Marie-Ange Mouret-Reynier
University of Auvergne
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Marie-Ange Mouret-Reynier.
Cancer Biology & Therapy | 2010
Mathilde Bayet-Robert; Fabrice Kwiatowski; Marianne Leheurteur; Françoise Gachon; Eloïse Planchat; Catherine Abrial; Marie-Ange Mouret-Reynier; Xavier Durando; Chantal Barthomeuf; Philippe Chollet
Background. Since the improvement of chemotherapy with safe molecules is needed for better efficacy without supplementary toxicity, we investigated the feasibility and tolerability of the combination of docetaxel and curcumin, a polyphenolic derivative extracted from Curcuma longa Patients and methods. Patients with advanced or metastatic breast cancer were eligible. Docetaxel (100 mg/m2) was administered as a 1-hour i.v. infusion every 3 weeks on d1 for 6 cycles. Curcumin was orally given from 500 mg/d for 7 consecutive days by cycle (from d-4 to d+2) and escalated until a dose-limiting toxicity should occur. The primary endpoint of this study was to determine the maximal tolerated dose of the combination of dose-escalating curcumin and standard dose of docetaxel chemotherapy in advanced and metastatic breast cancer patients. Secondary objectives included toxicity, safety, vascular endothelial growth factor and tumor markers measurements, and assessment of objective and clinical responses to the combination therapy. Results. Fourteen patients were accrued in this open-label phase I trial. At the last dose level of curcumin, 3 dose-limiting toxicities were observed and 2 out of 3 patients at this dose level refused to continue treatment, leading us to define the maximal tolerated dose of curcumin at 8000 mg/d. Eight patients out of 14 had measurable lesions according to RECIST criteria, with 5 PR and 3 SD. Some improvements as biological and clinical responses were observed in most patients. Conclusion. The recommended dose of curcumin is 6000 mg/d for 7 consecutive days every 3 weeks in combination with a standard dose of docetaxel. From the encouraging efficacy results, a comparative phase II trial of this regimen plus docetaxel versus docetaxel alone is ongoing in advanced and metastatic breast cancer patients.
Oncologist | 2008
Frédérique Penault-Llorca; Catherine Abrial; Ines Raoelfils; Philippe Chollet; Anne Cayre; Marie-Ange Mouret-Reynier; Emilie Thivat; Florence Mishellany; Pierre Gimbergues; Xavier Durando
The current study expands upon previous work using a database of 710 patients treated with neoadjuvant chemotherapy. First, we studied phenotypic characteristics of tumors before and after chemotherapy using the following factors: the mitotic index of the Scarff-Bloom-Richardson grade, Ki-67, cyclin D1, and cyclo-oxygenase-2. Second, the predictive value of these factors on response was assessed. Third, we measured the prognostic impact of these markers post-therapy in comparison with clinical and pathological responses according to the Chevallier and Sataloff classifications. Patients were treated using different neoadjuvant chemotherapy combinations, mainly in successive prospective phase II trials. They received a median number of six cycles (range, 1-9). After neoadjuvant chemotherapy, patients underwent surgery and radiotherapy. In cases of important residual disease, some received additional courses of chemotherapy. In addition, menopausal patients with hormone receptor-positive tumors received tamoxifen for 5 years. According to our analysis, we found significant variations before and after neoadjuvant chemotherapy only for cyclin D1 and the mitotic index. Concerning the predictive value of biomarkers for response, Ki-67 and the mitotic index were predictive on univariate analysis, both for objective clinical and pathological complete responses. Because these two factors were correlated, no multivariate analyses were conducted. We then assessed the prognostic impact of the biopathological factors. When the factors were measured before chemotherapy, all were prognostic. When evaluated after chemotherapy, the mitotic index, objective clinical response, and pathological complete response were prognostic. Because these factors were correlated, no multivariate model was done. The main clinical fact is that there were significant correlations between clinical and pathological responses and variations in the biological factors studied.
Lancet Oncology | 2014
Bruno Coudert; Jean-Yves Pierga; Marie-Ange Mouret-Reynier; Kaldoun Kerrou; Jean-Marc Ferrero; Thierry Petit; Pierre Kerbrat; Pierre-Francois Dupre; Thomas Bachelot; Philippe Gabelle; Sylvia Giard; David Coeffic; Philippe Bougnoux; Jean-Briac Prevost; Gilles Paintaud; Gilles Thibault; Juana Hernandez; Mathieu Coudert; Laurent Arnould; Alina Berriolo-Riedinger
BACKGROUND An effective and well tolerated treatment is needed for patients with early HER2-positive breast cancer who do not achieve a pathological complete response after neoadjuvant therapy. The AVATAXHER trial aimed to predict pathological complete response early with the use of PET and to investigate whether the addition of bevacizumab could improve the proportion of patients achieving a pathological complete response in patients unlikely to respond to treatment. METHODS AVATAXHER was a randomised, open-label, non-comparative, multicentre phase 2 study that enrolled women (≥18 years of age) with early-stage HER2-positive breast cancer from 26 oncology centres in France. Patients initially received two cycles of neoadjuvant docetaxel (100 mg/m(2) intravenously every 3 weeks) plus trastuzumab (8 mg/kg intravenously every 3 weeks then 6 mg/kg intravenously every 3 weeks for the second course). Before the first and second cycles, [(18)F]-fluorodeoxyglucose (FDG) PET was done and the change in standardised uptake value was used to predict pathological complete response in each patient. Patients who were predicted to be responders on PET continued to receive standard therapy. Predicted non-responders were randomly assigned (2:1) to receive four cycles of docetaxel (100 mg/m(2) intravenously every 3 weeks) and trastuzumab (6 mg/kg intravenously every 3 weeks) plus bevacizumab (15 mg/kg intravenously every 3 weeks; group A) or continue on docetaxel plus trastuzumab alone (group B). Randomisation was open label and was done by an adaptive minimisation method. Although investigators and patients were aware of group assignment, the anatomo-pathologist in charge of centralised review of surgical samples and lymph nodes was masked to treatment assignment. The primary endpoint was centrally assessed pathological complete response according to the Chevallier classification. Efficacy analyses were done in the intention-to-treat population. Safety analyses in this Article were done on all patients who received at least one dose of treatment starting from cycle 3. Survival outcomes are not yet mature. This study is registered with ClinicalTrials.gov (NCT01142778) and EUDRACT (2009-013410-26). FINDINGS Between May 19, 2010, and Oct 1, 2012, 152 patients were recruited for the study. Ten patients were subsequently excluded, leaving 142 patients in the intention-to-treat population. Of these 142 patients, 69 were predicted by [(18)F]-FDG PET to be treatment responders after two cycles of treatment. The 73 predicted non-responders were randomly assigned to group A (n=48) and group B (n=25). Pathological complete responses were noted in 37 (53·6%, 95% CI 41·2-65·7) of the PET responders, 21 (43·8%, 29·5-58·8) of those in group A, and six (24·0%, 9·4-45·1) of those in group B. Incidences of grade 3-4 adverse events were similar in all three groups. The most common grade 3-4 adverse events were neutropenia (four in PET responders, five in group A, and three in group B), febrile neutropenia (one, three, and one, respectively), and myalgia (four, none, and one, respectively). Overall, 24 serious adverse events were reported in 15 patients (PET responders: nine events in four [6%] of 67 patients; group A: 14 events in ten [21%] of 47 patients; group B: one event in one [4%] of 25 patients). No deaths occurred during the study. INTERPRETATION In patients with HER2-positive breast cancer, early PET assessment can help to identify non-responders to neoadjuvant docetaxel plus trastuzumab therapy. In these patients, the addition of bevacizumab can increase the proportion of patients achieving a pathological complete response. This potential new role for PET and the activity of bevacizumab in this setting need to be confirmed in larger phase 3 trials. FUNDING Roche France.
Human Pathology | 2008
Frédérique Penault-Llorca; Catherine Abrial; Ines Raoelfils; Anne Cayre; Marie-Ange Mouret-Reynier; Marianne Leheurteur; Xavier Durando; Jean-Louis Achard; Pierre Gimbergues; Philippe Chollet
Pathologic complete response (pCR) is linked to a better outcome, but its definition varies among working groups. We performed this study to validate different expressions of pCR as well as to determine the role of in situ and isolated tumor cell residues. A pathologic review was conducted on 710 operable patients with breast cancer to assess the residual disease in breast and in nodes according to the Chevallier (Ch) and Sataloffs (Sa) classifications. The pCR rate was 14.3% according to the Chevallier and 25.8% according to the Sataloffs classification. Overall survival and disease-free survival have been compared according to the pathologic response. There were significant differences between the pCR Ch(1+2) or Sa(A) and the non-pCR group. No significant difference was found between classes Ch(1) versus Ch(2) and between class Sa(A) without isolated cells versus class Sa(A) with isolated cells. Conversely, tumors histologically modified by chemotherapy were associated with a better prognosis than unmodified tumors. Finally, evidence of pCR in nodes was associated with a better prognosis. pCR should be defined as an absence of node invasion, and in the breast, either absence of tumor or tumor residue less than 5% of the tumor.
Annals of Oncology | 2014
J-M Nabholtz; C. Abrial; Marie-Ange Mouret-Reynier; M.M. Dauplat; B. Weber; J. Gligorov; A. M. Forest; O. Tredan; L. Vanlemmens; T. Petit; S. Guiu; I. van Praagh; C. Jouannaud; Pascale Dubray-Longeras; N. Tubiana-Mathieu; K.E. Benmammar; S. Kullab; M.R.K. Bahadoor; N. Radosevic-Robin; F. Kwiatkowski; A. Desrichard; A. Cayre; N. Uhrhammer; N. Chalabi; Philippe Chollet; Frédérique Penault-Llorca
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors for some of which the epithelial growth factor receptor (EGFR) pathway may play an important role. We investigated the efficacy and toxicity of an anti-EGFR antibody (panitumumab) combined with a standard neoadjuvant anthracycline-taxane-based chemotherapy in patients with operable, stage II-III, TNBC. PATIENTS AND METHODS Treatment in this multicentric neoadjuvant pilot study consisted of panitumumab (9 mg/kg) for eight cycles q.3 weeks combined with four cycles of 5-fluorouracil, epidoxorubicin and cyclophosphamide (FEC100: 500/100/500 mg/m(2)) q.3 weeks, followed by four cycles of docetaxel (T: 100 mg/m(2)) q.3 weeks. Following therapy, all patients underwent surgical resection. Pathologic complete response (pCR) in assessable patients was the main end point while clinical response, toxicity and ancillary studies were secondary end points. Paraffin-embedded and frozen tumor samples were systematically collected with the aim to identify predictive biomarkers of efficacy and resistance in order to select biologically defined subpopulations for potential further clinical development of the anti-EGFR antibody. RESULTS Sixty patients were included with 47 assessable for pathologic response. The pCR rates were 46.8% [95% confidence interval (CI): 32.5% to 61.1%] and 55.3% [95% CI: 41.1% to 69.5%] according, respectively, to Chevallier and Sataloff classifications. The complete clinical response (cCR) rate was 37.5%. Conservative surgery was carried out in 87% of cases. Toxicity was manageable. The association of high EGFR and low cytokeratin 8/18 expression in tumor cells on one hand and high density of CD8+ tumor-infiltrating lymphocytes on the other hand were significantly predictive of pCR. CONCLUSIONS Panitumumab in combination with FEC100 followed by docetaxel appears efficacious, with acceptable toxicity, as neoadjuvant therapy of operable TNBC. Several biomarkers could help define large subsets of patients with a high probability of pCR, suggesting a potential interest to further develop this combination in biologically defined subgroups of patients with TNBC. CLINICAL TRIAL NUMBER NCT00933517.
Ejso | 2011
E. Monrigal; J. Dauplat; P. Gimbergues; G. Le Bouedec; M. Peyronie; J.L. Achard; Philippe Chollet; Marie-Ange Mouret-Reynier; J.M. Nabholtz; C. Pomel
PURPOSE To evaluate the feasability of immediate breast reconstruction (IBR) following mastectomy after neoadjuvant chemotherapy (NACT) and radiation therapy (RT) for operable invasive breast cancer (OIBC), in terms of incidence of local complications, locoregional control and survival. PATIENTS AND METHODS From 1990 to 2008, 210 patients were treated by NACT, RT and mastectomy with IBR for OIBC. One hundred and seven patients underwent a latissimus dorsi flap with implant (LDI), 56 patients a transverse rectus abdominis musculocutaneous (TRAM) flap, 25 an autologous latissimus dorsi flap (ALD) and 22, a retropectoral implant (RI) reconstruction. RESULTS Forty-six (21.9%) early events were recorded: 20 necrosis, 9 surgical site infections and 6 haematomas, requiring further surgery in 23 patients. More necrosis were observed with TRAM flap reconstructions (p = 0.000004), requiring more surgical revision than LD reconstructions. Seromas represented 42% of early complications in LD reconstructions. Fifty-five patients presented with late complications (26.2%) with mainly implant complications (capsular contracture, infection, dislocation, deflation) (23.6%), requiring reintervention in 14 cases. There were more delayed surgical revisions in RI reconstructions (p = 0.0005). The 5 years overall and disease-free survival rates were respectively 86.7% and 75.6%. Sixty-four patients presented at least one recurrence (30.5%) with 5 local, 9 locoregional and 54 distant relapses. CONCLUSION This therapeutic sequence does not seem to increase the IBR morbidity nor alter disease-free and overall survival.
British Journal of Cancer | 2003
Philippe Chollet; Sophie Amat; E. Belembaogo; Hervé Curé; M. De Latour; J. Dauplat; G. Le Bouëdec; Marie-Ange Mouret-Reynier; J.P. Ferrière; Frédérique Penault-Llorca
The Nottingham prognostic index (NPI), based on tumour size in breast, node involvement and Scarff–Bloom–Richardson (SBR) grading, has been shown to constitute a definitive prognostic factor of primary operable breast cancer in the adjuvant setting. We performed a retrospective study to evaluate the prognostic value of this index in 163 patients after neoadjuvant chemotherapy. Secondly, we examined the influence on survival of a revised NPI, only based on residual tumour size in breast and SBR grading in 228 patients, and consequently called breast grading index (BGI). The prognostic value of these two indices was also evaluated by replacing the SBR grade with the MSBR grade, a French modified SBR grading; the modified NPI (MNPI) and modified BGI (MBGI) were, respectively, obtained in 153 and 222 patients. At a median follow-up of 9.3 years, survival was significantly related to these four indices (P<0.001). Multivariate analysis revealed that MBGI was the only one which retained a prognostic influence on disease-free survival (P<0.02). In conclusion, the ‘amount’ of residual tumour in breast and/or nodes, as defined by NPI and revised indices, confers a determinant prognosis after neoadjuvant chemotherapy, inviting an alternative postsurgical treatment for a subgroup of patients with a decreased survival.
Expert Opinion on Pharmacotherapy | 2009
Jean-Marc Nabholtz; Marie-Ange Mouret-Reynier; Xavier Durando; Isabelle Van Praagh; Sana Al-Sukhun; Jean-Pierre Ferriere; Philippe Chollet
The hormonal therapy of patients with endocrine-sensitive early breast cancer has mainly consisted, for several decades, of the gold standard tamoxifen. The efficacy and favorable toxicity profiles of third-generation aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, in advanced disease led to their development in early breast cancer. Recent results consistently show the superiority of these agents over tamoxifen. Adjuvant trials evaluated AIs using four different therapeutic approaches: (1) Upfront strategy: randomization of newly diagnosed patients: tamoxifen for 5 years versus AI for 5 years. (2) Sequencial strategy: randomization of newly diagnosed patients: tamoxifen (2 – 3 years) followed by AI or the inverse for a total of 5 years versus upfront AI for 5 years. (3) Switch strategy: delayed randomization (or analysis) after 2 – 3 years of tamoxifen (patients free of disease): 2 – 3 years of tamoxifen versus 2 – 3 years of AI (total treatment 5 years). (4) Extended strategy: delayed randomization after 5 years of tamoxifen (patients free of disease): 2 – 5 years of AI versus placebo. Overall, AIs show evidence of superiority over tamoxifen in the adjuvant setting with proven improved efficacy and better toxicity profile. Despite some common characteristics, a body of evidence on AIs indicates some specific differences between the three agents in mechanism of action, pharmacokinetics, efficacy as well as toxicity profiles. Consequently, these hormonal agents may not be considered interchangeable in clinical practice. This review explores available results from AI trials and tries to define their present role in the adjuvant management of postmenopausal patients with breast cancer.
Cancer Journal | 2008
Philippe Chollet; Catherine Abrial; Xavier Durando; Emilie Thivat; Olivier Tacca; Marie-Ange Mouret-Reynier; Marianne Leheurteur; Fabrice Kwiatkowski; J. Dauplat; Frédérique Penault-Llorca
Purpose:Several factors have been shown to correlate with prognosis of patients with breast carcinoma. Among the most useful are node involvement, tumor size, and pathologic grade. These factors retained their prognostic value when assessed after neoadjuvant chemotherapy. Methods:Previously we used a revised Nottingham prognostic index and defined 3 related indices (breast grading index, modified Nottingham prognostic index, and modified breast grading index) that were also significantly related to overall survival and disease-free survival. To assess the postchemotherapy risk globally, we have combined the 3 pathologic factors to design a specific classification to evaluate residual disease. This new classification includes 4 risk levels (levels 1–4) according to residual disease magnitude after neoadjuvant chemotherapy in 710 patients with operable breast cancer. Results:This classification resulted in significantly different results for overall survival (P < 10−7) and disease-free survival (P = 8.3 × 10−7). Conclusion:This classification should help us in the selection of subgroups of patients for further adjuvant treatment.
Annals of Oncology | 2016
Suzette Delaloge; David Pérol; C. Courtinard; Etienne Brain; Bernard Asselain; Thomas Bachelot; M. Debled; V. Dieras; M Campone; Christelle Levy; William Jacot; V. Lorgis; Corinne Veyret; F. Dalenc; J-M Ferrero; L. Uwer; Pierre Kerbrat; Anthony Gonçalves; Marie-Ange Mouret-Reynier; Thierry Petit; C. Jouannaud; Laurence Vanlemmens; G. Chenuc; T. Guesmia; M. Robain; C. Cailliot
BACKGROUND Bevacizumab combined with paclitaxel as first-line chemotherapy for patients with HER2-negative metastatic breast cancer (MBC) has led to mixed results in randomized trials, with an improvement in progression-free survival (PFS) but no statistically significant overall survival (OS) benefit. Real-life data could help in assessing the value of this combination. PATIENTS AND METHODS This study aimed to describe the outcome following first-line paclitaxel with or without bevacizumab in the French Epidemiological Strategy and Medical Economics (ESME) database of MBC patients, established in 2014 by Unicancer. The primary and secondary end points were OS and PFS, respectively. RESULTS From 2008 to 2013, 14 014 MBC patient files were identified, including 10 605 patients with a HER2-negative status. Of these, 3426 received paclitaxel and bevacizumab (2127) or paclitaxel (1299) as first-line chemotherapy. OS adjusted for major prognostic factors was significantly longer in the paclitaxel and bevacizumab group compared with paclitaxel [hazard ratio (HR) 0.672, 95% confidence interval (CI) 0.601-0.752; median survival time 27.7 versus 19.8 months]. Results were consistent in all supportive analyses (using a propensity score for adjustment and as a matching factor for nested case-control analyses) and sensitivity analyses. Similar results were observed for the adjusted PFS, favoring the combination (HR 0.739, 95% CI 0.672-0.813; 8.1 versus 6.4 months). CONCLUSIONS In this large-scale, real-life setting, patients with HER2-negative MBC who received paclitaxel plus bevacizumab as first-line chemotherapy had a significantly better OS and PFS than those receiving paclitaxel. Despite robust methodology, real-life data are exposed to important potential biases, and therefore, results need to be treated with caution. Our data cannot therefore support extension of current use of bevacizumab in MBC.