Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Extracell Vesicle ; 32024 Jun.
Article in English | MEDLINE | ID: mdl-38872853

ABSTRACT

Antibodies are critical tools for research into extracellular vesicles (EVs) and other extracellular nanoparticles (ENPs), where they can be used for their identification, characterization, and isolation. However, the lack of a centralized antibody platform where researchers can share validation results thus minimizing wasted personnel time and reagents, has been a significant obstacle. Moreover, because the performance of antibodies varies among assay types and conditions, detailed information on assay variables and protocols is also of value. To facilitate sharing of results on antibodies that are relevant to EV/ENP research, the EV Antibody Database has been developed by the investigators of the Extracellular RNA Communication Consortium (ERCC). Hosted by the ExRNA Portal (https://exrna.org/resources/evabdb/), this interactive database aggregates and shares results from antibodies that have been tested by research groups in the EV/ENP field. Currently, the EV Antibody Database includes modules for antibodies tested for western Blot, EV Flow Cytometry, and EV Sandwich Assays, and holds 110 records contributed by 6 laboratories from the ERCC. Detailed information on antibody sources, assay conditions, and results is provided, including negative results. We encourage ongoing expert input and community feedback to enhance the database's utility, making it a valuable resource for comprehensive validation data on antibodies and protocols in EV biology.

2.
Cancer Immunol Res ; 11(5): 629-645, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36881002

ABSTRACT

The composition of the tumor immune microenvironment (TIME) is considered a key determinant of patients' response to immunotherapy. The mechanisms underlying TIME formation and development over time are poorly understood. Glioblastoma (GBM) is a lethal primary brain cancer for which there are no curative treatments. GBMs are immunologically heterogeneous and impervious to checkpoint blockade immunotherapies. Utilizing clinically relevant genetic mouse models of GBM, we identified distinct immune landscapes associated with expression of EGFR wild-type and mutant EGFRvIII cancer driver mutations. Over time, accumulation of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) was more pronounced in EGFRvIII-driven GBMs and was correlated with resistance to PD-1 and CTLA-4 combination checkpoint blockade immunotherapy. We determined that GBM-secreted CXCL1/2/3 and PMN-MDSC-expressed CXCR2 formed an axis regulating output of PMN-MDSCs from the bone marrow leading to systemic increase in these cells in the spleen and GBM tumor-draining lymph nodes. Pharmacologic targeting of this axis induced a systemic decrease in the numbers of PMN-MDSC, facilitated responses to PD-1 and CTLA-4 combination checkpoint blocking immunotherapy, and prolonged survival in mice bearing EGFRvIII-driven GBM. Our results uncover a relationship between cancer driver mutations, TIME composition, and sensitivity to checkpoint blockade in GBM and support the stratification of patients with GBM for checkpoint blockade therapy based on integrated genotypic and immunologic profiles.


Subject(s)
Brain Neoplasms , Glioblastoma , Myeloid-Derived Suppressor Cells , Animals , Mice , Glioblastoma/therapy , Glioblastoma/drug therapy , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Programmed Cell Death 1 Receptor , Cell Line, Tumor , Immunotherapy , Mutation , Tumor Microenvironment/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy
4.
iScience ; 25(8): 104653, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35958027

ABSTRACT

The extracellular RNA communication consortium (ERCC) is an NIH-funded program aiming to promote the development of new technologies, resources, and knowledge about exRNAs and their carriers. After Phase 1 (2013-2018), Phase 2 of the program (ERCC2, 2019-2023) aims to fill critical gaps in knowledge and technology to enable rigorous and reproducible methods for separation and characterization of both bulk populations of exRNA carriers and single EVs. ERCC2 investigators are also developing new bioinformatic pipelines to promote data integration through the exRNA atlas database. ERCC2 has established several Working Groups (Resource Sharing, Reagent Development, Data Analysis and Coordination, Technology Development, nomenclature, and Scientific Outreach) to promote collaboration between ERCC2 members and the broader scientific community. We expect that ERCC2's current and future achievements will significantly improve our understanding of exRNA biology and the development of accurate and efficient exRNA-based diagnostic, prognostic, and theranostic biomarker assays.

5.
Nat Immunol ; 23(6): 971-984, 2022 06.
Article in English | MEDLINE | ID: mdl-35624211

ABSTRACT

Glioblastoma (GBM) is an incurable primary malignant brain cancer hallmarked with a substantial protumorigenic immune component. Knowledge of the GBM immune microenvironment during tumor evolution and standard of care treatments is limited. Using single-cell transcriptomics and flow cytometry, we unveiled large-scale comprehensive longitudinal changes in immune cell composition throughout tumor progression in an epidermal growth factor receptor-driven genetic mouse GBM model. We identified subsets of proinflammatory microglia in developing GBMs and anti-inflammatory macrophages and protumorigenic myeloid-derived suppressors cells in end-stage tumors, an evolution that parallels breakdown of the blood-brain barrier and extensive growth of epidermal growth factor receptor+ GBM cells. A similar relationship was found between microglia and macrophages in patient biopsies of low-grade glioma and GBM. Temozolomide decreased the accumulation of myeloid-derived suppressor cells, whereas concomitant temozolomide irradiation increased intratumoral GranzymeB+ CD8+T cells but also increased CD4+ regulatory T cells. These results provide a comprehensive and unbiased immune cellular landscape and its evolutionary changes during GBM progression.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Animals , Brain Neoplasms/metabolism , ErbB Receptors , Glioblastoma/metabolism , Humans , Mice , Sequence Analysis, RNA , Single-Cell Analysis , Temozolomide/therapeutic use , Tumor Microenvironment/genetics
6.
Oncogene ; 40(15): 2682-2696, 2021 04.
Article in English | MEDLINE | ID: mdl-33707748

ABSTRACT

Focal amplification of epidermal growth factor receptor (EGFR) and its ligand-independent, constitutively active EGFRvIII mutant form are prominent oncogenic drivers in glioblastoma (GBM). The EGFRvIII gene rearrangement is considered to be an initiating event in the etiology of GBM, however, the mechanistic details of how EGFRvIII drives cellular transformation and tumor maintenance remain unclear. Here, we report that EGFRvIII demonstrates a reliance on PDGFRA co-stimulatory signaling during the tumorigenic process in a genetically engineered autochthonous GBM model. This dependency exposes liabilities that were leveraged using kinase inhibitors treatments in EGFRvIII-expressing GBM patient-derived xenografts (PDXs), where simultaneous pharmacological inhibition of EGFRvIII and PDGFRA kinase activities is necessary for anti-tumor efficacy. Our work establishes that EGFRvIII-positive tumors have unexplored vulnerabilities to targeted agents concomitant to the EGFR kinase inhibitor repertoire.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/pathology , HEK293 Cells , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors
7.
Adv Biosyst ; 4(12): e2000069, 2020 12.
Article in English | MEDLINE | ID: mdl-32815324

ABSTRACT

Extracellular vesicles (EVs) offer a vehicle for diagnostic and therapeutic utility. EVs carry bioactive cargo and an accrued interest in their characterization has emerged. Efforts at identifying EV-enriched protein or RNA led to a surprising realization that EVs are excessively heterogeneous in nature. This diversity is originally attributed to vesicle sizes but it is becoming evident that different classes of EVs vehiculate distinct molecular cargos. Therefore, one of the current challenges in EV research is their selective isolation in quantities sufficient for efficient downstream analyses. Many protocols have been developed; however, reproducibility between research groups can be difficult to reach and inter-studies analyses of data from different isolation protocols are unmanageable. Therefore, there is an unmet need to optimize and standardize methods and protocols for the isolation and purification of EVs. This review focuses on the diverse techniques and protocols used over the years to isolate and purify EVs with a special emphasis on their adequacy for proteomics applications. By combining recent advances in specific isolation methods that yield superior quality of EV preparations and mass spectrometry techniques, the field is now prepared for transformative advancements in establishing distinct categorization and cargo identification of subpopulations based on EV surface markers.


Subject(s)
Biomarkers , Extracellular Vesicles , Proteome , Animals , Biomarkers/analysis , Biomarkers/chemistry , Biomarkers/metabolism , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Humans , Mass Spectrometry , Mice , Proteome/analysis , Proteome/chemistry , Proteome/metabolism , Proteomics
8.
Clin Neuropathol ; 39(3): 126-134, 2020.
Article in English | MEDLINE | ID: mdl-31841105

ABSTRACT

The molecular alterations underlying progression of low-grade glial/glioneuronal tumors remain to be elucidated. We present a case of a 56-year-old male with an enhancing left temporal lobe tumor. Histology revealed a high-grade glioma adjacent to a low-grade glioneuronal component with abundant Rosenthal fibers, focal eosinophilic granular bodies, and CD34-positive neurons. The tumor was negative for IDH1 (R132H), BRAF-V600E, and the KIAA1549-BRAF fusion. Comparative genomic hybridization detected a large amplification (> 15 copies) of the Son of Sevenless 1 (SOS1) gene, a component of the MAPK pathway. Although activating mutations in the MAPK pathway occur frequently in gliomas and glioneuronal tumors, SOS1 gene amplification has not been reported previously. This case indicates another potential mechanism for MAPK activation in glial tumors.


Subject(s)
Astrocytoma/genetics , Glioma/pathology , Mutation/genetics , SOS1 Protein/genetics , Astrocytoma/diagnosis , Astrocytoma/pathology , Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Comparative Genomic Hybridization/methods , Glioma/genetics , Humans , Male , Middle Aged , Neoplasm Grading , Proto-Oncogene Proteins B-raf/genetics
9.
Cell Rep ; 27(13): 3972-3987.e6, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242427

ABSTRACT

Cancer extracellular vesicles (EVs) are highly heterogeneous, which impedes our understanding of their function as intercellular communication agents and biomarkers. To deconstruct this heterogeneity, we analyzed extracellular RNAs (exRNAs) and extracellular proteins (exPTNs) from size fractionation of large, medium, and small EVs and ribonucleoprotein complexes (RNPs) from mouse glioblastoma cells by RNA sequencing and quantitative proteomics. mRNA from medium-sized EVs most closely reflects the cellular transcriptome, whereas small EV exRNA is enriched in small non-coding RNAs and RNPs contain precisely processed tRNA fragments. The exPTN composition of EVs and RNPs reveals that they are closely related by vesicle type, independent of their cellular origin, and single EV analysis reveals that small EVs are less heterogeneous in their protein content than larger ones. We provide a foundation for better understanding of segregation of macromolecules in glioma EVs through a catalog of diverse exRNAs and exPTNs.


Subject(s)
Extracellular Vesicles/metabolism , Glioblastoma/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Animals , Cell Line, Tumor , Extracellular Vesicles/pathology , Glioblastoma/pathology , Mice
10.
Life Sci Alliance ; 1(3): e201800029, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30456354

ABSTRACT

Activation of the platelet-derived growth factor receptors (PDGFRs) gives rise to some of the most important signaling pathways that regulate mammalian cellular growth, survival, proliferation, and differentiation and their misregulation is common in a variety of diseases. Herein, we present a comprehensive and detailed map of PDGFR signaling pathways assembled from literature and integrate this map in a bioinformatics protocol designed to extract meaningful information from large-scale quantitative proteomics mass spectrometry data. We demonstrate the usefulness of this approach using a new genetically engineered mouse model of PDGFRα-driven glioma. We discovered that acute PDGFRα stimulation differs considerably from chronic receptor activation in the regulation of protein translation initiation. Transient stimulation activates several key components of the translation initiation machinery, whereas the clinically relevant chronic activity of PDGFRα is associated with a significant shutdown of translational members. Our work defines a step-by-step approach to extract biologically relevant insights from global unbiased phospho-protein datasets to uncover targets for therapeutic assessment.

11.
Nat Commun ; 9(1): 3116, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082792

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive primary brain cancer that includes focal amplification of PDGFRα and for which there are no effective therapies. Herein, we report the development of a genetically engineered mouse model of GBM based on autocrine, chronic stimulation of overexpressed PDGFRα, and the analysis of GBM signaling pathways using proteomics. We discover the tubulin-binding protein Stathmin1 (STMN1) as a PDGFRα phospho-regulated target, and that this mis-regulation confers sensitivity to vinblastine (VB) cytotoxicity. Treatment of PDGFRα-positive mouse and a patient-derived xenograft (PDX) GBMs with VB in mice prolongs survival and is dependent on STMN1. Our work reveals a previously unconsidered link between PDGFRα activity and STMN1, and highlight an STMN1-dependent cytotoxic effect of VB in GBM.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stathmin/metabolism , Vinblastine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle , Cell Survival , Cells, Cultured , Computational Biology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Magnetic Resonance Imaging , Male , Mice , Neoplasm Transplantation , Phosphorylation , Proteomics , Signal Transduction
12.
Cancer Res ; 76(10): 2876-81, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27013191

ABSTRACT

A lack of experimental models of tumor heterogeneity limits our knowledge of the complex subpopulation dynamics within the tumor ecosystem. In high-grade gliomas (HGG), distinct hierarchical cell populations arise from different glioma stem-like cell (GSC) subpopulations. Extracellular vesicles (EV) shed by cells may serve as conduits of genetic and signaling communications; however, little is known about how HGG heterogeneity may impact EV content and activity. In this study, we performed a proteomic analysis of EVs isolated from patient-derived GSC of either proneural or mesenchymal subtypes. EV signatures were heterogeneous, but reflected the molecular make-up of the GSC and consistently clustered into the two subtypes. EV-borne protein cargos transferred between proneural and mesenchymal GSC increased protumorigenic behaviors in vitro and in vivo Clinically, analyses of HGG patient data from the The Cancer Genome Atlas database revealed that proneural tumors with mesenchymal EV signatures or mesenchymal tumors with proneural EV signatures were both associated with worse outcomes, suggesting influences by the proportion of tumor cells of varying subtypes in tumors. Collectively, our findings illuminate the heterogeneity among tumor EVs and the complexity of HGG heterogeneity, which these EVs help to maintain. Cancer Res; 76(10); 2876-81. ©2016 AACR.


Subject(s)
Brain Neoplasms/pathology , Carcinogenesis , Extracellular Vesicles/pathology , Glioma/pathology , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/metabolism , Cell Proliferation , Extracellular Vesicles/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Proteomics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 112(11): 3493-8, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733882

ABSTRACT

Oncogenic c-ros oncogene1 (ROS1) fusion kinases have been identified in a variety of human cancers and are attractive targets for cancer therapy. The MET/ALK/ROS1 inhibitor crizotinib (Xalkori, PF-02341066) has demonstrated promising clinical activity in ROS1 fusion-positive non-small cell lung cancer. However, emerging clinical evidence has shown that patients can develop resistance by acquiring secondary point mutations in ROS1 kinase. In this study we characterized the ROS1 activity of PF-06463922, a novel, orally available, CNS-penetrant, ATP-competitive small-molecule inhibitor of ALK/ROS1. In vitro, PF-06463922 exhibited subnanomolar cellular potency against oncogenic ROS1 fusions and inhibited the crizotinib-refractory ROS1(G2032R) mutation and the ROS1(G2026M) gatekeeper mutation. Compared with crizotinib and the second-generation ALK/ROS1 inhibitors ceritinib and alectinib, PF-06463922 showed significantly improved inhibitory activity against ROS1 kinase. A crystal structure of the PF-06463922-ROS1 kinase complex revealed favorable interactions contributing to the high-affinity binding. In vivo, PF-06463922 showed marked antitumor activity in tumor models expressing FIG-ROS1, CD74-ROS1, and the CD74-ROS1(G2032R) mutation. Furthermore, PF-06463922 demonstrated antitumor activity in a genetically engineered mouse model of FIG-ROS1 glioblastoma. Taken together, our results indicate that PF-06463922 has potential for treating ROS1 fusion-positive cancers, including those requiring agents with CNS-penetrating properties, as well as for overcoming crizotinib resistance driven by ROS1 mutation.


Subject(s)
Drug Resistance, Neoplasm/genetics , Lactams, Macrocyclic/pharmacology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Aminopyridines , Animals , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Proliferation/drug effects , Crizotinib , Crystallography, X-Ray , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Glioma/pathology , Humans , Lactams , Lactams, Macrocyclic/chemistry , Mice , Models, Molecular , Signal Transduction/drug effects
14.
Endocrinology ; 155(9): 3661-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24971615

ABSTRACT

The initial segment (IS) of the epididymis plays an essential role in male fertility. The IS epithelium is undifferentiated and nonfunctional at birth. Prior to puberty, the epithelium undergoes differentiation that leads to the formation of a fully functional organ. However, the mechanistic details of this program are not well understood. To explore this further, we used genetic engineering to create a kinase dead allele of the ROS1 receptor tyrosine kinase in mice and studied the effects of ROS1 tyrosine kinase activity on the differentiation of the IS epithelium. We show that the expression and activation of ROS1 coincides with the onset of differentiation and is exclusively located in the IS of the maturing and adult mouse epididymides. Here we demonstrate that the differentiation of the IS is dependent on the kinase activity of ROS1 and its downstream effector MEK1/2-ERK1/2 signaling axis. Using genetic engineering, we show that germ line ablation of ROS1 kinase activity leads to a failure of the IS epithelium to differentiate, and as a consequence sperm maturation and infertility were dramatically perturbed. Pharmacological inhibition of ROS1 kinase activity in the developing epididymis, however, only delayed differentiation transiently and did not result in infertility. Our results demonstrate that ROS1 kinase activity and the ensuing MEK1/2-ERK1/2 signaling are necessary for the postnatal development of the IS epithelium and that a sustained ablation of ROS1 kinase activity within the critical window of terminal differentiation abrogate the function of the epididymis and leads to sterility.


Subject(s)
Cell Differentiation , Epididymis/cytology , Epididymis/enzymology , Epithelial Cells/enzymology , MAP Kinase Signaling System , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Epididymis/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Male , Mice , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Spermatozoa/cytology , Spermatozoa/enzymology , Spermatozoa/growth & development , Spermatozoa/metabolism
15.
Cancer Res ; 71(23): 7198-206, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21987724

ABSTRACT

Glioblastoma multiforme (GBM) is characterized by overexpression of epidermal growth factor receptor (EGFR) and loss of the tumor suppressors Ink4a/Arf. Efforts at modeling GBM using wild-type EGFR in mice have proven unsuccessful. Here, we present a unique mouse model of wild-type EGFR-driven gliomagenesis. We used a combination of somatic conditional overexpression and ligand-mediated chronic activation of EGFR in cooperation with Ink4a/Arf loss in the central nervous system of adult mice to generate tumors with the histopathologic and molecular characteristics of human GBMs. Sustained, ligand-mediated activation of EGFR was necessary for gliomagenesis, functionally substantiating the clinical observation that EGFR-positive GBMs from patients express EGFR ligands. To gain a better understanding of the clinically disappointing EGFR-targeted therapies for GBM, we investigated the molecular responses to EGFR tyrosine kinase inhibitor (TKI) treatment in this model. Gefitinib treatment of primary GBM cells resulted in a robust apoptotic response, partially conveyed by mitogen-activated protein kinase (MAPK) signaling attenuation and accompanied by BIM(EL) expression. In human GBMs, loss-of-function mutations in the tumor suppressor PTEN are a common occurrence. Elimination of PTEN expression in GBM cells posttumor formation did not confer resistance to TKI treatment, showing that PTEN status in our model is not predictive. Together, these findings offer important mechanistic insights into the genetic determinants of EGFR gliomagenesis and sensitivity to TKIs and provide a robust discovery platform to better understand the molecular events that are associated with predictive markers of TKI therapy.


Subject(s)
Brain Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p16/deficiency , ErbB Receptors/metabolism , Glioblastoma/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enzyme Activation , ErbB Receptors/genetics , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Ligands , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects
16.
Glia ; 59(8): 1155-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21305617

ABSTRACT

The recently published comprehensive profiles of genomic alterations in glioma have led to a refinement in our understanding of the molecular events that underlie this cancer. Using state-of-the-art genomic tools, several laboratories have created and characterized accurate genetically engineered mouse models of glioma based on specific genetic alterations observed in human tumors. These in vivo brain tumor models faithfully recapitulate the histopathology, etiology, and biology of gliomas and provide an exceptional experimental system to discover novel therapeutic targets and test therapeutic agents. This review focuses on mouse models of glioma with a special emphasis on genetically engineered models developed around key genetic glioma signature mutations in the PDGFR, EGFR, and NF1 genes and pathways. The resulting animal models have provided insight into many fundamental and mechanistic facets of tumor initiation, maintenance and resistance to therapeutic intervention and will continue to do so in the future.


Subject(s)
Brain Neoplasms/genetics , Disease Models, Animal , Glioma/genetics , Animals , Brain Neoplasms/classification , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioma/classification , Humans , Mice , Mice, Transgenic , Mutation/genetics , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Signal Transduction/genetics
17.
Mol Cancer Ther ; 9(9): 2618-26, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20643786

ABSTRACT

Glioblastoma multiforme (GBM) has an abysmal prognosis. We now know that the epidermal growth factor receptor (EGFR) signaling pathway and the loss of function of the tumor suppressor genes p16Ink4a/p19ARF and PTEN play a crucial role in GBM pathogenesis: initiating the early stages of tumor development, sustaining tumor growth, promoting infiltration, and mediating resistance to therapy. We have recently shown that this genetic combination is sufficient to promote the development of GBM in adult mice. Therapeutic agents raised against single targets of the EGFR signaling pathway have proven rather inefficient in GBM therapy, showing the need for combinatorial therapeutic approaches. An effective strategy for concurrent disruption of multiple signaling pathways is via the inhibition of the molecular chaperone heat shock protein 90 (Hsp90). Hsp90 inhibition leads to the degradation of so-called client proteins, many of which are key effectors of GBM pathogenesis. NXD30001 is a novel second generation Hsp90 inhibitor that shows improved pharmacokinetic parameters. Here we show that NXD30001 is a potent inhibitor of GBM cell growth in vitro consistent with its capacity to inhibit several key targets and regulators of GBM biology. We also show the efficacy of NXD30001 in vivo in an EGFR-driven genetically engineered mouse model of GBM. Our findings establish that the Hsp90 inhibitor NXD30001 is a therapeutically multivalent molecule, whose actions strike GBM at the core of its drivers of tumorigenesis and represent a compelling rationale for its use in GBM treatment.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactones/pharmacology , Oximes/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Growth Processes/drug effects , Disease Models, Animal , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactones/pharmacokinetics , Male , Mice , Mice, Transgenic , Oximes/pharmacokinetics , Signal Transduction/drug effects , Tumor Cells, Cultured
18.
Genesis ; 47(10): 659-66, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19603508

ABSTRACT

Genetically engineered, Cre/LoxP-conditional mouse models of cancer are designed to investigate the genetic contributors of tumorigenesis and are well suited to assess therapeutic treatment responses. The capacity to serially visualize tumor burden in a noninvasive fashion would greatly strengthen their applications. We report the generation of a bioluminescent reporter strain that allows monitoring of tumor development in preexisting conditional mouse tumor models. We demonstrate that, in a Cre-dependent glioblastoma multiforme model, tumor initiation and progression is readily monitored over time and that luminescent output is related to tumor volume. Our results show that this reporter strain may be combined with various Cre/loxP mouse tumor models to allow for noninvasive longitudinal monitoring of tumor growth and therapeutic response in vivo.


Subject(s)
Cell Transformation, Neoplastic/pathology , Genes, Reporter , Glioblastoma/pathology , Integrases/genetics , Luciferases/analysis , Luminescent Measurements/methods , Animals , Cell Transformation, Neoplastic/chemistry , Female , Glioblastoma/chemistry , Luciferases/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
19.
Biosens Bioelectron ; 24(11): 3252-7, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19442510

ABSTRACT

Biopsies provide required information to diagnose cancer but, because of their invasiveness, they are difficult to use for managing cancer therapy. The ability to repeatedly sample the local environment for tumor biomarker, chemotherapeutic agent, and tumor metabolite concentrations could improve early detection of metastasis and personalized therapy. Here we describe an implantable diagnostic device that senses the local in vivo environment. This device, which could be left behind during biopsy, uses a semi-permeable membrane to contain nanoparticle magnetic relaxation switches. A cell line secreting a model cancer biomarker produced ectopic tumors in mice. The transverse relaxation time (T(2)) of devices in tumor-bearing mice was 20+/-10% lower than devices in control mice after 1 day by magnetic resonance imaging (p<0.01). Short term applications for this device are numerous, including verification of successful tumor resection. This may represent the first continuous monitoring device for soluble cancer biomarkers in vivo.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques/instrumentation , Choriocarcinoma/immunology , Choriocarcinoma/mortality , Immunoassay/instrumentation , Magnetics/instrumentation , Monitoring, Ambulatory/instrumentation , Prostheses and Implants , Animals , Biomarkers, Tumor/immunology , Cell Line, Tumor , Choriocarcinoma/pathology , Equipment Design , Equipment Failure Analysis , Female , Mice , Mice, Nude , Sensitivity and Specificity
20.
Proc Natl Acad Sci U S A ; 106(8): 2712-6, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196966

ABSTRACT

Glioblastoma multiforme (GBM) is a highly lethal brain tumor for which little treatment is available. The epidermal growth factor receptor (EGFR) signaling pathway is thought to play a crucial role in GBM pathogenesis, initiating the early stages of tumor development, sustaining tumor growth, promoting infiltration, and mediating resistance to therapy. The importance of this pathway is highlighted in the fact that EGFR is mutationally activated in over 50% of GBM tumors. Consistent with this, we show here that concomitant activation of wild-type and/or mutant (vIII) EGFR and ablation of Ink4A/Arf and PTEN tumor suppressor gene function in the adult mouse central nervous system generates a fully penetrant, rapid-onset high-grade malignant glioma phenotype with prominent pathological and molecular resemblance to GBM in humans. Studies of the activation of signaling events in these GBM tumor cells revealed notable differences between wild-type and vIII EGFR-expressing cells. We show that wild-type EGF receptor signals through its canonical pathways, whereas tumors arising from expression of mutant EGFR(vIII) do not use these same pathways. Our findings provide critical insights into the role of mutant EGFR signaling function in GBM tumor biology and set the stage for testing of targeted therapeutic agents in the preclinical models described herein.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Genes, Tumor Suppressor , Glioblastoma/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Immunohistochemistry , Mice , Mice, Transgenic , PTEN Phosphohydrolase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL