd(0) catalysis in Suzuki type cross couplings.15 The reaction times ranged from 1–18 hours under reflux conditions. In this present study we report a direct more efficient approach to 7-aminoquinoline quinones starting from commercially available 7-amino-8-hydroxyquinoline under microwave conditions where the reaction times are shorter. Computational, metabolism and cytotoxicity studies on the quinoline-5,8-diones are also described.

CHEMISTRY

The synthesis commenced with the nitration of 5-chloro-8-hydroxyquinoline under HNO3/H2SO4 according to a procedure reported by Musser et al.16 to give the 5-chloro-7-nitro-8-hydroxyquinoline (1) in good yield (79%). Hydrogenation under Pd/C-catalysis at 40–50 psi not only reduced the nitro group to the free amine but also removed the chloride to provide the desired 7-amino-8-hydroxyquinoline (2) in excellent yield (99%). A direct approach to the amino alcohol 2 involves the heating of a mixture of 8-hydroxyquinoline and N-methyl-N-phenylhydrazine at 90°C, albeit very low yields were obtained.17Our attempt to synthesize the amino alcohol by heating in a microwave between 130–160°C did not improve the yield. Acetylation proceeded smoothly where both the amino and hydroxyl groups were protected. The resulting diacetylated product (3) was hydrolyzed in MeOH/H2O under reflux to form 7-acetamido-8-hydroxyquinoline. Subsequent benzylation of the free hydroxyl was effected by reacting with BnBr/K2CO3 in DMF at 50°C for 24 hrs to give the 7-acetamido-8-benzyloxyquinoline (4) in 90% yield. Oxidation using mCPBA in 1,2-dichloroethane at rt for 48 hrs gave the N-oxide (5) in 82% yield.18 The key intermediate in the synthesis, the 2-chloro-7-acetamido-8-benzyloxyquinoline (6), was obtained in 62% yield by refuxing the N-oxide with POCl3 in CHCl3.19 The high regioselectivity of the reaction can be rationalized in terms of sterics as well as formation of an oxyphosphorane adduct anion in a rapid concerted mechanism.20 We also attempted refluxing the N-oxide 5 with SO2Cl2 as reported in literature,9 but only resulted in massive decomposition of the starting material. Deprotection of the benzyl group was effected with BCl3•SMe2 in CH2Cl2 and subsequent oxidation using Fremy’s salt (potassium nitrosodisulfonate-(KO3S)2NO) gave the 7-acetamido-2-chloro-quinoline-5,8-dione (8) in 71% yield.7 The results are summarized in Scheme 1 below.

Scheme 1a.

Scheme 1a

aReagents and conditions: (i) HNO3/H2SO4; (ii) H2/Pd-C, MeOH, 40–50psi, overnight; (iii) CH3COCl, DIEA, THF, 2hrs.; (iv) H2O-MeOH, reflux, 1hr; (v) BnBr, K2CO3, DMF, 50°C, 24 hrs; (vi) mCPBA, ClCH2CH2Cl, 48hrs; (vii) POCl3, CHCl3, reflux, 2 hrs; (viii) BCl3·SMe2, CH2Cl2, overnight; (ix) Fremy's salt, rt, 1 hr; (x) RB(OH)2, Pd(PPh3)4, 110–140°C, µW 20–25 min.

After the successful formation of the quinolinequinone 8, the stage was now set for Suzuki coupling chemistry. This was accomplished by reaction with different boronic acids under Pd(0) catalysis in a microwave as illustrated in Table 1 below. Generally, the reactions were complete within 20–30 minutes in good yields except for the arylboronate ester where only 27% of the product (16) was obtained. The mechanistic details of the reaction have been well studied in which case oxidative addition, transmetallation and reductive elimination being the most critical steps.21 Interestingly, the 7-amino-2-(2-pyridyl)quinoline-5,8-dione was prepared in 9 steps starting from 3-hydroxybenzoic acid where the key step was a Friedlander condensation of 2-acetylpyridine and 2-amino-3-benzyloxy-4-bromobenzaldehyde to give the 8-benzyloxy-7-bromo-2-(2’-pyridyl)quinoline.22 Although this seems an attractive strategy, the method lacks the flexibility needed to create a library of lavendamycin analogs.

Table 1.

Suzuki coupling products

graphic file with name nihms475173t1.jpg

R-X Reaction conditions R Yield (%)
graphic file with name nihms475173t2.jpg Pd(PPh3)4, DME/Na2CO3
140°C, 20 min
graphic file with name nihms475173t3.jpg (9) 70
graphic file with name nihms475173t4.jpg Pd(PPh3)4, DME/Na2CO3
120°C, 20 min
graphic file with name nihms475173t5.jpg (10) 41
graphic file with name nihms475173t6.jpg Pd(PPh3)4, DME/Na2CO3
120°C, 20 min
graphic file with name nihms475173t7.jpg (11) 51
graphic file with name nihms475173t8.jpg Pd(PPh3)4, DME/Na2CO3
110°C, 25 min
graphic file with name nihms475173t9.jpg (12) 67
graphic file with name nihms475173t10.jpg *Pd(PPh3)4, p-dioxane
120°C, 20 min
graphic file with name nihms475173t11.jpg (13) 71
graphic file with name nihms475173t12.jpg Pd(PPh3)4, DME/Na2CO3
110°C, 25 min
graphic file with name nihms475173t13.jpg (14) 53
graphic file with name nihms475173t14.jpg Pd(PPh3)4, DME/Na2CO3
120°C, 20 min
graphic file with name nihms475173t15.jpg (15) 42
graphic file with name nihms475173t16.jpg PdCl2(dppf), p-dioxane,K3PO4
120°C,30 min
graphic file with name nihms475173t17.jpg (16) 27
*

Stille coupling reaction

The final step in the synthesis involved the removal of the acetate protecting group which was effected by reaction with H2SO4-MeOH at rt. The Boc-protected derivatives were also subjected to TFA/CH2Cl2 at rt for 2 hrs to provide the 7-acetamido derivatives.

ELECTROCHEMISTRY

Eletrochemistry was performed to compare the electrochemical behavior of the quinolinequinones with their reduction rates by NQO1, and the data are shown in Table 2. Tetrahydrofuran was used as the solvent for all compounds except 15, which was run in DMSO. The compounds were run against a Ag/AgCl electrode cathodic and anodic peak potentials, Epc and Epa, respectively, were measured at a potential sweep rate of 50 mV/sec, and the midpoint of the peak potentials was used to determine E1/2 values, (Epc + Epa)/2. Unfortunately, many of the analogues did not show reversible electrochemistry, and in some cases, there were multiple reduction peaks. This makes interpretation of the numbers somewhat difficult, but some conclusions can be drawn. For instance, most of the acetylated quinolinequinones had a reduction peak between −1.08V and −1.18V, an indication that they are easier to reduce than the non-acetylated compounds due to the presence of this electron-withdrawing group. This is consistent with what we reported previously for lavendamycins. However, there was no correlation between reduction potentials and reduction rates by NQO1, in line with previous publications on this topic.68, 23, 24 This suggests that steric interactions are more likely to be predictive of substrate efficiency than reduction potentials.

Table 2.

Reduction ratesa and oxygen consumptionb as a result of quinoline-5,8-dione metabolism by recombinant human NQO1 and electrochemical reduction potentials vs ferrocenec. Reduction rates were determined by spectrophotometric cytochrome c assay and oxygen uptake by oxygen electrode.

graphic file with name nihms475173t18.jpg

No. R1 R2 Reduction rate by
hNQO1
(µmol cyt c reduced
/min/mg NQO1
Oxygen
Consumption
(µmol/min/mg
NQO1)
Reduction Potential
(E1/2(V) vs Fc)
9 CH3CO graphic file with name nihms475173t19.jpg 4.5 +/− 0.5 5.2 +/− 1.0 −1.93 nrd
10 CH3CO graphic file with name nihms475173t20.jpg 25 +/− 4 - −1.17, −1.92 nr
11 CH3CO graphic file with name nihms475173t21.jpg 7.0 +/− 0.3 - −1.18, −1.68, −1.77
13 CH3CO graphic file with name nihms475173t22.jpg 480 +/− 200 34 +/− 3 −1.90 nr
15 CH3CO graphic file with name nihms475173t23.jpg 16 +/− 1 - −1.08, −1.36, −1.58
17 CH3CO graphic file with name nihms475173t24.jpg 2.8 +/− 0.2 - −1.10, −1.60, −1.94
18 CH3CO graphic file with name nihms475173t25.jpg 31 +/− 9 - -
19 H graphic file with name nihms475173t26.jpg 78 +/− 7 - -
20 H graphic file with name nihms475173t27.jpg 170 +/− 30 - −1.99 nr
21 H graphic file with name nihms475173t28.jpg 80 +/− 8 - −1.84 nr
22 H graphic file with name nihms475173t29.jpg 18 +/− 6 - −1.53, −1.65
23 H graphic file with name nihms475173t30.jpg 71 +/− 13 - −1.85 nr
24 H graphic file with name nihms475173t31.jpg 120 +/− 10 8.5 +/− 1.6 -
a

Spectrophotometric assay using cytochrome c as terminal electron acceptor (550 nm).

b

Oxygen concentration monitored using an oxygen electrode.

c

E1/2 values calculated as (Epc+Epa)/2 are average values from voltammograms recorded potential sweep rate of 50 mV/sec. Epc = cathodic peak potential; Epa = anodic peak potential.

d

nr = non-reversible, anodic peak only.

NMR SPECTROSCOPY AND SPECTROPHOTOMETRY

Complexation of zinc(II) triflate by compounds 13, 19, 22, and 23 was studied using 1H NMR spectroscopy. No new peaks were observed in NMR spectra, indicating that free and complexed forms of zinc(II) triflate were in a rapid exchange relative to NMR timescale. The aromatic region of the NMR spectrum of compound 19 in THF-D8 at room temperature is shown below in Figure 2.

Figure 2.

Figure 2

Aromatic proton region NMR spectra of 19 upon addition of increasing equivalent of Zn(SO3CF3)2 in [2H8]THF. Note the change in δ̣ of H-2’ and H-3 on addition of Zn2+. Equivalents of Zn2+: A=0, B=1, C=2, D=3, E=4, F=5 and G=10.

There was a small difference in chemical shifts of H-2’ (moving upfield) and H-3 (moving downfield) after addition of one equivalent zinc(II) triflate to the NMR solution (Table S1 and Figure 2) whereas the changes in δ of the other protons were barely noticeable. The biggest change in δ of H-2’ (−0.04 ppm) and H-3’ (+0.07 ppm) occurs after addition of ten equivalents of Zn(SO3CF3)2. This suggests that weak binding occurs at low Zn2+ concentration.

In contrast, addition of only one equivalent of Zn(SO3CF3)2 to compound 22 caused larger chemical shift variations of all the protons (Table S2 and Figure 3). Increasing the amount of Zn2+ (2–10 equiv.) added to compound 22 made little or no difference in δ afterwards (>0.01 ppm). This means that the quinoline derivative binds the Zn2+ more efficiently than compound 19 and only one equivalent of Zn2+ is enough to cause chemical shift variations.

Figure 3.

Figure 3

Aromatic protons region NMR spectra of 22 upon addition of increasing equivalent of Zn(SO3CF3)2 in [2H8]THF. Equivalents of Zn2+: A=0, B=1, C=2, D=3, E=4, F=5 and G=10.

Similar observations were made with compounds 23 (Table S3 and Figure 4) and 13 (Table S4). This is consistent with the results reported by Long and Harding25 where they demonstrated that the 1:1 bipyridyl complex of streptonigrin was the major complex formed at room temperature by perfoming an NMR study in [2H8]THF with addition of Zn2+. Titration of compound 23 with Zn2+ in a mixture of dimethylsulfoxide-methanol (1:3) was monitored by a spectrophotometer as reported in literature.26 A plot of Δλ355 against Zn2+ concentration gave a moderate affinity constant of 1.41×104 M−1 for compound 23 binding with Zn2+.

Figure 4.

Figure 4

Aromatic proton region NMR spectra of 23 upon addition of increasing equivalent of Zn(SO3CF3)2 in [2H8]THF. Equivalents of Zn2+: A=0, B=1, C=2, D=3, E=4, F=5 and G=10.

RESULTS AND DISCUSSION

Quinolinequinone metabolism by recombinant human NQO1 was examined using a spectrophotometric assay that employs cytochrome c as the terminal electron acceptor. Initial rates of reduction (µmol cytochrome c reduced/min/mg NQO1) were calculated from the linear portion (0–30 s) of the reaction graphs. The 7-acetamido-2-(2-pyridinyl) compound 13 displayed the highest reduction rate by NQO1 (Table 2), although it was the only acetylated analogue with a high reduction rate. In all other cases, 7-amino compounds had much higher reduction rates than corresponding 7-acetamido compounds with identical substituents at the quinoline-2-position. Although unusual, higher rates for acetylated analogues have been observed in other series.6, 11 With regard to the aromatic substituents at the quinoline-2-position, no clear trend in reduction rates was observed except that bulkier groups generally decreased reduction rates. Oxygen consumption is a measure of the ability of the reduced quinone (hydroquinone) to redox cycle following reduction by NQO1. This could lead to production of toxic reactive oxygen species and ultimately to cell death. Oxygen consumption was measured for select quinolinequinones, and the trend, if not the magnitude, mirrored the reduction rates. (Table 2).

Cell survival was measured using the MTT colorimetric assay. In previous work, we demonstrated that IC50 values generated from standard clonogenic assays and MTT assays were positively correlated suggesting that the MTT assay is a reliable indicator of cytotoxicity.6 We utilized MDA-MB-468 human breast cancer cells stably transfected with human NQO1 cDNA (MDA468-NQ16) along with the non-transfected wild type cells (MDA468) to compare the cytotoxicity of the quinolinequinones (Table 3).27

Table 3.

Cytotoxicity of quinoline-5,8-diones toward MDA468-WT (NQO1-deficient) and MDA468-NQ16 (NQO1-rich) human breast cancer cell lines.

graphic file with name nihms475173t32.jpg

No R1 R2 IC50 (µM)
MDA468-WT
IC50 (µM)
MDA468-NQ16
Selectivity Ratio
IC50 (WT)/
IC50 NQ16
9 CH3CO graphic file with name nihms475173t33.jpg 1.7 +/− 0.8 2.4 +/− 1.9 0.73
10 CH3CO graphic file with name nihms475173t34.jpg 3.3 +/− 0.1 6.3 +/− 0.2 0.52
11 CH3CO graphic file with name nihms475173t35.jpg 0.80 +/− 0.33 0.64 +/− 0.41 1.2
13 CH3CO graphic file with name nihms475173t36.jpg 0.53 +/− 0.27 2.2 +/− 0.5 0.24
17 CH3CO graphic file with name nihms475173t37.jpg 7.4 +/− 5.0 19.1 +/− 5.9 0.39
19 H graphic file with name nihms475173t38.jpg 5.3 +/− 0.8 17 +/− 5 0.31
20 H graphic file with name nihms475173t39.jpg 5.6 +/− 1.3 15 +/− 2 0.37
21 H graphic file with name nihms475173t40.jpg 4.8 +/− 0.9 10 +/− 1 0.47
22 H graphic file with name nihms475173t41.jpg 0.14 +/− 0.02 0.19 +/− 0.04 0.75
23 H graphic file with name nihms475173t42.jpg 19 +/− 12 5.3 +/− 2.1 3.5
24 H graphic file with name nihms475173t43.jpg 4.5 +/− 1.9 17 +/− 2 0.26

Quinolinequinone cytotoxicity (IC50) to MDA468 cells was generally in the single digit micromolar range following 2-h exposures with some in the high nanomolar range (11, 13, 22). Surprisingly, selectivity ratios [IC50 (MDA468) / IC50 (MDA468-NQ16)] were generally <1 meaning that the quinolinequinones were less cytotoxic to the NQO1-rich MDA468-NQ16 cells rather than more cytotoxic. This suggests that NQO1 was protective to the cells rather than functioning as an activating enzyme.27 Only two compounds (11, 23) were selectively cytotoxic to the MDA468-NQ16 cells. The reason for the general absence of selective cytotoxicity with this particular series of compounds is unclear, but it is consistent with NQO1’s primary role as a detoxification enzyme.27

Molecular docking of the quinolinequinones in the NQO1 active site was performed using Sybyl 8.1.1 and GOLD 5.1 for scoring. Three good NQO1 substrates (13, 20, 24) and three poor NQO1 substrates (9, 11, 17) were docked and scored using ChemPLP and ChemScore (Table 4). The highest scores representing a good fit for the model were found for 20 and 24 consistent with the metabolism data. The exception again was 13, which scored the lowest, but was the best substrate. Interatomic distances between quinolinequinone carbonyl groups and FAD N5 and His161 were shortest for 20, but all were within a reasonable distance for hydride transfer from FAD when the dynamic effects of the quinone-enzyme interaction are considered. Figure 5 shows possible docking conformations for 20 and 11 with NQO1. All quinolinequinones orient with the quinone ring above the FAD isoalloxazine ring as needed for hydride transfer.

Table 4.

Computational parameters for selected quinoline-5,8-diones.

graphic file with name nihms475173t44.jpg

No R1 R2 ChemPLP ChemScore C=O8…
NH5 (Ǻ)
C=O5…
His161NE2 (Ǻ)
9 CH3CO graphic file with name nihms475173t45.jpg 63.2 22.6 3.9 3.6
11 CH3CO graphic file with name nihms475173t46.jpg 63.8 22.6 4.7 3.3
13 CH3CO graphic file with name nihms475173t47.jpg 57.6 21.4 4.3 3.5
17 CH3CO graphic file with name nihms475173t48.jpg 63.8 22.1 4.2 3.3
20 H graphic file with name nihms475173t49.jpg 72.8 26.0 3.6 3.2
24 H graphic file with name nihms475173t50.jpg 67.3 22.7 4.1 3.3

Figure 5.

Figure 5

Quinolinequinones docked in NQO1 active site: 20, cyan; 11, magenta; FAD, green.

The mechanism of action of lavendamycin and streptonigrin is not clearly understood. However previous studies demonstrated that quinone moieties are reduced by NQO1 to the corresponding hydroquinones which undergo auto-oxidation producing activated oxygen species including not only the semiquinone derivatives but also superoxide and hydroxyl radicals.28 In addition, both streptonigrin and lavendamycin chelate divalent cationic metal ions. This property might confer to streptonigrin and lavendamycin the ability to shuttle iron cations into the cells which in turn can catalyze production of reactive oxygen species through a Fenton reaction. On the other hand, this chelation can result in depletion of intracellular cationic metals which might result in cell death.29 Generation of the semiquinone radical, after reduction of the quinone to the hydroquinone followed by auto-oxidation, results in a decrease of activity in 9 compounds. The best NQO1 substrates are less active compounds (13, 20, 24) in NQO1 expressing cells. In contrast, poor NQO1 substrates such as compound 22 or 11, exhibit the best activity in both cancer cells expressing NQO1 and not expressing NQO1. According to the NMR experiments, the quinoline derivative 22 and compound 13 binds the Zn2+ more efficiently than compound 19; and only one equivalent of Zn2+ is enough to cause important chemical shift variations. Similar observations were made with compound 23, which was less cytotoxic than compound 22. Even though metal chelation by these compounds is still a plausible mechanism to explain their activity against breast cancer cells, another mode of action cannot be discarded. Most active compounds (11, 13 and 22) are potential tridentate ligands for metals. Compound 23 exhibits lower activity than the corresponding acetylated amino analogue 13. It was proposed that metals can assist tautomeric shift from the active quinone analogues to the quinoid analogue which has an isoelectronic structure with the biologically inactive azastreptonigrin.24 This tautomeric shift can explain the decrease of activity of the amino derivative compared to the amido derivative. In our series of aryl substituted quinonequinolines the active molecule is the quinone derivatives and not the semiquinone derivatives. A similar mode of action than the bidentate metal ligands derivatives 8-hydroxyquinoline is currently under investigation.30, 31

CONCLUSIONS

A ten-step synthetic scheme led to good yields for quinolinequinone analogs of lavendamycin projected as NQO1-directed antitumor agents. Unexpectedly, ten of eleven analogs demonstrated excellent cytotoxicity (IC50 values of single digit micromolar or better) towards MDA468 breast cancer cells, but only two were selectively cytotoxic to NQO1-expressing MDA468-NQ16 cells. Compounds 22 and 11 are poor NQO1 substrates and exhibit the best activity against breast cancer cells. In our novel series of aryl substituted quinonequinolines the active molecule appears to be the quinone derivatives and not the semiquinone derivatives resulting from NQO1 reduction, suggesting that the mode of action of this novel series differs from lavendamycin and involves an unidentified target. Quinolinequinone derivatives 11, 13 and 22 cytotoxicities (IC50) to MDA468 cells were in the high nanomolar range. Our results seem to indicate that compounds 11, 13 and 22 effects could be also, at least partially, mediated by metal chelation. These aryl quinonequinoline derivatives represent a promising class of cytotoxic agents with a potential therapeutic value.

EXPERIMENTAL SECTION

Cell Culture

MDA-MB-468 (MDA468) human breast cancer cells and stably NQO1-transfected MDA468-NQ1632 were a gift from Dr. David Ross (University of Colorado-Denver, Denver, CO). MDA468 cells had no measurable NQO1 activiy whereas activity in MDA468-NQ16 cells was 1070 nmol/min/mg total cell protein using dichlorophenolindophenol (DCPIP) as the standard electron acceptor. Cells were grown in RPMI 1640 medium with L-glutamine and penicillin/streptomycin, and supplemented with 10% fetal bovine serum (FBS). Cell culture medium and supplements were obtained from Invitrogen (Carlsbad, CA). The cells were incubated at 37 °C under a humidified atmosphere containing 5% CO2.

Spectrophotometric cytochrome c assay

Quinolinequinone reduction was monitored using a spectrophotometric assay in which the rate of reduction of cytochrome c was quantified at 550 nm. Briefly, the assay mixture contained cytochrome c (70 µM), NADH (1 mM), recombinant human NQO1 (0.1–10 µg) (gift from Dr. David Ross, University of Colorado-Denver, Denver, CO) and quinonlinequinones (25 µM) in a final volume of 1 mL Tris-HCl (25 mM, pH 7.4) containing 0.7 mg/mL BSA and 0.1% Tween-20. Reactions were carried out at room temperature and started by the addition of NADH. Rates of reduction were calculated from the initial linear part of the reaction curve (0–30 s) and results were expressed in terms of µmol of cytochrome c reduced/min/mg of NQO1 using a molar extinction coefficient of 21.1 mM−1 cm−1 for cytochrome c. All reactions were carried out at least in triplicate.

Oxygen Consumption

Oxygen concentration was monitored using a MI-730 Micro-Oxygen Electrode (Microelectrodes, Bedford, NH) with concentrations adjusted for temperature (25 °C). Assay mixtures contained 25 µM quinonlinequinones, 200 µM NADH and 1 µg/mL NQO1 in a 2 mL Tris-Hcl-BSA/Tween (0.1%) buffer system. Reactions were started with NADH and measured over 3 minute intervals at room temperature. All reactions were carried out in triplicate.

Electrochemistry

Cyclic Voltammagrams were collected for 10 analogues using a BAS CV-50W electrochemical analyzer using a standard 3 electrode cell. Experiments were performed using an Ag/AgCl reference electrode, a glossy carbon working electrode and a platinum wire auxiliary electrode. The reported potentials are referenced by the Ferrocene (0/+) couple in the solvent used, primarily THF, which occurs at +.60V vs. Ag/AgCl. The compounds were run at concentrations of 1mM in THF, except compound 15 which was run in DMSO, with a.1M concentration of tetrabutylammonium hexafluorophosphate as a supporting electrolyte. All samples were purged and run under an Ar atmosphere during the course of the experiment, and the electrodes were washed and wiped down between each sample. Each CV was collected at a sweep rate of 50mV/s in the potential range of 0V to −2V at room temperature of 21°C.

NMR spectroscopy

One-dimensional 1H NMR spectra were recorded at room temperature on Bruker Avance IIITM spectrometer (The Woodlands, Texas) at 400 MHz using a 5-mm probe and a simple pulse-acquire sequence. Acquisition parameters consisted of spectral width of 4000 Hz with an acquisition time 3.98 s, number of scans of 128, and relaxation delay of 1 s. Complexes were prepared in a mixture of CDCl3 and THF-D8.

Cell Viability Assay

Growth inhibition was determined using the MTT colorimetric assay. Cells were plated in 96-well plates at a density of 10,000 cells/mL and allowed to attach overnight (16 h). Quinolinequinone solutions were applied in medium for 2 hours, removed and replaced with fresh medium, and the plates were incubated at 37 °C under a humidified atmosphere containing 5% CO2 for 3–5 days. MTT (50 µg) was added and the cells were incubated for another 4 hours. Medium/MTT solutions were removed carefully by aspiration, the MTT formazan crystals were dissolved in 100 µL DMSO, and absorbance was determined on a plate reader at 560 nm. IC50 values (concentration at which cell survival equals 50% of control) were determined from semi-log plots of percent of control vs. concentration. Selectivity ratios were defined as the IC50 value for the MDA468 cell line divided by the IC50 value for the MDA468-NQ16 cell line.

Molecular Modeling

For docking purposes, the crystallographic coordinates of the human NQO1 complex with 3-(hydroxymethyl)-5-(2-methylaziridin-1-yl)-1-methyl-2-phenylindole-4,7-dione (25) were obtained from the Brookhaven Database (PDB code 1H6933 and resolution 1.86Ǻ) and was edited accordingly to provide a monomer of the protein. The protein complex was then minimized within Sybyl 7.3 (Tripos Ltd., St Louis) while holding all heavy atoms stationary. The ligand was then removed to leave the receptor complex which was used for the subsequent docking studies. For preparation of ligand structures, fragments from Sybyl 8.1.1 were used to construct the compounds and all symmetric compounds were prepared as monoanionic ligands. Ligands were subject to 1000 iterations of energy minimization using the Powell method with MMFF94s force field standard method. For computational docking, the GOLD 5.1 software was used in combination with the ChemPLP34 scoring function (rescoring with ChemScore.35 The active site was defined as being any volume within 8Ǻ of the quinone scaffold of 25 in its crystal pose in 1H69. Each GA run comprised using the default parameters of: 100000 genetic operations on an initial population of 100 members divided into five subpopulations with weights for crossover, mutation, and migration being set to 95, 95, and 10, respectively. GOLD allows a user-definable number of GA runs per ligand, each of which starts from a different orientation. For these experiments, the number of GA runs was set to 10, and scoring of the docked poses was performed with the ChemPLP scoring function using ChemScore rescore. Each GOLD run was saved and the strongest scoring binding pose of each ligand (subject to a rmsd default distance threshold of 1.5Ǻ) was compared to that of the reference ligand position observed in the crystal structure. The best output pose (orientations) of the ligands generated were analyzed based on its ChemPLP/ChemScore score, feasibility of hydride transfer process and H-bonding to the enzyme. The best pose(s) were visualized using PyMOL Molecular Graphics System version 1.3.

Chemistry

All moisture sensitive reactions were performed in an inert, dry atmosphere of argon in flame dried glassware. Air sensitive liquids were transferred via syringe or cannula through rubber septa. Reagent grade solvents were used for extraction and flash chromatography. THF was distilled from Na/benzophenone under argon; dichloromethane (CH2Cl2) and chloroform (CHCl3) were distilled from CaH2 under argon. All other reagents and solvents which were purchased from commercial sources, were used directly without further purification. The progress of reactions was checked by analytical thin-layer chromatography (Sorbent Technologies, Silica G TLC plates w/UV 254). The plates were visualized first with UV illumination followed by charring with ninhydrin (0.3% ninhydrin (w/v), 97:3 EtOH-AcOH). Flash column chromatography was performed using prepacked Biotage SNAP cartridges on a Biotage Isolera One instrument. Microwave reactions were performed using a Biotage Initiator instrument. The solvent compositions reported for all chromatographic separations are on a volume/volume (v/v) basis. 1HNMR spectra were recorded at 400 or 500 MHz and are reported in parts per million (ppm) on the δ scale relative to tetramethylsilane as an internal standard. 13CNMR spectra were recorded at 100 or 125 MHz and are reported in parts per million (ppm) on the δ scale relative to CDCl3 (δ 77.00). Melting points were determined on a Stuart melting point apparatus from Bibby Scientific Limited and are uncorrected. High Resolution mass spectrometry (HRMS) was performed on a Waters/Micromass LCT-TOF instrument. All compounds were more than 95% pure.

5-chloro-8-hydroxy-7-nitroquinoline (1)

This compound was prepared according to the literature12 procedure to yield a yellow solid, 4.40 g (79%). M.p. 198–200°C, [lit.12, m.p. 192–194°C]; 1H NMR (500 MHz, DMSO) δ 9.09 (dd, J = 4.2, 0.5 Hz, 1H), 8.58 (dd, J = 8.5, 0.8 Hz, 1H), 8.18 (s, 1H), 7.94 (dd, J = 8.5, 4.3 Hz, 1H). 13C NMR (126 MHz, DMSO) δ 150.5, 150.1, 139.9, 133.6, 132.3, 128.5, 125.9, 122.0, 117.9. HRMS (TOF MS ES+) for C9H6ClN2O3+ (MH+) calcd. 225.0067, found 225.0055.

7-Amino-8-hydroxyquinoline (2)

Compound 1 (2.4 g, 10.69 mmol) was placed in a hydrogenation apparatus equipped with a magnetic stir bar and methanol added. Pd/C (150 mg) in a small amount of MeOH (60 mL) was added and stirring commenced. H2 gas was introduced at a pressure of 40–50 psi and reacted at rt overnight. TLC showed full conversion. The black solution was filtered using a celite pad and concentrated under reduced pressure to yield 2 as a black oil, 99% yield. 1H NMR (500 MHz, CDCl3) δ 8.66 (dd, J = 4.4, 1.6 Hz, 1H), 8.03 (dd, J = 8.2, 1.6 Hz, 1H), 7.24 (d, J = 8.7 Hz, 1H), 7.17 (dd, J = 8.2, 4.4 Hz, 1H), 7.10 (d, J = 8.7 Hz, 1H). 13C NMR (126 MHz, CDCl3) δ 148.0, 137.9, 136.6, 136.1, 132.1, 122.4, 119.3, 118.5, 117.7. HRMS (TOF MS ES+) for C9H9N2O+ (MH+) calcd. 161.0715, found 161.0707.

7-acetamido-8-acetyloxyquinoline (3)

Compound 2 (330 mg, 2.06 mmol) was dissolved in dried THF (10 mL) and DIEA added with stirring. AcCl (176 µL) in 1mL THF was added drop wise while stirring and reacted at rt for 2 hrs. Then concentrated under reduced pressure followed by redissolving in CH2Cl2 (20 mL) and water (10 mL). The two layers were allowed to partition and extracted 2× 20 mL CH2Cl2. The combined organic layers were dried over MgSO4, filtered and concentrated under reduced pressure. Then purified on a Biotage SNAP cartridge (25 g) at a flow rate of 25 mL/min to yield an orange solid, 382 mg (76%); m.p. 151–153°C; 1H NMR (500 MHz, CDCl3) δ 8.85 (dd, J = 4.1, 1.3 Hz, 1H), 8.49 (d, J = 9.1 Hz, 1H), 8.13 (dd, J = 8.3, 1.5 Hz, 1H), 7.70 (d, J = 9.1 Hz, 1H), 7.67 (s, 1H), 7.36 (dd, J = 8.2, 4.2 Hz, 1H), 2.56 (s, 1H), 2.04 (s, 1H); 13C NMR (126 MHz, CDCl3) δ 169.7, 168.5, 150.6, 140.7, 135.8, 134.9, 130.8, 125.8, 125.6, 121.3, 120.6, 24.5, 21.0; HRMS (TOF MS ES+) for C13H13N2O3+ (MH+) calcd. 245.0926, found 245.0923.

7-acetamido-8-benzyloxyquinoline (4)

To a solution of 3 (1.2 g, 4.91 mmol) in MeOH (100 mL) was added water (10 mL) and the reaction stirred under reflux for 1 hr. The black solution was concentrated and in vacuo and flash chromatographed on a KP-Sil 100 g Biotage SNAP cartridge using MeOH: DCM as the solvent (0–5% MeOH). A white solid (0.9 g) obtained and used for the next step directly. Rf= 0.11 (5% MeOH:CH2Cl2).

To a solution of 7-acetamido-8-hydroxyquinoline (2.27 g, 11.23 mmol) in 40 mL DMF was added K2CO3 (2.33 g, 16.80 mmol) and BnBr (2 mL, 16.80 mmol) respectively. The reaction was stirred at 50°C for 24 hrs after which TLC showed almost all the starting material was consumed. The reaction mixture was diluted with 30 mL CH2Cl2, filtered with a pad of celite and concentrated under reduced pressure. The residue was loaded onto a 100 g Biotage SNAP cartridge by dissolving in a small amount of CH2Cl2 and eluted with EtOAc:heptane gradient (0–50%). Yield 2.95 g (90%) of a yellow oil was obtained. Rf= 0.50 (60% EtOAc:heptane). 1H NMR (500 MHz, CDCl3) δ 8.95 (dd, J = 4.2, 1.7 Hz, 1H), 8.58 (d, J = 9.0 Hz, 1H), 8.14 (dd, J = 8.3, 1.7 Hz, 1H), 7.77 (s, 1H), 7.57 (d, J = 9.0 Hz, 1H), 7.40 – 7.35 (m, 6H), 5.49 (s, 2H), 1.93 (s, 4H). 13C NMR (126 MHz, CDCl3) δ 168.3, 150.0, 142.0, 141.0, 137.4, 136.2, 132.0, 128.9, 128.8, 128.8, 126.0, 124.0, 120.0, 120.0, 77.3, 24.6. HRMS (TOF MS ES+) for C18H17N2O2+ (MH+) calcd. 293.1290, found 293.1264.

7-acetamido-8-(benzyloxy)quinoline-1-oxide (5)

The starting material (4) (428 mg, 1.46 mmol) was dissolved in 4.3 mL 1,2-dichloroethane with stirring. The mCPBA (340 mg, 1.76 mmol) was added (0.5 M) and the reaction stirred at rt for 48 hrs. TLC showed almost all the starting material was consumed. The precipitated mCPBA was filtered and washed with 5 mL 1, 2-dichloroethane. The filtrate was concentrated under reduced pressure and flash chromatographed on a KP-sil 100 g Biotage SNAP cartridge using a 5% MeOH: DCM gradient at a flow rate of 25 mL/min to yield a yellow solid, 373 mg (82%). M.p. 145–147°C; Rf 0.24 (5%MeOH:DCM). 1H NMR (500 MHz, DMSO) δ 9.45 (s, 1H), 8.46 (d, J = 6.1 Hz, 1H), 8.20 (d, J = 8.9 Hz, 1H), 7.81 (d, J = 8.3 Hz, 1H), 7.77 (d, J = 9.0 Hz, 1H), 7.58 – 7.50 (2H), 7.40 – 7.30 (aromatic, 4H). 13C NMR (126 MHz, DMSO) δ 168.9, 139.8, 138.1, 137.1, 136.4, 133.3, 129.8, 129.1, 128.1, 128.0, 124.7, 124.4, 120.8, 77.7, 23.8. HRMS (TOF MS ES+) for C18H17N2O3+ (MH+) calcd. 309.1239, found 309.1227.

7-acetamido-8-benzyloxy-2-chloroquinoline (6)

Phosphoryl chloride (280 µL, 3.0 mmol) in CHCl3 (1.0 mL) was added to a stirred solution of the oxide 5 (770 mg, 2.50 mmol) in 21 mL CHCl3 and stirred for 15 min. The mixture was then refluxed for 2 hrs, cooled and poured into ice (50 g) and the pH adjusted to 12 with NaOH (aq.). The aq. layer was extracted with 2 × 50 mL CH2Cl2, washed with 2 × 20 mL H2O, dried over MgSO4, filtered and concentrated under reduced pressure to yield a brown oil. Then purified on a HP-Sil 25 g Biotage SNAP cartridge using EtOAc:heptane gradient (0–50%) as the solvent. Yield 504 mg (62%) of an off-white solid was obtained. Rf= 0.58 (60% EtOAc:heptane); M.P. 92–94°C; 1H NMR (500 MHz, CDCl3) δ 8.60 (d, J = 9.0 Hz, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.81 (s, 1H), 7.54 (d, J = 9.0 Hz, 1H), 7.45 – 7.35 (m, 1H), 7.32 (d, J = 8.5 Hz, 1H), 5.48 (s, 1H), 1.96 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 168.4, 150.5, 141.4, 140.3, 139.0, 137.2, 133.0, 128.9, 128.8, 128.8, 124.3, 123.3, 121.1, 120.1, 77.4, 24.7. HRMS (TOF MS ES+) for C18H16ClN2O2+ (MH+) calcd. 327.0900, found 327.0936.

7-acetamido-2-chloro-8-hydroxyquinoline (7)

To a solution of 6 (330 mg, 1.01 mmol) in CH2Cl2 (10.1 mL) under an Ar atmosphere was added BCl3•SMe2 (10.1 mL) via a syringe and stirred at rt overnight. TLC showed the reaction was complete. The reaction was then quenched with saturated NaHCO3(aq.) and extracted with 2x20 mL CH2Cl2. The organic layers were combined, dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified on 50 g KP-Sil Biotage SNAP cartridge using a MeOH: CH2Cl2 gradient (0–5% MeOH) at a flow rate of 25 mL/minute to give a yellow solid, 198 mg (82%). M.P. 176–178°C; Rf= 0.50 (5% MeOH:CH2Cl2). 1H NMR (400 MHz, CDCl3) δ 8.60 (d, J = 9.0 Hz, 1H), 8.05 (d, J = 8.5 Hz, 1H), 7.82 (brs, 1H), 7.72 (s, 1H), 7.35 (d, J = 9.0 Hz, 1H), 7.30 (d, J = 8.5 Hz, 1H), 2.29 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 168.6, 149.7, 138.9, 138.2, 137.1, 124.4, 123.3, 121.5, 121.3, 118.0, 24.9. HRMS (TOF MS ES+) for C11H10ClN2O2+ (MH+) calcd. 237.0431, found 237.0424.

7-Acetamido-2-chloroquinoline-5,8-dione (8)

To a solution of 7 (300 mg, 1.27 mmol) in acetone (30 mL) was added a solution of Fremy’s salt in NaH2PO4 buffer (0.3 M, 30 mL) and the mixture stirred at rt for 1hr. A further solution of Fremy’s salt in the buffer (0.3M, 30 mL) was added and stirring continued for 2 hrs. The acetone was removed under reduced pressure and the residue extracted with 2 × 50 mL CH2Cl2. The CH2Cl2 phases were combined, dried over MgSO4 and concentrated under reduced pressure. The residue was purified on a 25 g HP-Sil Biotage SNAp cartridge using EtOAc:heptanes gradient (0–60%) to obtain a yellow solid, 225 mg (71% over 2 steps); m.p. 224–226°C (decomposes into a black mass), Rf= 0.49 (60% EtOAc:heptane). 1H NMR (500 MHz, CDCl3) δ 8.41 (s, 1H), 8.39 (d, J = 8.2 Hz, 1H), 7.97 (s, 1H), 7.74 (d, J = 8.2 Hz, 1H), 2.34 (s, 3H). 13C NMR (126 MHz, CDCl3) δ 183.4, 178.1, 169.5, 156.7, 145.9, 140.4, 137.2, 129.9, 128.0, 116.3, 25.1. HRMS (TOF MS ES+) for C11H8ClN2O3+ (MH+) calcd. 251.0223, found 250.0203.

General procedure for Suzuki coupling under microwave conditions

The 7-acetamido-2-chloroquinoline-5,8-dione 8 (21 mg, 0.08 mmol) was dissolved in 4 mL dimethoxyethane (DME) and degassed under reduced pressure. The palladium (0) catalyst, Pd(PPh3)4 (10 mg, 0084 mmol) was added and the solution degassed further. The mixture was stirred under Ar atmosphere for 10 minutes. Na2CO3 solution (0.2 mL, 2.0 M) was added followed by the boronic acid (0.126 mmol). The mixture was then heated using a Biotage microwave initiator at 110–140°C for 20 minutes. After cooling, TLC showed all the starting material was consumed. The reaction mixture was poured into DCM and washed with 2 × 10 mL water. Then dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified on HP-Sil 25 g Biotage SNAP cartridge using EtOAc:heptane gradient (0–50%) at a flow rate of 20 mL/min. For very polar products, MeOH:CH2Cl2 (0–10%MeOH) was used as solvent for purification.

7-acetamido-2-(4-(trifluoromethyl)phenyl)quinoline-5,8-dione (9)

Yield 21 mg (70%) of a yellow solid was obtained. Rf= 0.47 (50% EtOAc:heptane); m.p. 250°C(decomposes); 1H NMR (500 MHz, CDCl3) δ 8.53 (d, J = 8.2 Hz, 1H), 8.45 (s, 1H), 8.27 (d, J = 8.1 Hz, 2H), 8.17 (d, J = 8.2 Hz, 1H), 7.99 (s, 1H), 7.80 (d, J = 8.2 Hz, 2H), 2.35 (s, 4H).13C NMR (126 MHz, CDCl3) δ 184.1, 179.1, 169.5, 160.1, 146.1, 140.6, 135.7, 128.3, 128.0, 126.0, 126.0, 126.0, 125.3, 116.5, 25.2; HRMS (TOF MS ES+) for C18H12F3NS2O3+(MH+) calcd. 361.0800, found 361.0834.

7-acetamido-2-(3-pyridinyl))quinoline-5,8-dione (10)

Yield 21 mg (41%) of a yellow solid obtained, Rf= 0.19 (5% MeOH:DCM); m.p. >300°C(decomposes); 1H NMR (500 MHz, CDCl3) δ 9.29 (s, 1H), 8.72 (d, J = 3.9 Hz, 1H), 8.56 (d, J = 8.2 Hz, 1H),8.55 (m, 1H), 8.21 (d, J = 8.2 Hz, 1H), 8.00 (s, 1H), 7.56 (dd, J = 8.0, 4.9 Hz, 1H), 2.35 (s, 3H). 13C NMR (126 MHz, CDCl3) δ 184.2, 179.0, 170.4, 158.9, 150.7, 148.1, 146.1, 140.9, 135.7, 135.7, 128.1, 125.3, 116.6, 24.6 ; HRMS (TOF MS ES+) for C16H12N3O3+MH+) calcd. 294.0879, found 294.0914. 7-amino-2-(3-pyridinyl)quinoline-5,8-dione: 6 mg (12%) of a red solid was obtained. Rf= 0.13 (5% MeOH:DCM); m.p. 195–197°C (decomposes, turns black); 1H NMR (500 MHz, CDCl3) δ 9.29 (d, J = 1.7 Hz, 1H), 8.67 (dd, J = 4.9, 1.4 Hz, 1H), 8.58 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.52 (d, J = 8.2 Hz, 1H), 8.22 (d, J = 8.2 Hz, 1H), 7.59 (ddd, J = 8.0, 4.9, 0.7 Hz, 1H), 6.07 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 181.9, 179.8, 157.0, 150.4, 149.6, 147.6, 146.3, 135.4, 135.0, 133.5, 129.4, 124.7, 123.8, 102.1. HRMS (TOF MS ES+) for C14H10N3O2+MH+) calcd. 252.0773, found 252.0795.

7-acetamido-2-(8’-quinolinyl)quinoline-5,8-dione (11)

Yield 31 mg (51%) of a yellow solid was obtained. Rf= 0.25 (70% EtOAc:heptane), crystallized from MeOH/CH2Cl2; m.p. 295°C (decomposes); 1H NMR (500 MHz, CDCl3) δ 8.93 (dd, J = 4.2, 1.8 Hz, 1H), 8.53 (d, J = 8.1 Hz, 1H), 8.48 (d, J = 8.1 Hz, 1H), 8.34 (dd, J = 8.3, 1.8 Hz, 1H), 8.22 (dd, J = 7.2, 1.4 Hz, 1H), 8.04 (dd, J = 8.2, 1.4 Hz, 1H), 8.00 (s, 1H), 7.76 (dd, J = 8.1, 7.3 Hz, 1H), 7.54 (dd, J = 8.3, 4.2 Hz, 1H), 2.34 (s, 3H). 13C NMR (126 MHz, CDCl3) δ 184.8, 179.1, 170.9, 161.9, 150.5, 145.7, 145.1, 140.9, 136.7, 136.5, 133.3, 131.9, 131.7, 130.2, 128.4, 127.6, 126.3, 121.4, 116.5, 24.2. HRMS (TOF MS ES+) C20H14N3O3+ (MH+) calcd. 344.1035, found 344.1022.

7-acetamido-2-(2-(1-tert-butoxycarbonylindolyl))quinoline-5,8-dione (12)

Yield 63 mg (67%) of an orange solid was obtained. Rf= 0.40 (50% EtOAc:heptane); m.p. 191–193°C (decomposes); 1H NMR (500 MHz, CDCl3) δ 8.47 (s, 1H), 8.45 (d, J = 8.1 Hz, 1H), 8.15 (d, J = 8.4 Hz, 1H), 7.97 (s, 1H), 7.89 (d, J = 8.1 Hz, 1H), 7.60 (d, J = 7.8 Hz, 1H), 7.40 (t, J = 7.8 Hz, 1H), 7.27 (dd, J = 9.1, 5.9 Hz, 1H), 6.98 (s, 1H), 2.33 (s, 3H), 1.41 (s, 9H). 13C NMR (126 MHz, CDCl3) δ 184.2, 179.0, 169.6, 157.4, 149.7, 145.3, 140.4, 138.1, 137.5, 134.2, 128.6, 127.9, 127.6, 126.0, 123.3, 121.5, 116.5, 115.2, 114.0, 84.2, 27.8, 25.1. HRMS (TOF MS ES+) C24H22N3O5+(MH+) calcd. 432.1559, found 432.1568.

7-acetamido-2-(2-pyridinyl)quinoline-5,8-dione (13)

Yield 37 mg (71%) of a yellow solid was obtained. Rf= 0.19 (5% MeOH:CH2Cl2), crystallized from MeOH/CH2Cl2; m.p. 255–258°C (decomposes); 1H NMR (500 MHz, DMSO) δ 10.08 (s, 1H), 8.78 (ddd, J = 4.8, 1.6, 0.8 Hz, 1H), 8.53 (d, J = 7.9 Hz, 1H), 8.46 (d, J = 8.2 Hz, 1H), 8.08 (td, J = 7.7, 1.8 Hz, 1H), 7.77 (s, 1H), 7.58 (ddd, J = 7.5, 4.7, 1.1 Hz, 1H), 2.28 (s, 3H). 13C NMR (126 MHz, DMSO) δ 184.6, 178.4, 171.5, 158.6, 153.6, 149.8, 146.4, 142.5, 137.8, 135.0, 128.5, 125.5, 124.6, 121.7, 115.3, 24.7. HRMS (TOF MS ES+) C16H12N3O3+ (MH+) calcd. 294.0879, found 294.0914.

7-acetamido-2-(2-(1-tert-butoxycarbonylpyrrolyl))quinoline-5,8-dione (14)

Yield 36 mg (53%) of a yellow solid was obtained. Rf= 0.30 (50% EtOAc:heptane); m.p. 191–193°C (decomposes), recrystallized from methanol; 1H NMR (500 MHz, CDCl3) δ 8.42 (s, 1H), 8.39 (d, J = 8.2 Hz, 1H), 7.95 (s, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.42 (dd, J = 3.2, 1.7 Hz, 1H), 6.64 (dd, J = 3.4, 1.7 Hz, 1H), 6.29 (t, J = 3.3 Hz, 1H), 2.32 (s, 3H), 1.43 (s, 9H). 13C NMR (126 MHz, CDCl3) δ 184.3, 179.2, 169.5, 156.9, 148.8, 145.3, 140.3, 134.0, 132.5, 128.0, 127.3, 125.5, 118.6, 116.4, 111.2, 84.4, 27.7, 25.1. HRMS (TOF MS ES+) C20H20N3O5+ (MH+) calcd. 382.1403, found 382.1381.

7-acetamido-2-(4-pyrazolyl))quinoline-5,8-dione (15)

Yield 31 mg (42%) of a brown solid was obtained. Rf= 0.33 (5% MeOH:CH2Cl2); m.p. 270°C (decomposes), recrystallized from methanol; 1H NMR (500 MHz, DMSO) δ 13.34 (s, 1H), 9.97 (s, 1H), 8.55 (s, 1H), 8.25 (d, J = 8.2 Hz, 1H), 8.21 (s, 1H), 8.11 (d, J = 8.2 Hz, 1H), 7.69 (s, 1H), 2.26 (s, 3H). 13C NMR (126 MHz, DMSO) δ 184.6, 178.6, 171.4, 156.1, 146.6, 142.1, 134.2, 126.1, 123.8, 121.2, 115.1, 24.6. HRMS (TOF MS ES+) C14H11N4O3+ (MH+) calcd. 283.0831, found 283.0846.

7-acetamido-2-(3-(2-acetamido-pyridinyl))quinoline-5,8-dione (16)

The quinone 8 (71 mg, 0.28 mmol) was dissolved in 2 mL 1,4-dioxane and degassed under reduced pressure. PdCl2(dppf) (20 mg), K3PO4 (238 mg) and the boronate were added and the solution degassed further. The mixture was stirred under Ar atmosphere for 10 minutes. The mixture was then heated heated using a Biotage microwave initiator at 120°C for 30 minutes. After cooling, the reaction mixture was poured into CH2Cl2 and washed with 2 × 10 mL water and extracted 2× 30 mL DCM. The combined organic phases were dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified on a HP-Sil 25 g Biotage SNAP cartridge using MeOH:CH2Cl2 gradient (0–5%) at a flow rate of 20 mL/min. Yield 23 mg (23%) of a brown solid was obtained. Rf= 0.32 (5% MeOH:CH2Cl2); m.p. 249°C (decomposes); 1H NMR (500 MHz, DMSO) δ 10.82 (s, 1H), 10.04 (s, 1H), 9.17 (d, J = 2.5 Hz, 1H), 8.60 (dd, J = 8.8, 2.5 Hz, 1H), 8.45 (d, J = 8.3 Hz, 1H), 8.38 (d, J = 8.2 Hz, 1H), 8.27 (d, J = 8.8 Hz, 1H), 7.75 (s, 1H), 2.28 (s, 3H), 2.14 (s, 3H). 13C NMR (126 MHz, DMSO) δ 184.6, 178.5, 171.5, 169.7, 157.4, 153.6, 147.3, 146.6, 142.4, 137.0, 134.8, 128.0, 127.2, 124.1, 115.3, 113.0, 24.7, 24.0. HRMS (TOF MS ES+) C18H15N4O4+ (MH+) calcd. 351.1093, found 351.1064.

7-acetamido-2-(2-indolyl)quinoline-5,8-dione (17)

The starting material 12 (39 mg, 0.09 mmol) was dissolved in 2.5 mL CH2Cl2 and cooled to 0°C using an ice bath. Trifluoroacetic acid (140 µL) was the added dropwise and reacted at rt for 2 hrs. TLC showed full conversion. Then quenched with sat. NaHCO3 (10 mL) and extracted 2×20 mL CH2Cl2. The organic layers were combined, dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified on a HP-Sil 25 g Biotage SNAP cartridge using EtOAc:heptane gradient (0–70%) at a flow rate of 20 mL/min. Yield 17 mg (59%) of a red solid was obtained after recrystallization from MeOH. M.p. 185°C, decomposes; Rf= 0.38 (70% EtOAc:heptane). 1H NMR (500 MHz, CDCl3) δ 8.35 (d, J = 8.3 Hz, 1H), 8.14 (d, J = 8.3 Hz, 1H), 7.92 (s, 1H), 7.67 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 8.3 Hz, 1H), 7.29 (t, J = 7.6 Hz, 1H), 7.23 (s, 1H), 7.13 (t, J = 7.4 Hz, 1H), 2.34 (s, 3H). 13C NMR (126 MHz, CDCl3) δ 184.2, 180.4, 170.5, 154.7, 145.4, 140.3, 137.8, 134.7, 134.4, 128.3, 126.6, 124.4, 124.4, 121.5, 120.2, 117.0, 111.7, 104.4, 24.4. HRMS (TOF MS ES+) C19H14N3O3+ (MH+) calcd. 332.1035, found 332.1030.

7-acetamido-2-(2-(pyrrolyl))quinoline-5,8-dione (18)

The starting material 14 (30 mg, 0.08 mmol) was dissolved in 3 mL CH2Cl2 and cooled to 0°C using an ice bath. Trifluoroacetic acid (150 µL) was the added dropwise and reacted at rt for 2 hrs. TLC showed full conversion. Then quenched with sat. NaHCO3 (10 mL) and extracted 2×20 mL CH2Cl2. The organic layers were combined, dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified on a HP-Sil 25 g Biotage SNAP cartridge using EtOAc:heptane gradient (0–50%) at a flow rate of 20 mL/min. Yield 21 mg (93%) of a red solid was obtained after recrystallization from MeOH. M.P. 255°C, decomposes. Rf= 0.11 (50% EtOAc:heptane). 1H NMR (500 MHz, DMSO) δ 11.65 (s, 1H), 9.95 (s, 1H), 8.20 (d, J = 8.4 Hz, 1H), 8.05 (d, J = 8.4 Hz, 1H), 7.69 (s, 1H), 7.07 – 7.04 (m, 2H), 6.28 – 6.22 (m, 1H), 2.27 (s, 3H). 13C NMR (126 MHz, DMSO) δ 184.6, 178.7, 171.4, 154.0, 146.6, 141.9, 133.9, 130.1, 125.3, 123.8, 121.8, 115.2, 111.9, 110.4, 24.7. HRMS (TOF MS ES+) C15H12N3O3+ (MH+) calcd. 282.0879, found 282.0909.

General procedure for removal of the acetate group with MeOH-H2SO4

To the starting material (0.1 mmol) in a 20 mL vial was added 175µL of H2SO4 in 3.0 mL MeOH and stirred at rt for 3 hrs. The red solution was then neutralized with 5 mL 5% NaHCO3 (aq.) and extracted with 5 × 10 mL CH2Cl2. The combined organic extracts were dried over MgSO4, filtered and concentrated under reduced pressure. Then purified on a HP-Sil 25 g Biotage SNAP cartridge using EtOAc:heptanes (0–70%) or MeOH:CH2Cl2 gradient (0–5%) at a flow rate of 20 mL/min.

7-Amino-2-(4-(trifluoromethyl)phenyl)quinoline-5,8-dione (19)

The general procedure was used to obtain 6.0 mg (67%) of a red solid; Rf= 0.38 (60% EtOAc:heptane); m.p. 151–153°C (decomposes, turns black); 1H NMR (500 MHz, CDCl3) δ 8.28 (d, J = 8.2 Hz, 1H), 8.05 (d, J = 8.2 Hz, 2H), 7.95 (d, J = 8.2 Hz, 1H), 7.58 (d, J = 8.3 Hz, 2H), 5.84 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 182.2, 180.1, 158.7, 150.3, 146.3, 140.6, 135.1, 129.4, 127.7, 125.5, 125.5, 125.1, 105.8, 102.4. HRMS (TOF MS ES+) C16H10F3N2O2+ (MH+) calcd. 319.0694, found 319.0666.

7-amino-2-(3-pyridinyl)quinoline-5,8-dione (20)

The general procedure was used to obtain 10 mg (83%) of a red solid. Rf= 0.16 (5% MeOH:CH2Cl2); m.p. 195–197°C (decomposes, turns black). 1H NMR (500 MHz, CDCl3) δ 9.29 (d, J = 1.7 Hz, 1H), 8.67 (dd, J = 4.9, 1.4 Hz, 1H), 8.58 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.52 (d, J = 8.2 Hz, 1H), 8.22 (d, J = 8.2 Hz, 1H), 7.59 (ddd, J = 8.0, 4.9, 0.7 Hz, 1H), 6.07 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 181.9, 179.8, 157.0, 150.4, 149.6, 147.6, 146.3, 135.4, 135.0, 133.5, 129.4, 124.7, 123.8, 102.1. HRMS (TOF MS ES+) for C14H10N3O2+MH+) calcd. 252.0773, found 252.0795.

7-amino-2-(2-indolyl)quinoline-5,8-dione (21)

The general procedure was used to obtain 19 mg (63%) of a dark-brown solid. Rf= 0.22 (70% EtOAc:heptane); m.p. 235°C decomposes. 1H NMR (500 MHz, CDCl3) δ 8.33 (d, J = 8.3 Hz, 1H), 8.10 (d, J = 8.3 Hz, 1H), 7.67 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 8.2 Hz, 1H), 7.39 (s, 1H), 7.27 (ddd, J = 8.1, 7.1, 1.1 Hz, 1H), 7.19 (s, 1H), 7.12 (td, J = 7.5, 0.8 Hz, 1H), 6.01 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 182.6, 181.6, 153.6, 149.5, 145.9, 137.6, 135.0, 134.2, 128.3, 128.0, 124.1, 124.0, 121.3, 120.0, 111.7, 103.5, 102.9. HRMS (TOF MS ES+) for C17H12N3O2+ (MH+) calcd. 290.0930, found 290.0900.

7-amino-2-(8-quinolinyl)quinoline-5,8-dione (22)

The general procedure was used to obtain 55 mg (71%) of a brown solid. Rf= 0.29 (5% MeOH:CH2Cl2); m.p. 243–245°C, recrystallized from MeOH. 1H NMR (500 MHz, CD3OD) δ 8.92 (dd, J = 4.2, 1.8 Hz, 1H), 8.49 (d, J = 8.1 Hz, 1H), 8.40 (d, J = 8.1 Hz, 1H), 8.33 (dd, J = 8.3, 1.8 Hz, 1H), 8.21 (dd, J = 7.2, 1.5 Hz, 1H), 8.02 (dd, J = 8.2, 1.4 Hz, 1H), 7.75 (dd, J = 8.1, 7.3 Hz, 1H), 7.53 (dd, J = 8.3, 4.2 Hz, 1H), 6.06 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 182.8, 180.1, 160.5, 150.3, 150.2, 146.2, 145.2, 136.7, 136.7, 133.2, 131.6, 131.5, 129.8, 129.1, 128.4, 126.3, 121.2, 102.4. HRMS (TOF MS ES+) for C18H12N3O2+ (MH+) calcd. 302.0930, found 302.0939.

7-amino-2-(2-pyridinyl)quinoline-5,8-dione (23)

The general procedure was used to obtain 16 mg (76%) of a red solid. Rf= 0.25 (20% MeOH:CH2Cl2), recrystallized from MeOH. 1H NMR (500 MHz, DMSO) δ 8.75 (d, J = 4.1 Hz, 1H), 8.72 (d, J = 8.2 Hz, 1H), 8.50 (d, J = 7.9 Hz, 1H), 8.40 (d, J = 8.1 Hz, 1H), 8.05 (t, J = 7.7 Hz, 1H), 7.58 – 7.53 (m, 1H), 5.89 (s, 1H). HRMS (TOF MS ES+) for C14H10N3O2+ (MH+) calcd. 252.0773 found 252.0749.

7-Amino-2-(2-pyrrolyl)quinoline-5,8-dione (24)

The general procedure was used to obtain 11 mg (78%) of a red solid. Rf= 0.37 (5% MeOH:CH2Cl2); m.p. 230°C (decomposes), recrystallized from MeOH. 1H NMR (500 MHz, CDCl3) δ 8.23 (d, J = 8.4 Hz, 1H), 7.84 (d, J = 8.4 Hz, 1H), 7.06 (dd, J = 2.5, 1.3 Hz, 1H), 6.91 (dd, J = 3.7, 1.3 Hz, 1H), 6.32 (dd, J = 3.7, 2.6 Hz, 1H), 5.97 (s, 1H). 13C NMR (126 MHz, CDCl3) δ 183.0, 181.7, 153.5, 149.4, 145.7, 133.9, 129.9, 126.6, 122.7, 122.3, 110.8, 110.3, 102.5. HRMS (TOF MS ES+) for C13H10N3O2+ (MH+) calc. 240.0773, found 240.0779.

Supplementary Material

1_si_001

ACKNOWLEDGEMENT

This work was supported by NIH grants P30NS055022 (PD and CMK) and P20RR017670 (HDB). We thank Dr. Ayesha Sharmin for experimental assistance and the CBSD NIH CoBRE (P20GM103546), which supports the BioSpectroscopy Core and the Molecular Computational Core Facility. Marvin was used for drawing, displaying and characterizing chemical structures, substructures and reactions included in the supporting information, Marvin 5.11.5, 2013, ChemAxon (http://www.chemaxon.com).

ABBREVIATIONS

NQO1

NAD(P)H: quinone oxidoreductase 1

MeOH

Methanol

BnBr

Benzyl bromide

DME

1, 2-Dimethoxyethane

dppf

1,1’-Bis(diphenylphosphino)ferrocene

Epc

cathodic peak potential

Epa

anodic peak potential

Footnotes

ASSOCIATED CONTENT

Supporting Information. Additional characterization data, 1H and 13C-NMR spectra. This material is available free of charge via the Internet at http://pubs.acs.org.

REFERENCES

  • 1.Balitz DMBJA, Bradner WT, Doyle TW, O'Herron FA, Nettleton DE. Isolation of lavendamycin. A new antibiotic from Streptomyces lavendulae. J. Antibiot. 1982;35:259–265. doi: 10.7164/antibiotics.35.259. [DOI] [PubMed] [Google Scholar]
  • 2.Doyle TW, Balitz DM, Grulich RE, Nettleton DE, Gould SJ, Tann C-h, Moews AE. Structure determination of lavendamycin- a new antitumor antibiotic from streptomyces lavendulae. Tetrahedron Lett. 1981;22:4595–4598. [Google Scholar]
  • 3.P RKVCW. Streptonigrin, an antitumor substance. I. Isolation and characterization. Antibiot. Annu. 1959;7:950–953. [PubMed] [Google Scholar]
  • 4.Rao KV, Biemann K, Woodward RB. The Structure of Streptonigrin. J. Am. Chem. Soc. 1963;85:2532–2533. [Google Scholar]
  • 5.Podeszwa B, Niedbala H, Polanski J, Musiol R, Tabak D, Finster J, Serafin K, Milczarek M, Wietrzyk J, Boryczka S, Mol W, Jampilek J, Dohnal J, Kalinowski DS, Richardson DR. Investigating the antiproliferative activity of quinoline-5,8-diones and styrylquinolinecarboxylic acids on tumor cell lines. Bioorg. Med. Chem. Lett. 2007;17:6138–6141. doi: 10.1016/j.bmcl.2007.09.040. [DOI] [PubMed] [Google Scholar]
  • 6.Hassani M, Cai W, Holley DC, Lineswala JP, Maharjan BR, Ebrahimian GR, Seradj H, Stocksdale MG, Mohammadi F, Marvin CC, Gerdes JM, Beall HD, Behforouz M. Novel Lavendamycin Analogues as Antitumor Agents: Synthesis, in Vitro Cytotoxicity, Structure–Metabolism, and Computational Molecular Modeling Studies with NAD(P)H:Quinone Oxidoreductase 1. J. Med. Chem. 2005;48:7733–7749. doi: 10.1021/jm050758z. [DOI] [PubMed] [Google Scholar]
  • 7.Fryatt T, Pettersson HI, Gardipee WT, Bray KC, Green SJ, Slawin AMZ, Beall HD, Moody CJ. Novel quinolinequinone antitumor agents: structure-metabolism studies with NAD(P)H:quinone oxidoreductase (NQO1) Bioorg. Med. Chem. 2004;12:1667–1687. doi: 10.1016/j.bmc.2004.01.021. [DOI] [PubMed] [Google Scholar]
  • 8.Beall HD, Winski S, Swann E, Hudnott AR, Cotterill AS, O'Sullivan N, Green SJ, Bien R, Siegel D, Ross D, Moody CJ. Indolequinone Antitumor Agents: Correlation between Quinone Structure, Rate of Metabolism by Recombinant Human NAD(P)H:Quinone Oxidoreductase, and in Vitro Cytotoxicity1. J. Med. Chem. 1998;41:4755–4766. doi: 10.1021/jm980328r. [DOI] [PubMed] [Google Scholar]
  • 9.Swann E, Barraja P, Oberlander AM, Gardipee WT, Hudnott AR, Beall HD, Moody CJ. Indolequinone Antitumor Agents: Correlation between Quinone Structure and Rate of Metabolism by Recombinant Human NAD(P)H:Quinone Oxidoreductase. Part 21. J. Med. Chem. 2001;44:3311–3319. doi: 10.1021/jm010884c. [DOI] [PubMed] [Google Scholar]
  • 10.Hassani M, Cai W, Koelsch KH, Holley DC, Rose AS, Olang F, Lineswala JP, Holloway WG, Gerdes JM, Behforouz M, Beall HD. Lavendamycin Antitumor Agents: Structure-Based Design, Synthesis, and NAD(P)H:Quinone Oxidoreductase 1 (NQO1) Model Validation with Molecular Docking and Biological Studies. J. Med. Chem. 2008;51:3104–3115. doi: 10.1021/jm701066a. [DOI] [PubMed] [Google Scholar]
  • 11.Cai W, Hassani M, Karki R, Walter ED, Koelsch KH, Seradj H, Lineswala JP, Mirzaei H, York JS, Olang F, Sedighi M, Lucas JS, Eads TJ, Rose AS, Charkhzarrin S, Hermann NG, Beall HD, Behforouz M. Synthesis, metabolism and in vitro cytotoxicity studies on novel lavendamycin antitumor agents. Bioorg. Med. Chem. 2010;18:1899–1909. doi: 10.1016/j.bmc.2010.01.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Colucci MA, Couch GD, Moody CJ. Natural and synthetic quinones and their reduction by the quinone reductase enzyme NQO1: from synthetic organic chemistry to compounds with anticancer potential. Org. Biomol. Chem. 2008;6:637–656. doi: 10.1039/b715270a. [DOI] [PubMed] [Google Scholar]
  • 13.Behforouz M, Haddad J, Cai W, Arnold MB, Mohammadi F, Sousa AC, Horn MA. Highly Efficient and Practical Syntheses of Lavendamycin Methyl Ester and Related Novel Quinolindiones. J. Org. Chem. 1996;61:6552–6555. doi: 10.1021/jo960794t. [DOI] [PubMed] [Google Scholar]
  • 14.Prieto M, Zurita E, Rosa E, Muñoz L, Lloyd-Williams P, Giralt E. Arylboronic Acids and Arylpinacolboronate Esters in Suzuki Coupling Reactions Involving Indoles. Partner Role Swapping and Heterocycle Protection. J. Org. Chem. 2004;69:6812–6820. doi: 10.1021/jo0491612. [DOI] [PubMed] [Google Scholar]
  • 15.Collot V, Dallemagne P, Bovy PR, Rault S. Suzuki-type cross-coupling reaction of 3-iodoindazoles with aryl boronic acids: A general and flexible route to 3-arylindazoles. Tetrahedron. 1999;55:6917–6922. [Google Scholar]
  • 16.Musser JH, Jones H, Sciortino S, Bailey K, Coutts SM, Khandwala A, Sonnino-Goldman P, Leibowitz M, Wolf P, Neiss ES. Synthesis and antiallergic activities of 1,3-oxazolo[4,5-h]quinolines. J. Med. Chem. 1985;28:1255–1259. doi: 10.1021/jm00147a023. [DOI] [PubMed] [Google Scholar]
  • 17.Tang Q, Zhang C, Luo M. A New Method for N–N Bond Cleavage of N,N-Disubstituted Hydrazines to Secondary Amines and Direct Ortho Amination of Naphthol and Its Analogues. J. Am. Chem. Soc. 2008;130:5840–5841. doi: 10.1021/ja711153b. [DOI] [PubMed] [Google Scholar]
  • 18.Campeau L-C, Stuart DR, Leclerc J-P, Bertrand-Laperle Mg, Villemure E, Sun H-Y, Lasserre S, Guimond N, Lecavallier M, Fagnou K. Palladium-Catalyzed Direct Arylation of Azine and Azole N-Oxides: Reaction Development, Scope and Applications in Synthesis. J. Am. Chem. Soc. 2009;131:3291–3306. doi: 10.1021/ja808332k. [DOI] [PubMed] [Google Scholar]
  • 19.LaMontagne MP, Blumbergs P, Smith DC. Antimalarials. 16. Synthesis of 2-substituted analogs of 8-[4-amino-1-methylbutyl)amino]-6-methoxy-4-methyl-5-[3-trifluoromethyl)phenoxy]quinoline as candidate antimalarials. J. Med. Chem. 1989;32:1728–1732. doi: 10.1021/jm00128a010. [DOI] [PubMed] [Google Scholar]
  • 20.Rodríguez JG, de los Rios C, Lafuente A. Synthesis of n-chloroquinolines and n-ethynylquinolines (n=2, 4, 8): homo and heterocoupling reactions. Tetrahedron. 2005;61:9042–9051. [Google Scholar]
  • 21.Miyaura N, Suzuki A. Palladium-Catalyzed Cross-Coupling Reactions of Organoboron Compounds. Chem. Rev. 1995;95:2457–2483. [Google Scholar]
  • 22.Boger DL, Duff SR, Panek JS, Yasuda M. Inverse electron demand Diels-Alder reactions of heterocyclic azadienes. Studies on the total synthesis of lavendamycin: investigative studies on the preparation of the CDE .beta.-carboline ring system and AB quinoline-5,8-quinone ring system. J. Org. Chem. 1985;50:5782–5789. [Google Scholar]
  • 23.Buffinton G, Ollinger K, Brunmark A, Cadenas E. DT-diaphorase-catalysed reduction of 1,4-naphthoquinone derivatives and glutathionyl-quinone conjugates. Effect of substituents on autoxidation rates. Biochem. J. 1989;257:561–571. doi: 10.1042/bj2570561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gibson NW, Hartley JA, Butler J, Siegel D, Ross D. Relationship between DT-diaphorase-mediated metabolism of a series of aziridinylbenzoquinones and DNA damage and cytotoxicity. Mol. Pharmacol. 1992;42:531–536. [PubMed] [Google Scholar]
  • 25.Long GV, Harding MM. A proton nuclear magnetic resonance study of the interaction of zinc(II) with the antitumour drug streptonigrin. J. Chem. Soc. Dalton Trans. 1996:549–552. [Google Scholar]
  • 26.Wei X, Ming L-J. Comprehensive 2D 1H NMR Studies of Paramagnetic Lanthanide(III) Complexes of Anthracycline Antitumor Antibiotics. Inorg. Chem. 1998;37:2255–2262. doi: 10.1021/ic970741+. [DOI] [PubMed] [Google Scholar]
  • 27.Ross D, Kepa JK, Winski SL, Beall HD, Anwar A, Siegel D. NAD(P)H:quinone oxidoreductase 1 (NQO1): chemoprotection, bioactivation, gene regulation and genetic polymorphisms. Chem. Biol. Interact. 2000;129:77–97. doi: 10.1016/s0009-2797(00)00199-x. [DOI] [PubMed] [Google Scholar]
  • 28.Harding MM, Long GV. Interaction of the antitumor antibiotic streptonigrin with metal ions and DNA. Curr. Med. Chem. 1997;4:405–420. [Google Scholar]
  • 29.Budimir A. Metal ions, Alzheimer's disease and chelation therapy. Acta Pharm. 2011;61:1–14. doi: 10.2478/v10007-011-0006-6. [DOI] [PubMed] [Google Scholar]
  • 30.Kraus J-L, Conti F, Madonna S, Tchoghandjian A, Beclin C. Alternative responses of primary tumor cells and glioblastoma cell lines to N,N-bis-(8-hydroxyquinoline-5-yl methyl)-benzyl substituted amines: cell death versus P53-independent senescence. Int. J. Oncol. 2010;37:1463–1470. doi: 10.3892/ijo_00000798. [DOI] [PubMed] [Google Scholar]
  • 31.King ONF, Li XS, Sakurai M, Kawamura A, Rose NR, Ng SS, Quinn AM, Rai G, Mott BT, Beswick P, Klose RJ, Oppermann U, Jadhav A, Heightman TD, Maloney DJ, Schofield CJ, Simeonov A. Quantitative high-throughput screening identifies 8-hydroxyquinolines as cell-active histone demethylase inhibitors. PLoS One. 2010;5:e15535. doi: 10.1371/journal.pone.0015535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yan C, Kepa JK, Siegel D, Stratford IJ, Ross D. Dissecting the role of multiple reductases in bioactivation and cytotoxicity of the antitumor agent 2,5-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone (RH1) Mol. Pharmacol. 2008;74:1657–1665. doi: 10.1124/mol.108.050401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Faig M, Bianchet MA, Winski S, Hargreaves R, Moody CJ, Hudnott AR, Ross D, Amzel LM. Structure-Based Development of Anticancer Drugs. Complexes of NAD(P)H:Quinone Oxidoreductase 1 with Chemotherapeutic Quinones. Structure. 2001;9:659–667. doi: 10.1016/s0969-2126(01)00636-0. [DOI] [PubMed] [Google Scholar]
  • 34.Verdonk ML, Giangreco I, Hall RJ, Korb O, Mortenson PN, Murray CW. Docking Performance of Fragments and Druglike Compounds. J. Med. Chem. 2011;54:5422–5431. doi: 10.1021/jm200558u. [DOI] [PubMed] [Google Scholar]
  • 35.Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD. Improved protein-ligand docking using GOLD. Proteins. 2003;52:609–623. doi: 10.1002/prot.10465. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1_si_001

RESOURCES