Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Endocr Relat Cancer ; 26(3): 355-366, 2019 03.
Article in English | MEDLINE | ID: mdl-30645190

ABSTRACT

Pharmacological inhibition of the sonic hedgehog (SHH) pathway can be beneficial against certain cancers but detrimental in others. Adamantinomatous craniopharyngioma (ACP) is a relevant pituitary tumour, affecting children and adults, that is associated with high morbidity and increased mortality in long-term follow-up. We have previously demonstrated overactivation of the SHH pathway in both human and mouse ACP. Here, we show that this activation is ligand dependent and induced by the expression of SHH protein in a small proportion of tumour cells. We investigate the functional relevance of SHH signalling in ACP through MRI-guided preclinical studies using an ACP mouse model. Treatment with vismodegib, a clinically approved SHH pathway inhibitor, results in a significant reduction in median survival due to premature development of highly proliferative and vascularised undifferentiated tumours. Reinforcing the mouse data, SHH pathway inhibition in human ACP leads to a significant increase in tumour cell proliferation both ex vivo, in explant cultures, and in vivo, in a patient-derived xenograft model. Together, our results demonstrate a protumourigenic effect of vismodegib-mediated SHH pathway inhibition in ACP.


Subject(s)
Craniopharyngioma/physiopathology , Hedgehog Proteins/antagonists & inhibitors , Adolescent , Animals , Cell Proliferation , Child , Child, Preschool , Disease Models, Animal , Humans , Male , Mice , Pituitary Neoplasms , Signal Transduction
2.
Br J Cancer ; 111(2): 375-85, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24892448

ABSTRACT

BACKGROUND: Dichloroacetate (DCA) has been found to have antitumour properties. METHODS: We investigated the cellular and metabolic responses to DCA treatment and recovery in human colorectal (HT29, HCT116 WT and HCT116 Bax-ko), prostate carcinoma cells (PC3) and HT29 xenografts by flow cytometry, western blotting, electron microscopy, (1)H and hyperpolarised (13)C-magnetic resonance spectroscopy. RESULTS: Increased expression of the autophagy markers LC3B II was observed following DCA treatment both in vitro and in vivo. We observed increased production of reactive oxygen species (ROS) and mTOR inhibition (decreased pS6 ribosomal protein and p4E-BP1 expression) as well as increased expression of MCT1 following DCA treatment. Steady-state lactate excretion and the apparent hyperpolarised [1-(13)C] pyruvate-to-lactate exchange rate (k(PL)) were decreased in DCA-treated cells, along with increased NAD(+)/NADH ratios and NAD(+). Steady-state lactate excretion and k(PL) returned to, or exceeded, control levels in cells recovered from DCA treatment, accompanied by increased NAD(+) and NADH. Reduced k(PL) with DCA treatment was found in HT29 tumour xenografts in vivo. CONCLUSIONS: DCA induces autophagy in cancer cells accompanied by ROS production and mTOR inhibition, reduced lactate excretion, reduced k(PL) and increased NAD(+)/NADH ratio. The observed cellular and metabolic changes recover on cessation of treatment.


Subject(s)
Autophagy/drug effects , Colorectal Neoplasms/drug therapy , Dichloroacetic Acid/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , HCT116 Cells , HT29 Cells , Humans , Lactic Acid/metabolism , Mice , Mice, Nude , Microscopy, Electron , NAD/metabolism , Random Allocation , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
3.
Br J Cancer ; 110(7): 1727-32, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24569471

ABSTRACT

BACKGROUND: Magnetic resonance elastography (MRE) is an emerging imaging technique that affords non-invasive quantitative assessment and visualization of tissue mechanical properties in vivo. METHODS: In this study, MRE was used to quantify (kPa) the absolute value of the complex shear modulus |G*|, elasticity Gd and viscosity Gl of SW620 human colorectal cancer xenografts before and 24 h after treatment with either 200 mg kg(-1) of the vascular disrupting agent ZD6126 (N-acetylcolchinol-O-phosphate) or vehicle control, and the data were compared with changes in water diffusivity measured by diffusion-weighted magnetic resonance imaging. RESULTS: A heterogeneous distribution of |G*|, Gd and Gl was observed pre-treatment with an intertumoral coefficient of variation of 13% for |G*|. There were no significant changes in the vehicle-treated cohort. In contrast, ZD6126 induced a significant decrease in the tumour-averaged |G*| (P<0.01), Gd (P<0.01) and Gl (P<0.05), and this was associated with histologically confirmed central necrosis. This reduction in tumour viscoelasticity occurred at a time when no significant change in tumour apparent diffusion coefficient (ADC) was observed. CONCLUSIONS: These data demonstrate that MRE can provide early imaging biomarkers for treatment-induced tumour necrosis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Elasticity Imaging Techniques/methods , Organophosphorus Compounds/therapeutic use , Animals , Biomechanical Phenomena , Colonic Neoplasms/diagnostic imaging , Elasticity , Female , Humans , Mice , Mice, Nude , Necrosis/chemically induced , Shear Strength , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 106(12): 1960-6, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22596237

ABSTRACT

BACKGROUND: Non-invasive quantitative imaging biomarkers are essential for the evaluation of novel targeted therapeutics. Before deployment in clinical trials, such imaging biomarkers require qualification, typically through pre-clinical identification of imaging-pathology correlates. METHODS: First, in investigating imaging biomarkers of invasion, the response of orthotopic murine PC3 prostate xenografts to the Src inhibitor saracatinib was assessed using susceptibility contrast MRI. Second, the longitudinal response of chemically induced rat mammary adenocarcinomas to the VEGFR2 inhibitor vandetanib was monitored by intrinsic susceptibility MRI, to identify the time window of transient vascular normalisation. RESULTS: No significant differences in fractional blood volume (%), vessel calibre (µm), native T(1) (ms) or apparent water diffusion coefficient were determined, despite reduced expression of activated Fak and paxillin in the saracatinib cohort. Treatment with vandetanib elicited a 60% antitumour response (P<0.01), 80% inhibition in vessel density (P<0.05) and reduction in hypoxia (P<0.05). There was, however, no significant change in tumour baseline R(2)* (s(-1)) or carbogen-induced ΔR(2)* with treatment. CONCLUSION: Reporting negative imaging biomarker responses is important, to avoid the risk of clinical trials using the same biomarkers being undertaken with a false expectation of success, and the abandonment of promising new therapeutics based on a false-negative imaging biomarker response being mistaken for a true-negative.


Subject(s)
Benzodioxoles/therapeutic use , Blood Vessels/pathology , Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/drug therapy , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Biomarkers, Tumor , Blood Vessels/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , False Negative Reactions , Female , Humans , Male , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Targeted Therapy , Neoplasm Transplantation , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Rats
5.
Br J Cancer ; 106(10): 1638-47, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22498643

ABSTRACT

BACKGROUND: Hypoxia-inducible factor-1 (HIF-1) mediates the transcriptional response to hypoxic stress, promoting tumour progression and survival. This study investigated the acute effects of the small-molecule HIF-pathway inhibitor NSC-134754. METHODS: Human PC-3LN5 prostate cancer cells were treated with NSC-134754 for 24 h in hypoxia. Orthotopic prostate tumour-bearing mice were treated with a single dose of NSC-134754 for 6, 24 or 48 h. Treatment response was measured using magnetic resonance spectroscopy and imaging. Ex-vivo histological validation of imaging findings was also sought. RESULTS: In vitro, NSC-134754 significantly reduced lactate production and glucose uptake (P<0.05), while significantly increasing intracellular glucose (P<0.01) and glutamine uptake/metabolism (P<0.05). Increased glutamine metabolism was independent of c-Myc, a factor also downregulated by NSC-134754. In vivo, a significantly higher tumour apparent diffusion coefficient was determined 24 h post-treatment (P<0.05), with significantly higher tumour necrosis after 48 h (P<0.05). NSC-134754-treated tumours revealed lower expression of HIF-1α and glucose transporter-1, at 6 and 24 h respectively, while a transient increase in tumour hypoxia was observed after 24 h. Vessel perfusion/flow and vascular endothelial growth factor levels were unchanged with treatment. CONCLUSION: NSC-134754 induces metabolic alterations in vitro and early anti-tumour activity in vivo, independent of changes in vascular function. Our data support the further evaluation of NSC-134754 as an anti-cancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Isoquinolines/pharmacology , Animals , Blood Vessels/drug effects , Blood Vessels/physiology , Cell Hypoxia , Cell Line, Tumor , Diffusion Magnetic Resonance Imaging , Glucose/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Humans , Isoenzymes/antagonists & inhibitors , L-Lactate Dehydrogenase/antagonists & inhibitors , Lactate Dehydrogenase 5 , Male , Mice , Necrosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/analysis
6.
Neuroscience ; 179: 56-61, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21277354

ABSTRACT

Prion protein (PrP) is abundant in the nervous system, but its role remains uncertain. Prion diseases depend on an aggregation of the protein that is likely to interfere with its normal function. Loss of function does not in itself cause neurodegeneration, but whether it contributes to the clinical features of the disease remains an open question. Patients with classical Creutzfeldt-Jakob disease (CJD) have a higher than expected incidence of epilepsy. To study the mechanisms by which loss of PrP function may underlie changes in vulnerability to epilepsy in disease, we used several acute epilepsy models: we applied a variety of convulsant treatments (zero-magnesium, bicuculline, and pentylenetetrazol) to slices in vitro from PrP knockout (Prnp0/0) and control mice. In all three epilepsy models, we found that longer delays and/or higher concentrations of convulsants were necessary to generate spontaneous epileptiform activity in Prnp0/0 mice. These results together indicate an increased seizure threshold in Prnp0/0 mice, suggesting that loss of PrP function cannot explain a predisposition to seizures initiation in CJD.


Subject(s)
Brain/metabolism , Epilepsy/metabolism , Prions/metabolism , Animals , Convulsants/pharmacology , Creutzfeldt-Jakob Syndrome/complications , Creutzfeldt-Jakob Syndrome/metabolism , Disease Models, Animal , Epilepsy/etiology , Mice , Mice, Knockout , Organ Culture Techniques , Patch-Clamp Techniques
7.
Br J Cancer ; 104(1): 83-90, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21139581

ABSTRACT

BACKGROUND: Progressive tumour growth is dependent on the development of a functional tumour vasculature and highly regulated by growth factors and cytokines. Nitric oxide (NO) is a free radical, produced both by tumour and host cells, and functions as a signalling molecule downstream of several angiogenic factors. Both pro- and antitumourigenic properties have been attributed to NO. METHODS: The expression of the inducible isoform of NO synthase (iNOS) was knocked down in the C6 glioma cell line using constitutive expression of antisense RNA, and the effect of tumour-derived NO on tumour progression and angiogenesis was investigated. RESULTS: Tumours in which iNOS expression was decreased displayed significantly reduced growth rates compared with tumours derived from parental C6 cells. Quantitative non-invasive magnetic resonance imaging and fluorescence microscopy of tumour uptake of Hoechst 33342, and haematoxylin and eosin staining, revealed significantly impaired vascular development and function in antisense iNOS tumours compared with control in vivo, primarily associated with the more necrotic tumour core. Decreased iNOS expression had no effect on tumour VEGF expression. CONCLUSION: Nitric oxide derived from tumour iNOS is an important modulator of tumour progression and angiogenesis in C6 gliomas and further supports the therapeutic strategy of inhibiting iNOS for the treatment of cancer.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Glioma/blood supply , Glioma/enzymology , Neovascularization, Pathologic/enzymology , Nitric Oxide Synthase Type II/physiology , Animals , Blotting, Western , Brain Neoplasms/pathology , DNA, Antisense/pharmacology , Disease Progression , Female , Gene Silencing/physiology , Glioma/pathology , Immunoenzyme Techniques , Magnetic Resonance Imaging , Mice , Mice, Nude , Microscopy, Fluorescence , Nitric Oxide/metabolism , Rats , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
8.
Br J Cancer ; 98(12): 1985-92, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18493233

ABSTRACT

Oesophageal adenocarcinoma, which arises from an acquired columnar lesion, Barrett's metaplasia, is rising in incidence more rapidly than any other cancer in the Western world. Elevated expression of c-MYC has been demonstrated in oesophageal adenocarcinoma; however, the expression of other members of the MYC/MAX/MAD network has not been addressed. The aims of this work were to characterise the expression of c-MYC, MAX and the MAD family in adenocarcinoma development and assess the effects of overexpression on cellular behaviour. mRNA expression in samples of Barrett's metaplasia and oesophageal adenocarcinoma were examined by qRT-PCR. Semi-quantitative immunohistochemistry and western blotting were used to examine cellular localisation and protein levels. Cellular proliferation and mRNA expression were determined in SEG1 cells overexpressing c-MYCER or MAD1 using a bromodeoxyuridine assay and qRT-PCR, respectively. Consistent with previous work expression of c-MYC was deregulated in oesophageal adenocarcinoma. Paradoxically, increased expression of putative c-MYC antagonists MAD1 and MXI1 was observed in tumour specimens. Overexpression of c-MYC and MAD proteins in SEG1 cells resulted in differential expression of MYC/MAX/MAD network members and reciprocal changes in proliferation. In conclusion, the expression patterns of c-MYC, MAX and the MAD family were shown to be deregulated in the oesophageal cancer model.


Subject(s)
Adenocarcinoma/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, myc , Repressor Proteins/genetics , Blotting, Western , Cell Line , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...