Next Article in Journal
Chemical Composition, Antioxidant and Anti-Inflammatory Activities of Clary Sage and Coriander Essential Oils Produced on Polluted and Amended Soils-Phytomanagement Approach
Next Article in Special Issue
In Silico Structure-Guided Optimization and Molecular Simulation Studies of 3-Phenoxy-4-(3-trifluoromethylphenyl)pyridazines as Potent Phytoene Desaturase Inhibitors
Previous Article in Journal
Imidazopyridazine Acetylcholinesterase Inhibitors Display Potent Anti-Proliferative Effects in the Human Neuroblastoma Cell-Line, IMR-32
Previous Article in Special Issue
Molecular Modifications and Control of Processes to Facilitate the Synergistic Degradation of Polybrominated Diphenyl Ethers in Soil by Plants and Microorganisms Based on Queuing Scoring Method
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Crystal Structure and Bioactivity of Phenazine-1-carboxylic Acylhydrazone Derivatives

1
Research Center for Molecular Medical Engineering, Department of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, China
2
Department of Biology & Engineering, Guizhou Medical University, Guiyang 550004, China
3
Department of Chemistry, University of Hull, Cottingham Road, Hull HU6 7RX, UK
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(17), 5320; https://doi.org/10.3390/molecules26175320
Submission received: 26 July 2021 / Revised: 26 August 2021 / Accepted: 27 August 2021 / Published: 1 September 2021
(This article belongs to the Special Issue Molecular Structure Section in China)

Abstract

:
A phenazine-1-carboxylic acid intermediate was synthesized from the reaction of aniline and 2-bromo-3-nitro-benzoic acid. It was then esterified and reacted with hydrazine hydrate to afford phenazine-1-carboxylic hydrazine. Finally, 10 new hydrazone compounds 3a3j were obtained by the condensation reaction of phenazine-1-carboxylic acid hydrazide and the respective aldehyde-containing compound. The structures were characterized by 1H and 13C NMR spectroscopy, MS and single crystal X-ray diffraction. The antitumor activity of the target compounds in vitro (HeLa and A549) was determined by thiazolyl blue tetrazolium bromide. The results showed that compound (E)-N′-(2-hydroxy-4-(2-(piperidine-1-yl) ethoxy) benzyl) phenazine-1-carbonyl hydrazide 3d exhibited good cytotoxic activity.

1. Introduction

Acylhydrazone is formed by the condensation of hydrazine and an aldehyde or ketone, containing both oxygen and nitrogen atoms that are involved in the formation of hydrogen bonds in living organisms and increase the affinity between receptors, thereby, inhibiting numerous physiological and chemical processes in vivo [1,2,3,4,5]. Acylhydrazone compounds are special in terms of both their structure and properties and have been the focus of considerable research by chemists. The -CONH-N=CH- group has a large conjugate system, exhibits favorable stability and coordination ability [6,7,8], and can readily form chelates with iron/ferrous ions in the body. By changing the form of iron in this way, it prevents the absorption and utilization of iron by cells, thereby changing the distribution of iron in the body. In other words, it can regulate the distribution and metabolism of iron in the body. In particular, it can reduce cancer cell and tumor tissue iron content or can reduce the effective use of iron by cancer cells. It has the ability to make cancer cells and tissues iron “hungry” and can affect the activity of enzymes containing iron. In this way, the cancer cells′ energy metabolism, DNA replication, and transcription as well as protein synthesis and folding can be retarded or inhibited. The importance is highlighted because the division and growth can lead to the death [9,10,11,12,13]. Therefore, many hydrazone compounds are potential anticancer drugs because of their good anticancer activity [14].
Phenazine-1-carboxylic acid (PCA) is an antibiotic that can be used to protect crops against a broad spectrum of soil-borne fungal Pseudomonas spp. [15]. It is widely found in the metabolites of many microorganisms, such as streptomyces [16] and Pseudomonads [17]. PCA can be isolated from the fermentation broth of Pseudomonas M-18 [18]. It was found to have antituberculous bacterial properties and can inhibit seaweed growth [19,20]. PCA was confirmed by the Chinese pesticide naming unit, named as Shenazinamycin, which is an antibiotic secreted by Pseudomonas fluoresceae through biological culture. It has the dual functions of a broad-spectrum of inhibition of plant pathogens and the promotion of plant growth [21]. Rewcastle et al. synthesized a series of PCA analogs on the basis of original natural products and developed lead compounds possessing antilung cancer [22] and leukemia [23] properties and is currently in Phase II clinical trials [24]. PCA and its analogs isolated from secondary metabolites of Streptomyces sp. IFM 11694 can overcome the resistance of human gastric adenocarcinoma cells to TRAIL, a tumor necrosis factor related apoptotic inducing ligand, showing good antitumor activity [25]. However, the synthesis and bioactivity of PCA based acylhydrazones have not yet been reported. Therefore, we first synthesized PCA by a chemical synthesis method and then synthesized 10 new hydrazone compounds 3a3j (Scheme 1) based on PCA. We also investigated the antitumor activity of the target compounds in vitro. Chemical synthesis of phenazine natural products by means of structural modification or new synthesis of phenazine products are also emerging. The antitumor activity and structure activity relationship are also attracting increasing attention, and the mechanism of action may be related to interfering with respiratory chain, inhibiting topoisomerase activity and DNA breaking [26,27].

2. Results and Discussion

2.1. Synthesis and Crystal Structures of the Target Compounds

Phenazine biosynthesis has attracted attention since its early studies in the second half of the 20th century [28]. Ever since the total synthesis of PCA was reported by Kogl and Postowsky [29] in 1930, new reactions and synthetic strategies have emerged over the following half century, and the yield and route have been continuously improved and optimized. The synthetic route to the target compounds 3a3j is shown in Scheme 1, and the structure and yields are shown in Table 1. According to reference [30], PCA was synthesized without the need for column chromatography during the synthesis process. Instead, relatively pure PCA was obtained by recrystallization from acetonitrile and ethanol (1:1). Then, PCA was esterified in concentrated sulfuric acid, and reacted with hydrazine hydrate to form acylhydrazine. Finally, the acylhydrazine compounds were condensed with aldehyde-containing compounds to afford the target compounds 3a3j. 3a3j were dissolved in the mixed solvent system of chloroform and ethanol; the ratio of chloroform to ethanol is 1:1. Heating with frequent agitation, boiling to effect solution, and then slowing evaporation at room temperature resulted in 9 sets of crystals suitable for X-ray diffraction. The single crystal X-ray diffraction was performed, and the crystal structures revealed that the aldehyde compounds had reacted successfully with the amino group of the hydrazide. In all crystal structures, the hydrogen on the amide nitrogen atom and the nitrogen atom on the phenazine ring formed intramolecular N-H...N hydrogen bonds. The aldehydes containing ortho hydroxyl groups reacted with acylhydrazine to afford acylhydrazone compounds. Hydrogen atoms of the ortho hydroxyl groups and the imine nitrogen atoms also formed O-H...N intramolecular hydrogen bonds; thus, both the hydroxyl and carbonyl groups are on the same side, but on the other side of the phenazine ring (See Figure 1 for an example of the crystal structure of 3b; hydrogen bond distances are shown in Table S4).

2.2. Anticancer Activity of 3a3f in Hela and A549 Cell Lines

In our study, results are compared with cisplatin (20 μmol/L). Hela and A549 cells were seeded onto 96-well plates at the proper density and treated with 3a3j at final concentrations for 24 h; cell viability was analyzed via an MTT assay. There was a pool of three different sets of experiments, each repeated in triplicate. Statistical comparisons were conducted. The results are shown in Table 2.
The activity showed that the compounds 3b3f were effective in the Hela and A549 cell lines, among which 3d had the strongest effect on Hela and A549 cells. Compound 3d revealed better cytotoxic activity than cisplatin, with IC50 values of 5.5 ± 1.3 and 2.8 ± 1.53 μmol/L against HeLa and A549, respectively, with statistical significance (*** p < 0.001, significantly different from the value of control group, n = 3).
The antitumor activity of phenazine compounds has been widely studied, and can induce cell apoptosis and cycle arrest. The mechanism of action is related to the inhibition of topoisomerase and the induction of reactive oxygen species. DNA, which is regarded as an important genetic material of cell life, has become the target of many antitumor drugs. Phenazine compounds have planar macromolecular structures for intramolecular hydrogen bonding. The shorter the hydrogen bond distance, the more stable the structure. It may bind to DNA in a way similar to ethidium bromide and campinetin [31].
According to previous reports, compounds that bear hydrazide-hydrazone moiety have received considerable attention due to their biological value in the development of novel antimicrobials and analgesic, anti-inflammatory, and anticancer agents [32]. The compound PAC-1 (Figure 2) was reported to enhance the activity of procaspase-3 in vitro and induce apoptosis in tumor cells in vivo [33]. The cells were treated with different concentrations (0, 0.1, 1, 10, 100, and 1000 μmol/L) of synthesized compounds. Picolinoylhydrazone derivative (1) (Figure 3) is a potent growth inhibitor of lung, leukemia and ovarian cancer cell lines, with IC50 values below 10 μmol/L [34]. A bis-isatin hydrazones of imidazolidine-2, 4-dione (2) (Figure 4) showed good antiproliferative activity against the human colon cancer cell line HCT-116.
A series of novel pyrano[3,2-a]phenazine derivatives (Figure 5), designed as hybrid molecules of phenazine and pyran pharmocophores and then treated with compounds at concentrations from 0 to 50 μmol/L. Cytotoxic evaluation indicates that many compounds exhibited cytotoxicity against HCT116, MCF7, HepG2 and A549 cancer cell lines in vitro, in which compounds 1c, 1i, 2e, and 2l were found to have excellent antiproliferative activity against the HepG2 cancer cell line [35]. This provided the experimental basis for the further mechanism of 3a3j and its influence on tumor cells.
Compared with the above structures, 3a3j all have a hydrazine structure and IC50 values of 3d below 10 μmol/L. Significantly, 3d showed more potency than the positive control drug, which is a potent growth inhibitor of HeLa and A549 cells. It can be seen from Table 2 that the hydrazine obtained by the condensation reaction of phenazine-1-hydrazine with the salicylaldehyde and salicylaldehyde analogs have certain antitumor activity, while the hydrazine obtained by the condensation reaction of other aldehydes with phenazine-1-hydrazine has no activity. The specific antitumor activity mechanism, whether by inhibiting enzyme activity or not, needs further study.

3. Materials and Methods

3.1. Instruments and Reagents

1H/13C NMR spectra were measured on a Inova-400-Bruker AV600 NMR spectrometer, with d-DMSO as solvent and tetramethylsilane as internal reference (600 MHz), and the J value was in Hz. High resolution mass spectrometry (HRMS) was performed using a GCT premier CAB048 mass spectrometer. Data for the crystal structures were collected on a Bruker Smart Apex single crystal diffractometer. Melting points were measured on a Yanaco microscopic melting point meter, and the temperature was not corrected. Hela and A549 cells were replaced in a fresh culture medium and incubated for 24 h in a humidified atmosphere of 95% air and 5% CO2 at 37 °C (Thermo-Fisher, New York, NY, USA). MTT assay was analyzed by Epoch Continuous Wavelength Microplate (Bio-Tek, Burlington, VT, USA) and RPMI-1640 medium (Gibco, New York, NY, USA). Solvents and reagents were obtained commercially and were of analytical grade.

3.2. Determination of the Crystal Structures of the Compounds

The target compounds were dissolved in chloroform and slowly evaporated for 5–10 days to obtain crystals of appropriate size. A Bruker Smart Apex CCD single crystal diffractometer and a graphite monochromator with monochromatized Mo rays (λ = 0.071073 nm) were used to collect single crystal diffraction data within a certain θ range by φ-ω scanning mode. The diffraction intensity data were corrected by empirical absorption and LP. The crystal structures were solved by direct methods. All non-hydrogen coordinates and their anisotropic thermal parameters were modified by the full matrix least square method. All calculations were completed with the Shelx-97 program [36]. Crystallographic data and refinement details for 3a3j are given in Tables S1–S3. CCDC: 2102942, 3a. CCDC: 2102943, 3b. CCDC: 2102944, 3c. CCDC: 2102945, 3e. CCDC: 2102946, 3f. CCDC: 2102947, 3g. CCDC: 2102948, 3h. CCDC: 2102949, 3i. CCDC: 2102950, 3j. These data can be obtained free of charge from the Cambridge Crystallographic Data Centre www.ccdc.cam.ac.uk/data_request/cif (accessed on 13 August 2021).

3.3. Synthesis of Compounds

3.3.1. Synthesis of PCA

Aniline (2.5 g, 26.8 mmol), 2-bromo-3-nitrobenzoic acid (6.3 g, 26.6 mmol), CuI (1.3 g, 6.8 mmol) and triethylamine (12.5 mL, 90.0 mmol) were weighed out and combined with glycol (50.0 mL, 899.1 mmol), dissolved in a 100 mL three-mouth flask and the system heated at 95 °C for 3 h. On cooling to room temperature, it was poured into a 0.2 mol/L sodium hydroxide (300 mL, 60 mmol) solution. Work-up included silica gel adsorption decolorization and extraction and filtration to afford a red solution, which was then acidified with dilute hydrochloric acid to pH = 3, affording the product 3-nitro-2-(phenylamino) benzoic acid. This product was pure enough to be directly used in the next reaction.
The solution of 3-nitro-2-(phenylamino) benzoic acid (4.0 g, 15.5 mmol) and NaBH4 (4.8 g, 126.0 mmol) was dissolved in 2 mol/L sodium hydroxide (450 mL, 900 mmol), refluxed for 4 h, and then the sodium salt solution of phenazine carboxylic acid was obtained after cooling. A green solid was obtained by adjusting the pH of the solution with dilute hydrochloric acid to pH = 3. The solid was recrystallized with a mixture of acetonitrile and ethanol (1:1) to obtain yellow-green crystalline PCA.

3.3.2. Synthesis of Phenazine-1-hydrazide

Concentrated sulfuric acid (1 mL) was slowly added to 100 mL of anhydrous ethanol in an ice bath, restored to room temperature, and then PCA (2.24 g, 10 mmol) was added to the solution, which was refluxed for 21 h. Then the solution was cooled to room temperature. The concentrated solution was steamed to 50 mL, and the solution was poured into 200 mL of ice water. With saturated potash, the pH was adjusted to pH = 10, and then extraction with dichloromethane (60 mL × 4) was conducted. The organic phase was collected and combined, and then dried overnight with anhydrous sodium sulfate and filtered and rotary steamed to remove the dichloromethane to afford a black solution. Then, 40 mL anhydrous ethanol and 5 mL hydrazine hydrate were added to the black solution, and the system was refluxed for 8 h. After cooling, the solids were separated out and filtered, and phenazine-1-hydrazine was obtained after washing with anhydrous ethanol for three times and drying.

3.3.3. Synthesis of the Acylhydrazone Compounds

Ten aldehydes were weighed out, and each was dissolved in 30 mL anhydrous ethanol with phenazine-1-hydrazine at a molar ratio of 1:1. Following refluxing for 8 h and upon cooling, a solid separated out, and filtration followed by ethanol washing for 3 times to obtain a brown solid. The solid was recrystallized with chloroform and ethanol and filtered and dried to obtain the acylhydrazone compounds.
(E)-N′-(isoquinoline-3-methyl)phenazine-1-carbonyl hydrazide (3a): brown solid 0.23 g, yield 60%, m. p. 256–257 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.28 (s, 1H, NH), δ 8.72 (s, 1H, isoquinoline-H), δ 8.60 (d, J = 6.0 Hz, 1H, quinoline-H), 8.53 (d, J = 12.0 Hz, 1H, isoquinoxaline-H), 8.5. (d, J = 6.0 Hz, 1H, quinoline-H), 8.48 (d, J = 6.0 Hz, 1H, isoquinoxaline-H), 8.34 (t, J = 12.0 Hz, 1H, isoquinoline-H), 8.23 (s, 1H, N=CH), 8.14–8.08 (d, J = 6.0 Hz, 2H, quinoline-H), 8.7–8.04 (m, 2H, quinoline-H), 7.92–7.82 (m, 2H, isoquinoline-H), 7.68 (d, J = 12.0 Hz, 1H, isoquinoline-H). 13C NMR (150 MHz, d-DMSO): δC (ppm): 162.0, 153.7, 148.4, 147.3, 142.8, 142.5, 141.6, 139.8, 136.7, 133.3, 132.8, 132.0, 131.8, 131.5, 130.3, 130.0, 129.6, 129.1, 128.8, 128.0, 127.9, 127.3, 117.6. HRMS (ESI+) m/z Calcd for C23H15N5O [M + H]+ 378.1322, found: 378.1352.
(E)-N′-(2-hydroxybenzyl)phenazine-1-carbonyl hydrazide (3b): brownish yellow powder 0.22 g, yield 65%, m. p. 270–271 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.28 (s, 1H, NH), 11.36 (s, 1H, OH), 8.82 (s, 1H, N=CH), 8.63 (d, J = 6.0, 1H, quinoxaline-H), 8.57–8.47 (d, J = 12.0 Hz, 2H, quinoxaline-H), 8.34 (d, J = 12.0 Hz, 1H, quinoxaline-H) 8.16–8.06 (m, 3H, quinoxaline-H), 7.64 (d, J = 6.0 Hz, 1H, benzylidenimin-H), 7.36 (t, J = 6.0 Hz, 1H, benzylidenimin-H), 7.00 (d, J = 6.0 Hz, 1H, benzylidenimin-H), 6.99 (t, J = 6.0 Hz, 1H, benzylidenimin-H). 13C NMR (150 MHz, d-DMSO): δC (ppm): 161.3, 157.5, 149.3, 142.8, 142.5, 141.5, 139.7, 133.6, 132.9, 132.0, 131.8, 131.5, 130.7, 130.3, 129.8, 129.4, 129.2, 119.3, 118.5, 116.4. HRMS (ESI+) m/z Calcd for C20H14N4O2 [M + H]+ 343.1211, found: 343.1195.
(E)-N′-(3,5-dibromo-2-hydroxybenzy)phenazine-1-carbonyl hydrazide (3c): yellow powder 0.32 g, yield 65%, m. p. 269–270 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.70 (s, 1H, NH), 12.64 (s, 1H, OH), 8.80 (s, 1H, N=CH), 8.67 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.54 (d, J = 12.0 Hz, 2H, quinoxaline-H), 8.36 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.18–8.06 (m, 3H, quinoxaline-H), 7.88 (s, 2H, benzylidenimin-H). Due to the poor solubility of compound 3c, we could not obtain 13C-spectral data, HRMS (ESI+) m/z Calcd for C20H12Br2N4O2 [M + H]+ 498.9310, found: 498.9400.
(E)-N′-(2-hydroxy-4-(2-(piperidine-1-yl)ethoxy)benzyl) phenazine-1-carbonyl hydrazide (3d): orange yellow powder 0.26 g, yield was 55%, m. p. 203–204 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.24 (s, 1H, NH), 11.66 (s, 1H, OH), 8.73 (s, 1H, N=CH), 8.63 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.55–8.45 (d, J = 6.0 Hz, 2H, quinoxaline-CH), 8.34 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.18–7.92 (m, 3H, quinoxaline-H), 7.50 (s, 1H, benzylidenimin-H), 6.58 (s, 1H, benzylidenimin-H), 6.54 (d, J = 6.0 Hz, 1H, benzylidenimin-H), 4.12–2.62 (t, J = 6.0 Hz, 4H, methylene-H), 2,48–1.39 (m, 10H, piperidine-H). 13C NMR (150 MHz, d-DMSO): δC (ppm): 161.4, 160.8, 159.5, 149.8, 142.7, 142.6, 141.4, 139.7, 133.6, 132.9, 132.0, 131.8, 131.4, 130.5, 130.3, 129.4, 129.1, 111.5, 107.6, 106.9, 101.6, 101.2, 65.7, 57.2, 54.3, 25.5, 223.8. HR-MS (ESI+) m/z Calcd for C27H27N5O3 [M]+ 470.2102, found: 470.2194.
(E)-N′-(2-hydroxy-3-methoxybenzyl)phenazine-1-carbonyl hydrazide (3e): brown solid 0.25 g, yield 68%, m. p. 190–191 °C; 1H NMR(600 MHz, d-DMSO): δH (ppm): 13.24 (s, 1H, OH), 11.10 (s, 1H, NH), 8.81 (s, 1H, N=CH), 8.63–8.26 (d, J = 12.0 Hz, 4H, quinoxaline-H), 8.19–7.92 (m, 3H, quinoxaline-H), 7.25–7.08 (d, J = 12.0 Hz, 2H, benzylidenimin-H), 6.92 (t, J = 12.0 Hz, 1H, benzylidenimin-CH), 3.85 (s, 3H, OCH3). 13C NMR (150 MHz, CHCl3): δC (ppm): 161.2, 149.1, 147.8, 147.2, 142.7, 142.5, 141.4, 139.7, 133.6, 132.9, 132.0, 131.7, 130.6, 129.4, 129.1, 121.1, 118.9, 118.6, 113.9, 79.0, 55.7. HRMS (ESI+) m/z Calcd for C21H16N4O3 [M + H]+ 373.1102, found: 373.1298.
(E)-N′-(2,4-dihydroxybenzyl)phenazine-1-carbon hydrazide (3f): yellow-brown powder 0.25 g, the yield of 70%, m. p. 297–298 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.18 (s, 1H, OH), 11.54 (s, 1H, OH), 10.04 (s, 1H, NH), 8.67 (s, 1H, N=CH), 8.61 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.50-8.27 (d, J = 12.0 Hz, 3H, quinoxaline-H), 8.09-8.02 (m, 3H, quinoxaline-H), 7.40 (d, J = 6.0 Hz, 1H, benzylidenimin-H), 6.42 (s, 1H, benzylidenimin-H), 6.37 (d, J = 6 Hz, 1H, benzylidenimin-H). 13C NMR (150 MHz, d-DMSO): δC (ppm):160.8, 160.6, 159.6, 150.1, 142.7, 142.5, 141.2, 139.6, 133.7, 132.9, 132.0, 131.7, 131.7, 130.3, 130.3, 129.3, 129.1, 110.4, 107.7, 102.6. HRMS (ESI+) m/z Calcd for C20H14N4O3 [M + H]+ 359.1102, found: 359.1140.
(E)-N′-(pyridine-2-methyl)phenazine-1-carbonyl hydrazide (3g): reddish-brown crystalline 0.20 g, yield 60%, m. p. 220–221 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.10 (s, 1H, NH), 8.67 (d, J = 6.0 Hz, 1H, 2-pyridine-H), 8.56–8.49 (d, J = 6.0 Hz, 2H, quinoxaline-H), 8.48 (s, 1H, N=CH), 8.33 (t, J = 12.0 Hz, 1H, quinoxaline-H), 8.12 (d, J = 6.0 Hz, 1H, 2-pyridine-H), 8.07 (t, J = 6.0 Hz, 1H, quinoxaline-H), 8.06–8.02 (m, 2H, quinoxaline-H), 7.95 (t, J = 12.0 Hz, 2H, 2-pyridine-H ), 7.47 (s, 1H, N=CH). 13C NMR (150 MHz, d-DMSO): δC (ppm): 161.9, 153.1, 149.5, 148.4, 142.8, 142.5, 141.6, 139.8, 136.8, 133.0, 132.7, 132.0, 131.8, 131.7, 130.3, 129.5, 129.1, 124.5, 120.7. HRMS (ESI+) m/z Calcd for C19H13N5O [M + H]+ 328.1102, found: 328.1196.
(E)-N′-(pyridine-3-methyl)phenazine-1-carbonyl hydrazide (3h): yellowish brown crystal 0.20 g, yield 60%, m. p. 230–231 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.37 (s, 1H, NH), 8.97 (s, 1H, 3-pyridine-H), 8.67 (s, 1H, N=CH), 8.64 (d, J = 6.0 Hz, 1H, 3-pyridine-H), 8.53 (d, J = 6.0 Hz, 1H, 3-pyridine-H), 8.48 (d, J = 6.0 Hz, 1H, quinoxaline-H) 8.34–8.24 (d, J = 6.0 Hz, 2H, quinoxaline-H), 8.11 (d, J = 6.0 Hz, 1H, 3-pyridine-H), 8.11–8.03 (m, 3H, quinoxaline-H), 7.55 (t, J = 6.0 Hz, 1H, 3-pyridine-H). 13C NMR (150 MHz, d-DMSO): δC (ppm): 161,4, 150.7, 148.8, 145.8, 142.7, 142.6, 141.3, 139.7, 133.7, 133.5, 133.0, 132.0, 131.8, 130.7, 130.3, 130.2, 129.4, 129.2, 124.0. HRMS (ESI+) m/z Calcd for C19H13N5O [M + H]+ 328.1102, found: 328.1200.
(E)-N′-(pyridine-4-methyl)phenazine-1-carbonyl hydrazide (3i): yellow powder 0.20 g, yield 60%, m. p. 243–244 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.42 (s, 1H, NH), 8.72 (d, J = 6.0 Hz, 2H, 4-pyridine-H), 8.62 (s, 1H, N=CH), 8.57–8.47 (d, J = 6.0 Hz, 2H, quinoxaline-H), 8.37–8.32 (d, 2H, quinoxaline-H), 8.14–8.05 (m, 3H, quinoxaline-H), 7.78 (d, J = 6.0 Hz, 2H, 4-pyridine-H). 13 C NMR (150 MHz, CHCl3): δC (ppm): 161.3, 151.1, 148.5, 143.4, 141.0, 140.2, 136.6, 134.6, 132.3, 131.4, 130.2, 130.0, 128.6, 127.8, 122.6, 119.0, 118.0, 114.1, 56.3. HRMS (ESI+) m/z Calcd for C19H13N5O [M + H]+ 328.1102, found: 328.1198.
(E)-N′-((2-hydroxynaphthalene-1-yl)methyl)phenazine-1-carbonyl hydrazide (3j): orange powder 0.24 g, yield 62%, m. p. 276–277 °C; 1H NMR (600 MHz, d-DMSO): δH (ppm): 13.45 (s, 1H, OH), 13.01 (s, 1H, NH), 9.67 (s, 1H, N=CH), 8.72 (d, J = 6.0 Hz, 1H, quinoxaline-H), 8.69 (d, J = 6.0 Hz, 1H, 1-naphthalene-H), 8.52–8.32 (d, J = 12 Hz, 3H, quinoxaline-H), 8.16–8.09 (m, 3H, quinoxaline-H), 7.98–7.91 (d, J = 12.0 Hz, 2H, 1-naphthalene-H), 7.65–7.43 (t, J = 6.0 Hz, 2H, 1-naphthalene-H), 7.28 (d, J = 6.0 Hz, 1-naphthalene-H). 13C NMR (150 MHz, d-DMSO): δC (ppm): 161.0, 158.2, 147.7, 142.6, 141.4, 139.7, 134.1, 133.3, 132.8, 132.1, 131.8, 131.7, 130.3, 130.0, 129.8, 129.1, 128.8, 127.7, 127.6, 123.5, 121.2, 118.9, 108.6,79.1. HRMS (ESI+) m/z Calcd for C24H16N4O2 [M + H]+ 393.1112, found: 393.1350.

3.4. Antitumor Activity Assays

Using cisplatin as a positive control, the in vitro antitumor activity of the target compounds was determined by MTT assays [8]. A549 cells were cultured in RPMI 1640 medium at 37 °C with 5% CO2 and 95% air, supplemented with 10% (v/v) bovine calf serum and 80 μg/mL gentamicin. Hela cells were cultured in DMEM (high glucose) medium under the same condition as above. The logarithmic growth phase cells were inoculated in a 96-well culture plate at a concentration containing the medium at the cell density is 5 × 103 cells per well. After 24 h of culture, the cells were attached to the well, and the old culture medium was discarded. The compounds to be tested were dissolved in DMSO, and the final concentration was diluted in the culture medium. The complete culture medium containing different concentrations of compounds were replaced, six concentration gradients were set (2.5 to 100 μmol/L). Each concentration had 3 complex holes, the zeroing group containing only the culture medium. After treatment for 48 h, 20 μL MTT (5 mg/mL) was added to each well, and the culture was continued for 4 h. The medium was centrifuged at 3000 rpm for 10 min, and 150 μL DMSO was added. The absorbance was measured at 570 nm by a microplate analyzer, and the IC50 value was calculated.

3.5. Statistical Analysis

Data were presented as from at least three independent experiments. Results were reported as mean ± SD and analyzed by Student’s t test. Results were considered significant when a p value < 0.05 was obtained.

4. Conclusions

PCA intermediates were synthesized from aniline and 2-bromo-3-nitrobenzoic acid. Then, PCA was esterified, and acylhydrazide was synthesized. Finally, 10 new hydrazine-1-carboxylic acid compounds containing aldehyde groups were obtained by the condensation reaction of PCA with hydrazine-containing compounds. Their structures were characterized by 1H and 13C NMR spectroscopy, MS and single crystal X-ray diffraction. The in vitro antitumor activity of the target compounds (HeLa and A549) was tested by MTT assays. The results showed that the compounds exhibited good cytotoxic activity, particularly 3d, which has potential to be used for further studies.

Supplementary Materials

The following are available online: Figures S1–S10: 1H NMR of compound 3a to 3j. Figure S11–S19: 13C NMR of compound 3a to 3j. Figures S20–S29: HRMS of compound 3a to 3j. Table S1: Crystal data and structure refinement for the compound 3a3c. Table S2: Crystal data and structure refinement for the compound 3e3g. Table S3: Crystal data and structure refinement for the compound 3h3k. Table S4: Hydrogen bond parameters [Å and °] in the crystal structure of compounds 3a3j.

Author Contributions

S.W., X.L., F.L. and H.L. conducted the experiments; H.X., X.Y., N.W. and Y.H. analyzed the experiment data; L.S. analyzed the X-ray structure; C.R. and Q.Z. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by and the National Natural Science Foundation of China (22065009, 22066007) and the Guizhou Provincial Natural Science Foundation (grant number [2019] 2792, grant number 19NSP042).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of extracts are available from the authors.

References

  1. Huff, S.E.; Mohammed, F.A.; Yang, M.; Agrawal, P.; Pink, J.; Harris, M.E.; Dealwis, C.G.; Viswanathan, R. Structure-Guided synthesis and mechanistic studies reveal sweetspots on naphthyl salicyl hydrazone scaffold as non-Nucleosidic competitive, reversible inhibitors of human ribonucleotide reductase. J. Med. Chem. 2018, 61, 666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shareef, M.A.; Devi, G.P.; Routhu, S.R.; Kumar, C.G.; Kamal, A.; Babu, B.N. New imidazo[2,1-b]thiazole-based aryl hydrazones: Unravelling their synthesis and antiproliferative and apoptosis-inducing potential. RSC Med. Chem. 2020, 11, 1178. [Google Scholar] [CrossRef] [PubMed]
  3. Reznichenko, O.; Cucchiarini, A.; Gabelica, V.; Granzhan, A. Quadruplex DNA-guided ligand selection from dynamic combinatorial libraries of acylhydrazones. Org. Biomol. Chem. 2021, 19, 379. [Google Scholar] [CrossRef]
  4. Xi, W.; Song, F.Q.; Xia, X.L.; Song, X.Q. Tuned structure and DNA binding properties of metal complexes based on a new 4-acylpyrazolone derivative. New J. Chem. 2020, 44, 2281. [Google Scholar] [CrossRef]
  5. Boechat, N.; Ferreira, V.F.; Ferreira, S.B.; Ferreira, M.L.G.; Silva, F.C.; Bastos, M.M.; Costa, M.S.; Lourenc, M.C.S.; Pinto, A.C.; Krettli, A.U.; et al. Novel 1,2,3-Triazole Derivatives for Use against Mycobacterium tuberculosis H37Rv (ATCC 27294) Strain. J. Med. Chem. 2011, 54, 5988. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, X.Q.; Cai, Y.D.; Jiang, W.; Guo, P.; Fang, J.K.; Liu, J.L.; Tong, M.L.; Bao, X. A Multi-Stimuli-Responsive Fe(II) SCO Complex Based on an Acylhydrazone Ligand. Inorg. Chem. 2019, 58, 999–1002. [Google Scholar] [CrossRef]
  7. Ong, Y.C.; Roy, S.; Andrews, P.C.; Gasser, G. Metal Compounds against Neglected Tropical Diseases. Chem. Rev. 2019, 119, 730. [Google Scholar] [CrossRef]
  8. Li, B.; He, T.; Fan, Y.Q.; Yuan, X.C.; Qiu, H.Y.; Yin, S.C. Recent developments in the construction of metallacycle/metallacage-cored supramolecular polymers via hierarchical self-assembly. Chem. Commun. 2019, 55, 8036. [Google Scholar]
  9. Richardson, D.R.; Kalinowski, D.S.; Richardson, V.; Sharpe, P.C.; Lovejoy, D.B.; Islam, M.; Bernhardt, P.V. 2-Acetylpyridine Thiosemicarbazones are Potent Iron Chelators and Antiproliferative Agents: Redox Activity, Iron Complexation and Characterization of their Antitumor Activity. J. Med. Chem. 2009, 52, 1459–1470. [Google Scholar] [CrossRef]
  10. Stefani, C.; Jansson, P.J.; Gutierrez, E.; Bernhardt, P.V.; Richardson, D.R.; Kalinowski, D.S. Alkyl Substituted 2′-Benzoylpyridine Thiosemicarbazone Chelators with Potent and Selective Anti-Neoplastic Activity: Novel Ligands that Limit Methemoglobin Formation. J. Med. Chem. 2013, 56, 357–370. [Google Scholar] [CrossRef]
  11. Stefani, C.; Moorthy, G.P.; Lovejoy, D.B.; Jansson, P.J.; Kalinowski, D.S.; Sharpe, P.C.; Bernhardt, P.V.; Richardson, D.R. Halogenated 2′-benzoylpyridine thiosemicarbazone (XBpT) chelators with potent and selective anti-neoplastic activity: Relationship to intracellular redox activity. J. Med. Chem. 2011, 54, 6936–6948. [Google Scholar] [CrossRef]
  12. Kalinowski, D.S.; Yu, Y.; Sharpe, P.C.; Islam, M.; Liao, Y.T.; Lovejoy, D.B.; Kumar, N.; Bernhardt, P.V.; Richardson, D.R. Design, Synthesis, and Characterization of Novel Iron Chelators: Structure−Activity Relationships of the 2-Benzoylpyridine Thiosemicarbazone Series and Their 3-Nitrobenzoyl Analogues as Potent Antitumor Agents. J. Med. Chem. 2007, 50, 3716–3729. [Google Scholar] [CrossRef]
  13. Hamperl, S.; Bocek, M.J.; Saldivar, J.C.; Swigut, T.; Cimprich, K.A. Transcription-replication conflict orientation modulates R-loop levels and activates distinct DNA damage responses. Cell 2017, 170, 774–786.e19. [Google Scholar] [CrossRef] [Green Version]
  14. Onnis, V.; Demurtas, M.; Deplano, A.; Balboni, G.; Baldisserotto, A.; Manfredini, S.; Pacifico, S.; Liekens, S.; Balzarini, J. Design, Synthesis and Evaluation of Antiproliferative Activity of New Benzimidazolehydrazones. Molecules 2016, 21, 579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ligon, J.M.; Hill, D.S.; Hammer, P.E.; Torkewitz, N.R.; Hofmann, D.; Kempf, H.S.; Van Pee, K.H. Natural products with antifungal activity from Pseudomonas biocontrol bacteria. Pest Manag. Sci. 2000, 56, 688–695. [Google Scholar] [CrossRef]
  16. Abdelfattah, M.; Toume, K.; Ishibashi, M. Isolation and structure elucidation of izuminosides A–C: A rare phenazine glycosides from Streptomyces sp. IFM 11260. J. Antibiot. 2011, 64, 271–275. [Google Scholar] [CrossRef] [PubMed]
  17. Jasim, B.; Anisha, C.; Rohini, S.; Kurian, J.M.; Jyothis, M.; Radhakrishnan, E.K. Phenazine carboxylic acid production and rhizome protective effect of endophytic Pseudomonas aeruginosa isolated from Zingiber officinale. World J. Microbiol. Biotechnol. 2014, 30, 1649–1654. [Google Scholar] [CrossRef]
  18. Hu, H.B.; Xu, Y.Q.; Cheng, F.; Zhang, X.H.; Hur, B.K. Isolation and characterization of a new Pseudomonas strain produced both phenazine 1-carboxylic acid and pyoluteorin. J. Microbiol. Biotechnol. 2005, 15, 86–90. [Google Scholar]
  19. Yamashita, S.; Sawazaki, T.; Kawasaki, M.; Nakamura, G.; Anzai, K.; Isono, K. A new antiyeast substance, cerevioccidin, produced by streptomycete. J. Antibiot. 1955, 8, 42–43. [Google Scholar]
  20. Nelson, C.D.; Toohey, J.I. Method of Controlling the Growth of Noxious Plants. U.S. Patent US3367765A, 6 February 1968. [Google Scholar]
  21. Hane, M.; Wijaya, H.C.; Nyon, Y.A.; Sakihama, Y.; Hashimoto, M.; Matsuura, H.; Hashidoko, Y. Phenazine-1-carboxylic acid (PCA) produced by Paraburkholderia phenazinium CK-PC1 aids postgermination growth of Xyris complanata seedlings with germination induced by Penicillium rolfsii Y-1. Biosci. Biotechnol. Biochem. 2021, 85, 77–84. [Google Scholar] [CrossRef]
  22. Spicer, J.A.; Gamage, S.A.; Rewcastle, G.W.; Finlay, G.J.; Bridewell, D.; Baguley, B.C. Bis(phenazine-1-carboxamides): Structure-activity relationships for a new class of dual topoisomerase i/ii-directed anticancer drugs. J. Med. Chem. 2000, 43, 1350–1358. [Google Scholar] [CrossRef]
  23. Abdel-Mageed, W.M.; Milne, B.F.; Wagner, M.; Schumacher, M.; Sandor, P.; Pathomaree, W.; Goodfellow, M.; Bull, A.T.; Horikoshi, K.; Ebel, R.; et al. Dermacozines, a new phenazine family from deep-sea dermacocci isolated from a Mariana Trench sediment. Org. Biomol. Chem. 2010, 8, 2352–2362. [Google Scholar] [CrossRef]
  24. Rewcastle, G.W.; Denny, W.A.; Baguley, B.C. Potential antitumor agents. 51. Synthesis and antitumor activity of substituted phenazine-1-carboxamides. J. Med. Chem. 1987, 30, 843–851. [Google Scholar] [CrossRef]
  25. Abdelfattah, M.S.; Ishikawa, N.; Karmakar, U.K.; Yamaku, K.; Ishibashi, M. New phenazine analogues from Streptomyces sp. ifm 11694 with trail resistance-overcoming activities. J. Antibiot. 2015, 69, 446–450. [Google Scholar] [CrossRef]
  26. Laursen, J.B.; Nielsen, J. Phenazine natural products: Biosynthesis, synthetic analogues, and biological activity. Chem. Rev. 2004, 104, 1663–1686. [Google Scholar] [CrossRef]
  27. Cimmino, A.; Evidente, A.; Mathieu, V.; Mathieu, V.; Andolfi, A.; Lefranc, F.; Kornienko, A.; Kiss, R. Phenazines and cancer. Nat. Prod. Rep. 2012, 29, 487–501. [Google Scholar] [CrossRef]
  28. Guttenberger, N.; Blankenfeldt, W.; Breinbauer, R. Recent developments in the isolation, biological function, biosynthesis, and synthesis of phenazine natural products. Bioorg. Med. Chem. 2017, 25, 6149–6166. [Google Scholar] [CrossRef] [PubMed]
  29. Kögl, F.; Postowsky, J.J. Über das grüne Stoffwechselprodukt des Bacillus chlororaphis. Justus Liebigs Ann. Chem. 1930, 480, 280–297. [Google Scholar] [CrossRef]
  30. Raju, B.C.; Prasad, K.V.; Saidachary, G.; Sridhar, B. A novel approach for c-c, c-n, and c-o bond formation reactions: A facile synthesis of benzophenazine, quinoxaline, and phenoxazine derivatives via ring opening of benzoxepines. Org. Lett. 2014, 45, 420–423. [Google Scholar] [CrossRef] [PubMed]
  31. Wang, S.L.; Wu, F.Y.; Cheng, C.; Zhang, G.; Liu, Y.P.; Jiang, B.; Shi, F.; Tu, S.J. Multicomponent synthesis of poly-substituted benzo[a]pyrano[2,3-c]phenazine derivatives under microwave heating. ACS Comb. Sci. 2011, 13, 135–139. [Google Scholar] [CrossRef] [PubMed]
  32. Rollas, S.; Küçükgüzel, S.G. Biological activities of hydrazone derivatives. Molecules 2007, 12, 1910–1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Peterson, Q.P.; Goode, D.R.; West, D.C.; Ramsey, K.N.; Lee, J.; Hergenrother, P.J. Pac-1 activates procaspase-3 in vitro through relief of zinc-mediated inhibition. J. Mol. Biol. 2009, 388, 144–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, D.; Ma, Y.; Liu, Y.; Liu, Z.P. Synthesis of sulfonylhydrazone- and acylhydrazone-substituted 8-ethoxy-3-nitro-2h-chromenes as potent antiproliferative and apoptosis inducing agents. Arch. Pharm. 2014, 347, 576–588. [Google Scholar] [CrossRef] [PubMed]
  35. Lu, Y.; Yan, Y.; Wang, L.; Wang, X.; Gao, J.; Xi, T.; Wang, Z.; Jiang, F. Design, facile synthesis and biological evaluations of novel pyrano[3,2-a]phenazine hybrid molecules as antitumor agents. Eur. J. Med. Chem. 2017, 127, 928–943. [Google Scholar] [CrossRef]
  36. Sheldrick, G.M. SHELX-97: Programs for Crystal Structure Analysis; University of Göttingen: Göttingen, Germany, 1997. [Google Scholar]
Scheme 1. Synthetic route to the acylhydrazone compounds 3a3j.
Scheme 1. Synthetic route to the acylhydrazone compounds 3a3j.
Molecules 26 05320 sch001
Figure 1. Crystal structure of compound 3b (dotted line represents hydrogen bond).
Figure 1. Crystal structure of compound 3b (dotted line represents hydrogen bond).
Molecules 26 05320 g001
Figure 2. The structure of PAC.
Figure 2. The structure of PAC.
Molecules 26 05320 g002
Figure 3. The structure of 1. (IC50 = 8.07 μmol/L, A549; IC50 = 0.76 μmol/L, KG-1; IC50 = 0.18 μmol/L, A2780).
Figure 3. The structure of 1. (IC50 = 8.07 μmol/L, A549; IC50 = 0.76 μmol/L, KG-1; IC50 = 0.18 μmol/L, A2780).
Molecules 26 05320 g003
Figure 4. The structure of 2. (IC50 = 23.73 μmol/L, HCT-116).
Figure 4. The structure of 2. (IC50 = 23.73 μmol/L, HCT-116).
Molecules 26 05320 g004
Figure 5. The structure of pyrano[3,2-a]phenazine derivatives. (1c: R1, R2, R4, R5 = H, R3 = Cl. 1i: R1, R4, R5 = H, R2, R3 = Cl. 2e: R1 = Br, R2, R3, R4, R5=H. 2l: R1, R2 = Cl, R3, R4, R5 = H).
Figure 5. The structure of pyrano[3,2-a]phenazine derivatives. (1c: R1, R2, R4, R5 = H, R3 = Cl. 1i: R1, R4, R5 = H, R2, R3 = Cl. 2e: R1 = Br, R2, R3, R4, R5=H. 2l: R1, R2 = Cl, R3, R4, R5 = H).
Molecules 26 05320 g005
Table 1. Structure, yield and crystal structures of compounds 3a3j.
Table 1. Structure, yield and crystal structures of compounds 3a3j.
CompoundMolecular FormulaCrystal StructureYield%
3a Molecules 26 05320 i001 Molecules 26 05320 i00260
3b Molecules 26 05320 i003 Molecules 26 05320 i00465
3c Molecules 26 05320 i005 Molecules 26 05320 i00665
3d Molecules 26 05320 i007none55
3e Molecules 26 05320 i008 Molecules 26 05320 i00968
3f Molecules 26 05320 i010 Molecules 26 05320 i01170
3g Molecules 26 05320 i012 Molecules 26 05320 i01360
3h Molecules 26 05320 i014 Molecules 26 05320 i01560
3i Molecules 26 05320 i016 Molecules 26 05320 i01760
3j Molecules 26 05320 i018 Molecules 26 05320 i01962
Table 2. Anti-tumor activities of compounds 3a3j [IC50/(μmol/L)].
Table 2. Anti-tumor activities of compounds 3a3j [IC50/(μmol/L)].
Compd.HelaStatistical Analysis (Compared with the Control Group)A549Statistical Analysis (Compared with the Control Group)
3a>100 >100
3b26.6 ± 4.8** p < 0.0122.4 ± 3.7** p < 0.01
3c34.4 ± 4.1** p < 0.0124.7 ± 4.7** p < 0.01
3d5.5 ± 1.3*** p < 0.0012.8 ± 1.53*** p < 0.001
3e36.3 ± 2.3*** p < 0.00130.5 ± 1.9*** p < 0.001
3f32.8 ± 3.9** p < 0.0128.6 ± 3.5** p < 0.01
3g>100 >100
3h>100 >100
3i>100 >100
3j>100 >100
cisplatin20.4 ± 4.8** p < 0.0111.6 ± 2.25*** p < 0.001
** denotes significance (p < 0.01) vs. control, *** denotes significance difference (p < 0.001) vs. control.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, S.; Liang, X.; Luo, F.; Liu, H.; Shen, L.; Yang, X.; Huang, Y.; Xu, H.; Wu, N.; Zhang, Q.; et al. Synthesis, Crystal Structure and Bioactivity of Phenazine-1-carboxylic Acylhydrazone Derivatives. Molecules 2021, 26, 5320. https://doi.org/10.3390/molecules26175320

AMA Style

Wu S, Liang X, Luo F, Liu H, Shen L, Yang X, Huang Y, Xu H, Wu N, Zhang Q, et al. Synthesis, Crystal Structure and Bioactivity of Phenazine-1-carboxylic Acylhydrazone Derivatives. Molecules. 2021; 26(17):5320. https://doi.org/10.3390/molecules26175320

Chicago/Turabian Style

Wu, Shouting, Xi Liang, Fang Luo, Hua Liu, Lingyi Shen, Xianjiong Yang, Yali Huang, Hong Xu, Ning Wu, Qilong Zhang, and et al. 2021. "Synthesis, Crystal Structure and Bioactivity of Phenazine-1-carboxylic Acylhydrazone Derivatives" Molecules 26, no. 17: 5320. https://doi.org/10.3390/molecules26175320

Article Metrics

Back to TopTop