Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Journal subject
Affiliation country
Publication year range
1.
Stem Cells ; 31(7): 1266-77, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592496

ABSTRACT

Brain tumors represent the leading cause of childhood cancer mortality, of which medulloblastoma (MB) is the most frequent malignant tumor. Recent studies have demonstrated the presence of several MB molecular subgroups, each distinct in terms of prognosis and predicted therapeutic response. Groups 1 and 2 are characterized by relatively good clinical outcomes and activation of the Wnt and Shh pathways, respectively. In contrast, groups 3 and 4 ("non-Shh/Wnt MBs") are distinguished by metastatic disease, poor patient outcome, and lack a molecular pathway phenotype. Current gene expression platforms have not detected brain tumor-initiating cell (BTIC) self-renewal genes in groups 3 and 4 MBs as BTICs typically comprise a minority of tumor cells and may therefore go undetected on bulk tumor analyses. Since increasing BTIC frequency has been associated with increasing tumor aggressiveness and poor patient outcome, we investigated the subgroup-specific gene expression profile of candidate stem cell genes within 251 primary human MBs from four nonoverlapping MB transcriptional databases (Amsterdam, Memphis, Toronto, Boston) and 74 NanoString-subgrouped MBs (Vancouver). We assessed the functional relevance of two genes, FoxG1 and Bmi1, which were significantly enriched in non-Shh/Wnt MBs and showed these genes to mediate MB stem cell self-renewal and tumor initiation in mice. We also identified their transcriptional regulation through reciprocal promoter occupancy in CD15+ MB stem cells. Our work demonstrates the application of stem cell data gathered from genomic platforms to guide functional BTIC assays, which may then be used to develop novel BTIC self-renewal mechanisms amenable to therapeutic targeting.


Subject(s)
Cerebellar Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Medulloblastoma/metabolism , Neoplastic Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Forkhead Transcription Factors/genetics , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Polycomb Repressive Complex 1/genetics , Prognosis , Promoter Regions, Genetic , Signal Transduction , Transcriptome
2.
Clin Cancer Res ; 21(23): 5324-37, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26152745

ABSTRACT

PURPOSE: Clonal evolution of cancer may be regulated by determinants of stemness, specifically self-renewal, and current therapies have not considered how genetic perturbations or properties of stemness affect such functional processes. Glioblastoma-initiating cells (GICs), identified by expression of the cell surface marker CD133, are shown to be chemoradioresistant. In the current study, we sought to elucidate the functional role of CD133 in self-renewal and identify compounds that can specifically target this CD133(+) treatment-refractory population. EXPERIMENTAL DESIGN: Using gain/loss-of-function studies for CD133 we assessed the in vitro self-renewal and in vivo tumor formation capabilities of patient-derived glioblastoma cells. We generated a CD133 signature combined with an in silico screen to find compounds that target GICs. Self-renewal and proliferation assays on CD133-sorted samples were performed to identify the preferential action of hit compounds. In vivo efficacy of the lead compound pyrvinium was assessed in intracranial GIC xenografts and survival studies. Lastly, microarray analysis was performed on pyrvinium-treated GICs to discover core signaling events involved. RESULTS: We discovered pyrvinium, a small-molecule inhibitor of GIC self-renewal in vitro and in vivo, in part through inhibition of Wnt/ß-catenin signaling and other essential stem cell regulatory pathways. We provide a therapeutically tractable strategy to target self-renewing, chemoradioresistant, and functionally important CD133(+) stem cells that drive glioblastoma relapse and mortality. CONCLUSIONS: Our study provides an integrated approach for the eradication of clonal populations responsible for cancer progression, and may apply to other aggressive and heterogeneous cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glycoproteins/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Peptides/antagonists & inhibitors , Pyrvinium Compounds/pharmacology , AC133 Antigen , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Proliferation , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Regulatory Networks , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/mortality , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Peptides/genetics , Peptides/metabolism , Prognosis , Signal Transduction/drug effects , Spheroids, Cellular , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Natl Cancer Inst ; 105(8): 551-62, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23418195

ABSTRACT

BACKGROUND: Brain metastases are most common in adults with lung cancer, predicting uniformly poor patient outcome, with a median survival of only months. Despite their frequency and severity, very little is known about tumorigenesis in brain metastases. METHODS: We applied previously developed primary solid tumor-initiating cell models to the study of brain metastases from the lung to evaluate the presence of a cancer stem cell population. Patient-derived brain metastases (n = 20) and the NCI-H1915 cell line were cultured as stem-enriching tumorspheres. We used in vitro limiting-dilution and sphere-forming assays, as well as intracranial human-mouse xenograft models. To determine genes overexpressed in brain metastasis tumorspheres, we performed comparative transcriptome analysis. All statistical analyses were two-sided. RESULTS: Patient-derived brain metastasis tumorspheres had a mean sphere-forming capacity of 33 spheres/2000 cells (SD = 33.40) and median stem-cell frequency of 1/60 (range = 0-1/141), comparable to that of primary brain tumorspheres (P = .53 and P = .20, respectively). Brain metastases also expressed CD15 and CD133, markers suggestive of a stemlike population. Through intracranial xenotransplantation, brain metastasis tumorspheres were found to recapitulate the original patient tumor heterogeneity. We also identified several genes overexpressed in brain metastasis tumorspheres as statistically significant predictors of poor survival in primary lung cancer. CONCLUSIONS: For the first time, we demonstrate the presence of a stemlike population in brain metastases from the lung. We also show that NCI-H1915 tumorspheres could be useful in studying self-renewal and tumor initiation in brain metastases. Our candidate genes may be essential to metastatic stem cell populations, where pathway interference may be able to transform a uniformly fatal disease into a more localized and treatable one.


Subject(s)
Brain Neoplasms/mortality , Brain Neoplasms/secondary , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplastic Stem Cells , Transcriptome , Adult , Animals , Cell Line, Tumor , Female , Flow Cytometry , Humans , Male , Mice , RNA, Neoplasm/analysis , Sequence Analysis, RNA , Survival Analysis , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL