Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 11(11): 2904-2923, 2021 11.
Article in English | MEDLINE | ID: mdl-34039636

ABSTRACT

Glioblastoma (GBM) is highly resistant to chemotherapies, immune-based therapies, and targeted inhibitors. To identify novel drug targets, we screened orthotopically implanted, patient-derived glioblastoma sphere-forming cells using an RNAi library to probe essential tumor cell metabolic programs. This identified high dependence on mitochondrial fatty acid metabolism. We focused on medium-chain acyl-CoA dehydrogenase (MCAD), which oxidizes medium-chain fatty acids (MCFA), due to its consistently high score and high expression among models and upregulation in GBM compared with normal brain. Beyond the expected energetics impairment, MCAD depletion in primary GBM models induced an irreversible cascade of detrimental metabolic effects characterized by accumulation of unmetabolized MCFAs, which induced lipid peroxidation and oxidative stress, irreversible mitochondrial damage, and apoptosis. Our data uncover a novel protective role for MCAD to clear lipid molecules that may cause lethal cell damage, suggesting that therapeutic targeting of MCFA catabolism may exploit a key metabolic feature of GBM. SIGNIFICANCE: MCAD exerts a protective role to prevent accumulation of toxic metabolic by-products in glioma cells, actively catabolizing lipid species that would otherwise affect mitochondrial integrity and induce cell death. This work represents a first demonstration of a nonenergetic role for dependence on fatty acid metabolism in cancer.This article is highlighted in the In This Issue feature, p. 2659.


Subject(s)
Acyl-CoA Dehydrogenase , Glioblastoma , Lipid Peroxidation , Mitochondria , Acyl-CoA Dehydrogenase/metabolism , Apoptosis , Fatty Acids/metabolism , Glioblastoma/enzymology , Glioblastoma/genetics , Humans , Mitochondria/metabolism , Oxidative Stress
3.
Cell Metab ; 30(3): 525-538.e8, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31303424

ABSTRACT

Advances in DNA sequencing technologies have reshaped our understanding of the molecular basis of cancer, providing a precise genomic view of tumors. Complementary biochemical and biophysical perspectives of cancer point toward profound shifts in nutrient uptake and utilization that propel tumor growth and major changes in the structure of the plasma membrane of tumor cells. The molecular mechanisms that bridge these fundamental aspects of tumor biology remain poorly understood. Here, we show that the lysophosphatidylcholine acyltransferase LPCAT1 functionally links specific genetic alterations in cancer with aberrant metabolism and plasma membrane remodeling to drive tumor growth. Growth factor receptor-driven cancers are found to depend on LPCAT1 to shape plasma membrane composition through enhanced saturated phosphatidylcholine content that is, in turn, required for the transduction of oncogenic signals. These results point to a genotype-informed strategy that prioritizes lipid remodeling pathways as therapeutic targets for diverse cancers.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Gene Amplification , Neoplasms/genetics , Neoplasms/metabolism , Oncogenes/genetics , Phospholipids/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , A549 Cells , Animals , Cell Survival/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Genotype , Heterografts , Humans , Mice , Mice, Nude , PC-3 Cells , Signal Transduction/genetics , Transfection
5.
Nature ; 569(7757): 570-575, 2019 05.
Article in English | MEDLINE | ID: mdl-31019297

ABSTRACT

Precision oncology hinges on linking tumour genotype with molecularly targeted drugs1; however, targeting the frequently dysregulated metabolic landscape of cancer has proven to be a major challenge2. Here we show that tissue context is the major determinant of dependence on the nicotinamide adenine dinucleotide (NAD) metabolic pathway in cancer. By analysing more than 7,000 tumours and 2,600 matched normal samples of 19 tissue types, coupled with mathematical modelling and extensive in vitro and in vivo analyses, we identify a simple and actionable set of 'rules'. If the rate-limiting enzyme of de novo NAD synthesis, NAPRT, is highly expressed in a normal tissue type, cancers that arise from that tissue will have a high frequency of NAPRT amplification and be completely and irreversibly dependent on NAPRT for survival. By contrast, tumours that arise from normal tissues that do not express NAPRT highly are entirely dependent on the NAD salvage pathway for survival. We identify the previously unknown enhancer that underlies this dependence. Amplification of NAPRT is shown to generate a pharmacologically actionable tumour cell dependence for survival. Dependence on another rate-limiting enzyme of the NAD synthesis pathway, NAMPT, as a result of enhancer remodelling is subject to resistance by NMRK1-dependent synthesis of NAD. These results identify a central role for tissue context in determining the choice of NAD biosynthetic pathway, explain the failure of NAMPT inhibitors, and pave the way for more effective treatments.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Amplification , NAD/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Cell Death , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/enzymology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism
6.
Cancer Cell ; 35(3): 504-518.e7, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30827889

ABSTRACT

Ionizing radiation (IR) and chemotherapy are standard-of-care treatments for glioblastoma (GBM) patients and both result in DNA damage, however, the clinical efficacy is limited due to therapeutic resistance. We identified a mechanism of such resistance mediated by phosphorylation of PTEN on tyrosine 240 (pY240-PTEN) by FGFR2. pY240-PTEN is rapidly elevated and bound to chromatin through interaction with Ki-67 in response to IR treatment and facilitates the recruitment of RAD51 to promote DNA repair. Blocking Y240 phosphorylation confers radiation sensitivity to tumors and extends survival in GBM preclinical models. Y240F-Pten knockin mice showed radiation sensitivity. These results suggest that FGFR-mediated pY240-PTEN is a key mechanism of radiation resistance and is an actionable target for improving radiotherapy efficacy.


Subject(s)
Brain Neoplasms/therapy , Cell Nucleus/metabolism , Glioma/therapy , PTEN Phosphohydrolase/metabolism , Pyrimidines/administration & dosage , Radiation Tolerance/drug effects , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Brain Neoplasms/metabolism , DNA Repair/drug effects , Female , Glioma/metabolism , Humans , Male , Mice , Phosphorylation/drug effects , Pyrimidines/pharmacology , Rad51 Recombinase/metabolism , Tyrosine/metabolism , Xenograft Model Antitumor Assays
7.
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
8.
J Vis Exp ; (134)2018 04 26.
Article in English | MEDLINE | ID: mdl-29757276

ABSTRACT

Tumorigenicity is the capability of cancer cells to form a tumor mass. A widely used approach to determine if the cells are tumorigenic is by injecting immunodeficient mice subcutaneously with cancer cells and measuring the tumor mass after it becomes visible and palpable. Orthotopic injections of cancer cells aim to introduce the xenograft in the microenvironment that most closely resembles the tissue of origin of the tumor being studied. Brain cancer research requires intracranial injection of cancer cells to allow the tumor formation and analysis in the unique microenvironment of the brain. The in vivo imaging of intracranial xenografts monitors instantaneously the tumor mass of orthotopically engrafted mice. Here we report the use of fluorescence molecular tomography (FMT) of brain tumor xenografts. The cancer cells are first transduced with near infrared fluorescent proteins and then injected in the brain of immunocompromised mice. The animals are then scanned to obtain quantitative information about the tumor mass over an extended period of time. Cell pre-labeling allows for cost effective, reproducible, and reliable quantification of the tumor burden within each mouse. We eliminated the need for injecting imaging substrates, and thus reduced the stress on the animals. A limitation of this approach is represented by the inability to detect very small masses; however, it has better resolution for larger masses than other techniques. It can be applied to evaluate the efficacy of a drug treatment or genetic alterations of glioma cell lines and patient-derived samples.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/physiology , Glioblastoma/diagnostic imaging , Tomography, X-Ray Computed/methods , Animals , Brain Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Fluorescence , Glioblastoma/pathology , Heterografts , Humans , Mice , Mice, Nude
10.
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
11.
Nat Commun ; 8: 15223, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497778

ABSTRACT

Glioblastoma (GBM) is the most lethal type of human brain cancer, where deletions and mutations in the tumour suppressor gene PTEN (phosphatase and tensin homolog) are frequent events and are associated with therapeutic resistance. Herein, we report a novel chromatin-associated function of PTEN in complex with the histone chaperone DAXX and the histone variant H3.3. We show that PTEN interacts with DAXX and, in turn PTEN directly regulates oncogene expression by modulating DAXX-H3.3 association on the chromatin, independently of PTEN enzymatic activity. Furthermore, DAXX inhibition specifically suppresses tumour growth and improves the survival of orthotopically engrafted mice implanted with human PTEN-deficient glioma samples, associated with global H3.3 genomic distribution changes leading to upregulation of tumour suppressor genes and downregulation of oncogenes. Moreover, DAXX expression anti-correlates with PTEN expression in GBM patient samples. Since loss of chromosome 10 and PTEN are common events in cancer, this synthetic growth defect mediated by DAXX suppression represents a therapeutic opportunity to inhibit tumorigenesis specifically in the context of PTEN deletion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chromatin/metabolism , Glioblastoma/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cells, Cultured , Chromatin/genetics , Co-Repressor Proteins , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Molecular Chaperones , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Protein Binding , RNA Interference , Transplantation, Heterologous
12.
Cancer Cell ; 30(5): 683-693, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27746144

ABSTRACT

Small-molecule inhibitors targeting growth factor receptors have failed to show efficacy for brain cancers, potentially due to their inability to achieve sufficient drug levels in the CNS. Targeting non-oncogene tumor co-dependencies provides an alternative approach, particularly if drugs with high brain penetration can be identified. Here we demonstrate that the highly lethal brain cancer glioblastoma (GBM) is remarkably dependent on cholesterol for survival, rendering these tumors sensitive to Liver X receptor (LXR) agonist-dependent cell death. We show that LXR-623, a clinically viable, highly brain-penetrant LXRα-partial/LXRß-full agonist selectively kills GBM cells in an LXRß- and cholesterol-dependent fashion, causing tumor regression and prolonged survival in mouse models. Thus, a metabolic co-dependency provides a pharmacological means to kill growth factor-activated cancers in the CNS.


Subject(s)
Brain Neoplasms/drug therapy , Cholesterol/metabolism , Glioblastoma/drug therapy , Indazoles/administration & dosage , Liver X Receptors/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Glioblastoma/metabolism , Humans , Indazoles/pharmacology , Mice , Treatment Outcome
13.
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
14.
Mol Cell ; 60(2): 307-18, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26455392

ABSTRACT

Epidermal growth factor receptor (EGFR) gene amplification and mutations are the most common oncogenic events in glioblastoma (GBM), but the mechanisms by which they promote aggressive tumor growth are not well understood. Here, through integrated epigenome and transcriptome analyses of cell lines, genotyped clinical samples, and TCGA data, we show that EGFR mutations remodel the activated enhancer landscape of GBM, promoting tumorigenesis through a SOX9 and FOXG1-dependent transcriptional regulatory network in vitro and in vivo. The most common EGFR mutation, EGFRvIII, sensitizes GBM cells to the BET-bromodomain inhibitor JQ1 in a SOX9, FOXG1-dependent manner. These results identify the role of transcriptional/epigenetic remodeling in EGFR-dependent pathogenesis and suggest a mechanistic basis for epigenetic therapy.


Subject(s)
Brain Neoplasms/genetics , Epigenesis, Genetic , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Glioblastoma/genetics , Nerve Tissue Proteins/genetics , SOX9 Transcription Factor/genetics , Adult , Animals , Azepines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , ErbB Receptors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcriptome , Triazoles/pharmacology
15.
Cell ; 160(3): 489-502, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619690

ABSTRACT

Protein kinase C (PKC) isozymes have remained elusive cancer targets despite the unambiguous tumor promoting function of their potent ligands, phorbol esters, and the prevalence of their mutations. We analyzed 8% of PKC mutations identified in human cancers and found that, surprisingly, most were loss of function and none were activating. Loss-of-function mutations occurred in all PKC subgroups and impeded second-messenger binding, phosphorylation, or catalysis. Correction of a loss-of-function PKCß mutation by CRISPR-mediated genome editing in a patient-derived colon cancer cell line suppressed anchorage-independent growth and reduced tumor growth in a xenograft model. Hemizygous deletion promoted anchorage-independent growth, revealing that PKCß is haploinsufficient for tumor suppression. Several mutations were dominant negative, suppressing global PKC signaling output, and bioinformatic analysis suggested that PKC mutations cooperate with co-occurring mutations in cancer drivers. These data establish that PKC isozymes generally function as tumor suppressors, indicating that therapies should focus on restoring, not inhibiting, PKC activity.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/genetics , Animals , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Genes, Tumor Suppressor , Heterografts , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice, Nude , Models, Molecular , Mutation , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase C/metabolism , Protein Structure, Tertiary
16.
J Cell Physiol ; 229(10): 1359-68, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24477641

ABSTRACT

PED/PEA-15 is a death effector domain (DED) family member with a variety of effects on cell growth and metabolism. To get further insight into the role of PED in cancer, we aimed to find new PED interactors. Using tandem affinity purification, we identified HSC70 (Heat Shock Cognate Protein of 70 kDa)-which, among other processes, is involved in chaperone-mediated autophagy (CMA)-as a PED-interacting protein. We found that PED has two CMA-like motifs (i.e., KFERQ), one of which is located within a phosphorylation site, and demonstrate that PED is a bona fide CMA substrate and the first example in which phosphorylation modifies the ability of HSC70 to access KFERQ-like motifs and target the protein for lysosomal degradation. Phosphorylation of PED switches its function from tumor suppression to tumor promotion, and we show that HSC70 preferentially targets the unphosphorylated form of PED to CMA. Therefore, we propose that the up-regulated CMA activity characteristic of most types of cancer cell enhances oncogenesis by shifting the balance of PED function toward tumor promotion. This mechanism is consistent with the notion of a therapeutic potential for targeting CMA in cancer, as inhibition of this autophagic pathway may help restore a physiological ratio of PED forms.


Subject(s)
Autophagy , HSC70 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins , Cell Line, Tumor , HEK293 Cells , HSC70 Heat-Shock Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lysosomes/metabolism , Male , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Protein Transport , Proteolysis , RNA Interference , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Suppressor Proteins/genetics
17.
Cancer Res ; 74(5): 1429-39, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24436148

ABSTRACT

The EGF receptor (EGFR) is amplified and mutated in glioblastoma, in which its common mutation (ΔEGFR, also called EGFRvIII) has a variety of activities that promote growth and inhibit death, thereby conferring a strong tumor-enhancing effect. This range of activities suggested to us that ΔEGFR might exert its influence through pleiotropic effectors, and we hypothesized that microRNAs might serve such a function. Here, we report that ΔEGFR specifically suppresses one such microRNA, namely miR-9, through the Ras/PI3K/AKT axis that it is known to activate. Correspondingly, expression of miR-9 antagonizes the tumor growth advantage conferred by ΔEGFR. Silencing of FOXP1, a miR-9 target, inhibits ΔEGFR-dependent tumor growth and, conversely, de-repression of FOXP1, as a consequence of miR-9 inhibition, increases tumorigenicity. FOXP1 was sufficient to increase tumor growth in the absence of oncogenic ΔEGFR signaling. The significance of these findings is underscored by our finding that high FOXP1 expression predicts poor survival in a cohort of 131 patients with glioblastoma. Collectively, these data suggest a novel regulatory mechanism by which ΔEGFR suppression of miR-9 upregulates FOXP1 to increase tumorigenicity.


Subject(s)
Carcinogenesis/genetics , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , MicroRNAs/genetics , Signal Transduction/genetics , Animals , Carcinogenesis/pathology , Humans , Mice , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Tumor Cells, Cultured , Up-Regulation , ras Proteins/genetics
18.
Cancer Biol Med ; 10(4): 192-205, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24349829

ABSTRACT

Receptor tyrosine kinases (RTKs) such as the epidermal growth factor receptor (EGFR) regulate cellular homeostatic processes. EGFR activates downstream signaling cascades that promote tumor cell survival, proliferation and migration. Dysregulation of EGFR signaling as a consequence of overexpression, amplification and mutation of the EGFR gene occurs frequently in several types of cancers and many become dependent on EGFR signaling to maintain their malignant phenotypes. Consequently, concerted efforts have been mounted to develop therapeutic agents and strategies to effectively inhibit EGFR. However, limited therapeutic benefits to cancer patients have been derived from EGFR-targeted therapies. A well-documented obstacle to improved patient survival is the presence of EGFR-inhibitor resistant tumor cell variants within heterogeneous tumor cell masses. Here, we summarize the mechanisms by which tumors resist EGFR-targeted therapies and highlight the emerging role of microRNAs (miRs) as downstream effector molecules utilized by EGFR to promote tumor initiation, progression and that play a role in resistance to EGFR inhibitors. We also examine evidence supporting the utility of miRs as predictors of response to targeted therapies and novel therapeutic agents to circumvent EGFR-inhibitor resistance mechanisms.

19.
Clin Cancer Res ; 19(20): 5722-32, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24030701

ABSTRACT

PURPOSE: mTOR pathway hyperactivation occurs in approximately 90% of glioblastomas, but the allosteric mTOR inhibitor rapamycin has failed in the clinic. Here, we examine the efficacy of the newly discovered ATP-competitive mTOR kinase inhibitors CC214-1 and CC214-2 in glioblastoma, identifying molecular determinants of response and mechanisms of resistance, and develop a pharmacologic strategy to overcome it. EXPERIMENTAL DESIGN: We conducted in vitro and in vivo studies in glioblastoma cell lines and an intracranial model to: determine the potential efficacy of the recently reported mTOR kinase inhibitors CC214-1 (in vitro use) and CC214-2 (in vivo use) at inhibiting rapamycin-resistant signaling and blocking glioblastoma growth and a novel single-cell technology-DNA Encoded Antibody Libraries-was used to identify mechanisms of resistance. RESULTS: Here, we show that CC214-1 and CC214-2 suppress rapamycin-resistant mTORC1 signaling, block mTORC2 signaling, and significantly inhibit the growth of glioblastomas in vitro and in vivo. EGFRvIII expression and PTEN loss enhance sensitivity to CC214 compounds, consistent with enhanced efficacy in strongly mTOR-activated tumors. Importantly, CC214 compounds potently induce autophagy, preventing tumor cell death. Genetic or pharmacologic inhibition of autophagy greatly sensitizes glioblastoma cells and orthotopic xenografts to CC214-1- and CC214-2-induced cell death. CONCLUSIONS: These results identify CC214-1 and CC214-2 as potentially efficacious mTOR kinase inhibitors in glioblastoma, and suggest a strategy for identifying patients most likely to benefit from mTOR inhibition. In addition, this study also shows a central role for autophagy in preventing mTOR-kinase inhibitor-mediated tumor cell death, and suggests a pharmacologic strategy for overcoming it.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glioblastoma/drug therapy , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/metabolism , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Carcinogenesis ; 34(4): 725-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455378

ABSTRACT

Chemotherapy and molecularly targeted approaches represent two very different modes of cancer treatment and each is associated with unique benefits and limitations. Both types of therapy share the overarching limitation of the emergence of drug resistance, which prevents these drugs from eliciting lasting clinical benefit. This review will provide an overview of the various mechanisms of resistance to each of these classes of drugs and examples of drug combinations that have been tested clinically. This analysis supports the contention that understanding modes of resistance to both chemotherapy and molecularly targeted therapies may be very useful in selecting those drugs of each class that will have complementing mechanisms of sensitivity and thereby represent reasonable combination therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Precision Medicine
SELECTION OF CITATIONS
SEARCH DETAIL
...