Next Article in Journal
Effects of Statin Dose, Class, and Use Intensity on All-Cause Mortality in Patients with Type 2 Diabetes Mellitus
Previous Article in Journal
Pharmacotherapy for Pulmonary Hypertension in Infants with Bronchopulmonary Dysplasia: Past, Present, and Future
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Cytotoxic Activity of α-Aminophosphonic Derivatives Coming from the Tandem Kabachnik–Fields Reaction and Acylation

1
Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, 1521 Budapest, Hungary
2
ELKH-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University (ELTE), 1117 Budapest, Hungary
3
Department of Genetics, Cell and Immunobiology, Semmelweis University, 1089 Budapest, Hungary
4
TargetEx Biosciences, Ltd., 2120 Dunakeszi, Hungary
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(4), 506; https://doi.org/10.3390/ph16040506
Submission received: 24 February 2023 / Revised: 17 March 2023 / Accepted: 27 March 2023 / Published: 28 March 2023
(This article belongs to the Topic Advances in Anti-Cancer Drugs)

Abstract

:
Encouraged by the significant cytotoxic activity of simple α-aminophosphonates, a molecular library comprising phosphonoylmethyl- and phosphinoylmethyl-α-aminophosphonates, a tris derivative, and N-acylated species was established. The promising aminophosphonate derivatives were subjected to a comparative structure–activity analysis. We evaluated 12 new aminophosphonate derivatives on tumor cell cultures of different tissue origins (skin, lung, breast, and prostate). Several derivatives showed pronounced, even selective cytostatic effects. According to IC50 values, phosphinoylmethyl-aminophosphonate derivative 2e elicited a significant cytostatic effect on breast adenocarcinoma cells, but it was even more effective against prostatic carcinoma cells. Based on our data, these new compounds exhibited promising antitumor activity on different tumor types, and they might represent a new group of alternative chemotherapeutic agents.

1. Introduction

Within organophosphorus compounds [1,2], α-aminophosphonic derivatives are of special importance due to their real and potential biological activity. This is not surprising if the structural analogy between α-aminoalkylphosphonic acids and α-aminocarboxylic acids is considered [3,4,5]. The α-aminophosphonic acid derivatives possess different properties, e.g., they may inhibit enzymes and GABA-receptors, and they may be anti-metabolites [6]. As a consequence, the compounds under discussion may reveal anticancer [7], antibiotic [6], antiviral and HIV [8], anti-inflammatory [9], antimalarial [10], antiasthma [11], antidiabetic [12], as well as antihypertensive effects [13]. Diaryl α-aminophosphonate derivatives are selective and highly potent inhibitors of serine proteases and, hence, can mediate the patho–physical processes of cancer growth, metastasis, osteoarthritis, or heart failure [14].
Various α-aminophosphonate derivatives were investigated and identified as antiproliferative and/or potential anticancer agents. While the diaryl or dialkyl phosphonate ester unit remained unchanged, in most of the cases, different bioactive moieties, such as coumarin [15] or peptidomimetic structures [16], were linked to the amino group. Alternatively, the secondary carbon atom was substituted with heterocycles [17] or nucleobases [18]. In addition, the introduction of trifluoromethyl groups into the arylamino group increased the antiproliferative and apoptosis-inducing properties [19].
The α-aminophopshonic derivatives may be prepared by the Kabachnik–Fields condensation of primary or secondary amines, oxo compounds, such as aldehyde or ketones, and dialkyl phosphites or related derivatives [20,21,22,23]. The three-component reaction has been an excellent model for green chemical studies [24,25,26,27,28]. A modification of the Kabachnik–Fields condensation is the bis(phospha-Mannich) reaction when a primary amine interacts with two equivalents of an aldehyde and a similar quantity of the >P(O)H reactant to afford the corresponding bis derivatives [29,30,31,32]. A special variation is when an α-aminophosphonic derivative is reacted further with another aldehyde and >P(O)H reagent to furnish a phosphonoylmethyl-α-aminophosphonate or a phosphinoylmethyl-α-aminophosphonate [33].
In this article, we wish to report the cytotoxic activity of our products prepared using the tandem Kabachnik–Fields reaction [33]. Even the starting α-aminophosphonates revealed considerable anticancer effects [34].

2. Results and Discussion

2.1. Synthesis of α-Aminophosphonate Derivatives

The α-benzylamino-benzylphosphonate derivatives (1ac) obtained in the Kabachnik–Fields reaction of substituted benzaldehydes, benzylamine, and diethyl phosphite [33] were reacted further in another phospha-Mannich condensation applying paraformaldehyde and diethyl phosphite or secondary phosphine oxide to afford phosphonoylmethyl- or phosphinoylmethyl-benzylamino-benzylphosphonates (2af) (Scheme 1) [33]. Compound 1d (X = MeO) was also prepared from anisic aldehyde but was not reacted further. Aminophosphonate 1e was obtained using aniline instead of benzylamine in condensation with benzaldehyde and diethyl phosphite.
A debenzylation of benzylaminophosphonate 1a afforded aminophosphonate 3 [33] that was converted to acylated derivatives 4a and 4b (Scheme 2) [33].
A bis(α,α’-phosphonoylbenzyl)amine (5) was prepared starting from benzaldehyde, ammonium acetate, and diethyl phosphite (Scheme 3) [33].
α-Amino-benzylphosphonate 3 was a suitable starting material also for tris(phosphonoylmethyl)amine derivative 6 (Scheme 4) [33].
The compounds subjected to bioactivity tests are shown in Figure 1.

2.2. In Vitro Cytostatic Effect of α-Aminophosphonic Derivatives on Human Tumor Cell Lines

From among the above synthesized α-aminophosphonate derivatives, the cytotoxic activity of N-benzyl- and N-phenyl α-aminophopshonates 1be, phosphonoyl-α-aminophosphonates 2a and 2b, phopshinoyl-α-aminophosphonates 2df, as well as α-(acylamino)phosphonates 4a and 4b, along with bis(diethylphosphonoylphenylmethyl)amine 5 and tris derivative 6 were subjected to investigations on different cancer cells.
To provide structure–activity relations, the in vitro cytostatic activity was evaluated on four human cell lines of different origins: MDA-MB-231 human breast adenocarcinoma [35], A431 human epidermoid carcinoma [36], PC-3 human prostate adenocarcinoma [37], and Ebc-1 human lung squamous cell carcinoma [38].
Dose dependence of the cytostatic effect was studied by treating the cells with the compounds for 24 h, and after removing the active agents, the cells were cultured for another 72 h. The cytostatic effect was expressed in the percentage of the untreated control (Table 1 and Table 2). Compound 6 was insoluble in even DMSO and the aqueous medium applied; therefore, it was excluded from in vitro experiments.
We found that at c = 50 µM, compounds 2a and 2b elicited the most pronounced cytostatic effect on the A431 cell line (cytostasis = 43.7 ± 3.5% and 45.1 ± 6.9%, respectively) (Table 2). Compound 2f proved to be the most cytostatic on both A431 (92.8 ± 0.3%) and Ebc-1 cells (88.5 ± 0.3%). At c = 250 µM, the compound that killed the cells to the greatest extent was 2f (cytostasis = 90.6 ± 0.3% on MDA-MB 231 cells, 92.8 ± 0.3% on A431 cells, 87.3 ± 0.7% on PC-3 cells, and 90.4 ± 0.3%on Ebc-1 cells at c = 250 um). In general, we can say that A431 human epidermoid carcinoma and Ebc-1 human lung squamous cell carcinoma cells were the most sensitive to the treatment, as most of the compounds already showed a significant cytostatic effect on these cells at a concentration of 50 µM (Table 1 and Table 2).
For those compounds where the cytostatic effect exceeded 50% in certain concentrations, the IC50 value that was characteristic of the compound on the appropriate cell line was also determined (Table 3). The cell line on which most of the compounds showed a cytostatic effect over 50% was MDA-MB 231 human breast carcinoma. Considering the IC50 values, the most effective was phosphinoylmethyl-aminophosphonate 2e on MDA-MB 231 cells (IC50 = 55.1 µM) and PC-3 cells (IC50 = 29.4 µM); its phenyl analog 2d on MDA-MB 231 cells (IC50 = 45.8 µM), and phosphonoylmethyl-aminophosphonate 2b on A431 cell line (IC50 = 53.2 µM). The determined IC50 values were also below 100 µM in the case of the following >P(O)CH2-aminophosphonates: 2f (on MDA-MB 231 and Ebc-1 cells), 2b (on PC-3 cells), and 2a (on A431 cells).
We investigated the in vitro cytostatic effect of 12 novel α-aminophosphonic derivatives on four human tumor cell lines of different tissue origins (skin, lung, breast, and prostate). To compare the in vitro activity, we employed the end-point type MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay [39,40,41]. We determined concentration-dependent and cell-selective effects. At a lower concentration (c = 50 µM), Ebc-1 human lung squamous cell carcinoma and A431 human epidermoid carcinoma cells proved to be more sensitive, whereas, in higher concentration (c = 250 µM), MDA-MB-231 human breast adenocarcinoma and PC-3 human prostate adenocarcinoma cells were inhibited to the greatest extent. According to IC50 data, phosphinoylmethyl-aminophosphonate derivative 2e elicited a significant cytostatic effect on MDA-MB 231 breast adenocarcinoma cell line, but it was slightly even more effective against PC-3 cells. 2e Compound and its phenyl analog (2d) showed almost the same cytostatic effect on MDA-MB 231 cells. Compared to the previous studies [42,43,44,45,46], IC50 values of those derivatives obtained from the 72 h cytotoxicity data moved in the same range (22.9–352.9 µM) as the cytostasis data we determined after a 24 h treatment and a subsequent 72 h culturing (29.4–169.2 µM).
It is a challenge for us to continue exploring the cytotoxic effect of newer α-aminophospohonates, especially those of optically active derivatives. In order to be able to fulfill this plan, suitable resolution methods have to be elaborated. So far, we have been successful in separating racemic α-hydroxyphosphonates into their enantiomers [47].

2.3. In Silico Target Assessment of the Major Cytotoxic Hit Compounds

Since the mid-2000s, searchable annotated molecular databases have been available that contain biological activities, frequently together with the protein targets they act on [48].
BindingDB [49] (https://www.bindingdb.org, accessed on 31 January 2023), currently containing 107,154 active compounds with biological data, gives the opportunity to predict the protein targets of compounds found active in cellular assays based on the “similar structure—similar property” principle. Some recent examples from the literature where similarity search was used for initial target assessment are in [50,51,52]. We can identify structurally similar compounds that interact with numerous protein targets, and further analysis would reveal the putative protein targets of cell-based hit compounds.
The similarity search on the best aminophosphonate cytotoxic hit compound (2b, 2d, 2e) was carried out using InstantJChem software (ChemAxon, Budapest, Hungary) (Figure 2), which uses 2D molecular fingerprints for comparison. Compounds were considered similar if the Tanimoto coefficient was ≥0.5. Although 0.5 represents a moderate similarity threshold, it provides a broad preliminary target pool for more specific and detailed analysis. Of course, in vitro biological screening could only validate the prediction.
The number of bioactivity data with putative targets is, all together: 71 (2b: 58, 2d/2e: 25). (The lower number is due to the overlap of findings) (Table 4).
The resulting putative protein targets can be analyzed and attempted to link to disease states represented by the cell lines (e.g., MDA-MB-231 human breast, PC-3 prostate, and A431 skin cancer) where the best aminophosphonate cytotoxic hits were identified. Thus, at first, the predicted targets were classified as either cancer or non-cancer-associated targets. Secondly, the cancer-related targets were further investigated to determine whether the cell lines express or even overexpress such proteins and whether their overexpression could promote the progression of the tumor. All the identified cancer-related targets meet the specific OncoScore criteria and are listed among the cancer-associated genes (see Supplementary Materials of Ref. [53]). The small molecules that inhibit any of such targets could contribute to decreasing tumor growth as the aminophosphonate hits expressed cytotoxic/antiproliferative effects on the cancer cell lines.
The similar compounds related to our cytotoxic hits, together with their annotated protein targets, are shown in Table 5.
Short description of the putative targets.
  • Furin
Proprotein convertases, including furin, have been implicated in the activation of a wide variety of various biological pathways and could facilitate tumor formation and progression. The expression of furin has been confirmed in various cancers such as head and neck cell carcinoma, breast cancer, etc. [53].
Several peptidomimetic and related small molecule inhibitors have been identified over the years. In general, phosphates and phosphonates could often mimic the peptide bonds; thus, they are replaceable in peptidomimetic inhibitor design.
  • Prostatic Acid Phosphatase
Prostatic acid phosphatase (PAP) originates in the prostate and is normally present in small amounts in the blood. Elevated levels of PAP in patients indicate the progression of various tumors such as prostate cancer [55], testicular cancer, leukemia, etc. Various α-benzylaminobenzylphosphonic acid derivatives have been developed over the years using structure-based drug design methods that led to the identification of low nanomolar inhibitors [56].
  • Tyrosine Phosphatase Enzyme Family
The protein tyrosine phosphatase enzyme family (PTPs) removes phosphate groups from proteins. PTPs are considered potential drug targets against several diseases, including cancer [57], obesity, diabetes, etc. [58]. Several PTPs overexpressed in human cancers could act as targets (oncogene) or “anti-targets” in tumor therapy since they could also suppress tumor progression upon overexpression [59].
Based on recent data, the most important anticancer target is PTPN11 (SHP2). Overexpression of SHP2 is associated with an increased risk of leukemia [60], breast cancer [61], and skin cancer cell lines [62]. Several PTPN11 (SHP2) inhibitors have been developed so far, with an increasing focus on selectivity toward the anti-target PTPs [63]. Most of the 2nd generation compounds lack P-containing moieties; however, their structure partially mimics the transition state complex of the enzyme–substrate interaction.
In summary, based on the above in silico target prediction, we could conclude that our hit compounds that showed cytotoxicity against breast, prostate, and skin cancer cell lines might act through the above-described protein targets by applying the similarity principle. Detailed molecular dynamics and docking studies and, most importantly, target-based bioassays would confirm the above prediction. These future efforts are out of the scope of the present paper.

3. Materials and Methods

3.1. Synthesis of α-Aminophosphonate Derivatives

The phosphonoylmethyl- and phosphinoylmethyl-benzylamino-benzylphosphonates 2af, acylated derivatives 4a and 4b, along with bis- and tris derivatives 5 and 6, respectively, were synthesized as described earlier [32].

3.2. Cell Lines and Culture Conditions—In Vitro Cytotoxicity Assays of Carcinoma Cell Lines

In vitro cytostatic effect of the compounds was determined on MDA-MB-231 human breast adenocarcinoma [36], A431 human epidermoid carcinoma [37], PC-3 human prostate adenocarcinoma [38], and Ebc-1 human lung squamous cell carcinoma [39] cell lines. Cells were cultured in DMEM medium supplemented with 10% FBS, 2 mM L-glutamine, 50 IU/mL penicillin/50 μg/mL streptomycin antibiotic cocktail, 1 mM sodium pyruvate, and 1% non-essential amino acid mixture at 37 °C in a humidified atmosphere with 5% CO2. At the confluent state, cells were plated into 96-well tissue culture plates with the initial cell number of 5.0 × 103 cells/well. Cells were treated with the compounds after 24 h in serum-free medium containing 1.0 v/v% DMSO at 2, 10, 50, and 250 μM concentrations. Control cells were treated with serum-free medium only or with DMSO (c = 1.0 v/v %) at the same conditions. After overnight incubation, cells were washed twice with a serum-free medium; then, they were cultured for another 72 h in complete culturing medium at 37 °C. Following that, MTT-solution (at c = 0.37 mg/mL final concentration) was added to each well. The respiratory chain [40] and other electron transport systems [41] reduce MTT and, thereby, form non-water-soluble violet formazan crystals within the cell [42]. After 3 h of incubation with MTT, the cells were centrifuged for 5 min at 900× g, and then the supernatant was removed. The obtained formazan crystals were dissolved in DMSO (100 µL), and the optical density (OD) of the samples was measured at λ = 540 nm and 620 nm, respectively, using ELISA Reader (iEMS Reader, Labsystems, Vantaa, Finland). OD620 values were subtracted from OD540 values. The amount of these crystals serves as an estimate for the number of mitochondria and, hence, the number of living cells in the well [43]. The percent of cytostasis was calculated with the following equation:
Cytostatic effect (%) = [1−(ODtreated/ODcontrol)] × 100
where values ODtreated and ODcontrol corresponds to the optical densities of the treated and the control wells, respectively. In each case, two independent experiments were carried out with 4 parallel measurements; statistical analysis of data was performed using Student’s t-test at the 95% confidence level. The 50% inhibitory concentration (IC50) values were determined from the dose–response curves, which were defined using Microcal™ Origin 2018 software: cytostasis was plotted as a function of concentration, on which a sigmoidal curve was fitted. Based on this curve, the half-maximal inhibitory concentration (IC50) value was determined, which was expressed in micromolar units.

4. Conclusions

We have designed, synthesized, and in vitro evaluated 12 novel aminophosphonate derivatives, the majority of which comprised species with two P-functions. The data from cytostatic activity on skin, lung, breast, and prostate tissue-originated tumor cultures revealed that these compounds could be classified as new α-aminophosphonic derivatives with promising antitumor properties due to the pronounced and selective cytostatic effect. Our results are consistent with the observations of the past two decades, which proved that some α-aminophosphonic derivatives could have antitumor and genotoxic effects on different tumor cell cultures in vitro. According to IC50 values, phosphinoylmethyl-aminophosphonate 2e elicited a remarkable cytostatic effect on breast adenocarcinoma and on prostatic carcinoma cells. Moreover, our study revealed that Ebc-1, MDA-MB231, and A431 carcinoma cultures were generally sensitive to the treatments with the compounds, and there was significant inhibition detected at the 50–100 μM concentration range.
In silico target prediction proposed that the identified cytotoxic compounds would act through the interaction with furin, prostatic acid phosphatase, and tyrosine phosphatase protein targets that are highly expressed in the tumor cell lines used in the present study.

Author Contributions

Conceptualization, G.K., S.B. and G.D.; methodology, P.R.V., R.O.S. and S.B.; investigation, P.R.V., R.O.S. and S.B; resources, G.K. and S.B.; data curation, R.O.S., S.B. and G.D.; writing—original draft preparation, G.K., R.O.S., S.B. and G.D.; writing—review and editing, G.K., S.B. and G.D.; supervision, G.K. and S.B.; project administration, G.K.; funding acquisition, G.K. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by the National Research, Development, and Innovation Office (K134318). Project no. RRF-2.3.1-21-2022-00015 was implemented with the support provided by the European Union.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Acknowledgments

The assistance of Éva Sári in the preparative work is acknowledged. We are grateful for the availability of the following software (InstJChem, JChemforExcel, ChemAxon, Budapest).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Quin, L.D. A Guide to Organophosphorus Chemistry; Wiley & Sons: New York, NY, USA, 2000; ISBN 978-0-471-31824-8. [Google Scholar]
  2. Keglevich, G. (Ed.) Organophosphorus Chemistry—Novel Developments; De Gruyter: Berlin, Germany, 2018; ISBN 978-3-11-053453-5. [Google Scholar]
  3. Abdou, M.M. Synopsis of recent synthetic methods and biological applications of phosphinic acid derivatives. Tetrahedron 2020, 76, 131251. [Google Scholar] [CrossRef]
  4. Rodriguez, J.B.; Gallo-Rodriguez, C. The role of the phosphorus atom in drug design. ChemMedChem 2019, 14, 190–216. [Google Scholar] [CrossRef] [Green Version]
  5. Sevrain, C.M.; Berchel, M.; Couthon, H.; Jaffres, P.-A. Phosphonic acid: Preparation and applications. Beilstein J. Org. Chem. 2017, 13, 2186–2213. [Google Scholar] [CrossRef]
  6. Mucha, A.; Kafarski, P.; Berlicki, L. Remarkable potential of the α-aminophosphonate/phosphinate structural motif in medicinal chemistry. J. Med. Chem. 2011, 54, 5955–5980. [Google Scholar] [CrossRef] [PubMed]
  7. Kafarski, P.; Lejczak, B. Aminophosphonic acids of potential medical importance. Curr. Med. Chem. Anticancer Agents 2001, 1, 301–312. [Google Scholar] [CrossRef]
  8. Kang, S.-U.; Shi, Z.-D.; Worthy, K.M.; Bindu, L.K.; Dharmawardana, P.G.; Choyke, S.J.; Bottaro, D.P.; Fisher, R.J.; Burke, T.R., Jr. Examination of phosphoryl-mimicking functionalities within a macrocyclic Grb2 SH2 domain-binding platform. J. Med. Chem. 2005, 48, 3945–3948. [Google Scholar] [CrossRef] [Green Version]
  9. Robbins, B.L.; Srinivas, R.V.; Kim, C.; Bischofberger, N.; Fridland, A. Anti-human immunodeficiency virus activity and cellular 372 metabolism of a potential prodrug of the acyclic nucleoside phosphonate 9-R-(2-phosphonomethoxypropyl)adenine (PMPA), bis(isopropyloxymethylcarbonyl)PMPA. Antimicrob. Agents Chemother. 1998, 42, 612–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Lassaux, P.; Hamel, M.; Gulea, M.; Delbruck, H.; Mercuri, P.S.; Horsfall, L.; Dehareng, D.; Kupper, M.; Frere, J.-M.; Hoffmann, K.; et al. Mercaptophosphonate compounds as broad-spectrum inhibitors of the metallo-β-lactamases. J. Med. Chem. 2010, 53, 4862–4876. [Google Scholar] [CrossRef] [PubMed]
  11. Haemers, T.; Wiesner, J.; Van Poecke, S.; Goeman, J.; Henschker, D.; Beck, E.; Jomaa, H.; Van Calenbergh, S. Synthesis of α-substituted fosmidomycin analogues as highly potent Plasmodium falciparum growth inhibitors. Bioorg. Med. Chem. Lett. 2006, 16, 1888–1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Maryanoff, B.E. Inhibitors of serine proteases as potential therapeutic agents: The road from thrombin to tryptase to cathepsin G. J. Med. Chem. 2004, 47, 769–787. [Google Scholar] [CrossRef]
  13. Sienczyk, M.; Oleksyszyn, J. Irreversible inhibition of serine proteases—Design and in vivo activity of diaryl alpha-aminophosphonate derivatives. Curr. Med. Chem. 2009, 16, 1673–1687. [Google Scholar] [CrossRef]
  14. Combs, A.P. Recent advances in the discovery of competitive protein tyrosine phosphatase 1B inhibitors for the treatment of diabetes, obesity, and cancer. J. Med. Chem. 2010, 53, 2333–2344. [Google Scholar] [CrossRef]
  15. Li, Y.J.; Wang, C.Y.; Ye, M.Y.; Yao, G.Y.; Wang, H.S. Novel coumarin-containing aminophosphonatesas antitumor agent: Synthesis, cytotoxicity, DNA-binding and apoptosis evaluation. Molecules 2015, 20, 14791–14809. [Google Scholar] [CrossRef] [Green Version]
  16. Loredo-Calderón, E.L.; Velázquez-Martínez, C.A.; Ramírez-Cabrera, M.A.; Her-nández-Fernández, E.; Rivas-Galindo, V.M.; Arredondo Espinoza, E.; López-Cortina, S.T. Synthesis of novel α-aminophosphonates under microwave irradiation, biological evaluation as antiproliferative agents and apoptosis inducers. Med. Chem. Res. 2019, 28, 2067–2078. [Google Scholar] [CrossRef]
  17. Yu, Y.C.; Kuang, W.B.; Huang, R.Z.; Fang, Y.L.; Zhang, Y.; Chen, Z.F.; Ma, X.L. Design, synthesis and pharmacological evaluation of new 2-oxo-quinoline derivatives containing α-aminophosphonates as potential antitumor agents. MedChemComm 2017, 8, 1158–1172. [Google Scholar] [CrossRef] [PubMed]
  18. Bahrami, F.; Panahi, F.; Daneshgar, F.; Yousefi, R.; Shahsavani, M.B.; Khalafi-Nezhad, A. Synthesis of new α-aminophosphonate derivatives incorporating benzimidazole, theophylline and adenine nucleobases using l-cysteine functionalized magnetic nanoparticles (LCMNP) as magnetic reusable catalyst: Evaluation of their anticancer properties. RSC Adv. 2016, 6, 5915–5924. [Google Scholar] [CrossRef]
  19. Deshmukh, S.U.; Kharat, K.R.; Yadav, A.R.; Shisodia, S.U.; Damale, M.G.; Sangshetti, J.N.; Pawar, R.P. Synthesis of Novel α-Aminophosphonate Derivatives, Biological Evaluation as Potent Antiproliferative Agents and Molecular Docking. ChemistrySelect 2018, 3, 5552–5558. [Google Scholar] [CrossRef]
  20. Kukhar, V.P.; Hudson, H.R. Aminophosphonic and Aminophosphinic Acids: Chemistry and Biological Activity; John Wiley & Sons: Chichester, UK, 2000; ISBN 0471891495. [Google Scholar]
  21. Shastri, R.A. Review on the synthesis of α-aminophosphonate derivatives. Chem. Sci. Trans. 2019, 8, 359–367. [Google Scholar] [CrossRef] [Green Version]
  22. Keglevich, G.; Bálint, E. The Kabachnik–Fields reaction; mechanism and synthetic use. Molecules 2012, 17, 12821–12835. [Google Scholar] [CrossRef] [Green Version]
  23. Varga, P.R.; Keglevich, G. Synthesis of α-aminophosphonates and related derivatives; The last decade of the Kabachnik–Fields reaction. Molecules 2021, 26, 2511. [Google Scholar] [CrossRef] [PubMed]
  24. Kafarski, P.; Gorniak, M.G.; Andrasiak, I. Kabachnik-Fields reaction under green conditions—A critical overview. Curr. Green Chem. 2015, 2, 218–222. [Google Scholar] [CrossRef]
  25. Ranu, B.C.; Hajra, A. A simple and green procedure for the synthesis of α-aminophosphonate by a one-pot three-component condensation of carbonyl compound, amine and diethyl phosphite without solvent and catalyst. Green Chem. 2002, 4, 551–554. [Google Scholar] [CrossRef]
  26. Kabachnik, M.M.; Zobnina, E.V.; Beletskaya, I.P. Catalyst-free microwave-assisted synthesis of α-aminophosphonates in a three-component system: R1C(O)R2-(EtO)2P(O)H-RNH2. Synlett 2005, 2005, 1393–1396. [Google Scholar] [CrossRef]
  27. Mu, X.-J.; Lei, M.-Y.; Zou, J.-P.; Zhang, W. Microwave-assisted solvent-free and catalyst-free Kabachnik–Fields reactions for α-amino phosphonates. Tetrahedron Lett. 2006, 47, 1125–1127. [Google Scholar] [CrossRef]
  28. Keglevich, G.; Szekrényi, A. Eco-friendly accomplishment of the extended Kabachnik–Fields reaction; a solvent- and catalyst-free microwave-assisted synthesis of α-aminophosphonates and α-aminophosphine oxides. Lett. Org. Chem. 2008, 5, 616–622. [Google Scholar] [CrossRef]
  29. Bálint, E.; Fazekas, E.; Pinter, G.; Szöllősy, A.; Holczbauer, T.; Czugler, M.; Keglevich, G. Synthesis and utilization of the bis(> P(O)CH2)amine derivatives obtained by the double Kabachnik–Fields reaction with cyclohexylamine; Quantum chemical and X-ray study of the related bidentate chelate platinum complexes. Curr. Org. Chem. 2012, 16, 547–554. [Google Scholar] [CrossRef]
  30. Bálint, E.; Fazekas, E.; Pongrácz, P.; Kollár, L.; Drahos, L.; Holczbauer, T.; Czugler, M.; Keglevich, G. N-Benzyl and N-aryl bis(phospha-Mannich adducts): Synthesis and catalytic activity of the related bidentate chelate platinum complexes in hydroformylation. J. Organomet. Chem. 2012, 717, 75–82. [Google Scholar] [CrossRef]
  31. Bálint, E.; Fazekas, E.; Drahos, L.; Keglevich, G. The synthesis of N,N-bis(dialkoxyphosphinoylmethyl)- and N,N-bis(diphenylphosphinoylmethyl)glycine esters by the microwave-assisted double Kabachnik–Fields reaction. Heteroatom Chem. 2013, 24, 510–515. [Google Scholar] [CrossRef]
  32. Bálint, E.; Tripolszky, A.; Hegedűs, L.; Keglevich, G. Microwave-assisted synthesis of N,N-bis(phosphinoylmethyl)amines and N,N,N-tris(phosphinoylmethyl)amines bearing different substituents on the phosphorus atoms. Belstein J. Org. Chem. 2019, 15, 469–473. [Google Scholar] [CrossRef] [Green Version]
  33. Varga, P.R.; Karaghiosoff, K.; Sári, É.V.; Simon, A.; Hegedűs, L.; Drahos, L.; Keglevich, G. New N-acyl-, as well as N-phosphonoylmethyl- and N-phoshinoylmethyl-α-amino-benzylphosphonates by acylation and a tandem Kabachnik–Fields protocol. Org. Biomol. Chem. 2023, 21, 1709–1718. [Google Scholar] [CrossRef]
  34. Varga, P.R.; Dinnyési, E.; Tóth, S.; Szakács, G.; Keglevich, G. Optimized synthesis and cytotoxic activity of α-aminophosphonates against a multidrug resistant uterine sarcoma cell line. Lett. Drug Des. Discov. 2023, 20, 365–371. [Google Scholar] [CrossRef]
  35. Cailleau, R.; Olivé, M.; Cruciger, Q.V. Long-term human breast carcinoma cell lines of metastatic origin: Preliminary characterization. In Vitro 1978, 14, 911–915. [Google Scholar] [CrossRef]
  36. Haigler, H.; Ash, J.F.; Singer, S.J.; Cohen, S. Visualization by fluorescence of the binding and internalization of epidermal growth factor in human carcinoma cells A-431. Proc. Natl. Acad. Sci. USA 1978, 75, 3317–3321. [Google Scholar] [CrossRef] [Green Version]
  37. Kaighn, M.E.; Narayan, K.S.; Ohnuki, Y.; Lechner, J.F.; Jones, L.W. Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Investig. Urol. 1979, 17, 16–23. [Google Scholar]
  38. Fujita, T.; Kiyama, M.; Tomizawa, Y.; Kohno, T.; Yokota, J. Comprehensive analysis of p53 gene mutation characteristics in lung carcinoma with special reference to histological subtypes. Int. J. Oncol. 1999, 15, 927–961. [Google Scholar] [CrossRef]
  39. Slater, T.F.; Sawyerand, B.; Strauli, U. Studies on succinate-tetrazolium reductase systems: III. Points of coupling of four different tetrazolium salts III. Points of coupling of four different tetrazolium salts. Biochim. Biophys. Acta 1963, 77, 383–393. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, Y.B.; Peterson, D.A.; Kimuraand, H.; Schubert, D.J. Mechanism of cellular 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J. Neurochem. 1997, 69, 581–593. [Google Scholar] [CrossRef] [PubMed]
  41. Altman, F.P. Tetrazolium salts and formazans. Prog. Histochem. Cytochem. 1976, 9, 1–56. [Google Scholar] [CrossRef] [PubMed]
  42. Denizot, F.; Lang, R.J. Rapid colorimetric assay for cell growth and survival. Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J. Immunol. Methods 1986, 89, 271–277. [Google Scholar] [CrossRef]
  43. Orsini, F.; Sello, G.; Sisti, M. Aminophosphonic acids and derivatives. Synthesis and biological applications. Curr. Med. Chem. 2010, 17, 264–289. [Google Scholar] [CrossRef]
  44. Kraicheva, I.; Bogomilova, A.; Tsacheva, I.; Momekov, G.; Troev, K. Synthesis, NMR characterization and in vitro antitumor evaluation of new aminophosphonic acid diesters. Eur. J. Med. Chem. 2009, 44, 3363–3367. [Google Scholar] [CrossRef] [PubMed]
  45. Naydenova, E.; Topashka-Ancheva, M.; Todorov, P.; Yordanova, T.; Troev, K. Novel α-aminophosphonic acids. Design, characterization, and biological activity. Bioorg. Med. Chem. 2006, 14, 2190–2196. [Google Scholar] [CrossRef] [PubMed]
  46. Varga, P.R.; Belovics, A.; Bagi, P.; Tóth, S.; Szakács, G.; Bősze, S.; Szabó, R.; Drahos, L.; Keglevich, G. Efficient synthesis of acylated, dialkyl α-hydroxy-benzylphosphonates and their anticancer activity. Molecules 2022, 27, 2067. [Google Scholar] [CrossRef] [PubMed]
  47. Rádai, Z.; Bagi, P.; Czugler, M.; Karaghiosoff, K.; Keglevich, G. Optical resolution of dimethyl α-hydroxy-arylmethylphosphonates via diastereomer complex formation using calcium hydrogen O,O’-dibenzoyl-(2R,3R)-tartrate; X-ray analysis of the complexes and products. Symmetry 2020, 12, 758. [Google Scholar] [CrossRef]
  48. Jenkins, J.L.; Bender, A.; Davies, J.W. In silico target fishing: Predicting biological targets from chemical structure. Drug Discov. Today 2006, 3, 413–421. [Google Scholar] [CrossRef]
  49. Nicola, G.; Liu, T.; Hwang, L.; Gilson, M. BindingDB: A protein-ligand database for drug discovery. Biophys. J. 2012, 102, 61a. [Google Scholar] [CrossRef] [Green Version]
  50. Venugopala, K.N.; Uppar, V.; Chandrashekharappa, S.; Abdallah, H.H.; Pillay, M.; Deb, P.K.; Morsy, M.A.; Aldhubiab, B.E.; Attimarad, M.; Nair, A.B.; et al. Cytotoxicity and antimycobacterial properties of pyrrolo [1, 2-a] quinoline derivatives: Molecular target identification and molecular docking studies. Antibiotics 2020, 9, 233. [Google Scholar] [CrossRef] [PubMed]
  51. Ur Rehman, N.; Halim, S.A.; Khan, M.; Hussain, H.; Yar Khan, H.; Khan, A.; Abbas, G.; Rafiq, K.; Al-Harrasi, A. Antiproliferative and carbonic anhydrase II inhibitory potential of chemical constituents from Lycium shawii and aloe vera: Evidence from in silico target fishing and in vitro testing. Pharmaceuticals 2020, 13, 94. [Google Scholar] [CrossRef]
  52. Poirier, M.; Awale, M.; Roelli, M.A.; Giuffredi, G.T.; Ruddigkeit, L.; Evensen, L.; Stooss, A.; Calarco, S.; Lorens, J.B.; Charles, R.P.; et al. Identifying lysophosphatidic acid acyltransferase β (LPAAT-β) as the target of a nanomolar angiogenesis inhibitor from a phenotypic screen using the polypharmacology browser PPB2. ChemMedChem 2019, 14, 224–236. [Google Scholar] [CrossRef]
  53. Piazza, R.; Ramazzotti, D.; Spinelli, R.; Pirola, A.; De Sano, L.; Ferrari, P.; Magistroni, V.; Cordani, N.; Sharma, N.; Gambacorti-Passerini, C. OncoScore: A novel, Internet-based tool to assess the oncogenic potential of genes. Sci. Rep. 2017, 7, s46290. [Google Scholar] [CrossRef] [Green Version]
  54. He, Z.; Khatib, A.M.; Creemers, J.W. The proprotein convertase furin in cancer: More than an oncogene. Oncogene 2022, 41, 1252–1262. [Google Scholar] [CrossRef]
  55. Alpert, E.; Akhavan, A.; Gruzman, A.; Hansen, W.J.; Lehrer-Graiwer, J.; Hall, S.C.; Johansen, E.; McAllister, S.; Gulati, M.; Lin, M.F.; et al. Multifunctionality of prostatic acid phosphatase in prostate cancer pathogenesis. Biosci. Rep. 2021, 41, BSR20211646. [Google Scholar] [CrossRef]
  56. Ortlund, E.; LaCount, M.W.; Lebioda, L. Crystal structures of human prostatic acid phosphatase in complex with a phosphate ion and α-benzylaminobenzylphosphonic acid update the mechanistic picture and offer new insights into inhibitor design. Biochemistry 2003, 42, 383–389. [Google Scholar] [CrossRef]
  57. Bollu, L.R.; Mazumdar, A.; Savage, M.I.; Brown, P.H. Molecular pathways: Targeting protein tyrosine phosphatases in cancertargeting protein tyrosine phosphatases in cancer. Clin. Cancer Res. 2017, 23, 2136–2142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Barr, A.J. Protein tyrosine phosphatases as drug targets: Strategies and challenges of inhibitor development. Future Med. Chem. 2010, 2, 1563–1576. [Google Scholar] [CrossRef] [PubMed]
  59. Sivaganesh, V.; Sivaganesh, V.; Scanlon, C.; Iskander, A.; Maher, S.; Lê, T.; Peethambaran, B. Protein tyrosine phosphatases: Mechanisms in cancer. Int. J. Mol. Sci. 2021, 22, 12865. [Google Scholar] [CrossRef] [PubMed]
  60. Xu, R.; Yu, Y.; Zheng, S.; Zhao, X.; Dong, Q.; He, Z.; Liang, Y.; Lu, Q.; Fang, Y.; Gan, X.; et al. Overexpression ofShp2 tyrosine phosphatase is implicated in leukemogenesis in adult human leukemia. Blood 2005, 106, 3142–3149. [Google Scholar] [CrossRef]
  61. Nunes-Xavier, C.E.; Martín-Pérez, J.; Elson, A.; Pulido, R. Protein tyrosine phosphatases as novel targets in breast cancer therapy. Biochim. Biophys. Acta Rev. Cancer. 2013, 1836, 11–226. [Google Scholar] [CrossRef] [PubMed]
  62. Lin, C.C.; Suen, K.M.; Jeffrey, P.A.; Wieteska, L.; Lidster, J.A.; Bao, P.; Curd, A.P.; Stainthorp, A.; Seiler, C.; Koss, H.; et al. Receptor tyrosine kinases regulate signal transduction through a liquid-liquid phase separated state. Mol. Cell. 2022, 82, 1089–1106. [Google Scholar] [CrossRef]
  63. Song, Y.; Wang, S.; Zhao, M.; Yang, X.; Yu, B. Strategies targeting protein tyrosine phosphatase SHP2 for cancer therapy. J. Med. Chem. 2022, 65, 3066–3079. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Synthesis of phosphonoylmethyl- and phosphinoylmethyl-α-aminophosphonates 2a–f.
Scheme 1. Synthesis of phosphonoylmethyl- and phosphinoylmethyl-α-aminophosphonates 2a–f.
Pharmaceuticals 16 00506 sch001
Scheme 2. Preparation of α-acylamino-benzylphosphonates 4a and 4b.
Scheme 2. Preparation of α-acylamino-benzylphosphonates 4a and 4b.
Pharmaceuticals 16 00506 sch002
Scheme 3. Synthesis of bis(α,α’-phosphonoylbenzyl)amine 5.
Scheme 3. Synthesis of bis(α,α’-phosphonoylbenzyl)amine 5.
Pharmaceuticals 16 00506 sch003
Scheme 4. Preparation of a tris(phosphonoylmethyl)amine derivative 6.
Scheme 4. Preparation of a tris(phosphonoylmethyl)amine derivative 6.
Pharmaceuticals 16 00506 sch004
Figure 1. Molecule library of the compounds subjected to bioactivity screening.
Figure 1. Molecule library of the compounds subjected to bioactivity screening.
Pharmaceuticals 16 00506 g001
Figure 2. The workflow of the in silico target prediction.
Figure 2. The workflow of the in silico target prediction.
Pharmaceuticals 16 00506 g002
Table 1. In vitro cytostatic effect of α-aminophosphonic derivatives on human tumor cell lines.
Table 1. In vitro cytostatic effect of α-aminophosphonic derivatives on human tumor cell lines.
CompoundCytostasis (%) at c = 50 µM
Cell Line
MDA-MB 231PC-3Ebc-1A431
1b
1c
1d
1e <10%
2a 10–20%
2b 20–30%
2d 30–40%
2e 40–50%
2f >50%
4a
4b
5
Table 2. Cytostatic effect of α-aminophosphonic derivatives compounds on human tumor cell lines. Cells were treated for 24 h and then cultured for another 72 h. Cell viability was determined with an MTT assay, and the percent of cytostasis was calculated.
Table 2. Cytostatic effect of α-aminophosphonic derivatives compounds on human tumor cell lines. Cells were treated for 24 h and then cultured for another 72 h. Cell viability was determined with an MTT assay, and the percent of cytostasis was calculated.
Cell LineCompound2a2b2d2e2f4a4b5
Pharmaceuticals 16 00506 i001Pharmaceuticals 16 00506 i002Pharmaceuticals 16 00506 i003Pharmaceuticals 16 00506 i004Pharmaceuticals 16 00506 i005Pharmaceuticals 16 00506 i006Pharmaceuticals 16 00506 i007Pharmaceuticals 16 00506 i008
c (µM)Cytostasis (%) ± SD
MDA-MB 2312−4.5 ± 8.9−8.0 ± 3.336.7 ± 4.531.1 ± 1.238.8 ± 0.85.5 ± 5.917.9 ± 16.223.8 ± 13.4
10−9.0 ± 0.934.3 ± 6.928.7 ± 1.931.1 ± 3.532.5 ± 3.334.6 ± 6.221.7 ± 18.226.7 ± 9.6
503.7 ± 1.629.5 ± 1.60.00 ± 3.310.3 ± 4.725.7 ± 1.626.9 ± 4.622.4 ± 3.421.5 ± 4.9
25058.8 ± 3.239.0 ± 0.690.5 ± 0.890.2 ± 0.690.6 ± 0.3n.d.n.d.n.d.
PC-32n.d.32.3 ± 3.810.9 ± 8.36.9 ± 6.9−0.3 ± 2.88.9 ± 3.824.3 ± 4.316.1 ± 9.7
107.1 ± 5.121.1 ± 8.20.4 ± 6.03.4 ± 10.1−4.5 ± 5.424.5 ± 6.819.3 ± 2.316.9 ± 2.9
5010.3 ± 4.812.3 ± 4.9n.d.89.3 ± 0.9n.d.33.0 ± 4.620.7 ± 3.713.0 ± 2.4
250n.d.69.0 ± 3.189.8 ± 0.589.5 ± 0.787.3 ± 0.7n.d.n.d.n.d.
Ebc-1232.2 ± 6.617.6 ± 2.711.8 ± 4.011.6 ± 2.526.2 ± 7.84.2 ± 5.99.8 ± 8.53.6 ± 6.3
1023.8 ± 2.818.6 ± 2.49.1 ± 4.621.9 ± 1.837.0 ± 1.35.8 ± 7.512.0 ± 10.08.5 ± 8.5
5039.0 ± 1.837.8 ± 6.817.3 ± 1.419.1 ± 6.088.5 ± 0.314.2 ± 5.66.8 ± 0.411.1 ± 0.6
25086.3 ± 1.188.5 ± 0.257.4 ± 1.854.9 ± 2.790.4 ± 0.3n.d.n.d.n.d.
A4312n.d.11.1 ± 0.211.9 ± 6.314.7 ± 5.29.1 ± 8.73.3 ± 3.8−10.0 ± 9.22.7 ± 10.5
1017.3 ± 6.49.2 ± 6.77.8 ± 2.118.7 ± 5.40.3 ± 14.72.1 ± 4.0−5.1 ± 11.16.6 ± 10.1
5043.7 ± 3.545.1 ± 6.925.8 ± 3.331.1 ± 6.392.8 ± 0.38.7 ± 2.65.6 ± 1.2−0.4 ± 7.8
25089.1 ± 1.591.2 ± 0.452.4 ± 0.432.2 ± 0.993.3 ± 0.4n.d.n.d.n.d.
n.d.: Not determined.
Table 3. IC50 of the compounds on human tumor cell lines. The 50% inhibitory concentration (IC50) was determined from the dose–response curves fitted on the cytostasis data and is expressed in micromolar units.
Table 3. IC50 of the compounds on human tumor cell lines. The 50% inhibitory concentration (IC50) was determined from the dose–response curves fitted on the cytostasis data and is expressed in micromolar units.
CompoundCell Line
MDA-MB 231PC-3Ebc-1A431
IC50 (µM)
2a169.2>250>25087.5
2b>25073.8107.153.2
2d45.8114.8166.6143.8
2e55.129.4138.6>250
2f82.6128.297.9115.6
Table 4. Classification of the putative protein targets of the cytotoxic hit compounds (2b, 2d, 2e).
Table 4. Classification of the putative protein targets of the cytotoxic hit compounds (2b, 2d, 2e).
Cancer-Related TargetsNon-Cancer-Related Targets
FurinTyrosyl-DNA phosphodiesterase 1
Prostatic acid phosphataseCorticotropin-releasing-factor binding protein
Tyrosine phosphatase non-receptor type 2 (TCPTP)Muscarinic acetylcholine receptor M1/2
Tyrosine phosphatase non-receptor type 6 (SHP1)Solute carrier family 22 member 1
Tyrosine phosphatase non-receptor type 9 (PTP-MEG2)C-C chemokine receptor type 3
Tyrosine phosphatase non-receptor type 11 (SHP2)Acetylcholinesterase
Table 5. Target annotation of known bioactive compounds found similar to the cytotoxic hits (2b, 2d, 2e). Note: the annotated bioactivity databases often lack chirality information.
Table 5. Target annotation of known bioactive compounds found similar to the cytotoxic hits (2b, 2d, 2e). Note: the annotated bioactivity databases often lack chirality information.
Cancer-Associated
Protein + A1:D5
Expression/
Overexpression in
Cancer
Similar StructureRef.
Furinbreast, head/neck,
gastric cancer
Pharmaceuticals 16 00506 i009
weak inhibitor
[54]
Prostatic acid phosphatase (PAP)prostate cancerPharmaceuticals 16 00506 i010
highly potent inhibitor
[55,56]
Tyrosine phosphatase non-receptor type 11 (SHP2),
type 2 (TCPTP)
breast, skin cancer,
leukemia
Pharmaceuticals 16 00506 i011
inhibitor of TCPTP and SHP2 in various affinities
[57,58,59,60,61,62,63]
Tyrosine phosphatase non-receptor type 11 (SHP2),
type 2 (TCPTP),
type 9 (PTP-MEG2)
breast, skin cancer,
leukemia
Pharmaceuticals 16 00506 i012
inhibitor of TCPTP, SHP2 and PTP-MEG2 in various affinities
[57,58,59,60,61,62,63]
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

Varga, P.R.; Szabó, R.O.; Dormán, G.; Bősze, S.; Keglevich, G. Cytotoxic Activity of α-Aminophosphonic Derivatives Coming from the Tandem Kabachnik–Fields Reaction and Acylation. Pharmaceuticals 2023, 16, 506. https://doi.org/10.3390/ph16040506

AMA Style

Varga PR, Szabó RO, Dormán G, Bősze S, Keglevich G. Cytotoxic Activity of α-Aminophosphonic Derivatives Coming from the Tandem Kabachnik–Fields Reaction and Acylation. Pharmaceuticals. 2023; 16(4):506. https://doi.org/10.3390/ph16040506

Chicago/Turabian Style

Varga, Petra R., Rita Oláhné Szabó, György Dormán, Szilvia Bősze, and György Keglevich. 2023. "Cytotoxic Activity of α-Aminophosphonic Derivatives Coming from the Tandem Kabachnik–Fields Reaction and Acylation" Pharmaceuticals 16, no. 4: 506. https://doi.org/10.3390/ph16040506

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop