Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Inorg Biochem ; 153: 103-113, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26492162

ABSTRACT

Silver(I) complexes of coumarin-based ligands and one of their phenanthroline (phen) adducts have been prepared and characterized using microanalytical data, molar conductivity, IR, (1)H and (13)C NMR, UV-Vis, and atomic absorption (AAS) spectroscopies. The binding modes of the coumarin-based ligands and the most probable structure of their Ag(I) complexes were predicted by means of molecular modeling and calculations of their IR, NMR, and absorption spectra using density functional theory (DFT). The cytotoxicity of the compounds studied against human-derived hepatic carcinoma cells (Hep-G2) and a renal cancer cell line (A498) showed that the complexes were more cytotoxic than the clinically used chemotherapeutic, mitoxantrone. The compounds showed little interaction with DNA and also did not show nuclease activity but manifested excellent superoxide dismutase activity which may indicate that their mechanism of action is quite different to many metal-based therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Coumarins/pharmacology , Phenanthrolines/pharmacology , Silver/chemistry , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Coordination Complexes/chemistry , Coumarins/chemistry , DNA/chemistry , Hep G2 Cells , Humans , Ligands , Mitoxantrone/pharmacology , Models, Molecular , Phenanthrolines/chemistry , Solubility
2.
J Inorg Biochem ; 124: 70-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23603796

ABSTRACT

The successful design and synthesis of a novel Pt complex of the histone deacteylase inhibitor belinostat are reported. Molecular modelling assisted in the identification of a suitable malonate derivative of belinostat (mal-p-Bel) for complexation to platinum. Reaction of [Pt(NH3)2(H2O)2](NO3)2 with the disodium salt of mal-p-Bel gave cis-[Pt(NH3)2(mal-p-Bel-2H)] (where -2H indicates that mal-p-Bel is doubly deprotonated) in excellent yield. An in vitro cytotoxicity study revealed that cis-[Pt(NH3)2(mal-p-Bel-2H)] possesses (i) considerable cytotoxicity against reported ovarian cancer cell lines, (ii) enhanced cytotoxicity relative to the previously reported Pt histone deacetylase inhibitor conjugate, cis-[Pt(II)(NH3)2(malSAHA-2H)] and (iii) favourable cyto-selective properties as compared to cisplatin and belinostat.


Subject(s)
Cell Proliferation/drug effects , Cytotoxins , Histone Deacetylase Inhibitors , Hydroxamic Acids , Platinum Compounds , Sulfonamides , Cell Line, Tumor , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , Female , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Platinum Compounds/chemical synthesis , Platinum Compounds/chemistry , Platinum Compounds/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology
3.
Eur J Pharmacol ; 689(1-3): 45-55, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22705894

ABSTRACT

This study determined the cytotoxic, cyto-selective and mutagenic potential of novel quinolinone Schiff base ligands and their corresponding copper(II) complexes in human-derived hepatic carcinoma cells (Hep-G2) and non-malignant human-derived hepatic cells (Chang). Results indicated that complexation of quinolinone Schiff bases with copper served to significantly enhance cytotoxicity. Here, the complex of (7E)-7-(3-ethoxy-2-hydroxybenzylideamino)-4-methylquinolin-2(1H)-one (TV117-FM) exhibited the lowest IC(50) value (17.9 µM) following 96 h continuous exposure, which was comparable to cisplatin (15.0 µM). However, results revealed that TV117-FM lacked cytoselectivity over non-malignant cells. Additionally, the complex was minimally effluxed from cells via Pglycoprotein (P-gp) and was shown to be non-mutagenic in the Standard Ames test. Furthermore, BrdU incorporation assays showed that it was capable of inhibiting DNA synthesis in a concentrationand time-dependent manner. However, inhibition was not as a consequence of DNA intercalation, as illustrated in electrophoretic mobility shift assays. Interestingly, it was shown that the ligand was capable of inhibiting the action of topoisomerase II, but this was lost following complexation. This indicated that the mechanism of action of the novel copper(II) complex was different from that of the parent ligand and suggests that TV117-FM may have a therapeutic role to play in the treatment of hepatocellular carcinoma. Studies are currently underway to elucidate the exact in vitro mechanism of action of this novel, metal-based anti-cancer agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Copper/therapeutic use , Liver Neoplasms/drug therapy , Mutagenesis/drug effects , Quinolones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , CHO Cells , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cell Survival/physiology , Copper/chemistry , Copper/pharmacology , Cricetinae , Cricetulus , HeLa Cells , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mutagenesis/physiology , Quinolones/chemistry , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Schiff Bases/chemistry , Schiff Bases/pharmacology , Schiff Bases/therapeutic use
4.
J Inorg Biochem ; 105(6): 793-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21497577

ABSTRACT

The first examples of Pt complexes of the well known anti-epilepsy drug and histone deacetylase inhibitor, valproic acid (VPA), are reported. Reaction of the Pt(II) am(m)ine precursors trans-[PtCl(2)(NH(3))(py)] and trans-[PtCl(2)(py)(2)] with silver nitrate and subsequently sodium valproate gave trans-[Pt(VPA(-1H))(2)(NH(3))(py)] and trans-[Pt(VPA(-1H))(2)(py)(2)], respectively. The valproato ligands in both complexes are bound to the Pt(II) centres via the carboxylato functionality and in a monodentate manner. The X-ray crystal structure of trans-[Pt(VPA(-1H))(2)(NH(3))(py)] is described. Replacement of the dichlorido ligands in trans-[PtCl(2)(py)(2)] and trans-[PtCl(2)(NH(3))(py)] by valproato ligands (VPA(-1H)) to yield trans-[Pt(VPA(-1H))(2)(py)(2)] and trans-[Pt(VPA(-1H))(2)(NH(3))(py)] respectively, significantly enhanced cytotoxicity against A2780 (parental) and A2780 cisR (cisplatin resistant) ovarian cancer cells. The mutagenicity of trans-[Pt(VPA(-1H))(2)(NH(3))(py)] and trans-[Pt(VPA(-1H))(2)(py)(2)] was determined using the Ames test and is also reported.


Subject(s)
Antineoplastic Agents/chemical synthesis , Histone Deacetylase Inhibitors/chemical synthesis , Mutagens/chemical synthesis , Organoplatinum Compounds/chemistry , Valproic Acid/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Crystallography, X-Ray , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/toxicity , Humans , Inhibitory Concentration 50 , Ligands , Male , Mutagens/chemistry , Mutagens/toxicity , Organoplatinum Compounds/toxicity , Rats , Rats, Sprague-Dawley
5.
Eur J Pharmacol ; 602(2-3): 203-14, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19041861

ABSTRACT

The central objective of the current study was to investigate the potential in vitro anti-proliferative effect of 4-hydroxy-3-nitro-coumarin (hncH), and the mixed-ligand silver (I) complex of 4-oxy-3-nitro-coumarin-bis(phenanthroline), [Ag(hnc)(phen)(2)] using four human-derived model cell lines. In addition, selected mechanistic studies were carried out using the most sensitive of the four cell lines. Results obtained show that the complex could decrease the proliferation of all four cell lines including neoplastic renal and hepatic, namely A-498 and HepG(2) cells, respectively, along with two non-neoplastic renal and hepatic cell lines, HK-2 and Chang, respectively. Furthermore, non-neoplastic hepatic cells (Chang) appeared to be less sensitive to the effect of the complex, but this effect was not replicated in the non-neoplastic renal (HK-2) cells. Based on IC(50) values [Ag(hnc)(phen)(2)] was shown to be almost four times more potent than cisplatin, using HepG(2) cells. In addition, the observed anti-proliferative effect was shown to be both dose- and time-dependent. Furthermore, the complex was shown to decrease DNA synthesis, but did not intercalate with it. Moreover, there was no evidence that P-glycoprotein-mediated multi-drug resistance was likely to decrease anti-proliferative activity. Cytological stains, analysis of genomic DNA, and biochemical assays [caspase-3 and -9 and cleaved poly(ADP-ribose)-polymerase protein] showed that cell death appeared to result from apoptosis, with the possibility of secondary necrosis. Additionally, flow cytometric analysis showed that the complex functioned through an alteration in cell cycle progression. Taken together, [Ag(hnc)(phen)(2)] has been shown to be a more potent anti-proliferative agent than cisplatin, capable of altering key biochemical events leading to cell death. Additional mechanistic studies are underway to probe more fully its mechanism of action.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Neoplasms/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Silver/chemistry , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism
6.
Eur J Pharmacol ; 569(1-2): 16-28, 2007 Aug 13.
Article in English | MEDLINE | ID: mdl-17585902

ABSTRACT

The central objective of the current study was to investigate the potential in vitro anti-proliferative effect of the parent ligand, 4-methylcoumarin-6,7-dioxyacyeic acid (4-MecdoaH(2)), and its copper (II) complex, bis(phenanthroline4-methylcoumarin-6,7-dioxacetatocopper(II) ([Cu(4-Mecdoa)(phen)(2)]) using four human model cell lines. In addition, selected mechanistic studies were carried out using the most sensitive of the four cell lines. Results obtained show that the complex could alter proliferation of both human neoplastic renal (A-498) and hepatic (HepG2) cells. Furthermore, non-neoplastic hepatic (CHANG) cells appeared to be less sensitive. However, this effect was not duplicated with non-neoplastic renal (HK-2) cells, a profile shared by cisplatin. The observed anti-proliferative effect appeared to be dose-and time-dependent, and could be attributed to the complex, rather than any of the free components i.e. the 1,10-phenanthroline or coumarin ligand, or the simple metal salt. Furthermore, the complex was shown to decrease DNA synthesis, but did not intercalate with it. Based on IC(50) values, [Cu(4-Mecdoa)(phen)(2)] was shown to be almost 12 times more potent than cisplatin. Moreover, there was no evidence that P-glycoprotein-mediated multi-drug resistance was likely to decrease anti-proliferative activity. Cytological stains, analysis of genomic DNA, and biochemical assays [caspase-3 and -9 and cleaved poly(ADP-ribose)-polymerase protein], showed that cell death could switch between apoptosis and necrosis, and this effect appeared to be concentration-dependent. Additionally, flow cytometric analysis showed that the complex functioned through an alteration in cell cycle progression. Taken together, [Cu(4-Mecdoa)(phen)(2)] has been shown to be a more potent anti-proliferative agent than either the ligand or cisplatin, and is capable of altering key biochemical events leading to the execution of apoptotic and/or necrotic cell death, suggesting that it is worthy of further investigation.


Subject(s)
Acetates/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Copper/pharmacology , Coumarins/pharmacology , Organometallic Compounds/pharmacology , Acetates/chemistry , Animals , CHO Cells , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Copper/chemistry , Coumarins/chemistry , Cricetinae , Cricetulus , DNA/biosynthesis , DNA/metabolism , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Neoplasms/metabolism , Neoplasms/pathology , Organometallic Compounds/chemistry , Poly(ADP-ribose) Polymerases/metabolism
7.
J Inorg Biochem ; 101(8): 1108-19, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17555821

ABSTRACT

Two novel coumarin-based ligands, coumarin-6,7-dioxyacetic acid (1) (cdoaH(2)) and 4-methylcoumarin-6,7-dioxyacetic acid (2) (4-MecdoaH(2)), were reacted with copper(II) and manganese(II) salts to give [Cu(cdoa)(H(2)O)(2)].1.5H(2)O (3), [Cu(4-Mecdoa)(H(2)O)(2)] (4), [Mn(cdoa)(H(2)O)(2)] (5) and [Mn(4-Mecdoa)(H(2)O)(2)].0.5H(2)O (6). The metal complexes, 3-6, were characterised by elemental analysis, IR and UV-Vis spectroscopy, and magnetic susceptibility measurements and were assigned a polymeric structure. 1 and 2 react with Cu(II) in the presence of excess 1,10-phenanthroline (phen) giving [Cu(cdoa)(phen)(2)].8.8H(2)O (7) and [Cu(4-Mecdoa)(phen)(2)].13H(2)O (8), respectively. The X-ray crystal structures of 7 and 8 confirmed trigonal bipyramidal geometries, with the metals bonded to the four nitrogen atoms of the two chelating phen molecules and to a single carboxylate oxygen of the dicarboxylate ligand. The complexes were screened for their antimicrobial activity against a number of microbial species, including methicillin-resistant Staphylococcus aureus (MRSA), Escherichia coli and Candida albicans. The metal-free ligands 1 and 2 were active against all of the microbes. Complexes 3-6 demonstrated no significant activity whilst the phen adducts 7 and 8 were active against MRSA (MIC(80)=12.1microM), E. coli (MIC(80)=14.9microM) and Patonea agglumerans (MIC(80)=12.6microM). Complex 7 also demonstrated anti-Candida activity (MIC(80)=22microM) comparable to that of the commercially available antifungal agent ketoconazole (MIC(80)=25microM).


Subject(s)
Anti-Infective Agents/chemical synthesis , Copper , Manganese , Phenanthrolines/chemistry , Umbelliferones/chemical synthesis , Anti-Infective Agents/pharmacology , Candida albicans/drug effects , Candida albicans/growth & development , Candida albicans/metabolism , Chlorates/chemistry , Copper/chemistry , Crystallization , Crystallography, X-Ray , Ergosterol/antagonists & inhibitors , Ergosterol/metabolism , Escherichia coli/drug effects , Escherichia coli/growth & development , Manganese/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Umbelliferones/pharmacology , Water/chemistry
8.
Chem Biol Interact ; 168(2): 143-58, 2007 Jun 30.
Article in English | MEDLINE | ID: mdl-17512508

ABSTRACT

The central objective of the current study was to investigate the potential in vitro anti-proliferative properties of the parent ligand, coumarin-dioxy-acetic acid (cdoaH(2)), and its copper complex, copper-coumarin-dioxyacetic acetate-phenathroline ([Cu(cdoa)(phen)(2)]) using four human-derived model cell lines, two neoplastic and two non-neoplastic. In addition, selected mechanistic studies were carried out using one of the neoplastic-derived model cell lines, Hep-G2. Results obtained show that the complex, rather than the ligand, could alter the proliferation of both human neoplastic renal (A-498) and hepatic (Hep-G2) cells. Furthermore, hepatic non-neoplastic cells (Chang) appeared to be less sensitive. However, this effect was not mirrored in non-neoplastic renal (HK-2) cells, a profile shared with cisplatin. The observed anti-proliferative effect appeared to be concentration- and time-dependant, and could be attributed to the complex, rather than any of the component parts, i.e. 1,10-phenanthroline, the coumarin ligand, or the simple metal salt. Furthermore, the complex was shown to decrease DNA synthesis, but did not intercalate with it. Based on IC(50) values, [Cu(cdoa)(phen)(2)] was shown to be almost six times more potent than cisplatin. Moreover, there was no evidence to show that P-glycoprotein (P-gp)-mediated multi-drug resistance (MDR) was likely to play a role in decreasing the anti-proliferative activity of the complex. Cytological stains, analysis of genomic DNA, and biochemical assays [caspase-3 and -9 and cleaved poly(ADP-ribose)-polymerase protein], suggested that cell death could switch between apoptosis and necrosis, and this effect appeared to be concentration-dependent. Additionally, flow cytometric analysis showed that the complex functioned through an alteration in cell cycle progression. Taken together, [Cu(cdoa)(phen)(2)] has been shown to be a more potent anti-proliferative agent than either the ligand or cisplatin, and is capable of altering key biochemical events leading to the execution of apoptotic and/or necrotic cell death, suggesting that it is worthy of further investigation.


Subject(s)
Apoptosis/drug effects , Copper/pharmacology , Coumarins/pharmacology , Neoplasms/drug therapy , Organometallic Compounds/pharmacology , Phenanthrolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , CHO Cells , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , Cricetulus , DNA Replication/drug effects , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , Humans , Neoplasms/pathology
9.
J Inorg Biochem ; 101(6): 881-92, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17397929

ABSTRACT

Cu(BZA)(2)(EtOH)(0.5) (1) was generated by the reaction of copper(II) hydroxide with benzoic acid (BZAH). [Cu(TBZH)(2)(BZA)](BZA).0.5TBZH.H(2)O (2) and [Cu(2-PyBZIMH)(2-PyBZIM)(BZA)].1.66EtOH (3) were obtained when 1 reacted with Thiabendazole (TBZH) and 2-(2-pyridyl)benzimidazole (2-PyBZIMH), respectively. [Cu(BZA)(2)(phen)(H(2)O)] (4) was isolated from the reaction of benzoic acid and 1,10-phenanthroline (phen) with copper(II)acetate dihydrate. Molecular structures of 2, 3 and 4 were determined crystallographically. 2 and 3 are hydrogen bonded dimers and trimers, respectively. The copper centres in complexes 2 and 3 are bis-chelate derivatives that have N(4)O ligation and their geometry is very similar being approximately square-pyramidal. However whereas in complex 2 both TBZH ligands are neutral in 3 one of the 2-PyBZIMH chelators is deprotonated on each copper. The structural results for 4 represent a re-examination of this crystallographically known compound for which no hydrogen atom coordinates have been previously reported. It crystallises as a hydrogen bonded dimmer and is a mono-chelate of phen with each copper centre possessing N(2)O(3) ligation and square pyramidal geometry. The catalase and superoxide dismutase (SOD) activities of the four complexes along with those of the known phenanthroline complexes [Cu(mal)(phen)(2)] and [Cu(phendione)(3)](ClO(4))(2) (malH(2)=malonic acid and phendione=1,10-phenanthroline-5,6-dione) were investigated. Complexes 1-4, the metal free ligands and a simple copper(II) salt were assessed for their cancer chemotherapeutic potential against the hepatocellular carcinoma (Hep-G(2)) and kidney adenocarcinoma (A-498) cell lines. TBZH, 2-PyBZIMH and benzoic acid when uncoordinated to a metal centre offer poor chemotherapeutic potential. copper(II) benzoate is significantly more active than the free acid. The bis-chelate derivatives [Cu(TBZH)(2)(BZA)](BZA).0.5TBZH.H(2)O (2) and [Cu(2-PyBZIMH)(2-PyBZIM)(BZA)].1.66EtOH (3) elicit a significant cytotoxic response to the cancer cell lines tested. Replacing TBZH and 2-PyBZIMH with phen to give [Cu(BZA)(2)(phen)(H(2)O)] (4) does not significantly increase the anti-cancer activity.


Subject(s)
Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Benzoates/chemistry , Chelating Agents/chemistry , Molecular Mimicry , Nitrogen/chemistry , Phenanthrolines/chemistry , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Humans , Hydrogen Bonding , Ligands , Molecular Structure , Phenanthrolines/pharmacology , Spectrophotometry, Infrared
10.
Toxicol In Vitro ; 21(5): 801-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17350222

ABSTRACT

The anti-fungal activity and mode of action of a range of silver(I)-coumarin complexes was examined. The most potent silver(I)-coumarin complexes, namely 7-hydroxycoumarin-3-carboxylatosilver(I), 6-hydroxycoumarin-3-carboxylatosilver(I) and 4-oxy-3-nitrocoumarinbis(1,10-phenanthroline)silver(I), had MIC80 values of between 69.1 and 4.6 microM against the pathogenic yeast Candida albicans. These compounds also reduced respiration, lowered the ergosterol content of cells and increased the trans-membrane leakage of amino acids. A number of the complexes disrupted cytochrome synthesis in the cell and induced the appearance of morphological features consistent with cell death by apoptosis. These compounds appear to act by disrupting the synthesis of cytochromes which directly affects the cell's ability to respire. A reduction in respiration leads to a depletion in ergosterol biosynthesis and a consequent disruption of the integrity of the cell membrane. Disruption of cytochrome biosynthesis may induce the onset of apoptosis which has been shown previously to be triggered by alteration in the location of cytochrome c. Silver(I)-coumarin complexes demonstrate good anti-fungal activity and manifest a mode of action distinct to that of the conventional azole and polyene drugs thus raising the possibility of their use when resistance to conventional drug has emerged or in combination with such drugs.


Subject(s)
Antifungal Agents , Candida albicans/drug effects , Coumarins/pharmacology , Silver Compounds/pharmacology , Candida albicans/metabolism , Cell Membrane/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Crystallography, X-Ray , Cytochromes/metabolism , DNA, Fungal/isolation & purification , Ergosterol/metabolism , Microbial Sensitivity Tests , Microscopy, Electron , Oxygen Consumption/drug effects , Silver Compounds/chemical synthesis , Silver Compounds/chemistry
11.
Cancer Lett ; 247(2): 224-33, 2007 Mar 18.
Article in English | MEDLINE | ID: mdl-16740357

ABSTRACT

The chemotherapeutic potential of 1,10-phenanthroline (phen), and three of its transition metal complexes, namely [Cu(phen)(2)(mal)]x2H(2)O, [Mn(phen)(2)(mal)]x2H(2)O and [Ag(2)(phen)(3)(mal)]x2H(2)O (malH(2)=malonic acid) was determined using two human carcinoma cell lines (A-498 and Hep-G2). Phen and the three metal-phen complexes induced a concentration-dependent cytotoxic effect, with metal complexes demonstrating the greatest cytotoxic response. In comparative studies, IC(50) values show cytotoxicity of between 3 and 18 times greater than that observed for the metal-based anti-cancer agent, cisplatin. All of the phen-based complexes inhibited DNA synthesis which did not appear to be mediated through intercalation. Also, the potential cancer chemotherapeutic application of these compounds was seen to be enhanced by results obtained from Ames tests, which showed all of the test agents and their phase I metabolites were non-mutagenic. Taken together, these results suggest that phen and the three metal-phen complexes may have a therapeutic role to play in the successful treatment and management of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Malonates/chemistry , Metals/chemistry , Phenanthrolines/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Replication/drug effects , Humans , Mutagenicity Tests , Phenanthrolines/chemistry , Phenanthrolines/pharmacology
12.
Cancer Lett ; 248(2): 321-31, 2007 Apr 18.
Article in English | MEDLINE | ID: mdl-16996681

ABSTRACT

The chemotherapeutic potential of coumarin-3-carboxylic acid (C-3-COOH) and a series of three hydroxylated coumarin-3-carboxylic acid ligands, namely 6-hydroxy-coumarin-3-carboxylic acid (6-OH-C-3-COOH), 7-hydroxy-coumarin-3-carboxylic acid (7-OH-C-3-COOH) and 8-hydroxy-coumarin-3-carboxylic acid (8-OH-C-3-COOH), along with their corresponding silver-based complexes, namely 6-hydroxycoumarin-3-carboxylatosilver (6-OH-C-COO-Ag), 7-hydroxycoumarin-3-carboxylatosilver (7-OH-C-COO-Ag) and 8-hydroxycoumarin-3-carboxylatosilver (8-OH-C-COO-Ag), was determined using two human-derived carcinoma (A-498 and Hep-G2), along with two non-carcinoma human-derived cell lines (CHANG and HK-2). All of the ligands and their silver complexes induced a concentration-dependent cytotoxic effect. Furthermore, hydroxylation of C-3-COOH and its subsequent complexation with silver led to the production of a series of compounds with dramatically enhanced cytotoxicity, with 6-OH-C-3-COO-Ag having the greatest activity. Additionally, all of the metal-based complexes were selectively cytotoxic to both carcinoma-derived cell lines, relative to normal renal and hepatic cells. In comparative studies with cisplatin, and based on the IC(50) values obtained with Hep-G2 cells, it appeared that the coumarin-silver complexes were between 2 and 5.5 times more cytotoxic than cisplatin. All of the coumarin-silver complexes inhibited DNA synthesis, which did not appear to be mediated through intercalation. Furthermore, results obtained from Ames tests showed that all of the test agents and their phase I metabolites were non-mutagenic. Taken together, these findings suggest that both hydroxylation particularly in the 6th position and complexation with silver, served to significantly augment the cytotoxic properties of C-3-COOH, to yield a compound which acts as a cyto-selective agent, as it is a significant killer of cancer, relative to normal cells. We suggest that this group of compounds may have a therapeutic role to play in the successful treatment and management of cancer in man.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Coumarins/pharmacology , Silver Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Coumarins/chemistry , DNA/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Electrophoretic Mobility Shift Assay , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Mutagenicity Tests , Silver Compounds/chemical synthesis , Silver Compounds/chemistry
13.
Cancer Lett ; 250(1): 128-39, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17126993

ABSTRACT

Previously our research group has studied the anti-proliferative effects of a series of hydroxylated derivatives and silver (I) complexes of coumarin-3-carboxylic acid (C-3-COOH) using two human-derived carcinoma cell lines (A-498 and Hep-G2). Results obtained suggested that both hydroxylation and complexation with silver served to significantly augment the cytotoxic properties of C-3-COOH, to yield a compound, namely 6-hydroxycoumarin-3-carboxylatosilver (6-OH-C-COO-Ag) which could act as a potent and cyto-selective agent, capable of killing cancer cells, and with limited toxicity to cells derived from normal tissue. Here we seek to expand on these findings by probing the molecular mechanism underlying this effect. Results from cytological staining clearly illustrated cellular changes consistent with the induction of apoptotic cell death and which occurred 24 h post-drug-treatment. Additionally, electrophoretic analysis of genomic DNA showed the presence of a ladder pattern, characteristic of apoptotic cell death. This result was subsequently confirmed using a selection of biochemical assays, where increased activity of pro-apoptotic caspases 3 and 9, and increased cleavage of poly(ADP-ribose)-polymerase protein (PARP) were observed. This result was further underpinned by the appearance of a sub-G(1) peak, representing hypo-diploid cells, using flow cytometric analysis. Furthermore, 6-OH-C-COO-Ag was seen to function through an alteration in the percentage of cells entering the G(0)/G(1) phase of cell cycle. Consequently, 6-OH-C-COO-Ag has been shown to a more potent and selective anti-cancer agent than cisplatin, capable of altering key biochemical events leading to the execution of apoptotic cell death as early as 24 h post-treatment, suggesting that it may represent a novel therapeutic agent for the safe and effective treatment of cancer in man.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Cycle/drug effects , Cell Death/drug effects , Liver Neoplasms/drug therapy , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Cells, Cultured , Coumarins/pharmacology , DNA Fragmentation , Dose-Response Relationship, Drug , Humans , Liver/drug effects , Liver Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Tumor Cells, Cultured
14.
Chem Biol Interact ; 164(1-2): 115-25, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17030029

ABSTRACT

The anti-cancer chemotherapeutic potential of 1,10-phenanthroline-5,6-dione (phendione), [Cu(phendione)(3)](ClO(4))(2).4H(2)O and [Ag(phendione)(2)]ClO(4) were determined using four human cells lines, i.e. two neoplastic (A-498 and Hep-G2) and two non-neoplastic (CHANG and HK-2). All of the phendione derivatives induced a concentration-dependant decrease in the viability of the four cell lines, with [Cu(phendione)(3)](ClO(4))(2).4H(2)O displaying greatest activity. In comparative studies, IC(50) values obtained with the two neoplastic cell lines showed a cytotoxic response which was between 3 and 35 times greater than that observed for the metal-based anti-cancer agent, cisplatin. Furthermore, metal-phendione complexes, rather than simple solvated metal ions, were responsible for the observed cytotoxicity. Despite the high level of potency associated with these compounds they did not display an apparent cyto-selective profile, as they reduced the viability of both neoplastic and non-neoplastic cells. However, selected mechanistic studies showed that phendione and its metal complexes inhibited DNA synthesis which did not appear to be mediated through intercalation. Ames testing highlighted that all three compounds and their phase I metabolites were non-mutagenic, unlike cisplatin. Taken together, these results suggest that phendione and its Cu(II) and Ag(I) complexes may be capable of acting as highly effective anti-cancer therapies, which with careful administration could provide very potent and effective alternatives to cisplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/pharmacology , Epithelial Cells , Organometallic Compounds/pharmacology , Perchlorates/pharmacology , Phenanthrolines/pharmacology , Silver Compounds/pharmacology , Cell Line/drug effects , Cisplatin/pharmacology , DNA/antagonists & inhibitors , DNA/biosynthesis , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Mutagenicity Tests
15.
Bioinorg Chem Appl ; : 80283, 2006.
Article in English | MEDLINE | ID: mdl-17497019

ABSTRACT

2,2-dimethylpentanedioic acid (2dmepdaH(2)) and 3,3-dimethylpentanedioic acid (3dmepdaH(2)) reacted with copper(II) acetate to give [Cu(2dmepda)(H(2)O)(3)](2) (1) and [Cu(3dmepda)(H(2)O)(3)](2) (2). Reaction of (1) and (2) with 1,10-phenanthroline and 2,2'-bipyridine yielded [Cu(2dmepda)(phen)(H(2)O)](2)0.5phen (3), [Cu(2dmepda)(bipy)(H(2)O)](2) (4), [Cu(2dmepda)(bipy)(EtOH)](2). 2EtOH (4A), [Cu(3dmepda)(phen)(H(2)O)](2) (5), and [Cu(3dmepda)(bipy)(H(2)O)](2). (6). The structures of (4A) and (6) each consists of a [Cu(bipy)(dicarboxylate)(solvent)](2) dimer. The superoxide dismutase (SOD) mimetic activity of the novel copper complexes and their manganese analogues was investigated. The dimethyl sulphoxide(DMSO) soluble complexes (1)-(4) and (6) were assessed for their cancer chemotherapeutic potential towards hepatocellular carcinoma and kidney adenocarcinoma cell lines. The 1,10-phenanthroline containing complex [Cu(2dmepda)(phen)(H(2)O)](2)0.5phen (3) was the most potent with activity that compares well to that of cisplatin.

16.
Eur J Pharm Sci ; 26(1): 16-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15996858

ABSTRACT

6-Nitro-7-hydroxycoumarin (6-NO2-7-OHC) and 3,6,8-trinitro-7-hydroxycoumarin (3,6,8-NO2-7-OHC) have previously been shown to be potent and selective anti-proliferative agents in a human melanoma cell line. These agents functioned by decreasing DNA synthesis, through an inhibition of the S phase regulatory protein, cyclin A. However, the key molecular target(s) for these drugs remained undefined. Here, we attempted to elucidate the exact nature of the relationship between drug exposure and signal transduction, particularly their effects on the mitogen activated protein kinase (MAPK) cascades, and the consequent effect on cell growth, death and differentiation. Comparative studies were carried out using 7-hydroxycoumarin (7-OHC). Both nitro-derivatives were found to alter the phosphorylation status of ERK1/ERK2 and p38. However, 7-OHC exerted this effect only at higher concentrations and longer incubation times. Also, none of the three drugs had any effect on SAPK phosphorylation. Tyrosinase activity assays and morphological studies were used to show drug-induced effects on cellular differentiation. Unlike 7-OHC, both 6-NO2-7-OHC and 3,6,8-NO2-7-OHC caused a dramatic increase in tyrosinase activity in a manner similar to the cAMP elevating agent, forskolin. Also, the MEK inhibitor (PD98059) in combination with nitro-derivatives stimulated an even greater increase in tyrosinase activity when compared to either drug. In addition, the p38 inhibitor (SB203580) reduced the activity of both drugs. Morphological examination of treated cells showed nitro-derivatives caused changes consistent with altered cellular differentiation. Taken together, we have established that exposure of human malignant melanoma cells to these drugs leads to a modulation of p38 MAP kinase phosphorylation. This implies that these drugs may function by altering both melanogenesis and cellular differentiation. However, their effect on the levels of these proteins rather than their phosphorylation status remains to be determined. Therefore, additional studies are underway in order to identify the exact binding partners for these drugs.


Subject(s)
Antineoplastic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Melanins/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Nitro Compounds/pharmacology , Umbelliferones/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor/drug effects , Colforsin/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Melanoma , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Pyridines/pharmacology
17.
Cancer Lett ; 214(1): 43-54, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15331172

ABSTRACT

6-Nitro-7-hydroxycoumarin (6-NO2-7-OHC) and 3,6,8-trinitro-7-hydroxycoumarin (3,6,8-NO2-7-OHC) have previously been shown to be potent and selective anti-proliferative agents to the human skin cell line, SK-MEL-31. Here, we investigate the reversibility of their cytotoxicity, along with their effects on DNA synthesis and cell cycle events. Comparative studies were carried out using the main metabolite of coumarin in man, 7-hydroxycoumarin (7-OHC). 6-NO2-7-OHC and 3,6,8-NO2-7-OHC, were found to be irreversible cytotoxic agents, unlike 7-OHC. All three derivatives inhibited DNA synthesis, but 7-OHC was only nitro-derivatives which acted in an irreversible manner. Flow cytometric studies demonstrated that both nitro-derivatives caused a dose- and time-dependant S phase accumulation. 7-OHC exerted a similar effect, but appeared to be less potent. Finally, the two nitro-derivatives caused a dose-dependant inhibition of the S phase regulatory protein, cyclin A. Consequently, these and other nitro-derivatives of 7-OHC may represent novel therapeutic agents for the treatment of malignant melanoma as they are capable of selective and irreversible cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Cycle/drug effects , DNA/biosynthesis , Melanoma/pathology , Nitro Compounds/pharmacology , Nitro Compounds/toxicity , Skin Neoplasms/pathology , Umbelliferones/pharmacology , Umbelliferones/toxicity , Cell Cycle/physiology , Cyclin A/biosynthesis , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Tumor Cells, Cultured
18.
J Inorg Biochem ; 98(8): 1361-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15271512

ABSTRACT

[Ag(2)(NH(3))(2)(salH)(2)] (salH(2)=salicylic acid) was synthesised from salicylic acid and Ag(2)O in concentrated aqueous NH(3) and the dimeric Ag(I) complex was characterised using X-ray crystallography. The complex is centrosymmetric with each metal coordinated to a salicylate carboxylate oxygen and to an ammonia nitrogen atom in an almost linear fashion. The two [Ag(NH(3))(salH)] units in the complex are linked by an Ag-Ag bond. Whilst metal-free salH(2) did not prevent the growth of the fungal pathogen Candida albicans [Ag(2)(NH(3))(2)(salH)(2)], [Ag(2)(salH)(2)] and some simple Ag(I) salts greatly inhibited cell reproduction. SalH(2), [Ag(2)(NH(3))(2)(salH)(2)] [Ag(2)(salH)(2)] and AgClO(4) produced a dose-dependent cytotoxic response against the three human derived cancer cell lines, Cal-27, Hep-G2 and A-498, with the Ag(I)-containing reagents being the most effective.


Subject(s)
Antifungal Agents , Antineoplastic Agents , Salicylic Acid , Silver Compounds , Animals , Antifungal Agents/chemistry , Antifungal Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Candida albicans/metabolism , Candidiasis/drug therapy , Cell Line, Tumor , Crystallography, X-Ray , Humans , Molecular Structure , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Salicylic Acid/chemistry , Salicylic Acid/therapeutic use , Silver Compounds/chemistry , Silver Compounds/therapeutic use
19.
J Inorg Biochem ; 98(6): 1023-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15149811

ABSTRACT

Thiabendazole (TBZH) reacts with iron(III) nitrate causing protonation of the ligand to yield the nitrate salt [TBZH(2)NO(3)] (1). Reaction of TBZH with copper(II) acetate results in the deprotonation of the ligand yielding [Cu(TBZ)2.(H2O)2] (2). Reactions of TBZH with the chloride, nitrate and butanedioate salts of copper(II) yields [Cu(TBZH)2Cl]Cl.H2O.EtOH (3), [Cu(TBZH)(2)(NO(3))(2)] (4) and [Cu(TBZH)(O(2)C-CH(2)CH(2)-CO(2))] (5), respectively. The TBZH acts as a neutral chelating ligand in 3-5. Molecular structures of 1 and 3 were determined crystallographically. In 1, the asymmetric unit contains one TBZH(2)(+) cation and one NO(3)(-) anion. The structure of 3 comprises a five coordinate copper centre with the metal bound to two chelating TBZH ligands and one chloride. The geometry is best described as trigonal bipyramidal. Hydrogen bonding connects the complex cation with the uncoordinated chloride anion and the water and ethanol solvate molecules. Compound 1 and the copper complexes 2-5, the metal free ligands and a number of simple copper(II) salts were each tested for their ability to inhibit the growth of Candida albicans. The metal free TBZH and its nitrate salt (1) exhibited very poor activity. Complex 2, in which the TBZH is present as an anionic ligand (TBZ(-)), exhibits moderate activity towards the pathogen. Chelation of the neutral TBZH to copper centres (complexes 3-5) results in potent anti-candida activity. The dimethyl sulphoxide (DMSO) soluble complexes 3 and 4, along with metal free TBZH were assessed for their cancer chemotherapeutic potential towards two human epithelial-derived cancer model cell lines. Complexes 3 and 4 displayed similar dose-dependent cytotoxicity in both cell lines with IC(50) values of approximately 50 microM, which were found to be significantly lower than that for metal free TBZH.


Subject(s)
Antifungal Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Thiabendazole/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/toxicity , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Candida albicans/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Copper/chemistry , Humans , Organometallic Compounds/toxicity , Thiabendazole/analogs & derivatives , Thiabendazole/toxicity
20.
Biochem Pharmacol ; 67(9): 1779-88, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15081877

ABSTRACT

Daphnetin has been shown to be a potent in vitro anti-proliferative agent to the human renal cell carcinoma (RCC) cell line, A-498. In the present study, we investigated its effects on mitogen-activated protein kinase (MAPK) signalling along with cell cycle events and cellular differentiation. Daphnetin-activated p38, however, higher concentrations were required to inhibit ERK1/ERK2. In addition, it did not activate SAPK or induce apoptosis, but instead inhibited S phase cell cycle transition of A-498 cells at low concentrations and time of exposure. In addition, a late G(1), early S phase inhibition was observed at higher concentrations and time of exposure, indicating that the mechanism of daphnetin-induced differentiation was concentration dependent. Increased expression of the epithelial differentiation markers cytokeratins 8 and 18, correlated with increasing concentrations of daphnetin, while pre-treatment with a specific p38-inhibitor, served to limit this effect. There was no evidence that P-glycoprotein (P-gp) mediated multi-drug resistance (MDR) played a role in the anti-proliferative activity of daphnetin. Consequently, we concluded that p38 MAP kinase is intrinsically involved in mediating the effect of daphnetin in A-498 cells, suggesting that this drug may act by promotion of cellular maturation, and consequently may represent a novel low toxic approach for the treatment of poorly differentiated RCCs.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Differentiation/drug effects , Mitogen-Activated Protein Kinases/physiology , Umbelliferones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Cycle/drug effects , Drug Resistance, Multiple/drug effects , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...