Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Sci Adv ; 8(6): eabm2382, 2022 02 11.
Article En | MEDLINE | ID: mdl-35138907

Fusion genes represent a class of attractive therapeutic targets. Thousands of fusion genes have been identified in patients with cancer, but the functional consequences and therapeutic implications of most of these remain largely unknown. Here, we develop a functional genomic approach that consists of efficient fusion reconstruction and sensitive cell viability and drug response assays. Applying this approach, we characterize ~100 fusion genes detected in patient samples of The Cancer Genome Atlas, revealing a notable fraction of low-frequency fusions with activating effects on tumor growth. Focusing on those in the RTK-RAS pathway, we identify a number of activating fusions that can markedly affect sensitivity to relevant drugs. Last, we propose an integrated, level-of-evidence classification system to prioritize gene fusions systematically. Our study reiterates the urgent clinical need to incorporate similar functional genomic approaches to characterize gene fusions, thereby maximizing the utility of gene fusions for precision oncology.


Neoplasms , Gene Fusion , Genome , Genomics , Humans , Neoplasms/genetics , Precision Medicine
2.
Elife ; 92020 04 09.
Article En | MEDLINE | ID: mdl-32270762

The melanoma-associated antigen family A (MAGEA) antigens are expressed in a wide variety of malignant tumors but not in adult somatic cells, rendering them attractive targets for cancer immunotherapy. Here we show that a number of cancer-associated MAGEA mutants that undergo proteasome-dependent degradation in vitro could negatively impact their utility as immunotherapeutic targets. Importantly, in pancreatic ductal adenocarcinoma cell models, MAGEA6 suppresses macroautophagy (autophagy). The inhibition of autophagy is released upon MAGEA6 degradation, which can be induced by nutrient deficiency or by acquisition of cancer-associated mutations. Using xenograft mouse models, we demonstrated that inhibition of autophagy is critical for tumor initiation whereas reinstitution of autophagy as a consequence of MAGEA6 degradation contributes to tumor progression. These findings could inform cancer immunotherapeutic strategies for targeting MAGEA antigens and provide mechanistic insight into the divergent roles of MAGEA6 during pancreatic cancer initiation and progression.


Antigens, Neoplasm/physiology , Autophagy/physiology , Carcinoma, Pancreatic Ductal/etiology , Neoplasm Proteins/physiology , Pancreatic Neoplasms/etiology , Animals , Antigens, Neoplasm/genetics , Autophagy/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Mice , Mutation , Neoplasm Proteins/genetics , Pancreatic Neoplasms/pathology , Proteasome Endopeptidase Complex/physiology
3.
Cancer Cell ; 33(3): 450-462.e10, 2018 03 12.
Article En | MEDLINE | ID: mdl-29533785

The functional impact of the vast majority of cancer somatic mutations remains unknown, representing a critical knowledge gap for implementing precision oncology. Here, we report the development of a moderate-throughput functional genomic platform consisting of efficient mutant generation, sensitive viability assays using two growth factor-dependent cell models, and functional proteomic profiling of signaling effects for select aberrations. We apply the platform to annotate >1,000 genomic aberrations, including gene amplifications, point mutations, indels, and gene fusions, potentially doubling the number of driver mutations characterized in clinically actionable genes. Further, the platform is sufficiently sensitive to identify weak drivers. Our data are accessible through a user-friendly, public data portal. Our study will facilitate biomarker discovery, prediction algorithm improvement, and drug development.


Biomarkers, Tumor/genetics , Mutation/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Algorithms , Genomics , High-Throughput Nucleotide Sequencing/methods , Humans , Precision Medicine , Proteomics
4.
Cancer Res ; 77(13): 3502-3512, 2017 07 01.
Article En | MEDLINE | ID: mdl-28512244

Oncogenic gene fusions drive many human cancers, but tools to more quickly unravel their functional contributions are needed. Here we describe methodology permitting fusion gene construction for functional evaluation. Using this strategy, we engineered the known fusion oncogenes, BCR-ABL1, EML4-ALK, and ETV6-NTRK3, as well as 20 previously uncharacterized fusion genes identified in The Cancer Genome Atlas datasets. In addition to confirming oncogenic activity of the known fusion oncogenes engineered by our construction strategy, we validated five novel fusion genes involving MET, NTRK2, and BRAF kinases that exhibited potent transforming activity and conferred sensitivity to FDA-approved kinase inhibitors. Our fusion construction strategy also enabled domain-function studies of BRAF fusion genes. Our results confirmed other reports that the transforming activity of BRAF fusions results from truncation-mediated loss of inhibitory domains within the N-terminus of the BRAF protein. BRAF mutations residing within this inhibitory region may provide a means for BRAF activation in cancer, therefore we leveraged the modular design of our fusion gene construction methodology to screen N-terminal domain mutations discovered in tumors that are wild-type at the BRAF mutation hotspot, V600. We identified an oncogenic mutation, F247L, whose expression robustly activated the MAPK pathway and sensitized cells to BRAF and MEK inhibitors. When applied broadly, these tools will facilitate rapid fusion gene construction for subsequent functional characterization and translation into personalized treatment strategies. Cancer Res; 77(13); 3502-12. ©2017 AACR.


Neoplasms/genetics , Oncogene Fusion , Oncogene Proteins, Fusion/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Female , Fusion Proteins, bcr-abl/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Nude , Mutagenesis, Site-Directed/methods , Proto-Oncogene Proteins B-raf/genetics
5.
Nat Commun ; 7: 10500, 2016 Jan 25.
Article En | MEDLINE | ID: mdl-26806015

As we enter the era of precision medicine, characterization of cancer genomes will directly influence therapeutic decisions in the clinic. Here we describe a platform enabling functionalization of rare gene mutations through their high-throughput construction, molecular barcoding and delivery to cancer models for in vivo tumour driver screens. We apply these technologies to identify oncogenic drivers of pancreatic ductal adenocarcinoma (PDAC). This approach reveals oncogenic activity for rare gene aberrations in genes including NAD Kinase (NADK), which regulates NADP(H) homeostasis and cellular redox state. We further validate mutant NADK, whose expression provides gain-of-function enzymatic activity leading to a reduction in cellular reactive oxygen species and tumorigenesis, and show that depletion of wild-type NADK in PDAC cell lines attenuates cancer cell growth in vitro and in vivo. These data indicate that annotating rare aberrations can reveal important cancer signalling pathways representing additional therapeutic targets.


Pancreatic Neoplasms/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Carcinogenesis , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Mutation , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Pancreatic Neoplasms
6.
Cancer Res ; 75(24): 5341-54, 2015 Dec 15.
Article En | MEDLINE | ID: mdl-26627007

Large-scale sequencing efforts are uncovering the complexity of cancer genomes, which are composed of causal "driver" mutations that promote tumor progression along with many more pathologically neutral "passenger" events. The majority of mutations, both in known cancer drivers and uncharacterized genes, are generally of low occurrence, highlighting the need to functionally annotate the long tail of infrequent mutations present in heterogeneous cancers. Here we describe a mutation assessment pipeline enabled by high-throughput engineering of molecularly barcoded gene variant expression clones identified by tumor sequencing. We first used this platform to functionally assess tail mutations observed in PIK3CA, which encodes the catalytic subunit alpha of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) frequently mutated in cancer. Orthogonal screening for PIK3CA variant activity using in vitro and in vivo cell growth and transformation assays differentiated driver from passenger mutations, revealing that PIK3CA variant activity correlates imperfectly with its mutation frequency across breast cancer populations. Although PIK3CA mutations with frequencies above 5% were significantly more oncogenic than wild-type in all assays, mutations occurring at 0.07% to 5.0% included those with and without oncogenic activities that ranged from weak to strong in at least one assay. Proteomic profiling coupled with therapeutic sensitivity assays on PIK3CA variant-expressing cell models revealed variant-specific activation of PI3K signaling as well as other pathways that include the MEK1/2 module of mitogen-activated protein kinase pathway. Our data indicate that cancer treatments will need to increasingly consider the functional relevance of specific mutations in driver genes rather than considering all mutations in drivers as equivalent.


Breast Neoplasms/genetics , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Female , Heterografts , Humans , Immunoblotting , Mice , Mice, Nude , Mutagenesis, Site-Directed , Phenotype , Polymerase Chain Reaction , Transduction, Genetic
7.
Cancer Res ; 73(3): 1142-55, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23221384

Moesin is an ERM family protein that connects the actin cytoskeleton to transmembrane receptors. With the identification of the ERM family protein NF2 as a tumor suppressor in glioblastoma, we investigated roles for other ERM proteins in this malignancy. Here, we report that overexpression of moesin occurs generally in high-grade glioblastoma in a pattern correlated with the stem cell marker CD44. Unlike NF2, moesin acts as an oncogene by increasing cell proliferation and stem cell neurosphere formation, with its ectopic overexpression sufficient to shorten survival in an orthotopic mouse model of glioblastoma. Moesin was the major ERM member activated by phosphorylation in glioblastoma cells, where it interacted and colocalized with CD44 in membrane protrusions. Increasing the levels of moesin competitively displaced NF2 from CD44, increasing CD44 expression in a positive feedback loop driven by the Wnt/ß-catenin signaling pathway. Therapeutic targeting of the moesin-CD44 interaction with the small-molecule inhibitor 7-cyanoquinocarcinol (DX-52-1) or with a CD44-mimetic peptide specifically reduced the proliferation of glioblastoma cells overexpressing moesin, where the Wnt/ß-catenin pathway was activated. Our findings establish moesin and CD44 as progression markers and drugable targets in glioblastoma, relating their oncogenic effects to activation of the Wnt/ß-catenin pathway.


Brain Neoplasms/pathology , Glioma/pathology , Hyaluronan Receptors/physiology , Microfilament Proteins/physiology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Amino Acid Sequence , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins/physiology , Disease Progression , Humans , Membrane Proteins/physiology , Microfilament Proteins/analysis , Molecular Sequence Data , Neurofibromin 2/metabolism , Phosphorylation
8.
Genome Res ; 22(11): 2120-9, 2012 Nov.
Article En | MEDLINE | ID: mdl-23028188

Endometrial cancer is the most common gynecological malignancy, with more than 280,000 cases occurring annually worldwide. Although previous studies have identified important common somatic mutations in endometrial cancer, they have primarily focused on a small set of known cancer genes and have thus provided a limited view of the molecular basis underlying this disease. Here we have developed an integrated systems-biology approach to identifying novel cancer genes contributing to endometrial tumorigenesis. We first performed whole-exome sequencing on 13 endometrial cancers and matched normal samples, systematically identifying somatic alterations with high precision and sensitivity. We then combined bioinformatics prioritization with high-throughput screening (including both shRNA-mediated knockdown and expression of wild-type and mutant constructs) in a highly sensitive cell viability assay. Our results revealed 12 potential driver cancer genes including 10 tumor-suppressor candidates (ARID1A, INHBA, KMO, TTLL5, GRM8, IGFBP3, AKTIP, PHKA2, TRPS1, and WNT11) and two oncogene candidates (ERBB3 and RPS6KC1). The results in the "sensor" cell line were recapitulated by siRNA-mediated knockdown in endometrial cancer cell lines. Focusing on ARID1A, we integrated mutation profiles with functional proteomics in 222 endometrial cancer samples, demonstrating that ARID1A mutations frequently co-occur with mutations in the phosphatidylinositol 3-kinase (PI3K) pathway and are associated with PI3K pathway activation. siRNA knockdown in endometrial cancer cell lines increased AKT phosphorylation supporting ARID1A as a novel regulator of PI3K pathway activity. Our study presents the first unbiased view of somatic coding mutations in endometrial cancer and provides functional evidence for diverse driver genes and mutations in this disease.


Endometrial Neoplasms/genetics , Exome , Genes, Tumor Suppressor , Genomics , High-Throughput Nucleotide Sequencing , Oncogenes , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Female , Humans , Mutation , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , RNA, Small Interfering , Sequence Analysis, DNA , Systems Biology
9.
J Neurosurg ; 116(4): 810-20, 2012 Apr.
Article En | MEDLINE | ID: mdl-22283189

OBJECT: Chordomas are locally aggressive bone tumors known to arise from the remnants of the notochord. Because chordomas are rare, molecular studies aimed at developing new therapies are scarce and new approaches are needed. Chordoma cells and cancer stem-like cells share similar characteristics, including self-renewal, differentiation, and resistance to chemotherapy. Therefore, it seems possible that chordomas might contain a subpopulation of cancer stem-like cells. The aim of this study is to determine whether cancer stem-like cells might be present in chordomas. METHODS: In this study, the authors used gene expression analysis for common cancer stem-like cellmarkers, including c-myc, SSEA-1, oct4, klf4, sox2, nanog, and brachyury, and compared chordoma cells and tissues with nucleus pulposus tissues (disc degenerated nontumorigenic tissues). Differentiation through agents such as all-trans retinoic acid and osteogenic differentiation medium was induced to the chordoma cells. Additionally, U-CH1 cells were sorted via magnetic cell sorting for stem cell markers CD133 and CD15. After separation, positive and negative cells for these markers were grown in a nonadherent environment, soft agar, to determine whether the presence of these cancer stem-like cells might be responsible for initiating chordoma. The results were compared with those of untreated cells in terms of migration, proliferation, and gene expression by using reverse transcriptase polymerase chain reaction. RESULTS: The results indicate that chordoma cells might be differentiating and committing into an osteogenic lineage when induced with the osteogenic differentiation agent. Chordoma cells that are induced with retinoic acid showed slower migration and proliferation rates when compared with the untreated cells. Chordoma cells that were found to be enriched by cancer stem-like cell markers, namely CD133 and CD15, were able to live in a nonadherent soft agar medium, demonstrating a self-renewal capability. To the authors' knowledge, this is the first time that cancer stem-like cell markers were also found to be expressed in chordoma cells and tissues. CONCLUSIONS: Cancer stem-like cell detection might be an important step in determining the recurrent and metastatic characteristics of chordoma. This finding may lead to the development of new approaches toward treatments of chordomas.


Chordoma/pathology , Neoplastic Stem Cells/pathology , Spinal Cord Neoplasms/pathology , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Child , Chordoma/genetics , Female , Gene Expression Profiling , Humans , Kruppel-Like Factor 4 , Male , Middle Aged , Spinal Cord Neoplasms/genetics , Young Adult
...