Back to Journals » Drug Design, Development and Therapy » Volume 11

Isatin-benzoazine molecular hybrids as potential antiproliferative agents: synthesis and in vitro pharmacological profiling

Authors Abdel-Aziz HA , Eldehna WM , Keeton AB, Piazza GA, Kadi AA, Attwa MW , Abdelhameed AS , Attia MI 

Received 21 April 2017

Accepted for publication 26 June 2017

Published 9 August 2017 Volume 2017:11 Pages 2333—2346

DOI https://doi.org/10.2147/DDDT.S140164

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Manfred Ogris



Hatem A Abdel-Aziz,1 Wagdy M Eldehna,2 Adam B Keeton,3 Gary A Piazza,3 Adnan A Kadi,4 Mohamed W Attwa,4 Ali S Abdelhameed,4 Mohamed I Attia4,5

1Department of Applied Organic Chemistry, National Research Centre, Giza, 2Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt; 3Department of Oncologic Sciences and Pharmacology, Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; 4Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; 5Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt

Abstract: In continuation of our endeavor with respect to the development of potent and effective isatin-based anticancer agents, we adopted the molecular hybridization approach to design and synthesize four different sets of isatin-quinazoline (6a–f and 7a–e)/phthalazine (8a–f)/quinoxaline (9a–f) hybrids. The antiproliferative activity of the target hybrids was assessed towards HT-29 (colon), ZR-75 (breast) and A-549 (lung) human cancer cell lines. Hybrids 8b–d emerged as the most active antiproliferative congener in this study. Compound 8c induced apoptosis via increasing caspase 3/7 activity by about 5-fold in the A-549 human cancer cell line. In addition, it exhibited an increase in the G1 phase and a decrease in the S and G2/M phases in the cell cycle effect assay. Furthermore, it displayed an inhibitory concentration 50% value of 9.5 µM against multidrug-resistant NCI-H69AR lung cancer cell line. The hybrid 8c was also subjected to in vitro metabolic investigations through its incubation with rat liver microsomes and analysis of the resulting metabolites with the aid of liquid chromatography-mass spectrometry.

Keywords: isatins, hybridization approach, antiproliferative, apoptosis

Introduction

In the current medical era, molecular hybridization approach has stood out as a valuable and important structural modification tool useful for the discovery and development of better therapies for diverse human diseases, mostly for cancer.1 The growing endeavors to discover hybrid drugs resulting from the combination of two or more haptophoric moieties of different bioactive substances have brought a new hope for the treatment of multifactorial disorders in recent years. Moreover, hybrid drugs can potentially overcome most of the pharmacokinetic drawbacks encountered by conventional anticancer drugs as well as provide combination therapies in a single multifunctional therapeutic agent at the target molecule conferring a more powerful, selective and safer drug compared to conventional classic treatments.24

Isatin (1H-indole-2,3-dione) is an endogenous compound found in many organisms, which was first isolated in 1988.5 As a privileged scaffold, isatin has emerged as an attractive and promising nucleus in the development of novel anticancer agents.6 Sunitinib (I) (Sutent™, Figure 1), granted the US Food and Drug Administration (FDA) approval in 2006, is an isatin-based orally active multi-targeted tyrosine kinase inhibitor used for the management of imatinib-resistant gastrointestinal stromal tumors and metastatic renal-cell carcinoma.79 By 2014, Nintedanib (II, Vargatef™, Figure 1), an orally available triple angiokinase inhibitor, was approved in the US for the treatment of idiopathic pulmonary fibrosis. One year later, the European Medicines Agency approved Nintedanib (II) as a second-line treatment in combination with docetaxel for non-small cell lung cancer of adenocarcinoma.10,11 Also, semaxanib and orantinib are other examples for isatin-based anticancer agents that are being used in clinical trials and possess multiple tyrosine kinase receptor inhibitory activities.12

Figure 1 Chemical structures of clinically used anticancer agents.

Over the last decade, several studies suggested the significance of developing isatin-based hybrids as promising anticancer agents;13 among them, isatin-chromene VII,14 isatin-pyridine VIII,15 bis-isatin IX,16,17 isatin-benzoxazole X,18 isatin-benzimidazole XI,19 isatin-benzothiazole,20 isatin-thiazolidine/thiazolidinone,2125 isatin-4-piperazinylquinoline26 and isatin-pyrazoline27 hybrids were reported (Figure 2).

Figure 2 Structures of some reported isatin-based hybrids with promising anticancer activity and structures of the target hybrids 6a–f, 7a–e, 8a–f and 9a–f.

On the other hand, quinazolines constitute the cornerstone for a number of tyrosine kinase inhibitors such as the reversible EGFR-inhibitor. Erlotinib (III, Tarceva™, Figure 1), as well as the dual VEGFR-2-EGFR inhibitor Vandetanib (IV, Caprelsa™, Figure 1), is indicated for the treatment of symptomatic or progressive medullary thyroid cancer in patients with unresectable locally advanced or metastatic disease.28,29 Also, phthalazine is another attractive scaffold forming the backbone of certain promising antitumor lead candidates. Among them, Vatalanib (V, PTK787, Figure 1) is an orally active VEGFR-1 and VEGFR-2 inhibitor undergoing phase III clinical trials for the treatment of colorectal cancer.30,31 Olaparib (VI, Lynparza, Figure 1), the first approved phthalazine-based anticancer drug, is an oral small molecule poly (ADP-ribose) polymerase inhibitor that was approved in 2014 for the treatment of BRCA-mutated ovarian cancer.32 Recently, much attention has been paid to anticancer drug discovery based on the quinoxaline nucleus as an important heterocyclic one that exhibited interesting biological activities.33 It has been documented that several isatin-quinazoline and isatin-phthalazine hybrids displayed promising anticancer activities.34,35

In the light of the aforementioned findings and in continuation of our endeavor with respect to the development of potent and effective isatin-based anticancer agents,35,36 we adopted the molecular hybridization approach to design and synthesize four different sets of isatin-quinazolines (6a–f and 7a–e)/phthalazines (8a–f)/quinoxaline (9a–f) hybrids (Figure 2). All the synthesized hybrids (6a–f, 7a–e, 8a–f and 9a–f) were in vitro evaluated for their antiproliferative activity against three human cancer cell lines, namely human colon cancer HT-29 cell line, breast cancer ZR-75 cell line and lung cancer A-549 cell line. Moreover, the most active congeners were further assessed for their apoptosis induction potential using human cancer A-549 cell line, via evaluation of their effects on the expression of caspase 3/7 as well as on the cell cycle progression, to obtain mechanistic insights into their anticancer activity. Furthermore, their antiproliferative activity against multidrug-resistant lung cancer NCI-H69AR cell line was evaluated. Finally, the most active candidates were subjected to in vitro metabolic investigations through their incubation with rat liver microsomes (RLMs) and analysis of the resulting metabolites with the aid of liquid chromatography-mass spectrometry (LC-MS).

Materials and methods

General

Melting points of the synthesized compounds were measured with a Stuart melting point apparatus (Staffordshire, UK) and were uncorrected. Infrared (IR) spectra were recorded as KBr disks using FT-IR Spectrum BX apparatus (Perkin Elmer, Shelton, CT, USA). Mass spectra were recorded using Agilent Quadrupole 6120 LC-MS with electrospray ionization (ESI) source (Agilent Technologies, Palo Alto, CA, USA). NMR spectra were recorded on a Bruker NMR spectrometer (Bruker Biospin, Billerica, MA, USA). 1H spectra were run at 500 or 700 MHz, and 13C spectra were run at 125 or 175 MHz in deuterated dimethyl sulfoxide (DMSO-d6). Chemical shifts are expressed in δ values (ppm) using the solvent peak as internal standard. All coupling constant (J) values are given in hertz. The abbreviations used are as follows: s, singlet; d, doublet; m, multiplet. Elemental analyses were carried out at Microanalytical Centre, Cairo University, Egypt. High-resolution mass spectrometry (HRMS) measurements were performed on an LTQ-Orbitrap XL coupled to matrix-assisted laser desorption ionization (MALDI). Reaction courses and product mixtures were routinely monitored by thin layer chromatography on silica-gel precoated F254 Merck plates (Merck Millipore, Billerica, MA, USA). Unless otherwise noted, all solvents and reagents were commercially available and used without further purification. Compounds 1,37 2,35 3,38 and 439 were prepared according to the reported method. All cell lines were purchased from the American Type Culture Collection (ATCC) as follows: HT-29: (ATCC® HTB-38™); ZR75: (ATCC® CRL-1500™); A549: (ATCC® CRM-CCL-185™); IEC-6: (ATCC® CRL-1592™); 3T3-Swiss albino (ATCC® CCL-92™); MCF 10A (ATCC® CRL-10317™); H69AR (ATCC® CRL-11351™).

Chemistry

General procedure for preparation of the target hybrids 6a–f, 7a–e, 8a–f and 9a–f

The appropriate indoline-2,3-dione derivative 5a–f (1 mmol) was added to a suspension of each hydrazinyl intermediate 1–4 (1 mmol) in ethanol (10 mL) and a catalytic amount of glacial acetic acid. The reaction mixture was refluxed for 1 h. The precipitate formed was collected by filtration while hot, washed with hot ethanol, dried and re-crystallized from DMF/ethanol to furnish the desired hybrids.

3-(2-(6,7-Dimethoxyquinazolin-4-yl)hydrazono)indolin-2-one (6a) – orange powder (yield 75%), m.p. 297°C–299°C; IR (KBr, ν cm−1): 3,411 (NH) and 1,699 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.96 (s, 3H, OCH3), 4.00 (s, 3H, OCH3), 6.87–6.89 (m, 2H, Ar-H), 7.17 (t, 1H, Ar-H, J =8.9 Hz), 7.21 (s, 1H, Ar-H), 7.74 (s, 1H, Ar-H), 8.06 (s, 1H, Ar-H), 8.25 (d, 1H, Ar-H, J =8.8 Hz), 10.61 (s, 1H, NH), 12.36 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 55.8 (OCH3), 56.58 (OCH3), 104.2, 108.6, 111.1, 111.2, 112.9, 114.4, 114.5, 117.4, 118.9, 139.6, 144.3, 149.6, 155.4, 157.2, 158.5, 166.4 (C=O); MS (ESI) m/z: 350.0 [M+H]+; Anal. calcd. for C18H15N5O3 (349.12): C, 61.89; H, 4.33; N, 20.05; found C, 62.13; H, 4.28; N, 20.11; HRMS (MALDI) calcd. for C18H15N5O3: 350.1253, found: 350.1224 [M+H]+.

3-(2-(6,7-Dimethoxyquinazolin-4-yl)hydrazono)-5-fluoroindolin-2-one (6b) – orange powder (yield 73%), m.p. >300°C; IR (KBr, ν cm−1): 3,420 (NH) and 1,700 (C=O); 1H NMR (DMSO-d6) δ ppm: 4.01 (s, 3H, OCH3), 4.03 (s, 3H, OCH3), 7.00–7.39 (m, 3H, Ar-H), 7.73 (s, 1H, Ar-H), 8.07 (s, 1H, Ar-H), 8.72 (s, 1H, Ar-H), 11.33 (s, 1H, NH), 13.65 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 56.0, 56.5, 104.4, 108.5, 110.4 (3JF-C =9.0 Hz), 111.9, (2JF-C =27.0 Hz), 113.0 (2JF-C =25.5 Hz), 118.4 (2JF-C =8.0 Hz), 120.4, 122.3, 128.0, 131.5, 133.4, 144.4, 149.5, 153.8 (1JF-C =238.0 Hz), 155.5, 166.2; MS (ESI) m/z: 368.0 [M+H]+; Anal. calcd. for C18H14FN5O3 (367.11): C, 58.85; H, 3.84; N, 19.07; found C, 59.09; H, 3.77; N, 19.13; HRMS (MALDI) calcd. for C18H14FN5O3: 368.1159, found: 368.1151 [M+H]+.

5-Chloro-3-(2-(6,7-dimethoxyquinazolin-4-yl)hydrazono)indolin-2-one (6c) – orange powder (yield 80%), m.p. >300°C; IR (KBr, ν cm−1): 3,421 (NH) and 1,706 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.96 (s, 3H, OCH3), 4.00 (s, 3H, OCH3), 6.90 (d, 1H, Ar-H, J =8.3 Hz), 7.21 (s, 1H, Ar-H), 7.36 (d, 1H, Ar-H, J =8.3 Hz), 7.72 (s, 1H, Ar-H), 8.08 (s, 1H, Ar-H), 8.57 (s, 1H, Ar-H), 10.71 (s, 1H, NH), 12.45 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 55.7 (OCH3), 56.6 (OCH3), 104.0, 108.5, 111.78, 112.9, 119.7, 125.7, 127.4, 130.5, 141.9, 142.5, 144.0, 149.5, 152.4, 153.7, 155.4, 166.0 (C=O); MS (ESI) m/z: 384.0 [M+H]+; Anal. calcd. for C18H14ClN5O3 (383.08): C, 56.33; H, 3.68; N, 18.25; found C, 56.21; H, 3.72; N, 18.17; HRMS (MALDI) calcd. for C18H14ClN5O3: 384.0863, found: 384.0853 [M+H]+.

5-Bromo-3-(2-(6,7-dimethoxyquinazolin-4-yl)hydrazono)indolin-2-one (6d) – orange powder (yield 85%), m.p. >300°C; IR (KBr, ν cm−1): 3,420 (NH) and 1,717 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.96 (s, 3H, OCH3), 4.01 (s, 3H, OCH3), 6.86 (d, 1H, Ar-H, J =8.3 Hz), 7.21 (s, 1H, Ar-H), 7.48 (d, 1H, Ar-H, J =8.3 Hz), 7.72 (s, 1H, Ar-H), 8.08 (s, 1H, Ar-H), 8.73 (s, 1H, Ar-H), 10.73 (s, 1H, NH), 12.45 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 55.9 (OCH3), 56.6 (OCH3), 104.0, 108.4, 112.3, 112.9, 113.5, 120.2, 130.3, 133.3, 142.3, 142.9, 143.6, 144.3, 149.6, 155.5, 155.8, 165.9 (C=O); MS (ESI) m/z: 428.0 [M+H]+; Anal. calcd. for C18H14BrN5O3 (427.03): C, 50.48; H, 3.30; N, 16.35; found C, 50.61; H, 3.26; N, 16.28; HRMS (MALDI) calcd. for C18H14BrN5O3: 428.0358, found: 428.0353 [M+H]+.

3-(2-(6,7-Dimethoxyquinazolin-4-yl)hydrazono)-5-methoxyindolin-2-one (6e) – red powder (yield 72%), m.p. >300°C; IR (KBr, ν cm−1): 3,413 (NH) and 1,699 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.81 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 3.95 (s, 3H, OCH3), 6.80 (d, 1H, Ar-H, J =8.4 Hz), 6.90 (d, 1H, Ar-H, J =8.4 Hz), 7.19 (s, 1H, Ar-H), 7.76 (s, 1H, Ar-H), 8.06 (s, 1H, Ar-H), 8.71 (s, 1H, Ar-H), 10.40 (s, 1H, NH), 12.23 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 55.6 (OCH3), 55.7 (OCH3), 56.5 (OCH3), 104.2, 108.6, 111.1, 112.5, 113.0, 118.0, 118.7, 137.2, 143.2, 143.7, 144.1, 149.4, 154.1, 154.9, 155.2, 166.4 (C=O); MS (ESI) m/z: 380.0 [M+H]+; Anal. calcd. for C19H17N5O4 (379.13): C, 60.15; H, 4.52; N, 18.46; found C, 59.91; H, 4.58; N, 18.59; HRMS (MALDI) calcd. for C19H17N5O4: 380.1359, found: 380.1345 [M+H]+.

3-(2-(6,7-Dimethoxyquinazolin-4-yl)hydrazono)-5-methylindolin-2-one (6f) – orange powder (yield 75%), m.p. >300°C; IR (KBr, ν cm−1): 3,421 (NH) and 1,700 (C=O); 1H NMR (DMSO-d6) δ ppm: 2.31 (s, 3H, CH3), 3.95 (s, 3H, OCH3), 4.01 (s, 3H, OCH3), 6.77 (d, 1H, Ar-H, J =7.5 Hz), 7.12 (d, 1H, Ar-H, J =7.0 Hz), 7.18 (s, 1H, Ar-H), 7.77 (s, 1H, Ar-H), 8.02 (s, 1H, Ar-H), 8.69 (s, 1H, Ar-H), 10.48 (s, 1H, NH), 12.32 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 21.3 (CH3), 55.7 (OCH3), 56.5 (OCH3), 104.2, 108.6, 110.1, 113.1, 118.6, 128.9, 130.8, 131.8, 141.2, 143.8, 144.8, 149.4, 153.3, 155.1, 156.3, 166.4 (C=O); MS (ESI) m/z: 364.0 [M+H]+; Anal. calcd. for C19H17N5O3 (363.13): C, 62.80; H, 4.72; N, 19.27; found C, 63.03; H, 4.66; N, 19.15; HRMS (MALDI) calcd. for C19H17N5O3: 364.1409, found: 364.1422 [M+H]+.

3-(2-(2-(3,4-Dimethoxyphenyl)quinazolin-4-yl)hydrazono)indolin-2-one (7a).35

3-(2-(2-(3,4-Dimethoxyphenyl)quinazolin-4-yl)hydrazono)-5-fluoroindolin-2-one (7b).35

5-Chloro-3-(2-(2-(3,4-dimethoxyphenyl)quinazolin-4-yl)hydrazono)indolin-2-one (7c).35

5-Bromo-3-(2-(2-(3,4-dimethoxyphenyl)quinazolin-4-yl)hydrazono)indolin-2-one (7d) – red powder (yield 80%), m.p. 295°C–297°C; IR (KBr, ν cm−1): 3,421 (NH) and 1,718 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.87 (s, 3H, OCH3), 3.92 (s, 3H, OCH3), 6.96 (d, 1H, Ar-H, J =8.0 Hz), 7.15 (d, 1H, Ar-H, J =8.0 Hz), 7.55 (d, 1H, Ar-H, J =8.5 Hz), 7.73 (t, 1H, Ar-H, J =7.0 Hz), 7.85 (s, 1H, Ar-H), 7.94–7.97 (m, 2H, Ar-H), 8.12 (s, 1H, Ar-H), 8.18 (d, 1H, Ar-H, J =8.5 Hz), 8.52–8.53 (m, 1H, Ar-H), 11.49 (s, 1H, NH), 13.81 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 56.2 (OCH3), 56.3 (OCH3), 110.7, 110.9, 112.1, 118.1, 120.6, 122.4, 125.1, 125.3, 127.7, 128.0, 128.4, 132.3, 132.5, 134.9, 143.3, 146.0, 147.4, 149.3, 152.5, 166.2 (C=O); MS (ESI) m/z: 504 [M+H]+; Anal. calcd. for C24H18BrN5O3 (503.06): C, 57.16; H, 3.60; N, 13.89; found C, 57.29; H, 3.63; N, 13.79; HRMS (MALDI) calcd. for C24H18BrN5O3: 504.0671, found: 504.0653 [M+H]+.

3-(2-(2-(3,4-Dimethoxyphenyl)quinazolin-4-yl)hydrazono)-5-methoxyindolin-2-one (7e) – orange powder (yield 79%), m.p. 263°C–265°C; IR (KBr, ν cm−1): 3,412 (NH) and 1,718 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.76 (s, 3H, OCH3), 3.89 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 6.83 (d, 1H, Ar-H, J =8.5 Hz), 6.96 (d, 1H, Ar-H, J =8.5 Hz), 7.14 (s, 1H, Ar-H), 7.19 (d, 1H, Ar-H, J =9.0 Hz), 7.81 (t, 1H, Ar-H, J =7.5 Hz), 7.96 (s, 1H, Ar-H), 8.06–8.09 (m, 4H, Ar-H), 11.31 (s, 1H, NH), 13.08 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 55.8 (OCH3), 56.2 (OCH3), 56.3 (OCH3), 110.0, 110.8, 111.5, 113.3, 114.4, 117.6, 120.9, 123.1, 124.8, 125.8, 127.1, 127.9, 129.1, 131.8, 133.0, 135.2, 142.5, 144.7, 148.2, 149.1, 154.8, 166.3 (C=O); MS (ESI) m/z: 456 [M+H]+; Anal. calcd. for C25H21N5O4 (455.16): C, 65.93; H, 4.65; N, 15.38; found C, 66.17; H, 4.59; N, 15.52; HRMS (MALDI) calcd. for C25H21N5O4: 456.1672, found: 456.1662 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)indolin-2-one (8a) – orange powder (yield 75%), m.p. 259°C–261°C; IR (KBr, ν cm−1): 3,412 (NH) and 1,700 (C=O); 1H NMR (DMSO-d6) δ ppm: 4.36 (s, 2H, CH2), 6.89 (d, 1H, Ar-H, J =7.5 Hz), 7.06 (t, 1H, Ar-H, J =7.5 Hz), 7.19 (t, 1H, Ar-H, J =7.5 Hz), 7.29–7.36 (m, 5H, Ar-H), 7.86–7.98 (m, 3H, Ar-H), 8.45 (d, 1H, Ar-H, J =7.5 Hz), 8.63 (d, 1H, Ar-H, J =7.5 Hz), 10.63 (s, 1H, NH), 12.89 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH2), 111.6, 115.6, 118.3, 120.3, 122.3, 126.2, 127.0, 127.6, 127.8, 128.9, 129.1, 131.4, 132.9, 134.0, 138.6, 143.3, 144.0, 148.4, 156.5, 166.7; MS (ESI) m/z: 380.0 [M+H]+; Anal. calcd. for C23H17N5O (379.14): C, 72.81; H, 4.52; N, 18.46; found C, 73.01; H, 4.48; N, 18.55; HRMS (MALDI) calcd. for C23H17N5O: 380.1511, found: 380.1525 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)-5-fluoroindolin-2-one (8b) – orange powder (yield 79%), m.p. 275°C–277°C; IR (KBr, ν cm−1): 3,411 (NH) and 1,705 (C=O); 1H NMR (DMSO-d6) δ ppm: 4.39 (s, 2H, CH2), 6.88–7.36 (m, 7H, Ar-H), 7.90–8.01 (m, 3H, Ar-H), 8.19 (d, 1H, Ar-H, J =8.5 Hz), 8.61 (d, 1H, Ar-H, J =7.0 Hz), 10.62 (s, 1H, NH), 12.93 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH2), 111.1 (3JF-C =8.8 Hz), 114.0 (2JF-C =24.5 Hz), 117.3 (2JF-C =25.0 Hz), 118.7 (3JF-C =8.8 Hz), 125.9, 126.2, 126.6, 127.0, 127.6, 128.9, 129.1, 133.1, 134.1, 138.6, 139.4, 143.3, 148.9, 152.8, 157.3 (1JF-C =234.5 Hz), 166.5; MS (ESI) m/z: 398.0 [M+H]+; Anal. calcd. for C23H16FN5O (397.13): C, 69.51; H, 4.06; N, 17.62; found C, 69.32; H, 4.13; N, 17.51; HRMS (MALDI) calcd. for C23H16FN5O: 398.1417, found: 398.1405 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)-5-chloroindolin-2-one (8c) – orange powder (yield 80%), m.p. 295°C–297°C; IR (KBr, ν cm−1): 3,410 (NH) and 1,700 (C=O); 1H NMR (DMSO-d6) δ ppm: 4.39 (s, 2H, CH2), 6.91 (d, 1H, Ar-H, J =8.5 Hz), 7.21 (t, 1H, Ar-H, J =7.5 Hz), 7.30–7.36 (m, 5H, Ar-H), 7.90–8.02 (m, 3H, Ar-H), 8.42 (s, 1H, Ar-H), 8.56 (d, 1H, Ar-H, J =7.0 Hz), 10.73 (s, 1H, NH), 12.96 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH2), 111.8, 119.5, 125.7, 126.3, 126.6, 126.7, 127.0, 127.7, 128.9, 129.0, 129.1, 130.5, 133.1, 134.2, 138.6, 141.8, 142.7, 149.0, 152.8, 166.2 (C=O); MS (ESI) m/z: 414.0 [M+H]+; Anal. calcd. for C23H16ClN5O (413.10): C, 66.75; H, 3.90; N, 16.92; found C, 66.97; H, 3.83; N, 17.05; HRMS (MALDI) calcd. for C23H16ClN5O: 414.1122, found: 414.1146 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)-5-bromoindolin-2-one (8d) – orange powder (yield 86%), m.p. 298°C–299°C; IR (KBr, ν cm−1): 3,412 (NH) and 1,716 (C=O); 1H NMR (DMSO-d6) δ ppm: 4.39 (s, 2H, CH2), 6.86 (d, 1H, Ar-H, J =8.5 Hz), 7.21 (t, 1H, Ar-H, J =7.5 Hz), 7.30–7.36 (m, 3H, Ar-H), 7.47 (d, 1H, Ar-H, J =8.5 Hz), 7.90–8.02 (m, 3H, Ar-H), 8.20 (s, 1H, Ar-H), 8.53–8.56 (m, 2H, Ar-H), 10.74 (s, 1H, NH), 12.69 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH2), 112.3, 113.5, 120.0, 126.4, 126.6, 127.0, 127.7, 128.8, 128.9, 129.1, 129.5, 133.0, 133.3, 134.2, 138.5, 142.1, 142.6, 149.0, 152.8, 166.0 (C=O); MS (ESI) m/z: 458.0 [M+H]+; Anal. calcd. for C23H16BrN5O (457.05): C, 60.28; H, 3.52; N, 15.28; found C, 60.46; H, 3.47; N, 15.40; HRMS (MALDI) calcd. for C23H16BrN5O: 458.0617, found: 458.0605 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)-5-methoxyindolin-2-one (8e) – red powder (yield 74%), m.p. >300°C; IR (KBr, ν cm−1): 3,420 (NH) and 1,707 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.79 (s, 3H, OCH3), 4.37 (s, 2H, CH2), 6.80 (d, 1H, Ar-H, J =8.5 Hz), 6.91 (d, 1H, Ar-H, J =8.5 Hz), 7.19 (t, 1H, Ar-H, J =7.5 Hz), 7.29–7.36 (m, 4H, Ar-H), 7.87–7.99 (m, 3H, Ar-H), 8.08 (s, 1H, Ar-H), 8.56 (d, 1H, Ar-H, J =8.0 Hz), 10.44 (s, 1H, NH), 12.88 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH2), 55.8 (OCH3), 110.7, 112.1, 113.5, 116.9, 120.1, 123.7, 126.4, 127.1, 127.6, 128.8, 129.0, 129.2, 131.7, 133.8, 138.5, 141.7, 144.2, 146.2, 149.9, 154.5, 166.5 (C=O); MS (ESI) m/z: 410.0 [M+H]+; Anal. calcd. for C24H19N5O2 (409.15): C, 70.40; H, 4.68; N, 17.10; found C, 70.64; H, 4.63; N, 16.98; HRMS (MALDI) calcd. for C24H19N5O2: 410.1617, found: 410.1644 [M+H]+.

3-(2-(4-Benzylphthalazin-1-yl)hydrazono)-5-methylindolin-2-one (8f) – orange powder (yield 71%), m.p. 253°C–255°C; IR (KBr, ν cm−1): 3,350 (NH) and 1,697 (C=O); 1H NMR (DMSO-d6) δ ppm: 2.34 (s, 3H, CH3), 4.36 (s, 2H, CH2), 6.78 (d, 1H, Ar-H, J =7.5 Hz), 7.12 (d, 1H, Ar-H, J =8.0 Hz), 7.21 (t, 1H, Ar-H, J =7.5 Hz), 7.29–7.36 (m, 4H, Ar-H), 7.88–7.97 (m, 3H, Ar-H), 8.26 (s, 1H, Ar-H), 8.60 (d, 1H, Ar-H, J =7.5 Hz), 10.52 (s, 1H, NH), 12.84 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 21.4 (CH3), 38.1 (CH2), 110.1, 118.5, 125.8, 126.1, 126.8, 127.0, 127.5, 128.3, 128.9, 129.1, 130.8, 131.7, 132.8, 133.8, 138.6, 141.2, 144.6, 148.2, 152.1, 166.6 (C=O); MS (ESI) m/z: 394.0 [M+H]+; Anal. calcd. for C24H19N5O (393.16): C, 73.27; H, 4.87; N, 17.80; found C, 73.39; H, 4.91; N, 17.72; HRMS (MALDI) calcd. for C24H19N5O: 394.1668, found: 394.1646 [M+H]+.

1-Methyl-3-(2-(2-oxoindolin-3-ylidene)hydrazinyl)quinoxalin-2(1H)-one (9a) – orange powder (yield 80%), m.p. >300°C; IR (KBr, ν cm−1): 3,347 (NH) and 1,711 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.71 (s, 3H, CH3), 6.95 (d, 1H, Ar-H, J =7.5 Hz), 7.10 (t, 1H, Ar-H, J =7.5 Hz), 7.33–7.39 (m, 2H, Ar-H), 7.46 (t, 1H, Ar-H, J =7.5 Hz), 7.54 (d, 1H, Ar-H, J =8.0 Hz), 7.64 (d, 1H, Ar-H, J =7.5 Hz), 7.71 (d, 1H, Ar-H, J =7.5 Hz), 10.72 (s, 1H, NH), 11.24 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 29.8 (CH3), 111.5, 115.3, 120.1, 121.0, 122.9, 124.7, 126.6, 131.5, 132.6, 137.8, 140.5, 142.0, 143.2, 150.5, 162.3, 168.3; MS (ESI) m/z: 320.0 [M+H]+; Anal. calcd. for C17H13N5O2 (319.11): C, 63.94; H, 4.10; N, 21.93; found C, 64.17; H, 4.07; N, 21.84; HRMS (MALDI) calcd. for C17H13N5O2: 320.1148, found: 320.1133 [M+H]+.

3-(2-(5-Fluoro-2-oxoindolin-3-ylidene)hydrazinyl)-1-methylquinoxalin-2(1H)-one (9b) – red powder (yield 75%), m.p. >300°C; IR (KBr, ν cm−1): 3,412 (NH) and 1,701 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.71 (s, 3H, CH3), 6.94–6.97 (m, 1H, Ar-H), 7.18 (t, 1H, Ar-H, J =7.0 Hz), 7.37 (t, 1H, Ar-H, J =7.5 Hz), 7.43–7.50 (m, 2H, Ar-H), 7.55 (d, 1H, Ar-H, J =8.0 Hz), 7.71 (d, 1H, Ar-H, J =7.5 Hz), 11.26 (s, 1H, NH), 13.70 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 29.7 (CH3), 111.4 (3JF-C =9.0 Hz), 115.2 (2JF-C =28.8 Hz), 118.5 (2JF-C =28.00 Hz), 119.0 (3JF-C =9.0 Hz), 121.9, 123.9, 125.7, 128.2, 131.5, 138.3, 140.4, 147.1, 150.8, 156.8 (1JF-C =239.5 Hz), 163.5, 165.9; MS (ESI) m/z: 338.0 [M+H]+; Anal. calcd. for C17H12FN5O2 (337.10): C, 60.53; H, 3.59; N, 20.76; found C, 60.74; H, 3.62; N, 20.67; HRMS (MALDI) calcd. for C17H12FN5O2: 338.1053, found: 338.1041 [M+H]+.

3-(2-(5-Chloro-2-oxoindolin-3-ylidene)hydrazinyl)-1-methylquinoxalin-2(1H)-one (9c) – orange powder (yield 81%), m.p. >300°C; IR (KBr, ν cm−1): 3,411 (NH) and 1,698 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.71 (s, 3H, CH3), 6.97 (d, 1H, Ar-H, J =8.5 Hz), 7.38–7.41 (m, 2H, Ar-H), 7.50 (t, 1H, Ar-H, J =7.5 Hz), 7.56 (d, 1H, Ar-H, J =8.5 Hz), 7.62 (s, 1H, Ar-H), 7.72 (d, 1H, Ar-H, J =8.0 Hz), 11.37 (s, 1H, NH), 13.65 (s, 1H, NH), 13C NMR (DMSO-d6) δ ppm: 29.8 (CH3), 112.1, 115.4, 117.5, 120.3, 122.8, 124.7, 126.1, 128.3, 130.7, 132.1, 134.7, 140.9, 142.7, 150.9, 163.3, 165.5; MS (ESI) m/z: 354.0 [M+H]+; Anal. calcd. for C17H12ClN5O2 (353.07): C, 57.72; H, 3.42; N, 19.80; found C, 57.88; H, 3.38; N, 19.91; HRMS (MALDI) calcd. for C17H12ClN5O2: 354.0758, found: 354.0757 [M+H]+.

3-(2-(5-Bromo-2-oxoindolin-3-ylidene)hydrazinyl)-1-methylquinoxalin-2(1H)-one (9d) – orange powder (yield 84%), m.p. >300°C; IR (KBr, ν cm−1): 3,411 (NH) and 1,708 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.72 (s, 3H, CH3), 6.93 (d, 1H, Ar-H, J =8.5 Hz), 7.38 (t, 1H, Ar-H, J =7.5 Hz), 7.48–7.58 (m, 3H, Ar-H), 7.73–7.74 (m, 2H, Ar-H), 11.44 (s, 1H, NH), 13.64 (s, 1H, NH), 13C NMR (DMSO-d6) δ ppm: 29.8 (CH3), 112.6, 115.4, 117.1, 119.7, 122.8, 124.0, 127.6, 128.3, 131.2, 133.4, 141.2, 143.1, 147.8, 150.8, 163.1, 165.4; MS (ESI) m/z: 398.0 [M+H]+; Anal. calcd. for C17H12BrN5O2 (397.02): C, 51.27; H, 3.04; N, 17.59; found C, 51.09; H, 3.07; N, 17.70; HRMS (MALDI) calcd. for C17H12BrN5O2: 398.0253, found: 398.0243 [M+H]+.

3-(2-(5-Methoxy-2-oxoindolin-3-ylidene)hydrazinyl)-1-methylquinoxalin-2(1H)-one (9e) – red powder (yield 78%), m.p. >300°C; IR (KBr, ν cm−1): 3,413 (NH) and 1,707 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.69 (s, 3H, CH3), 3.79 (s, 3H, OCH3), 6.80–6.94 (m, 2H, Ar-H), 7.14–7.51 (m, 4H, Ar-H), 7.69 (d, 1H, Ar-H, J =8.0 Hz), 10.53 (s, 1H, NH), 13.70 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 38.1 (CH3), 55.8 (OCH3), 110.8, 113.6, 116.5, 118.9, 125.5, 126.8, 127.6, 128.9, 129.1, 132.9, 133.9, 137.0, 138.6, 148.4, 154.9, 166.5; MS (ESI) m/z: 350.0 [M+H]+; Anal. calcd. for C18H15N5O3 (349.12): C, 61.89; H, 4.33; N, 20.05; found C, 62.11; H, 4.29; N, 19.94; HRMS (MALDI) calcd. for C18H15N5O3: 350.1253, found: 350.1239 [M+H]+.

1-Methyl-3-(2-(5-methyl-2-oxoindolin-3-ylidene)hydrazinyl)quinoxalin-2(1H)-one (9f) – orange powder (yield 77%), m.p. >300°C; IR (KBr, ν cm−1): 3,411 (NH) and 1,700 (C=O); 1H NMR (DMSO-d6) δ ppm: 3.70 (s, 3H, CH3), 2.32 (s, 3H, CH3), 6.82 (d, 1H, Ar-H, J =7.5 Hz), 7.14–7.16 (m, 2H, Ar-H), 7.36 (d, 1H, Ar-H, J =7.0 Hz), 7.44 (s, 1H, Ar-H), 7.52 (d, 1H, Ar-H, J =7.5 Hz), 7.68 (d, 1H, Ar-H, J =7.0 Hz), 11.11 (s, 1H, NH), 13.63 (s, 1H, NH); 13C NMR (DMSO-d6) δ ppm: 21.0 (CH3), 29.8 (CH3), 111.3, 115.4, 118.1, 121.4, 124.6, 127.2, 131.8, 132.5, 134.6, 137.4, 140.7, 146.3, 152.4, 157.8, 162.5, 164.1; MS (ESI) m/z: 334.0 [M+H]+; Anal. calcd. for C18H15N5O2 (333.12): C, 64.86; H, 4.54; N, 21.01; found C, 64.69; H, 4.61; N, 21.12; HRMS (MALDI) calcd. for C18H15N5O2: 334.1304, found: 350. 334.1322 [M+H]+.

Pharmacological evaluation

The details of the experimental protocols are provided in Supplementary materials.

Metabolic investigations

The study protocol was approved by the Research Ethics Committee at College of Pharmacy, King Saud University. Animals were maintained according to the guidelines of Animal Care Center, College of Pharmacy, King Saud University, and approved by the Local Animal Care and Use Committee of King Saud University. The details of the experimental protocols4042 are provided in Supplementary materials.

Results and discussion

Chemistry

The synthetic pathway employed to prepare the target isatin derivatives is outlined in Scheme 1. The target compounds 6a–f, 7a–e, 8a–f and 9a–f were obtained by the reaction of the appropriate indoline-2,3-diones 5a–f with the hydrazinyl intermediates 1–4 in refluxed ethanol in the presence of a catalytic amount of glacial acetic acid with 70%–86% yields (Scheme 1).

Scheme 1 Synthesis of targets hydrazino-isatins 6a–f, 7a–e, 8a–f and 9a–f and the chemical structures of the intermediates 1–4. Reagents and conditions: (i) Ethanol/glacial acetic acid (catalytic)/reflux for 1 h.

IR spectra of the target compounds 6a–f, 7a–e, 8a–f and 9a–f showed absorption bands due to the NH groups in the region 3,347–3,421 cm−1, in addition to carbonyl bands in the region 1,697–1,718 cm−1. Their 1H NMR spectra showed two singlet signals attributable to NH protons of the isatin and the hydrazine function (=N–NH–) in the region δ 10.40–11.44 and 11.24–13.81 ppm. Also, the methoxy (–OCH3) protons of compounds 6a–f appeared as singlet signals around δ 4.00 ppm, while the methoxy protons of derivatives 7a–e appeared around δ 3.80 ppm in the 1H NMR spectra. Furthermore, the (–CH2) protons of benzylic moiety of 8a–f appeared as a singlet signal in the range δ 4.36–4.37 ppm, while in case of 9a–f the signals of the aliphatic protons (N–CH3) were observed as singlets near to δ 3.70 ppm.

Pharmacological evaluation

Antiproliferative activity

A total of 23 compounds were analyzed for cancer cell growth inhibitory activity. These studies were carried out using cells derived from human lung, colon and breast tumors (A-549, HT-29 and ZR-75 cells, respectively). This initial assessment of activity tested each compound in quadruplicate at a single concentration of 30 μM, if solubility permitted. As indicated in Table 1, compounds 8b–d are the most potent congeners, inhibiting growth of all three cell lines with average growth inhibition values of 93.8, 96.5 and 96.4%, respectively, at a test concentration of 30 μM. The rest of the compounds showed an average growth inhibition values from 6.1%–81.2% at the tested concentration levels.

Table 1 Antiproliferative (cell growth inhibitory activity at 30 μM concentration) activity of the target compounds 6a–f, 7a–e, 8a–f and 9a–f against HT-29, ZR-75 and A-549 cell lines
Note: *Tested concentration was 10 μM.

The most active promising compounds 8b–d in the preliminary antiproliferative screening were subjected to quantitative inhibitory concentration 50% (IC50) determination for their cell growth inhibitory activity towards A-549, HT-29 and ZR-75 cancer cell lines and the results are presented in Table 2.

Table 2 IC50 of antiproliferative activity of the selected compounds 8b–d and sunitinib against HT-29, ZR-75 and A-549 cell lines
Abbreviation: IC50, inhibitory concentration 50%.

Compound 8c bearing 4-benzylphthalazine moiety exhibited the best average IC50 value of 5.53 μM, as compared with the positive control, sunitinib, which showed an average IC50 =8.11 μM. Therefore, compound 8c was subjected to deeper pharmacological investigations in order to gain insight into its pharmacological profile.

Apoptosis and caspase 3/7 activity

Compound 8c was analyzed for apoptosis-inducing activity in cancer cells. These studies were carried out using cells derived from human lung (A-549). This further assessment of activity tested compounds in quadruplicate at concentrations equivalent to IC50 value to inhibit growth and a concentration 3-fold above the IC50 concentrations over a time course ranging from 2–48 h. As indicated in Figure 3, compound 8c at 5 μM increased caspase activity by 3-fold after 16 h of treatment and to over 5-fold after 24 h of treatment at a concentration of 15 μM.

Figure 3 Caspase 3/7 activity of compound 8c.
Abbreviation: IC50, inhibitory concentration 50%.

Cell cycle effects

Compound 8c was analyzed for effects on various aspects of the cell cycle progression in human cancer cells. These studies were carried out using cells derived from lung adenocarcinoma (A-549). This follow-up assessment of activity tested compounds using immunofluorescent imaging of phosphorylated Rb protein and total DNA content of each cell to assess phase of cell cycle. The ability of test compounds to affect cell cycle distribution and Rb phosphorylation was tested over a range of concentrations less than 100 nM to 50 μM. As shown in Figure 4, compound 8c produced dose-dependent effects on the tested parameters; however, compound 7d displayed no effects on the tested parameters (not shown). Compound 8c caused a significant reduction in the total cell number after 24 h of treatment with IC50 value =10.19 μM and with IC50 value =5.11 μM after 48 h (Table 3).

Figure 4 Cell cycle effects of compound 8c after 24 and 48 h of incubation.

Table 3 IC50 for reductions in the total cell number and cell cycle effects of compound 8c and sunitinib
Abbreviation: IC50, inhibitory concentration 50%.

In addition, compound 8c caused an increase in the percentage of cells in the G1 phase of the cell cycle with corresponding decrease in S and G2/M phases. This suggests that part of the compound effects on growth may be attributable to the decreased rate of progression through the cell cycle and corresponding decrease in proliferation. By contrast, sunitinib caused a reduction in the percentage of cells in G1, with corresponding increases in S or G2/M phases. Arrest in G2 may represent a checkpoint blockade, whereas mitotic arrest may, in some cases, lead to mitotic catastrophe and subsequent programmed death of cells with multiple or aberrant nuclei.

As with other cell cycle parameters, levels of phosphorylated Rb protein were substantially reduced in a dose-dependent manner by the control and the test compound 8c. After 24 h of treatment, the IC50 value was lower than the IC50 value for reductions in the cell number caused by compound 8c (Table 3). This may support the hypothesis that inhibition of cyclin-dependent kinases by isatin compounds plays a role in their growth inhibitory activity. However, the correlation is less apparent at the 48-h time point. Furthermore, compound 8c was analyzed for effects on total cellular levels of phosphorylated tyrosine residues in human cancer cells. These studies were carried out using cells derived from lung adenocarcinoma (A-549) and immunofluorescent imaging. The ability of the test compounds to affect acute serum stimulation of tyrosine phosphorylation was tested over a range of concentrations less than 100 to 50 μM. Compound 8c had no significant effect on P-Tyr labeling.

Selectivity

As an indicator of the selectivity for tumor cells, compound 8c was analyzed for its cell growth inhibitory activity in three non-tumorigenic cell lines. IEC-6 cells derived from rat intestine exhibit morphologic and karyotypic features of normal intestinal epithelial cells.43 Cultures derived from human fibrocystic mammary tissue (MCF-10A) are non-tumorigenic and exhibit features of primary cultures of breast tissue including dome formation.44 Fibroblasts derived from embryonic tissue from mice (Swiss 3t3 fibroblasts) are both non-tumorigenic and contact inhibited.45 For comparison, A-549 human non-small cell lung cancer (NSCLC) cell line was included. This assessment of growth inhibitory activity tested compounds in quadruplicate at maximum concentrations of 25 μM, followed by 10 serially diluted concentrations. As demonstrated in Figure 5 and Table 4, compound 8c inhibited growth in both normal and tumor cell lines by >50%. Compound 8c not only inhibited NSCLC with IC50 value =1.27 μM but also inhibited non-tumor cells less potently with 4.8-fold selectivity value. For the control compound, sunitinib, there was a modest degree of selectivity (1.4-fold difference between mean IC50 in non-tumor cell lines versus the NSCLC cells).

Figure 5 Selectivity profile of sunitinib and compound 8c.

Table 4 Selectivity for compound 8c and sunitinib toward tumor and non-tumorigenic cell lines
Abbreviations: IC50, inhibitory concentration 50%; NSCLC, non-small cell lung cancer.

Multidrug-resistant lung cancer cell line

Compound 8c was analyzed for cancer cell growth inhibitory activity in a sensitive NSCLC cell line (A-549) and a multidrug-resistant lung cancer cell line (NCI-H69AR) that expresses the ABCC1 efflux pump protein. This assessment of activity tested compound 8c in quadruplicate at maximum concentrations of 25 μM, followed by 10 serially diluted concentrations. As illustrated in Figure 6 and summarized in Table 5, compound 8c inhibited growth in both sensitive and resistant cancer cell lines with IC50 values =1.3 and 9.5 μM, respectively, being 7.5-fold less sensitive toward the resistant NCI-H69AR cell line, indicating that this compound may be subjected to efflux by ABCC1. Sunitinib showed a lesser degree of fold resistance being 1.9-fold less sensitive toward the resistant NCI-H69AR cell line.

Figure 6 Activity of sunitinib and compound 8c against sensitive and resistant cancer cell lines.

Table 5 Cancer cell growth inhibitory activity of compound 8c and sunitinib toward sensitive (A-549) and resistant NCI-H69AR cancer cell lines
Abbreviation: IC50, inhibitory concentration 50%.

Metabolic investigations

The study of drug metabolism is a core part of the process of drug discovery and development; it has evolved from being a complementary step to that process to becoming crucial to it.46 Nowadays, metabolic profiles of new drugs have to be investigated prior to any clinical use of such drugs. This approach has been prejudiced by data accumulation assuming that poor pharmacokinetics is the main reason for failure of drug substances, in which the metabolic liability of a drug molecule is the primary determinant.47,48 In this study, comparison of the extracted ion chromatograms between incubations with or without RLMs as well as comparison of the product ion mass spectra of the postulated metabolites of 8c allowed the detection of ten metabolites. Such metabolites resulted from the incubation of 7d and 8c with RLMs that involved various metabolic reaction types, namely, demethylation for 7d and isomerization, reduction, hydroxylation and oxidation for 8c (Figure 7 and Scheme 2). Table 6 summarizes the product ions, retention times and metabolic reactions for the in vitro phase I 8c metabolites.

Figure 7 A representative product ion spectrum of compound 8c (retention time =36.40 min).

Scheme 2 Postulated in vitro metabolic pathway of compound 8c.

Table 6 In vitro RLMs metabolites of compound 8c
Abbreviations: RLM, rat liver microsome; Arom, aromatic.

Conclusion

Quinazoline-isatin hybrids 6a–f and 7a–e, phthalazine-isatin hybrids 8a–f and 1-methylquinoxaline-isatin hybrids 9a–f were synthesized and characterized with different spectroscopic techniques. The preliminary in vitro antiproliferative activity of the synthesized compounds against various human cancer cell lines revealed that compounds 8b–d were the most active candidates. Therefore, they were subjected to quantitative IC50 determination. Detailed pharmacological investigations were carried out on the most promising compound 8c in order to gain insight into its pharmacological profile. Compound 8c induced apoptosis through increasing caspase 3/7 activity by about 5-fold at 15 μM concentration using human cancer A-549 cell line. In addition, it displayed an increase in the G1 phase and a decrease in the S and G2/M phases in the cell cycle effects assay and it showed IC50 value of 9.5 μM against resistant NCI-H69AR cancer cell lines. In vitro metabolic profiling of compound 8c predicted its possible metabolites. Overall, the current study demonstrated that the new chemical entity 8c might be harnessed for cancer therapy after integration of the required preclinical studies.

Supporting materials

The details of the experimental methods that were adopted for the pharmacological investigations of the prepared compounds, the protocols that were used for metabolic studies and representative NMR (1H and 13C) spectra of the target compounds are provided as Supplementary materials.

Acknowledgment

This project was funded by the National Plan for Science, Technology and Innovation (MAARIFAH), King Abdulaziz City for Science and Technology, Kingdom of Saudi Arabia, Award Number 11-MED1924-02.

Disclosure

The authors report no conflicts of interest in this work.


References

1.

Bérubé G. An overview of molecular hybrids in drug discovery. Expert Opin Drug Discov. 2016;11(3):281–305.

2.

Fortin S, Bérubé G. Advances in the development of hybrid anticancer drugs. Expert Opin Drug Discov. 2013;8(8):1029–1047.

3.

Viegas-Junior C, Danuello A, da Silva Bolzani V, Barreiro EJ, Fraga CA. Molecular hybridization: a useful tool in the design of new drug prototypes. Curr Med Chem. 2007;14(17):1829–1852.

4.

Meunier B. Hybrid molecules with a dual mode of action: dream or reality? Acc Chem Res. 2008;41(1):69–77.

5.

Glover V, Halket JM, Watkins PJ, Clow A, Goodwin BL, Sandler M. Isatin: identity with the purified endogenous monoamine oxidase inhibitor tribulin. J Neurochem. 1988;51(2):656–659.

6.

Vine KL, Matesic L, Locke JM, Ranson M, Skropeta D. Cytotoxic and anticancer activities of isatin and its derivatives: a comprehensive review from 2000–2008. Anticancer Agents Med Chem. 2009;9(4):397–414.

7.

Sun L, Liang C, Shirazian S, et al. Discovery of 5-[5-fluoro-2-oxo-1,2-dihydroindol-(3Z)-ylidenemethyl]-2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-diethylaminoethyl)amide, a novel tyrosine kinase inhibitor targeting vascular endothelial and platelet-derived growth factor receptor tyrosine kinase. J Med Chem. 2003;46(7):1116–1119.

8.

Prenen H, Cools J, Mentens N, et al. Efficacy of the kinase inhibitor SU11248 against gastrointestinal stromal tumor mutants refractory to imatinib mesylate. Clin Cancer Res. 2006;12(8):2622–2627.

9.

Motzer RJ, Michaelson MD, Redman BG, et al. Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol. 2006;24(1):16–24.

10.

McCormack PL. Nintedanib: first global approval. Drugs. 2015;75(1):129–139.

11.

Roth GJ, Heckel A, Colbatzky F, et al. Design, synthesis, and evaluation of indolinones as triple angiokinase inhibitors and the discovery of a highly specific 6-methoxycarbonyl-substituted indolinone (BIBF 1120). J Med Chem. 2009;52(14):4466–4480.

12.

Krug M, Hilgeroth A. Recent advances in the development of multi-kinase inhibitors. Mini Rev Med Chem. 2008;8(13):1312–1327.

13.

Ibrahim HS, Abou-Seri SM, Abdel-Aziz HA. 3-Hydrazinoindolin-2-one derivatives: chemical classification and investigation of their targets as anticancer agents. Eur J Med Chem. 2016;122:366–381.

14.

Abdel-Aziz HA, Elsaman T, Al-Dhfyan A, Attia MI, Al-Rashood KA, Al-Obaid AR. Synthesis and anticancer potential of certain novel 2-oxo-N′-(2-oxoindolin-3-ylidene)-2H-chromene-3-carbohydrazides. Eur J Med Chem. 2013;70:358–363.

15.

Eldehna WM, Altoukhy A, Mahrous H, Abdel-Aziz HA. Design, synthesis and QSAR study of certain isatin-pyridine hybrids as potential anti-proliferative agents. Eur J Med Chem. 2015;90:684–694.

16.

Hassan TA, Kadi AA, Abdel-Aziz HA. Novel N,N′-hydrazino-bis-isatin derivatives with selective activity against multidrug-resistant cancer cells. United States Patent US US20120252860. 2012.

17.

Ibrahim HS, Abou-seri SM, Ismail NS, Elaasser MM, Aly MH, Abdel-Aziz HA. Bis-isatin hydrazones with novel linkers: synthesis and biological evaluation as cytotoxic agents. Eur J Med Chem. 2016;108:415–422.

18.

Gudipati R, Anreddy RN, Manda S. Synthesis, anticancer and antioxidant activities of some novel N-(benzo[d]oxazol-2-yl)-2-(7-or 5-substituted-2-oxoindolin-3-ylidene)-hydrazinecarboxamide derivatives. J Enzyme Inhib Med Chem. 2011;26(6):813–818.

19.

Taher AT, Khalil NA, Ahmed EM. Synthesis of novel isatin-thiazoline and isatin-benzimidazole conjugates as anti-breast cancer agents. Arch Pharm Res. 2011;34(10):1615–1621.

20.

Solomon VR, Hu C, Lee H. Hybrid pharmacophore design and synthesis of isatin–benzothiazole analogs for their anti-breast cancer activity. Bioorg Med Chem. 2009;17(21):7585–7592.

21.

Havrylyuk D, Kovach N, Zimenkovsky B, Vasylenko O, Lesyk R. Synthesis and anticancer activity of isatin-based pyrazolines and thiazolidines conjugates. Arch Pharm (Weinheim). 2011;344(8):514–522.

22.

Ramshid PK, Jagadeeshan S, Krishnan A, Mathew M, Nair SA, Pillai MR. Synthesis and in vitro evaluation of some isatin-thiazolidinone hybrid analogues as anti-proliferative agents. Med Chem. 2010;6(5):306–312.

23.

Kaminskyy D, Khyluk D, Vasylenko O, Zaprutko L, Lesky R. A facile synthesis and anticancer activity evaluation of spiro [thiazolidinone-isatin] conjugates. Sci Pharm. 2011;79(4):763–777.

24.

Havrylyuk D, Zimenkovsky B, Vasylenko O, Gzella A, Lesyk R. Synthesis of new 4-thiazolidinone-, pyrazoline-, and isatin-based conjugates with promising antitumor activity. J Med Chem. 2012;55(20):8630–8641.

25.

Wang S, Zhao Y, Zhang G, Lv Y, Zhang N, Gong P. Design, synthesis and biological evaluation of novel 4-thiazolidinones containing indolin-2-one moiety as potential antitumor agent. Eur J Med Chem. 2011;46(8):3509–3518.

26.

Solomon VR, Hu C, Lee H. Design and synthesis of anti-breast cancer agents from 4-piperazinylquinoline: a hybrid pharmacophore approach. Bioorg Med Chem. 2010;18(4):1563–1572.

27.

Sharma M, Sharma S, Buddhiraja A, Saxena AK, Nepali K, Bedi PMS. Synthesis and cytotoxicity studies of 3, 5-diaryl N-acetyl pyrazoline-isatin hybrids. Med Chem Res. 2014;23(10):4337–4344.

28.

Cohen MH, Johnson JR, Chen YF, Sridhara R, Pazdur R. FDA drug approval summary: erlotinib (Tarceva) tablets. Oncologist. 2005;10(7):461–466.

29.

Thornton K, Kim G, Maher VE, et al. Vandetanib for the treatment of symptomatic or progressive medullary thyroid cancer in patients with unresectable locally advanced or metastatic disease: US Food and Drug Administration drug approval summary. Clin Cancer Res. 2012;18(14):3722–3730.

30.

Bold G, Altmann KH, Frei J, et al. New anilinophthalazines as potent and orally well absorbed inhibitors of the VEGF receptor tyrosine kinases useful as antagonists of tumor-driven angiogenesis. J Med Chem. 2000;43(12):2310–2323.

31.

Dumas J, Dixon JA. VEGF receptor kinase inhibitors: phthalazines, anthranilamides and related structures. Expert Opin Ther Pat. 2005;15(6):647–658.

32.

Deeks ED. Olaparib: first global approval. Drugs. 2015;75(2):231–240.

33.

Pinheiro AC, Mendonça Nogueira TC, de Souza MV. Quinoxaline nucleus: a promising scaffold in anti-cancer drug discovery. Anticancer Agents Med Chem. 2016;16(10):1339–1352.

34.

Wu WY, Cao SL, Mao BB, et al. Synthesis and antiproliferative evaluation of hybrids of indolin-2-one and quinazoline-4-(3H)-one linked via imine bond. Lett Drug Des Discov. 2013;10(1):61–66.

35.

Fares M, Eldehna WM, Abou-Seri SM, Abdel-Aziz HA, Aly MH, Tolba MF. Design, synthesis and in vitro antiproliferative activity of novel isatin-quinazoline hybrids. Arch Pharm (Weinheim). 2015;348(2):144–154.

36.

Eldehna WM, Fares M, Ceruso M, et al. Amido/ureido substituted benzenesulfonamides-isatin conjugates as low nanomolar/subnanomolar inhibitors of the tumor-associated carbonic anhydrase isoform XII. Eur J Med Chem. 2016;110:259–266.

37.

He J, Wang X, Zhao X, Liang Y, He H, Fu L. Synthesis and antitumor activity of novel quinazoline derivatives containing thiosemicarbazide moiety. Eur J Med Chem. 2012;54:925–930.

38.

El-Feky S, Bayoumy BE. Cyclocondensation of 4-benzyl-1-hydrazinophthalazine. J Prakt Chem. 1990;332(6):1041–1048.

39.

Makino K, Sakata G, Morimoto K, Ochiai Y. A facile synthesis of novel tricyclic compounds, tetrazoloquinoxalines and 1, 2, 4-triazoloquinoxalines. Heterocycles. 1985;23(8):2025–2034.

40.

Gill HJ, Tingle MD, Park BK. N-Hydroxylation of dapsone by multiple enzymes of cytochrome P450: implications for inhibition of haemotoxicity. Br J Clin Pharmacol. 1995;40(6):531–538.

41.

Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193(1):265–275.

42.

Billings RE. Sex differences in rats in the metabolism of phenytoin to 5-(3, 4-dihydroxyphenyl)-5-phenylhydantoin. J Pharmacol Exp Ther. 1983;225(3):630–636.

43.

Quaroni A, Wands J, Trelstad RL, Isselbacher KJ. Epithelioid cell cultures from rat small intestine. Characterization by morphologic and immunologic criteria. J Cell Biol. 1979;80(2):248–265.

44.

Soule HD, Maloney TM, Wolman SR, et al. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990;50(18):6075–6086.

45.

Todaro GJ, Green H. Quantitative studies of the growth of mouse embryo cells in culture and their development into established lines. J Cell Biol. 1963;17:299–313.

46.

Kumar GN, Surapaneni S. Role of drug metabolism in drug discovery and development. Med Res Rev. 2001;21(5):397–411.

47.

Caldwell GW. Compound optimization in early- and late-phase drug discovery: acceptable pharmacokinetic properties utilizing combined physicochemical, in vitro and in vivo screens. Curr Opin Drug Discov Devel. 2000;3(1):30–41.

48.

Kennedy T. Managing the drug discovery/development interface. Drug Discov Today. 1997;2(10):436–444.

Creative Commons License © 2017 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.