Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(11): e82111, 2013.
Article in English | MEDLINE | ID: mdl-24260588

ABSTRACT

A novel methodology is described for the efficient and divergent synthesis of pseudodisaccharides, molecules comprising of amino carbasugar analogues linked to natural sugars. The methodology is general and enables the introduction of diversity both at the carbasugar and the natural sugar components of the pseudodisaccharides. Using this approach, a series of pseudodisaccharides are synthesised that mimic the repeating backbone unit of heparan sulfate, and are tested for inhibition of heparanase, a disease-relevant enzyme that hydrolyses heparan sulfate. A new homology model of human heparanase is described based on a family 79 ß-glucuronidase. This model is used to postulate a computational rationale for the observed activity of the different pseudodisaccharides and provide valuable information that informs the design of potential inhibitors of this enzyme.


Subject(s)
Disaccharides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Glucuronidase/chemistry , Heparitin Sulfate/chemistry , Catalytic Domain , Disaccharides/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Glucuronidase/antagonists & inhibitors , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Mimicry , Small Molecule Libraries
2.
Curr Cancer Drug Targets ; 12(8): 925-39, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22463390

ABSTRACT

Polysialic acid (polySia) is a carbohydrate polymer critical for neuronal cell migration and axon pathfinding in embryonic development. Besides brain regions requiring persistent neuronal plasticity, polySia is essentially absent from the adult body. However, polySia is aberrantly re-expressed on many tumours, where it decorates the surface of NCAM (neuronal cell adhesion molecule) and modulates cell adhesion, migration and invasion. PolySia-NCAM expression is strongly associated with poor clinical prognosis and correlates with aggressive and invasive disease in many cancers, including lung cancer, neuroblastoma and gliomas. The synthesis of polySia is mediated by two polysialyltransferases (polySTs), ST8SiaIV (PST) and particularly ST8SiaII (STX) in cancer cells. The demonstration that polyST knock-down negates events associated with tumour cell dissemination indicates that PST and STX are validated targets. Selective inhibition of polySTs therefore presents a therapeutic opportunity to inhibit tumour invasion and metastasis.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Sialic Acids/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/metabolism , Animals , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Drug Design , Enzyme Inhibitors/pharmacology , Glioma/metabolism , Glioma/pathology , Glycocalyx/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasms/drug therapy , Neural Cell Adhesion Molecules/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Prognosis , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Sialic Acids/biosynthesis , Sialyltransferases/genetics
3.
J Proteome Res ; 11(5): 2863-75, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22416669

ABSTRACT

Three dimensional multicell tumor spheroids (MCTS) provide an experimental model where the influence of microenvironmental conditions on protein expression can be determined. Sequential trypsin digestion of HT29 colon carcinoma MCTS enabled segregation into four populations comprising proliferating cells from the surface (SL), an intermediate region (IR), nonproliferating hypoxic cells from the perinecrotic region (PN), and a necrotic core (NC). Total protein was extracted from each population and subjected to iTRAQ-based quantitative proteomics analysis. From a total of 887 proteins identified, 209 were observed to be up-regulated and 114 were down-regulated in the PN and NC regions relative to the SL. Among the up-regulated proteins, components of glycolysis, TCA cycle, lipid metabolism, and steroid biosynthesis increased progressively toward the PN and NC regions. Western blotting, immunohistochemistry, and enzyme assays confirmed that significant changes in the expression of proteins involved in cellular metabolism occur in the nonproliferating fraction of cells within the viable rim. The presence of full length, functional proteins within the NC was unexpected, and further analysis demonstrated that this region contains cells that are undergoing autophagy. This study has identified possible targets that may be suitable for therapeutic intervention, and further studies to validate these are required.


Subject(s)
Proteome/metabolism , Proteomics/methods , Spheroids, Cellular/metabolism , Autophagy , Cell Proliferation , Citric Acid Cycle , Enzyme Activation , Enzyme Assays , Glycolysis , HT29 Cells , Humans , Hypoxia/metabolism , Immunohistochemistry , Lipid Metabolism , Malate Dehydrogenase/metabolism , Proteolysis , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spheroids, Cellular/pathology , Trypsin/metabolism , Tumor Cells, Cultured , Tumor Microenvironment
4.
Curr Cancer Drug Targets ; 9(4): 519-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519320

ABSTRACT

The integrin family of cell surface receptors integrates cell-extracellular matrix interactions with the cell cytoskeleton and signalling across the cell membrane, resulting in an important role in cell adhesion, mobility and migration, proliferation, and survival. Changes in the number and identity of integrin receptors are common in cancer cells resulting in alteration of the ability of malignant cells to interact with the extracellular matrix, and promoting migration as well as facilitating survival outside the tumour normal environment. beta(3) integrins are potentially involved in every step of the metastatic process and expression of both alpha(IIb)beta(3) and alpha(n)beta(3) is correlated with metastatic ability of tumour cells. The recognition of the RGD binding motif common to the disintegrins and natural integrin ligands such as fibrinogen allowed the development of small molecule beta(3) integrin antagonists, progressing from linear peptides containing the RGD sequence to cyclic peptides with well-defined conformation, and hence to small molecule peptidomimetics with improved pharmacological properties. In this review, we summarize the role of the beta(3)-subfamily of integrins when expressed in normal and tumour tissue, the development of small-molecule antagonists of beta(3) integrins and their potential anti-cancer applications.


Subject(s)
Integrin beta3/physiology , Integrin beta3/therapeutic use , Integrins/antagonists & inhibitors , Neoplasm Metastasis/physiopathology , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Disintegrins/pharmacology , Drug Delivery Systems , Humans , Integrin beta3/genetics , Integrins/physiology , Models, Biological , Molecular Structure , Neoplasms/metabolism , Peptides/pharmacology , Peptides, Cyclic/pharmacology , Polymorphism, Single Nucleotide
5.
Ann Oncol ; 18(6): 1098-103, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442658

ABSTRACT

BACKGROUND: AQ4N (1,4-bis[[2-(dimethylamino)ethyl] amino]-5,8-dihydroxyanthracene-9, 10-dione bis-N-oxide dihydrochloride) is a prodrug which is selectively activated within hypoxic tissues to AQ4, a topoisomerase II inhibitor and DNA intercalator. PATIENTS AND METHODS: In the phase I study, 22 patients with oesophageal carcinoma received an i.v. infusion of AQ4N (22.5-447 mg/m(2)) followed, 2 weeks later, by further infusion and radiotherapy. Pharmacokinetics and lymphocyte AQ4N and AQ4 levels were measured after the first dose. At 447 mg/m(2), biopsies of tumour and normal tissue were taken after AQ4N administration. RESULTS: Drug-related adverse events were blue discolouration of skin and urine, grade 2-3 lymphopenia, grade 1-3 fatigue, grade 1-2 anaemia, leucopenia and nausea. There were no drug-related serious adverse events (SAEs). Three patients had reductions in tumour volume >50%, nine had stable disease. Pharmacokinetics indicated predictable clearance. Plasma area under the curve (AUC) at 447 mg/m(2) exceeded AQ4N concentrations in mice at therapeutic doses and tumour biopsies contained concentrations of AQ4 greater than those in normal tissue. Tumour concentrations of AQ4 exceeded in vitro IC(50) values for most cell lines investigated. CONCLUSIONS: No dose-limiting toxic effects were observed and a maximum tolerated dose was not established. Tumour AQ4 concentrations and plasma AUC at 447 mg/m(2) exceeded active levels in preclinical models. This dose was chosen for future studies with radiotherapy.


Subject(s)
Anthraquinones/pharmacokinetics , Anthraquinones/toxicity , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/radiotherapy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Aged , Aged, 80 and over , Anthraquinones/administration & dosage , Area Under Curve , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Combined Modality Therapy , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Patient Selection
6.
Cancer Gene Ther ; 13(6): 598-605, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16410820

ABSTRACT

Drug metabolizing transgene products, which activate bioreductive cytotoxins, can be used to target treatment-resistant hypoxic tumors. The prodrug AQ4N is bioreduced in hypoxic cells by cytochrome P450s (CYPs) to the cytotoxin AQ4. Previously we have shown that intra-tumoral injection of CYP3A4 and CYP2B6 transgenes with AQ4N and radiation inhibits tumor growth. Here we examine the ability of other CYPs, in particular CYP1A1, to metabolize AQ4N, and to enhance radiosensitization. Metabolism of AQ4N was assessed using microsomes prepared from baculovirus-infected cells transfected with various CYP isoforms. AQ4N metabolism was most efficient with CYP1A1 (66.7 nmol/min/pmol) and 2B6 (34.4 nmol/min/pmol). Transient transfection of human CYP1A1+/-CYP reductase (CYPRED) was investigated in hypoxic RIF-1 mouse cells in vitro using the alkaline comet assay. There was a significant increase in DNA damage following transient transfection of CYP1A1 compared to non-transfected cells; inclusion of CYPRED provided no additional effect. In vivo, a single intra-tumoral injection of a CYP1A1 construct in combination with AQ4N (100 mg/kg i.p.) and 20 Gy X-rays caused a 16-day delay in tumor regrowth compared to tumors receiving AQ4N plus radiation and empty vector (P=0.0344). The results show the efficacy of a CYP1A1-mediated GDEPT strategy for bioreduction of AQ4N.


Subject(s)
Anthraquinones/pharmacology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/pharmacology , DNA, Neoplasm/drug effects , Genetic Therapy/methods , Prodrugs/pharmacology , Animals , Anthraquinones/metabolism , Blotting, Western , Cell Hypoxia/drug effects , Cell Line, Tumor , Combined Modality Therapy , Cytochrome P-450 CYP1A1/metabolism , DNA Damage/drug effects , Mice , Molecular Structure , Prodrugs/metabolism , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/pharmacology , Radiotherapy , Tumor Cells, Cultured
7.
Adv Drug Deliv Rev ; 57(1): 153-67, 2005 Jan 02.
Article in English | MEDLINE | ID: mdl-15518927

ABSTRACT

Many anticancer drugs require interaction with DNA or chromatin components of tumor cells to achieve therapeutic activity. Quantification and exploration of drug targeting dynamics can be highly informative in the rational development of new therapies and in the drug discovery pipeline. The problems faced include the potential infrequency and transient nature of critical events, the influence of micropharmacokinetics on the drug-target equilibria, the dependence on preserving cell function to demonstrate dynamic processes in situ, the need to map events in functional cells and the confounding effects of cell-to-cell heterogeneity. We demonstrate technological solutions in which we have integrated two-photon laser scanning microscopy (TPLSM) to track drug delivery in subcellular compartments, with the mapping of sites of critical molecular interactions. We address key design concepts for the development of modular tools used to uncover the complexity of drug targeting in single cells. First, we describe the combination of two-photon excitation with fluorescence lifetime imaging microscopy (FLIM) to map the nuclear docking of the anticancer drug topotecan (TPT) at a subset of DNA sites in nuclear structures of live breast tumor cells. Secondly, we demonstrate how we incorporate the smart design of a two-photon 'dark' DNA binding probe, such as DRAQ5, as a well-defined quenching probe to uncover sites of drug interaction. Finally, we discuss the future perspectives on introducing these modular kinetic assays in the high-content screening arena and the interlinking of the consequences of drug-target interactions with cellular stress responses.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Microscopy, Fluorescence/methods , Models, Molecular , Technology, Pharmaceutical , Topotecan/administration & dosage , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Humans , Topotecan/metabolism , Topotecan/pharmacokinetics
8.
Eur J Cancer ; 40(14): 2127-34, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15341988

ABSTRACT

We describe the direct identification of multiple cytochrome P450 (CYP) enzymes in healthy and cancerous tissue. CYPs in human liver colorectal metastases were compared with those in the surrounding liver using a mass spectrometry-based proteomic approach. Nano-scale reversed phase liquid chromatography combined with electrospray ionisation tandem mass spectrometry has been used to identify CYPs with no pre-selection of the proteins anticipated. Fourteen distinct CYP enzymes from the subfamilies 1A, 2A, 2B, 2C, 2D, 2E, 3A, 4A, 4F, 8B and 27A were positively identified; 13 in the liver samples and 12 in the tumour tissue. It was found that three of the colorectal metastases expressed essentially the same drug-metabolising pattern of CYPs as the surrounding liver, whilst three tumour samples from different individuals showed no CYP expression. This was likely in at least one case to be due to low sample mass. The CYP expression profile in an individual tumour is likely to be an important determinant in predicting the outcome of cancer chemotherapy.


Subject(s)
Colorectal Neoplasms/enzymology , Cytochrome P-450 Enzyme System/metabolism , Liver Neoplasms/enzymology , Adult , Aged , Colorectal Neoplasms/secondary , Electrophoresis, Polyacrylamide Gel , Female , Humans , Male , Microsomes, Liver/enzymology , Middle Aged , Proteome/metabolism
9.
Drug Metab Dispos ; 32(4): 382-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039290

ABSTRACT

Nanoscale reversed-phase liquid chromatography (LC) combined with electrospray ionization-tandem mass spectrometry (ESI-MS/MS) has been used as a method for the direct identification of multiple cytochrome P450 (P450) isoforms found in male and female rat liver. In this targeted proteomic approach, rat liver microsomes were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis followed by in-gel tryptic digestion of the proteins present in the 48- to 62-kDa bands. The resultant peptides were extracted and analyzed by LC-ESI-MS/MS. P450 identifications were made by searching the MS/MS data against a rat protein database containing 21,576 entries including 47 P450s using Sequest software (Thermo Electron, Hemel Hempstead, UK). Twenty-four P450 isoforms from the subfamilies 1A, 2A, 2B, 2C, 2D, 2E, 3A, 4A, 4F, CYP17, and CYP19 were positively identified in rat liver.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Microsomes, Liver/chemistry , Nanotechnology/methods , Protein Isoforms/chemistry , Proteomics/methods , Amino Acid Sequence , Animals , Chromatography, Liquid/methods , Cytochrome P-450 Enzyme System/isolation & purification , Female , Male , Microsomes, Liver/enzymology , Protein Isoforms/isolation & purification , Rats , Sex Factors , Spectrometry, Mass, Electrospray Ionization/methods
10.
Curr Pharm Des ; 8(15): 1335-47, 2002.
Article in English | MEDLINE | ID: mdl-12052211

ABSTRACT

The expression of drug metabolising cytochrome P450s (CYPs) notably 1A, 1B, 2C, 3A, 2D subfamily members have been identified in a wide range of human cancers. Individual tumour types have distinct P450 profiles as studied by detection of P450 activity, identification of immunoreactive CYP protein and detection of CYP mRNA. Selected P450s, especially CYP1B1, are overexpressed in tumours including cancers of the lung, breast, liver, gastrointestinal tract, prostate, bladder. Several prodrug anti-tumour agents have retrospectively been identified as P450 substrates for which tumour CYP activation may hitherto have been underestimated. Those in clinical use include prodrug alkylating agents (cyclophosphamide, ifosphamide, dacarbazine, procarbazine), Tegafur, a prodrug fluoropyrimidine, methoxymorphylinodoxorubicin, a metabolically activated anthracycline, as well as flutamide and tamoxifen, two non-steroidal hormone receptor antagonists that are significantly more active following CYP-hydroxylation. More exciting is the prospect of developing new agents designed to be selectively dependent on tumour CYP activation. This can be illustrated with P450 activation of the 2-(4-aminophenyl)benzothiazoles exclusively in CYP1A1 inducible tumours. Also of interest is the bioreductive antitumour prodrug AQ4N, a CYP3A substrate that is activated to a cytotoxic metabolite specifically in hypoxic tumour regions.


Subject(s)
Antineoplastic Agents/chemistry , Cytochrome P-450 Enzyme System/biosynthesis , Neoplasms/metabolism , Prodrugs/chemistry , Antineoplastic Agents/metabolism , Cytochrome P-450 Enzyme System/analysis , Drug Design , Humans , Isoenzymes/analysis , Isoenzymes/biosynthesis , Oxidation-Reduction , Prodrugs/metabolism , Substrate Specificity
11.
Br J Cancer ; 86(5): 774-8, 2002 Mar 04.
Article in English | MEDLINE | ID: mdl-11875742

ABSTRACT

Resveratrol is a cancer preventative agent that is found in red wine. Piceatannol is a closely related stilbene that has antileukaemic activity and is also a tyrosine kinase inhibitor. Piceatannol differs from resveratrol by having an additional aromatic hydroxy group. The enzyme CYP1B1 is overexpressed in a wide variety of human tumours and catalyses aromatic hydroxylation reactions. We report here that the cancer preventative agent resveratrol undergoes metabolism by the cytochrome P450 enzyme CYP1B1 to give a metabolite which has been identified as the known antileukaemic agent piceatannol. The metabolite was identified by high performance liquid chromatography analysis using fluorescence detection and the identity of the metabolite was further confirmed by derivatisation followed by gas chromatography-mass spectrometry studies using authentic piceatannol for comparison. This observation provides a novel explanation for the cancer preventative properties of resveratrol. It demonstrates that a natural dietary cancer preventative agent can be converted to a compound with known anticancer activity by an enzyme that is found in human tumours. Importantly this result gives insight into the functional role of CYP1B1 and provides evidence for the concept that CYP1B1 in tumours may be functioning as a growth suppressor enzyme.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents/chemistry , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Stilbenes/chemistry , Stilbenes/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Chemoprevention , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1B1 , Gas Chromatography-Mass Spectrometry , Humans , Neoplasms/enzymology , Neoplasms/prevention & control , Resveratrol , Stilbenes/pharmacology , Tumor Cells, Cultured , Wine
12.
Br J Cancer ; 85(4): 625-9, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11506506

ABSTRACT

AQ4N is a bioreductive drug that can significantly enhance the anti-tumour effect of radiation and cyclophosphamide. The aim of this study was to examine the ability of AQ4N to potentiate the anti-tumour effect of cisplatin and to compare it to the chemopotentiation effect of tirapazamine. In the T50/80 murine tumour model, AQ4N (50-100 mg/kg) was administered 30 min, 2.5 or 6 h prior to cisplatin (4 mg/kg or 8 mg/kg); this produced an anti-tumour effect that was approximately 1.5 to 2 times greater than that achieved by a single 4 or 8 mg/kg dose of cisplatin. Tirapazamine (25 mg/kg) administered 2.5 h prior to cisplatin (4 mg/kg) resulted in a small increase in anti-tumour efficacy. AQ4N was also successful in enhancing the anti-tumour effect of cisplatin in the SCCVII and RIF-1 murine tumour models. This resulted in an increased cell kill of greater than 3 logs in both models; this was a greater cell kill than that observed for tirapazamine with cisplatin. Combination of cisplatin with AQ4N or tirapazamine resulted in no additional bone marrow toxicity compared to cisplatin administered alone. In conclusion, AQ4N has the potential to improve the clinical efficacy of cisplatin.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Prodrugs/pharmacology , Animals , Anthraquinones/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , Disease Models, Animal , Drug Interactions , Drug Screening Assays, Antitumor , Female , Mammary Neoplasms, Animal/drug therapy , Mice , Prodrugs/pharmacokinetics , Tirapazamine , Triazines/pharmacokinetics , Triazines/pharmacology
13.
Biochem Pharmacol ; 62(4): 461-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11448456

ABSTRACT

Evidence suggests that DT-diaphorase is involved in the activation and mechanism of cytotoxicity of the investigational indoloquinone anticancer drug EO9 under aerobic conditions. Data also implicate a role for other enzymes including NADPH: cytochrome P450 reductase, especially in low DT-diaphorase tumour cells and under hypoxic conditions. Here, we used purified rat NADPH: cytochrome P450 reductase to provide additional evidence in support of a role for this enzyme in activation of EO9 to generate free radical and DNA-damaging species. Electron spin resonance spectrometry studies showed that NADPH: cytochrome P450 reductase reduced EO9 to a free radical species, including a drug radical (most likely the semiquinone) and reactive oxygen species. Plasmid DNA experiments showed that reduction of EO9 catalysed by NADPH: cytochrome P450 reductase results in single-strand breaks in DNA. The information obtained may contribute to the understanding of the molecular mechanism of DNA damage and cytotoxicity exerted by EO9 and may be useful in the design of future bioreductive drugs.


Subject(s)
Aziridines/metabolism , DNA/drug effects , Free Radicals/pharmacology , Indolequinones , Indoles/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Catalysis , DNA/metabolism , DNA Damage , Electron Spin Resonance Spectroscopy , Indoles/pharmacology , Rats
14.
Drug Metab Dispos ; 29(4 Pt 1): 422-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259326

ABSTRACT

AQ4N (1,4-bis-[[2-(dimethylamino-N-oxide)ethyl]amino]5,8-dihydroxyanthracene-9,10-dione) is in a class of bioreductive agents incorporating the aliphatic N-oxide functionality and is well documented as a very effective enhancer of radiotherapy and chemotherapy. The compound is shortly to enter Phase I clinical trials in the United Kingdom, and this study describes the preclinical pharmacokinetics and metabolism of AQ4N in mice. AQ4N was administered by i.v. injection at doses of 200, 100, and 20 mg/kg and was quantified by high-performance liquid chromatography and liquid chromatography/mass spectroscopy. There was a linear increase in the maximum plasma concentration (Cmax) proportional to dose with a Cmax of 1171 microg/ml at the maximum tolerated dose of 200 mg/kg. The area under plasma concentration versus time curve (AUC) increased disproportionately with dose from 14.1 microg/h/ml at 20 mg/kg to 247 microg/h/ml at 200 mg/kg with a subsequent decrease in clearance. Terminal elimination half-lives ranged from 0.64 to 0.83 h. The spectra of the two major metabolites matched those from authentic standards with the molecular ions [M + H]+ being detected at m/z 445.4 (AQ4N), m/z 429.5 (AQ4 mono-N-oxide) and m/z 413.5 (AQ4). Only low concentrations of the toxic metabolite (AQ4) were detected in plasma at all three doses, with the AUC and Cmax at 200 mg/kg being 3.54 microg/h/ml and 3.7 microg/ml, respectively, representing <2% of AQ4N. Concentrations of the intermediate AQ4 M represented 8, 10, and 18% of those for AQ4N at the doses of 20,100, and 200 mg/kg. The concentrations necessary for a therapeutic response in vivo have been described in this pharmacokinetic study.


Subject(s)
Anthraquinones/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Anthraquinones/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Female , Mice , Prodrugs/metabolism , Radiation-Protective Agents/metabolism , Radiation-Protective Agents/pharmacokinetics
15.
Bioorg Med Chem Lett ; 11(3): 351-3, 2001 Feb 12.
Article in English | MEDLINE | ID: mdl-11212108

ABSTRACT

Peptide-1-[N-[2-succinamidylethyl]amino]anthraquinones containing five seven amino acid residues including the KCR motif important in AP-1 protein binding to DNA have been synthesised as potential transcription factor inhibitors. These anthraquinone-peptides showed DNA intercalative binding and inhibition of AP-1 protein binding to its DNA consensus sequence.


Subject(s)
Anthraquinones/pharmacology , Peptide Fragments/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Amino Acid Motifs , Anthraquinones/chemical synthesis , Binding Sites , Combinatorial Chemistry Techniques , Consensus Sequence , DNA/metabolism , Electrophoresis , Humans , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Binding/drug effects , Structure-Activity Relationship , Temperature
16.
Br J Cancer ; 83(12): 1589-93, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11104551

ABSTRACT

Preclinical studies demonstrate that in vivo AQ4N enhances the anti-tumour effects of radiation and chemotherapeutic agents with a dose-modifying factor of approximately 2.0. With careful scheduling no, or very little, additional normal tissue toxicity should be observed. AQ4N is a bioreductive prodrug of a potent, stable, reduction product which binds non-covalently to DNA, facilitating antitumour activity in both hypoxic and proximate oxic tumour cells. AQ4N is clearly different in both its mechanism of action and potential bystander effect compared to previously identified bioreductive drugs. In particular AQ4N is the only bioreductive prodrug topoisomerase II inhibitor to enter clinical trials. Targeting this enzyme, which is crucial to cell division, may help sensitize tumours to repeated (fractionated) courses of radiotherapy. This is because in principle, the bioreduction product of AQ4N can inhibit the topoisomerase activity of hypoxic cells as they attempt to re-enter the cell cycle.


Subject(s)
Anthraquinones/therapeutic use , Antineoplastic Agents/therapeutic use , Prodrugs/therapeutic use , Radiation-Protective Agents/therapeutic use , Animals , Cell Hypoxia , Humans , Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy
17.
Biochem Pharmacol ; 60(12): 1933-5, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11108810

ABSTRACT

Tirapazamine (SR 4233) is a bioreductive antitumour drug in Phase III clinical trial which is activated in hypoxic tumour regions to generate a cytotoxic species. Electron paramagnetic resonance (EPR) spectrometry was used to investigate directly the formation of free radicals as the result of tirapazamine reduction by NADPH-supplemented liver microsomes. Under anaerobic conditions, the tirapazamine nitroxide free radical EPR signal was not evident over a range of rat or human liver microsomal protein (1-5 mg) concentrations. However, in combination with 1,1',5, 5'-dimethylpyrolline-1-N-oxide (DMPO), a spin trap for short-lived free radicals, tirapazamine resulted in formation of a 1:1:1:1:1:1 spectrum with hyperfine splitting A(N) = 15.8 G A(H) = 22.3 G consistent with generation of DMPO-R, a carbon-centered radical adduct. Addition of DMSO increased the signal intensity of the carbon-centred radical by at least twofold. The hyperfine splitting constants associated with DMPO-R could be indicative of a tirapazamine carbon-centred radical per se or, more likely, carbon radicals from endogenous materials (or DMSO) in the biological matrix as a result of oxidative attack by the tirapazamine primary radical. Formation of DMPO-OH, the hydroxyl radical spin adduct, by tirapazamine in the absence of air indicates that liberation of a hydroxyl radical may be a consequence of tirapazamine bioreduction under anaerobic conditions. The reactivity of tirapazamine free radicals with endogenous microsomal substances to generate reactive carbon-centred radicals indicates that tirapazamine may disrupt a wide range of cellular activities.


Subject(s)
Antineoplastic Agents/metabolism , Free Radicals/metabolism , Microsomes, Liver/metabolism , Triazines/metabolism , Anaerobiosis , Animals , Electron Spin Resonance Spectroscopy/methods , In Vitro Techniques , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Time Factors , Tirapazamine
18.
Cytometry ; 40(4): 280-91, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10918279

ABSTRACT

BACKGROUND: The multiparameter fluorometric analysis of intact and fixed cells often requires the use of a nuclear DNA discrimination signal with spectral separation from visible range fluorochromes. We have developed a novel deep red fluorescing bisalkylaminoanthraquinone, DRAQ5 (Ex(lambdamax) 646 nm; Em(lambdamax) 681 nm; Em(lambdarange) 665->800 nm), with high affinity for DNA and a high capacity to enter living cells. We describe here the spectral characteristics and applications of this synthetic compound, particularly in relation to cytometric analysis of the cell cycle. METHODS: Cultured human tumor cells were examined for the ability to nuclear locate DRAQ5 using single and multiphoton laser scanning microscopy (LSM) and multiparameter flow cytometry. RESULTS: Multiparameter flow cytometry shows that the dye can rapidly report the cellular DNA content of live and fixed cells at a resolution level adequate for cell cycle analysis and the cycle-specific expression of cellular proteins (e.g., cyclin B1). The preferential excitation of DRAQ5 by laser red lines (633/647 nm) was found to offer a means of fluorescence signal discrimination by selective excitation, with greatly reduced emission overlap with UV-excitable and visible range fluophors as compared with propidium iodide. LSM reveals nuclear architecture and clearly defines chromosomal elements in live cells. DRAQ5 was found to permit multiphoton imaging of nuclei using a 1,047-nm emitting mode-locked YLF laser. The unusual spectral properties of DRAQ5 also permit live cell DNA analysis using conventional 488 nm excitation and the single-photon imaging of nuclear fluorescence using laser excitation between 488 nm and low infrared (IR; 780 nm) wavelengths. Single and multiphoton microscopy studies revealed the ability of DRAQ5 to report three-dimensional nuclear structure and location in live cells expressing endoplasmic reticulum targeted-GFP, MitoTracker-stained mitochondria, or a vital cell probe for free zinc (Zinquin). CONCLUSION: The fluorescence excitation and emission characteristics of DRAQ5 in living and fixed cells permit the incorporation of the measurement of cellular DNA content into a variety of multiparameter cytometric analyses.


Subject(s)
DNA Probes , DNA, Neoplasm/analysis , Fluorescent Dyes , Melanoma/genetics , Nitrogen Oxides , Anthraquinones , CDC2 Protein Kinase/metabolism , Cell Cycle , Cyclin B/metabolism , Cyclin B1 , Diagnostic Imaging/methods , Flow Cytometry/methods , Fluorescent Antibody Technique , Gene Expression , Green Fluorescent Proteins , Humans , Infrared Rays , Luminescent Proteins/metabolism , Melanoma/metabolism , Melanoma/pathology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Molecular Structure , Quinolones/metabolism , Spectrophotometry/methods , Tosyl Compounds/metabolism , Tumor Cells, Cultured
19.
J Chromatogr B Biomed Sci Appl ; 742(2): 239-45, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10901128

ABSTRACT

A simple, highly selective and reproducible reversed-phase high-performance liquid chromatography method has been developed for the analysis of the new anti-cancer pro-drug AQ4N. The sample pre-treatment involves a simple protein precipitation protocol, using methanol. Chromatographic separations were performed using a HiChrom HIRPB (25 cmX4.6 mm I.D.) column, with mobile phase of acetonitrile-ammonium formate buffer (0.05 M) (22:78, v/v), with final pH adjusted to 3.6 with formic acid. The flow-rate was maintained at 1.2 ml min(-1). Detection was via photodiode array performed in the UV range at 242 nm and, since the compounds are an intense blue colour, in the visible range at 612 nm. The structurally related compound mitoxantrone was used as internal standard. The validated quantification range of the method was 0.05-10.0 microg ml(-1) in mouse plasma. The inter-day relative standard deviations (RSDs) (n=5) ranged from 18.4% and 12.1% at 0.05 microg ml(-1) to 2.9% and 3.3% at 10.0 microg ml(-1) for AQ4N and AQ4, respectively. The intra-day RSDs for supplemented mouse plasma (n=6) ranged from 8.2% and 14.2% at 0.05 microg ml(-1) to 7.6% and 11.5% at 10.0 microg ml(-1) for AQ4N and AQ4, respectively. The overall recovery of the procedure for AQ4N was 89.4 +/- 1.77% and 76.1 +/- 7.26% for AQ4. The limit of detection was 50 ng ml(-1) with a 100 microl sample volume. The method described provides a suitable technique for the future analysis of low levels of AQ4N and AQ4 in clinical samples.


Subject(s)
Anthraquinones/blood , Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Prodrugs/pharmacokinetics , Animals , Anthraquinones/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Blood Proteins/metabolism , Calibration , Female , Mice , Protein Binding , Reference Standards
20.
Br J Cancer ; 82(12): 1984-90, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10864207

ABSTRACT

AQ4 (1,4-Bis-[[2-(dimethylamino-N-oxide)ethyl]amino]5,8-dihydroxyanthrace ne-9, 10-dione) is a prodrug designed to be excluded from cell nuclei until bioreduced in hypoxic cells to AQ4, a DNA intercalator and topoisomerase II poison. Thus, AQ4N is a highly selective bioreductive drug that is activated in, and is preferentially toxic to, hypoxic cells in tumours. Five murine tumours (MAC16, MAC26, NT, SCCVII and RIF-1) have been used to investigate the anti-tumour effects of AQ4N. In only one tumour (MAC16) was AQ4N shown to be active as a single agent. However, when combined with methods to increase the hypoxic tumour fraction in RIF-1 (by physical clamping) and MAC26 tumours (using hydralazine) there was a substantial enhancement in anti-tumour effect. Notably, RIF-1 tumours treated with AQ4N (250 mg kg(-1)) followed 15 min later by physically occluding the blood supply to the tumour for 90 min, resulted in a 13-fold increase in growth delay. When combined with radiation or chemotherapy, AQ4N substantially increased the effectiveness of these modalities in a range of in vivo model systems. AQ4N potentiates the action of radiation in both a drug and radiation dose-dependent manner. Further the enhancement observed is schedule-independent with AQ4N giving similar effects when given at any time within 16 h before or after the radiation treatment. In combination with chemotherapy it is shown that AQ4N potentiates the activity of cyclophosphamide, cisplatin and thiotepa. Both the chemotherapeutic drugs and AQ4N are given at doses which individually are close to their estimated maximum tolerated dose (data not included) which provides indirect evidence that in the combination chemotherapy experiments there is some tumour selectivity in the enhanced action of the drugs.


Subject(s)
Anthraquinones/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Hypoxia/drug effects , Drug Administration Schedule , Drug Synergism , Male , Mice , Mice, Inbred Strains , Prodrugs/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...