1
|
Facile synthesis of heterobimetallic [FeII(µ-diphosphine)RuII] and homobimetallic [FeII(µ-diphosphine)FeII] complexes and their in vitro cytotoxic activity on cisplatin-resistant cancer cells. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
2
|
Nabiyeva T, Marschner C, Blom B. Synthesis, structure and anti-cancer activity of osmium complexes bearing π-bound arene substituents and phosphane Co-Ligands: A review. Eur J Med Chem 2020; 201:112483. [PMID: 32592914 DOI: 10.1016/j.ejmech.2020.112483] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 12/24/2022]
Abstract
While many examples of osmium complexes, as anti-cancer agents, have been reported and some reviews have been devoted to this topic, a particularly interesting and synthetically accessible sub-class of these compounds namely those bearing a π- bound arene and phosphane co-ligand have escaped review. These complexes have made a surprisingly late entry in the literature (2005) in terms of anti-cancer investigations. This is somewhat surprising considering the plethora of analogous complexes that have been reported for the lighter analogue, ruthenium. Herein we review all complexes, neutral and ionic, bearing the "(ƞ6-arene)Os(PR3)" moiety focusing on their synthesis, reactivity, structural features (by X-ray diffraction analysis) as well as anti-cancer biological activity. An attempt is made throughout the article to contrast these to each other and to analogous Ru systems, and a full summary of all existing in vitro biological data is presented.
Collapse
Affiliation(s)
- Tomiris Nabiyeva
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Kapoenstraat 2, PO Box 616, 6200, MD, Maastricht, the Netherlands
| | - Christoph Marschner
- Institut für Anorganische Chemie, Technische Universität Graz, Stremayrgasse 9, A-8010, Graz, Austria
| | - Burgert Blom
- Maastricht Science Programme, Faculty of Science and Engineering, Maastricht University, Kapoenstraat 2, PO Box 616, 6200, MD, Maastricht, the Netherlands.
| |
Collapse
|
3
|
Franco Machado J, Machuqueiro M, Marques F, Robalo MP, Piedade MFM, Garcia MH, Correia JDG, Morais TS. Novel "ruthenium cyclopentadienyl"-peptide conjugate complexes against human FGFR(+) breast cancer. Dalton Trans 2020; 49:5974-5987. [PMID: 32314752 DOI: 10.1039/d0dt00955e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this work we explored the possibility of improving the selectivity of a cytotoxic Ru complex [RuCp(PPh3)(2,2'-bipy)][CF3SO3] (where Cp = η5-cyclopentadienyl) TM34 towards FGFR(+) breast cancer cells. Molecular dynamics (MD) simulations of TM34 in a phosphatidylcholine membrane model pinpointed the cyclopentadienyl group as a favorable derivatization position for the peptide conjugation approach. Three new Ru(ii) complexes presenting a functionalized η5-cyclopentadienyl were synthesized, namely [Ru(η5-C5H4COOH)(2,2'-bipy)(PPh3)][CF3SO3] (TM281) and its precursors, [Ru(η5-C5H4COOCH2CH3)(η2-2,2'-bipy)(PPh3)][CF3SO3] (3) and [Ru(η5-C5H4COOCH2CH3)(PPh3)2Cl] (2). Complex TM281 was prepared by the hydrolysis of the ethyl ester group appended to the η5-cyclopentadienyl ligand of complex 3 with K2CO3 in water/acetonitrile, followed by mild protonation using an ion exchange resin. The newly synthesized complexes were fully characterized by NMR, FTIR and UV-vis spectroscopic techniques. Also, electrochemical studies were carried out by means of cyclic voltammetry in order to evaluate the stability of the compounds. Single crystal X-ray diffraction studies were carried out for compounds 3 and TM281 which crystallized in the monoclinic system, space group P21/n. The unprecedented synthesis and characterization of three half-sandwich ruthenium(ii)-cyclopentadienyl peptide conjugates and their preliminary biological evaluation against human FGFR(+) and FGFR(-) breast cancer cells are also reported.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Synthesis, structure and biological evaluation of ruthenium(III) complexes of triazolopyrimidines with anticancer properties. J Biol Inorg Chem 2019; 25:109-124. [PMID: 31741123 DOI: 10.1007/s00775-019-01743-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Six novel ruthenium(III) complexes of general formula [RuCl3(L)3] (1,3,5) and [RuCl3(H2O)(L)2] (2,4,6), where L stands for three different triazolopyrimidine-derived ligands, are reported. The compounds have been structurally characterized (IR, EPR, SCXRD), and their magnetic moments have been determined. The single-crystal X-ray diffraction study revealed a slightly distorted octahedral geometry of the Ru(III) complexes with mer configuration in 1 and 5, and fac configuration in 3. In 2 and 4, three chloride ions are in mer configuration and the two triazolopyrimidines are oriented trans mutually with the water molecule playing the role of the sixth ligand. All complexes have been thoroughly screened for their in vitro cytotoxicity against human breast cancer cell line MCF-7, human cervical cancer cell line HeLa, and L929 murine fibroblast cells, uncovering among others that the most lipophilic complexes 5 and 6, containing the bulky ligand dptp (5,7-diphenyl-1,2,4-triazolo[1,5-a]pyrimidine), display high cytotoxic activity against MCF-7, and HeLa cells. Moreover, it was also revealed that during the interaction of the complexes 1-6 with the cancer MCF-7 cell line, reactive oxygen species are released intracellularly, which could indicate that they are involved in cell apoptosis. Furthermore, extensive studies have been carried out to reveal the mechanism by which complexes 1-6 interact with DNA, albumin, and apotransferrin. The biological studies were complemented by detailed kinetic studies of the hydrolysis of the complexes in the pH range 5-8, to determine the stability of the complexes in solution. Six novel ruthenium(III) complexes with triazolopyrimidine derivatives demonstrated the potential for use as anticancer agents by maintaining the toxic effect on MCF-7 and HeLa cells.
Collapse
|
5
|
Guan S, Pan T, Zhang Y, Zeng Z, Mu L, Zhu D, Chang B, Zheng K, Qian J, Xie Q, Mei W, Tang W, Bai M. Synthesis, DNA-binding, and antitumor activity of polypyridyl-ruthenium(II) complexes [Ru(L)2(DClPIP)] (L = bpy, phen; DClPIP = 2-(2,4-dichlorophenyl)-1H-imidazo[4,5-f][1, 10]phenanthroline). J COORD CHEM 2019. [DOI: 10.1080/00958972.2019.1630614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Shouhai Guan
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Pan
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanyang Zhang
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaolin Zeng
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Luwen Mu
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Duo Zhu
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boyang Chang
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kangdi Zheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiesheng Qian
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xie
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjie Mei
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenjie Tang
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mingjun Bai
- Department of Vascular Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
NAMI-A and KP1019/1339, Two Iconic Ruthenium Anticancer Drug Candidates Face-to-Face: A Case Story in Medicinal Inorganic Chemistry. Molecules 2019; 24:molecules24101995. [PMID: 31137659 PMCID: PMC6571951 DOI: 10.3390/molecules24101995] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 01/23/2023] Open
Abstract
NAMI-A ((ImH)[trans-RuCl4(dmso-S)(Im)], Im = imidazole) and KP1019/1339 (KP1019 = (IndH)[trans-RuCl4(Ind)2], Ind = indazole; KP1339 = Na[trans-RuCl4(Ind)2]) are two structurally related ruthenium(III) coordination compounds that have attracted a lot of attention in the medicinal inorganic chemistry scientific community as promising anticancer drug candidates. This has led to a considerable amount of studies on their respective chemico-biological features and to the eventual admission of both to clinical trials. The encouraging pharmacological performances qualified KP1019 mainly as a cytotoxic agent for the treatment of platinum-resistant colorectal cancers, whereas the non-cytotoxic NAMI-A has gained the reputation of being a very effective antimetastatic drug. A critical and strictly comparative analysis of the studies conducted so far on NAMI-A and KP1019 allows us to define the state of the art of these experimental ruthenium drugs in terms of the respective pharmacological profiles and potential clinical applications, and to gain some insight into the inherent molecular mechanisms. Despite their evident structural relatedness, deeply distinct biological and pharmacological profiles do emerge. Overall, these two iconic ruthenium complexes form an exemplary and unique case in the field of medicinal inorganic chemistry.
Collapse
|
7
|
Caterino M, Herrmann M, Merlino A, Riccardi C, Montesarchio D, Mroginski MA, Musumeci D, Ruffo F, Paduano L, Hildebrandt P, Kozuch J, Vergara A. On the pH-Modulated Ru-Based Prodrug Activation Mechanism. Inorg Chem 2019; 58:1216-1223. [PMID: 30614697 DOI: 10.1021/acs.inorgchem.8b02667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The RuIII-based prodrug AziRu efficiently binds to proteins, but the mechanism of its release is still disputed. Herein, in order to test the hypothesis of a reduction-mediated Ru release from proteins, a Raman-assisted crystallographic study on AziRu binding to a model protein (hen egg white lysozyme), in two different oxidation states, RuII and RuIII, was carried out. Our results indicate Ru reduction, but the Ru release upon reduction is dependent on the reducing agent. To better understand this process, a pH-dependent, spectroelectrochemical surface-enhanced Raman scattering (SERS) study was performed also on AziRu-functionalized Au electrodes as a surrogate and simplest model system of RuII- and RuIII-based drugs. This SERS study provided a p Ka of 6.0 ± 0.4 for aquated AziRu in the RuIII state, which falls in the watershed range of pH values separating most cancer environments from their physiological counterparts. These experiments also indicate a dramatic shift of the redox potential E0 by >600 mV of aquated AziRu toward more positive potentials upon acidification, suggesting a selective AziRu reduction in cancer lumen but not in healthy ones. It is expected that the nature of the ligands (e.g., pyridine vs imidazole, present in well-known RuIII complex NAMI-A) will modulate the p Ka and E0, without affecting the underlying reaction mechanism.
Collapse
Affiliation(s)
- Marco Caterino
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Mona Herrmann
- Institut für Chemie , Technische Universität Berlin , Straße des 17 Juni 135 , Berlin 10623 , Germany
| | - Antonello Merlino
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Claudia Riccardi
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Maria A Mroginski
- Institut für Chemie , Technische Universität Berlin , Straße des 17 Juni 135 , Berlin 10623 , Germany
| | - Domenica Musumeci
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Francesco Ruffo
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Luigi Paduano
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy
| | - Peter Hildebrandt
- Institut für Chemie , Technische Universität Berlin , Straße des 17 Juni 135 , Berlin 10623 , Germany
| | - Jacek Kozuch
- Institut für Chemie , Technische Universität Berlin , Straße des 17 Juni 135 , Berlin 10623 , Germany
| | - Alessandro Vergara
- Department of Chemical Sciences , University of Naples Federico II , via Cinthia , Naples I-80126 , Italy.,CEINGE, Biotecnologie Avanzate s.c.a.r.l.m. , via G Salvatore , Naples I-80131 , Italy
| |
Collapse
|
8
|
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]
|
9
|
Ravi Kumar V, Nagababu P, Srinivas G, Rajender Reddy M, Vinoda Rani M, Ravi M, Satyanarayana S. Investigation of DNA/BSA binding of three Ru(II) complexes by various spectroscopic methods, molecular docking and their antimicrobial activity. J COORD CHEM 2017. [DOI: 10.1080/00958972.2017.1407410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | - Penumaka Nagababu
- CSIR-National Environmental Engineering Research Institute (NEERI) Kolkata Zonal Centre, Kolkata, India
- Inorganic & Physical Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - G. Srinivas
- Department of Chemistry, Osmania University, Hyderabad, India
| | | | - M. Vinoda Rani
- Department of Physics, Osmania University, Hyderabad, India
| | - Mudavath Ravi
- Department of Chemistry, Osmania University, Hyderabad, India
| | | |
Collapse
|
10
|
Oszajca M, Mrugała B, Brindell M. Aqueous behavior and reactivity towards nitric oxide of NAMI-A type complexes bearing bulky N-heterocyclic ligands. Inorganica Chim Acta 2017. [DOI: 10.1016/j.ica.2016.09.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
11
|
Bergamo A, Pelillo C, Chambery A, Sava G. Influence of components of tumour microenvironment on the response of HCT-116 colorectal cancer to the ruthenium-based drug NAMI-A. J Inorg Biochem 2017; 168:90-97. [DOI: 10.1016/j.jinorgbio.2016.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
|
12
|
Biological activity and binding properties of [Ru(II)(dcbpy)2Cl2] complex to bovine serum albumin, phospholipase A2 and glutathione. Biometals 2016; 29:921-33. [PMID: 27515969 DOI: 10.1007/s10534-016-9964-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/31/2016] [Indexed: 02/02/2023]
Abstract
Ruthenium compounds are highly regarded as metallo-drug candidates. Many studies have focused their attention on the interaction between ruthenium complexes with their possible biological targets. The interaction of ruthenium complexes with transport proteins, enzymes and peptides is of great importance for understanding their biodistribution and mechanism of action, therefore, the development of an anti-cancer therapy involving ruthenium complexes has recently shifted from DNA targeting towards protein targeting. With the aim of gaining insight into possible interactions between ruthenium complexes with biologically relevant proteins, we have studied the interaction of cis-dichlorobis(2,2'-bipyridyl-4,4'-dicarboxylic acid)ruthenium(II) complex [Ru(II)(dcbpy)2Cl2], which previously showed good potency in photo-dynamic chemotherapy, with bovine serum albumin (BSA), phospholipase A2 (PLA2) and glutathione (GSH). Binding constants and possible number of binding sites to mentioned proteins and peptide are investigated by ultraviolet-visible spectroscopy and Matrix-Assisted Laser Desorption Ionization Mass Spectrometry (MALDI TOF MS). The complex binding affinities were in the following order: PLA2 > BSA > GSH. Moreover, genotoxic profile of the complex, tested on peripheral blood lymphocytes as a model system, was also promising.
Collapse
|
13
|
Castellarin A, Zorzet S, Bergamo A, Sava G. Pharmacological Activities of Ruthenium Complexes Related to Their NO Scavenging Properties. Int J Mol Sci 2016; 17:ijms17081254. [PMID: 27490542 PMCID: PMC5000652 DOI: 10.3390/ijms17081254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is considered responsible for the growth of primary tumours and of their metastases. With the present study, the effects of three ruthenium compounds, potassiumchlorido (ethylendiamminotetraacetate)rutenate(III) (RuEDTA), sodium (bis-indazole)tetrachloro-ruthenate(III), Na[trans-RuCl₄Ind₂] (KP1339) and trans-imidazoledimethylsulphoxidetetrachloro-ruthenate (NAMI-A), are studied in vitro in models mimicking the angiogenic process. The ruthenium compounds reduced the production and the release of nitrosyls from either healthy macrophages and immortalized EA.hy926 endothelial cells. The effects of NAMI-A are qualitatively similar and sometimes quantitatively superior to those of RuEDTA and KP1339. NAMI-A reduces the production and release of nitric oxide (NO) by the EA.hy926 endothelial cells and correspondingly inhibits their invasive ability; it also strongly inhibits the angiogenesis in matrigel sponges implanted subcutaneously in healthy mice. Taken together, these data support the anti-angiogenic activity of the tested ruthenium compounds and they contribute to explain the selective activity of NAMI-A against solid tumour metastases, the tumour compartment on which angiogenesis is strongly involved. This anti-angiogenic effect may also contribute to the inhibition of the release of metastatic cells from the primary tumour. Investigations on the anti-angiogenic effects of NAMI-A at this level will increase knowledge of its pharmacological properties and it will give a further impulse to the development of this class of innovative metal-based drugs.
Collapse
Affiliation(s)
- Anna Castellarin
- Callerio Foundation Onlus, via A. Fleming 22-31, 34127 Trieste, Italy.
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Sonia Zorzet
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Alberta Bergamo
- Callerio Foundation Onlus, via A. Fleming 22-31, 34127 Trieste, Italy.
| | - Gianni Sava
- Callerio Foundation Onlus, via A. Fleming 22-31, 34127 Trieste, Italy.
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| |
Collapse
|
14
|
N. Deepika, C. Shobha Devi, Y. Praveen Kumar, K. Laxma Reddy, P. Venkat Reddy, D. Anil Kumar, Surya S. Singh, S. Satyanarayana. DNA-binding, cytotoxicity, cellular uptake, apoptosis and photocleavage studies of Ru(II) complexes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 160:142-53. [DOI: 10.1016/j.jphotobiol.2016.03.061] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/28/2022]
|
15
|
Rojas S, Carmona FJ, Maldonado CR, Barea E, Navarro JAR. RAPTA-C incorporation and controlled delivery from MIL-100(Fe) nanoparticles. NEW J CHEM 2016. [DOI: 10.1039/c5nj02741a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The properties of MIL-100(Fe) nanoparticles as vehicles of a non-conventional half-sandwich ruthenium(ii) metallodrug in simulated intravenous conditions have been investigated.
Collapse
Affiliation(s)
- Sara Rojas
- Departamento de Química Inorgánica
- Universidad de Granada
- Granada
- Spain
| | | | | | - Elisa Barea
- Departamento de Química Inorgánica
- Universidad de Granada
- Granada
- Spain
| | | |
Collapse
|
16
|
Mu C, Chang SW, Prosser KE, Leung AWY, Santacruz S, Jang T, Thompson JR, Yapp DTT, Warren JJ, Bally MB, Beischlag TV, Walsby CJ. Induction of Cytotoxicity in Pyridine Analogues of the Anti-metastatic Ru(III) Complex NAMI-A by Ferrocene Functionalization. Inorg Chem 2015; 55:177-90. [PMID: 26652771 DOI: 10.1021/acs.inorgchem.5b02109] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of novel ferrocene (Fc) functionalized Ru(III) complexes was synthesized and characterized. These compounds are derivatives of the anti-metastatic Ru(III) complex imidazolium [trans-RuCl4(1H-imidazole) (DMSO-S)] (NAMI-A) and are derived from its pyridine analogue (NAMI-Pyr), with direct coupling of Fc to pyridine at the 4 or 3 positions, or at the 4 position via a two-carbon linker, which is either unsaturated (vinyl) or saturated (ethyl). Electron paramagnetic resonance (EPR) and UV-vis spectroscopic studies of the ligand exchange processes of the compounds in phosphate buffered saline (PBS) report similar solution behavior to NAMI-Pyr. However, the complex with Fc substitution at the 3 position of the coordinated pyridine shows greater solution stability, through resistance to the formation of oligomeric species. Further EPR studies of the complexes with human serum albumin (hsA) indicate that the Fc groups enhance noncoordinate interactions with the protein and help to inhibit the formation of protein-coordinated species, suggesting the potential for enhanced bioavailability. Cyclic voltammetry measurements demonstrate that the Fc groups modestly reduce the reduction potential of the Ru(III) center as compared to NAMI-Pyr, while the reduction potentials of the Fc moieties of the four compounds vary by 217 mV, with the longer linkers giving significantly lower values of E1/2. EPR spectra of the compounds with 2-carbon linkers show the formation of a high-spin Fe(III) species (S = 5/2) in PBS with a distinctive signal at g = 4.3, demonstrating oxidation of the Fe(II) ferrocene center and likely reflecting degradation products. Density functional theory calculations and paramagnetic (1)H NMR describe delocalization of spin density onto the ligands and indicate that the vinyl linker could be a potential pathway for electron transfer between the Ru and Fe centers. In the case of the ethyl linker, electron transfer is suggested to occur via an indirect mechanism enabled by the greater flexibility of the ligand. In vitro assays with the SW480 cell line reveal cytotoxicity induced by the ruthenium ferrocenylpyridine complexes that is at least an order of magnitude higher than the unfunctionalized complex, NAMI-Pyr. Furthermore, migration studies with LNCaP cells reveal that Fc functionalization does not reduce the ability of the compounds to inhibit cell motility. Overall, these studies demonstrate that NAMI-A-type compounds can be functionalized with redox-active ligands to produce both cytotoxic and anti-metastatic activity.
Collapse
Affiliation(s)
| | | | | | - Ada W Y Leung
- Department of Experimental Therapeutics, BC Cancer Agency , 675 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | | | | | - Donald T T Yapp
- Department of Experimental Therapeutics, BC Cancer Agency , 675 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Marcel B Bally
- Department of Experimental Therapeutics, BC Cancer Agency , 675 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | | |
Collapse
|
17
|
Leijen S, Burgers SA, Baas P, Pluim D, Tibben M, van Werkhoven E, Alessio E, Sava G, Beijnen JH, Schellens JHM. Phase I/II study with ruthenium compound NAMI-A and gemcitabine in patients with non-small cell lung cancer after first line therapy. Invest New Drugs 2014; 33:201-14. [PMID: 25344453 DOI: 10.1007/s10637-014-0179-1] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/15/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND This phase I/II study determined the maximal tolerable dose, dose limiting toxicities, antitumor activity, the pharmacokinetics and pharmacodynamics of ruthenium compound NAMI-A in combination with gemcitabine in Non-Small Cell Lung Cancer patients after first line treatment. METHODS Initial dose escalation of NAMI-A was performed in a 28 day cycle: NAMI-A as a 3 h infusion through a port-a-cath at a starting dose of 300 mg/m(2) at day 1, 8 and 15, in combination with gemcitabine 1,000 mg/m(2) at days 2, 9 and 16. Subsequently, dose escalation of NAMI-A in a 21 day schedule was explored. At the maximal tolerable dose level of this schedule an expansion group was enrolled of which 15 patients were evaluable for response. RESULTS Due to frequent neutropenic dose interruptions in the third week, the 28 day schedule was amended into a 21 day schedule. The maximal tolerable dose was 300 and 450 mg/m(2) of NAMI-A (21 day schedule). Main adverse events consisted of neutropenia, anemia, elevated liver enzymes, transient creatinine elevation, nausea, vomiting, constipation, diarrhea, fatigue, and renal toxicity. CONCLUSION NAMI-A administered in combination with gemcitabine is only moderately tolerated and less active in NSCLC patients after first line treatment than gemcitabine alone.
Collapse
Affiliation(s)
- Suzanne Leijen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Apoptosis of Dalton's lymphoma due to in vivo treatment with emodin is associated with modulations of hydrogen peroxide metabolizing antioxidant enzymes. Cell Biochem Biophys 2014; 67:439-49. [PMID: 22038303 DOI: 10.1007/s12013-011-9305-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The evolving concept of pro-oxidative mechanism-based antitumor activity of emodin (1,3,8-trihydroxy-6-methyl anthraquinone), derived mainly from in vitro studies, needs to be defined for in vivo tumor models. The present article describes apoptosis and regression of Dalton's lymphoma (DL) in mice by emodin vis a vis modulations of hydrogen peroxide (H2O2) metabolizing antioxidant enzymes in the tumor cells in vivo. A non-toxic dose (40 mg/kg bw) of emodin, given intraperitoneally to the DL bearing mice daily up to 12th post DL transplantation day, caused a significant decline (P < 0.05) in the number of viable DL cells and could significantly increase life span of the DL mice (P < 0.01). A significant decline in Bcl2/Bax ratio consistent with the release of mitochondrial cytochrome c release in DL cells from emodin-treated DL mice suggested that emodin could induce mitochondrial pathway of apoptosis in the DL cells in vivo. Apoptosis of DL cells by emodin was further confirmed by the appearance of smaller DNA fragments on DNA ladder analysis. Over activation of both, the Cu-Zn-superoxide dismutases (SOD1) and Mn-SOD (SOD2), has been found correlated with the tumor suppression. Emodin caused significant increases in the expression and activity of SOD1 and SOD2 in the DL cells. H2O2 produced by SODs is degraded by catalase and glutathione peroxidase in the cells. Both these enzymes were observed to be declined significantly with a concomitant increment in H2O2 concentration (P < 0.01) in the DL cells from emodin-treated DL mice. It is concluded that emodin is able to induce mitochondrial pathway of apoptosis in the DL cells in vivo via reciprocal modulations of H2O2 producing and degrading antioxidant enzymes.
Collapse
|
19
|
Dinda J, Adhikary SD, Roymahapatra G, Nakka KK, Santra MK. Synthesis, structure, electrochemistry and cytotoxicity studies of Ru(II) and Pt(II)–N-heterocyclic carbene complexes of CNC-pincer ligand. Inorganica Chim Acta 2014. [DOI: 10.1016/j.ica.2013.12.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
20
|
Wang JQ, Zhang PY, Qian C, Hou XJ, Ji LN, Chao H. Mitochondria are the primary target in the induction of apoptosis by chiral ruthenium(II) polypyridyl complexes in cancer cells. J Biol Inorg Chem 2013; 19:335-48. [PMID: 24287874 DOI: 10.1007/s00775-013-1069-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/12/2013] [Indexed: 12/22/2022]
Abstract
A series of novel chiral ruthenium(II) polypyridyl complexes (Δ-Ru1, Λ-Ru1, Δ-Ru2, Λ-Ru2, Δ-Ru3, Λ-Ru3) were synthesized and evaluated to determine their antiproliferative activities. Colocalization, inductively coupled plasma mass spectrometry, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay studies showed that these ruthenium(II) complexes accumulated preferentially in the mitochondria and exhibited cytotoxicity against various cancer cells in vitro. The complex Δ-Ru1 is of particular interest because it was found to have half-maximal inhibitory concentrations comparable to those of cisplatin and better activity than cisplatin against a cisplatin-resistant cell line, A549-CP/R. Δ-Ru1 induced alterations in the mitochondrial membrane potential and triggered intrinsic mitochondria-mediated apoptosis in HeLa cells, which involved the regulation of Bcl-2 family members and the activation of caspases. Taken together, these data suggest that Δ-Ru1 may be a novel mitochondria-targeting anticancer agent.
Collapse
Affiliation(s)
- Jin-Quan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | | | | | | | | | | |
Collapse
|
21
|
Morais TS, Santos FC, Jorge TF, Côrte-Real L, Madeira PJA, Marques F, Robalo MP, Matos A, Santos I, Garcia MH. New water-soluble ruthenium(II) cytotoxic complex: biological activity and cellular distribution. J Inorg Biochem 2013; 130:1-14. [PMID: 24145065 DOI: 10.1016/j.jinorgbio.2013.09.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 12/30/2022]
Abstract
A novel water soluble organometallic compound, [RuCp(mTPPMSNa)(2,2'-bipy)][CF3SO3] (TM85, where Cp=η(5)-cyclopentadienyl, mTPPMS=diphenylphosphane-benzene-3-sulfonate and 2,2'-bipy=2,2'-bipyridine) is presented herein. Studies of interactions with relevant proteins were performed to understand the behavior and mode of action of this complex in the biological environment. Electrochemical and fluorescence studies showed that TM85 strongly binds to albumin. Studies carried out to study the formation of TM85 which adducts with ubiquitin and cytochrome c were performed by electrospray ionization mass spectrometry (ESI-MS). Antitumor activity was evaluated against a variety of human cancer cell lines, namely A2780, A2780cisR, MCF7, MDAMB231, HT29, PC3 and V79 non-tumorigenic cells and compared with the reference drug cisplatin. TM85 cytotoxic effect was reduced in the presence of endocytosis modulators at low temperatures, suggesting an energy-dependent mechanism consistent with endocytosis. Ultrastructural analysis by transmission electron microscopy (TEM) revealed that TM85 targets the endomembranar system disrupting the Golgi and also affects the mitochondria. Disruption of plasma membrane observed by flow cytometry could lead to cellular damage and cell death. On the whole, the biological activity evaluated herein combined with the water solubility property suggests that complex TM85 could be a promising anticancer agent.
Collapse
Affiliation(s)
- Tânia S Morais
- Centro de Ciências Moleculares e Materiais, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cheng CC, Lee WL, Su JG, Liu CL. Covalent Interaction of Ru(terpy)(tmephen)Cl+with DNA: A Potential Ruthenium-Based Anticancer Drug. J CHIN CHEM SOC-TAIP 2013. [DOI: 10.1002/jccs.200000025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
23
|
Morais TS, Santos FC, Corte-Real L, Garcia MH. Exploring the effect of the ligand design on the interactions between [Ru(η(5)-C5H5)(PPh3)(N,O)][CF3SO3] complexes and human serum albumin. J Inorg Biochem 2013; 129:94-101. [PMID: 24096130 DOI: 10.1016/j.jinorgbio.2013.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 09/10/2013] [Accepted: 09/10/2013] [Indexed: 01/02/2023]
Abstract
Ruthenium complexes hold a great potential in chemotherapy as an alternative to the classical platinum based drugs. The organometallic compounds studied in the present work were previously found to exhibit important anticancer activities. Here we have investigated the binding of three ruthenium compounds, namely [Ru(η(5)-C5H5)(PPh3)(bopy)][CF3SO3] 1, [Ru(η(5)-C5H5)(PPh3)(2-ap)][CF3SO3] 2, and [Ru(η(5)-C5H5)(PPh3)(isoquinpk)][CF3SO3] 3 (bopy=2-benzoylpyridine; 2-ap=2-acetylpyridine; isoquinpk=1-isoquinolinyl phenyl ketone) to fatty acid human serum albumin (HSA) and fatty acid-free human serum albumin (HSA(faf)) at physiological pH7.4. The influence of the substituent groups on the heteroaromatic (N,O) coordinated ligand was also studied by fluorescence spectroscopy to get information about this binding. The Stern-Volmer quenching constants (KSV) were calculated at 293, 298 and 310K, with the corresponding thermodynamic parameters ∆G, ∆H and ∆S as well. The fluorescence quenching method was used to determine the number of binding sites (n) and association constants (Ka) at the same temperatures. The binding site to HSA was confirmed by competitive studies of the ruthenium compounds with warfarin.
Collapse
Affiliation(s)
- Tânia S Morais
- Centro de Ciências Moleculares e Materiais, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | | | | | | |
Collapse
|
24
|
Synthesis, DNA-binding, Cytotoxicity, Photo Cleavage, Antimicrobial and Docking Studies of Ru(II) Polypyridyl Complexes. J Fluoresc 2013; 23:897-908. [DOI: 10.1007/s10895-013-1209-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 02/24/2013] [Indexed: 11/24/2022]
|
25
|
Hummer AA, Heffeter P, Berger W, Filipits M, Batchelor D, Büchel GE, Jakupec MA, Keppler BK, Rompel A. X-ray absorption near edge structure spectroscopy to resolve the in vivo chemistry of the redox-active indazolium trans-[Tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019). J Med Chem 2013; 56:1182-96. [PMID: 23282017 PMCID: PMC3579476 DOI: 10.1021/jm301648f] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
Indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (1, KP1019) and
its analogue
sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]
(2, KP1339) are promising redox-active anticancer drug
candidates that were investigated with X-ray absorption near edge
structure spectroscopy. The analysis was based on the concept of the
coordination charge and ruthenium model compounds representing possible
coordinations and oxidation states in vivo. 1 was investigated
in citrate saline buffer (pH 3.5) and in carbonate buffer (pH 7.4)
at 37 °C for different time intervals. Interaction studies on 1 with glutathione in saline buffer and apo-transferrin in
carbonate buffer were undertaken, and the coordination of 1 and 2 in tumor tissues was studied too. The most likely
coordinations and oxidation states of the compound under the above
mentioned conditions were assigned. Microprobe X-ray fluorescence
of tumor thin sections showed the strong penetration of ruthenium
into the tumor tissue, with the highest concentrations near blood
vessels and in the edge regions of the tissue samples.
Collapse
Affiliation(s)
- Alfred A Hummer
- Institut für Biophysikalische Chemie, Universität Wien, Althanstrasse 14, 1090 Wien, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Morais TS, Santos F, Côrte-Real L, Marques F, Robalo MP, Madeira PJA, Garcia MH. Biological activity and cellular uptake of [Ru(η5-C5H5)(PPh3)(Me2bpy)][CF3SO3] complex. J Inorg Biochem 2013; 122:8-17. [PMID: 23416310 DOI: 10.1016/j.jinorgbio.2013.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 01/08/2023]
Abstract
Anticancer activity of the new [Ru(η(5)-C5H5)(PPh3)(Me2bpy)][CF3SO3] (Me2bpy = 4,4'-dimethyl-2,2'-bipyridine) complex was evaluated in vitro against several human cancer cell lines, namely A2780, A2780CisR, HT29, MCF7, MDAMB231 and PC3. Remarkably, the IC50 values, placed in the nanomolar and sub-micromolar range, largely exceeded the activity of cisplatin. Binding to human serum albumin, either HSA (human serum albumin) or HSA(faf) (fatty acid-free human serum albumin) does not affect the complex activity. Fluorescence studies revealed that the present ruthenium complex strongly quench the intrinsic fluorescence of albumin. Cell death by the [Ru(η(5)-C5H5)(PPh3)(Me2bpy)][CF3SO3] complex was reduced in the presence of endocytosis modulators and at low temperature, suggesting an energy-dependent mechanism consistent with endocytosis. On the whole, the biological activity evaluated herein suggests that the complex could be a promising anticancer agent.
Collapse
Affiliation(s)
- Tânia S Morais
- Centro de Ciências Moleculares e Materiais, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | | | | | | | | | | | | |
Collapse
|
27
|
Mijatović A, Šmit B, Rilak A, Petrović B, Čanović D, Bugarčić ŽD. NMR kinetic studies of the interactions between [Ru(terpy)(bipy)(H2O)]2+ and some sulfur-donor ligands. Inorganica Chim Acta 2013. [DOI: 10.1016/j.ica.2012.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Schmitt SM, Frezza M, Dou QP. New applications of old metal-binding drugs in the treatment of human cancer. Front Biosci (Schol Ed) 2012; 4:375-91. [PMID: 22202066 DOI: 10.2741/274] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significant advances in the use of metal complexes, precipitated by platinum, have fostered a renewed interest in harnessing their rich potential in the treatment of cancer. In addition to platinum-based complexes, the anticancer properties of other metals such as ruthenium have been realized, and ruthenium-based compounds are currently being investigated in clinical trials. Since the process of drug development can be expensive and cumbersome, finding new applications of existing drugs may provide effective means to expedite the regulatory process in bringing new drugs to the clinical setting. Encouraging findings from laboratory studies reveal significant anticancer activity from different classes of metal-chelating compounds, such as disulfiram, clioquinol, and dithiocarbamate derivatives that are currently approved for the treatment of various pathological disorders. Their use as coordination complexes with metals such as copper, zinc, and gold that target the ubiquitin-proteasome pathway have shown significant promise as potential anticancer agents. This review discusses the unique role of several selected metals in relation to their anti-cancer properties as well as the new therapeutic potential of several previously approved metal-chelating drugs. In vitro and in vivo experimental evidence along with mechanisms of action (e.g., via targeting the tumor proteasome) will also be discussed with anticipation of strengthening this exciting new concept.
Collapse
Affiliation(s)
- Sara M Schmitt
- Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
29
|
Webb MI, Chard RA, Al-Jobory YM, Jones MR, Wong EWY, Walsby CJ. Pyridine analogues of the antimetastatic Ru(III) complex NAMI-A targeting non-covalent interactions with albumin. Inorg Chem 2011; 51:954-66. [PMID: 22224431 DOI: 10.1021/ic202029e] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of pyridine-based derivatives of the antimetastatic Ru(III) complex imidazolium [trans-RuCl(4)(1H-imidazole)(DMSO-S)] (NAMI-A) have been synthesized along with their sodium-ion compensated analogues. These compounds have been characterized by X-ray crystallography, electron paramagnetic resonance (EPR), NMR, and electrochemistry, with the goal of probing their noncovalent interactions with human serum albumin (hsA). EPR studies show that the choice of imidazolium ligands and compensating ions does not strongly influence the rates of ligand exchange processes in aqueous buffer solutions. By contrast, the rate of formation and persistence of interactions of the complexes with hsA is found to be strongly dependent on the properties of the axial ligands. The stability of noncovalent binding is shown to correlate with the anticipated ability of the various pyridine ligands to interact with the hydrophobic binding domains of hsA. These interactions prevent the oligomerization of the complexes in solution and limit the rate of covalent binding to albumin amino acid side chains. Electrochemical studies demonstrate relatively high reduction potentials for these complexes, leading to the formation of Ru(II) species in aqueous solutions containing biological reducing agents, such as ascorbate. However, EPR measurements indicate that while noncovalent interactions with hsA do not prevent reduction, covalent binding produces persistent mononuclear Ru(III) species under these conditions.
Collapse
Affiliation(s)
- Michael I Webb
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | | | | | | | | | | |
Collapse
|
30
|
Yata PK, Shilpa M, Nagababu P, Reddy MR, Kotha LR, Gabra NM, Satyanarayana S. Study of DNA Light Switch Ru(II) Complexes : Synthesis, Characterization, Photocleavage and Antimicrobial Activity. J Fluoresc 2011; 22:835-47. [DOI: 10.1007/s10895-011-1018-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/21/2011] [Indexed: 11/29/2022]
|
31
|
DNA- and RNA-binding and enhanced DNA-photocleavage properties of a ferrocenyl-containing ruthenium(II) complex. J Inorg Biochem 2011; 107:104-10. [PMID: 22178672 DOI: 10.1016/j.jinorgbio.2011.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 09/12/2011] [Accepted: 10/17/2011] [Indexed: 11/24/2022]
Abstract
The interaction of [Ru(bpy)(2)(fip)](PF(6))(2) {bpy=2,2'-bipyridine, fip=2-ferrocenyl-1H-imidazo[4,5-f][1,10]-phenanthroline} with calf thymus DNA and yeast tRNA was investigated comparatively by UV-visible absorption and luminescence spectrophotometric titrations, steady-state emission quenching by [Fe(CN)(6)](4-), ethidium bromide competition experiment, DNA thermal denaturation, viscosity measurements and salt effect studies. The results suggest that the complex binds to the DNA more strongly than to the RNA. The density functional theory calculations were also carried out in order to better understand the nucleic acid binding properties. Agarose gel electrophoresis showed that the complex exhibited enhanced DNA-photocleavage capacity on pUC 18 plasmid DNA under irradiation at 360 nm as compared with a ferrocenyl-free analogous complex.
Collapse
|
32
|
Beckford F, Thessing J, Woods J, Didion J, Gerasimchuk N, Gonzalez-Sarrias A, Seeram NP. Synthesis and structure of [(η(6)-p-cymene)Ru(2-anthracen-9-ylmethylene-N-ethylhydrazinecarbothioamide)Cl]Cl; biological evaluation, topoisomerase II inhibition and reaction with DNA and human serum albumin. Metallomics 2011; 3:491-502. [PMID: 21347491 PMCID: PMC3271380 DOI: 10.1039/c1mt00003a] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have synthesized and evaluated the biological properties of a compound of the type [η(6)-p-cymene)Ru(EtATSC)Cl]Cl (1) where EtATSC = 2-anthracen-9-ylmethylene-N-ethylhydrazinecarbothioamide, a thiosemicarbazone. The complex has been characterized by elemental analysis, spectroscopically (NMR, UV-Vis, and IR) and structurally by XRD. The in vitro anticancer activity of 1 has been evaluated against two human colon cancer cell lines. The IC(50) value for activity against HCT-116 was 224 ± 7 μM and 205 ± 5 μM against the Caco-2 cell line. The proficiency of 1 as an antibacterial agent was also evaluated against six bacterial strains. The minimum inhibitory concentration for Bacillus cereus was determined to be 5 μM and for Enterococcus faecalis it was 20 μM. At the maximum concentration tested the complex showed no activity against the Gram-negative strains. The complex binds strongly to human serum albumin with a binding constant of 1.37 ± 0.02 M(-1) at 308 K on a single binding site. It is also a strong binder to DNA with an apparent binding constant of 2.82 × 10(5) M(-1) at 308 K. 1 shows very good activity as a catalytic inhibitor of human topoisomerase II at concentrations as low as 20 μM.
Collapse
Affiliation(s)
- Floyd Beckford
- Science Division, Lyon College, Batesville, AR 72501, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
In vitro and in vivo biological activity screening of Ru(III) complexes involving 6-benzylaminopurine derivatives with higher pro-apoptotic activity than NAMI-A. J Inorg Biochem 2011; 105:937-48. [PMID: 21536006 DOI: 10.1016/j.jinorgbio.2011.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 11/21/2022]
Abstract
A series of novel octahedral ruthenium(III) complexes involving 6-benzylaminopurine (L) derivatives as N-donor ligands has been prepared by the reaction of [(DMSO)(2)H][trans-RuCl(4)(DMSO)(2)] with the corresponding L derivative. The complexes 1-12 have the general compositions trans-[RuCl(4)(DMSO)(n-Cl-LH)]⋅xSol (1-3), trans-[RuCl(4)(DMSO)(n-Br-LH)]·xSol (4-6), trans-[RuCl(4)(DMSO)(n-OMe-LH)]·xSol (7-9) and trans-[RuCl(4)(DMSO)(n-OH-LH)]·xSol (10-12); n=2, 3, and 4, x=0-1.5; and Sol = H(2)O, DMSO, EtOH and/or (Me)(2)CO. The complexes have been thoroughly characterized by elemental analysis, UV-visible, FTIR, Raman, and EPR spectroscopy, ES+(positive ionization electrospray) mass spectrometry, thermal analysis, cyclic voltammetry, magnetic and conductivity measurements. The X-ray molecular structure of trans-[RuCl(4)(DMSO)(3-Br-LH)]⋅(Me)(2)CO (5) revealed the distorted octahedral coordination in the vicinity of the central atom, and also confirmed that the 3-Br-L ligand is present as the N3-protonated N7-H tautomer and is coordinated to Ru(III) through the N9 atom of the purine moiety. The tested complexes have been found to be in vitro non-cytotoxic against K562, G361, HOS and MCF7 human cancer cell lines with IC(50)>100μM in contrast to the moderate results regarding the antiradical activity with IC(50)≈10(-3)M. On the contrary, in vivo antitumor activity screening showed that the prepared Ru(III) complexes possess higher pro-apoptotic activity than NAMI-A. The reduction of Ru(III) to Ru(II) and Ru(II)-species formation in tumor tissues was confirmed by means of a simple method of detection and visualization of intracellular Ru(II) by fluorescence microscopy. The originality of this method is based on the preparation of a Ru(II)-bipyridine complex in situ.
Collapse
|
34
|
Moreno V, Font-Bardia M, Calvet T, Lorenzo J, Avilés FX, Garcia MH, Morais TS, Valente A, Robalo MP. DNA interaction and cytotoxicity studies of new ruthenium(II) cyclopentadienyl derivative complexes containing heteroaromatic ligands. J Inorg Biochem 2010; 105:241-9. [PMID: 21194624 DOI: 10.1016/j.jinorgbio.2010.10.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 11/27/2022]
Abstract
Four ruthenium(II) complexes with the formula [Ru(η(5)-C(5)H(5))(PP)L][CF(3)SO(3)], being (PP = two triphenylphosphine molecules), L = 1-benzylimidazole, ; (PP = two triphenylphosphine molecules), L = 2,2'bipyridine, ; (PP = two triphenylphosphine molecules), L = 4-Methylpyridine, ; (PP = 1,2-bis(diphenylphosphine)ethane), L = 4-Methylpyridine, , were prepared, in view to evaluate their potentialities as antitumor agents. The compounds were completely characterized by NMR spectroscopy and their crystal and molecular structures were determined by X-ray diffraction. Electrochemical studies were carried out giving for all the compounds quasi-reversible processes. The images obtained by atomic force microscopy (AFM) suggest interaction with pBR322 plasmid DNA. Measurements of the viscosity of solutions of free DNA and DNA incubated with different concentrations of the compounds confirmed this interaction. The cytotoxicity of compounds 1234 was much higher than that of cisplatin against human leukemia cancer cells (HL-60 cells). IC(50) values for all the compounds are in the range of submicromolar amounts. Apoptotic death percentage was also studied resulting similar than that of cisplatin.
Collapse
Affiliation(s)
- Virtudes Moreno
- Department de Química Inorgànica, Universitat de Barcelona, Martí y Franquès 1-11, 08028, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Silveira-Lacerda EDP, Vilanova-Costa CAST, Hamaguchi A, Pavanin LA, Goulart LR, Homsi-Brandenburgo MI, Dos Santos WB, Soares AM, Nomizo A. The ruthenium complex cis-(dichloro)tetraammineruthenium(III) chloride presents selective cytotoxicity against murine B cell lymphoma (A-20), murine ascitic sarcoma 180 (S-180), human breast adenocarcinoma (SK-BR-3), and human T cell leukemia (Jurkat) tumor cell lines. Biol Trace Elem Res 2010; 135:98-111. [PMID: 19727575 DOI: 10.1007/s12011-009-8498-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 08/06/2009] [Indexed: 11/30/2022]
Abstract
The aim of present study was to verify the in vitro antitumor activity of a ruthenium complex, cis-(dichloro)tetraammineruthenium(III) chloride (cis-[RuCl(2)(NH(3))(4)]Cl) toward different tumor cell lines. The antitumor studies showed that ruthenium(III) complex presents a relevant cytotoxic activity against murine B cell lymphoma (A-20), murine ascitic sarcoma 180 (S-180), human breast adenocarcinoma (SK-BR-3), and human T cell leukemia (Jurkat) cell lines and a very low cytotoxicity toward human peripheral blood mononuclear cells. The ruthenium(III) complex decreased the fraction of tumor cells in G0/G1 and/or G2-M phases, indicating that this compound may act on resting/early entering G0/G1 cells and/or precycling G2-M cells. The cytotoxic activity of a high concentration (2 mg mL(-1)) of cis-[RuCl(2)(NH(3))(4)]Cl toward Jurkat cells correlated with an increased number of annexin V-positive cells and also the presence of DNA fragmentation, suggesting that this compound induces apoptosis in tumor cells. The development of new antineoplastic medications demands adequate knowledge in order to avoid inefficient or toxic treatments. Thus, a mechanistic understanding of how metal complexes achieve their activities is crucial to their clinical success and to the rational design of new compounds with improved potency.
Collapse
Affiliation(s)
- Elisângela de Paula Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas-ICB I-Sala 200, Universidade Federal de Goiás-UFG, Campus Samambaia Campus II, Goiânia, Goiás, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Antonarakis ES, Emadi A. Ruthenium-based chemotherapeutics: are they ready for prime time? Cancer Chemother Pharmacol 2010; 66:1-9. [PMID: 20213076 DOI: 10.1007/s00280-010-1293-1] [Citation(s) in RCA: 367] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 02/12/2010] [Indexed: 11/28/2022]
Abstract
Since the discovery of cis-platinum, many transition metal complexes have been synthesized and assayed for antineoplastic activity. In recent years, ruthenium-based molecules have emerged as promising antitumor and antimetastatic agents with potential uses in platinum-resistant tumors or as alternatives to platinum. Ruthenium compounds theoretically possess unique biochemical features allowing them to accumulate preferentially in neoplastic tissues and to convert to their active state only after entering tumor cells. Intriguingly, some ruthenium agents show significant activity against cancer metastases but have minimal effects on primary tumors. Two ruthenium-based drugs, NAMI-A and KP1019, have reached human clinical testing. This review will highlight the chemical properties, mechanism of action, preclinical data, and early phase clinical results of these two lead ruthenium compounds. Other promising ruthenium agents will also be reviewed with emphasis on the novel ruthenium compound ONCO4417, and DW1/2 that has demonstrated Pim-1 kinase inhibition in preclinical systems. Further development of these and other ruthenium agents may rely on novel approaches including rational combination strategies as well as identification of potential pharmacodynamic biomarkers of drug activity aiding early phase clinical studies.
Collapse
Affiliation(s)
- Emmanuel S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street, CRB1-191, Baltimore, MD 21231, USA
| | | |
Collapse
|
37
|
Silveira-Lacerda EDP, Vilanova-Costa CAST, Pereira FDC, Hamaguchi A, Pavanin LA, Goulart LR, Homsi-Brandenburgo MI, Soares AM, dos Santos WB, Nomizo A. The ruthenium complex cis-(Dichloro)Tetraammineruthenium(III) chloride presents immune stimulatory activity on human peripheral blood mononuclear cells. Biol Trace Elem Res 2010; 133:270-83. [PMID: 19590831 DOI: 10.1007/s12011-009-8440-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 06/22/2009] [Indexed: 11/28/2022]
Abstract
Ruthenium compounds in general are well suited for medicinal applications. They have been investigated as immunosuppressants, nitric oxide scavengers, antimicrobial agents, and antimalarials. The aim of this study is to evaluate the immunomodulatory activity of cis-(dichloro)tetraammineruthenium(III) chloride (cis-[RuCl(2)(NH(3))(4)]Cl) on human peripheral blood mononuclear cells (PBMC). The cytotoxic studies performed here revealed that the ruthenium(III) complex presents a cytotoxic activity towards normal human PBMC, only at very high concentration. Results also showed that cis-[RuCl(2)(NH(3))(4)]Cl presents a dual role on PBMC stimulating proliferation and interleukin-2 (IL-2) production at low concentration and inducing cytotoxicity, inability to proliferate, and inhibiting IL-2 production at high concentration. The noncytotoxic activity of cis-[RuCl(2)(NH(3))(4)]Cl at low concentration towards PBMC, which correlates with the small number of annexin V positive cells and also the absence of DNA fragmentation, suggest that this compound does not induce apoptosis on PBMC. For the first time, we show that, at low concentration (10-100 microg L(-1)), the cis-[RuCl(2)(NH(3))(4)]Cl compound induces peripheral blood lymphocytes proliferation and also stimulates them to IL-2 production. These results open a new potential applicability of ruthenium(III) complexes as a possible immune regulatory compound acting as immune suppressor at high concentration and as immune stimulator at low concentration.
Collapse
Affiliation(s)
- Elisângela de Paula Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas (ICB I), Universidade Federal de Goiás, Sala 200, Campus Samambaia (Campus II), Cx. Postal: 131, Goiânia, Goiás, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Structural characterization and cytotoxicity studies of ruthenium(II)–dmso–chloro complexes of chalcone and flavone derivatives. Polyhedron 2010. [DOI: 10.1016/j.poly.2009.11.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Reaction of the Anticancer Organometallic Ruthenium Compound, [(eta-p-Cymene)Ru(ATSC)Cl]PF(6) with Human Serum Albumin. ACTA ACUST UNITED AC 2010; 2010:1-7. [PMID: 20671814 DOI: 10.1155/2010/975756] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The reaction of [(eta(6)-p-cymene)Ru(ATSC)Cl]PF(6) (ATSC = 9-anthraldehyde thiosemicarbazone) with human serum albumin was investigated at different temperatures using fluorescence and infrared spectrophotometry. The binding constant, K, for the reaction was determined using a number of different methods. Using a modified Stern-Volmer equation, K was determined to be 9.09 x 10(4), 12.1 x 10(4), and 13.1 x 10(4) M(-1) at 293 K, 298 K, and 308 K, respectively. A thermodynamic analysis showed that the reaction is spontaneous with DeltaG being negative. The enthalpy of reaction DeltaH = 16.5kJ mol(-1) and the entropy of reaction DeltaS = 152 Jmol(-1)K(-1). The values of DeltaH and DeltaS suggest that hydrophobic forces are dominant in the mode of interaction and that the process is mostly entropy driven.
Collapse
|
40
|
Beckford FA, Shaloski M, Leblanc G, Thessing J, Lewis-Alleyne LC, Holder AA, Li L, Seeram NP. Microwave synthesis of mixed ligand diimine-thiosemicarbazone complexes of ruthenium(II): biophysical reactivity and cytotoxicity. Dalton Trans 2009:10757-64. [PMID: 20023905 DOI: 10.1039/b915081a] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel microwave-assisted synthetic method has been used to synthesise a series of mixed ligand ruthenium(II) compounds containing diimine as well as bidentate thiosemicarbazone ligands. The compounds contain the diimine 1,10-phenanthroline (phen) or 2,2'-bipyridine (bpy) and the thiosemicarbazone is derived from 9-anthraldehyde. Based on elemental analyses and spectroscopic data, the compounds are best formulated as [(phen)(2)Ru(thiosemicarbazone)](PF(6))(2) and [(phen)(2)Ru(thiosemicarbazone)](PF(6))(2) where thiosemicarbazone = 9-anthraldehydethiosemicarbazone, 9-anthraldehyde-N(4)-methylthiosemicarbazone, and 9-anthraldehyde-N(4)-ethylthiosemicarbazone. Fluorescence competition studies with ethidium bromide, along with viscometric measurements suggests that the complexes bind calf thymus DNA (CTDNA) relatively strongly via an intercalative mode possibly involving the aromatic rings of the diimine ligands. The complexes show good cytotoxic profiles against MCF-7 and MDA-MB-231 (breast adenocarcinoma) as well as HCT 116 and HT-29 (colorectal carcinoma) cell lines.
Collapse
|
41
|
Corral E, Hotze ACG, den Dulk H, Leczkowska A, Rodger A, Hannon MJ, Reedijk J. Ruthenium polypyridyl complexes and their modes of interaction with DNA: is there a correlation between these interactions and the antitumor activity of the compounds? J Biol Inorg Chem 2008; 14:439-48. [PMID: 19085018 PMCID: PMC3036821 DOI: 10.1007/s00775-008-0460-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 12/02/2008] [Indexed: 11/04/2022]
Abstract
Various interaction modes between a group of six ruthenium polypyridyl complexes and DNA have been studied using a number of spectroscopic techniques. Five mononuclear species were selected with formula [Ru(tpy)L1L2](2−n)+, and one closely related dinuclear cation of formula [{Ru(apy)(tpy)}2{μ-H2N(CH2)6NH2}]4+. The ligand tpy is 2,2′:6′,2″-terpyridine and the ligand L1 is a bidentate ligand, namely, apy (2,2′-azobispyridine), 2-phenylazopyridine, or 2-phenylpyridinylmethylene amine. The ligand L2 is a labile monodentate ligand, being Cl−, H2O, or CH3CN. All six species containing a labile L2 were found to be able to coordinate to the DNA model base 9-ethylguanine by 1H NMR and mass spectrometry. The dinuclear cationic species, which has no positions available for coordination to a DNA base, was studied for comparison purposes. The interactions between a selection of four representative complexes and calf-thymus DNA were studied by circular and linear dichroism. To explore a possible relation between DNA-binding ability and toxicity, all compounds were screened for anticancer activity in a variety of cancer cell lines, showing in some cases an activity which is comparable to that of cisplatin. Comparison of the details of the compound structures, their DNA binding, and their toxicity allows the exploration of structure–activity relationships that might be used to guide optimization of the activity of agents of this class of compounds.
Collapse
Affiliation(s)
- Eva Corral
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
42
|
Koiri RK, Trigun SK, Mishra L, Pandey K, Dixit D, Dubey SK. Regression of Dalton’s lymphoma in vivo via decline in lactate dehydrogenase and induction of apoptosis by a ruthenium(II)-complex containing 4-carboxy N-ethylbenzamide as ligand. Invest New Drugs 2008; 27:503-16. [DOI: 10.1007/s10637-008-9202-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 11/12/2008] [Indexed: 12/18/2022]
|
43
|
Garza-Ortiz A, Maheswari PU, Siegler M, Spek AL, Reedijk J. Ruthenium(III) Chloride Complex with a Tridentate Bis(arylimino)pyridine Ligand: Synthesis, Spectra, X-ray Structure, 9-Ethylguanine Binding Pattern, and In Vitro Cytotoxicity. Inorg Chem 2008; 47:6964-73. [DOI: 10.1021/ic8005579] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ariadna Garza-Ortiz
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Bijvoet Center for Biomolecular Research, Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Palanisamy Uma Maheswari
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Bijvoet Center for Biomolecular Research, Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Maxime Siegler
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Bijvoet Center for Biomolecular Research, Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Anthony L. Spek
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Bijvoet Center for Biomolecular Research, Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Jan Reedijk
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Bijvoet Center for Biomolecular Research, Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
44
|
|
45
|
Victor EG, Zanette F, Aguiar MR, Aguiar CS, Cardoso DC, Cristiano MP, Streck EL, Paula MMS. Effect of ruthenium complexes on the activities of succinate dehydrogenase and cytochrome oxidase. Chem Biol Interact 2007; 170:59-66. [PMID: 17707358 DOI: 10.1016/j.cbi.2007.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 07/04/2007] [Accepted: 07/05/2007] [Indexed: 11/17/2022]
Abstract
In this article, we report the effects of acute administration of ruthenium complexes, trans-[RuCl(2)(nic)(4)] (nic=3-pyridinecarboxylic acid) 180.7 micromol/kg (complex I), trans-[RuCl(2)(i-nic)(4)] (i-nic=4-pyridinecarboxylic acid) 13.6 micromol/kg (complex II), trans-[RuCl(2)(dinic)(4)] (dinic=3,5-pyridinedicarboxylic acid) 180.7 micromol/kg (complex III) and trans-[RuCl(2)(i-dinic)(4)]Cl (i-dinic=3,4-pyridinedicarboxylic acid) 180.7 micromol/kg (complex IV) on succinate dehydrogenase (SDH) and cytochrome oxidase (COX) activities in brain (hippocampus, striatum and cerebral cortex), heart, skeletal muscle, liver and kidney of rats. Our results showed that complex I inhibited SDH activity in hippocampus, cerebral cortex, heart and liver; and inhibited COX in heart and kidney. Complex II inhibited SDH in heart and hippocampus; COX was inhibited in hippocampus, heart, liver and kidney. SDH activity was inhibited by complex III in heart, muscle, liver and kidney. However, COX activity was increased in hippocampus, striatum, cerebral cortex and kidney. Complex IV inhibited SDH activity in muscle and liver; COX activity was inhibited in kidney and increased in hippocampus, striatum and cerebral cortex. In a general manner, the complexes tested in this work decrease the activities of SDH and COX in heart, skeletal muscle, liver and kidney. In brain, complexes I and II were shown to be inhibitors and complexes III and IV activators of these enzymes. In vitro studies showed that the ruthenium complexes III and IV did not alter COX activity in kidney, but activated the enzyme in hippocampus, striatum and cerebral cortex, suggesting that these complexes present a direct action on COX in brain.
Collapse
Affiliation(s)
- Eduardo G Victor
- Laboratório de Síntese de Complexos Multifuncionais, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Metastases are more decisive for tumour prognosis than primary lesions, because of their multiple locations, low accessibility to surgery and/or radiotherapy, and generally poor responsiveness to chemotherapy. The metastasis should therefore be the primary target for drug therapy. Among ruthenium complexes, NAMI-A is a leading compound that shows selective effects for solid tumour metastases related to a mechanism of action involving the inhibition of the processes of tumour invasiveness. NAMI-A opens an avenue to new perspectives in cancer chemotherapy. This includes novel compounds directed at targets selectively expressed by tumour metastases, thus reducing the typical side effects of the current metal-based drugs that are active via their unselective DNA interaction.
Collapse
Affiliation(s)
- A Bergamo
- Callerio Foundation Onlus, via A Fleming 22-31, Trieste, Italy
| | | |
Collapse
|
47
|
Corral E, Hotze AC, Tooke DM, Spek AL, Reedijk J. Ruthenium polypyridyl complexes containing the bischelating ligand 2,2′-azobispyridine. Synthesis, characterization and crystal structures. Inorganica Chim Acta 2006. [DOI: 10.1016/j.ica.2005.05.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Reisner E, Arion VB, Rufińska A, Chiorescu I, Schmid WF, Keppler BK. Isomeric [RuCl2(dmso)2(indazole)2] complexes: ruthenium(ii)-mediated coupling reaction of acetonitrile with 1H-indazole. Dalton Trans 2005:2355-64. [PMID: 15995743 DOI: 10.1039/b503650j] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reaction of the antitumor complex trans-[Ru(III)Cl4(Hind)2]- (Hind = indazole) with an excess of dimethyl sulfoxide (dmso) in acetone afforded the complex trans,trans,trans-[Ru(II)Cl2(dmso)2(Hind)2] (1). Two other isomeric compounds trans,cis,cis-[Ru(II)Cl2(dmso)2(Hind)2] (2) and cis,cis,cis-[Ru(II)Cl2(dmso)2(Hind)2] (3) have been obtained on refluxing cis-[Ru(II)Cl(2)(dmso)(4)] with 2 equiv. of indazole in ethanol and methanol, respectively. Isomers 1 and 2 react with acetonitrile yielding the complexes trans-[Ru(II)Cl2(dmso)(Hind){HN=C(Me)ind}].CH3CN (4.CH3CN) and trans,cis-[Ru(II)Cl2(dmso)2{HN=C(Me)ind}].H2O (5.H2O), respectively, containing a cyclic amidine ligand resulting from insertion of the acetonitrile C triple bond N group in the N1-H bond of the N2-coordinated indazole ligand in the nomenclature used for 1H-indazole. These are the first examples of the metal-assisted iminoacylation of indazole. The products isolated have been characterized by elemental analysis, IR spectroscopy, UV-vis spectroscopy, electrospray mass-spectrometry, thermogravimetry, differential scanning calorimetry, 1H NMR spectroscopy, and solid-state 13C CP MAS NMR spectroscopy. The isomeric structures of 1-3 and the presence of a chelating amidine ligand in 4 and 5 have been confirmed by X-ray crystallography. The electrochemical behavior of 1-5 and the formation of 5 have been studied by cyclic voltammetry.
Collapse
Affiliation(s)
- Erwin Reisner
- Institute of Inorganic Chemistry-Bioinorganic, Environmental and Radiochemistry, Faculty of Chemistry, University of Vienna, A-1090 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
49
|
Srivastava RS, Fronczek FR, Romero LM. Formation and molecular structure of novel Ru(III) dimers of a symmetric antitumor drug analogue. Inorganica Chim Acta 2004. [DOI: 10.1016/j.ica.2004.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
50
|
Pluim D, van Waardenburg RCAM, Beijnen JH, Schellens JHM. Cytotoxicity of the organic ruthenium anticancer drug Nami-A is correlated with DNA binding in four different human tumor cell lines. Cancer Chemother Pharmacol 2004; 54:71-8. [PMID: 15034754 DOI: 10.1007/s00280-004-0773-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2003] [Accepted: 01/19/2004] [Indexed: 10/26/2022]
Abstract
PURPOSE The cytotoxicity, intracellular accumulation and DNA adduct formation of the ruthenium complex imidazolium trans-imidazoledimethylsulfoxide tetrachlororuthenate (ImH[ trans-RuCl(4)(DMSO)Im], Nami-A) were compared in vitro with those of cisplatin in four human tumor cell lines: Igrov-1, 2008, MCF-7, and T47D. METHODS Cytotoxicity was assessed in vitro using a growth inhibition assay. Accumulation was determined by flameless atomic absorption spectroscopy (AAS). GG and AG intrastrand adducts were measured using the (32)P-postlabeling assay. RESULTS Nami-A was on average 1053 times less cytotoxic than cisplatin. The cytotoxicity of cisplatin was linearly related to both intracellular platinum accumulation and DNA binding, while the cytotoxicity of Nami-A was significantly related only to DNA binding and not to intracellular ruthenium accumulation. The levels of accumulation of Nami-A measured as ruthenium and of cisplatin measured as platinum were correlated linearly with the incubation concentration over a concentration range of 0 to 600 micro M of both drugs. Ruthenium intracellular accumulation and DNA binding were on average 4.8 and 42 times less, respectively, than those of cisplatin. In addition, the numbers of GG and AG intrastrand adducts induced by Nami-A were 418 and 51 times fewer, respectively. Nami-A and cisplatin had the same binding capacity to calf thymus DNA. Nami-A was 25-40% less bound to cellular proteins than cisplatin. CONCLUSIONS There was no saturation of the uptake and DNA binding capacity of either Nami-A or cisplatin. Furthermore, the low binding of Nami-A to cellular DNA cannot simply be explained by a lower capacity to bind to DNA, because the absolute level of binding in vitro to calf thymus DNA was the same for Nami-A and cisplatin. Finally, the lower cytotoxicity of Nami-A on a molar basis than that of cisplatin can at least partly be explained by its reduced reactivity to DNA in intact cells.
Collapse
Affiliation(s)
- Dick Pluim
- Division of Experimental Therapy and Medical Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | | | | | | |
Collapse
|