Network


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

Hotspot


Dive into the research topics where Sarah Dumont is active.

Publication


Featured researches published by Sarah Dumont.


Lancet Oncology | 2016

Safety and efficacy of regorafenib in patients with advanced soft tissue sarcoma (REGOSARC): a randomised, double-blind, placebo-controlled, phase 2 trial

Olivier Mir; Thomas Brodowicz; Antoine Italiano; Jennifer Wallet; Jean-Yves Blay; François Bertucci; Christine Chevreau; Sophie Piperno-Neumann; Emmanuelle Bompas; Sébastien Salas; Christophe Perrin; Corinne Delcambre; Bernadette Liegl-Atzwanger; Maud Toulmonde; Sarah Dumont; Isabelle Ray-Coquard; Stéphanie Clisant; Sophie Taieb; C. Guillemet; Maria Rios; Olivier Collard; Laurence Bozec; Didier Cupissol; Esma Saada-Bouzid; Christine Lemaignan; Wolfgang Eisterer; Nicolas Isambert; Loic Chaigneau; Axel Le Cesne; Nicolas Penel

BACKGROUND Regorafenib is a multikinase inhibitor with proven activity in refractory gastrointestinal stromal tumours and chemotherapy-refractory advanced colorectal cancers. We assessed this agents efficacy and safety in patients with metastatic soft tissue sarcomas previously treated with anthracycline. METHODS In this randomised, double-blind, phase 2 trial undertaken in France and Austria, we enrolled patients aged 18 years and older with advanced soft tissue sarcomas who had received previous doxorubicin or other anthracycline treatment. These patients were randomly assigned (1:1) into one of the following four cohorts: liposarcoma, leiomyosarcoma, synovial sarcoma, and other sarcomas. Participants were treated with oral regorafenib (160 mg per day 3 weeks on and 1 week off) or matched placebo. Patients receiving placebo were offered optional crossover in case of centrally confirmed disease progression. The random allocation schedule was computer-generated with permuted blocks of four patients, with two stratification factors: country (France or Austria) and previous exposure to pazopanib (yes or no). Eligibility criteria included patients with histologically proven advanced and inoperable soft tissue sarcomas with intolerance or failure to doxorubicin or other anthracycline-based chemotherapy and at least one unidimensionally or bidimensionally measurable lesion according to Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1). The primary endpoint was RECIST-based progression-free survival after central radiological review in the intention-to-treat population. Patients, physicians, and radiologists of the panel were masked to treatment allocation. This study is still open for recruitment for an additional stratum (patients previously treated with pazopanib) and registered with ClinicalTrials.gov, NCT01900743. FINDINGS From Aug 5, 2013, to Nov 26, 2014, 182 patients were randomly assigned to one of four cohorts and included in the final analysis. At the cutoff date (Jan 7, 2016), the number of required events was reached for the four cohorts. In the liposarcoma cohort, progression-free survival was 1·1 months (95% CI 0·9-2·3) with regorafenib versus 1·7 months (0·9-1·8) with placebo (HR 0·89 [95% CI 0·48-1·64] p=0·70). In the leiomyosarcoma cohort, progression-free survival was 3·7 months (95% CI 2·5-5·0) with regorafenib versus 1·8 (1·0-2·8) months with placebo (HR 0·46 [95% CI 0·46-0·80] p=0·0045). In the synovial sarcoma cohort, progression-free survival was 5·6 months (95% CI 1·4-11·6) with regorafenib versus 1·0 (0·8-1·4) with placebo (HR 0·10 [95% CI 0·03-0·35] p<0·0001). In the other sarcoma cohort, progression-free survival was 2·9 months (95% CI 1·0-7·8) with regorafenib versus 1·0 (0·9-1·9) with placebo (HR 0·46 [95% CI 0·25-0·81] p=0·0061). Before crossover, the most common clinically significant grade 3 or higher adverse events were arterial hypertension (17 [19%] events in the 89 patients in the regorafenib group vs two [2%] events in the 92 patients in the placebo group), hand and foot skin reaction (14 [15%] vs no events) and asthenia (12 [13%] vs six [6%]). One treatment-related death occurred in the regorafenib group due to liver failure. INTERPRETATION Regorafenib has an important clinical antitumour effect in non-adipocytic soft tissue sarcomas, improving progression-free survival. Regorafenib should be further evaluated in this setting, and its therapeutic role has to be defined in the context of the growing therapeutic armamentarium, already including one approved multikinase inhibitor, pazopanib. FUNDING Bayer HealthCare.


Journal of Neuropathology and Experimental Neurology | 2016

Clinical, Imaging, Histopathological and Molecular Characterization of Anaplastic Ganglioglioma

Marc Zanello; Mélanie Pagès; Arnault Tauziède-Espariat; Raphaël Saffroy; Stéphanie Puget; Ludovic Lacroix; Edouard Dezamis; Bertrand Devaux; Fabrice Chrétien; Felipe Andreiuolo; Christian Sainte-Rose; Michel Zerah; Frédéric Dhermain; Sarah Dumont; Guillaume Louvel; Jean-François Meder; Jacques Grill; Christelle Dufour; Johan Pallud; Pascale Varlet

Anaplastic ganglioglioma (AGG) is a rare and malignant variant of ganglioglioma. According to the World Health Organization classification version 2016, their histopathological grading criteria are still ill-defined. The aim of the present study was to assess the clinical, imaging, histopathological, and molecular characteristics and outcomes of AGGs in a large consecutive and retrospective adult and pediatric case series. Eighteen patients with AGGs (13 adults and 5 children) were identified (14 de novo and 4 secondary) from a cohort of 222 gangliogliomas (GG) (8%) treated at our institution between 2000 and 2015. AGGs represented a very aggressive disease with poor outcome (median progression-free survival, 10 months; median overall survival, 27 months). They were located in the temporal lobe only in 22% and presented with seizures (44%) or increased intracranial pressure (44%) at diagnosis. Concerning histopathological and molecular data, they shared morphological characteristics and BRAF V600E mutation (39%) with their benign counterparts but also showed hTERT promoter mutation (61%), p53 accumulation (39%), ATRX loss (17%), or p.K27M H3F3A mutation (17%). AGGs are malignant neoplasms requiring aggressive oncological treatment. In the perspective of targeted therapies, AGGs should be screened for BRAF V600E, hTERT, ATRX, and mutations of histone genes.


Radiotherapy and Oncology | 2016

Delaying standard combined chemoradiotherapy after surgical resection does not impact survival in newly diagnosed glioblastoma patients.

Guillaume Louvel; Philippe Metellus; Georges Noel; Sophie Peeters; Jacques Guyotat; J. Duntze; Pierre-Jean Le Reste; Phong Dam Hieu; Thierry Faillot; Fabien Litre; Nicolas Desse; Antoine Petit; Evelyne Emery; Jimmy Voirin; Johann Peltier; François Caire; Jean-Rodolphe Vignes; Jean-Luc Barat; Olivier Langlois; Philippe Menei; Sarah Dumont; Marc Zanello; Edouard Dezamis; Frédéric Dhermain; Johan Pallud

BACKGROUND To assess the influence of the time interval between surgical resection and standard combined chemoradiotherapy on survival in newly diagnosed and homogeneously treated (surgical resection plus standard combined chemoradiotherapy) glioblastoma patients; while controlling confounding factors (extent of resection, carmustine wafer implantation, functional status, neurological deficit, and postoperative complications). METHODS From 2005 to 2011, 692 adult patients (434 men; mean of 57.5 ± 10.8 years) with a newly diagnosed glioblastoma were enrolled in this retrospective multicentric study. All patients were treated by surgical resection (65.5% total/subtotal resection, 34.5% partial resection; 36.7% carmustine wafer implantation) followed by standard combined chemoradiotherapy (radiotherapy at a median dose of 60 Gy, with daily concomitant and adjuvant temozolomide). Time interval to standard combined chemoradiotherapy was analyzed as a continuous variable and as a dichotomized variable using median and quartiles thresholds. Multivariate analyses using Cox modeling were conducted. RESULTS The median progression-free survival was 10.3 months (95% CI, 10.0-11.0). The median overall survival was 19.7 months (95% CI, 18.5-21.0). The median time to initiation of combined chemoradiotherapy was 1.5 months (25% quartile, 1.0; 75% quartile, 2.2; range, 0.1-9.0). On univariate and multivariate analyses, OS and PFS were not significantly influenced by time intervals to adjuvant treatments. On multivariate analysis, female gender, total/subtotal resection and RTOG-RPA classes 3 and 4 were significant independent predictors of improved OS. CONCLUSIONS Delaying standard combined chemoradiotherapy following surgical resection of newly diagnosed glioblastoma in adult patients does not impact survival.


The American Journal of Surgical Pathology | 2016

PAX7 Expression in Rhabdomyosarcoma, Related Soft Tissue Tumors, and Small Round Blue Cell Neoplasms

Gregory W. Charville; Sushama Varma; Erna Forgó; Sarah Dumont; Eduardo Zambrano; Jonathan C. Trent; Alexander J. Lazar; Matt van de Rijn

Rhabdomyosarcoma, the most common soft tissue malignancy of childhood, is a morphologically variable tumor defined by its phenotype of skeletal muscle differentiation. The diagnosis of rhabdomyosarcoma often relies in part on the identification of myogenic gene expression using immunohistochemical or molecular techniques. However, these techniques show imperfect sensitivity and specificity, particularly in scant tissue biopsies. Here, we expand the toolkit for rhabdomyosarcoma diagnosis by studying the expression of PAX7, a transcriptional regulator of mammalian muscle progenitor cells implicated in the pathogenesis of rhabdomyosarcoma. Immunohistochemical analysis of tissue microarrays using a monoclonal anti-PAX7 antibody was used to characterize PAX7 expression in 25 non-neoplastic tissues, 109 rhabdomyosarcomas, and 697 small round blue cell or other soft tissue tumors. Among non-neoplastic tissues, PAX7 was specifically expressed in adult muscle progenitor cells (satellite cells). In embryonal rhabdomyosarcoma, PAX7 expression was positive in 52 of 63 cases (83%), negative in 9 of 63 cases (14%), and focal in 2 of 63 cases (3%). PAX7-positive embryonal rhabdomyosarcoma cases included several showing focal or negative myogenin expression. PAX7 expression in alveolar rhabdomyosarcoma was positive in 6 of 31 cases (19%), negative in 14 of 31 cases (45%), and focal in 11 of 31 cases (36%). In addition, PAX7 was expressed in 5 of 7 pleomorphic rhabdomyosarcomas (71%) and 6 of 8 spindle cell rhabdomyosarcomas (75%). Among histologic mimics, only Ewing sarcoma showed PAX7 expression (7/7 cases, 100%). In contrast, expression of PAX7 was not seen in the large majority (688/690, 99.7%) of examined cases of other soft tissue tumors, small round blue cell neoplasms, and leukemias/lymphomas. In summary, immunohistochemical analysis of PAX7 expression may be a useful diagnostic tool in the assessment of skeletal muscle differentiation in human tumors.


Bulletin Du Cancer | 2016

Mise au point sur les glioblastomes : des thérapies ciblant les cellules tumorales aux thérapies ciblant les cellules immunitaires

Antoine Schernberg; Aurélien Marabelle; Christophe Massard; Jean-Pierre Armand; Sarah Dumont; Eric Deutsch; Frédéric Dhermain

INTRODUCTION Glioblastoma (GBM) is associated with a poor prognosis. This review will discuss different directions of treatment, mostly regarding immunotherapies and combinatorial approaches. DEVELOPMENT Standard treatment for newly diagnosed GBM is maximal and safe surgical resection followed by concurrent radiochemotherapy (RCT) based on temozolomide, allowing 14.6 months median survival. Nowadays, no combination with molecular-targeted therapy had significantly improved prognosis. Phases I and II data are emerging, highlighting the potential efficacy of associations with other therapies. Studies have suggested the potential of targeting tumor stem cells, at less partially responsible for resistance to RCT. There is now some evidence that immunotherapy is also relevant for brain tumors. Treatment strategies have mainly explored vaccines strategies, such as the dendritic cell, heat shock protein or EGFRvIII vaccines. Of the work initiated in melanoma, immune checkpoints inhibitors have exhibited stimulating results. Others trials have demonstrated potential of autologous stimulated lymphocytes. Moreover, strong data indicates that radiation therapy has the potential to promote immunogenicity and create a sort of in situ personalized vaccine. CONCLUSION These data provide strong evidence to support the potential of associating combinatorial targeted and/or immunotherapeutic regimens in patients with GBM that may change patient outcome.


Journal of Neuro-oncology | 2017

Extent of resection and Carmustine wafer implantation safely improve survival in patients with a newly diagnosed glioblastoma: a single center experience of the current practice

Alexandre Roux; Sophie Peeters; Marc Zanello; Rabih Bou Nassif; Georges Abi Lahoud; E. Dezamis; Eduardo Parraga; Emmanuelle Lechapt-Zalcmann; Frédéric Dhermain; Sarah Dumont; Guillaume Louvel; Fabrice Chrétien; Xavier Sauvageon; Bertrand Devaux; Catherine Oppenheim; Johan Pallud

For newly diagnosed glioblastomas treated with resection in association with the standard combined chemoradiotherapy, the impact of Carmustine wafer implantation remains debated regarding postoperative infections, quality of life, and feasibility of adjuvant oncological treatments. To assess together safety, tolerance and efficacy of Carmustine wafer implantation and of extent of resection for glioblastoma patients in real-life experience. Observational retrospective monocentric study including 340 consecutive adult patients with a newly diagnosed supratentorial glioblastoma who underwent surgical resection with (n = 123) or without (n = 217) Carmustine wafer implantation as first-line oncological treatment. Carmustine wafer implantation and extent of resection did not significantly increase postoperative complications, including postoperative infections (p = 0.269, and p = 0.446, respectively). Carmustine wafer implantation and extent of resection did not significantly increase adverse events during adjuvant oncological therapies (p = 0.968, and p = 0.571, respectively). Carmustine wafer implantation did not significantly alter the early postoperative Karnofsky performance status (p = 0.402) or the Karnofsky performance status after oncological treatment (p = 0.636) but a subtotal or total surgical resection significantly improved those scores (p < 0.001, and p < 0.001, respectively). Carmustine wafer implantation, subtotal and total resection, and standard combined chemoradiotherapy were independently associated with longer event-free survival (adjusted Hazard Ratio (aHR), 0.74 [95% CI 0.55–0.99], p = 0.043; aHR, 0.70 [95% CI 0.54–0.91], p = 0.009; aHR, 0.40 [95% CI 0.29–0.55], p < 0.001, respectively) and with longer overall survival (aHR, 0.69 [95% CI 0.49–0.96], p = 0.029; aHR, 0.52 [95% CI 0.38–0.70], p < 0.001; aHR, 0.58 [95% CI 0.42–0.81], p = 0.002, respectively). Carmustine wafer implantation in combination with maximal resection, followed by standard combined chemoradiotherapy is safe, efficient, and well-tolerated in newly diagnosed supratentorial glioblastomas in adults.


Annals of Oncology | 2015

Pharmacokinetic and pharmacodynamic end points with anti-VEGFR: the more it hurts, the more it works?

Olivier Mir; Sarah Dumont; Jean-Pierre Armand

Pharmacokinetic and pharmacodynamic end points with anti-VEGFR: the more it hurts, the more it works? For the past decade, an increasing number of publications and international meeting abstracts have suggested a relationship between the occurrence of specific toxicities and the anti-tumour activity of molecular targeted agents. Indeed, these agents are supposed to be targeted against specific kinases and signal transducers in cancer cells and their micro-environment, but their targets are also expressed in normal tissues, leading to a broad range of clinical and biological toxicities. In particular, hyperten-sion has been suggested as a potential surrogate marker for the activity of anti-vascular endothelial growth factor (VEGF) agents bevacizumab, sunitinib, sorafenib and axitinib [1]. A majority of previously published studies found a relationship between blood pressure (BP) elevation (with various thresholds) and treatment activity (based on response rate, progression-free survival or overall survival) [2–8]. Theoretically, five hypotheses may account for these results, but also for the contradictory results of large cohort analyses [9]: • Patients with prolonged survival are more likely to develop hypertension because of length-bias (i.e. patients must live long enough to develop hypertension), • Heterogeneity in the definition of hypertension, in the definition of activity, or in the study population, • Hypertension and response to anti-VEGF agents are favoured by a common set of prognostic factors, • Hypertension truly predicts the activity of anti-VEGF agents, • Chance (which cannot be definitively ruled out, even in large phase III studies). Strategies to manage the length-bias issue are the time-dependent covariate approach, which was used in only one study of bevacizu-mab in non-small-cell lung cancer patients [10], and the landmark method. Using the latter, patients who died or discontinued treatment before the landmark time point are excluded from analyses, while patients who develop hypertension after the landmark time point are included in the same group than patients who never developed hypertension. Different landmark time points were used in previous studies, and all of them were determined empirically. However, we should keep in mind that the earliest landmark points are probably the more helpful for treatment adjustments. As far as the definition of hypertension is concerned, it should be borne in mind that toxicity was graded according to the NCI-CTC v2.0 or v3.0 in most previous studies, both classifications being based on therapeutic interventions but not specifically on BP levels. The methods used for BP monitoring were also poorly reported, and only …


Anti-Cancer Drugs | 2017

Trabectedin in advanced desmoplastic round cell tumors: a retrospective single-center series

Benjamin Verret; Charles Honoré; Sarah Dumont; Philippe Terrier; Julien Adam; Andrea Cavalcanti; Isabelle Sourrouille; Guillaume Klausner; Marion Ahlenc-Gelas; Nicolas Kiavue; Gabriella Domitrescu; Lamiae El Amarti; Olivier Mir; Axel Le Cesne

Desmoplastic small round cell tumor (DSRCT) is a rare and aggressive malignancy that occurs with unpredictable chemosensitivity and limited treatment options in the advanced setting. Prognosis is poor, and exploring new treatment options for such diseases is difficult because of its rarity. Clinical activity of trabectedin for advanced DSRCT was scarcely reported in the literature. Here, we report a series of six patients treated with trabectedin for an unresectable DSRCT. After receiving trabectedin, two patients had stable disease with a time to progression of 3 and 3.5 months; four patients experienced disease progression after one cycle, two of them could receive one and two patients another line regiment. Four patients experienced grade 3–4 adverse events, two grade 3 thrombocytopenia, and one neutropenic fever. Prognosis was poor with a median overall survival of 4 (range: 2–14) months. In our experience, trabectedin had limited activity in advanced DSRCT. Further studies are warranted to find effective treatments.


Scientific Reports | 2017

A Retrospective Multicentric Study of Ewing Sarcoma Family of Tumors in Patients Older Than 50: Management and Outcome

Pauline Rochefort; Antoine Italiano; Valérie Laurence; Nicolas Penel; Audrey Lardy-Cleaud; Olivier Mir; Christine Chevreau; François Bertucci; Emmanuelle Bompas; Loic Chaigneau; Dominique Levy; Thomas Ryckewaert; Sarah Dumont; Pierre Meeus; Dominique Ranchère; Jean-Yves Blay; Philippe Cassier

Ewing’s sarcoma family of tumors (EFTs) is a group of rare and aggressive tumors. Data on EFTs in patients (pts) ≥ 50 years are limited and these pts are often not eligible for clinical trials. Some, but not all, studies have reported inferior outcome for older pts with EFTs. We conducted an IRB-approved retrospective analysis among centers of the French Sarcoma Group on pts diagnosed with EFTs at age ≥50 between 2000 and 2012. Clinical features, treatment modality and outcomes were analyzed. Seventy-seven pts were identified, including 36 females (46.8%) and the median age at diagnosis was 56 years (range: 50–86). The primary tumor was located in soft tissue in 59 pts (76.6%). Fifty-six pts (72.7%) had localized disease, among them 49 (87.5%) received chemotherapy in addition to local therapy. Their estimated 3-yr OS and event-free survival (EFS) rates were respectively 73.3% and 62.2%. Recurrence occurred in 43 pts. The estimated 3-yr OS rate was 37% in pts with metastatic disease at presentation. EFTs in pts ≥50 years are more likely to originate from soft tissue and their outcomes appear to be worse than that of younger pts treated with modern protocols.


British Journal of Neurosurgery | 2017

Epileptic seizures in anaplastic gangliogliomas

Marc Zanello; Mélanie Pagès; Alexandre Roux; Sophie Peeters; Edouard Dezamis; Stéphanie Puget; Bertrand Devaux; C. Sainte-Rose; Michel Zerah; Guillaume Louvel; Sarah Dumont; Jean-François Meder; Jacques Grill; Gilles Huberfeld; Fabrice Chrétien; Eduardo Parraga; Xavier Sauvageon; Pascale Varlet; Johan Pallud

Abstract Aim: Prevalence and predictors of epileptic seizures are unknown in the malignant variant of ganglioglioma. Methods: In a retrospective exploratory dataset of 18 supratentorial anaplastic World Health Organization grade III gangliogliomas, we studied: (i) the prevalence and predictors of epileptic seizures at diagnosis; (ii) the evolution of seizures during tumor evolution; (iii) seizure control rates and predictors of epilepsy control after oncological treatments. Results: Epileptic seizures prevalence progresses throughout the natural course of anaplastic gangliogliomas: 44% at imaging discovery, 67% at histopathological diagnosis, 69% following oncological treatment, 86% at tumor progression, and 100% at the end-of-life phase. The medical control of seizures and their refractory status worsened during the tumor’s natural course: 25% of uncontrolled seizures at histopathological diagnosis, 40% following oncological treatment, 45.5% at tumor progression, and 45.5% at the end-of-life phase. Predictors of seizures at diagnosis appeared related to the tumor location (i.e. temporal and/or cortical involvement). Prognostic parameters of seizure control after first-line oncological treatment were temporal tumor location, eosinophilic granular bodies, TP53 mutation, and extent of resection. Prognostic parameters of seizure control at tumor progression were a history of epileptic seizures at diagnosis, seizure control after first-line oncological treatment, eosinophilic granular bodies, and TP53 mutation. Conclusion: Epileptic seizures are frequently observed in anaplastic gangliogliomas and both prevalence and medically refractory status worsen during the tumor’s natural course. Both oncological and antiepileptic treatments should be employed to improve the control of epileptic seizures and the quality of life of patients harboring an anaplastic ganglioglioma.

Collaboration


Dive into the Sarah Dumont's collaboration.

Top Co-Authors

Avatar

Olivier Mir

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Johan Pallud

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Antoine Italiano

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc Zanello

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge