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










Publication year range
1.
J Med Chem ; 64(5): 2678-2690, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33621096

ABSTRACT

A glucose-based vector for targeting cancer cells conjugated to a tris(methylpyridyl)amine (tpa) ligand to generate targeted chaperone and caging complexes for active anticancer agents is described. The ligand, tpa(CONHPEGglucose)1, inhibits hexokinase, suggesting that it will be phosphorylated in the cell. A Co(III) complex incorporating this ligand and coumarin-343 hydroximate (C343ha), [Co(C343ha){tpa(CONHPEGglucose)1}]Cl, is shown to exhibit glucose-dependent cellular accumulation in DLD-1 colon cancer cells. Cellular accumulation of [Co(C343ha){tpa(CONHPEGglucose)1}]+ is slower than for the glucose null and glucosamine analogues, and the glucose complex also exhibits a lower ability to inhibit antiproliferative activity. Distributions of cobalt (X-ray fluorescence) and C343ha (visible light fluorescence) in DLD-1 cancer cell spheroids are consistent with uptake of [Co(C343ha){tpa(CONHPEGglucose)1}]+ by rapidly dividing cells, followed by release and efflux of C343ha and trapping of the Co{tpa(CONHPEGglucose)1} moiety. The Co{tpa(CONHPEGglucose)1} moiety is shown to have potential for the caged and targeted delivery of highly toxic anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Warburg Effect, Oncologic/drug effects , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cobalt/chemistry , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hexokinase/antagonists & inhibitors , Humans , Spheroids, Cellular/drug effects
2.
Metallomics ; 12(10): 1599-1615, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33084707

ABSTRACT

cis- and trans-Platinum(iv) complexes with diaminetetracarboxylate coordination spheres possess the highly desirable property of exhibiting unusual resistance to reduction by blood serum components and endogenous reductants such as ascorbate. At the same time they are rapidly reduced in the intracellular environment of cancer cells. Consequently, they can potentially be tuned to remain intact in vivo until arrival at the tumour target where they are rapidly reduced to yield the active platinum(ii) species. However, in order to achieve this, uptake must be largely restricted to tumour cells and therefore uptake by healthy cells including red blood cells must be prevented. In this proof of concept study, we report on the effect of net charge as a means of controlling the uptake by red blood cells. Using 1H NMR spectroscopy we found that modifying the net charge of the complex does not influence the rate of reduction of the complexes by an excess of ascorbate. Using XANES spectroscopy we found that modifying the net charge of the platinum(iv) complexes decreased the extent of reduction in whole blood, although probably not to the degree needed for the optimal delivery to tumours. Therefore, it is likely to be necessary to adopt higher charges and/or additional strategies to keep platinum(iv) prodrugs out of blood cells.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Ascorbic Acid/metabolism , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacokinetics , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/pharmacology , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Serum/metabolism , Static Electricity
3.
Chem Sci ; 11(10): 2627-2639, 2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32206266

ABSTRACT

There is a dire need for new antimicrobial compounds to combat the growing threat of widespread antibiotic resistance. With a currently very scarce drug pipeline, consisting mostly of derivatives of known antibiotics, new classes of antibiotics are urgently required. Metal complexes are currently in clinical development for the treatment of cancer, malaria and neurodegenerative diseases. However, only little attention has been paid to their application as potential antimicrobial compounds. We report the evaluation of 906 metal-containing compounds that have been screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD) for antimicrobial activity. Metal-bearing compounds display a significantly higher hit-rate (9.9%) when compared to the purely organic molecules (0.87%) in the CO-ADD database. Out of 906 compounds, 88 show activity against at least one of the tested strains, including fungi, while not displaying any cytotoxicity against mammalian cell lines or haemolytic properties. Herein, we highlight the structures of the 30 compounds with activity against Gram-positive and/or Gram-negative bacteria containing Mn, Co, Zn, Ru, Ag, Eu, Ir and Pt, with activities down to the nanomolar range against methicillin resistant S. aureus (MRSA). 23 of these complexes have not been reported for their antimicrobial properties before. This work reveals the vast diversity that metal-containing compounds can bring to antimicrobial research. It is important to raise awareness of these types of compounds for the design of truly novel antibiotics with potential for combatting antimicrobial resistance.

5.
Chembiochem ; 20(22): 2876-2882, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31102568

ABSTRACT

Cancer is currently one of the deadliest diseases worldwide. Based on the high incidence of this disease, the side effects associated with current chemotherapies and the appearance of drug resistance, considerable efforts have been directed towards the development of new anticancer drugs with new modes of action. Metal-based compounds are particularly attractive candidates due to their metabolic mechanisms, which differ substantially from those of organic drugs. Of special interest in this context are organometallic ruthenium(II) complexes of the type [Ru(η6 -arene)(pta)Cl2 ] (arene: p-cymene, toluene, benzene, etc.; pta: 1,3,5-triaza-7-phosphaadamantane), which are abbreviated to RAPTA. Complementary to chemotherapy, photoactivated chemotherapy is a technique that has received increasing attention towards the development of treatment for numerous kinds of cancer. With this in mind, a photoactive RAPTA-type complex bearing azide ligands has been designed. The diazide complex, [Ru(η6 -p-cymene)pta-(N3 )2 ], is inert in water, but slowly releases the azide ligand upon exposure to light. Consequently, the in vitro cytotoxicity of the complex in the dark and upon light exposure at λ=450 nm in human cervical carcinoma (HeLa) and noncancerous retinal pigment epithelium (RPE-1) cells was investigated. Although the cytotoxicity of the complex was found to be modest in the dark, an increase in toxicity upon light exposure was observed.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Prodrugs/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , Coordination Complexes/chemical synthesis , Coordination Complexes/radiation effects , Coordination Complexes/toxicity , Drug Screening Assays, Antitumor , Epithelial Cells/drug effects , HeLa Cells , Humans , Prodrugs/chemical synthesis , Prodrugs/radiation effects , Prodrugs/toxicity , Retinal Pigment Epithelium/cytology , Ruthenium/chemistry , Ultraviolet Rays
6.
J Inorg Biochem ; 179: 146-153, 2018 02.
Article in English | MEDLINE | ID: mdl-29180165

ABSTRACT

CHS-828 (N-(6-(4-chlorophenoxy)hexyl)-N'-cyano-N″-4-pyridyl guanidine) is an anticancer agent with low bioavailability and high systemic toxicity. Here we present an approach to improve the therapeutic profile of the drug using photolabile ruthenium complexes to generate light-activated prodrugs of CHS-828. Both prodrug complexes are stable in the dark but release CHS-828 when irradiated with visible light. The complexes are water-soluble and accumulate in tumour cells in very high concentrations, predominantly in the mitochondria. Both prodrug complexes are significantly less cyototoxic than free CHS-828 in the dark but their toxicity increases up to 10-fold in combination with visible light. The cellular responses to light treatment are consistent with release of the cytotoxic CHS-828 ligand.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Cyanides/pharmacology , Guanidines/pharmacology , Prodrugs/pharmacology , Ruthenium/chemistry , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Coordination Complexes/chemical synthesis , Coordination Complexes/radiation effects , Cyanides/radiation effects , DNA/chemistry , Guanidines/radiation effects , Humans , Inhibitory Concentration 50 , Light , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Prodrugs/chemical synthesis , Prodrugs/radiation effects , Reactive Oxygen Species/metabolism
7.
Dalton Trans ; 46(45): 15897-15907, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29116280

ABSTRACT

Cobalt(iii) chaperones are a promising class of bioreductive prodrugs under investigation for the delivery of cytotoxic ligands to hypoxic solid tumours. Here we investigate a series of cobalt complexes as chaperones for hydroxamic acid ligands, comparing the properties of the cyclic cyclen (1,4,7,10-tetraazacyclododecane) ancillary ligand with the tripodal tpa (tris-(2-pyridylmethyl)amine) and tren (tris-(2-aminoethyl)amine). A small library of complexes containing several different hydroxamic acids, including the MMP inhibitor Marimistat and the fluorescent ligand C343haH2, were prepared and their pKa values, reduction potentials, and in some cases X-ray crystal structures, were determined. The antiproliferative actitivity of the series was evaluated against DLD-1 colon cancer cells and the cellular accumulation of the fluorescent C343haH2 complexes was monitored by ICPMS and confocal fluorescence microscopy, revealing that the nature of the ancillary ligand significantly influences the complexes' properties, cytotoxicity and cellular distribution.


Subject(s)
Cobalt/chemistry , Coordination Complexes/chemistry , Hydroxamic Acids/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/toxicity , Crystallography, X-Ray , Cyclams , Electrochemical Techniques , Heterocyclic Compounds/chemistry , Humans , Hydroxamic Acids/metabolism , Kinetics , Ligands , Microscopy, Confocal , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Conformation , Oxidation-Reduction , Spectrometry, Fluorescence
8.
Inorg Chem ; 56(16): 9860-9868, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28766939

ABSTRACT

Dense tumors are resistant to conventional chemotherapies due to the unique tumor microenvironment characterized by hypoxic regions that promote cellular dormancy. Bioreductive drugs that are activated in response to this hypoxic environment are an attractive strategy for therapy with anticipated lower harmful side effects in normoxic healthy tissue. Cobalt bioreductive pro-drugs that selectively release toxic payloads upon reduction in hypoxic cells have shown great promise as anticancer agents. However, the bioreductive response in the tumor microenvironment must be better understood, as current techniques for monitoring bioreduction to Co(II) such as X-ray absorption near-edge structure and extended X-ray absorption fine structure provide limited information on speciation and require synchrotron radiation sources. Here, we present magnetic resonance imaging (MRI) as an accessible and powerful technique to monitor bioreduction by treating the cobalt complex as an MRI contrast agent and monitoring the change in water signal induced by reduction from diamagnetic Co(III) to paramagnetic Co(II). Cobalt pro-drugs built upon the tris(2-pyridylmethyl)amine ligand scaffold with varying charge were investigated for distribution and activity in a 3D tumor spheroid model by laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) and MRI. In addition, paramagnetic 1H NMR spectroscopy of spheroids enabled determination of the speciation of activated Co(II)TPAx complexes. This study demonstrates the utility of MRI and associated spectroscopy techniques for understanding bioreductive cobalt pro-drugs in the tumor microenvironment and has broader implications for monitoring paramagnetic metal-based therapies.


Subject(s)
Cobalt/chemistry , Contrast Media/pharmacology , Coordination Complexes/pharmacology , Prodrugs/pharmacology , Spheroids, Cellular/physiology , Animals , Cattle , Contrast Media/chemical synthesis , Coordination Complexes/chemical synthesis , Humans , Ligands , Mass Spectrometry/methods , Prodrugs/chemical synthesis , Proton Magnetic Resonance Spectroscopy/methods , Sheep , Tumor Cells, Cultured , Tumor Hypoxia/physiology , Water/chemistry
9.
Chimia (Aarau) ; 71(3): 112-119, 2017 Mar 29.
Article in English | MEDLINE | ID: mdl-28351456

ABSTRACT

Metal-based drugs with novel targets and modes of action are increasingly being developed as alternatives to classical platinum(ii) chemotherapeutics. Imaging methods in tumour cells and tissues offer valuable insights into the behaviour of these novel complexes; however, mapping the distribution of metal ions and complexes within cellular environments remains challenging. The advantages and limitations of three modes of imaging: synchrotron radiation-induced X-ray fluorescence, mass spectrometry, and fluorescence microscopy are discussed in this review, with particular emphasis on their use in imaging ruthenium-based drugs.


Subject(s)
Mass Spectrometry/methods , Microscopy, Fluorescence/methods , Pharmaceutical Preparations/analysis , Ruthenium/pharmacokinetics , Spectrometry, X-Ray Emission/methods , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Humans , Lasers , Pharmaceutical Preparations/chemistry , Ruthenium/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Synchrotrons
10.
Chemistry ; 21(43): 15224-34, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26471438

ABSTRACT

Light-activated prodrugs offer the potential for highly selective tumour targeting. However, the application of many photoactivated chemotherapeutics is limited by a requirement for oxygen, or for short activation wavelengths that can damage surrounding tissue. Herein, we present a series of cobalt(III)-curcumin prodrugs that can be activated by visible light under both oxygenated and hypoxic conditions. Furthermore, the photoproduct can be controlled by the activation wavelength: green light yields free curcumin, whereas blue light induces photolysis of curcumin to a phototoxic product. Confocal fluorescence microscopy and phototocytotoxicity studies in DLD-1 and MCF-7 tumour cells demonstrated that the cobalt(III) prodrugs are nontoxic in the dark but accumulate in significant concentrations in the cell membrane. When cells were treated with light for 15  min, the cytotoxicity of the cobalt complexes increased by up to 20-fold, whereas free curcumin exhibited only a two-fold increase in cytotoxicity. The nature of the ancillary ligand and cobalt reduction potential were found to strongly influence the stability and biological activity of the series.


Subject(s)
Cobalt/chemistry , Curcumin/chemistry , Prodrugs/chemistry , Cell Hypoxia , Cell Line, Tumor , Cobalt/pharmacology , Curcumin/pharmacology , Humans , Ligands , Light , Prodrugs/pharmacology
11.
Chem Commun (Camb) ; 51(74): 14038-41, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26248575

ABSTRACT

We report a novel ruthenium(II) complex for selective release of the imidazole-based drug econazole. While the complex is highly stable and luminescent in the dark, irradiation with green light induces release of one of the econazole ligands, which is accompanied by a turn-off luminescence response and up to a 34-fold increase in cytotoxicity towards tumour cells.


Subject(s)
Cell Tracking/methods , Coordination Complexes/chemistry , Drug Delivery Systems/methods , Light , Luminescent Agents/chemistry , Ruthenium/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Drug Liberation , Econazole/administration & dosage , Econazole/pharmacokinetics , Econazole/pharmacology , Humans , Microscopy, Fluorescence , Molecular Structure , Photochemotherapy , Spectrophotometry, Atomic
12.
Metallomics ; 6(8): 1324-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850462

ABSTRACT

The unique properties of transition metal complexes, such as environment-responsive ligand exchange kinetics, diverse photochemical and photophysical properties, and the ability to form specific interactions with biomolecules, make them interesting platforms for selective drug delivery. This minireview will focus on recent examples of rationally designed complexes with bioactive ligands, exploring the different roles of the metal, and mechanisms of ligand release. Developments in the techniques used to study the mechanisms of action of metal-drug complexes will also be discussed, including X-ray protein crystallography, fluorescence lifetime imaging, and X-ray absorption spectroscopy.


Subject(s)
Coordination Complexes/chemistry , Transition Elements/chemistry , Crystallography, X-Ray , Drug Delivery Systems , Ligands , Molecular Structure , X-Ray Absorption Spectroscopy
13.
J Med Chem ; 55(24): 11013-21, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23199008

ABSTRACT

The rational design of prodrugs for selective accumulation and activation in tumor microenvironments is one of the most promising strategies for minimizing the toxicity of anticancer drugs. Manipulation of the charge of the prodrug represents a potential mechanism to selectively deliver the prodrug to the acidic tumor microenvironment. Here we present delivery of a fluorescent coumarin using a cobalt(III) chaperone to target hypoxic regions, and charged ligands for pH selectivity. Protonation or deprotonation of the complexes over a physiologically relevant pH range resulted in pH dependent accumulation of the fluorophore in colon cancer cells. Furthermore, in a spheroid solid tumor model, the anionic complexes exhibited preferential release of the fluorophore in the acidic/hypoxic region. By fine-tuning the physicochemical properties of the cobalt-chaperone moiety, we have demonstrated selective drug release in the acidic and hypoxic tumor microenvironment.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cobalt , Coordination Complexes/chemical synthesis , Coumarins/chemical synthesis , Prodrugs/chemical synthesis , Tumor Microenvironment , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Hypoxia , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coumarins/chemistry , Coumarins/pharmacology , Drug Screening Assays, Antitumor , Fluorescent Dyes , Humans , Hydrogen-Ion Concentration , Hydroxamic Acids/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Structure-Activity Relationship
14.
Dalton Trans ; 40(5): 1158-67, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21165516

ABSTRACT

Chelating neutral (N,O) and cationic (N,N) first- and second-generation ruthenium(II) arene metallodendrimers based on poly(propyleneimine) dendrimer scaffolds were obtained from dinuclear arene ruthenium precursors by reactions with salicylaldimine and iminopyridyl dendritic ligands, respectively. The N,N cationic complexes were isolated as hexafluorophosphate salts and were characterised by NMR and IR spectroscopy, and MALDI-TOF mass spectrometry. Related mononuclear complexes were obtained in a similar manner and their molecular structures have been determined by X-ray diffraction analysis. The cytotoxicities of the mono- and multinuclear complexes were established using A2780 and A2780cisR human ovarian carcinoma cancer cell lines.


Subject(s)
Cell Proliferation/drug effects , Chelating Agents/pharmacology , Dendrimers/pharmacology , Polypropylenes/chemistry , Ruthenium Compounds/chemistry , Animals , Cell Line , Mice
15.
Dalton Trans ; 39(35): 8248-55, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20689885

ABSTRACT

The self-assembly of 2,4,6-tris(pyridin-4-yl)-1,3,5-triazine (tpt) triangular panels with p-cymene (p-Pr(i)C(6)H(4)Me) ruthenium building blocks and 2,5-dioxydo-1,4-benzoquinonato (dobq) bridges, in the presence of a functionalised pyrenyl derivative (pyrene-R), affords the triangular prismatic host-guest compounds [(pyrene-R) [symbol: see text] Ru(6)(p-Pr(i)C(6)H(4)Me)(6)(tpt)(2)(dobq)(3)](6+) ([(pyrene-R) [symbol: see text] 1](6+)). The inclusion of eight mono-substituted pyrenyl derivatives including biologically relevant structures (a = 1-pyrenebutyric acid, b = 1-pyrenebutanol, c = 1-pyrenemethylamine, d = 1-pyrenemethylbutanoate, e = 1-(4,6-dichloro-1,3,5-triazin-2-yl)pyrene, f = N-hexadecylpyrene-1-sulfonamide, g = pyrenyl ethacrynic amide and h = 2-(pyren-1-ylmethylcarbamoyl) phenyl acetate), and a di-substituted pyrenyl derivative (i = 1,8-bis(3-methyl-butyn-1-yl-3-ol)pyrene), has been accomplished. The carceplex nature of these systems with the pyrenyl moiety being firmly encapsulated in the hydrophobic cavity of the cage with the functional groups pointing outwards was confirmed by NMR ((1)H, 2D, DOSY) spectroscopy and electrospray ionization mass spectrometry (ESI-MS). The cytotoxicities of these water-soluble compounds have been established using human ovarian A2780 cancer cells. All the host-guest systems are more cytotoxic than the empty cage itself [1][CF(3)SO(3)](6) (IC(50) = 23 microM), the most active carceplex [f [symbol: see text] 1][CF(3)SO(3)](6) is an order of magnitude more cytotoxic.


Subject(s)
Antineoplastic Agents/chemistry , Pyrenes/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Magnetic Resonance Spectroscopy , Pyrenes/administration & dosage , Pyrenes/toxicity , Ruthenium/chemistry , Spectrometry, Mass, Electrospray Ionization , Water/chemistry
17.
J Biol Inorg Chem ; 15(6): 919-27, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20364440

ABSTRACT

The anticancer ruthenium-arene compound [Ru(eta(6)-C(6)H(5)CF(3))(pta)Cl(2)] (where pta is 1,3,5-triaza-7-phosphatricyclo[3.3.1.1]decane), termed RAPTA-CF3, with the electron-withdrawing alpha,alpha,alpha-trifluorotoluene ligand, is one of the most cytotoxic RAPTA compounds known. To rationalize the high observed cytotoxicity, the hydrolysis of RAPTA-CF3 in water and brine (100 mM sodium chloride) and its reactions with the protein ubiquitin and a double-stranded oligonucleotide (5'-GTATTGGCACGTA-3') were studied using NMR spectroscopy, high-resolution Fourier transform ion cyclotron resonance mass spectrometry, and gel electrophoresis. The aquation of the ruthenium-chlorido complex was accompanied by a loss of the arene ligand, independent of the chloride concentration, which is a special property of the compound not observed for other ruthenium-arene complexes with relatively stable ruthenium-arene bonds. Accordingly, the mass spectra of the biomolecule reaction mixtures contained mostly [Ru(pta)]-biomolecule adducts, whereas [Ru(pta)(arene)] adducts typical of other RAPTA compounds were not observed in the protein or DNA binding studies. Gel electrophoresis experiments revealed a significant degree of decomposition of the oligonucleotide, which was more pronounced in the case of RAPTA-CF3 compared with RAPTA-C. Consequently, facile arene loss appears to be responsible for the increased cytotoxicity of RAPTA-CF3.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Hydrocarbons, Aromatic/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Antineoplastic Agents/pharmacology , Base Sequence , Electrons , Hydrolysis , Ligands , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Organometallic Compounds/pharmacology , Toluene/analogs & derivatives , Toluene/chemistry
18.
Inorg Chem ; 49(5): 2239-46, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20131860

ABSTRACT

A series of compounds of general formula [Ru(eta(6)-arene)(pta)(PR(3))Cl]BF(4) (arene = p-cymene or 4-phenyl-2-butanol; pta = 1,3,5-triaza-7-phosphatricyclo[3.3.1.1]decane, PR(3) = PPh(2)(p-C(6)H(4)C(2)H(4)C(8)F(17)), PPh(p-C(6)H(4)C(2)H(4)C(8)F(17))(2), P(p-C(6)H(4)C(2)H(4)C(6)F(13))(3), PPh(3) or P(p-C(6)H(4)F)(3)) have been prepared and characterized by spectroscopic methods. The structure of [Ru(eta(6)-p-cymene)(pta)Cl(P(p-C(6)H(4)F)(3))]BF(4) has also been established in the solid state by X-ray crystallography. The cytotoxicities of the compounds were determined in the A2780 and A2780 cisplatin-resistant cell lines revealing that the fluorinated phosphines significantly increase antiproliferative activity relative to their bis-chloride precursors. Two of the complexes were found to be thermoresponsive, that is, showing poor water solubility at 37 degrees C and good solubility at 42 degrees C, the temperature of a heated tumor, providing a method of tumor targeting. Incubation at 42 degrees C for 2 h resulted in improved cytotoxicities for two of the complexes.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Halogenation , Hot Temperature , Neoplasms/drug therapy , Phosphines/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Neoplasms/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use , Ruthenium/chemistry , Solubility , Water/chemistry
19.
Inorg Chem ; 48(17): 8233-44, 2009 Sep 07.
Article in English | MEDLINE | ID: mdl-19655717

ABSTRACT

Electrochemical and spectroelectrochemical techniques were employed to study in detail the formation and so far unreported spectroscopic properties of soluble electroactive molecular chains with nonbridged metal-metal backbones, namely, [{Ru(0)(CO)(PrCN)(bpy)}(m)](n) (m = 0, -1) and [{Ru(0)(CO)(bpy)Cl}(m)](n) (m = -1, -2; bpy = 2,2'-bipyridine). The precursors cis-(Cl)-[Ru(II)(CO)(MeCN)(bpy)Cl(2)] (in PrCN) and mer-[Ru(II)(CO)(bpy)Cl(3)](-) (in tetrahydrofuran (THF) and PrCN) undergo one-electron reductions to reactive radicals [Ru(II)(CO)(MeCN)(bpy(*-))Cl(2)](-) and [Ru(II)(CO)(bpy(*-))Cl(3)](2-), respectively. Both [bpy(*-)]-containing species readily electropolymerize on concomitant dissociation of two chloride ligands and consumption of a second electron. Along this path, mer-to-fac isomerization of the bpy-reduced trichlorido complex (supported by density functional theory calculations) and a concentration-dependent oligomerization process contribute to the complex reactivity pattern. In situ spectroelectrochemistry (IR, UV/vis) has revealed that the charged polymer [{Ru(0)(CO)(bpy)Cl}(-)](n) is stable in THF, but in PrCN it converts readily to [Ru(0)(CO)(PrCN)(bpy)](n). An excess of chloride ions retards this substitution at low temperatures. Both polymetallic chains are completely soluble in the electrolyte solution and can be reduced reversibly to the corresponding [bpy(*-)]-containing species.

SELECTION OF CITATIONS
SEARCH DETAIL
...