Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genes Dev ; 38(5-6): 273-288, 2024 04 17.
Article in English | MEDLINE | ID: mdl-38589034

ABSTRACT

Glioblastoma is universally fatal and characterized by frequent chromosomal copy number alterations harboring oncogenes and tumor suppressors. In this study, we analyzed exome-wide human glioblastoma copy number data and found that cytoband 6q27 is an independent poor prognostic marker in multiple data sets. We then combined CRISPR-Cas9 data, human spatial transcriptomic data, and human and mouse RNA sequencing data to nominate PDE10A as a potential haploinsufficient tumor suppressor in the 6q27 region. Mouse glioblastoma modeling using the RCAS/tv-a system confirmed that Pde10a suppression induced an aggressive glioma phenotype in vivo and resistance to temozolomide and radiation therapy in vitro. Cell culture analysis showed that decreased Pde10a expression led to increased PI3K/AKT signaling in a Pten-independent manner, a response blocked by selective PI3K inhibitors. Single-nucleus RNA sequencing from our mouse gliomas in vivo, in combination with cell culture validation, further showed that Pde10a suppression was associated with a proneural-to-mesenchymal transition that exhibited increased cell adhesion and decreased cell migration. Our results indicate that glioblastoma patients harboring PDE10A loss have worse outcomes and potentially increased sensitivity to PI3K inhibition.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Animals , Mice , Glioblastoma/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Haploinsufficiency , Glioma/genetics , PTEN Phosphohydrolase/genetics , Phosphoric Diester Hydrolases/genetics , Cell Line, Tumor , Brain Neoplasms/genetics
2.
Genes Dev ; 37(3-4): 86-102, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36732025

ABSTRACT

Glioblastomas (GBMs) are heterogeneous, treatment-resistant tumors driven by populations of cancer stem cells (CSCs). However, few molecular mechanisms critical for CSC population maintenance have been exploited for therapeutic development. We developed a spatially resolved loss-of-function screen in GBM patient-derived organoids to identify essential epigenetic regulators in the SOX2-enriched, therapy-resistant niche and identified WDR5 as indispensable for this population. WDR5 is a component of the WRAD complex, which promotes SET1 family-mediated Lys4 methylation of histone H3 (H3K4me), associated with positive regulation of transcription. In GBM CSCs, WDR5 inhibitors blocked WRAD complex assembly and reduced H3K4 trimethylation and expression of genes involved in CSC-relevant oncogenic pathways. H3K4me3 peaks lost with WDR5 inhibitor treatment occurred disproportionally on POU transcription factor motifs, including the POU5F1(OCT4)::SOX2 motif. Use of a SOX2/OCT4 reporter demonstrated that WDR5 inhibitor treatment diminished cells with high reporter activity. Furthermore, WDR5 inhibitor treatment and WDR5 knockdown altered the stem cell state, disrupting CSC in vitro growth and self-renewal, as well as in vivo tumor growth. These findings highlight the role of WDR5 and the WRAD complex in maintaining the CSC state and provide a rationale for therapeutic development of WDR5 inhibitors for GBM and other advanced cancers.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors , Neoplastic Stem Cells/pathology , Intracellular Signaling Peptides and Proteins/genetics
3.
Genes Dev ; 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36008139

ABSTRACT

YAP1 is a transcriptional coactivator regulated by the Hippo signaling pathway, including NF2. Meningiomas are the most common primary brain tumors; a large percentage exhibit heterozygous loss of chromosome 22 (harboring the NF2 gene) and functional inactivation of the remaining NF2 copy, implicating oncogenic YAP activity in these tumors. Recently, fusions between YAP1 and MAML2 have been identified in a subset of pediatric NF2 wild-type meningiomas. Here, we show that human YAP1-MAML2-positive meningiomas resemble NF2 mutant meningiomas by global and YAP-related gene expression signatures. We then show that expression of YAP1-MAML2 in mice induces tumors that resemble human YAP1 fusion-positive and NF2 mutant meningiomas by gene expression. We demonstrate that YAP1-MAML2 primarily functions by exerting TEAD-dependent YAP activity that is resistant to Hippo signaling. Treatment with YAP-TEAD inhibitors is sufficient to inhibit the viability of YAP1-MAML2-driven mouse tumors ex vivo. Finally, we show that expression of constitutively active YAP1 (S127/397A-YAP1) is sufficient to induce similar tumors, suggesting that the YAP component of the gene fusion is the critical driver of these tumors. In summary, our results implicate YAP1-MAML2 as a causal oncogenic driver and highlight TEAD-dependent YAP activity as an oncogenic driver in YAP1-MAML2 fusion meningioma as well as NF2 mutant meningioma in general.

4.
Cell ; 156(3): 603-616, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485463

ABSTRACT

Glioblastomas (GBMs) are the most common and malignant primary brain tumors and are aggressively treated with surgery, chemotherapy, and radiotherapy. Despite this treatment, recurrence is inevitable and survival has improved minimally over the last 50 years. Recent studies have suggested that GBMs exhibit both heterogeneity and instability of differentiation states and varying sensitivities of these states to radiation. Here, we employed an iterative combined theoretical and experimental strategy that takes into account tumor cellular heterogeneity and dynamically acquired radioresistance to predict the effectiveness of different radiation schedules. Using this model, we identified two delivery schedules predicted to significantly improve efficacy by taking advantage of the dynamic instability of radioresistance. These schedules led to superior survival in mice. Our interdisciplinary approach may also be applicable to other human cancer types treated with radiotherapy and, hence, may lay the foundation for significantly increasing the effectiveness of a mainstay of oncologic therapy. PAPERCLIP:


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Radiation Dosage , Animals , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Mice , Models, Biological
5.
Genes Dev ; 34(15-16): 1051-1064, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32675324

ABSTRACT

YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1, YAP1-FAM118B, YAP1-TFE3, and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/genetics , Oncogene Proteins, Fusion/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Nuclear Localization Signals , Nucleotide Motifs , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/chemistry , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
6.
Proc Natl Acad Sci U S A ; 121(7): e2310430121, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38315854

ABSTRACT

Phase separation (PS) drives the formation of biomolecular condensates that are emerging biological structures involved in diverse cellular processes. Recent studies have unveiled PS-induced formation of several transcriptional factor (TF) condensates that are transcriptionally active, but how strongly PS promotes gene activation remains unclear. Here, we show that the oncogenic TF fusion Yes-associated protein 1-Mastermind like transcriptional coactivator 2 (YAP-MAML2) undergoes PS and forms liquid-like condensates that bear the hallmarks of transcriptional activity. Furthermore, we examined the contribution of PS to YAP-MAML2-mediated gene expression by developing a chemogenetic tool that dissolves TF condensates, allowing us to compare phase-separated and non-phase-separated conditions at identical YAP-MAML2 protein levels. We found that a small fraction of YAP-MAML2-regulated genes is further affected by PS, which include the canonical YAP target genes CTGF and CYR61, and other oncogenes. On the other hand, majority of YAP-MAML2-regulated genes are not affected by PS, highlighting that transcription can be activated effectively by diffuse complexes of TFs with the transcriptional machinery. Our work opens new directions in understanding the role of PS in selective modulation of gene expression, suggesting differential roles of PS in biological processes.


Subject(s)
Phase Separation , Transcriptome , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Oncogenes
7.
Genes Dev ; 32(7-8): 512-523, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29632085

ABSTRACT

Glioblastoma is the most frequently occurring and invariably fatal primary brain tumor in adults. The vast majority of glioblastomas is characterized by chromosomal copy number alterations, including gain of whole chromosome 7 and loss of whole chromosome 10. Gain of whole chromosome 7 is an early event in gliomagenesis that occurs in proneural-like precursor cells, which give rise to all isocitrate dehydrogenase (IDH) wild-type glioblastoma transcriptional subtypes. Platelet-derived growth factor A (PDGFA) is one gene on chromosome 7 known to drive gliomagenesis, but, given its location near the end of 7p, there are likely several other genes located along chromosome 7 that select for its increased whole-chromosome copy number within glioblastoma cells. To identify other potential genes that could select for gain of whole chromosome 7, we developed an unbiased bioinformatics approach that identified homeobox A5 (HOXA5) as a gene whose expression correlated with gain of chromosome 7 and a more aggressive phenotype of the resulting glioma. High expression of HOXA5 in glioblastoma was associated with a proneural gene expression pattern and decreased overall survival in both human proneural and PDGF-driven mouse glioblastoma. Furthermore, HOXA5 overexpression promoted cellular proliferation and potentiated radioresistance. We also found enrichment of HOXA5 expression in recurrent human and mouse glioblastoma at first recurrence after radiotherapy. Overall, this study implicates HOXA5 as a chromosome 7-associated gene-level locus that promotes selection for gain of whole chromosome 7 and an aggressive phenotype in glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 7 , Glioblastoma/genetics , Homeodomain Proteins/metabolism , Phosphoproteins/metabolism , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Proliferation , Chromosome Duplication , Glioblastoma/mortality , Glioblastoma/pathology , Glioblastoma/radiotherapy , Homeodomain Proteins/genetics , Humans , Isocitrate Dehydrogenase/genetics , Mice , Neoplasm Recurrence, Local , Phosphoproteins/genetics , Radiation Tolerance , Transcription Factors
8.
Genes Dev ; 31(8): 774-786, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28465358

ABSTRACT

Gliomas harboring mutations in isocitrate dehydrogenase 1/2 (IDH1/2) have the CpG island methylator phenotype (CIMP) and significantly longer patient survival time than wild-type IDH1/2 (wtIDH1/2) tumors. Although there are many factors underlying the differences in survival between these two tumor types, immune-related differences in cell content are potentially important contributors. In order to investigate the role of IDH mutations in immune response, we created a syngeneic pair mouse model for mutant IDH1 (muIDH1) and wtIDH1 gliomas and demonstrated that muIDH1 mice showed many molecular and clinical similarities to muIDH1 human gliomas, including a 100-fold higher concentration of 2-hydroxygluratate (2-HG), longer survival time, and higher CpG methylation compared with wtIDH1. Also, we showed that IDH1 mutations caused down-regulation of leukocyte chemotaxis, resulting in repression of the tumor-associated immune system. Given that significant infiltration of immune cells such as macrophages, microglia, monocytes, and neutrophils is linked to poor prognosis in many cancer types, these reduced immune infiltrates in muIDH1 glioma tumors may contribute in part to the differences in aggressiveness of the two glioma types.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/immunology , Glioma/genetics , Glioma/immunology , Immune System/physiopathology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Animals , Brain Neoplasms/enzymology , Chemotaxis/genetics , DNA Methylation , Disease Models, Animal , Glioma/enzymology , Humans , Leukocyte Common Antigens/metabolism , Leukocytes/pathology , Mice , Mutation , Neutrophil Infiltration/genetics , Neutrophils/pathology
9.
Dev Biol ; 475: 205-221, 2021 07.
Article in English | MEDLINE | ID: mdl-33428889

ABSTRACT

YAP1 is a transcriptional co-activator whose activity is controlled by the Hippo signaling pathway. In addition to important functions in normal tissue homeostasis and regeneration, YAP1 has also prominent functions in cancer initiation, aggressiveness, metastasis, and therapy resistance. In this review we are discussing the molecular functions of YAP1 and its roles in cancer, with a focus on the different mechanisms of de-regulation of YAP1 activity in human cancers, including inactivation of upstream Hippo pathway tumor suppressors, regulation by intersecting pathways, miRNAs, and viral oncogenes. We are also discussing new findings on the function and biology of the recently identified family of YAP1 gene fusions, that constitute a new type of activating mutation of YAP1 and that are the likely oncogenic drivers in several subtypes of human cancers. Lastly, we also discuss different strategies of therapeutic inhibition of YAP1 functions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
10.
Br J Cancer ; 127(3): 422-435, 2022 08.
Article in English | MEDLINE | ID: mdl-35418212

ABSTRACT

BACKGROUND: Angiogenin is a multifunctional secreted ribonuclease that is upregulated in human cancers and downregulated or mutationally inactivated in neurodegenerative diseases. A role for angiogenin in glioblastoma was inferred from the inverse correlation of angiogenin expression with patient survival but had not been experimentally investigated. METHODS: Angiogenin knockout mice were generated and the effect of angiogenin deficiency on glioblastoma progression was examined. Angiogenin and plexin-B2 genes were knocked down in glioblastoma cells and the changes in cell proliferation, invasion and vascular association were examined. Monoclonal antibodies of angiogenin and small molecules were used to assess the therapeutic activity of the angiogenin-plexin-B2 pathway in both genetic and xenograft animal models. RESULTS: Deletion of Ang1 gene prolonged survival of PDGF-induced glioblastoma in mice in the Ink4a/Arf-/-:Pten-/- background, accompanied by decreased invasion, vascular association and proliferation. Angiogenin upregulated MMP9 and CD24 leading to enhanced invasion and vascular association. Inhibition of angiogenin or plexin-B2, either by shRNA, monoclonal antibody or small molecule inhibitor, decreases sphere formation of patient-derived glioma stem cells, reduces glioblastoma proliferation and invasion and inhibits glioblastoma growth in both genetic and xenograft animal models. CONCLUSIONS: Angiogenin and its receptor, plexin-B2, are a pair of novel regulators that mediate invasion, vascular association and proliferation of glioblastoma cells. Inhibitors of the angiogenin-plexin-B2 axis have therapeutic potential against glioblastoma.


Subject(s)
Glioblastoma , Nerve Tissue Proteins , Ribonuclease, Pancreatic , Animals , Cell Line, Tumor , Cell Proliferation , Glioblastoma/drug therapy , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
11.
Oncologist ; 27(4): 272-284, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35380712

ABSTRACT

Within the last decade, the science of molecular testing has evolved from single gene and single protein analysis to broad molecular profiling as a standard of care, quickly transitioning from research to practice. Terms such as genomics, transcriptomics, proteomics, circulating omics, and artificial intelligence are now commonplace, and this rapid evolution has left us with a significant knowledge gap within the medical community. In this paper, we attempt to bridge that gap and prepare the physician in oncology for multiomics, a group of technologies that have gone from looming on the horizon to become a clinical reality. The era of multiomics is here, and we must prepare ourselves for this exciting new age of cancer medicine.


Subject(s)
Artificial Intelligence , Neoplasms , Genomics , Humans , Medical Oncology , Neoplasms/genetics , Neoplasms/therapy , Proteomics
12.
Mol Syst Biol ; 17(9): e10426, 2021 09.
Article in English | MEDLINE | ID: mdl-34486798

ABSTRACT

Although 15-20% of COVID-19 patients experience hyper-inflammation induced by massive cytokine production, cellular triggers of this process and strategies to target them remain poorly understood. Here, we show that the N-terminal domain (NTD) of the SARS-CoV-2 spike protein substantially induces multiple inflammatory molecules in myeloid cells and human PBMCs. Using a combination of phenotypic screening with machine learning-based modeling, we identified and experimentally validated several protein kinases, including JAK1, EPHA7, IRAK1, MAPK12, and MAP3K8, as essential downstream mediators of NTD-induced cytokine production, implicating the role of multiple signaling pathways in cytokine release. Further, we found several FDA-approved drugs, including ponatinib, and cobimetinib as potent inhibitors of the NTD-mediated cytokine release. Treatment with ponatinib outperforms other drugs, including dexamethasone and baricitinib, inhibiting all cytokines in response to the NTD from SARS-CoV-2 and emerging variants. Finally, ponatinib treatment inhibits lipopolysaccharide-mediated cytokine release in myeloid cells in vitro and lung inflammation mouse model. Together, we propose that agents targeting multiple kinases required for SARS-CoV-2-mediated cytokine release, such as ponatinib, may represent an attractive therapeutic option for treating moderate to severe COVID-19.


Subject(s)
Antiviral Agents/pharmacology , Cytokines/metabolism , Host-Pathogen Interactions/physiology , Animals , Azetidines/pharmacology , Host-Pathogen Interactions/drug effects , Humans , Imidazoles/pharmacology , Interleukin-1 Receptor-Associated Kinases/metabolism , Janus Kinase 1/metabolism , Lipopolysaccharides/toxicity , Machine Learning , Male , Mice , Mice, Inbred C57BL , Neutrophils/virology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Pyridazines/pharmacology , RAW 264.7 Cells , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Sulfonamides/pharmacology
13.
Glia ; 69(9): 2059-2076, 2021 09.
Article in English | MEDLINE | ID: mdl-33638562

ABSTRACT

Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Glioma , Sarcoma , Animals , Avian Sarcoma Viruses/genetics , Brain Neoplasms/pathology , Glioma/pathology , Humans , Oncogenic Viruses , Receptors, Virus , Tumor Microenvironment
14.
Curr Issues Mol Biol ; 41: 381-468, 2021.
Article in English | MEDLINE | ID: mdl-32938804

ABSTRACT

The therapeutic promise of oncolytic viruses (OVs) rests on their ability to both selectively kill tumor cells and induce anti-tumor immunity. The potential of tumors to be recognized and eliminated by an effective anti-tumor immune response has been spurred on by the discovery that immune checkpoint inhibition can overcome tumor-specific cytotoxic T cell (CTL) exhaustion and provide durable responses in multiple tumor indications. OV-mediated tumor destruction is now recognized as a powerful means to assist in the development of anti-tumor immunity for two important reasons: (i) OVs, through the elicitation of an anti-viral response and the production of type I interferon, are potent stimulators of inflammation and can be armed with transgenes to further enhance anti-tumor immune responses; and (ii) lytic activity can promote the release of tumor-associated antigens (TAAs) and tumor neoantigens that function as in situ tumor-specific vaccines to elicit adaptive immunity. Oncolytic herpes simplex viruses (oHSVs) are among the most widely studied OVs for the treatment of solid malignancies, and Amgen's oHSV Imlygic® for the treatment of melanoma is the only OV approved in major markets. Here we describe important biological features of HSV that make it an attractive OV, clinical experience with HSV-based vectors, and strategies to increase applicability to cancer treatment.


Subject(s)
Immune Checkpoint Inhibitors/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Viruses/immunology , Simplexvirus/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , Humans , Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes, Cytotoxic/immunology
15.
Glia ; 68(12): 2486-2502, 2020 12.
Article in English | MEDLINE | ID: mdl-32621641

ABSTRACT

Radiation therapy is part of the standard of care for gliomas and kills a subset of tumor cells, while also altering the tumor microenvironment. Tumor cells with stem-like properties preferentially survive radiation and give rise to glioma recurrence. Various techniques for enriching and quantifying cells with stem-like properties have been used, including the fluorescence activated cell sorting (FACS)-based side population (SP) assay, which is a functional assay that enriches for stem-like tumor cells. In these analyses, mouse models of glioma have been used to understand the biology of this disease and therapeutic responses, including the radiation response. We present combined SP analysis and single-cell RNA sequencing of genetically-engineered mouse models of glioma to show a time course of cellular response to radiation. We identify and characterize two distinct tumor cell populations that are inherently radioresistant and also distinct effects of radiation on immune cell populations within the tumor microenvironment.


Subject(s)
Brain Neoplasms , Glioma , Stem Cells , Animals , Brain Neoplasms/radiotherapy , Mice , Neoplastic Stem Cells , Single-Cell Analysis , Tumor Microenvironment
16.
Neurosurg Focus ; 49(4): E11, 2020 10.
Article in English | MEDLINE | ID: mdl-33002863

ABSTRACT

Glioblastoma (GBM) is the most common type of malignant primary brain tumor in adults. It is a uniformly fatal disease (median overall survival 16 months) even with aggressive resection and an adjuvant temozolomide-based chemoradiation regimen. Age remains an independent risk factor for a poor prognosis. Several factors contribute to the dismal outcomes in the elderly population with GBM, including poor baseline health status, differences in underlying genomic alterations, and variability in the surgical and medical management of this subpopulation. The latter arises from a lack of adequate representation of elderly patients in clinical trials, resulting in limited data on the response of this subpopulation to standard treatment. Results from retrospective and some prospective studies have indicated that resection of only contrast-enhancing lesions and administration of hypofractionated radiotherapy in combination with temozolomide are effective strategies for optimizing survival while maintaining baseline quality of life in elderly GBM patients; however, survival remains dismal relative to that in a younger cohort. Here, the authors present historical context for the current strategies used for the multimodal management (surgical and medical) of elderly patients with GBM. Furthermore, they provide insights into elderly GBM patient-specific genomic signatures such as isocitrate dehydrogenase 1/2 (IDH1/2) wildtype status, telomerase reverse transcriptase promoter (TERTp) mutations, and somatic copy number alterations including CDK4/MDM2 coamplification, which are becoming better understood and could be utilized in a clinical trial design and patient stratification to guide the development of more effective adjuvant therapies specifically for elderly GBM patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/surgery , Clinical Decision-Making , Genomics , Glioblastoma/genetics , Glioblastoma/surgery , Humans , Prognosis , Prospective Studies , Quality of Life , Retrospective Studies
17.
J Neurooncol ; 144(2): 403-407, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31325145

ABSTRACT

PURPOSE: Perifosine (PRF) is an oral alkylphospholipid with antineoplastic effects and reasonable tolerability. It inhibits signaling through the PI3/AKT axis and other cascades of biologic importance in glioblastoma, and has promising pre-clinical activity in vitro and in vivo. Therefore, we conducted a phase II open-label single-arm clinical trial of perifosine for patients with recurrent glioblastoma (GBM). METHODS: We planned to accrue up to 30 adults with recurrent GBM with a minimum Karnofsky Performance Status of 50 following radiotherapy but without other restrictions on the number or types of prior therapy. Concurrent p450 stimulating hepatic enzyme inducing anticonvulsants were prohibited. Patients were treated with a loading dose of 600 mg PRF (in 4 divided doses on day 1) followed by 100 mg daily until either disease progression or intolerable toxicity. The primary endpoint was the 6-month progression free survival (PFS6) rate, with at least 20% considered promising. Accrual was continuous but if 0 of the first 12 patients with GBM reached PFS6, then further accrual would terminate for futility. Patients with other high grade gliomas were accrued concurrently to an exploratory cohort. RESULTS: Treatment was generally well tolerated; gastrointestinal toxicities were the most common side effects, although none resulted in treatment discontinuation. However, there was limited to no efficacy in GBM (n = 16): the PFS6 rate was 0%, median PFS was 1.58 months [95% CI (1.08, 1.84)], median overall survival was 3.68 months [95% CI (2.50, 7.79)], with no radiographic responses. There was a confirmed partial response in one patient with anaplastic astrocytoma (n = 14). CONCLUSIONS: PRF is tolerable but ineffective as monotherapy for GBM. Preclinical data suggests synergistic effects of PRF in combination with other approaches, and further study is ongoing.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Phosphorylcholine/analogs & derivatives , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adult , Aged , Brain Neoplasms/pathology , Female , Follow-Up Studies , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Phosphorylcholine/therapeutic use , Prognosis , Prospective Studies , Survival Rate , Young Adult
18.
Proc Natl Acad Sci U S A ; 113(19): 5394-9, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27118839

ABSTRACT

We show that visualizing large molecular and clinical datasets enables discovery of molecularly defined categories of highly similar patients. We generated a series of linked 2D sample similarity plots using genome-wide single nucleotide alterations (SNAs), copy number alterations (CNAs), DNA methylation, and RNA expression data. Applying this approach to the combined glioblastoma (GBM) and lower grade glioma (LGG) The Cancer Genome Atlas datasets, we find that combined CNA/SNA data divide gliomas into three highly distinct molecular groups. The mutations commonly used in clinical evaluation of these tumors are regionally distributed in these plots. One of the three groups is a mixture of GBM and LGG that shows similar methylation and survival characteristics to GBM. Altogether, our approach identifies eight molecularly defined glioma groups with distinct sequence/expression/methylation profiles. Importantly, we show that regionally clustered samples are enriched for specific drug targets.


Subject(s)
Data Mining/methods , Databases, Genetic , Datasets as Topic , High-Throughput Nucleotide Sequencing/methods , Neoplasm Proteins/genetics , User-Computer Interface , Biomarkers, Tumor/genetics , Brain Neoplasms , Computer Graphics , Genetic Predisposition to Disease/genetics , Glioma , Humans
19.
EMBO J ; 33(18): 1984-6, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25061226

ABSTRACT

Heterogeneity within tumors is becoming increasingly recognized as an important cause of treatment failure in cancer. Two recent studies use fate-mapping and limiting dilution transplantation assays to identify SRY (sex determining region Y)-box 2 (Sox2) as cancer stem-cell marker and driver of cancer stemness.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cerebellar Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , SOXB1 Transcription Factors/metabolism , Skin Neoplasms , Animals , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL