Next Article in Journal
A Structural Approach to the Strength Evaluation of Linear Chalcogen Bonds
Next Article in Special Issue
Design, Synthesis, Molecular Modeling, and Biological Evaluation of Novel Pyrimidine Derivatives as Potential Calcium Channel Blockers
Previous Article in Journal
Cell-Free Expression of a Therapeutic Protein Serratiopeptidase
Previous Article in Special Issue
New Bicyclic Pyridine-Based Hybrids Linked to the 1,2,3-Triazole Unit: Synthesis via Click Reaction and Evaluation of Neurotropic Activity and Molecular Docking
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Anticancer Properties of 3-Dietoxyphosphorylfuroquinoline-4,9-dione

by
Joanna Drogosz-Stachowicz
1,
Katarzyna Gach-Janczak
1,
Marek Mirowski
2,
Jacek Pietrzak
2,
Tomasz Janecki
3 and
Anna Janecka
1,*
1
Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
2
Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
3
Institute of Organic Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(7), 3128; https://doi.org/10.3390/molecules28073128
Submission received: 28 February 2023 / Revised: 27 March 2023 / Accepted: 29 March 2023 / Published: 31 March 2023
(This article belongs to the Special Issue Biologically Active Heterocyclic Compounds)

Abstract

:
Herein, the antitumor activity of a novel synthetic analog with 5,8-quinolinedione scaffold, diethyl (2-(2-chlorophenyl)-4,9-dioxo-4,9-dihydrofuro [3,2-g]quinolin-3-yl)phosphonate (AJ-418) was investigated on two breast cancer cell lines. This analog was selected from a small library of synthetic quinolinediones on the basis of its strong antiproliferative activity against MCF-7 and MDA-MB-231 cells and 4-5-fold lower cytotoxicity towards healthy MCF-10A cells. The morphology of MCF-7 and MDA-MB-231 cancer cells treated with AJ-418 changed drastically, while non-tumorigenic MCF-10A cells remained unaffected. In MCF-7 cells, after 24 h incubation, the increased number of apoptotic cells coincided with a decrease in proliferation and cell viability. The 24 h treatment of MDA-MB-231 cells with the tested compound reduced their cell viability and proliferation rate; however, a significant pro-apoptotic effect was visible only after longer incubation times (48 h and 72 h). Then, the maximum tolerated dose (MTD) of compound AJ-418 in C3H mice after subcutaneous administration was determined to be 160 mg/kg, showing that this analog was well tolerated and can be further evaluated to assess its potential therapeutic effect in tumor-bearing mice.

1. Introduction

Heterocyclic compounds play an important role in the development of anticancer agents [1] because the ability of heteroatoms to participate in hydrogen bonding with the target protein can improve the selectivity and potency of the molecules [2]. The scaffold of quinolinediones is often found in nature and used for the development of new, biologically active substances. Among many possible isomeric forms, the most common are 5,8-quinolinediones (Figure 1), which are present in several structures of natural and synthetic compounds [3]. Two naturally occurring antitumor antibiotics based on 5,8-quinolinedione structure are streptonigrin [4] and lavendamycin [5] (Figure 1). Streptonigrin was found to be a potent anti-leukemia agent. However, a high degree of toxicity caused the termination of its development at phase II clinical trials [6]. The use of lavendamycin in cancer therapy failed, due to poor water solubility and low specificity of this compound. The study of lavendamycin-based compounds designed to overcome these drawbacks has been an area of research [7].
Several synthetic compounds containing 5,8-quinolinedione moiety that exhibit anticancer activity have been described in the literature. Ling and co-workers obtained a series of 6- and 7-arylamino-5,8-quinolinediones that were tested against drug sensitive (HeLeS3) and multidrug resistant (KB-vin) cell lines [8]. Other mono and disubstituted derivatives of the 5,8-quinolinedione were synthesized and showed high cytotoxic activity against several tumor cell lines [9,10,11]. More recently, a series of diversely substituted furoquinolinediones has been synthesized and some of these analogs turned out to be inhibitors of tyrosyl-DNA phosphodiesterase-2 (TDP2), the enzyme that repairs topoisomerase II (TOP2)-mediated DNA damage [12].
On the other hand, heterocycles containing a phosphorus substituent have received a lot of attention mainly due to their applications in chemical synthesis and their increasing usefulness in drug design [13]. A number of phosphorus-containing drugs have been designed as prodrugs in the form of phosphotriesters, phosphonates, phosphinates and phosphorus oxides to reduce side effects and toxicity and achieve greater selectivity and bioavailability. Such prodrugs are believed to have a higher polarity and enable the formation of stronger hydrogen bonds in vivo compared to unmodified molecules [14]. Several important drugs have been designed on this basis, fludarabine phosphate with anticancer activity being one example [15]. In addition, it has been shown that the presence of a dialkoxy- or diaryloxyphosphoryl group can improve the bioavailability of drugs [16], as well as slow down the progression of cancer by inhibiting, among others, farnesyl transferases [17] or purine nucleoside phosphorylases [18].
In our previous paper [19] we reported the synthesis of 3-diethoxyphosphorylfuroquinoline-4,9-diones combining furoquinoline-5,8-dione skeleton with diethoxyphosphoryl moiety. All obtained compounds proved to be highly cytotoxic for breast cancer adenocarcinoma MCF-7 and promyelocytic leukemia HL-60 cells, with IC50 values in the low µM range. The mode of action of the most potent analogs was investigated in HL-60 cells. These analogs enhanced intracellular ROS generation and NAD(P)H:quinone oxidoreductase 1 (NQO1) depletion, which led to reduced cell proliferation, DNA damage and apoptosis.
In the extension of this research, herein, we investigated the influence of diethyl (2-(2-chlorophenyl)-4,9-dioxo-4,9-dihydrofuro[3,2-g]quinolin-3-yl)phosphonate (AJ-418) (Figure 2) on cancerous mammary cell lines, MCF-7 and MDA-MB-231, in comparison with the non-tumorigenic MCF-10A cells. Then, the maximum tolerated dose (MTD) of the new analog in C3H mice was assessed in vivo.

2. Results

2.1. In Vitro Data

2.1.1. Cytotoxic Activity against Breast Cancer and Non-Tumorigenic Cell Lines (MTT Assay)

The cytotoxicity of AJ-418 for MCF-7, MDA-MB-231 and non-tumorigenic MCF-10A cells was assessed after 48 h and 24 h incubation in the concentration range of 0–2.5 µM using an MTT assay. The obtained IC50 values (Table 1) were similar for both cancer cell lines and 4- to 5-fold higher for the MCF-10A non-tumorigenic breast cell line, indicating that the tested compound showed some selectivity of action.

2.1.2. Morphology of MCF-7, MDA-MB-231 and MCF-10A Cells Incubated with AJ-418

Morphology changes in MCF-7 and MDA-MB-231 cancer cells, and for comparison in non-tumorigenic MCF-10A breast cells, were assessed after staining these cells with Giemsa dye that stains nuclei dark blue and cytoplasm blue or pink. The control cells (untreated) grew well and exhibited epithelial features, forming a monolayer. The morphology of MCF-7 and MDA-MB-231 cells treated with AJ-418 for 24 h changed, the cells became more heterogeneous in shape and size, and a reduction in cell number was observed. Importantly, the morphology of non-tumorigenic MCF-10A breast cells treated with the tested compound at either 0.5 µM or 1 µM concentrations did not change compared to the control (Figure 3).

2.1.3. Apoptosis Induction

Apart from morphological changes, the typical features of apoptotic cell death are phosphatidylserine (PS) translocation to extracellular membrane, caspase-3 activation and DNA damage.
The translocation of PS in MCF-7 and MDA-MB-231 cells treated with AJ-418 was assessed using double-staining with Annexin V and propidium iodide (PI). As shown in Figure 4, after 24 h, AJ-418 did not significantly increase the number of MCF-7 cells in the early phase of apoptosis, while increased late apoptosis events from 14.9 ± 0.57% in control (untreated) cells to 47.7 ± 0.6% and 60.8 ± 1.4% for 0.5 and 1 µM concentrations, respectively. In MDA-MB-231 cells, 24 h incubation with AJ-418 induced early apoptosis only in a small percent of cell population. Longer incubation (48 h and 72 h) led to the induction of both early and late apoptosis, which was more pronounced when a higher concentration of AJ-418 (1 µM) was used. At this concentration, the tested compound enhanced the percentage of apoptotic cells to 7.1 ± 0.21% and 6.7 ± 0.16%, for early apoptotic occurrence and to 16.3 ± 0.83% and 39.9 ± 0.44% for late apoptosis, after 48 h and 72 h incubation, respectively. Thus, the pro-apoptotic effect of compound AJ-418 in MDA-MB-231 cells was dose and time dependent (Figure 4).
The induction of apoptosis was also evaluated by detecting a cleaved fragment of PARP (poly(ADP-ribose) polymerase) with a mass of 89 kDa, generated by cleavage of PARP1 by caspase 3 and 7 during caspase-mediated apoptosis [20]. AJ-418 at 0.5 and 1 µM concentrations dose-dependently increased the population of cleaved PARP in MCF-7 cells to 28.7 ± 2.95% and 36.2 ± 0.64%, respectively, compared to controls (2.7 ± 0.52%). In MDA-MB-231 cells, these values were lower, 1.6 ± 0.18% and 1.2 ± 0.2% of the cell population, respectively, compared to controls (0.5 ± 0.07%) (Figure 5). The results suggest that after 24 h, AJ-418 at the tested concentrations caused significant apoptosis in MCF-7 cells, while in MDA-MB-231 cells, only a weak pro-apoptotic effect was observed.

2.1.4. DNA Damage Assessment

Apoptosis induced by many anticancer drugs may be a consequence of DNA damage [21]. Chemical genotoxins that target DNA can inhibit DNA replication, which leads to the collapse of replication forks and the formation of DNA double-strand breaks (DSBs). An early cells’ response to DNA damage includes the histone variant H2AX phosphorylation on Ser139, which leads to structural modifications at the site of damage. Phosphorylated H2AX has been shown to promote DNA repair and play a role in genome stability, cell cycle checkpoint response, and tumor suppression [22]. Therefore, the measurement of histone H2AX phosphorylation level can be used to detect the genotoxic effects of potential anticancer agents.
To assess the ability of AJ-418 to phosphorylate H2AX, MCF-7 and MDA-MB-231, cells were incubated with the tested compound for 24 h at 0.5 and 1 µM concentrations. AJ-418 caused a significant H2AX phosphorylation, indicative of DNA damage in both MCF-7 and MDA-MB-231 cell lines (Figure 6). AJ-418 phosphorylated H2AX in 33.2 ± 2.14% and 48.4 ± 0.18% of MCF-7 cells, respectively, compared to controls (2.7 ± 0.03%) and was slightly less genotoxic to MDA-MB-231 cells, 33.0 ± 3.19% and 36.9 ± 2.03%, respectively, compared to controls (0.8 ± 0.13%).

2.1.5. Antiproliferative Activity

The ability of AJ-418 to inhibit MCF-7 and MDA-MB-231 cell proliferation was evaluated by exposure of cells to 5-bromodeoxyuridine (BrdU), a thymidine analog that incorporates into newly synthesized DNA during the S-phase of the cell cycle. The obtained results showed that the tested compound significantly inhibited the proliferation of both cell lines. Analog AJ-418 at 0.5 and 1 µM concentrations caused the decrease in BrdU-incorporating MCF-7 cells from 37.9 ± 2.8% in the control to 26.9 ± 2.24% and 7.4 ± 2.02%, respectively, while in MDA-MB-231 cells, AJ-418 reduced proliferation from 56.0 ± 1.26% in the control to 32.6 ± 0.76% and 22.7 ± 1.86%, respectively (Figure 7).

2.1.6. Cell Cycle Analysis

Cytotoxic action of anticancer drugs may be exerted, among others, by the arrest of the cell cycle. Changes in the distribution of the cell cycle phases after treatment with AJ-418 were observed using flow cytometry analysis. After 24 h incubation with this compound, MCF-7 and MDA-MB-231 cells were treated with a fluorescent dye (4,6-diamidino-2-phenylindole, DAPI), which quantitatively stains DNA. The fluorescence intensity of the stained cells corresponds to the amount of DNA they contain and indicates the position in the cell cycle in the major phases (G0/G1 vs. S vs. G2/M). Representative histograms showing changes in the cell cycle distribution are presented in Figure 8. AJ-418 at 0.5 µM and 1 µM concentrations caused a significant increase in the number of MCF-7 cells in the S and G2/M phases (from 18.2% and 12.0% in the control to 27.5% and 19.0%, respectively), and in MDA-MB-231 cells (from 20.1% and 16.9% in the control to 25.4% and 20.3% in treated cells, respectively). At the higher concentration (1 µM), there was a significant increase in the number of MCF-7 cells primarily in the sub G0/G1 phase (from 1.8% in the control to 14.6%). In contrast, in MDA-MB-231 cells, the higher concentration of AJ-418 did not result in significant changes in the cell cycle distribution compared to controls.

2.2. In Vivo Studies—Evaluation of Maximum Tolerated Dose in C3H Mice

The aim of the in vivo studies was to estimate the toxicity of the tested compound in C3H mice. To determine the maximum tolerated dose (MTD) of compound AJ-418 in C3H mice after subcutaneous (s.c.) administration, on the first day of the experiment, a group of three mice was given an initial dose of the compound, 20 mg/kg, after which the animals were observed for 48 h. During this period no signs of compound toxicity, including weight loss, changes in appearance, body posture, mobility or changes at the site of administration were observed. Then, the other four groups of mice (n = 3) were administered sequentially higher doses of the compound, 40 mg/kg, 80 mg/kg, 160 mg/kg and 240 mg/kg. Animals were observed for 48 h between doses and no adverse effects were detected. Within an hour after administration of a dose of 240 mg/kg, no negative symptoms were seen in the animals that could suggest a toxic effect of the tested compound. However, the next morning all mice (n = 3) were found dead. Therefore, the penultimate dose of 160 mg/kg administered s.c. was considered the MTD of compound AJ-418 in C3H mice (Figure 9).

3. Discussion

Cancer tumors often consist of different types of cells; therefore, the phenotype of cancer cells is commonly considered an important parameter to determine the degree of their malignancy, and consequently, the response that can be expected from the applied therapy [23]. Here, the effect of AJ-418 on cell viability and proliferation was analyzed on two human breast cancer cell lines, MCF-7 and MDA-MB-231, with different proliferative potential.
MCF-7 cells express estrogen receptors (ER), progesterone receptors (PR) and epidermal growth factor receptors (EGFR). It was shown that estrogens stimulate the proliferation of breast cancer cells in vitro and in vivo [24,25]. ER activation is considered an important factor in breast cancer progression [26], whereas treatment with anti-estrogen chemotherapy drugs (e.g., tamoxifen) can reduce the growth of cultures by inhibiting proliferation and inducing apoptosis [27].
The MDA-MB-231 is defined as a triple-negative breast cancer (TNBC) cell line, as it lacks ER, PR and EGFR. This cell line is commonly used to model late-stage breast cancer [28]. TNBC is characterized by high invasiveness, high metastatic potential and short-term response to available therapies, with the tendency to relapse and with poor prognosis [29]. Since TNBC tumors do not express ER, PR, and EGFR, they are not sensitive to endocrine therapy or EGF therapy, and standardized TNBC treatment regimens are not available. Therefore, the development of new strategies for the treatment of TNBC continues to be the goal of many studies [30,31].
The human non-tumorigenic breast cell line MCF-10A is widely used for comparison in various studies, due to its structural similarity to the normal human mammary epithelium. MCF-10A cells are controlled by hormones and growth factors, although they do not express ER or show signs of terminal differentiation or senescence [32]. In mice, these cells were found to be non-tumorigenic [33].
In this report, it was shown that AJ-418 was cytotoxic to MCF-7 and MDA-MB-231 cells in a concentration-dependent manner but had no apparent effect on the growth of MCF-10A cells, nor did it induce noticeable changes in their morphology. The increase in the number of apoptotic MCF-7 cells observed after incubation with AJ-418 coincided with a decrease in proliferation and cell viability. In MCF-7, but not in MDA-MB-231 cells, AJ-418 caused a significant increase in the number of cells in the sub-G0/G1 phase, with reduced DNA content, indicating their programmed death. In MDA-MB-231 cells, a significant pro-apoptotic effect was visible only after a longer incubation time (72 h). This observation is in agreement with the fact that the MDA-MB-231 cell line represents one of the most aggressive, therapy-resistant and metastatic cancers.
The cytotoxic effect of anticancer agents often correlates with DNA damage and double-strand breaks (DSBs) [21]. AJ-418 led to a significant increase in DSBs in almost half of the MCF-7 cell population and was slightly less genotoxic to MDA-MB-231 cells. The exposure of MCF-7 cells to the tested compound led to an increase in γH2AX, leading to DNA damage and apoptosis. Interestingly, in MDA-MB-231 cells, despite a similar level of DNA damage, apoptosis occurred in a smaller number of cells, which probably indicates that DSBs could be repaired and did not immediately lead to cell death. These results show that similar to other anticancer drugs, AJ-418 exerts a stronger therapeutic effect on MCF-7 cells, while higher efficiency of DNA repair in MDA-MB-231 cells may be one of the factors contributing to their chemoresistance [34].
In order to determine the in vivo toxicity of AJ-418, this analog was administered to C3H mice. The C3H strain was obtained by Strong in 1920 [35]. Female C3H mice that are neonatally infected with the murine breast cancer virus develop tumors spontaneously and with high frequency [36]. The MTD of AJ-418 in C3H mice after a single s.c. administration, was determined to be 160 mg/kg b.w., indicating that this analog was well tolerated.
In summary, AJ-418 was cytotoxic to MCF-7 and MDA-MB-231 tumor cells, but had no apparent effect on the growth of non-tumorigenic MCF-10A cells. In MCF-7 cells, the tested analog induced the formation of DSBs, which led to the cell cycle arrest in the S and G2/M phases, and finally to apoptosis. In MDA-MB-231 cells, AJ-418 increased the number of cells with DSBs, but significant differences in the cell cycle distribution were not observed and apoptosis occurred after longer incubation times, indicating that this cell line was more resistant than MCF-7 to the action of AJ-418. In vivo, the new analog was well tolerated in female C3H mice (Figure 10).
These findings indicate that synthetic 3-dietoxyphosphorylfuroquinoline-4,9-dione AJ-418 can be further modified and studied in vivo as a potential anticancer agent for breast cancer therapy.

4. Materials and Methods

4.1. Cell Cultures and Sample Preparation

Breast cancer MCF-7 and MDA-MB-231 cell lines were purchased from the European Collection of Cell Cultures (ECACC) and cultured in Minimum Essential Medium Eagle (MEME, Sigma–Aldrich, St. Louis, MO, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS) (Biological Industries, Beit-Haemek, Israel), 2 mM glutamine, Men Non-essential amino acid solution and antibiotics (100 μg/mL streptomycin and 100 U/mL penicillin), all from Sigma–Aldrich (Sigma–Aldrich, St. Louis, MO, USA). MCF-10A were purchased from the American Type Culture Collection (ATCC) and cultured using MEGM Mammary Epithelial BulletKit. Cells were maintained at 37 °C in a 5% CO2 atmosphere and grown until 80% confluent.
For biological experiments, the tested compound was dissolved in DMSO (Sigma–Aldrich, Louis, MO, USA) and further diluted in culture medium to obtain less than 0.1% DMSO concentration. In each experiment, controls without and with DMSO were performed. DMSO in 0.1% concentration had no effect on the observed parameters.

4.2. MTT Assay

The MTT (3-(4,5-dimethyldiazol-2-yl)-2,5 diphenyl tetrazolium bromide) assay was performed according to Mosmann [37]. The cells were incubated with the tested compound for 24 h or 48 h. The absorbance was measured at 560 nm using FlexStation 3 Multi-Mode Microplate Reader (Molecular Devices, LLC, San Jose, CA, USA) and compared with control (untreated cells). Experiments were performed in triplicate. The IC50 values were calculated from concentration–response curves.

4.3. Assessment of Morphological Changes

Cells (MCF- 7, MDA-MB-231, MCF-10A) were seeded in 6-well plates at 2 × 105 cells/well in 2 mL of culture medium and incubated for 24 h. After another 24 h incubation with the tested compound, the old medium was removed and the cells were stained by adding 1 mL of Giemsa reagent (Sigma–Aldrich, St. Louis, MO, USA) at a 1:2 dilution in deionized water. The plates were incubated at 37 °C for 2 h and then rinsed thoroughly with distilled water. Morphological changes in cells treated with the tested compound were observed and photographed using a digital camera (MoticMoticam 2300, Motic China Group Co.,Ltd., Xiamen, China) connected to a phase-contrast microscope.

4.4. Apoptosis Detection by Annexin V and Propidium Iodide Staining

The induction of apoptosis was determined using a FITC Annexin V Apoptosis Detection Kit I (BD Biosciences, San Jose, CA, USA). The percentage of apoptotic cells was assessed using flow cytometry with CytoFLEX (Beckman Coulter, Inc., Brea, CA, USA). Data analysis was performed using Kaluza Analysis Software v2 (Beckman Coulter, Inc., Brea, CA, USA).

4.5. Evaluation of Cell Cycle Distribution, Proliferation, Apoptosis and DNA Damage Using the “Apoptosis, DNA Damage and Cell Proliferation Kit”

Cell cycle kinetics, apoptosis and DNA damage were assessed using the “Apoptosis, DNA Damage and Cell Proliferation Kit” (BD Biosciences, San Jose, CA, USA) according to the manufacturer’s protocol. MCF-7 and MDA-MB-231 cells were seeded into bottles at 2 × 105 cells/mL in 10 mL of medium culture and left for 24 h. Then, the tested compound in the specific concentrations was added. Cells incubated without the tested compound served as controls. After 24 h incubation, the cells were treated with a BrdU solution in a final concentration of 10 µM in culture medium and incubated for 8 h. Cells were trypsinized, collected using centrifugation, fixed, permeabilized and then incubated with DNase (300 µg/mL in PBS) for 1 h at 37 °C to expose the incorporated BrdU. Then, the cells were stained with Anti-BrdU, Anti-H2AX (pS139) and Anti-Cleaved PARP (Asp214) antibodies for 20 min in the dark at room temperature. Finally, the cells were resuspended in DAPI solution (1 µg/mL in staining buffer) to stain total DNA for cell cycle analysis. Cells were analyzed using flow cytometry using CytoFLEX (Beckman Coulter, Inc.). The analysis of data was performed using Kaluza Analysis Software v2 (Beckman Coulter, Inc.).

4.6. Animals

Animal care and all experimental procedures were performed in accordance with the Medical University of Lodz recommendations, described in the Guide for the Care and Use of Laboratory Animals and complied with the ARRIVE guidelines [38].
All experiments on living animals were carried out according to protocols approved by the Local Ethical Committee No: 41/ŁB 180/2020 20 July 2020.
Female C3H mice (Department of Genetics and Laboratory Animal Breeding, Oncology Center, Institute of Maria Skłodowska-Curie, Warsaw, Poland), weighing 22–25 g were used in the study. The animals were housed under standard conditions (22 ± 1 °C, 12 h light/dark cycle) with food and water ad libitum for 7 days before the experiments.

4.7. Determination of the Maximum Tolerated Dose (MTD)

Fifteen C3H mice were randomly assigned to groups of n = 3 animals. The first group was given s.c. an initial dose (20 mg/kg). This dose was determined on the basis of bibliographic data on the MTD values of similar cytotoxic drugs in mice [39]. The first dose could be escalated or de-escalated depending on the absence or presence of severe toxicity. Subsequent groups were treated with increasing doses of the tested compound. If none of the three animals in a group developed dose-limiting toxicity (DLT), the next three animals were given the next higher dose. However, if one of the three mice developed dose-limiting toxicity, the next three animals would be given the compound at the same dose level. Dose escalation continued until at least two subjects per group experienced dose-limiting toxicity. Following the administration of each dose, mice were observed for 48 h and the following parameters and endpoints were assessed: mortality, clinical signs and body weight. The MTD in this study was defined as the highest dose that will be tolerated and will not cause serious life-threatening toxicity in an animal for the duration of the study. The MTD was set as the dose preceding the toxic dose.
The solution of the tested compound was administered with a 25-gauge needle in a volume of 0.1 mL/10g b.w. The injection was performed s.c. in the interscapular area.
All surviving animals after the experimental time were euthanized by cervical dislocation. The animals were sacrificed individually, in isolation from other animals, and anesthetized by intraperitoneal injection with a mixture of ketamine (50 mg/kg b.w.) and xylazine (5 mg/kg b.w.).

4.8. Statistical Analysis

For the in vitro studies, all data were expressed as mean ± SEM. Statistical analyzes were performed using Prism 6.0 (GraphPad Software Inc., San Diego, CA, USA). Statistical significance was assessed using one-way ANOVA, followed by the Student–Newman–Keuls post-hoc multiple comparison test (for comparisons of three or more groups) or the Student’s t-test (for comparisons of two groups). The values * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant.

Author Contributions

Conceptualization, J.D.-S. and A.J.; Investigation, J.D.-S.; Methodology, J.D.-S., M.M. and T.J.; Software, J.D.-S., K.G.-J. and J.P.; Writing—original draft, J.D.-S. and A.J.; Writing—review & editing, K.G.-J., M.M., T.J. and A.J. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by grants 503/1-156-02/503-11-001-19-00 and 503/3-015-02/503-31-001-19-00 from the Medical University of Lodz, Poland.

Institutional Review Board Statement

The animal study protocol was approved by the Ethical Committee of the Medical University of Lodz No: 41/ŁB 180/2020 20 July 2020.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank veterinarian Piotr Kamola from the Animal Facility of the Medical University of Lodz for his help in conducting the animal studies and for valuable suggestions related to the determination of the clinical toxicity of the investigated compound. We would also like to thank Jacek Szymański, Head of the Research Laboratory CoreLab, for conducting the flow cytometry analysis.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Jampilek, J. Heterocycles in medicinal chemistry. Molecules 2019, 24, 3839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mancini, I.; Vigna, J.; Sighel, D.; Defant, A. Hybrid molecules containing naphthoquinone and quinolinedione scaffolds as antineoplastic agents. Molecules 2022, 27, 4948. [Google Scholar] [CrossRef] [PubMed]
  3. Kadela-Tomanek, M.; Bębenek, E.; Chrobak, E.; Boryczka, S. 5, 8-Quinolinedione scaffold as a promising moiety of bioactive agents. Molecules 2019, 24, 4115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rao, K.V.; Cullen, W.P. Streptonigrin, an antitumor substance. I. Isolation and characterization. Antibiot. Annu. 1959, 7, 950–953. [Google Scholar]
  5. Balitz, D.M.; Bush, J.A.; Bradner, W.T.; Doyle, T.W.; O’Herron, F.A.; Nettleton, D.E. Isolation of lavendamycin, a new antibiotic from Streptomyces lavendulae. J. Antibiot. 1982, 35, 259–265. [Google Scholar] [CrossRef] [Green Version]
  6. Bolzán, A.D.; Bianchi, M.S. Genotoxicity of streptonigrin: A review. Mutat. Res. 2001, 488, 25–37. [Google Scholar] [CrossRef]
  7. Hassani, M.; Cai, W.; Koelsch, K.H.; Holley, D.C.; Rose, A.S.; Olang, F.; Lineswala, J.P.; Holloway, W.G.; Gerdes, J.M.; Behforouz, M.; et al. Lavendamycin antitumor agents: Structure-based design, synthesis, and NAD(P)H:quinone oxidoreductase 1 (NQO1) model validation with molecular docking and biological studies. J. Med. Chem. 2008, 51, 3104–3115. [Google Scholar] [CrossRef]
  8. Ling, Y.; Yang, Q.X.; Teng, Y.N.; Chen, S.; Gao, W.J.; Guo, J.; Hsu, P.L.; Liu, Y.; Morris-Natschke, S.L.; Hung, C.C.; et al. Development of novel amino-quinoline-5,8-dione derivatives as NAD(P)H:quinone oxidoreductase 1 (NQO1) inhibitors with potent antiproliferative activities. Eur. J. Med. Chem. 2018, 154, 199–209. [Google Scholar] [CrossRef]
  9. Kadela-Tomanek, M.; Jastrzębska, M.; Pawełczak, B.; Bębenek, E.; Chrobak, E.; Latocha, M.; Boryczka, S. Alkynyloxy derivatives of 5, 8-quinolinedione: Synthesis, in vitro cytotoxicity studies and computational molecular modeling with NAD (P) H:Quinone oxidoreductase 1. Eur. J. Med. Chem. 2017, 126, 969–982. [Google Scholar] [CrossRef]
  10. Zhou, B.-Q.; Huang, X.-L.; Qin, Q.-P.; Wang, Z.-F.; Wu, X.-Y.; Tan, M.-X.; Liang, H. Transition metal complexes with 6,7-dichloro-5,8-quinolinedione as mitochondria-targeted anticancer agents. Polyhedron 2020, 181, 114–482. [Google Scholar]
  11. Hsu, T.S.; Chen, C.; Lee, P.T.; Chiu, S.J.; Liu, H.F.; Tsai, C.C.; Chao, J.I. 7-Chloro-6-piperidin-1-yl-quinline-5, 8-dione (PT-262), a novel synthetic compound induces lung carcinoma cell death associated with inhibiting ERK and CDC2 phosphorylation via a p53-independent pathway. Cancer Chemother. Pharmacol. 2008, 62, 799–808. [Google Scholar] [CrossRef]
  12. Yu, L.-M.; Hu, Z.; Chen, Y.; Ravji, A.; Lopez, S.; Plescia, C.B.; Yu, Q.; Yang, H.; Abdelmalak, M.; Saha, S.; et al. Synthesis and structure-activity relationship of furoquinolinediones as inhibitors of tyrosyl-DNA phosphodiesterase 2 (TDP2). Eur. J. Med. Chem. 2018, 151, 777–802. [Google Scholar] [CrossRef]
  13. Chen, L.; Liu, X.Y.; Zou, Y.X. Recent advances in the construction of phosphorus-substituted heterocycles, 2009–2019. Adv. Synth. Catal. 2020, 362, 1724–1818. [Google Scholar] [CrossRef]
  14. Yu, H.; Yang, H.; Shi, E.; Tang, W. Development and clinical application of phosphorus-containing drugs. Med. Drug Discov. 2020, 8, 100063. [Google Scholar] [CrossRef]
  15. Rodriguez, G. Fludarabine phosphate. Investig. New Drugs 1994, 12, 75–92. [Google Scholar] [CrossRef]
  16. Wiemer, A.J.; Wiemer, D.F. Prodrugs of phosphonates and phosphates: Crossing the membrane barrier. Top. Curr. Chem. 2015, 360, 115–160. [Google Scholar]
  17. Bagchi, S.; Rathee, P.; Jayaprakash, V.; Banerjee, S. Farnesyl transferase inhibitors as potential anticancer agents. Mini Rev. Med. Chem. 2018, 18, 1611–1623. [Google Scholar] [CrossRef]
  18. Al-Kali, A.; Gandhi, V.; Ayoubi, M.; Keating, M.; Ravandi, F. Forodesine: Review of preclinical and clinical data. Future Oncol. 2010, 6, 1211–1217. [Google Scholar] [CrossRef]
  19. Modranka, J.; Drogosz-Stachowicz, J.; Pietrzak, A.; Janecka, A.; Janecki, T. Synthesis and structure–activity relationship study of novel 3-diethoxyphosphorylfuroquinoline-4, 9-diones with potent antitumor efficacy. Eur. J. Med. Chem. 2021, 219, 113–429. [Google Scholar] [CrossRef]
  20. Mashimo, M.; Onishi, A.; Uno, A.; Tanimichi, A.; Nobeyama, M.; Mori, T.; Yamada, S.; Negi, S.; Bu, X.; Kato, J. The 89-kDa PARP1 cleavage fragment serves as a cytoplasmic PAR carrier to induce AIF-mediated apoptosis. J. Biol. Chem. 2021, 296, 100046–100058. [Google Scholar] [CrossRef]
  21. Roos, W.P.; Kaina, B. DNA damage-induced cell death by apoptosis. Trends Mol. Med. 2006, 12, 440–450. [Google Scholar] [CrossRef]
  22. Fernandez-Capetillo, O.; Lee, A.; Nussenzweig, M.; Nussenzweig, A. H2AX: The histone guardian of the genome. DNA Repair 2004, 3, 959–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Marusyk, A.; Almendro, V.; Polyak, K. Intra-tumour heterogeneity: A looking glass for cancer? Nat. Rev. Cancer 2012, 12, 323–334. [Google Scholar] [CrossRef] [PubMed]
  24. Liao, X.H.; Lu, D.I.; Wang, N.; Liu, L.Y.; Wang, Y.; Li, Y.Q.; Yan, T.B.; Sun, X.G.; Hu, P.; Zhang, T.C. Estrogen receptor alpha mediate proliferation of breast cancer MCF-7 cells via a p21/PCNA/E2F1-dependent pathway. FEBS J. 2014, 17, 581–590. [Google Scholar]
  25. Zeng, L.; Li, W.; Chen, C.S. Breast cancer animal models and applications. Zool. Res. 2020, 41, 477–494. [Google Scholar] [CrossRef]
  26. Lamb, C.A.; Vanzulli, S.I.; Lanari, C. Hormone receptors in breast cancer: More than estrogen receptors. Medicina 2019, 79, 540–545. [Google Scholar]
  27. Zheng, A.; Kallio, A.; Harkonen, P. Tamoxifen-induced rapid death of MCF-7 breast cancer cells is mediated via extracellularly signal-regulated kinase signaling and can be abrogated by oestrogen. Endocrinology 2007, 148, 2764–2777. [Google Scholar] [CrossRef]
  28. Akekawatchai, C.; Roytrakul, S.; Kittisenachai, S.; Isarankura-Na-Ayudhya, P.; Jitrapakdee, S. Protein profiles associated with anoikis resistance of metastatic MDA-MB-231 breast cancer cells. Asian Pac. J. Cancer Prev. 2016, 17, 1007–1012. [Google Scholar] [CrossRef] [Green Version]
  29. Lee, K.S.; Lee, D.H.; Chun, S.Y.; Nam, K.S. Metastatic potential in MDA-MB-231 human breast cancer cells is inhibited by proton beam irradiation via the Akt/nuclear factor-kappaB signaling pathway. Mol. Med. Rep. 2014, 10, 1007–1012. [Google Scholar] [CrossRef] [Green Version]
  30. Lyons, T.G. Targeted therapies for triple-negative breast cancer. Curr. Treat. Options Oncol. 2019, 20, 82–94. [Google Scholar] [CrossRef]
  31. Yin, L.; Duan, J.J.; Bian, X.W.; Yu, S. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61–73. [Google Scholar] [CrossRef] [PubMed]
  32. Tait, L.; Soule, H.D.; Russo, J. Ultrastructural and immunocytochemical characterization of an immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990, 50, 6087–6094. [Google Scholar] [PubMed]
  33. Soule, H.D.; Maloney, T.M.; Wolman, S.R.; Peterson, W.D.; Brenz, R.; McGrath, C.M.; Russo, J.; Pauley, R.J.; Jones, R.F.; Brooks, S.C. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990, 50, 6075–6086. [Google Scholar] [PubMed]
  34. Lee, K.J.; Mann, E.; Wright, G.; Piett, C.G.; Nagel, Z.D.; Gassman, N.R. Exploiting DNA repair defects in triple negative breast cancer to improve cell killing. Ther. Adv. Med. Oncol. 2020, 12, 1758835920958354–1758835920958370. [Google Scholar] [CrossRef]
  35. McLaren, A.N.N.E.; Taita, A. Cytoplasmic isocitrate dehydrogenase variation within the C3H inbred strain. Gen. Res. 1969, 14, 93–94. [Google Scholar] [CrossRef] [Green Version]
  36. Shao, R.; Dao, M.L.; Day, N.K.; Good, R.A. Dietary manipulation of mammary tumor development in adult C3H/Bi Mice. Exp. Biol. Med. 1990, 193, 313–317. [Google Scholar] [CrossRef]
  37. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  38. Kilkenny, C.; Browne, W.; Cuthill, I.C.; Emerson, M.; Altman, D.G. NC3Rs Reporting Guidelines Working Group. Animal research: Reporting in vivo experiments: The ARRIVE guidelines. Br. J. Pharmacol. 2010, 160, 1577–1579. [Google Scholar] [CrossRef]
  39. Aston, W.J.; Hope, D.E.; Nowak, A.K.; Robinson, B.W.; Lake, R.A.; Lesterhuis, W.J. A systematic investigation of the maximum tolerated dose of cytotoxic chemotherapy with and without supportive care in mice. BMC Cancer 2017, 17, 684–693. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Structures of 5,8-quinolinedione and anticancer antibiotics containing this motif.
Figure 1. Structures of 5,8-quinolinedione and anticancer antibiotics containing this motif.
Molecules 28 03128 g001
Figure 2. Structure of diethyl (2-(2-chlorophenyl)-4,9-dioxo-4,9-dihydrofuro[3,2-g]quinolin-3-yl)phosphonate (AJ-418).
Figure 2. Structure of diethyl (2-(2-chlorophenyl)-4,9-dioxo-4,9-dihydrofuro[3,2-g]quinolin-3-yl)phosphonate (AJ-418).
Molecules 28 03128 g002
Figure 3. Morphological changes in MCF-7, MDA-MB-231 and MCF-10A cells after 24 h incubation with AJ-418 at concentrations of 0.5 µM and 1 µM. After staining with Giemsa dye, the cells were photographed under a light microscope with a built-in camera (magnification 40×).
Figure 3. Morphological changes in MCF-7, MDA-MB-231 and MCF-10A cells after 24 h incubation with AJ-418 at concentrations of 0.5 µM and 1 µM. After staining with Giemsa dye, the cells were photographed under a light microscope with a built-in camera (magnification 40×).
Molecules 28 03128 g003
Figure 4. Induction of apoptosis by AJ-418 in MCF-7 and MDA-MB-231 cells. Cells were exposed to the tested compound at 0.5 µM and 1 µM concentrations, MCF-7 for 24 h and in MDA-MB-231 for 24, 48 and 72 h, then stained with annexin V and PI and analyzed using flow cytometry. Representative cytograms and quantification of apoptotic cell death are shown in the figure; each column represents the mean ± SEM of three independent experiments; * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Figure 4. Induction of apoptosis by AJ-418 in MCF-7 and MDA-MB-231 cells. Cells were exposed to the tested compound at 0.5 µM and 1 µM concentrations, MCF-7 for 24 h and in MDA-MB-231 for 24, 48 and 72 h, then stained with annexin V and PI and analyzed using flow cytometry. Representative cytograms and quantification of apoptotic cell death are shown in the figure; each column represents the mean ± SEM of three independent experiments; * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Molecules 28 03128 g004
Figure 5. Percentage of the cell population containing cleaved PARP. MCF-7 and MDA-MB-231 cells were exposed to AJ-418 at 0.5 µM and 1 µM for 24 h, then labeled with Anti-Cleaved-PARP antibody (Asp214) and analyzed using flow cytometry. Data represent means ± SEM of three independent experiments. Statistical significance was determined using one-way ANOVA and the Student–Newman–Kleus post-hoc multiple comparison test; *** p < 0.001, ** p < 0.01, * p < 0.05 represent statistically significant differences in comparison to controls, ns—no statistical significance.
Figure 5. Percentage of the cell population containing cleaved PARP. MCF-7 and MDA-MB-231 cells were exposed to AJ-418 at 0.5 µM and 1 µM for 24 h, then labeled with Anti-Cleaved-PARP antibody (Asp214) and analyzed using flow cytometry. Data represent means ± SEM of three independent experiments. Statistical significance was determined using one-way ANOVA and the Student–Newman–Kleus post-hoc multiple comparison test; *** p < 0.001, ** p < 0.01, * p < 0.05 represent statistically significant differences in comparison to controls, ns—no statistical significance.
Molecules 28 03128 g005
Figure 6. Induction of DNA damage by AJ-418 in MCF-7 and MDA-MB-231 cells. Cells were exposed to the tested compound at 0.5 µM and 1 µM concentration for 24 h, stained with anti-H2AX antibody (pS139) and analyzed using flow cytometry. Error bars represent means ± SEM of three independent experiments. *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Figure 6. Induction of DNA damage by AJ-418 in MCF-7 and MDA-MB-231 cells. Cells were exposed to the tested compound at 0.5 µM and 1 µM concentration for 24 h, stained with anti-H2AX antibody (pS139) and analyzed using flow cytometry. Error bars represent means ± SEM of three independent experiments. *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Molecules 28 03128 g006
Figure 7. Effect of AJ-418 on the proliferation of MCF-7 and MDA-MB-231 cells. Cells were exposed to AJ-418 at 0.5 µM and 1 µM concentrations for 24 h, stained with BrdU and analyzed using flow cytometry. Error bars represent means of three replicates ± SEM. * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls.
Figure 7. Effect of AJ-418 on the proliferation of MCF-7 and MDA-MB-231 cells. Cells were exposed to AJ-418 at 0.5 µM and 1 µM concentrations for 24 h, stained with BrdU and analyzed using flow cytometry. Error bars represent means of three replicates ± SEM. * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls.
Molecules 28 03128 g007
Figure 8. Effect of AJ-418 on cell cycle distribution in MCF-7 and MDA-MB-231 cells. Cells were exposed to AJ-418 at 0.5 µM and 1 µM concentrations for 24 h, stained with DAPI and analyzed using flow cytometry. Representative histograms and quantification are shown in the figure; error bars represent means ± SEM of three replicates. * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Figure 8. Effect of AJ-418 on cell cycle distribution in MCF-7 and MDA-MB-231 cells. Cells were exposed to AJ-418 at 0.5 µM and 1 µM concentrations for 24 h, stained with DAPI and analyzed using flow cytometry. Representative histograms and quantification are shown in the figure; error bars represent means ± SEM of three replicates. * p < 0.05, ** p < 0.01 and *** p < 0.001 were considered statistically significant compared to controls, ns—no statistical significance.
Molecules 28 03128 g008
Figure 9. Diagram showing the determination of the maximum tolerated dose (MTD). Each box represents a group of 3 mice that were given the tested compound at the specific dose. After each administration, the mice were observed for 48 h.
Figure 9. Diagram showing the determination of the maximum tolerated dose (MTD). Each box represents a group of 3 mice that were given the tested compound at the specific dose. After each administration, the mice were observed for 48 h.
Molecules 28 03128 g009
Figure 10. Schematic representation of the effect of quinolinedione AJ-418 on MCF-7 and MDA-231 cells and evaluation of MTD in C3H mice.
Figure 10. Schematic representation of the effect of quinolinedione AJ-418 on MCF-7 and MDA-231 cells and evaluation of MTD in C3H mice.
Molecules 28 03128 g010
Table 1. Cell viability of MCF-7, MDA-MB-231 and MCF-10A cells after 48 h and 24 h incubation with AJ-418, assessed using the MTT test.
Table 1. Cell viability of MCF-7, MDA-MB-231 and MCF-10A cells after 48 h and 24 h incubation with AJ-418, assessed using the MTT test.
Cell LineIC50 (µM) a
AJ-418Doxorubicin
48 h24 h48 h
MCF-70.10 ± 0.010.46 ± 0.010.89 ± 0.12
MDA-MB-2310.12 ± 0.010.49 ± 0.010.68 ± 0.08
MCF-10A0.50 ± 0.011.82 ± 0.041.2 ± 0.05
a Compound concentration required to inhibit cell viability by 50% after 48 h or 24 h treatment. Data are expressed as the mean ± SEM from the concentration–response curves of at least three experiments.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Drogosz-Stachowicz, J.; Gach-Janczak, K.; Mirowski, M.; Pietrzak, J.; Janecki, T.; Janecka, A. Anticancer Properties of 3-Dietoxyphosphorylfuroquinoline-4,9-dione. Molecules 2023, 28, 3128. https://doi.org/10.3390/molecules28073128

AMA Style

Drogosz-Stachowicz J, Gach-Janczak K, Mirowski M, Pietrzak J, Janecki T, Janecka A. Anticancer Properties of 3-Dietoxyphosphorylfuroquinoline-4,9-dione. Molecules. 2023; 28(7):3128. https://doi.org/10.3390/molecules28073128

Chicago/Turabian Style

Drogosz-Stachowicz, Joanna, Katarzyna Gach-Janczak, Marek Mirowski, Jacek Pietrzak, Tomasz Janecki, and Anna Janecka. 2023. "Anticancer Properties of 3-Dietoxyphosphorylfuroquinoline-4,9-dione" Molecules 28, no. 7: 3128. https://doi.org/10.3390/molecules28073128

Article Metrics

Back to TopTop