Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Nat Genet ; 56(1): 60-73, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38049664

ABSTRACT

In this study, the impact of the apolipoprotein B mRNA-editing catalytic subunit-like (APOBEC) enzyme APOBEC3B (A3B) on epidermal growth factor receptor (EGFR)-driven lung cancer was assessed. A3B expression in EGFR mutant (EGFRmut) non-small-cell lung cancer (NSCLC) mouse models constrained tumorigenesis, while A3B expression in tumors treated with EGFR-targeted cancer therapy was associated with treatment resistance. Analyses of human NSCLC models treated with EGFR-targeted therapy showed upregulation of A3B and revealed therapy-induced activation of nuclear factor kappa B (NF-κB) as an inducer of A3B expression. Significantly reduced viability was observed with A3B deficiency, and A3B was required for the enrichment of APOBEC mutation signatures, in targeted therapy-treated human NSCLC preclinical models. Upregulation of A3B was confirmed in patients with NSCLC treated with EGFR-targeted therapy. This study uncovers the multifaceted roles of A3B in NSCLC and identifies A3B as a potential target for more durable responses to targeted cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Up-Regulation/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Cytidine Deaminase/genetics , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism
2.
Front Res Metr Anal ; 6: 748095, 2021.
Article in English | MEDLINE | ID: mdl-35005422

ABSTRACT

The current digital content industry is heavily oriented towards building platforms that track users' behaviour and seek to convince them to stay longer and come back sooner onto the platform. Similarly, authors are incentivised to publish more and to become champions of dissemination. Arguably, these incentive systems are built around public reputation supported by a system of metrics, hard to be assessed. Generally, the digital content industry is permeable to non-human contributors (algorithms that are able to generate content and reactions), anonymity and identity fraud. It is pertinent to present a perspective paper about early signs of track and persuasion in scholarly communication. Building our views, we have run a pilot study to determine the opportunity for conducting research about the use of "track and persuade" technologies in scholarly communication. We collected observations on a sample of 148 relevant websites and we interviewed 15 that are experts related to the field. Through this work, we tried to identify 1) the essential questions that could inspire proper research, 2) good practices to be recommended for future research, and 3) whether citizen science is a suitable approach to further research in this field. The findings could contribute to determining a broader solution for building trust and infrastructure in scholarly communication. The principles of Open Science will be used as a framework to see if they offer insights into this work going forward.

3.
J Thorac Dis ; 12(5): 2883-2895, 2020 May.
Article in English | MEDLINE | ID: mdl-32642201

ABSTRACT

Comprehensive characterization of the genomic landscape of epidermal growth factor receptor (EGFR)-mutated lung cancers have identified patterns of secondary mutations beyond the primary oncogenic EGFR mutation. These include concurrent pathogenic alterations affecting p53 (60-65%), RTKs (5-10%), PIK3CA/KRAS (3-23%), Wnt (5-10%), and cell cycle (7-25%) pathways as well as transcription factors such as MYC and NKX2-1 (10-15%). The majority of these co-occurring alterations were detected or enriched in samples collected from patients at resistance to tyrosine kinase inhibitor (TKI) treatment, indicating a potential functional role in driving resistance to therapy. Of note, these co-occurring tumor genomic alterations are not necessarily mutually exclusive, and evidence suggests that multiple clonal and sub-clonal cancer cell populations can co-exist and contribute to EGFR TKI resistance. Computational tools aimed to classify, track and predict the evolution of cancer clonal populations during therapy are being investigated in pre-clinical models to guide the selection of combination therapy switching strategies that may delay the development of treatment resistance. Here we review the most frequently identified tumor genomic alterations that co-occur with mutated EGFR and the evidence that these alterations effect responsiveness to EGFR TKI treatment.

4.
Nat Genet ; 49(12): 1693-1704, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106415

ABSTRACT

A widespread approach to modern cancer therapy is to identify a single oncogenic driver gene and target its mutant-protein product (for example, EGFR-inhibitor treatment in EGFR-mutant lung cancers). However, genetically driven resistance to targeted therapy limits patient survival. Through genomic analysis of 1,122 EGFR-mutant lung cancer cell-free DNA samples and whole-exome analysis of seven longitudinally collected tumor samples from a patient with EGFR-mutant lung cancer, we identified critical co-occurring oncogenic events present in most advanced-stage EGFR-mutant lung cancers. We defined new pathways limiting EGFR-inhibitor response, including WNT/ß-catenin alterations and cell-cycle-gene (CDK4 and CDK6) mutations. Tumor genomic complexity increases with EGFR-inhibitor treatment, and co-occurring alterations in CTNNB1 and PIK3CA exhibit nonredundant functions that cooperatively promote tumor metastasis or limit EGFR-inhibitor response. This study calls for revisiting the prevailing single-gene driver-oncogene view and links clinical outcomes to co-occurring genetic alterations in patients with advanced-stage EGFR-mutant lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Clonal Evolution , Cyclin-Dependent Kinases/genetics , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Neoplasm Staging , Protein Kinase Inhibitors/pharmacology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/genetics
5.
Int J Syst Evol Microbiol ; 67(6): 2046-2052, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28639933

ABSTRACT

Two strains, D5088T and D5095, representing a novel yeast species belonging to the genus Saccharomyces were isolated from oak tree bark and surrounding soil located at an altitude of 1000 m above sea level in Saint Auban, France. Sequence analyses of the internal transcribed spacer (ITS) region and 26S rRNA D1/D2 domains indicated that the two strains were most closely related to Saccharomyces mikatae and Saccharomyces paradoxus. Genetic hybridization analyses showed that both strains are reproductively isolated from all other Saccharomyces species and, therefore, represent a distinct biological species. The species name Saccharomyces jurei sp. nov. is proposed to accommodate these two strains, with D5088T (=CBS 14759T=NCYC 3947T) designated as the type strain.


Subject(s)
Phylogeny , Plant Bark/microbiology , Quercus/microbiology , Saccharomyces/classification , DNA, Fungal/genetics , DNA, Ribosomal Spacer/genetics , France , Mycological Typing Techniques , RNA, Ribosomal/genetics , Saccharomyces/genetics , Saccharomyces/isolation & purification , Sequence Analysis, DNA
6.
Mol Cell ; 67(1): 128-138.e7, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28648777

ABSTRACT

Mutations in cancer reprogram amino acid metabolism to drive tumor growth, but the molecular mechanisms are not well understood. Using an unbiased proteomic screen, we identified mTORC2 as a critical regulator of amino acid metabolism in cancer via phosphorylation of the cystine-glutamate antiporter xCT. mTORC2 phosphorylates serine 26 at the cytosolic N terminus of xCT, inhibiting its activity. Genetic inhibition of mTORC2, or pharmacologic inhibition of the mammalian target of rapamycin (mTOR) kinase, promotes glutamate secretion, cystine uptake, and incorporation into glutathione, linking growth factor receptor signaling with amino acid uptake and utilization. These results identify an unanticipated mechanism regulating amino acid metabolism in cancer, enabling tumor cells to adapt to changing environmental conditions.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain Neoplasms/enzymology , Cysteine/metabolism , Glioblastoma/enzymology , Glutamine/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Amino Acid Transport System y+/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Glutathione/biosynthesis , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Mutation , Phosphorylation , Protein Binding , Proteomics/methods , RNA Interference , Serine , TOR Serine-Threonine Kinases/genetics , Tandem Mass Spectrometry , Time Factors , Transfection , Tumor Microenvironment
7.
Nat Genet ; 49(1): 87-96, 2017 01.
Article in English | MEDLINE | ID: mdl-27869830

ABSTRACT

Metastasis is the leading cause of death in people with lung cancer, yet the molecular effectors underlying tumor dissemination remain poorly defined. Through the development of an in vivo spontaneous lung cancer metastasis model, we show that the developmentally regulated transcriptional repressor Capicua (CIC) suppresses invasion and metastasis. Inactivation of CIC relieves repression of its effector ETV4, driving ETV4-mediated upregulation of MMP24, which is necessary and sufficient for metastasis. Loss of CIC, or an increase in levels of its effectors ETV4 and MMP24, is a biomarker of tumor progression and worse outcomes in people with lung and/or gastric cancer. Our findings reveal CIC as a conserved metastasis suppressor, highlighting new anti-metastatic strategies that could potentially improve patient outcomes.


Subject(s)
Adenovirus E1A Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/pathology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Adenovirus E1A Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Gene Expression Profiling , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Matrix Metalloproteinases, Membrane-Associated/genetics , Mice , Mice, SCID , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Cells, Cultured
8.
Cancer Cell ; 29(4): 563-573, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27070703

ABSTRACT

Intratumoral heterogeneity of signaling networks may contribute to targeted cancer therapy resistance, including in the highly lethal brain cancer glioblastoma (GBM). We performed single-cell phosphoproteomics on a patient-derived in vivo GBM model of mTOR kinase inhibitor resistance and coupled it to an analytical approach for detecting changes in signaling coordination. Alterations in the protein signaling coordination were resolved as early as 2.5 days after treatment, anticipating drug resistance long before it was clinically manifest. Combination therapies were identified that resulted in complete and sustained tumor suppression in vivo. This approach may identify actionable alterations in signal coordination that underlie adaptive resistance, which can be suppressed through combination drug therapy, including non-obvious drug combinations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Single-Cell Analysis/methods , Adaptation, Physiological , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Butadienes/administration & dosage , Dasatinib/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Gene Expression Profiling , Genes, erbB-1 , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Models, Biological , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/physiology , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitriles/administration & dosage , Pyrazines/administration & dosage , Selection, Genetic , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology , Xenograft Model Antitumor Assays
9.
Elife ; 42015 Dec 23.
Article in English | MEDLINE | ID: mdl-26701914

ABSTRACT

Conflict over parental investment between parent and offspring is predicted to lead to selection on genes expressed in offspring for traits influencing maternal investment, and on parentally expressed genes affecting offspring behaviour. However, the specific genetic variants that indirectly modify maternal or offspring behaviour remain largely unknown. Using a cross-fostered population of mice, we map maternal behaviour in genetically uniform mothers as a function of genetic variation in offspring and identify loci on offspring chromosomes 5 and 7 that modify maternal behaviour. Conversely, we found that genetic variation among mothers influences offspring development, independent of offspring genotype. Offspring solicitation and maternal behaviour show signs of coadaptation as they are negatively correlated between mothers and their biological offspring, which may be linked to costs of increased solicitation on growth found in our study. Overall, our results show levels of parental provisioning and offspring solicitation are unique to specific genotypes.


Subject(s)
Behavior, Animal , Genetic Variation , Maternal Behavior , Animals , Female , Mice
10.
J Clin Invest ; 125(4): 1591-602, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25798620

ABSTRACT

The mechanistic target of rapamycin (mTOR) is hyperactivated in many types of cancer, rendering it a compelling drug target; however, the impact of mTOR inhibition on metabolic reprogramming in cancer is incompletely understood. Here, by integrating metabolic and functional studies in glioblastoma multiforme (GBM) cell lines, preclinical models, and clinical samples, we demonstrate that the compensatory upregulation of glutamine metabolism promotes resistance to mTOR kinase inhibitors. Metabolomic studies in GBM cells revealed that glutaminase (GLS) and glutamate levels are elevated following mTOR kinase inhibitor treatment. Moreover, these mTOR inhibitor-dependent metabolic alterations were confirmed in a GBM xenograft model. Expression of GLS following mTOR inhibitor treatment promoted GBM survival in an α-ketoglutarate-dependent (αKG-dependent) manner. Combined genetic and/or pharmacological inhibition of mTOR kinase and GLS resulted in massive synergistic tumor cell death and growth inhibition in tumor-bearing mice. These results highlight a critical role for compensatory glutamine metabolism in promoting mTOR inhibitor resistance and suggest that rational combination therapy has the potential to suppress resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzophenanthridines/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/physiology , Glioblastoma/drug therapy , Glutaminase/physiology , Glutamine/metabolism , Indoles/pharmacology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aged , Animals , Benzophenanthridines/administration & dosage , Benzophenanthridines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Citric Acid Cycle , Drug Synergism , Energy Metabolism , Gas Chromatography-Mass Spectrometry , Glioblastoma/metabolism , Glioblastoma/pathology , Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , Glutaminase/biosynthesis , Glutaminase/genetics , Glycolysis , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Ketoglutaric Acids/metabolism , Magnetic Resonance Spectroscopy , Male , Metabolome/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Purines/administration & dosage , Purines/therapeutic use , RNA, Small Interfering/pharmacology , Rotarod Performance Test , Signal Transduction/drug effects , Signal Transduction/physiology , Temporal Lobe/metabolism , Xenograft Model Antitumor Assays
11.
Cancer Discov ; 4(8): 876-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25092745

ABSTRACT

Single-cell sequencing approaches are needed to characterize the genomic diversity of complex tumors, shedding light on their evolutionary paths and potentially suggesting more effective therapies. In this issue of Cancer Discovery, Francis and colleagues develop a novel integrative approach to identify distinct tumor subpopulations based on joint detection of clonal and subclonal events from bulk tumor and single-nucleus whole-genome sequencing, allowing them to infer a subclonal architecture. Surprisingly, the authors identify convergent evolution of multiple, mutually exclusive, independent EGFR gain-of-function variants in a single tumor. This study demonstrates the value of integrative single-cell genomics and highlights the biologic primacy of EGFR as an actionable target in glioblastoma.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/genetics , High-Throughput Nucleotide Sequencing/methods , Single-Cell Analysis/methods , Humans
12.
Science ; 343(6166): 72-6, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24310612

ABSTRACT

Intratumoral heterogeneity contributes to cancer drug resistance, but the underlying mechanisms are not understood. Single-cell analyses of patient-derived models and clinical samples from glioblastoma patients treated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) demonstrate that tumor cells reversibly up-regulate or suppress mutant EGFR expression, conferring distinct cellular phenotypes to reach an optimal equilibrium for growth. Resistance to EGFR TKIs is shown to occur by elimination of mutant EGFR from extrachromosomal DNA. After drug withdrawal, reemergence of clonal EGFR mutations on extrachromosomal DNA follows. These results indicate a highly specific, dynamic, and adaptive route by which cancers can evade therapies that target oncogenes maintained on extrachromosomal DNA.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glioblastoma/drug therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Central Nervous System Neoplasms/genetics , DNA/genetics , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Glioblastoma/genetics , Humans , Mice , Mutation , Neoplasm Transplantation , Quinazolines/therapeutic use , Single-Cell Analysis , Tumor Cells, Cultured , Withholding Treatment
13.
Cell Metab ; 18(5): 726-39, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24140020

ABSTRACT

Aerobic glycolysis (the Warburg effect) is a core hallmark of cancer, but the molecular mechanisms underlying it remain unclear. Here, we identify an unexpected central role for mTORC2 in cancer metabolic reprogramming where it controls glycolytic metabolism by ultimately regulating the cellular level of c-Myc. We show that mTORC2 promotes inactivating phosphorylation of class IIa histone deacetylases, which leads to the acetylation of FoxO1 and FoxO3, and this in turn releases c-Myc from a suppressive miR-34c-dependent network. These central features of activated mTORC2 signaling, acetylated FoxO, and c-Myc levels are highly intercorrelated in clinical samples and with shorter survival of GBM patients. These results identify a specific, Akt-independent role for mTORC2 in regulating glycolytic metabolism in cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , Glioblastoma/metabolism , Glycolysis , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , Acetylation/drug effects , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Death/drug effects , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Glucose/pharmacology , Glycolysis/drug effects , Histone Deacetylases/metabolism , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , MicroRNAs/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
14.
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
15.
Cell Metab ; 17(6): 1000-1008, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23707073

ABSTRACT

Alternative splicing contributes to diverse aspects of cancer pathogenesis including altered cellular metabolism, but the specificity of the process or its consequences are not well understood. We characterized genome-wide alternative splicing induced by the activating EGFRvIII mutation in glioblastoma (GBM). EGFRvIII upregulates the heterogeneous nuclear ribonucleoprotein (hnRNP) A1 splicing factor, promoting glycolytic gene expression and conferring significantly shorter survival in patients. HnRNPA1 promotes splicing of a transcript encoding the Myc-interacting partner Max, generating Delta Max, an enhancer of Myc-dependent transformation. Delta Max, but not full-length Max, rescues Myc-dependent glycolytic gene expression upon induced EGFRvIII loss, and correlates with hnRNPA1 expression and downstream Myc-dependent gene transcription in patients. Finally, Delta Max is shown to promote glioma cell proliferation in vitro and augment EGFRvIII expressing GBM growth in vivo. These results demonstrate an important role for alternative splicing in GBM and identify Delta Max as a mediator of Myc-dependent tumor cell metabolism.


Subject(s)
Alternative Splicing/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterografts , Humans , Mice , Mice, SCID , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering
16.
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
17.
Proc Natl Acad Sci U S A ; 110(11): 4339-44, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440206

ABSTRACT

Despite their nearly universal activation of mammalian target of rapamycin (mTOR) signaling, glioblastomas (GBMs) are strikingly resistant to mTOR-targeted therapy. We analyzed GBM cell lines, patient-derived tumor cell cultures, and clinical samples from patients in phase 1 clinical trials, and find that the promyelocytic leukemia (PML) gene mediates resistance to mTOR-targeted therapies. Direct mTOR inhibitors and EGF receptor (EGFR) inhibitors that block downstream mTOR signaling promote nuclear PML expression in GBMs, and genetic overexpression and knockdown approaches demonstrate that PML prevents mTOR and EGFR inhibitor-dependent cell death. Low doses of the PML inhibitor, arsenic trioxide, abrogate PML expression and reverse mTOR kinase inhibitor resistance in vivo, thus markedly inhibiting tumor growth and promoting tumor cell death in mice. These results identify a unique role for PML in mTOR and EGFR inhibitor resistance and provide a strong rationale for a combination therapeutic strategy to overcome it.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Nuclear Proteins/metabolism , Oxides/pharmacology , TOR Serine-Threonine Kinases/biosynthesis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Arsenic Trioxide , Cell Line, Tumor , ErbB Receptors/biosynthesis , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
18.
Front Genet ; 3: 198, 2012.
Article in English | MEDLINE | ID: mdl-23060902

ABSTRACT

Recombinant inbred (RI) systems such as the BXD mouse family represent a population with defined genetic architecture and variation that approximates those of natural populations. With the development of novel RI lines and sophisticated methods that conjointly analyze phenotype, gene sequence, and expression data, RI systems such as BXD are a timely and powerful tool to advance the field of behavioral ecology. The latter traditionally focused on functional questions such as the adaptive value of behavior but largely ignored underlying genetics and mechanisms. In this perspective, we argue that using RI systems to address questions in behavioral ecology and evolutionary biology has great potential to advance research in these fields. We outline key questions and how they can be tackled using RI systems and BXD in particular. The unique opportunity to analyze genetic and phenotypic data from studies conducted in different laboratories and at different times is a key benefit of RI systems and may lead the way to a better understanding of how adaptive phenotypes arise from genetic and environmental factors.

19.
Neuro Oncol ; 14(8): 1050-61, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22670012

ABSTRACT

To optimize the development of stem cell (SC)-based therapies for the treatment of glioblastoma (GBM), we compared the pathotropism of 2 SC sources, human mesenchymal stem cells (hMSCs) and fetal neural stem cells (fNSCs), toward 2 orthotopic GBM models, circumscribed U87vIII and highly infiltrative GBM26. High resolution and contrast-enhanced (CE) magnetic resonance imaging (MRI) were performed at 14.1 Tesla to longitudinally monitor the in vivo location of hMSCs and fNSCs labeled with the same amount of micron-size particles of iron oxide (MPIO). To assess pathotropism, SCs were injected in the contralateral hemisphere of U87vIII tumor-bearing mice. Both MPIO-labeled SC types exhibited tropism to tumors, first localizing at the tumor edges, then in the tumor masses. MPIO-labeled hMSCs and fNSCs were also injected intratumorally in mice with U87vIII or GBM26 tumors to assess their biodistribution. Both SC types distributed throughout the tumor in both GBM models. Of interest, in the U87vIII model, areas of hyposignal colocalized first with the enhancing regions (ie, regions of high vascular permeability), consistent with SC tropism to vascular endothelial growth factor. In the GBM26 model, no rim of hyposignal was observed, consistent with the infiltrative nature of this tumor. Quantitative analysis of the index of dispersion confirmed that both MPIO-labeled SC types longitudinally distribute inside the tumor masses after intratumoral injection. Histological studies confirmed the MRI results. In summary, our results indicate that hMSCs and fNSCs exhibit similar properties regarding tumor tropism and intratumoral dissemination, highlighting the potential of these 2 SC sources as adequate candidates for SC-based therapies.


Subject(s)
Ferric Compounds , Fetal Stem Cells , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells , Neoplasms, Experimental , Neural Stem Cells , Animals , Brain Neoplasms/pathology , Cell Movement/physiology , Female , Fluorescent Antibody Technique , Glioblastoma/pathology , Humans , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Confocal , Neuroimaging , Stem Cell Transplantation
20.
Cancer Discov ; 1(6): 524-38, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22145100

ABSTRACT

UNLABELLED: Although it is known that mTOR complex 2 (mTORC2) functions upstream of Akt, the role of this protein kinase complex in cancer is not well understood. Through an integrated analysis of cell lines, in vivo models, and clinical samples, we demonstrate that mTORC2 is frequently activated in glioblastoma (GBM), the most common malignant primary brain tumor of adults. We show that the common activating epidermal growth factor receptor (EGFR) mutation (EGFRvIII) stimulates mTORC2 kinase activity, which is partially suppressed by PTEN. mTORC2 signaling promotes GBM growth and survival and activates NF-κB. Importantly, this mTORC2-NF-κB pathway renders GBM cells and tumors resistant to chemotherapy in a manner independent of Akt. These results highlight the critical role of mTORC2 in the pathogenesis of GBM, including through the activation of NF-κB downstream of mutant EGFR, leading to a previously unrecognized function in cancer chemotherapy resistance. These findings suggest that therapeutic strategies targeting mTORC2, alone or in combination with chemotherapy, will be effective in the treatment of cancer. SIGNIFICANCE: This study demonstrates that EGFRvIII-activated mTORC2 signaling promotes GBM proliferation, survival, and chemotherapy resistance through Akt-independent activation of NF-κB. These results highlight the role of mTORC2 as an integrator of two canonical signaling networks that are commonly altered in cancer, EGFR/phosphoinositide-3 kinase (PI3K) and NF-κB. These results also validate the importance of mTORC2 as a cancer target and provide new insights into its role in mediating chemotherapy resistance, suggesting new treatment strategies.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , NF-kappa B/metabolism , TOR Serine-Threonine Kinases/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , NF-kappa B/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL