Network


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

Hotspot


Dive into the research topics where M. Barrie is active.

Publication


Featured researches published by M. Barrie.


Journal of Clinical Oncology | 2001

Safety and Efficacy of Temozolomide in Patients With Recurrent Anaplastic Oligodendrogliomas After Standard Radiotherapy and Chemotherapy

Oliver-Louis Chinot; Stéphane Honoré; Henry Dufour; M. Barrie; Dominique Figarella-Branger; Xavier Muracciole; Diane Braguer; Pierre-Marie Martin; François Grisoli

PURPOSEnMost primary oligodendrogliomas and mixed gliomas (oligoastrocytoma) respond to treatment with procarbazine, lomustine, and vincristine (PCV), with response rates of approximately 80%. However, limited data on second-line treatments are available in patients with recurrent tumors. A novel second-generation alkylating agent, temozolomide, has recently demonstrated efficacy and safety in patients with recurrent glioblastoma multiforme and anaplastic astrocytoma. This study describes the effects of temozolomide in patients with recurrent anaplastic oligodendroglioma (AO) and anaplastic mixed oligoastrocytoma (AOA).nnnPATIENTS AND METHODSnForty-eight patients with histologically confirmed AO or AOA who had received previous PCV chemotherapy were treated with temozolomide (150 to 200 mg/m2/d for 5 days per 28-day cycle). The primary end point was objective response. Secondary end points included progression-free survival (PFS), time to progression, overall survival (OS), safety, and tolerability.nnnRESULTSnEight patients (16.7%) experienced a complete response, 13 patients (27.1%) experienced a partial response (objective response rate, 43.8%), and 19 patients (39.6%) experienced stable disease. For the entire treatment group, median PFS was 6.7 months and median OS was 10 months. For objective responders, median PFS was 13.1 months and median OS was 16 months. For complete responders, PFS was more than 11. 8 months and OS was more than 26 months. Response correlated with improved survival. Temozolomide was safe and well tolerated. Twelve patients developed grade 1/2 thrombocytopenia and three patients developed grade 3/4 thrombocytopenia.nnnCONCLUSIONnTemozolomide is safe and effective in the treatment of recurrent AO and AOA.


Neuro-oncology | 2012

FDG-PET predicts survival in recurrent high-grade gliomas treated with bevacizumab and irinotecan

Cécile Colavolpe; Olivier Chinot; Philippe Metellus; Julien Mancini; M. Barrie; Céline Béquet-Boucard; Emeline Tabouret; Olivier Mundler; Dominique Figarella-Branger; Eric Guedj

Prognosis of recurrent high-grade glioma (HGG) is poor, although bevacizumab has been documented in that context. This study aimed to determine the independent prognostic value of fluorodeoxyglucose (FDG)-PET on progression-free survival (PFS) and overall survival (OS) of recurrent HGG after combined treatment with bevacizumab and irinotecan, compared with other documented prognostic variables. Twenty-five adult patients with histologically proven HGG were included at recurrence. Brain FDG-PET imaging was performed within 6 weeks of starting chemotherapy with bevacizumab and irinotecan. Response based on MRI was assessed every 2 months according to revised assessment in Neuro-Oncology (RANO) criteria. Median PFS and OS were 4 months (range, 0.9-10.4 months) and 7.2 months (range, 1.2-41.7 months), respectively. At 6 months, PFS and OS rate were 16.0% and 72.0%. FDG uptake was the most powerful predictor of both PFS and OS, using either univariate or multivariate analysis, among all variables tested: histological grade, Karnofsky performance status, steroid intake, and number of previous treatments. Moreover, FDG uptake was also prognostic of response to bevacizumab-based therapy. This study provides the first evidence that pretreatment FDG-PET can serve as an imaging biomarker in recurrent HGG for predicting survival following anti-angiogenic therapy with bevacizumab.


The Lancet Haematology | 2015

Methotrexate and temozolomide versus methotrexate, procarbazine, vincristine, and cytarabine for primary CNS lymphoma in an elderly population: an intergroup ANOCEF-GOELAMS randomised phase 2 trial

Antonio Omuro; Olivier Chinot; Luc Taillandier; Hervé Ghesquières; Carole Soussain; Vincent Delwail; Thierry Lamy; Remy Gressin; Sylvain Choquet; Pierre Soubeyran; Aymeri Huchet; Alexandra Benouaich-Amiel; Sophie Lebouvier-Sadot; Emmanuel Gyan; Valérie Touitou; M. Barrie; Monica Sierra del Rio; Alberto Gonzalez-Aguilar; Caroline Houillier; Daniel Delgadillo; L. Lacomblez; Marie Laure Tanguy; Khê Hoang-Xuan

BACKGROUNDnNo standard chemotherapy regimen exists for primary CNS lymphoma, reflecting an absence of randomised studies. We prospectively tested two promising methotrexate-based regimens, one more intensive and a milder regimen, for primary CNS lymphoma in the elderly population, who account for most patients.nnnMETHODSnIn this open-label, randomised phase 2 trial, done in 13 French institutions, we enrolled immunocompetent patients who had neuroimaging and histologically confirmed newly diagnosed primary CNS lymphoma, were aged 60 years and older, and had a Karnofsky performance scale score of 40 or more. Participants were stratified by Karnofsky performance scale score (<60 vs ≥60) and treating institution and randomly assigned (1:1) to receive methotrexate (3·5 g/m(2)) with temozolomide (150 mg/m(2)) or methotrexate (3·5 g/m(2)), procarbazine (100 mg/m(2)), vincristine (1·4 mg/m(2)), and cytarabine (3 mg/m(2)). Neither regimen included radiotherapy; both included prophylactic G-CSF and corticosteroids. The primary endpoint was 1-year progression-free survival. Analysis was intent to treat, in a non-comparative phase 2 trial design. This study is registered with ClinicalTrials.gov, number NCT00503594.nnnFINDINGSnBetween July 16, 2007, and March 25, 2010, 98 patients were enrolled, of whom 95 were randomly assigned and analysed; 48 to methotrexate with temozolomide and 47 to methotrexate, procarbazine, vincristine, and cytarabine. 1-year progression-free survival was 36% (95% CI 22-50) in the methotrexate, procarbazine, vincristine, and cytarabine group and 36% (22-50) in the methotrexate with temozolomide group; median progression-free survival was 9·5 months (95% CI 5·3-13·8) versus 6·1 months (3·8-11·9), respectively. Objective responses were noted in 82% (95% CI 68-92) of patients in the methotrexate, procarbazine, vincristine, and cytarabine group versus 71% (55-84) of patients in the methotrexate with temozolomide group. Median overall survival was 31 months (95% CI 12·2-35·8) in the methotrexate, procarbazine, vincristine, and cytarabine group and 14 months (8·1-28·4) in the methotrexate with temozolomide group. No differences were noted in toxic effects between the two groups. The most common grades 3 and 4 toxicities in both groups were liver dysfunction (21 [4%] in the the methotrexate and temozolomide group and 18 [38%] in the methotrexate, procarbazine, vincristine, and cytarabine group), lymphopenia (14 [29%] and 14 [30%]), and infection (six [13%] and seven [15%]). To date, 33 (69%) patients in the methotrexate and temozolomide group have died, versus 31 (55%) in the methotrexate, procarbazine, vincristine and cytarabine group. Quality-of-life evaluation (QLQ-C30 and BN20) showed improvements in most domains (p=0·01-0·0001) compared with baseline in both groups. Prospective neuropsychological testing showed no evidence of late neurotoxicity.nnnINTERPRETATIONnIn this study of two different methotrexate-based combination regimens in elderly patients, the efficacy endpoints tended to favour the methotrexate, procarbazine, vincristine, and cytarabine group. Both regimens were associated with similar, moderate toxicity, but quality of life improved with time, suggesting pursuing treatment in these poor prognosis patients is worthwhile. New alternatives are needed to improve response duration in this population.nnnFUNDINGnSchering-Plough/Merck and French Government.


Journal of Neuro-oncology | 2013

Limited impact of prognostic factors in patients with recurrent glioblastoma multiforme treated with a bevacizumab-based regimen.

E. Tabouret; M. Barrie; A. Thiebaut; M. Matta; Celine Boucard; D. Autran; Anderson Loundou; O. Chinot

Bevacizumab has demonstrated activity in patients with recurrent glioblastoma. However, the impact of prognostic factors associated with recurrent glioblastoma treated with cytotoxic agents has not been determined in patients treated with bevacizumab. To analyze the prognostic factors and clinical benefits of bevacizumab and irinotecan treatment in patients with recurrent glioblastoma. This monocentric study retrospectively analyzed all patients with recurrent glioblastoma who were treated with at least one cycle of bevacizumab and irinotecan at our institution from April 2007 to May 2010. Multivariate analysis was used to analyze prognostic factors for overall survival (OS) from the initiation of bevacizumab administration. Among the 100 patients that were identified (M/F: 65/35), the median age was 57.9xa0years (range: 18–76). Karnofsky Performance Status (KPS) was <70 in 44 patients and ≥70 in 56 patients; 83xa0% of the patients were on steroids. The median tumor area was 2012xa0mm2. The median progression free survival was 3.9xa0months (CI 95xa0%: 3.4–4.3). The median OS was 6.5xa0months (CI 95xa0%: 5.6–7.4). Multivariate analysis revealed that OS was affected by KPS (pxa0=xa00.024), but not by gender, age, steroid treatment, number of previous lines of treatment, tumor size, or time from initial diagnosis. KPS was improved in 30 patients, including 14/44 patients with an initial KPS <70. The median duration of maintained functional independence (KPS ≥70) was 3.75xa0months (CI 95xa0%: 2.9–4.6). The median OS from initial diagnosis was 18.9xa0months (CI 95xa0%: 17.5–20.3). In patients with recurrent glioblastoma treated with bevacizumab, KPS was revealed as the only factor to impact OS. The clinical benefits associated with this regimen appear valuable. A positive impact of bevacizumab administration on OS of patients with glioblastoma multiforme is suggested.


Journal of Neuro-oncology | 2016

Coping with a newly diagnosed high-grade glioma: patient-caregiver dyad effects on quality of life.

Karine Baumstarck; Tanguy Leroy; Zeinab Hamidou; E. Tabouret; Patrizia Farina; M. Barrie; Chantal Campello; Gregorio Petrirena; Olivier Chinot; Pascal Auquier

Patients with high-grade gliomas (HGG) and their caregivers have to confront a very aggressive disease that produces major lifestyle disruptions. There is an interest in studying the ability of patients and their caregivers to cope with the difficulties that affect quality of life (QoL). We examine, in a sample of patient-caregiver dyads in the specific context of newly diagnosed cases of HGG, whether the QoL of patients and caregivers is influenced by the coping processes they and their relatives use from a specific actor–partner interdependence model (APIM). This cross-sectional study involved 42 dyads with patients having recent diagnoses of HGG and assessed in the time-frame between diagnosis and treatment initiation. The self-reported data included QoL (Patient-Generated Index, EORTC QLQ-C30, and CareGiver Oncology QoL), emotional status, and coping strategies (BriefCope). The APIM was used to test the dyadic effects of coping strategies on QoL. Coping strategies, such as social support, avoidance, and problem solving, exhibited evidence of either an actor effect (degree to which the individual’s coping strategies are associated with their own QoL) or partner effect (degree to which the individual’s coping strategies are associated with the QoL of the other member of the dyad) for patients or caregivers. For positive-thinking coping strategies, actor and partner effect were not observed. This study emphasizes that the QoL for patients and their caregivers was directly related to the coping strategies they used. This finding suggests that targeted interventions should be offered to help patients and their relatives to implement more effective coping strategies.


Journal of Neuro-oncology | 2017

Relationship between magnetic resonance imaging characteristics and plasmatic levels of MMP2 and MMP9 in patients with recurrent high-grade gliomas treated by Bevacizumab and Irinotecan

Patrizia Farina; Emeline Tabouret; Pierre Lehmann; M. Barrie; Gregorio Petrirena; Chantal Campello; Celine Boucard; Thomas Graillon; Nadine Girard; Olivier Chinot

Matrix metalloproteases MMP2 and MMP9 are involved in cancer angiogenesis and invasion. We recently demonstrated that plasma MMP2 and MMP9 levels could both predict response to bevacizumab in patients with recurrent high-grade glioma (HGG). We examined the potential relationship between MMP2/MMP9 plasma levels and glioma imaging characteristics. In this retrospective, monocentric study, MRI before bevacizumab administration for HGG patients was independently analyzed for contrast enhancement (CE) and FLAIR sequences. Contemporary MMP2 and MMP9 plasma levels were assessed using ELISA kits. We analyzed 28 patients with a median Karnofsky Performance Status of 70 (range 50–80). A diffuse pattern was observed in 14 patients (50%). We did not observe any correlation between baseline imaging features and plasma levels of MMP2 or MMP9. We found no association between baseline MMP levels and diffuse MRI patterns. In univariate analyses, diffuse pattern, multi-focal disease, tumor diameter, surface area, and volume had no impact on outcome, while the number of lobes involved in CE and crossing of the midline by CE were associated with a worse progression-free survival (pu2009=u20090.072 and pu2009=u20090.012, respectively) and overall survival (pu2009=u20090.012 and pu2009<u20090.001, respectively). In patients with recurrent high-grade glioma treated with a bevacizumab-based regimen, our exploratory analysis of multiple MRI tumor characteristics at baseline failed to detect a relationship between imaging feature and plasma levels of MMP2 and MMP9. Our results suggests that number of lobes involved in CE and crossing of the midline by CE are associated with outcome although the potential prognostic versus predictive role of these markers warrant further investigation.


Journal of Neuro-oncology | 2016

Neuro-oncological patients admitted in intensive-care unit: predictive factors and functional outcome.

E. Tabouret; Celine Boucard; R. Devillier; M. Barrie; S. Boussen; D. Autran; O. Chinot; Nicolas Bruder

The prognosis of oncology patients admitted to the intensive care unit (ICU) is considered poor. Our objective was to analyze the characteristics and predictive factors of death in the ICU and functional outcome following ICU treatment for neuro-oncology patients. A retrospective study was conducted on all patients with primary brain tumor admitted to our institutional ICU for medical indications. Predictive impact on the risk of death in the ICU was analyzed as well as the functional status was evaluated prior and following ICU discharge. Seventy-one patients were admitted to the ICU. ICU admission indications were refractory seizures (41xa0%) and septic shock (17xa0%). On admission, 16xa0% had multi-organ failure. Ventilation was necessary for 41xa0% and catecholamines for 13xa0%. Twenty-two percent of patients died in the ICU. By multivariate analysis, predictive factors associated with an increased risk of ICU death were: non-neurological cause of admission [pxa0=xa00.045; odds ratio (OR) 5.405], multiple organ failure (pxa0=xa00.021; OR 8.027), respiratory failure (pxa0=xa00.006; OR 9.615), and hemodynamic failure (pxa0=xa00.008; OR 10.111). In contrast, tumor type (pxa0=xa00.678) and disease control status (pxa0=xa00.380) were not associated with an increased risk of ICU death. Among the 35 evaluable patients, 77xa0% presented with a stable or improved Karnofsky performance status following ICU hospitalization compared with the ongoing status before discharge. In patients with primary brain tumor admitted to the ICU, predictive factors of death appear to be similar to those described in non-oncology patients. ICU hospitalization is generally not associated with a subsequent decrease in the functional status.


Journal of Clinical Oncology | 2011

IDH mutation status impact on in vivo hypoxia biomarkers expression: New insights from a clinical, nuclear imaging, and immunohistochemical study in 34 patients with glioma.

P. Metellus; Carole Colin; David Taïeb; E. Guedj; I. Nanni-Metellus; Béma Coulibaly; C. Colavolpe; Stéphane Fuentes; Henry Dufour; M. Barrie; Olivier Chinot; L'Houcine Ouafik; Dominique Figarella-Branger

2078 Background: Mutations in the gene encoding isocitrate dehydrogenase enzyme isoforms 1 (IDH1) and 2 (IDH2) have recently been identified in a large proportion of glial tumors of the CNS, but their mechanistic role in tumor development and progression remains unclear. Here, we assessed the actual impact of IDH1 and IDH2 mutations in patients harboring WHO grade 2 and 3 gliomas.nnnMETHODSnWe sequenced IDH1 at codon 132 and IDH2 at codon 172 in 34 patients with WHO grade 2 and 3 gliomas who benefited from a preoperative 18F-FDG positron emission tomography (PET). Immunohistochemical expression of Hypoxia Inducible Factor alpha (HIF-1 a), Carbonic Anhydrase IX (CAIX) and Glucose Transporter 1 (GLUT1) along with the R132HIDH1 mutation was assessed in all cases as well as 1p19q deletion status and p53 expression.nnnRESULTSnHIF-1α expression was found in 15% of IDH-mutated compared to 14.3% of IDH-nonmutated tumors (P=0.954). Also, GLUT-1 positive staining was found in 5.% of IDH-mutated and in 7.1% of IDH-nonmutated tumors (P=0.794). Finally, CA-IX expression was found in 15% of IDH-mutated and in 7.1% of IDH-nonmutated tumors (P=0.484). The combined expression of these three hypoxic markers was found in 2 WHO grade 3 tumors, one of which was IDH-mutated whereas the other was IDH-nonmutated (P=0.794). In IDH-mutated tumors, the median SUVmax ratio was 2.24 vs. 2.15 in IDH-nonmutated tumors (P=0.775).nnnCONCLUSIONSnTogether, these data suggest the absence of correlation between IDH mutation status and in vivo hypoxic biomarkers expression in WHO grade 2 and 3 gliomas.


Journal of Clinical Oncology | 2005

Phase II study of temozolomide (TMZ) administered on a 7 days on-7 days off regimen as primary treatment before radiotherapy (RT) in inoperable newly diagnosed glioblastoma multiforme (GBM)

O. Chinot; M. Barrie; A. Cournede; Henry Dufour; Dominique Figarella-Branger; Diane Braguer; J.-C. Peragut; François Grisoli


Journal of Clinical Oncology | 2006

Methotrexate (MTX), procarbazine and CCNU for primary central nervous system lymphoma (PCNSL) in patients younger than 60: Can radiotherapy (RT) be deferred?

Antonio Omuro; Luc Taillandier; Olivier Chinot; C. Carnin; M. Barrie; Carole Soussain; Véronique Leblond; Khê Hoang-Xuan

Collaboration


Dive into the M. Barrie's collaboration.

Top Co-Authors

Avatar

Celine Boucard

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Olivier Chinot

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

O. Chinot

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

E. Tabouret

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Henry Dufour

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Didier Autran

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge