Synthesis and biological evaluation of novel quinoline analogs of ketoprofen as multidrug resistance protein 2 (MRP2) inhibitors

Document Type : Original Article

Authors

1 Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran

2 Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

Abstract

Objective(s): A new series of quinoline analogs of ketoprofen was designed and synthesized as multidrug resistance protein 2 (MRP2) inhibitors using ketoprofen as the lead compounds.
Materials and Methods:  The cytotoxic activity of the compounds was evaluated againt two cancer cell lines including A2780/RCIS (MRP2-overexpressing ovarian carcinoma), A2780, drug-sensitive ovarian carcinoma using MTT assay. Compounds showing low toxicity in MTT test were selected to investigate their MRP inhibition activity. MRP2 inhibitory potency was evaluated by determination of the uptake amount of fluorescent 5-carboxy fluorescein diacetate (5-CFDA) substrate, by A2780/RCIS in the presence of the selected compounds. Mode of interaction between synthesized ligands and homology modeled MRP2 was investigated by MOE software.
Results: Compound 6d, a 4-carboxy quinoline possessing dimethoxy phenyl in position 2 of quinoline ring, showed the most MRP2 inhibition activity among all the quinolines and more than the reference drug ketoprofen. MRP2 inhibition activity of compound 7d was less in comparison to that of compound 6d, indicating that carboxyl group in position 4 of quinoline may interact with MRP2.  Docking studies showed that compound 7d methyl ester of 6d, interacted less compared to its parent 6d, which is consistent with biological results.
Conclusion: This study indicates that 6- or 8-benzoyl-2-arylquinoline is a suitable scaffold to design MRP2 inhibitors. The position of benzoyl in quinoline ring is important in inhibition of MRP2. Generally, MRP2 inhibition activity of compound 7d was less in comparison to that of 6d, indicating that carboxyl group in position 4 of quinoline may interact with MRP2.

Keywords


1. Siegel RL, Miller KD, Jemal A. CA Cancer J Clin 2015;65:5-29.
2. Cozzi P. The discovery of a new potential anticancer drug: a case history. Farmaco 2003;58:213-220.
3. Liang XJ, Chen C, Zhao Y, Wang PC. Circumventing tumor resistance to chemotherapy by nanotechnology. Methods Mol Biol 2010;596:467-488.
4. Beretta GL, Cassinelli G, Pennati M, Zuco V, Gatti L. Overcoming ABC transporter-mediated multidrug resistance: The dual role of tyrosine kinase inhibitors as multitargeting agents. Eur J Med Chem 2017;142:271-289.
5. Staud F, Pavek P. Breast cancer resistance protein (BCRP/ABCG2). Int J Biochem Cell Biol 2005;37:720-725.
6. Hosseinzadeh H, Mazaheri F, Ghodsi R. Pharmacological effects of a synthetic quinoline, a hybrid of tomoxiprole and naproxen, against acute pain and inflammation in mice: a behavioral and docking study. Iran J Basic Med Sci  2017;20:446-450.
7. Zarghi A, Arfaei S. Selective COX-2 Inhibitors: A Review of Their Structure-Activity Relationships. Iran J Pharm Res 2011;10:655-683.
8. Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues. J Natl Cancer Inst 2002;94:252-266.
9. Perkovic I, Butula I, Kralj M, Martin-Kleiner I, Balzarini J, Hadjipavlou-Litina D, et al. Novel NSAID 1-acyl-4-cycloalkyl/arylsemicarbazides and 1-acyl-5-benzyloxy/hydroxy carbamoylcarbazides as potential anticancer agents and antioxidants. Eur J Med Chem 2012;51:227-238.
10. Amin R, Kamitani H, Sultana H, Taniura S, Islam A, Sho A, et al. Aspirin and indomethacin exhibit antiproliferative effects and induce apoptosis in T98G human glioblastoma cells. Neurol Res 2003;25:370-376.
11. Carrett-Dias M, Votto AP, Filgueira Dde M, Almeida DV, Vallochi AL, D’Oca MG, et al. Anti-MDR and antitumoral action of acetylsalicylic acid on leukaemic cells. Biosci Rep 2011;31:391-398.
12. Draper MP, Martell RL, Levy SB. Indomethacin-mediated reversal of resistance and drug efflux in human and murine cell lines overexpressing MRP, but not P-glycoprotein. Br J Cancer 1997;75:810-815.
13. Duffy CP, Elliott CJ, O’Connor RA, Heenan MM, Coyle S, Cleary IM, et al. Enhancement of chemotherapeutic drug toxicity to human tumour cells in vitro by a subset of non-steroidal anti-inflammatory drugs (NSAIDs). Eur J Cancer 1998;34:1250-1259.
14. El-Sheikh AA, van den Heuvel JJ, Koenderink JB, Russel FG. Interaction of nonsteroidal anti-inflammatory drugs with multidrug resistance protein (MRP) 2/ABCC2- and MRP4/ABCC4-mediated methotrexate transport. J Pharmacol Exp  2007;320:229-235.
15. Gruber BM, Bubko I, Krzyszton-Russjan J, Anuszewska EL. Synergistic action of doxorubicin and sulindac in human cervix carcinoma cells - studies on possible mechanisms. Med Sci Mon Int Med J Exp Clin Res 2010;16: 45-51.
16. Maguire AR, Plunkett SJ, Papot S, Clynes M, O’Connor R, Touhey S. Synthesis of indomethacin analogues for evaluation as modulators of MRP activity. Bioorg Med Chem 2001;9:745-462.
17. O’Connor R, Heenan M, Connolly L, Larkin A, Clynes M. Increased anti-tumour efficacy of doxorubicin when combined with sulindac in a xenograft model of an MRP-1-positive human lung cancer. Anticancer Res 2004;24:457-464.
18. O’Connor R, O’Leary M, Ballot J, Collins CD, Kinsella P, Mager DE, et al. A phase I clinical and pharmacokinetic study of the multidrug resistance protein-1 (MRP-1) inhibitor sulindac, in combination with epirubicin in patients with advanced cancer. Cancer Chemother Pharmacol 2007;59:79-87.
19. Roller A, Bahr OR, Streffer J, Winter S, Heneka M, Deininger M, et al. Selective potentiation of drug cytotoxicity by NSAID in human glioma cells: the role of COX-1 and MRP. Biochem Biophys Res Commun 1999;259:600-605.
20. Rosenbaum C, Rohrs S, Muller O, Waldmann H. Modulation of MRP-1-mediated multidrug resistance by indomethacin analogues. J Med Chem 2005;48:1179-1187.
21. Touhey S, O’Connor R, Plunkett S, Maguire A, Clynes M. Structure-activity relationship of indomethacin analogues for MRP-1, COX-1 and COX-2 inhibition. identification of novel chemotherapeutic drug resistance modulators. Eur J Cancer 2002;38:1661-1670.
22. Zhang L, Liu L, Zheng C, Wang Y, Nie X, Shi D, et al. Synthesis and biological evaluation of novel podophyllotoxin-NSAIDs conjugates as multifunctional anti-MDR agents against resistant human hepatocellular carcinoma Bel-7402/5-FU cells. Eur J Med Chem 2017;131:81-91.
23. Anuchapreeda S, Thanarattanakorn P, Sittipreechacharn S, Tima S, Chanarat P, Limtrakul P. Inhibitory effect of curcumin on MDR1 gene expression in patient leukemic cells. Arch Pharm Res 2006;29:866-873.
24. Nakamichi N, Ishimoto T, Yamauchi Y, Masuo Y, Kato Y. Screening to identify multidrug resistance-associated protein inhibitors with neuroblastoma-selective cytotoxicity. Biol Pharm Bull 2016;39:1638-1645.
25. O’CONNOR R. The pharmacology of cancer resistance. Anticancer Res 2007;27:1267-1272.
26. El-Sheikh AA, van den Heuvel JJ, Koenderink JB, Russel FG. Interaction of nonsteroidal anti-inflammatory drugs with multidrug resistance protein (MRP) 2/ABCC2-and MRP4/ABCC4-mediated methotrexate transport. J Pharmacol Exp 2007;320:229-235.
27. Rijpma SR, van den Heuvel JJ, van der Velden M, Sauerwein RW, Russel FG, Koenderink JB. Atovaquone and quinine anti-malarials inhibit ATP binding cassette transporter activity. Malar J 2014;13:359-367.
28. Nakamura T, Oka M, Aizawa K, Soda H, Fukuda M, Terashi K, et al. Direct interaction between a quinoline derivative, MS-209, and multidrug resistance protein (MRP) in human gastric cancer cells. Biochem Biophys Res Commun 1999;255:618-624.
29. Wu CP, Klokouzas A, Hladky SB, Ambudkar SV, Barrand MA. Interactions of mefloquine with ABC proteins, MRP1 (ABCC1) and MRP4 (ABCC4) that are present in human red cell membranes. Biochem Pharmacol 2005;70:500-510.
30. Gekeler V, Ise W, Sanders KH, Ulrich WR, Beck J. The leukotriene LTD4 receptor antagonist MK571 specifically modulates MRP associated multidrug resistance. Biochem Biophys Res Commun 1995;208:345-352.
31. Karthikeyan C, Malla R, Ashby CR, Jr., Amawi H, Abbott KL, Moore J, et al. Pyrimido[1’’,2’’:1,5]pyrazolo[3,4-b]quinolines: Novel compounds that reverse ABCG2-mediated resistance in cancer cells. Cancer Lett 2016;376:118-126.
32. Behbahani FS, Tabeshpour J, Mirzaei S, Golmakaniyoon S, Tayarani-Najaran Z, Ghasemi A, et al. Arch Pharm 2019;352:1800307–1800318.
33. Ghodsi R, Azizi E, Ferlin MG, Pezzi V, Zarghi A. Design, synthesis and biological evaluation of 4-(Imidazolylmethyl)-2-aryl-quinoline derivatives as aromatase inhibitors and anti-breast cancer agents. Lett Drug Des Discov 2016;13:89-97.
34. Ghodsi R, Azizi E, Zarghi A. Design, synthesis and biological evaluation of4-(Imidazolylmethyl)- 2-(4-methylsulfonyl phenyl)-quinoline derivatives as selective COX-2 inhibitors and in-vitro anti-breast cancer agents. Iran J Basic Med Sci 2016;15:169-177.
35. Golmakaniyoon S, Askari VR, Abnous K, Zarghi A, Ghodsi R. Synthesis, characterization and in-vitro evaluation of novel naphthoquinone derivatives and related imines: Identification of new anticancer leads. Iran J Pharma Res 2019;18:16-29.
36. Jafari F, Baghayi H, Lavaee P, Hadizadeh F, Soltani F, Moallemzadeh H, et al. Design, synthesis and biological evaluation of novel benzo- and tetrahydrobenzo-[h]quinoline derivatives as potential DNA-intercalating antitumor agents. Eur J Med Chem 2019;164:292-303.
37. Karimikia E, Behravan J, Zarghi A, Ghandadi M, Malayeri SO, Ghodsi R. Colchicine-like β-acetamidoketones as inhibitors of microtubule polymerization: Design, synthesis and biological evaluation of in vitro anticancer activity. Iran J Basic Med Sci 2019;22:1138-1146.
38. Malayeri SO, Abnous K, Arab A, Akaberi M, Mehri S, Zarghi A, et al. Design, synthesis and biological evaluation of 7-(aryl)-2,3-dihydro-[1,4]dioxino[2,3-g]quinoline derivatives as potential Hsp90 inhibitors and anticancer agents. Bioorg Med Chem 2017;25:1294-1302.
39. Malayeri SO, Tayarani-Najaran Z, Behbahani FS, Rashidi R, Delpazir S, Ghodsi R. Synthesis and biological evaluation of benzo[b]furo[3,4-e][1,4]diazepin-1-one derivatives as anticancer agents. Bioorg Chem 2018;80:631-638.
40. Mirzaei S, Eisvand F, Hadizadeh F, Mosaffa F, Ghasemi A, Ghodsi R. Design, synthesis and biological evaluation of novel 5,6,7-trimethoxy-N-aryl-2-styrylquinolin-4-amines as potential anticancer agents and tubulin polymerization inhibitors. Bioorg Chem 2020;98:103711.
41. Mirzaei S, Hadizadeh F, Eisvand F, Mosaffa F, Ghodsi R. Synthesis, structure-activity relationship and molecular docking studies of novel quinoline-chalcone hybrids as potential anticancer agents and tubulin inhibitors. J Mol Struct 2020;1202: 127310.
42. Zarghi A, Ghodsi R. Design, synthesis, and biological evaluation of ketoprofen analogs as potent cyclooxygenase-2 inhibitors. Bioorg Med Chem 2010;18:5855-5860.
43. Aboutorabzadeh SM, Mosaffa F, Hadizadeh F, Ghodsi R. Design, synthesis, and biological evaluation of 6-methoxy-2-arylquinolines as potential P-glycoprotein inhibitors. Iran J Basic Med Sci 2018;21:9-18.
44. Mahdizadeh S, Karimi G, Behravan J, Arabzadeh S, Lage H, Kalalinia F. Crocin suppresses multidrug resistance in MRP overexpressing ovarian cancer cell line. Daru 2016;24:17-24.
45. Xing L, Hu Y, Lai Y. Advancement of structure-activity relationship of multidrug resistance-associated protein 2 interactions. AAPSJ 2009;11:406-413.
46. Nies AT, König J, Cui Y, Brom M, Spring H, Keppler D. Structural requirements for the apical sorting of human multidrug resistance protein 2 (ABCC2). Eur J Biochem 2002;269:1866-1876.
47. Williamson G, Aeberli I, Miguet L, Zhang Z, Sanchez MB, Crespy V, et al. Interaction of positional isomers of quercetin glucuronides with the transporter ABCC2 (cMOAT, MRP2). Drug Metab Dispos 2007;35:1262-1268.