Next Article in Journal
A Trisbenzimidazole Phosphoramidite Building Block Enables High-Yielding Syntheses of RNA-Cleaving Oligonucleotide Conjugates
Previous Article in Journal
Improved Detection of Molecular Markers of Atherosclerotic Plaques Using Sub-Millimeter PET Imaging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

5-Oxo-hexahydroquinoline Derivatives and Their Tetrahydroquinoline Counterparts as Multidrug Resistance Reversal Agents

1
Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
2
Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz 71348-53734, Iran
3
Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
4
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
*
Authors to whom correspondence should be addressed.
Molecules 2020, 25(8), 1839; https://doi.org/10.3390/molecules25081839
Submission received: 9 February 2020 / Revised: 28 March 2020 / Accepted: 31 March 2020 / Published: 16 April 2020
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Cancer is a leading cause of death worldwide. Multidrug resistance (MDR) is a main reason of chemotherapy failure in many patients and is often related to overexpression of ATP-binding cassette (ABC) transporters, including P-glycoprotein (P-gp/ABCB1). Agents that are capable of modulation of the activity of these transporters might be effective in overcoming MDR. In this study, a new set of 1,4,5,6,7,8-hexahydro 5-oxo quinoline-3-carboxamide derivatives bearing 4-methylthiazole moiety and their tetrahydroquinoline counterparts were synthesized. MDR reversal activity of these 16 newly synthesized derivatives was tested in P-gp overexpressing MES-SA-DX5 human uterine sarcoma cells by flow cytometric determination of Rhodamine123 efflux. The effect of the most potent compounds in induction of apoptosis and alterations of cell cycle was examined in these cells by a flow cytometric method. Inherent cytotoxicity of the synthesized compounds was evaluated against MCF-7, A-549 and K562 cancer cell lines, as well as MES-SA-DX5 and their parental non-resistant MES-SA and also HEK-293 non-cancerous cells by MTT assay. Compounds A1 and A2 with 5-oxo-hexahydroquinoline structure bearing 2,4-dichlorophenyl and 4-bromophenyl moieties, respectively, and their tetrahydroquinoline counterparts B1 and B2 significantly blocked P-gp efflux, induced apoptosis and showed the highest cytotoxicities against MES-SA-DX5 cells. However, only A2 and B2 compounds were relatively selective against cancer and MDR cells as compared to non-resistant and non-cancerous cells. These findings demonstrate that 5-oxo-hexahydroquinoline and 5-oxo-tetrahydroquinoline derivatives represent promising agents with therapeutic potential in drug resistant cancers.

1. Introduction

Cancer is a major cause of death and disability worldwide and despite considerable diagnostic and therapeutic advancements in recent years, it is still the cause of more than 8 million deaths in the world every year [1]. The resistance of malignant tumor cells to multiple structurally and mechanistically unrelated classes of anticancer agents is recognized as multidrug resistance (MDR). This phenomenon very often occurs in advanced cancer cases and is a major cause of failure of therapies [2,3]. Different mechanisms are involved in MDR: Very often it is caused by the overexpression of P-glycoprotein (P-gp), also named multidrug resistance protein-1 (MDR1), which is encoded by ATP-binding cassette (ABC) subfamily B member 1 (ABCB1) gene [4]. P-gp has been one of the first members of ABC transporters to be studied [5]. It functions as an efflux pump extruding several substrate molecules and plays an important physiological role in cleaning cells from xenobiotics and maintaining the integrity of blood-brain barrier [6]. However, overexpression of P-gp in cancer cells leads to reduced accumulation of cytotoxic drugs and even targeted therapeutics resulting in resistance against these agents [3,7].
In this context, targeting P-gp by small-molecule inhibitors seems to be a promising approach for overcoming MDR and restoring chemosensitivity in tumor cells [4]. In spite of numerous considerable efforts using different classes of compounds, there is no approved MDR-targeting drug available yet [3,8,9]. Hence, new P-gp inhibitors with high potency and selectivity are much needed to be developed.
One important class of candidate compounds capable of MDR reversal, is 1,4-dihydropyridine derivatives. 1,4-DHPs possess several important pharmacological properties including calcium-channel-blocking, bronchodilator, anti-ischemic and antitumor as well as MDR reversal effects [10,11,12,13,14,15]. Several agents structurally related to 1,4-DHP derivatives have been reported as P-gp inhibitors [16,17,18,19,20,21].
We have recently designed and synthesized a set of 5-oxohexahydroquinoline [22] and tetrahydroquinolinone derivatives [20] closely related structures to 1,4-DHPs, as MDR reversal agents. Here, in another attempt to respond to the crucial need for novel cancer therapeutic agents and as part of our research program toward the discovery of useful MDR reversal compounds, we report the synthesis and biological evaluation a set of sixteen 5-oxo-hexahydroquinoline derivatives. The findings of the study showed that some of these compounds are capable of reversing MDR in P-gp overexpressing cancer cells.

2. Materials and Methods

2.1. Chemistry

All the reagents were bought from Sigma Aldrich chemical Co. or Merck and used without further purification. Melting points were recorded by a hot stage apparatus (Electrothermal, Essex, UK) and reported without correction. 1H-NMR spectra were determined by a Bruker FT-400 MHz spectrometer (Bruker Daltonics, Bremen, Germany) in DMSO-d6. All the chemical shifts were reported as (d) values (ppm) against tetramethylsilane as an internal standard. The MS spectra were recorded using an Agilent 7890A spectrometer at 70 eV. Elemental analyses were performed on a TESCAN-Vega3 CNO analyzer (TESCAN, Brno, Czech Republic).

General Procedure of the Synthesis

2-Methylthiazol-4-amine (5 mmol) was mixed with 2,2,6-trimethyl-4H-1,3-dioxin-4-one (6 mmol) in 10 mL xylene. The mixture was refluxed for 2–4 h. The reaction procedure was monitored using TLC (thin layer chromatography). At the end of reaction, the precipitate was removed and washed with petroleum ether (30–50 mL). The obtained product, N-(2-methylthiazol-4-yl)-3-oxobutanamide, cyclohexane-1,3-dione and different aryl aldehydes were mixed in equimolar and excess amount of ammonium acetate was added. The mixture was refluxed in ethanol for 24 h. The oxidized products were achieved with high purity via oxidation for 24–48 h in the presence of MnO2 and ethanol as solvent. The acquired products of each step were characterized using 1H-NMR, 13C-NMR, mass and IR spectroscopy. All spectra are shown in Figures S1–S64, Supplementary Materials.
N-(2-Methylthiazol-4-yl)-3-oxobutanamide (a): Yield: 86%, yellow powder, mp: 173–175 °C, 1H-NMR (DMSO-d6) δ: 11.65 (1H, brs, amide N-H), 7.26 (1H, Apparent s, thiazole C5-H), 3.76 (2H, s, COCH2CO), 2.35 (3H, s, CH3CO), 2.21 (3H, Apparent, thiazole-CH3); IR (KBr) ν (cm−1): 3179 (N-H, amide), 3053 (C-H, aromatic), 2974, 2899 (C-H, aliphatic), 1723 (C=O, ketone), 1672 (C=O, amide); MS m/z (%): 198(29) [M+], 141(8), 114(100), 72(23), 43(65).
4-(2,4-Dichlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A1): Yield 95%; yellow powder; mp. 195 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.92 (s, 1H, amide N-H), 8.95 (s, 1H, NH-DHP), 7.33 (d, 1H, J = 2.0 Hz, C3-H), 7.30 (d, 1H, J = 8.5 Hz, C5-H), 7.23 (d, 1H, J = 8.5 Hz, C6-H), 6.67 (s, 1H, thiazole-H), 5.35 (s, 1H, DHP C4-H), 2.23 (s, 3H, thiazole-CH3), 2.21-2.09 (m, 2H, cyclohexanone C6-H), 1.93 (s, 3H, DHP-CH3), 1.91-1.79 (m, 2H, cyclohexanone C8-H), 1.82-1.80 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.19, 153.37, 144.15, 136.18, 132.67, 132.61. 131.38, 128.45, 127.89, 109.25, 108.61. 107.78, 37.18, 36.03, 26.84, 25.01, 21.52, 21.36, 19.02, 17.36, 17.12. IR (KBr) ν (cm−1): 3197 (DHP N-H), 3080 (C-H, aromatic), 2947 (C-H, aliphatic), 1675, 1608 (C=O, amide, ketone); MS m/z (%): 449 (30) [M + 2]+, 448 (8) [M + 1]+, 447 (22) [M], 412 (19), 334 (100), 298 (16), 272 (28), 162 (35), 115 (29), Anal. Calcd: C, 56.26; N, 9.37; O, 7.14. Found: C, 56.19; N, 9.25; O, 7.09.
4-(4-Bromophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A2): Yield 92%; yellow powder; mp. 273 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.80 (s, 1H, amide N-H), 8.97 (s, 1H, NH-DHP), 7.38 (d, 2H, J = 8.5 Hz, C3-H, C5-H), 7.13 (d, 2H, J = 8.5 Hz, C2- H, C6-H), 6.66 (s, 1H, thiazole-H), 5.08 (s, 1H, DHP C4-H), 2.48-2.46 (m, 2H, cyclohexanone C8-H), 2.24 (s, 3H, thiazole-CH3), 2.20–2.14 (m, 2H, cyclohexanone C6-H), 2.10 (s, 3H, DHP-CH3), 1.92–1.72 (2 × m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.59, 152.46, 146.72, 138.75, 131.30, 130.19, 119.37, 109.89, 107.92, 107.88, 107.83, 56.49, 37.40, 37.20, 26.75, 21.24, 19.02, 17.72, 17.30. IR (KBr) ν (cm−1): 3191 (DHP N-H), 3055 (C-H, aromatic), 2945 (C-H, aliphatic), 1667, 1607 (C=O, amide, ketone) ; MS m/z (%): 459 (34) [M + 2]+, 457 (34) [M], 346 (74), 344 (80), 317 (34), 300 (26), 264 (19), 162 (100), Anal. Calcd: C, 55.03; N, 9.17; O, 6.98. Found: C, 54.99; N, 9.03; O, 6.73.
4-(3-Chlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A3): Yield 88%; yellow powder; mp. 253 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.83 (s, 1H, amide N-H), 9.00 (s, 1H, NH-DHP), 7.22 (t, 1H, J = 8.0 Hz, C4-H), 7.18 (s, 1H, C2-H), 7.14 (t, 2H, J = 6.0 Hz, C5-H, C6-H), 6.67 (s, 1H, thiazole-H), 5.12 (s, 1H, DHP C4-H), 2.48–2.47 (m, 2H, cyclohexanone C8-H), 2.23 (s, 3H, thiazole-CH3), 2.21–2.17 (m, 2H, cyclohexanone C6-H), 2.13 (s, 3H, DHP-CH3), 1.92-1.74 (2 × m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.65, 152.63, 149.69, 139.02, 139.00, 137.63, 133.10, 130.32, 127.74, 126.69, 126.30, 109.73, 107.70, 56.49, 37.65, 37.17, 26.75, 21.22, 19.01, 17.77, 17.27. IR (KBr) ν (cm−1): 3189 (DHP N-H), 3065 (C-H, aromatic), 2970 (C-H, aliphatic), 1670, 1604 (C=O, amide, ketone), 1548, 1378 (C-NO2 aromatic); MS m/z (%): 415 (21) [M + 2]+, 413 (51) [M], 302 (47), 300 (100), 264 (36), 115 (37), Anal. Calcd: C, 60.94; N, 10.15; O, 7.73. Found: C, 60.82; N, 10.08; O, 7.46.
2-Methyl-N-(2-methylthiazol-4-yl)-5-oxo-4-(3,4,5-trimethoxyphenyl)-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A4): Yield 81%; yellow powder; mp. 160 °C; 1H-NMR (500MHz, DMSO-d6) δ: 8.99 (s, 1H, NH-DHP), 6.67 (s, 1H, thiazole-H), 6.41 (s, 2H, C2-H, C6-H), 5.09 (s, 1H, DHP C4-H), 3.62 (s, 6H, -OCH3), 3.57 (s, 3H, -OCH3), 2.56–2.51(m, 3H, thiazole-CH3), 2.26–2.23 (m, 5H, cyclohexanone C6-H, cyclohexanone C8-H), 2.09 (s, 3H, DHP-CH3), 1.96–1.81 (2 × m, 2H, cyclohexanone C7-H).13C-NMR (125MHz, DMSO-d6): 194.77, 167.75, 152.91, 162.67, 146.76, 143.03, 138.06, 136.05, 109.52, 108.65, 107.67, 104.78, 60.25, 56.48, 55.95, 37.74, 37.34, 26.83, 21.42, 19.01, 17.67, 17.30. IR (KBr) ν (cm−1): 3305 (DHP N-H), 2932 (C-H, aromatic), 2895 (C-H, aliphatic), 1609 (C=O, amide, ketone), 1267, 1002 (C-O, aromatic); MS m/z (%): 469 (19) [M]+, 356 (34), 324 (20), 300 (15), 218 (16), 170 (18), 156 (100), Anal. Calcd: C, 61.39, N, 8.95; O, 17.04. Found: C, 61.28; N, 8.75; O, 16.94.
4-(4-Chlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A5): Yield 90%; yellow powder; mp. 253 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.79 (s, 1H, amide N-H), 8.97 (s, 1H, NH-DHP), 7.24 (d, 2H, J = 8.0 Hz, C3-H, C5-H), 7.18 (d, 2H, J = 8.0 Hz, C2- , C6- H), 6.67 (s, 1H, thiazole-H), 5.09 (s, 1H, DHP C4-H), 2.48–2.46(m, 2H, cyclohexanone C8-H), 2.23 (s, 3H, thiazole-CH3), 2.23–2.14 (m, 2H, cyclohexanone C8-H), 2.10 (s, 3H, DHP-CH3), 1.92–1.75 (2 × m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.68, 172.20, 152.45, 139.28, 130.82, 129.76, 128.38, 123.93, 109.94, 43.70, 43.68, 42.70, 38.94, 37.20, 26.75, 21.24, 17.72, 17.05. IR (KBr) ν (cm−1): 3269 (DHP N-H), 3195 (C-H, aromatic), 2956 (C-H, aliphatic), 1667, 1608 (C=O, amide, ketone); MS m/z (%): 415 (10) [M + 2]+, 413 (3) [M]+, 300 (44), 298 (100), 267 (30), 239 (39), Anal. Calcd: C, 60.94; N, 10.15; O, 7.73. Found: C, 60.86; N, 10.04; O, 7.41.
4-(3-Ethoxy-4-hydroxyphenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A6): Yield 85%; yellow powder; mp. 251 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.63 (s, 1H, amide N-H), 8.85 (s, 1H, NH-DHP), 8.56 (s, 1H, OH), 6.71 (s, 1H, C2- H ), 6.62 (s, 1H, thiazole-H), 6.59 (d, 1H, J = 8.0 Hz, C5- H ), 6.52 (d, 1H, J = 8.0 Hz, C6-H), 4.98 (s, 1H, DHP C4-H), 3.91–3.76 (m, 2H, CH3-CH2-O), 2.48-2.45 (m, 2H, cyclohexanone C8-H), 2.24–2.19 (m, 5H, cyclohexanone C6-H, thiazole-CH3), 2.09 (s, 3H, DHP-CH3), 1.93–1.78 (2 × m, 2H, cyclohexanone C7-H). 1.23 (t, 3H, J = 7.0, CH3-CH2-O). 13C-NMR (125MHz, DMSO-d6): 194.66, 167.45, 155.99, 151.91, 146.97, 146.38, 145.36, 138.58, 138.29, 119.98, 115.64, 113.70, 110.50, 108.62, 107.73, 64.04, 37.34, 36.97, 26.79, 21.35, 17.70, 17.37, 15.12. (IR (KBr) ν (cm−1): 3282 (DHP N-H), 3067 (C-H, aromatic), 2990 (C-H, aliphatic), 1666, 1614 (C=O, amide, ketone); MS m/z (%): 439 (1) [M]+, 310 (10), 285 (57), 268 (14), 162 (100), Anal. Calcd: C, 62.85; N, 9.56; O, 14.56. Found: C, 62.45; N, 9.32; O, 14.18.
4-(4-Methoxyphenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A7): Yield 78%; yellow powder; mp. 185 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.82 (s, 1H, amide N-H), 8.89 (s, 1H, NH-DHP), 7.08 (d, 2H, J = 8.0 Hz, C5-H, C3-H), 6.74 (d, 2H, J = 8.0 Hz, C2- H, C6- H), 6.66 (s, 1H, thiazole-H), 5.04 (s, 1H, DHP C4-H), 3.66 (s, 3H, CH3-O), 2.47–2.44 (m, 2H, cyclohexanone C8-H), 2.23–2.17 (m, 5H, cyclohexanone C6-H, thiazole-CH3), 2.10 (s, 3H, DHP-CH3), 1.91–1.72 (2 × m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.56, 172.48, 157.83, 151.89, 139.72, 139.67, 138.54, 128.88, 113.81, 110.68, 108.51, 107.69, 55.31, 37.28, 36.74, 26.76, 21.52, 21.29, 17.71, 17.31. (IR (KBr) ν (cm−1): 3196 (DHP N-H), 3076 (C-H, aromatic), 2943 (C-H, aliphatic), 1675, 1608 (C=O, amide, ketone); MS m/z (%): 407 (15) [M − 2]+, 294 (100), 251 (5), 223 (4), Anal. Calcd: C, 64.53; N, 10.26; O, 11.72. Found: C, 64.41; N, 10.20; O, 11.58.
4-(3,4-Dimethoxyphenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide (A8): Yield 73%; yellow powder; mp. 222 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.69 (s, 1H, amide N-H), 8.88 (s, 1H, NH-DHP), 6.76 (d, 2H, J = 8.0 Hz, C5-H , thiazole-H), 6.66–6.64 (m, 2H, C2-H , C6-H ), 5.05 (s, 1H, DHP C4-H), 3.65 (s, 3H, CH3-O), 3.58 (s, 3H, CH3-O), 2.49–2.46 (m, 2H, cyclohexanone C8-H), 2.24–2.19 (m, 5H, cyclohexanone C6-H, thiazole-CH3), 2.10 (s, 3H, DHP-CH3), 1.94–1.76 (2 × m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 194.66, 167.97, 167.53, 152.10, 148.62, 147.43, 140.14, 138.35, 119.62, 112.12, 111.96, 110.28, 78.20, 55.88, 55.61, 53.32, 53.13, 37.32, 37.07, 26.70, 26.79, 21.35, 17.71, 17.33. (IR (KBr) ν (cm−1): 3229 (DHP N-H), 2937 (C-H, aromatic), 2837 (C-H, aliphatic), 1654, 1620 (C=O, amide, ketone); MS m/z (%): 437 (25) [M − 2]+, 324 (100), 308 (3), 280 (8). Anal. Calcd: C, 63.14; N, 9.60; O, 14.63. Found: C, 63.03; N, 9.46; O, 14. 46.
4-(2,4-Dichlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B1): Yield 35%; yellow powder; mp. 248 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.49 (s, 1H, amide N-H), 7.58 (s, 1H, C5-H), 7.30 (d, 2H, J = 8.5Hz, C2-H, C3-H), 6.76 (s, 1H, thiazole-H), 3.17 (m, 2H, cyclohexanone C8-H), 2.60-2.55 (m, 2H, cyclohexanone C6-H), 2.50 (s, 3H, thiazole-CH3), 2.24 (s, 3H, DHP-CH3), 2.08 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.06, 164.86, 164.46, 158.40, 156.61, 147.39, 143.96, 135.67, 133.25, 133.09, 131.16, 128.60, 127.04, 123.70, 108.88, 57.92, 33.23, 23.31, 21.43, 19.02, 17.23. IR (KBr) ν (cm−1): 3179 (DHP N-H), 3062 (C-H, aromatic), 2953 (C-H, aliphatic), 1692, 1640 (C=O, amide, ketone); MS m/z (%): 447 (2) [M + 2]+, 445 (6) [M], 410 (50), 332 (92), 296 (19), 272 (28), 165 (18), 98 (17), Anal. Calcd: C, 56.51; N, 9.41; O, 7.17. Found: C, 56.35; N, 9.32; O, 7.01.
4-(4-Bromophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B2): Yield 30%; yellow powder; mp. 261 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.42 (s, 1H, amide N-H), 7.47 (d, 2H, J = 7.0Hz, C3-H, C5-H), 7.08 (d, 2H, J = 7.0Hz, C2-H, C6-H), 6.76 (s, 1H, thiazole-H), 3.14 (t, 2H, J = 6.0Hz, cyclohexanone C8-H), 2.58 (t, 2H, cyclohexanone C6-H), 2.47 (s, 3H, thiazole-CH3), 2.23 (s, 3H, DHP-CH3), 2.11–2.08 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.37, 165.07, 164.94, 157.79, 156.67, 147.34, 146.86, 136.86, 131.48, 130.80, 130.34, 124.08, 121.20, 108.80, 79.63, 33.36, 23.12, 21.39, 17.24. IR (KBr) ν (cm−1): 3182 (DHP N-H), 3048 (C-H, aromatic), 2947 (C-H, aliphatic), 1693, 1644 (C=O, amide, ketone); MS m/z (%): 457 (21) [M + 2]+, 455 (20) [M]+, 342 (100), 262 (7), 235 (25), 162 (16), Anal. Calcd: C, 55.27; N, 9.21; O, 7.01. Found: C, 55.17; N, 9.16; O, 6.84.
4-(3-Chlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B3): Yield 25%; yellow powder; mp. 241 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.41 (s, 1H, amide N-H), 7.31–7.27 (m, 2H, C4-H, C5-H), 7.20 (s, 1H, C2-H), 7.09 (d, 1H, J = 7.0Hz, C6-H), 6.75 (s, 1H, thiazole-H), 3.15 (t, 2H, J=6.0Hz, cyclohexanone C8-H), 2.67–2.51 (m, 2H, cyclohexanone C6-H), 2.47 (s, 3H, thiazole-CH3), 2.23 (s, 3H, DHP-CH3), 2.12–2.08 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.26, 164.95, 157.63, 156.68, 147.31, 146.36, 139.61, 132.41, 131.50, 129.69, 128.08, 127.74, 126.80,123.92, 108.76, 56.49, 33.37, 23.11, 21.35, 19.01, 17.22. IR (KBr) ν (cm−1): 3154 (DHP N-H), 3062 (C-H, aromatic), 2924 (C-H, aliphatic), 1694, 1672 (C=O, amide, ketone); MS m/z (%): 413 (32) [M + 2]+, 411 (13) [M], 300 (58), 298 (100), 280 (12), 165 (21), Anal. Calcd: C, 61.24; N, 10.20; O, 7.77; Found: C, 61.13; N, 10.16; O, 7.56.
2-Methyl-N-(2-methylthiazol-4-yl)-5-oxo-4-(3,4,5-trimethoxyphenyl)-5,6,7,8-tetrahydroquinoline-3-carboxamide (B4): Yield 30%; yellow powder; mp. 238 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.39 (s, 1H, amide N-H), 6.76 (s, 1H, thiazole-H), 6.46 (s, 2H, C2-H, C6-H), 3.61 (s, 6H, -OCH3), 3.58 (s, 3H, -OCH3), 3.15 (t, 2H, J = 6.0Hz, cyclohexanone C8-H), 2.67 (t, 2H, J = 6.0Hz, cyclohexanone C6-H), 2.47 (s, 3H, thiazole-CH3), 2.23 (s, 3H, DHP-CH3), 2.13–2.10 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.29, 165.47, 164.69, 157.22, 156.88, 152.46, 147.94, 147.28, 137.13, 132.97, 131.83, 124.52, 108.60,106.26, 102.78,79.63, 60.38, 56.18, 33.36, 23.06, 21.47, 17.26. IR (KBr) ν (cm−1): 2994 (C-H, aromatic), 2960 (C-H, aliphatic), 1693, 1664 (C=O, amide, ketone); MS m/z (%): 467 (40) [M]+, 354 (100), 338 (16), 323 (21), 311 (68), Anal. Calcd: C, 61.66; N, 8.99; O, 17.11. Found: C, 61.46; N, 8.75; O, 17.07.
4-(4-Chlorophenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B5): Yield 27%; yellow powder; mp. 251 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.41 (s, 1H, amide N-H), 7.33 (d, 2H, J = 2.0Hz, C3-H, C5-H), 7.15 (d, 2H, J = 2.0Hz, C2-H, C6-H), 6.75 (s, 1H, thiazole-H), 3.14 (m, 2H, cyclohexanone C8-H), 2.50(s, 3H, thiazole-CH3 overlapped with DMSO peak), 2.57 (m, 2H, cyclohexanone C6-H), 2.22 (s, 3H, DHP-CH3), 2.09 (s, m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.36, 165.10, 164.93, 157.58, 156.68, 147.33, 146.86, 136.46, 132.53, 131.59, 130.05, 127.89, 124.11, 108.77, 30.36, 23.11, 21.39, 21.27, 17.21. (IR (KBr) ν (cm−1): 3079 (C-H, aromatic), 2967 (C-H, aliphatic), 1692, 1681 (C=O, amide, ketone); MS m/z (%): 413 (10) [M + 2]+, 411 (28) [M + 1]+, 300 (61), 280 (13), 268(6), 165(21), Anal. Calcd: C, 61.24; N, 10.20; O, 7.77. Found: C, 61.12; N, 10.11; O, 7.24.
4-(3-Ethoxy-4-hydroxyphenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B6): Yield 29%; yellow powder; mp. 267 °C; 1H-NMR (500MHz, DMSO-d6) δ: 11.62 (s, 1H, amide N-H), 8.55 (s, 1H, OH), 6.72 (s, 1H, C2-H), 6.66 (s, 1H, thiazole-H), 6.59 (d, 1H, J = 8.0 Hz, C5-H), 6.52 (d, 1H, J = 8.0 Hz, C6-H), 3.90-3.78 (m, 2H, CH3-CH2-O), 2.48–2.45 (m, 2H, cyclohexanone C8-H), 2.23 (s, 3H, C6-H, thiazole-CH3), 2.21-2.19 (m, 2H, cyclohexanone), 2.09 (s, 3H, DHP-CH3), 1.93–1.78 (2 × m, 2H, cyclohexanone C7-H), 1,23(t, 3H, J = 7.0, CH3-CH2-O). 13C-NMR (125MHz, DMSO-d6): 194.54, 152.26, 151.76, 146.45, 146.29, 140.34, 138.59, 138.10, 134.89, 118.42, 115.54, 112.32, 110.69, 108.44, 107.74, 56.02, 37.32, 36.79, 30.83, 26.78, 21.28, 17.72, 17.34. IR (KBr) ν (cm−1): 3320 (O-H), 3277 (C-H, aromatic), 2925 (C-H, aliphatic), 1667, 1602 (C=O, amide, ketone); MS m/z (%): 439 (5) [M + 2]+, 326 (8), 299 (49), 268 (23), 188 (74), 162 (100), Anal. Calcd: C, 63.00; N, 9.58; O, 14.59. Found: C, 62.87; N, 9.36; O, 14.48.
4-(4-Methoxyphenyl)-2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B7): Yield 25%; yellow powder; mp. 166 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.35 (s, 1H, amide N-H), 7.05 (d, 2H, J = 8.0 Hz, C5-H, C3-H), 6.81 (d, 2H, J = 8.0Hz, C2-H, C6-H), 6.74 (s, 1H, thiazole-H), 3.71 (s, 3H, CH3-O),3.37(s, 3H, thiazole-CH3), 3.12 (t, 2H, cyclohexanone C8-H), 2.57 (t, 2H, cyclohexanone C6-H), 2.22 (s, 3H, DHP-CH3), 2.09-2.07 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.44, 165.52, 164.61, 158.92, 157.27, 156.85, 148.01, 147.27, 131.96, 129.62, 124.74, 113.33, 108.64, 79.63, 55.37, 33.40, 23.07, 21.48, 19.02, 17.24. (IR (KBr) ν (cm−1): 3011 (C-H, aromatic), 2924 (C-H, aliphatic), 1691, 1609 (C=O, amide, ketone); MS m/z (%): 407 (4) [M]+, 294 (21), 408 (26), 269 (62), 252 (19), 162 (100), Anal. Calcd: C, 64.85; N, 10.31; O, 11.78. Found: C, 64.65; N, 10.41; O, 11.64.
4-(3,4-Dimethoxyphenyl)-2-methyl-N-(5-methylthiazol-2-yl)-5-oxo-5,6,7,8-tetrahydroquinoline-3-carboxamide (B8): Yield 30%; yellow powder; mp. 219 °C; 1H-NMR (500MHz, DMSO-d6) δ: 12.42 (s, 1H, amide N-H), 6.83 (d, 1H, J = 8.0Hz, C6-H), 6.79 (s, 1H, J = 8.0Hz, C2-H), 6.76 (s, 1H, thiazole-H), 6.64 (d, 1H, J = 8.0Hz, C5-H), 3.71 (s, 3H, CH3-O), 3.60 (s, 3H, CH3-O), 3.12 (t, 2H, cyclohexanone C8-H), 2.59-2.55 (m, 2H, cyclohexanone C6-H), 2.45 (s, 3H, thiazole-CH3), 2.23 (s, 3H, DHP-CH3), 2.09-2.11-2.08 (m, 2H, cyclohexanone C7-H). 13C-NMR (125MHz, DMSO-d6): 197.45, 165.58, 164.66, 157.18, 156.88, 148.50, 148.05, 147.93, 147.32, 147.25, 131.94, 129.73, 124.84, 121.10, 112.47, 111.34, 108.65, 63.26, 55.76, 33.37, 23.05, 21.49, 17.25. (IR (KBr) ν (cm−1): 3117 (C-H, aromatic), 2936 (C-H, aliphatic), 1667, 1604 (C=O, amide, ketone); MS m/z (%): 437 (1) [M + 2]+, 407 (4), 294 (18), 269 (65), 252 (15), 162 (100), Anal. Calcd: C, 63.14; N, 9.60; O, 14.63. Found: C, 62.59; N, 9.40; O, 14.82.

2.2. Biological Evaluation

2.2.1. Cell Lines

Cell lines used for cytotoxicity assays included MCF-7 (human breast adenocarcinoma), A549 (human lung adenocarcinoma), K562 (human chronic myelogenous leukemia) and HEK-293 (human embryonic kidney) cells and were obtained from the Iranian Biological Resource Center, Tehran, Iran. MES-SA-DX5 multidrug resistant human uterine sarcoma, over-expressing P-gp and their parental non-resistant MES-SA cell lines were purchased from Sigma-Aldrich (St. Louis, MO, USA). All cells, except for HEK-293, were cultured in RPMI 1640 supplemented with 10% FBS and 100 units/mL penicillin-streptomycin at 37 °C in humidified air containing 5% CO2. HEK-293 cells were maintained in DMEM-F12 medium containing L-glutamine 2 mM supplemented with FBS and penicillin-streptomycin. MES-SA-DX5 cells were maintained in media containing 100 nM doxorubicin in order to ensure that only drug-resistant cells continue to grow.

2.2.2. Reagents

RPMI 1640 and DMEM-F12 growth mediua, L-glutamine, penicillin and streptomycin were purchased from Biosera (Nuaille, France). Fetal bovine serum (FBS) was obtained from Gibco (Carlsbad, CA, USA). Doxorubicin, cisplatin and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were purchased from EBEWE Pharma (Unterach, Austria) and Sigma Aldrich (St. Louis, MO, USA).

2.2.3. MDR Reversal Assay

Rhodamine 123 (Rh123) is a well-known substrate of P-gp and it is readily pumped out of the cells by this ABC transporter. The amount of retained Rh123 in the cells indicates the efflux function of the target transporter. Flow cytometric analysis was used for assessment of intracellular Rh123 as previously described [23].
Briefly, a suspension of the multidrug resistant uterine sarcoma MES-SA-DX5 cells at a density of 5 × 105 cells/mL in growth medium was prepared. Four-hundred microliters (400 µL) of the synthesized compounds with final concentration of 5, 10, and 25 µM were added to 500 µL of the cell suspension. Verapamil was also tested as a positive control. After an incubation of 20 min, 50 µL of Rh123 at a final concentration of 5 µM was added and incubated for further 30 min at 37 °C. The cells were then centrifuged and washed twice with ice-cold PBS, and then resuspended in PBS and kept on ice. Rh123 accumulated in cells was then quantified by a FACSCalibur flow cytometer (Becton Dickinson, San Jose, CA, USA) with excitation and emission wavelengths of 488 and 530 nm, respectively. Geometric means of fluorescence intensity values were determined using Flowing Software (http://flowingsoftware.btk.fi/).

2.2.4. Cell Cycle Analysis

The analysis of cells in different phases of cell cycle and sub-G1 was performed using propidium iodide (PI)-RNase assay by flow cytometry. MES-SA-DX5 cells were seeded in 12-well plates (1 × 106 cells/well) and treated with different concentrations of synthesized compounds for 24 h. The cells were then collected, washed with PBS, and fixed with 70% ethanol overnight at –20 °C. After at least 24 h, fixed cells were washed with PBS and subsequently stained with a DNA staining solution containing PI 20 μg/mL and RNase 200 μg/mL at room temperature for 30 min in the dark. Twenty thousand cells of each sample were analyzed using a BD FACSCalibur flow cytometer (BD Biosciences, USA) and the amount of the cells in sub-G1, G0/G1, S, and G2/M phases were estimated using CellQuest (Becton Dickinson, San Jose, CA, USA) software.

2.2.5. Cell Viability Assay

Cytotoxicity of the test compounds were evaluated by MTT assay [24]. In this method, reduction of MTT, a yellow tetrazolium salt to the purple formazan, by cellular dehydrogenase enzymes is determined as an index of cell viability. Cancer cells were seeded into 96-well plates (3–5 × 103 cells/well) and incubated for 24 h at 37 °C. Afterwards, four different concentrations of synthesized derivatives were added in triplicate and the cells were incubated at 37 °C for another 72 h, after which the media was replaced with MTT solution at a concentration of 0.5 mg/mL was added for MTT assay. After another 4h, formazan crystals were dissolved in 200 μL DMSO for 90 min and absorbance was measured at a wavelength of 570 nm with background correction at 650 nm using a microplate reader (model 680, Bio-Rad, Hercules, CA, USA) and IC50 for each compound was calculated with CurveExpert version 1.34 for Windows.

2.3. Statistical Analysis

All experiments were carried out in triplicate and were repeated 3–5 times. The results are reported as mean ± S.E.M.. The differences between various treatments were analyzed by one-way ANOVA with an LSD post hoc test. Statistical analysis was performed using the SPSS program version 14.0 (Chicago, IL, USA) for Windows.

3. Results and Discussion

3.1. Synthesis

The synthetic routes for the target compounds A1A8 and B1B8 are depicted in Scheme 1 and the structures are presented in Table 1. Compound 1-((2-methylthiazol-4-yl) amino) pentane-2,4-dione was synthesized by the reaction of the commercially available 2-methylthiazol-4-amine with 2,2,6-trimethyl-4H-1,3-dioxin-4-one in xylene under reflux condition. According to Scheme 1, the final products were synthesized by treatment of obtained intermediate with 1,3-cyclohexadione and different appropriate arylaldehydes in the presence of excess amounts of ammonium acetate in refluxing ethanol. In order to synthesis the tetrahydroquinoline derivatives; the corresponding hexahydroquinoline compound was oxidized in the presence of MnO2 in ethanol under reflux condition for 24–48 h.

3.2. Biological Evaluations

3.2.1. MDR Reversal Assay

Accumulation of Rhodamine123 (Rh123), caused by inhibition of P-gp-mediated efflux, was determined by flow cytometry in the multidrug resistant uterine sarcoma MES-SA-DX5 cell line. Verapamil was used as a positive control. MES-SA-DX5 is a resistant cell line, which overexpresses P-gp as a result of constant exposure to doxorubicin. Alterations in the amount of the fluorescent Rh123 retained inside the MES-SA-DX5 cells can be plausibly related to the inhibition of the activity of P-gp efflux pump in the cells (Figure 1). The ratio of the geometric mean value of cells treated with synthesized compounds to the geometric mean value of the control untreated cells are show in Figure 2. The findings demonstrate a clear dose-dependent effect at 5, 10, and 25 µM on Rh123 accumulation for most of the compounds. Compounds A1, A2, and B2 showed the highest P-gp inhibitory activities.

3.2.2. Effect of Synthesized Derivatives on Cell Cycle

The effect of the most potent compounds on the cell cycle distribution and apoptosis induction in MES-SA-DX5 resistant cells was evaluated using propidium iodide (PI)-RNase assay by flow cytometry. Table 2 shows the distribution of the cells in the sub-G1, G0/G1, S and G2/M phases of the cell cycle. The results indicate that the number of cells in sub-G1 phase, which shows the apoptotic cells, is significantly increased after treatment with 100 µM concentrations of A1, A2, A3, A5, B1, B2, and B5 compounds. Therefore, it could be concluded that these compounds cause cytotoxicity in drug resistant MES-SA-DX5 cells by induction of apoptosis in these cells. Furthermore, treatment of MES-SA-DX5 cells with compounds A1 and B1 caused a considerable increase in G2/M and a decrease in G0/G1 phase cells. Representative histograms of the most potent derivatives are shown in Figure 3.

3.2.3. Cell Viability Assay

Cell viability of different cancer cell lines exposed to synthesized derivatives was evaluated using the MTT reduction assay. The average IC50 values in tested cell lines are summarized in Table 3. In general, the compounds had the lowest IC50 values against K562 cells, which is usually expected in hematological malignancies compared to solid tumors.
Interestingly, among tested solid tumors, the synthesized derivatives A1, A2, A5, B1, and B2 were more effective against MES-SA-DX5 cells. These findings are particularly interesting if we note that MES-SA-DX5 is a multidrug resistant cell line and any compound effective against these cancer cells could have high potential as therapeutic agents against drug resistant tumors. We also tested the more promising derivatives against parental non-resistant MES-SA cells and the IC50 values were invariably higher in these cells compared to MES-SA-DX5-resistant cells. It should be noted that parental MES-SA cells are more than 200 times more sensitive to doxorubicin compared to resistant cells.
Since these compounds have a moderate effect against cancer cell lines, it can be deduced that they should possess an inherent cytotoxicity. On the other hand, they also have a pronounced P-gp inhibitory effect (Figure 2). Indeed, the combination of these two biological properties may lead to an additive effect in resistant cells. In other words, the P-gp inhibitory activity contributes to the retention of the derivatives within the cell, where they can better exert their antiproliferative action.
The effect of the active compounds was also tested against HEK-293 non-cancerous cell line. Compounds A2, A3, A5, B2, B3, and B5 were derivatives with IC50 values higher than 50 µM in these normal cells. Putting together all MTT results, it can be concluded that compounds A2 and B2 are the most promising agents, which show relative selectivity of action against resistant cells compared to non-resistant and also non-cancerous cell lines.

3.2.4. Structure-Activity Relationships

Considering the MDR reversal and cytotoxicity results, the following structure-activity relationships could be construed for the synthesized compounds:

3.2.5. Comparison between 1,4-Dihydropyridine and Pyridine Structures

The findings in MES-DX-5 drug resistant cells indicated that oxidation of 1,4-dihydropyridine ring in A (hexahydroquinoline) compounds could not significantly alter the MDR reversal effect of derivatives as compared with B (tetrahydroquinoline) series. As it can be observed in Figure 1 and Figure 2, A compounds show only a slight overall superiority over B agents. Both groups of hexahydroquinoline and tetrahydroquinoline derivatives in general demonstrated weak to moderate cytotoxic effects against MES-SA-DX5 cells in the MTT assay (Table 3).
On the other hand, oxidation of 1,4-dihydropyridine ring to pyridine counterpart resulted in an increase of the cytotoxic potential of compounds against K562 cells in most cases; e.g., B1 (IC50 = 6.7 µM), B2 (IC50 = 10.1 µM) and B5 (IC50 = 10.4 µM) demonstrated superior activity compared to their pyridine counterparts.

3.2.6. Influence of the Substituted Group at C4 Position of the Phenyl Ring

The MDR reversal results indicated that most of the compounds bearing halogen moieties including Cl (A1, A3, A5, B1, B5) and Br (A2 and B2) on the phenyl ring showed superior MDR reversal effect compared to the compounds bearing other substitutes, such as methoxy, methyl, and hydroxyl, in both A and B series.
As for the cytotoxic effect, the findings showed that the best compounds especially against K562 cells were those bearing halogen moieties on the para position of phenyl ring (A1, A2, B1, B2) compared to compounds bearing other substitutes in both A and B series. It should also be mentioned that meta-chlorinated derivatives (A3 and B3) demonstrated weaker cytotoxic activity in cell lines compared to para-chlorinated counterparts (A5 and B5).

4. Conclusions

A series of 2-methyl-N-(2-methylthiazol-4-yl)-5-oxo-1,4,5,6,7,8-hexahydroquinoline-3-carboxamide derivatives (A series) and their tetrahydroquinoline analogs (B series) were synthetized and evaluated for MDR reversal and cytotoxic properties. Examination of MDR reversal in MES-SA-DX5 drug resistant cells by flow cytometric detection of Rh123 showed that compounds especially those bearing Cl and Br moieties in both hexahydroquinoline and tetrahydroquinoline subgroups are able to reverse MDR in a dose dependent manner. The flow cytometric analysis of cell cycle distribution in MES-SA-DX5 cells after treatment with some of the synthesized compounds showed that especially compounds A1 and A2 with 5-oxo-hexahydroquinoline structure bearing 2,4-dichlorophenyl and 4-bromophenyl moieties, respectively, and their tetrahydroquinoline counterparts B1 and B2 induce apoptosis in MDR cells. Compounds A2 and B2 did not show any effect against parental non-resistant MES-SA cells and had higher IC50 values against HEK-293 non-cancerous cells compared to cancer cells. The results of this report show that the synthesized derivatives deserve further studies for discovery of novel MDR reversal agents.

Supplementary Materials

Chemical characterization data (1H NMR, 13C NMR, Mass and IR spectra) of synthesized compounds A1A8 and B1B8 are available online, Tables S1–S64.

Author Contributions

Conceptualization, M.K., N.E. and O.F.; methodology, O.S., M.K., N.E. and O.F.; software, O.S., N.E. and O.F.; validation, M.K., N.E. and O.F.; formal analysis, O.S., M.D., M.M and M.T.; investigation, O.S., M.D., M.M and M.T.; resources, L.S., N.E. and O.F.; data curation, O.S., M.M and M.T.; writing—original draft preparation, O.S., M.D., M.T., M.M.; writing—review and editing, M.K., L.S., N.E., O.F.; visualization, O.S., M.D., M.M and M.T.; supervision, M.K., N.E. and O.F.; project administration, M.K., N.E. and O.F.; funding acquisition, M.K., L.S. and O.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institute for Medical Research Development (NIMAD), Tehran, Iran, grant number 943656 and also the Shiraz University of Medical Sciences, Vice Chancellor for Research, Shiraz, Iran.

Acknowledgments

Part of this study was performed within the Pharm. D. thesis of Mojtaba Dehghani at Shiraz University of Medical Sciences.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations:

P-gp, P-glycoprotein; MDR, multidrug resistance; ABC, ATP binding cassette; Rh123, Rhodamine 123; DMSO, dimethylsulfoxide; TLC, thin layer chromatography; MTT, 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide reagent; DHPs, dihydropyridines

References

  1. Fitzmaurice, C.; Akinyemiju, T.F.; Al Lami, F.H.; Alam, T.; Alizadeh-Navaei, R.; Allen, C.; Alsharif, U.; Alvis-Guzman, N.; Amini, E.; Anderson, B.O.; et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2016: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2018, 4, 1553–1568. [Google Scholar] [PubMed]
  2. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP–dependent transporters. Nat. Rev. Cancer. 2002, 2, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer. 2018, 18, 452–464. [Google Scholar] [CrossRef] [PubMed]
  4. Breier, A.; Gibalova, L.; Seres, M.; Barancik, M.; Sulova, Z. New insight into p-glycoprotein as a drug target. Anticancer Agents Med Chem. 2013, 13, 159–170. [Google Scholar] [CrossRef]
  5. Juliano, R.L.; Ling, V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochimica et Biophysica Acta (BBA)-Biomembranes 1976, 455, 152–162. [Google Scholar] [CrossRef]
  6. Teodori, E.; Dei, S.; Martelli, C.; Scapecchi, S.; Gualtieri, F. The functions and structure of ABC transporters: Implications for the design of new inhibitors of Pgp and MRP1 to control multidrug resistance (MDR). Curr. Drug Targets 2006, 7, 893–909. [Google Scholar] [CrossRef]
  7. Li, X.; Yuan, H.; Wu, J.; Li, J.; Qu, X.; Xu, W.; Tang, W. Strategies to overcome or circumvent P-glycoprotein mediated multidrug resistance. Curr. Med. Chem. 2008, 15, 470–476. [Google Scholar] [CrossRef]
  8. Kuppens, I.E.; Witteveen, E.O.; Jewell, R.C.; Radema, S.A.; Paul, E.M.; Mangum, S.G.; Beijnen, J.H.; Voest, E.E.; Schellens, J.H. A phase I, randomized, open-label, parallel-cohort, dose-finding study of elacridar (GF120918) and oral topotecan in cancer patients. Clin. Cancer Res. 2007, 13, 3276–3285. [Google Scholar] [CrossRef] [Green Version]
  9. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Inhibition of P-glycoprotein (ABCB1)-and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1®. Biochem. Pharmacol. 2008, 75, 1302–1312. [Google Scholar] [CrossRef] [Green Version]
  10. Miri, R.; Javidnia, K.; Mirkhani, H.; Hemmateenejad, B.; Sepeher, Z.; Zalpour, M.; Behzad, T.; Khoshneviszadeh, M.; Edraki, N.; Mehdipour, A.R. Synthesis, QSAR and calcium channel modulator activity of new hexahydroquinoline derivatives containing nitroimidazole. Chem. Biol. Drug Des. 2007, 70, 329–336. [Google Scholar] [CrossRef]
  11. Miri, R.; Javidnia, K.; Sarkarzadeh, H.; Hemmateenejad, B. Synthesis, study of 3D structures, and pharmacological activities of lipophilic nitroimidazolyl-1, 4-dihydropyridines as calcium channel antagonist. Bioorg. Med. Chem. 2006, 14, 4842–4849. [Google Scholar] [CrossRef] [PubMed]
  12. Edraki, N.; Mehdipour, A.R.; Khoshneviszadeh, M.; Miri, R. Dihydropyridines: Evaluation of their current and future pharmacological applications. Drug Discov. Today 2009, 14, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
  13. Tomassoni, D.; Lanari, A.; Silvestrelli, G.; Traini, E.; Amenta, F. Nimodipine and its use in cerebrovascular disease: Evidence from recent preclinical and controlled clinical studies. Clin. Exp. Hypertens 2008, 30, 744–766. [Google Scholar] [CrossRef] [PubMed]
  14. Khedkar, S.A.; Auti, P.B. 1,4-Dihydropyridines: A class of pharmacologically important molecules. Mini Rev. Med. Chem. 2014, 14, 282–390. [Google Scholar] [CrossRef]
  15. Viradiya, D.; Mirza, S.; Shaikh, F.; Kakadiya, R.; Rathod, A.; Jain, N.; Rawal, R.; Shah, A. Design and Synthesis of 1,4-dihydropyridine Derivatives as Anti-Cancer Agent. Anticancer Agents Med. Chem. 2017, 17, 1003–1013. [Google Scholar] [CrossRef]
  16. Steiger, S.A.; Li, C.; Backos, D.S.; Reigan, P.; Natale, N. Dimeric isoxazolyl-1, 4-dihydropyridines have enhanced binding at the multi-drug resistance transporter. Bioorg. Med. Chem. 2017, 25, 3223–3234. [Google Scholar] [CrossRef]
  17. Krauze, A.; Grinberga, S.; Krasnova, L.; Adlere, I.; Sokolova, E.; Domracheva, I.; Shestakova, I.; Andzans, Z.; Duburs, G. Thieno [2, 3-b] pyridines—a new class of multidrug resistance (MDR) modulators. Bioorg. Med. Chem. 2014, 22, 5860–5870. [Google Scholar] [CrossRef]
  18. Shekari, F.; Sadeghpour, H.; Javidnia, K.; Saso, L.; Nazari, F.; Firuzi, O.; Miri, R. Cytotoxic and multidrug resistance reversal activities of novel 1, 4-dihydropyridines against human cancer cells. Eur. J. Pharmacol. 2015, 746, 233–244. [Google Scholar] [CrossRef]
  19. Radadiya, A.; Khedkar, V.; Bavishi, A.; Vala, H.; Thakrar, S.; Bhavsar, D.; Shah, A.; Coutinho, E. Synthesis and 3D-QSAR study of 1, 4-dihydropyridine derivatives as MDR cancer reverters. Eur. J. Med. Chem. 2014, 74, 375–387. [Google Scholar] [CrossRef]
  20. Ranjbar, S.; Firuzi, O.; Edraki, N.; Shahraki, O.; Saso, L.; Khoshneviszadeh, M.; Miri, R. Tetrahydroquinolinone derivatives as potent P-glycoprotein inhibitors: Design, synthesis, biological evaluation and molecular docking analysis. MedChemComm 2017, 8, 1919–1933. [Google Scholar] [CrossRef]
  21. Firuzi, O.; Javidnia, K.; Mansourabadi, E.; Saso, L.; Mehdipour, A.R.; Miri, R. Reversal of multidrug resistance in cancer cells by novel asymmetrical 1,4-dihydropyridines. Arch. Pharm. Res. 2013, 36, 1392–1402. [Google Scholar] [CrossRef]
  22. Shahraki, O.; Edraki, N.; Khoshneviszadeh, M.; Zargari, F.; Ranjbar, S.; Saso, L.; Firuzi, O.; Miri, R. Novel 5-oxo-hexahydroquinoline derivatives: Design, synthesis, in vitro P-glycoprotein-mediated multidrug resistance reversal profile and molecular dynamics simulation study. Drug Des. Dev. Ther. 2017, 11, 407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ranjbar, S.; Khonkarn, R.; Moreno, A.; Baubichon-Cortay, H.; Miri, R.; Khoshneviszadeh, M.; Saso, L.; Edraki, N.; Falson, P.; Firuzi, O. 5-Oxo-hexahydroquinoline derivatives as modulators of P-gp, MRP1 and BCRP transporters to overcome multidrug resistance in cancer cells. Toxicol. Appl. Pharmacol. 2019, 362, 136–149. [Google Scholar] [CrossRef] [PubMed]
  24. Gaspar, A.; Mohabbati, M.; Cagide, F.; Razzaghi-Asl, N.; Miri, R.; Firuzi, O.; Borges, F. Searching for new cytotoxic agents based on chromen-4-one and chromane-2,4-dione scaffolds. Res. Pharm. Sci. 2019, 14, 74–83. [Google Scholar] [PubMed]
Sample Availability: Samples of the compounds are available from the authors.
Scheme 1. Synthesis of the 5-oxo-hexahydroquinoline derivatives and their tetrahydroquinoline counterparts. Reagents and conditions: (i) xylene, reflux, 2–4 h; (ii) CH3COONH4, ethanol, reflux, 24 h; (iii) MnO2, ethanol, reflux, 24–48 h.
Scheme 1. Synthesis of the 5-oxo-hexahydroquinoline derivatives and their tetrahydroquinoline counterparts. Reagents and conditions: (i) xylene, reflux, 2–4 h; (ii) CH3COONH4, ethanol, reflux, 24 h; (iii) MnO2, ethanol, reflux, 24–48 h.
Molecules 25 01839 sch001
Figure 1. Flow cytometric detection of Rhodamine123 efflux in MES-SA-DX5 drug resistant cells. Cells were suspended in RPMI 1640 and treated with synthesized derivatives A1, A2, B1, and B2. After incubation for 20 min, 5 μM Rh123 was added and the cells incubated for another 20 min at 37 °C. Afterwards, the cells were centrifuged and washed twice with ice-cold PBS and resuspended in PBS. The fluorescence signal caused by the presence of Rh123 inside the cells was measured using a flow cytometer with excitation and emission wavelengths of 488 nm and 530 nm, respectively. Representative histograms of cells treated with different concentrations of test compounds are shown.
Figure 1. Flow cytometric detection of Rhodamine123 efflux in MES-SA-DX5 drug resistant cells. Cells were suspended in RPMI 1640 and treated with synthesized derivatives A1, A2, B1, and B2. After incubation for 20 min, 5 μM Rh123 was added and the cells incubated for another 20 min at 37 °C. Afterwards, the cells were centrifuged and washed twice with ice-cold PBS and resuspended in PBS. The fluorescence signal caused by the presence of Rh123 inside the cells was measured using a flow cytometer with excitation and emission wavelengths of 488 nm and 530 nm, respectively. Representative histograms of cells treated with different concentrations of test compounds are shown.
Molecules 25 01839 g001aMolecules 25 01839 g001b
Figure 2. MDR reversal assessed by flow cytometric detection of Rhodamine123 efflux in MES-SA-DX5 cells. Cells were suspended in RPMI 1640 and treated with verapamil (Ver, positive control) or synthesized derivatives. After incubation for 20 min, 5 μM Rh123 was added, the cells were incubated for another 20 min at 37 °C and then centrifuged and washed twice with ice-cold PBS and resuspended in PBS. The fluorescence caused by the presence of Rh123 in cells was measured using a flow cytometer with excitation and emission wavelengths of 488 nm and 530 nm, respectively. The values represent the mean ± S.E.M. of 3–5 experiments. * The difference between treated and controlled cells are statistically significant (p ˂ 0.05).
Figure 2. MDR reversal assessed by flow cytometric detection of Rhodamine123 efflux in MES-SA-DX5 cells. Cells were suspended in RPMI 1640 and treated with verapamil (Ver, positive control) or synthesized derivatives. After incubation for 20 min, 5 μM Rh123 was added, the cells were incubated for another 20 min at 37 °C and then centrifuged and washed twice with ice-cold PBS and resuspended in PBS. The fluorescence caused by the presence of Rh123 in cells was measured using a flow cytometer with excitation and emission wavelengths of 488 nm and 530 nm, respectively. The values represent the mean ± S.E.M. of 3–5 experiments. * The difference between treated and controlled cells are statistically significant (p ˂ 0.05).
Molecules 25 01839 g002
Figure 3. Effect of synthesized derivatives on cell cycle alterations in MES-SA-DX5 drug resistant cells. The cells in different phases of cell cycle were monitored using propidium iodide (PI)-RNase assay by flow cytometry. MES-SA-DX5 cells were seeded in 12-well plates (1 × 106 cells/well) and treated with different concentrations of synthesized compounds for 24 h. The cells were then collected, washed with PBS and fixed with 70% ethanol overnight at –20 °C. After at least 24 h, fixed cells were washed with PBS and subsequently stained with a DNA staining solution containing PI 20 μg/mL and RNase 200 μg/mL at room temperature for 30 min in the dark. Twenty thousand cells of each sample were analyzed using a FACS Calibur flow cytometer (BD Biosciences, USA). Representative histograms of cells treated with synthesized compounds are shown. An increase in sub-G1 phase cells could be observed after treatment with A1, A2, B1, and B2 compounds.
Figure 3. Effect of synthesized derivatives on cell cycle alterations in MES-SA-DX5 drug resistant cells. The cells in different phases of cell cycle were monitored using propidium iodide (PI)-RNase assay by flow cytometry. MES-SA-DX5 cells were seeded in 12-well plates (1 × 106 cells/well) and treated with different concentrations of synthesized compounds for 24 h. The cells were then collected, washed with PBS and fixed with 70% ethanol overnight at –20 °C. After at least 24 h, fixed cells were washed with PBS and subsequently stained with a DNA staining solution containing PI 20 μg/mL and RNase 200 μg/mL at room temperature for 30 min in the dark. Twenty thousand cells of each sample were analyzed using a FACS Calibur flow cytometer (BD Biosciences, USA). Representative histograms of cells treated with synthesized compounds are shown. An increase in sub-G1 phase cells could be observed after treatment with A1, A2, B1, and B2 compounds.
Molecules 25 01839 g003
Table 1. Chemical structures of synthesized 5-oxo-hexahyroquinoline derivatives and their tetrahydroquinoline counterparts.
Table 1. Chemical structures of synthesized 5-oxo-hexahyroquinoline derivatives and their tetrahydroquinoline counterparts.
Molecules 25 01839 i001
CompoundArMWCompoundArMW
A1 Molecules 25 01839 i002448.36B1 Molecules 25 01839 i003446.35
A2 Molecules 25 01839 i004458.37B2 Molecules 25 01839 i005456.36
A3 Molecules 25 01839 i006413.92B3 Molecules 25 01839 i007411.90
A4 Molecules 25 01839 i008469.56B4 Molecules 25 01839 i009467.54
A5 Molecules 25 01839 i010413.92B5 Molecules 25 01839 i011411.90
A6 Molecules 25 01839 i012439.53B6 Molecules 25 01839 i013437.51
A7 Molecules 25 01839 i014409.50B7 Molecules 25 01839 i015407.49
A8 Molecules 25 01839 i016439.53B8 Molecules 25 01839 i017437.51
Table 2. Flow cytometric analysis of cell cycle distribution of MES-SA-DX5 cells treated with synthesized compounds.
Table 2. Flow cytometric analysis of cell cycle distribution of MES-SA-DX5 cells treated with synthesized compounds.
CompoundSub G1G0/G1SG2/M
Control1.99 ± 1.252.71 ± 0.820.29 ± 0.724.99 ± 0.6
A1 (10 µM)0.61 ± 0.252.32 ± 1.821.85 ± 1.025.12 ± 1.7
A1 (25 µM)0.71 ± 0.253.30 ± 0.920.96 ± 1.225.01 ± 0.7
A1 (100 µM)4.23 ± 0.6*39.18 ± 1.721.12 ± 1.635.48 ± 2.7
A2 (10 µM)1.21 ± 0.756.45 ± 1.118.90 ± 0.223.45 ± 0.2
A2 (25 µM)1.71 ± 1.159.47 ± 3.018.53 ± 1.720.23 ± 0.7
A2 (100 µM)6.88 ± 3.0*50.57 ± 2.819.27 ± 0.318.37 ± 2.6
A3 (10 µM)0.51 ± 0.150.35 ± 4.518.23 ± 2.623.25 ± 1.6
A3 (25 µM)0.97 ± 0.256.18 ± 1.219.70 ± 2.023.08 ± 1.0
A3 (100 µM)3.59 ± 0.7*53.64 ± 1.413.31 ± 1.029.46 ± 1.8
A5 (10 µM)0.65 ± 0.256.07 ± 0.319.03 ± 0.924.20 ± 1.2
A5 (25 µM)0.70 ± 0.358.03 ± 0.718.90 ± 0.922.33 ± 0.5
A5 (100 µM)3.17 ± 0.6*48.92 ± 1.418.58 ± 0.629.32 ± 1.2
B1 (10 µM)0.64 ± 0.149.10 ± 3.721.27 ± 2.129.03 ± 2.1
B1 (25 µM)0.91 ± 0.051.13 ± 2.420.90 ± 2.027.03 ± 0.6
B1 (100 µM)5.08 ± 0.7*24.00 ± 4.125.17 ± 0.745.77 ± 3.4
B2 (10 µM)0.96 ± 0.153.00 ± 3.621.80 ± 1.524.13 ± 2.4
B2 (25 µM)1.57 ± 0.455.83 ± 2.620.30 ± 1.722.23 ± 1.4
B2 (100 µM)4.25 ± 0.5*47.00 ± 0.619.48 ± 1.829.27 ± 1.8
B5 (10 µM)0.62 ± 0.153.87 ± 0.718.13 ± 1.927.27 ± 2.6
B5 (25 µM)0.69 ± 0.155.43 ± 1.317.73 ± 1.326.07 ± 2.7
B5 (100 µM)1.91 ± 0.3*57.58 ± 0.913.62 ± 0.826.87 ± 0.9
Doxorubicin (2.5 µM)1.08 ± 0.234.23 ± 3.312.57 ± 1.452.10 ± 4.4
Doxorubicin (5 µM)1.81 ± 0.321.50 ± 2.512.57 ± 1.764.13 ± 4.2
Values represent mean ± S.E.M. of 3–5 independent experiments. * The difference with control untreated cells was significantly different (p ˂ 0.05). Significantly increased sub-G1 values are shown in boldface for emphasis.
Table 3. Cytotoxic activity of synthetic compounds assessed by the MTT reduction assay.
Table 3. Cytotoxic activity of synthetic compounds assessed by the MTT reduction assay.
CompoundIC50 (µM) in Cultured Cells
MCF-7A-549K562MES-SA-DX5MES-SAHEK-293
A172.5 ± 11.948.8 ± 1.315.0 ± 0.540.1 ± 4.764.1 ± 19.428.6 ± 4.3
A256.7 ± 3.687.1 ± 6.731.0 ± 2.731.5 ± 1.5>10063.1 ± 7.4
A3>100>10056.4 ± 7.078.0 ± 4.0>10093.7 ± 11.9
A4>100>100>100>100--
A572.9 ± 12.387.4 ± 1.5 31.5 ± 3.340.3 ± 3.0 69.5 ± 12.571.1 ± 10.1
A6>100>100>100>100--
A7>100>100>100>100--
A8>100>100>100>100--
B147.1 ± 2.160.0 ± 3.06.7 ± 0.639.8 ± 1.655.6 ± 2.439.9 ± 5.4
B277.7 ± 8.7 86.8 ± 5.010.1 ± 1.543.6 ± 2.8 >10083. 2 ± 10.6
B3>100>10050.2 ± 2.6 85.2 ± 7.4 >100>100
B4>100>100>100>100--
B589.5 ± 3.591.0 ± 4.610.4 ± 1.562.3 ± 5.5>10064.2 ± 1.1
B6>100>100>100>100--
B7>100>10032.9 ± 5.1 >100--
B8>100>10067.5 ± 8.0 >100--
Doxorubicin0.115 ± 0.042 0.575 ± 0.127 0.042 ± 0.006 2.2 ± 0.30.010 ± 0.0020.009 ± 0.002
Cisplatin26.3 ±7.914.0 ± 0.53.3 ± 2.38.6 ± 0.51.2 ± 0.11.1 ± 0.5
IC50 values are presented as mean ± S.E.M. of 3–5 independent experiments.

Share and Cite

MDPI and ACS Style

Shahraki, O.; Khoshneviszadeh, M.; Dehghani, M.; Mohabbati, M.; Tavakkoli, M.; Saso, L.; Edraki, N.; Firuzi, O. 5-Oxo-hexahydroquinoline Derivatives and Their Tetrahydroquinoline Counterparts as Multidrug Resistance Reversal Agents. Molecules 2020, 25, 1839. https://doi.org/10.3390/molecules25081839

AMA Style

Shahraki O, Khoshneviszadeh M, Dehghani M, Mohabbati M, Tavakkoli M, Saso L, Edraki N, Firuzi O. 5-Oxo-hexahydroquinoline Derivatives and Their Tetrahydroquinoline Counterparts as Multidrug Resistance Reversal Agents. Molecules. 2020; 25(8):1839. https://doi.org/10.3390/molecules25081839

Chicago/Turabian Style

Shahraki, Omolbanin, Mehdi Khoshneviszadeh, Mojtaba Dehghani, Maryam Mohabbati, Marjan Tavakkoli, Luciano Saso, Najmeh Edraki, and Omidreza Firuzi. 2020. "5-Oxo-hexahydroquinoline Derivatives and Their Tetrahydroquinoline Counterparts as Multidrug Resistance Reversal Agents" Molecules 25, no. 8: 1839. https://doi.org/10.3390/molecules25081839

Article Metrics

Back to TopTop