Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
Oncogene ; 36(43): 6049-6058, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28759044

ABSTRACT

Glioblastomas represent the most aggressive glioma grade and are associated with a poor patient prognosis. The current standard of care, consisting of surgery, radiation and chemotherapy, only results in a median survival of 14 months, underscoring the importance of developing effective new therapeutic strategies. Among the challenges in treating glioblastomas are primary resistance and the rapid emergence of recurrent disease, which can result from tumor cell-intrinsic mechanisms in addition to tumor microenvironment (TME)-mediated extrinsic resistance. Using a PDGF-B-driven proneural glioma mouse model, we assessed a panel of tyrosine kinase inhibitors with different selectivity profiles. We found that PLX3397, an inhibitor of colony stimulating factor-1 receptor (CSF-1R), blocks glioma progression, markedly suppresses tumor cell proliferation and reduces tumor grade. By contrast, the multi-targeted tyrosine kinase inhibitors dovitinib and vatalanib, which directly target tumor cells, exert minimal anti-tumoral effects in vivo, despite killing glioma cells in vitro, suggesting a TME-mediated resistance mechanism may be involved. Interestingly, PLX3397 interferes with tumor-mediated education of macrophages and consequently restores the sensitivity of glioma cells to tyrosine kinase inhibitors in vivo in preclinical combination trials. Our findings thus demonstrate that microenvironmental alteration by CSF-1R blockade renders tumor cells more susceptible to receptor tyrosine kinase inhibition in a preclinical glioblastoma model, which may have important translational relevance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Aminopyridines/administration & dosage , Animals , Becaplermin , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Glioma/genetics , Glioma/pathology , Humans , Mice , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-sis/genetics , Pyrroles/administration & dosage , Tumor Microenvironment/drug effects
3.
PLoS One ; 11(7): e0158634, 2016.
Article in English | MEDLINE | ID: mdl-27462980

ABSTRACT

BACKGROUND: A critical challenge in the management of Glioblastoma Multiforme (GBM) tumors is the accurate diagnosis and assessment of tumor progression in a noninvasive manner. We have identified Membrane-type 1 matrix metalloproteinase (MT1-MMP) as an attractive biomarker for GBM imaging since this protein is actively involved in tumor growth and progression, correlates with tumor grade and is closely associated with poor prognosis in GBM patients. Here, we report the development of an immunoPET tracer for effective detection of MT1-MMP in GBM models. METHODS: An anti-human MT1-MMP monoclonal antibody (mAb), LEM2/15, was conjugated to p-isothiocyanatobenzyl-desferrioxamine (DFO-NCS) for 89Zr labeling. Biodistribution and PET imaging studies were performed in xenograft mice bearing human GBM cells (U251) expressing MT1-MMP and non-expressing breast carcinoma cells (MCF-7) as negative control. Two orthotopic brain GBM models, patient-derived neurospheres (TS543) and U251 cells, with different degrees of blood-brain barrier (BBB) disruption were also used for PET imaging experiments. RESULTS: 89Zr labeling of DFO-LEM2/15 was achieved with high yield (>90%) and specific activity (78.5 MBq/mg). Biodistribution experiments indicated that 89Zr-DFO-LEM2/15 showed excellent potential as a radiotracer for detection of MT1-MMP positive GBM tumors. PET imaging also indicated a specific and prominent 89Zr-DFO-LEM2/15 uptake in MT1-MMP+ U251 GBM tumors compared to MT1-MMP- MCF-7 breast tumors. Results obtained in orthotopic brain GBM models revealed a high dependence of a disrupted BBB for tracer penetrance into tumors. 89Zr-DFO-LEM2/15 showed much higher accumulation in TS543 tumors with a highly disrupted BBB than in U251 orthotopic model in which the BBB permeability was only partially increased. Histological analysis confirmed the specificity of the immunoconjugate in all GBM models. CONCLUSION: A new anti MT1-MMP-mAb tracer, 89Zr-DFO-LEM2/15, was synthesized efficiently. In vivo validation showed high-specific-contrast imaging of MT1-MMP positive GBM tumors and provided strong evidence for utility of MT1-MMP-targeted immunoPET as an alternate to nonspecific imaging of GBM.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , Matrix Metalloproteinase 14/metabolism , Positron-Emission Tomography/methods , Animals , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/metabolism , Brain Neoplasms/enzymology , Cell Line, Tumor , Glioblastoma/enzymology , Humans , Matrix Metalloproteinase 14/immunology , Mice , Mice, Nude , Neoplasm Transplantation , Prognosis , X-Ray Microtomography
4.
Genes Brain Behav ; 8(1): 60-71, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18823404

ABSTRACT

Costello syndrome (CS) is a rare congenital disorder caused by germline activation of H-Ras oncogenes. A mouse model of CS generated by introduction of an oncogenic Gly12Val mutation in the mouse H-Ras locus using homologous recombination in embryonic stem (ES) cells has been recently described. These mice phenocopied some of the abnormalities observed in patients with CS, including facial dysmorphia and cardiomyopathies. We investigated here their neurological and behavioral phenotype. The analysis of H-Ras(G12V) mice revealed phenotypes that resembled the hyperemotivity, hypersensibility and cognitive impairments observed in children with CS. Stronger neurological deficits were found in the analysis of mice homozygous for this mutation than in the analysis of heterozygous mice, suggesting the existence of a gene dose effect. These mice represent the first mouse model for CS, offering an experimental tool to study the molecular and physiological alterations underlying the neurological manifestations of CS and to test new therapies aimed at preventing or ameliorating the cognitive and emotional impairments associated to this condition.


Subject(s)
Genes, ras/genetics , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Oncogenes/genetics , Animals , Anxiety/genetics , Anxiety/psychology , Brain/pathology , Cognition Disorders/genetics , Cognition Disorders/psychology , Fear/psychology , Gene Dosage , Germ-Line Mutation , Humans , Maze Learning/physiology , Mice , Mice, Mutant Strains , Motor Activity/genetics , Motor Activity/physiology , Phenotype , Point Mutation/genetics , Point Mutation/physiology , Postural Balance/physiology , Reaction Time/physiology , Reflex, Startle/physiology , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...