Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
CNS Neurosci Ther ; 20(5): 429-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24483339

ABSTRACT

AIMS: Down-regulation of AJAP1 in glioblastoma multiforme (GBM) has been reported. However, the expression profiles of AJAP1 in gliomas and the underlying mechanisms of AJAP1 function on invasion are still poorly understood. METHODS: The gene profiles of AJAP1 in glioma patients were studied among four independent cohorts. Confocal imaging was used to analyze the AJAP1 localization. After AJAP1 overexpression in GBM cell lines, cellular polarity, cytoskeleton distribution, and antitumor effect were investigated in vitro and in vivo. RESULTS: AJAP1 expression was significantly decreased in gliomas compared with normal brain in REMBRANDT and CGCA cohorts. Additionally, low AJAP1 expression was associated with worse survival in GBMs in REMBRANDT and TCGA U133A cohorts and was significantly associated with classical and mesenchymal subtypes of GBMs among four cohorts. Confocal imaging indicated AJAP1 localized in cell membranes in low-grade gliomas and AJAP1-overexpressing GBM cells, but difficult to assess in high-grade gliomas due to its absence. AJAP1 overexpression altered the cytoskeleton and cellular polarity in vitro and inhibited the tumor growth in vivo. CONCLUSIONS: AJAP1 is dysregulated at an early stage of gliomagenesis and may suppress glioma cell invasion and proliferation, which suggests that AJAP1 may be a potential diagnostic and prognostic marker for gliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Cell Adhesion Molecules/metabolism , Cytoskeleton/metabolism , Glioma/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cohort Studies , Cytoskeleton/ultrastructure , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , Kaplan-Meier Estimate , Mice, Nude , Microscopy, Confocal , Neoplasm Staging , Neoplasm Transplantation
2.
J Neurooncol ; 98(1): 1-7, 2010 May.
Article in English | MEDLINE | ID: mdl-19898744

ABSTRACT

A major obstacle in glioblastoma (GBM) therapy is the restrictive nature of the blood-brain barrier (BBB). Convection-enhanced delivery (CED) is a novel method of drug administration which allows direct parenchymal infusion of therapeutics, bypassing the BBB. MR1-1 is a novel recombinant immunotoxin that targets the GBM tumor-specific antigen EGFRvIII and can be delivered via CED infusion. However, drug distribution via CED varies dramatically, which necessitates active monitoring. Gadolinium conjugated to diethylenetriamine penta-acetic acid (Gd-DTPA) is a commonly used MRI contrast agent which can be co-infused with therapies using CED and may be useful in monitoring infusion leak and early distribution. Forty immunocompetent rats were implanted with intracerebral cannulas that were connected to osmotic pumps and subsequently randomized into four groups that each received 0.2% human serum albumin (HSA) mixed with a different experimental infusion: (1) 25 ng/ml MR1-1; (2) 0.1 micromol/ml Gd-DTPA; (3) 25 ng/ml MR1-1 and 0.1 micromol/ml Gd-DTPA; (4) 250 ng/ml MR1-1 and 0.1 micromol/ml Gd-DTPA. The rats were monitored clinically for 6 weeks then necropsied and histologically assessed for CNS toxicity. All rats survived the entirety of the study without clinical or histological toxicity attributable to the study drugs. There was no statistically significant difference in weight change over time among groups (P > 0.999). MR1-1 co-infused with Gd-DTPA via CED is safe in the long-term setting in a pre-clinical animal model. Our data supports the use of Gd-DTPA, as a surrogate tracer, co-infused with MR1-1 for drug distribution monitoring in patients with GBM.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Contrast Media/pharmacokinetics , Convection , Drug Delivery Systems , Gadolinium DTPA/pharmacokinetics , Immunotoxins/pharmacokinetics , Animals , Antibodies, Monoclonal/administration & dosage , Brain/drug effects , Contrast Media/administration & dosage , Dose-Response Relationship, Drug , Epidermal Growth Factor/immunology , Gadolinium DTPA/administration & dosage , Immunocompetence , Immunotoxins/administration & dosage , Magnetic Resonance Imaging , Protein Precursors/immunology , Rats , Rats, Inbred F344 , Tissue Distribution
3.
Nat Genet ; 41(4): 465-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19270706

ABSTRACT

We used high-resolution SNP genotyping to identify regions of genomic gain and loss in the genomes of 212 medulloblastomas, malignant pediatric brain tumors. We found focal amplifications of 15 known oncogenes and focal deletions of 20 known tumor suppressor genes (TSG), most not previously implicated in medulloblastoma. Notably, we identified previously unknown amplifications and homozygous deletions, including recurrent, mutually exclusive, highly focal genetic events in genes targeting histone lysine methylation, particularly that of histone 3, lysine 9 (H3K9). Post-translational modification of histone proteins is critical for regulation of gene expression, can participate in determination of stem cell fates and has been implicated in carcinogenesis. Consistent with our genetic data, restoration of expression of genes controlling H3K9 methylation greatly diminishes proliferation of medulloblastoma in vitro. Copy number aberrations of genes with critical roles in writing, reading, removing and blocking the state of histone lysine methylation, particularly at H3K9, suggest that defective control of the histone code contributes to the pathogenesis of medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Cerebellar Neoplasms/enzymology , Gene Amplification , Gene Deletion , Genes, Tumor Suppressor , Genome, Human , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine/metabolism , Medulloblastoma/enzymology , Polymorphism, Single Nucleotide , Protein Processing, Post-Translational/genetics , Sequence Deletion
4.
Cancer ; 106(10): 2224-32, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16586497

ABSTRACT

BACKGROUND: Exercise may represent a supportive intervention that may complement existing neurooncologic therapies and address a multitude of therapy-induced debilitating side effects in patients with brain tumors. Given the limited evidence, the authors conducted a survey to examine the exercise patterns of brain tumor patients across the cancer trajectory. METHODS: Using a cross-sectional design, 386 brain tumor patients who received treatment at the Brain Tumor Center at Duke University were sent a questionnaire that assessed self-reported exercise behavior prior to diagnosis, during adjuvant therapy, and after the completion of therapy. RESULTS: The response rate was 28% (106 of 383 patients). Descriptive analyses indicated that 42%, 38%, and 41% of participants, respectively, met national exercise prescription guidelines prior to diagnosis, during treatment, and after the completion of adjuvant therapy. Repeated measures analyses indicated no significant changes in the majority of exercise behavior outcomes over the cancer trajectory. However, exploratory analyses indicated that males and younger participants may be at the greatest risk of reducing exercise levels after a brain tumor diagnosis. These analyses remained unchanged after controlling for relevant demographic and medical covariates. CONCLUSIONS: A relatively high percentage of brain tumor patients are exercising at recommended levels across the cancer trajectory. Moreover, these patients have unique exercise patterns that may be modified by select demographic variables. This preliminary study provides important informative data for future studies examining the potential role of exercise in patients diagnosed with neurologic malignancies.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/epidemiology , Exercise/physiology , Health Behavior , Adult , Age Distribution , Aged , Analysis of Variance , Brain Neoplasms/therapy , Cross-Sectional Studies , Disease-Free Survival , Female , Humans , Life Style , Male , Middle Aged , Probability , Prognosis , Reference Values , Risk Assessment , Sex Distribution , Sickness Impact Profile , Surveys and Questionnaires , Survival Analysis
5.
Cancer Chemother Pharmacol ; 56(6): 585-93, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16012865

ABSTRACT

Substance P receptor (SPR), a G protein-coupled receptor (GPCR), is found in human glioblastomas, and has been implicated in their growth. Consistent with a role for SPR in cell growth, activation of SPR in U373 MG human glioblastoma cells leads to the phosphorylation of mitogen-activated protein kinases [extracellular signal-regulated kinase 1 and 2 (ERK1/2)] and stimulation of cell proliferation. The purpose of the present study was to elucidate the pathway through which these actions occur. Using either the epidermal growth factor receptor (EGFR) kinase inhibitor, AG 1478, or a small-interfering RNA (siRNA) directed against human EGFR, we found that transactivation of EGFR by SPR is only marginally involved in SP-dependent ERK1/2 phosphorylation. Src, however, is shown to be a major component of SPR signaling because the Src kinase inhibitor, PP2, and a kinase-dead Src mutant both inhibit SP-dependent ERK1/2 phosphorylation. We also report that SPR stimulates the phosphorylation of protein kinase Cdelta(PKCdelta), and that this stimulation is blocked by PP2. SP-dependent ERK1/2 phosphorylation is also blocked by rottlerin, a PKCdelta inhibitor, and the calcium scavenger, BAPTA/AM. Finally, rottlerin and PP2 were both found to inhibit the growth of several glioblastoma cell lines, underscoring the potential of these agents to block glioblastoma growth.


Subject(s)
Glioblastoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C-delta/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , Acetophenones/pharmacology , Benzopyrans/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Protein Kinase C-delta/antagonists & inhibitors , Pyrimidines/pharmacology , Quinazolines , RNA, Small Interfering/pharmacology , Tyrphostins/pharmacology
6.
Nucl Med Biol ; 31(3): 345-55, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15028247

ABSTRACT

We have obtained encouraging responses in recent Phase I studies evaluating (131)I-labeled human/murine chimeric 81C6 anti-tenascin monoclonal antibody (ch81C6) administered into surgically-created tumor resection cavities in brain tumor patients. However, because the blood clearance is slow, hematologic toxicity has been higher than seen with murine 81C6 (mu81C6). In the current study, a series of paired-label experiments were performed in athymic mice bearing subcutaneous D-245 MG human glioma xenografts to compare the biodistribution of the fragment ch81C6 F(ab')(2) labeled using Iodogen to a) intact ch81C6, b) mu81C6, and c) ch81C6 F(ab')(2) labeled using N-succinimidyl 3-[(131)I]iodobenzoate. Tumor retention of radioiodine activity for the F(ab')(2) fragment was comparable to that for intact ch81C6 for the first 24 h and to that for mu81C6 for the first 48 h; as expected, blood and other normal tissue levels declined faster for ch81C6 F(ab')(2.) Radiation dosimetry calculations suggest that (131)I-labeled ch81C6 F(ab')(2) may warrant further evaluation as a targeted radiotherapeutic for the treatment of brain tumors.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Glioma/metabolism , Iodine Radioisotopes/pharmacokinetics , Radioimmunotherapy/methods , Animals , Antibodies, Monoclonal/therapeutic use , Body Burden , Drug Evaluation, Preclinical , Feasibility Studies , Glioma/radiotherapy , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/therapeutic use , Iodine Radioisotopes/therapeutic use , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Radiation Dosage , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Tissue Distribution , Tumor Cells, Cultured
7.
Int J Radiat Oncol Biol Phys ; 54(4): 1259-75, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419456

ABSTRACT

PURPOSE: To establish the theoretical framework and study the feasibility of (211)At-labeled anti-tenascin chimeric 81C6 monoclonal antibody (mAb) as anti-vascular endoradiotherapy for the treatment of glioblastoma multiforme (GBM) tumors. METHODS AND MATERIALS: The morphology of blood vessels from histologic images was analyzed and used along with reaction-diffusion equations to assess the activity concentration of (211)At-labeled chimeric 81C6 mAb in GBM tumor and normal-brain tissue. Alpha particle microdosimetry was then used to assess the survival probability and average absorbed dose for tumor and normal tissue endothelial cells (ECs) per unit vascular cumulated activity concentration q(source) (MBq-s g(-1)). In turn, these survival probabilities were used to assess the probability of failure Phi for a single vessel. Furthermore, using the vessel density, the specific tumor control probability per unit mass of tumor tissue (tcp) and the specific normal-tissue complication probability per unit mass of normal-brain tissue (ntcp) were estimated. The specific tumor control probability, tcp, was used to assess the overall tumor control probability (TCP) as a function of tumor mass. RESULTS: The levels of (211)At-labeled ch81C6 mAb cumulated activity concentration in GBM tumor tissue were approximately five times higher than that in normal-brain tissue. Thus, the average absorbed dose to tumor ECs was higher than that of normal tissue ECs, and the survival probability for GBM ECs was lower than for normal-brain tissue ECs. Consequently, the resulting vessel-failure probability, Phi, for GBM tumor and for normal-brain tissue differ considerably, yielding a q(source) range between 10(3) and 10(4) MBq-s g(-1). CONCLUSIONS: This theoretical analysis demonstrated that (211)At-labeled chimeric 81C6 is an effective anti-vascular therapy for the treatment of GBM tumors, yielding a tcp higher than 0.999 for vascular cumulated activity concentrations q(source) higher than 1 x 10(4) MBq-s g(-1), while yielding a low probability for normal-brain tissue damage.


Subject(s)
Alpha Particles/therapeutic use , Astatine/therapeutic use , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Radioimmunotherapy , Tenascin/immunology , Boron Neutron Capture Therapy , Brain/radiation effects , Humans , Probability , Radiometry
SELECTION OF CITATIONS
SEARCH DETAIL
...