Next Article in Journal
Synthesis of ω-Methylsulfinyl- and ω-Methylsulfonylalkyl Glucosinolates
Previous Article in Journal
Cyto- and Genotoxicity of Selected Plant Extracts and Microbial Metabolites with Confirmed Activity Against Phytopathogens of Potato Seed (Solanum tuberosum L.)
Previous Article in Special Issue
Thermal Characterisation and Toxicity Profile of Potential Drugs from a Class of Disubstituted Heterofused Triazinones
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Anticancer Potential of Azatetracyclic Derivatives: In Vitro Screening and Selective Cytotoxicity of Azide and Monobrominated Compounds

by
Costel Moldoveanu
1,
Ionel I. Mangalagiu
1,2,
Gheorghita Zbancioc
1,*,
Ramona Danac
1,*,
Gabriela Tataringa
3 and
Ana Maria Zbancioc
3
1
Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1, 700506 Iasi, Romania
2
Institute of Interdisciplinary Research—CERNESIM Centre, Alexandru Ioan Cuza University of Iasi, 11 Carol I, 700506 Iasi, Romania
3
Faculty of Pharmacy, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 16 University Street, 700115 Iasi, Romania
*
Authors to whom correspondence should be addressed.
Molecules 2025, 30(3), 702; https://doi.org/10.3390/molecules30030702
Submission received: 22 December 2024 / Revised: 26 January 2025 / Accepted: 27 January 2025 / Published: 5 February 2025

Abstract

:
This study investigated the antiproliferative activity of three classes of benzo[f]pyrrolo[1,2-a]quinoline azatetracyclic derivatives. All compounds were screened against 60 cancer cell lines at a single dose of 10 μM. When we compared the activity of the three classes of azatetracyclic derivatives (azide, monobrominated and dibrominated), we found that the dibrominated compounds were less active, while the azides were the most active molecules. Compounds 3b and 5a, showing the best growth inhibition profile of all the drugs evaluated, were selected for the second stage of a full five-dose testing. According to the results of the in vitro screening, compounds 3b and 5a exhibit good to moderate anticancer activity (in micromolar range) against all nine cancer sub-panels, with compound 5a being more selective than compound 3b. Both compounds presented better activity than phenstatin on T–47D breast cancer cells, with compound 3b also being more active on SK–MEL–28 melanoma cells, while compound 5a was more active than phenstatin on COLO 205 colon cancer cells. As for the probable mechanism of action, the benzoquinoline derivatives could act as PI5P4Kα and PI5P4Kβ inhibitors or topoisomerase II inhibitors.

1. Introduction

Due to their various useful features, including complexation [1,2,3], luminescence [1,2,4], biological activities [4,5,6,7], and semiconductors [8], benzoquinolines are versatile molecules that have been used in several applications [1,2,3]. The benzoquinoline scaffold is a great option for the creation of distinctive bioactive molecules because these polycyclic compounds are also found naturally in morphine alkaloids, sterols, or hormones [9].
In addition, the combination of two or more heterocycle rings can create new classes of ligands, many of which have intriguing new features, such as anticancer activity. Analogs of nitidine and fagaronine with substituted benzo[c]phenanthrolines, for instance, demonstrated cytotoxic effects linked to DNA intercalation and caused arrests in the G2/M phases [10]. Our team recently revealed various fused pyrrolophenanthrolines or pyrrolobenzoquinoline that exhibited moderate antiproliferative activity [11,12].
The treatment for a variety of cancer kinds can be difficult and include stem cell transplantation, immunotherapy, hormone treatment, radiation, chemotherapy, surgery, and often, a combination of these [13,14,15]. Chemotherapy is a cancer treatment that can increase life expectancy. When the first anticancer drug was introduced into the market in 1949, the average lifespan was 46.8 years; in recent years, more than 160 anticancer drugs have been approved for clinical usage, bringing the average lifespan up to 71.4 years [16,17]. An estimated 9.7 million people died from cancer and 20 million new cases were reported in 2022. However, the drugs on the market now have a low rate of effectiveness and a host of drawbacks [18,19,20]. Even with the significant advancements in chemotherapy, cancer continues to be the primary cause of death globally. Thus, it is imperative for novel anticancer medications to be developed for use in treatment.
In light of the aforementioned considerations, our study focuses on the study of azaheterocyclic scaffolds in potential anticancer compounds [21]. Herein, we studied new hybrid compounds that include a benzo[f]quinoline core with possible anticancer potential.
In earlier studies, quinoline derivatives with polycyclic skeletons have been shown to exhibit a variety of biological activities, including anticancer properties [12,22,23]. Furthermore, quinoline- or benzo[f]quinoline-based substances are ATP synthase and Topo II inhibitors in terms of their mode of action [22,23,24]. Given the aforementioned factors, it seems reasonable to assume that target polycyclic benzo[f]quinoline functionalized derivatives, which had a similar structure to those previously tested, will function similarly in terms of their mechanism of action and antitumor activity (Scheme 1).
The synthesis of target functionalized azatetracyclic derivatives was realized as presented in our previous research [25]. Figure 1 shows the molecular structures of the compounds tested as anticancer agents against different cell lines.

2. Results and Discussion

2.1. Anticancer Activity

The anticancer activity of the synthesized compounds was evaluated by the National Cancer Institute (NCI), USA, through its screening program for anticancer medications. The in vitro NCI 60 cell line screen provides a comprehensive assessment of the compounds’ efficacy against various cancer cell types, making it a valuable resource for drug development. It comprises 60 distinct human tumor cell lines that represent a range of malignancies, such as kidney, lung, colon, brain, ovary, breast, prostate, and leukemia. The screening was conducted in accordance with the NCI protocol, which is included in the accompanying documentation [26,27,28,29,30,31,32].
In the first step of the screening procedure, each selected chemical is evaluated at a single dose of 10 μM against all 60 cell lines [26]. The mean graph showing the outcomes of this screen can be examined with the COMPARE application [29]. The results are expressed and the growth, relative to the drug-free control and the initial cell count using the percentage growth inhibition, or PGI, is indicated. This method can be used to detect growth inhibition (values range from 0 to 100) and death (values less than 0). If the number was 30, for example, 70% of growth would be inhibited, and if the number was −30, 30% of the cells would be dead.
Ten synthesized compounds, including four azatetracyclic monobromoderivatives (1a,b and 3a,b), two azatetracyclic dibromoderivatives (2a and 4a) and four azatetracyclic azides (5a,b and 6a,b), were evaluated in a first single-dose anticancer assay (at a concentration of 10−5 M). Table 1 contains the representative results that were obtained for all tested substances.
Compounds 3b and 5a were very effective in inhibiting the growth of many cancer cell lines. Compound 5a showed the best efficacy in terms of growth inhibition against the following cell lines: CCRF–CEM (with 89% lethality); HL–60 (TB) (with 98% lethality); MOLT–4 (with 99% lethality) and RPMI–8226 (with 87% lethality) leukemia cell lines, A549/ATCC (with 40% lethality); EKVX (with 44% lethality); HOP–62 (with 20% lethality); HOP–92 (with 6% lethality); NCI–H226 (with 2% lethality); NCI–H23 (with 2% lethality); NCI–H322M (with 15% lethality) and NCI–460 (with 88% lethality) non-small cell lung cancer lines, COLO 205 (with 44% lethality) and HCC–2998 (with 9% lethality) colon cancer cell lines, SF–295 (with 15% lethality); SF–539 (with 1% lethality) and SNB–75 (with 8% lethality) CNS cancer cell lines, SK–OV–3 (with 61% lethality) ovarian cancer cell line, A498 (with 47% lethality) and ACHN (with 7% lethality) renal cancer cell lines, DU–145 (with 2% lethality) prostate cancer cell line and HS 578T (with 19% lethality) and MDA–MB–468 (with 52% lethality) breast cancer cell lines.
Additionally, compound 3b demonstrated cytotoxic activity against the following cell lines: CCRF–CEM (with 4% lethality) leukemia cell line, NCI–H522 (with 55% lethality) non-small cell lung cancer line and LOX IMVI (with 5% lethality) melanoma cell line.
Regarding the leukemia cell line, compounds 5b and 6b show significant anticancer activity with great lethality against three (HL–60 (TB); MOLT–4; SR) and four (CCRF–CEM; MOLT–4; RPMI–8226; SR) cancer cell lines, respectively.
The analysis of the three azatetracyclic cycloadduct classes revealed a clear cancer cells inhibition hierarchy: azides demonstrated the highest activity, followed by monobrominated derivatives, while dibrominated compounds showed the lowest growth inhibition levels.
Considering the structure–activity relationship (SAR), a number of preliminary conclusions can be drawn (Figure 2).
Among the cycloadducts of type A, the unsubstituted azide 5a (R1 = H) exhibited the best inhibitory potential. In general, derivatives with R1 = H showed superior anticancer activity across most cell lines compared to compounds with R1 = Me. The presence of an azido group (R2 = N3) enhanced activity against leukemia and non-small cell lung cancer cells specifically. Dibrominated compounds exhibited the lowest potency against all cancer types, except breast cancer.
For type B cycloadducts, the methyl-substituted monobrominated derivative 3b (R1 = Me) demonstrated the highest activity across all cancer types except leukemia. In contrast to type A, compounds bearing R1 = Me provided better inhibitory properties compared to the ones with R1 = H. The azido substitution (R2 = N3) selectively enhanced the activity against leukemia cells. Dibrominated derivatives showed the lowest activity across all cancer cell lines, which was a similar behavior as in the case of type A compounds.
Based on their specific anticancer profiles, compounds 3b and 5a were selected for comprehensive five-dose evaluation studies. Phenstatin served as a reference compound under identical testing conditions. Table 2 shows a selection of the GI50 values.
According to the results of the in vitro screening, compound 3b exhibits good to moderate anticancer activity against the cancer cell lines from all nine sub-panels, with GI50 values ranging from 2.79 to 39.9 µM (the GI50 value was no longer determined if it was higher than 42.2 µM). Specifically, compound 3b had the best GI50 and TGI (the concentration that results in total growth inhibition) values against RXF 393 renal cancer cells (2.79 µM and 36.7 µM, respectively). Additionally, promising GI50 values were found against all leukemia cell lines, on three breast cancer cells (MCF7; MDA-MB-231/ATCC; BT-549), on two melanoma cells (LOX IMVI; MALME–3M), and on two colon cancer cells (HCT–15; SW-620).
Compound 5a exhibited anticancer activity on fewer cell lines, compared to compound 3b, on the 60 cancer cell lines from all nine sub-panels, with GI50 values ranging from 1.41 to 29.8 µM. In particular, compound 5a had the best GI50 and TGI values against A549/ATCC non-small cell lung cancer (1.41 µM and 6.76 µM, respectively).
When comparing the anticancer activity of the two compounds tested with the control phenstatin, both compounds showed better activity on T-47D breast cancer cells; compound 3b was more active than phenstatin on SK-MEL-28 melanoma cells, while compound 5a was more active than phenstatin on COLO 205 colon cancer cells.
In order to determine a possible mode of action of compounds 3b and 5a, we used in silico platform-approved and Investigational Oncology Agents COMPARE (IOA COMPARE) of NCI [29]. This NCI platform is built on top of the NCI 60 screening data for a set of 183 FDA-approved oncology drugs and 820 investigational agents. The similarity of pattern to that of the analyzed compound is expressed quantitatively as a Pearson correlation coefficient. The results obtained with the COMPARE algorithm indicate that compounds high in this ranking (correlation coefficient > 0.5) may possess a mechanism of action similar to that of the seed compound. When analyzing compound 3b with IOA COMPARE, we found a best-fitting profile (correlation coefficient = 0.68) with (R)-8-cyclopentyl-7-(cyclopentylmethyl)-2-((3,5-dichloro-4-hydroxyphenyl)amino)-5-methyl-7,8-dihydropteridin-6(5H)-one, known as CC260, a highly potent and selective noncovalent dual inhibitor for both phosphatidylinositol 5-phosphate 4-kinases PIP4Kα and PIP4Kβ. In the case of compound 5a, the best-fitting profile was obtained for (S)-4,4′-(propane-1,2-diyl)bis(piperazine-2,6-dione, Dexrazoxane), an FDA-approved cardioprotective drug used to ameliorate cardiac toxicity seen in anthracycline-based (e.g., doxorubicin, daunorubicin, epirubicin) chemotherapy recipients for cancer by fusing with free and bound iron. Dexrazoxane also acts as a DNA topoisomerase II inhibitor, which happens to be the same target of the DNA topoisomerase II anticancer agent (e.g., the anthracyclines), antagonizing the formation of the topoisomerase II cleavage complex and also rapidly degrading topoisomerase II beta [33].
The mean graphs comparison of the two compounds 3b and 5a are presented in Figures S13 and S14, respectively (Supplementary Materials) [34].

2.2. In Silico ADME and Toxicity Profile

Given the promising anticancer activity shown in our results, it becomes essential to further evaluate the drug candidates’ pharmacokinetic and toxicological profile. Thus, we performed an in silico ADMET (absorption, distribution, metabolism, excretion, and toxicity) study to theoretically assess the potential of compounds 3b and 5a as therapeutic agents. The predicted parameters, including molecular properties, pharmacokinetics, drug-likeness, and medicinal chemistry, are summarized in Table 3.
Compounds 3b and 5a exhibit favorable drug-like properties, fully complying with both Lipinski’s rule of five and Veber’s guidelines. Specifically, these compounds meet all the criteria for molecular properties: they have no violations of Lipinski’s rule, maintain fewer than 10 rotatable bonds, and possess a topological polar surface area below 140 Å2.
Compound 3b and 5a exhibit limited aqueous solubility, with a Log S (ESOL) value of ~−6.00. While the compounds present three Brenk structural alerts, these structural features do not necessarily preclude their potential pharmaceutical utility and may, in fact, contribute to their unique molecular characteristics.
Derivatives 3b and 5a exhibit promising pharmacokinetic characteristics, as detailed in Table 3. The compounds are predicted to have high gastrointestinal absorption, though they cannot permeate the blood–brain barrier (BBB). Furthermore, both compounds are not anticipated to act as a P-glycoprotein (P-gp) substrate, which can be beneficial for targeted drug delivery.
The charts derived from the Swiss ADME QSAR web tool, depicting the oral bioavailability predictions for compounds 3b and 5a, are illustrated in Figure 3.
The chart evaluates six key molecular parameters for drug-likeness: lipophilicity (LIPO), molecular size, polarity (POLAR), insolubility (INSOLU), unsaturation (INSATU), and molecular flexibility (FLEX). The chart represents these parameters with a red line. If the red line is integrated into the pink area, the molecule is considered drug-like. Compounds 3b and 5a demonstrated partial matches, meeting three (compound 3b) or four (compound 5a) of the six parameters while both exhibiting violations in two areas: INSATU (the ratio of sp3-hybridized carbon atoms to the total number of carbon atoms) and LIPO (with an XLOGP3 value falling outside the acceptable range of −0.7 to +5.0). Despite these minor deviations, the estimated results, together, suggest a promising pharmacokinetic and drug-likeness profile for both compounds.
The compound’s toxicity profile was analyzed using in silico predictions that generate probability scores for both active (Pa) and inactive (Pi) states across different biological targets (Table 4).
As shown in Table 4, compound 5a demonstrated the predicted cytotoxicity (defined by Pa > Pi and Pa > 0.3) against multiple cancer cell lines, notably against all six lines that were experimentally tested in the NCI panel: SF-539 glioblastoma, UACC-62 melanoma, SN12C renal carcinoma, HOP-62 non-small cell lung carcinoma, OVCAR-3 ovarian carcinoma and T47D breast carcinoma (Table 3). The predictions for compound 5b indicated potential cytotoxic activity against three cancer cell lines: MOLT-3, CCRF-CEM and NCI-H187. In this case, our experimental testing also confirmed the cytotoxicity against CCRF-CEM leukemia cells (Table 3), providing a validation of the computational predictions. Importantly, the prediction algorithm did not indicate any significant toxicity against normal human cell lines, suggesting potential selective anticancer activity for both compounds 3b and 5a. However, further experimental validation would be needed to confirm this selectivity.

3. Experimental Section

3.1. Synthetic Procedure

All ten tested compounds were synthesized according to our procedure previously reported in the literature [25].

3.2. Biological Testing

Cell Proliferation Assay

The in vitro biological studies were conducted by the National Cancer Institute (NCI, Bethesda, MD, USA) as part of the Developmental Therapeutics Program (DTP).
The Developmental Therapeutics Program (DTP) at the NCI has successfully guided late-stage preclinical therapeutics through the critical stages of development for more than 50 years. As a result, this initiative has been responsible for the discovery and development of more than 70% of the anticancer drugs used in modern therapy [30].
This screen works with 60 different human tumor cell lines representing leukemia, melanoma and malignancies of the ovary, breast, prostate, kidney, colon, and brain [31]. The goal is to prioritize synthetic chemicals or natural product samples that selectively inhibit the growth of or kill specific tumor cell lines for further evaluation. This screen is unique in that the biological response pattern generated by the complex dose–response of a given compound in 60 cell lines can be used by the COMPARE program in pattern recognition algorithms [29].
The evaluation of all compounds against the 60 cell lines at a single dose of 10−5 M is the first screening stage.
The standard NCI/DTP in vitro cancer screening methodology was used [32].
All 60 human malignant tumor cell lines were cultured in RPMI (Roswell Park Memorial Institute) 1640 medium containing 5% fetal bovine serum and 2 × 10−3 M L-glutamine. For a standard screening test, 100 μL of cancer cells are injected into 96-well microtiter plates (at plating densities ranging from 5000 to 40,000 cells/well, depending on the cell division rate of certain cell lines). After cell inoculation, the microtiter plates are incubated for 24 h under specified conditions before the addition of experimental drugs: 37 °C, 5% CO2, 95% air and 100% relative humidity. Two plates for each cell line are fixed in situ with TCA after 24 h to obtain measurements of each cell population at the time of test substance (Tz) addition. Prior to use, the test chemicals are frozen after being solubilized in DMSO (dimethylsulphoxide) at 400 times the intended final maximum test concentration. After the test substance has been added, a portion of the frozen concentrate is thawed, diluted two-fold and then mixed with a medium containing 50 µg/mL gentamicin. The required final drug concentration (10−5 M) is achieved by adding 100 µL of the test substance to the microtiter wells already containing 100 µL of medium. After the addition of the test chemical, the microtiter plates are incubated for a further 48 h under the conditions described above. The addition of cold TCA completes the test in the case of adherent cells. Cancer cells are fixed by the addition of 50 µL of cold 50 % (w/v) TCA (final concentration, 10% TCA). This is followed by incubation at 4 °C for 60 min. The plates are air dried after five rinses with tap water and the supernatant is discarded. After drying, 100 µL of a fluorescent dye solution (sulforhodamine B) is added to each well. The plates are then incubated at 25 °C for 10 min. The plates are washed five times with a 1% acetic acid solution to remove unbound fluorophore and then air dried. A 10 × 10−3 M Trisma base is used to solubilize the bound fluorophore and the absorbance is measured at 515 nm using an automated plate reader. The procedure is the same for the suspension cells, except that settled cells at the bottom of the wells are fixed by adding 50 µL of TCA 80% solution (final concentration, 16% TCA) to the test.
Calculate the percentage growth inhibition (PGI) from the seven absorbance readings as follows:
[(Ti − Tz)/(C − Tz)] × 100 (for concentrations for which Ti >/= Tz);
[(Ti − Tz)/Tz] × 100 (for concentrations for which Ti < Tz).
where Ti = test growth in the presence of the drug at the concentration level, C = control growth, and Tz = time zero.
Three dose–response parameters are derived for each chemical tested.

3.3. In Silico ADMET Predictions

The ADME in silico evaluation of the compounds 3b and 5a was performed using the SwissADME web tool (http://swissadme.ch/index.php (accessed on 10 December 2024)) in terms of molecular properties, pharmacokinetics, drug-likeness, and medicinal chemistry [35].
The in silico toxicological evaluation for the most active compounds 3b and 5a was performed using the webservice Cell Line Cytotoxicity Predictor (CLC-Pred) (https://www.way2drug.com/clc-pred/ (accessed on 13 January 2025)), which screens for in silico cytotoxicity on a panel of 278 tumor cells and 27 normal human cell lines from different tissues [36].

4. Conclusions

In conclusion, we report herein the anticancer potential of three classes of azatetracyclic derivatives (monobrominated, dibrominated and azide). Testing using a single-dose (10−5 M) experiment showed azide 5a being the most active compound, showing very good lethality against the majority of the cell lines tested (with a lethality in the range of 2–99%). Very good anticancer activity was also demonstrated by the monobrominated azatetracyclic derivative 3b, but also by all other azide derivatives 5b and 6ab that showed significant selective lethality for the leukemia and non-small cell lung cancer line. In addition, compound 3b showed selective growth inhibitory activity against the LOX IMVI melanoma cell line.
An SAR analysis revealed distinct inhibitory patterns for cycloadducts A and B. In type A compounds, cancer cell growth inhibition was optimized by an unsubstituted position (R1 = H) and azido substitution (R2 = N3), with compound 5a showing the highest potency. Conversely, type B compounds benefited from methyl substitution (R1 = Me), exemplified by the potent monobrominated derivative 3b. Notably, dibromination consistently reduced the activity across both series, while azido substitution enhanced the activity against specific cancer types, particularly leukemia.
The five-dose testing of compounds 3b and 5a shows anticancer activity in the micromolar range across all nine cancer sub-panels, with compound 5a proving more selective for several cancer cell lines than compound 3b. Both compounds also showed better activity on T-47D breast cancer cells than the control phenstatin. Compound 3b was more active than phenstatin on SK–MEL–28 melanoma cells, while compound 5a was more active than phenstatin on COLO 205 colon cancer cells. The IOA COMPARE NCI’s platform suggested that the anticancer activity of the benzoquinolines derivative could be achieved by the inhibition of PI5P4K kinase for compound 3b, or by topoisomerase II inhibition in the case of compound 5a. The in silico ADMET showed interesting pharmacokinetic properties and toxicity profiles. Considering their potential, compounds 3b and 5a could be further optimized.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30030702/s1, Figure S1: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 1a; Figure S2: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 1b; Figure S3: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 2a; Figure S4: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 3a; Figure S5: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 3b; Figure S6: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 4a; Figure S7: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 5a; Figure S8: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 5b; Figure S9: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 6a; Figure S10: Anticancer activity (single-dose (10−5 M) assay) of the azatetracyclic derivative 6b; Figure S11: Anticancer activity (five-dose assay) of the azatetracyclic derivative 3b; Figure S12: Anticancer activity (five-dose assay) of the azatetracyclic derivative 5a; Figure S13: Comparison of mean graphs of compounds 3b and CC260; Figure S14: Comparison of mean graphs of compounds 5a and dexrazoxane. Full single-dose and five-dose screen results of the tested compounds and the COMPARE mean graphs of compounds 3b and 5a are provided in the supporting information.

Author Contributions

The study design, synthesis, and writing were performed by R.D., A.M.Z., I.I.M., G.Z. and C.M. The interpretation of the anticancer activity of the tested compounds was performed by A.M.Z., G.T., G.Z., R.D. and C.M. The in silico ADMET study was performed by R.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant provided by the Romanian Ministry of Education and Research, CNCS—UEFISCDI, project number PN-III-P4-ID-PCE-2020-0371, within PNCDI III.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

The authors acknowledge the infrastructure support from Operational Program Competitiveness 2014–2020, Axis 1, under POC/448/1/1 Research infrastructure projects for public R&D institutions/Sections F 2018, through the Research Center with Integrated Techniques for Atmospheric Aerosol Investigation in Romania (RECENT AIR) project, under grant agreement MySMIS no. 127324. The authors are thankful to UEFISCDI Bucharest, Romania, project PN-III-P1-1.1-TE-2016-1205 and PN-III-P4-ID-PCE-2020-0371, for the financial support provided.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bencini, A.; Lippolis, V. 1,10-Phenanthroline: A versatile building block for the construction of ligands for various purposes. Coord. Chem. Rev. 2010, 254, 2096–2180. [Google Scholar] [CrossRef]
  2. Accorsi, G.; Listorti, A.; Yoosaf, K.; Armaroli, N. 1,10-Phenanthrolines: Versatile building blocks for luminescent molecules, materials and metal complexes. Chem. Soc. Rev. 2009, 38, 1690–1700. [Google Scholar] [CrossRef]
  3. Viganor, L.; Howe, O.; McCarron, P.; McCann, M.; Devereux, M. The Antibacterial Activity of Metal Complexes Containing 1,10-phenanthroline: Potential as Alternative Therapeutics in the Era of Antibiotic Resistance. Curr. Top. Med. Chem. 2017, 17, 1280–1302. [Google Scholar] [CrossRef]
  4. Yoshikawa, N.; Yamazaki, S.; Kakimoto, Y.; Eguchi, S.; Yokoyama, R.; Kanehisa, N.; Tohnai, N.; Nakata, E.; Takashima, H. Emission properties of 1,10-phenanthroline derivatives induced by protonation of a nitrogen atom. J. Mol. Struct. 2021, 1242, 130728. [Google Scholar] [CrossRef]
  5. Sall, C.; Yapi, A.-D.; Desbois, N.; Chevalley, S.; Chezal, J.-M.; Tan, K.; Teulade, J.-C.; Valentin, A.; Blache, Y. Design, synthesis, and biological activities of conformationally restricted analogs of primaquine with a 1,10-phenanthroline framework. Bioorg. Med. Chem. Lett. 2008, 18, 4666–4669. [Google Scholar] [CrossRef] [PubMed]
  6. Nielsen, M.C.; Larsen, A.F.; Abdikadir, F.H.; Ulven, T. Phenanthroline-2,9-bistriazoles as selective G-quadruplex ligands. Eur. J. Med. Chem. 2014, 72, 119–126. [Google Scholar] [CrossRef] [PubMed]
  7. Wesselinova, D.; Neykov, M.; Kaloyanov, N.; Toshkova, R.; Dimitrov, G. Antitumour activity of novel 1,10-phenanthroline and 5-amino-1,10-phenanthroline derivatives. Eur. J. Med. Chem. 2009, 44, 2720–2723. [Google Scholar] [CrossRef]
  8. Leontie, L.; Druta, I.; Rotaru, A.; Apetroaei, N.; Rusu, G.I. Electronic transport properties of some new monoquaternary salts of 4,4′-bipyridine in thin films. Synth. Met. 2009, 159, 642–648. [Google Scholar] [CrossRef]
  9. Castedo, L.; Tojo, G. Phenanthrene Alkaloids. In The Alkaloids; Brossi, A., Ed.; Academic Press Inc.: San Diego, CA, USA, 1990; pp. 99–139. [Google Scholar]
  10. Genès, C.; Lenglet, G.; Depauw, S.; Nhili, R.; Pradoa, S.; David-Cordonnier, M.-H.; Michela, S.; Tillequina, F.; Porée, F.-H. Synthesis and biological evaluation of N-substituted benzo[c]phenanthrolines and benzo[c]phenanthrolinones as antiproliferative agents. Eur. J. Med. Chem. 2011, 46, 2117–2131. [Google Scholar] [CrossRef] [PubMed]
  11. Al Matarneh, C.M.; Amarandi, R.M.; Craciun, A.M.; Mangalagiu, I.I.; Zbancioc, G.; Danac, R. Design, Synthesis, Molecular Modelling and Anticancer Activities of New Fused Phenanthrolines. Molecules 2020, 25, 527. [Google Scholar] [CrossRef] [PubMed]
  12. Zbancioc, G.; Mangalagiu, I.I.; Moldoveanu, C. The Effective Synthesis of New Benzoquinoline Derivatives as Small Molecules with Anticancer Activity. Pharmaceuticals 2024, 17, 52. [Google Scholar] [CrossRef] [PubMed]
  13. Abraham, J.; Staffurth, J. Hormonal therapy for cancer. Medicine 2016, 44, 30–33. [Google Scholar] [CrossRef]
  14. Esfahani, K.; Roudaia, L.; Buhlaiga, N.; Del Rincon, S.V.; Papneja, N.; Miller, W.H., Jr. A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol. 2020, 27 (Suppl. S2), 87–97. [Google Scholar] [CrossRef] [PubMed]
  15. Czogała, W.; Czogała, M.; Kwiecinska, K.; Bik-Multanowski, M.; Tomasik, P.; Hałubiec, P.; Łazarczyk, A.; Miklusiak, K.; Skoczen, S. The Expression of Genes Related to Lipid Metabolism and Metabolic Disorders in Children before and after Hematopoietic StemCell Transplantation—A Prospective Observational Study. Cancers 2021, 13, 3614. [Google Scholar] [CrossRef]
  16. Kinch, M.S. An analysis of FDA–approved drugs for oncology. Drug Discov. Today 2014, 19, 1831–1835. [Google Scholar] [CrossRef] [PubMed]
  17. Magalhaes, L.G.; Ferreira, L.L.G.; Andricopulo, A.D. Recent Advances and Perspectives in Cancer Drug Design. An. Acad. Bras. Cienc. 2018, 90, 1233–1236. [Google Scholar] [CrossRef]
  18. Barreca, M.; Spanò, V.; Raimondi, M.V.; Tarantelli, C.; Spriano, F.; Bertoni, F.; Barraja, P.; Montalbano, A. Recurrence of the oxazolemotif in tubulin colchicine site inhibitors with anti-tumor activity. Eur. J Med. Chem. Rep. 2021, 1, 100004. [Google Scholar] [CrossRef]
  19. Barreca, M.; Spanò, V.; Rocca, R.; Bivacqua, R.; Abel, A.C.; Maruca, A.; Montalbano, A.; Raimondi, M.V.; Tarantelli, C.; Gaudio, E.; et al. Development of [1,2]oxazoloisoindoles tubulin polymerization inhibitors: Further chemical modifications and potential therapeutic effects against lymphomas. Eur. J. Med. Chem. 2022, 243, 114744. [Google Scholar] [CrossRef]
  20. Vieira, A.M.G.; Silvestre, O.F.; Silva, B.F.B.; Ferreira, C.J.O.; Lopes, I.; Gomes, A.C.; Espiña, B.; Sárria, M.P. pH-sensitive nanoliposomes for passive and CXCR-4-mediated marine yessotoxin delivery for cancer therapy. Nanomedicine 2022, 17, 717–739. [Google Scholar] [CrossRef] [PubMed]
  21. Shrivastava, N.; Naim, M.J.; Alam, M.J.; Nawaz, F.; Ahmed, S.; Alam, O. Benzimidazole Scaffold as Anticancer Agent: Synthetic Approaches and Structure-Activity Relationship. Arch. Pharm. 2017, 350, e201700040. [Google Scholar] [CrossRef]
  22. Amin, N.H.; El-Saadi, M.T.; Abdel-Fattah, M.M.; Mohammed, A.A.; Said, E.G. Development of certain aminoquinazoline scaffolds as potential multitarget anticancer agents with apoptotic and anti-proliferative effects: Design, synthesis and biological evaluation. Bioorg. Chem. 2023, 135, 106496. [Google Scholar] [CrossRef]
  23. Ilakiyalakshmi, M.; Arumugam Napoleon, A.A. Review on recent development of quinoline for anticancer activities. Arab. J. Chem. 2022, 15, 104168. [Google Scholar] [CrossRef]
  24. Park, G.Y.; Wilson, J.J.; Song, Y.; Lippard, S.J. Phenanthriplatin, a monofunctional DNA–binding platinum anticancer drugcandidate with unusual potency and cellular activity profile. Proc. Natl. Acad. Sci. USA 2012, 109, 11987–11992. [Google Scholar] [CrossRef]
  25. Zbancioc, G.; Ciobanu, C.-I.; Mangalagiu, I.I.; Moldoveanu, C. Ultrasound-Assisted Synthesis of Fluorescent Azatetracyclic Derivatives: An Energy-Efficient Approach. Molecules 2022, 27, 3180. [Google Scholar] [CrossRef] [PubMed]
  26. Shoemaker, H.R. The NCI60 human tumor cell line anticancer drug screen. Nat. Rev. Cancer 2006, 6, 813–823. [Google Scholar] [CrossRef]
  27. Skehan, P.; Storeng, R.; Scudiero, D.; Monks, A.; McMahon, J.; Vistica, D.; Warren, J.T.; Bokesch, H.; Kenney, S.; Boyd, M.R. Newcolorimetric cytotoxicity assay for anticancer–drug screening. J. Natl. Cancer Inst. 1990, 82, 1107–1112. [Google Scholar] [CrossRef] [PubMed]
  28. Boyd, R.B. The NCI In Vitro Anticancer Drug Discovery Screen. In Anticancer Drug Development Guide; Teicher, B.A., Ed.; Cancer Drug Discovery and Development; Humana Press: Totowa, NJ, USA, 1997; pp. 23–42. [Google Scholar]
  29. The COMPARE Program. Available online: https://dtp.cancer.gov/databases_tools/compare.htm (accessed on 12 January 2025).
  30. Development. Available online: https://next.cancer.gov/developmentResources/default.htm (accessed on 22 October 2024).
  31. Cell Lines in the In Vitro Screen. Available online: https://dtp.cancer.gov/discovery_development/nci-60/cell_list.htm (accessed on 14 November 2024).
  32. The Standard NCI/DTP Methodology of the In Vitro Cancer Screen. Available online: https://dtp.cancer.gov/discovery_development/nci-60/methodology.htm (accessed on 23 October 2024).
  33. Cruz, M.; Duarte-Rodrigues, J.; Campelo, M. Cardiotoxicity in anthracycline therapy: Prevention strategies. Rev. Port. Cardiol. 2016, 35, 359–371. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, S.; Chandra Tjin, C.; Gao, X.; Xue, Y.; Jiao, H.; Zhang, R.; Wu, M.; He, Z.; Ellman, J.; Ha, Y. Pharmacological inhibition of PI5P4Kα/β disrupts cell energy metabolism and selectively kills p53-null tumor cells. Proc. Natl. Acad. Sci. USA 2021, 118, e2002486118. [Google Scholar] [CrossRef]
  35. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef]
  36. Lagunin, A.A.; Dubovskaja, V.I.; Rudik, A.V.; Pogodin, P.V.; Druzhilovskiy, D.S.; Gloriozova, T.A.; Filimonov, D.A.; Sastry, N.G.; Poroikov, V.V. CLC-Pred: A freely available web-service for in silico prediction of human cell line cytotoxicity for drug-like compounds. PLoS ONE 2018, 12, e0191838. [Google Scholar] [CrossRef]
Scheme 1. Design in the class of benzo[f]quinolines derivatives with anticancer activity.
Scheme 1. Design in the class of benzo[f]quinolines derivatives with anticancer activity.
Molecules 30 00702 sch001
Figure 1. Molecular structures of benzo[f]quinoline derivatives.
Figure 1. Molecular structures of benzo[f]quinoline derivatives.
Molecules 30 00702 g001
Figure 2. SAR diagram in the series of benzo[f]pyrrolo[1,2-a]quinoline azatetracyclic derivatives. (A—cycloadduct from methyl propiolate; B—cycloadduct from dimethyl acetylenedicarboxylate).
Figure 2. SAR diagram in the series of benzo[f]pyrrolo[1,2-a]quinoline azatetracyclic derivatives. (A—cycloadduct from methyl propiolate; B—cycloadduct from dimethyl acetylenedicarboxylate).
Molecules 30 00702 g002
Figure 3. SwissADME bioavailability chart of compounds 3b and 5a.
Figure 3. SwissADME bioavailability chart of compounds 3b and 5a.
Molecules 30 00702 g003
Table 1. Single-dose (10−5 M) assay results of the functionalized benzo[f]quinolinium derivatives against 60 NCI human cancer cell lines, expressed as the percentage growth inhibition (PGI%).
Table 1. Single-dose (10−5 M) assay results of the functionalized benzo[f]quinolinium derivatives against 60 NCI human cancer cell lines, expressed as the percentage growth inhibition (PGI%).
Cell TypeCompound/Percentage Growth Inhibition (PGI%) a
1a1b2a3a3b4a5a5b6a6b
Leukemia
CCRF–CEM87656528100 (4)  b42100 (89)  b5968100 (19)  b
HL–60 (TB)51182476110100 (98)  b100 (38)  b3366
K–5625833483696440523566
MOLT–4946878459935100 (99)  b100 (96)  b88100 (93)  b
RPMI–8226622946319729100 (87)  b8455100 (30)  b
SR988787449765-100 (59)  b100 (6)  b100 (97)  b
Non–small Cell Lung Cancer
A549/ATCC6946608500100 (40)  b5160
EKVX4014571140100 (44)  b90100 (1)  b36
HOP–6252254413330100 (20)  b---
HOP–924726460470100 (6)  b561527
NCI–H2264722360390100 (2)  b100 (5)  b4780
NCI–H234917317672100 (2)  b551835
NCI–H322M186114170100 (15)  b0321
NCI–4606160820830100 (88)  b771749
NCI–H52241284027100 (55)  b10934641100 (44)  b
Colon Cancer
COLO 205620450570100 (44)  b35010
HCC–2998380570330100 (9)  b1905
HCT–116652351586977471949
HCT–156139663191189232519
HT295616371477567201324
SW–620453455691296461034
CNS Cancer
SF–26826432031177761647
SF–2956322470220100 (15)  b---
SF–5395727462833100 (1)  b451036
SNB–194517421548072531930
SNB–7510234060100 (8)  b725049
U25161505630771375---
Melanoma
LOX IMVI69585956100 (5)  b2871523664
MDA–MB–4352602117904390010
SK–MEL–20001074061202487
Ovarian Cancer
OVCAR–30024484192342358
OVCAR–869446220851674652644
SK–OV–3389250190100 (61)  b25174
Renal Cancer
786–0481236359081551828
A4980000130100 (47)  b531818
ACHN8331640570100 (7)  b653131
CAKI–1473042329477522353
RXF 3933400068089---
SN12C493347465093702422
TK–10161235042099613343
Prostate Cancer
DU–1453135480290100 (2)  b381014
Breast Cancer
MCF794619530943887635852
HS 578T4031560350100 (19)  b100 (12)  b6176
T–47D432499406423-994842
MDA–MB–468520100 (8)  b15500100 (52)  b---
a values are reported as a one-dose assay; the percentage growth inhibition (PGI) is growth relative to the no-drug control, and relative to the number of cells at time zero; b cytotoxic effect; percentage lethality is shown in brackets; bold highlighted values are the best in the series.
Table 2. Results of the five-dose in vitro human cancer cell growth inhibition a for compounds 3b and 5a and the control, phenstatin.
Table 2. Results of the five-dose in vitro human cancer cell growth inhibition a for compounds 3b and 5a and the control, phenstatin.
Cell TypeGI50 (μM) b
Compound 3bCompound 5aPhenstatin
Leukemia
CCRF–CEM3.9224.70.034
HL–60 (TB)5.3912.70.011
K–5626.48>42.2-
MOLT–43.306.58-
RPMI–82267.4313.6-
SR5.878.32<0.010
Non-small Cell Lung Cancer
A549/ATCC>42.21.41-
EKVX>42.28.77-
HOP–6233.1>42.20.073
HOP–9215.2>42.2-
NCI–H22625.6>42.2-
NCI–H2312.9>42.2-
NCI–H322M>42.212.8-
NCI–46017.83.070.033
NCI–H5223.07>42.20.027
Colon Cancer
COLO 20538.84.184.86
HCC–299839.96.73-
HCT–11611.0>42.20.038
HCT–156.17>42.2<0.010
HT2920.6>42.22.95
KM12>42.2>42.2<0.010
SW–6207.26>42.2<0.010
CNS Cancer
SF–26834.4>42.2-
SF–29528.3>42.20.367
SF–53910.9>42.20.011
SNB–19>42.2>42.2-
SNB–7524.7>42.2<0.010
U25118.3>42.20.043
Melanoma
LOX IMVI4.35>42.20.013
MALME–3M6.10>42.2-
M1431.3>42.2<0.010
MDA–MB–43516.1>42.2<0.010
SK–MEL–217.9>42.20.520
SK–MEL–2825.7>42.265.20
SK–MEL–513.0>42.20.040
UACC–257>42.2>42.2-
UACC–6211.9>42.20.448
Ovarian Cancer
IGROV133.2>42.20.18
OVCAR–316.9>42.20.021
OVCAR–425.6>42.2-
OVCAR–522.5>42.2-
OVCAR–814.9>42.20.042
NCI/ADR–RES17.7>42.20.012
SK–OV–3>42.211.9-
Renal Cancer
786–021.1>42.20.905
A498>42.25.712.28
ACHN22.2>42.20.042
CAKI–121.227.8-
RXF 3932.79>42.20.016
SN12C17.229.8-
TK–10>42.2>42.2-
UO–3124.9>42.20.074
Prostate cancer
PC–317.8>42.20.045
DU–145>42.225.30.039
Breast cancer
MCF74.5824.50.033
MDA–MB–231/ATCC4.72>42.20.029
HS 578T18.8>42.20.031
BT–5496.18>42.20.034
T–47D11.518.130.4
MDA–MB–46813.27.252.71
a the data were obtained from NCI’s in vitro 60 cell five-dose screening. b GI50—the molar concentration of the tested compound causing the 50% growth inhibition of tumor cells. The GI50 value was no longer determined if it was more than 42.2 µM. Bold highlighted values are the best in the series.
Table 3. In silico prediction of ADME parameters for compounds 3b and 5a.
Table 3. In silico prediction of ADME parameters for compounds 3b and 5a.
ADME Parameter3b5a
Physicochemical Properties
Molecular weight468.30 g/mol358.35 g/mol
Log Po/w (MLOGP)3.241.94
Number of H-bond acceptors56
Number of H-bond donors00
Number of rotatable bonds65
TPSA74.08 Å297.53 Å2
Pharmacokinetics
Gastrointestinal (GI) absorptionhighhigh
Blood–brain barrier (BBB) permeantnono
P-gp substratenono
Drug-likeness
Log S (ESOL)−6.55−6.02
Water solubility classpoorly solublepoorly soluble
Lipinski ruleno violationno violation
Veber ruleno violationno violation
Bioavailability0.550.55
Medicinal Chemistry
PAINS alerts01 alert: azo
Brenk alerts3 alerts: alkyl_halide, more than 2 esters, polycyclic
aromatic hydrocarbon
4 alerts: azido_group, diazo_group, polycyclic aromatic hydrocarbon, quaternary nitrogen
Synthetic accessibility3.652.54
Table 4. Results of the prediction for cytotoxicity of compounds 3b and 5a.
Table 4. Results of the prediction for cytotoxicity of compounds 3b and 5a.
3b
PaPiCell LineCell Type
0.5670.001MOLT-3T-lymphoblastic leukemia
0.3810.033CCRF-CEMChildhood T acute lymphoblastic leukemia
0.4020.069NCI-H187Small cell lung carcinoma
5a
PaPiCell LineCell Type
0.4370.019SF-539Glioblastoma
0.4330.021UACC-62Melanoma
0.4190.028SN12CRenal carcinoma
0.4190.028HOP-62Non-small cell lung carcinoma
0.3790.036OVCAR-3Ovarian adenocarcinoma
0.3180.060T47DBreast carcinoma
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

Moldoveanu, C.; Mangalagiu, I.I.; Zbancioc, G.; Danac, R.; Tataringa, G.; Zbancioc, A.M. Anticancer Potential of Azatetracyclic Derivatives: In Vitro Screening and Selective Cytotoxicity of Azide and Monobrominated Compounds. Molecules 2025, 30, 702. https://doi.org/10.3390/molecules30030702

AMA Style

Moldoveanu C, Mangalagiu II, Zbancioc G, Danac R, Tataringa G, Zbancioc AM. Anticancer Potential of Azatetracyclic Derivatives: In Vitro Screening and Selective Cytotoxicity of Azide and Monobrominated Compounds. Molecules. 2025; 30(3):702. https://doi.org/10.3390/molecules30030702

Chicago/Turabian Style

Moldoveanu, Costel, Ionel I. Mangalagiu, Gheorghita Zbancioc, Ramona Danac, Gabriela Tataringa, and Ana Maria Zbancioc. 2025. "Anticancer Potential of Azatetracyclic Derivatives: In Vitro Screening and Selective Cytotoxicity of Azide and Monobrominated Compounds" Molecules 30, no. 3: 702. https://doi.org/10.3390/molecules30030702

APA Style

Moldoveanu, C., Mangalagiu, I. I., Zbancioc, G., Danac, R., Tataringa, G., & Zbancioc, A. M. (2025). Anticancer Potential of Azatetracyclic Derivatives: In Vitro Screening and Selective Cytotoxicity of Azide and Monobrominated Compounds. Molecules, 30(3), 702. https://doi.org/10.3390/molecules30030702

Article Metrics

Back to TopTop