Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters










Publication year range
2.
Br J Radiol ; 82(974): 89-104, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19168690

ABSTRACT

Biological effects of radiation are manifest over timescales extending to years. However, many chemical events are complete in milliseconds; after this time, adding oxygen to irradiated hypoxic cells no longer enhances radiosensitivity. This does not mean that damage pathways cannot be modified; the potential gain from chemical modulation of early events is as large as any associated with later pathways, and the prognostic importance of variations in levels of small molecules active in fast free radical pathways is as important as any associated with genetic make-up. Reactive oxygen species are much invoked in the wider context, but are frequently undefined and seldom measured unambiguously. Radiation chemistry has much to offer to both radiation and free radical biology. An appreciation of the interlinked parameters of time, spatial distribution and yield is well developed, as are methods to generate specific radicals in known concentrations and to monitor their reactions directly. Intense clinical interest in the 1980s in hypoxic cell radiosensitizers, developed from radiation chemical studies, has waned, but the goal of eliminating hypoxic radioresistance remains attractive. Nitric oxide may be more important than oxygen in determining hypoxic radiosensitivity, and radiation chemistry provides the tools to understand the mechanisms and the limitations of in vitro models. Imaging hypoxia in tumours relies heavily on free radical chemistry and radiolysis methods to understand the mechanistic basis for diagnostic agents. Quantitation of the chemical reactivity of free radicals is a cornerstone of radiation chemistry via the language, concepts and mathematics of chemical kinetics, which are equally applicable to understanding the molecular pathways in radiobiology.


Subject(s)
Radiobiology/methods , Radiochemistry/methods , Artifacts , Fluorescent Dyes , Free Radicals/metabolism , Humans , Nitric Oxide/physiology , Nitrosation , Oxidative Stress , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism
3.
Clin Oncol (R Coll Radiol) ; 19(6): 397-417, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478086

ABSTRACT

Radiosensitizers are intended to enhance tumour cell killing while having much less effect on normal tissues. Some drugs target different physiological characteristics of the tumour, particularly hypoxia associated with radioresistance. Oxygen is the definitive hypoxic cell radiosensitizer, the large differential radiosensitivity of oxic vs hypoxic cells being an attractive factor. The combination of nicotinamide to reduce acute hypoxia with normobaric carbogen breathing is showing clinical promise. 'Electron-affinic' chemicals that react with DNA free radicals have the potential for universal activity to combat hypoxia-associated radioresistance; a nitroimidazole, nimorazole, is clinically effective at tolerable doses. Hypoxia-specific cytotoxins, such as tirapazamine, are valuable adjuncts to radiotherapy. Nitric oxide is a potent hypoxic cell radiosensitizer; variations in endogenous levels might have prognostic significance, and routes to deliver nitric oxide specifically to tumours are being developed. In principle, many drugs can be delivered selectively to hypoxic tumours using either reductase enzymes or radiation-produced free radicals to activate drug release from electron-affinic prodrugs. A redox-active agent based on a gadolinium chelate is being evaluated clinically. Pyrimidines substituted with bromine or iodine are incorporated into DNA and enhance free radical damage; fluoropyrimidines act by different mechanisms. A wide variety of drugs that influence the nature or repair of DNA damage are being evaluated in conjunction with radiation; it is often difficult to define the mechanisms underlying chemoradiation regimens. Drugs being evaluated include topoisomerase inhibitors (e.g. camptothecin, topotecan), and the hypoxia-activated anthraquinone AQ4N; alkylating agents include temozolomide. Drugs involved in DNA repair pathways being investigated include the potent poly(ADP ribose)polymerase inhibitor, AG14,361. Proteins involved in cell signalling, such as the Ras family, are attractive targets linked to radioresistance, as are epidermal growth factor receptors and linked kinases (drugs including vandetanib [ZD6,474], cetuximab and gefitinib), and cyclooxygenase-2 (celecoxib). The suppression of radioprotective thiols seems to offer more potential with alkylating agents than with radiotherapy, although it remains a strategy worthy of exploration.


Subject(s)
Neoplasms/radiotherapy , Radiation-Sensitizing Agents/chemistry , Animals , Cell Communication/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cytotoxins/metabolism , Cytotoxins/radiation effects , DNA Repair/radiation effects , DNA, Neoplasm/radiation effects , Free Radical Scavengers/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/radiation effects , Neoplasms/physiopathology , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Radiation-Protective Agents/radiation effects , Radiation-Sensitizing Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects
5.
Biochem Soc Trans ; 31(Pt 6): 1337-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14641058

ABSTRACT

Superoxide radicals are produced in trace amounts by the mitochondrial respiratory chain. Most are removed rapidly by superoxide dismutase in the matrix. Superoxide is also known to react with glutathione. Reported values of the rate constant for this reaction range from 10(2) to in excess of 10(5) M(-1).s(-1). The magnitude of this rate constant has important physiological implications because, if it is at the upper end of the reported range, a significant proportion of mitochondrial superoxide will evade removal by superoxide dismutase, and will oxidize glutathione to the potentially harmful glutathionyl radical. Using EPR spectroscopy to monitor competition between glutathione and the spin trap 5,5-dimethyl-1-pyrroline N-oxide for reaction with superoxide, we have estimated that the rate constant for the reaction between superoxide and glutathione is only approximately 200 M(-1).s(-1). Hence superoxide dismutase will always out-compete glutathione for reaction with the superoxide radical, thereby preventing formation of the glutathionyl radical.


Subject(s)
Free Radical Scavengers/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Superoxides/metabolism , Electron Spin Resonance Spectroscopy , Kinetics , Spin Labels
6.
Int J Radiat Oncol Biol Phys ; 51(2): 460-4, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11583019

ABSTRACT

PURPOSE: To test the hypothesis that there is a link between plasma glutathione (GSH) or other antioxidants (uric acid, ascorbate) and the severity of radiation mucositis following radiation treatment of tumors of the head and neck. PATIENTS AND METHODS: Patients with carcinomas of the head-and-neck region were treated with the continuous hyperfractionated accelerated radiotherapy (CHART) regimen (54 Gy in 36 fractions over 12 days). Samples of blood plasma were analyzed for GSH, cysteine, urate, and ascorbate by high-pressure liquid chromatography. Patients were graded for dysphagia and requirement for analgesics. The areas under the curves of scores over 2-6 weeks following treatment were computed, and Spearman's rank-correlation coefficient was used to test for an association between plasma GSH levels (or those of other antioxidants) and mucositis. RESULTS: The pretreatment plasma GSH level in 18 patients scored in the study was 1.0 +/- 0.7 M. Analysis of these and the dysphagia scores produced a correlation coefficient of 0.22 (confidence interval -0.28, 0.61; p = 0.39). No correlation was seen between mucositis severity and other measures of plasma antioxidants: cysteine (7.6 +/- 1.7 M), cysteine + GSH (8.6 +/- 1.9 M), uric acid (317 +/- 86 M), ascorbate (29 +/- 20 M), or whole-blood GSH concentrations (1,010 +/- 239 M). CONCLUSION: The measurements of approximately micromolar levels of plasma GSH, or about 10 M cysteine + GSH (almost all of the total nonprotein thiols), are consistent with most other published data for either healthy adults or cancer patients; however, the values reported in an earlier study, suggesting a link between GSH and mucositis, are much higher. The hypothesis of a possible link between radiation mucositis and plasma-free (nonprotein) thiols was not supported.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Glutathione/blood , Mouth Mucosa/radiation effects , Mouth Neoplasms/radiotherapy , Stomatitis/blood , Ascorbic Acid/blood , Cysteine/blood , Dose Fractionation, Radiation , Humans , Linear Models , Sensitivity and Specificity , Stomatitis/etiology , Uric Acid/blood
7.
Curr Med Chem ; 8(7): 739-61, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375747

ABSTRACT

Hypoxia is a feature of some regions of many tumours, ischaemic events, and arthritis. Drugs activated in hypoxia have wide potential application, particularly in overcoming the resistance of hypoxic tumour cells to radiotherapy. Key features of such drugs include redox properties appropriate for activation by reductase enzymes (typically flavoproteins), and oxygen-sensitive reduction chemistry such that normal levels of oxygen inhibit or reverse reduction. In many cases this selectivity is achieved by a fast, free-radical reaction in which the drug radical (often an obligate intermediate in drug reduction) reduces oxygen to form superoxide radicals and thus 'futile cycles' the drug in normoxic tissues. However, this enhances cellular oxidative stress, which may be linked to normal tissue toxicity. Appropriate redox properties are found with nitroarene, quinone, or aromatic N-oxide moieties. A particularly promising and versatile exploitation of bioreductive activation is for reduction of such 'triggers' to activate release of an 'effector', an agent that can obviously be active against diverse conditions associated with hypoxia. The same approach can also be used in diagnosis of hypoxia. Much information concerning the reactions of intermediates in drug action and the quantitative prediction of redox properties of analogues has been accrued. Drug design can be mechanism-led, with the wealth of literature quantifying redox properties of drug candidates a rich source of potential new leads. There is a clear appreciation of the kinetic factors that limit drug efficacy or selectivity. Thus the potential for rapid expansion of these concepts to diverse diseases is considerable.


Subject(s)
Drug Design , Flavoproteins/metabolism , Hypoxia/diagnosis , Hypoxia/drug therapy , Oxidative Stress/drug effects , Drug Delivery Systems/methods , Electron Transport/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Humans , Hypoxia/physiopathology , Neoplasms/blood supply , Neoplasms/diagnosis , Oxidation-Reduction , Quinones/chemistry , Quinones/pharmacology , Tirapazamine , Triazines/chemistry , Triazines/pharmacology
8.
Biochem Biophys Res Commun ; 282(1): 329-33, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11264011

ABSTRACT

The generation of reactive oxygen species has been suggested to occur at increased rates during apoptosis, but the validity and significance of this remain contentious. In several key studies levels of reactive oxygen species have been monitored using the intracellular probe dichlorofluorescin (DCFH(2)), which undergoes oxidation to the fluorescent dichlorofluorescein (DCF). We report here that cytochrome c, which is released from mitochondria during cell death, is a potent catalyst of DCF formation. In a model system using xanthine oxidase to generate superoxide radicals, the rate of DCF formation was insensitive to changes in the rate of superoxide production over a 17-fold range, but extremely sensitive to nanomolar concentrations of cytochrome c. Thus we conclude that the DCF fluorescence observed in dying cells is a reflection of increased cytosolic cytochrome c. Moreover, we suggest that the suppression of DCF formation by the anti-apoptotic oncoprotein Bcl-2, which has been suggested to have antioxidant properties, can be explained on the basis of its prevention of mitochondrial cytochrome c release.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Fluoresceins/metabolism , Reactive Oxygen Species , Catalysis , Electron Spin Resonance Spectroscopy , Oxidation-Reduction
9.
Biochem Pharmacol ; 61(2): 129-36, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11163327

ABSTRACT

Indole-3-acetic acid (IAA) and some derivatives can be oxidised by horseradish peroxidase (HRP) to cytotoxic species. Upon treatment with IAA/HRP, liposomes undergo lipid peroxidation, strand breaks and adducts are formed in supercoiled plasmid DNA, and mammalian cells in culture lose colony-forming ability. IAA is only toxic after oxidative decarboxylation; no effects are seen when IAA or HRP is incubated independently in these systems at equivalent concentrations. Toxicity is similar in both hamster fibroblasts and some human tumour cells. The effect of IAA/HRP is thought to be due in part to the formation of 3-methylene-2-oxindole, which may conjugate with DNA bases and protein thiols. Our hypothesis is that IAA/HRP could be used as the basis for targeted cancer therapy involving antibody-, polymer-, or gene-directed approaches. HRP can thus be targeted to a tumour allowing non-toxic IAA delivered systemically to be activated only in the tumour. Exposure to newly synthesised analogues of IAA shows a range of four orders of magnitude difference in cellular toxicity but no structure-activity relationships are apparent, in contrast to well-defined redox dependencies of oxidation by HRP intermediates or rates of decarboxylation of radical-cation intermediates.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Indoleacetic Acids/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/metabolism , Biotransformation , Drug Delivery Systems , Drug Screening Assays, Antitumor , Horseradish Peroxidase/administration & dosage , Horseradish Peroxidase/metabolism , Horseradish Peroxidase/pharmacology , Humans , Indoleacetic Acids/administration & dosage , Indoleacetic Acids/metabolism , Lipid Peroxidation , Oxidation-Reduction , Prodrugs/administration & dosage , Prodrugs/metabolism , Tumor Cells, Cultured
10.
Mol Cancer Ther ; 1(2): 151-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-12467232

ABSTRACT

We have previously proposed the plant enzyme horseradish peroxidase (HRP) and the plant hormone indole-3-acetic acid (IAA) as an enzyme/prodrug combination for cancer gene therapy. In the current study, we evaluated the potential of HRP/IAA for gene-directed enzyme/prodrug therapy in three human tumor cell lines (T24 bladder carcinoma, MCF-7 breast adenocarcinoma, and FaDu nasopharyngeal squamous carcinoma) and one endothelial cell line (HMEC-1). The action of 10 IAA analogues in combination with HRP was studied in vitro in normoxic conditions as well as in the extreme tumor conditions of anoxia. Compounds characterized by prompt normoxic or anoxic cytotoxic activation and high HRP transfectant killing or selectivity were identified. Some variations were observed in the response of cells of different origin, with IAA, 1-Me-IAA, and 5-Br-IAA representing the most promising candidates for HRP gene therapy. In particular, 5-Br-IAA showed a very prompt and selective activation in anoxia. A strong bystander effect was produced by activated IAA and analogues because 70-90% cell kill was obtained when only 5% of the cells expressed the HRP enzyme. These results indicate that HRP/IAA represents an effective system for enzyme/prodrug-based anticancer approaches, and further improvements could be achieved by the use of novel IAA derivatives.


Subject(s)
Genetic Therapy/methods , Horseradish Peroxidase/genetics , Indoleacetic Acids/pharmacology , Prodrugs/pharmacology , Tumor Cells, Cultured/drug effects , Blotting, Western , Cell Division/drug effects , Cell Hypoxia , Combined Modality Therapy , Humans , Oxygen/metabolism , Plasmids , Transfection , Tumor Cells, Cultured/metabolism , Tumor Stem Cell Assay
11.
Cancer Gene Ther ; 7(11): 1414-20, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11129283

ABSTRACT

This paper demonstrates the potential for utilizing the plant enzyme, horseradish peroxidase (HRP), in a gene-directed enzyme prodrug therapy context. Human T24 bladder carcinoma cells transfected with a mammalian expression vector containing the HRP cDNA were selectively sensitized to the nontoxic plant hormone, indole-3-acetic acid (IAA). The HRP/IAA-induced cell kill was effective in normoxic and anoxic conditions. The activated drug is a long-lived species able to cross cell membranes, and cell contact appears not to be required for a bystander effect to take place. These preliminary results suggest that the delivery of the HRP gene to human tumors followed by IAA treatment may provide a novel cancer gene-directed enzyme prodrug therapy approach, with potential to target hypoxic cells.


Subject(s)
Enzyme Therapy , Genetic Therapy/methods , Horseradish Peroxidase/genetics , Indoleacetic Acids/metabolism , Neoplasms/therapy , Prodrugs , Blotting, Western , Cell Division/drug effects , Cell Separation , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Fluorescent Antibody Technique , Horseradish Peroxidase/metabolism , Humans , Hypoxia , Indoleacetic Acids/toxicity , Inhibitory Concentration 50 , Models, Chemical , Oxygen/metabolism , Plasmids/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
12.
Bioorg Med Chem ; 8(5): 1065-73, 2000 May.
Article in English | MEDLINE | ID: mdl-10882018

ABSTRACT

Two pyridine analogues of the metal complexing region of the anticancer drug bleomycin and two related but deactivated prodrugs have been linked to a 2,6-diphenylpyridine derivative as a DNA binding unit. The 2,6-diphenylpyridine system is structurally related to known amplifiers of the cytotoxicity of bleomycin. The conjugates were found to bind to DNA more strongly than bleomycin-A2 and were more cytotoxic than the corresponding compounds lacking the DNA binding unit. On exposure of a mixture of cells and prodrugs to hypoxia and then air, the prodrug containing the nitrohistidine unit was not bioreductively activated but the prodrug having an N-oxide group was bioreductively activated. This result represents a novel approach to the improvement of the therapeutic ratio of bleomycin analogues.


Subject(s)
Bleomycin/chemical synthesis , Hydroxyl Radical/chemistry , Animals , Bleomycin/analogs & derivatives , Cell Survival/drug effects , Cricetinae , Cricetulus
13.
Photochem Photobiol ; 70(3): 348-52, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10483363

ABSTRACT

Human skin biopsies (hair-bearing scalp skin and non-hair-bearing breast skin) were treated with t-butylhydroperoxide, irradiated with UV light (UVR) or irradiated with 694 nm ruby laser red light. Free-radical production and oxidative stress were assessed with electron spin resonance spectroscopy (ESR) using the ascorbate radical as a marker. In comparison with both UVR and t-butyl-hydroperoxide (which readily induce the ascorbate radical in hair-bearing and hairless skin), 694 nm red light does not result in the formation of the ascorbate radical in detectable concentrations. Spin-trapping experiments with the spin trap 5,5-dimethyl-1-pyrroline N-oxide (DMPO) showed that while free radicals could be detected after treatment of skin with t-butylhydroperoxide, radicals could not be trapped after laser treatment. Treatment of lasered skin (containing DMPO) with t-butylhydroperoxide produced radical adducts as well as the ascorbate radical, demonstrating that the laser neither depletes endogenous ascorbate nor the preadministered spin trap. It is concluded that 694 nm red light does not induce oxidative stress in human skin in levels comparable either to t-butyl hydroperoxide or UV light.


Subject(s)
Hair Removal/adverse effects , Lasers/adverse effects , Oxidative Stress , Skin/metabolism , Skin/radiation effects , Cyclic N-Oxides , Electron Spin Resonance Spectroscopy , Free Radicals/metabolism , Humans , Spin Labels , Spin Trapping , Ultraviolet Rays , tert-Butylhydroperoxide/metabolism , tert-Butylhydroperoxide/toxicity
14.
Bioorg Med Chem Lett ; 9(9): 1267-72, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10340612

ABSTRACT

2-Nitroimidazoles were synthesised substituted with aspirin or salicylic acid, as leaving groups linked through the (imidazol-5-yl)methyl position. Activation of aqueous solutions by CO2*- (a model one-electron reductant) resulted in release of aspirin or salicylate, probably via the 2-hydroxyaminoimidazole. The analogous 2-nitroimidazole with bromide as leaving group eliminated bromide in < 1 ms via the radical-anion.


Subject(s)
Aspirin/chemistry , Cell Hypoxia/drug effects , Nitroimidazoles/chemical synthesis , Prodrugs/chemistry , Animals , Bromides/chemistry , Humans , Salicylates/chemistry , Time Factors
15.
Bioorg Med Chem Lett ; 9(1): 113-8, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9990467

ABSTRACT

A series of regioisomeric derivatives of a 1-methylindole-4,7-dione were synthesised, substituted with a 2-acetoxybenzoate leaving group linked through the (indol-2-yl)methyl or (indol-3-yl)methyl (or propenyl) positions. Reductive elimination of the leaving group occurred from the (indol-3-yl)methyl derivatives but not the 2-substituted regioisomers, indicating that only the C-3 position may be utilised in bioreductively-activated drug delivery, which was demonstrated with an aspirin prodrug.


Subject(s)
Aspirin/chemistry , Indoles/chemistry , Prodrugs/chemistry , Quinones/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal , Arthritis/metabolism , Aspirin/metabolism , Free Radicals/metabolism , Humans , Hypoxia/drug therapy , Hypoxia/metabolism , Indoles/metabolism , Neoplasms/metabolism , Oxidation-Reduction , Prodrugs/metabolism , Quinones/metabolism , Structure-Activity Relationship
16.
Biochem Pharmacol ; 57(4): 375-82, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9933025

ABSTRACT

This study aimed to explore the mechanisms and molecular parameters which control the cytotoxicity of derivatives of indole-3-acetic acid (IAA) when oxidatively activated by horseradish peroxidase (HRP). Lipid peroxidation was measured in liposomes, damage to supercoiled plasmid DNA assessed by gel electrophoresis, free radical intermediates detected by EPR following spin trapping, binding of IAA-derived products demonstrated by 3H labelling, stable products measured by HPLC, and cytotoxicity in hamster fibroblasts measured by clonogenic survival. IAA, and nine analogues more easily oxidized by HRP, caused lipid peroxidation in liposomes, but not detectably in membranes of hamster fibroblasts, and were cytotoxic after HRP activation to varying degrees. Cytotoxicity was not correlated with activation rate. The hydrophilic vitamin E analogue, Trolox, inhibited cytotoxicity, whereas loading fibroblasts with vitamin E was ineffective, consistent with an oxidative mechanism in which radical precursors to damage are intercepted by Trolox in the aqueous phase. However, two known oxidation products were nontoxic (the 3-carbinol and 3-aldehyde, both probably produced from 3-CH2OO* peroxyl radicals via the 3-CH*2 [skatolyl] radical following decarboxylation of the radical cation). The skatolyl radical from IAA was shown by EPR with spin trapping to react with DNA; electrophoresis showed binding to occur. Treatment of hamster fibroblasts with 5-3H-IAA/HRP resulted in intracellular bound 3H. Together with earlier results, the new data point to unknown electrophilic oxidation products, reactive towards intracellular targets, being involved in cytotoxicity of the IAA/HRP combination, rather than direct attack of free radicals, excited states, or membrane lipid peroxidation.


Subject(s)
DNA, Superhelical/drug effects , Horseradish Peroxidase/pharmacology , Indoleacetic Acids/toxicity , Lipid Peroxidation/drug effects , Animals , Cell Line , Cell Survival , Colony-Forming Units Assay , Cricetinae , Cricetulus , DNA Adducts/biosynthesis , Free Radicals , Indoleacetic Acids/metabolism
17.
Int J Radiat Oncol Biol Phys ; 42(4): 917-20, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9845122

ABSTRACT

PURPOSE: The study aimed to identify suitable prodrugs that could be used to test the hypothesis that peroxidase activity in cells, either endogenous or enhanced by immunological targeting, can activate prodrugs to cytotoxins. We hypothesized that prototype prodrugs based on derivatives of indole-3-acetic acid (IAA), when activated by peroxidase enzymes (e.g., from horseradish, HRP) should produce peroxyl radicals, with deleterious biological consequences. METHODS AND MATERIALS: V79 hamster cells were incubated with IAA or derivatives +/- HRP and cytotoxicity assessed by a clonogenic assay. To assess the toxicity of stable oxidation products, prodrugs were also oxidized by HRP without cells, and the products then added to cells. RESULTS: The combination of prodrug and enzyme resulted in cytotoxicity, but neither indole nor enzyme in isolation was toxic under the conditions used. Although lipid peroxidation was stimulated in liposomes by the prodrug/enzyme treatment, it could not be measured in mammalian cells. Adding oxidized prodrugs to cells resulted in cytotoxicity. CONCLUSIONS: Although the hypothesis that prodrugs of this type could enhance oxidative stress via lipid peroxidation was not established, the results nonetheless demonstrated oxidatively-activated cytotoxicity via indole acetic acid prodrugs, and suggested these as a new type of substrate for antibody-directed enzyme-prodrug therapy (ADEPT). The hypothesized free-radical fragmentation intermediates were demonstrated, but lipid peroxidation associated with peroxyl radical formation was unlikely to be the major route to cytotoxicity.


Subject(s)
Indoleacetic Acids/metabolism , Peroxidases/metabolism , Prodrugs/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Line/metabolism , Cricetinae , Cricetulus , HL-60 Cells/metabolism , Horseradish Peroxidase/metabolism , Humans , Indoleacetic Acids/therapeutic use , Oxidation-Reduction , Prodrugs/therapeutic use
18.
Anticancer Drug Des ; 13(6): 635-53, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9755722

ABSTRACT

The factors influencing the kinetics of the oxygen-sensitive reduction of indolequinones, including those bearing leaving groups in the (indol-3-yl)methyl position, have been studied. The hydroquinones derived from some representative indolequinones were found to autoxidize slowly in oxygenated solution at rates (effective rate constant with O2 approximately 40-300 M-1 s-1) that cannot compete with the reductive elimination of leaving groups. The rates of reaction between hydroquinone and O2 were even slower in the presence of approximately 4 microM superoxide dismutase (effective rate constant approximately 2-7 M-1 s-1), indicating the role of superoxide radicals in hydroquinone autoxidation. Since the release of the leaving groups from the hydroquinones is not significantly oxygen-sensitive, tumour selectivity requires specific reduction by enzymes that are overexpressed in some tumours. Conversely, the release of leaving groups from semiquinone radicals is inhibited by oxygen too efficiently unless the semiquinone reacts with targets on a timescale of milliseconds. Modification of redox properties has been explored with the aim of changing this oxygen sensitivity. The new 2-phenylindolequinones are approximately 60-100 mV higher in reduction potential than 2-alkyl derivatives but this is insufficient to decrease the rate of electron transfer from semiquinone to oxygen to a degree which might confer hypoxia-selective cytotoxicity. These results are discussed in the context of toxicity of EO9 and related compounds towards hypoxic rather than anoxic cells.


Subject(s)
Cell Hypoxia/drug effects , Indolequinones , Indoles/chemistry , Quinones/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aziridines/chemistry , Aziridines/pharmacology , Chromatography, High Pressure Liquid , Humans , Hydroquinones/chemistry , Indoles/pharmacology , Kinetics , Molecular Structure , Oxidation-Reduction , Oxygen/chemistry , Pulse Radiolysis , Quinones/pharmacology , Structure-Activity Relationship , Superoxide Dismutase/pharmacology , Superoxides/chemistry
19.
J Med Chem ; 41(15): 2720-31, 1998 Jul 16.
Article in English | MEDLINE | ID: mdl-9667963

ABSTRACT

A series of indolequinones bearing a variety of leaving groups at the (indol-3-yl)methyl position was synthesized by functionalization of the corresponding 3-(hydroxymethyl)indolequinone, and the resulting compounds were evaluated in vitro as bioreductively activated cytotoxins. The elimination of a range of functional groups-carboxylate, phenol, and thiol-was demonstrated upon reductive activation under both chemical and quantitative radiolytic conditions. Only those compounds which eliminated such groups under both sets of conditions exhibited significant hypoxia selectivity, with anoxic:oxic toxicity ratios in the range 10-200. With the exception of the 3-hydroxymethyl derivative, radiolytic generation of semiquinone radicals and HPLC analysis indicated that efficient elimination of the leaving group occurred following one-electron reduction of the parent compound. The active species in leaving group elimination was predominantly the hydroquinone rather than the semiquinone radical. The resulting iminium derivative acted as an alkylating agent and was efficiently trapped by added thiol following chemical reduction and by either water or 2-propanol following radiolytic reduction. A chain reaction in the radical-initiated reduction of these indolequinones (not seen in a simpler benzoquinone) in the presence of a hydrogen donor (2-propanol) was observed. Compounds that were unsubstituted at C-2 were found to be up to 300 times more potent as cytotoxins than their 2-alkyl-substituted analogues in V79-379A cells, but with lower hypoxic cytotoxicity ratios.


Subject(s)
Antineoplastic Agents , Indoles , Quinones , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/radiation effects , Cell Death/drug effects , Cell Hypoxia , Chromatography, High Pressure Liquid , Cricetinae , Drug Screening Assays, Antitumor , Free Radicals/chemistry , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacology , Indoles/radiation effects , Kinetics , Oxidation-Reduction , Pulse Radiolysis , Quinones/chemical synthesis , Quinones/chemistry , Quinones/pharmacology , Quinones/radiation effects , Structure-Activity Relationship , Tumor Cells, Cultured
20.
Free Radic Biol Med ; 24(1): 1-10, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9436608

ABSTRACT

The oxidative denitrification of the antitumour agent hydroxyguanidine (HOG) has been investigated by radiolysis methods and EPR spectroscopy. The azide radical (N3.), a model one-electron oxidant, reacts with HOG with the rate constant 5.1 x 10(9) dm3 mol(-1) s(-1) to yield the guanidino carbon-centred radical (HOG.) which rapidly eliminates nitric oxide (k = 3.1 x 10[3] s[-1]) with the concomitant formation of urea. The HOG. undergoes conjugation with molecular oxygen to form a peroxyl radical (HOGOO.) with a rate constant 8.8 x 10(8) dm3 mol(-1) s(-1). The HOGOO. radical also eliminates nitric oxide but may act as a precursor to the peroxynitrite (ONOO-) ion. The oxidation of HOG by the dibromide radical (Br2.-) was found to release nitric oxide with a yield of 95% relative to Br2.- as determined from the combined yields of inorganic nitrite, nitrate and a HOG/nitric oxide-adduct. This study provides a possible mechanistic basis for the oxidative denitrification of HOG which may contribute to the observed toxicity of the drug both in vitro and in vivo and for the oxidation of nonphysiological hydroxyguanidines to NO. via nitric oxide synthase-independent pathways.


Subject(s)
Antineoplastic Agents/metabolism , Guanidines/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Catalysis , Electron Spin Resonance Spectroscopy , Free Radicals , Hydroxylamines , Linear Models , Oxidation-Reduction , Peroxidase/metabolism , Pulse Radiolysis
SELECTION OF CITATIONS
SEARCH DETAIL
...