Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Genes Dev ; 31(12): 1212-1227, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28724615

ABSTRACT

In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glioblastoma/physiopathology , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction/genetics , Animals , Cell Communication , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-6/metabolism , Mice , Mice, Nude , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Ther ; 22(9): 1678-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24895995

ABSTRACT

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMß2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Glioblastoma/immunology , Herpesvirus 1, Human/genetics , Macrophages/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Chemokine CCL2/metabolism , Female , Genetic Vectors/administration & dosage , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Macrophage Activation , Mice , Neoplasm Transplantation , Oncolytic Viruses/genetics
3.
Front Immunol ; 11: 327, 2020.
Article in English | MEDLINE | ID: mdl-32161596

ABSTRACT

Overcoming tolerance to tumor-associated antigens remains a hurdle for cancer vaccine-based immunotherapy. A strategy to enhance the anti-tumor immune response is the inclusion of adjuvants to cancer vaccine protocols. In this report, we generated and systematically screened over twenty gene-based molecular adjuvants composed of cytokines, chemokines, and T cell co-stimulators for the ability to increase anti-tumor antigen T cell immunity. We identified several robust adjuvants whose addition to vaccine formulations resulted in enhanced T cell responses targeting the cancer antigens STEAP1 and TERT. We further characterized direct T cell stimulation through CD80-Fc and indirect T cell targeting via the dendritic cell activator Flt3L-Fc. Mechanistically, intramuscular delivery of Flt3L-Fc into mice was associated with a significant increase in infiltration of dendritic cells at the site of administration and trafficking of activated dendritic cells to the draining lymph node. Gene expression analysis of the muscle tissue confirmed a significant up-regulation in genes associated with dendritic cell signaling. Addition of CD80-Fc to STEAP1 vaccine formulation mimicked the engagement provided by DCs and increased T cell responses to STEAP1 by 8-fold, significantly increasing the frequency of antigen-specific cells expressing IFNγ, TNFα, and CD107a for both CD8+ and CD4+ T cells. CD80-Fc enhanced T cell responses to multiple tumor-associated antigens including Survivin and HPV, indicating its potential as a universal adjuvant for cancer vaccines. Together, the results of our study highlight the adjuvanting effect of T cell engagement either directly, CD80-Fc, or indirectly, Flt3L-Fc, for cancer vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , B7-1 Antigen/immunology , Cancer Vaccines/immunology , Membrane Proteins/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Tetraspanin 28/immunology , Animals , Antigens, Neoplasm , B7-1 Antigen/genetics , Cell Movement/immunology , Cytokines/metabolism , DNA/genetics , Dendritic Cells/immunology , Female , Humans , Immunotherapy/methods , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Plasmids/genetics , Vaccines, DNA/genetics , Vaccines, DNA/immunology
4.
Cancer Cell ; 34(1): 163-177.e7, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29990498

ABSTRACT

We explored the clinical and pathological impact of epidermal growth factor receptor (EGFR) extracellular domain missense mutations. Retrospective assessment of 260 de novo glioblastoma patients revealed a significant reduction in overall survival of patients having tumors with EGFR mutations at alanine 289 (EGFRA289D/T/V). Quantitative multi-parametric magnetic resonance imaging analyses indicated increased tumor invasion for EGFRA289D/T/V mutants, corroborated in mice bearing intracranial tumors expressing EGFRA289V and dependent on ERK-mediated expression of matrix metalloproteinase-1. EGFRA289V tumor growth was attenuated with an antibody against a cryptic epitope, based on in silico simulation. The findings of this study indicate a highly invasive phenotype associated with the EGFRA289V mutation in glioblastoma, postulating EGFRA289V as a molecular marker for responsiveness to therapy with EGFR-targeting antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Magnetic Resonance Imaging , Mutation, Missense , Adolescent , Adult , Aged , Animals , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Child , Child, Preschool , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genetic Predisposition to Disease , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Image Interpretation, Computer-Assisted , Infant , Infant, Newborn , Machine Learning , Male , Matrix Metalloproteinase 1/metabolism , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phenotype , Phosphorylation , Predictive Value of Tests , Protein Domains , Retrospective Studies , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Young Adult
5.
Neuro Oncol ; 18(7): 914-8, 2016 07.
Article in English | MEDLINE | ID: mdl-26755074

ABSTRACT

With the evolution of technology, there is now a deeper understanding of glioblastoma as an inter- and intraheterogeneous disease comprising a multitude of genetically and epigenetically different cancer cells. Greater characterization of glioblastoma at the molecular level has improved its initial pathophysiological staging and classification. With this knowledge comes the hope that more efficacious therapies to combat this highly lethal disease are on the horizon. One possibility for intervention is represented by the targeting of epidermal growth factor receptor (EGFR), which is amplified and mutated in a large subset of patients. In this review, we provide a brief overview of EGFR and its mutated form, EGFR variant III, describing the downstream cellular pathways activated by each receptor, available animal models, therapeutic strategies to inhibit the receptor, and possible intervention routes to efficiently target this receptor and prevent the emergence of resistant mechanisms which to date have hampered a successful therapeutic outcome.


Subject(s)
Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Animals , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Humans , Signal Transduction/drug effects
6.
Cancer Res ; 75(2): 394-404, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25432173

ABSTRACT

EGFR is the most common genetically altered oncogene in glioblastoma (GBM), but small-molecule EGFR tyrosine kinase inhibitors (TKI) have failed to yield durable clinical benefit. Here, we show that in two novel model systems of acquired resistance to EGFR TKIs, elevated expression of urokinase plasminogen activator (uPA) drives signaling through the MAPK pathway, which results in suppression of the proapoptotic BCL2-family member protein BIM (BCL2L11). In patient-derived GBM cells and genetic GBM models, uPA is shown to suppress BIM levels through ERK1/2 phosphorylation, which can be reversed by siRNA-mediated knockdown of uPA. TKI-resistant GBMs are resensitized to EGFR TKIs by pharmacologic inhibition of MEK or a BH3 mimetic drug to replace BIM function. A link between the uPA-uPAR-ERK1/2 pathway and BIM has not been previously demonstrated in GBM, and involvement of this signaling axis in resistance provides rationale for a new strategy to target EGFR TKI-resistant GBM.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Brain Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , Glioblastoma/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Bcl-2-Like Protein 11 , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , ErbB Receptors/genetics , Erlotinib Hydrochloride , Female , Gefitinib , Glioblastoma/drug therapy , Glioblastoma/genetics , Heterografts , Humans , Mice , Mice, Nude , Quinazolines/pharmacology , Signal Transduction/drug effects
7.
Oncotarget ; 5(20): 9703-9, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25210852

ABSTRACT

SapC-DOPS is a novel nanotherapeutic that has been shown to target and induce cell death in a variety of cancers, including glioblastoma (GBM). GBM is a primary brain tumor known to frequently demonstrate resistance to apoptosis-inducing therapeutics. Here we explore the mode of action for SapC-DOPS in GBM, a treatment being developed by Bexion Pharmaceuticals for clinical testing in patients. SapC-DOPS treatment was observed to induce lysosomal dysfunction of GBM cells characterized by decreased glycosylation of LAMP1 and altered proteolytic processing of cathepsin D independent of apoptosis and autophagic cell death. We observed that SapC-DOPS induced lysosomal membrane permeability (LMP) as shown by LysoTracker Red and Acridine Orange staining along with an increase of sphingosine, a known inducer of LMP. Additionally, SapC-DOPS displayed strong synergistic interactions with the apoptosis-inducing agent TMZ. Collectively our data suggest that SapC-DOPS induces lysosomal cell death in GBM cells, providing a new approach for treating tumors resistant to traditional apoptosis-inducing agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Nanostructures/administration & dosage , Phosphatidylserines/pharmacology , Saposins/pharmacology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Lysosomes/drug effects , Mice , Mice, Nude , Random Allocation , Saposins/administration & dosage , Temozolomide , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL