1
|
Sayala J, Srivastava E, Kumar P, Shukla N, Kumar A, Patra AK. Photocytotoxic kinetically stable ruthenium(II)- N, N-donor polypyridyl complexes of oxalate with anticancer activity against HepG2 liver cancer cells. Dalton Trans 2024; 53:4580-4597. [PMID: 38349214 DOI: 10.1039/d3dt04058e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Liver cancer is one of the leading causes of death that motivating scientists worldwide to synthesize novel chemotherapeutics. Ru(II)-polypyridyl complexes are extensively studied for possible therapeutic and cellular applications due to their tunable coordination chemistry, structural diversity, ligand-exchange kinetics, accessible redox states, and rich photophysical or photochemical properties. Herein, we have synthesized a series of Ru(II) polypyridyl complexes [RuII(N^N)2(ox)] (1-3), where ox is oxalate (C2O42-) and N^N is 1,10-phenanthroline (phen) (1), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq) (2), and dipyrido[3,2,-a:2',3'-c]phenazine (dppz) (3). Oxalate (ox2-) was opted as a bioactive dioxo ligand to prevent facile hydrolysis in aqueous media, thereby increasing the stability of the Ru(II)-polypyridyl complexes in physiological media. We thoroughly characterized all the complexes using ESI-MS, FT-IR, UV-vis, and 1H NMR spectroscopy and other physicochemical methods. The complexes were stable under physiological conditions and under low-energy green LED light (λirr = 530 nm). However, the photoirradiation of complexes resulted in the efficient generation of singlet oxygen (1O2) as a major reactive oxygen species (ROS). The role of the extended aromatic conjugation of the N^N-donor ligands in the complexes was demonstrated by their binding propensities with CT-DNA and bovine serum albumin (BSA). Both DNA intercalation and groove binding were evidenced, while tryptophan (Trp) and tyrosine (Tyr) binding site preferences were revealed from the synchronous fluorescence spectra (SFS) of BSA. The cytotoxic profiling of the complexes performed on hepatocellular carcinoma cells (HepG2) in the dark and in the presence of green light indicated their dose-dependent cytotoxicity. The [RuII(N^N)2(ox)] complexes exhibited enhanced photocytotoxicity mediated by efficient generation of cytotoxic 1O2 and effective interaction with DNA. All the complexes were internalized by the HepG2 liver cancer cells efficiently and localized to the cytoplasm and nucleus. The complexes exhibited potent anti-proliferative, anti-clonogenic, and anti-migratory effects on the cancer cells, suggesting their potential for therapeutic applications.
Collapse
Affiliation(s)
- Juhi Sayala
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Ekta Srivastava
- Department of Biological Science & Bioengineering Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Priyaranjan Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Nitin Shukla
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| | - Ashok Kumar
- Department of Biological Science & Bioengineering Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashis K Patra
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India.
| |
Collapse
|
2
|
Skoczynska A, Lewinski A, Pokora M, Paneth P, Budzisz E. An Overview of the Potential Medicinal and Pharmaceutical Properties of Ru(II)/(III) Complexes. Int J Mol Sci 2023; 24:ijms24119512. [PMID: 37298471 DOI: 10.3390/ijms24119512] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
This review examines the existing knowledge about Ru(II)/(III) ion complexes with a potential application in medicine or pharmacy, which may offer greater potential in cancer chemotherapy than Pt(II) complexes, which are known to cause many side effects. Hence, much attention has been paid to research on cancer cell lines and clinical trials have been undertaken on ruthenium complexes. In addition to their antitumor activity, ruthenium complexes are under evaluation for other diseases, such as type 2 diabetes, Alzheimer's disease and HIV. Attempts are also being made to evaluate ruthenium complexes as potential photosensitizers with polypyridine ligands for use in cancer chemotherapy. The review also briefly examines theoretical approaches to studying the interactions of Ru(II)/Ru(III) complexes with biological receptors, which can facilitate the rational design of ruthenium-based drugs.
Collapse
Affiliation(s)
- Anna Skoczynska
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Andrzej Lewinski
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Mateusz Pokora
- International Center of Research on Innovative Biobased Materials (ICRI-BioM)-International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Piotr Paneth
- International Center of Research on Innovative Biobased Materials (ICRI-BioM)-International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Medical University of Lodz, 90-151 Lodz, Poland
| |
Collapse
|
3
|
Chen J, Liu H, Chen Y, Hu H, Huang C, Wang Y, Liang L, Liu Y. Iridium(III) complexes inhibit the proliferation and migration of BEL-7402 cells through the PI3K/AKT/mTOR signaling pathway. J Inorg Biochem 2023; 241:112145. [PMID: 36709684 DOI: 10.1016/j.jinorgbio.2023.112145] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
Iridium(III) complexes are largely studied as anti-cancer complexes due to their excellent anti-cancer activity. In this article, two new iridium(III) complexes [Ir(piq)2(THPIP)]PF6 (THPIP = 2,4-di-tert-butyl-6-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)phenol, piq = deprotonated 1-phenylisoquinoline) (Ir1) and [Ir(bzq)2(THPIP)]PF6 (bzq = deprotonated benzo[h]quinolone) (Ir2) were synthesized. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays showed that complex Ir1 exhibits moderate activity (IC50 = 29.9 ± 4.6 μM) and Ir2 shows high cytotoxicity (IC50 = 9.8 ± 1.8 μM) against BEL-7402 cells. Further studies on the mechanism showed that Ir1 and Ir2 induced apoptosis by changing the mitochondrial membrane potential, Ca2+ release, ROS accumulation, and cell cycle arrest at the S phase. The complexes can effectively inhibit cell colony formation and migration. The expression of B-cell lymphoma-2 (Bcl-2) family proteins, PI3K (phosphatidylinositol 3-kinase), AKT (protein kinase B), mTOR (mammalian target of rapamycin), and p-mTOR was studied by immunoblotting. Complexes Ir1 and Ir2 downregulated the expression of anti-apoptotic protein Bcl-2 and increased the expression of autophagy-related proteins of Beclin-1 and LC3-II. Further experiments showed that the complexes inhibited the production of glutathione (GSH) and increased the amounts of malondialdehyde (MDA). Fluorescence of HMGB1 was significantly increased. We also investigated the effect of the complexes on the expression of genes using RNA-sequence analysis, we further calculated the lowest binding energies between the complexes and proteins using molecular docking. Taken together, the above results indicated that complexes Ir1 and Ir2 induce apoptosis in BEL-7402 cells through a ROS-mediated mitochondrial dysfunction and inhibition of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Haimei Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yichuan Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Shaban NZ, Aboelsaad AM, Awad D, Abdulmalek SA, Shaban SY. Therapeutic effect of dithiophenolato chitosan nanocomposites against carbon tetrachloride-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:8487-8502. [PMID: 34487322 DOI: 10.1007/s11356-021-15834-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/02/2021] [Indexed: 06/13/2023]
Abstract
Our previous study showed that dithiophenolate (DTP) and its chitosan nanoparticles (DTP-CSNPs) have abilities to bind with DNA helixes. So in this study, their lethal doses (LD50) and therapeutic roles against rat liver injuries induced by carbon tetrachloride (CCl4) were evaluated. The study focused on the determination of the markers of oxidative stress (OS) and apoptosis and compare the results with those of cisplatin treatment. The results revealed that LD50 values of DTP and DTP-CSNPs are 2187.5 and 1462.5 mg/kg, respectively. Treatment with DPT and DPT-CSNPs after CCl4 administration reduced liver injuries, induced by CCl4, and improved liver functions and architecture through the reduction of OS and apoptosis. Where the oxidant marker was decreased with elevations of antioxidant markers. Also, there was an elevation in Bcl-2 value, with decreases in caspase-8, Bax, and Bax/Bcl-2 ratio. DPT-CSNPs treatment gave preferable results than those treated with DPT. Moreover, DTP and DPT-CSNPs treatment gave better results than cisplatin treatment. The administration of healthy rats with low doses of DTP and DTP-CSNPs for 14 days had no effect. Otherwise, the study on HepG2 cell line showed that DTP and DPT-CSNPs inhibited cell growth by arresting cells in the G2/M phase and inducing cell death. In conclusion, DTP and DTP-CSNPs have antiapoptotic and anti-oxidative stress toward hepatotoxicity induced by CCl4. Moreover, DTP and DTP-CSNPs have anticancer activity against the HepG2 cell line. Generally, DTP-CSNPs are more effective than DTP. So, they can be used in the pharmacological fields, especially DTP-CSNPs.
Collapse
Affiliation(s)
- Nadia Z Shaban
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Ahmed M Aboelsaad
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Doaa Awad
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Shaymaa A Abdulmalek
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Shaban Y Shaban
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| |
Collapse
|
5
|
Hairat S, Zaki M. Half sandwiched RutheniumII complexes: En Route towards the targeted delivery by Human Serum Albumin (HSA). J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
6
|
Sudhindra P, Ajay Sharma S, Roy N, Moharana P, Paira P. Recent advances in cytotoxicity, cellular uptake and mechanism of action of ruthenium metallodrugs: A review. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
7
|
Ferraro MG, Piccolo M, Misso G, Maione F, Montesarchio D, Caraglia M, Paduano L, Santamaria R, Irace C. Breast Cancer Chemotherapeutic Options: A General Overview on the Preclinical Validation of a Multi-Target Ruthenium(III) Complex Lodged in Nucleolipid Nanosystems. Cells 2020; 9:E1412. [PMID: 32517101 PMCID: PMC7349411 DOI: 10.3390/cells9061412] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review we have showcased the preclinical development of original amphiphilic nanomaterials designed for ruthenium-based anticancer treatments, to be placed within the current metallodrugs approach leading over the past decade to advanced multitarget agents endowed with limited toxicity and resistance. This strategy could allow for new options for breast cancer (BC) interventions, including the triple-negative subtype (TNBC) with poor therapeutic alternatives. BC is currently the second most widespread cancer and the primary cause of cancer death in women. Hence, the availability of novel chemotherapeutic weapons is a basic requirement to fight BC subtypes. Anticancer drugs based on ruthenium are among the most explored and advanced next-generation metallotherapeutics, with NAMI-A and KP1019 as two iconic ruthenium complexes having undergone clinical trials. In addition, many nanomaterial Ru complexes have been recently conceived and developed into anticancer drugs demonstrating attractive properties. In this field, we focused on the evaluation of a Ru(III) complex-named AziRu-incorporated into a suite of both zwitterionic and cationic nucleolipid nanosystems, which proved to be very effective for the in vivo targeting of breast cancer cells (BBC). Mechanisms of action have been widely explored in the context of preclinical evaluations in vitro, highlighting a multitarget action on cell death pathways which are typically deregulated in neoplasms onset and progression. Moreover, being AziRu inspired by the well-known NAMI-A complex, information on non-nanostructured Ru-based anticancer agents have been included in a precise manner.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| |
Collapse
|
8
|
Riccardi C, Musumeci D, Trifuoggi M, Irace C, Paduano L, Montesarchio D. Anticancer Ruthenium(III) Complexes and Ru(III)-Containing Nanoformulations: An Update on the Mechanism of Action and Biological Activity. Pharmaceuticals (Basel) 2019; 12:E146. [PMID: 31561546 PMCID: PMC6958509 DOI: 10.3390/ph12040146] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
The great advances in the studies on metal complexes for the treatment of different cancer forms, starting from the pioneering works on platinum derivatives, have fostered an increasingly growing interest in their properties and biomedical applications. Among the various metal-containing drugs investigated thus far, ruthenium(III) complexes have emerged for their selective cytotoxic activity in vitro and promising anticancer properties in vivo, also leading to a few candidates in advanced clinical trials. Aiming at addressing the solubility, stability and cellular uptake issues of low molecular weight Ru(III)-based compounds, some research groups have proposed the development of suitable drug delivery systems (e.g., taking advantage of nanoparticles, liposomes, etc.) able to enhance their activity compared to the naked drugs. This review highlights the unique role of Ru(III) complexes in the current panorama of anticancer agents, with particular emphasis on Ru-containing nanoformulations based on the incorporation of the Ru(III) complexes into suitable nanocarriers in order to enhance their bioavailability and pharmacokinetic properties. Preclinical evaluation of these nanoaggregates is discussed with a special focus on the investigation of their mechanism of action at a molecular level, highlighting their pharmacological potential in tumour disease models and value for biomedical applications.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Carlo Irace
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, I-80131 Naples, Italy.
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| |
Collapse
|
9
|
Piccolo M, Misso G, Ferraro MG, Riccardi C, Capuozzo A, Zarone MR, Maione F, Trifuoggi M, Stiuso P, D'Errico G, Caraglia M, Paduano L, Montesarchio D, Irace C, Santamaria R. Exploring cellular uptake, accumulation and mechanism of action of a cationic Ru-based nanosystem in human preclinical models of breast cancer. Sci Rep 2019; 9:7006. [PMID: 31065032 PMCID: PMC6505035 DOI: 10.1038/s41598-019-43411-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
According to WHO, breast cancer incidence is increasing so that the search for novel chemotherapeutic options is nowadays an essential requirement to fight neoplasm subtypes. By exploring new effective metal-based chemotherapeutic strategies, many ruthenium complexes have been recently proposed as antitumour drugs, showing ability to impact on diverse cellular targets. In the framework of different molecular pathways leading to cell death in human models of breast cancer, here we demonstrate autophagy involvement behind the antiproliferative action of a ruthenium(III)-complex incorporated into a cationic nanosystem (HoThyRu/DOTAP), proved to be hitherto one of the most effective within the suite of nucleolipidic formulations we have developed for the in vivo transport of anticancer ruthenium(III)-based drugs. Indeed, evidences are implicating autophagy in both cancer development and therapy, and anticancer interventions endowed with the ability to trigger this biological response are currently considered attractive oncotherapeutic approaches. Moreover, crosstalk between apoptosis and autophagy, regulated by finely tuned metallo-chemotherapeutics, may provide novel opportunities for future improvement of cancer treatment. Following this line, our in vitro and in vivo preclinical investigations suggest that an original strategy based on suitable formulations of ruthenium(III)-complexes, inducing sustained cell death, could open new opportunities for breast cancer treatment, including the highly aggressive triple-negative subtype.
Collapse
Affiliation(s)
- Marialuisa Piccolo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Claudia Riccardi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
| | - Antonella Capuozzo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Mayra Rachele Zarone
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
| | - Paola Stiuso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Gerardino D'Errico
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
- CSGI - Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy.
- CSGI - Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy.
| | - Carlo Irace
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy.
| | - Rita Santamaria
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| |
Collapse
|
10
|
Lin K, Zhao ZZ, Bo HB, Hao XJ, Wang JQ. Applications of Ruthenium Complex in Tumor Diagnosis and Therapy. Front Pharmacol 2018; 9:1323. [PMID: 30510511 PMCID: PMC6252376 DOI: 10.3389/fphar.2018.01323] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/29/2018] [Indexed: 12/27/2022] Open
Abstract
Ruthenium complexes are a new generation of metal antitumor drugs that are currently of great interest in multidisciplinary research. In this review article, we introduce the applications of ruthenium complexes in the diagnosis and therapy of tumors. We focus on the actions of ruthenium complexes on DNA, mitochondria, and endoplasmic reticulum of cells, as well as signaling pathways that induce tumor cell apoptosis, autophagy, and inhibition of angiogenesis. Furthermore, we highlight the use of ruthenium complexes as specific tumor cell probes to dynamically monitor the active biological component of the microenvironment and as excellent photosensitizer, catalyst, and bioimaging agents for phototherapies that significantly enhance the diagnosis and therapeutic effect on tumors. Finally, the combinational use of ruthenium complexes with existing clinical antitumor drugs to synergistically treat tumors is discussed.
Collapse
Affiliation(s)
- Ke Lin
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zi-Zhuo Zhao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hua-Ben Bo
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Juan Hao
- Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Jin-Quan Wang
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
11
|
Bergamo A, Dyson PJ, Sava G. The mechanism of tumour cell death by metal-based anticancer drugs is not only a matter of DNA interactions. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.01.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Yao D, Wang P, Zhang J, Fu L, Ouyang L, Wang J. Deconvoluting the relationships between autophagy and metastasis for potential cancer therapy. Apoptosis 2018; 21:683-98. [PMID: 27003389 DOI: 10.1007/s10495-016-1237-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a highly conserved lysosome-dependent degradation process that may digest some long-lived proteins and damaged organelles. As an essential homeostasis maintaining system in normal cells, autophagy plays a key role in several pathological settings, especially cancer. Metastasis, known as a crucial hallmark of cancer progression, is the primary cause of cancer lethality. The role of autophagy in metastasis is quite complex as supportive evidence has indicated both pro-metastatic and anti-metastatic functions of autophagy. Autophagy can inhibit metastasis by restricting necrosis and mediating autophagic cell death, whereas it may also promote metastasis by enhancing cancer cell fitness in response to stress. Moreover, the function of autophagy is context- and stage-dependent. Specifically, during the early steps of metastasis, autophagy mainly serves as a suppressor, while it plays a pro-metastatic role in the later steps. Here, we focus on highlighting the dual roles of autophagy in metastasis and address the molecular mechanisms involved in this process, which may provide a new insight into cancer biology. While, we also summarize several anti-metastatic agents manipulating autophagy, in the hope of shedding light on exploration of potential novel drugs for future cancer therapy.
Collapse
Affiliation(s)
- Dahong Yao
- State Key Laboratory of Biotherapy & Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Peiqi Wang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy & Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Leilei Fu
- State Key Laboratory of Biotherapy & Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy & Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jinhui Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
13
|
Zeng L, Gupta P, Chen Y, Wang E, Ji L, Chao H, Chen ZS. The development of anticancer ruthenium(ii) complexes: from single molecule compounds to nanomaterials. Chem Soc Rev 2017; 46:5771-5804. [PMID: 28654103 PMCID: PMC5624840 DOI: 10.1039/c7cs00195a] [Citation(s) in RCA: 729] [Impact Index Per Article: 104.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer is rapidly becoming the top killer in the world. Most of the FDA approved anticancer drugs are organic molecules, while metallodrugs are very scarce. The advent of the first metal based therapeutic agent, cisplatin, launched a new era in the application of transition metal complexes for therapeutic design. Due to their unique and versatile biochemical properties, ruthenium-based compounds have emerged as promising anti-cancer agents that serve as alternatives to cisplatin and its derivertives. Ruthenium(iii) complexes have successfully been used in clinical research and their mechanisms of anticancer action have been reported in large volumes over the past few decades. Ruthenium(ii) complexes have also attracted significant attention as anticancer candidates; however, only a few of them have been reported comprehensively. In this review, we discuss the development of ruthenium(ii) complexes as anticancer candidates and biocatalysts, including arene ruthenium complexes, polypyridyl ruthenium complexes, and ruthenium nanomaterial complexes. This review focuses on the likely mechanisms of action of ruthenium(ii)-based anticancer drugs and the relationship between their chemical structures and biological properties. This review also highlights the catalytic activity and the photoinduced activation of ruthenium(ii) complexes, their targeted delivery, and their activity in nanomaterial systems.
Collapse
Affiliation(s)
- Leli Zeng
- College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Flamme M, Cressey PB, Lu C, Bruno PM, Eskandari A, Hemann MT, Hogarth G, Suntharalingam K. Induction of Necroptosis in Cancer Stem Cells using a Nickel(II)-Dithiocarbamate Phenanthroline Complex. Chemistry 2017; 23:9674-9682. [PMID: 28556445 DOI: 10.1002/chem.201701837] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Indexed: 12/27/2022]
Abstract
The cytotoxic properties of a series of nickel(II)-dithiocarbamate phenanthroline complexes is reported. The complexes 1-6 kill bulk cancer cells and cancer stem cells (CSCs) with micromolar potency. Two of the complexes, 2 and 6, kill twice as many breast cancer stem cell (CSC)-enriched HMLER-shEcad cells as compared to breast CSC-depleted HMLER cells. Complex 2 inhibits mammosphere formation to a similar extent as salinomycin (a CSC-specific toxin). Detailed mechanistic studies suggest that 2 induces CSC death by necroptosis, a programmed form of necrosis. Specifically, 2 triggers MLKL phosphorylation, oligomerization, and translocation to the cell membrane. Additionally, 2 induces necrosome-mediated propidium iodide (PI) uptake and mitochondrial membrane depolarisation, as well as morphological changes consistent with necroptotosis. Strikingly, 2 does not evoke necroptosis by intracellular reactive oxygen species (ROS) production or poly(ADP) ribose polymerase (PARP-1) activation.
Collapse
Affiliation(s)
- Marie Flamme
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | - Paul B Cressey
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | - Chunxin Lu
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | - Peter M Bruno
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Arvin Eskandari
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | - Michael T Hemann
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Graeme Hogarth
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | | |
Collapse
|
15
|
Chow MJ, Alfiean M, Pastorin G, Gaiddon C, Ang WH. Apoptosis-independent organoruthenium anticancer complexes that overcome multidrug resistance: self-assembly and phenotypic screening strategies. Chem Sci 2017; 8:3641-3649. [PMID: 30155208 PMCID: PMC6094174 DOI: 10.1039/c7sc00497d] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/22/2017] [Indexed: 01/20/2023] Open
Abstract
Multidrug resistance is a major impediment to chemotherapy and limits the efficacies of conventional anticancer drugs. A strategy to bypass multidrug resistance is to develop new drug candidates capable of inducing apoptosis-independent programmed cell death. However, cellular pathways implicated in alternative programmed cell death are not well-elucidated and multifactorial, making a target-based discovery approach a challenge. Here, we show that a coordination-directed three-component assembly and phenotypic screening strategy could be employed as a viable alternative for the identification of apoptosis-independent organoruthenium anticancer agents. Through an on-plate synthesis and screening of 195 organoruthenium complexes against apoptosis-sensitive and -resistant cancers, we identified two apoptosis-independent hits. Subsequent validation of the two hits showed a lack of induction of apoptotic biomarkers, a caspase-independent activity and an equal efficacy in both apoptosis-sensitive and -resistant colorectal cancers. This validated their apoptosis-independent modes-of-action, paving the way as potential candidates for the treatment of highly-refractory cancer phenotypes.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore .
| | - Mohammad Alfiean
- School of Chemistry and Life Sciences , Nanyang Polytechnic , Singapore
| | - Giorgia Pastorin
- Department of Pharmacy , National University of Singapore , 3 Science Drive 3 , 117543 Singapore
- NUS Graduate School for Integrative Sciences and Engineering , Singapore . ; Tel: +65 6516 5131
| | - Christian Gaiddon
- Université de Strasbourg , Strasbourg , France
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France
| | - Wee Han Ang
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore .
- NUS Graduate School for Integrative Sciences and Engineering , Singapore . ; Tel: +65 6516 5131
| |
Collapse
|
16
|
Zhang C, Lai SH, Yang HH, Xing DG, Zeng CC, Tang B, Wan D, Liu YJ. Photoinduced ROS regulation of apoptosis and mechanism studies of iridium(iii) complex against SGC-7901 cells. RSC Adv 2017. [DOI: 10.1039/c7ra00732a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A new iridium(iii) complex, Ir(ppy)2(FBPIP)]PF6 (Ir-1), was synthesized and characterized. The anticancer activity of the complex was investigated by cytotoxicity in vitro, apoptosis, cell invasion, autophagy, cell cycle arrest and western blot.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Shang-Hai Lai
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Hui-Hui Yang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - De-Gang Xing
- School of Basic Course
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Chuan-Chuan Zeng
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Bing Tang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Dan Wan
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Yun-Jun Liu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
- Guangdong Cosmetics Engineering & Technology Research Center
| |
Collapse
|
17
|
Liu J, Ren H, Liu B, Zhang Q, Li M, Zhu R. Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells. Oncol Lett 2016; 12:4385-4392. [PMID: 28101201 PMCID: PMC5228182 DOI: 10.3892/ol.2016.5301] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 08/26/2016] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC), which is a type of malignant tumor, is the fifth most common cancer in men and ninth in women worldwide. The aim of the present study was to investigate the antitumor effect of diosmetin (DIOS) in hepatocellular carcinoma HepG2 cells. The proliferation, apoptosis and autophagy rates of HepG2 cells were measured following treatment with DIOS. The effects of DIOS treatment on HepG2 cell proliferation and apoptosis rates were analyzed using MTT assays and Annexin V staining, respectively. The effect of DIOS treatment on autophagy levels was assessed using transmission electron microscopy, green fluorescent protein (GFP)-microtubule-associated protein 1 light chain (LC3) transfection and LysoTracker Red staining. Furthermore, bafilomycin A1 (BA1), an autophagy inhibitor, was used to assess the association between DIOS and cell autophagy, proliferation and apoptosis. In addition, the expression of autophagy-related proteins [mammalian target of rapamycin (mTOR), phosphatidylinositol 3-kinase, P70S6K, phosphoinositide-dependent kinase-1, extracellular signal-regulated kinase, 5′-AMP-activated protein kinase and Akt] and apoptosis-related proteins [B-cell lymphoma (Bcl)-2-associated X protein, Bak, p53, Bcl-2 and caspase-3] were analyzed by western blotting. The results revealed that DIOS significantly inhibited proliferation (P<0.01) and induced apoptosis (P<0.001) in HepG2 cells. It was also demonstrated that DIOS triggered autophagy by regulating the mTOR pathway in HepG2 cells. Notably, following treatment of HepG2 cells with the autophagy inhibitor, BA1, the expression of apoptosis-related proteins, including Bax, Bak and p53, were significantly decreased (P<0.05), and cell viability was recovered to a certain extent. In conclusion, DIOS inhibits cell proliferation and induces apoptosis in HepG2 cells via regulation of the mTOR pathway. Thus, the results of the current study indicate that DIOS may present a potential therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Jie Liu
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Hao Ren
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Bin Liu
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Qingyu Zhang
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Mingyi Li
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Runzhi Zhu
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
18
|
Koceva-Chyła A, Matczak K, Hikisz MP, Durka MK, Kochel MK, Süss-Fink G, Furrer J, Kowalski K. Insights into the in vitro Anticancer Effects of Diruthenium-1. ChemMedChem 2016; 11:2171-2187. [DOI: 10.1002/cmdc.201600315] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Aneta Koceva-Chyła
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Karolina Matczak
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Paweł Hikisz
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Kamil Durka
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Msc. Krzysztof Kochel
- Department of Medical Biophysics; Faculty of Biology and Environmental Protection; University of Łódź; Pomorska St. 141/143 90236 Łódź Poland
| | - Georg Süss-Fink
- Institut de Chimie; Université de Neuchâtel; Avenue de Bellevaux 51 2000 Neuchâtel Switzerland
| | - Julien Furrer
- Department für Chemie und Biochemie; Universität Bern; Freiestrasse 3 3012 Bern Switzerland
| | - Konrad Kowalski
- Department of Organic Chemistry; Faculty of Chemistry; University of Łódź; Tamka St. 12 91403 Łódź Poland
| |
Collapse
|
19
|
Chow MJ, Licona C, Pastorin G, Mellitzer G, Ang WH, Gaiddon C. Structural tuning of organoruthenium compounds allows oxidative switch to control ER stress pathways and bypass multidrug resistance. Chem Sci 2016; 7:4117-4124. [PMID: 30155055 PMCID: PMC6013925 DOI: 10.1039/c6sc00268d] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/25/2016] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) is a major impediment to the success of chemotherapy in many cancer types. One particular MDR mechanism is the inherent or acquired adaptation of the cellular survival pathways that render malignant cells resistant to apoptotic cell death. Since most drugs act through apoptosis, compounds capable of inducing alternative forms of programmed cell death (PCD) can potentially be harnessed to bypass MDR. We investigated two organoruthenium complexes, RAS-1H and RAS-1T, and demonstrated that although they both induced non-apoptotic PCD through ER stress pathways, their modes-of-action were drastically different despite modest structural variations. RAS-1T acted through ROS-mediated ER stress while RAS-1H was ROS-independent. We further showed that they were more efficacious against apoptosis-resistant cells compared to clinical drugs including oxaliplatin. This work provides the basis for underpinning ER stress modulation using metal complexes to bypass apoptosis resistance.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore . ; Tel: +65 6516 5131
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
| | - Cynthia Licona
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| | - Giorgia Pastorin
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
- Department of Pharmacy , National University of Singapore , 18 Science Drive 4 , 117543 Singapore
| | - Georg Mellitzer
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| | - Wee Han Ang
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore . ; Tel: +65 6516 5131
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
| | - Christian Gaiddon
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| |
Collapse
|
20
|
Kljun J, Anko M, Traven K, Sinreih M, Pavlič R, Peršič Š, Ude Ž, Codina EE, Stojan J, Lanišnik Rižner T, Turel I. Pyrithione-based ruthenium complexes as inhibitors of aldo-keto reductase 1C enzymes and anticancer agents. Dalton Trans 2016; 45:11791-800. [PMID: 27357845 DOI: 10.1039/c6dt00668j] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Four ruthenium complexes of clinically used zinc ionophore pyrithione and its oxygen analog 2-hydroxypyridine N-oxide were prepared and evaluated as inhibitors of enzymes of the aldo-keto reductase subfamily 1C (AKR1C). A kinetic study assisted with docking simulations showed a mixed type of inhibition consisting of a fast reversible and a slow irreversible step in the case of both organometallic compounds 1A and 1B. Both compounds also showed a remarkable selectivity towards AKR1C1 and AKR1C3 which are targets for breast cancer drug design. The organoruthenium complex of ligand pyrithione as well as pyrithione itself also displayed toxicity on the hormone-dependent MCF-7 breast cancer cell line with EC50 values in the low micromolar range.
Collapse
Affiliation(s)
- Jakob Kljun
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ruthenium Complexes Induce HepG2 Human Hepatocellular Carcinoma Cell Apoptosis and Inhibit Cell Migration and Invasion through Regulation of the Nrf2 Pathway. Int J Mol Sci 2016; 17:ijms17050775. [PMID: 27213353 PMCID: PMC4881594 DOI: 10.3390/ijms17050775] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/28/2016] [Accepted: 05/05/2016] [Indexed: 11/17/2022] Open
Abstract
Ruthenium (Ru) complexes are currently the focus of substantial interest because of their potential application as chemotherapeutic agents with broad anticancer activities. This study investigated the in vitro and in vivo anticancer activities and mechanisms of two Ru complexes—2,3,7,8,12,13,17,18-Octaethyl-21H,23H-porphine Ru(II) carbonyl (Ru1) and 5,10,15,20-Tetraphenyl-21H,23H-porphine Ru(II) carbonyl (Ru2)—against human hepatocellular carcinoma (HCC) cells. These Ru complexes effectively inhibited the cellular growth of three human hepatocellular carcinoma (HCC) cells, with IC50 values ranging from 2.7–7.3 μM. In contrast, the complexes exhibited lower toxicity towards L02 human liver normal cells with IC50 values of 20.4 and 24.8 μM, respectively. Moreover, Ru2 significantly inhibited HepG2 cell migration and invasion, and these effects were dose-dependent. The mechanistic studies demonstrated that Ru2 induced HCC cell apoptosis, as evidenced by DNA fragmentation and nuclear condensation, which was predominately triggered via caspase family member activation. Furthermore, HCC cell treatment significantly decreased the expression levels of Nrf2 and its downstream effectors, NAD(P)H: quinone oxidoreductase 1 (NQO1) and heme oxygenase 1 (HO1). Ru2 also exhibited potent in vivo anticancer efficacy in a tumor-bearing nude mouse model, as demonstrated by a time- and dose-dependent inhibition on tumor growth. The results demonstrate the therapeutic potential of Ru complexes against HCC via Nrf2 pathway regulation.
Collapse
|
22
|
Lai SH, Li W, Wang XZ, Zhang C, Zeng CC, Tang B, Wan D, Liu YJ. Apoptosis, autophagy, cell cycle arrest, cell invasion and BSA-binding studies in vitro of ruthenium(ii) polypyridyl complexes. RSC Adv 2016. [DOI: 10.1039/c6ra11391e] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Four new ruthenium(ii) polypyridyl complexes were synthesized and characterized. The anticancer activity was investigated by cytotoxicity in vitro, apoptosis, comet assay, ROS, autophagy, cell invasion and western blot analysis.
Collapse
Affiliation(s)
- Shang-Hai Lai
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Wei Li
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Xiu-Zhen Wang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Cheng Zhang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Chuan-Chuan Zeng
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Bing Tang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Dan Wan
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| | - Yun-Jun Liu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P.R. China
| |
Collapse
|