Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Publication year range
1.
Nature ; 576(7785): 112-120, 2019 12.
Article in English | MEDLINE | ID: mdl-31748746

ABSTRACT

The evolutionary processes that drive universal therapeutic resistance in adult patients with diffuse glioma remain unclear1,2. Here we analysed temporally separated DNA-sequencing data and matched clinical annotation from 222 adult patients with glioma. By analysing mutations and copy numbers across the three major subtypes of diffuse glioma, we found that driver genes detected at the initial stage of disease were retained at recurrence, whereas there was little evidence of recurrence-specific gene alterations. Treatment with alkylating agents resulted in a hypermutator phenotype at different rates across the glioma subtypes, and hypermutation was not associated with differences in overall survival. Acquired aneuploidy was frequently detected in recurrent gliomas and was characterized by IDH mutation but without co-deletion of chromosome arms 1p/19q, and further converged with acquired alterations in the cell cycle and poor outcomes. The clonal architecture of each tumour remained similar over time, but the presence of subclonal selection was associated with decreased survival. Finally, there were no differences in the levels of immunoediting between initial and recurrent gliomas. Collectively, our results suggest that the strongest selective pressures occur during early glioma development and that current therapies shape this evolution in a largely stochastic manner.


Subject(s)
Glioma/genetics , Adult , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Disease Progression , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Polymorphism, Single Nucleotide , Recurrence
2.
Genomics ; 114(2): 110289, 2022 03.
Article in English | MEDLINE | ID: mdl-35124175

ABSTRACT

Notch signalling pathway, particularly its ligand delta-ligand 3 (DLL3), is important in glioma, however, little is known about DLL3 regulation and prognostic effects. Immunohistochemistry on a cohort of 163 gliomas revealed DLL3 upregulation in IDH1 mutant gliomas, where it was associated with a favourable prognosis (HR[95% CI]: 0.28[0.09-0.87]; p = 0.021). We investigated the epigenetic regulation of DLL3, and identified individual CpG sites correlating with DLL3 mRNA expression, which were significant prognostic markers in LGG. In silico analysis revealed that infiltrating immune cells significantly correlated with DLL3 expression, methylation and somatic copy number alterations. The prognostic effects of DLL3 expression was significantly affected by infiltration of immune cells. RNA Sequencing of 83 LGGs and GO Term analysis of differentially expressed genes showed that low DLL3 expression was related to ciliogenesis, which was confirmed by TCGA LGG analysis. Thus, DLL3 may play an important role in the immune microenvironment and prognosis of LGGs.


Subject(s)
Brain Neoplasms , Glioma , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Epigenesis, Genetic , Glioma/genetics , Glioma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Prognosis , Tumor Microenvironment/genetics
3.
Cancer Causes Control ; 33(5): 749-757, 2022 May.
Article in English | MEDLINE | ID: mdl-35184245

ABSTRACT

PURPOSE: High-grade disease accounts for ~ 70% of all glioma, and has a high mortality rate. Few modifiable exposures are known to be related to glioma risk or mortality. METHODS: We examined associations between lifetime physical activity and physical activity at different ages (15-18 years, 19-29 years, 30-39 years, last 10 years) with the risk of glioma diagnosis, using data from a hospital-based family case-control study (495 cases; 371 controls). We followed up cases over a median of 25 months to examine whether physical activity was associated with all-cause mortality. Physical activity and potential confounders were assessed by self-administered questionnaire. We examined associations between physical activity (metabolic equivalent [MET]-h/wk) and glioma risk using unconditional logistic regression and with all-cause mortality in cases using Cox regression. RESULTS: We noted a reduced risk of glioma for the highest (≥ 47 MET-h/wk) versus lowest (< 24 METh/wk) category of physical activity for lifetime activity (OR = 0.58, 95% CI: 0.38-0.89) and at 15-18 years (OR = 0.57, 95% CI: 0.39-0.83). We did not observe any association between physical activity and all-cause mortality (HR for lifetime physical activity = 0.91, 95% CI: 0.64-1.29). CONCLUSION: Our findings are consistent with previous research that suggested physical activity during adolescence might be protective against glioma. Engaging in physical activity during adolescence has many health benefits; this health behavior may also offer protection against glioma.


Subject(s)
Exercise , Glioma , Adolescent , Case-Control Studies , Follow-Up Studies , Glioma/epidemiology , Humans , Risk Factors
4.
J Neurooncol ; 147(1): 37-47, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32002804

ABSTRACT

PURPOSE: The phenotypic and genotypic landscapes in multifocal glioblastoma (MF GBM) cases can vary greatly among lesions. In a MF GBM patient, the rapid development of a secondary lesion was investigated to determine if a unique genetic signature could account for the apparent increased malignancy of this lesion. METHODS: The primary (G52) and secondary (G53) tumours were resected to develop patient derived models followed by functional assays and multiplatform molecular profiling. RESULTS: Molecular profiling revealed G52 was wild-type for TP53 while G53 presented with a TP53 missense mutation. Functional studies demonstrated increased proliferation, migration, invasion and colony formation in G53. CONCLUSION: This data suggests that the TP53 mutation led to gain-of-function phenotypes and resulted in greater overall oncogenic potential of G53.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Tumor Suppressor Protein p53/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gain of Function Mutation , Glioblastoma/pathology , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Mutation , Tumor Cells, Cultured
5.
Int J Cancer ; 145(12): 3425-3435, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31373686

ABSTRACT

DNA methylation has long been recognized as a tumor-promoting factor when aberrantly regulated in the promoter region of genes. However, the effect of intragenic DNA methylation remains poorly understood on the clinical aspects of cancer. Here, we first evaluated the significance of intragenic DNA methylation for survival outcomes of cancer patients in a genome-wide manner. Glioblastoma patients with hypermethylated intragenic regions exhibited better survival than hypomethylated patients. Enrichment analyses of intragenic DNA methylation profiles with epigenetic signatures prioritized the intragenic DNA methylation of ZMIZ1 as a possible glioblastoma prognostic marker that is independent of MGMT methylation in IDH1 wild-type patients. This intragenic region harbored molecular signatures of alternative transcription across many cell types. Furthermore, we found that the intragenic region of ZMIZ1 can serve as a molecular marker in multiple cancers including astrocytomas, bladder cancer and renal cell carcinoma according to DNA methylation status. Finally, in vitro and in vivo experiments uncovered the role of ZMIZ1 as a driver of tumor cell migration. Altogether, our results identify ZMIZ1 as a prognostic marker in cancer and highlight the clinical significance of intragenic methylation in cancer.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Methylation/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genome-Wide Association Study/methods , Mice, Nude , Prognosis , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics
6.
J Neurooncol ; 141(1): 1-12, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30392086

ABSTRACT

BACKGROUND: Immune checkpoint inhibition (ICI) is an emerging immunotherapy for metastatic brain disease (MBD). Current management options include stereotactic radiosurgery (SRS), which has been shown to confer prognostic benefit in combination with ICI. However, the effect, if any, of ICI timing on this benefit is currently unclear. The aim of this study was to evaluate the effect of concurrent ICI with SRS on survival outcomes in MBD compared to non-concurrent ICI administered before or after SRS. METHODS: Searches of 7 electronic databases from inception to April 2018 were conducted following the appropriate guidelines. 1210 articles were identified for screening. Kaplan Meier estimation of 12-month overall survival (OS), local progression free survival (LPFS) and distant progression free survival (DPFS) were pooled as odd ratios (ORs) and analyzed using the random effects model. RESULTS: A total of 8 retrospective observational cohort studies satisfied selection criteria. Compared to non-concurrent ICI, concurrent ICI with SRS conferred a significant 12-month OS benefit (OR = 1.74; p = 0.011), and comparable 12-month LPFS (OR = 2.09; p = 0.154) and DPFS (OR = 0.88; p = 0.839). These significances were reflected in the subgroup of melanoma metastases. CONCLUSION: Based on the trends of our findings, there appears to exist an optimal time window around SRS of which ICI may confer the most survival benefit. However, current literature is limited by a number of clinical parameters requiring further delineation which limits the certainty of these findings. Larger, prospective, and randomized studies will assist in identifying the time period for which ICI can provide the best outcome in MBD managed with SRS.


Subject(s)
Brain Neoplasms/therapy , Cell Cycle Checkpoints/drug effects , Immunotherapy/methods , Radiosurgery/methods , Brain Neoplasms/immunology , Brain Neoplasms/secondary , Combined Modality Therapy/methods , Humans , Kaplan-Meier Estimate , Treatment Outcome
7.
J Neurooncol ; 139(2): 231-238, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29767813

ABSTRACT

INTRODUCTION: There are many potential biomarkers in glioblastoma (GBM), and meta-analyses represent the highest level of evidence when inferring their prognostic significance. It is possible however, that inherent design properties of the studies included in these meta-analyses may affect the pooled hazard ratio (HR) of the meta-analyses. This meta-epidemiological study aims to investigate the potential bias of three study-level properties in meta-analyses of GBM biomarkers currently published in the literature. METHODS: Seven electronic databases from inception to December 2017 were searched for meta-analyses evaluating different GBM biomarkers, which were screened against specific criteria. Study-level data were extracted from each meta-analysis, and analyzed using logistic regression to yield the ratio of HR (RHR) summary statistic. RESULTS: Nine meta-analyses investigating different GBM biomarkers were included. Of all the meta-analyses, the HRs of two studies were significantly underestimated by older studies; they investigated biomarkers IDH1 (RHR = 1.145; p = 0.017) and CD133 (RHR = 0.850; p = 0.013). Study-level size and design showed non-significant trends towards affecting the overall HR in all included meta-analyses. CONCLUSIONS: This meta-epidemiological study demonstrated that study-level year can already significantly affect the pooled HR of GBM biomarkers reported by meta-analyses. It is possible that in the future, more study-level properties will exert significant effect. In terms of future GBM biomarker meta-analyses, special consideration of bias should be given to these study-level properties as potential sources of effect on the prognostic pooled HR.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Brain Neoplasms/epidemiology , Brain Neoplasms/metabolism , Epidemiologic Studies , Glioblastoma/epidemiology , Glioblastoma/metabolism , Humans , Prognosis
8.
J Transl Med ; 15(1): 61, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28314386

ABSTRACT

BACKGROUND: The O 6 -methylguanine methyltransferase (MGMT) gene is frequently unmethylated in patients with glioblastoma (GBM), rendering them non-responsive to the standard treatment regime of surgery followed by concurrent radiotherapy (RT) and temozolomide. Here, we investigate the efficacy of adding a PARP inhibitor, veliparib, to radiotherapy to treat MGMT unmethylated GBM. METHODS: The inhibition of PARP with veliparib (ABT-888), a potent and orally bioavailable inhibitor in combination with RT was tested on a panel of patient derived cell lines (PDCLs) and patient-derived xenografts (PDX) models generated from GBM patients with MGMT unmethylated tumors. RESULTS: The combination of veliparib and RT inhibited colony formation in the majority of PDCLs tested. The PDCL, RN1 showed significantly reduced levels of the homologous repair protein, Mre11 and a heightened response to PARP inhibition measured by increased apoptosis and decreased colony formation. The oral administration of veliparib (12.5 mg/kg, twice daily for 5 days in a 28-day treatment cycle) in combination with whole brain RT (4 Gy) induced apoptosis (Tunel staining) and decreased cell proliferation (Ki67 staining) in a PDX of MGMT unmethylated GBM. Significantly longer survival times of the PDX treated with the combination treatment were recorded compared to RT only or veliparib only. CONCLUSIONS: Our results demonstrate preclinical efficacy of targeting PARP at multiple levels and provide a new approach for the treatment of MGMT unmethylated GBM.


Subject(s)
Benzimidazoles/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , DNA Methylation/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/drug effects , Benzimidazoles/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Mice, Nude , Survival Analysis
9.
J Neurooncol ; 132(3): 359-372, 2017 05.
Article in English | MEDLINE | ID: mdl-28293764

ABSTRACT

Glioblastoma (GBM) is the most common malignant brain tumour in adults. Current prognosis with standard treatment is poor. Immunotherapy is a new paradigm in tumour management. Specifically, recent advances in the field of immune checkpoint molecules have led to dramatic results in many cancers. Inhibition of one particular, programmed cell death-1 (PD-1) has recently been shown to be highly effective in melanoma and non-small cell lung cancer. There has also been recent data to suggest potential benefit in GBM. There also appears to be a relationship between immune checkpoint inhibition and hypermutation, in particular with the mismatch repair process. In this review we look at the current knowledge of immune checkpoint inhibitors with a focus on the PD-1 pathway. We will also review the evidence of PD-1 inhibition in GBM and the role of hypermutation in PD-1 inhibition.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Glioblastoma/drug therapy , Glioblastoma/immunology , Immunotherapy/methods , Animals , Antineoplastic Agents, Immunological/pharmacology , Humans , Phenotype , Tumor Escape/drug effects , Tumor Escape/immunology
10.
J Neurooncol ; 127(3): 427-34, 2016 May.
Article in English | MEDLINE | ID: mdl-26900075

ABSTRACT

Glioblastoma (GBM) is the most aggressive malignant brain tumor in adults. Improvements in the treatment of GBM have remained static since the advent of the standard therapy which includes radiation with concurrent and adjuvant temozolomide treatment. Developing treatment and diagnostic or companion biomarker combinations is transforming the way we treat numerous cancers. However, can this emerging paradigm be also effective for GBM? Can GBM be treated the same way as other cancers? Here we review the challenges for a personalized molecular targeted therapeutic approach in GBM. The specific challenges for establishing a personalized molecular targeted medicine program for GBM patients include overcoming the blood brain barrier, unravelling the intra- and inter-heterogeneity that exists and the importance of developing more relevant animal models that recapitulate a patient's GBM tumor.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Brain Neoplasms/genetics , Glioblastoma/genetics , Humans , Mutation/genetics , Neoplasm Proteins/genetics
11.
Tumour Biol ; 36(1): 153-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25534238

ABSTRACT

The survival trends for glioblastoma (GBM) patients have remained largely static, reflecting a lack of improvement in the therapeutic options for patients. Less than 5 % of newly diagnosed GBM survives more than 5 years. Tumor relapse is nearly universal and the majority of patients do not respond to further systemic therapy. The results from phase II studies conducted with recurrent GBM patients have not translated to successful confirmatory studies and thus we have reached a significant roadblock in the development of new treatments for patients with recurrent GBM. The development of new, active, and potentially targeted drugs for the treatment of recurrent GBM represents a major unmet need. The incorporation of diagnostic/companion biomarker combinations into the phase II studies and appropriate stratification of the patients is lagging significantly behind other larger cancer groups such as breast, non-small cell lung cancer, and melanoma. We herein carried out a systematic review of the phase II clinical studies conducted in patients with recurrent GBM (2010-2013 inclusive) to assess the degree of biomarker incorporation within the clinical trial design.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplasm Recurrence, Local/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Clinical Trials, Phase II as Topic , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Neoplasm Recurrence, Local/prevention & control , Treatment Outcome
12.
J Biol Chem ; 288(52): 37355-64, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24265321

ABSTRACT

Studies in cell culture and mouse models of cancer have indicated that the soluble sphingolipid metabolite sphingosine 1-phosphate (S1P) promotes cancer cell proliferation, survival, invasiveness, and tumor angiogenesis. In contrast, its metabolic precursor ceramide is prodifferentiative and proapoptotic. To determine whether sphingolipid balance plays a significant role in glioma malignancy, we undertook a comprehensive analysis of sphingolipid metabolites in human glioma and normal gray matter tissue specimens. We demonstrate, for the first time, a systematic shift in sphingolipid metabolism favoring S1P over ceramide, which increases with increasing cancer grade. S1P content was, on average, 9-fold higher in glioblastoma tissues compared with normal gray matter, whereas the most abundant form of ceramide in the brain, C18 ceramide, was on average 5-fold lower. Increased S1P content in the tumors was significantly correlated with increased sphingosine kinase 1 (SPHK1) and decreased sphingosine phosphate phosphatase 2 (SGPP2) expression. Inhibition of S1P production by cultured glioblastoma cells, using a highly potent and selective SPHK1 inhibitor, blocked angiogenesis in cocultured endothelial cells without affecting VEGF secretion. Our findings validate the hypothesis that an altered ceramide/S1P balance is an important feature of human cancers and support the development of SPHK1 inhibitors as antiangiogenic agents for cancer therapy.


Subject(s)
Brain Neoplasms/metabolism , Ceramides/biosynthesis , Glioblastoma/metabolism , Lipid Metabolism , Lysophospholipids/biosynthesis , Neovascularization, Pathologic/metabolism , Sphingosine/analogs & derivatives , Angiogenesis Inhibitors/therapeutic use , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Ceramides/genetics , Enzyme Inhibitors/therapeutic use , Follow-Up Studies , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Lysophospholipids/genetics , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/biosynthesis , Sphingosine/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Tumour Biol ; 35(2): 1459-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24068568

ABSTRACT

Reduced levels of specific microRNA in cancer are frequently reported and associated with attenuated cancer genes and associated pathways. We previously reported a loss of miR-124a in glioblastoma (GBM) patient specimens; however, the upstream causes of this loss are largely unknown. Loss of miR-124a has been attributed to hypermethylation while other studies have shown miR-124a to be regulated by the repressor-element-1-silencing transcription factor (REST, also known as neuron-restrictive silencing factor). This current study looked at both epigenetic and transcription factor regulation as potential mechanisms resulting in the loss of miR-124a expression in GBM patient specimens and cell lines. Hypermethylation of miR-124a was observed in 82 % of GBM patient specimens (n = 56). In vitro miR-124a expression levels also increased after treatment of several patient-derived cell lines with 5-aza-2'-deoxycytidine. Additionally, we also demonstrated a positive interaction between REST activity and miR-124a using a luciferase-binding assay and we correlated the reciprocal expression of REST and miR-124a in our clinical cohort. This result indicates that miR-124a expression may also be modulated through the upstream targeting of REST. Preclinical studies involving inhibitors of REST and treatment with demethylating agents with the intent to increase miR-124a levels could be interesting.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , Aged , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioblastoma/pathology , Humans , Middle Aged
14.
Neurooncol Adv ; 5(1): vdad124, 2023.
Article in English | MEDLINE | ID: mdl-37841696

ABSTRACT

Background: There is an immunologic rationale to evaluate immunotherapy in the older glioblastoma population, who have been underrepresented in prior trials. The NUTMEG study evaluated the combination of nivolumab and temozolomide in patients with glioblastoma aged 65 years and older. Methods: NUTMEG was a multicenter 2:1 randomized phase II trial for patients with newly diagnosed glioblastoma aged 65 years and older. The experimental arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant nivolumab and temozolomide. The standard arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant temozolomide. The primary objective was to improve overall survival (OS) in the experimental arm. Results: A total of 103 participants were randomized, with 69 in the experimental arm and 34 in the standard arm. The median (range) age was 73 (65-88) years. After 37 months of follow-up, the median OS was 11.6 months (95% CI, 9.7-13.4) in the experimental arm and 11.8 months (95% CI, 8.3-14.8) in the standard arm. For the experimental arm relative to the standard arm, the OS hazard ratio was 0.85 (95% CI, 0.54-1.33). In the experimental arm, there were three grade 3 immune-related adverse events which resolved, with no unexpected serious adverse events. Conclusions: Due to insufficient evidence of benefit with nivolumab, the decision was made not to transition to a phase III trial. No new safety signals were identified with nivolumab. This complements the existing series of immunotherapy trials. Research is needed to identify biomarkers and new strategies including combinations.

15.
Anal Chem ; 84(1): 470-4, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22111688

ABSTRACT

Selected reaction monitoring (SRM) is a mass spectrometry-based approach commonly used to increase analytical sensitivity and selectively for specific compounds in complex metabolomic samples. While the goal of well-designed SRM methods is to monitor for unique precursor-product ion pairs, in practice this is not always possible due to the diversity of the metabome and the resolution limits of mass spectrometers that are capable of SRM. Isobaric or near-isobaric precursor ions with different chromatographic properties but identical product ions often arise in complex samples. Without analytical standards, such metabolites will go undetected by conventional data analysis methods. Furthermore, a single SRM method may include simultaneous monitoring of tens to hundreds of different metabolites across multiple samples making quantification of all detected ions a challenging task. To facilitate the analysis of SRM data from complex metabolomic samples, we have developed the Metabolite Mass Spectrometry Analysis Tool (MMSAT). MMSAT is a web-based tool that objectively quantifies every metabolite peak detected in a set of samples and aligns peaks across multiple samples to enable quantitative comparison of each metabolite between samples. The analysis incorporates quantification of multiple peaks/ions that have different chromatographic retention times but are detected within a single SRM transition. We compare the performance of MMSAT against existing tools using a human glioblastoma tissue extract and illustrate its ability to automatically quantify multiple precursors within each of three different transitions. The Web-interface and source code is avaliable at http://www.cancerresearch.unsw.edu.au/crcweb.nsf/page/MMSAT .


Subject(s)
Automation , Mass Spectrometry/methods , Algorithms , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Humans
16.
J Neurooncol ; 109(1): 35-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22528800

ABSTRACT

Increasing evidence suggests that an inflammatory microenvironment promotes invasion by glioblastoma (GBM) cells. Together with p38 mitogen-activated protein kinase (MAPK) activation being regarded as promoting inflammation, we hypothesized that elevated inflammatory cytokine secretion and p38 MAPK activity contribute to expansion of GBMs. Here we report that IL-1ß, IL-6, and IL-8 levels and p38 MAPK activity are elevated in human glioblastoma specimens and that p38 MAPK inhibitors attenuate the secretion of pro-inflammatory cytokines by microglia and glioblastoma cells. RNAi knockdown and immunoprecipitation experiments suggest that the p38α MAPK isoform drives inflammation in GBM cells. Importantly, p38 MAPK inhibition strongly reduced invasion of U251 glioblastoma cells in an inflammatory microenvironment, providing evidence for a p38 MAPK-regulated link between inflammation and invasiveness in GBM pathophysiology.


Subject(s)
Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/pathology , Interleukin-1beta/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Wound Healing/drug effects , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Immunoprecipitation , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 14/genetics , Neoplasm Invasiveness , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
17.
Biochem Biophys Res Commun ; 410(3): 574-80, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21683064

ABSTRACT

Sphingolipids are ubiquitous components of eukaryotic cells and sphingolipid metabolites, such as the long chain base phosphate (LCB-P), sphingosine 1 phosphate (S1P) and ceramide (Cer) are important regulators of apoptosis in animal cells. This study evaluated the role of LCB-Ps in regulating apoptotic-like programmed cell death (AL-PCD) in plant cells using commercially available S1P as a tool. Arabidopsis cell cultures were exposed to a diverse array of cell death-inducing treatments (including Cer) in the presence of S1P. Rates of AL-PCD and cell survival were recorded using vital stains and morphological markers of AL-PCD. Internal LCB-P levels were altered in suspension cultured cells using inhibitors of sphingosine kinase and changes in rates of death in response to heat stress were evaluated. S1P reduced AL-PCD and promoted cell survival in cells subjected to a range of stresses. Treatments with inhibitors of sphingosine kinase lowered the temperature which induced maximal AL-PCD in cell cultures. The data supports the existence of a sphingolipid rheostat involved in controlling cell fate in Arabidopsis cells and that sphingolipid regulation of cell death may be a shared feature of both animal apoptosis and plant AL-PCD.


Subject(s)
Apoptosis , Arabidopsis/physiology , Ceramides/physiology , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Arabidopsis/cytology , Arabidopsis/drug effects , Cells, Cultured , Ceramides/pharmacology , Heat-Shock Response , Lysophospholipids/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/pharmacology , Sphingosine/physiology
18.
Cancers (Basel) ; 13(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34771529

ABSTRACT

BACKGROUND: Identification of prognostic biomarkers in cancers is a crucial step to improve overall survival (OS). Although mutations in tumour protein 53 (TP53) is prevalent in astrocytoma, the prognostic effects of TP53 mutation are unclear. METHODS: In this retrospective study, we sequenced TP53 exons 1 to 10 in a cohort of 102 lower-grade glioma (LGG) subtypes and determined the prognostic effects of TP53 mutation in astrocytoma and oligodendroglioma. Publicly available datasets were analysed to confirm the findings. RESULTS: In astrocytoma, mutations in TP53 codon 273 were associated with a significantly increased OS compared to the TP53 wild-type (HR (95% CI): 0.169 (0.036-0.766), p = 0.021). Public datasets confirmed these findings. TP53 codon 273 mutant astrocytomas were significantly more chemosensitive than TP53 wild-type astrocytomas (HR (95% CI): 0.344 (0.13-0.88), p = 0.0148). Post-chemotherapy, a significant correlation between TP53 and YAP1 mRNA was found (p = 0.01). In O (6)-methylguanine methyltransferase (MGMT) unmethylated chemotherapy-treated astrocytoma, both TP53 codon 273 and YAP1 mRNA were significant prognostic markers. In oligodendroglioma, TP53 mutations were associated with significantly decreased OS. CONCLUSIONS: Based on these findings, we propose that certain TP53 mutant astrocytomas are chemosensitive through the involvement of YAP1, and we outline a potential mechanism. Thus, TP53 mutations may be key drivers of astrocytoma therapeutic efficacy and influence survival outcomes.

19.
Cancers (Basel) ; 13(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557152

ABSTRACT

We have developed a platform, termed PathoFusion, which is an integrated system for marking, training, and recognition of pathological features in whole-slide tissue sections. The platform uses a bifocal convolutional neural network (BCNN) which is designed to simultaneously capture both index and contextual feature information from shorter and longer image tiles, respectively. This is analogous to how a microscopist in pathology works, identifying a cancerous morphological feature in the tissue context using first a narrow and then a wider focus, hence bifocal. Adjacent tissue sections obtained from glioblastoma cases were processed for hematoxylin and eosin (H&E) and immunohistochemical (CD276) staining. Image tiles cropped from the digitized images based on markings made by a consultant neuropathologist were used to train the BCNN. PathoFusion demonstrated its ability to recognize malignant neuropathological features autonomously and map immunohistochemical data simultaneously. Our experiments show that PathoFusion achieved areas under the curve (AUCs) of 0.985 ± 0.011 and 0.988 ± 0.001 in patch-level recognition of six typical pathomorphological features and detection of associated immunoreactivity, respectively. On this basis, the system further correlated CD276 immunoreactivity to abnormal tumor vasculature. Corresponding feature distributions and overlaps were visualized by heatmaps, permitting high-resolution qualitative as well as quantitative morphological analyses for entire histological slides. Recognition of more user-defined pathomorphological features can be added to the system and included in future tissue analyses. Integration of PathoFusion with the day-to-day service workflow of a (neuro)pathology department is a goal. The software code for PathoFusion is made publicly available.

20.
Front Mol Biosci ; 8: 633344, 2021.
Article in English | MEDLINE | ID: mdl-33996894

ABSTRACT

Medulloblastoma is the most common malignant childhood brain tumor, and 5-year overall survival rates are as low as 40% depending on molecular subtype, with new therapies critically important. As radiotherapy and chemotherapy act through the induction of DNA damage, the sensitization of cancer cells through the inhibition of DNA damage repair pathways is a potential therapeutic strategy. The poly-(ADP-ribose) polymerase (PARP) inhibitor veliparib was assessed for its ability to augment the cellular response to radiation-induced DNA damage in human medulloblastoma cells. DNA repair following irradiation was assessed using the alkaline comet assay, with veliparib inhibiting the rate of DNA repair. Veliparib treatment also increased the number of γH2AX foci in cells treated with radiation, and analysis of downstream pathways indicated persistent activation of the DNA damage response pathway. Clonogenicity assays demonstrated that veliparib effectively inhibited the colony-forming capacity of medulloblastoma cells, both as a single agent and in combination with irradiation. These data were then validated in vivo using an orthotopic implant model of medulloblastoma. Mice harboring intracranial D425 medulloblastoma xenografts were treated with vehicle, veliparib, 18 Gy multifractionated craniospinal irradiation (CSI), or veliparib combined with 18 Gy CSI. Animals treated with combination therapy exhibited reduced tumor growth rates concomitant with increased intra-tumoral apoptosis observed by immunohistochemistry. Kaplan-Meier analyses revealed a statistically significant increase in survival with combination therapy compared to CSI alone. In summary, PARP inhibition enhanced radiation-induced cytotoxicity of medulloblastoma cells; thus, veliparib or other brain-penetrant PARP inhibitors are potential radiosensitizing agents for the treatment of medulloblastoma.

SELECTION OF CITATIONS
SEARCH DETAIL