Synthesis, chemical characterization, PARP inhibition, DNA binding and cellular uptake of novel ruthenium(II)-arene complexes bearing benzamide derivatives in human breast cancer cells

https://doi.org/10.1016/j.jinorgbio.2020.111155Get rights and content

Highlights

  • Poly(ADP-ribose) polymerase-1 (PARP-1) is a key player in repairing single-strand DNA breaks

  • Ru(II) arene complexes exhibit good efficiency in inhibiting PARP-1 activity.

  • Ru complexes exhibit good antiproliferative activity against breast cancer cells.

  • Ru(II) arene complexes display notable nuclear-targeting properties.

  • Ru(II) arene complexes interfere with the DNA replication.

Abstract

Inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1) showed remarkable clinical efficacy in BRCA-mutated tumors. Based on the rational drug design, derivatives of PARP inhibitor 3-aminobenzamide (3-AB), 2-amino-4-methylbenzamide (L1) and 3-amino-N-methylbenzamide (L2), were coordinated to the ruthenium(II) ion, to form potential drugs affecting DNA and inhibiting PARP enzyme. The four conjugated complexes of formula: C1 [(ƞ6-toluene)Ru(L1)Cl]PF6, C2 [(ƞ6-p-cymene)Ru(L1)Cl]PF6, C3 [(ƞ6-toluene)Ru(L2)Cl2] and C4 [(ƞ6-p-cymene)Ru(L2)Cl2], have been synthesized and characterized. Colorimetric 3-(4.5-dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide (MTT) assay showed the highest antiproliferative activity of C1 in HCC1937, MDA-MB-231, and MCF-7 breast cancer cells. Efficiency of inhibition of PARP-1 enzymatic activity in vitro decreased in order: C2 > C4 > 3-AB>C1 > C3. ICP-MS study of intracellular accumulation and distribution in BRCA1-mutated HCC1937 revealed that C1-C4 entered cells within 24 h. The complex C1 showed the highest intracellular accumulation, nuclear-targeting properties, and exhibited the highest DNA binding (39.2 ± 0.6 pg of Ru per μg of DNA) that resulted in the cell cycle arrest in the S phase.

Graphical abstract

Designing the single molecule that modulates multiple and specific targets, is paradigmin antitumor drug discovery. Here, we describe the synthesis, characterization, and in vitro antitumor activity of ruthenium(II)-arene complexes conjugated with 3-aminobenzamide derivatives, affecting poly(ADP-ribose) polymerase 1 enzymatic activity in vitro and interacting with DNA in human breast cancer cells.

Unlabelled Image
  1. Download : Download high-res image (116KB)
  2. Download : Download full-size image

Introduction

Rapid increase in the number of cancer deaths worldwide makes drug development research highly important. Nowadays, it is well known that novel antitumor drugs developed to replace cisplatin need to outweigh severe adverse effects and accompanying resistance induced by cisplatin. [[1], [2], [3]]. Ruthenium-based compounds are one of the most promising metal-based anticancer drug candidates [4]. The synthetic chemistry of ruthenium complexes is very well established and gives access to a large variety of compounds [5]. Among all synthesized ruthenium complexes so far, two ruthenium(III) complexes, NAMI-A (imidazolium-trans-[tetrachloro-(dimethylsulfoxide)imidazole ruthenium(III)]), which acts on metastases of solid tumors [6], and KP1339 (sodium-trans-[tetrachlorobis(1H-indazole)ruthenium(III)]), which is effective against platinum-resistant tumors [7], completed phase II clinical trials [[8], [9], [10], [11], [12], [13]]. Organometallic ruthenium(II)-arene compounds with completely different metallodrug scaffold, in which three of the coordination sites are occupied by η6-coordinated arene that stabilizes Ru+2 oxidation state, are being under intensive development [14]. RAPTA-C ([Ru(η6-p-cymene)(pta)Cl2], pta = 1,3,5-triaza-7-phosphatricyclo[3.3.1.1]decane), RAED-C ([Ru(η6-p-cymene)(en)Cl][PF6], en = ethylenediamine), and RAPTA-T ([Ru(η6-toluene)(pta)Cl2]), showed to be effective in reducing the number and weight of lung metastases [[15], [16], [17], [18], [19]]. To date, a myriad of structurally different ruthenium(II)-arene complexes have been prepared, with some of them exhibiting notable anticancer activities in vitro [[20], [21], [22], [23], [24], [25], [26]]. Ruthenium(II) have similar ligand exchange kinetics to platinum(II), and the octahedral geometry of ruthenium complexes offers unique possibilities for binding to nucleic acids [27,28].

The numerous still ongoing studies of mechanisms of action of cisplatin and ruthenium-based drugs emphasize the existence of more variable cellular targets of these agents beyond nuclear DNA [[29], [30], [31], [32]]. The particular interest is in investigating enzyme inhibition by metal complexes [33]. In that aspect, Kilpin and Dyson proposed the classification of metal-based enzyme inhibitors to several groups [34], depending on whether the activity of metal complexes relies mostly on the chemical properties of metal centre, bioactive ligand, or was influenced by the combination of both.

Particularly interesting family of enzymes present in eukaryotes are poly(ADP-ribose) polymerases (PARPs), which are involved in various cellular processes, including DNA repair, chromatin remodeling, transcriptional regulation, and cell death [[35], [36], [37]]. PARP-1 is one of the most abundant chromatin-bound protein, accounting for >90% of total PARP activity [38]. It is considered to be a key player in DNA base excision repair (BER) and repair of single-strand DNA breaks (SSBs) in response to ionizing radiation, oxidative stress, or DNA-binding agents [39,40]. Structurally, PARP-1 is comprised of three distinct functional domains: (1) N-terminal DNA-binding domain containing three zinc fingers important for the PARP-1 binding to DNA breaks, (2) central automodification domain serving as an acceptor of adenosine diphosphate ribose (ADP-ribose) moieties, and (3) C-terminal catalytic domain forming active site of PARP-1 (“PARP signature”) that transfers ADP-ribose subunits from nicotinamide adenine dinucleotide (NAD+) to protein acceptors (Fig. 1) [41,42].

PARP-1 utilizes beta nicotinamide adenine dinucleotide (β-NAD+) as a substrate to covalently add poly(ADP-ribose) (PAR) chains onto itself and other nuclear acceptor proteins, in a process termed PARylation [43]. The role of PARP-1 in DNA damage response and cell death regulation prompted the development of potent small molecules named PARP inhibitors (PARPi) [39]. Generally, PARPi are designated as competitive inhibitors, since they impair the catalytic activity of PARP-1 by interacting with the NAD+ binding site [44]. Nicotinamide, benzamide, and 3-aminobenzamide (3-AB) were identified as the first generation of “classical” PARPi [45]. Despite the limited potency and specificity to be used in clinics, these compounds are significant for research purposes [46]. Structure-activity relationship analysis showed that potent PARPi should have an electron-rich aromatic or polyaromatic heterocyclic system, bearing a pharmacophore with cis-configurated carboxamide, imide, or a carbamoyl group [47]. To date, four PARPi of the third generation (olaparib, rucaparib, niraparib, and talazoparib) were approved by the United States Food and Drug Administration (U.S. FDA) (Fig. 2) [42,[48], [49], [50]], as single-agent therapy, for targeting breast cancer susceptibility genes (BRCA)-mutated breast, ovarian, prostate, and pancreatic cancers [51]. PARPi are the first clinically approved drugs designed to exploit synthetic lethality concept in tumors harboring mutations in BRCA1 or BRCA2 genes, responsible for the repair of double-strand DNA breaks (DSBs) by homologous recombination (HR) [52,53]. Synthetic lethality is a genetic concept based on the idea that defect in either one of two genes has little effect on the cell or organism, but a combination of defects in both genes results in cell death [54]. The cells with impaired HR pathway are dependent on alternative ways for DNA repair and survival, and thereby PARPi became promising therapy for BRCA-mutated cancers.

Although recent reports suggested that the application of PARPi could be extended to “BRCAness” tumors with defects in HR repair genes other than BRCA1 and BRCA2 [[55], [56], [57]], the acquired resistance caused by arising secondary mutations in HR repair genes that restore the function of HR repair pathway limits the clinical efficacy of PARPi as single agents [58]. Therefore, combination therapy is a reasonable approach to improve the utility of these inhibitors to serve as chemopotentiators of commonly used cytotoxic chemotherapeutics, such as cisplatin [[59], [60], [61], [62]]. Unfortunately, multicomponent drug cocktails can display adverse effects caused by complex pharmacokinetics and unpredictable drug-drug interactions. Hence, multi-targeting single drugs offer some advantages of pharmacokinetic simplicity and improved outcomes [35].

Growing tendency in the drug design area is to connect metals with pharmacophoric moieties of bioactive ligand, to achieve different spectra of biological activity and improve the properties of both constituents [29,32,63,64]. Reports up to date showed that different ruthenium(II)-arene complexes entered tumor cells efficiently and bound DNA [24,65]. Therefore, they may present appropriate scaffold to bring bioactive ligands with PARP-1 inhibitory potential more closely to their targets in the cell. Additionally, the study performed by Mendes et al has shown that compounds based on platinum, ruthenium, or gold may display high PARP-1 inhibitory activity, supporting the model whereby displacement of zinc from the zinc finger motif of PARP-1 by metal ions led to decreased PARP-1 activity [66].

In the present study, novel ruthenium(II)-arene complexes carrying 3-AB derivatives as ligands, were synthesized and examined for the inhibitory potential against the catalytic activity of PARP-1. Further, their growth inhibitory effects were investigated in a panel of human breast cancer cell lines, which were either BRCA1-mutant, triple-negative, or hormone-responsive. To reveal the mechanism of action and intracellular targets, we analyzed cell cycle progression, binding to DNA, cellular uptake, and distribution across cellular compartments of treated cells. The influence of the particular structural changes in the molecules on the different aspects of anticancer potential is discussed.

Section snippets

Material and methods

RuCl3·3H2O was purchased from Johnson Matthey (London, UK). A 2-amino-4-methylbenzamide (L1) and 3-amino-N-methylbenzamide (L2) were purchased from Sigma Aldrich. [Ru(η6-p-cymene)Cl2]2 was prepared according to a published procedure [67]. [Ru(η6-toluene)Cl2]2 was prepared according to a published procedure [68]. Solvents were obtained from different companies and used without further purification. Infrared spectra were recorded on Nicolet 6700 FTIR spectrometer, using the ATR technique. 1H and

Cell culture

MDA-MB-231 and MCF-7 cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM) (Sigma-Aldrich Co), while HCC1937, MDA-MB-453, MDA-MB-361, and BEAS-2B cells were maintained in Roswell Park Memorial Institute (RPMI) 1640 nutrient medium (Sigma-Aldrich Co). All media were supplemented with 10% fetal calf serum (FCS) (pH 7.2) (Sigma-Aldrich Co), 2 mM l-glutamine, 25 mM 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid (HEPES, Capricorn), and Penicillin-Streptomycin solution

Synthesis and characterization

Four new ruthenium(II)-arene complexes (C1-C4) were synthesized in the reaction of 3-aminobenzamide analogues, 2-amino-4-methylbenzamide (L1) and 3-amino-N-methylbenzamide (L2), to get potentially good PARP inhibitors (Scheme 1). Starting Ru(II) complexes were Ru(II) dimmers with toluene or p-cymene moiety. Synthesis of Ru(II)-p-cymene-based complexes with L1 and L2 has to be done in an oxygen-free atmosphere, as it was previously reported for similar complexes [65]. All syntheses were

Conclusions

In the present study, four new ruthenium(II)-arene complexes, C1-C4, carrying derivatives of 3-aminobenzamide, with anticipated multifunctional potential, were synthesized and tested for their anticancer activity in vitro. Complexes exhibited efficiency in inhibiting PARP-1 activity, at micromolar concentrations, in the following order: C2 > C4 > 3-AB>C1 > C3. Complexes bearing η6-p-cymene moiety (C2 and C4), demonstrated better PARP-1 inhibition than their starting complex C6 and the benchmark

Abbreviations

NAMI-A Imidazolium-trans-[tetrachloro(dimethylsulfoxide)imidazoleruthenium(III)]

KP1339 Sodium-trans-[tetrachlorobis(1H-indazole)ruthenium(III)]

RAPTA-C Ruthenium(II)-dichloro(1,3,5-triaza-7-phosphaadamantane)(η6-p-cymene)

RAED-C Ruthenium(II)chloro(η6-p-cymene)ethylenediamine-hexafluorophosphate

RAPTA-T Ruthenium(II)-dichloro(1,3,5-triaza-7-phosphaadamantane)(η6-toluene)

PARP Poly(ADP-ribose) polymerase

ADP Adenosine diphosphate

BER Base excision repair

SSBs Single-strand breaks

β-NAD+ Beta nicotinamide adenine

Acknowledgements

This work was financed by Ministry of Education, Science and Technological Development of the Republic of Serbia, contract numbers: 451-03-68/2020-14/200168 and 451-03-68/2020-14/200043.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (104)

  • Z. Yuan et al.

    Olaparib hydroxamic acid derivatives as dual PARP and HDAC inhibitors for cancer therapy

    Bioorg. Med. Chem.

    (2017)
  • P.G. Jain et al.

    Medicinal chemistry approaches of poly ADP-ribose polymerase 1 (PARP1) inhibitors as anticancer agents - a recent update

    Eur. J. Med. Chem.

    (2019)
  • M. De Vos et al.

    The diverse roles and clinical relevance of PARPs in DNA damage repair: current state of the art

    Biochem. Pharmacol.

    (2012)
  • A.T.A. Boraei et al.

    Discovery of novel functionalized 1,2,4-triazoles as PARP-1 inhibitors in breast cancer: design, synthesis and antitumor activity evaluation

    Eur. J. Med. Chem.

    (2019)
  • N.J. Curtin et al.

    Therapeutic applications of PARP inhibitors: anticancer therapy and beyond

    Mol. Asp. Med.

    (2013)
  • A. Ashworth et al.

    Genetic interactions in cancer progression and treatment

    Cell

    (2011)
  • A.K. Byrum et al.

    Defining and modulating 'BRCAness'

    Trends Cell Biol.

    (2019)
  • A. Drean et al.

    PARP inhibitor combination therapy

    Crit. Rev. Oncol. Hematol.

    (2016)
  • E. Goodfellow et al.

    Design, synthesis and biological activity of novel molecules designed to target PARP and DNA

    Bioorg. Med. Chem. Lett.

    (2017)
  • Z. Wang et al.

    Multi-targeted organometallic ruthenium(II)–arene anticancer complexes bearing inhibitors of poly(ADP-ribose) polymerase-1: a strategy to improve cytotoxicity

    J. Inorg. Biochem.

    (2014)
  • S.B. Jensen et al.

    Facile preparation of η6-p-cymene ruthenium diphosphine complexes. Crystal structure of [(η6-p-cymene)Ru(dppf)cl]PF6

    J. Organomet. Chem.

    (1998)
  • W.J. Geary

    The use of conductivity measurements in organic solvents for the characterisation of coordination compounds

    Coordin. Chem. Rev.

    (1971)
  • S. Grgurić-Šipka et al.

    Ruthenium(II)–arene complexes with functionalized pyridines: synthesis, characterization and cytotoxic activity

    Eur. J. Med. Chem.

    (2010)
  • P.A. Kenny et al.

    HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment

    Mol. Oncol.

    (2007)
  • R.M. Neve et al.

    A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes

    Cancer Cell

    (2006)
  • O. Tari et al.

    Synthesis, characterization and DNA binding studies of platinum(II) complexes with benzimidazole derivative ligands

    Bioorg. Chem.

    (2017)
  • D.D. Von Hoff et al.

    Toxic effects of cis-dichlorodiammineplatinum(II) in man

    Cancer Treat. Rep.

    (1979)
  • L. Astolfi et al.

    Correlation of adverse effects of cisplatin administration in patients affected by solid tumours: a retrospective evaluation

    Oncol. Rep.

    (2013)
  • R. Oun et al.

    The side effects of platinum-based chemotherapy drugs: a review for chemists

    Dalton Trans.

    (2018)
  • P.V. Simpson et al.

    Metal-based antitumor compounds: beyond cisplatin

    Future Med. Chem.

    (2019)
  • G. Sava et al.

    Na[trans-RuCl4(DMSO)Im], a metal complex of ruthenium with antimetastatic properties

    Clin. Exp. Metastasis

    (1992)
  • M.R. Berger et al.

    Efficacy of new ruthenium complexes against chemically induced autochthonous colorectal carcinoma in rats

    Anticancer Res.

    (1989)
  • R. Trondl et al.

    NKP-1339, the first ruthenium-based anticancer drug on the edge to clinical application

    Chem. Sci.

    (2014)
  • N.R. Dickson et al.

    A phase I dose-escalation study of NKP-1339 in patients with advanced solid tumors refractory to treatment

    J. Clin. Oncol.

    (2011)
  • J.M. Rademaker-Lakhai et al.

    A phase I and pharmacological study with imidazolium-trans-DMSO-imidazole-tetrachlororuthenate, a novel ruthenium anticancer agent

    Clin. Cancer Res.

    (2004)
  • S. Leijen et al.

    Phase I/II study with ruthenium compound NAMI-A and gemcitabine in patients with non-small cell lung cancer after first line therapy

    Investig. New Drugs

    (2015)
  • E. Alessio et al.

    NAMI-A and KP1019/1339, two iconic ruthenium anticancer drug candidates face-to-face: a case story in medicinal inorganic chemistry

    Molecules

    (2019)
  • G. Suss-Fink

    Areneruthenium complexes as anticancer agents

    Dalton Trans.

    (2010)
  • Y.K. Yan et al.

    Organometallic chemistry, biology and medicine: ruthenium arene anticancer complexes

    Chem. Commun. (Camb)

    (2005)
  • P.J. Dyson

    Systematic Design of a Targeted Organometallic Antitumour Drug in pre-clinical development

    Chimia

    (2007)
  • C. Scolaro et al.

    In vitro and in vivo evaluation of ruthenium(II)−Arene PTA complexes

    J. Med. Chem.

    (2005)
  • A. Bergamo et al.

    Modulation of the metastatic progression of breast cancer with an organometallic ruthenium compound

    Int. J. Oncol.

    (2008)
  • B. Biersack

    Anticancer activity and modes of action of (arene) ruthenium(II) complexes coordinated to C-, N-, and O-ligands

    Mini Rev. Med. Chem.

    (2016)
  • B.J. Reedijk

    Metal-ligand exchange kinetics in platinum and ruthenium complexes

    Platin. Met. Rev.

    (2008)
  • G. Sava et al.

    Metal-based antitumour drugs in the post-genomic era: what comes next?

    Dalton Trans.

    (2011)
  • M. Raudenska et al.

    Unexpected therapeutic effects of cisplatin

    Metallomics

    (2019)
  • G. Palermo et al.

    Fighting cancer with transition metal complexes: from naked DNA to protein and chromatin targeting strategies

    ChemMedChem

    (2016)
  • R.G. Kenny et al.

    Toward multi-targeted platinum and ruthenium drugs—a new paradigm in Cancer drug treatment regimens?

    Chem. Rev.

    (2019)
  • G. Gasser et al.
  • K.J. Kilpin et al.

    Enzyme inhibition by metal complexes: concepts, strategies and applications

    Chem. Sci.

    (2013)
  • Cited by (14)

    • Exploring heterometallic bridged Pt(II)-Zn(II) complexes as potential antitumor agents

      2023, Journal of Inorganic Biochemistry
      Citation Excerpt :

      PARP1 and PARP2 are activated in response to DNA damaging agents such as alkylating agents (cisplatin), topoisomerase inhibitors (etoposide) etc. that cause DNA single-strand break (SSBR), and have been recognized as promising target in various cancers which exhibit defective DDR pathways. We investigated whether PARP1/2 may mediate chemosensitivity to novel Pt(II)-L-Zn(II) complexes [52–56]. The effect of heterometallic Pt(II)-L-Zn(II) complexes C1 or trans-C2 on cell cycle progression in HCT116 cells was examined using propidium iodide (PI) staining and flow cytometry analysis.

    • Effect of substituents on the binding of Ru(II) polypyridyl complexes [Ru(bpy)<inf>2</inf>L]<sup>2+</sup> (L = pnpip and mnpip) to poly(U-A•U) triplex

      2022, Dyes and Pigments
      Citation Excerpt :

      Poly(U-A•U) triplex was prepared by incubating a 1:1 M ratio mixture of poly(U) and poly(A)•poly(U) at 90 °C for 30 min followed by 24 h annealing at room temperature in pH 7.0 phosphate buffer (6 mmol/L Na2HPO4, 2 mmol/L NaH2PO4, 1 mmol/L Na2EDTA, 19 mmol/L NaCl). The formation of the triplex was confirmed by its characteristic CD spectrum (Fig. S1) and melting profile (Fig. S2) [28]. Polynucleotide concentrations were determined spectrophotometrically using the molar extinction coefficients (which were expressed in terms of nucleotide phosphates): ε260 = 9350 M−1 cm−1 for poly(U), ϵ260 = 7140 M−1 cm−1 for poly(A)•poly(U), and ε257 = 5900 M−1 cm−1 for poly(U-A•U) [29].

    • Target based chemotherapeutic advancement of ruthenium complexes

      2021, Coordination Chemistry Reviews
      Citation Excerpt :

      Compounds possessing benzamide ligand show lower cytotoxicity values. Cellular distribution studies revealed that the complexes mostly accumulate in cytosol and membrane/organelle fraction [44]. Malignant tumor usually shows upregulation of lipoxygenase (LOX) and cyclooxygenase (COX-1 and COX-2) [45].

    • Design concepts of half-sandwich organoruthenium anticancer agents based on bidentate bioactive ligands

      2021, Coordination Chemistry Reviews
      Citation Excerpt :

      Derivatives of 3-aminobenzamide and nicotinamide inhibit poly(ADP-ribose) polymerase (PARP), an enzyme involved in DNA repair and other transcription processes, and related PARP inhibitors have been approved by the FDA for clinical use [80,81]. A derivative of the first generation PARP inhibitors, i.e., 2-amino-4-methylbenzamide, was treated with [Ru(cym)Cl2]2 to prepare NN-27a and NN-27b (Fig. 20; Fig. 1, concept i) [81]. While the complexes showed more potent PARP inhibition than the respective ligands, antiproliferation assays indicated minor activity with NN-27b being the most potent compound.

    • Monodentately-coordinated bioactive moieties in multimodal half-sandwich organoruthenium anticancer agents

      2021, Coordination Chemistry Reviews
      Citation Excerpt :

      Notably, both complexes showed improved bioactivity over their free ligands which highlights the potential for synergistic targeting properties through transcription inhibition and DNA binding. Complexes N-30a and N-30b containing a 3-aminobenzamide-derived PARP inhibitor (N-ah) (Fig. 20) were assessed for inhibition of cell growth against five human breast cancer cell lines (Hcc1937, MDA-MB-231, MDA-MB-453, MDA-MB-361, MCF-7) as well as non-cancerous bronchial epithelium BEAS-2B cells [173]. Both complexes showed moderate anticancer activity across all cell lines without selectivity for cancer cells (Table S1).

    View all citing articles on Scopus
    View full text