Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nature ; 572(7767): 67-73, 2019 08.
Article in English | MEDLINE | ID: mdl-31043743

ABSTRACT

Study of the origin and development of cerebellar tumours has been hampered by the complexity and heterogeneity of cerebellar cells that change over the course of development. Here we use single-cell transcriptomics to study more than 60,000 cells from the developing mouse cerebellum and show that different molecular subgroups of childhood cerebellar tumours mirror the transcription of cells from distinct, temporally restricted cerebellar lineages. The Sonic Hedgehog medulloblastoma subgroup transcriptionally mirrors the granule cell hierarchy as expected, while group 3 medulloblastoma resembles Nestin+ stem cells, group 4 medulloblastoma resembles unipolar brush cells, and PFA/PFB ependymoma and cerebellar pilocytic astrocytoma resemble the prenatal gliogenic progenitor cells. Furthermore, single-cell transcriptomics of human childhood cerebellar tumours demonstrates that many bulk tumours contain a mixed population of cells with divergent differentiation. Our data highlight cerebellar tumours as a disorder of early brain development and provide a proximate explanation for the peak incidence of cerebellar tumours in early childhood.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Evolution, Molecular , Fetus/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Transcription, Genetic , Animals , Cerebellar Neoplasms/classification , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/metabolism , Child , Female , Fetus/cytology , Glioma/classification , Glioma/genetics , Glioma/pathology , Humans , Medulloblastoma/classification , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Sequence Analysis, RNA , Single-Cell Analysis , Time Factors , Transcriptome/genetics
2.
J Neurooncol ; 143(3): 417-428, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115870

ABSTRACT

PURPOSE: Glioblastoma (GBM) is the most aggressive adult brain cancer, with a 15 month median survivorship attributed to the existence of treatment-refractory brain tumor initiating cells (BTICs). In order to better understand the mechanisms regulating the tumorigenic properties of this population, we studied the role of the polycomb group member BMI1 in our patient-derived GBM BTICs and its relationship with CD133, a well-established marker of BTICs. METHODS: Using gain and loss-of-function studies for Bmi1 in neural stem cells (NSCs) and patient-derived GBM BTICs respectively, we assessed in vitro self-renewal and in vivo tumor formation in these two cell populations. We further explored the BMI1 transcriptional regulatory network through RNA sequencing of different GBM BTIC populations that were knocked down for Bmi1. RESULTS: There is a differential role of BMI1 in CD133-positive cells, notably involving cell metabolism. In addition, we identified pivotal targets downstream of BMI1 in CD133+ cells such as integrin alpha 2 (ITGA2), that may contribute to regulating GBM stem cell properties. CONCLUSIONS: Our work sheds light on the association of three genes with CD133-BMI1 circuitry, their importance as downstream effectors of the BMI1 signalling pathway, and their potential as future targets for tackling GBM treatment-resistant cell populations.


Subject(s)
AC133 Antigen/metabolism , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/metabolism , AC133 Antigen/genetics , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Acta Neuropathol ; 134(6): 923-940, 2017 12.
Article in English | MEDLINE | ID: mdl-28766011

ABSTRACT

Brain metastases (BM) are the most common brain tumor in adults and are a leading cause of cancer mortality. Metastatic lesions contain subclones derived from their primary lesion, yet their functional characterization is limited by a paucity of preclinical models accurately recapitulating the metastatic cascade, emphasizing the need for a novel approach to BM and their treatment. We identified a unique subset of stem-like cells from primary human patient brain metastases, termed brain metastasis-initiating cells (BMICs). We now establish a BMIC patient-derived xenotransplantation (PDXT) model as an investigative tool to comprehensively interrogate human BM. Using both in vitro and in vivo RNA interference screens of these BMIC models, we identified SPOCK1 and TWIST2 as essential BMIC regulators. SPOCK1 in particular is a novel regulator of BMIC self-renewal, modulating tumor initiation and metastasis from the lung to the brain. A prospective cohort of primary lung cancer specimens showed that SPOCK1 was overexpressed only in patients who ultimately developed BM. Protein-protein interaction network mapping between SPOCK1 and TWIST2 identified novel pathway interactors with significant prognostic value in lung cancer patients. Of these genes, INHBA, a TGF-ß ligand found mutated in lung adenocarcinoma, showed reduced expression in BMICs with knockdown of SPOCK1. In conclusion, we have developed a useful preclinical model of BM, which has served to identify novel putative BMIC regulators, presenting potential therapeutic targets that block the metastatic process, and transform a uniformly fatal systemic disease into a locally controlled and eminently more treatable one.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/physiopathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Female , Humans , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Prospective Studies , Proteoglycans/genetics , Proteoglycans/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
4.
J Neurooncol ; 126(1): 57-67, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26498281

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults with average disease relapse at 9 months and median survival rarely extending beyond 15 months. Brain tumor stem cells (BTSCs) have been implicated in not only initiating GBM but also conferring resistance to therapy. However, it is not clear whether the BTSC population that initiates tumor growth is also responsible for GBM recurrence. In this study, we have developed a novel in vitro treatment model to profile the evolution of primary treatment-naïve GBM BTSCs through chemoradiotherapy. We report that our in vitro model enriched for a CD15+/CD133- BTSC population, mirroring the phenotype of BTSCs in recurrent GBM. We also show that in vitro treatment increased stem cell gene expression as well as self-renewal capacity of primary GBMs. In addition, the chemoradiotherapy-refractory gene signature obtained from gene expression profiling identified a hyper-aggressive subtype of glioma. The delivery of in vitro chemoradiotherapy to primary GBM BTSCs models several aspects of recurrent GBM biology, and could be used as a discovery and drug-screening platform to uncover new biological drivers and therapeutic targets in GBM.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Aged , Aged, 80 and over , Analysis of Variance , Antigens, CD/metabolism , Antinematodal Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Self Renewal/physiology , Dose-Response Relationship, Drug , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tumor Cells, Cultured
5.
Cancers (Basel) ; 15(10)2023 May 13.
Article in English | MEDLINE | ID: mdl-37345085

ABSTRACT

We aimed to evaluate the expression of the "targetable" androgen receptor (AR) in breast cancer brain metastases (BrM). An established, retrospective 57-patient cohort with metastatic breast cancer who underwent surgery for BrM at the Sunnybrook Odette Cancer Centre between 1999-2013 was studied. AR expression in BrM samples was assessed in triplicate using immunohistochemistry (IHC). AR positive status was defined as nuclear AR expression ≥ 10% by IHC using the SP107 antibody. The median age of patients was 52 years (range 32-85 years). 28 (49%) of BrM were HER2+, 17 (30%) were hormone receptor positive (HR+)/HER2-, and 12 (21%) were triple negative breast cancers (TNBCs). 56% (n = 32/57) of BrM were AR positive, and median AR expression was 20% (CI 1.6-38.3%). AR expression was different across breast cancer subtypes; AR was most frequently expressed in HER2+ (n = 21/28), followed by HR+/HER2- (n = 9/17), and lowest in TNBC (n = 2/12) BrM (p = 0.003). Patients with AR positive versus AR negative BrM had similar overall survival (12.5 vs. 7.9 months, p = 0.6), brain-specific progression-free survival (8.0 vs. 5.1 months, p = 0.95), and time from breast cancer diagnosis to BrM diagnosis (51 vs. 29 months, p = 0.16). AR is expressed in the majority of breast cancer BrM and represents a potential therapeutic target.

6.
Neurooncol Adv ; 4(1): vdac154, 2022.
Article in English | MEDLINE | ID: mdl-36299795

ABSTRACT

Background: To evaluate the potential intracranial efficacy of immunotherapy among patients with breast cancer brain metastases (BrM), we analyzed the immunohistochemical expression of programmed death-ligand 1 (PD-L1), a predictive biomarker of response to immunotherapy. Methods: In this single-center retrospective cohort study, consecutive patients with breast cancer BrM (immunotherapy naïve) who underwent surgery for BrM at Sunnybrook Health Sciences Center between July 1999 and June 2013 were identified. PD-L1 expression by immunohistochemistry (IHC) was assessed on BrM samples in triplicate; PD-L1 positive status was defined as PD-L1 expression ≥1% on tumor-infiltrating cells as a percentage of tumor area using the Ventana SP142 antibody. Estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER2) status was determined using 2018 ASCO/CAP guidelines. Results: The median patient age at the time of BrM diagnosis was 52 (range 32-85). PD-L1 expression using the SP42 antibody was identified in 9 out of 59 (15.3%) breast cancer BrM. The frequency of PD-L1 positive BrM by subtype is as follows: TNBC (n = 3/12, 25.0%), HER2+/HR- (n = 3/14, 21.4%), HR+/HER2- (n = 2/18, 11.1%), and HER2+/HR+ (n = 1/14, 7.1%). 24-month brain-specific progression-free survival was 66.7% (95% CI 37.9%-100%) among patients with PD-L1 positive BrM versus 42% (95% CI 26.6%-67.3%) among those with PD-L1 negative BrM (log-rank P-value .142). Conclusions: One in 7 patients in our cohort had PD-L1 positive BrM; this proportion was highest (25%) among those with TNBC. Intracranial efficacy of immunotherapy warrants further study, particularly among patients with treatment-naïve metastatic TNBC, for whom extracranial efficacy has already been established.

7.
Cell Rep ; 40(13): 111420, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36170831

ABSTRACT

Recurrence of solid tumors renders patients vulnerable to advanced, treatment-refractory disease state with mutational and oncogenic landscape distinctive from initial diagnosis. Improving outcomes for recurrent cancers requires a better understanding of cell populations that expand from the post-therapy, minimal residual disease (MRD) state. We profile barcoded tumor stem cell populations through therapy at tumor initiation, MRD, and recurrence in our therapy-adapted, patient-derived xenograft models of glioblastoma (GBM). Tumors show distinct patterns of recurrence in which clonal populations exhibit either a pre-existing fitness advantage or an equipotency fitness acquired through therapy. Characterization of the MRD state by single-cell and bulk RNA sequencing reveals a tumor-intrinsic immunomodulatory signature with prognostic significance at the transcriptomic level and in proteomic analysis of cerebrospinal fluid (CSF) collected from patients with GBM. Our results provide insight into the innate and therapy-driven dynamics of human GBM and the prognostic value of interrogating the MRD state in solid cancers.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm, Residual/genetics , Neoplastic Stem Cells/pathology , Proteomics
8.
Neurooncol Adv ; 3(1): vdab144, 2021.
Article in English | MEDLINE | ID: mdl-34765972

ABSTRACT

BACKGROUND: Glioblastoma (GBM), the most common and aggressive primary brain tumour in adults, has been classified into three subtypes: classical, mesenchymal, and proneural. While the original classification relied on an 840 gene-set, further clarification on true GBM subtypes uses a 150-gene signature to accurately classify GBM into the three subtypes. We hypothesized whether a machine learning approach could be used to identify a smaller gene-set to accurately predict GBM subtype. METHODS: Using a supervised machine learning approach, extreme gradient boosting (XGBoost), we developed a classifier to predict the three subtypes of glioblastoma (GBM): classical, mesenchymal, and proneural. We tested the classifier on in-house GBM tissue, cell lines, and xenograft samples to predict their subtype. RESULTS: We identified the five most important genes for characterizing the three subtypes based on genes that often exhibited high Importance Scores in our XGBoost analyses. On average, this approach achieved 80.12% accuracy in predicting these three subtypes of GBM. Furthermore, we applied our five-gene classifier to successfully predict the subtype of GBM samples at our centre. CONCLUSION: Our 5-gene set classifier is the smallest classifier to date that can predict GBM subtypes with high accuracy, which could facilitate the future development of a five-gene subtype diagnostic biomarker for routine assays in GBM samples.

9.
Sci Adv ; 7(50): eabi5568, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34878832

ABSTRACT

Medulloblastoma (MB) remains a leading cause of cancer-related mortality among children. The paucity of MB samples collected at relapse has hindered the functional understanding of molecular mechanisms driving therapy failure. New models capable of accurately recapitulating tumor progression in response to conventional therapeutic interventions are urgently needed. In this study, we developed a therapy-adapted PDX MB model that has a distinct advantage of generating human MB recurrence. The comparative gene expression analysis of MB cells collected throughout therapy led to identification of genes specifically up-regulated after therapy, including one previously undescribed in the setting of brain tumors, bactericidal/permeability-increasing fold-containing family B member 4 (BPIFB4). Subsequent functional validation resulted in a markedly diminished in vitro proliferation, self-renewal, and longevity of MB cells, translating into extended survival and reduced tumor burden in vivo. Targeting endothelial nitric oxide synthase, a downstream substrate of BPIFB4, impeded growth of several patient-derived MB lines at low nanomolar concentrations.

10.
Cell Stem Cell ; 26(6): 832-844.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32464096

ABSTRACT

CD133 marks self-renewing cancer stem cells (CSCs) in a variety of solid tumors, and CD133+ tumor-initiating cells are known markers of chemo- and radio-resistance in multiple aggressive cancers, including glioblastoma (GBM), that may drive intra-tumoral heterogeneity. Here, we report three immunotherapeutic modalities based on a human anti-CD133 antibody fragment that targets a unique epitope present in glycosylated and non-glycosylated CD133 and studied their effects on targeting CD133+ cells in patient-derived models of GBM. We generated an immunoglobulin G (IgG) (RW03-IgG), a dual-antigen T cell engager (DATE), and a CD133-specific chimeric antigen receptor T cell (CAR-T): CART133. All three showed activity against patient-derived CD133+ GBM cells, and CART133 cells demonstrated superior efficacy in patient-derived GBM xenograft models without causing adverse effects on normal CD133+ hematopoietic stem cells in humanized CD34+ mice. Thus, CART133 cells may be a therapeutically tractable strategy to target CD133+ CSCs in human GBM or other treatment-resistant primary cancers.


Subject(s)
Brain Neoplasms , Glioblastoma , AC133 Antigen , Animals , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Immunotherapy , Mice , Neoplastic Stem Cells
11.
Methods Mol Biol ; 1869: 155-168, 2019.
Article in English | MEDLINE | ID: mdl-30324522

ABSTRACT

The ability to elucidate the phenotype of brain tumor initiating cell (BTIC) in the context of bulk tumor in glioblastoma multiforme (GBM) provides significant therapeutic benefits for therapeutic evaluation. For the identification of such an elusive and rare subpopulation of cells, a single cell analysis technology with deep profiling capabilities known as Mass Cytometry (CyTOF) can prove to be highly useful. CyTOF circumvents the spectral overlap limitations of traditional flow cytometry by replacing fluorophores with metal isotope tags, allowing the accurate detection of significantly more parameters at the same time. In this chapter, we demonstrate that synthetic antibodies can be conjugated with metal isotope tags for CyTOF analysis, resulting in the development of a highly tailored, custom multi-parameter panel. This toolset was used to stain patient-derived GBM cells, which was analyzed via CyTOF. Analysis software viSNE and SPADE were applied to study the co-expression patterns of the Eph Receptor (EphR) family and several putative BTIC markers in GBM, resulting in the identification of a distinct group of cells consistent with a BTIC subpopulation. This approach can be readily adapted to the detection of cancer stem-like cells in other cancer types.


Subject(s)
Brain Neoplasms/pathology , Ephrins/metabolism , Flow Cytometry/methods , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Antibodies/metabolism , Cell Survival , Humans , Microspheres , Staining and Labeling , Tumor Cells, Cultured
12.
Oncogene ; 38(10): 1702-1716, 2019 03.
Article in English | MEDLINE | ID: mdl-30348991

ABSTRACT

Medulloblastoma (MB) is the most frequent malignant pediatric brain tumor, representing 20% of newly diagnosed childhood central nervous system malignancies. Although advances in multimodal therapy yielded a 5-year survivorship of 80%, MB still accounts for the leading cause of childhood cancer mortality. In this work, we describe the epigenetic regulator BMI1 as a novel therapeutic target for the treatment of recurrent human Group 3 MB, a childhood brain tumor for which there is virtually no treatment option beyond palliation. Current clinical trials for recurrent MB patients based on genomic profiles of primary, treatment-naive tumors will provide limited clinical benefit since recurrent metastatic MBs are highly genetically divergent from their primary tumor. Using a small molecule inhibitor against BMI1, PTC-028, we were able to demonstrate complete ablation of self-renewal of MB stem cells in vitro. When administered to mice xenografted with patient tumors, we observed significant reduction in tumor burden in both local and metastatic compartments and subsequent increased survival, without neurotoxicity. Strikingly, serial in vivo re-transplantation assays demonstrated a marked reduction in tumor initiation ability of recurrent MB cells upon re-transplantation of PTC-028-treated cells into secondary recipient mouse brains. As Group 3 MB is often metastatic and uniformly fatal at recurrence, with no current or planned trials of targeted therapy, an efficacious targeted agent would be rapidly transitioned to clinical trials.


Subject(s)
Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Polycomb Repressive Complex 1/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Child , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Small Molecule Libraries/pharmacology , Treatment Outcome , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Res ; 78(17): 5124-5134, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29986997

ABSTRACT

Brain metastases (BM) result from the spread of primary tumors to the brain and are a leading cause of cancer mortality in adults. Secondary tissue colonization remains the main bottleneck in metastatic development, yet this "premetastatic" stage of the metastatic cascade, when primary tumor cells cross the blood-brain barrier and seed the brain before initiating a secondary tumor, remains poorly characterized. Current studies rely on specimens from fully developed macrometastases to identify therapeutic options in cancer treatment, overlooking the potentially more treatable "premetastatic" phase when colonizing cancer cells could be targeted before they initiate the secondary brain tumor. Here we use our established brain metastasis initiating cell (BMIC) models and gene expression analyses to characterize premetastasis in human lung-to-BM. Premetastatic BMIC engaged invasive and epithelial developmental mechanisms while simultaneously impeding proliferation and apoptosis. We identified the dopamine agonist apomorphine to be a potential premetastasis-targeting drug. In vivo treatment with apomorphine prevented BM formation, potentially by targeting premetastasis-associated genes KIF16B, SEPW1, and TESK2 Low expression of these genes was associated with poor survival of patients with lung adenocarcinoma. These results illuminate the cellular and molecular dynamics of premetastasis, which is subclinical and currently impossible to identify or interrogate in human patients with BM. These data present several novel therapeutic targets and associated pathways to prevent BM initiation.Significance: These findings unveil molecular features of the premetastatic stage of lung-to-brain metastases and offer a potential therapeutic strategy to prevent brain metastases. Cancer Res; 78(17); 5124-34. ©2018 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Neoplasm Metastasis/drug therapy , Apomorphine/pharmacology , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Dopamine/metabolism , Dopamine Agonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kinesins/genetics , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Protein Serine-Threonine Kinases/genetics , Selenoprotein W/genetics
14.
Methods Mol Biol ; 1692: 1-16, 2018.
Article in English | MEDLINE | ID: mdl-28986882

ABSTRACT

The Cancer Stem Cell (CSC) hypothesis postulates the existence of a small population of cancer cells with intrinsic properties allowing for resistance to conventional radiochemotherapy regiments and increased metastatic potential. Clinically, the aggressive nature of CSCs has been shown to correlate with increased tumor recurrence, metastatic spread, and overall poor patient outcome across multiple cancer subtypes. Traditionally, isolation of CSCs has been achieved through utilization of cell surface markers, while the functional differences between CSCs and remaining tumor cells have been described through proliferation, differentiation, and limiting dilution assays. The generated insights into CSC biology have further highlighted the importance of studying intratumoral heterogeneity through advanced functional assays, including CRISPR-Cas9 screens in the search of novel targeted therapies. In this chapter, we review the discovery and characterization of cancer stem cells populations within several major cancer subtypes, recent developments of novel assays used in studying therapy resistant tumor cells, as well as recent developments in therapies targeted at cancer stem cells.


Subject(s)
Neoplastic Stem Cells/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Humans , Immunotherapy/methods , Leukemia/therapy
15.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29945963

ABSTRACT

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Subject(s)
Ephrin-A2/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Ephrin-A2/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Neoplastic Stem Cells/pathology , Prognosis , Radiation , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, EphA3 , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/genetics , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
16.
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
17.
Oncotarget ; 6(29): 27461-77, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26314961

ABSTRACT

Brain metastases (BM) represent the most common tumor to affect the adult central nervous system. Despite the increasing incidence of BM, likely due to consistently improving treatment of primary cancers, BM remain severely understudied. In this study, we utilized patient-derived stem cell lines from lung-to-brain metastases to examine the regulatory role of STAT3 in brain metastasis initiating cells (BMICs). Annotation of our previously described BMIC regulatory genes with protein-protein interaction network mapping identified STAT3 as a novel protein interactor. STAT3 knockdown showed a reduction in BMIC self-renewal and migration, and decreased tumor size in vivo. Screening of BMIC lines with a library of STAT3 inhibitors identified one inhibitor to significantly reduce tumor formation. Meta-analysis identified the oncomir microRNA-21 (miR-21) as a target of STAT3 activity. Inhibition of miR-21 displayed similar reductions in BMIC self-renewal and migration as STAT3 knockdown. Knockdown of STAT3 also reduced expression of known downstream targets of miR-21. Our studies have thus identified STAT3 and miR-21 as cooperative regulators of stemness, migration and tumor initiation in lung-derived BM. Therefore, STAT3 represents a potential therapeutic target in the treatment of lung-to-brain metastases.


Subject(s)
Brain Neoplasms/secondary , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cell Movement , Genes, Regulator , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Protein Interaction Mapping , Proteomics , RNA, Small Interfering/metabolism , Stem Cells/cytology
19.
Methods Mol Biol ; 1210: 37-49, 2014.
Article in English | MEDLINE | ID: mdl-25173159

ABSTRACT

The generation of xenograft models, which support the growth of human tissue in animals, forms an important part of a researcher's tool kit and enhances the ability to understand the initiation and development of cancer in vivo. Especially in the context of the brain tumor-initiating cell (BTIC), a xenograft model allows for careful characterization of BTIC roles in tumor initiation, growth, and relapse. Here, we detail a set of procedures which describe the isolation, enrichment, and intracranial injection of human BTICs from patient samples to generate xenograft models of a human brain tumor.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Transformation, Neoplastic , Heterografts , Neoplastic Stem Cells/metabolism , Animals , Disease Models, Animal , Flow Cytometry , Humans , Immunohistochemistry , Mice , Primary Cell Culture , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL