Network


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

Hotspot


Dive into the research topics where Caroline Dartigeas is active.

Publication


Featured researches published by Caroline Dartigeas.


Blood | 2008

Rituximab combined with chemotherapy and interferon in follicular lymphoma patients: results of the GELA-GOELAMS FL2000 study

Gilles Salles; Nicolas Mounier; Sophie de Guibert; Franck Morschhauser; Chantal Doyen; Jean-François Rossi; Corinne Haioun; Pauline Brice; Beatrice Mahe; Reda Bouabdallah; Bruno Audhuy; Christophe Fermé; Caroline Dartigeas; Pierre Feugier; Catherine Sebban; Luc Xerri; Charles Foussard

The FL2000 study was undertaken to evaluate the combination of the anti-CD20 monoclonal antibody rituximab with chemotherapy plus interferon in the first-line treatment of follicular lymphoma patients with a high tumor burden. Patients were randomly assigned to receive either 12 courses of the chemotherapy regimen CHVP (cyclophosphamide, adriamycin, etoposide, and prednisolone) plus interferon-alpha2a (CHVP+I arm) over 18 months or 6 courses of the same chemotherapy regimen combined with 6 infusions of 375 mg/m(2) rituximab and interferon for the same time period (R-CHVP+I arm). After a median follow-up of 5 years, event-free survival estimates were, respectively, 37% (95% confidence interval [CI], 29%-44%) and 53% (95% CI, 45%-60%) in the CHVP+I and R-CHVP+I arm (P = .001). Five-year overall survival estimates were not statistically different in the CHVP+I (79%; 95% CI, 72%-84%) and R-CHVP+I (84%; 95% CI, 78%-84%) arms. In a multivariate regression analysis, event-free survival was significantly influenced by both the Follicular Lymphoma International Prognostic Index score (hazard ratio = 2.08; 95% CI, 1.6%-2.8%) and the treatment arm (hazard ratio = 0.59; 95% CI, 0.44%-0.78%). With a 5-year follow-up, the combination of rituximab with CHVP+I provides superior disease control in follicular lymphoma patients despite a shorter duration of chemotherapy. This studys clinical trial was registered at the National Institutes of Health website as no. NCT00136552.


Blood | 2011

Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine

S. Thepot; Bruno Quesnel; Francois Dreyfus; Odile Beyne-Rauzy; Pascal Turlure; Norbert Vey; Christian Recher; Caroline Dartigeas; Laurence Legros; Jacques Delaunay; Celia Salanoubat; Sorin Visanica; Aspasia Stamatoullas; Françoise Isnard; Anne Marfaing-Koka; Stéphane de Botton; Youcef Chelghoum; Anne-Laure Taksin; Isabelle Plantier; Shanti Ame; S. Boehrer; Claude Gardin; C.L. Beach; L. Ades; Pierre Fenaux

Prognostic factors for response and survival in higher-risk myelodysplastic syndrome patients treated with azacitidine (AZA) remain largely unknown. Two hundred eighty-two consecutive high or intermediate-2 risk myelodysplastic syndrome patients received AZA in a compassionate, patient-named program. Diagnosis was RA/RARS/RCMD in 4%, RAEB-1 in 20%, RAEB-2 in 54%, and RAEB-t (AML with 21%-30% marrow blasts) in 22%. Cytogenetic risk was good in 31%, intermediate in 17%, and poor in 47%. Patients received AZA for a median of 6 cycles (1-52). Previous low-dose cytosine arabinoside treatment (P = .009), bone marrow blasts > 15% (P = .004), and abnormal karyotype (P = .03) independently predicted lower response rates. Complex karyotype predicted shorter responses (P = .0003). Performance status ≥ 2, intermediate- and poor-risk cytogenetics, presence of circulating blasts, and red blood cell transfusion dependency ≥ 4 units/8 weeks (all P < 10(-4)) independently predicted poorer overall survival (OS). A prognostic score based on those factors discriminated 3 risk groups with median OS not reached, 15.0 and 6.1 months, respectively (P < 10(-4)). This prognostic score was validated in an independent set of patients receiving AZA in the AZA-001 trial (P = .003). Achievement of hematological improvement in patients who did not obtain complete or partial remission was associated with improved OS (P < 10(-4)). In conclusion, routine tests can identify subgroups of patients with distinct prognosis with AZA treatment.


Blood | 2010

Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM)

Sylvain Thépot; Valerie Seegers; Emmanuel Raffoux; Bruno Quesnel; Yasmine Chait; Lucile Sorin; Francois Dreyfus; Thomas Cluzeau; Jacques Delaunay; Laurence Sanhes; Virginie Eclache; Caroline Dartigeas; Pascal Turlure; Stephanie Harel; Celia Salanoubat; Jean-Jacques Kiladjian; Pierre Fenaux; Lionel Ades

Transformation of Philadelphia (Ph)-negative myeloproliferative neoplasms (MPNs) to myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) is associated with poor response to chemotherapy and short survival. Fifty-four patients with Ph-negative MPN (including 21 essential thrombocythemia [ET], 21 polycythemia vera [PV], 7 primary myelofibrosis, and 5 unclassified MPN) who had progressed to AML (n = 26) or MDS (n = 28) were treated with azacitidine in a patient-named program. Overall response rate was 52% (24% complete response [CR], 11% partial response [PR], 8% marrow CR or CR with incomplete recovery of cytopenias, 9% hematologic improvement) and median response duration was 9 months. Prognostic factors were for overall response the underlying MPN (71% vs 33% responses in ET and PV, respectively; P = .016); prognostic factors for CR achievement were the underlying MPN (14% CR for PV vs 43% for ET; P = .040) and World Health Organization classification at transformation (36% vs 12% CR in MDS and AML, respectively, P = .038). Recurrence of chronic phase features of the initial MPN was observed in 39% of the responders. Median overall survival was 11 months. Azacitidine gives encouraging results in Ph-negative MPN having progressed to AML or MDS, but response duration is short, and consolidation treatments have to be evaluated.


Journal of Clinical Oncology | 2013

Results of a Randomized Trial of Chlorambucil Versus Fludarabine for Patients With Untreated Waldenström Macroglobulinemia, Marginal Zone Lymphoma, or Lymphoplasmacytic Lymphoma

Véronique Leblond; Steve Johnson; Sylvie Chevret; Adrian Copplestone; Simon Rule; Olivier Tournilhac; John F. Seymour; Russell Patmore; David Wright; Pierre Morel; Marie-Sarah Dilhuydy; Sara Willoughby; Caroline Dartigeas; Marion Malphettes; Bruno Royer; Maeve Ewings; Guy Pratt; Julie Lejeune; Sylvain Choquet; Roger Owen

PURPOSE Treatment options for patients with Waldenström macroglobulinemia (WM) and closely related disorders include alkylating agents, purine analogs, and monoclonal antibodies. No large randomized studies have yet been reported comparing any of these approaches. PATIENTS AND METHODS The randomized WM1 study (Trial Comparing Chlorambucil to Fludarabine in Patients With Advanced Waldenström Macroglobulinemia) was undertaken in 101 centers in five countries enrolling 414 eligible patients (339 with WM, 37 with non-mucosa-associated lymphoid tissue marginal zone lymphoma, and 38 with lymphoplasmacytic lymphoma) who were randomly assigned to receive chlorambucil or fludarabine. The primary end point was the overall response rate (ORR). RESULTS On the basis of intent-to-treat analysis, the ORR was 47.8% (95% CI, 40.9% to 54.8%) in the fludarabine arm versus 38.6% (95% CI, 32.0% to 45.7%) in the chlorambucil arm (P = .07). With a median follow-up of 36 months (interquartile range, 18 to 58 months), median progression-free survival (PFS), and duration of response (DR) were significantly improved in the fludarabine arm compared with the chlorambucil arm: PFS, 36.3 versus 27.1 months (P = .012) and DR, 38.3 versus 19.9 months (P < .001). In patients with WM, median overall survival (OS) was not reached in the fludarabine arm versus 69.8 months in the chlorambucil arm (95% CI, 61.6 to 79.8 months; P = .014). Grade 3 to 4 neutropenia was significantly higher among patients treated with fludarabine (36%) compared with patients treated with chlorambucil (17.8%; P < .001). Second malignancies were significantly more frequent in the chlorambucil arm with 6-year cumulative incidence rate of 20.6% versus 3.7% in the fludarabine arm (P = .001). CONCLUSION In the complete intent-to-treat study population, fludarabine significantly improved PFS compared with chlorambucil, and in patients with WM, it improved OS.


American Journal of Hematology | 2014

Azacitidine in untreated acute myeloid leukemia: a report on 149 patients.

Sylvain Thépot; Valerie Seegers; Christian Recher; Emmanuel Raffoux; Bruno Quesnel; Jacques Delaunay; Thomas Cluzeau; Anne Marfaing Koka; Aspasia Stamatoullas; M.P. Chaury; Caroline Dartigeas; Stéphane Cheze; Anne Banos; Pierre Morel; Isabelle Plantier; Anne-Laure Taksin; Jean Pierre Marolleau; Cécile Pautas; Xavier Thomas; F. Isnard; Blandine Beve; Yasmine Chait; Agnès Guerci; Norbert Vey; Francois Dreyfus; Lionel Ades; Norbert Ifrah; Hervé Dombret; Pierre Fenaux; Claude Gardin

Limited data are available on azacitidine (AZA) treatment and its prognostic factors in acute myeloid leukemia (AML). One hundred and forty‐nine previously untreated AML patients considered ineligible for intensive chemotherapy received AZA in a compassionate patient‐named program. AML diagnosis was de novo, post‐myelodysplastic syndromes (MDS), post‐MPN, and therapy‐related AML in 51, 55, 13, and 30 patients, respectively. Median age was 74 years, median white blood cell count (WBC) was 3.2 × 109/L and 58% of the patients had ≥30% marrow blasts. Cytogenetics was adverse in 60 patients. Patients received AZA for a median of five cycles (range 1–31). Response rate (including complete remission/CR with incomplete recovery/partial remission) was 27.5% after a median of three cycles (initial response), and 33% at any time (best response). Only adverse cytogenetics predicted poorer response. Median overall survival (OS) was 9.4 months. Two‐year OS was 51% in responders and 10% in non‐responders (P<0.0001). Adverse cytogenetics, WBC >15 × 109/L and ECOG‐PS ≥2 predicted poorer OS, while age and marrow blast percentage had no impact. Using MDS IWG 2006 response criteria, among patients with stable disease, those with hematological improvement had no significant survival benefit in a 7 months landmark analysis. Outcomes observed in this high‐risk AML population treated with AZA deserve comparison with those of patients treated intensively in prospective studies. Am. J. Hematol. 89:410–416, 2014.


Blood | 2012

The revised IPSS is a powerful tool to evaluate the outcome of MDS patients treated with azacitidine: the GFM experience

Mathilde Lamarque; Sophie Raynaud; Sylvain Thépot; Bruno Quesnel; Francois Dreyfus; Odile Beyne Rauzy; Pascal Turlure; Norbert Vey; Christian Recher; Caroline Dartigeas; Laurence Legros; Jacques Delaunay; Sorin Visanica; Aspasia Stamatoullas; Pierre Fenaux; Lionel Ades

To the editor: The International Prognostic Scoring System (IPSS), which is based on cytogenetics, BM blast percentage, and number of cytopenias, has played a major role in prognosis assessment in myelodysplastic syndrome (MDS) since its publication in 1997. The recently published revised IPSS (


Leukemia Research | 2013

Azacitidine in the treatment of therapy related myelodysplastic syndrome and acute myeloid leukemia (tMDS/AML): A report on 54 patients by the Groupe Francophone Des Myelodysplasies (GFM)

Cecile Bally; Sylvain Thépot; Bruno Quesnel; Norbert Vey; Francois Dreyfus; Jehane Fadlallah; Pascal Turlure; Stéphane de Botton; Caroline Dartigeas; Benoit de Renzis; Pierre Fenaux; Lionel Ades

The effect of azacitidine (AZA) in therapy related MDS and AML (t-MDS/AML) is not well established. 54 patients (42 t-MDS and 12 t-AML), 71% of whom had complex karyotype, received AZA for at least one cycle (median 4 cycles). The overall response rate (ORR) was 39% in the whole cohort and 62% in patients who received ≥4 cycles. One, 2 and 3 year OS was 36%, 14% and 8% respectively. Female gender (p=0.01) and ECOG 0-1 (p=0.04) were associated with significantly better OS, while karyotype and marrow blast percentage had no significant impact. By comparison with de novo MDS/AML treated in the same program, t-MDS/AML had a similar response rate (38% vs 45% in de novo MDS/AML, p=0.53), but significantly shorter OS (2 year OS of 14% vs 33.9%, p=0.0005). However, in a multivariate analysis performed in all patients (de novo and therapy related cases), only complex karyotype and high IPSS, and not etiology (i.e. de novo versus therapy related), had a significant impact on OS. Nine (15%) patients received allogeneic stem cell transplantation, 4 of whom were still alive.


Annals of Oncology | 2012

Combination of rituximab, bortezomib, doxorubicin, dexamethasone and chlorambucil (RiPAD+C) as first-line therapy for elderly mantle cell lymphoma patients: results of a phase II trial from the GOELAMS

Roch Houot; S. Le Gouill; M. Ojeda Uribe; Christiane Mounier; Stéphane Courby; Caroline Dartigeas; Kamal Bouabdallah; M. Alexis Vigier; Marie-Pierre Moles; O. Tournilhac; Nina Arakelyan; Philippe Rodon; A. El Yamani; Laurent Sutton; Luc-Matthieu Fornecker; D. Assouline; Harousseau Jl; Hervé Maisonneuve; Sylvie Caulet-Maugendre; Remy Gressin

BACKGROUND There is no consensual first-line chemotherapy for elderly patients with mantle cell lymphoma (MCL). The GOELAMS (Groupe Ouest-Est des Leucémies Aiguës et Maladies du Sang) group previously developed the (R)VAD+C regimen (rituximab, vincristine, doxorubicin, dexamethasone and chlorambucil), which appeared as efficient as R-CHOP (rituximab, cyclophosphamide, doxorubicine, vincristine, prednisone) while less toxic. Based on this protocol, we now added bortezomib (RiPAD+C: rituximab, bortezomib, doxorubicin, dexamethasone and chlorambucil) given its efficacy in relapsed/refractory MCL patients. The goal of the current phase II trial was to evaluate the feasibility and efficacy of the RiPAD+C regimen as frontline therapy for elderly patients with MCL. PATIENTS AND METHODS Patients between 65 and 80 years of age with newly diagnosed MCL received up to six cycles of RiPAD+C. RESULTS Thirty-nine patients were enrolled. Median age was 72 years (65-80). After four cycles of RiPAD+C, the overall response rate was 79%, including 51% complete responses (CRs). After six cycles, CR rate increased up to 59%. After a 27-month follow-up, median progression-free survival (PFS) is 26 months and median overall survival has not been reached. Four patients (10%) discontinued the treatment because of a severe toxicity and seven patients (18%) experienced grade 3 neurotoxicity. CONCLUSION The bortezomib-containing RiPAD+C regimen results in high CR rates and prolonged PFS with predictable and manageable toxic effects in elderly patients with MCL.


Leukemia Research | 2015

Azacitidine for the treatment of relapsed and refractory AML in older patients

Sylvain Thépot; Céline Berthon; Jacques Delaunay; D. Bouscary; Thomas Cluzeau; Pascal Turlure; Thomas Prebet; Caroline Dartigeas; Jean-Pierre Marolleau; Christian Recher; Isabelle Plantier; Aspasia Stamatoullas; Alain Devidas; Anne-Laure Taksin; Romain Guièze; Denis Caillot; Norbert Vey; Lionel Ades; Norbert Ifrah; Hervé Dombret; Pierre Fenaux; Claude Gardin

The prognosis of patients older than 50 with relapsed or refractory AML is dismal. Azacitidine has been investigated in older AML patients. Here we report the outcome of 130 patients older than 50 years included in a multicenter patient named program of azacitidine after relapse (n=67) or induction failure (n=63) of intensive chemotherapy. Median age was 67 years, cytogenetic risk was high in 28% and performance status ≥2 in 15% of cases. Most (72%) patients received azacitidine at the standard schedule (75mg/m(2)/d, 7 days/month) for a median of 4 courses. The overall response rate was 17% (CR: 10%, CRi: 7%). Median overall survival was 8.4 months. Achievement of CR/CRi was associated with prolonged survival (P=0.0001), whereas hematological improvement according to MDS criteria, achieved in 36% of patients with resistant disease, did not improve survival. In multivariate analysis, high risk cytogenetics (P=0.022) and peripheral blasts >10% (P<0.0001) at onset of azacitidine were independently predictive of poor prognosis. Combining these two factors, we identified a subgroup of 48% of patients with intermediate risk cytogenetics and peripheral blasts ≤10% and a median OS of 11.3 months. These results warrant further investigation of azacitidine-based regimens in this subgroup of patients.


The New England Journal of Medicine | 2017

Rituximab after Autologous Stem-Cell Transplantation in Mantle-Cell Lymphoma

Steven Le Gouill; Catherine Thieblemont; Lucie Oberic; Anne Moreau; Krimo Bouabdallah; Caroline Dartigeas; Gandhi Damaj; Thomas Gastinne; Vincent Ribrag; Pierre Feugier; Olivier Casasnovas; Hacene Zerazhi; Corinne Haioun; Hervé Maisonneuve; Roch Houot; Fabrice Jardin; Eric Van Den Neste; Olivier Tournilhac; Katell Le Dû; Franck Morschhauser; Guillaume Cartron; Luc-Matthieu Fornecker; Danielle Canioni; Mary Callanan; Marie C. Béné; Gilles Salles; Hervé Tilly; Thierry Lamy; Remy Gressin; Olivier Hermine

BACKGROUND Mantle‐cell lymphoma is generally incurable. Despite high rates of complete response after initial immunochemotherapy followed by autologous stem‐cell transplantation, patients have relapses. We investigated whether rituximab maintenance therapy at a dose of 375 mg per square meter of body‐surface area administered every 2 months for 3 years after transplantation would prolong the duration of response. METHODS In a phase 3 trial involving 299 patients who were younger than 66 years of age at diagnosis, we randomly assigned 240 patients to receive rituximab maintenance therapy or to undergo observation after autologous stem‐cell transplantation (120 patients per group); 59 patients did not undergo randomization. The primary end point was event‐free survival (with an event defined as disease progression, relapse, death, allergy to rituximab, or severe infection) after transplantation among patients who underwent randomization. RESULTS After four courses of immunochemotherapy induction (rituximab, dexamethasone, cytarabine, and a platinum derivative [R‐DHAP]), the overall response rate was 89%, and the complete response rate 77%. Transplantation was performed in 257 patients. The median follow‐up from randomization after transplantation was 50.2 months (range, 46.4 to 54.2). Starting from randomization, the rate of event‐free survival at 4 years was 79% (95% confidence interval [CI], 70 to 86) in the rituximab group versus 61% (95% CI, 51 to 70) in the observation group (P=0.001). The rate of progression‐free survival at 4 years was 83% (95% CI, 73 to 88) in the rituximab group versus 64% (95% CI, 55 to 73) in the observation group (P<0.001). The rate of overall survival was 89% (95% CI, 81 to 94) in the rituximab group versus 80% (95% CI, 72 to 88) in the observation group (P=0.04). According to a Cox regression unadjusted analysis, the rate of overall survival at 4 years was higher in the rituximab group than in the observation group (hazard ratio for death, 0.50; 95% CI, 0.26 to 0.99; P=0.04). CONCLUSIONS Rituximab maintenance therapy after transplantation prolonged event‐free survival, progression‐free survival, and overall survival among patients with mantle‐cell lymphoma who were younger than 66 years of age at diagnosis. (Funded by Roche and Amgen; LyMa ClinicalTrials.gov number, NCT00921414.)

Collaboration


Dive into the Caroline Dartigeas'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

Norbert Vey

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge