Enriching Chemical Space of Bioactive Scaffolds by New Ring Systems: Benzazocines and Their Metal Complexes as Potential Anticancer Drugs

The search for new scaffolds of medicinal significance combined with molecular shape enhances their innovative potential and continues to attract the attention of researchers. Herein, we report the synthesis, spectroscopic characterization (1H and 13C NMR, UV–vis, IR), ESI-mass spectrometry, and single-crystal X-ray diffraction analysis of a new ring system of medicinal significance, 5,6,7,9-tetrahydro-8H-indolo[3,2-e]benzazocin-8-one, and a series of derived potential ligands (HL1–HL5), as well as ruthenium(II), osmium(II), and copper(II) complexes (1a, 1b, and 2–5). The stability of compounds in 1% DMSO aqueous solutions has been confirmed by 1H NMR and UV–vis spectroscopy measurements. The antiproliferative activity of HL1–HL5 and 1a, 1b, and 2–5 was evaluated by in vitro cytotoxicity tests against four cancer cell lines (LS-174, HCT116, MDA-MB-361, and A549) and one non-cancer cell line (MRC-5). The lead compounds HL5 and its copper(II) complex 5 were 15× and 17×, respectively, more cytotoxic than cisplatin against human colon cancer cell line HCT116. Annexin V-FITC apoptosis assay showed dominant apoptosis inducing potential of both compounds after prolonged treatment (48 h) in HCT116 cells. HL5 and 5 were found to induce a concentration- and time-dependent arrest of cell cycle in colon cancer cell lines. Antiproliferative activity of 5 in 3D multicellular tumor spheroid model of cancer cells (HCT116, LS-174) superior to that of cisplatin was found. Moreover, HL5 and 5 showed notable inhibition potency against glycogen synthase kinases (GSK-3α and GSK-3β), tyrosine-protein kinase (Src), lymphocyte-specific protein-tyrosine kinase (Lck), and cyclin-dependent kinases (Cdk2 and Cdk5) (IC50 = 1.4–6.1 μM), suggesting their multitargeted mode of action as potential anticancer drugs.

[1]  G. Gasser,et al.  Red-Absorbing Ru(II) Polypyridyl Complexes with Biotin Targeting Spontaneously Assemble into Nanoparticles in Biological Media. , 2022, Inorganic chemistry.

[2]  V. Brabec,et al.  Ruthenium(II)–Tris-pyrazolylmethane Complexes Inhibit Cancer Cell Growth by Disrupting Mitochondrial Calcium Homeostasis , 2022, Journal of medicinal chemistry.

[3]  Richard D. Taylor,et al.  Rings in Clinical Trials and Drugs: Present and Future , 2022, Journal of medicinal chemistry.

[4]  W. Ang,et al.  Elucidation of Structure–Activity Relationships in Indolobenzazepine-Derived Ligands and Their Copper(II) Complexes: the Role of Key Structural Components and Insight into the Mechanism of Action , 2022, Inorganic chemistry.

[5]  G. Spengler,et al.  Highly Antiproliferative Latonduine and Indolo[2,3-c]quinoline Derivatives: Complex Formation with Copper(II) Markedly Changes the Kinase Inhibitory Profile , 2022, Journal of medicinal chemistry.

[6]  J. Sessler,et al.  Metal-based anticancer agents as immunogenic cell death inducers: the past, present, and future. , 2022, Chemical Society reviews.

[7]  L. Massai,et al.  Proteomics as a tool to disclose the cellular and molecular mechanisms of selected anticancer gold compounds , 2021, Coordination Chemistry Reviews.

[8]  I. Baranowska-Bosiacka,et al.  Inhibitors of Cyclin-Dependent Kinases: Types and Their Mechanism of Action , 2021, International journal of molecular sciences.

[9]  Ting Guo,et al.  Recent Advances in the Discovery of Multitargeted Tyrosine Kinase Inhibitors as Anticancer Agents , 2020, ChemMedChem.

[10]  P. Perego,et al.  The rediscovery of platinum-based cancer therapy , 2020, Nature reviews. Cancer.

[11]  A. Tesei,et al.  Modeling neoplastic disease with spheroids and organoids , 2020, Journal of Hematology & Oncology.

[12]  B. El Hassouni,et al.  A Brief Guide to Performing Pharmacological Studies In Vitro: Reflections from the EORTC-PAMM Course “Preclinical and Early-phase Clinical Pharmacology” , 2019, AntiCancer Research.

[13]  S. Mobin,et al.  Mitochondrial Localization of Highly Fluorescent and Photostable BODIPY-Based Ruthenium(II), Rhodium(III), and Iridium(III) Metal Complexes. , 2019, Inorganic chemistry.

[14]  D. Colin,et al.  Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma , 2019, Scientific Reports.

[15]  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 , 2019, Molecules.

[16]  S. Radulović,et al.  Antitumor activity of organoruthenium complexes with chelate aromatic ligands, derived from 1,10-phenantroline: Synthesis and biological activity. , 2019, Journal of inorganic biochemistry.

[17]  Seon Woong Lee,et al.  BODIPY-based Ru(II) and Ir(III) organometallic complexes of avobenzone, a sunscreen material: Potent anticancer agents. , 2018, Journal of inorganic biochemistry.

[18]  H. Rupasinghe,et al.  Kinase-targeted cancer therapies: progress, challenges and future directions , 2018, Molecular Cancer.

[19]  A. Buschini,et al.  Effects of polar substituents on the biological activity of thiosemicarbazone metal complexes. , 2018, Journal of inorganic biochemistry.

[20]  Azah Kamilah Muda,et al.  Similarity Measure for Molecular Structure: A Brief Review , 2017 .

[21]  Richard D. Taylor,et al.  Combining Molecular Scaffolds from FDA Approved Drugs: Application to Drug Discovery. , 2017, Journal of medicinal chemistry.

[22]  Sasanka Deka,et al.  Novel mitochondria targeted copper(ii) complexes of ferrocenyl terpyridine and anticancer active 8-hydroxyquinolines showing remarkable cytotoxicity, DNA and protein binding affinity. , 2017, Dalton transactions.

[23]  Christine Unger,et al.  Comparison of cancer cells in 2D vs 3D culture reveals differences in AKT–mTOR–S6K signaling and drug responses , 2017, Journal of Cell Science.

[24]  Arindam Mukherjee,et al.  Anticancer activity of a chelating nitrogen mustard bearing tetrachloridoplatinum(iv) complex: better stability yet equipotent to the Pt(ii) analogue. , 2016, Dalton transactions.

[25]  G. Devi,et al.  Three-dimensional culture systems in cancer research: Focus on tumor spheroid model. , 2016, Pharmacology & therapeutics.

[26]  L. Kenner,et al.  Comparison of cancer cells cultured in 2 D vs 3 D reveals differences in AKT / mTOR / S 6-kinase signaling and drug response , 2016 .

[27]  C. Unger,et al.  Three-dimensional and co-culture models for preclinical evaluation of metal-based anticancer drugs , 2015, Investigational New Drugs.

[28]  Agnieszka K. Witkiewicz,et al.  The history and future of targeting cyclin-dependent kinases in cancer therapy , 2015, Nature Reviews Drug Discovery.

[29]  S. Radulović,et al.  Synthesis, characterization and cytotoxic activity of novel platinum(II) iodido complexes. , 2014, European journal of medicinal chemistry.

[30]  P. Sadler,et al.  Organoiridium Complexes: Anticancer Agents and Catalysts , 2014, Accounts of chemical research.

[31]  Richard D. Taylor,et al.  Rings in drugs. , 2014, Journal of medicinal chemistry.

[32]  V. Arion,et al.  Metal‐Based Indolobenzazepines and Indoloquinolines: From Moderate CDK Inhibitors to Potential Antitumor Drugs , 2013 .

[33]  W. Berger,et al.  Metal–Arene Complexes with Indolo[3,2-c]-quinolines: Effects of Ruthenium vs Osmium and Modifications of the Lactam Unit on Intermolecular Interactions, Anticancer Activity, Cell Cycle, and Cellular Accumulation , 2013, Organometallics.

[34]  Daniel V LaBarbera,et al.  The multicellular tumor spheroid model for high-throughput cancer drug discovery , 2012, Expert opinion on drug discovery.

[35]  P. Dyson,et al.  Metal-based antitumour drugs in the post-genomic era: what comes next? , 2011, Dalton transactions.

[36]  Xiaoyuan Chen,et al.  Apoptosis Imaging: Beyond Annexin V , 2010, The Journal of Nuclear Medicine.

[37]  Simone Göschl,et al.  Structure-activity relationships of highly cytotoxic copper(II) complexes with modified indolo[3,2-c]quinoline ligands. , 2010, Inorganic chemistry.

[38]  D. Dalvie,et al.  Chapter 6:Influence of Aromatic Rings on ADME Properties of Drugs , 2010 .

[39]  W. Berger,et al.  Organometallic indolo[3,2-c]quinolines versus indolo[3,2-d]benzazepines: synthesis, structural and spectroscopic characterization, and biological efficacy , 2010, JBIC Journal of Biological Inorganic Chemistry.

[40]  Tingjun Hou,et al.  Drug and Drug Candidate Building Block Analysis , 2010, J. Chem. Inf. Model..

[41]  M. Jakupec,et al.  Highly cytotoxic copper(II) complexes with modified paullone ligands. , 2010, Inorganic chemistry.

[42]  C. Humblet,et al.  Escape from flatland: increasing saturation as an approach to improving clinical success. , 2009, Journal of medicinal chemistry.

[43]  F. de Vos,et al.  Synthesis, in vitro and in vivo evaluation, and radiolabeling of aryl anandamide analogues as candidate radioligands for in vivo imaging of fatty acid amide hydrolase in the brain. , 2009, Journal of medicinal chemistry.

[44]  Martin Krug,et al.  Recent advances in the development of multi-kinase inhibitors. , 2008, Mini reviews in medicinal chemistry.

[45]  G. Sheldrick A short history of SHELX. , 2008, Acta crystallographica. Section A, Foundations of crystallography.

[46]  P. Cohen,et al.  The selectivity of protein kinase inhibitors: a further update. , 2007, The Biochemical journal.

[47]  W. Berger,et al.  Metal-based paullones as putative CDK inhibitors for antitumor chemotherapy. , 2007, Journal of medicinal chemistry.

[48]  L. Kèlland,et al.  The resurgence of platinum-based cancer chemotherapy , 2007, Nature Reviews Cancer.

[49]  Carlo Riccardi,et al.  Analysis of apoptosis by propidium iodide staining and flow cytometry , 2006, Nature Protocols.

[50]  Peter Ertl,et al.  Quest for the rings. In silico exploration of ring universe to identify novel bioactive heteroaromatic scaffolds. , 2006, Journal of medicinal chemistry.

[51]  Dong Wang,et al.  Cellular processing of platinum anticancer drugs , 2005, Nature Reviews Drug Discovery.

[52]  Gavin Harper,et al.  Drug rings database with web interface. A tool for identifying alternative chemical rings in lead discovery programs. , 2003, Journal of medicinal chemistry.

[53]  P. Cohen,et al.  The specificities of protein kinase inhibitors: an update. , 2003, The Biochemical journal.

[54]  G. Schneider,et al.  Scaffold architecture and pharmacophoric properties of natural products and trade drugs: application in the design of natural product-based combinatorial libraries. , 2001, Journal of combinatorial chemistry.

[55]  G L Snyder,et al.  Paullones are potent inhibitors of glycogen synthase kinase-3beta and cyclin-dependent kinase 5/p25. , 2000, European journal of biochemistry.

[56]  P. Cohen,et al.  Specificity and mechanism of action of some commonly used protein kinase inhibitors. , 2000, The Biochemical journal.

[57]  L. Meijer,et al.  Paullones, a series of cyclin-dependent kinase inhibitors: synthesis, evaluation of CDK1/cyclin B inhibition, and in vitro antitumor activity. , 1999, Journal of medicinal chemistry.

[58]  C. Reutelingsperger,et al.  Annexin V-affinity assay: a review on an apoptosis detection system based on phosphatidylserine exposure. , 1998, Cytometry.

[59]  G. Bemis,et al.  The properties of known drugs. 1. Molecular frameworks. , 1996, Journal of medicinal chemistry.

[60]  M. N. Burnett,et al.  ORTEP-III: Oak Ridge Thermal Ellipsoid Plot Program for crystal structure illustrations , 1996 .

[61]  R. Supino MTT assays. , 1995, Methods in molecular biology.

[62]  H. Fujita [Anticancer drugs]. , 1995, Nihon rinsho. Japanese journal of clinical medicine.

[63]  A. W. Addison,et al.  Synthesis, structure, and spectroscopic properties of copper(II) compounds containing nitrogen–sulphur donor ligands; the crystal and molecular structure of aqua[1,7-bis(N-methylbenzimidazol-2′-yl)-2,6-dithiaheptane]copper(II) perchlorate , 1984 .

[64]  C. Jørgensen "Symbiotic" Ligands, Hard and Soft Central Atoms , 1964 .

[65]  Highly Antiproliferative Latonduine and Indolo[2,3c]quinoline Derivatives: Complex Formation with Copper(II) Markedly Changes the Kinase Inhibitory Profile , 2022 .