Network


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

Hotspot


Dive into the research topics where Caroline Dehais is active.

Publication


Featured researches published by Caroline Dehais.


Neurology | 2009

α-Internexin expression identifies 1p19q codeleted gliomas

François Ducray; Emmanuelle Crinière; Ahmed Idbaih; Karima Mokhtari; Yannick Marie; Sophie Paris; Soledad Navarro; Florence Laigle-Donadey; Caroline Dehais; Joëlle Thillet; Khê Hoang-Xuan; J. Y. Delattre; Marc Sanson

Background: α-Internexin (INA) is a proneural gene encoding a neurofilament interacting protein that is upregulated in some gliomas, particularly oligodendrogliomas. Methods: INA expression was evaluated by immunohistochemistry in a series of 122 gliomas, and correlated to the 1p19q codeletion, a favorable prognostic marker of oligodendroglial tumors. Results: INA expression was strong (>10% positive cells) in 22 cases (22 oligodendroglial tumors and 0 astrocytic tumors), weak (<10% cells) in 14 cases (12 oligodendroglial tumors, 2 glioblastoma with an oligodendroglial component, and 0 astrocytic tumors), and negative in 86 cases (49 oligodendroglial tumors, 9 glioblastoma with an oligodendroglial component, and 28 astrocytic tumors). Among the 27 tumors exhibiting the 1p19q codeletion (all with an oligodendroglial phenotype), INA was detected in 96% (26/27, 18 strong, 8 weak) as compared to 11% (10/95, 4 strong, 6 weak) in the tumors without 1p19q codeletion (with an oligodendroglial or an astrocytic phenotype) (p < 0.001). In oligodendroglial tumors, INA expression specificity for 1p19q codeletion was 86%, sensitivity 96%, positive predictive value 76%, and negative predictive value was 98%. The prognostic impact of INA expression could be evaluated in grade III oligodendroglial tumors. Similar to 1p19q deletion, positive INA expression was correlated with better progression-free survival (52.6 vs 8.7 months [p = 0.001]) and overall survival (121.1 vs 31.4 months [p = 0.0001]). Conclusion: α-Internexin (INA) expression appears to be a simple, reliable prognostic marker and a surrogate marker of 1p19q codeletion.


Neuro-oncology | 2014

Contrast enhancement in 1p/19q-codeleted anaplastic oligodendrogliomas is associated with 9p loss, genomic instability, and angiogenic gene expression

German Reyes-Botero; Caroline Dehais; Ahmed Idbaih; Nadine Martin-Duverneuil; Marion Lahutte; Catherine Carpentier; Eric Letouzé; Olivier Chinot; Hugues Loiseau; Jérôme Honnorat; Carole Ramirez; Elisabeth Moyal; Dominique Figarella-Branger; François Ducray; Christine Desenclos; Henri Sevestre; Philippe Menei; Sophie Michalak; Edmond Nader; Joel Godard; Gabriel Viennet; Antoine F. Carpentier; Sandrine Eimer; Phong Dam-Hieu; Isabelle Quintin-Roué; Jean-Sébastien Guillamo; Emmanuelle Lechapt-Zalcman; Jean-Louis Kemeny; Pierre Verrelle; Thierry Faillot

BACKGROUND The aim of this study was to correlate MRI features and molecular characteristics in anaplastic oligodendrogliomas (AOs). METHODS The MRI characteristics of 50 AO patients enrolled in the French national network for high-grade oligodendroglial tumors were analyzed. The genomic profiles and IDH mutational statuses were assessed using high-resolution single-nucleotide polymorphism arrays and direct sequencing, respectively. The gene expression profiles of 25 1p/19q-codeleted AOs were studied on Affymetrix expression arrays. RESULTS Most of the cases were frontal lobe contrast-enhanced tumors (52%), but the radiological presentations of these cases were heterogeneous, ranging from low-grade glioma-like aspects (26%) to glioblastoma-like aspects (22%). The 1p/19q codeletion (n = 39) was associated with locations in the frontal lobe (P = .001), with heterogeneous intratumoral signal intensities (P = .003) and with no or nonmeasurable contrast enhancements (P = .01). The IDH wild-type AOs (n = 7) more frequently displayed ringlike contrast enhancements (P = .03) and were more frequently located outside of the frontal lobe (P = .01). However, no specific imaging pattern could be identified for the 1p/19q-codeleted AO or the IDH-mutated AO. Within the 1p/19q-codeleted AO, the contrast enhancement was associated with larger tumor volumes (P = .001), chromosome 9p loss and CDKN2A loss (P = .006), genomic instability (P = .03), and angiogenesis-related gene expression (P < .001), particularly for vascular endothelial growth factor A and angiopoietin 2. CONCLUSION In AOs, the 1p/19q codeletion and the IDH mutation are associated with preferential (but not with specific) imaging characteristics. Within 1p/19q-codeleted AO, imaging heterogeneity is related to additional molecular alterations, especially chromosome 9p loss, which is associated with contrast enhancement and larger tumor volume.


Neurology | 2009

Supratentorial low-grade gliomas in older patients

Gentian Kaloshi; Dimitri Psimaras; Karima Mokhtari; Caroline Dehais; Caroline Houillier; Yannick Marie; Florence Laigle-Donadey; Sophie Taillibert; R. Guillevin; Nadine Martin-Duverneuil; Marc Sanson; Khê Hoang-Xuan; J. Y. Delattre

Background: Low-grade gliomas (LGG) are thought to be very rare in elderly patients (>60 years) and have not been thoroughly studied. Methods: A series of 62 elderly (≥60 years of age) LGG patients were identified in a department database collecting information on pathologically identified adult supratentorial LGG. The clinical, radiologic, pathologic, and therapeutic data of these patients were analyzed and compared to those of 704 younger LGG patients (<60 years). Results: Comparisons between older and younger groups showed that elderly patients more often presented with a clinical deficit (p < 0.0001), a lower Karnofsky performance status (p = 0.0002), a larger tumor on MRI (p = 0.03), and a lower rate of tumor resection (p < 0.0001). Chemotherapy was more often used as first line treatment (p = 0.001). Among the patients who died of progressive disease, 55% of the elderly patients had not received radiotherapy compared to 11% in the younger group (p < 0.0001). Survival was shorter in older patients (p < 0.0001), with a 5-year survival rate of 40%. An astrocytic phenotype (p = 0.0097), increasing age (p = 0.0049), and a tumor crossing the midline (p = 0.028) were negative prognostic factors in the older group. Conclusion: We found that 8% of low-grade gliomas (LGG) occur in older patients (≥60 years of age). The clinical–radiologic picture of LGG in the elderly population differs from younger patients. Although long-term survival occurs, the course is generally more severe because elderly patients accumulate negative prognostic factors and because they are probably undertreated.


Neuro-oncology | 2014

Mitotic index, microvascular proliferation, and necrosis define 3 groups of 1p/19q codeleted anaplastic oligodendrogliomas associated with different genomic alterations

Dominique Figarella-Branger; Karima Mokhtari; Caroline Dehais; Anne Jouvet; Emmanuelle Uro-Coste; Carole Colin; Catherine Carpentier; François Ducray; Ahmed Idbaih

BACKGROUND The aim of this study was to correlate histological features and molecular characteristics in anaplastic oligodendrogliomas (AOs). METHODS The histological characteristics of 203 AO patients, enrolled in the French national network POLA, were analyzed. The genomic profiles of 191 cases were studied using genomic arrays. IDH mutational status was assessed by immunohistochemistry and direct sequencing. RESULTS 1p/19q codeletion was present in 79% of cases and was associated with alpha-internexin expression (P < 10(-4)), IDH1/2 mutation (P < 10(-4)), chromosome 4 loss (P < 10(-3)), and better overall survival (P < 10(-4)). Based on mitotic index, microvascular proliferation (MVP), and necrosis, 3 groups of 1p/19q codeleted AOs were identified: (group 1) AO with more than 5 mitoses per 10-HPF, no MVP, and no necrosis; (group 2) AO with MVP and no necrosis; and (group 3) AO with MVP and necrosis. Compared with group 1, groups 2 and 3 AOs had a higher mean Ki-67 proliferation index and a higher rate of 9p and 9q losses. Compared with group 2, group 3 AOs had a higher number of chromosomal alterations including chromosome 4 loss. In the subgroup of 157 1p/19q codeleted AOs, chromosomal instability was associated with shorter progression-free survival (P = .024) and shorter overall survival (P = .023). CONCLUSIONS The present study shows that oligodendrogliomas with classic histological features remain a molecularly heterogeneous entity and should be stratified according to 1p/19q status because of its major prognostic relevance. Moreover, 1p/19q codeleted AOs are also heterogeneous. Interestingly, mitotic index, MVP, and necrosis help to classify them into 3 groups associated with distinct genomic alterations.


PLOS ONE | 2012

SNP Array Analysis Reveals Novel Genomic Abnormalities Including Copy Neutral Loss of Heterozygosity in Anaplastic Oligodendrogliomas

Ahmed Idbaih; François Ducray; Caroline Dehais; Célia Courdy; Catherine Carpentier; Simon de Bernard; Emmanuelle Uro-Coste; Karima Mokhtari; Anne Jouvet; Jérôme Honnorat; Olivier Chinot; Carole Ramirez; Patrick Beauchesne; Alexandra Benouaich-Amiel; Joel Godard; Sandrine Eimer; Fabrice Parker; Emmanuelle Lechapt-Zalcman; Philippe Colin; Delphine Loussouarn; Thierry Faillot; Phong Dam-Hieu; Selma Elouadhani-Hamdi; Luc Bauchet; Olivier Langlois; Caroline Le Guerinel; Denys Fontaine; Elodie Vauleon; Philippe Menei; Marie Janette Motsuo Fotso

Anaplastic oligodendrogliomas (AOD) are rare glial tumors in adults with relative homogeneous clinical, radiological and histological features at the time of diagnosis but dramatically various clinical courses. Studies have identified several molecular abnormalities with clinical or biological relevance to AOD (e.g. t(1;19)(q10;p10), IDH1, IDH2, CIC and FUBP1 mutations). To better characterize the clinical and biological behavior of this tumor type, the creation of a national multicentric network, named “Prise en charge des OLigodendrogliomes Anaplasiques (POLA),” has been supported by the Institut National du Cancer (InCA). Newly diagnosed and centrally validated AOD patients and their related biological material (tumor and blood samples) were prospectively included in the POLA clinical database and tissue bank, respectively. At the molecular level, we have conducted a high-resolution single nucleotide polymorphism array analysis, which included 83 patients. Despite a careful central pathological review, AOD have been found to exhibit heterogeneous genomic features. A total of 82% of the tumors exhibited a 1p/19q-co-deletion, while 18% harbor a distinct chromosome pattern. Novel focal abnormalities, including homozygously deleted, amplified and disrupted regions, have been identified. Recurring copy neutral losses of heterozygosity (CNLOH) inducing the modulation of gene expression have also been discovered. CNLOH in the CDKN2A locus was associated with protein silencing in 1/3 of the cases. In addition, FUBP1 homozygous deletion was detected in one case suggesting a putative tumor suppressor role of FUBP1 in AOD. Our study showed that the genomic and pathological analyses of AOD are synergistic in detecting relevant clinical and biological subgroups of AOD.


Nature Communications | 2016

Integrated multi-omics analysis of oligodendroglial tumours identifies three subgroups of 1p/19q co-deleted gliomas

Aurélie Kamoun; Ahmed Idbaih; Caroline Dehais; Nabila Elarouci; Catherine Carpentier; Eric Letouzé; Carole Colin; Karima Mokhtari; Anne Jouvet; Emmanuelle Uro-Coste; Nadine Martin-Duverneuil; Marc Sanson; Jean-Yves Delattre; Dominique Figarella-Branger; Aurélien de Reyniès; François Ducray; Clovis Adam; Marie Andraud; Marie-Hélène Aubriot-Lorton; Luc Bauchet; Patrick Beauchesne; Franck Bielle; Claire Blechet; Mario Campone; Antoine F. Carpentier; Ioana Carpiuc; Dominique Cazals-Hatem; Marie-Pierre Chenard; Danchristian Chiforeanu; Olivier Chinot

Oligodendroglial tumours (OT) are a heterogeneous group of gliomas. Three molecular subgroups are currently distinguished on the basis of the IDH mutation and 1p/19q co-deletion. Here we present an integrated analysis of the transcriptome, genome and methylome of 156 OT. Not only does our multi-omics classification match the current classification but also reveals three subgroups within 1p/19q co-deleted tumours, associated with specific expression patterns of nervous system cell types: oligodendrocyte, oligodendrocyte precursor cell (OPC) and neuronal lineage. We confirm the validity of these three subgroups using public datasets. Importantly, the OPC-like group is associated with more aggressive clinical and molecular patterns, including MYC activation. We show that the MYC activation occurs through various alterations, including MYC genomic gain, MAX genomic loss, MYC hypomethylation and microRNA-34b/c down-regulation. In the lower grade glioma TCGA dataset, the OPC-like group is associated with a poorer outcome independently of histological grade. Our study reveals previously unrecognized heterogeneity among 1p/19q co-deleted tumours.


Neuro-oncology | 2010

Chromosome 9p and 10q losses predict unfavorable outcome in low-grade gliomas

Caroline Houillier; Karima Mokhtari; Catherine Carpentier; Emmanuelle Crinière; Yannick Marie; Audrey Rousseau; Gentian Kaloshi; Caroline Dehais; Julien Laffaire; Florence Laigle-Donadey; Khê Hoang-Xuan; Marc Sanson; Jean-Yves Delattre

The loss of chromosomes 1p-19q is the only prognostic molecular alteration identified in low-grade gliomas (LGGs) to date. Search for loss of heterozygosity (LOH) on chromosomes 1p, 9p, 10q, and 19q was performed in a series of 231 LGGs. Loss of chromosomes 1p-19q was strongly correlated with prolonged progression-free survival (PFS) and overall survival (OS) in univariate and multivariate analyses. LOH on 9p and 10q were associated with shortened PFS (P = .01 and .03, respectively) on univariate analysis. On multivariate analysis, LOH on 9p remained significant for PFS (P = .05), whereas LOH on 10q had a significant effect on OS (P = .02). Search for LOH 9p and 10q appears to be a useful complement to analysis of chromosomes 1p-19q in LGGs.


Nature Communications | 2015

TCF12 is mutated in anaplastic oligodendroglioma.

Karim Labreche; Iva Simeonova; Vincent Gleize; Daniel Chubb; Eric Letouzé; Yasser Riazalhosseini; Sara E. Dobbins; Nabila Elarouci; François Ducray; Diana Zelenika; Christopher P. Wardell; Mathew Frampton; Olivier Saulnier; Tomi Pastinen; Sabrina Hallout; Dominique Figarella-Branger; Caroline Dehais; Ahmed Idbaih; Karima Mokhtari; Jean-Yves Delattre; Emmanuelle Huillard; G. Mark Lathrop; Marc Sanson; Richard S. Houlston

Anaplastic oligodendroglioma (AO) are rare primary brain tumours that are generally incurable, with heterogeneous prognosis and few treatment targets identified. Most oligodendrogliomas have chromosomes 1p/19q co-deletion and an IDH mutation. Here we analysed 51 AO by whole-exome sequencing, identifying previously reported frequent somatic mutations in CIC and FUBP1. We also identified recurrent mutations in TCF12 and in an additional series of 83 AO. Overall, 7.5% of AO are mutated for TCF12, which encodes an oligodendrocyte-related transcription factor. Eighty percent of TCF12 mutations identified were in either the bHLH domain, which is important for TCF12 function as a transcription factor, or were frameshift mutations leading to TCF12 truncated for this domain. We show that these mutations compromise TCF12 transcriptional activity and are associated with a more aggressive tumour type. Our analysis provides further insights into the unique and shared pathways driving AO.


Cancer | 2014

Diagnostic and prognostic value of preoperative combined GFAP, IGFBP‐2, and YKL‐40 plasma levels in patients with glioblastoma

Jaime Gállego Pérez-Larraya; Sophie Paris; Ahmed Idbaih; Caroline Dehais; Florence Laigle-Donadey; Soledad Navarro; Laurent Capelle; Karima Mokhtari; Yannick Marie; Marc Sanson; Khê Hoang-Xuan; Jean-Yves Delattre; Alain Mallet

Circulating proteins released by tumor cells have recently been investigated as potential single surrogate biomarkers for glioblastoma multiforme (GBM). The aim of the current hypothesis‐generating study was to evaluate the diagnostic and prognostic role of preoperative insulin‐like growth factor‐binding protein 2 (IGFBP‐2), chitinase‐3‐like protein 1 (YKL‐40), and glial fibrillary acidic protein (GFAP) plasma levels in patients with GBM, both as single markers and as a combined profile.


Neurology | 2015

Allelic loss of 9p21.3 is a prognostic factor in 1p/19q codeleted anaplastic gliomas

Agusti Alentorn; Caroline Dehais; François Ducray; Catherine Carpentier; Karima Mokhtari; Dominique Figarella-Branger; Olivier Chinot; Elisabeth Cohen-Moyal; Carole Ramirez; Hugues Loiseau; Selma Elouahdani-Hamdi; Patrick Beauchesne; Olivier Langlois; Christine Desenclos; Jean-Sébastien Guillamo; Phong Dam-Hieu; François Ghiringhelli; Philippe Colin; Joel Godard; Fabrice Parker; Frédéric Dhermain; Antoine F. Carpentier; Jean-Sebastien Frenel; Philippe Menei; Luc Bauchet; Thierry Faillot; Mélanie Fesneau; Denys Fontaine; Marie-Jeannette Motuo-Fotso; Elodie Vauleon

Objectives: We aimed to study the potential clinical relevance of 9p allelic loss, with or without copy number variation, in 1p/19q codeleted anaplastic oligodendroglial tumors (AOTs). Methods: This study enrolled 216 patients with 1p/19q codeleted AOT. The prognostic value of 9p allelic loss was investigated using a French nation-wide prospective registry, POLA (prise en charge des tumeurs oligodendrogliales anaplasiques) and high-density single nucleotide polymorphism arrays. We validated our results using the Repository of Molecular Brain Neoplasia Data (REMBRANDT) dataset. Results: The minimal common region of allelic loss in chromosome arm 9p was 9p21.3. Allelic loss of 9p21.3, detected in 41.7% of tumors, was associated with shorter progression-free and overall survival rates in univariate (p = 0.008 and p < 0.001, respectively) and multivariate analyses (p = 0.009 and p = 0.009, respectively). This finding was validated in the REMBRANDT dataset in univariate and multivariate analysis (p = 0.01 and p = 0.01, respectively). Conclusion: Our study highlights a novel potential prognostic biomarker in 1p/19q codeleted AOT. Further prospective studies are warranted to investigate our finding.

Collaboration


Dive into the Caroline Dehais'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carole Colin

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge