Network


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

Hotspot


Dive into the research topics where Norbert Ifrah is active.

Publication


Featured researches published by Norbert Ifrah.


The New England Journal of Medicine | 1996

A Prospective, Randomized Trial of Autologous Bone Marrow Transplantation and Chemotherapy in Multiple Myeloma

Michel Attal; Jean-Luc Harousseau; Anne-Marie Stoppa; Jean-Jacques Sotto; Jean-Gabriel Fuzibet; Jean-François Rossi; Philippe Casassus; Hervé Maisonneuve; Thierry Facon; Norbert Ifrah; Catherine Payen; Régis Bataille

Background The median survival of patients with myeloma after conventional chemotherapy is three years or less. Promising results have been reported with high-dose therapy supported by autologous bone marrow transplantation. We conducted a randomized study comparing conventional chemotherapy and high-dose therapy. Methods Two hundred previously untreated patients under the age of 65 years who had myeloma were randomly assigned at the time of diagnosis to receive either conventional chemotherapy or high-dose therapy and autologous bone marrow transplantation. Results The response rate among the patients who received high-dose therapy was 81 percent (including complete responses in 22 percent and very good partial responses in 16 percent), whereas it was 57 percent (complete responses in 5 percent and very good partial responses in 9 percent) in the group treated with conventional chemotherapy (P<0.001). The probability of event-free survival for five years was 28 percent in the high-dose group and 10 perce...


Blood | 2013

Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia

Eric Jourdan; Nicolas Boissel; Sylvie Chevret; Eric Delabesse; Aline Renneville; Pascale Cornillet; Odile Blanchet; Jean-Michel Cayuela; Christian Recher; Emmanuel Raffoux; Jacques Delaunay; Arnaud Pigneux; Claude-Eric Bulabois; Céline Berthon; Cécile Pautas; Norbert Vey; Bruno Lioure; Xavier Thomas; Isabelle Luquet; Christine Terré; Philippe Guardiola; Marie C. Béné; Claude Preudhomme; Norbert Ifrah; Hervé Dombret

Not all patients with core binding factor acute myeloid leukemia (CBF-AML) display a good outcome. Modern risk factors include KIT and/or FLT3 gene mutations and minimal residual disease (MRD) levels, but their respective values have never been prospectively assessed. A total of 198 CBF-AML patients were randomized between a reinforced and a standard induction course, followed by 3 high-dose cytarabine consolidation courses. MRD levels were monitored prospectively. Gene mutations were screened at diagnosis. Despite a more rapid MRD decrease after reinforced induction, induction arm did not influence relapse-free survival (RFS) (64% in both arms; P = .91). Higher WBC, KIT, and/or FLT3-ITD/TKD gene mutations, and a less than 3-log MRD reduction after first consolidation, were associated with a higher specific hazard of relapse, but MRD remained the sole prognostic factor in multivariate analysis. At 36 months, cumulative incidence of relapse and RFS were 22% vs 54% (P < .001) and 73% vs 44% (P < .001) in patients who achieved 3-log MRD reduction vs the others. These results suggest that MRD, rather than gene mutations, should be used for future treatment stratifications in CBF-AML patients. This trial was registered at EudraCT as #2006-005163-26 and at www.clinicaltrials.gov as #NCT 00428558.


Lancet Oncology | 2014

Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials

Robert Kerrin Hills; Sylvie Castaigne; Frederick R. Appelbaum; Jacques Delaunay; Stephen H. Petersdorf; Megan Othus; Elihu H. Estey; Hervé Dombret; Sylvie Chevret; Norbert Ifrah; Jean-Yves Cahn; Christian Recher; Lucy Chilton; Anthony V. Moorman; Alan Kenneth Burnett

BACKGROUND Gemtuzumab ozogamicin was the first example of antibody-directed chemotherapy in cancer, and was developed for acute myeloid leukaemia. However, randomised trials in which it was combined with standard induction chemotherapy in adults have produced conflicting results. We did a meta-analysis of individual patient data to assess the efficacy of adding gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia. METHODS We searched PubMed for reports of randomised controlled trials published in any language up to May 1, 2013, that included an assessment of gemtuzumab ozogamicin given to adults (aged 15 years and older) in conjunction with the first course of intensive induction chemotherapy for acute myeloid leukaemia (excluding acute promyelocytic leukaemia) compared with chemotherapy alone. Published data were supplemented with additional data obtained by contacting individual trialists. The primary endpoint of interest was overall survival. We used standard meta-analytic techniques, with an assumption-free (or fixed-effect) method. We also did exploratory stratified analyses to investigate whether any baseline features predicted a greater or lesser benefit from gemtuzumab ozogamicin. FINDINGS We obtained data from five randomised controlled trials (3325 patients); all trials were centrally randomised and open label, with overall survival as the primary endpoint. The addition of gemtuzumab ozogamicin did not increase the proportion of patients achieving complete remission with or without complete peripheral count recovery (odds ratio [OR] 0·91, 95% CI 0·77-1·07; p=0·3). However, the addition of gemtuzumab ozogamicin significantly reduced the risk of relapse (OR 0·81, 0·73-0·90; p=0·0001), and improved overall survival at 5 years (OR 0·90, 0·82-0·98; p=0·01). At 6 years, the absolute survival benefit was especially apparent in patients with favourable cytogenetic characteristics (20·7%; OR 0·47, 0·31-0·73; p=0·0006), but was also seen in those with intermediate characteristics (5·7%; OR 0·84, 0·75-0·95; p=0·005). Patients with adverse cytogenetic characteristics did not benefit (2·2%; OR 0·99, 0·83-1·18; p=0·9). Doses of 3 mg/m(2) were associated with fewer early deaths than doses of 6 mg/m(2), with equal efficacy. INTERPRETATION Gemtuzumab ozogamicin can be safely added to conventional induction therapy and provides a significant survival benefit for patients without adverse cytogenetic characteristics. These data suggest that the use of gemtuzumab ozogamicin should be reassessed and its licence status might need to be reviewed. FUNDING None.Background Gemtuzumab Ozogamicin (GO) was the first example of antibody directed chemotherapy in cancer and developed for Acute Myeloid Leukaemia. Its role has been unclear. Five randomised trials where it was combined with standard induction chemotherapy in adults have produced different results. In an effort to clarify the level of benefit, if any, and in which patients outcomes might be improved, an individual patient data meta-analysis of these 5 trials has been undertaken.


British Journal of Haematology | 1997

Avascular necrosis of bone after allogeneic bone marrow transplantation: analysis of risk factors for 4388 patients by the Société Française de Greffe de Moëlle (SFGM)

Gérard Socié; Jean-Yves Cahn; J. Carmelo; Jean-Paul Vernant; Jean-Pierre Jouet; Norbert Ifrah; Noel Milpied; Mauricette Michallet; Bruno Lioure; J. L. Pico; Francis Witz; L. Molina; Alain Fischer; V. J. Bardou; Eliane Gluckman; Josy Reiffers

Increasing numbers of patients are surviving after allogeneic bone marrow transplantation and are therefore at risk for developing late complications. Among these complications, avascular necrosis of bone has been reported, but only two single‐centre studies included sufficient patients to enable analysis of the risk factors for developing avascular necrosis. In this multicentre retrospective study the aim was to assess risk factors for this complication in a large number of patients.


Blood | 2014

Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia

Kheira Beldjord; Sylvie Chevret; Vahid Asnafi; Françoise Huguet; Marie-Laure Boulland; Thibaut Leguay; Xavier Thomas; Jean-Michel Cayuela; Nathalie Grardel; Yves Chalandon; Nicolas Boissel; Beat Schaefer; Eric Delabesse; Hélène Cavé; Patrice Chevallier; Agnès Buzyn; Thierry Fest; Oumedaly Reman; Jean-Paul Vernant; Véronique Lhéritier; Marie C. Béné; Marina Lafage; Elizabeth Macintyre; Norbert Ifrah; Hervé Dombret

With intensified pediatric-like therapy and genetic disease dissection, the field of adult acute lymphoblastic leukemia (ALL) has evolved recently. In this new context, we aimed to reassess the value of conventional risk factors with regard to new genetic alterations and early response to therapy, as assessed by immunoglobulin/T-cell receptor minimal residual disease (MRD) levels. The study was performed in 423 younger adults with Philadelphia chromosome-negative ALL in first remission (265 B-cell precursor [BCP] and 158 T-cell ALL), with cumulative incidence of relapse (CIR) as the primary end point. In addition to conventional risk factors, the most frequent currently available genetic alterations were included in the analysis. A higher specific hazard of relapse was independently associated with postinduction MRD level ≥10(-4) and unfavorable genetic characteristics (ie, MLL gene rearrangement or focal IKZF1 gene deletion in BCP-ALL and no NOTCH1/FBXW7 mutation and/or N/K-RAS mutation and/or PTEN gene alteration in T-cell ALL). These 2 factors allowed definition of a new risk classification that is strongly associated with higher CIR and shorter relapse-free and overall survival. These results indicate that genetic abnormalities are important predictors of outcome in adult ALL not fully recapitulated by early response to therapy. Patients included in this study were treated in the multicenter GRAALL-2003 and GRAALL-2005 trials. Both trials were registered at http://www.clinicaltrials.gov as #NCT00222027 and #NCT00327678, respectively.


British Journal of Haematology | 1999

Allogeneic bone marrow transplantation in aggressive non-Hodgkin's lymphoma (excluding Burkitt and lymphoblastic lymphoma): a series of 73 patients from the SFGM database

Nathalie Dhedin; Giraudier S; Gaulard P; Helene Esperou; Norbert Ifrah; Mauricette Michallet; Noel Milpied; Bernard Rio; Jean-Yves Cahn; Molina L; Laporte Jl; François Guilhot; Mathieu Kuentz

The place of allogeneic bone marrow transplantation (BMT) in the treatment of aggressive non‐Hodgkins lymphoma (NHL) remains controversial. We conducted a retrospective study of French experience in allografting NHL between 1984 and 1994. To improve the homogeneity of the study population, cases of low‐grade, Burkitt and lymphoblastic NHL were excluded. 73 patients were included in the analysis. Median age at transplantation was 35 years (range 9–61 years); 64 patients were in stage IV and 45 had bone marrow involvement at diagnosis. At the time of transplantation, 46 patients had sensitive disease (25 in complete remission; CR).


Haematologica | 2010

Mutations in TP53 are exclusively associated with del(17p) in multiple myeloma

Laurence Lodé; Marion Eveillard; Valérie Trichet; Thierry Soussi; Soraya Wuilleme; Steven Richebourg; Florence Magrangeas; Norbert Ifrah; Loic Campion; Catherine Traulle; François Guilhot; Denis Caillot; Gerald Marit; Claire Mathiot; Thierry Facon; Michel Attal; Jean-Luc Harousseau; Philippe Moreau; Stephane Minvielle; Hervé Avet-Loiseau

Deletion of the 17p13 chromosomal region [del(17p)] is associated with a poor outcome in multiple myeloma. Most of the studies have targeted the TP53 gene for deletion analyses, although no study showed that this gene is the deletion target. In order to address this issue, we sequenced the TP53 gene in 92 patients with multiple myeloma at diagnosis, 54 with a del(17p) and 38 lacking del(17p). At least one mutation was found in 20 patients, all of them presenting a del(17p). The analysis of the mutation location showed that virtually all of them occurred in highly conserved domains involved in the DNA-protein interactions. In conclusion, we showed that 37% of the myeloma patients with del(17p) present a TP53 mutation versus 0% in patients lacking the del(17p). The prognostic significance of these mutations remains to be evaluated.


Blood | 2015

Role of allogeneic stem cell transplantation in adult patients with Ph-negative acute lymphoblastic leukemia.

Anne Huynh; Sébastien Maury; Reza Tabrizi; Kheira Beldjord; Vahid Asnafi; Xavier Thomas; Patrice Chevallier; Stéphanie Nguyen; Valérie Coiteux; Jean-Henri Bourhis; Yosr Hichri; Martine Escoffre-Barbe; Oumedaly Reman; Carlos Graux; Yves Chalandon; Didier Blaise; Urs Schanz; Véronique Lhéritier; Jean-Yves Cahn; Hervé Dombret; Norbert Ifrah

Because a pediatric-inspired Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) protocol yielded a markedly improved outcome in adults with Philadelphia chromosome-negative ALL, we aimed to reassess the role of allogeneic stem cell transplantation (SCT) in patients treated in the GRAALL-2003 and GRAALL-2005 trials. In all, 522 patients age 15 to 55 years old and presenting with at least 1 conventional high-risk factor were candidates for SCT in first complete remission. Among these, 282 (54%) received a transplant in first complete remission. At 3 years, posttransplant cumulative incidences of relapse, nonrelapse mortality, and relapse-free survival (RFS) were estimated at 19.5%, 15.5%, and 64.7%, respectively. Time-dependent analysis did not reveal a significant difference in RFS between SCT and no-SCT cohorts. However, SCT was associated with longer RFS in patients with postinduction minimal residual disease (MRD) ≥10(-3) (hazard ratio, 0.40) but not in good MRD responders. In B-cell precursor ALL, SCT also benefitted patients with focal IKZF1 gene deletion (hazard ratio, 0.42). This article shows that poor early MRD response, in contrast to conventional ALL risk factors, is an excellent tool to identify patients who may benefit from allogeneic SCT in the context of intensified adult ALL therapy. Trial GRAALL-2003 was registered at www.clinicaltrials.gov as #NCT00222027; GRAALL-2005 was registered as #NCT00327678.


Biology of Blood and Marrow Transplantation | 2013

Long-term follow-up of the imatinib GRAAPH-2003 study in newly diagnosed patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: a GRAALL study.

Aline Tanguy-Schmidt; Philippe Rousselot; Yves Chalandon; Jean-Michel Cayuela; Sandrine Hayette; Martine Escoffre; Françoise Huguet; Delphine Rea; Jean-Yves Cahn; Jean-Paul Vernant; Norbert Ifrah; Hervé Dombret; Xavier Thomas

We report here the results of the GRAAPH-2003 trial with long-term follow-up in 45 patients with de novo Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Imatinib-based strategy improved the 4-year overall survival (OS) up to 52% versus 20% in the pre-imatinib LALA-94 trial (P = .0001). Despite the selection in patients who actually underwent transplantation, these results suggest that allogeneic or autologous stem cell transplants (SCTs) still have a place in overcoming the poor prognosis of Ph+ ALL in the era of imatinib therapy. OS was 50% after allogeneic SCT (24 patients), 33% in patients without a transplantation (9 patients), and 80% after autologous SCT (10 patients without allogeneic donor or >55 years, including 7 patients in complete molecular response).


Blood | 2011

Phase 1/2 study to assess the safety, efficacy, and pharmacokinetics of barasertib (AZD1152) in patients with advanced acute myeloid leukemia

Bob Löwenberg; Petra Muus; Gert J. Ossenkoppele; Philippe Rousselot; Jean Yves Cahn; Norbert Ifrah; Giovanni Martinelli; S. Amadori; Ellin Berman; Pieter Sonneveld; Mojca Jongen-Lavrencic; Sophie Rigaudeau; Paul Stockman; Alison Goudie; Stefan Faderl; Elias Jabbour; Hagop M. Kantarjian

The primary objective of this 2-part phase 1/2 study was to determine the maximum-tolerated dose (MTD) of the potent and selective Aurora B kinase inhibitor barasertib (AZD1152) in patients with newly diagnosed or relapsed acute myeloid leukemia (AML). Part A determined the MTD of barasertib administered as a continuous 7-day infusion every 21 days. In part B, the efficacy of barasertib was evaluated at the MTD. In part A, 32 patients were treated with barasertib 50 mg (n = 3), 100 mg (n = 3), 200 mg (n = 3), 400 mg (n = 4), 800 mg (n = 7), 1200 mg (n = 6), and 1600 mg (n = 6). Dose-limiting toxicities (stomatitis/mucosal inflammation events) were reported in the 800 mg (n = 1), 1200 mg (n = 1), and 1600 mg (n = 2) groups. The MTD was defined as 1200 mg. In part B, 32 patients received barasertib 1200 mg. In each part of the study, 8 of 32 patients had a hematologic response according to Cheson AML criteria. The most commonly reported grade ≥ 3 events were febrile neutropenia (n = 24) and stomatitis/mucosal inflammation (n = 16). We concluded that the MTD of barasertib is 1200 mg in patients with relapsed or newly diagnosed AML. Toxicity was manageable and barasertib treatment resulted in an overall hematologic response rate of 25%. This study is registered at www.ClinicalTrials.gov as NCT00497991.

Collaboration


Dive into the Norbert Ifrah's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Didier Blaise

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Bruno Lioure

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thibaut Leguay

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Vahid Asnafi

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne Huynh

University of Toulouse

View shared research outputs
Researchain Logo
Decentralizing Knowledge