1,683
Views
13
CrossRef citations to date
0
Altmetric
Short Communication

Quinoline-based imidazole-fused heterocycles as new inhibitors of 15-lipoxygenase

, , , , , , , , & show all
Pages 205-209 | Received 20 Jan 2016, Accepted 21 Jun 2016, Published online: 17 Jul 2016

Abstract

A series of 2-chloro-quinoline-based imidazopyridines 6a–l and imidazothiazoles 6m–o bearing a bulky alkylamine side chain were synthesized as soybean 15-LOX inhibitors. The target compounds 6a–o were prepared via one-pot reaction of 2-chloroquinoline-3-carbaldehyde (3), heteroaromatic amidine 4, and alkyl isocyanides 5, in the presence of NH4Cl. All compounds showed significant anti-15-LOX activity (IC50 values ≤40 μM). Among the title compounds, the imidazo[2,1-b]thiazole derivative 6n bearing a tert-butylamine moiety showed the highest activity against soybean 15-LOX enzyme.

Introduction

Polyunsaturated fatty acids such as arachidonic acid are implicated in the control of several physiological processes. Lipoxygenases (LOXs) are a class of nonheme iron-containing enzymes which regio- and stereospecifically catalyze the hydroperoxidation of polyunsaturated fatty acidsCitation1. In humans, three major families of LOXs were found as 5-LOX, 12-LOX, and 15-LOX isoforms. These isoforms (5-, 12-, or 15-LOX) initiate the biosynthesis of leukotrienes, lipoxins, and other compounds by oxidizing the C-5, C-12, and C-15 positions of the key substrate, arachidonic acidCitation2,Citation3. For example, 15-LOX oxidizes arachidonic acid to produce mainly 15(S)-5Z,8Z,11Z,13E-hydroperoxyeicosatetraenic acid (15-HPETE). The bioactive metabolites of 15-LOX hydroperoxidation (e.g. HETE and leukotriene A4) are found to be potent signal transduction modifiers which affect the inflammatory processesCitation4. Furthermore, 15-LOX has been implicated in neurodegenerative diseasesCitation5, atherosclerosisCitation6,Citation7, chronic obstructive pulmonary disease (COPD), and a variety of cancersCitation8.

Consequently, small molecules affecting the 15-LOX pathway might be therapeutically useful in chronic inflammatory diseases, cardiovascular disorders, and some types of tumors. Thus, medicinal chemists have attended extensively to find new inhibitors of 15-LOX. A part of attentions has been focused on small molecules containing fused heterocyclic systems including indolizineCitation9,Citation10 and imidazo-fused heterocyclesCitation11,Citation12 previously described (I, ) as a potential inhibitors of 15-LOX. In a study by Wisniewska et al., imidazo[1,2-a]pyridine-3-yl-amine analog (EP6, II) was evaluated as a 5-LOX inhibitorCitation13. On the other hand, a series of quinoline-based compounds were reported as potent inhibitors of LOXCitation14–18. These findings convinced us to design a new core containing quinoline and imidazole-fused system as a new 15-LOX inhibitors. Thus, we describe here, synthesis, in vitro anti-15-LOX activity and docking study of [2-(2-chloro-quinolin-3-yl)-imidazo[1,2-a]pyridin-3-yl]amines 6a–l and [6-(2-chloro-quinolin-3-yl)-imidazo[2,1-b]thiazol-5-yl]amines 6m–o (). Following our interests on the chemistry aspects of this core meaning finding new catalysis for the synthesis of imidazo[1,2-a]pyridine and imidazo[2,1-b]thiazol-quinoline derivatives and their further coupling reactionCitation19,Citation20, herein, we report the synthesis of new quinolin imidazo[1,2-a]pyridine derivatives and also evaluate their soybean 15-LOX inhibitory activity.

Figure 1. Structures of reported LOX inhibitors I and II, and designed compounds 6a–o as new 15-LOX inhibitors.

Figure 1. Structures of reported LOX inhibitors I and II, and designed compounds 6a–o as new 15-LOX inhibitors.

Experimental

15-LOX inhibition assay

To evaluate 15-LOX (Lipoxidase from Glycine max, soybean) inhibitory activity of new synthesized compounds, the stock solution of the compounds were prepared by dissolving them in 1 mL of DMSO. To prepare substrate solution (stock concentration = 38 mM), 12 μL linoleic acid was dissolved in 988 μL ethanol. This solution should be used the same day it is made. The final concentration of substrate will be 122 μM. Five different concentrations of each compound were tested in triplicate to obtain the inhibition range between 20 and 80%. The test solution was a mixture of 3 mL phosphate buffer (0.1 M, pH = 8), 50 μL enzyme solution (final concentration: 167 U/mL), and 50 μL of target compound solution. Being incubated for 4 min, the substrate (Linoleic acid, final concentration: 122 μM) was added, and the change in absorbance was measured for 60s at 234 nm. A control test was done with the same volume of DMSO (50 μL) to eliminate the effect of DMSO on enzyme activity.

Molecular modeling and docking stimulation

All docking simulations were performed using Autodock Vina (ver. 1.1.1). First, the 3D structure of soybean LOX in complex with 13(S)-hydroproxy-9(Z)-2,11(E)-octadecadienoic acid (code ID: 1IK3) was retrieved from protein databank (www.pdb.org). Then, the co-crystallized ligand and water molecules were removed, and the protein was converted to pdbqt format using Autodock Tools (1.5.4). To prepare the ligands for docking, the 2D chemical structure of ligands was sketched using MarvinSketch 5.8.3, 2012, ChemAxon (http://www.chemaxon.com) and then converted to 3D format by Openbabel (ver 2.3.1). Finally, pdbqt format of ligands was prepared using an Autodock Tools python script, prepare_ligand4.py. The docking parameters were set as follow: size_x= 20; size_y = 20; size_z= 20; center_x =19.693; center_y = 0.054; center_z= 17.628. The exhaustiveness was set to 100, and the max number of retrieved final docked poses was set to 15 using num_modes parameter. The other docking parameters were left as default. Finally, the most favorable docked poses in terms of free binding energy were selected for analyzing of enzyme–inhibitor interactions.

Chemistry

Commercially available chemicals and reagents were purchased from Merck and Fluka Chemical Company and used without further purification. Melting points are measured with a Kofler hot stage apparatus and are uncorrected. 1H and 13C NMR spectra were run on a Bruker FT-400 in CDCl3, using TMS as an internal standard. IR spectra were recorded on a Shimadzu 470 spectrophotometer (KBr disks). MS were recorded with an Agilent Technology (HP) mass spectrometer operating at an ionization potential of 70 eV. Elemental analysis was performed with an Elementar Analysensysteme GmbH VarioELCHNS mode.

General procedure for the synthesis of compounds 6a–o

A mixture of 2-chloroquinolin-3-carbaldehyde 3 (1.0 mmol), heteroaromatic amidine 4a–f (1.0 mmol), appropriate alkyl isocyanide (1.2 mmol), and NH4Cl (1.0 mmol) in toluene (5 mL) was heated under reflux for 12–24 h. After completion of the reaction, as indicated by TLC, the solvent was evaporated under reduced pressure, and the residue was recrystallized from petroleum ether–EtOAc to afford target compounds 6a–o in 65–93% yields.

[2-(2-Chloro-quinolin-3-yl)-imidazo[1,2-a]pyridin-3-yl]- (1,1,3,3-tetramethyl-butyl)-amine (6c)

Yield: 0.32 g (80%); pale yellow solid; mp 168–170 °C; IR (KBr): 3321, 2919, 2848, 1631, 1497, 754 cm−1; 1H NMR (400 MHz, CDCl3): δ 0.94 (s, 9H, C(CH3)3), 1.29 (s, 6H, C(CH3)2), 1.42 (s, 2H, CH2), 3.48 (s, 1H, NH), 6.94 (dd, J = 6.8, 4.0 Hz, 1H, H6), 7.63 (t, J = 7.5 Hz, 1H, H6′), 7.80 (t, J = 7.5 Hz, 1 H, H7′), 7.93 (d, J = 8.0 Hz, 1H, H5′), 8.09 (d, J = 8.4 Hz, 1H, H8′), 8.52 (dd, J = 6.8, 1.6 Hz, 1H, H7), 8.58–8.59 (m, 2H, H8, H5), 8.63 (s, 1H, H4′); 13C NMR (100 MHz, CDCl3): δ 29.0, 31.5, 31.7, 56.4, 59.4, 108.1, 112.0, 126.2, 127.2, 127.5, 127.9, 128.8, 129.0, 130.9, 131.2, 136.0, 141.8, 145.1, 148.0, 149.1, 150.0. MS: m/z (%) 408 (15, [M + 2]+), 406 (49, M+). Anal. Calcd for C24H27ClN4: C, 70.83; H, 6.69; N, 13.77. Found: C, 70.97; H, 6.49; N, 13.84.

[2-(2-Chloro-quinolin-3-yl)-8-methyl-imidazo[1,2-a]pyridin-3-yl]-cyclohexyl-amine (6d)

Yield: 0.35 g (91%); pale yellow solid; mp 143–145 °C; IR (KBr): 3327, 2971, 1640, 1568, 781 cm−1; 1H NMR (400 MHz, CDCl3): δ 1.01–1.69 (m, 10 H, 5CH2, cyclohexyl), 2.41 (s, 3H, CH3), 2.67 (s, 1H, NCH), 3.32 (s, 1H, NH), 6.79 (t, J = 6.4 Hz, 1H, H6), 7.02 (d, J = 6.4 Hz, 1H, H7), 7.61 (t, J = 7.6 Hz, 1H, H6′), 7.78–7.82 (m, 1H, H7′), 7.92 (d, J = 8.0 Hz, 1H, H5′), 8.10 (d, J = 8.4 Hz, 1H, H8′), 8.24 (d, J = 6.4 Hz, 1H, H5), 8.61 (s, 1H, H4′); 13C NMR (100 MHz, CDCl3): δ 21.4, 24.6, 25.6, 33.8, 56.7, 111.6, 121.7, 123.4, 125.7, 127.0, 127.1, 127.3, 127.4, 128.4, 129.7, 130.5, 135.6, 140.8, 141.3, 147.1, 149.0. MS: m/z (%) 392 (19, [M + 2]+), 390 (58, M+). Anal. Calcd for C23H23ClN4: C, 70.67; H, 5.93; N, 14.33. Found: C, 70.45; H, 6.13; N, 14.12.

[6-Chloro-2-(2-chloro-quinolin-3-yl)-imidazo[1,2-a]pyridin-3-yl]-(1,1,3,3-tetramethyl-butyl)-amine (6k)

Yield: 0.33 g (75%); yellow solid; mp 145–147 °C; IR (KBr): 3274, 2828, 1624, 1607, 1330, 772 cm−1; 1H NMR (400 MHz, CDCl3): δ 0.91 (s, 6H, C(CH3)2), 0.96 (s, 9H, C(CH3)3), 1.48 (s, 2H, CH2), 3.39 (s, 1H, NH), 7.19 (dd, J = 9.6, 2.0 Hz, 1H, H7), 7.54 (dd, J = 9.6, 0.8 Hz, 1H, H8), 7.61–7.65 (m, 1H, H6′), 7.77–7.81 (m, 1H, H7′), 7.93 (d, J = 8.0 Hz, 1H, H5′), 8.11 (d, J = 8.0 Hz, 1H, H8′), 8.24 (dd, J = 2.0, 0.8 Hz, 1 H, H5), 8.51 (s, 1H, H4′); 13C NMR (100 MHz, CDCl3):δ 30.0, 31.5, 31.7, 56.5, 59.2, 118.6, 120.7, 121.0, 126.1, 127.4, 127.7, 127.9, 128.0, 128.5, 129.0, 131.1, 134.9, 140.7, 141.0, 147.4, 149.0. MS: m/z (%) 444 (10, [M + 4]+), 442 (58, [M + 2]+), 440 (90, M+). Anal. Calcd for C24H26Cl2N4: C, 65.31; H, 5.94; N, 12.69. Found: C, 65.24; H, 5.83; N, 12.76.

Results and discussion

Chemistry

In the synthetic route to target compounds 6a–o, initially aniline (1) was converted to N-phenylacetamide (2) via the acetylation reaction in the presence of acetyl chloride and potassium carbonate under mild condition (Scheme 1). Then, 2-chloroquinolin-3-carbaldehyde (3) was prepared using the Vilsmeier–Haak reaction in the presence of POCl3 in DMFCitation21. The final compounds 6a–o were synthesized via one-pot condensation reaction of aldehyde 3, an appropriate isocyanide 5a–c and various heteroaromatic amidine 4a–f, in the presence of catalytic amount of ammonium chloride in toluene. After recrystallization from petroleum ether–EtOAc, pure compounds 6a–o were obtained in 65–93% yields.

Scheme 1. Preparation of 2-chloro-quinoline-based imidazopyridines and imidazothiazoles.

Scheme 1. Preparation of 2-chloro-quinoline-based imidazopyridines and imidazothiazoles.

15-LOX inhibitory activity

The inhibitory activity of synthesized compounds was determinedCitation22 against 15-LOX, and the obtained IC50 values (IC50 expressed as mean ± SD of three independent experiments) were listed in . All compounds showed significant inhibitory activity with the IC50 values ≤40 μM. Among them, compound 6n possessing IC50 value of 11.5 μM was the most potent compound. Furthermore, compounds 6g and 6i with IC50 values of 15.3 and 14.1 μM were more active than remaining compounds. As seen in , compounds 6a–l were imidazopyridine derivatives, and compounds 6m–o had imidazothiazole substructure. The most potent compound 6n was imidazothiazole derivative. On the other hand, the potent compounds 6 g and 6i were imidazopyridine analogs. In the imidazopyridine series, introduction of bromo or chloro substituent at 6-position decreased the inhibitory activity. The effect of methyl group at 7 or 8 position of imidazopyridine ring depended on the alkyl side chain connected to the amine group. For example, while 7-methyl-imidazopyridine derivative 6i bearing a 1,1,3,3-tetramethyl-butylamine residue was more potent than 6c, but 7-methyl-imidazopyridine 6h containing a tert-butyl group found to be as potent as 6b. The comparison of 7- and 8-methyl regioisomers revealed that 7-methyl derivatives 6g and 6i exhibiting more potent activity in respect to their 8-methyl analogs 6d and 6f. In contrast, (7-methyl-imidazopyridin-3-yl)amine 6h was less potent than its 8-methyl regioisomer 6e.

Table 1. Used amidine (4) and isocyanide (5) derivatives for the one-pot synthesis of compounds 6a–o including their 15-Lox inhibitory activity.

Docking study

The docking study was performed to clarify the binding mode of the target compounds in the active site of 15-LOX. For this purpose, the tested compounds were docked onto the active site of enzyme using Autodock Vina (ver. 1.1.1)Citation23,Citation24. Then, the best docked poses in terms of free binding energy were further analyzed to clarify interactions between ligands and the 15-LOX enzyme. Because of similar orientation of compounds in the active site of 15-LOX, further analysis was performed on the most active compound 6n. As shown in , the target compound was laid near the Fe3+ ion in the 15-LOX active site. In this position, the ligand interacted with Fe3+ ion through a π-cation interaction via phenyl ring of its 2-chloroquinoline moiety. A careful inspection of the binding pocket indicated that this moiety oriented toward a hydrophobic cavity comprised of Trp519, Ile557, Leu560, Ala561, and Leu565. The ligand also established another remarkable hydrophobic interaction via the orientation of tert-butylamino group toward a hydrophobic pocket including side chains of Leu515, Val571, Ile572, Phe576, and Val769.

Figure 2. The best docked pose of compound 6n in the active site of 15-LOX.

Figure 2. The best docked pose of compound 6n in the active site of 15-LOX.

Conclusions

We synthesized a series of 2-chloro-quinoline-based imidazopyridines 6a–l and imidazothiazoles 6m–o bearing a bulky alkylamine side chain as soybean 15-LOX inhibitors. The in vitro evaluation of title compounds against 15-LOX demonstrated that all compounds had significant inhibitory activity (IC50 values ≤40 μM). The most potent compound 6n with IC50 value of 11.5 μM was belong to the imidazo[2,1-b]thiazole series. However, the imidazopyridine derivatives 6g and 6i showed substantial inhibition against 15-LOX (IC50 values ≤15.3 μM). The limited SAR study revealed that the effect of substituent on imidazopyridine ring depended on the attached alkylamine side chain. The docking study indicated that the target compound 6n was laid near the Fe3+ ion in the 15-LOX active site. A π-cation interaction of 2-chloroquinoline with Fe3+ ion and hydrophobic interactions had important roles in the favorable binding of the inhibitor with the enzyme active site.

Supporting information

Experimental details and 1H and 13C NMR spectra are available, via the supplementary content section of this article’s web page.

Declaration of interest

The authors report no declarations of interest. This work was supported and funded by Research council of Tehran University of Medical Sciences (TUMS); Grant no: 95-01-92-31756; and Iran National Science Foundation (INSF).

Supplementary materials online only – For review only at proofing stage

Supplemental material

IENZ_1206087_Supplementary_Material.pdf

Download PDF (558 KB)

References

  • Solomon EI, Zhou J, Neese F, Pavel EG. New insights from spectroscopy into the structure/function relationships of lipoxygenases. Chem Biol 1997;4:795–808
  • Kuhn H, Thiele BJ. The diversity of the lipoxygenase family. Many sequence data but little information on biological significance. FEBS Lett 1999;449:7–11
  • Serhan CN, Petasis NA. Resolvins and protectins in inflammation resolution. Chem Rev 2011;111:5922–43
  • Kashfi K. Anti-inflammatory agents as cancer therapeutics. In: Enna SJ, Williams M, eds. Contemporary aspects of biomedical research: drug discovery. Amsterdam: Academic Press; 2009:31–56
  • Pratico D, Zhukareva V, Yao Y, et al 12/15-Lipoxygenase is increased in Alzheimer’s disease: possible involvement in brain oxidative stress. Am J Pathol 2004;164:1655–62
  • Cyrus T, Witztum JL, Rader DJ, et al Disruption of the 12/15-lipoxygenase gene diminishes atherosclerosis in apo E-deficient mice. J Clin Invest 1999;103:1597–604
  • Harats D, Shaish A, George J, et al Overexpression of 15-lipoxygenase in vascular endothelium accelerates early atherosclerosis in LDL receptor-deficient mice. Arteriosler Thromb Vasc Biol 2000;20:2100–5
  • Kuhn H, Walther M, Kuban RJ. Mammalian arachidonate 15-lipoxygenases: structure, function, and biological implications. Prostaglandins Other Lipid Mediat 2002;68–69:263–90
  • Gundersen LL, Malterud KE, Negussie AH, et al Indolizines as novel potent inhibitors of 15-lipoxygenase. Bioorg Med Chem 2003;11:5409–15
  • Teklu S, Gundersen LL, Larsen T, et al Indolizine 1-sulfonates as potent inhibitors of 15-lipoxygenase from soybeans. Bioorg Med Chem 2005;13:3127–39
  • Barazandeh Tehrani M, Emami S, Asadi M, et al Imidazo[2,1-b]thiazole derivatives as new inhibitors of 15-lipoxygenase. Eur J Med Chem 2014;87:759–64
  • Hofmann B, Franke L, Proschak E, et al Scaffold-hopping cascade yields potent inhibitors of 5-lipoxygenase. ChemMedChem 2008;3:1535–8
  • Wisniewska JM, Rödl CB, Kahnt AS, et al Molecular characterization of EP6-a novel imidazo[1,2-a]pyridine based direct 5-lipoxygenase inhibitor. Biochem Pharmacol 2012;83:228–40
  • Dubé D, Blouin M, Brideau C, et al Quinolines as potent 5-lipoxygenase inhibitors: synthesis and biological profile of L-746,530. Bioorg Med Chem Lett 1998;8:1255–60
  • Werz O, Greiner C, Koeberle A, et al Novel and potent inhibitors of 5-lipoxygenase product synthesis based on the structure of pirinixic acid. J Med Chem 2008;51:5449–53
  • Golitzer K, Fabian J, Frohberg P, Drutkowski G. Pyrrolo[2,3-c]quinolones and pyrrolo[3,4-d]qunolines-synthesis and investigation of lipoxygenase inhibition. Pharmazie 2002;57:243–9
  • White JD, Yager KM, Stappenbeck F. Synthesis and conformation of 2-[[3-(1-hydroxyhexyl)phenoxy]methyl]quinolone, a 5-lipoxygenase inhibitor and leukotriene antagonist. J Med Chem 1993;58:3466–8
  • Musser JH, Kreft AF. 5-lipoxygenase: properties, pharmacology, and the quinolinyl(bridged)aryl class of inhibitors. J Med Chem 1992;35:2501–24
  • Mouradzadegun A, Ma’mani L, Mahdavi M, et al Sulfamic acid-functionalized hydroxyapatite encapsulated g-Fe2O3 nanoparticles as a magnetically recoverable catalyst for synthesis of N-fused imidazole-quinoline conjugates under solvent-free condition. RSC Adv 2015;5:83530–7
  • Dianat S, Mahdavi M, Moghimi S, et al Combined isocyanide-based multi-component Ullmann-type reaction: an efficient access to novel nitrogen-containing pentacyclic compounds. Mol Divers 2015;19:797–805
  • Baruah B, Bhuyan PJ. Synthesis of some complex pyrano[2,3-b]- and pyrido[2,3-b]quinolines from simple acetanilides via intramolecular domino hetero Diels–Alder reactions of 1-oxa-1,3-butadienes in aqueous medium. Tetrahedron 2009;65:7099–104
  • Malterud KE, Rydland KM. Inhibitors of 15-lipoxygenase from orange peel. J Agric Food Chem 2000;48:5576–80
  • Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010;31:455–61
  • Mahdavi M, Shirazi MS, Taherkhani R, et al Synthesis, biological evaluation and docking study of 3-aroyl-1-(4-sulfamoylphenyl)thiourea derivatives as 15-lipoxygenase inhibitors. Eur J Med Chem 2014;82:308–13

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.