Research Article | Volume: 12, Issue: 11, November, 2022

Discovery of novel trihybrids based on salicylic acid/isoleucine/N-acylhydrazone: A promising therapeutic opportunity in colorectal cancer

David Preciado Gustavo Moreno Wilson Cardona Andres Felipe Yepes   

Open Access   

Published:  Nov 05, 2022

DOI: 10.7324/JAPS.2022.121102
Abstract

Twelve novel trihybrids bearing salicylic acid/isoleucine/N-acylhydrazone (SA-Ile-NAH) features were designed, synthetized, and evaluated for anti-colorectal cancer (CRC) effects against SW480 human colon adenocarcinoma cells. SA-Ile-NAH hybrids 4a–l were obtained in moderate to excellent yields (54%–94%). Regarding biological results, trihybrids 4a, 4c, 4f, and 4j showed 2- to 8-fold higher cytotoxic activities after 48 hours of exposure than the standard chemotherapeutic 5-fluorouracil (5-FU) (174. 30 ± 19.10), with inhibitory concentration (IC50) values of 20.43 ± 1.88 to 78.15 ± 6.73 μM, respectively. Of those, 2,5-dimethoxy-substituited trihybrid 4j is highlighted for its antiproliferative response with viability below 11% in the low micromolar range after 8 days of treatment, as well as for having no significant toxic effects on nontumorigenic cells (IC50 = 102.40 ± 4.64; IS = 1.31) beyond 48 hours of treatment. Finally, theoretical drug-likeness studies suggest that the promising 4j exhibits optimal biopharmaceutical indices. From a therapeutic perspective, merging key pharmacophoric features of SA-Ile-NAH provides a new medicinal scaffold for developing new chemopreventive agents against CRC.


Keyword:     Colorectal cancer salicylic acid isoleucine N-acylhydrazone cytotoxicity antiproliferative activity


Citation:

Preciado D, Moreno G, Cardona W, Yepes AF. Discovery of novel trihybrids based on salicylic acid/isoleucine/N-acylhydrazone: A promising therapeutic opportunity in colorectal cancer. J Appl Pharm Sci, 2022; 12(11):010–020. https://doi.org/10.7324/JAPS.2022.121102

Copyright: © The Author(s). This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

HTML Full Text

INTRODUCTION

Cancer is the leading cause of death worldwide, accounting for nearly 10 million deaths in 2020, or nearly 1 in 6 deaths. It is estimated that the number of deaths may increase up to 28.4 million in 2040. Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), followed by lung (11.4%), colorectal (10.0%), prostate (7.3%), and stomach (5.6%) cancers (Ferlay et al., 2020; Sung et al., 2021). Packages of effective and resource-sensitive preventative and therapeutic interventions are available for cancer; however, serious side effects, resistance, and long-term sequelae offered by these modern treatments continue to be major problems in cancer therapies. To date, colorectal cancer (CRC) is the third most deadly and fourth most commonly diagnosed cancer in the world. In fact, CRC is the third leading cause of cancer death in both men and women, and the second overall among men and women combined (GLOBOCAN, 2021; Høydahl et al., 2020; Siegel et al., 2021). Because standard treatments for CRC usually involves surgery to remove the cancer tumor, followed by adjuvant chemotherapy or radiation therapy, and because of the high toxicity associated with standard chemotherapy regimens together with drug resistance, high doses, and low tumor-specific selectivity, there is an urgent need to search for novel, more potent, safe, and selective treatment strategies in CRC based on targeted therapies able to interfere with closely related signaling pathways in CRC.

Marketed by Bayer under the trade name Aspirin, acetylsalicylic acid (ASA) used currently for minor pain relief and fever reduction has also demonstrated to have chemopreventive effects in cancer incidence and mortality associated with CRC (Bagheri et al., 2018; Burn et al., 2020; Cuzick et al., 2009; Patrignani and Patrono, 2016; Peleg et al., 1994; Spiegel, 2020; Thun et al., 1991). As for ASA, its metabolite salicylic acid (SA), displayed antiproliferative and proapoptotic effects in CRC in in vitro and in vivo models (Brennan et al., 2021; Deb et al., 2011; Elder et al., 1996; Elder and Paraskeva 1999; Giardina and Inan, 1998; Gökçe et al., 2017; Pathi et al., 2012; Paterson and Lawrence, 2001). Similarly, isoleucine has been revealed to exhibit high potency against several cancer cell lines, including lines of human colorectal carcinoma cells (Ananieva and Wilkinson, 2018; Lieu et al., 2020; Murata et al., 2007; Wakshlag et al., 2006). Furthermore, reports of diagnosed CRC patients suggest that isoleucine is linked to survival after diagnosis. This finding not only suggests isoleucine as a possible prognostic biomarker in CRC patients, but that those patients might need more of this amino acid (Bener et al., 2006; Delphan et al., 2018; Long et al., 2020; Wang et al., 1997). In addition, the antitumoral effectiveness of N-acylhydrazone-containing compounds, especially to prevent proliferation in human colorectal cancerous cells, has been extensively reported (Al-awar et al., 2019; Dasgupta et al., 2020; Huff et al., 2018; Iliev et al., 2019; Song et al., 2011; Thota et al., 2018). In this regard, N-acylhydrazones derivatives have drawn special attention for anticancer treatment, due to their ability for inhibiting the growth of a range of tumor cells including lung, colon, pancreas, and breast cancer, at micromolar levels (Buss et al., 2002; Chen et al., 2018; Chaston et al., 2003).

From a therapeutic point of view, due to the complexity of cancer and the abnormal activation of several signaling pathways in its progression, the multitarget-directed ligands strategy is considered to be an effective way to treat this disease. In an extension to this approach, merging of two (or more than two) antitumoral pharmacophoric features from known bioactive compounds covalently linked to a single molecule (hybrid molecules) offers new avenues for the development of improved chemotherapeutic cancer candidates (Ali et al., 2013; Viegas-Junior et al., 2007). Thus, considering the high antitumoral effectiveness found for SA, isoleucine and the N-acylhydrazone have not been combined into a single drug candidate for anticancer agent, and so we designed for the first time a series of molecular hybrids featuring structurally moieties of SA, N-acylhydrazone, and isoleucine into parent molecules (Fig. 1). To further analyze their structure–activity relationships (SARs), the phenyl ring substitution on N-acylhydrazone framework was used. We hypothesize that these new hybrids would exert a valuable antitumor response, in a synergistic effect, leading to tumor cell apoptosis in human colon carcinoma SW480 cells, which would likely open a new clinical possibility for CRC management. The most promising hybrid was also chosen to be investigated for its pharmacokinetics profile using in silico-derived parameters.


MATERIALS AND METHODS

General information

All chemicals used were obtained from commercial suppliers and used without further purification. Thin-layer chromatography (TLC, silica gel 60 F254) was used to follow reactions and was visualized in an iodine chamber or with a UV lamp (λ = 254 nm). Melting points were determined using a Stuart SMP10 Digital Melting Point instrument. Microwave-assisted reactions were conducted in CEM Focused Microwave. Nuclear Magnetic Spectroscopy (NMR) spectra of compounds measured on a Varian instrument spectrometer (300 MHz 1H NMR and 75 MHz 13C NMR), 1H and 13C NMR spectra were recorded in DMSO-d6, CDCl3, or CD3OD solutions and chemical shift values are in parts per million (ppm) using the Me4Si as internal standard.

Preparation of methylisoleucinate (1)

Thionyl chloride (1.5 mmol) was dissolved into dry methanol (10 ml) cooled to −10°C and the resulting mixture was stirred for 5 minutes. Afterward, the solution was treated with isoleucine (1.0 mmol), stirred for 10 minutes, maintained at −10°C for 2 hours, and then kept at room temperature for another 24 hours. After completion of the reaction, the reaction mixture was poured into ether (100 ml) and cooled for 2 hours collecting the interfacial precipitate which was filtered and oven-dried at 45°C for 24 hours; pure product 1 was obtained in a quantitatively yield (95%) as colorless needles: mp 96°C–98°C. 1H NMR (300 MHz, CDCl3) δ 3.90 (dd, J = 10.2, 4.2 Hz, 1H), 3.74 (s, 1H, CH3O), 1.67–1.39 (m, 1H), 1.34–1.03 (m, 1H), 0.94 (d, J = 6.9 Hz, 3H, CH3), 0.92 (t, J = 8.0 Hz, 3H, CH3). 13C NMR (75 MHz, CDCl3) δ 168.9 (C=O), 56.8 (CH-NH), 52.2 (CH3O), 36.4 (CH), 25.3 (CH2), 13.5 (CH3), 10.6 (CH3).

Figure 1. Design strategy of the novel trihybrids bearing salicylic acid (in red), isoleucine (in blue), and N-acylhydrazone (in green) features as anticancer agents.

[Click here to view]

Synthesis of methyl (2-hydroxybenzoyl)isoleucinate (2)

The present synthetic procedure is similar to a previously reported method (Castrillón et al., 2019). Triethylamine (4 mmol) and SA (1 mmol) in Tetrahydrofuran (THF) (10 ml) were stirred for 15 minutes. 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate, Hexafluorophosphate Benzotriazole Tetramethyl Uronium (HBTU) (1.5 mmol) was added to this mixture and stirred for a further 20 minutes. Isoleucine ester 1 (1.2 mmol) was then added and allowed to stir for 24 hours. Upon completion, the reaction mixture solvent was evaporated to dryness under reduced pressure and the residue was purified on a silica gel chromatography using mixtures hexane-ethyl acetate (1:1 ratio) as eluent yielding pure intermediate 2 in a good yield (71%) as a viscous yellow oil. 1H NMR (300 MHz, CDCl3) δ 7.46 (d, J = 8.2 Hz, 1H, 1-H), 7.40 (t, J = 8.2 Hz, 1H, 3-H), 6.98 (d, J = 8.2 Hz, 1H, 4-H), 6.88 (t, J = 8.2 Hz, 1H, 2-H), 4.78 (dd, J = 8.3, 4.9 Hz, 1H), 3.79 (s, 3H, OCH3), 2.13–1.90 (m, 1H), 1.64–1.36 (m, 1H), 1.39–1.05 (m, 1H), 1.07–0.82 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 172.3 (C=O), 169.6 (C=O), 161.6 (Ar-O), 134.6 (Ar), 125.6 (Ar), 118.8 (Ar), 118.7 (Ar), 114.0 (Ar), 55.4 (CH-NH), 52.6 (CH3O), 38.2 (CH), 25.4 (CH2), 14.8 (CH3), 11.8 (CH3).

Synthesis of N-((2S,3S)-1-hydrazineyl-3-methyl-1-oxopentan-2-yl)-2-hydroxybenzamide (3)

The present synthetic procedure is similar to a previously reported method (Coa et al., 2015; Vergara et al., 2017). Compound 2 (1 mmol) and MeOH (1 ml) were mixed. Subsequently, hydrazine hydrate (5 mmol, 98% solution) was added and the resulting mixture was stirred in a preheated oil bath at 65°C for 24 hours when analysis by TLC indicated the end of the reaction. After being cooled to room temperature, the solvent was evaporated under reduced pressure and the residue was poured onto ice. The resulting precipitate was filtered, washed with ice water, and oven-dried at 45°C for 24 hours, affording the corresponding N-acylhydrazide 3 as a white solid (93%); mp 111°C–113°C. 1H NMR (300 MHz, Methanol-d4) δ 7.90 (td, J = 8.0, 1.8 Hz, 1H), 7.38 (td, J = 8.0, 7.4, 1.8 Hz, 1H), 6.92 (m, 2H), 3.33 (dd, J = 8.1, 3.9 Hz, 1H), 2.10–1.92 (m, 1H), 1.70–1.44 (m, 1H), 1.04–0.92 (m, 6H, 2 × CH3). 13C NMR (75 MHz, Methanol-d4) δ 171.7 (C=O), 168.1 (C=O), 158.3 (Ar-O), 133.4 (Ar), 129.0 (Ar), 119.0 (Ar), 116.7 (Ar), 116.5 (Ar), 56.6 (CH-NH), 36.8 (CH), 24.8 (CH2), 14.5 (CH3), 9.9 (CH3).

General procedure for synthesis of trihybrids (4a–l)

The present synthetic procedure is similar to a previously reported method (Coa et al., 2015; Vergara et al., 2017). The appropriate aromatic aldehyde (1 mmol), acylhydrazide 3 (1 mmol), and a few drops of acetic acid in ethanol were deposited into a CEM microwave reaction vessel. After that, the mixture was irradiated in a CEM Discover microwave synthesizer at 200 W in the open-vessel mode attached to a reflux condenser at 80°C with vigorous stir during 10 minutes. After being cooled to room temperature, the solvent was evaporated under reduced pressure and the oily residue was purified by preparative TLC over silica gel 60 F 254 (Merck) using an mixture of hexane:AcOEt (10:8–1:1) as eluent, furnishing the target hybrids 4a–l in the yields as described below.

2-hydroxy-N-(1-(2-((2-hydroxynaphthalen-1-yl)methylene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)benzamide (4a). Pale yellow solid; yield = 94%; mp = 258°C–260°C; 1H NMR (300 MHz, DMSO-d6) δ 9.22 (s, 1H, N=C-H), 7.93–7.72 (m, 2H), 7.67–7.48 (m, 2H), 7.32 (t, J = 8.1 Hz, 1H), 7.14–7.00 (m, 2H), 6.85–6.66 (m, 2H), 6.42 (t, J = 7.8 Hz, 1H), 4.60 (d, J = 6.9 Hz, 1H), 2.06–1.92 (m, 1H), 1.70–1.43 (m, 2H), 1.06–0.71 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 172.7 (C=O), 168.2 (C=O), 159.2 (Ar-O), 157.2 (Ar-O), 140.6 (C=N), 137.4, 133.6, 132.6, 130.4, 129.1, 127.1, 126.8, 125.9, 124.3, 120.0, 118.8, 118.6, 117.4, 133.5, 56.6, 37.4, 25.1, 16.1 (CH3), 11.4 (CH3).

2-hydroxy-N-(1-(2-(2-hydroxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)benzamide (4b). Pale yellow solid; yield = 75%; mp = 280°C–282°C; 1H NMR (300 MHz, DMSO-d6) δ 8.11 (s, 1H, N=C-H), 7.64 (d, J = 7.8 Hz, 1H), 7.29–6.77 (m, 4H), 6.56 (d, J = 8.2 Hz, 2H), 6.30 (t, J = 7.1 Hz, 1H), 4.40 (d, J = 6.8 Hz, 1H), 2.18–1.75 (m, 1H), 1.42–1.08 (m, 2H), 0.98–0.70 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 172.7 (C=O), 168.7 (C=O), 165.8 (Ar-O), 156.7 (Ar-O), 152.5 (C=N), 149.0, 138.2, 137.9, 133.7, 131.9, 118.3, 114.8, 106.1, 104.6, 102.2, 62.8, 37.7, 26.4, 15.9 (CH3), 11.4 (CH3).

N-(1-(2-(2,3-dihydroxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4c). Pale yellow solid; yield = 58%; mp = 197°C–199°C; 1H NMR (300 MHz, DMSO-d6) δ 8.85 (s, 1H, N = C-H), 8.34 (d, J = 7.5 Hz, 1H), 7.87 (t, J = 7.5 Hz, 1H), 7.38 (t, J = 7.5 Hz, 1H), 7.06 (d, J = 8.1 Hz, 1H), 6.85 (d, J = 7.2 Hz, 1H), 6.81 (t, J = 7.2 Hz, 1H), 6.75 (d, J = 8.1 Hz, 1H), 4.48 (d, J = 7.1 Hz, 1H), 2.10–1.78 (m, 1H), 1.68–1.27 (m, 2H), 1.00–0.64 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 168.4 (C=O), 168.2 (C=O), 157.5 (Ar-O), 147.3 (Ar-O), 145.4 (Ar-O), 145.3 (C=N), 134.1, 130.1, 122.7, 121.1, 120.2, 120.0, 119.4, 118.7, 117.5, 56.7, 36.8, 24.9, 15.6 (CH3), 11.2 (CH3).

N-(1-(2-(2,4-dihydroxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4d). Pale yellow solid; yield = 54%; mp = 196°C–198°C; 1H NMR (300 MHz, DMSO-d6) δ 8.00 (s, 1H, N = C-H), 7.67 (dt, J = 7.9, 2.4 Hz, 1H), 7.20 (m, 2H), 7.88 (t, J = 8.2 Hz, 1H, 2-H), 7.01 (d, J = 3.2 Hz, 1H), 6.66 (dd, J = 8.5, 3.2 Hz, 1H), 6.52 (t, J = 7.4 Hz, 1H), 6.09 (d, J = 8.5 Hz, 1H), 4.34 (d, J = 6.8 Hz, 1H), 2.16–1.86 (m, 1H), 1.43–1.14 (m, 2H), 0.93 (d, J = 7.4 Hz, 3H, CH3), 0.83 (t, J = 7.6 Hz, 3H, CH3). 13C NMR (75 MHz, DMSO-d6) δ 171.5 (C=O), 171.0 (C=O), 168.9 (Ar-O), 153.9 (Ar-O), 151.4 (Ar-O), 142.1 (C=N), 135.5, 134.1, 134.1, 129.6, 122.2, 122.1, 117.7, 114.2, 110.5, 57.2, 36.9, 24.7, 15.5 (CH3), 11.2 (CH3).

N-(1-(2-(2,5-dihydroxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4e). Pale yellow solid; yield = 86%; mp = 201°C–203°C; 1H NMR (300 MHz, DMSO-d6) δ 8.24 (s, 1H, N=C-H) 7.73 (td, J = 7.3, 1.7 Hz, 1H), 7.26 (dd, J = 7.3, 2.4 Hz, 1H), 6.89 (d, J = 2.7 Hz, 1H), 6.84 (dd, J = 7.5, 1.7 Hz, 1H), 6.77 (d, J = 8.4 Hz, 1H), 6.73 (dd, J = 8.4, 2.7 Hz, 1H), 6.59 (t, J = 7.5 Hz, 1H), 4.41 (d, J = 6.6 Hz, 0H), 2.24–1.90 (m, 1H), 1.47–1.20 (m, 2H), 1.01 (d, J = 6.9 Hz, 3H), 0.89 (t, J = 7.1 Hz, 3H). 13C NMR (75 MHz, DMSO-d6) δ 172.8 (C=O), 168.7 (C=O), 165.7 (Ar-O), 155.1 (Ar-O), 150.2 (Ar-O), 145.6 (C=N), 133.0, 130.1, 120.5, 119.3, 118.3, 118.0, 117.7, 116.2, 115.9, 55.9, 37.1, 25.0, 15.9 (CH3), 11.9 (CH3).

N-(1-(2-(3,4-dihydroxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4f). Yellow solid; yield = 64%; mp = 188°C–191°C; 1H NMR (300 MHz, DMSO-d6) δ 7.98 (s, 1H, N=C-H), 7.79 (t, J = 7.6 Hz, 1H), 7.36 (t, J = 7.6 Hz, 1H), 7.22 (s, 1H), 6.99 (d, J = 7.7 Hz, 1H), 6.96–6.85 (m, 2H), 6.78 (d, J = 7.7 Hz, 1H), 4.34 (d, J = 7.6 Hz, 1H), 1.93–1.79 (m, 1H), 1.36–1.06 (m, 2H), 0.88–0.78 (m, 6H). 13C NMR (75 MHz, DMSO-d6) δ 169.0 (C=O), 167.6 (C=O), 157.2 (Ar-O), 152.5 (Ar-O), 149.9 (Ar-O), 147.8 (C=N), 145.1, 134.4, 130.0, 125.7, 122.1, 117.3, 117.0, 116.0, 113.4, 57.1, 36.8, 24.9, 14.8 (CH3), 10.7 (CH3).

2-hydroxy-N-(3-methyl-1-oxo-1-(2-(2,3,4-trihydroxybenzylidene)hydrazineyl)pentan-2-yl) benzamide (4g). Yellow solid; yield = 68%; mp = 126°C–128°C; 1H NMR (300 MHz, DMSO-d6) δ 8.22 (s, 1H, N=C-H), 7.85 (d, J = 8.5 Hz, 1H), 7.37 (t, J = 7.7 Hz, 1H), 7.06–6.84 (m, 2H), 6.77 (dd, J = 8.5, 2.4 Hz, 1H), 6.40 (d, J = 8.7 Hz, 1H), 4.38 (d, J = 7.2 Hz, 1H), 1.96–1.71 (m, 1H), 1.43–1.07 (m, 2H), 0.94–0.73 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 168.1 (C=O), 168.0 (C=O), 157.4 (Ar-O), 157.1 (Ar-O), 151.1 (Ar-O), 148.8 (Ar-O), 147.2 (C=N), 132.6, 130.2, 122.4, 120.2, 117.5, 117.1, 111.0, 108.2, 56.9, 36.9, 25.0, 15.6 (CH3), 10.9 (CH3).

N-(1-(2-(2,3-dimethoxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4h). Pale yellow solid; yield = 91%; mp = 118°C–120°C; 1H NMR (300 MHz, DMSO-d6) δ 8.55 (s, 1H, N=C-H), 8.32 (d, J = 7.6 Hz, 1H), 7.92 (t, J = 7.6 Hz, 1H), 7.44 (t, J = 7.6 Hz, 1H), 7.27 (m, 2H), 7.11 (m, 2H), 4.44 (d, J = 7.0 Hz, 1H), 3.79 (s, 3H, OMe), 3.78 (s, 3H, OMe), 2.05–1.91 (m, 1H), 1.65–1.40 (m, 2H), 1.02–0.88 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6). δ 166.9 (C=O), 166.7 (C=O), 161.2 (Ar-O), 153.2 (Ar-OMe), 153.1 (Ar-OMe), 148.3 (C=N), 143.0, 133.2, 130.4, 128.1, 124.8, 118.6, 118.2, 117.4, 114.4, 61.6 (CH3O), 57.0 (CH3O), 56.2, 37.1, 27.0, 16.0 (CH3), 11.3 (CH3).

N-(1-(2-(2,4-dimethoxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4i). Pale yellow solid; yield = 61%; mp = 120°C–122°C; 1H NMR (300 MHz, DMSO-d6) δ 8.52 (s, 1H, N=C-H), 7.91 (d, J = 8.1 Hz, 1H), 7.76 (dd, J = 8.6, 2.2 Hz, 1H), 7.74 (d, J = 8.4 Hz, 1H), 6.97 (t, J = 8.1 Hz, 1H), 6.93 (d, J = 7.7 Hz, 1H), 6.75 (d, J = 2.1 Hz, 1H), 6.65 (dd, J = 8.4, 2.1 Hz, 1H), 3.88 (d, J = 6.8 Hz, 1H), 3.85 (s, 3H, OMe), 3.82 (s, 3H OMe), 2.07–1.91 (m, 1H), 1.65–1.38 (m, 2H), 0.94 (d, J = 6.8 Hz, 3H), 0.89 (t, J = 7.2 Hz, 3H). 13C NMR (75 MHz, DMSO-d6) δ 167.9 (C=O), 167.4 (C=O), 162.9 (Ar-O), 159.5 (Ar-OMe), 159.4(Ar-OMe), 143.1 (C=N), 133.3, 130.4, 127.1, 118.4, 118.3, 115.5, 115.3, 106.8, 98.7, 56.2 (CH3O), 56.1 (CH3O), 55.9, 37.3, 27.0, 15.9 (CH3), 11.4 (CH3).

N-(1-(2-(2,5-dimethoxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4j). Pale yellow solid; yield = 60%; mp = 127°C–129°C; 1H NMR (300 MHz, DMSO-d6) δ 8.59 (s, 1H, N=C-H), 8.33 (d, J = 7.5 Hz, 1H), 7.89 (d, J = 7.5 Hz, 1H), 7.32 (d, J = 3.1 Hz, 1H), 7.04 (d, J = 8.9 Hz, 2H), 7.00 (td, J = 8.6, 2.9 Hz, 1H), 6.92 (d, J = 8.6 Hz, 1H), 3.81 (s, 3H, OMe), 3.76 (d, J = 6.9 Hz, 1H), 3.74 (s, 3H, OMe), 2.07–1.89 (m, 1H), 1.65–1.39 (m, 2H), 0.97–0.87 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 168.3 (C=O), 167.4 (C=O), 153.7 (Ar-O), 152.7 (Ar-OMe), 152.4 (Ar-OMe), 142.9 (C=N), 133.1, 130.2, 123.2, 118.7, 118.4, 118.2, 117.4, 113.8, 109.4, 56.7 (CH3O), 55.9 (CH3O), 55.7, 37.2, 25.2, 16.0 (CH3), 11.4 (CH3).

N-(1-(2-(3,4-dimethoxybenzylidene)hydrazineyl)-3-methyl-1-oxopentan-2-yl)-2-hydroxy benzamide (4k). White solid; yield = 60%; mp = 108°C–110°C; 1H NMR (300 MHz, DMSO-d6) δ 8.18 (s, 1H, N=C-H), 7.96 (dd, J = 8.3, 1.4 Hz, 1H), 7.39–7.25 (m, 1H), 7.17 (dd, J = 7.5, 2.1 Hz, 1H), 7.01 (t, J = 8.3 Hz, 1H), 6.96 (dd, J = 7.5, 2.1 Hz, 1H), 6.83 (t, J = 7.5 Hz, 1H), 4.44 (d, J = 7.4 Hz, 1H), 3.79 (s, 6H, 2 × OCH3), 2.07–1.90 (m, 1H), 1.67–1.34 (m, 1H), 1.02–0.74 (m, 6H, 2 × CH3). 13C NMR (75 MHz, DMSO-d6) δ 173.1 (C=O), 173.0 (C=O), 168.0 (Ar-O), 167.4 (Ar-OMe), 166.5 (Ar-OMe), 151.1 (C=N), 149.4, 147.9, 127.3, 122.4, 118.5, 118.0, 117.4, 111.8, 108.6, 57.0 (CH3O), 56.0 (CH3O), 55.9, 37.0, 25.2, 15.9 (CH3), 11.3 (CH3).

2-hydroxy-N-(1-(2-((3-hydroxy-5-(hydroxymethyl)-2-methylpyridin-4-yl)methylene) hydrazineyl)-3-methyl-1-oxopentan-2-yl)benzamide (4l). Yellow solid; yield = 86%; mp = 198°C–200°C; 1H NMR (300 MHz, DMSO-d6) δ 8.64 (s, 1H, N=C-H), 7.77 (d, J = 7.8 Hz, 1H), 7.19 (s, 1H, H pyridine), 7.09 (t, J = 8.0 Hz, 1H), 6.75 (d, J = 8.1 Hz, 1H), 6.43 (d, J = 8.0 Hz, 1H), 4.46 (s, 2H, CH2O), 4.44 (d, J = 6.9 Hz, 1H), 2.29 (s, 3H, CH3-Py), 2.01–1.87 (m, 1H), 1.54–1.25 (m, 2H), 0.99 (d, J = 6.5 Hz, 3H), 0.93 (t, J = 7.4 Hz, 3H). 13C NMR (75 MHz, DMSO-d6) δ 168.0 (C=O), 166.2 (C=O), 158.3 (Ar-O), 154.1 (Py-O), 150.2 (Py-CH3), 146.8 (C=N), 139.3 (C=Npy), 133.8, 132.6, 130.4, 128.9, 120.2, 118.5, 118.2, 68.6 (-CH2O-Py), 60.2, 37.5, 25.1, 16.0 (CH3-Py), 15.5 (CH3), 11.3 (CH3).

Biological activity assays

Cell lines and culture medium

Nonmalignant cells (CHO-K1) along with the adenocarcinoma colon cancer cell line (SW480) were used for biological experiments. Cultures were obtained from The European Collection of Authenticated Cell Cultures (England), cultivated with horse serum, 1% nonessential amino acids (Gibco Invitrogen, Carlsbad, CA), 10% heat-inactivated (56°C), and maintained in Dulbecco’s Modified Eagle Medium. Serum was reduced to 3% and the medium was supplemented with insulin (10 mg/ml), selenium (5 ng/ml), and transferrin (5 mg/ml) (Herrera et al., 2018).

Cytotoxic activity

The colorimetric sulforhodamine B (SRB) assay was used to evaluate the cytotoxicity activity of the synthesized trihybrids as well as the reference compounds. Using 96-well tissue culture plates, cells were plated at a seeding density of 20,000 cells/well at 37°C and incubated in a humidified atmosphere containing 5% CO2. All cultures were allowed to grow for 24 hours, and then treated with vehicle control containing 1% (v/v) DMSO or increasing concentrations (0.01–200 μM) of the target trihybrids, as well 5-FU (the standard drug), pyridoxal isonicotinoyl hydrazone (PIH), parental compounds (SA and isoleucine), and the equimolar mixture of these. After cell fixation with trichloroacetic acid (50% v/v, 1 hour at 4°C) (Merck), cell proteins were determined by staining with 0.4% (w/v) SRB (Sigma-Aldrich, United States), and afterward unbound SRB was removed washed with 1% acetic acid and left for air-drying. After that, 10 mM Tris base was used to dissolve protein-bound SRB. The absorbance measured at 492 nm in a microplate reader (Mindray MR-96A) (Perez et al., 2014). All experiments were conducted at least three times.

Antiproliferative ativity

The assessment of antiproliferative activity of the most promising compound was also tested through SRB assay. Typically, after the cells were seeded at an initial density of 2,500 cells/well in 96-well tissue culture plates, they were incubated in the same conditions described earlier for cytotoxicity. Afterward, cultures were allowed to grow for a period of 24 hours and then treated with DMSO (vehicle control, 1%) or increasing concentrations of the hit-trihybrid 4j [6.25–100 μM, ranges dependent on the inhibitory concentration (IC50) values] for 0, 2, 4, 6, and 8 days. The culture medium is changed every 48 hours. After the incubation time, cells were fixed, stained, and read as previously reported (Castrillón et al., 2019).

Statistical analysis

Data obtained from three independent experiments was reported as mean ± standard error (SE). One-way analysis of variance and Dunnett’s test were used to evaluated statistical differences between treated cells and control group (nontreated). p-values lesser than 0.05 (typically ≤ 0.05) were considered statistically significant. To analyze experimental data, GraphPad Prism software V.7 for Windows was used.

Theoretical drug-likeness and pharmacokinetic indices

Ten biopharmaceutical relevant properties were screened for the most promising trihybrid 4j. To that, MW, topographical polar surface area (TPSA), and rotatable bonds descriptors were calculated by using Molinspiration Chemoinfomatics Software. Meanwhile, human serum albumin (LogKHSA), apparent predicted intestinal permeability (app. Caco-2 and MDCK models), and gastrointestinal uptake and absorption (%GI) were estimated using the online PreADMET 2.0 program. In addition, the ALOGPS 2.1 algorithm was used to calculate the lipophilicity parameter, log Po/w.


RESULTS AND DISCUSSION

Chemistry

Novel salicylic acid/isoleucine/N-acylhydrazone (SA-Ile-NAH) hybrids were synthetized through a four-step (i–iv) divergent sequence of reactions shown in Scheme 1. To summarize, commercially available isoleucine as starting material was esterified to the corresponding methyl ester using thionyl chloride (SOCl2) in methanol solvent by a modified Fischer esterification mechanism (Castrillón et al., 2019) to give ester 1 in quantitative yields, which was subjected to one-pot coupling procedure with available SA using HBTU as amide bond promoter and triethylamine (Et3N) as a Lewis base to obtain 2 in good yields (71%) (Castrillón et al., 2019; Herrera et al., 2021). Subsequently, this intermediate reacted with hydrazine hydrate via nucleophilic substitution to obtain in high yield (93%) the desired N-acylhydrazide 3 (Coa et al., 2015; Vergara et al., 2017). Finally, the target SA-Ile-NAH hybrids 4a–l were obtained in moderate to excellent yields (54%–94%) via Brønsted acid-catalyzed imine formation between the hydrazide 3 with the corresponding aromatic aldehydes (ArCHO) in ethanol under conditions of microwave heating, as described in Scheme 1 (Coa et al., 2015; Vergara et al., 2017). The structures of all compounds have been fully characterized by a combined 1H-NMR and 13C-NMR study with carbon atom types (C, CH, CH2, and CH3), determined by using the distortionless enhancement by polarization transfer or attached proton test pulse sequence. The signals were assigned using two-dimensional heteronuclear correlations (correlation spectroscopy and heteronuclear single quantum correlation). The 1H-NMR spectra of hybrids 4a–l dissolved in DMSO-d6 showed a characteristic singlet peak within the 7.98–9.22 ppm range which is assigned to the proton of the azomethine group (-CH=N-). The isoleucine moiety was recognized by their two methyl groups which give a characteristic row of double signals within the 0.64–1.06 ppm range, while the signals corresponding to α-CH (~4.5 ppm) and CH2 aliphatic protons were registered in the range of δ = 1.4–2.0 ppm. 1H-NMR spectra also confirm the presence of aromatic protons which were located in the downfield region (6.70–8.60 ppm). Analysis of the 13C NMR data for the 4a–l hybrids showed a peak in the range of 140–152 ppm which confirmed the presence of azomethine carbon in the hybrid structure. Moreover, the 13C-NMR spectrum shows 5 signals in the 10–60 ppm upfield region, which was assigned to the carbon atoms of the aliphatic chain in the isoleucine portion. In addition, 13C-NMR spectra of hybrids show two picks around 170 and 167 ppm, respectively, which confirms the presence of both carbonyl groups (C=O). Finally, aromatic carbons appear between 120 and 170 ppm. (see Supplementary Online Material for further details)

Scheme 1. Reagents and conditions: (i) SOCl2, MeOH, 0°C; then rt, 48 hours, 95%. (ii) HBTU, Et3N, THF, rt, 24 hours, 71%. (iii) NH2NH2.H2O, 98%, MeOH, reflux, 24 hours, 93%. (iv) AcOH (cat), EtOH, reflux, 10 minutes, (μw), 54%–94%.

[Click here to view]

Biological activity

Cytotoxic effect of SA-Ile-NAH hybrids on SW480 and CHO-K1 cell lines

The cytotoxic effect of the synthesized SA-Ile-NAH hybrids using an in vitro model of human colorectal adenocarcinoma (SW480) and the nonmalignant CHO-K1 cell line were measured by the SRB colorimetric assay. In addition to the trihybrid compounds, 5-FU and the promising PIH were also included as positive controls, as well as the parent scaffolds SA and isoleucine and their equimolar mixture. Cytotoxicity data for the compounds are reported as half-maximal IC50 values and listed in Table 1.

In general, most trihybrids exhibited comparable cytotoxic activity to the reference drug 5-FU and PIH, being significantly more active than the parental subunits (SA and isoleucine) and their respective unlinked equimolar mixture, displaying promising in vitro cytotoxic effects in a concentration range of 20.43 ± 1.88 and 168.2 ± 20.33 μM after 48 hours of treatment. Furthermore, as can be seen in Table 1, most of those compounds increased the cytotoxic response over time as evidenced by the reduction in the IC50 values at 48 hours (see values at 24 vs. 48 hours). It is worth noting that the proposed molecular design allowed obtaining molecules with improved cytotoxic properties with respect to their respective parental compounds (SA and isoleucine), considerably higher activity than the equimolar mixture, and exhibited cytotoxic potency similar to that of 5-FU and PIH, thereby demonstrating a remarkable advantage of the molecular hybridization strategy.

Despite trihybrids 4a-c, 4f, 4h, 4i, and 4l improving the efficacy over malignant cells (SW480), a decrease in selectivity due to the high toxicity on nonmalignant cells (CHO-K1) was also observed after 48 hours of treatment. It was, however, observed that the 2,5-dimethoxy-substituited trihybrid 4j highlighted not only for its good cytotoxic effect (IC50 = 78.15 ± 6.73 μM) but has no significant toxicity to nontumor cells (IS = 1.31). More importantly, a microscopic inspection (Fig. 2A and B) revealed that hybrid 4j induced marked cell morphology changes related to the size and shape in SW480 colorectal cell line which is highly associated with cell death process, while a typical and healthy shape was observed after treatment with DMSO as control vehicle. Moreover, hybrid 4j was able to induce a visible reduction in the total number of cells in a dose-dependent manner suggesting that this compound is able to induce a short-term loss of cell viability by either cytostatic or cytotoxic effect on tumor cells.

Table 1. Cytotoxic effect of SA-Ile-NAH hybrids against SW480 and CHO-K1 cell lines after 24 and 48 hours of treatment.

[Click here to view]

Studies of the SAR were conducted at 48 hours after incubation. Results revealed a synergistic action of the parent subunits when they are linked to form a single structure in the hybrid. Hybrid 4a exhibited better cytotoxicity than compound 4b, which shows the importance of the naphthyl group over the phenyl group for activity. The change of the phenyl group by the pyridyl group causes a slight decrease in the activity (4b vs. 4l). The presence of a hydroxyl group at the C-3 position of the aromatic ring increased the activity (4b vs. 4c). However, when this group changes position, the activity decreases (4c vs. 4d and 4e). Hybrid 4g, a combination of compounds 4b and 4f, was not active, showing that there was no synergistic effect in SW480 cells, which was reflected in decreased activity compared to the initial hybrids (Fig. 3).

The methylation effect is not clear, because when the hybrids 4c and 4h are compared, it is possible to observe a decrease in activity. However, an opposite behavior is observed when the other hybrids are compared (4d vs. 4i and 4e vs. 4j); in these cases, the activity increases. These findings are in good agreement with other reports, where compounds combining structural moieties of SA and hydrazone displayed a promising anticancer potential (Alam et al., 2017; Jongstra et al., 2006; Misko et al., 2019). Moreover, similar results were reported by Herrera et al. (2021) who also measured good cytotoxic and selective activity of hybrids of SA incorporating an S-allylcysteine fragment, using human colorectal adenocarcinoma SW480 cell lines as model. We hypothesized that the cytotoxic effect on SW480 cells observed after SA-Ile-NAH hybrids exposure could be associated to the fact that these compounds would be able to target essential bioreceptors through formation of intermolecular hydrogen bonds, oxidative stress triggered by reactive free-radical species formation, and/or the ability of metal complexation. However, further experimental and computational investigations need to be conducted in order to elucidate the most plausible antiproliferative mechanism of these promising trihybrids. According to the aforementioned findings, merging SA, isoleucine, and hydrazone moieties into a unique structural core provides promising compounds that could be considered a novel interesting therapeutical scaffold for developing new antitumoral compounds, particularly in CRC. In addition, further investigations by introducing electron-withdrawing substituents and electron-donating core combinations in the aromatic ring attached to the hydrazone moiety will be conducted aiming at improving the selectivity of most of the tested compounds. Studies of how these electronics effects can affect the antiproliferative potency, not only in CRC but other most commonly diagnosed cancers at even higher risk, are also needed.

Figure 2. Images of growth control of SW480 cancer cell line (magnification: 20×). (A). Vehicle control. (DMSO) treatment. (B). After hybrid 4j exposure (48 hours posttreatment)

[Click here to view]

Antiproliferative effect of 4j on SW480 cells

The antiproliferative effect of the most promising compound 4j highlighted by the selectivity and cytotoxic effect was further evaluated to know if it can preserve its activity against SW480 cells after periods of time ranging from 2 to 8 days of treatment (Table 2).

To achieve this goal, SW480 adenocarcinoma cells were treated with increasing concentrations of 6.25, 12.5, 25, 50, and 100 μM of 4j for 8 days and the cell viability was measured by the colorimetric SRB assay. As shown in Figure 4, our results in comparison with the control showed that trihybrid 4j inhibited the proliferation of human colorectal adenocarcinoma cells (SW480), in dose- and time-dependent manner. Thus, treatment of tumor cells with 4j reduced the number of viable cells and prevented the exponential growth of the primary tumor cell line examined. Particularly, after 4 days of treatment with 25–100 μM, this hybrid significantly reduced the viability of the SW480 cells compared to untreated control cells, with p-values lesser than 0.001. Notably, when SW480 cells were treated with a low concentration of 4j (12.5 μM), the number of tumor cell colonies was also significantly reduced within 4 days of treatment. All these findings demonstrated that hit compound 4j caused a time- and dose-dependent loss of SW480 cell viability confirming the previous results of viability and suggesting that trihybrid 4j may in vitro induce either cytostatic/cytotoxic response on the colon cancer cell line model. We conclude here that, the synthetized SA-Ile-NAH hybrids compounds, particularly compound 4j, emerge as attractive building blocks in the search of new anti-CRC agents.

Figure 3. SAR for the cytotoxic activity of the novel SA-Ile-NAH hybrid.

[Click here to view]

Table 2. Data viability (%) of the five concentrations evaluated against SW480 cell.

[Click here to view]

Figure 4. Antiproliferative effect of hybrid 4j against SW480 cell line; Data are presented as the mean ± SE of at least three independent experiments (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). Vehicle control was assumed as 100% viable.

[Click here to view]

Theoretical pharmacokinetic and drug-likeness properties of promising trihybrid 4j

Drug-likeness and pharmacokinetics inspections represent a valuable strategy to complete the journey from initial discovery to the marketplace of novel drug candidates in pharmaceutical research, particularly in assessing the quality of novel antitumor scaffolds. Here, for the most promising trihybrid 4j, we have predicted 10 biopharmaceutical indices that have the most profound influence on drug-like properties of a molecule, such as molecular weight, polar surface area, number of hydrogen bond acceptors/donors, octanol–water partition coefficient, binding-serum albumin, human intestinal permeation (Caco-2 and MDCK cells models), and a number of rotatable bonds. The drug-likeness and pharmacokinetic predictions provided by the SwissADME and pre-ADMET software are listed in Table 3. Our results show that a favorable pharmacokinetics profile was found for the examined hybrid when compared to 95% of oral U.S. Food and Drug Administration-marketed drugs. 4j did not violate any of Lipinski’s rule of five (Lipinski et al., 1997). Moreover, the degree of lipophilicity (expressed as LogPo/w) was in good agreement with the optimal quality parameters for oral lipid-based formulations (–2.0 to 6.0) (Ditzinger et al., 2019). Furthermore, 4j displayed an optimal human intestinal absorption (%GI) number larger than 80%, and good predicted permeability data in the range of 536–1,077 nm/second when traditional Caco-2 and MDCK cells models were used, respectively (Broccatelli et al., 2016; Pham-The et al., 2018; Press and Di Grandi, 2008). Meanwhile, the compound exhibits a computational TPSA value of 106.75 Å2 within the ideal value of less than 140 Å2 (Ertl et al., 2000). We predict that 4j would bind favorably to human serum albumin (expressed as logKHSA), within the recommended therapeutic range suggested for potential drug candidates (–1.5 to 1.5) (Colmenarejo, 2003; Zhivkova, 2015). Finally, the pan-assay interference compounds (PAINS) filter, which provides early alerts for potential toxicity as part of successful drug discovery, showed that the promising 4j hybrid can be regarded as valid starting points in cancer drug discovery.

Taken altogether, merging salicylic acid, N-acylhydrazone, and isoleucine scaffolds into a new single chemical structure provides bioactive compounds with an optimal pharmacokinetic profile. Accordingly, novel trihybrid scaffolds based on SA-Ile-NAH should be taken into account for further investigations focused on the design of new oral antitumoral drug candidates to combat CRC.

Table 3. Lipinski’s rule and pharmacokinetic indices for the hit compound 4j.

[Click here to view]


CONCLUSION

A novel series of SA-Ile-NAH trihybrids was designed, synthesized, and screened for cytotoxic effect on SW480 human colon adenocarcinoma cells. Most of these hybrids were more potent than the reference drug (5-FU) and the equimolar mixture as well as than the parental subunits (SA and isoleucine). In particular, the 2,5-dimethoxy-substituited trihybrid 4j was the most promising compound because it not only displayed good cytotoxic potency against SW480 human colon cancer cells, but also expressed remarkable antiproliferative activity at lower concentrations, and no significant toxic effects on nontumorigenic cells. Moreover, 4j showed an optimal drug-likeness/pharmacokinetic profile. Considering all these results, trihybrids based on SA-Ile-NAH emerge as a promising therapeutic opportunity against CRC in the forthcoming years.


FUNDING

This work was financially supported by Universidad de Antioquia through the Committee for Research Development (Comité para el Desarrollo y la Investigación) CIEN–CODI–Vicerrectoría de Investigación (Primer proyecto), project number 2018-21112.


CONFLICT OF INTEREST

The authors declare that there is no conflict of interest.


ETHICAL APPROVAL

This study does not involve the use of animals or human subjects.


AUTHOR CONTRIBUTIONS

All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; agreed to submit to the current journal; gave final approval of the version to be published; and agree to be accountable for all aspects of the work. All the authors are eligible to be an author as per the international committee of medical journal editors (ICMJE) requirements/guidelines


DATA AVAILABILITY

All data generated and analyzed are included within this research article.


PUBLISHER’S NOTE

This journal remains neutral with regard to jurisdictional claims in published institutional affiliation.


REFERENCES

 Alam MS, Choi SU, Lee DU. Synthesis, anticancer, and docking studies of salicyl-hydrazone analogues: A novel series of small potent tropomyosin receptor kinase A inhibitors. Bioorg Med Chem, 2017; 25(1):389–96. CrossRef

 Al-awar R, Mamai A, Zhang A, inventors. Acyl hydrazone linkers, methods and uses thereof. WO2019109188A1, 2019 June 13.

 Ali I, Wani WA, Haque A, Saleem K. Glutamic acid and its derivatives: candidates for rational design of anticancer drugs. Future Med Chem, 2013; 5(8):961–78. CrossRef

 Ananieva EA, Wilkinson AC. Branched-chain amino acid metabolism in cancer. Curr Opin Clin Nutr Metab Care, 2018; 21(1):64–70. CrossRef

 Bagheri M, Hesari A, Sarabi P, Rahimi H, Baazm M, Ghasemi F. Antiproliferative effect of aspirin on colorectal cancer cell line. Iran J Tox, 2018; 12(5):1–4. CrossRef

 Bener A, Dogan M, Azab A, Rashed A, Siddiqui M. Short communication: amino acid profiles among colorectal cancer patients. Biomed Res, 2006; 17(2):149–54.

 Brennan CA, Nakatsu G, Gallini Comeau CA, Drew DA, Glickman JN. Aspirin modulation of the colorectal cancer-associated microbe Fusobacterium nucleatum. mBio, 2021; 12(2):e00547–21. CrossRef

 Broccatelli F, Salphati L, Plise E. Predicting passive permeability of drug-like molecules from chemical structure: where are we? Mol Pharm, 2016; 13(12):4199–208. CrossRef

 Burn J, Sheth H, Elliott F, Reed L, Macrae F, Mecklin JP. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10-year follow-up and registry-based 20-year data in the CAPP2 study: a double-blind, randomised, placebo-controlled trial. Lancet, 2020; 395(10240):1855–63. CrossRef

 Buss JL, Hermes-Lima M, Ponka P. Pyridoxal isonicotinoyl hydrazone and its analogues. Adv Exp Med Biol, 2002; 509:205–29. CrossRef

 Castrillón W, Herrera-R A, Prieto LJ, Conesa-Milián L, Carda M, Naranjo T, Maldonado ME, Cardona-G W. Synthesis and in-vitro evaluation of S-allyl cysteine ester-caffeic acid amide hybrids as potential anticancer agents. Iran J Pharm Res, 2019; 18(4):1770–89.

 Chaston TB, Lovejoy DB, Watts RN, Richardson DR. Examination of the antiproliferative activity of iron chelators: multiple cellular targets and the different mechanism of action of triapine compared with desferrioxamine and the potent pyridoxal isonicotinoyl hydrazone analogue 311. Clin Cancer Res, 2003; 9(1):402–14.

 Chen YL, Kong X, Xie Y, Hider RC. The interaction of pyridoxal isonicotinoyl hydrazone (PIH) and salicylaldehyde isonicotinoyl hydrazone (SIH) with iron. J Inorg Biochem, 2018; 180:194–203. CrossRef

 Coa JC, Castrillón W, Cardona W, Carda M, Ospina V, Muñoz JA, Vélez ID, Robledo SM. Synthesis, leishmanicidal, trypanocidal and cytotoxic activity of quinoline-hydrazone hybrids. Eur J Med Chem, 2015; 101:746–53. CrossRef

 Colmenarejo G. In silico prediction of drug-binding strengths to human serum albumin. Med Res Rev, 2003; 23(3):275–301. CrossRef

 Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P. Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention. Lancet Oncol, 2009; 10(5):501–7. CrossRef

 Dasgupta S, Karim S, Banerjee S, Saha M, Das Saha K, Das D. Designing of novel zinc(ii) Schiff base complexes having acyl hydrazone linkage: study of phosphatase and anti-cancer activities. Dalton Trans, 2020; 49(4):1232–40. CrossRef

 Deb J, Dibra H, Shan S, Rajan S, Manneh J, Kankipati CS. Activity of aspirin analogues and vanillin in a human colorectal cancer cells. Oncol Rep, 2011; 26(3):557–65.

 Delphan M, Lin T, Liesenfeld DB, Nattenmüller J, Böhm JT, Gigic B. Associations of branched-chain amino acids with parameters of energy balance and survival in colorectal cancer patients: results from the ColoCare Study. Metabolomics, 2018; 2018(14):22–31. CrossRef

 Ditzinger F, Price DJ, Ilie AR, Köhl NJ, Jankovic S, Tsakiridou G. Lipophilicity and hydrophobicity considerations in bio-enabling oral formulations approaches—a PEARRL review. J Pharm Pharmacol, 2019; 71:464–82. CrossRef

 Elder DJ, Hague A, Hicks DJ, Paraskeva C. Differential growth inhibition by the aspirin metabolit salicylate in human colorectal tumor cell lines. Cancer Res, 1996; 56(10):2273–6.

 Elder DJ, Paraskeva C. Induced apoptosis in the prevention of colorectal cancer by non-steroidal anti-inflammatory drugs. Apoptosis, 1999; 4(5):365–72. CrossRef

 Ertl P, Rohde B, Selzer P. Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. J Med Chem, 2000; 43(20):3714–7. CrossRef

 Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M. Global cancer observatory: cancer today. International Agency for Research on Cancer, Lyon, France, 2020. Availabel via https://gco.iarc.fr/today (Accessed 1 February 2022).

 Giardina C, Inan MS. Nonsteroidal anti-inflammatory drugs, short-chain fatty acids, and reactive oxygen metabolism in human colorectal cancer cells. Biochim Biophys Acta, 1998; 1401(3):277–88. CrossRef

 GLOBOCAN. Cancer incidence and mortality, 2020. Available via https://www.uicc.org/news/globocan-2020-new-global-cancer-data (Accessed 19 April 2021).

 Gökçe K, ??kan N, Türker N, Ka?it M. Evaluating combined effect of naringin and salicylic acid on colon cancer cell culture. Turkish Med Stud J, 2017; 4:17–24 CrossRef

 Herrera A, Castrillón W, Otero E, Ruiz E, Carda M, Agut R. Synthesis and antiproliferative activity of 3- and 7-styrylcoumarins. Med Chem Res, 2018; 27:1893–905. CrossRef

 Herrera-R A, Castrillón W, Pastrana M, Yepes AF, Cardona-G W. Promising hybrids derived from S-allylcysteine and NSAIDs fragments against colorectal cancer: synthesis, in-vitro evaluation, drug-likeness and in-silico ADME/tox studies. Iran J Pharm Res, 2021; (3):351–67.

 Høydahl Ø, Edna TH, Xanthoulis A, Lydersen S, Endreseth BH. Long-term trends in colorectal cancer: incidence, localization, and presentation. BMC Cancer, 2020; 20(1):1077–90. CrossRef

 Huff SE, Mohammed FA, Yang M, Agrawal P, Pink J, Harris ME. Structure-guided synthesis and mechanistic studies reveal sweetspots on naphthyl salicyl hydrazone scaffold as non-nucleosidic competitive. J Med Chem, 2018; 61(3):666–80. CrossRef

 Iliev I, Kontrec D, Detcheva R, Georgieva M, Balacheva A, Galic N. Cancer cell growth inhibition by aroylhydrazone. Biotechnol. Biotec. Equip, 2019; 33(1):756–63. CrossRef

 Jongstra J, Kotra LP, Wrana JL, inventors. Salicylic acid hydrazones as inhibitors of the erk mapkinase for the treatment of cancer. WO 2006136008 A1, 2006, December 28.

 Lieu EL, Nguyen T, Rhyne S. Amino acids in cancer. Exp Mol Med, 2020; 52:15–30. CrossRef

 Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery. Adv Drug Deliv Rev, 1997; 23:3–25. CrossRef

 Long L, Yang W, Liu L, Tobias DK, Katagiri R, Wu K. Dietary intake of branched-chain amino acids and survival after colorectal cancer diagnosis. Int J Cancer, 2020; 148(10):2471–80. CrossRef

 Misko TA, Liu YT, Harris ME, Oleinick NL, Pink J, Lee HY. Structure-guided design of anti-cancer ribonucleotide reductase inhibitors. J Enzyme Inhib Med Chem, 2019; 34(1):438–50. CrossRef

 Murata K, Moriyama M. Isoleucine, an essential amino acid, prevents liver metastases of colon cancer by antiangiogenesis. Cancer Res, 2007; 67(7):3263–8. CrossRef

 Paterson JR, Lawrence JR. Salicylic acid: a link between aspirin, diet and the prevention of colorectal cancer. QJM, 2001; 94(8):445–48. CrossRef

 Pathi S, Jutooru I, Chadalapaka G, Nair V, Lee SO, Safe S. Aspirin inhibits colon cancer cell and tumor growth and downregulates specificity protein (Sp) transcription factors. PLoS One, 2012; 7(10):e48208–17. CrossRef

 Patrignani P, Patrono C. Aspirin and cancer. J Am Coll Cardiol, 2016; 68(9):967–76. CrossRef

 Peleg II, Maibach HT, Brown SH, Wilcox CM. Aspirin and nonsteroidal anti-inflammatory drug use and the risk of colorectal cancer. Arch Intern Med, 1994; 154(4):394–99. CrossRef

 Perez J, Maldonado M, Rojano B, Alzate F, Saez J, Cardona W. Comparative Antioxidant, Antiproliferative and Apoptotic effects of Ilex laurina and Ilex paraguariensis on Colon Cancer Cells Trop J Pharm Res, 2014; 13(8):1279–86. CrossRef

 Pham-The H, Cabrera-Pérez MÁ, Nam NH, Castillo-Garit JA, Rasulev B, Le-Thi-Thu H, Casañola-Martin GM. In Silico Assessment of ADME properties: advances in Caco-2 cell monolayer permeability modeling. Curr Top Med Chem, 2018; 18(26):2209–29. CrossRef

 Press B, Di Grandi D. Permeability for intestinal absorption: Caco-2 assay and related issues. Curr Drug Metab, 2008; 9(9):893–900. CrossRef

 Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin, 2021; 71:7–33. CrossRef

 Song S, Christova T, Perusini S, Alizadeh S, Bao RY, Miller BW. Wnt inhibitor screen reveals iron dependence of β-catenin signaling. Cancer Res, 2011; 71(24):7628–39. CrossRef

 Spiegel R. Aspirin as a painkiller in patients with a high cardiovascular risk profile. Nat Rev Cardiol, 2020; 17(9):609–19. CrossRef

 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021; 71:209–49. CrossRef

 Thota S, Rodrigues DA, Pinheiro PSM, Lima LM, Fraga CAM. N-Acylhydrazones as drugs. Bioorg Med Chem Lett, 2018; 28(17):2797–306. CrossRef

 Thun MJ, Namboodiri MM, Heath CW Jr. Aspirin use and reduced risk of fatal colon cancer. N Engl J Med, 1991; 325(23):1593–96. CrossRef

 Vergara S, Carda M, Agut R, Yepes LM, Robledo SM, Cardona W. Synthesis, antiprotozoal activity and cytotoxicity in U-937 macrophages of triclosan–hydrazone hybrids. Med Chem Res, 2017; 26:3262–73. CrossRef

 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):1829–52. CrossRef

 Wakshlag JJ, Kallfelz FA, Wakshlag RR, Davenport GM. The effects of branched-chain amino acids on canine neoplastic cell proliferation and death. J Nutr, 2006; 136(7):2007–10. CrossRef

 Wang LB, Zhang SZ, Ding KF, Zheng S. A study of the free amino acids uptake in Colon Cancer. Zhejiang Y, 1997; 19:208–9.

 Zhivkova ZD. Studies on drug-human serum albumin binding: the current state of the matter. Curr Pharm Des, 2015; 21(14):1817–30. CrossRef


SUPPLEMENTARY MATERIAL

1H and 13C NMR spectra of the intermediate compounds 1–3 as well as target hybrids 4a–l are provided in the supporting information.

Reference

Alam MS, Choi SU, Lee DU. Synthesis, anticancer, and docking studies of salicyl-hydrazone analogues: A novel series of small potent tropomyosin receptor kinase A inhibitors. Bioorg Med Chem, 2017; 25(1):389-96.https://doi.org/10.1016/j.bmc.2016.11.005

Al-awar R, Mamai A, Zhang A, inventors. Acyl hydrazone linkers, methods and uses thereof. WO2019109188A1, 2019 June 13.

Ali I, Wani WA, Haque A, Saleem K. Glutamic acid and its derivatives: candidates for rational design of anticancer drugs. Future Med Chem, 2013; 5(8):961-78.https://doi.org/10.4155/fmc.13.62

Ananieva EA, Wilkinson AC. Branched-chain amino acid metabolism in cancer. Curr Opin Clin Nutr Metab Care, 2018; 21(1):64-70.https://doi.org/10.1097/MCO.0000000000000430

Bagheri M, Hesari A, Sarabi P, Rahimi H, Baazm M, Ghasemi F. Antiproliferative effect of aspirin on colorectal cancer cell line. Iran J Tox, 2018; 12(5):1-4.https://doi.org/10.32598/IJT.12.5.474.1

Bener A, Dogan M, Azab A, Rashed A, Siddiqui M. Short communication: amino acid profiles among colorectal cancer patients. Biomed Res, 2006; 17(2):149-54.

Brennan CA, Nakatsu G, Gallini Comeau CA, Drew DA, Glickman JN. Aspirin modulation of the colorectal cancer-associated microbe Fusobacterium nucleatum. mBio, 2021; 12(2):e00547-21.https://doi.org/10.1128/mBio.00547-21

Broccatelli F, Salphati L, Plise E. Predicting passive permeability of drug-like molecules from chemical structure: where are we? Mol Pharm, 2016; 13(12):4199-208.https://doi.org/10.1021/acs.molpharmaceut.6b00836

Burn J, Sheth H, Elliott F, Reed L, Macrae F, Mecklin JP. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10-year follow-up and registry-based 20-year data in the CAPP2 study: a double-blind, randomised, placebo-controlled trial. Lancet, 2020; 395(10240):1855-63.https://doi.org/10.1016/S0140-6736(20)30366-4

Buss JL, Hermes-Lima M, Ponka P. Pyridoxal isonicotinoyl hydrazone and its analogues. Adv Exp Med Biol, 2002; 509:205-29.https://doi.org/10.1007/978-1-4615-0593-8_11

Castrillón W, Herrera-R A, Prieto LJ, Conesa-Milián L, Carda M, Naranjo T, Maldonado ME, Cardona-G W. Synthesis and in-vitro evaluation of S-allyl cysteine ester-caffeic acid amide hybrids as potential anticancer agents. Iran J Pharm Res, 2019; 18(4):1770-89.

Chaston TB, Lovejoy DB, Watts RN, Richardson DR. Examination of the antiproliferative activity of iron chelators: multiple cellular targets and the different mechanism of action of triapine compared with desferrioxamine and the potent pyridoxal isonicotinoyl hydrazone analogue 311. Clin Cancer Res, 2003; 9(1):402-14.

Chen YL, Kong X, Xie Y, Hider RC. The interaction of pyridoxal isonicotinoyl hydrazone (PIH) and salicylaldehyde isonicotinoyl hydrazone (SIH) with iron. J Inorg Biochem, 2018; 180:194-203.https://doi.org/10.1016/j.jinorgbio.2017.12.007

Coa JC, Castrillón W, Cardona W, Carda M, Ospina V, Muñoz JA, Vélez ID, Robledo SM. Synthesis, leishmanicidal, trypanocidal and cytotoxic activity of quinoline-hydrazone hybrids. Eur J Med Chem, 2015; 101:746-53.https://doi.org/10.1016/j.ejmech.2015.07.018

Colmenarejo G. In silico prediction of drug-binding strengths to human serum albumin. Med Res Rev, 2003; 23(3):275-301.https://doi.org/10.1002/med.10039

Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P. Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention. Lancet Oncol, 2009; 10(5):501-7.https://doi.org/10.1016/S1470-2045(09)70035-X

Dasgupta S, Karim S, Banerjee S, Saha M, Das Saha K, Das D. Designing of novel zinc(ii) Schiff base complexes having acyl hydrazone linkage: study of phosphatase and anti-cancer activities. Dalton Trans, 2020; 49(4):1232-40.https://doi.org/10.1039/C9DT04636D

Deb J, Dibra H, Shan S, Rajan S, Manneh J, Kankipati CS. Activity of aspirin analogues and vanillin in a human colorectal cancer cells. Oncol Rep, 2011; 26(3):557-65.

Delphan M, Lin T, Liesenfeld DB, Nattenmüller J, Böhm JT, Gigic B. Associations of branched-chain amino acids with parameters of energy balance and survival in colorectal cancer patients: results from the ColoCare Study. Metabolomics, 2018; 2018(14):22-31.https://doi.org/10.1007/s11306-017-1314-8

Ditzinger F, Price DJ, Ilie AR, Köhl NJ, Jankovic S, Tsakiridou G. Lipophilicity and hydrophobicity considerations in bio-enabling oral formulations approaches-a PEARRL review. J Pharm Pharmacol, 2019; 71:464-82.https://doi.org/10.1111/jphp.12984

Elder DJ, Hague A, Hicks DJ, Paraskeva C. Differential growth inhibition by the aspirin metabolit salicylate in human colorectal tumor cell lines. Cancer Res, 1996; 56(10):2273-6.

Elder DJ, Paraskeva C. Induced apoptosis in the prevention of colorectal cancer by non-steroidal anti-inflammatory drugs. Apoptosis, 1999; 4(5):365-72.https://doi.org/10.1023/A:1009696505108

Ertl P, Rohde B, Selzer P. Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. J Med Chem, 2000; 43(20):3714-7.https://doi.org/10.1021/jm000942e

Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M. Global cancer observatory: cancer today. International Agency for Research on Cancer, Lyon, France, 2020. Availabel via https://gco.iarc.fr/today (Accessed 1 February 2022).

Giardina C, Inan MS. Nonsteroidal anti-inflammatory drugs, short-chain fatty acids, and reactive oxygen metabolism in human colorectal cancer cells. Biochim Biophys Acta, 1998; 1401(3):277-88.https://doi.org/10.1016/S0167-4889(97)00140-7

GLOBOCAN. Cancer incidence and mortality, 2020. Available via https://www.uicc.org/news/globocan-2020-new-global-cancer-data (Accessed 19 April 2021).

Gökçe K, ??kan N, Türker N, Ka?it M. Evaluating combined effect of naringin and salicylic acid on colon cancer cell culture. Turkish Med Stud J, 2017; 4:17-24https://doi.org/10.4274/tmsj.2017.04.02.0001

Herrera A, Castrillón W, Otero E, Ruiz E, Carda M, Agut R. Synthesis and antiproliferative activity of 3- and 7-styrylcoumarins. Med Chem Res, 2018; 27:1893-905.https://doi.org/10.1007/s00044-018-2202-0

Herrera-R A, Castrillón W, Pastrana M, Yepes AF, Cardona-G W. Promising hybrids derived from S-allylcysteine and NSAIDs fragments against colorectal cancer: synthesis, in-vitro evaluation, drug-likeness and in-silico ADME/tox studies. Iran J Pharm Res, 2021; (3):351-67.

Høydahl Ø, Edna TH, Xanthoulis A, Lydersen S, Endreseth BH. Long-term trends in colorectal cancer: incidence, localization, and presentation. BMC Cancer, 2020; 20(1):1077-90.https://doi.org/10.1186/s12885-020-07582-x

Huff SE, Mohammed FA, Yang M, Agrawal P, Pink J, Harris ME. Structure-guided synthesis and mechanistic studies reveal sweetspots on naphthyl salicyl hydrazone scaffold as non-nucleosidic competitive. J Med Chem, 2018; 61(3):666-80.https://doi.org/10.1021/acs.jmedchem.7b00530

Iliev I, Kontrec D, Detcheva R, Georgieva M, Balacheva A, Galic N. Cancer cell growth inhibition by aroylhydrazone. Biotechnol. Biotec. Equip, 2019; 33(1):756-63.https://doi.org/10.1080/13102818.2019.1608302

Jongstra J, Kotra LP, Wrana JL, inventors. Salicylic acid hydrazones as inhibitors of the erk mapkinase for the treatment of cancer. WO 2006136008 A1, 2006, December 28.

Lieu EL, Nguyen T, Rhyne S. Amino acids in cancer. Exp Mol Med, 2020; 52:15-30.https://doi.org/10.1038/s12276-020-0375-3

Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery. Adv Drug Deliv Rev, 1997; 23:3-25.https://doi.org/10.1016/S0169-409X(96)00423-1

Long L, Yang W, Liu L, Tobias DK, Katagiri R, Wu K. Dietary intake of branched-chain amino acids and survival after colorectal cancer diagnosis. Int J Cancer, 2020; 148(10):2471-80.https://doi.org/10.1002/ijc.33449

Misko TA, Liu YT, Harris ME, Oleinick NL, Pink J, Lee HY. Structure-guided design of anti-cancer ribonucleotide reductase inhibitors. J Enzyme Inhib Med Chem, 2019; 34(1):438-50.https://doi.org/10.1080/14756366.2018.1545226

Murata K, Moriyama M. Isoleucine, an essential amino acid, prevents liver metastases of colon cancer by antiangiogenesis. Cancer Res, 2007; 67(7):3263-8.https://doi.org/10.1158/0008-5472.CAN-06-3739

Paterson JR, Lawrence JR. Salicylic acid: a link between aspirin, diet and the prevention of colorectal cancer. QJM, 2001; 94(8):445-48.https://doi.org/10.1093/qjmed/94.8.445

Pathi S, Jutooru I, Chadalapaka G, Nair V, Lee SO, Safe S. Aspirin inhibits colon cancer cell and tumor growth and downregulates specificity protein (Sp) transcription factors. PLoS One, 2012; 7(10):e48208-17.https://doi.org/10.1371/journal.pone.0048208

Patrignani P, Patrono C. Aspirin and cancer. J Am Coll Cardiol, 2016; 68(9):967-76.https://doi.org/10.1016/j.jacc.2016.05.083

Peleg II, Maibach HT, Brown SH, Wilcox CM. Aspirin and nonsteroidal anti-inflammatory drug use and the risk of colorectal cancer. Arch Intern Med, 1994; 154(4):394-99.https://doi.org/10.1001/archinte.1994.00420040050009

Perez J, Maldonado M, Rojano B, Alzate F, Saez J, Cardona W. Comparative Antioxidant, Antiproliferative and Apoptotic effects of Ilex laurina and Ilex paraguariensis on Colon Cancer Cells Trop J Pharm Res, 2014; 13(8):1279-86.https://doi.org/10.4314/tjpr.v13i8.12

Pham-The H, Cabrera-Pérez MÁ, Nam NH, Castillo-Garit JA, Rasulev B, Le-Thi-Thu H, Casañola-Martin GM. In Silico Assessment of ADME properties: advances in Caco-2 cell monolayer permeability modeling. Curr Top Med Chem, 2018; 18(26):2209-29.https://doi.org/10.2174/1568026619666181130140350

Press B, Di Grandi D. Permeability for intestinal absorption: Caco-2 assay and related issues. Curr Drug Metab, 2008; 9(9):893-900.https://doi.org/10.2174/138920008786485119

Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin, 2021; 71:7-33.https://doi.org/10.3322/caac.21654

Song S, Christova T, Perusini S, Alizadeh S, Bao RY, Miller BW. Wnt inhibitor screen reveals iron dependence of β-catenin signaling. Cancer Res, 2011; 71(24):7628-39.https://doi.org/10.1158/0008-5472.CAN-11-2745

Spiegel R. Aspirin as a painkiller in patients with a high cardiovascular risk profile. Nat Rev Cardiol, 2020; 17(9):609-19.https://doi.org/10.1038/s41569-020-0398-4

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021; 71:209-49.https://doi.org/10.3322/caac.21660

Thota S, Rodrigues DA, Pinheiro PSM, Lima LM, Fraga CAM. N-Acylhydrazones as drugs. Bioorg Med Chem Lett, 2018; 28(17):2797- 306.https://doi.org/10.1016/j.bmcl.2018.07.015

Thun MJ, Namboodiri MM, Heath CW Jr. Aspirin use and reduced risk of fatal colon cancer. N Engl J Med, 1991; 325(23):1593-96.https://doi.org/10.1056/NEJM199112053252301

Vergara S, Carda M, Agut R, Yepes LM, Robledo SM, Cardona W. Synthesis, antiprotozoal activity and cytotoxicity in U-937 macrophages of triclosan-hydrazone hybrids. Med Chem Res, 2017; 26:3262-73.https://doi.org/10.1007/s00044-017-2019-2

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):1829-52.https://doi.org/10.2174/092986707781058805

Wakshlag JJ, Kallfelz FA, Wakshlag RR, Davenport GM. The effects of branched-chain amino acids on canine neoplastic cell proliferation and death. J Nutr, 2006; 136(7):2007-10.https://doi.org/10.1093/jn/136.7.2007S

Wang LB, Zhang SZ, Ding KF, Zheng S. A study of the free amino acids uptake in Colon Cancer. Zhejiang Y, 1997; 19:208-9.

Zhivkova ZD. Studies on drug-human serum albumin binding: the current state of the matter. Curr Pharm Des, 2015; 21(14):1817-30.https://doi.org/10.2174/1381612821666150302113710

Article Metrics
144 Views 200 Downloads 344 Total

Year

Month

Related Search

By author names