Elsevier

Bioorganic & Medicinal Chemistry

Volume 25, Issue 20, 15 October 2017, Pages 5452-5460
Bioorganic & Medicinal Chemistry

Exploring the C^N^C theme: Synthesis and biological properties of tridentate cyclometalated gold(III) complexes

https://doi.org/10.1016/j.bmc.2017.08.001Get rights and content

Abstract

A family of cyclometalated Au(III) complexes featuring a tridentate C^N^C scaffold has been synthesized and characterized. Microwave assisted synthesis of the ligands has also been exploited and optimized. The biological properties of the thus formed compounds have been studied in cancer cells and demonstrate generally moderate antiproliferative effects. Initial mechanistic insights have also been gained on the gold complex [Au(C^N^C)(GluS)] (3), and support the idea that the thioredoxin system may be a target for this family of compounds together with other relevant intracellular thiol-containing molecules.

Introduction

Gold-based organometallic complexes as anti-cancer agents have become increasingly popular in recent years, and several reviews have been published highlighting their structural diversity and biological activity.1 In fact, organometallic complexes display numerous attractive features. For example, while the organic ligand allows for the introduction of stereospecificity and alteration of the physicochemical properties by choosing different functional groups, the metal–carbon (M–C) bond provides strong trans influence and, in the case of π-bonded aromatic arene and cyclopentadienyl ligands, can act both as electron donors and acceptors. Furthermore, variations of the organic moiety are accessible, allowing the “fine-tuning” of the physiochemical properties of the respective metal compound. Finally, by choosing specific targeting moieties, either incorporation into the ligand or tethering to the metal centre can be achieved in a modular approach.

Typical classes of organometallics include metallocenes, metallo-arenes, metallo-carbonyls, metallo-carbenes (e.g. N-heterocyclic carbenes, NHC), alkynyl complexes, etc.2 featuring both mononuclear and multinuclear scaffolds. All these compound families have been extensively applied in catalysis during the last decades.3 However, their medicinal use has been considered only much more recently.1(c), 2, 4

Concerning Au(I) organometallics, in 2008 Berners-Price et al. synthesized a series of mononuclear cationic Au(I) biscarbene complexes that show remarkable cytotoxicity in vitro and are able to induce mitochondrial damage.5 Since then, the effects of Au(I) NHC complexes on cell metabolism and their interference with pathways relevant to cancer cell proliferation have been studied in a broad number of cases.1(a), 4(b), 6

In this context, to explore the design of Au(III) compounds for biological applications, cyclometalation is a convenient method to stabilize the otherwise easily reduced Au(III) centre, and numerous cyclometalated scaffolds have been synthesized, including C^N, C^N^C, C^N^N and C^N^S.1(b), 1(c), 7 Cyclometalated Au(III) C^N compounds of general formula [(Au(damp)X2] (Fig. 1) with anticancer properties were first investigated by Parish, Buckley et al. in 1996,8 featuring a 2[dimethylamino)methyl]-phenyl (damp) backbone. The compounds display cytotoxic activity which is comparable to that of cisplatin, against a variety cancer cell lines. This activity is also accompanied by high selectivity and cytotoxicity in vitro which translates to moderate activity in vivo.

In 1996, the synthesis of an Au(III) 2-benzylpyridine derivative [Au(pyb-H)Cl2] (pyb-H = C^N cyclometalated 2-benzylpyridine) (Fig. 1) was reported by Cinellu et al.9 In 2015 Casini, Cinellu et al. synthesized a structural analogue of this complex replacing the chlorido ligand, trans to the nitrogen atom, with 1,3,5-triazaphosphaadamantane (PTA).10 The resulting compound displays good cytotoxic activity against various cancer cell lines such as A2780 (human ovarian adenocarcinoma). Furthermore, dinuclear oxo-bridged Au(III) C^N^N complexes of formula [(C^N^N)2Au2(µ-O)](PF6)2 (with C^N^N = 6-(1-methylbenzyl)-2,2‘-bipyridine or 6-(1,1-dimethylbenzyl)-2,2′-bipyridine) showing moderate cytotoxicity against various cancer cell lines were synthesized.11 (Fig. 1).

The field of Au(III) C^N^C complexes with anticancer properties has been closely examined by Che and co-workers1(b), 12 For example, dinuclear complexes of the type [Aum(C^N^C)mL]n+ (with HC^N^CH = 2,6-diphenylpyridine; m = 1–3; n = 0–3) showed higher cytotoxic activity against various cancer cell lines than their mononuclear counterparts.12

The highest cytotoxicities were observed for complex [Au2(C^N^C)2(μ-dppp)](OTf)2 (Fig. 1), relating to the cytotoxicity of the free 1,2-bis(diphenylphosphino)propane (dppp) ligand. By replacing the phosphane moiety with an NHC ligand as in the cationic C^N^C stabilized complexes [Aun(R–C^N^C)n(NHC)]n+, a general decrease in cytotoxic effects has been noted,13 supporting the idea that indeed phosphine ligand-mediated cytotoxicity plays a major role in the overall anticancer properties. Nevertheless, within this series, the mononuclear complex [Au(C^N^C)(IMe)]CF3SO3 (IMe = 1,3-dimethylimidazol-2-ylidene) (Fig. 1) shows higher cytotoxic activity than its dinuclear analogue, and a high degree of selectivity towards human cancer cells compared to normal lung fibroblasts (CCD-19Lu). Through DNA interaction studies it was demonstrated that the compound induces DNA strand breaks and can cause subsequent cell death through the stabilization of Topoisomerase-linked DNA.14 Treatment of nude mice bearing PLC tumors (hepatocellular carcinoma) with [Au(C^N^C)(IMe)]CF3SO3 at 10 mg/kg/week for 28 days significantly suppressed (47%) tumor growth when compared with that of the vehicle control.13

Of note from the same group, is the application of supramolecular polymers, self-assembled from cyclometalated Au(III) C^N^C complexes, as anticancer agents. The mononuclear complex [Au(C^N^C)(4-dpt)]+ (C^N^C = 2,6-diphenylpyridine, 4-dpt = 2,4-diamino-6-(4-pyridyl)-1,3,5-triazine) (Fig. 1) was chosen due to the ability of the antiangiogenic 4-dpt ligand to form intramolecular hydrogen bonds and to establish π-π interactions, leading to supramolecular complex formation by self-assembly at ambient temperatures.15 The supramolecular polymer displays high cytotoxic activity towards B16 cells (murine cancer).

In terms of possible mechanisms of action, cytotoxic Au(III) complexes can interact with protein targets,16 including those constituting the thioredoxin system, often overexpressed in tumor cells and involved in maintaining the intracellular redox balance.17 Among the enzymes included in this system, the seleno-protein thioredoxin reductase (TrxR) contains a cysteine-selenocysteine redox pair at the C-terminal active site, and the solvent-accessible selenolate group, arising from enzymatic reduction, constitutes a likely target for “soft” metal ions such as gold. In addition, Au(III) complexes can interfere with thiols including glutathione, the most abundant intracellular reducing agent.

Within the scope of new medicinally relevant organometallic Au(III) compounds, two new cyclometalated Au(III) complexes of the general formula [Au(III)(C^N^C)X] (with C^N^C = 2,6-diphenylpyridine and X = Cl, thio-β-d-glucose-tetraacetate (GluS), 1,3,5-triazaphosphaadamantane (PTA)) were synthesized. The complexes were prepared starting from the literature known chlorido precursor [Au(III)(C^N^C)Cl]18 (Fig. 2, Fig. 1, 1), substituting the chlorido ligand with PTA (2) and thio-β-d-glucose-tetraacetate (GluSH) (3), respectively. The PTA ligand was chosen to increase the water solubility, while the GluS ligand was selected as modulator of the hydrophilic/lipophilic character, as well as a possible facilitator of the compound’s uptake via interactions with the GLUT1 transporter. A second series of analogous C^N^C complexes – Au(III)[(C^NR^C)] – was synthesized starting from the corresponding 2,4,6-triarylpyridine ligands, but featuring different substituents in the para position of the phenylpyridine (R = OH, F, Br, NO2) (Fig. 2, Fig. 4, Fig. 5, Fig. 6, 47) in order to evaluate the effects of this ligand modification on the biological properties. Notably, complex 5 has already been described by Che et al. in 2013 as a precursor for photoactive functionalized bis-cyclometalated alkynyl gold(III) complexes suitable as phosphorescent organic light-emitting diodes (OLEDs).19 However, the authors did not evaluate the biological activity of this complex. All the cyclometalated Au(III) complexes reported herein were studied for their antiproliferative effects against different human cancer cell lines, including some that are resistant to cisplatin. Moreover, compounds 13 were also tested for their inhibitory properties of TrxR on the purified protein and in cell lysates. The effects of the new complexes on the oxidation state of Trx were also investigated by Western blot analysis. Discrimination between the oxidation of the GSH/GSSG system and the Trx system can be very informative in terms of mechanisms of toxicity since different cellular pathways are controlled by GSH and Trx. Thus, estimation of the glutathione content was performed in treated cells.

Section snippets

Synthesis and structural characterization

To achieve the synthesis of the new compounds, the transmetalation pathway via the respective Hg(II) precursor was followed as described in Fig. 2. Thus, treatment of 2,6-diphenylpyridine or para substituted 2,4,6-triarylpyridines with mercury(II) acetate, followed by salt metathesis with LiCl, affords the organomercury(II) precursors (see Supplementary material for Hg complexes characterization, Figs. S16–S24). Subsequent transmetalation with K[AuCl4] in refluxing acetonitrile, adapting

Conclusions

Two new Au(III)[C^N^C] complexes, [Au(C^N^C)(PTA)][PF6] (2) and [Au(C^N^C)(GluS)] (3), and three new Au(III)[C^N(R)^C] complexes with R = Ph-p-OH (4), Ph-p-Br (6), Ph-p-NO2 (7) have been synthesized. Microwave assisted ligand synthesis for the para substituted 2,4,6-triaryl ligands for complex 47 was optimized concerning applied temperature, power and reaction time. The compounds have been examined for their antiproliferative effects in a small panel of human cancer cells. In general, the

General remarks

Reactions were carried out under purified argon using standard Schlenk techniques. Solvents were dried, degassed and stored over molecular sieve and under argon before use. Microwave reactions were carried out in a CEM Focused Microwave™ Synthesis System or a Biotage® Initiator Robot Eight and Sixty. All physico-chemical analytics were performed at the Technische Universität München and the University of Cardiff. UV−vis spectra were recorded on a Perkin-Elmer Lambda 650 UV/vis spectrophotometer.

Acknowledgements

Authors thank Cardiff University and Technische Universität München (TUM) Graduate School for funding, as well as the Department of Sciences, Technology, and Innovation COLCIENCIAS (Colombia) for a fellowship to N.E.O. A.C. acknowledges a Hans Fischer Senior Fellowship of the TUM – Institute for Advanced Study, funded by the German Excellence Initiative and the European Union Seventh Framework Programme under grant agreement n° 291763. Authors acknowledge Dr Alexander Pöthig’s support with

References (31)

  • J.J. Zhang et al.

    Organogold (III) supramolecular polymers for anticancer treatment

    Angew Chem Int Ed

    (2012)
  • J. Fernandez-Cestau et al.

    Synthesis and luminescence modulation of pyrazine-based gold (III) pincer complexes

    Chem Commun

    (2015)
  • M.E. Anderson

    [70] Determination of glutathione and glutathione disulfide in biological samples

    Methods Enzymol

    (1985)
  • M. Cinellu et al.

    Gold Organometallics with Biological Properties

    T. Zou et al.

    Chemical biology of anticancer gold (III) and gold (I) complexes

    Chem Soc Rev

    (2015)
    B. Bertrand et al.

    A golden future in medicinal inorganic chemistry: the promise of anticancer gold organometallic compounds

    Dalton Trans

    (2014)
    S. Nobili et al.

    Gold compounds as anticancer agents: chemistry, cellular pharmacology, and preclinical studies

    Med Res Rev

    (2010)
  • G. Gasser et al.

    Organometallic Anticancer Compounds

    J Med Chem

    (2011)
  • L.-A. Schaper et al.

    Synthesis and Application of Water-Soluble NHC Transition-Metal Complexes

    Angew Chem Int Ed

    (2013)
    H.D. Velazquez et al.

    N-heterocyclic carbene transition metal complexes for catalysis in aqueous media

    Chem Soc Rev

    (2012)
  • E. Meggers

    Targeting proteins with metal complexes

    Chem Commun

    (2009)
    L. Oehninger et al.

    N-Heterocyclic carbene metal complexes in medicinal chemistry

    Dalton Trans

    (2013)
    G. Jaouen et al.

    Ferrocifen type anti cancer drugs

    Chem Soc Rev

    (2015)
    A. Gautier et al.

    Advances in metal–carbene complexes as potent anti-cancer agents

    Metallomics

    (2012)
    L. Mercs et al.

    Beyond catalysis: N-heterocyclic carbene complexes as components for medicinal, luminescent, and functional materials applications

    Chem Soc Rev

    (2010)
  • J.L. Hickey et al.

    Mitochondria-Targeted Chemotherapeutics: The Rational Design of Gold(I) N-Heterocyclic Carbene Complexes That Are Selectively Toxic to Cancer Cells and Target Protein Selenols in Preference to Thiols

    J Am Chem Soc

    (2008)
  • J.K. Muenzner et al.

    Adjusting the DNA interaction and anticancer activity of Pt (II) N-heterocyclic carbene complexes by steric shielding of the trans leaving group

    J Med Chem

    (2015)
  • N. Cutillas et al.

    Anticancer cyclometalated complexes of platinum group metals and gold

    Coord Chem Rev

    (2013)
  • R.G. Buckley et al.

    Antitumor properties of some 2-[(dimethylamino) methyl] phenylgold (III) complexes

    J Med Chem

    (1996)
  • M.A. Cinellu et al.

    Synthesis and characterization of gold(III) adducts and cyclometallated derivatives with 6-benzyl- and 6-alkyl-2,2[prime or minute]-bipyridines

    J Chem Soc, Dalton Trans

    (1996)
  • B. Bertrand et al.

    Exploring the potential of gold(iii) cyclometallated compounds as cytotoxic agents: variations on the C^N theme

    Dalton Trans

    (2015)
  • C. Gabbiani et al.

    Mechanistic studies on two dinuclear organogold(III) compounds showing appreciable antiproliferative properties and a high redox stability

    Metallomics

    (2011)
  • C.K.L. Li et al.

    Anticancer cyclometalated [AuIIIm (C∧ N∧ C) mL] n+ compounds: synthesis and cytotoxic properties

    Chem Eur J

    (2006)
  • Cited by (32)

    • Organometallic Chemistry of Gold-Based Drugs

      2022, Comprehensive Organometallic Chemistry IV: Volume 1-15
    • Synthesis, structural characterization, docking simulation and in vitro antiproliferative activity of the new gold(III) complex with 2-pyridineethanol

      2021, Journal of Inorganic Biochemistry
      Citation Excerpt :

      Calculated values of SI clearly indicated that Au complex could be a potential candidate for an anticancer drug, since it exhibited high cytotoxicity against studied cancer cells (MCF7, A549) and low toxicity toward healthy lines (HEK298T, HaCat). In general, the anticancer activity of the synthesized complex against the A549 and MCF7 human cancer cell lines exhibits better or similar activity than observed in analogous gold-based complexes [98–102]. However, more investigations should be undertaken to highlight the exact molecular mechanism responsible for the observed selective cytotoxicity.

    • Mechanisms underlying the cytotoxic activity of syn/anti-isomers of dinuclear Au(I) NHC complexes

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Considering the mitochondrial enzyme TrxR as well-known target of cationic gold(I) NHC complexes, preliminary mechanistic studies were conducted for both cytotoxic isomers synAu2(MesLOH)(PF6)2 and anti,mixAu2(MesLOH)(PF6)2 including auranofin to analyse their inhibition properties particularly focusing on the comparison between both isomers [28,45]. These inhibition studies were performed using a colorimetric method (5,5′-Dithiobis-2-nitrobenzoic acid, DTNB assay) monitoring the reduction of DTNB to TNB (5-thio-2-nitrobenzoic acid) in presence of the enzyme and NADPH [45–47]. The corresponding IC50 values were determined by the use of an appropriate range of concentrations and are summarized in Fig. 7.

    • Cyclometallated gold(III) complexes against colon cancer. X-ray structure of [Au(C,NPhenylpyridine)(OAc)<inf>2</inf>]

      2020, Journal of Organometallic Chemistry
      Citation Excerpt :

      Moving to the different derivatives with acetate or amino acid groups (Fig. 9) only the complex C8 ([Au(ppy)(Phe)]Cl) showed cytotoxic properties (1.32 ± 0.12 μM). A poorly cytotoxic effect from some gold(III) complexes is not surprising as it was showed in other published works [9]. Besides, this cell line allows us to measure the selectivity of our complexes because, Caco-2 cells, after reaching the confluence (15 days after seeding), experiment a spontaneous and homogeneous enterocytic differentiation [65,66].

    View all citing articles on Scopus
    View full text