Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1208: 79-98, 2021.
Article in English | MEDLINE | ID: mdl-34260023

ABSTRACT

Autophagy is a lysosome-dependent degradation process. During autophagy, cytoplasmic components are sequestered and catabolized to supply nutrition and energy under starvation conditions. Recent work has demonstrated that many cargos can be specifically recognized and then eliminated via the core mechanism of autophagy which is termed as selective autophagy. The cargo recognition program provides the basis for the specific degradation of selective autophagy; thus, the exploration of the interaction between the cargo and the receptor is the key for revealing the underlying mechanism. Also, receptor protein complexes are required in various selective autophagy subtypes which process and guide the cargo to the core mechanism. Ubiquitination and phosphorylation are the main methods to modulate the affinity of the receptor toward cargo. Although many key processes of selective autophagy subtypes have been discovered and intensively studied, the precise ways in which the mechanisms of cargo recognition function remain mostly elusive. A fuller mechanistic understanding of selective autophagy will be important for efforts to promote disease treatment and drug development.


Subject(s)
Autophagy , Lysosomes , Carrier Proteins , Cytosol , Ubiquitination
2.
Cancer Res ; 80(4): 709-718, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31806641

ABSTRACT

The mTOR signaling is dysregulated prominently in human cancers including glioblastoma, suggesting mTOR as a robust target for therapy. Inhibitors of mTOR have had limited success clinically, however, in part because their mechanism of action is cytostatic rather than cytotoxic. Here, we tested three distinct mTOR kinase inhibitors (TORKi) PP242, KU-0063794, and sapanisertib against glioblastoma cells. All agents similarly decreased proliferation of glioblastoma cells, whereas PP242 uniquely induced apoptosis. Apoptosis induced by PP242 resulted from off-target cooperative inhibition of JAK2 and protein kinase C alpha (PKCα). Induction of apoptosis was also decreased by additional on-target inhibition of mTOR, due to induction of autophagy. As EGFR inhibitors can block PKCα, EGFR inhibitors erlotinib and osimertinib were tested separately in combination with the JAK2 inhibitor AZD1480. Combination therapy induced apoptosis of glioblastoma tumors in both flank and in patient-derived orthotopic xenograft models, providing a preclinical rationale to test analogous combinations in patients. SIGNIFICANCE: These findings identify PKCα and JAK2 as targets that drive apoptosis in glioblastoma, potentially representing a clinically translatable approach for glioblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Acrylamides/pharmacology , Acrylamides/therapeutic use , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Female , Glioblastoma/pathology , Humans , Indoles/pharmacology , Indoles/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Mice , Morpholines/pharmacology , Morpholines/therapeutic use , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
4.
Cancer Res ; 78(24): 6785-6794, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30401716

ABSTRACT

: Amplification of the EGFR gene and its truncation mutant EGFRvIII are hallmarks of glioblastoma. Although coexpression of EGFR and EGFRvIII confers a growth advantage, how EGFR and EGFRvIII influence the tumor microenvironment remains incompletely understood. Here, we show that EGFR and EGFRvIII cooperate to induce macrophage infiltration via upregulation of the chemokine CCL2. EGFRvIII was significantly enriched in glioblastoma patient samples with high CCL2, and knockout of CCL2 in tumors coexpressing EGFR and EGFRvIII led to decreased infiltration of macrophages. KRAS was a critical signaling intermediate for EGFR- and EGFRvIII-induced expression of CCL2. Our results illustrate how EGFR and EGFRvIII direct the microenvironment in glioblastoma. SIGNIFICANCE: Full-length EGFR and truncated EGFRvIII work through KRAS to upregulate the chemokine CCL2 and drive macrophage infiltration in glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Chemokine CCL2/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , Macrophages/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Microglia/metabolism , Neoplasm Transplantation , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Microenvironment , Up-Regulation
5.
Oncogene ; 37(12): 1561-1575, 2018 03.
Article in English | MEDLINE | ID: mdl-29321659

ABSTRACT

Amplification of epidermal growth factor receptor (EGFR) and its active mutant EGFRvIII occurs frequently in glioblastoma (GBM). While EGFR and EGFRvIII play critical roles in pathogenesis, targeted therapy with EGFR-tyrosine kinase inhibitors (TKIs) or antibodies has only shown limited efficacy in patients. Here we discuss signaling pathways mediated by EGFR/EGFRvIII, current therapeutics, and novel strategies to target EGFR/EGFRvIII-amplified GBM.


Subject(s)
Brain Neoplasms/therapy , ErbB Receptors/physiology , Glioblastoma/therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Brain Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , Glioblastoma/genetics , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Signal Transduction/drug effects , Signal Transduction/genetics
6.
Clin Cancer Res ; 24(1): 14-21, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28696243

ABSTRACT

Glioblastoma is the most common and aggressive adult brain cancer. Tumors show frequent dysregulation of the PI3K-mTOR pathway. Although a number of small molecules target the PI3K-AKT-mTOR axis, their preclinical and clinical efficacy has been limited. Reasons for treatment failure include poor penetration of agents into the brain and observations that blockade of PI3K or AKT minimally affects downstream mTOR activity in glioma. Clinical trials using allosteric mTOR inhibitors (rapamycin and rapalogs) to treat patients with glioblastoma have also been unsuccessful or uncertain, in part, because rapamycin inefficiently blocks the mTORC1 target 4EBP1 and feeds back to activate PI3K-AKT signaling. Inhibitors of the mTOR kinase (TORKi) such as TAK-228/MLN0128 interact orthosterically with the ATP- and substrate-binding pocket of mTOR kinase, efficiently block 4EBP1 in vitro, and are currently being investigated in the clinical trials. Preclinical studies suggest that TORKi have poor residence times of mTOR kinase, and our data suggest that this poor pharmacology translates into disappointing efficacy in glioblastoma xenografts. RapaLink-1, a TORKi linked to rapamycin, represents a drug with improved pharmacology against 4EBP1. In this review, we clarify the importance of 4EBP1 as a biomarker for the efficacy of PI3K-AKT-mTOR inhibitors in glioblastoma. We also review mechanistic data by which RapaLink-1 blocks p-4EBP1 and discuss future clinical strategies for 4EBP1 inhibition in glioblastoma. Clin Cancer Res; 24(1); 14-21. ©2017 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Biomarkers, Tumor , Glioblastoma/metabolism , Phosphoproteins/antagonists & inhibitors , Animals , Cell Cycle Proteins , Cell Proliferation , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
J Neurooncol ; 131(3): 495-505, 2017 02.
Article in English | MEDLINE | ID: mdl-27848137

ABSTRACT

BRAFV600E is a common finding in glioma (about 10-60% depending on histopathologic subclassification). BRAFV600E monotherapy shows modest preclinical efficacy against BRAFV600E gliomas and also induces adverse secondary skin malignancies. Here, we examine the molecular mechanism of intrinsic resistance to BRAFV600E inhibition in glioma. Furthermore, we investigate BRAFV600E/MEK combination therapy that overcomes intrinsic resistance to BRAFV600E inhibitor and also prevents BRAFV600E inhibitor induced secondary malignancies. Immunoblotting and Human Phospho-Receptor Tyrosine Kinase Array assays were used to interrogate MAPK pathway activation. The cellular effect of BRAFV600E and MEK inhibition was determined by WST-1 viability assay and cell cycle analysis. Flanked and orthotopic GBM mouse models were used to investigate the in vivo efficacy of BRAFV600E/MEK combination therapy and the effect on secondary malignancies. BRAFV600E inhibition leads to recovery of ERK phosphorylation. Combined BRAFV600E and MEK inhibition prevents reactivation of the MAPK signaling, which correlates with decreased cell viability and augmented cell cycle arrest. Similarly, mice bearing BRAFV600E glioma showed reduced tumor growth when treated with a combination of BRAFV600E and MEK inhibitor compared to BRAFV600E inhibition alone. Additional benefit of BRAFV600E/MEK inhibition was reflected by reduced cutaneous squamous-cell carcinoma (cSCC) growth (a surrogate for RAS-driven secondary maligancies). In glioma, recovery of MAPK signaling upon BRAF inhibition accounts for intrinsic resistance to BRAFV600E inhibitor. Combined BRAFV600E and MEK inhibition prevents rebound of MAPK activation, resulting in enhanced antitumor efficacy and also reduces the risk of secondary malignancy development.


Subject(s)
Antineoplastic Agents/administration & dosage , Glioma/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Benzamides/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Diphenylamine/administration & dosage , Diphenylamine/analogs & derivatives , Disease Models, Animal , Female , Glioma/drug therapy , Glioma/genetics , Humans , Indoles/administration & dosage , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mutation , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Survival Analysis , Xenograft Model Antitumor Assays
8.
Cancer Cell ; 24(4): 438-49, 2013 Oct 14.
Article in English | MEDLINE | ID: mdl-24135280

ABSTRACT

EGFRvIII, a frequently occurring mutation in primary glioblastoma, results in a protein product that cannot bind ligand, but signals constitutively. Deducing how EGFRvIII causes transformation has been difficult because of autocrine and paracrine loops triggered by EGFRvIII alone or in heterodimers with wild-type EGFR. Here, we document coexpression of EGFR and EGFRvIII in primary human glioblastoma that drives transformation and tumorigenesis in a cell-intrinsic manner. We demonstrate enhancement of downstream STAT signaling triggered by EGFR-catalyzed phosphorylation of EGFRvIII, implicating EGFRvIII as a substrate for EGFR. Subsequent phosphorylation of STAT3 requires nuclear entry of EGFRvIII and formation of an EGFRvIII-STAT3 nuclear complex. Our findings clarify specific oncogenic signaling relationships between EGFR and EGFRvIII in glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Alleles , Cell Line, Tumor , Cell Nucleus/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasm Transplantation , Phosphorylation , Signal Transduction
9.
Sci Signal ; 6(283): ra55, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23838182

ABSTRACT

The gene that encodes the epidermal growth factor receptor (EGFR) is frequently overexpressed or mutated in human cancers, including glioblastoma. However, the efficacy of EGFR-targeted small-molecule inhibitors or monoclonal antibodies in glioblastomas that also have mutation or deletion of the gene encoding phosphatase and tensin homolog (PTEN) has been modest. We found that EGFR signaling was blocked by a small molecule (G5-7) that selectively inhibited Janus kinase 2 (JAK2)-mediated phosphorylation and activation of EGFR and STAT3 (signal transducer and activator of transcription 3) by binding to JAK2, thereby decreasing the activity of downstream signaling by mTOR (mammalian target of rapamycin) and inducing cell cycle arrest. G5-7 inhibited the proliferation of PTEN-deficient glioblastoma cell lines harboring a constitutively active variant of EGFR (U87MG/EGFRvIII) and human glioblastoma explant neurosphere cultures, but the drug only weakly inhibited the proliferation of either glioblastoma cell lines that were wild type for EGFR and stably transfected with PTEN (U87MG/PTEN) or normal neural progenitor cells and astrocytes. Additionally, G5-7 reduced vascular endothelial growth factor (VEGF) secretion and endothelial cell migration and induced apoptosis in glioblastoma xenografts, thereby suppressing glioblastoma growth in vivo. Furthermore, G5-7 was more potent than EGFR or JAK2 inhibitors that interfere with either ligand or adenosine 5'-triphosphate (ATP) binding at impeding glioblastoma cell proliferation, demonstrating that this allosteric JAK2 inhibitor may be an effective clinical strategy.


Subject(s)
Cell Proliferation/drug effects , Glioma/drug therapy , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Deletion , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Heterografts , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Kinase Inhibitors/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
10.
Proc Natl Acad Sci U S A ; 109(31): 12722-7, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802621

ABSTRACT

Malignant glioma, the most common primary brain tumor, is generally incurable. Although phosphatidylinositol-3-kinase (PI3K) signaling features prominently in glioma, inhibitors generally block proliferation rather than induce apoptosis. Starting with an inhibitor of both lipid and protein kinases that induced prominent apoptosis and that failed early clinical development because of its broad target profile and overall toxicity, we identified protein kinase targets, the blockade of which showed selective synthetic lethality when combined with PI3K inhibitors. Prioritizing protein kinase targets for which there are clinical inhibitors, we demonstrate that cyclin-dependent kinase (CDK)1/2 inhibitors, siRNAs against CDK1/2, and the clinical CDK1/2 inhibitor roscovitine all cooperated with the PI3K inhibitor PIK-90, blocking the antiapoptotic protein Survivin and driving cell death. In addition, overexpression of CDKs partially blocked some of the apoptosis caused by PIK-75. Roscovitine and PIK-90, in combination, were well tolerated in vivo and acted in a synthetic-lethal manner to induce apoptosis in human glioblastoma xenografts. We also tested clinical Akt and CDK inhibitors, demonstrating induction of apoptosis in vitro and providing a preclinical rationale to test this combination therapy in patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Glioma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Female , Glioma/enzymology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Roscovitine , Xenograft Model Antitumor Assays
11.
Cancer Discov ; 2(5): 450-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22588882

ABSTRACT

UNLABELLED: Although mutational activation of the epidermal growth factor receptor (EGFR) features prominently in glioma and non-small cell lung cancer (NSCLC), inhibitors of EGFR improve survival only in patients with NCSLC. To understand how mutations in EGFR influence response to therapy, we generated glioma cells expressing either glioma- or NSCLC-derived alleles and quantified kinase-site occupancy by clinical inhibitors with the use of a novel affinity probe and kinetic methodology. At equivalent doses, erlotinib achieved lower kinase-site occupancy in glioma-derived EGFRvIII compared with NSCLC-derived EGFR mutants. Kinase-site occupancy correlated directly with cell-cycle arrest. EGFRvIII released erlotinib rapidly compared with wild-type EGFR, whereas NSCLC-derived mutants released erlotinib slowly. SIGNIFICANCE: These data suggest that kinase-site occupancy is a biomarker for efficacy of EGFR inhibitors, that rapid binding and release of erlotinib in glioma-derived EGFRvIII opposes the blockade of downstream signaling, and that slower cycling of erlotinib within the active site of NSCLC-derived mutants underlies their improved clinical response.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Glioma/metabolism , Humans , Kinetics
12.
Methods Mol Biol ; 821: 349-59, 2012.
Article in English | MEDLINE | ID: mdl-22125077

ABSTRACT

Amplification of the gene encoding the epidermal growth factor receptor (EGFR) occurs commonly in glioblastoma (GBM), leading to activation of downstream kinases, including phosphatidylinositol 3'-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR). A serine-threonine kinase, mTOR controls cell growth by regulating mRNA translation, metabolism, and autophagy; acting as both a downstream effector and upstream regulator of PI3K. These signaling functions are distributed between at least two distinct complexes, mTORC1 and mTORC2 with respect to pathway specificity. We have investigated mTOR signaling in glioma cells with the allosteric mTORC1 inhibitor rapamycin, the mTORC1/2 inhibitor Ku-0063794, a dual PI3K/mTORC1/2 kinase inhibitor PI-103, and siRNA against raptor, rictor, or mTOR, and evaluated the value of mTOR inhibitors for the treatment of glioblastoma.


Subject(s)
Glioblastoma/enzymology , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , ErbB Receptors/metabolism , Furans/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1 , Morpholines/pharmacology , Multiprotein Complexes , Oncogene Protein v-akt/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
13.
Autophagy ; 7(5): 536-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21266843

ABSTRACT

Signaling through phosphatidylinositol 3-kinase (PtdIns3K)-Akt-mTOR is frequently activated in cancers including glioblastoma multiforme (GBM), where this kinase network regulates survival. It is thus surprising that inhibitors of these pathways induce minimal cell death in glioma. We showed that the dual PtdIns3K-mTOR inhibitor PI-103 induces autophagy in therapy-resistant, PTEN-mutant glioma, with blockade of mTOR complex 1 (mTORC1) and complex 2 (mTORC2) contributing independently to autophagy. Inhibition of autophagosome maturation synergizes with PI-103 to induce apoptosis through the Bax-dependent intrinsic mitochondrial pathway, indicating that PI-103 induces autophagy as a survival pathway in this setting. Not all inhibitors of PtdIns3K-Akt-mTOR signaling synergize with inhibitors of autophagy. The allosteric mTORC1 inhibitor rapamycin fails to induce apoptosis in conjunction with blockade of autophagy, due to feedback-activation of Akt. Apoptosis in the setting of rapamycin therapy requires concurrent inhibition of both autophagy and of PtdIns3K-Akt. Moreover, the clinical PtdIns3K-mTOR inhibitor NVP-BEZ235 cooperates with the clinical lysosomotropic autophagy inhibitor chloroquine to induce apoptosis in PTEN-mutant glioma xenografts in vivo, offering a therapeutic approach translatable to patients.


Subject(s)
Autophagy/physiology , Brain Neoplasms/pathology , Glioma/pathology , Oncogene Protein v-akt/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Brain Neoplasms/genetics , Cell Survival/drug effects , Cell Survival/genetics , Glioma/genetics , Humans , Models, Biological , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
14.
Sci Signal ; 3(147): ra81, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21062993

ABSTRACT

Although the phosphatidylinositol 3-kinase to Akt to mammalian target of rapamycin (PI3K-Akt-mTOR) pathway promotes survival signaling, inhibitors of PI3K and mTOR induce minimal cell death in PTEN (phosphatase and tensin homolog deleted from chromosome 10) mutant glioma. Here, we show that the dual PI3K-mTOR inhibitor PI-103 induces autophagy in a form of glioma that is resistant to therapy. Inhibitors of autophagosome maturation cooperated with PI-103 to induce apoptosis through the mitochondrial pathway, indicating that the cellular self-digestion process of autophagy acted as a survival signal in this setting. Not all inhibitors of mTOR synergized with inhibitors of autophagy. Rapamycin delivered alone induced autophagy, yet cells survived inhibition of autophagosome maturation because of rapamycin-mediated activation of Akt. In contrast, adenosine 5'-triphosphate-competitive inhibitors of mTOR stimulated autophagy more potently than did rapamycin, with inhibition of mTOR complexes 1 and 2 contributing independently to induction of autophagy. We show that combined inhibition of PI3K and mTOR, which activates autophagy without activating Akt, cooperated with inhibition of autophagy to cause glioma cells to undergo apoptosis. Moreover, the PI3K-mTOR inhibitor NVP-BEZ235, which is in clinical use, synergized with the lysosomotropic inhibitor of autophagy, chloroquine, another agent in clinical use, to induce apoptosis in glioma xenografts in vivo, providing a therapeutic approach potentially translatable to humans.


Subject(s)
Autophagy/drug effects , Furans/pharmacology , Glioma/drug therapy , Glioma/metabolism , Oncogene Protein v-akt/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Synergism , Flow Cytometry , Glioma/genetics , Histological Techniques , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Mitochondria/metabolism , Mutation/genetics , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Quinolines/metabolism , Quinolines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transplantation, Heterologous
15.
Curr Top Microbiol Immunol ; 347: 279-96, 2010.
Article in English | MEDLINE | ID: mdl-20535652

ABSTRACT

Gliomas represent the most common primary brain tumor and among the most aggressive of cancers. Patients with glioma typically relapse within a year of initial diagnosis. Recurrent glioma is associated with acquired therapeutic resistance. Although neurosurgical resection, radiation and chemotherapy provide clear benefit, survival remains disappointing. It is, therefore, critical that we identify effective medical therapies and appropriate tumor biomarkers in patients at initial presentation, to promote durable responses in glioma. Pathways linking receptor tyrosine kinases, PI3 kinase, Akt, and mTOR feature prominently in this disease and represent therapeutic targets. Small molecules that inhibit one or more of these kinases are now being introduced into the clinic and may have some activity. Disappointingly, however, preclinical studies demonstrate these agents to be primarily cytostatic rather than cytotoxic to glioma cells. Here, we detail activation of the EGFR-PI3K-Akt-mTOR signaling network in glioma, review class I PI3K inhibitors, discuss roles for Akt, PKC and mTOR, and the importance of biomarkers. We further delineate attempts to target both single and multiple components within the EGFR-PI3K-Akt-mTOR axes. Lastly, we discuss the need to combine targeted therapies with cytotoxic chemotherapy, radiation and with inhibitors of survival signaling to improve outcomes in glioma.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm , Receptor, EphB3
16.
Genes Dev ; 24(10): 1059-72, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20478998

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Sonic Hedgehog (SHH) signaling drives a minority of MB, correlating with desmoplastic pathology and favorable outcome. The majority, however, arises independently of SHH and displays classic or large cell anaplastic (LCA) pathology and poor prognosis. To identify common signaling abnormalities, we profiled mRNA, demonstrating misexpression of MYCN in the majority of human MB and negligible expression in normal cerebella. We clarified a role in pathogenesis by targeting MYCN (and luciferase) to cerebella of transgenic mice. MYCN-driven MB showed either classic or LCA pathologies, with Shh signaling activated in approximately 5% of tumors, demonstrating that MYCN can drive MB independently of Shh. MB arose at high penetrance, consistent with a role for MYCN in initiation. Tumor burden correlated with bioluminescence, with rare metastatic spread to the leptomeninges, suggesting roles for MYCN in both progression and metastasis. Transient pharmacological down-regulation of MYCN led to both clearance and senescence of tumor cells, and improved survival. Targeted expression of MYCN thus contributes to initiation, progression, and maintenance of MB, suggesting a central role for MYCN in pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Medulloblastoma/physiopathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cell Cycle/physiology , Cellular Senescence/physiology , Cerebellum/metabolism , Down-Regulation , Gene Expression Profiling , Genomic Instability , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/pathology , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics
17.
Sci Signal ; 2(55): ra4, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19176518

ABSTRACT

Amplification of the gene encoding the epidermal growth factor (EGF) receptor (EGFR) occurs commonly in glioblastoma, leading to activation of downstream kinases including phosphatidylinositol 3'-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR). Here, we show that phosphorylation of mTOR and its downstream substrate rpS6 (ribosomal protein S6) are robust biomarkers for the antiproliferative effect of EGFR inhibitors. Inhibition of EGFR signaling correlated with decreased abundance of phosphorylated mTOR (p-mTOR) and rpS6 (p-rpS6) in cells wild type for the gene encoding PTEN (phosphatase and tensin homolog on chromosome 10), a negative regulator of PI3K. In contrast, inhibition of EGFR signaling failed to affect p-mTOR or p-rpS6 in cells mutant for PTEN, which are resistant to EGFR inhibitors. Although the abundance of phosphorylated Akt (p-Akt) decreased in response to inhibition of EGFR signaling, Akt was dispensable for signaling between EGFR and mTOR. We identified an Akt-independent pathway linking EGFR to mTOR that was critically dependent on protein kinase C (PKC). Consistent with these observations, the abundance of EGFR generally correlated with phosphorylation of rpS6 and PKC in primary human glioblastoma tumors, and correlated poorly with phosphorylation of Akt. Inhibition of PKC led to decreased viability of glioma cells regardless of PTEN or EGFR status, suggesting that PKC inhibitors should be tested in glioma. These findings underline the importance of signaling between EGFR and mTOR in glioma, identify PKCalpha as essential to this network, and question the necessity of Akt as a critical intermediate coupling EGFR and mTOR in glioma.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Protein Kinase C/metabolism , Protein Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/pharmacology , Humans , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
18.
Brain Pathol ; 19(1): 112-20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19076776

ABSTRACT

The PI3 kinase (PI3K) family plays a complex role in cell biology and metabolism. Signaling through the PI3Ks is frequently activated in many human cancers, including glioblastoma, because of gain-of-function mutations in PIK3CA or loss of PTEN. Experiments involving genetic mouse models and small molecule inhibitors have helped to elucidate the roles of the regulatory and catalytic subunits of PI3K in metabolism and cancer. Downstream of PI3K is Akt, a critical effector of growth, proliferation and survival. The suggested dependence of glioblastoma tumors on PI3K signaling implies that PI3K inhibitors should lead to effective killing of these cancer cells, but that has been shown not to be the case. The engagement of other survival pathways in response to PI3K inhibition prompts the need to develop combination therapies that promote cytotoxicity in cancer cells.


Subject(s)
Disease Models, Animal , Glioma/therapy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Class I Phosphatidylinositol 3-Kinases , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/therapy , Glioma/genetics , Glioma/metabolism , Humans , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics
19.
EMBO Rep ; 9(9): 923-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18660750

ABSTRACT

In multicellular epithelial tissues, the orientation of polarity of each cell must be coordinated. Previously, we reported that for Madin-Darby canine kidney cells in three-dimensional collagen gel culture, blockade of beta1-integrin by the AIIB2 antibody or expression of dominant-negative Rac1N17 led to an inversion of polarity, such that the apical surfaces of the cells were misorientated towards the extracellular matrix. Here, we show that this process results from the activation of RhoA. Knockdown of RhoA by short hairpin RNA reverses the inverted orientation of polarity, resulting in normal cysts. Inhibition of RhoA downstream effectors, Rho kinase (ROCK I) and myosin II, has similar effects. We conclude that the RhoA-ROCK I-myosin II pathway controls the inversion of orientation of epithelial polarity caused by AIIB2 or Rac1N17. These results might be relevant to the hyperactivation of RhoA and disruption of normal polarity frequently observed in human epithelial cancers.


Subject(s)
Epithelial Cells/metabolism , Myosin Type II/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Blotting, Western , Cell Line , Cell Polarity , Epithelial Cells/cytology , Humans , Myosin Type II/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
20.
Cancer Res ; 67(17): 7960-5, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804702

ABSTRACT

We have shown previously that blockade of epidermal growth factor receptor (EGFR) cooperates with a pan-selective inhibitor of phosphoinositide-3-kinase (PI3K) in EGFR-driven glioma. In this communication, we tested EGFR-driven glioma differing in PTEN status, treating with the EGFR inhibitor erlotinib and a novel dual inhibitor of PI3Kalpha and mTOR (PI-103). Erlotinib blocked proliferation only in PTEN(wt) cells expressing EGFR. Although erlotinib monotherapy showed little effect in PTEN(mt) glioma, PI-103 greatly augmented the antiproliferative efficacy of erlotinib in this setting. To address the importance of PI3K blockade, we showed in PTEN(mt) glioma that combining PI-103 and erlotinib was superior to either monotherapy or to therapy combining erlotinib with either rapamycin (an inhibitor of mTOR) or PIK-90 (an inhibitor of PI3Kalpha). These experiments show that a dual inhibitor of PI3Kalpha and mTOR augments the activity of EGFR blockade, offering a mechanistic rationale for targeting EGFR, PI3Kalpha, and mTOR in the treatment of EGFR-driven, PTEN-mutant glioma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , ErbB Receptors/antagonists & inhibitors , Furans/pharmacology , Glioma/drug therapy , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Cell Cycle/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Erlotinib Hydrochloride , Furans/administration & dosage , Glioma/genetics , Glioma/pathology , Humans , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Quinazolines/administration & dosage , TOR Serine-Threonine Kinases , Treatment Outcome , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...