Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Clin Cancer Res ; 28(8): 1595-1602, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35165100

ABSTRACT

PURPOSE: Tyrosine kinase inhibitors (TKI) have poor efficacy in patients with glioblastoma (GBM). Here, we studied whether this is predominantly due to restricted blood-brain barrier penetration or more to biological characteristics of GBM. PATIENTS AND METHODS: Tumor drug concentrations of the TKI sunitinib after 2 weeks of preoperative treatment was determined in 5 patients with GBM and compared with its in vitro inhibitory concentration (IC50) in GBM cell lines. In addition, phosphotyrosine (pTyr)-directed mass spectrometry (MS)-based proteomics was performed to evaluate sunitinib-treated versus control GBM tumors. RESULTS: The median tumor sunitinib concentration of 1.9 µmol/L (range 1.0-3.4) was 10-fold higher than in concurrent plasma, but three times lower than sunitinib IC50s in GBM cell lines (median 5.4 µmol/L, 3.0-8.5; P = 0.01). pTyr-phosphoproteomic profiles of tumor samples from 4 sunitinib-treated versus 7 control patients revealed 108 significantly up- and 23 downregulated (P < 0.05) phosphopeptides for sunitinib treatment, resulting in an EGFR-centered signaling network. Outlier analysis of kinase activities as a potential strategy to identify drug targets in individual tumors identified nine kinases, including MAPK10 and INSR/IGF1R. CONCLUSIONS: Achieved tumor sunitinib concentrations in patients with GBM are higher than in plasma, but lower than reported for other tumor types and insufficient to significantly inhibit tumor cell growth in vitro. Therefore, alternative TKI dosing to increase intratumoral sunitinib concentrations might improve clinical benefit for patients with GBM. In parallel, a complex profile of kinase activity in GBM was found, supporting the potential of (phospho)proteomic analysis for the identification of targets for (combination) treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Indoles , Proteomics , Pyrroles/therapeutic use , Sunitinib/therapeutic use
2.
Cancers (Basel) ; 12(10)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023148

ABSTRACT

In this Special Issue of Cancer, a series of 10 papers (seven papers, three reviews) on Neurofibromatosis is presented by international leaders in this field of research [...].

3.
J Neurooncol ; 142(2): 337-345, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30680510

ABSTRACT

PURPOSE: The highly vascular malignant brain tumor glioblastoma (GBM) appears to be an ideal target for anti-angiogenic therapy; however, clinical trials to date suggest the VEGF antibody bevacizumab affects only progression-free survival. Here we analyze a group of patients with GBM who received bevacizumab treatment at recurrence and are stratified according to tumor molecular and genomic profile (TCGA classification), with the goal of identifying molecular predictors of the response to bevacizumab. METHODS: We performed a retrospective review of patients with a diagnosis of glioblastoma who were treated with bevacizumab in the recurrent setting at our hospital, from 2006 to 2014. Treatment was discontinued by the treating neuro-oncologists, based on clinical and radiographic criteria. Pre- and post-treatment imaging and genomic subtype were available on 80 patients. We analyzed time on bevacizumab and time to progression. EGFR gene amplification was determined by FISH. RESULTS: Patients with classical tumors had a significantly shorter time on bevacizumab than mesenchymal, and proneural patients (2.7 vs. 5.1 vs. 6.4 and 6.0 months respectively, p = 0.011). Classical subtype and EGFR gene amplification were significantly associated with a shorter time to progression both in univariate (p < 0.001 and p = 0.007, respectively) and multivariate analysis (both p = 0.010). CONCLUSION: EGFR gene amplification and classical subtype by TCGA analysis are associated with significantly shorter time to progression for patients with recurrent GBM when treated with bevacizumab. These findings can have a significant impact on decision-making and should be further validated prospectively.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , ErbB Receptors/genetics , Female , Follow-Up Studies , Gene Amplification , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Pharmacogenomic Variants , Retrospective Studies , Survival Analysis
4.
Neurosurgery ; 83(1): 76-85, 2018 07 01.
Article in English | MEDLINE | ID: mdl-28973417

ABSTRACT

BACKGROUND: Thalamic glioblastomas (GBMs) represent a significant neurosurgical challenge. In view of the low incidence of these tumors, outcome data and management strategies are not well defined. OBJECTIVE: To identify the natural history and factors associated with survival in patients with thalamic glioblastoma. METHODS: A retrospective review of all patients with thalamic glioblastoma over a 10-yr period was performed. Presenting clinical, radiological, and outcome data were collected. Chi-squared and Fisher's exact tests were used to compare clinical characteristics across tumor groups. Cox proportional hazard models were utilized to investigate variables of interest with regard to overall survival. RESULTS: Fifty-seven patients met inclusion criteria, with a median age of 53 and median Karnofsky Performance Scale (KPS) score of 80. The most common presenting symptoms were weakness, confusion, and headache. Hydrocephalus was present in 47% of patients preoperatively. Stereotactic biopsy was performed in 47 cases, and 10 patients underwent craniotomy. The median overall survival was 12.2 mo. Higher KPS, younger age, and cerebrospinal fluid (CSF) diversion were correlated with better overall survival univariately, respectively, while the presence of language deficits at initial presentation was associated with poorer survival. In multivariate analysis, the only significant predictor of survival was presenting KPS. CONCLUSION: The overall survival of patients with thalamic glioblastoma is comparable to unresectable lobar supratentorial GBMs. Younger patients and those with good presenting functional status had improved survival. Midbrain involvement by the tumor is not a negative prognostic factor. Improved therapies are needed, and patients should be considered for early trial involvement and aggressive upfront therapy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/surgery , Female , Glioblastoma/mortality , Glioblastoma/surgery , Humans , Middle Aged , Proportional Hazards Models , Retrospective Studies , Treatment Outcome , Young Adult
5.
J Neurooncol ; 129(2): 221-30, 2016 09.
Article in English | MEDLINE | ID: mdl-27444431

ABSTRACT

Surgery followed by chemoradiation and adjuvant chemotherapy is standard of care for patients with a glioblastoma (GBM). Due to its limited benefit, an upfront method to predict dismal outcome would prevent unnecessary toxic treatment. We searched for a predictive blood derived biomarker in a cohort of 55 patients with GBM. Increasing age (HR 1.03, 95 % CI 1.01-1.06), and postoperative tumor residue (HR 1.07, 95 % CI 1.02-1.15) were independently associated with unfavourable progression free survival (PFS) in these patients. Corticosteroid use before start of chemoradiaton was strongly predictive for outcome (HR 3.26, 95 % CI 1.67-6.39) with a mean PFS and OS in patients using corticosteroids of 7.3 and 14.6 months, versus 16.1 and 21.6 months in patients not using corticosteroids (p = 0.0005, p < 0.0067 respectively). Despite earlier reports, blood concentrations of YKL-40, Fetuin-a and haptoglobin were not predictive for response. In addition, serum peptide profiles, determined by MALDI-TOF mass spectroscopy, were not predictive as well. In conclusion, further biomarker discovery studies are needed to predict treatment outcome for patients with GBM in the near future.


Subject(s)
Brain Neoplasms/blood , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Glioblastoma/blood , Glioblastoma/therapy , alpha-2-HS-Glycoprotein/metabolism , Adolescent , Adrenal Cortex Hormones/blood , Adult , Aged , Blood Platelets/pathology , Chitinase-3-Like Protein 1/blood , Cohort Studies , Disease-Free Survival , Female , Haptoglobins/metabolism , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Proteomics , Treatment Outcome , Young Adult
6.
Curr Protoc Stem Cell Biol ; Chapter 3: Unit3.5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22135084

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most aggressive brain tumors. GBM cell lines used in laboratory studies are frequently passaged in various culture media at high proliferation rates, resulting in significant genetic and molecular alterations. Thus, data obtained in cell lines are often inapplicable to patient tumors. Furthermore, recent studies suggest that there is a stem cell-like hierarchy among GBM cell populations and a crucial role for tumor vasculature in stem cells, as well as tumor growth, which cannot be reproduced in cell line cultures. Our laboratory has developed a novel three-dimensional (3D) organotypic "explant" system of surgical GBM specimens that preserves tumor cells in their original milieu, as well as the cytoarchitecture of the tumor stroma. Our previous study on the role of Notch inhibition has demonstrated a definitive effect on the tumor endothelium that could only be highlighted by this system. In this unit, we describe a detailed protocol for preparing GBM explants, and discuss strengths, as well as limitations of the explant system as an in vitro 3D model of GBM.


Subject(s)
Glioblastoma/pathology , Organ Culture Techniques/methods , Single-Cell Analysis/methods , Humans , Suspensions , Tumor Cells, Cultured
7.
Nature ; 468(7325): 829-33, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21102433

ABSTRACT

Glioblastoma (GBM) is among the most aggressive of human cancers. A key feature of GBMs is the extensive network of abnormal vasculature characterized by glomeruloid structures and endothelial hyperplasia. Yet the mechanisms of angiogenesis and the origin of tumour endothelial cells remain poorly defined. Here we demonstrate that a subpopulation of endothelial cells within glioblastomas harbour the same somatic mutations identified within tumour cells, such as amplification of EGFR and chromosome 7. We additionally demonstrate that the stem-cell-like CD133(+) fraction includes a subset of vascular endothelial-cadherin (CD144)-expressing cells that show characteristics of endothelial progenitors capable of maturation into endothelial cells. Extensive in vitro and in vivo lineage analyses, including single cell clonal studies, further show that a subpopulation of the CD133(+) stem-like cell fraction is multipotent and capable of differentiation along tumour and endothelial lineages, possibly via an intermediate CD133(+)/CD144(+) progenitor cell. The findings are supported by genetic studies of specific exons selected from The Cancer Genome Atlas, quantitative FISH and comparative genomic hybridization data that demonstrate identical genomic profiles in the CD133(+) tumour cells, their endothelial progenitor derivatives and mature endothelium. Exposure to the clinical anti-angiogenesis agent bevacizumab or to a γ-secretase inhibitor as well as knockdown shRNA studies demonstrate that blocking VEGF or silencing VEGFR2 inhibits the maturation of tumour endothelial progenitors into endothelium but not the differentiation of CD133(+) cells into endothelial progenitors, whereas γ-secretase inhibition or NOTCH1 silencing blocks the transition into endothelial progenitors. These data may provide new perspectives on the mechanisms of failure of anti-angiogenesis inhibitors currently in use. The lineage plasticity and capacity to generate tumour vasculature of the putative cancer stem cells within glioblastoma are novel findings that provide new insight into the biology of gliomas and the definition of cancer stemness, as well as the mechanisms of tumour neo-angiogenesis.


Subject(s)
Cell Differentiation , Endothelial Cells/pathology , Glioblastoma/blood supply , Glioblastoma/pathology , Neovascularization, Pathologic/pathology , Neural Stem Cells/pathology , AC133 Antigen , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antigens, CD/metabolism , Bevacizumab , Cadherins/deficiency , Cadherins/metabolism , Cell Line, Tumor , Cell Lineage , Chromosome Aberrations , Coculture Techniques , Endothelial Cells/metabolism , Female , Glioblastoma/genetics , Glycoproteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Integrin beta4/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neural Stem Cells/metabolism , Peptides/metabolism , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Stem Cells ; 28(6): 1019-29, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20506127

ABSTRACT

Glioblastoma multiforme (GBM) is a highly heterogeneous malignant tumor. Recent data suggests the presence of a hierarchical organization within the GBM cell population that involves cancer cells with stem-like behavior, capable of repopulating the tumor and contributing to its resistance to therapy. Tumor stem cells are thought to reside within a vascular niche that provides structural and functional support. However, most GBM studies involve isolated tumor cells grown under various culture conditions. Here, we use a novel three-dimensional organotypic "explant" system of surgical GBM specimens that preserves cytoarchitecture and tumor stroma along with tumor cells. Notch inhibition in explants results in decreased proliferation and self-renewal of tumor cells but is also associated with a decrease in endothelial cells. When endothelial cells are selectively eliminated from the explants via a toxin conjugate, we also observed a decrease in self-renewal of tumor stem cells. These findings support a critical role for tumor endothelial cells in GBM stem cell maintenance, mediated at least in part by Notch signaling. The explant system further highlighted differences in the response to radiation between explants and isolated tumor neurospheres. Combination treatment with Notch blockade and radiation resulted in a substantial decrease in proliferation and in self-renewal in tumor explants while radiation alone was less effective. This data suggests that the Notch pathway plays a critical role in linking angiogenesis and cancer stem cell self-renewal and is thus a potential therapeutic target. Three-dimensional explant systems provide a novel approach for the study of tumor and microenvironment interactions.


Subject(s)
Cell Separation/methods , Endothelial Cells/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Tissue Culture Techniques/methods , Apoptosis , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Endothelial Cells/cytology , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
9.
Eur J Cancer ; 45(17): 3074-80, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19734041

ABSTRACT

AIM OF THE STUDY: Tumour angiogenesis and invasion are key features of glioblastoma multiforme (GBM). Angiogenesis inhibitors increase progression-free survival (PFS) of recurrent GBM patients. VEGF inhibition controls the bulk tumour growth by inhibition of angiogenesis, but does not inhibit the invasive tumour component. We investigated if invasive tumour growth can be controlled by combining anti-VEGF treatment with irradiation of tumour plus surrounding brain in an orthotopic murine model for GBM. METHODS AND MATERIALS: GBM cell line U251-NG2 was inoculated through a guide screw in the right frontal lobe of 53 athymic nude mice. Pegaptanib (a slow-releasing aptamer against VEGF) was injected in the tumour bed either or not followed by irradiation treatment with implanted I-125 seeds. Pegaptanib and/or irradiation were compared with sham-treated controls, resulting in four groups of 10-15 mice each. After 6 weeks of treatment, histological analysis was performed on all brains. RESULTS: VEGF inhibition by locally deposited pegaptanib decreased tumour blood vessel density, and increased tumour hypoxia. Pegaptanib treatment still allowed the formation of tumour satellites. Irradiation decreased tumour size and suppressed formation of satellites. Combined pegaptanib plus irradiation further increased PFS. Tumour size directly correlated with PFS. CONCLUDING STATEMENT: The anti-tumour effects of local VEGF inhibition are partially circumvented by the formation of invasive tumour satellites. Additional irradiation is effective in slowing down proliferation of these invasive tumour components.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Aptamers, Nucleotide/therapeutic use , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Combined Modality Therapy , Disease Models, Animal , Disease-Free Survival , Female , Glioblastoma/blood supply , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Oncol Rep ; 18(3): 709-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17671724

ABSTRACT

Glioblastoma multiforme (GBM) is a radioresistant tumor. Tumor neoangiogenesis is an important mechanism for tumor sustenance. Angiogenesis is primarily mediated by vascular endothelial growth factor (VEGF), and earlier studies have suggested that VEGF protects human umbilical vein endothelial cells (HUVECs) against high doses of radiation. We tried to extend these findings to other endothelial cell lines and clinically relevant irradiation doses. Therefore, four different endothelial cell lines (HUVEC-C, primary HUVEC-P, an immortalized HUVEC cell line: EC-RF24, and bovine retina endothelial cells: BREC) were cultured without or with recombinant human VEGF165 (rhVEGF165). Cells were irradiated with gamma-rays from a 137Cs-source. Radiosensitivity was determined by proliferation or clonogenic assay. Apoptosis was assayed by flow cytometric determination of the sub-G1 population or by counting nuclear fragmentation. We found that the biologically active rhVEGF165 was able to improve clonogenic survival of HUVEC-C after 2 and 5 Gy. However, rhVEGF165 could not significantly alter the radiosensitivity of all cell lines studied in proliferation assays. rhVEGF165 only slightly reduced apoptosis in HUVEC-C after 3 Gy. In conclusion, the radioprotective effect from rhVEGF165 was found on different endothelial cell lines after clinically relevant radiation doses was negligible. We therefore hypothesize that the high VEGF-levels found in GBM in vivo do not reduce the radiosensitivity of endothelial cells, which is thought to contribute to the strong radioresistance of the tumor vasculature.


Subject(s)
Endothelium, Vascular/physiology , Endothelium, Vascular/radiation effects , Radiation Protection , Vascular Endothelial Growth Factor A/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cesium Radioisotopes , Dose-Response Relationship, Radiation , Humans , Infant, Newborn , Neovascularization, Pathologic , RNA/genetics , RNA/isolation & purification , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...