1,921
Views
44
CrossRef citations to date
0
Altmetric
Research Article

Synthesis and potential antitumor activity of 7-(4-substituted piperazin-1-yl)-4-oxoquinolines based on ciprofloxacin and norfloxacin scaffolds: in silico studies

, , , , , , & show all
Pages 796-809 | Received 30 Apr 2015, Accepted 30 May 2015, Published online: 30 Jul 2015

Abstract

The potential antitumor activities of a series of 7-(4-substituted piperazin-1-yl)fluoroquinolone derivatives (114a,b) using ciprofloxacin and norfloxacin as scaffolds are described. These compounds exhibit potent and broad spectrum antitumor activities using 60 human cell lines in addition to the inherent antibacterial activity. Compounds 1a, 2a, 3b, 6b and 7a were found to be the most potent, while 2b, 5b, and 6a were found to have an average activity. The results of this study demonstrated that compounds 1a, 2a, 3b, 6b and 7a (mean GI50; 2.63–3.09 µM) are nearly 7-fold more potent compared with the positive control 5-fluorouracil (mean GI50; 22.60 µM). More interestingly, compounds 1a, 2a, 3b, 6b and 7a have an almost antitumor activity similar to gefitinib (mean GI50; 3.24 µM) and are nearly 2-fold more potent compared to erlotinib (mean GI50; 7.29 µM). In silico study and ADME-Tox prediction methodology were used to study the antitumor activity of the most active compounds and to identify the structural features required for antitumor activity.

Introduction

Cancer is continuing to be a major health problem worldwide to treat and is the leading cause of human deathCitation1–4. The development of novel anticancer agents remains an important and a challenge goal in medicinal chemistry and therefore, developing new effective anticancer drugs is an important strategy in cancer treatmentCitation3,Citation4. The antibacterial fluoroquinolones have been found to be one of the fastest growing groups of drugs in recent yearsCitation5–8. Quinolones are known for their antibacterial and antitumor activities through alteration of the normal functions of bacterial gyrase, and are found to be a topoisomerase II inhibitor in humansCitation9–18. Ciprofloxacin (), a commonly used broad-spectrum fluoroquinolone antibiotic, has shown anticancer activity in several cancer cell linesCitation19,Citation20. Other fluoroquinolone derivatives such as levofloxacin and ofloxacin have also been shown to inhibit the growth of cell bladder cancer cell lines ()Citation21. Voreloxin is a quinolone derivative that shows potent cytotoxicity towards eukaryotic cancer cell lines without antibacterial activity ()Citation22,Citation23. Voreloxin inhibits topoisomerase II and intercalates DNA and it is currently being evaluated in a Phase 2 clinical trial for ovarian cancerCitation22,Citation23.

Figure 1. Reported antitumor quinolones and designed N-piperazinyl fluoroquinolones 114ab.

Figure 1. Reported antitumor quinolones and designed N-piperazinyl fluoroquinolones 1–14ab.

Recently we reported the synthesis and antibacterial activity of bulky arylsulfonylfluoroquinolones based on ciprofloxacin and norfloxacin scaffoldsCitation6. It was found that compounds carrying dimethoxy or dichloro substituents exhibited excellent antibacterial activity compared with ciprofloxacin as a reference drug. From the detailed analysis of the results of our studies, we conclude that antibacterial activity of these compounds significantly depends on the electronic effect of methoxy and chloro groups.

We thus initiated a screening program to search for novel utility of 7-(4-substituted piperazin-1-yl)fluoroquinolones, such as 7-(4-arylsulfonylpiperazin-1-yl)fluoroquinolones, 7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-fluoroquinolones and 7-(4-alkylpiperazin-1-yl)fluoroquinolones, as potential antitumor agents in addition to the reported antibacterial activity.Citation6,Citation24 To the best of our knowledge, there are no reports concerning antitumor activity of bulky arylsulfonylfluoroquinolones were reported.

In this context, the present work describes the investigation of the antitumor properties of 7-(4-arylulfonylpiperazin-1-yl)fluoroquinolones (17ab), 7-(4-alkylpiperazin-1-yl)fluoroquinolones (813ab) and 7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-fluoroquinolones (14ab) based on ciprofloxacin and norfloxacin scaffolds and the achievement of a better antitumor profile at lower concentrations using 60 human cancer cell lines and keeping the inherent antibacterial activity. Moreover, in silico study and ADME-T prediction were used to identify the structural features required for the antitumor properties of the designed compounds. The rationale for testing of these 7-(4-substituted piperazin-1-yl)fluoroquinolones as antitumor agents was the following: (i) delineate the structure-activity relationship (SAR) for the antitumor activity of the arylsulfonylfluoroquinolones with compounds incorporating ciprofloxacin and norfloxacin scaffolds; (ii) investigate and compare the antitumor activity of 4-arylsulfonyl, 4-alkylpiperazinyl and 4-phenylsulfonylcarbamoyl derivatives; (iii) compare the efficacy of the mono and dichloro of arylsulfonylfluoroquinolones versus the methoxy derivatives for the inhibitory power against various tumor cell lines, in compounds incorporating the same scaffold. Furthermore, derivatives incorporating the bulkier trimethylbenzenesulfonyl moieties were also included in the study, in order to explore as much chemical space as possible.

Materials and methods

Chemistry

Melting points (uncorrected) were recorded on Barnstead 9100 Electrothermal melting apparatus at the Pharmaceutical Chemistry Department, King Saud University, Riyadh, Saudi Arabia. IR spectra were recorded on a FT-IR Perkin-Elmer spectrometer at Research Center, King Saud University, Riyadh, Saudi Arabia. Nuclear magnetic resonance (1H and 13C NMR) spectra were recorded on Bruker 700 MHz or 500 MHz spectrometer using CD3OD, CDCl3 and DMSO-d6 as solvents at Research Center, King Saud University, Riyadh, Saudi Arabia. The chemical shifts are expressed in δ ppm using TMS as internal standard. Mass spectra were recorded on a Clarus 600 GC/MS (Middletown, CT) and Varian, TQ 320 GC/MS/MS mass spectrometers (West Sussex, UK) at Research Center, King Saud University, Riyadh, Saudi Arabia. Elemental analysis was carried out for C, H and N, at Research Center, King Saud University, Riyadh, Saudi Arabia. Solvent evaporation was performed under reduced pressure using Buchan Rotatory Evaporator at the Pharmaceutical Chemistry Department, King Saud University, Riyadh, Saudi Arabia. Thin layer chromatography was performed on precoated (0.25 mm) silica gel GF254 plates (E. Merck, Darmstadt, Germany), compounds were detected with 254 nm UV lamp. Silica gel (60–230 mesh) was employed for routine column chromatography separations. Compounds 7-arylsulfonyl-piperazin-1-ylfluoroquinolones (17a,b) were prepared according to the reported procedureCitation6,Citation24.

General method for synthesis of ciprofloxacin and norfloxacin containing 2-(morpholin-4-yl)ethyl and 2-(piperidin-1-yl)ethyl fragments (8, 9ab)

A mixture of norfloxacin (a) or ciprofloxacin (b) (1 mmol) and K2CO3 (1.1 mmol) was stirred in DMF (10 mL) at room temperature for 20 min. To the resulted mixture, the 1-(2-chloroethyl)piperidine or 4-(2-chloroethyl)morpholine (1.1 mmol) in DMF (5 mL) was added dropwise over a period of 10 min. The reaction mixture was further stirred at room temperature for 24 h. The separated solid was then filtered, washed with cold water, dried and crystallized from the appropriate solvent.

1-Ethyl-6-fluoro-4-oxo-7-(4-(2-(morpholin-1-yl)ethyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (8a)

White powder, 52% yield; mp 217–219 °C (MeOH/CH2Cl2); IR (KBr) ν max/cm−1: 3440 (OH), 1719 (C=O), 1641 (C=O). 1HNMR (CDCl3): δ 1.54–1.58 (3H, t, J = 7.5 Hz), 2.54-2.63 (8H, m), 2.75–2.76 (4H, d, J = 2.5 Hz), 3.35–3.36 (4H, d, J = 3.5 Hz), 3.75–3.82 (4H, d, J = 5.0 Hz), 4.31–4.36 (2H, q, J = 7.5 Hz), 6.88–6.89 (1H, d, J = 6.5 Hz), 8.02–8.08 (1H, dd, J = 12.5, 7.5 Hz), 8.67–8.68 (1H, d, J = 6.0 Hz), 15.29 (1H, s, br); 13CNMR (CDCl3): δ 14.44, 49.81, 49.85, 53.23, 54.15, 55.49, 56.34, 66.93, 103.72, 108.32, 112.81, 120.52, 137.11, 146.07, 147.08, 152.52, 154.52, 167.23, 176.97; C22H29FN4O4 m/z: 432.5 (34.6%). Anal. Calcd: C, 61.10; H, 6.76; N, 12.95. Founded: C, 61.31; H, 6.74; N, 12.84.

1-Cyclopropyl-6-fluoro-7-(4-(2-morpholinoethyl)piperazin-1-yl)-4-oxo-1,4-dihydroquinoline-3-carboxylic (8b)

White powder, 66% yield; mp 145–147 °C (MeOH/CH2Cl2); IR(KBr) ν max/cm−1: 3441 (OH), 1733 (C=O), 1628 (C=O). 1HNMR (CDCl3): δ 1.16 (2H, s), 1.35–1.36 (2H, d, J = 6.0 Hz), 2.61 (4H, s), 2.80–2.82 (2H, t, J = 7.5 Hz), 3.24 (2H, s), 3.30 (2H, s), 3.45 (1H, s), 3.63 (2H, s), 3.74 (4H, s), 3.78 (2H, s), 4.46–4.47 (2H, d, J = 6.0 Hz), 7.28 (1H, s), 8.15 (1H, s), 8.56 (1H, s); 13CNMR (CDCl3): δ 8.29, 35.34, 39.69, 45.33, 49.28, 50.82, 63.43, 105.50, 108.27, 112.59, 120.57, 138.96, 145.36, 147.64, 160.80, 166.78, 177.07; C23H29FN4O4 m/z: 444.2 (5.5%). Anal. Calcd: C, 62.15; H, 6.58; N, 12.60. Founded: C, 62.24; H, 6.52; N, 12.40.

1-Ethyl-6-fluoro-4-oxo-7-(4-(2-(piperidin-1-yl)ethyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (9a)

Yellow powder, 69% yield; mp 259–260 °C (MeOH/CH2Cl2); IR(KBr) ν max/cm−1: 3440 (OH), 1730 (C=O), 1636 (C=O). 1HNMR (CDCl3/CD3OD): δ 1.56–1.59 (3H, t, J = 7.0 Hz), 1.71 (2H, s), 1.94 (4H, s), 2.79 (4H, s), 2.88 (2H, s), 3.30–3.33 (6H, d, J = 13.5 Hz), 3.43 (4H, s), 4.51–4.52 (2H, d, J = 7.0 Hz), 7.08–7.09 (1H, d, J = 6.5 Hz), 7.90–7.93 (1H, d, J = 13.5 Hz), 8.80 (1H, s); 13CNMR (CDCl3/CD3OD): δ 15.24, 22.89, 24.12, 50.85, 51.13, 53.51, 53.95, 54.43, 54.93, 106.24, 108.66, 113.00, 121.35, 138.88, 147.43, 149.22, 154.01, 169.72, 178.22; C23H31FN4O3 m/z: 430.6 (28.7%). Anal. Calcd: C, 64.17; H, 7.26; N, 13.01. Founded: C, 64.28; H, 6.17; N, 13.09.

1-Cyclopropyl-6-fluoro-4-oxo-7-(4-(2-(piperidin-1-yl)ethyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (9b)

Yellow powder, 73% yield; mp 265–266 °C (MeOH/CH2Cl2); IR(KBr) ν max/cm−1: 3443 (OH), 1737 (C=O), 1640 (C=O). 1HNMR (CDCl3/CD3OD): δ 1.23 (2H, s), 1.44–1.45 (2H, d, J = 6.0 Hz), 1.71 (2H, s), 1.92 (4H, s), 2.80 (4H, s), 2.87 (2H, s), 3.28–3.29 (6H, dd, J = 5.5, 7.0 Hz), 3.45 (4H, s), 3.76 (1H, s), 7.54–7.55 (1H, d, J = 7.0 Hz), 7.86–7.89 (1H, d, J = 13.0 Hz), 8.77 (1H, s); 13CNMR (CDCl3/CD3OD): δ 8.85, 22.96, 24.24, 37.01, 50.71, 53.62, 53.95, 54.53, 54.94, 107.00, 108.33, 112.63, 112.83, 140.79, 149.20, 169.61; C24H31FN4O3 m/z: 442.7 (5.6%). Anal. Calcd: C, 65.14; H, 7.06; N, 12.66. Founded: C, 65.35; H, 7.15; N, 12.23.

General method for synthesis of norfloxacin and ciprofloxacin containing phthalimide fragments (10, 11ab)

A mixture of norfloxacin (a) or ciprofloxacin (b) (1.0 mmol) and K2CO3 (1.1 mmol) was stirred in DMF (10 mL) at room temperature for 20 min. To the resulted mixture, the 2-(2-bromoethyl)isoindoline-1,3-dione or 2-(3-bromopropyl)isoindoline-1,3-dione (1.1 mmol) in DMF (5 mL) was added drop-wise over a period of 10 min. The reaction mixture was further stirred at room temperature for 24 h. The separated solid was then filtered, washed with cold water, dried and crystallized from the appropriate solvent.

7-(4-(3-(1,3-Dioxoisoindolin-2-yl)ethyl)piperazin-1-yl)-1-ethyl-5-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (10a)

White powder, 64% yield; mp 250–252 °C (CH2Cl2); IR(KBr) ν max/cm−1: 3441 (OH), 1731 (C=O), 1719 (C=O), 1637 (C=O). 1HNMR (CDCl3): δ 1.24–1.30 (3H, t, J = 13.5 Hz), 2.51 (1H, s), 2.65 (5H, s), 3.26 (4H, s), 3.77 (2H, s), 4.51–4.64 (2H, q, J = 6.5 Hz), 7.15–7.16 (1H, d, J = 6.5 Hz), 7.85--7.92 (5H, m), 8.94 (1H, s), 15.36 (1H, s, br); Citation13CNMR (CDCl3): δ 14.44, 28.16, 34.93, 36.89, 39.28, 53.64, 55.62, 62.72, 103.89, 107.99, 112.54, 123.38, 128.99, 131.29, 132.04, 134.10, 134.30, 147.27, 152.52, 167.17, 168.14, 168.56, 176.96; C26H25FN4O5 m/z: 492.5 (7.7%). Anal. Calcd: C, 63.41; H, 5.12; N, 11.38. Founded: C, 63.29; H, 5.22; N, 11.56.

1-Cyclopropyl-7-(4-(2-(1,3-dioxoisoindolin-2-yl)ethyl)piperazin-1-yl)-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (10b)

White powder, 61% yield; mp 189–192 °C (CH2Cl2). 1HNMR (CDCl3): δ 1.16–1.18 (2H, d, J = 5.5 Hz), 1.34–1.36 (2H, d, J = 6.0 Hz), 2.43 (2H, s), 3.36 (4H, s), 3.63 (6H, m), 3.98 (1H, s), 6.66 (1H, s), 7.48–7.50 (4H, d, J = 6.0 Hz), 8.15–8.16 (1H, d, J = 12.5 Hz), 8.41 (1H, s), 15.12 (1H, s, br); 13CNMR (CDCl3): δ 8.16, 35.03, 37.79, 45.08, 49.12, 55.51, 57.01, 63.32, 103.88, 109.07, 112.78, 123.01, 133.38, 134.40, 135.53, 149.65, 151.50, 165.19, 167.00, 168.82, 174.77; C27H25FN4O5 m/z: 504.5 (15.6%). Anal. Calcd: C, 64.28; H, 4.99; N, 11.11. Founded: C, 64.38; H, 5.10; N, 10.99.

7-(4-(3-(1,3-Dioxoisoindolin-2-yl)propyl)piperazin-1-yl)-1-ethyl-5-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (11a)

White powder, 63% yield; mp 205-206 oC (Hexane/CH2Cl2). 1HNMR (CDCl3): δ 1.59–1.61 (3H, t, J = 7.0 Hz), 1.92–1.94 (2H, t, J = 6.5 Hz), 2.54 (2H, s), 2.65 (4H, s), 3.23 (4H, s), 3.82--3.84 (2H, t, J = 7.0 Hz), 4.33-4.34 (2H, d, J = 7.0 Hz), 6.77–6.78 (1H, d, J = 6.5 Hz), 7.72–7.74 (2H, dd, J = 5.0, 8.0 Hz), 7.86–7.88 (2H, dd, J = 5.5, 8.5 Hz), 8.01-8.04 (1H, d, J = 13.0 Hz), 8.68 (1H, s), 15.14 (1H, s, br); 13CNMR (CDCl3): δ14.48, 25.20, 36.42, 49.76, 52.65, 52.68, 103.63, 108.33, 112.62, 120.49, 123.19, 132.27, 133.96, 137.09, 146.06, 147.09, 152.50, 154.50, 167.27, 168.52, 176.99; C27H27FN4O5 m/z: 506.5 (6.4%). Anal. Calcd: C, 64.02; H, 5.37; N, 11.06. Founded: C, 64.11; H, 5.35; N, 11.09.

1-Cyclopropyl-7-(4-(3-(1,3-dioxoisoindolin-2-yl)propyl)piperazin-1-yl)-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (11b)

White powder, 59% yield; mp 210–212 °C (Hexane/CH2Cl2); IR(KBr) ν max/cm−1: 3437 (OH), 1726 (C=O), 1711 (C=O), 1627 (C=O).1HNMR (CDCl3): δ 1.31–1.33 (4H, t, J = 5.5 Hz), 1.84–1.93 (2H, m), 2.14–2.16 (2H, t, J = 6.0 Hz), 3.60–3.62 (2H, t, J = 6.0 Hz), 3.78-4.01 (7H, m), 4.29–4.31 (2H, t, J = 6.0 Hz), 7.16–7.17 (1H, d, J = 6.5 Hz), 7.77–7.92 (5H, m), 8.68 (1H, s), 15.10 (1H, s, br); 13CNMR (CDCl3): δ 8.13, 27.70, 31.33, 34.56, 36.49, 49.74, 52.79, 55.82, 59.06, 61.59, 104.71, 109.83, 112.95, 123.15, 132.09, 133.93, 137.94, 144.44, 148.32, 152.27, 164.98, 166.31, 168.48, 172.99; C28H27FN4O5 m/z: 518.8 (9.7%). Anal. Calcd: C, 64.86; H, 5.25; N, 10.80. Founded: C, 64.76; H, 5.20; N, 10.89.

General method for synthesis of ciprofloxacin and norfloxacin containing 4-(piperidin-1-ylmethyl) fragments (12, 13ab)

To a mixture of norfloxacin (a) or ciprofloxacin (b) (1.0 mmol) and the piperidine or 4-benzylpiperidine (1.0 mmol) in methanol (10 mL), 1 mL of formaline (37%) was added. The reaction mixture was heated overnight at a reflux temperature. The reaction mixture was cooled, the solvent was removed under vacuum, the solid obtained washed with water, dried and re-crystallised from an appropriate solvent.

1-Ethyl-6-fluoro-4-oxo-7-(4-(piperidin-1-ylmethyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (12a)

Yellow powder, 74% yield; mp >300 oC (MeOH); IR(KBr) ν max/cm−1: 3438 (OH), 1728 (C=O), 1629 (C=O). 1HNMR (CDCl3): δ 1.14-1.15 (9H, d, J = 7.0 Hz), 1.52-1.53 (3H, d, J = 7.0 Hz), 2.72 (1H, s), 2.84 (3H, s), 3.28 (4H, s), 3.47–3.49 (1H, d, J = 7.0 Hz), 3.64–3.65 (1H, d, J = 6.5 Hz), 4.07 (1H, s), 4.25–4.27 (2H, t, J = 7.0 Hz), 6.76–6.77 (1H, d, J = 6.5 Hz), 7.94–7.96 (1H, d, J = 13.5 Hz), 8.59 (1H, s), 15.05 (1H, s, br); 13CNMR (CDCl3): δ 14.45, 18.45, 30.94, 49.20, 49.96, 51.07, 58.45, 63.43, 64.40, 88.07, 103.77, 108.31, 112.82, 120.39, 137.136, 146.28, 147.06, 152.53, 154.53, 167.26, 176.98; C22H29FN4O3 m/z: 416.9 (2.5%). Anal. Calcd: C, 63.44; H, 7.02; N, 13.45. Founded: C, 63.79; H, 6.81; N, 13.47.

1-Cyclopropyl-6-fluoro-4-oxo-7-(4-(piperidin-1-ylmethyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (12b)

Yellow powder, 79% yield; mp 256–258 °C (MeOH). 1HNMR (CDCl3): δ 1.14–1.18 (6H, m), 1.31–1.33 (4H, d, J = 6.0 Hz), 2.09 (2H, s), 2.72 (2H, s), 2.84 (4H, s), 3.30 (4H, s), 3.46–3.50 (2H, q, J = 7.0 Hz), 4.07 (1H, s), 7.28–7.29 (1H, d, J = 6.5 Hz), 7.83–7.87 (1H, dd, J = 8.5, 12.5 Hz), 8.62–8.64 (1H, d, J = 6.5 Hz), 14.96 (1H, s, br); 13CNMR (CDCl3): δ 8.21, 15.28, 18.44, 30.93, 35.31, 49.21, 49.88, 51.09, 58.39, 64.39, 88.09, 104.82, 107.96, 112.34, 119.53, 139.08, 145.97, 147.31, 152.66, 154.66, 166.96, 177.01; C23H29FN4O3 m/z: 428.6 (8.3%). Anal. Calcd: C, 64.47; H, 6.82; N, 13.08. Founded: C, 64.57; H, 6.82; N, 13.14.

7-(4-((4-Benzylpiperidin-1-yl)methyl)piperazin-1-yl)-1-ethyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (13a)

White powder, 89% yield; mp 285–287 °C (MeOH); IR(KBr) ν max/cm−1: 3440 (OH), 1736 (C=O), 1641 (C=O). 1HNMR (CDCl3): δ 1.14-1.17 (8H, d, J = 7.0 Hz), 1.51–1.52 (4H, d, J = 7.0 Hz), 2.71 (3H, s), 2.86 (3H, s), 3.26 (4H, s), 3.46–3.48 (1H, d, J = 7.0 Hz), 4.09 (1H, s), 4.24–4.26 (2H, t, J = 7.0 Hz), 6.77–6.79 (1H, d, J = 6.5 Hz), 7.20–7.26 (5H, m), 7.90–7.94 (1H, dd, J = 8.5, 5.0 Hz), 8.56 (1H, s), 15.06 (1H, s, br); 13CNMR (CDCl3): δ 14.44, 29.69, 49.20, 49.75, 49.95, 51.05, 58.42, 64.39, 88.07, 103.77, 108.25, 112.57, 112.75, 120.32, 120.38, 128.14, 129.12, 137.13, 146.19, 146.27, 147.04, 152.52, 154.52, 167.25, 176.93; C29H35FN4O3 m/z: 506.4 (6.1%). Anal. Calcd: C, 68.75; H, 6.96; N, 11.06. Founded: C, 68.91; H, 7.02; N, 11.21.

7-(4-((4-Benzylpiperidin-1-yl)methyl)piperazin-1-yl)-1-cyclopropyl-6-fluoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid (13b)

Yellow powder, 86% yield; mp 260–262 °C (MeOH). 1HNMR (CDCl3): δ 1.03–1.22 (6H, m), 1.31 (2H, s), 1.55–1.57 (2H, d, J = 6.0 Hz), 2.23–2.39 (1H, m), 2.45–2.47 (2H, d, J = 7.0 Hz), 2.65 (1H, s), 2.72 (1H, s), 2.76–2.91 (4H, m), 3.27–3.31 (4H, d, J = 15.0 Hz), 3.48–3.48 (2H, d, J = 3.5 Hz), 4.11–4.22 (1H, m), 7.059 (2H, s), 7.09–7.10 (1H, d, J = 6.0 Hz), 7.18–7.19 (2H, d, J = 6.5 Hz), 7.25–7.27 (1H, d, J = 7.0 Hz), 7.80–7.85 (1H, m), 8.60–8.61 (1H, J = 4.5 Hz); 13CNMR (CDCl3): δ 7.18, 30.70, 31.24, 34.30, 36.67, 37.33, 42.24, 48.97, 50.07, 51.21, 80.09, 103.72, 106.86, 111.18, 118.45, 124.74, 127.12, 128.08, 138.05, 139.61, 145.03, 146.27, 151.63, 153.63, 165.98, 166.05, 175.94; C30H35FN4O3 m/z: 518.6 (5.5%). Anal. Calcd: C, 69.48; H, 6.80; N, 10.80. Founded: C, 69.61; H, 6.84; N, 10.57.

General method for synthesis of ciprofloxacin and norfloxacin containing 4-(phenylsulfonyl)carbamoyl fragments (14ab)

Benzenesulfonyl isocyanate (1.83 g, 1.34 ml, 0.01 mol) was added dropwise to a stirred solution of norfloxacin (a) or ciprofloxacin (b) (0.01 mol) and triethylamine (1.0 mL, 0.01 mol) in dry toluene (50 mL). The reaction mixture was heated under reflux for 3 h, then evaporated in vacuo and the obtained residue was triturated with ice water, filtered, dried and crystallized from methanol to afford N-(phenylsulfonyl)carbamoyl derivatives.

1-Ethyl-6-fluoro-4-oxo-7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (14a)

Yellow powder, 81% yield; mp 255–257 oC (MeOH); IR(KBr) ν max/cm−1: 3446 (OH), 3260 (NH), 1718 (C=O), 1684 (C=O), 1628 (C=O). 1HNMR (CDCl3/TFA): δ 1.36–1.42 (3H, dd, J = 6.5, 8.0 Hz), 3.66 (4H, s), 3.85 (4H, s), 4.73 (2H, s), 7.28 (3H, s), 7.48–7.51 (1H, d, J = 14.5 Hz), 7.93–7.94 (1H, d, J = 6.0 Hz), 8.08–8.22 (2H, t, J = 14.5 Hz), 9.13 (1H, s); 13CNMR (CDCl3/TFA): δ 13.49, 44.03, 46.20, 46.75, 52.29, 63.43, 104.81, 115.67, 116.12, 126.31, 127.45, 129.36, 133.32, 138.52, 141.03, 142.02, 146.56, 159.93, 160.27, 169.46, 170.99; C23H23FN4O6S m/z: 502.4 (5.7%). Anal. Calcd: C, 54.97; H, 4.61; N, 11.15. Founded: C, 55.17; H, 4.60; N, 11.02.

1-Cyclopropyl-6-fluoro-4-oxo-7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid (14b)

Yellow powder, 78% yield; mp 185–187 °C (MeOH); IR(KBr) ν max/cm−1: 3446 (OH), 3264 (NH), 1718 (C=O), 1682 (C=O), 1628 (C=O). 1HNMR (CDCl3/TFA): δ 1.30 (2H, s), 1.51–1.53 (2H, d, J = 6.0 Hz), 3.51 (4H, s), 3.57 (4H, s), 3.79 (1H, s), 7.28 (2H, s), 7.44–7.46 (1H, dd, J = 2.0, 8.5 Hz), 7.52–7.54 (1H, d, J = 6.0 Hz), 7.59–7.60 (1H, d, J = 1.5 Hz), 8.00–8.03 (2H, t, J = 3.0 Hz), 8.97 (1H, s); 13CNMR (CDCl3/TFA): δ 8.29, 36.84, 45.40, 49.57, 105.39, 106.18, 112.18, 115.94, 127.64, 132.98, 133.24, 134.05, 139.83, 140.27, 146.75, 148.38, 153.18, 155.21, 159.06, 169.89, 174.51; C24H23FN4O6S m/z: 515.0 (4.6%). Anal. Calcd: C, 56.02; H, 4.51; N, 10.89. Founded: C, 55.84; H, 4.51; N, 11.00.

Antitumor screening

A primary anticancer assay was performed for an approximately 60 human tumor cell line panel derived from nine neoplastic diseases, in accordance with the protocol of the Drug Evaluation Branch, National Cancer Institute, Bethesda, MDCitation25.

Three dose response parameters were calculated for each compound including GI50 (the drug concentration resulting in a 50% lower net protein increase in the treated cells measured by SRB staining), TGI (the drug concentration resulting in total growth inhibition), and LC50 (concentration of drug resulting in a 50% reduction in the measured protein at the end of the drug treatment compared to that at the beginning). Values were calculated for each of these three parameters if the level of activity was reached; however, if the effect was not reached or was exceeded, the value for that parameter was expressed as more or less for the maximum or minimum concentration tested. The lowest values are obtained with the most sensitive cell lines. The compounds having GI50 ≤100 μM were declared to be activeCitation25.

Results and discussion

Chemistry

Scheme 1 outlines the synthetic pathway used to obtain compounds (114a,b). Compounds 17a,b were prepared according to our previous report by the reaction of norfloxacin (a) or ciprofloxacin (b) with the appropriate arylsulfonyl chloride in acetone in the presence of K2CO3 at room temperature for 24 hCitation6. On the other hand, compounds 811a,b were prepared by the reaction of alkyl halides with norfloxacin (a) or ciprofloxacin (b) in DMF containing anhydrous K2CO3. Mannich bases 12, 13a,b were prepared (74–89% yield) by heating at reflux temperature of norfloxacin (a) or ciprofloxacin (b) and piperidine derivatives as secondary amine with excess formaldehyde in methanol. Moreover, compounds 14a,b were also prepared in a better yield (79–85%) through the reaction of norfloxacin (a) or ciprofloxacin (b) with benzenesulfonyl isocyanate in toluene (Scheme 1). The structural formulae and the purity of the synthesized compounds were checked by thin layer chromatography and spectral analysis. The NMR spectra were identified by their chemical shifts, multiplicities and coupling constants. In general, 1H NMR spectra showed the characteristic chemical shifts for the fluoroquinolone nucleus.

Scheme 1. Synthesis of 7-(4-substituted piperazin-1-yl)-4-oxoquinolines (1–14a,b).

Scheme 1. Synthesis of 7-(4-substituted piperazin-1-yl)-4-oxoquinolines (1–14a,b).

Antitumor activity

The designed compounds 114a,b in addition to the main scaffold ciprofloxacin (b) were tested for their in vitro antitumor activity. Sixteen compounds and ciprofloxacin (b) were selected by National Cancer Institute, Bethesda, MD () on the basis of degree of the structure variation and computer modelling techniques for evaluation of their antitumor activityCitation25. The selected compounds were subjected to in vitro antitumor assay against tumor cells in a full panel of 60-cell lines taken from nine different organs (lung, colon, breast, ovary, blood, kidney, skin, prostate and brain). The compounds were grouped into three series, including 7-(4-arenesulfonylpiperazin-1-yl)fluoroquinolone (17a,b), 7-(4-alkylpiperazin-1-yl)fluoroquinolones (8b, 10b, 11b and 12b) and 7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-fluoroquinolone (14b) (, Figure S1, Supplementary file), which were first evaluated at a single dose concentration of 10 µM, the percentages of growth inhibitions over the 60 tested cell lines were determined and the results were compared with 5-fluorouracil (5-Flu)Citation26, gefitinib (IressaTM)Citation27–29 and erlotinib (TarcevaTM)Citation30 as reference drugs. In the screening methodology, each cell line was inoculated and incubated for 24–47 h. Molecules were then added at a single concentration and the culture was incubated for further 48 h. End point determination were made with a protein binding dye, sulforhodamine (SRB)Citation25. Results for each tested molecule were reported as the percentage growth of the treated cells comparing to the untreated control cells. The screening results and percentages of growth inhibitions over sensitive cell lines are shown in and .

Table 1. Antitumor activity of the designed 7-(4-substituted piperazin-1-yl)fluoroquinolones at 10 µM concentration.

Table 2. Percentage of growth inhibition of the most active fluoroquinolones against individual cell lines.

The screening results of the selected molecules at 10 µM concentration showed that 7-(4-arylsulfonylpiperazin-1-yl)fluoroquinolones (1–7a,b) are more active compared with 7-(4-alkylpiperazin-1-yl)fluoroquinolones (8b, 10b, 11b and 12b) and 7-(4-((phenylsulfonyl)carbamoyl)piperazin-1-yl)-fluoroquinolone (14b) in addition to the parent ciprofloxacin (b) scaffold as indicated by the number of sensitive cell lines and cytotoxic effect (, Supporting Information Figure S1).

The synthesized 7-(4-arylsulfonylpiperazin-1-yl)fluoroquinolones (17a,b) displayed significant activity in the in vitro screening on the tested cell lines in 10 µM concentration with positive cytotoxic effect (PCE) of 6/58 ∼ 58/58. ( and , Figure S1, Supporting Information). These molecules showed a cytotoxic effect on the most of the cancer cell lines especially compounds 1a, 2a, 3b, 6b and 7a with mean growth inhibition percentages (MGI%) of 75.5%, 94.1%, 89.2%, 43.8% and 93.7% respectively. Arylsulfonylpiperazinyl fluoroquinolones (2b, 5b and 6a) showed moderate activity with MGI% of 17.3%, 7.3% and 18.3% respectively. Derivatives based on norfloxacin (a) scaffold were more active compared with the corresponding ciprofloxacin (b) derivatives as indicated by mean growth inhibition percentages (MGI% = 75.5, 1a; 3.5, 1b; 94.1, 2a and 17.3, 2b). Moreover the dimethoxy derivatives of arylsulfonylpiperazinyl fluoroquinolones (MGI% = 2.3, 4a; 1.8, 4b and 7.3, 5b) were less active than the corresponding dichloro derivatives of 7-arenesulfonyl-piperazinyl fluoroquinolones (MGI% = 94.1, 2a; 17.3, 2b and 89.2, 3b) emphasizing the importance of chloro fragment which may attributed to lipophilic character and electronic effect of the chloro derivatives. Replacement of the methoxy group with trimethyl substituent lead to improvement of the antitumor activity such as compounds 6a and 6b with mean growth inhibition percentages (MGI%) of 18.3% and 43.8% respectively. On the other hand, introducing 1-naphthalenesulfonyl moiety in compound 7a led to sharp increase of the antitumor activity (MGI% = 93.7) compared with parent ciprofloxacin (b) and molecules containing trimethylbenzenesulfonyl moiety such as compounds 6a and 6b with mean growth inhibition percentages (MGI%) of −1.4, 18.3 and 43.8% respectively (, Figure S1, Supporting Information).

By investigating the variation in selectivity and broad spectrum of the tested compounds over the full panel of cell lines, it was revealed that nearly all of arylsulfonylpiperazinyl fluoroquinolones (17a,b) ( and ) showed significant inhibition for the most cell lines used in this assay (leukemia, non-small cell lung (NSCLC), colon, CNS, melanoma, ovarian, renal and breast cancer cell lines) with growth inhibition reached to >100% ( and ), while alkylpiperazinyl fluoroquinolones (8b, 10b, 11b and 12b), phenylsulfonylcarbamoylpiperazinyl fluoroquinolone (14b) and ciprofloxacin (b) (, Figure S1, Supporting Information) are mainly active against renal cell lines (A498 and UO-31) with growth inhibition reached to 36%. It is clear that the agreement of the compounds under investigation in the inhibition of renal cell lines could be correlated to a similar inhibitory mechanism related to the common structural feature in ciprofloxacin core, while the selectivity of arylsulfonylpiperazinyl fluoroquinolones (17a,b) ( and , Figure S1, Supporting Information) over other cell lines is probably caused by the differences in the hydrocarbon skeleton around the core structure (ciprofloxacin scaffold).

Moreover, among the 7-arenesulfonyl-piperazin-1-ylfluoroquinolones (17a,b), compounds 1a, 2a, 2b, 3b, 6a, 6b and 7a showed broad spectrum and significant inhibition for cancer cells (58 cell lines; and , Figure S1, Supporting Information) and possessed a considerable cytotoxic activity against cell lines of leukemia (GI% = 60.9 – >100), non-small cell lung (GI% = 53.0 – >100), colon (GI% = 52.3 – >100), CNS (GI% = 53.5 – >100), melanoma (GI% = 51.5 – >100), ovarian (GI% = 57.7 – >100), renal (GI% = 56.3 – >100), prostate (GI% = 68.3 – 98.4) and breast (GI% = 58.8 – >100) cancer cells. This potential inhibition at the mentioned concentration indicates a high potency for the compounds 1a, 2a, 2b, 3b, 6a, 6b and 7a with a strong lethal effect over cancer cells.

Regarding the activity toward individual cell lines (); compounds 1a, 2a, 3b, 6b and 7a showed selective activity against leukemia cell lines such as CCRF-CEM (GI % values of >100, 97.2, 83.0, 62.6 and 87.9, respectively), HL-60 (GI % values of 91.0, >100, >100, 91.2 and >100, respectively), and PRMI-8226 (GI % values of 86.5, 90.9, 95.1, 77.6 and 93.5, respectively). Non-small cell lung; NCI-H23 cell line proved to be selectively sensitive to 1a, 2a, 3b, 6b and 7a with GI% values of >100, >100, 98.2, 42.6 and 99.9, respectively. In addition, compounds 1a, 2a and 7a proved to have equal susceptibility to the HOP-92 cell line with GI% value of >100. Concerning colon cancer; compounds 2a, 3b and 7a showed GI% values of >100 with colon COLO 205 cancer cells. On the other hand, compounds 1a, 2a, 3b, 6b and 7a verified sensitivity with GI% values of 92.1, 98.1, 99.7, 71.7 and 96.0 to colon HT-29 cancer cells. Respecting melanoma; compounds 1a, 2a, 3b, 6b and 7a are active against LOX IMVI cell lines with GI% values of 80.1, >100, 98.7, 70.3 and >100, respectively. Pertaining to renal cancer; compounds 1a, 2a, 3b and 7a were active against A498 cell line with GI% values of >100, >100, 86.2 and >100, respectively. Renal ACHN and UO-31 cell lines are sensitive to compounds 2a and 7a with GI% value of >100. Relating to breast cancer; MCF7, MDA-MB-231/ATCC, T-47D and MDA-MB-468 cell lines possess convinced response to compounds 1a, 2a, 3b, 6b and 7a with GI% range of 46.0 – >100. On the other hand, ovarian IGROV1, OVCAR-3, OVCAR-5 and NCI/ADR-RES cell lines are receptive to compound 1a, 2a, 3b, 6b and 7a with GI% range of 40.5 – >100.

Finally, compounds 1a, 2a, 3b, 6b and 7a were selected in advanced assay against a panel of approximately 60 tumor cell lines at 10-fold dilution of five concentration (100, 10, 1, 0.1, and 0.01 µM) and the results were compared with 5-fluorouracil (5-Flu), gefitinib (IressaTM) and erlotinib (TarcevaTM) as reference drugs ( and ; Figure S2, Supporting Information; http://dtp.nci.nih.gov/dtpstandard/dwindex/index.jsp). Based on the cytotoxicity assays, three antitumor activity dose-response parameters were calculated for tested molecules against each cell lines: GI50, molar concentration of the compound that inhibits 50% net cell growth; TGI, molar concentration of the compound leading to total inhibition; and LC50, molar concentration of the compounds leading to 50% net cell death. Furthermore mean graph midpoints (MG_MID) were calculated for each of the parameters, giving an average activity parameter over all cell lines for the tested molecules.

Table 3. Average antitumor activity of compounds 1a, 2a, 3b, 6b and 7a against tumor cell lines from nine different organs at 10-fold dilution of five concentrations; median growth inhibitory (GI50, µM), total growth inhibitory (TGI, µM) and median lethal (LC50, µM).

Table 4. Influence of compounds 1a, 2a, 3b, 6b and 7a on the growth of the individual tumor cell lines; median growth inhibitory (GI50, µM).

The screening data analysis indicated that compounds 1a, 2a, 3b, 6b and 7a possessed potent in vitro antitumor activity, with GI50 values across the 60 cell lines ranging from 1.22 to 5.37 µM ( and ; Figure S2, Supporting Information). Mean GI50 of compounds 1a, 2a, 3b, 6b and 7a in comparison with 5-fluorouracil (5-Flu), gefitinib (IressaTM) and erlotinib (TarcevaTM) as standard antitumor drugs are given in and Figure S2, Supporting Information.

Compounds 1a, 2a, 3b, 6b and 7a ( and ) exhibited remarkable growth inhibitory activity pattern against leukemia (GI50 = 2.89, 3.07, 3.02, 2.57 and 3.48 µM), non-small cell lung cancer (GI50 = 2.87, 2.49, 3.07, 2.88 and 3.04 µM), colon cancer (GI50 = 3.38, 3.15, 3.79, 3.46 and 3.39 µM), CNS (GI50 = 3.57, 3.03, 3.68, 2.95 and 3.09 µM), breast cancer (GI50 = 2.95, 2.51, 3.04, 2.85 and 2.81 µM), ovarian cancer (GI50 = 3.16, 2.96, 3.39, 2,89 and 3.07 µM), melanoma cancer (GI50 = 2.56, 2.30, 2.54, 22.31 and 3.20 µM), prostate cancer (GI50 = 3.21, 2.62, 3.29, 3.40 and 3.71 µM) and renal cancer (GI50 = 2.93, 2.73, 3.19, 2.78 and 3.50 µM), respectively. Compounds 1a, 2a, 3b, 6b and 7a (mean GI50; 2.63–3.09 µM) are nearly seven-fold more potent compared with the positive control 5-Flu (mean GI50; 22.60 µM). More interestingly, compounds 1a, 2a, 3b, 6b and 7a have an almost antitumor activity similar to gefitinib (mean GI50; 3.24 µM) and are nearly 2-fold more potent compared to erlotinib (mean GI50; 7.29 µM).

The structure correlation study revealed that (1) potent antitumor activity of tested compounds depended on the presence of a combination of quinolone scaffold and arylsulfonyl fragment in one molecule; such as arylsulfonylquinolones (17ab) (7.3–94.1 MGI%) were more active in comparison with alkylated quinolones 8b, 10b and 11b (−4.1 MGI%); (2) attachment of a halogen atom to arylsulfonyl derivative such as compounds 1, 2ab allowed sharp increase of activity (17.3–94.1 MGI%) in comparison with ciprofloxacin scaffold (−1.4 MGI%); (3) introduction of a methoxy group at the arylsulfonyl fragment such as compounds 4a, 4b and 5b led to the decrease of activity; (4) the substituent in the arylsulfonyl fragment proved crucial and manipulated the antitumor activity; accordingly the introduction of an electron-withdrawing moiety, such as chloro fragment (compounds 1a, 2a, 2b and 3b), improved the antitumor activity in comparison with electron-donor moiety, such as methoxy group (compounds 4a, 4b and 5b), and methyl group (compounds 6a and 6b); (5) replacement of the steric bulky trimethylbenzenesulfonyl substituent (compounds 6a and 6b; MGI% = 18–43) with 1-naphthylsulfonyl moiety (compound 7a; MGI% = 93) led to improvement of the antitumor activity; (6) it is clear also that the compounds contain norfloxacin scaffold is more active in comparison with that contain ciprofloxacin scaffold such as compounds 1a and 1b (MGI% = 75.5 and 3.5, respectively).

In silico studies

Lipinski’s rule of five (the effect of lipophilic and steric parameters)Citation31–33

As a part of our study; the compliance of compounds to the Lipinski’s rule of five was evaluatedCitation31. In addition, the polar surface area (PSA) of the compounds was also calculated (), since it is another key property that has been linked to drug bioavailability, where passively absorbed compounds with a PSA > 140 Å2 are thought to have low oral bioavailabilityCitation32. The results disclosed in show that all of the synthesized compounds comply with these rules. Hence; theoretically, all of these compounds should present good passive oral absorption and differences in their bioactivity cannot be attributed to this property.

Table 5. The predicted ADME-Tox and calculated Lipinski’s rule of five of the tested compounds and reported antitumor agents.

The introduction of electron withdrawal/donating groups incorporating the arylsulfonyl fragment and the variation of the substituents on the N-piperazine moiety of the ciprofloxacin and norfloxacin scaffolds have allowed us to evaluate the influence of lipophilicity and steric parameters at the pharmacophoric part of the molecules. gathers physicochemical properties of some selected compounds such as ClogP (lipophilic factors), molar refractometry and polar surface area (steric factors) for each compound, determined by using iLab2 online program (https://ilab.acdlabs.com/iLab2/index.php). Although molar refractometry (MR) does not exert a significant effect on activity in tested compounds, an increase in potency was observed in compounds 1a, 2a, 3b, 6b and 7a with MR values of 120–132 cm3/mol. Regarding lipophilicity (logP); from the data gathered in , there is a clear influence of lipophilicity on antitumor activity compared to polar surface area for all compounds. The optimal lipophilicity for the most active compounds was found to lie in the range of 2.14∼2.87 ().

ADME-Tox evaluationCitation34,Citation35

To estimate the prospect of designed compounds as antitumor agents compared with the reported antitumor agents ciprofloxacin (b) and 5-Flu; their drug-likeness were calculated according to absorption, distribution, metabolism, elimination, toxicity (ADME-T) program, and defined human intestinal absorption (HIA) modelCitation34,Citation35. It was predicted that the examined compounds could be transported across the intestinal epithelium, and they can cross the blood–brain barrier and are of medium aqueous soluble. The values of HIA, BBB crossing and solubility prediction for all compounds are presented in using iLab2 online program (https://ilab.acdlabs.com/iLab2/index.php). In general, all compounds presented some advantages when compared to ciprofloxacin and 5-Flu. No marked differences, in human oral bioavailability (>70%), human intestinal absorption (100%), human jejunum permeability and health effects in rodent toxicity profiles, were observed among the compounds. However, the absorption related parameters call for attention, since the promising compounds were calculated to be at least as soluble as the reported compounds, and are predicted to have oral bioavailability and absorption significantly higher than that of the reported antitumor agents ciprofloxacin (b) and 5-Flu. Accordingly; it can be deduced from these results that the pharmacokinetic profile of the designed compounds is affected and modified by the presence of arylsulfonyl moiety connected to 7-piperazinyl moiety of fluoroquinolone scaffolds.

Toxicities, drug score and drug-likeness profiles

Currently there are many approaches that assess a compound drug-likeness based on topological descriptors, fingerprints of molecular drug-likeness structure keys or other propertiesCitation36,Citation37. In the Osiris program (http://www.organic-chemistry.org/prog/peo) the occurrence frequency of each fragment is determined within the collection created by shredding 3300 traded drugs as well as 15 000 commercially available chemicals (Fluka) yielding a complete list of all available fragmentsCitation36,Citation37. In this work, we used the Osiris program for calculating the fragment based drug-likeness of the active compounds also comparing them with voreloxin, ciprofloxacin, levofloxacin, 5-Flu, erlotinib and gefitinib (). Interestingly, the derivatives 1a, 2a, 2b, 3b, 5b and 7a presented better drug-likeness values (from 2.53 to 4.72) than ciprofloxacin, 5-Flu, erlotinib and gefitinib (2.07, −4.5, −6.73 and −2.62 respectively) and possessed similar results to levofloxacin and voreloxin (5.77 and 4.71 respectively). In this study we also verified the drug-scoreCitation37 as the theoretical data showed that compounds 1a, 2a, 2b, 3b, 5b, 6a, 6b and 7a presented values once again higher than 5-Flu, erlotinib and gefitinib (). Moreover, we used the Osiris program to predict the overall toxicity of the most active derivatives as it may point to the presence of some fragments generally responsible for the irritant, mutagenic, tumorigenic, or reproductive effects in these moleculesCitation36,Citation37. Interestingly, all of the active derivatives presented a low in silico toxicity risk profile, better than 5-Flu and similar to voreloxin, ciprofloxacin, levofloxacin, erlotinib and gefitinib (). These theoretical data reinforced the cytotoxicity experimental data described in this work pointing these compounds as lead compounds for further study.

Figure 2. In silico toxicity risks (upper panel), drug-Likeness (lower left panel) and drug-Score (lower right panel) of the reported and the most active antitumor fluoroquinolone derivatives compared with reference drugs 5-Flu, Erlotinib and Gefitinib (M, mutagenic; T, tumorigenic; I, irritant; R, reproductive).

Figure 2. In silico toxicity risks (upper panel), drug-Likeness (lower left panel) and drug-Score (lower right panel) of the reported and the most active antitumor fluoroquinolone derivatives compared with reference drugs 5-Flu, Erlotinib and Gefitinib (M, mutagenic; T, tumorigenic; I, irritant; R, reproductive).

Conclusion

Sixteen different quinolones based on ciprofloxacin and norfloxacin scaffolds were evaluated for their antitumor activity, where most of the tested compounds exhibited significant antitumor activity. Compounds 1a, 2a, 3b, 6b and 7a were possessed the most potent broad-spectrum antitumor activities (mean GI50; 2.63-3.09 µM) and were nearly seven-fold () more potent compared with the positive control 5-Flu (mean GI50; 22.60 µM). More interestingly, compounds 1a, 2a, 3b, 6b and 7a () have an almost antitumor activity similar to gefitinib (mean GI50; 3.24 µM) and are nearly 2-fold more potent compared to erlotinib (mean GI50; 7.29 µM). The results of this study demonstrated that the electronic effect in the arylsulfonyl fragment proved crucial and manipulated the antitumor activity. The designed 7-substituted 1-piperazinyl fluoroquinolones are compatible with Lipinski’s rule of five (molecular weight, ClogP, hydrogen bond-donating and accepting capabilities). In vitro antitumor activity, together with in silico studies, revealed that compounds 1a, 2a, 3b, 6b and 7a could be considered as promising leads for further development of more potent antitumor agents.

Figure 3. Outcomes of GI50 (µM) of compounds 1a, 2a, 3b, 6b, 7a and reference drugs 5-Flu, Erlotinib and Gefitinib using tumor cell lines from nine different.

Figure 3. Outcomes of GI50 (µM) of compounds 1a, 2a, 3b, 6b, 7a and reference drugs 5-Flu, Erlotinib and Gefitinib using tumor cell lines from nine different.

Supplementary material available online

Supplementary Figures S1 and S2

Acknowledgements

The authors would like to express their gratitude and thanks to the National Cancer Institute (NCI), Bethesda Maryland, USA, http://dtp.cancer.gov/ for doing the antitumor testing of the new compounds.

Declaration of interest

The authors extend their appreciation to the Deanship of Scientific Research at King Saud University for funding the work through the research group project No. RG-1435-046.

References

  • Avendaño C, Menéndez JC. Medicinal chemistry of anticancer drugs. Amsterdam, Netherlands: Elsevier; 2008
  • Harris CC, Hollstein M. Clinical implications of the p53 tumor-suppressor gene. N Engl J Med 1993;329:1318–27
  • Varmus H. The new era in cancer research. Science 2006;312:1162–5
  • Eckhardt S. Recent progress in the development of anticancer agents. Curr Med Chem 2002;2:419–39
  • Hooper DC, Rubinstein E. Quinolone antimicrobial agents. Washington, USA: ASM Press American Society for Microbiology; 2003
  • (a) Abdel-Aziz AA-M, Asiri YA, Al-Agamy MHM. Design, synthesis and antibacterial activity of fluoroquinolones containing bulky arenesulfonyl fragment: 2D-QSAR and docking study. Eur J Med Chem 2011;46:5487–97. (b) Nieto MJ, Alovero FL, Manzo RH, Mazzieri MR. A new class of fluoroquinolones: benzenesulfonamidefluoroquinolones (BSFQs), antibacterial activity and SAR studies. Eur J Med Chem 1999;34:209–14. (c) Nieto MJ, Alovero FL, Manzo RH, Mazzieri MR. Benzenesulfonamide analogs of fluoroquinolones. Antibacterial activity and QSAR studies. Eur J Med Chem 2005;40:361–9. (d) Alovero F, Barnes A, Nieto M, et al. Comparative study of new benzenesulphonamide fluoroquinolones structurally related to ciprofloxacin against selected ciprofloxacin-susceptible and -resistant Gram-positive cocci. J Antimicrob Chemother 2001;48:709–12
  • Chen YL, Fang KC, Sheu JY, Hsu SL, Tzeng CC. Synthesis and antibacterial evaluation of certain quinolone derivatives. J Med Chem 2001;44:2374–7
  • Mirzaei M, Foroumadi A. Synthesis and in-vitro antibacterial activity of N-piperazinyl quinolone derivatives with a 2-thienyl group. Pharm Pharm Commun 2000;6:351–4
  • Hooper DC. Mechanisms of action of antimicrobials: focus on fluoroquinolones. Clin Infect Dis 2001;32:S9–15
  • Blondeau JM. Fluoroquinolones: mechanism of action, classification, and development of resistance. Surv Ophtalmol 2004;49:S73–8
  • Anderson VE, Osheroff N. Type II topoisomerases as targets for quinolone antibacterials: turning Dr. Jekyll into Mr. Hyde. Curr Pharm Des 2001;7:337–53
  • Alovero F, Pan XS, Morris J, et al. Engineering the specificity of antibacterial fluoroquinolones: benzenesulfonamide modifications at C-7 of ciprofloxacin change its primary target in Streptococcus pneumoniae from topoisomerase IV to gyrase. Antimicrob Agents Chemother 2000;44:320–5
  • Pan XS, Hamlyn P, Talens-Visconti R, et al. Small-colony mutants of Staphylococcus aureus allow selection of gyrase-mediated resistance to dual-target fluoroquinolones. Antimicrob Agents Chemother 2002;46:2498–506
  • Heddle J, Maxwell A. Quinolone-binding pocket of DNA gyrase: role of GyrB. Antimicrob Agents Chemother 2002;46:1805–15
  • Fang KC, Chen YL, Sheu JY, et al. Synthesis, antibacterial, and cytotoxic evaluation of certain 7-substituted norfloxacin derivatives. J Med Chem 2000;43:3809–12
  • Azéma J, Guidetti B, Dewelle J, et al. 7-((4-Substituted)piperazin-1-yl) derivatives of ciprofloxacin: synthesis and in vitro biological evaluation as potential antitumor agents. Bioorg Med Chem Lett 2009;17:5396–407
  • Yamashita Y, Ashizawa T, Morimoto M, et al. Antitumor quinolones with mammalian topoisomerase II mediated DNA cleavage activity. Cancer Res 1992;52:2818–22
  • Paul M, Gafter-Gvili A, Fraser A, Leibovici L. The anti-cancer effects of quinolone antibiotics. Eur J Clin Microbiol Infect Dis 2007;26:825–31
  • Herold C, Ocker M, Ganslmayer M, et al. Ciprofloxacin induces apoptosis and inhibits proliferation of human colorectal carcinoma cells. Br J Cancer 2002;86:443–8
  • El-Rayes BF, Grignon R, Aslam N, et al. Ciprofloxacin inhibits cell growth and synergises the effect of etoposide in hormone resistant prostate cancer cells. Int J Oncol 2002;21:207–11
  • Yamakuchi M, Nakata M, Kawahara K, et al. New quinolones, ofloxacin and levofloxacin, inhibit telomerase activity in transitional cell carcinoma cell lines. Cancer Lett 1997;119:213–19
  • Advani RH, Hurwitz HI, Gordon MS, et al. Voreloxin, a first-in-class anticancer quinolone derivative, in relapsed/refractory solid tumors: a report on two dosing schedules. Clin Cancer Res 2010;16:2167–75
  • Hawtin RE, Stockett DE, Byl JA, et al. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons topoisomerase II. PLoS One 2010;5:e10186
  • Abdel-Aziz AA-M, El-Azab AS, Alanazi AM, et al. 7-substituted piperazin-1-yl fluoroquinolone compounds, their use in the treatment of cancer, pharmaceutical compositions and a method for preparation. EPO patent application pending No.: EP 14181731.2, filing date: August 21, 2014
  • (a) Grever MR, Schepartz SA, Chabner BA. The National Cancer Institute: cancer drug discovery and development program. Semin Oncol 1992;19:622--38. (b) Monks A, Scudiero D, Skehan PJ. Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. Natl Cancer Inst 1991;83:757–66. (c) Boyd MR, Paull KD. Some practical considerations and applications of the national cancer institute in vitro anticancer drug discovery screen. Drug Dev Res 1995;34:91–109
  • Le VM, Wang JJ, Yuan M, et al. An investigation of antitumor efficiency of novel sustained and targeted 5-fluorouracil Nanoparticles. Eur J Med Chem 2015;92:882–9
  • Barlesi F, Tchouhadjian C, Doddoli C, et al. Gefitinib (ZD1839, Iressa) in non-small-cell lung cancer: a review of clinical trials from a daily practice perspective. Fundam Clin Pharmacol 2005;19:385–93
  • Arteaga CL, Johnson DH. Tyrosine kinase inhibitors-ZD1839 (Iressa). Curr Opin Oncol 2001;13:491–8
  • Baker AJ, Gibson KH, Grundy W, et al. Studies leading to the identification of ZD1839 (IRESSA): an orally active, selective epidermal growth factor receptor tyrosine kinase inhibitor targeted to the treatment of cancer. Bioorg Med Chem Lett 2001;11:1911–14
  • Ganjoo KN, Wakelee H. Review of erlotinib in the treatment of advanced non-small cell lung cancer. BiolTargets Therapy 2007;1:335–46
  • Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001;46:3–26
  • Clark DE, Pickett SD. Computational methods for the prediction of ‘drug-likeness’. Drug Discov Today 2000;5:49–58
  • El-Azab AS, Al-Omar MA, Abdel-Aziz AA-M, et al. Design, synthesis and biological evaluation of novel quinazoline derivatives as potential antitumor agents: molecular docking study. Eur J Med Chem 2010;45:4188–98
  • Moda TL, Torres LG, Carrara AE, Andricopulo AD. PK/DB: database for pharmacokinetic properties and predictive in silico ADME models. Bioinformatics 2008;24:2270–1
  • El-Deeb IM, Bayoumi SM, El-Sherbeny MA, Abdel-Aziz AA-M. Synthesis and antitumor evaluation of novel cyclic arylsulfonylureas: ADME-T and pharmacophore prediction. Eur J Med Chem 2010;45:2516–30
  • Tetko IV. Computing chemistry on the web. Drug Discov Today 2005;10:1497–500
  • Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem 1999;1:55–68

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.