Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Toxicol Appl Pharmacol ; 434: 115822, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34896434

ABSTRACT

Our recent investigation directed to synthesize a novel ruthenium-phloretin complex accompanied by the study of antioxidant in addition to DNA binding capabilities, to determine the chemotherapeutic activity against breast carcinoma in vitro and in vivo. Ruthenium-phloretin complex was synthesized and characterized by different spectroscopic methods. The complex was further investigated to determine its efficacy in both MCF-7 and MDA-MB-231 human carcinoma cell lines and finally in an in vivo model of mammary carcinogenesis induced by DMBA in rats. Our studies confirm that the chelation of the metal and ligand was materialize by the 3-OH and 9-OH functional groups of the ligand and the complex is found crystalline and was capable of intercalating with CT-DNA. The complex was capable of reducing cellular propagation and initiate apoptotic events in MCF-7 and MDA-MB-231 breast carcinoma cell lines. Ruthenium-phloretin complex could modulate p53 intervene apoptosis in the breast carcinoma, initiated by the trail of intrinsic apoptosis facilitated through Bcl2 and Bax and at the same time down regulating the PI3K/Akt/mTOR pathway coupled with MMP9 regulated tumor invasive pathways. Ruthenium-phloretin chemotherapy could interrupt, revoke or suspend the succession of breast carcinoma by altering intrinsic apoptosis along with the anti-angiogenic pathway.


Subject(s)
Breast Neoplasms/drug therapy , Malus/chemistry , Phloretin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Ruthenium Compounds/pharmacology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/drug therapy , Mice , Neoplasms, Experimental , Phloretin/chemistry , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Ruthenium Compounds/chemistry , Ruthenium Compounds/toxicity , TOR Serine-Threonine Kinases , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Toxicol Appl Pharmacol ; 426: 115618, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34126112

ABSTRACT

WHO suggests that colon cancer incidences are rising steadily, propelling researchers to search for novel chemotherapeutic options. Metal-based chemotherapy is a potential forte to explore ruthenium-based complexes, exhibiting the capability to influence a variety of cellular targets. We discovered the chemotherapeutic effects of ruthenium-rifampicin complex on HT-29 and HCT-116 human colorectal cell lines and on a chemically developed murine colorectal cancer model. Complex was synthesized and characterized by analytical techniques and evaluation of antioxidant potential along with DNA binding capabilities. The complex minimizes cellular propagation and initiates apoptotic events in the colon cancer cell lines of HT-29 and HCT-116. The results of the in vivo study suggest that the complex has been successful in minimizing the wide spectrum of aberrant crypt foci and hyperplastic lesions, as well as encouraging elevated amounts of CAT, SOD and glutathione. Along with that, p53 could be modulated by the ruthenium-rifampicin complex to interfere with apoptosis in colon carcinoma, initiated by the intrinsic apoptotic trail facilitated through Bcl2 and Bax, thus controlling the Akt/mTOR/VEGF pathway coupled through the WNT/ß-catenin trail. Ruthenium-rifampicin chemotherapy could interrupt, retract or interrupt the progression of colorectal cancer through modifying intrinsic apoptosis including the antiangiogenic pathway, thereby achieving the function of a potential contender in chemotherapy in the near future.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Rifampin/therapeutic use , Ruthenium Compounds/therapeutic use , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Colon/drug effects , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Combinations , Female , HCT116 Cells , HT29 Cells , Humans , Male , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Rifampin/pharmacology , Rifampin/toxicity , Ruthenium Compounds/pharmacology , Ruthenium Compounds/toxicity , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment/drug effects , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Regul Toxicol Pharmacol ; 80: 233-40, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27375008

ABSTRACT

Recently, a series of thiazolo arene ruthenium complexes were found to be highly cytotoxic in vitro, on both cisplatin-sensitive and cisplatin-resistant ovarian cancer cells. The most active compound of the series, [(η(6)-p-cymene)Ru(L)Cl]Cl (L = 1-(2-(2-(3-chlorobenzylidene)hydrazinyl)-4-methylthiazol-5-yl)ethanone), was selected for an in vivo study in order to assess its safety profile. The ruthenium complex was administered to female Crl:WI rats orally, by gastric intubation and intraperitoneal injection. The hematological parameters and the histopathological changes in liver, kidneys, spleen and brain were investigated after a 14-days treatment. The substance was very well tolerated orally, with a LD50 value of over 2000 mg/kg body weight. Symptoms were observed only in the first day after intraperitoneal administration of the highest dose, with a LD50 value between 300 and 2000 mg/kg bw. The hematological profile was not modified at any of the tested doses, after both oral and intraperitoneal acute administration. Structural modifications (moderate lymphocytolysis) were identified only in the spleen at the highest tested dose. In conclusion, the thiazolo arene ruthenium complex was very well tolerated orally and had a low acute toxicity after intraperitoneal administration in Crl:WI rats The results justify further investigation to determine the in vivo therapeutic potential of this promising ruthenium complex.


Subject(s)
Antineoplastic Agents/toxicity , Organometallic Compounds/toxicity , Ruthenium Compounds/toxicity , Toxicity Tests, Acute/methods , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Biomarkers/blood , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Injections, Intraperitoneal , Lethal Dose 50 , Models, Animal , Organometallic Compounds/administration & dosage , Rats, Wistar , Risk Assessment , Ruthenium Compounds/administration & dosage , Spleen/drug effects , Spleen/pathology , Time Factors
4.
Mycoses ; 54(5): e323-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20557461

ABSTRACT

The in vitro antifungal activity of ruthenium dithiocarbamate compounds (1-5) was investigated and assessed for its activity against seven different species of Aspergillus (Aspergillus clavatus, Aspergillus flavus, Aspergillus fumigatus, Aspergillus niger, Aspergillus nomius, Aspergillus tamarii and Aspergillus terreus). Analysis of in vitro susceptibility was performed using broth microdilution assay following the Clinical and Laboratory Standards Institute guidelines for filamentous fungi. The cytotoxicity was evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Aspergillus clavatus and A. fumigatus were more susceptible species for complexes 1 and 2. Other complexes showed excellent minimum inhibitory concentration (4-64 µg ml(-1)) against most microorganisms. Complexes 1 and 2 are respectively 180- and 95-fold more active than the corresponding free ligands against A. clavatus and the complex 5 is 46-fold more active than free ligand against A. niger. Aspergillus niger was more susceptible to the action of the complexes 1 and 5 (16 µg ml(-1)). A low cytotoxic activity (IC(50) > 10(-6) mol l(-1) ) on normal mammalian cells (BHK-21) to the evaluated complexes was measured. Ruthenium complexes are promising antifungal agents against the development of novel effective drug against different species of Aspergillus; however, for A. nomius and A. terreus, they were not active in the highest concentration tested.


Subject(s)
Aspergillus/drug effects , Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacology , Thiocarbamates/chemistry , Thiocarbamates/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cricetinae , Microbial Sensitivity Tests , Ruthenium Compounds/toxicity , Thiocarbamates/toxicity
5.
Bioorg Med Chem ; 17(14): 5038-43, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19539479

ABSTRACT

Although effective against epimastigotes (proliferative form) and of low cytotoxicity in mammals, the aryl-4-oxothiazolylhydrazones (ATZ) display only limited activity against trypomastigotes (bloodstream form) of Trypanosoma cruzi. Considering the metal complexation approach with bioactive ligands as one possible strategy for improving the biological efficacy of ATZ, a set of eight new ruthenium-ATZ complexes (RuCl(2)ATZCOD, COD is 1,5-cyclooctadiene) were prepared, chemically and biologically characterized, including in vitro assays against epimastigotes and trypomastigote forms of the parasite and also assessment of cytotoxicity in mammals. Two of these complexes presented antitrypanosomal activity at non-cytotoxic concentrations on mammalian cells and of higher potency than its metal-free ligands, while the metallic precursor [RuCl(2)COD(MeCN)(2)] showed only moderate antitrypanosomal activity. Comparative analysis between the ruthenium complexes and metal-free ligands demonstrated the usefulness of this approach, with the establishment of new SAR data. Additional pharmacological tests, including a DNA bond assay, gave rise to the proposal of a single preliminary explanation for the molecular origin of the bioactivity.


Subject(s)
Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , DNA/metabolism , Hydrazones/chemical synthesis , Hydrazones/chemistry , Hydrazones/pharmacology , Mice , Mice, Inbred BALB C , Plasmids/metabolism , Ruthenium Compounds/chemical synthesis , Ruthenium Compounds/toxicity , Spleen/cytology , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacology , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/toxicity , Trypanosoma cruzi/growth & development
6.
Sci Rep ; 9(1): 4146, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858469

ABSTRACT

Sulpiride (SPR) is a selective antagonist of central dopamine receptors but has limited clinical use due to its poor pharmacokinetics. The aim of this study was to investigate how metal ligation to SPR may improve its solubility, intestinal permeability and prolong its half-life. The synthesis and characterisation of ternary metal complexes [Ru(p -cymene)(L)(SPR)]PF6 (L1 = (R)-(+)-2-amino-3-phenyl-1-propanol, L2 = ethanolamine, L3 = (S)-(+)-2-amino-1-propanol, L4 = 3-amino-1-propanol, L5 = (S)-(+)-2-pyrrolidinemethanol) are described in this work. The stability constant of the [Ru(p -cymene)(SPR)] complex was determined using Job's method. The obtained value revealed higher stability of the metal complex in the physiological pH than in an acidic environment such as the stomach. The ternary metal complexes were characterised by elemental analysis, Fourier transform infrared spectroscopy (FT-IR), 1H and 13C nuclear magnetic resonance (NMR), differential scanning calorimetry (DSC), thermal analyses, Ultraviolet-Visible (UV-Vis). Solubility studies showed higher aqueous solubility for complexed SPR than the free drug. Dissolution profiles of SPR from the metal complexes exhibited slower dissolution rate of the drug. Permeation studies through the pig's intestine revealed enhanced membrane permeation of the complexed drug. In vitro methyl thiazolyl tetrazolium (MTT) assay showed no noticeable toxic effects of the ternary metal complexes on Caco-2 cell line.


Subject(s)
Dopamine Antagonists/chemical synthesis , Ruthenium Compounds/chemical synthesis , Sulpiride/analogs & derivatives , Animals , Caco-2 Cells , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/toxicity , Humans , Intestinal Absorption , Propanolamines/chemistry , Ruthenium Compounds/pharmacokinetics , Ruthenium Compounds/toxicity , Swine
7.
Inorg Chem ; 47(15): 6964-73, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18593112

ABSTRACT

The synthetic, spectroscopic, structural, and biological studies of a bis(arylimino)pyridine Ru(III) chloride compound containing the ligand, 2,6-bis(2,4,6-trimethylphenyliminomethyl)pyridine are reported. The bis(arylimino)pyridine ligand, with three donor nitrogen atoms, was synthesized by condensation of 2,6-pyridinedicarboxaldehyde with 2,4,6-trimethylaniline. The Ru(III) complex, with formula [RuCl 3(L1)](H 2O) (RuL1), where L1 = 2,6-bis(2,4,6-trimethylphenyliminomethyl)pyridine, was structurally determined on the basis of analytical and spectroscopic (IR, UV-vis, ESI-MS) studies. A straightforward strategy to fully characterize the paramagnetic compound using advanced (1)H NMR is reported. This new complex is a prototype for a series of new anticancer Ru(III) and Ru(II) compounds with improved cytostatic properties; likely to be modified in a desirable manner due to the relatively facile ligand modification of the bis(imino)pyridines and their molecular architecture. The present Ru(III) complex is the first example of this family of Ru(III)/Ru(II) anticancer compounds with the aimed physicochemical characteristics. Although the ligand itself is moderately active in selected cell lines (EVSA-T and MCF-7), the activity of the [Ru(L1)Cl 3] complex has increased significantly for a broad range of cancer cell lines tested in vitro (IC 50 values = 11 approximately 17 microM). Reaction of the RuL1 species with the DNA model base 9-ethylguanine (9EtGua) was found to produce in a redox reaction the species trans-[Ru(II)(L1)(9EtGua) 2(H 2O)](ClO 4) 2 (abbreviated as RuL1-9EtGua), which was studied in solution and also in the solid state, by X-ray crystallography. The structure comprises the as yet unknown trans-bis(purine)Ru(II) unit.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Imines/chemistry , Pyridines/chemistry , Ruthenium Compounds/chemical synthesis , Ruthenium Compounds/pharmacology , Absorption , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Humans , Ruthenium Compounds/chemistry , Ruthenium Compounds/toxicity , Spectrum Analysis
8.
Bioresour Technol ; 99(17): 8207-11, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18448332

ABSTRACT

Electrochemical treatment of oleate using RuO2 and IrO2 type dimensionally stable anodes in alkaline medium was performed to develop a feasible anaerobic pre-treatment of fatty effluents. The results showed that the pre-treated solutions over RuO2 were faster degraded by anaerobic consortium than the raw oleate solutions or the electrolysed solutions using IrO2. In batch experiments carried out with pre-treated solutions over RuO2 (100-500mg/L), no lag phases were observed before the methane production onset. On the other hand, raw oleate and pre-treated oleate over IrO2 had originated lag phases of 0-140 and 0-210h, respectively. This study demonstrated that it is advantageous to apply the electrochemical treatment carried out on the RuO2 type DSA in order to achieve a faster biodegradation of lipid-containing effluent and consequently to obtain a faster methane production.


Subject(s)
Fats/metabolism , Ferric Compounds/chemistry , Oleic Acid/metabolism , Ruthenium Compounds/chemistry , Anaerobiosis , Bacteria/drug effects , Biodegradation, Environmental , Electrochemistry , Electrodes , Ferric Compounds/toxicity , Methane/metabolism , Oxidation-Reduction , Ruthenium Compounds/toxicity , Toxicity Tests
9.
Biomed Pharmacother ; 107: 1082-1092, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30257320

ABSTRACT

Anticancer potential of ruthenium complexes has been widely investigated, but safety evaluation studies are still scarce. Despite of ruthenium-based anticancer agents are known to cause fewer side effects compared to other metal-based drugs, these compounds are not fully free of toxicity, causing mainly nephrotoxicity. Based on the promising results from antitumor activity of the complexes [Ru(L-Met)(bipy)(dppb)]PF6 (RuMet) and [Ru(L-Trp)(bipy)(dppb)]PF6 (RuTrp), for the first time we investigated the toxicity profile of these complexes in rodent and zebrafish models. The acute oral toxicity was evaluated in Swiss mice. The mutagenic and genotoxic potential was determined by a combination of Micronucleus (MN) and Comet assay protocols, after exposure of Swiss mice to RuMet and RuTrp in therapeutic doses. Zebrafish embryos were exposed to these complexes, and their development observed up to 96 h post-fertilization. RuMet and RuTrp complexes showed low acute oral toxicity. Recorded behavioral changes were not recorded, nor were macroscopic morphological changes or structural modifications in the liver and kidneys. These complexes did not cause genetic toxicity, presenting a lack of micronuclei formation and low DNA damage induction in the cells from Swiss mice. In contradiction, cisplatin treatment exhibited high mutagenicity and genotoxicity. RuMet and RuTrp showed low toxicity in the embryo development of zebrafish. The RuMet and RuTrp complexes demonstrated low toxicity in the two study models, an interesting property in preclinical studies for novel anticancer agents.


Subject(s)
Antineoplastic Agents/toxicity , DNA Damage/drug effects , Embryonic Development/drug effects , Ruthenium Compounds/toxicity , Administration, Oral , Amino Acids/chemistry , Animals , Antineoplastic Agents/chemistry , Cisplatin/toxicity , Comet Assay , Female , Male , Mice , Micronucleus Tests , Ruthenium Compounds/chemistry , Toxicity Tests, Acute , Zebrafish
10.
Chem Biol Interact ; 167(2): 116-24, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17368594

ABSTRACT

Several metallic compounds recognized as potent antitumor agents, have been developed and tested in vivo and in vitro. In this work, we evaluated the toxic, therapeutic, and cytotoxic properties of the cis-dichloro-tetra-amine-ruthenium(III) chloride. Transplanted animals with Sarcoma 180 cells were treated with ruthenium(III) complex and injected i.p., at different time intervals. After the 15th day, tumoral postimplant, the animals were sacrificed and their lungs, kidneys, liver, and tumors were removed and processed for histopathological analysis. Blood samples were also taken for haematological and biochemical analyses. Interaction between the ruthenium complex and the DNA was also investigated. Besides being cytotoxic for the S180 cells, the metallic compound induced tumoral volume reduction and increased survival time of the animals treated. Serum levels of LDH, creatinine, and bilirubin increased, but no serious irreversible histopathological alterations were observed in the analyzed tissues. The compound did not cause anemia, but reduced the number of leukocytes in the treated animals. The absence of viable S180 cells, necrotic cells, and the presence of granulation tissue were observed in tumor tissue of treated animals. The Ru(III) complex, in the presence of the reduction agent, caused plasmid DNA to fragment. These results suggest that cis-RuCl(2)(NH(3))(4)Cl compound is a potent antitumoral drug in vitro and in vivo, which seems to involve binding to DNA molecule.


Subject(s)
Antineoplastic Agents/pharmacology , Ruthenium Compounds/pharmacology , Sarcoma 180/drug therapy , Animals , Antineoplastic Agents/toxicity , Bilirubin/blood , Blood Cell Count , Creatinine/blood , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Histocytochemistry , Hydro-Lyases/blood , Inhibitory Concentration 50 , Lethal Dose 50 , Male , Mice , Mice, Inbred BALB C , Plasmids/drug effects , Plasmids/metabolism , Ruthenium Compounds/toxicity , Sarcoma 180/blood , Sarcoma 180/genetics , Sarcoma 180/pathology
11.
Biochem Pharmacol ; 71(4): 408-15, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16360645

ABSTRACT

Novel ruthenium(II) organo-metallic compounds are active in ovarian cancer models [Aird RE, Cummings J, Ritchie AA, Muir M, Morris RE, Chen H, et al. In vitro and in vivo activity and cross resistance profiles of novel ruthenium(II) organometallic arene complexes in human ovarian cancer. Br J Cancer 2002;86(10):1652-7]. [(eta6-C6H5C6H5)Ru(en)Cl]+ (as a PF6 salt, where en=ethylenediamine (RM175)) has been evaluated in a 13-cell line panel. Particular sensitivity (approximately 10-fold lower than mean IC50) was noted in breast cancer and non-small cell lung cancer cell lines. In addition, IC50 in the A549 was 2 microM and RM175 (25 mg kg-1, days 1 and 5, i.p.) caused a significant (p=0.004) growth delay in a xenograft model. HC11 [(eta6-tetrahydroanthracene)Ru(en)Cl]PF6 was more potent in the A549 cell line (IC50 0.5 microM). HC11 (25 mg kg-1, days 1, 8 and 15, i.p.) was also active in vivo. Following RM175 25 mg kg-1, days 1 and 5, and 15 mg kg-1, days 1-5, HC11 25 and 40 mg kg-1, day 1, elevated alanine transaminase levels were detected, suggesting hepatotoxicity. No changes were observed in kidney or haematological parameters. In liver sections, multi-focal hepatic necrosis was seen, becoming confluent at high doses of HC11. In vitro studies confirmed that HC11 was more toxic than RM175 to fresh human hepatocytes and equitoxic to mithramycin. Liver toxicity may be related to the arene ligand and modification may reduce the potential for hepatic toxicity, while maintaining the anti-tumour activity seen.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Xenograft Model Antitumor Assays , Alanine Transaminase/blood , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Carcinoma, Large Cell/drug therapy , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Inhibitory Concentration 50 , Injections, Intraperitoneal , Liver/drug effects , Liver/metabolism , Liver/ultrastructure , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Nude , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , Ruthenium Compounds/chemistry , Ruthenium Compounds/toxicity , Weight Loss/drug effects
12.
J Inorg Biochem ; 146: 89-96, 2015 May.
Article in English | MEDLINE | ID: mdl-25708310

ABSTRACT

The antitumor activity of a ruthenium(II) polypyridyl complex, Δ-[Ru(bpy)2(HPIP)](ClO4)2 (Δ-Ru1, where bpy=2,2'-bipyridine, HPIP=2-(2-hydroxyphenyl)imidazo[4,5-f][1,10]phenanthroline), was evaluated. The in vivo experiments showed that Δ-Ru1 inhibited the growth of a human cervical carcinoma cell line (HeLa) xenotransplanted into nude mice with efficiency similar to that of cisplatin. Histopathology examination of the tumors from treated xenograft models was consistent with apoptosis in tumor cells. Importantly, in striking contrast with cisplatin, Δ-Ru1 did not cause any detectable side effects on the kidney, liver, peripheral neuronal system, or the hematological system at the pharmacologically effective dose. The preclinical studies reported here provide support for the clinical use of Δ-Ru1 as an exciting new drug candidate with lower toxicity than cisplatin, endowed with proapoptotic properties.


Subject(s)
Antineoplastic Agents/adverse effects , Organometallic Compounds/adverse effects , Ruthenium Compounds/adverse effects , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cisplatin/adverse effects , Female , HeLa Cells , Humans , Kidney/drug effects , Liver/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Organometallic Compounds/toxicity , Peripheral Nerves/drug effects , Ruthenium Compounds/pharmacokinetics , Ruthenium Compounds/toxicity
13.
J Inorg Biochem ; 83(2-3): 139-49, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11237253

ABSTRACT

Estimates of the net equilibrium binding constants for [(H2O)(NH3)5RuII]2+, [Cl(NH3)5RuIII]2+, cis-[(H2O)2(NH3)4RuII]2+ and cis-[Cl2(NH3)4RuIII]+ with apotransferrin (Tf) and holotransferrin (Fe2Tf) suggests that RuIII, but not RuII complexes bind with a higher affinity to the iron binding sites. Several other presumably histidyl imidazole sites bind with approximately the same affinity (Keff = 10(2) to 10(3) M(-1) to both RuII and RuIII. Compared to HeLa cells, an order of magnitude higher level of nuclear DNA binding ([Ru]DNA/[P]DNA) was required to achieve the same level of toxicity in Jurkat Tag cells, which probably relates to the substantially higher levels of cis-[Cl2(NH3)4Ru]+ needed to inhibit 50% of the cell growth in the Jurkat Tag cell line. Against Jurkat Tag cells, the toxicity of the pentaammineruthenium(III) group is enhanced by approximately two orders of magnitude upon binding primarily to the Fe-sites in apotransferrin, whereas the toxicity of the tetraammineruthenium(III) moiety is only marginally increased. Binding to Fe2Tf does not increase the toxicity of either group. Significant dissociation over 24 h of the ammineruthenium(III) ions from apotransferrin requires reduction to RuII.


Subject(s)
Antineoplastic Agents/pharmacology , Ruthenium Compounds/pharmacology , Transferrin/chemistry , Transferrin/pharmacology , Antineoplastic Agents/chemistry , Citric Acid/metabolism , DNA/metabolism , Drug Screening Assays, Antitumor , Glutathione/metabolism , Humans , Jurkat Cells , Kinetics , Oxidation-Reduction , Protein Binding , Ruthenium Compounds/chemical synthesis , Ruthenium Compounds/chemistry , Ruthenium Compounds/toxicity
14.
Chem Biol Interact ; 95(1-2): 109-26, 1995 Mar 30.
Article in English | MEDLINE | ID: mdl-7697744

ABSTRACT

A series of 18 ruthenium(III) complexes, structurally related to the selective antimetastatic drug Na[trans-RuCl4(DMSO)Im], and characterized by the presence of sulfoxide and nitrogen-donor ligands were tested on TLX5 lymphoma and some of them on MCa mammary carcinoma to evaluate the dependence of the degree of cytotoxicity and of antimetastatic activity on the chemical properties. In vitro cytotoxicity is present only at high concentrations (> 10(-4) M), depends upon lipophilicity and is markedly affected by the presence of 5% serum or plasma samples in the culture medium. The comparison of the effects on in vitro cytotoxicity with in vivo antitumor and antimetastatic action points out that these compounds reduce metastasis formation by a mechanism unrelated to a direct tumor cell cytotoxicity. If on one hand Na[trans-RuCl4(TMSO)Iq], the compound that shows the most potent in vitro cytotoxic effects, is the least effective against metastases, on the other Na[trans-RuCl4(DMSO)Im], the compound that better reduces metastasis formation, is rather devoid of cytotoxic effects on tumor cells kept in vitro. In particular, Na[trans-RuCl4(DMSO)Im] seems to distinguish between artificially induced metastases and spontaneous metastases and reduces only the former by a cytotoxic mechanism. Out of all the tested compounds, with the exception of Na[trans-RuCl4(DMSO)Ox], Na[trans-RuCl4(DMSO)Im] is confirmed to be the most selective antimetastatic agent of the group.


Subject(s)
Antineoplastic Agents/toxicity , Brain Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Lymphoma/drug therapy , Lymphoma/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Ruthenium Compounds/toxicity , Animals , Brain Neoplasms/secondary , Drug Screening Assays, Antitumor , Female , Mice , Mice, Inbred CBA , Neoplasm Transplantation
15.
J Biosci ; 26(2): 271-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11426063

ABSTRACT

Trans-imidazolium (bis imidazole) tetrachloro ruthenate (RuIm) and trans-indazolium (bis indazole) tetrachloro ruthenate (RuInd) are ruthenium coordination complexes, which were first synthesized and exploited for their anticancer activity. These molecules constitute two of the few most effective anticancer ruthenium compounds. The clinical use of these compounds however was hindered due to toxic side effects on the human body. Our present study on topoisomerase II poisoning by these compounds shows that they effectively poison the activity of topoisomerase II by forming a ternary cleavage complex of DNA, drug and topoisomerase II. The thymidine incorporation assays show that the inhibition of cancer cell proliferation correlates with topoisomerase II poisoning. The present study on topoisomerase II poisoning by these two compounds opens a new avenue for renewing further research on these compounds. This is because they could be effective lead candidates for the development of more potent and less toxic ruthenium containing topoisomerase II poisons. Specificity of action on this molecular target may reduce the toxic effects of these ruthenium-containing molecules and thus improve their therapeutic index.


Subject(s)
DNA/metabolism , Imidazoles/pharmacology , Indazoles/pharmacology , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Topoisomerase II Inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , DNA/chemistry , DNA Topoisomerases, Type II/metabolism , Humans , Imidazoles/toxicity , Indazoles/toxicity , Molecular Structure , Nucleic Acid Conformation , Organometallic Compounds/toxicity , Rats , Ruthenium Compounds/toxicity , Thymidine/metabolism , Tumor Cells, Cultured
16.
PLoS One ; 9(10): e105865, 2014.
Article in English | MEDLINE | ID: mdl-25329644

ABSTRACT

Over the past several decades, much attention has been focused on ruthenium complexes in antitumor therapy. Ruthenium is a transition metal that possesses several advantages for rational antitumor drug design and biological applications. In the present study, five ruthenium complexes containing amino acids were studied in vitro to determine their biological activity against sarcoma-180 tumor cells. The cytotoxicity of the complexes was evaluated by an MTT assay, and their mechanism of action was investigated. The results demonstrated that the five complexes inhibited the growth of the S180 tumor cell line, with IC50 values ranging from 22.53 µM to 50.18 µM, and showed low cytotoxicity against normal L929 fibroblast cells. Flow cytometric analysis revealed that the [Ru(gly)(bipy)(dppb)]PF6 complex (2) inhibited the growth of the tumor cells by inducing apoptosis, as evidenced by an increased number of Annexin V-positive cells and G0/G1 phase cell cycle arrest. Further investigation showed that complex 2 caused a loss of mitochondrial membrane potential; activated caspases 3, caspase-8, and caspase-9 and caused a change in the mRNA expression levels of caspase 3, caspase-9 as well as the bax genes. The levels of the pro-apoptotic Bcl-2 family protein Bak were increased. Thus, we demonstrated that ruthenium amino acid complexes are promising drugs against S180 tumor cells, and we recommend further investigations of their role as chemotherapeutic agents for sarcomas.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis , Ruthenium Compounds/toxicity , Sarcoma/metabolism , Amino Acids/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Mice , Ruthenium/chemistry , Ruthenium Compounds/chemical synthesis
17.
J Phys Chem B ; 118(36): 10653-61, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25133573

ABSTRACT

One of the major challenges in drug design is to identify compounds with potential toxicity toward target cells, preferably with molecular-level understanding of their mode of action. In this study, the antitumor property of a ruthenium complex, mer-[RuCl3(dppb)(VPy)] (dppb = 1,4-bis(diphenylphosphine)butane and VPy = 4-vinylpyridine) (RuVPy), was analyzed. Results showed that this compound led to a mortality rate of 50% of HEp-2 cell with 120 ± 10 µmol L(-1), indicating its high toxicity. Then, to prove if its mode of action is associated with its interaction with cell membranes, Langmuir monolayers were used as a membrane model. RuVPy had a strong effect on the surface pressure isotherms, especially on the elastic properties of both the zwitterionic dipalmitoylphosphatidylcholine (DPPC) and the negatively charged dipalmitoylphosphatidylglycerol (DPPG) phospholipids. These data were confirmed by polarization-modulated infrared reflection-absorption spectroscopy (PM-IRRAS). In addition, interactions between the positive group from RuVPy and the phosphate group from the phospholipids were corroborated by density functional theory (DFT) calculations, allowing the determination of the Ru complex orientation at the air-water interface. Although possible contributions from receptors or other cell components cannot be discarded, the results reported here represent evidence for significant effects on the cell membranes which are probably associated with the high toxicity of RuVPy.


Subject(s)
Cell Membrane/drug effects , Ruthenium Compounds/toxicity , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Air , Animals , Cell Line , Cell Membrane/metabolism , Cell Survival/drug effects , Chlorocebus aethiops , Elasticity , Humans , Membranes, Artificial , Models, Biological , Models, Chemical , Phosphatidylglycerols/chemistry , Pressure , Pyridines/toxicity , Spectrophotometry, Infrared , Water/chemistry
18.
Metallomics ; 6(12): 2204-12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25142071

ABSTRACT

Effective chemotherapy drugs for cancer that would inhibit tumor growth and suppress metastasis are currently lacking. In this study, a series of arene ruthenium complexes, [(η6-arene)Ru(H2iip)Cl]Cl (arene = p-cymene, RAWQ03; CH3C6H5, RAWQ04; and C6H6, RAWQ11), were synthesized and their inhibitory activity against tumor cells were evaluated. The results showed that the complex RAWQ11 inhibited the growth of MDA-MB-231 breast cancer cells by inducing S-phase arrest, which is closely related to the inhibition of cell mitosis-mediated cell nucleus damage. Further studies showed that RAWQ11 can inhibit the invasion and metastasis of MDA-MB-231 cells. The morphology of MDA-MB-231 cells changed, the number of focal adhesions decreased, and the stress fibers de-polymerized upon dealing with the complex RAWQ11. The FITC-gelatin assay confirmed that the formation of invadopodia in MDA-MB-231 cells was significantly blocked by RAWQ11. Furthermore, RAWQ11 can block the AKT signal pathway by upregulating the PTEN expression through binding and downregulating miR-21. These results demonstrated that this type of arene ruthenium(ii) complex can block the invadopodia formation by regulating the PTEN/AKT signal pathway mediated by miR-21 to inhibit the invasion and metastasis of breast cancer cells. Therefore, this complex can be used as a potential dual functional agent to inhibit the growth and metastasis of tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Surface Extensions/drug effects , Focal Adhesions/drug effects , Ruthenium Compounds/pharmacology , Stress Fibers/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cytoskeleton/drug effects , Humans , Microscopy, Fluorescence , Ruthenium Compounds/chemistry , Ruthenium Compounds/toxicity
19.
Zebrafish ; 7(1): 13-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20192849

ABSTRACT

N-Heterocyclic carbene (NHC) ligands have attracted great interest over the last decade for their use in the design of homogenous catalysts. NHC-based metal complexes have interesting potential biomedical applications, such as in antimicrobial and cancer therapy, which are beginning to be explored more fully. We have studied here the oxidant activities of a series of Ru(II) complexes in vitro and zebrafish (Danio rerio) have been used as a model in vivo to investigate and characterize the toxicity of some of these compounds. Dual behavior was observed for the NHC-based complexes as they behaved as antioxidants at low concentrations but showed pro-oxidant capacity at higher concentrations. Zebrafish embryos were exposed to Ru(II) complexes under several different conditions (0 or 24 h postfertilization, with or without the chorion) and various parameters, such as viability, edema, heart rate, blood coagulation, pigmentation, scoliosis, malformation, and hatching, were tested. In general, zebrafish embryos were not harmed by exposure to Ru(II) complexes whatever the experimental conditions. Several toxicity profiles were observed depending upon the chemical structure of the compound in question. Their characteristics as pro-oxidant and/or antioxidant agents together with their biosafety may point to their having biomedical applications as antitumoral or neuroprotective drugs.


Subject(s)
Heterocyclic Compounds/toxicity , Methane/analogs & derivatives , Oxidants/toxicity , Ruthenium Compounds/toxicity , Zebrafish/embryology , Animals , Heart/drug effects , Heart/embryology , Heterocyclic Compounds/chemistry , Larva/drug effects , Methane/chemistry , Methane/toxicity , Molecular Structure , Oxidants/chemistry , Ruthenium Compounds/chemistry , Structure-Activity Relationship
20.
Curr Med Chem ; 17(31): 3643-57, 2010.
Article in English | MEDLINE | ID: mdl-20846113

ABSTRACT

The discovery of the involvement of nitric oxide (NO) in several physiological and pathophysiological processes launched a spectacular increase in studies in areas such as chemistry, biochemistry, and pharmacology. As a consequence, the development of NO donors or scavengers for regulation of its concentration and bioavailability in vivo is required. In this sense, ruthenium nitrosyl ammines and aliphatic tetraazamacrocyles have attracted a lot of attention due to their unique chemical properties. These complexes are water soluble and stable in solution, not to mention that they can deliver NO when photochemically or chemically activated by the reduction of the coordinated nitrosonium (NO+). The tuning of the energies of the charge transfer bands, the redox potential, and the specific rate constants of NO liberation, in both solution and matrices, is desirable for the achievement of selective NO delivery to biological targets, hence making the ruthenium ammines and aliphatic tetraazamacrocyles a quite versatile platform for biological application purposes. These ruthenium nitrosyls have shown to be active in firing neurons in mouse hippocampus, performing redox reactions in mitochondria, acting in blood pressure control, exhibiting cytotoxic activities against trypanosomatids (T.cruzi and L.major) and tumor cells. This tailoring approach is explored here, being heavily supported by the accumulated knowledge on the chemistry and photochemistry of ruthenium complexes, which allows NO donors/scavengers systems to be custom made designed.


Subject(s)
Nitric Oxide Donors/pharmacology , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Ruthenium/pharmacology , Amines/chemistry , Amines/pharmacology , Animals , Ligands , Male , Mice , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide Donors/toxicity , Nitrous Oxide , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/toxicity , Oxidation-Reduction , Photochemical Processes , Rats , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/toxicity , Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacokinetics , Ruthenium Compounds/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL