Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mathilde Sibony is active.

Publication


Featured researches published by Mathilde Sibony.


American Journal of Pathology | 2003

Angiopoietin-Like 4 Is a Proangiogenic Factor Produced during Ischemia and in Conventional Renal Cell Carcinoma

Sébastien Le Jan; Céline Amy; Aurélie Cazes; Catherine Monnot; Noël Lamandé; Judith Favier; Josette Philippe; Mathilde Sibony; Jean-Marie Gasc; Pierre Corvol; Stéphane Germain

Ischemic and solid tumor tissues are less well perfused than normal tissue, leading to metabolic changes and chronic hypoxia, which in turn promotes angiogenesis. We identified human angiopoietin-like 4 (angptl4) as a gene with hypoxia-induced expression in endothelial cells. We showed that the levels of both mRNA and protein for ANGPTL4 increased in response to hypoxia. When tested in the chicken chorioallantoic membrane assay, ANGPTL4 induced a strong proangiogenic response, independently of vascular endothelial growth factor. In human pathology, ANGPTL4 mRNA is produced in ischemic tissues, in conditions such as critical leg ischemia. In tumors, ANGPTL4 is produced in the hypoxic areas surrounding necrotic regions. We observed particularly high levels of ANGPTL4 mRNA in tumor cells of conventional renal cell carcinoma. Other benign and malignant renal tumor cells do not produce ANGPTL4 mRNA. This molecule therefore seems to be a marker of conventional renal cell carcinoma. ANGPTL4, originally identified as a peroxisome proliferator-activated receptor alpha and gamma target gene, has potential for use as a new diagnostic tool and a potential therapeutic target, modulating angiogenesis both in tumors and in ischemic tissues. This study also suggests that ANGPTL4 may provide a link between metabolic disorders and hypoxia-induced angiogenesis.


Journal of Clinical Oncology | 2005

Promoter hypermethylation is associated with tumor location, stage, and subsequent progression in transitional cell carcinoma.

James Catto; A.R. Azzouzi; Ishtiaq Rehman; Kenneth M. Feeley; Simon S. Cross; Najla Amira; Gaëlle Fromont; Mathilde Sibony; Oliver Cussenot; Mark Meuth; Freddie C. Hamdy

PURPOSE Transitional cell carcinoma (TCC) is a pan-urothelial disease characterized by multiplicity. Although little is known about the molecular events in upper-tract TCC, similar carcinogenic mechanisms are thought to occur throughout the urinary tract. However, we have previously shown that distinct patterns of microsatellite instability occur in upper and lower urinary tract TCC, suggesting biologic differences between these tumors. Here we investigate the extent of promoter hypermethylation in TCC throughout the urinary tract. PATIENTS AND METHODS Tissue was obtained from 280 patients (median follow-up, 56 months) whose tumors comprised 116 bladder and 164 upper-tract tumors (UTT). Analysis for hypermethylation at 11 CpG islands, using methylation-sensitive polymerase chain reaction and bisulfite sequencing, was performed for each sample and compared with the tumors clinicopathologic details, microsatellite instability status, and subsequent behavior. RESULTS Promoter methylation was present in 86% of TCC and occurred both more frequently and more extensively in UTT (94%) than in bladder tumors (76%; P < .0001). Methylation was associated with advanced tumor stage (P = .0001) and higher tumor progression (P = .03) and mortality rates (P = .04), when compared with tumors without methylation. Multivariate analysis revealed that methylation at the RASSF1A and DAPK loci, in addition to tumor stage and grade, were associated with disease progression (P < .04). CONCLUSION Despite morphologic similarities, there are genetic and epigenetic differences between TCC in the upper and lower urinary tracts. Methylation occurs commonly in urinary tract tumors, may affect carcinogenic mechanisms, and is a prognostic marker and a potential therapeutic target.


The American Journal of Surgical Pathology | 2008

Renal translocation carcinomas: clinicopathologic, immunohistochemical, and gene expression profiling analysis of 31 cases with a review of the literature.

Philippe Camparo; Viorel Vasiliu; Vincent Molinié; Jérôme Couturier; Karl Dykema; David Petillo; Kyle A. Furge; Eva Comperat; Marick Laé; Raymonde Bouvier; Liliane Boccon-Gibod; Yves Denoux; Sophie Ferlicot; Eric Forest; Gaëlle Fromont; Marie C. Hintzy; Myriam Laghouati; Mathilde Sibony; Marie L. Tucker; Nina Weber; Bin Tean Teh; Annick Vieillefond

We report clinicopathologic features of a large series of renal translocation carcinomas from a multicentric study. Diagnosis was performed by cytogenetic examination of fresh material and/or by immunochemistry with antibodies directed against the C-terminal part of transcription factor E3 (TFE3) and native transcription factor EB (TFEB) proteins. Clinical data, follow-up, and histologic features were assessed. Antibodies against CK7, CD10, vimentin, epithelial membrane antigen, AE1-AE3, E-cadherin, α-methylacyl-coenzyme A racemase, melan A, and HMB45 were tested on tissue microarrays. Whole-genome microarray expression profiling was performed on 4 tumors. Twenty-nine cases were diagnosed as TFE3 and 2 as TFEB renal translocation carcinomas, including 13 males and 18 females, mean age 24.6 years. Two patients had a previous history of chemotherapy and 1 had a history of renal failure. Mean size of the tumor was 6.9 cm. Thirteen cases were ≥pT3 stage. Twelve cases were N+ or M+. Mean follow-up was 29.5 months. Three patients presented metastases and 5 have died. Mixed papillary and nested patterns with clear and/or eosinophilic cells represented the most consistent histologic appearance, with common foci of calcifications regardless of the type of translocation. Using a 30 mn incubation at room temperature, TFE3 immunostainings were positive in only 82% of our TFE3 translocation carcinomas. Both TFE3 and TFEB renal translocation carcinomas expressed CD10 and α-methylacyl-coenzyme A racemase in all cases. An expression of E-cadherin was observed in two-third of cases. Cytokeratins were expressed in less than one-third of cases. Melanocytic markers were expressed at least weakly in all cases except two. Unsupervised clustering on the basis of the gene expression profiling indicated a distinct subgroup of tumors. TRIM 63 glutathione S-transferase A1 and alanyl aminopeptidase are the main differentially expressed genes for this group of tumors. Our results suggest that these differentially expressed genes may serve as novel diagnostic or prognostic markers.


The New England Journal of Medicine | 2013

ARMC5 Mutations in Macronodular Adrenal Hyperplasia with Cushing's Syndrome

Guillaume Assié; Rossella Libé; Stéphanie Espiard; Marthe Rizk-Rabin; Anne Guimier; Windy Luscap; Olivia Barreau; Lucile Lefèvre; Mathilde Sibony; Laurence Guignat; S. Rodriguez; Karine Perlemoine; F. René-Corail; Franck Letourneur; Bilal Trabulsi; Alix Poussier; Nathalie Chabbert-Buffet; Françoise Borson-Chazot; Lionel Groussin; Xavier Bertagna; Constantine A. Stratakis; Bruno Ragazzon; Jérôme Bertherat; Abstr Act

BACKGROUND Corticotropin-independent macronodular adrenal hyperplasia may be an incidental finding or it may be identified during evaluation for Cushings syndrome. Reports of familial cases and the involvement of both adrenal glands suggest a genetic origin of this condition. METHODS We genotyped blood and tumor DNA obtained from 33 patients with corticotropin-independent macronodular adrenal hyperplasia (12 men and 21 women who were 30 to 73 years of age), using single-nucleotide polymorphism arrays, microsatellite markers, and whole-genome and Sanger sequencing. The effects of armadillo repeat containing 5 (ARMC5) inactivation and overexpression were tested in cell-culture models. RESULTS The most frequent somatic chromosome alteration was loss of heterozygosity at 16p (in 8 of 33 patients for whom data were available [24%]). The most frequent mutation identified by means of whole-genome sequencing was in ARMC5, located at 16p11.2. ARMC5 mutations were detected in tumors obtained from 18 of 33 patients (55%). In all cases, both alleles of ARMC5 carried mutations: one germline and the other somatic. In 4 patients with a germline ARMC5 mutation, different nodules from the affected adrenals harbored different secondary ARMC5 alterations. Transcriptome-based classification of corticotropin-independent macronodular adrenal hyperplasia indicated that ARMC5 mutations influenced gene expression, since all cases with mutations clustered together. ARMC5 inactivation decreased steroidogenesis in vitro, and its overexpression altered cell survival. CONCLUSIONS Some cases of corticotropin-independent macronodular adrenal hyperplasia appear to be genetic, most often with inactivating mutations of ARMC5, a putative tumor-suppressor gene. Genetic testing for this condition, which often has a long and insidious prediagnostic course, might result in earlier identification and better management. (Funded by Agence Nationale de la Recherche and others.).


Oncogene | 2003

Involvement of aquaporins in colorectal carcinogenesis

Chulso Moon; Se Jin Jang; Juna Lee; Mohammad O. Hoque; Mathilde Sibony; Barry Trink; Yoon Soo Chang; David Sidransky; Li Mao

Aquaporins (AQPs) are important in controlling water permeability. As AQP1 is known as a serum-responsive gene, we hypothesized that AQP expression may be involved in the development of human cancer. By reverse transcriptase–polymerase chain reaction analysis, expression of AQPs 1, 3, and 5 was found in seven colon and colorectal cancer cell lines. Western blot analysis confirmed their expression in four of these cell lines. In situ hybridization demonstrated that during colorectal carcinogenesis, the expression of AQPs 1 and 5 was induced in early-stage disease (early dysplasia) and maintained through the late stages of colon cancer development. Expression of AQPs 1 and 5 was maintained even in metastatic lesions in the liver. These findings demonstrate that the expression of several AQPs is found in tumor cells and is associated with an early stage of colorectal cancer development. These novel observations suggest that multiple AQP expression may be advantageous to tumorigenesis, which may lead to a better understanding of colorectal carcinogenesis.


Oncogene | 2003

Distinct patterns of microsatellite instability are seen in tumours of the urinary tract.

James Catto; A.R. Azzouzi; Najla Amira; Ishtiaq Rehman; Kenneth M. Feeley; Simon S. Cross; Gaëlle Fromont; Mathilde Sibony; Freddie C. Hamdy; Oliver Cussenot; Mark Meuth

To date, two forms of microsatellite instability (MSI) have been described in human cancer. MSI typical of hereditary nonpolyposis colon cancer (HNPCC), is due to deficient DNA mismatch repair (MMR) and is defined with mono- and dinucleotide repeat microsatellites. A second variety of instability is best seen at selective tetranucleotide repeats (EMAST; elevated microsatellite alterations at select tetranucleotides). While MSI occurs infrequently in bladder cancers, EMAST is common. Sporadic tumours with the largest proportion showing MSI are those found most frequently in HNPCC kindreds. While bladder cancer is not frequently seen in HNPCC, upper urinary tract tumours (UTTs) are. Having previously found a low frequency of MSI in bladder cancer, we sought to determine the relative levels of MSI and EMAST in transitional cell carcinoma (TCC) of the upper and lower urinary tracts. Microsatellite analysis was performed at 10 mono- and dinucleotide and eight tetranucleotide loci, in 89 bladder and 71 UTT TCC. Contrasting patterns of instability were seen in urinary tumours. In bladder cancer, MSI was rare and EMAST was common. The presence of EMAST was not related to tumour grade, stage, subsequent outcome or immunohistochemical expression of the MMR proteins. In UTT, while MSI occurred frequently, EMAST was seen less frequently than in bladder cancer. When TCC of the upper and lower urinary tracts are compared, MSI-H is more frequent in UTT and EMAST more frequent in bladder cancer. Our findings show that, as for colorectal cancer, the pattern of MSI varies with location in the urinary tract. In addition, we have confirmed that MSI and EMAST are discrete forms of MSI, and that the presence of EMAST does not affect tumour phenotype.


Science Translational Medicine | 2014

EGFR as a potential therapeutic target for a subset of muscle-invasive bladder cancers presenting a basal-like phenotype

Sandra Rebouissou; Isabelle Bernard-Pierrot; Aurélien de Reyniès; May-Linda Lepage; Clémentine Krucker; Elodie Chapeaublanc; Aurélie Hérault; Aurélie Kamoun; Aurélie Caillault; Eric Letouzé; Nabila Elarouci; Yann Neuzillet; Yves Denoux; Vincent Molinié; Dimitri Vordos; Agnès Laplanche; Pascale Maillé; Karina Ofualuka; Fabien Reyal; Anne Biton; Mathilde Sibony; Xavier Paoletti; Jennifer Southgate; Simone Benhamou; Thierry Lebret; Yves Allory; François Radvanyi

A subtype of aggressive human muscle-invasive bladder cancer expresses basal epithelial markers and is sensitive to EGFR inhibition in preclinical models. Bladder Cancer’s Basal Instincts Like most cancers, bladder tumors are much easier to treat when they do not invade deep into the tissue, accounting for the poor outcomes of patients with muscle-invasive bladder cancer. By performing genetic analysis on a large number of these tumors, Rebouissou et al. identified a specific subgroup of muscle-invasive bladder cancers expressing basal markers. Although these are aggressive tumors, the authors showed that they have a weak spot and are unusually dependent on the activity of a signaling pathway called epidermal growth factor receptor (EGFR). As a result, these tumors are sensitive to treatment with drugs that inhibit the EGFR pathway, which the authors confirmed in preclinical models. Muscle-invasive bladder carcinoma (MIBC) constitutes a heterogeneous group of tumors with a poor outcome. Molecular stratification of MIBC may identify clinically relevant tumor subgroups and help to provide effective targeted therapies. From seven series of large-scale transcriptomic data (383 tumors), we identified an MIBC subgroup accounting for 23.5% of MIBC, associated with shorter survival and displaying a basal-like phenotype, as shown by the expression of epithelial basal cell markers. Basal-like tumors presented an activation of the epidermal growth factor receptor (EGFR) pathway linked to frequent EGFR gains and activation of an EGFR autocrine loop. We used a 40-gene expression classifier derived from human tumors to identify human bladder cancer cell lines and a chemically induced mouse model of bladder cancer corresponding to human basal-like bladder cancer. We showed, in both models, that tumor cells were sensitive to anti-EGFR therapy. Our findings provide preclinical proof of concept that anti-EGFR therapy can be used to target a subset of particularly aggressive MIBC tumors expressing basal cell markers and provide diagnostic tools for identifying these tumors.


The American Journal of Surgical Pathology | 2014

Succinate Dehydrogenase (SDH)-deficient Renal Carcinoma: A Morphologically Distinct Entity: A Clinicopathologic Series of 36 Tumors From 27 Patients

Anthony J. Gill; Ondřej Hes; Thomas G. Papathomas; Monika Sedivcova; Puay Hoon Tan; Abbas Agaimy; Per Arne Andresen; Andrew Kedziora; Adele Clarkson; Christopher W. Toon; Loretta Sioson; Nicole Watson; Angela Chou; Julie Y. Paik; Roderick J. Clifton-Bligh; Bruce G. Robinson; Diana E. Benn; Kirsten Hills; Fiona Maclean; Nicolasine D. Niemeijer; Ljiljana Vlatkovic; Arndt Hartmann; Eleonora P. M. Corssmit; Geert J.L.H. van Leenders; Christopher G. Przybycin; Jesse K. McKenney; Cristina Magi-Galluzzi; Asli Yilmaz; Darryl Yu; Katherine D. Nicoll

Succinate dehydrogenase (SDH)-deficient renal carcinoma has been accepted as a provisional entity in the 2013 International Society of Urological Pathology Vancouver Classification. To further define its morphologic and clinical features, we studied a multi-institutional cohort of 36 SDH-deficient renal carcinomas from 27 patients, including 21 previously unreported cases. We estimate that 0.05% to 0.2% of all renal carcinomas are SDH deficient. Mean patient age at presentation was 37 years (range, 14 to 76 y), with a slight male predominance (M:F=1.7:1). Bilateral tumors were observed in 26% of patients. Thirty-four (94%) tumors demonstrated the previously reported morphology at least focally, which included: solid or focally cystic growth, uniform cytology with eosinophilic flocculent cytoplasm, intracytoplasmic vacuolations and inclusions, and round to oval low-grade nuclei. All 17 patients who underwent genetic testing for mutation in the SDH subunits demonstrated germline mutations (16 in SDHB and 1 in SDHC). Nine of 27 (33%) patients developed metastatic disease, 2 of them after prolonged follow-up (5.5 and 30 y). Seven of 10 patients (70%) with high-grade nuclei metastasized as did all 4 patients with coagulative necrosis. Two of 17 (12%) patients with low-grade nuclei metastasized, and both had unbiopsied contralateral tumors, which may have been the origin of the metastatic disease. In conclusion, SDH-deficient renal carcinoma is a rare and unique type of renal carcinoma, exhibiting stereotypical morphologic features in the great majority of cases and showing a strong relationship with SDH germline mutation. Although this tumor may undergo dedifferentiation and metastasize, sometimes after a prolonged delay, metastatic disease is rare in the absence of high-grade nuclear atypia or coagulative necrosis.


Virchows Archiv | 2005

Mucinous tubular and spindle cell carcinoma: a report of 15 cases and a review of the literature

Sophie Ferlicot; Yves Allory; Eva Comperat; Florence Mege-Lechevalier; Stéphanie Dimet; Mathilde Sibony; Jérôme Couturier; Annick Vieillefond

Mucinous tubular and spindle cell carcinomas are low-grade renal epithelial neoplasms, which were first recognized as a specific entity in the World Health Organization 2004 classification. Forty-five documented cases have been reported. We present 15 additional cases that were incidentally discovered in ten women and five men, with a mean age of 53 years. The tumor is characteristically made up of large eosinophilic regular spindle cells separated by a myxoid stroma with intercellular alcian-blue-positive clear droplets. In peripheral areas, elongated tubules and papillae covered by cubic cells are found. Until this entity had been defined, pathologists used to classify these tumors as variants of solid papillary carcinomas with compressed and elongated papillae, metanephric adenomas, and sarcomatoid carcinomas. In the literature, cytogenetic data indicate various chromosomal losses and gains, but no loss of 3p or trisomy 7 and/or trisomy 17. In two cases, we demonstrate chromosomal loss involving chromosomes 1, 4, 6, 11, 8, 13, 14, 15, 18, and 22. In our 15 cases, immunohistochemistry favored a distal tubule origin (EMA+, AE1/AE3+, CK7+, CK19+, E-cadherin+, AMACR+, and CD10−). Prognosis was favorable in our cases, while in the literature, two metastatic cases were reported. Further investigations are required to determine the frequency and true prognosis of these tumors, which are easily identifiable morphologically.


Clinical Cancer Research | 2015

Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting

Benoit Beuselinck; Sylvie Job; Etienne Becht; Alexandra Karadimou; Virginie Verkarre; Gabrielle Couchy; Nicolas A. Giraldo; Nathalie Rioux-Leclercq; Vincent Molinié; Mathilde Sibony; Reza Elaidi; Corinne Teghom; Jean Jacques Patard; Arnaud Mejean; Wolf-Herman Fridman; Aurélien de Reyniès; S. Oudard; Jessica Zucman-Rossi

Purpose: Selecting patients with metastatic clear-cell renal cell carcinoma (m-ccRCC) who might benefit from treatment with targeted tyrosine kinase inhibitors (TKI) is a challenge. Our aim was to identify molecular markers associated with outcome in patients with m-ccRCC treated with sunitinib. Experimental Design: We performed global transcriptome analyses on 53 primary resected ccRCC tumors from patients who developed metastatic disease and were treated with first-line sunitinib. We also determined chromosome copy-number aberrations, methylation status, and gene mutations in von Hippel–Lindau and PBRM1. Molecular data were analyzed in relation with response rate (RR), progression-free survival (PFS), and overall survival (OS). Validation was performed in 47 additional ccRCC samples treated in first-line metastatic setting with sunitinib. Results: Unsupervised transcriptome analysis identified 4 robust ccRCC subtypes (ccrcc1 to 4) related to previous molecular classifications that were associated with different responses to sunitinib treatment. ccrcc1/ccrcc4 tumors had a lower RR (P = 0.005) and a shorter PFS and OS than ccrcc2/ccrcc3 tumors (P = 0.001 and 0.0003, respectively). These subtypes were the only significant covariate in the multivariate Cox model for PFS and OS (P = 0.017 and 0.006, respectively). ccrcc1/ccrcc4 tumors were characterized by a stem-cell polycomb signature and CpG hypermethylation, whereas ccrcc3 tumors, sensitive to sunitinib, did not exhibit cellular response to hypoxia. Moreover, ccrcc4 tumors exhibited sarcomatoid differentiation with a strong inflammatory, Th1-oriented but suppressive immune microenvironment, with high expression of PDCD1 (PD-1) and its ligands. Conclusions: ccRCC molecular subtypes are predictive of sunitinib response in metastatic patients, and could be used for personalized mRCC treatment with TKIs, demethylating or immunomodulatory drugs. Clin Cancer Res; 21(6); 1329–39. ©2015 AACR.

Collaboration


Dive into the Mathilde Sibony's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Zerbib

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gaëlle Fromont

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bertrand Dousset

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge