Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
BMC Cardiovasc Disord ; 14: 44, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24708700

ABSTRACT

BACKGROUND: Optimal treatment of acute ST-elevation myocardial infarction (STEMI) involves rapid diagnosis, and transfer to a cardiac centre capable of percutaneous coronary intervention (PCI) for immediate mechanical revascularisation. Successful treatment requires rapid return of perfusion to the myocardium achieved by thromboaspiration, passivation of the culprit lesion with stent scaffolding and systemic inhibition of thrombosis and platelet activation. A delicate balance exists between thrombosis and bleeding and consequently anti-thrombotic and antiplatelet treatment regimens continue to evolve. The desire to achieve reperfusion as soon as possible, in the setting of high platelet reactivity, requires potent and fast-acting anti-thrombotic/anti-platelet therapies. The associated bleeding risk may be minimised by use of short-acting anti-thrombotic intravenous agents. However, effective oral platelet inhibition is required to prevent recurrent thrombosis. The interaction between baseline platelet reactivity, timing of revascularisation and effective inhibition of thrombosis is yet to be formally investigated. METHODS/DESIGN: We present a protocol for a prospective observational study in patients presenting with acute STEMI treated with primary PCI (PPCI) and receiving bolus/infusion bivalirudin and prasugrel therapy. The objective of this study is to describe variation in platelet reactivity, as measured by the multiplate platelet function analyser, at presentation, the end of the PPCI procedure and 1, 2, & 24 hours post-procedure. We intend to assess the prevalence of high residual platelet reactivity within 24 hours of PPCI in acute STEMI patients receiving prasugrel and bivalirudin. Additionally, we will investigate the association between high platelet reactivity before and after PPCI and the door-to-procedure completion time.This is a single centre study with a target sample size of 108 participants. DISCUSSION: The baseline platelet reactivity on presentation with a STEMI may impact on the effect of acute anti-thrombotic and anti-platelet therapy and expose patients to a heightened risk of bleeding or ongoing thrombosis. This study will define the baseline variation in platelet reactivity in a population of patients experiencing acute STEMI and assess the pharmacodynamic response to combined treatment with bivalirudin and prasugrel. The data obtained from this trial will be hypothesis generating for future trials testing alternative pharmacotherapies in the acute phase of treatment for STEMI. TRIAL REGISTRATION: This study has approval from Wiltshire research ethics committee (10/H0106/87) and is registered with current controlled trials (http://www.controlled-trials.com/ISRCTN82257414).


Subject(s)
Blood Platelets/drug effects , Drug Monitoring/methods , Myocardial Infarction/therapy , Peptide Fragments/therapeutic use , Percutaneous Coronary Intervention , Piperazines/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests , Point-of-Care Systems , Research Design , Thiophenes/therapeutic use , Blood Platelets/metabolism , Clinical Protocols , Coronary Thrombosis/blood , Coronary Thrombosis/etiology , Coronary Thrombosis/prevention & control , Drug Therapy, Combination , England , Hemorrhage/chemically induced , Hirudins/adverse effects , Humans , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Peptide Fragments/adverse effects , Percutaneous Coronary Intervention/adverse effects , Piperazines/adverse effects , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Prasugrel Hydrochloride , Predictive Value of Tests , Prospective Studies , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Thiophenes/adverse effects , Time Factors , Treatment Outcome
4.
PLoS One ; 7(1): e30389, 2012.
Article in English | MEDLINE | ID: mdl-22272341

ABSTRACT

BACKGROUND: The number and functional activity of circulating progenitor cells (CPCs) is altered in diabetic patients. Furthermore, reduced CPC count has been shown to independently predict cardiovascular events. Validation of CPCs as a biomarker for cardiovascular risk stratification requires rigorous methodology. Before a standard operation protocol (SOP) can be designed for such a trial, a variety of technical issues have to be addressed fundamentally, which include the appropriate type of red blood cell lysis buffer, FMO or isotype controls to identify rare cell populations from background noise, optimal antibody dilutions and conditions of sample storage. We herein propose improvements in critical steps of CPC isolation, antigenic characterization and determination of functional competence for final application in a prospective investigation of CPCs as a biomarker of outcome following acute myocardial infarction. METHODS AND FINDINGS: In this validation study, we refined the standard operating procedure (SOP) for flow cytometry characterisation and functional analysis of CPCs from the first 18 patients of the Progenitor Cell Response after Myocardial Infarction Study (ProMIS). ProMIS aims to verify the prognostic value of CPCs in patients with either ST elevation or non-ST elevation myocardial infarction with or without diabetes mellitus, using cardiac magnetic resonance imaging (MRI) for assessment of ventricular function as a primary endpoint. Results indicate crucial steps for SOP implementation, namely timely cell isolation after sampling, use of appropriate lysis buffer to separate blood cell types and minimize the acquisition events during flow cytometry, adoption of proper fluorophore combination and antibody titration for multiple antigenic detection and introduction of counting beads for precise quantification of functional CPC activity in migration assay. CONCLUSION AND SIGNIFICANCE: With systematic specification of factors influencing the enumeration of CPC by flow cytometry, the abundance and migration capacity of CPCs can be correctly assessed. Adoption of validated SOP is essential for refined comparison of patients with different comorbidities in the analysis of risk stratification.


Subject(s)
Cell Separation/methods , Myocardial Infarction/blood , Myocardial Infarction/pathology , Stem Cells/pathology , AC133 Antigen , Adult , Aged , Antigens, CD/analysis , Antigens, CD34/analysis , Cell Movement , Endolyn/analysis , Flow Cytometry , Glycoproteins/analysis , Hemolysis , Humans , Middle Aged , Peptides/analysis , Prospective Studies , Proto-Oncogene Proteins c-kit/analysis , Reproducibility of Results , Stem Cells/metabolism
6.
Cancer Prev Res (Phila) ; 2(7): 625-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19584076

ABSTRACT

Naturally occurring anthocyanins possess colorectal cancer chemopreventive properties in rodent models. We investigated whether mirtocyan, an anthocyanin-rich standardized bilberry extract, causes pharmacodynamic changes consistent with chemopreventive efficacy and generates measurable levels of anthocyanins in blood, urine, and target tissue. Twenty-five colorectal cancer patients scheduled to undergo resection of primary tumor or liver metastases received mirtocyan 1.4, 2.8, or 5.6 grams (containing 0.5-2.0 grams anthocyanins) daily for 7 days before surgery. Bilberry anthocyanins were analyzed by high performance liquid chromatography (HPLC) with visible or mass spectrometric detection. Proliferation was determined by immunohistochemistry of Ki-67 in colorectal tumor. Concentrations of insulin-like growth factor (IGF)-I were measured in plasma. Mirtocyan anthocyanins and methyl and glucuronide metabolites were identified in plasma, colorectal tissue, and urine, but not in liver. Anthocyanin concentrations in plasma and urine were roughly dose-dependent, reaching approximately 179 ng/gram in tumor tissue at the highest dose. In tumor tissue from all patients on mirtocyan, proliferation was decreased by 7% compared with preintervention values. The low dose caused a small but nonsignificant reduction in circulating IGF-I concentrations. In conclusion, repeated administration of bilberry anthocyanins exerts pharmacodynamic effects and generates concentrations of anthocyanins in humans resembling those seen in Apc(Min) mice, a model of FAP adenomas sensitive to the chemopreventive properties of anthocyanins. Studies of doses containing <0.5 gram bilberry anthocyanins are necessary to adjudge whether they may be appropriate for development as colorectal cancer chemopreventive agents.


Subject(s)
Anthocyanins/administration & dosage , Anticarcinogenic Agents/pharmacology , Colorectal Neoplasms/prevention & control , Vaccinium myrtillus/metabolism , Adenomatous Polyposis Coli/genetics , Administration, Oral , Aged , Aged, 80 and over , Animals , Anthocyanins/pharmacology , Cell Proliferation , Colorectal Neoplasms/secondary , Female , Humans , Ki-67 Antigen/biosynthesis , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Neoplasm Metastasis , Pilot Projects , Plant Extracts/pharmacology
7.
Cancer Res ; 69(7): 3052-9, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276345

ABSTRACT

Ethylene oxide (EO) is widely used in the chemical industry and is also formed in humans through the metabolic oxidation of ethylene, generated during physiologic processes. EO is classified as a human carcinogen and is a direct acting alkylating agent, primarily forming N7-(2-hydroxyethyl)guanine (N7-HEG). To conduct accurate human risk assessments, it is vital to ascertain the relative contribution of endogenously versus exogenously derived DNA damage and identify the sources of background lesions. We have therefore defined in vivo dose-response relationships over a concentration range relevant to human EO exposures using a dual-isotope approach. By combining liquid chromatography-tandem mass spectrometry and high-performance liquid chromatography-accelerator mass spectrometry analysis, both the endogenous and exogenous N7-HEG adducts were quantified in tissues of [(14)C]EO-treated rats. Levels of [(14)C]N7-HEG induced in spleen, liver, and stomach DNA increased in a linear manner from 0.002 to 4 adducts/10(8) nucleotides. More importantly, the extent of damage arising through this route was insignificant compared with the background abundance of N7-HEG naturally present. However, at the two highest doses, [(14)C]EO exposure caused a significant increase in endogenous N7-HEG formation in liver and spleen, suggesting that EO can induce physiologic pathways responsible for ethylene generation in vivo and thereby indirectly promote N7-HEG production. We present evidence for a novel mechanism of adduct formation to explain this phenomenon, involving oxidative stress and 1-aminocyclopropane-1-carboxylic acid as a potential biosynthetic precursor to ethylene in mammalian cells. Based on the proposed pathway, N7-HEG may have potential as a biomarker of cellular oxidative stress.


Subject(s)
DNA Adducts/biosynthesis , Ethylene Oxide/administration & dosage , Guanine/analogs & derivatives , Amino Acids, Cyclic/analysis , Amino Acids, Cyclic/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Carbon Radioisotopes , Chromatography, High Pressure Liquid , DNA Adducts/analysis , Dose-Response Relationship, Drug , Ethylene Oxide/pharmacokinetics , Guanine/analysis , Guanine/biosynthesis , Male , Oxidative Stress , Rats , Rats, Inbred F344 , Tandem Mass Spectrometry , Tissue Distribution
8.
Eur J Cancer ; 44(6): 898-906, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18343654

ABSTRACT

Silibinin, a flavonolignan from milk thistle seeds, possesses cancer chemopreventive properties in rodent models of carcinogenesis. We tested the hypotheses that silibinin or silipide, silibinin formulated with phospholipids, delays tumour development in TRAMP or Apc(Min) mice, genetic models of prostate or intestinal malignancies, respectively. Mice received silibinin or silipide with their diet (0.2% silibinin equivalents) from weaning. Intervention with silipide reduced the size of well differentiated TRAMP adenocarcinomas by 31%. Silipide and silibinin decreased the incidence of poorly differentiated carcinomas by 61% compared to mice on control diet. Silipide decreased plasma levels of insulin-like growth factor (IGF)-1 by 36%. Levels of circulating IGF binding protein (IGFBP)-3 in mice on silipide or silibinin were 3.9- or 5.9-fold, respectively, elevated over those in control TRAMP mice. In Apc(Min) mice silibinin, but not silipide, had only a marginal adenoma number-reducing effect. The results cautiously support the advancement of silipide to the stage of clinical investigation in prostate cancer.


Subject(s)
Adenoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Intestinal Neoplasms/prevention & control , Phosphatidylcholines/therapeutic use , Prostatic Neoplasms/prevention & control , Silymarin/therapeutic use , Adenoma/blood , Animals , Anticarcinogenic Agents/blood , Body Weight/drug effects , Chromatography, High Pressure Liquid , Drug Evaluation , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Neoplasms/blood , Male , Mice , Mice, Inbred C57BL , Phosphatidylcholines/blood , Prostatic Neoplasms/blood , Silybin , Silymarin/blood
9.
Rapid Commun Mass Spectrom ; 22(1): 19-28, 2008.
Article in English | MEDLINE | ID: mdl-18041793

ABSTRACT

A method has been developed for the simultaneous detection and quantitation of five different 2-hydroxyethyl-DNA (HE-DNA) adducts that could be formed as a result of exposure to ethylene oxide (EO). In addition to the major N7-HE-guanine (N7-HEG) adducts this assay can also measure the less prevalent but potentially more biologically significant N1-HE-2'-deoxyadenosine (N1-HEdA), O(6)-HE-2'-deoxyguanosine (O(6)-HEdG), N(6)-HE-2'-deoxyadenosine (N(6)-HEdA) and N3-HE-2'-deoxyuridine adducts (N3-HEdU). The method involves the isolation of HE adducts from the unmodified nucleosides by either neutral thermal hydrolysis or enzymatic digestion, followed by high-performance liquid chromatographic (HPLC) purification, before detection and quantification by liquid chromatography tandem mass spectrometry (LC/MS/MS) using selective reaction monitoring (SRM). The limits of detection were in the range 0.5-25 fmol for each individual adduct, making this one of the most sensitive assays available for the detection of N7-HEG. To illustrate the possible applications of the assay, it has been employed in the measurement of endogenous/background and EO-induced HE adducts in a variety of DNA samples.


Subject(s)
DNA Adducts/analysis , Ethylene Oxide/chemistry , Animals , Calibration , Chromatography, High Pressure Liquid , DNA Adducts/chemical synthesis , Liver/chemistry , Rats , Rats, Inbred F344 , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet , Tandem Mass Spectrometry
10.
Chem Res Toxicol ; 20(2): 290-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17263564

ABSTRACT

Ethylene oxide (EO) is one of the most widely used intermediates in the chemical industry. It is also formed endogenously as a result of cytochrome P450-mediated metabolism of ethylene, which is ubiquitous in the environment. Additionally, ethylene is generated in vivo during normal physiological processes such as methionine oxidation and lipid peroxidation; therefore, humans are continually exposed to EO. EO is classed by the IARC as carcinogenic to humans and reacts with DNA, primarily forming N7-(2-hydroxyethyl)guanine adducts (N7-HEG), which can be used as biomarkers of exposure and potential cancer risk. To assess the risks to humans associated with occupational exposure to low EO concentrations, it is necessary to establish the relative contribution of DNA damage arising from endogenous and exogenously derived EO. Using a newly developed highly sensitive LC-MS/MS assay with selected reaction monitoring that offers a limit of detection of 0.1 fmol of N7-HEG on column, we have established background levels of N7-HEG (1.1-3.5 adducts/10(8) nucleotides) in tissues of rats. Following intraperitoneal administration of a single dose or three daily doses of EO (0.01-1.0 mg/kg), N7-HEG adducts generally increased with dose, except at the lowest concentration where total N7-HEG levels were no different to that detected in control animals, indicating that any increase was negligible as compared to the endogenous damage already present. In the 3 day study, the kinetics of adduct removal were also investigated and in comparing N7-HEG formation in the two studies, DNA damage did not appear to accumulate with repeated administration.


Subject(s)
DNA Adducts/analysis , Ethylene Oxide/administration & dosage , Guanine/analogs & derivatives , Animals , Chromatography, Liquid/methods , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Ethylene Oxide/pharmacokinetics , Guanine/analysis , Injections, Intraperitoneal , Male , Molecular Structure , Rats , Rats, Inbred F344 , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Time Factors , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...