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 ; 59(1): 294-312, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26645570

ABSTRACT

Selenosemicarbazones show marked antitumor activity. However, their mechanism of action remains unknown. We examined the medicinal chemistry of the selenosemicarbazone, 2-acetylpyridine 4,4-dimethyl-3-selenosemicarbazone (Ap44mSe), and its iron and copper complexes to elucidate its mechanisms of action. Ap44mSe demonstrated a pronounced improvement in selectivity toward neoplastic relative to normal cells compared to its parent thiosemicarbazone. It also effectively depleted cellular Fe, resulting in transferrin receptor-1 up-regulation, ferritin down-regulation, and increased expression of the potent metastasis suppressor, N-myc downstream regulated gene-1. Significantly, Ap44mSe limited deleterious methemoglobin formation, highlighting its usefulness in overcoming toxicities of clinically relevant thiosemicarbazones. Furthermore, Cu-Ap44mSe mediated intracellular reactive oxygen species generation, which was attenuated by the antioxidant, N-acetyl-L-cysteine, or Cu sequestration. Notably, Ap44mSe forms redox active Cu complexes that target the lysosome to induce lysosomal membrane permeabilization. This investigation highlights novel structure-activity relationships for future chemotherapeutic design and underlines the potential of Ap44mSe as a selective anticancer/antimetastatic agent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Lysosomal Membrane Proteins/drug effects , Lysosomes/drug effects , Pyridines/chemical synthesis , Pyridines/pharmacology , Semicarbazones/chemical synthesis , Semicarbazones/pharmacology , Antioxidants/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Ferritins/drug effects , Genes, myc/drug effects , Humans , Iron/metabolism , Iron Chelating Agents/pharmacology , Methemoglobin/metabolism , Models, Molecular , Molecular Conformation , Permeability , Reactive Oxygen Species/metabolism , Receptors, Transferrin/drug effects , Structure-Activity Relationship
2.
J Biol Inorg Chem ; 20(2): 395-402, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25410832

ABSTRACT

The respiratory DMSO reductase from Rhodobacter capsulatus catalyzes the reduction of dimethyl sulfoxide to dimethyl sulfide. Herein, we have utilized this Mo enzyme as an enantioselective catalyst to generate optically pure sulfoxides (methyl p-tolyl sulfoxide, methyl phenyl sulfoxide and phenyl vinyl sulfoxide) from racemic starting materials. A hexaaminecobalt coordination compound in its divalent oxidation state was employed as the mediator of electron transfer between the working electrode and DMSO reductase to continually reactivate the enzyme after turnover. In all cases, chiral HPLC analysis of the reaction mixture revealed that the S-sulfoxide was reduced more rapidly leading to enrichment or isolation of the R isomer.


Subject(s)
Iron-Sulfur Proteins/chemistry , Molybdenum/chemistry , Oxidoreductases/chemistry , Rhodobacter capsulatus/enzymology , Sulfoxides/chemistry , Catalysis , Dimethyl Sulfoxide/chemistry , Oxidation-Reduction , Sulfides/chemistry
3.
J Med Chem ; 55(16): 7230-44, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22861499

ABSTRACT

We developed a series of second-generation di-2-pyridyl ketone thiosemicarbazone (DpT) and 2-benzoylpyridine thiosemicarbazone (BpT) ligands to improve the efficacy and safety profile of these potential antitumor agents. Two novel DpT analogues, Dp4e4mT and DpC, exhibited pronounced and selective activity against human lung cancer xenografts in vivo via the intravenous and oral routes. Importantly, these analogues did not induce the cardiotoxicity observed at high nonoptimal doses of the first-generation DpT analogue, Dp44mT. The Cu(II) complexes of these ligands exhibited potent antiproliferative activity having redox potentials in a range accessible to biological reductants. The activity of the copper complexes of Dp4e4mT and DpC against lung cancer cells was synergistic in combination with gemcitabine or cisplatin. It was demonstrated by EPR spectroscopy that dimeric copper compounds of the type [CuLCl](2), identified crystallographically, dissociate in solution to give monomeric 1:1 Cu:ligand complexes. These monomers represent the biologically active form of the complex.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Copper , Ketones/chemical synthesis , Lung Neoplasms/drug therapy , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , Dimerization , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Injections, Intravenous , Ketones/chemistry , Ketones/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Transferrin/metabolism , Transplantation, Heterologous
4.
Mol Pharmacol ; 82(1): 105-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22508546

ABSTRACT

Thiosemicarbazones are a group of compounds that have received comprehensive investigation as anticancer agents. The antitumor activity of the thiosemicarbazone, 3-amino-2-pyridinecarboxaldehyde thiosemicarbazone (3-AP; triapine), has been extensively assessed in more than 20 phase I and II clinical trials. These studies have demonstrated that 3-AP induces methemoglobin (metHb) formation and hypoxia in patients, limiting its usefulness. Considering this problem, we assessed the mechanism of metHb formation by 3-AP compared with that of more recently developed thiosemicarbazones, including di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT). This was investigated using intact red blood cells (RBCs), RBC lysates, purified oxyhemoglobin, and a mouse model. The chelation of cellular labile iron with the formation of a redox-active thiosemicarbazone-iron complex was found to be crucial for oxyhemoglobin oxidation. This observation was substantiated using a thiosemicarbazone that cannot ligate iron and also by using the chelator, desferrioxamine, that forms a redox-inactive iron complex. Of significance, cellular copper chelation was not important for metHb generation in contrast to its role in preventing tumor cell proliferation. Administration of Dp44mT to mice catalyzed metHb and cardiac metmyoglobin formation. However, ascorbic acid administered together with the drug in vivo significantly decreased metHb levels, providing a potential therapeutic intervention. Moreover, we demonstrated that the structure of the thiosemicarbazone is of importance in terms of metHb generation, because the DpT analog, di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), does not induce metHb generation in vivo. Hence, DpC represents a next-generation thiosemicarbazone that possesses markedly superior properties. This investigation is important for developing more effective thiosemicarbazone treatment regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Methemoglobin/metabolism , Pyridines/pharmacology , Thiosemicarbazones/pharmacology , Animals , Ascorbic Acid , Cell Proliferation/drug effects , Deferoxamine/pharmacology , Drug Interactions , Erythrocytes/drug effects , Humans , Hypoxia/chemically induced , Iron/metabolism , Iron Chelating Agents/pharmacology , Kinetics , Mice , Oxidation-Reduction/drug effects , Oxyhemoglobins/metabolism
5.
J Med Chem ; 54(19): 6936-48, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21846118

ABSTRACT

Iron chelators of the 2'-benzoylpyridine thiosemicarbazone (BpT) class show substantial potential as anticancer agents. To explore structure-activity relationships, new BpT analogues were designed that incorporated halogen substituents on the noncoordinating phenyl group (XBpTs). These XBpT ligands exhibited potent antiproliferative activity with some analogues exceeding that of the parent BpT compound. Importantly, there was an appreciable therapeutic index in vitro, as mortal cells were significantly less affected by these chelators relative to neoplastic cells. The addition of a halogen led to a halogen-specific increase in the redox potential of XBpT-Fe complexes. Probing for chelator-induced intracellular reactive oxygen species (ROS) with the fluorescent probe, 2',7'-dichlorofluorescein, revealed a 1.5-4.7-fold increase in fluorescence upon incorporation of Cl, Br, or I to the parent analogues. Furthermore, an important structure-activity relationship was deduced where the addition of halogens led to a positive correlation between intracellular ROS generation and antiproliferative activity in the more hydrophilic BpT parent compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Iron Chelating Agents/chemical synthesis , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ascorbic Acid/chemistry , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Electrochemistry , Fluoresceins , Fluorescent Dyes , Fluorometry , Humans , Hydrophobic and Hydrophilic Interactions , Iron Chelating Agents/chemistry , Molecular Structure , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology
6.
Chemistry ; 17(1): 151-60, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-21207612

ABSTRACT

Transformation of proteins and peptides to fibrillar aggregates rich in ß sheets underlies many diseases, but mechanistic details of these structural transitions are poorly understood. To simulate aggregation, four equivalents of a water-soluble, α-helical (65 %) amphipathic peptide (AEQLLQEAEQLLQEL) were assembled in parallel on an oxazole-containing macrocyclic scaffold. The resulting 4α-helix bundle is monomeric and even more α helical (85 %), but it is also unstable at pH 4 and undergoes concentration-dependent conversion to ß-sheet aggregates and amyloid fibrils. Fibrils twist and grow with time, remaining flexible like rope (>1 µm long, 5-50 nm wide) with multiple strings (2 nm), before ageing to matted fibers. At pH 7 the fibrils revert back to soluble monomeric 4α-helix bundles. During αâ†’ß folding we were able to detect soluble 3(10) helices in solution by using 2D-NMR, CD and FTIR spectroscopy. This intermediate satisfies the need for peptide elongation, from the compressed α helix to the fully extended ß strand/sheet, and is driven here by 3(10) -helix aggregation triggered in this case by template-promoted helical bundling and by hydrogen-bonding glutamic acid side chains. A mechanism involving α⇌α(4) ⇌(3(10) )(4) ⇌(3(10) )(n) ⇌(ß)(n) ⇋m(ß)(n) equilibria is plausible for this peptide and also for peptides lacking hydrogen-bonding side chains, with unfavourable equilibria slowing the αâ†’ß conversion.


Subject(s)
Amyloid/chemistry , Oligopeptides/chemical synthesis , Amino Acid Sequence , Amyloid/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemistry , Oxazoles/chemistry , Protein Structure, Tertiary
7.
Curr Top Med Chem ; 11(5): 591-607, 2011.
Article in English | MEDLINE | ID: mdl-21189130

ABSTRACT

An evaluation of existing and proposed Fe chelators, both synthetic and natural products, for the treatment of Fe-overload disease must address a number of issues. There are fundamental parameters that determine the efficacy of a drug: absorption, distribution, metabolism, clearance and toxicity. However, the administration of chelator for Fe overload aims to generate Fe complexes in vivo that are able to be excreted. Hence, the chemical and pharmacological properties of the complexes formed are equally important as the chelators themselves. The redox properties of the Fe complexes formed is particularly relevant to their toxicity. If both Fe(II) and Fe(III) oxidation states of the complexes are biologically accessible, then there is potential for the auto-catalytic production of deleterious free radicals, by Fenton-type chemistry. In addition, since the burden of Fe overload disease falls predominantly on some of the poorest economies, the cost of a drug must be considered, as well as the mode of delivery. There are also possible issues with the use of naturally occurring ligands, which may form Fe complexes capable of being utilised by opportunistic bacteria. This review will concentrate on recent developments in our chemical understanding of existing chelators approved or proposed for use and will also consider some of the candidates from natural sources that have been recently proposed.


Subject(s)
Biological Products/pharmacology , Chelation Therapy , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/pharmacology , Iron Overload/drug therapy , Animals , Biological Products/chemistry , Humans , Iron/chemistry , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Overload/metabolism , Oxidation-Reduction
8.
J Med Chem ; 53(15): 5759-69, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20597487

ABSTRACT

The novel chelators 2-acetylpyridine-4,4-dimethyl-3-thiosemicarbazone (HAp44mT) and di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (HDp44mT) have been examined to elucidate the structure-activity relationships necessary to form copper (Cu) complexes with pronounced antitumor activity. Electrochemical studies demonstrated that the Cu complexes of these ligands had lower redox potentials than their iron complexes. Moreover, the Cu complexes where the ligand/metal ratio was 1:1 rather than 2:1 had significantly higher intracellular oxidative properties and antitumor efficacy. Interestingly, the 2:1 complex was shown to dissociate to give significant amounts of the 1:1 complex that could be the major cytotoxic effector. Both types of Cu complex showed significantly more antiproliferative activity than the ligand alone. We also demonstrate the importance of the inductive effects of substituents on the carbonyl group of the parent ketone, which influence the Cu(II/I) redox potentials because of their proximity to the metal center. The structure-activity relationships described are important for the design of potent thiosemicarbazone Cu complexes.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chelating Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Copper , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/chemistry , Chelating Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Drug Screening Assays, Antitumor , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Ligands , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology
10.
J Med Chem ; 52(5): 1459-70, 2009 Mar 12.
Article in English | MEDLINE | ID: mdl-19216562

ABSTRACT

Through systematic structure-activity studies of the 2-benzoylpyridine thiosemicarbazone (HBpT), 2-(3-nitrobenzoyl)pyridine thiosemicarbazone (HNBpT) and dipyridylketone thiosemicarbazone (HDpT) series of iron (Fe) chelators, we identified structural features necessary to form Fe complexes with potent anticancer activity (J. Med. Chem. 2007, 50, 3716-3729). In this investigation, we generated the related 2-acetylpyridine thiosemicarbazone (HApT) analogues to examine the influence of the methyl group at the imine carbon. Four of the six HApT chelators had potent antitumor activity (IC(50): 0.001-0.002 microM) and Fe chelation efficacy that was similar to the most effective HBpT and HDpT ligands. The HApT Fe complexes had the lowest Fe(III/II) redox potentials of any thiosemicarbazone series we have generated. This property, in combination with their ability to effectively chelate cellular Fe, make the HApT series one of the most potent antiproliferative agents developed by our group.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Iron Chelating Agents/chemical synthesis , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ascorbic Acid/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Electrochemical Techniques , Humans , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Oxidation-Reduction , Pyridines/chemistry , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Transferrin/metabolism
11.
J Med Chem ; 52(2): 407-15, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19090766

ABSTRACT

We previously reported a series of di-2-pyridylketone thiosemicarbazone (HDpT) chelators that showed marked and selective antitumor activity (Whitnall, M.; et al. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 14901-14906). To further understand their biological efficacy, we report the characterization and activity of their Mn(II), Co(III), Ni(II), Cu(II), and Zn(II) complexes. The X-ray crystal structures of four divalent (Mn, Ni, Cu, and Zn) and one trivalent (Fe) complexes are reported. Electrochemistry shows the Fe(III/II) and Cu(II/I) potentials of the complexes may be redox-active within cells. Stability constants were also determined for the Mn(II), Ni(II), Cu(II), and Zn(II) complexes. All divalent complexes underwent transmetalation upon encountering Fe(II), to form low spin ferrous complexes. Importantly, the divalent Mn(II), Ni(II), Cu(II), and Zn(II) complexes of the HDpT analogues are equally active in preventing proliferation as their ligands, suggesting the complexes act as lipophilic vehicles facilitating intracellular delivery of the free ligand upon metal dissociation.


Subject(s)
Antineoplastic Agents/chemistry , Iron Chelating Agents/chemistry , Ketones/chemistry , Metals/chemistry , Thiosemicarbazones/chemistry , Cell Proliferation/drug effects , Crystallography, X-Ray , Electrochemistry , HL-60 Cells , Humans , Iron Chelating Agents/pharmacology , Models, Molecular , Molecular Structure , Spectrophotometry, Ultraviolet , Thiosemicarbazones/pharmacology
12.
J Med Chem ; 51(2): 331-44, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18159922

ABSTRACT

The design of novel Fe chelators with high Fe mobilization efficacy and low toxicity remains an important priority for the treatment of Fe overload disease. We have designed and synthesized the novel methyl pyrazinylketone isonicotinoyl hydrazone (HMPIH) analogs based on previously investigated aroylhydrazone chelators. The HMPIH series demonstrated high Fe mobilization efficacy from cells and showed limited to moderate antiproliferative activity. Importantly, this novel series demonstrated irreversible electrochemistry, which was attributed to the electron-withdrawing effects of the noncoordinating pyrazine N-atom. The latter functionality played a major role in forming redox-inactive complexes that prevent reactive oxygen species generation. In fact, the Fe complexes of the HMPIH series prevented the oxidation of ascorbate and hydroxylation of benzoate. We determined that the incorporation of electron-withdrawing groups is an important feature in the design of N, N, O-aroylhydrazones as candidate drugs for the treatment of Fe overload disease.


Subject(s)
Hydrazones/chemical synthesis , Iron Chelating Agents/chemical synthesis , Iron Overload/drug therapy , Isonicotinic Acids/chemical synthesis , Ketones/chemical synthesis , Pyrazines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ascorbic Acid/metabolism , Benzoates/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Humans , Hydrazones/chemistry , Hydrazones/pharmacology , Hydroxylation , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Iron Radioisotopes , Isonicotinic Acids/chemistry , Isonicotinic Acids/pharmacology , Ketones/chemistry , Ketones/pharmacology , Ligands , Oxidation-Reduction , Pyrazines/chemistry , Pyrazines/pharmacology , Structure-Activity Relationship
13.
J Biol Inorg Chem ; 13(1): 107-19, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17899222

ABSTRACT

2-Pyridinecarbaldehyde isonicotinoyl hydrazone (HPCIH) and di-2-pyridylketone isonicotinoyl hydrazone (HPKIH) are two Fe chelators with contrasting biological behavior. HPCIH is a well-tolerated Fe chelator with limited antiproliferative activity that has potential applications in the treatment of Fe-overload disease. In contrast, the structurally related HPKIH ligand possesses significant antiproliferative activity against cancer cells. The current work has focused on understanding the mechanisms of the Fe mobilization and antiproliferative activity of these hydrazone chelators by synthesizing new analogs (based on 2-acetylpyridine and 2-benzoylpyridine) that resemble both series and examining their Fe coordination and redox chemistry. The Fe mobilization activity of these compounds is strongly dependent on the hydrophobicity and solution isomeric form of the hydrazone (E or Z). Also, the antiproliferative activity of the hydrazone ligands was shown to be influenced by the redox properties of the Fe complexes. This indicated that toxic Fenton-derived free radicals are important for the antiproliferative activity for some hydrazone chelators. In fact, we show that any substitution of the H atom present at the imine C atom of the parent HPCIH analogs leads to an increase in antiproliferative efficacy owing to an increase in redox activity. These substituents may deactivate the imine R-C=N-Fe (R is Me, Ph, pyridyl) bond relative to when a H atom is present at this position preventing nucleophilic attack of hydroxide anion, leading to a reversible redox couple. This investigation describes novel structure-activity relationships of aroylhydrazone chelators that will be useful in designing new ligands or fine-tuning the activity of others.


Subject(s)
Cell Proliferation/drug effects , Iron Chelating Agents/pharmacology , Pyridines/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Humans , Iron Chelating Agents/chemistry , Iron Chelating Agents/metabolism , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Pyridines/chemistry , Pyridines/metabolism , Spectrometry, Mass, Electrospray Ionization
14.
J Med Chem ; 50(24): 6212-25, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17963372

ABSTRACT

Di-2-pyridylketone isonicotinoyl hydrazone Fe chelators utilize the N,N,O-donor set and have moderate anti-proliferative effects. Their closely related N,N,S-thiosemicarbazone analogues, namely, the di-2-pyridylketone thiosemicarbazones, exhibit markedly increased anti-proliferative and redox activity, and this was thought to be due to the inclusion of a sulfur donor atom (Richardson, D. R. et al. J. Med. Chem. 2006, 49, 6510-6521). To further examine the effect of donor atom identity on anti-proliferative activity, we synthesized thiohydrazone analogues of extensively examined aroylhydrazone chelators. The O,N,S-thiohydrazones exhibited decreased anti-proliferative effects compared to their parent aroylhydrazones and reduced redox activity. In contrast, the N,N,S-thiohydrazones showed vastly increased anti-proliferative activity compared to their hydrazone analogues, being comparable to potent thiosemicarbazones. Additionally, N,N,S-thiohydrazone complexes had reversible FeIII/II couples and exhibited increased redox activity. These observations demonstrate that the N,N,S-donor set is critical for potent anti-proliferative efficacy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Ferrous Compounds/chemical synthesis , Hydrazones/chemical synthesis , Iron Chelating Agents/chemical synthesis , Thiones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ascorbic Acid/metabolism , Benzoates/metabolism , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Electrochemistry , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Humans , Hydrazones/chemistry , Hydrazones/pharmacology , Hydroxylation , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Ligands , Molecular Structure , Oxidation-Reduction , Structure-Activity Relationship , Thiones/chemistry , Thiones/pharmacology , Transferrin/metabolism
15.
Dalton Trans ; (30): 3232-44, 2007 Aug 14.
Article in English | MEDLINE | ID: mdl-17893768

ABSTRACT

The potentially tridentate ligand 2-pyridinecarbaldehyde isonicotinoyl hydrazone (HPCIH) and its analogues are an emerging class of orally effective Fe chelators that show great promise for the treatment of Fe overload diseases. Herein, we present an extensive study of the Fe coordination chemistry of the HPCIH analogues including the first crystallographically characterised Fe(II) complex of these chelators. Unlike most other clinically effective Fe chelators, the HPCIH analogues bind Fe(II) and not Fe(III). In fact, these chelators form low-spin bis-ligand Fe(II) complexes, although NMR suggests that the complexes are close to the high-spin/low-spin crossover. All the Fe complexes show a high potential Fe(III/II) redox couple (> 500 mV vs. NHE) and cyclic voltammetry in aqueous or mixed aqueous/organic solvents is irreversible as a consequence of a rapid hydration reaction that occurs upon oxidation. A number of the HPCIH analogues show high activity at inducing Fe efflux from cells and also at preventing Fe uptake by cells from the serum Fe transport protein transferrin. As a class of ligands, these chelators are more effective at reducing Fe uptake from transferrin than inducing Fe mobilisation from cells. This may be related to their ability to intercept Fe(II) after its release from transferrin within the cell. Our studies indicate that their Fe chelation efficacy is due, at least in part, to the fact that these ligands and their Fe(II) complexes are neutral at physiological pH (7.4) and sufficiently lipophilic to permeate cell membranes.


Subject(s)
Chelating Agents/chemistry , DNA/drug effects , Hydrazones/chemistry , Iron Overload/drug therapy , Chelating Agents/pharmacology , Chemistry, Pharmaceutical , Crystallography, X-Ray , Electrochemistry , Molecular Structure
16.
J Med Chem ; 50(15): 3716-29, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17602603

ABSTRACT

Previously, we demonstrated that the potent antiproliferative activity of the di-2-pyridylketone thiosemicarbazone (DpT) series of Fe chelators was due to their ability to induce Fe depletion and form redox-active Fe complexes (Richardson, D. R.; et al. J. Med. Chem. 2006, 49, 6510-6521). We now examine the role of aromatic substituents on the antiproliferative and redox activity of novel DpT analogues, namely, the 2-benzoylpyridine thiosemicarbazone (BpT) and 2-(3-nitrobenzoyl)pyridine thiosemicarbazone (NBpT) series. Both series exhibited selective antiproliferative effects, with the majority having greater antineoplastic activity than their DpT homologues. This makes the BpT chelators the most active anticancer agents developed within our laboratory. The BpT series Fe complexes exhibit lower redox potentials than their corresponding DpT and NBpT complexes, highlighting their enhanced redox activity. The increased ability of BpT-Fe complexes to catalyze ascorbate oxidation and benzoate hydroxylation, relative to their DpT and NBpT analogues, suggested that redox cycling plays an important role in their antiproliferative activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Iron Chelating Agents/chemical synthesis , Nitrobenzoates/chemical synthesis , Pyridines/chemical synthesis , Thiosemicarbazones/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ascorbic Acid/chemistry , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Ferric Compounds/chemical synthesis , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Ferrous Compounds/chemical synthesis , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Humans , Iron/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Ligands , Molecular Structure , Nitrobenzoates/chemistry , Nitrobenzoates/pharmacology , Oxidation-Reduction , Protein Binding , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Transferrin/metabolism
17.
J Med Chem ; 49(22): 6510-21, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064069

ABSTRACT

There has been much interest in the development of iron (Fe) chelators for the treatment of cancer. We developed a series of di-2-pyridyl ketone thiosemicarbazone (HDpT) ligands which show marked and selective antitumor activity in vitro and in vivo. In this study, we assessed chemical and biological properties of these ligands and their Fe complexes in order to understand their marked activity. This included examination of their solution chemistry, electrochemistry, ability to mediate redox reactions, and antiproliferative activity against tumor cells. The higher antiproliferative efficacy of the HDpT series of chelators relative to the related di-2-pyridyl ketone isonicotinoyl hydrazone (HPKIH) analogues can be ascribed, in part, to the redox potentials of their Fe complexes which lead to the generation of reactive oxygen species. The most effective HDpT ligands as antiproliferative agents possess considerable lipophilicity and were shown to be charge neutral at physiological pH, allowing access to intracellular Fe pools.


Subject(s)
Antineoplastic Agents , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Iron/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Ascorbic Acid/chemistry , Benzoates/chemistry , Chemical Phenomena , Chemistry, Physical , Crystallography, X-Ray , DNA/genetics , Electrochemistry , Humans , Hydroxylation , Magnetic Resonance Spectroscopy , Models, Molecular , Oxidation-Reduction , Plasmids/genetics , Spectrophotometry, Infrared , Structure-Activity Relationship
18.
J Biol Inorg Chem ; 11(7): 930-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16868742

ABSTRACT

A selection of nine macrocyclic Fe(III/II) and Co(III/II) transition metal complexes has been chosen to serve as a universal set of mediator-titrants in redox potentiometry of protein samples. The potential range spanned by these mediators is approximately from +300 to -700 mV vs the normal hydrogen electrode, which covers the range of most protein redox potentials accessible in aqueous solution. The complexes employed exhibit stability in both their oxidized and their reduced forms as well as pH-independent redox potentials within the range 6 < pH < 9. The mediators were also chosen on the basis of their very weak visible absorption maxima in both oxidation states, which will enable (for the first time) optical redox potentiometric titrations of proteins with relatively low extinction coefficients. This has previously been impractical with organic mediators, such as indoles, viologens and quinones, whose optical spectra interfere strongly with those of the protein.


Subject(s)
Organometallic Compounds/chemistry , Proteins/chemistry , Spectrophotometry/methods , Transition Elements/chemistry , Cobalt/chemistry , Iron/chemistry , Molecular Structure , Oxidation-Reduction , Potentiometry , Titrimetry
19.
J Biol Inorg Chem ; 10(7): 761-77, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16193304

ABSTRACT

The search for orally effective drugs for the treatment of iron overload disorders is an important goal in improving the health of patients suffering diseases such as beta-thalassemia major. Herein, we report the syntheses and characterization of some new members of a series of N-aroyl-N'-picolinoyl hydrazine chelators (the H2IPH analogs). Both 1:1 and 1:2 Fe(III):L complexes were isolated and the crystal structures of Fe(HPPH)Cl2, Fe(4BBPH)Cl2, Fe(HAPH)(APH) and Fe(H3BBPH)(3BBPH) were determined (H2PPH=N,N'-bis-picolinoyl hydrazine; H2APH=N-4-aminobenzoyl-N'-picolinoyl hydrazine, H23BBPH=N-3-bromobenzoyl-N'-picolinoylhydrazine and H24BBPH=N-(4-bromobenzoyl)-N'-(picolinoyl)hydrazine). In each case, a tridentate N,N,O coordination mode of each chelator with Fe was observed. The Fe(III) complexes of these ligands have been synthesized and their structural, spectroscopic and electrochemical characterization are reported. Five of these new chelators, namely H2BPH (N-(benzoyl)-N'-(picolinoyl)hydrazine), H2TPH (N-(2-thienyl)-N'-(picolinoyl)-hydrazine), H2PPH, H23BBPH and H24BBPH, showed high efficacy at mobilizing 59Fe from cells and inhibiting 59Fe uptake from the serum Fe transport protein, transferrin (Tf). Indeed, their activity was much greater than that found for the chelator in current clinical use, desferrioxamine (DFO), and similar to that observed for the orally active chelator, pyridoxal isonicotinoyl hydrazone (H2PIH). The ability of the chelators to inhibit 59Fe uptake could not be accounted for by direct chelation of 59Fe from 59Fe-Tf. The most effective chelators also showed low antiproliferative activity which was similar to or less than that observed with DFO, which is important in terms of their potential use as agents to treat Fe-overload disease.


Subject(s)
Hydrazines/chemistry , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Carrier Proteins/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemical Phenomena , Chemistry, Physical , Crystallography, X-Ray , Deferoxamine/chemistry , Drug Screening Assays, Antitumor , Electrochemistry , Humans , Iron/chemistry , Iron/metabolism , Iron Radioisotopes/chemistry , Ligands , Lipids/chemistry , Models, Molecular , Potentiometry , Solutions , Structure-Activity Relationship , Transferrin/chemistry
20.
Inorg Chem ; 36(11): 2420-2425, 1997 May 21.
Article in English | MEDLINE | ID: mdl-11669880

ABSTRACT

The syntheses and characterisation of the new macrocyclic hexaamine trans-(5(S),7(S),12(R),14(R)-tetramethyl)-1,4,8,11-tetraazacyclotetradecane-6,13-diamine (L(6)) and its Co(III) complex are reported. The X-ray crystal structural analyses of [CoL(6)]Cl(2)(ClO(4)) [monoclinic, space group C2/c, a = 16.468(3) Å, b = 9.7156(7) Å, c = 15.070(3) Å, beta = 119.431(8) degrees, Z = 4] and the closely related cis-diamino-substituted macrocyclic complex [CoL(2)](ClO(4))(3). 2H(2)O (L(2) = cis-6,13-dimethyl-1,4,8,11-tetraazacyclotetradecane-6,13-diamine) [orthorhombic, space group Pna2(1), a = 16.8220(8) Å, b = 10.416(2) Å, c = 14.219(3) Å, Z = 4] reveal significant variations in the observed Co-N bond lengths and coordination geometries, which may be attributed to the trans or cis disposition of the pendent primary amines. The Co(III/II) self-exchange electron transfer rate constants for these and other closely related hexaamines have been determined, and variations of some 2 orders of magnitude are found between pairs of trans and cis isomeric Co(III) complexes.

SELECTION OF CITATIONS
SEARCH DETAIL
...