Network


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

Hotspot


Dive into the research topics where E. Marangoni is active.

Publication


Featured researches published by E. Marangoni.


Breast Cancer Research and Treatment | 2012

Modeling of response to endocrine therapy in a panel of human luminal breast cancer xenografts

P. Cottu; E. Marangoni; Franck Assayag; P. de Cremoux; Anne Vincent-Salomon; Ch. Guyader; L. De Plater; C. Elbaz; Narjesse Karboul; J.J. Fontaine; Sophie Chateau-Joubert; P. Boudou-Rouquette; S. Alran; Virginie Dangles-Marie; David Gentien; M.-F. Poupon; Didier Decaudin

Resistance to endocrine therapy is a major complication of luminal breast cancer and studies of the biological features of hormonal resistance are limited by the lack of adequate preclinical models. The aim of this study is to establish and characterize a panel of primary human luminal breast carcinoma xenografts, and to evaluate their response to endocrine therapies. Four hundred and twenty-three tumor fragments obtained directly from patients have been grafted in the interscapular fatpad of Swiss nude mice. After stable engraftment with estradiol supplementation, xenografted tumors have been validated by conventional pathology and immunohistochemistry examination, and additional molecular studies. In vivo tumor growth and response to different endocrine treatments were evaluated. We have engrafted 423 tumors including 314 ER+ tumors, and 8 new luminal breast cancer xenografts have been obtained (2.5%). Tumor take was much lower for luminal tumors than for non-luminal tumors (2.5 vs. 24.7%, Pxa0<xa00.0001), and was associated with two independent criteria, i.e., ER status (Pxa0<xa00.0001) and a high grade tumor (Pxa0=xa00.05). Histological and immunohistochemical analyses performed on patient’s tumors and xenografts showed striking similarities in the tumor morphology as well as in the expression level of ER, PR, and HER2. Response to hormone therapy, evaluated in 6 luminal models, showed different sensitivities, thus exhibiting heterogeneity similar to what is observed in the clinic. We have established a panel of primary human luminal breast cancer xenografts, recapitulating the biological and clinical behaviors of patient tumors, and therefore suitable for further preclinical experiments.


British Journal of Cancer | 2010

Establishment and characterisation of a new breast cancer xenograft obtained from a woman carrying a germline BRCA2 mutation.

L. De Plater; A. Laugé; Charlotte Guyader; M.-F. Poupon; Franck Assayag; P. de Cremoux; Anne Vincent-Salomon; Dominique Stoppa-Lyonnet; Brigitte Sigal-Zafrani; J.J. Fontaine; Rachel Brough; Christopher J. Lord; Alan Ashworth; P. Cottu; Didier Decaudin; E. Marangoni

Background:The BRCA2 gene is responsible for a high number of hereditary breast and ovarian cancers, and studies of the BRCA2 biological functions are limited by the lack of models that resemble the patients tumour features. The aim of this study was to establish and characterise a new human breast carcinoma xenograft obtained from a woman carrying a germline BRCA2 mutation.Methods:A transplantable xenograft was obtained by grafting a breast cancer sample into nude mice. The biological and genetic profiles of the xenograft were compared with that of the patients tumour using histology, immunohistochemistry (IHC), BRCA2 sequencing, comparative genomic hybridisation (CGH), and qRT–PCR. Tumour response to standard chemotherapies was evaluated.Results:Histological profile identified the tumour as a basal-like triple-negative breast cancer. Targeted BRCA2 DNA sequencing of the xenograft showed the presence of the mutation previously identified in the carrier. Comparative genomic hybridisation array profiles of the primary tumour and the xenograft revealed a high number of similar genetic alterations. The therapeutic assessment of the xenograft showed sensitivity to anthracyclin-based chemotherapy and resistance to docetaxel. The xenograft was also highly sensitive to radiotherapy and cisplatin-based treatments.Conclusions:This study describes a new human breast cancer xenograft obtained from a BRCA2-mutated patient. This xenograft provides a new model for the pre-clinical drug development and for the exploration of the drug response biological basis.


Molecular Cancer Therapeutics | 2016

Chromatin regulators as a guide for cancer treatment choice

Zachary A. Gurard-Levin; Laurence Wilson; Vera Pancaldi; Sophie Postel-Vinay; Fabricio G. Sousa; Cécile Reyes; E. Marangoni; David Gentien; Alfonso Valencia; Yves Pommier; Paul Cottu; Geneviève Almouzni

The limited capacity to predict a patients response to distinct chemotherapeutic agents is a major hurdle in cancer management. The efficiency of a large fraction of current cancer therapeutics (radio- and chemotherapies) is influenced by chromatin structure. Reciprocally, alterations in chromatin organization may affect resistance mechanisms. Here, we explore how the misexpression of chromatin regulators—factors involved in the establishment and maintenance of functional chromatin domains—can inform about the extent of docetaxel response. We exploit Affymetrix and NanoString gene expression data for a set of chromatin regulators generated from breast cancer patient-derived xenograft models and patient samples treated with docetaxel. Random Forest classification reveals specific panels of chromatin regulators, including key components of the SWI/SNF chromatin remodeler, which readily distinguish docetaxel high-responders and poor-responders. Further exploration of SWI/SNF components in the comprehensive NCI-60 dataset reveals that the expression inversely correlates with docetaxel sensitivity. Finally, we show that loss of the SWI/SNF subunit BRG1 (SMARCA4) in a model cell line leads to enhanced docetaxel sensitivity. Altogether, our findings point toward chromatin regulators as biomarkers for drug response as well as therapeutic targets to sensitize patients toward docetaxel and combat drug resistance. Mol Cancer Ther; 15(7); 1768–77. ©2016 AACR.


Clinical Cancer Research | 2017

Cytidine Deaminase Deficiency Reveals New Therapeutic Opportunities against Cancer

Hamza Mameri; Ivan Bièche; Didier Meseure; E. Marangoni; Géraldine Buhagiar-Labarchède; André Nicolas; Sophie Vacher; Rosine Onclercq-Delic; Vinodh N. Rajapakse; Sudhir Varma; William C. Reinhold; Yves Pommier; Mounira Amor-Guéret

Purpose: One of the main challenges in cancer therapy is the identification of molecular mechanisms mediating resistance or sensitivity to treatment. Cytidine deaminase (CDA) was reported to be downregulated in cells derived from patients with Bloom syndrome, a genetic disease associated with a strong predisposition to a wide range of cancers. The purpose of this study was to determine whether CDA deficiency could be associated with tumors from the general population and could constitute a predictive marker of susceptibility to antitumor drugs. Experimental Design: We analyzed CDA expression in silico, in large datasets for cancer cell lines and tumors and in various cancer cell lines and primary tumor tissues using IHC, PDXs, qRT-PCR, and Western blotting. We also studied the mechanism underlying CDA silencing and searched for molecules that might target specifically CDA-deficient tumor cells using in silico analysis coupled to classical cellular experimental approaches. Results: We found that CDA expression is downregulated in about 60% of cancer cells and tissues. We demonstrate that DNA methylation is a prevalent mechanism of CDA silencing in tumors. Finally, we show that CDA-deficient tumor cells can be specifically targeted with epigenetic treatments and with the anticancer drug aminoflavone. Conclusions: CDA expression status identifies new subgroups of cancers, and CDA deficiency appears to be a novel and relevant predictive marker of susceptibility to antitumor drugs, opening up new possibilities for treating cancer. Clin Cancer Res; 23(8); 2116–26. ©2016 AACR.


British Journal of Cancer | 2017

Clinical value of R-spondins in triple-negative and metaplastic breast cancers

Florence Coussy; F Lallemand; Sophie Vacher; A Schnitzler; W Chemlali; M Caly; A Nicolas; Sophie Richon; D Meseure; R El Botty; L De-Plater; L Fuhrmann; T Dubois; Sergio Roman-Roman; V Dangles-Marie; E. Marangoni; Ivan Bièche

Background:RSPO ligands, activators of the Wnt/β-catenin pathway, are overexpressed in different cancers. The objective of this study was to investigate the role of RSPOs in breast cancer (BC).Methods:Expression of RSPO and markers of various cancer pathways were measured in breast tumours and cell lines by qRT–PCR. The effect of RSPO on the Wnt/β-catenin pathway activity was determined by luciferase assay, western blotting, and qRT–PCR. The effect of RSPO2 inhibition on proliferation was determined by using RSPO2 siRNAs. The effect of IWR-1, an inhibitor of the Wnt/β-catenin pathway, was examined on the growth of an RSPO2-positive patient-derived xenograft (PDX) model of metaplastic triple-negative BC.Results:We detected RSPO2 and RSPO4 overexpression levels in BC, particularly in triple-negative BC (TNBC), metaplastic BC, and triple-negative cell lines. Various mechanisms could account for this overexpression: presence of fusion transcripts involving RSPO, and amplification or hypomethylation of RSPO genes. Patients with RSPO2-overexpressing tumours have a poorer metastasis-free survival (P=3.6 × 10−4). RSPO2 and RSPO4 stimulate Wnt/β-catenin pathway activity. Inhibition of RSPO expression in a TN cell line inhibits cell growth, and IWR-1 significantly inhibits the growth of an RSPO2-overexpressing PDX.Conclusions:RSPO overexpression could therefore be a new prognostic biomarker and therapeutic target for TNBC.


The Journal of Pathology | 2018

The iron chelator deferasirox synergises with chemotherapy to treat triple-negative breast cancers: Iron deprivation for breast cancer treatment

Sandrine Tury; Franck Assayag; Florian Bonin; Sophie Chateau-Joubert; Jean-Luc Servely; Sophie Vacher; Véronique Becette; Martial Caly; Audrey Rapinat; David Gentien; Pierre de la Grange; Anne Schnitzler; François Lallemand; E. Marangoni; Ivan Bièche; Céline Callens

To ensure their high proliferation rate, tumor cells have an iron metabolic disorder causing them to have increased iron needs, making them more susceptible to iron deprivation. This vulnerability could be a therapeutic target. In breast cancers, the development of new therapeutic approaches is urgently needed for patients with triple‐negative tumors, which frequently relapse after chemotherapy and suffer from a lack of targeted therapies. In this study, we demonstrated that deferasirox (DFX) synergises with standard chemotherapeutic agents such as doxorubicin, cisplatin and carboplatin to inhibit cell proliferation and induce apoptosis and autophagy in triple‐negative breast cancer (TNBC) cells. Moreover, the combination of DFX with doxorubicin and cyclophosphamide delayed recurrences in breast cancer patient‐derived xenografts without increasing the side‐effects of chemotherapies alone or altering the global iron storage of mice. Antitumor synergy of DFX and doxorubicin seems to involve downregulation of the phosphoinositide 3‐kinase and nuclear factor‐κB pathways. Iron deprivation in combination with chemotherapy could thus help to improve the effectiveness of chemotherapy in TNBC patients without increasing toxicity. Copyright


Clinical Cancer Research | 2018

Capecitabine efficacy is correlated with TYMP and RB expression in PDX established from triple-negative breast cancers

E. Marangoni; Cecile Laurent; Florence Coussy; Rania El Botty; Sophie Chateau-Joubert; Jean-Luc Servely; Ludmilla de Plater; Franck Assayag; Ahmed Dahmani; Elodie Montaudon; Fariba Nemati; Justine Fleury; Sophie Vacher; David Gentien; Audrey Rapinat; Pierre Foidart; Nor Eddine Sounni; Agnès Noël; Anne Vincent Salomon; Marick Laé; Didier Decaudin; Sergio Roman-Roman; Ivan Bièche; Martine Piccard; Fabien Reyal

Purpose: Triple-negative breast cancer (TNBC) patients with residual disease after neoadjuvant chemotherapy have a poor outcome. We developed patient-derived xenografts (PDX) from residual tumors to identify efficient chemotherapies and predictive biomarkers in a context of resistance to anthracyclines- and taxanes-based treatments. Experimental Design: PDX were established from residual tumors of primary breast cancer patients treated in neoadjuvant setting. TNBC PDX were treated by anthracyclines, taxanes, platins, and capecitabine. Predictive biomarkers were identified by transcriptomic and immunohistologic analysis. Downregulation of RB1 was performed by siRNA in a cell line established from a PDX. Results: Residual TNBC PDX were characterized by a high tumor take, a short latency, and a poor prognosis of the corresponding patients. With the exception of BRCA1/2-mutated models, residual PDX were resistant to anthracyclines, taxanes, and platins. Capecitabine, the oral prodrug of 5-FU, was highly efficient in 60% of PDX, with two models showing complete responses. Prior treatment of a responder PDX with 5-FU increased expression of thymidylate synthase and decreased efficacy of capecitabine. Transcriptomic and IHC analyses of 32 TNBC PDX, including both residual tumors and treatment-naïve derived tumors, identified RB1 and TYMP proteins as predictive biomarkers for capecitabine response. Finally, RB1 knockdown in a cell line established from a capecitabine-responder PDX decreased sensitivity to 5-FU treatment. Conclusions: We identified capecitabine as efficient chemotherapy in TNBC PDX models established from residual disease and resistant to anthracyclines, taxanes, and platins. RB1 positivity and high expression of TYMP were significantly associated with capecitabine response. Clin Cancer Res; 24(11); 2605–15. ©2018 AACR.


American Journal of Pathology | 2018

Medullary Breast Carcinoma, a Triple-Negative Breast Cancer Associated with BCLG Overexpression

Pierre Romero; Vanessa Benhamo; Gabrielle Deniziaut; Laetitia Fuhrmann; Frédérique Berger; Elodie Manié; Jaydutt Bhalshankar; Sophie Vacher; Cécile Laurent; E. Marangoni; Nadège Gruel; Gaëtan MacGrogan; Roman Rouzier; Olivier Delattre; Tatiana Popova; Fabien Reyal; Marc-Henri Stern; Dominique Stoppa-Lyonnet; Caterina Marchiò; Ivan Bièche; Anne Vincent-Salomon

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.


Cancer Research | 2016

Abstract P4-09-03: Prognostic and predictive value of COX2 in breast cancer, correlation with PIK3CA mutations

Sandrine Tury; Véronique Becette; Franck Assayag; Sophie Vacher; E. Marangoni; Ivan Bièche; Florence Lerebours; Céline Callens

Background: Cyclooxygenase-2 (COX2) is responsible for the synthesis of prostaglandins from arachidonic acid. This enzyme is weakly expressed in normal tissues and implicated in oncogenic and inflammatory processes. Treatment with a COX2 inhibitor (aspirin) increases overall survival of patients with colorectal cancer only for PIK3CA mutated tumors confirming an interaction between COX2 and the PI3K/AKT pathway. PIK3CA gene mutations are detected in 10-40% of BC depending on the molecular subtype. We hypothesized that COX inhibition could have an impact in Breast Cancer (BC) treatment. Methods: COX2 mRNA expression levels were analyzed in 446 BC samples and 61 patient-derived xenografts (PDX) using qRT-PCR. Protein expression of COX2 was studied by immunohistochemistry (IHC) in 26 BC and 14 PDX. The prognostic impact of COX2 expression level according to PIK3CA mutation status was also analyzed in BC patients. The activity of celecoxib, a selective COX2 inhibitor, was tested in two PDX of triple-negative BC: the HBCx50 PDX (PIK3CA wild-type, expressing COX2) and the HBCx4B PDX (PIK3CA mutated, expressing COX2). Results: COX2 transcript was under-expressed in 74% and overexpressed in 2% of the BC samples. COX2 overexpression is significantly associated with triple-negative subtype (11%, 7/68 cases, p Conclusion: In BC, COX2 underexpression is frequent and impact prognosis. BC overexpressing COX2 are rare and mainly belong to the triple-negative subtype. In vivo PDX studies show that the antitumoral effect of celecoxib may be restricted to BC expressing COX2 with PIK3CA mutation. Analyses of signaling pathways, expression levels of COX2 and its target proteins, tumor proliferation and apoptosis induction are ongoing on tumors and serum of mice to elucidate the antitumoral effect of celecoxib. This work could help to identify a subgroup of BC patients who may benefit from celecoxib especially as COX2 immunostaining and PIK3CA mutation status could routinely be used as COX2 inhibitor sensitivity biomarkers. These results need to be validated in a phase II clinical trial. Citation Format: Tury S, Becette V, Assayag F, Vacher S, Marangoni E, Bieche I, Lerebours F, Callens C. Prognostic and predictive value of COX2 in breast cancer, correlation with PIK3CA mutations. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P4-09-03.


Cancer Research | 2013

Abstract P5-09-07: Identification of resistance-specific gene expression signatures in a breast cancer patient-derived xenograft with acquired resistance to different endocrine therapies

Paul Cottu; Ivan Bièche; P de la Grange; David Gentien; Franck Assayag; Aurélie Thuleau; R El-Botty; Sophie Chateau-Joubert; M Huerre; Rana Hatem; S Richon; K Slimane; E. Marangoni

Background :nnAcquired resistance to endocrine treatments (ET) occurs in more than 70% of cases of luminal breast cancer (LBC). We used patient derived xenografts (PDX) models of LBC to study molecular changes associated with acquired resistance to different ET modalities.nnMethods :nnA PDX model of LBC, established from an early stage BRCA2-mutated breast cancer, was treated with different ET (tamoxifen, fulvestrant, oophorectomy and letrozole) during several months. Tumors escaping to therapies were re-engrafted and maintained under therapy. ET-resistant and parental hormono-responders tumors were analyzed with immunohistochemistry (IHC), RT-PCR and Affymetrix Gene Expression Arrays. Hormono-resistant tumors were additionally studied for their in vivo response to ET, mTOR and PARP inhibitors.nnResults :nnFrom the initially ET sensitive HBCx22 xenograft model (Cottu, BCRT 2012) two resistant models were obtained respectively to tamoxifen (HBCx22-TamR) and to estrogen deprivation (HBCx22-OvaR). Unsupervised clustering of gene expression showed a clear cut separation between parental, TamR and OvaR tumors. Genes differentially expressed in TamR and OvaR tumors compared to parental HBCx22 were only partially overlapping. Up-Regulated genes in both TamR and OvaR tumors (n = 302) were involved in response to wounding, nucleotide metabolism, immune system, adhesion and cell growth. Biological Processes (BP) specifically deregulated in OvaR tumors (n = 380) included embryonic development, antigen presentation, amino acid and lipid metabolism. The top BP specifically regulated in TamR tumors (n = 1059) were response to estrogen and steroid hormones, TGF-b signaling, hypoxia, regulation of cell proliferation, with several strongly up-regulated genes of the histone clusters 1 and 3. Ingenuity Transcription Factor Analysis predicted activation of NFKB, SP1, AP-1 and JUN, and inhibition of ESR1. RT-PCR and IHC analyses confirmed the down regulation of ER controlled genes in the TamR tumors. Expression of ER co-regulators determined by RT-PCR showed that GREB1 was strongly reduced in TamR, while PBX1, GATA3 and FOXA1 were inhibited in OvaR. IHC analysis showed a loss of PTEN expression in HBCx22, with high levels of p-AKT and p-RPS6 in both parental and TamR and OvaR tumors. In vivo ET showed that the TamR xenograft was resistant to all modalities of ET, while OvaR was resistant to estrogen deprivation while retaining some sensitivity to tamoxifen and fulvestrant. Treatment with the mTOR inhibitor RAD001 arrested tumor growth but did not show any additive effect when combined to ET in TamR or OvaR tumors. Conversely, the combination of RAD001 with Olaparib was highly synergistic and induced complete tumor response in 70% of mice.nnConclusions :nnAccording to the therapeutic selection, tumors derived from a PDX model of ER+ breast cancer show specific resistance patterns and gene expression profiles including disruption in the ER transcriptional program. The analysis of additional resistant tumors established from a second ER+ PDX will be presented at the meeting.nnCitation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P5-09-07.

Collaboration


Dive into the E. Marangoni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J.J. Fontaine

École nationale vétérinaire d'Alfort

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sophie Chateau-Joubert

École nationale vétérinaire d'Alfort

View shared research outputs
Researchain Logo
Decentralizing Knowledge