Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Commun ; 14(1): 2575, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142597

ABSTRACT

Noradrenergic and mesenchymal identities have been characterized in neuroblastoma cell lines according to their epigenetic landscapes and core regulatory circuitries. However, their relationship and relative contribution in patient tumors remain poorly defined. We now document spontaneous and reversible plasticity between the two identities, associated with epigenetic reprogramming, in several neuroblastoma models. Interestingly, xenografts with cells from each identity eventually harbor a noradrenergic phenotype suggesting that the microenvironment provides a powerful pressure towards this phenotype. Accordingly, such a noradrenergic cell identity is systematically observed in single-cell RNA-seq of 18 tumor biopsies and 15 PDX models. Yet, a subpopulation of these noradrenergic tumor cells presents with mesenchymal features that are shared with plasticity models, indicating that the plasticity described in these models has relevance in neuroblastoma patients. This work therefore emphasizes that intrinsic plasticity properties of neuroblastoma cells are dependent upon external cues of the environment to drive cell identity.


Subject(s)
Cell Plasticity , Neuroblastoma , Humans , Neuroblastoma/metabolism , Cell Line, Tumor , Tumor Microenvironment/genetics
2.
Genes Chromosomes Cancer ; 62(6): 367-372, 2023 06.
Article in English | MEDLINE | ID: mdl-36744846

ABSTRACT

Adipocytic tumors are the most common mesenchymal tumors in soft tissues. Among them, a diagnostic challenge relies in the distinction between lipoma and atypical lipomatous tumor (ALT)/well differentiated liposarcoma (WDLPS), as both entities are often undistinguishable not only from a radiological point of view, but also at the microscopic level and particularly when dealing with small tumor specimen. Thus, detection of recurrent MDM2 amplifications may be the only criteria to discriminate malignant tumors from lipomas. In this study, we report the case of a patient diagnosed with a well differentiated, adipocytic tumor located in the inferior limb and lacking MDM2 amplification, whose diagnosis was reclassified for ALT/WDLPS after identification of an alternative MDM4 amplification by comparative genomic hybridization profiling, whole exome sequencing and fluorescence in situ hybridization (FISH). Screening of a cohort of 37 large, deep-seated, well-differentiated adipocytic tumors previously classified as lipomas using RT-qPCR and FISH failed to detect other cases of MDM4-amplified ALT/WDLPS. This report shows that MDM4 amplification is an exceptional molecular event alternative to MDM2 amplification in ALT/WDLPS. This alteration should be considered and looked for in suspicious adipocytic tumors to optimize their surgical management.


Subject(s)
Lipoma , Liposarcoma , Humans , Liposarcoma/diagnosis , Liposarcoma/genetics , Liposarcoma/pathology , Gene Amplification , In Situ Hybridization, Fluorescence , Comparative Genomic Hybridization , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Lipoma/diagnosis , Lipoma/genetics , Lipoma/pathology , Biomarkers, Tumor/genetics , Proto-Oncogene Proteins/genetics , Cell Cycle Proteins/genetics
3.
Eur J Surg Oncol ; 49(6): 1053-1060, 2023 06.
Article in English | MEDLINE | ID: mdl-35151525

ABSTRACT

Retroperitoneal sarcomas (RPS) refer to a heterogeneous group of malignancies of mesenchymal origin developing from retroperitoneal tissues and vessels. The most frequent RPS are well differentiated/dedifferentiated liposarcomas and leiomyosarcomas, but other rare histological subtypes can be observed. Over the last decade, significant advances have been made in the pathological and molecular characterization of sarcomas. These advances have led to major changes in their diagnostic management as well as in the development of new therapeutic strategies based on tumor biology and microenvironment. This review describes the current knowledge and recent findings in the pathology and molecular biology of the most frequent RPS subtypes.


Subject(s)
Leiomyosarcoma , Liposarcoma , Retroperitoneal Neoplasms , Sarcoma , Soft Tissue Neoplasms , Humans , Sarcoma/genetics , Sarcoma/therapy , Sarcoma/diagnosis , Liposarcoma/pathology , Leiomyosarcoma/pathology , Retroperitoneal Neoplasms/genetics , Retroperitoneal Neoplasms/therapy , Retroperitoneal Neoplasms/diagnosis , Molecular Biology , Tumor Microenvironment
4.
Front Immunol ; 13: 1022136, 2022.
Article in English | MEDLINE | ID: mdl-36311728

ABSTRACT

CXCR6 is a receptor for the chemokine CXCL16, which exists as a membrane or soluble form. CXCR6 is a marker for resident memory T (TRM) cells that plays a role in immunosurveillance through their interaction with epithelial cells. The interaction of CXCR6 with CXCL16 expressed at the membrane of certain subpopulations of intratumor dendritic cells (DC) called DC3, ideally positions these CXCR6+ T cells to receive a proliferation signal from IL-15 also presented by DC3. Mice deficient in cxcr6 or blocking the interaction of CXCR6 with its ligand, experience a poorer control of tumor proliferation by CD8+ T cells, but also by NKT cells especially in the liver. Intranasal vaccination induces CXCL16 production in the lungs and is associated with infiltration by TRM expressing CXCR6, which are then required for the efficacy of anti-tumor vaccination. Therapeutically, the addition of CXCR6 to specific CAR-T cells enhances their intratumoral accumulation and prolongs survival in animal models of pancreatic, ovarian and lung cancer. Finally, CXCR6 is part of immunological signatures that predict response to immunotherapy based on anti-PD-(L)1 in various cancers. In contrast, a protumoral role of CXCR6+T cells has also been reported mainly in Non-alcoholic steatohepatitis (NASH) due to a non-antigen specific mechanism. The targeting and amplification of antigen-specific TRM expressing CXCR6 and its potential use as a biomarker of response to immunotherapy opens new perspectives in cancer treatment.


Subject(s)
Natural Killer T-Cells , Neoplasms , Mice , Animals , Receptors, CXCR6 , CD8-Positive T-Lymphocytes , Chemokine CXCL16 , Neoplasms/therapy
5.
Clin Cancer Res ; 28(22): 4983-4994, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36067339

ABSTRACT

PURPOSE: CD70 is a costimulatory molecule known to activate CD27-expressing T cells. CD27-CD70 interaction leads to the release of soluble CD27 (sCD27). Clear-cell renal cell carcinoma (ccRCC) expresses the highest levels of CD70 among all solid tumors; however, the clinical consequences of CD70 expression remain unclear. EXPERIMENTAL DESIGN: Tumor tissue from 25 patients with ccRCC was assessed for the expression of CD27 and CD70 in situ using multiplex immunofluorescence. CD27+ T-cell phenotypes in tumors were analyzed by flow cytometry and their gene expression profile were analyzed by single-cell RNA sequencing then confirmed with public data. Baseline sCD27 was measured in 81 patients with renal cell carcinoma (RCC) treated with immunotherapy (35 for training cohort and 46 for validation cohort). RESULTS: In the tumor microenvironment, CD27+ T cells interacted with CD70-expressing tumor cells. Compared with CD27- T cells, CD27+ T cells exhibited an apoptotic and dysfunctional signature. In patients with RCC, the intratumoral CD27-CD70 interaction was significantly correlated with the plasma sCD27 concentration. High sCD27 levels predicted poor overall survival in patients with RCC treated with anti-programmed cell death protein 1 in both the training and validation cohorts but not in patients treated with antiangiogenic therapy. CONCLUSIONS: In conclusion, we demonstrated that sCD27, a surrogate marker of T-cell dysfunction, is a predictive biomarker of resistance to immunotherapy in RCC. Given the frequent expression of CD70 and CD27 in solid tumors, our findings may be extended to other tumors.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , CD27 Ligand/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Immunotherapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Tumor Microenvironment
6.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36054452

ABSTRACT

BACKGROUND: High-risk neuroblastoma is a pediatric cancer with still a dismal prognosis, despite multimodal and intensive therapies. Tumor microenvironment represents a key component of the tumor ecosystem the complexity of which has to be accurately understood to define selective targeting opportunities, including immune-based therapies. METHODS: We combined various approaches including single-cell transcriptomics to dissect the tumor microenvironment of both a transgenic mouse neuroblastoma model and a cohort of 10 biopsies from neuroblastoma patients, either at diagnosis or at relapse. Features of related cells were validated by multicolor flow cytometry and functional assays. RESULTS: We show that the immune microenvironment of MYCN-driven mouse neuroblastoma is characterized by a low content of T cells, several phenotypes of macrophages and a population of cells expressing signatures of myeloid-derived suppressor cells (MDSCs) that are molecularly distinct from the various macrophage subsets. We document two cancer-associated fibroblasts (CAFs) subsets, one of which corresponding to CAF-S1, known to have immunosuppressive functions. Our data unravel a complex content in myeloid cells in patient tumors and further document a striking correspondence of the microenvironment populations between both mouse and human tumors. We show that mouse intratumor T cells exhibit increased expression of inhibitory receptors at the protein level. Consistently, T cells from patients are characterized by features of exhaustion, expressing inhibitory receptors and showing low expression of effector cytokines. We further functionally demonstrate that MDSCs isolated from mouse neuroblastoma have immunosuppressive properties, impairing the proliferation of T lymphocytes. CONCLUSIONS: Our study demonstrates that neuroblastoma tumors have an immunocompromised microenvironment characterized by dysfunctional T cells and accumulation of immunosuppressive cells. Our work provides a new and precious data resource to better understand the neuroblastoma ecosystem and suggest novel therapeutic strategies, targeting both tumor cells and components of the microenvironment.


Subject(s)
Neuroblastoma , Transcriptome , Animals , Child , Ecosystem , Humans , Mice , Neoplasm Recurrence, Local , Neuroblastoma/pathology , Tumor Microenvironment/genetics
7.
Nat Commun ; 13(1): 5399, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36104342

ABSTRACT

Cancer and cardiovascular disease (CVD) share common risk factors such as dyslipidemia, obesity and inflammation. However, the role of pro-atherogenic environment and its associated low-grade inflammation in tumor progression remains underexplored. Here we show that feeding C57BL/6J mice with a non-obesogenic high fat high cholesterol diet (HFHCD) for two weeks to induce mild dyslipidemia, increases the pool of circulating Ly6Chi monocytes available for initial melanoma development, in an IL-1ß-dependent manner. Descendants of circulating myeloid cells, which accumulate in the tumor microenvironment of mice under HFHCD, heighten pro-angiogenic and immunosuppressive activities locally. Limiting myeloid cell accumulation or targeting VEGF-A production by myeloid cells decrease HFHCD-induced tumor growth acceleration. Reverting the HFHCD to a chow diet at the time of tumor implantation protects against tumor growth. Together, these data shed light on cross-disease communication between cardiovascular pathologies and cancer.


Subject(s)
Dyslipidemias , Monocytes , Animals , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Dyslipidemias/pathology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Monocytes/pathology , Myeloid Cells/pathology , Tumor Microenvironment
8.
Mol Cell ; 82(13): 2458-2471.e9, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35550257

ABSTRACT

Many cancers are characterized by gene fusions encoding oncogenic chimeric transcription factors (TFs) such as EWS::FLI1 in Ewing sarcoma (EwS). Here, we find that EWS::FLI1 induces the robust expression of a specific set of novel spliced and polyadenylated transcripts within otherwise transcriptionally silent regions of the genome. These neogenes (NGs) are virtually undetectable in large collections of normal tissues or non-EwS tumors and can be silenced by CRISPR interference at regulatory EWS::FLI1-bound microsatellites. Ribosome profiling and proteomics further show that some NGs are translated into highly EwS-specific peptides. More generally, we show that hundreds of NGs can be detected in diverse cancers characterized by chimeric TFs. Altogether, this study identifies the transcription, processing, and translation of novel, specific, highly expressed multi-exonic transcripts from otherwise silent regions of the genome as a new activity of aberrant TFs in cancer.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion , Proto-Oncogene Protein c-fli-1 , Transcription Factors , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Genome/genetics , Genomics , Humans , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogenes/genetics , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Transcription Factors/genetics , Transcription, Genetic/genetics
9.
J Mol Diagn ; 23(10): 1380-1392, 2021 10.
Article in English | MEDLINE | ID: mdl-34325056

ABSTRACT

Cancers of unknown primary (CUP) are metastatic cancers for which the primary tumor is not found despite thorough diagnostic investigations. Multiple molecular assays have been proposed to identify the tissue of origin (TOO) and inform clinical care; however, none has been able to combine accuracy, interpretability, and easy access for routine use. We developed a classifier tool based on the training of a variational autoencoder to predict tissue of origin based on RNA-sequencing data. We used as training data 20,918 samples corresponding to 94 different categories, including 39 cancer types and 55 normal tissues. The TransCUPtomics classifier was applied to a retrospective cohort of 37 CUP patients and 11 prospective patients. TransCUPtomics exhibited an overall accuracy of 96% on reference data for TOO prediction. The TOO could be identified in 38 (79%) of 48 CUP patients. Eight of 11 prospective CUP patients (73%) could receive first-line therapy guided by TransCUPtomics prediction, with responses observed in most patients. The variational autoencoder added further utility by enabling prediction interpretability, and diagnostic predictions could be matched to detection of gene fusions and expressed variants. TransCUPtomics confidently predicted TOO for CUP and enabled tailored treatments leading to significant clinical responses. The interpretability of our approach is a powerful addition to improve the management of CUP patients.


Subject(s)
Deep Learning , Neoplasms, Unknown Primary/diagnosis , Neoplasms, Unknown Primary/genetics , RNA-Seq/methods , Transcriptome , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Data Accuracy , Female , Gene Fusion , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies
10.
Oncologist ; 26(7): 554-557, 2021 07.
Article in English | MEDLINE | ID: mdl-33797168

ABSTRACT

Cystic lymphangioma are rare benign vascular or lymphatic tumors, diagnosed mostly in newborns or children, that may become life-threatening because of local invasiveness. Surgical "en-bloc" resection with negative margins is the only curative treatment, but some patients are diagnosed with unresectable tumors. We describe the case of a young adult with giant unresectable mesenteric lymphangioma. Extensive pathological characterization as well as whole exome and transcriptome sequencing enabled us to identify mTOR pathway activation within endothelial tumor cells. The patient was treated with everolimus and experienced major partial response, leading to the surgical resection of the residual lesions. This case highlights the importance of molecular characterization of adult cystic lymphangioma for mTOR pathway activation because multidisciplinary therapeutic approaches, including neoadjuvant everolimus and secondary surgery, can lead to complete cure of this rare condition. KEY POINTS: The case of an adult patient diagnosed with giant unresectable mesenteric cystic lymphangioma, in which activation of the mTOR pathway was documented at both the pathological and transcriptomic levels, is reported. This patient showed major partial response to the mTOR inhibitor everolimus, which led to the successful resection of residual tumor lesions after 9 months of treatment. This report shows that mTOR targeting should be considered as neoadjuvant treatment in adult large cystic lymphangioma, as it can lead to complete surgery and cure of this rare condition.


Subject(s)
Everolimus , Lymphangioma, Cystic , TOR Serine-Threonine Kinases , Everolimus/therapeutic use , Humans , Lymphangioma, Cystic/drug therapy , Lymphangioma, Cystic/surgery , Mesentery , Neoadjuvant Therapy , TOR Serine-Threonine Kinases/genetics , Young Adult
11.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33692218

ABSTRACT

BACKGROUND: Resident memory T lymphocytes (TRM) are located in tissues and play an important role in immunosurveillance against tumors. The presence of TRM prior to treatment or their induction is associated to the response to anti-Programmed cell death protein 1 (PD-1)/Programmed death-ligand 1 (PD-L1) immunotherapy and the efficacy of cancer vaccines. Previous work by our group and others has shown that the intranasal route of vaccination allows more efficient induction of these cells in head and neck and lung mucosa, resulting in better tumor protection. The mechanisms of in vivo migration of these cells remains largely unknown, apart from the fact that they express the chemokine receptor CXCR6. METHODS: We used CXCR6-deficient mice and an intranasal tumor vaccination model targeting the Human Papillomavirus (HPV) E7 protein expressed by the TC-1 lung cancer epithelial cell line. The role of CXCR6 and its ligand, CXCL16, was analyzed using multiparametric cytometric techniques and Luminex assays.Human biopsies obtained from patients with lung cancer were also included in this study. RESULTS: We showed that CXCR6 was preferentially expressed by CD8+ TRM after vaccination in mice and also on intratumoral CD8+ TRM derived from human lung cancer. We also demonstrate that vaccination of Cxcr6-deficient mice induces a defect in the lung recruitment of antigen-specific CD8+ T cells, preferentially in the TRM subsets. In addition, we found that intranasal vaccination with a cancer vaccine is less effective in these Cxcr6-deficient mice compared with wild-type mice, and this loss of efficacy is associated with decreased recruitment of local antitumor CD8+ TRM. Interestingly, intranasal, but not intramuscular vaccination induced higher and more sustained concentrations of CXCL16, compared with other chemokines, in the bronchoalveolar lavage fluid and pulmonary parenchyma. CONCLUSIONS: This work demonstrates the in vivo role of CXCR6-CXCL16 axis in the migration of CD8+ resident memory T cells in lung mucosa after vaccination, resulting in the control of tumor growth. This work reinforces and explains why the intranasal route of vaccination is the most appropriate strategy for inducing these cells in the head and neck and pulmonary mucosa, which remains a major objective to overcome resistance to anti-PD-1/PD-L1, especially in cold tumors.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/pharmacology , Head and Neck Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Memory T Cells/drug effects , Receptors, CXCR6/deficiency , Vaccine Efficacy , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokine CXCL16/metabolism , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Humans , Immunologic Memory , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Memory T Cells/immunology , Memory T Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, CXCR6/genetics , Tumor Burden/drug effects , Tumor Microenvironment , Vaccination
12.
Cell Rep ; 30(6): 1767-1779.e6, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049009

ABSTRACT

EWSR1-FLI1, the chimeric oncogene specific for Ewing sarcoma (EwS), induces a cascade of signaling events leading to cell transformation. However, it remains elusive how genetically homogeneous EwS cells can drive the heterogeneity of transcriptional programs. Here, we combine independent component analysis of single-cell RNA sequencing data from diverse cell types and model systems with time-resolved mapping of EWSR1-FLI1 binding sites and of open chromatin regions to characterize dynamic cellular processes associated with EWSR1-FLI1 activity. We thus define an exquisitely specific and direct enhancer-driven EWSR1-FLI1 program. In EwS tumors, cell proliferation and strong oxidative phosphorylation metabolism are associated with a well-defined range of EWSR1-FLI1 activity. In contrast, a subpopulation of cells from below and above the intermediary EWSR1-FLI1 activity is characterized by increased hypoxia. Overall, our study reveals sources of intratumoral heterogeneity within EwS tumors.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/genetics , Transcription, Genetic/genetics , Cell Line, Tumor , Humans , Signal Transduction
13.
Am J Pathol ; 188(10): 2378-2391, 2018 10.
Article in English | MEDLINE | ID: mdl-30075151

ABSTRACT

Medullary breast carcinoma (MBC) is a rare subtype of triple-negative breast cancer with specific genomic features within the spectrum of basal-like carcinoma (BLC). In this study of 19 MBCs and 36 non-MBC BLCs, we refined the transcriptomic and genomic knowledge about this entity. Unsupervised and supervised analysis of transcriptomic profiles confirmed that MBC clearly differs from non-MBC BLC, with 92 genes overexpressed and 154 genes underexpressed in MBC compared with non-MBC BLC. Immunity-related pathways are the most differentially represented pathways in MBC compared with non-MBC BLC. The proapoptotic gene BCLG (official name BCL2L14) is by far the most intensely overexpressed gene in MBC. A quantitative RT-PCR validation study conducted in 526 breast tumors corresponding to all molecular subtypes documented the specificity of BCLG overexpression in MBC, which was confirmed at the protein level by immunohistochemistry. We also found that most MBCs belong to the immunomodulatory triple-negative breast cancer subtype. Using pan-genomic analysis, it was found that MBC harbors more losses of heterozygosity than non-MBC BLC. These observations corroborate the notion that MBC remains a distinct entity that could benefit from specific treatment strategies (such as deescalation or targeted therapy) adapted to this rare tumor type.


Subject(s)
Carcinoma, Medullary/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Triple Negative Breast Neoplasms/genetics , BRCA2 Protein/genetics , DNA, Neoplasm/metabolism , Female , Gene Expression Profiling , Genes, Neoplasm/genetics , Humans , Loss of Heterozygosity/genetics , RNA, Neoplasm/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics
14.
Breast Cancer Res ; 18(1): 23, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26887652

ABSTRACT

BACKGROUND: Polarity defects are a hallmark of most carcinomas. Cells from invasive micropapillary carcinomas (IMPCs) of the breast are characterized by a striking cell polarity inversion and represent an interesting model for the analysis of polarity abnormalities. METHODS: In-depth investigation of polarity proteins in 24 IMPCs and a gene expression profiling, comparing IMPC (n = 73) with invasive carcinomas of no special type (ICNST) (n = 51) have been performed. RESULTS: IMPCs showed a profound disorganization of the investigated polarity proteins and revealed major abnormalities in their subcellular localization. Gene expression profiling experiments highlighted a number of deregulated genes in the IMPCs that have a role in apico-basal polarity, adhesion and migration. LIN7A, a Crumbs-complex polarity gene, was one of the most differentially over-expressed genes in the IMPCs. Upon LIN7A over-expression, we observed hyperproliferation, invasion and a complete absence of lumen formation, revealing strong polarity defects. CONCLUSION: This study therefore shows that LIN7A has a crucial role in the polarity abnormalities associated with breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Polarity/genetics , Membrane Proteins/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Vesicular Transport Proteins
15.
Genes Dev ; 29(24): 2547-62, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26637281

ABSTRACT

Alterations of chromatin modifiers are frequent in cancer, but their functional consequences often remain unclear. Focusing on the Polycomb protein EZH2 that deposits the H3K27me3 (trimethylation of Lys27 of histone H3) mark, we showed that its high expression in solid tumors is a consequence, not a cause, of tumorigenesis. In mouse and human models, EZH2 is dispensable for prostate cancer development and restrains breast tumorigenesis. High EZH2 expression in tumors results from a tight coupling to proliferation to ensure H3K27me3 homeostasis. However, this process malfunctions in breast cancer. Low EZH2 expression relative to proliferation and mutations in Polycomb genes actually indicate poor prognosis and occur in metastases. We show that while altered EZH2 activity consistently modulates a subset of its target genes, it promotes a wider transcriptional instability. Importantly, transcriptional changes that are consequences of EZH2 loss are predominantly irreversible. Our study provides an unexpected understanding of EZH2's contribution to solid tumors with important therapeutic implications.


Subject(s)
Breast Neoplasms/enzymology , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , Polycomb Repressive Complex 2/metabolism , Animals , Animals, Genetically Modified , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Female , Histones/metabolism , Homeostasis/genetics , Humans , Male , Polycomb Repressive Complex 2/genetics , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics
16.
Oncotarget ; 6(34): 35616-24, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26426992

ABSTRACT

We describe the case of a woman carrying a germline pathogenic BRCA1 mutation diagnosed with a breast cancer overexpressing HER2. Clinical presentation of the tumor, HER2-positivity, genomic profile and loss of the mutated BRCA1 allele in tumor evidence that BRCA1 is not inactivated in this breast cancer. It represents the first biological demonstration for the existence of a sporadic HER2-positive breast cancer independent from BRCA loss of function in a woman carrier of a deleterious BRCA1 mutation. In a context where targeted therapies based on BRCA loss of function in the tumor are developed, such case could have direct implications.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Germ-Line Mutation/genetics , Sequence Deletion/genetics , Adult , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/therapy , Chemoradiotherapy , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Genotype , Humans , Pedigree , Polymorphism, Genetic , Receptor, ErbB-2/metabolism
17.
Breast Cancer Res ; 16(3): R46, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24887297

ABSTRACT

INTRODUCTION: Pure invasive micropapillary carcinoma (IMPC) is a special type of breast carcinoma characterised by clusters of cells presenting polarity abnormalities. The biological alterations underlying this pattern remain unknown. METHODS: Pangenomic analysis (n=39), TP53 (n=43) and PIK3CA (n=41) sequencing in a series of IMPCs were performed. A subset of cases was also analysed with whole-exome sequencing (n=4) and RNA sequencing (n=6). Copy number variation profiles were compared with those of oestrogen receptors and grade-matched invasive ductal carcinomas (IDCs) of no special type. RESULTS: Unsupervised analysis of genomic data distinguished two IMPC subsets: one (Sawtooth/8/16) exhibited a significant increase in 16p gains (71%), and the other (Firestorm/Amplifier) was characterised by a high frequency of 8q (35%), 17q (20% to 46%) and 20q (23% to 30%) amplifications and 17p loss (74%). TP53 mutations (10%) were more frequently identified in the amplifier subset, and PIK3CA mutations (4%) were detected in both subsets. Compared to IDC, IMPC exhibited specific loss of the 6q16-q22 region (45%), which is associated with downregulation of FOXO3 and SEC63 gene expression. SEC63 and FOXO3 missense mutations were identified in one case each (2%). Whole-exome sequencing combined with RNA sequencing of IMPC allowed us to identify somatic mutations in genes involved in polarity, DNAH9 and FMN2 (8% and 2%, respectively) or ciliogenesis, BBS12 and BBS9 (2% each) or genes coding for endoplasmic reticulum protein, HSP90B1 and SPTLC3 (2% each) and cytoskeleton, UBR4 and PTPN21 (2% each), regardless of the genomic subset. The intracellular biological function of the mutated genes identified by gene ontology analysis suggests a driving role in the clinicopathological characteristics of IMPC. CONCLUSION: In our comprehensive molecular analysis of IMPC, we identified numerous genomic alterations without any recurrent fusion genes. Recurrent somatic mutations of genes participating in cellular polarity and shape suggest that they, together with other biological alterations (such as epigenetic modifications and stromal alterations), could contribute to the morphological pattern of IMPC. Though none of the individual abnormalities demonstrated specificity for IMPC, whether their combination in IMPC may have a cumulative effect that drives the abnormal polarity of IMPC needs to be examined further with in vitro experiments.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Cell Polarity/genetics , Neoplasm Invasiveness/genetics , Axonemal Dyneins/genetics , Base Sequence , Breast/pathology , Breast Neoplasms/pathology , Calmodulin-Binding Proteins/genetics , Carcinoma, Ductal, Breast/pathology , Chaperonins , Class I Phosphatidylinositol 3-Kinases , Cytoskeletal Proteins/genetics , DNA Copy Number Variations , Exome/genetics , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Formins , Gene Amplification/genetics , Group II Chaperonins/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microfilament Proteins/biosynthesis , Molecular Chaperones , Mutation, Missense , Neoplasm Proteins/genetics , Nuclear Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA-Binding Proteins , Receptor, ErbB-2/biosynthesis , Receptors, Estrogen/biosynthesis , Retrospective Studies , Sequence Analysis, DNA , Sequence Analysis, RNA , Sequence Deletion/genetics , Serine C-Palmitoyltransferase/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases
18.
Carcinogenesis ; 35(3): 670-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24148822

ABSTRACT

RNA interference has boosted the field of functional genomics, by making it possible to carry out 'loss-of-function' screens in cultured cells. Here, we performed a small interfering RNA screening, in three breast cancer cell lines, for 101 candidate driver genes overexpressed in amplified breast tumors and belonging to eight amplicons on chromosomes 8q and 17q, investigating their role in cell survival/proliferation. This screening identified eight driver genes that were amplified, overexpressed and critical for breast tumor cell proliferation or survival. They included the well-described oncogenic driver genes for the 17q12 amplicon, ERBB2 and GRB7. Four of six other candidate driver genes-RAD21 and EIF3H, both on chromosome 8q23, CHRAC1 on chromosome 8q24.3 and TANC2 on chromosome 17q23-were confirmed to be driver genes regulating the proliferation/survival of clonogenic breast cancer cells presenting an amplification of the corresponding region. Indeed, knockdown of the expression of these genes decreased cell viability, through both cell cycle arrest and apoptosis induction, and inhibited the formation of colonies in anchorage-independent conditions, in soft agar. Strategies for inhibiting the expression of these genes or the function of the proteins they encode are therefore of potential value for the treatment of breast cancers presenting amplifications of the corresponding genomic region.


Subject(s)
Breast Neoplasms/genetics , Cell Division/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 8 , RNA, Small Interfering/genetics , Base Sequence , Breast Neoplasms/pathology , Cell Cycle Proteins , DNA Primers , DNA-Binding Proteins/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Nuclear Proteins/genetics , Nucleoproteins/genetics , Phosphoproteins/genetics , Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
PLoS One ; 8(10): e76496, 2013.
Article in English | MEDLINE | ID: mdl-24143191

ABSTRACT

BACKGROUND: The accurate prognosis definition to tailor treatment for early luminal invasive breast carcinoma patients remains challenging. MATERIALS AND METHODS: Two hundred fourteen early luminal breast carcinomas were genotyped with single nucleotide polymorphisms (SNPs) array to determine the number of chromosomal breakpoints as a marker of genomic instability. Proliferation was assessed by KI67 (immunohistochemistry) and genomic grade index (transcriptomic analysis). IHC3 (IHC4 score for HER2 negative tumors) was also determined. RESULTS: In the training set (109 cases), the optimal cut-off was 34 breakpoints with a specificity of 0.94 and a sensitivity of 0.57 (Area under the curve (AUC): 0.81[0.71; 0.91]). In the validation set (105 cases), the outcome of patients with > 34 breakpoints (11 events / 22 patients) was poorer (logrank test p < 0.001; Relative Risk (RR): 3.7 [1.73; 7.92]), than that of patients with < 34 breakpoints (19 events / 83 patients).Whereas genomic grade and KI67 had a significant prognostic value in univariate analysis in contrast to IHC3 that failed to have a statistical significant prognostic value in this series, the number of breakpoints remained the only significant parameter predictive of outcome (RR: 3.47, Confidence Interval (CI [1.29; 9.31], p = 0.014)) in multivariate analysis . CONCLUSION: Genomic instability, defined herein as a high number of chromosomal breakpoints, in early stage luminal breast carcinoma is a stronger prognostic marker than proliferation.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Genomic Instability , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Chromosome Breakpoints , Computational Biology , Disease-Free Survival , Female , Genotyping Techniques , Humans , Ki-67 Antigen/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Polymorphism, Single Nucleotide , Reproducibility of Results
20.
Am J Pathol ; 183(5): 1634-1644, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24051013

ABSTRACT

Amplification of the 8p11-12 chromosomal region is a common genetic event in many epithelial cancers. In breast cancer, several genes within this region have been shown to display oncogenic activity. Among these genes, the enzyme-encoding genes, PPAPDC1B and WHSC1L1, have been identified as potential therapeutic targets. We investigated whether PPAPDC1B and WHSC1L1 acted as general driver genes, thereby serving as therapeutic targets in other tumors with 8p11-12 amplification. By using publicly available genomic data from a panel of 883 cell lines derived from different cancers, we identified the cell lines presenting amplification of both WHSC1L1 and PPAPDC1B. In particular, we focused on cell lines derived from lung cancer and pancreatic adenocarcinoma and found a correlation between the amplification of PPAPDC1B and WHSC1L1 with their overexpression. Loss-of-function studies based on the use of siRNA and shRNA demonstrated that PPAPDC1B and WHSC1L1 played a major role in regulating the survival of pancreatic adenocarcinoma and small-cell lung cancer-derived cell lines, both in anchorage-dependent and anchorage-independent conditions, displaying amplification and overexpression of these genes. We also demonstrated that PPAPDC1B and WHSC1L1 regulated xenograft growth in these cell lines. Finally, quantitative RT-PCR experiments after PPAPDC1B and WHSC1L1 knockdown revealed exclusive PPAPDC1B and WHSC1L1 gene targets in small-cell lung cancer and pancreatic adenocarcinoma-derived cell lines compared with breast cancer.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 8/genetics , Gene Amplification , Histone-Lysine N-Methyltransferase/genetics , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Phosphatidate Phosphatase/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Breast Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...