Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neurooncol ; 163(3): 635-645, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37354357

ABSTRACT

PURPOSE: Medulloblastomas (MBs) constitute the most common malignant brain tumor in children and adolescents. MYC-amplified Group 3 MBs are characterized by disease recurrence, specifically in the leptomeninges, whereby patients with these metastatic tumors have a mortality rate nearing 100%. Despite limited research on such tumors, studies on MB metastases at diagnosis suggest targeting kinases to be beneficial. METHODS: To identify kinase inhibitors that eradicate cells driving therapy evasion and tumor dissemination, we utilized our established patient-derived xenograft (PDX) mouse-adapted therapy platform that models human MB metastatic recurrences following standard chemoradiotherapy. High-throughput screens of 640 kinase inhibitors were conducted against cells isolated from mouse spines in the PDX model and human fetal neural stem cells to reveal compounds that targeted these treatment-refractory, metastatic cells, whilst sparing healthy cells. Blood-brain barrier permeability assays and additional in vitro experimentation helped select top candidates for in vivo studies. RESULTS: Recurrent Group 3 MB PDX spine cells were therapeutically vulnerable to a selective checkpoint kinase 1 (CHK1) inhibitor and small molecular inhibitor of platelet-derived growth factor receptor beta (PDGFRß). Inhibitor-treated cells showed a significant reduction in MB stem cell properties associated with treatment failure. Mice also demonstrated survival advantage when treated with a CHK1 inhibitor ex vivo. CONCLUSION: We identified CHK1 and PDGFRß inhibitors that effectively target MB cells fueling treatment-refractory metastases. With limited research on effective therapies for Group 3 MB metastatic recurrences, this work highlights promising therapeutic options to treat these aggressive tumors. Additional studies are warranted to investigate these inhibitors' mechanisms and recommended in vivo administration.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Humans , Child , Mice , Animals , Adolescent , Medulloblastoma/pathology , Xenograft Model Antitumor Assays , Neoplasm Recurrence, Local/drug therapy , Brain Neoplasms/drug therapy , Disease Models, Animal , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cerebellar Neoplasms/pathology , Cell Line, Tumor
2.
Proc Natl Acad Sci U S A ; 120(8): e2205247120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36780531

ABSTRACT

Brain metastases (BM) are the most common brain neoplasm in adults. Current BM therapies still offer limited efficacy and reduced survival outcomes, emphasizing the need for a better understanding of the disease. Herein, we analyzed the transcriptional profile of brain metastasis initiating cells (BMICs) at two distinct stages of the brain metastatic cascade-the "premetastatic" or early stage when they first colonize the brain and the established macrometastatic stage. RNA sequencing was used to obtain the transcriptional profiles of premetastatic and macrometastatic (non-premetastatic) lung, breast, and melanoma BMICs. We identified that lung, breast, and melanoma premetastatic BMICs share a common transcriptomic signature that is distinct from their non-premetastatic counterparts. Importantly, we show that premetastatic BMICs exhibit increased expression of HLA-G, which we further demonstrate functions in an HLA-G/SPAG9/STAT3 axis to promote the establishment of brain metastatic lesions. Our findings suggest that unraveling the molecular landscape of premetastatic BMICs allows for the identification of clinically relevant targets that can possibly inform the development of preventive and/or more efficacious BM therapies.


Subject(s)
Brain Neoplasms , Breast Neoplasms , HLA-G Antigens , Lung Neoplasms , Melanoma , Adult , Humans , Adaptor Proteins, Signal Transducing , Brain/pathology , Brain Neoplasms/secondary , HLA-G Antigens/genetics , Lung/pathology , Lung Neoplasms/pathology , Melanoma/pathology , STAT3 Transcription Factor/genetics , Breast Neoplasms/pathology
4.
Nat Commun ; 13(1): 7506, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36473869

ABSTRACT

Pediatric medulloblastoma (MB) is the most common solid malignant brain neoplasm, with Group 3 (G3) MB representing the most aggressive subgroup. MYC amplification is an independent poor prognostic factor in G3 MB, however, therapeutic targeting of the MYC pathway remains limited and alternative therapies for G3 MB are urgently needed. Here we show that the RNA-binding protein, Musashi-1 (MSI1) is an essential mediator of G3 MB in both MYC-overexpressing mouse models and patient-derived xenografts. MSI1 inhibition abrogates tumor initiation and significantly prolongs survival in both models. We identify binding targets of MSI1 in normal neural and G3 MB stem cells and then cross referenced these data with unbiased large-scale screens at the transcriptomic, translatomic and proteomic levels to systematically dissect its functional role. Comparative integrative multi-omic analyses of these large datasets reveal cancer-selective MSI1-bound targets sharing multiple MYC associated pathways, providing a valuable resource for context-specific therapeutic targeting of G3 MB.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Animals , Mice , Humans , Proteomics , Medulloblastoma/genetics , RNA-Binding Proteins/genetics , Cerebellar Neoplasms/genetics , Nerve Tissue Proteins
5.
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
6.
Front Immunol ; 13: 905768, 2022.
Article in English | MEDLINE | ID: mdl-35874663

ABSTRACT

Glioblastomas (GBM), the most common malignant primary adult brain tumors, are uniformly lethal and are in need of improved therapeutic modalities. GBM contain extensive regions of hypoxia and are enriched in therapy resistant brain tumor-initiating cells (BTICs). Carbonic anhydrase 9 (CA9) is a hypoxia-induced cell surface enzyme that plays an important role in maintenance of stem cell survival and therapeutic resistance. Here we demonstrate that CA9 is highly expressed in patient-derived BTICs. CA9+ GBM BTICs showed increased self-renewal and proliferative capacity. To target CA9, we developed dual antigen T cell engagers (DATEs) that were exquisitely specific for CA9-positive patient-derived clear cell Renal Cell Carcinoma (ccRCC) and GBM cells. Combined treatment of either ccRCC or GBM cells with the CA9 DATE and T cells resulted in T cell activation, increased release of pro-inflammatory cytokines and enhanced cytotoxicity in a CA9-dependent manner. Treatment of ccRCC and GBM patient-derived xenografts markedly reduced tumor burden and extended survival. These data suggest that the CA9 DATE could provide a novel therapeutic strategy for patients with solid tumors expressing CA9 to overcome treatment resistance. .


Subject(s)
Brain Neoplasms , Carbonic Anhydrases , Carcinoma, Renal Cell , Glioblastoma , Kidney Neoplasms , Adult , Antigens, Neoplasm/therapeutic use , Brain Neoplasms/metabolism , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrases/metabolism , Carbonic Anhydrases/therapeutic use , Carcinoma, Renal Cell/therapy , Glioblastoma/therapy , Humans , Hypoxia , Immunotherapy , Kidney Neoplasms/therapy , T-Lymphocytes/metabolism
7.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35017149

ABSTRACT

PURPOSE: Glioblastoma (GBM) patients suffer from a dismal prognosis, with standard of care therapy inevitably leading to therapy-resistant recurrent tumors. The presence of cancer stem cells (CSCs) drives the extensive heterogeneity seen in GBM, prompting the need for novel therapies specifically targeting this subset of tumor-driving cells. Here, we identify CD70 as a potential therapeutic target for recurrent GBM CSCs. EXPERIMENTAL DESIGN: In the current study, we identified the relevance and functional influence of CD70 on primary and recurrent GBM cells, and further define its function using established stem cell assays. We use CD70 knockdown studies, subsequent RNAseq pathway analysis, and in vivo xenotransplantation to validate CD70's role in GBM. Next, we developed and tested an anti-CD70 chimeric antigen receptor (CAR)-T therapy, which we validated in vitro and in vivo using our established preclinical model of human GBM. Lastly, we explored the importance of CD70 in the tumor immune microenvironment (TIME) by assessing the presence of its receptor, CD27, in immune infiltrates derived from freshly resected GBM tumor samples. RESULTS: CD70 expression is elevated in recurrent GBM and CD70 knockdown reduces tumorigenicity in vitro and in vivo. CD70 CAR-T therapy significantly improves prognosis in vivo. We also found CD27 to be present on the cell surface of multiple relevant GBM TIME cell populations, notably putative M1 macrophages and CD4 T cells. CONCLUSION: CD70 plays a key role in recurrent GBM cell aggressiveness and maintenance. Immunotherapeutic targeting of CD70 significantly improves survival in animal models and the CD70/CD27 axis may be a viable polytherapeutic avenue to co-target both GBM and its TIME.


Subject(s)
Brain Neoplasms/therapy , CD27 Ligand/metabolism , Glioblastoma/therapy , Immunotherapy/methods , Proteomics/methods , Transcriptome/genetics , Tumor Microenvironment/immunology , Animals , Brain Neoplasms/immunology , Cell Proliferation , Glioblastoma/immunology , Humans , Male , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local , Prognosis
8.
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.

9.
STAR Protoc ; 2(2): 100436, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33912845

ABSTRACT

Recent data suggest that cells respond to infection by upregulating the antiviral cytokine interferon-beta (IFN-ß) in a fraction of infected cells. Approaches are thus needed to study these responses on a single-cell level rather than bulk population. Here, we describe a protocol to analyze the IFN-ß response of individual cells using flow cytometry and immunofluorescence microscopy. We show the heterogeneous IFN-ß response to inactivated Sendai virus and human cytomegalovirus, but this protocol can be adapted to other viruses. For complete details on the use and execution of this protocol, please refer to Hare et al. (2020).


Subject(s)
Fluorescent Dyes/metabolism , Interferon-beta , Single Molecule Imaging/methods , Single-Cell Analysis/methods , Telomerase/metabolism , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Dyes/analysis , Humans , Interferon-beta/analysis , Interferon-beta/metabolism , Telomerase/genetics
10.
Nat Commun ; 11(1): 4323, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859895

ABSTRACT

Medulloblastoma (MB) is defined by four molecular subgroups (Wnt, Shh, Group 3, Group 4) with Wnt MB having the most favorable prognosis. Since prior reports have illustrated the antitumorigenic role of Wnt activation in Shh MB, we aimed to assess the effects of activated canonical Wnt signaling in Group 3 and 4 MBs. By using primary patient-derived MB brain tumor-initiating cell (BTIC) lines, we characterize differences in the tumor-initiating capacity of Wnt, Group 3, and Group 4 MB. With single cell RNA-seq technology, we demonstrate the presence of rare Wnt-active cells in non-Wnt MBs, which functionally retain the impaired tumorigenic potential of Wnt MB. In treating MB xenografts with a Wnt agonist, we provide a rational therapeutic option in which the protective effects of Wnt-driven MBs may be augmented in Group 3 and 4 MB and thereby support emerging data for a context-dependent tumor suppressive role for Wnt/ß-catenin signaling.


Subject(s)
Cerebellar Neoplasms/therapy , Medulloblastoma/therapy , Wnt Proteins/pharmacology , Wnt Proteins/therapeutic use , Animals , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/pathology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Stem Cells , Wnt Proteins/genetics , Wnt Signaling Pathway , beta Catenin/therapeutic use
11.
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
12.
Oncogene ; 39(7): 1590-1599, 2020 02.
Article in English | MEDLINE | ID: mdl-31695152

ABSTRACT

Mechanistic insight into signaling pathways downstream of surface receptors has been revolutionized with integrated cancer genomics. This has fostered current treatment modalities, namely immunotherapy, to capitalize on targeting key oncogenic signaling nodes downstream of a limited number of surface markers. Unfortunately, rudimentary mechanistic understanding of most other cell surface proteins has reduced the clinical utility of these markers. CD133 has reproducibly been shown to correlate with disease progression, recurrence, and poor overall survivorship in the malignant adult brain tumor, glioblastoma (GBM). Using several patient-derived CD133high and CD133low GBMs we describe intrinsic differences in determinants of stemness, which we owe to a CD133-AKT-Wnt signaling axis in which CD133 functions as a putative cell surface receptor for AKT-dependent Wnt activation. These findings may have implications for personalized oncology trials targeting PI3K/AKT or Wnt as both pathways may be activated independent of their canonical drivers, leading to treatment resistance and disease relapse.


Subject(s)
AC133 Antigen/metabolism , Carcinogenesis , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway , Cell Line, Tumor , Glioblastoma/metabolism , Humans
13.
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
14.
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
15.
Methods Mol Biol ; 1869: 69-77, 2019.
Article in English | MEDLINE | ID: mdl-30324514

ABSTRACT

As a useful biotechnology, flow cytometry has revolutionized the field of cell analysis through its dynamic system that employs fluidics, optics, and electronics. It was first used to analyze DNA, but is often used to determine biomarker expression, as well as to characterize and sort cells, in accordance with various parameters. A common application of flow cytometry is the identification and isolation of a distinct cancer cell population, known as cancer stem cells (CSCs). Various biomarkers have been used to elucidate this proportion of cells within the brain, termed brain tumor initiating cells (BTICs). Here, we discuss methodology to prepare BTICs for flow cytometric analysis that includes the expression of markers.


Subject(s)
Brain Neoplasms/pathology , Flow Cytometry/methods , Neoplastic Stem Cells/pathology , Cell Adhesion , Cell Line, Tumor , Cell Survival , Fluorescent Dyes/metabolism , Humans , Staining and Labeling
16.
Methods Mol Biol ; 1869: 85-91, 2019.
Article in English | MEDLINE | ID: mdl-30324516

ABSTRACT

Differentiation is a central key capability of stem cells. Their ability to be multipotent and undergo self-renewal are key identifying features of stem cells. A differentiation assay allows for study of one of the essential features of stem cells, the ability to differentiate into all of the cell types of its lineage, in order to ensure that the cells cultured and utilized in key experiments indeed have stem cell properties. Neural stem cells when plated in differentiation media, differentiate into all three neural lineages: Neurons, Astrocytes, and Oligodendrocytes. Brain tumor initiating cells (BTICs) are cells present in brain tumors that possess stem cell properties and are able to self-renew and differentiate into neural lineages. In the current chapter, we discuss protocols involved in immunofluorescence staining and identification of differentiated cells from BTIC populations.


Subject(s)
Brain Neoplasms/pathology , Cell Culture Techniques/methods , Cell Differentiation , Neoplastic Stem Cells/pathology , Cell Membrane Permeability , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , Humans , Neural Stem Cells/metabolism
17.
Clin Cancer Res ; 25(6): 2001-2017, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30593514

ABSTRACT

PURPOSE: Stem-like cancer cells, with characteristic self-renewal abilities, remain highly refractory to various clinical interventions. As such, stemness-inhibiting entities, such as tumor suppressor p53, are therapeutically pursued for their anticancer activities. Interestingly, similar implications for tumor suppressor TAp73 in regulating stemness features within stem-like cancer cells remain unknown.Experimental Design: This study utilizes various in vitro molecular biology techniques, including immunoblotting, qRT-PCR, and mass spectrometry-based proteomics, and metabolomics approaches to study the role of TAp73 in human and murine embryonal carcinoma stem-like cells (ECSLC) as well as human breast cancer stem-like cells (BCSLC). These findings were confirmed using patient-derived brain tumor-initiating cells (BTIC) and in vivo xenograft models. RESULTS: TAp73 inhibition decreases the expression of stem cell transcription factors Oct4, Nanog, and Sox-2, as well as tumorsphere formation capacity in ECSLCs. In vivo, TAp73-deficient ECSLCs and BCSLCs demonstrate decreased tumorigenic potential when xenografted in mice. Mechanistically, TAp73 modifies the proline regulatory axis through regulation of enzymes GLS, OAT, and PYCR1 involved in the interconversion of proline-glutamine-ornithine. Further, TAp73 deficiency exacerbates glutamine dependency, enhances accumulation of reactive oxygen species through reduced superoxide dismutase 1 (SOD1) expression, and promotes differentiation by arresting cell cycle and elevating autophagy. Most importantly, the knockdown of TAp73 in CD133HI BTICs, separated from three different glioblastoma patients, strongly decreases the expression of prosurvival factors Sox-2, BMI-1, and SOD1, and profoundly decreases their self-renewal capacity as evidenced through their reduced tumorsphere formation ability. CONCLUSIONS: Collectively, we reveal a clinically relevant aspect of cancer cell growth and stemness regulation through TAp73-mediated redox-sensitive metabolic reprogramming.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Tumor Protein p73/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Self Renewal/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Oxidation-Reduction , RNA, Small Interfering/metabolism , Tumor Protein p73/genetics , Xenograft Model Antitumor Assays
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...