Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 113
1.
J Pharmacol Exp Ther ; 370(2): 242-251, 2019 08.
Article En | MEDLINE | ID: mdl-31189729

Adequate drug distribution through tumors is essential for treatment to be effective. Palbociclib is a cyclin-dependent kinase 4/6 inhibitor approved for use in patients with hormone receptor positive, human epidermal growth factor receptor 2 negative metastatic breast cancer. It has unusual physicochemical properties, which may significantly influence its distribution in tumor tissue. We studied the penetration and distribution of palbociclib in vitro, including the use of multicellular three-dimensional models and mathematical modeling. MCF-7 and DLD-1 cell lines were grown as single cell suspensions (SCS) and spheroids; palbociclib uptake and efflux were studied using liquid chromatography-tandem mass spectrometry. Intracellular concentrations of palbociclib for MCF-7 SCS (C max 3.22 µM) and spheroids (C max 2.91 µM) were 32- and 29-fold higher and in DLD-1, 13- and 7-fold higher, respectively, than the media concentration (0.1 µM). Total palbociclib uptake was lower in DLD-1 cells than MCF-7 cells in both SCS and spheroids. Both uptake and efflux of palbociclib were slower in spheroids than SCS. These data were used to develop a mathematical model of palbociclib transport that quantifies key parameters determining drug penetration and distribution. The model reproduced qualitatively most features of the experimental data and distinguished between SCS and spheroids, providing additional support for hypotheses derived from the experimental data. Mathematical modeling has the potential for translating in vitro data into clinically relevant estimates of tumor drug concentrations. SIGNIFICANCE STATEMENT: This study explores palbociclib uptake and efflux in single cell suspension and spheroid models of cancer. Large intracellular concentrations of palbociclib are found after drug exposure. The data from this study may aid understanding of the intratumoural pharmacokinetics of palbociclib, which is useful in understanding how drug distributes within tumor tissue and optimizing drug efficacy. Biomathematical modelling has the potential to derive intratumoural drug concentrations from plasma pharmacokinetics in patients.


Piperazines/metabolism , Pyridines/metabolism , Spheroids, Cellular/metabolism , Biological Transport , Cell Survival/drug effects , Humans , MCF-7 Cells , Models, Biological , Piperazines/pharmacology , Pyridines/pharmacology , Single-Cell Analysis , Spheroids, Cellular/drug effects
2.
RSC Adv ; 8(19): 10474-10477, 2018 Mar 13.
Article En | MEDLINE | ID: mdl-35540495

Amphiphilic block copolymers have been developed for the encapsulation of organometallic drugs. silver-N-heterocyclic carbene complexes have shown significant promise as anticancer and antibacterial compounds, and have been studied as the payload in these carriers. Simple modification of the N-heterocyclic carbene ligand structure enables solubility properties and interaction with the polymer to be tuned.

3.
R Soc Open Sci ; 4(5): 170014, 2017 May.
Article En | MEDLINE | ID: mdl-28573005

The tumour vasculature and microenvironment is complex and heterogeneous, contributing to reduced delivery of cancer drugs to the tumour. We have developed an in silico model of drug transport in a tumour cord to explore the effect of different drug regimes over a 72 h period and how changes in pharmacokinetic parameters affect tumour exposure to the cytotoxic drug doxorubicin. We used the model to describe the radial and axial distribution of drug in the tumour cord as a function of changes in the transport rate across the cell membrane, blood vessel and intercellular permeability, flow rate, and the binding and unbinding ratio of drug within the cancer cells. We explored how changes in these parameters may affect cellular exposure to drug. The model demonstrates the extent to which distance from the supplying vessel influences drug levels and the effect of dosing schedule in relation to saturation of drug-binding sites. It also shows the likely impact on drug distribution of the aberrant vasculature seen within tumours. The model can be adapted for other drugs and extended to include other parameters. The analysis confirms that computational models can play a role in understanding novel cancer therapies to optimize drug administration and delivery.

4.
Expert Opin Drug Metab Toxicol ; 13(7): 783-791, 2017 Jul.
Article En | MEDLINE | ID: mdl-28637373

INTRODUCTION: Apaziquone (also known as EO9 and QapzolaTM) is a prodrug that is activated to DNA damaging species by oxidoreductases (particularly NQO1) and has the ability to kill aerobic and/or hypoxic cancer cells. Areas covered: Whilst its poor pharmacokinetic properties contributed to its failure in phase II clinical trials when administered intravenously, these properties were ideal for loco-regional therapies. Apaziquone demonstrated good anti-cancer activity against non-muscle invasive bladder cancer (NMIBC) when administered intravesically to marker lesions and was well tolerated with no systemic side effects. However, phase III clinical trials did not reach statistical significance for the primary endpoint of 2-year recurrence in apaziquone over placebo although improvements were observed. Post-hoc analysis of the combined study data did indicate a significant benefit for patients treated with apaziquone, especially when the instillation of apaziquone was given 30 min or more after surgery. A further phase III study is ongoing to test the hypotheses generated in the unsuccessful phase III studies conducted to date. Expert opinion: Because of its specific pharmacological properties, Apaziquone is excellently suited for local therapy such as NMIBC. Future studies should include proper biomarkers.


Antineoplastic Agents/administration & dosage , Aziridines/administration & dosage , Indolequinones/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aziridines/pharmacokinetics , Aziridines/pharmacology , Humans , Indolequinones/pharmacokinetics , Indolequinones/pharmacology , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms/pathology
5.
Dalton Trans ; 44(7): 3265-70, 2015 Feb 21.
Article En | MEDLINE | ID: mdl-25597645

The complexes [RuCp*(PP)Cl] (Cp* = C5Me5; [], PP = dppm; [], PP = Xantphos), [RuCp(#)(PP)Cl] (Cp(#) = C5Me4(CH2)5OH; [], PP = dppm; [], PP = Xantphos) and [RuCp*(dppm)(CH3CN)][SbF6] [] were synthesized and evaluated in vitro as anticancer agents. Compounds gave nanomolar IC50 values against normoxic A2780 and HT-29 cell lines, and were also tested against hypoxic HT-29 cells, maintaining their high activity. Complex yielded an IC50 value of 0.55 ± 0.03 µM under a 0.1% O2 concentration.


Antineoplastic Agents/chemical synthesis , Chelating Agents/chemical synthesis , Ruthenium/chemistry , Crystallography, X-Ray , Diphosphates/chemical synthesis , Drug Evaluation, Preclinical/methods , HT29 Cells , Humans , Ligands
6.
Oncogenesis ; 3: e102, 2014 May 12.
Article En | MEDLINE | ID: mdl-24819061

Most cancer cells use aerobic glycolysis to fuel their growth. The enzyme lactate dehydrogenase-A (LDH-A) is key to cancer's glycolytic phenotype, catalysing the regeneration of nicotinamide adenine dinucleotide (NAD(+)) from reduced nicotinamide adenine dinucleotide (NADH) necessary to sustain glycolysis. As such, LDH-A is a promising target for anticancer therapy. Here we ask if the tumour suppressor p53, a major regulator of cellular metabolism, influences the response of cancer cells to LDH-A suppression. LDH-A knockdown by RNA interference (RNAi) induced cancer cell death in p53 wild-type, mutant and p53-null human cancer cell lines, indicating that endogenous LDH-A promotes cancer cell survival irrespective of cancer cell p53 status. Unexpectedly, however, we uncovered a novel role for p53 in the regulation of cancer cell NAD(+) and its reduced form NADH. Thus, LDH-A silencing by RNAi, or its inhibition using a small-molecule inhibitor, resulted in a p53-dependent increase in the cancer cell ratio of NADH:NAD(+). This effect was specific for p53(+/+) cancer cells and correlated with (i) reduced activity of NAD(+)-dependent deacetylase sirtuin 1 (SIRT1) and (ii) an increase in acetylated p53, a known target of SIRT1 deacetylation activity. In addition, activation of the redox-sensitive anticancer drug EO9 was enhanced selectively in p53(+/+) cancer cells, attributable to increased activity of NAD(P)H-dependent oxidoreductase NQO1 (NAD(P)H quinone oxidoreductase 1). Suppressing LDH-A increased EO9-induced DNA damage in p53(+/+) cancer cells, but importantly had no additive effect in non-cancer cells. Our results identify a unique strategy by which the NADH/NAD(+) cellular redox status can be modulated in a cancer-specific, p53-dependent manner and we show that this can impact upon the activity of important NAD(H)-dependent enzymes. To summarise, this work indicates two distinct mechanisms by which suppressing LDH-A could potentially be used to kill cancer cells selectively, (i) through induction of apoptosis, irrespective of cancer cell p53 status and (ii) as a part of a combinatorial approach with redox-sensitive anticancer drugs via a novel p53/NAD(H)-dependent mechanism.

7.
J R Soc Interface ; 11(94): 20131173, 2014 May 06.
Article En | MEDLINE | ID: mdl-24621814

The ability to predict how far a drug will penetrate into the tumour microenvironment within its pharmacokinetic (PK) lifespan would provide valuable information about therapeutic response. As the PK profile is directly related to the route and schedule of drug administration, an in silico tool that can predict the drug administration schedule that results in optimal drug delivery to tumours would streamline clinical trial design. This paper investigates the application of mathematical and computational modelling techniques to help improve our understanding of the fundamental mechanisms underlying drug delivery, and compares the performance of a simple model with more complex approaches. Three models of drug transport are developed, all based on the same drug binding model and parametrized by bespoke in vitro experiments. Their predictions, compared for a 'tumour cord' geometry, are qualitatively and quantitatively similar. We assess the effect of varying the PK profile of the supplied drug, and the binding affinity of the drug to tumour cells, on the concentration of drug reaching cells and the accumulated exposure of cells to drug at arbitrary distances from a supplying blood vessel. This is a contribution towards developing a useful drug transport modelling tool for informing strategies for the treatment of tumour cells which are 'pharmacokinetically resistant' to chemotherapeutic strategies.


Antineoplastic Agents/pharmacokinetics , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Biological Transport , Computer Simulation , Humans
8.
Br J Pharmacol ; 171(1): 224-36, 2014 Jan.
Article En | MEDLINE | ID: mdl-24117380

BACKGROUND AND PURPOSE: Hypoxia in tumours is known to cause resistance to conventional chemotherapeutic drugs. In contrast, little is known about the effects of hypoxia on targeted anti-cancer drugs. This study evaluated the effect of hypoxia on a series of clinically approved tyrosine kinase inhibitors (TKIs). EXPERIMENTAL APPROACH: The effect of hypoxia (0.1% oxygen) on the activity of conventional cytotoxic drugs (5-fluorouracil, doxorubicin and vinblastine), the hypoxia-activated prodrug tirapazamine and 9 TKIs was determined in a panel of cell lines. Where hypoxia had a marked effect on chemosensitivity, Western blot analysis was conducted to determine the effect of hypoxia on target expression and the effect of TKIs on cell signalling response under aerobic and hypoxic conditions. KEY RESULTS: Three patterns of chemosensitivity were observed: resistance under hypoxia, equitoxic activity against hypoxic and aerobic cells, and preferential cytotoxicity to hypoxic cells. Significant hypoxia selectivity (independent of HIF1) was observed in the case of dasatinib and this correlated with the ability of dasatinib to inhibit phosphorylation of Src at tyrosine 530. Sorafenib was significantly less effective under hypoxic conditions but resistance did not correlate with hypoxia-induced changes in Raf/MEK/ERK signalling. CONCLUSIONS AND IMPLICATIONS: Hypoxia influences the activity of TKIs but in contrast to conventional cytotoxic drugs, preferential activity against hypoxic cells can occur. The search for hypoxia-targeted therapies has been long and fruitless and this study suggests that some clinically approved TKIs could preferentially target the hypoxic fraction of some tumour types.


Antineoplastic Agents/pharmacology , Neoplasms/enzymology , Oxygen/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Hypoxia , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inhibitory Concentration 50 , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , MCF-7 Cells , Molecular Targeted Therapy , Neoplasms/pathology , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
11.
Gene Ther ; 20(1): 7-15, 2013 Jan.
Article En | MEDLINE | ID: mdl-22170342

Oncolytic viruses (OV) are promising treatments for cancer, with several currently undergoing testing in randomised clinical trials. Measles virus (MV) has not yet been tested in models of human melanoma. This study demonstrates the efficacy of MV against human melanoma. It is increasingly recognised that an essential component of therapy with OV is the recruitment of host antitumour immune responses, both innate and adaptive. MV-mediated melanoma cell death is an inflammatory process, causing the release of inflammatory cytokines including type-1 interferons and the potent danger signal HMGB1. Here, using human in vitro models, we demonstrate that MV enhances innate antitumour activity, and that MV-mediated melanoma cell death is capable of stimulating a melanoma-specific adaptive immune response.


Measles virus/immunology , Melanoma/immunology , Oncolytic Viruses/immunology , Cell Death/immunology , Cell Line, Tumor , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Measles virus/pathogenicity , Melanoma/pathology , Melanoma/virology , Oncolytic Viruses/pathogenicity , Up-Regulation
12.
J Theor Biol ; 313: 142-52, 2012 Nov 21.
Article En | MEDLINE | ID: mdl-22974970

The potential for the use of in-silico models of disease in progression monitoring is becoming increasingly recognised, as well as its contribution to the development of complete curative processes. In this paper we report the development of a hybrid cellular automaton model to mimic the growth of avascular tumours, including the infusion of a bioreductive drug to study the effects of protein binding on drug transportation. The growth model is operated within an extracellular tumour microenvironment. An artificial Neural Network based scheme was implemented that modelled the behaviours of each cell (proliferation, quiescence, apoptosis and/or movement) based on the complex heterogeneous microenvironment; consisting of oxygen, glucose, hydrogen ions, inhibitory factors and growth factors. To validate the growth model results, we conducted experiments with multicellular tumour spheroids. These results showed good agreement with the predicted growth dynamics. The outcome of the avascular tumour growth model suggested that tumour microenvironments have a strong impact on cell behaviour. To address the problem of cellular proteins acting as resistive factors preventing efficient drug penetration, a bioreactive drug (tirapazamine) was added to the system. This allowed us to study the drug penetration through multicellular layers of tissue after its binding to cellular proteins. The results of the in vitro model suggested that the proteins reduce the toxicity of the drug, reducing its efficacy for the most severely hypoxic fractions furthest from a functional blood vessel. Finally this research provides a unique comparison of in vitro tumour growth with an intelligent in silico model to measure bioreductive drug availability inside tumour tissue through a set of experiments.


Antineoplastic Agents/metabolism , Models, Biological , Neoplasms/blood supply , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Diffusion/drug effects , HT29 Cells , Humans , Neural Networks, Computer , Protein Binding/drug effects , Reproducibility of Results , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tirapazamine , Triazines/metabolism , Triazines/pharmacology
13.
J Proteome Res ; 11(5): 2863-75, 2012 May 04.
Article En | MEDLINE | ID: mdl-22416669

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.


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
14.
J Perinatol ; 32(5): 374-80, 2012 May.
Article En | MEDLINE | ID: mdl-21836549

OBJECTIVE: Approximately 40% of women who smoke tobacco quit smoking during pregnancy, yet up to 85% relapse after delivery. Those who resume smoking often do so by 2 to 8 weeks postpartum. Smoking mothers are more than twice as likely to quit breastfeeding by 10 weeks postpartum. The hospitalization of a newborn, while stressful, is an opportunity to emphasize the importance of a smoke-free environment for babies. Supporting maternal-infant bonding may reduce maternal stress and motivate mothers to remain smoke free and continue breastfeeding. The objective of this study was to reduce postpartum smoking relapse and prolong breastfeeding duration during the first 8 weeks postpartum in mothers who quit smoking just before or during pregnancy and have newborns admitted to the Neonatal Intensive Care Unit (NICU). STUDY DESIGN: This study was an Institutional Review Board-approved prospective randomized clinical trial. After informed consent, mothers of newborns admitted to the NICU were randomized to a control or intervention group. Both groups received weekly encouragement to remain smoke free and routine breastfeeding support. Mothers in the intervention group were also given enhanced support for maternal-infant bonding including information about newborn behaviors, and were encouraged to frequently hold their babies skin-to-skin. RESULT: More mothers were smoke free (81 vs 46%, P<0.001) and breastfeeding (86 vs 21%, P<0.001) in the intervention than in the control group at 8 weeks postpartum. CONCLUSION: Interventions to support mother-infant bonding during a newborn's hospitalization in the NICU are associated with reduced rates of smoking relapse and prolonged duration of breastfeeding during the first 8 weeks postpartum.


Intensive Care Units, Neonatal , Patient Education as Topic/methods , Postpartum Period , Smoking Prevention , Adult , Attitude to Health , Breast Feeding/statistics & numerical data , Female , Follow-Up Studies , Humans , Incidence , Kaplan-Meier Estimate , Maternal Welfare , Mother-Child Relations , Postnatal Care/methods , Pregnancy , Prospective Studies , Reference Values , Risk Assessment , Secondary Prevention , Smoking/adverse effects , Smoking Cessation/statistics & numerical data , Statistics, Nonparametric , Young Adult
15.
J Theor Biol ; 257(4): 598-608, 2009 Apr 21.
Article En | MEDLINE | ID: mdl-19183560

Inadequate drug delivery to tumours is now recognised as a key factor that limits the efficacy of anticancer drugs. Extravasation and penetration of therapeutic agents through avascular tissue are critically important processes if sufficient drug is to be delivered to be therapeutic. The purpose of this study is to develop an in silico model that will simulate the transport of the clinically used cytotoxic drug doxorubicin across multicell layers (MCLs) in vitro. Three cell lines were employed: DLD1 (human colon carcinoma), MCF7 (human breast carcinoma) and NCI/ADR-Res (doxorubicin resistant and P-glycoprotein [Pgp] overexpressing ovarian cell line). Cells were cultured on transwell culture inserts to various thicknesses and doxorubicin at various concentrations (100 or 50 microM) was added to the top chamber. The concentration of drug appearing in the bottom chamber was determined as a function of time by HPLC-MS/MS. The rate of drug penetration was inversely proportional to the thickness of the MCL. The rate and extent of doxorubicin penetration was no different in the presence of NCI/ADR-Res cells expressing Pgp compared to MCF7 cells. A mathematical model based upon the premise that the transport of doxorubicin across cell membrane bilayers occurs by a passive "flip-flop" mechanism of the drug between two membrane leaflets was constructed. The mathematical model treats the transwell apparatus as a series of compartments and the MCL is treated as a series of cell layers, separated by small intercellular spaces. This model demonstrates good agreement between predicted and actual drug penetration in vitro and may be applied to the prediction of drug transport in vivo, potentially becoming a useful tool in the study of optimal chemotherapy regimes.


Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Models, Biological , Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/administration & dosage , Biological Transport , Cell Membrane/metabolism , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Neoplasm Proteins/metabolism , Tumor Cells, Cultured
16.
Oncol Rep ; 14(4): 909-13, 2005 Oct.
Article En | MEDLINE | ID: mdl-16142350

HIF-1 is a heterodimer consisting of the HIF-1alpha and HIF-1beta subunits, and HIF-1alpha is the unique oxygen regulated subunit that determines HIF-1 activity. HIF-1alpha upgrades many gene products which include the glucose transporter protein 1 (Glut-1). Immunohistochemical studies using a monoclonal antibody specific for HIF-1alpha indicate that the overexpression of HIF-1alpha occurs in the most common forms of human cancer, including bladder cancer. The expression of Glut-1 in human bladder cancer is associated with poor prognosis and a low survival rate. To our knowledge, this is the first study to compare the expression of both HIF-1alpha and Glut-1 with clinicopathological characteristics in superficial and invasive human bladder cancer (all invasive bladder cancer patients received radical radiotherapy). The Kaplan-Meier survival analysis curve shows a significant association of HIF-1alpha expression with recurrence and survival in superficial bladder cancer and shows a significant association of Glut-1 with survival in invasive bladder cancer [chi2 (4)=10.52; Pr >chi2 =0.0012].


Glucose Transporter Type 1/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Urinary Bladder Neoplasms/metabolism , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Dimerization , Humans , Immunohistochemistry , Models, Statistical , Neoplasm Invasiveness , Oxygen/chemistry , Prognosis , Treatment Outcome , Urinary Bladder Neoplasms/pathology
17.
Int J Oncol ; 26(6): 1477-84, 2005 Jun.
Article En | MEDLINE | ID: mdl-15870859

Tumour hypoxia confers poor prognosis in a wide range of solid tumours, due to an increased malignancy, increased likelihood of metastasis and treatment resistance. Poorly oxygenated tumours are resistant to both radiation therapy and chemotherapy. However, although the link between radiation therapy and hypoxia is well established in a range of clinical studies, evidence of its influence on chemotherapy response is lacking. In this study, a panel of human tumour-derived xenografts that have been characterised previously for in vivo response to a large series of anti-cancer agents, and have been found to show chemosensitivities that correlate strongly with the parent tumour, were used to address this issue. Immunohistochemistry was carried out on formalin-fixed, paraffin-embedded sections of xenograft samples to detect expression of the intrinsic hypoxia marker Glut-1 and adducts of the bioreductive hypoxia marker pimonidazole. Glut-1 scores correlated significantly with T/C values for CCNU sensitivity (r = 0.439, P = 0.036, n = 23) and showed a borderline significant correlation with dacarbazine T/C (r = 0.405, P = 0.076, n = 20). However, there was no correlation between both Glut-1 and pimonidazole scores and T/C obtained for the bioreductive drug mitomycin C. The use of human tumour-derived xenografts offers a potentially useful way of using archival material to determine the influence of hypoxia and other tumour-microenvironmental factors on chemosensitivity without the direct use of human subjects.


Antineoplastic Agents, Alkylating/pharmacology , Monosaccharide Transport Proteins/physiology , Neoplasms, Experimental/drug therapy , Animals , Biomarkers , Cell Hypoxia , Endoplasmic Reticulum Chaperone BiP , Glucose Transporter Type 1 , Heat-Shock Proteins/physiology , Humans , Mice , Molecular Chaperones/physiology , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Transplantation, Heterologous
18.
Teratog Carcinog Mutagen ; Suppl 2: 13-29, 2003.
Article En | MEDLINE | ID: mdl-14691977

In order to determine differences in repair after treatment with DNA damaging agents, normal and cancer cells were selected for analysis of single strand breaks and DNA crosslinks using the Comet assay. Normal human lymphocytes, human colorectal adenocarcinoma SW620 cells, lung carcinoma A549, and H460 cell lines were exposed to an ethylating agent (ethylmethane sulfonate [EMS]), and a cross-linking agent (mitomycin C [MMC]). Differences in repair profiles of DNA damage demonstrated using the comet assay were observed in human lymphocytes and tumour cell lines with both mutagens. Results were also indicative that MMC repair is concentration-dependent. It was also apparent that normal cells repair DNA damage more readily than tumour cells. Repair also varied between different cell lines. To investigate the mechanistic differences of these two chemicals, flow cytometry studies were undertaken in tumour cells, namely cell cycle analysis and frequency of micronuclei induction (FMN). A G2M phase block was clearly evident following treatment with EMS at all concentrations tested. With MMC, an initial arrest of cells in G2M was accompanied by a build-up in S-phase over longer exposure periods. Also, at the highest mutagen doses there were different patterns of micronuclei induction. Thus, using the mutagens with different mechanisms of action highlighted the differences in repair patterns between normal and tumour cells.


Comet Assay/methods , DNA Repair , DNA, Neoplasm/drug effects , DNA/drug effects , Flow Cytometry/methods , Mutagens/toxicity , Cell Cycle/drug effects , Cells, Cultured , Cross-Linking Reagents/toxicity , DNA Damage , Ethyl Methanesulfonate/toxicity , Humans , Lymphocytes/drug effects , Micronuclei, Chromosome-Defective/drug effects , Mitomycin/toxicity , Neoplasms/genetics
20.
Br J Pharmacol ; 137(5): 701-9, 2002 Nov.
Article En | MEDLINE | ID: mdl-12381684

The bioreductive drug EO9 (3-hydroxy-5-aziridinyl-1-methyl-2[indole-4,7-dione]-prop-beta-en-alpha-ol) has good pharmacodynamic properties in vitro, modest anti-tumour activity in experimental tumour models, but failed to show activity in clinical trials. Understanding the reasons for its poor efficacy in vivo is important in terms of progressing second generation analogues into the clinic. In two human tumour xenografts, direct intra-tumoural injection resulted in improved anti-tumour activity compared with intravenous administration suggesting that drug delivery to tumours is suboptimal. Compared with Mitomycin C (MMC) and the experimental agent MeDZQ, EO9 was rapidly cleared from the systemic circulation (t1/2=1.8 min) whereas MMC and MeDZQ had significantly increased plasma t1/2 values (14 and 22 min respectively). These three compounds demonstrated similar pharmacodynamic properties in terms of potency towards the NQO1 (NAD(P)H:Quinone oxidoreductase) rich H460 cell line in vitro but differed significantly in their in vivo activity with growth delays of 17.7, 4.5 and 1.0 days for MMC, MeDZQ and EO9 respectively. EO9 was rapidly metabolized by red blood cells in vitro (t1/2=14.5 min) which must contribute to its rapid pharmacokinetic elimination in vivo whereas MMC and MeDZQ were metabolized at comparatively slower rates (t1/2>120 min and 77.0 min respectively). In conclusion, the development of second generation EO9 analogues should address the issue of drug delivery and analysis of drug metabolism by murine whole blood in vitro could be utilized as a preliminary screen to identify lead compounds that are likely to have improved pharmacokinetic profiles in vivo.


Aziridines/administration & dosage , Aziridines/chemistry , Indolequinones , Indoles/administration & dosage , Indoles/chemistry , Quinones/administration & dosage , Technology, Pharmaceutical/methods , Animals , Aziridines/blood , Drug Delivery Systems/methods , Humans , Indoles/blood , Mice , Mice, Nude , Quinones/blood , Quinones/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
...