Elsevier

European Journal of Medicinal Chemistry

Volume 90, 27 January 2015, Pages 766-774
European Journal of Medicinal Chemistry

Original article
In vitro anticancer activity of gold(III) complexes with some esters of (S,S)-ethylenediamine-N,N′-di-2-propanoic acid

https://doi.org/10.1016/j.ejmech.2014.12.019Get rights and content

Highlights

  • Five novel Au(III) complexes with (S,S)-R2eddip esters were prepared.

  • [AuCl2{(S,S)-(i-Bu)2eddip}]PF6 forms metal-protein adducts with BSA.

  • [AuCl2{(S,S)-(i-Am)2eddip}]PF6 is much more active and selective than cisplatin.

  • Complexes induced apoptotic mode of HeLa cell death.

Abstract

Five novel gold(III) complexes of general formulas [AuCl2{(S,S)-R2eddip}]PF6, ((S,S)-eddip = (S,S)-ethylenediamine-N,N′-di-2-propanoate, R = n-Bu, n-Pe, i-Bu, i-Am, cPe; 15, respectively) were synthesized and characterized by UV/Vis, IR and NMR spectroscopy and mass spectrometry. DFT calculations indicated that (R,R)-N,N′-configuration diastereoisomers were the most stable for 15. 3 is stable in DMSO for at least 24 h, but immediate hydrolysis in PBS occurs. 3 is readily reduced with ascorbic acid and forms adducts with bovine serum albumin (BSA). In vitro anticancer activity of the gold(III) complexes against human cervix adenocarcinoma HeLa, human myelogenous leukemia K562, human melanoma Fem-x tumor cell lines, as well as against non-cancerous human embryonic lung fibroblast cell line MRC-5 was determined using MTT assay. Complex 4 showed highest activity and selectivity (IC50(Fem-x) = 1.3 ± 0.2; IC50(MRC-5)/IC50(Fem-x) = 72.5 ± 12.4), 4 times more active and 28 times more selective than cisplatin. Complexes induced apoptotic mode of death in a time-dependent manner in HeLa cells.

Graphical abstract

Synthesis, characterization and biological activity of five novel gold(III) complexes with some R2edda-type esters, [AuCl2{(S,S)-R2eddip}]PF6, R = n-Bu, n-Pe, i-Bu, i-Am, cPe, 15, is described. [AuCl2{(S,S)-(i-Bu)2eddip}]PF6 forms metal-protein adducts with BSA. Antitumoral activity was tested in vitro on four tumor cell lines. Complex 4 showed highest activity and selectivity, 4 times more active and 28 times more selective than cisplatin. Apoptosis is the main mode of cell death induced by these compounds in HeLa cells.

  1. Download : Download full-size image

Introduction

The progress of medicinal inorganic chemistry includes several applications of a variety of metal complexes in medicine [1], [2], [3], [4]. To design a metal-based applicable anticancer drug is challenging. Any candidate for an antitumor agent needs to demonstrate its positive reactions with target biomolecules and favorable physiological responses to tumors before entering clinical trials. Cisplatin has made an impressive impact on cancer chemotherapy, and nowadays is frequently used in treatment of various types of cancers [5], [6], [7], [8], [9]. Its activity, as well as activity of new-generation platinum compounds, is compromised due to inevitable serious side effects such as nephrotoxicity and neurotoxicity, hair and hear loss and many others [10], [11], [12], [13]. Interest in medicinal chemistry of gold has been growing with the successful use of auranofin for treatment of rheumatoid arthritis [14], [15]. Gold(III) complexes have greatly attracted researchers' attention in the last decade for their outstanding cytotoxic actions against different tumor cells [16], [17], even against the cisplatin-resistant cell lines [18], [19], [20]. With square-planar geometry (d8 system), gold(III) complexes are isoelectronic and isostructural to platinum(II) complexes, thus they could show a model of binding to the biomolecules similarly to cisplatin [21], [22]. The strict relationship to platinum(II) compounds makes gold(III) complexes good candidates for development as anticancer drugs, although gold(III) complexes are not very stable under physiological conditions because of their high reduction potential and fast hydrolysis rate. These problems can possibly be circumvented by forming gold(III) compounds with one or more multidentate nitrogen-donor ligands to enhance their stability [23], [24], [25]. Recent findings by Messori et al. showed that most of the cytotoxic gold(III) complexes have a weak binding affinity to DNA, which is the primary target for platinum(II) antitumor drugs [22]. Also, it was found that cytotoxic gold(III) complexes have shown high reactivity toward different protein models [26]. Most of the known active gold(III) complexes might react through gold(I) species produced by gold(III) reduction in vivo [27]. However, there are exceptions where e.g. porphyrinato ligand markedly stabilizes the gold(III) ion against reduction diminishing the possibility to be reduced by biological reductants such as glutathione and ascorbic acid [28].

Generally, very few gold(III) compounds demonstrate anticancer activity in vivo [29]. Since the early investigations on [AuX2(damp)] (damp = 2-[(dimethylamino)methyl]-phenyl, X2/X = malonato/acetato) [30], [31], in vivo anticancer activity has been reported for only four other types of gold(III) compounds, gold(III) dithiocarbamate [32], gold(III) porphyrins [33], [34], cyclometallated gold(III) NHC [35] and gold(III) phosphine complex [36].

Recently, synthesis and characterization of gold(III) complexes with esters of cyclohexyl-functionalized ethylenediamine-N,N′-diacetate was reported (Fig. 1) [37]. The in vitro cytotoxic evaluation of the investigated complexes against tumor cell lines: human adenocarcinoma (HeLa cells), human myelogenuos leukemia (K562 cells) and against normal peripheral blood mononuclear cells (PBMC), showed that the cytotoxic action of gold(III) complexes with cyclohexyl-functionalized ethylenediamine-N,N′-diacetate esters, (R = i-Bu, i-Am), is fairly comparable to that of cisplatin [37].

Inspired by these promising results, five novel gold(III) complexes of N,N′ bidentate (S,S)-R2eddip ligands with general formulae [AuCl2{(S,S)-R2eddip}]PF6: ((S,S)-eddip = (S,S)-ethelendiamine-N,N’-di-2-propanoate; R = n-Bu, n-Pe, i-Bu, i-Am, cPe, 15, respectively) were synthesized. Density functional theory (DFT) analyses were performed for indication of the preferred configuration of nitrogen atoms. Stability of 3 in DMSO and in physiological medium (PBS) was examined, as well as possibility of reduction by ascorbic acid, by time-dependent UV/Vis spectrometry and 13C NMR spectroscopy. Interaction of a selected complex, 3 with bovine serum albumin (BSA) is monitored by UV/Vis spectrometry over time. All compounds were tested against cervix adenocarcnoma cell line (HeLa), human chronic myelogenous leukemia (K562), human melanoma (Fem-x) and non-cancerous cell line, human embryonic lung fibroblast (MRC-5) with the aim of assessing in vitro activity and selectivity. The mode of HeLa cell death induced by 15 was also studied, as well as cell cycle distribution of HeLa cells upon treatment with these complexes.

Section snippets

Chemistry

In the reaction of Na[AuCl4]·2H2O and an equimolar amount of corresponding (S,S)-R2eddip ligand, previously deprotonated with LiOH in methanol, and after addition of ammonium hexafluorophosphate to the reaction mixture, desired complexes 15 (Scheme 1) were obtained as yellow powders. The complexes are soluble in methanol, ethanol, acetone, dichloromethane, chloroform, dimethyl sulfoxide and acetonitrile.

Spectroscopic characterizations

IR spectra showed strong ν(Cdouble bondO) absorption stretching bands from 1732 to 1738 cm−1 (15) [38]

Conclusions

Synthesis of five novel gold(III) complexes, [AuCl2{(S,S)-R2eddip}]PF6, R = n-Bu, n-Pe, i-Bu, i-Am, cPe, 15, respectively, was described. The compounds were characterized by elemental analysis, UV/Vis, IR, NMR spectroscopy and mass spectrometry. Spectroscopic data suggest ligand chelation via nitrogen donor atoms. NMR spectra show presence of one isomer. DFT calculations indicate that (R,R)-N,N′- diastereoisomer is the most stable one. Complex 3 was stable in DMSO but in physiological medium

Materials and methods

The n-butyl, n-pentyl, isobutyl, isoamyl and cyclopentyl esters of (S,S)-ethylenediamine-N,N′-di-3-propanoic acid were synthesized according to described method [38], [40], [41], [42]. Na[AuCl4] was synthesized by the standard procedure [61].

Elemental analyses were performed on an Elemental Vario EL III microanalyzer. A Nicolet 6700 FT–IR spectrometer and ATR technique were used for recording mid-infrared spectra (4000–400 cm−1) for all complexes. Far-IR spectra were recorded at room

Acknowledgments

This research was supported by the Ministry of Education, Science and Technological Development of the Republic of Serbia, grant numbers 172035 and 175011. The authors thank D. Vučetić, Department of Chemistry, Institute of Chemistry, Technology and Metallurgy, University of Belgrade, for his help in NMR spectroscopy.

References (69)

  • M.J. Clarke

    Coord. Chem. Rev.

    (2002)
  • B. Rosenberg et al.

    J. Biol. Chem.

    (1967)
  • B. Lippert

    Coord. Chem. Rev.

    (1999)
  • U. Kalinowska-Lis et al.

    Coord. Chem. Rev.

    (2008)
  • I. Ott

    Coord. Chem. Rew

    (2009)
  • A. Bindoli et al.

    Coord. Chem. Rev.

    (2009)
  • A. Casini et al.

    J. Inorg. Biochem.

    (2008)
  • F. Abbate et al.

    Inorg. Chim. Acta

    (2000)
  • N. Pantelić et al.

    J. Inorg. Biochem.

    (2013)
  • B.B. Krajčinović et al.

    J. Inorg. Biochem.

    (2008)
  • B.B. Zmejkovski et al.

    Eur. J. Med. Chem.

    (2009)
  • G.P. Vasić et al.

    Inorg. Chim. Acta

    (2010)
  • A.A. Isab et al.

    Spectrochim. Acta A

    (2011)
  • G.N. Kaluđerović et al.

    Inorg. Chim. Acta

    (2008)
  • A.N. Wein et al.

    J. Inorg. Biochem.

    (2011)
  • R.W.-Y. Sun et al.

    Coord. Chem. Rev.

    (2009)
  • K. Palanichamy et al.

    Inorg. Chim. Acta

    (2006)
  • W. Bal et al.

    J. Inorg. Biochem.

    (1998)
  • B.P. Espósito et al.

    Coord. Chem. Rev.

    (2002)
  • C. Adamo et al.

    Chem. Phys. Lett.

    (1997)
  • T. Mosmann

    J. Immunol. Methods

    (1983)
  • M. Ohno et al.

    J. Immunol. Methods

    (1991)
  • B. Lippert

    Prog. Inorg. Chem.

    (1989)
  • E.R. Jamieson et al.

    Chem. Rev.

    (1999)
  • M.J. Clarke et al.

    Chem. Rev.

    (1999)
  • B. Rosenberg et al.

    Nature

    (1965)
  • L. Kelland

    Nat. Rev.

    (2007)
  • G.N. Kaluđerović et al.

    Bioneorg. Chem. Appl.

    (2012)
  • M.A. Jakupec et al.

    Rev. Physiol. Biochem.. Pharmacol.

    (2003)
  • G.N. Kaluđerović et al.

    Curr. Med. Chem.

    (2011)
  • S. Gómez et al.

    Bioinorg. Chem. Appl.

    (2012)
  • K.C. Dash et al.

    Metal Ions in Biological Systems

    (1982)
  • A. Casini et al.

    J. Med. Chem.

    (2006)
  • D. Aldinucci et al.

    Anti-Cancer Drug

    (2007)
  • Cited by (33)

    • Palladium(II) complexes: Structure, development and cytotoxicity from cisplatin analogues to chelating ligands with N stereocenters

      2022, Inorganica Chimica Acta
      Citation Excerpt :

      Data obtained are in accordance to the previous studies showing higher cytotoxicity of Pd(II) complexes when raising the lipophilicity of the O,O′-dialkyl esters groups. The studies conducted by Pantelić et al. have shown that the gold(III) complexes with N,N’-ethylenediamine bidentate ester ligands exhibit strong anticancer activity versus different tumor cell lines [68,69,118–120]. Namely, [AuCl2{(S,S)-(i-Am)2eddip}]PF6, ((S,S)-(i-Am)2eddip2- = O,O’-diisoamyl-(S,S)-ethylenediamine-N,N’-di-2-propanoate) have demonstrated 4 fold higher activity and 28 times more selectivity than cisplatin [118].

    • Antitumor potential of cisplatin loaded into SBA-15 mesoporous silica nanoparticles against B16F1 melanoma cells: in vitro and in vivo studies

      2021, Journal of Inorganic Biochemistry
      Citation Excerpt :

      Samples were investigated using flow cytometry after 48 h of treatment. In 6 well plate, cells were incubated with CP (IC50, 2 × IC50 doses) or SBA-15|CP (MC50, 2 × MC50 doses) for 48 h. Afterwards, Annexin V/PI or apostat procedure was applied as described in the literature [51]. In 6 well plate, cells were incubated with CP (IC50, 2 × IC50 doses) or SBA-15|CP doses (MC50, 2 × MC50 doses) for 48 h. Afterwards, AO assay was performed as previously described [52].

    • Delivery of [Ru(η<sup>6</sup>-p-cymene)Cl<inf>2</inf>{Ph<inf>2</inf>P(CH<inf>2</inf>)<inf>3</inf>SPh-κP}] using unfunctionalized and mercapto functionalized SBA-15 mesoporous silica: Preparation, characterization and in vitro study

      2018, Journal of Inorganic Biochemistry
      Citation Excerpt :

      Using nonplatinum-based anticancer metal compounds might have the impact on different targets in tumor cells, thus different cytotoxicity as well as toxicity is to be expected [7–10]. Some of these drugs contain tin(IV), titanium(IV), gallium(III) or gold(III) compounds as metal center, for instance [11–18]. Specially, ruthenium-based compounds seem to be very promising, not only because of their lower general toxicity but they also display a very good cytotoxic activity, moreover some of them exhibit an antimetastatic activity [19,20].

    • In vitro antitumor activity, metal uptake and reactivity with ascorbic acid and BSA of some gold(III) complexes with N,N′-ethylenediamine bidentate ester ligands

      2017, Journal of Inorganic Biochemistry
      Citation Excerpt :

      The data obtained using UV/Vis spectroscopy showed absorption around 320 nm, which can be assigned to Cl → Au charge transfer, by analogy to auric acid absorption spectra [61]. Similar electronic transitions are observed in analogous gold(III) complexes [45,46]. According to crystal field theory of d8 complexes, the lowest unoccupied molecular orbital (LUMO) is dx2 − y2, and therefore, the ligand to metal charge transfer could be due to pσ → dx2 − y2 transition [62].

    View all citing articles on Scopus
    View full text