Nicolas Ketterer
University of Lausanne
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Nicolas Ketterer.
Journal of Clinical Oncology | 2010
Christian Gisselbrecht; Bertram Glass; Nicolas Mounier; Devinder Gill; David C. Linch; Marek Trneny; André Bosly; Nicolas Ketterer; Ofer Shpilberg; Hans Hagberg; David Ma; Josette Briere; Craig H. Moskowitz; Norbert Schmitz
PURPOSE Salvage chemotherapy followed by high-dose therapy and autologous stem-cell transplantation (ASCT) is the standard treatment for relapsed diffuse large B-cell lymphoma (DLBCL). Salvage regimens have never been compared; their efficacy in the rituximab era is unknown. PATIENTS AND METHODS Patients with CD20(+) DLBCL in first relapse or who were refractory after first-line therapy were randomly assigned to either rituximab, ifosfamide, etoposide, and carboplatin (R-ICE) or rituximab, dexamethasone, high-dose cytarabine, and cisplatin (R-DHAP). Responding patients received high-dose chemotherapy and ASCT. RESULTS The median age of the 396 patients enrolled (R-ICE, n = 202; R-DHAP, n = 194) was 55 years. Similar response rates were observed after three cycles of R-ICE (63.5%; 95% CI, 56% to 70%) and R-DHAP (62.8%; 95 CI, 55% to 69%). Factors affecting response rates (P < .001) were refractory disease/relapse less than versus more than 12 months after diagnosis (46% v 88%, respectively), International Prognostic Index (IPI) of more than 1 versus 0 to 1 (52% v 71%, respectively), and prior rituximab treatment versus no prior rituximab (51% v 83%, respectively). There was no significant difference between R-ICE and R-DHAP for 3-year event-free survival (EFS) or overall survival. Three-year EFS was affected by prior rituximab treatment versus no rituximab (21% v 47%, respectively), relapse less than versus more than 12 months after diagnosis (20% v 45%, respectively), and IPI of 2 to 3 versus 0 to 1 (18% v 40%, respectively). In the Cox model, these parameters were significant (P < .001). CONCLUSION In patients who experience relapse more than 12 months after diagnosis, prior rituximab treatment does not affect EFS. Patients with early relapses after rituximab-containing first-line therapy have a poor prognosis, with no difference between the effects of R-ICE and R-DHAP.
Journal of Clinical Oncology | 2010
Anjali S. Advani; Bertrand Coiffier; Myron S. Czuczman; Martin Dreyling; James M. Foran; Eva Giné; Christian Gisselbrecht; Nicolas Ketterer; Sunita D. Nasta; A. Z. S. Rohatiner; Ingo G.H. Schmidt-Wolf; Martin Schuler; Jorge Sierra; Mitchell R. Smith; Gregor Verhoef; Jane N. Winter; Joseph Boni; Erik Vandendries; Mark Shapiro; Luis Fayad
PURPOSE Inotuzumab ozogamicin (CMC-544) is an antibody-targeted chemotherapy agent composed of a humanized anti-CD22 antibody conjugated to calicheamicin, a potent cytotoxic agent. This was a phase I study to determine the maximum-tolerated dose (MTD), safety, and preliminary efficacy of inotuzumab ozogamicin in an expanded MTD cohort of patients with relapsed or refractory CD22(+) B-cell non-Hodgkins lymphoma (NHL). PATIENTS AND METHODS Inotuzumab ozogamicin was administered intravenously as a single agent once every 3 or 4 weeks at doses ranging from 0.4 to 2.4 mg/m(2). Outcomes included MTD, safety, pharmacokinetics, response, progression-free survival (PFS), and overall survival. Results Seventy-nine patients were enrolled. The MTD was determined to be 1.8 mg/m(2). Common adverse events at the MTD were thrombocytopenia (90%), asthenia (67%), and nausea and neutropenia (51% each). The objective response rate at the end of treatment was 39% for the 79 enrolled patients, 68% for all patients with follicular NHL treated at the MTD, and 15% for all patients with diffuse large B-cell lymphoma treated at the MTD. Median PFS was 317 days (approximately 10.4 months) and 49 days for patients with follicular NHL and diffuse large B-cell lymphoma, respectively. CONCLUSION Inotuzumab ozogamicin has demonstrated efficacy against CD22(+) B-cell NHL, with reversible thrombocytopenia as the main toxicity.
Journal of Clinical Oncology | 2005
Michele Ghielmini; Shu-Fang Hsu Schmitz; Sergio Cogliatti; Francesco Bertoni; Ursula Waltzer; Martin F. Fey; Daniel C. Betticher; Hubert Schefer; Gabriella Pichert; Rolf A. Stahel; Nicolas Ketterer; Mario Bargetzi; Thomas Cerny
PURPOSE To evaluate the effect of single-agent rituximab given at the standard or a prolonged schedule in patients with newly diagnosed, or refractory or relapsed mantle cell lymphoma (MCL). PATIENTS AND METHODS After induction treatment with the standard schedule (375 mg/m2 weekly x 4), patients who were responding or who had stable disease at week 12 from the start of treatment were randomly assigned to no further treatment (arm A) or prolonged rituximab administration (375 mg/m2) every 8 weeks for four times (arm B). RESULTS The trial enrolled 104 patients. After induction, clinical response was 27% with 2% complete responses. Among patients with detectable t(11;14)-positive cells in blood and bone marrow at baseline, four of 20, and one of 14, respectively, became polymerase chain-reaction-negative after induction. Anemia was the only adverse predictor of response in the multivariate analysis. After a median follow-up of 29 months, response rate and duration of response were not significantly different between the two schedules in 61 randomly assigned patients. Median event-free survival (EFS) was 6 months in arm A versus 12 months in arm B; the difference was not significant (P = .1). Prolonged treatment seemed to improve EFS in the subgroup of pretreated patients (5 months in arm A v 11 months in arm B; P = .04). Thirteen percent of patients in arm A and 9% in arm B presented with grade 3 to 4 hematologic toxicity. CONCLUSION Single-agent rituximab is active in MCL, but the addition of four single doses at 8-week intervals does not seem to significantly improve response rate, duration of response, or EFS after treatment with the standard schedule.
British Journal of Haematology | 1998
Nicolas Ketterer; Gilles Salles; Isabelle Moullet; Charles Dumontet; Assia Eljaafari-Corbin; Pierre Tremisi; Catherine Thieblemont; Brigitte Durand; Eve-Marie Neidhardt-Berard; Hanadi Samaha; Dominique Rigal; Bertrand Coiffier
Peripheral blood progenitor cells (PBPC) were mobilized and harvested in 200 patients treated for non‐Hodgkins lymphoma (n = 148), Hodgkins disease (n = 22) and multiple myeloma (n = 30). The variables predicting the collection of a minimal (>2.5 × 106/kg) or a high (>10 × 106/kg) CD34+ cell count were analysed. Patients were mobilized with haemopoietic growth factors following either standard chemotherapy (n = 49) or high‐dose cyclophosphamide, given alone (n = 55) or combined with high‐dose VP16 (n = 86). 10 patients received haemopoietic growth factors only. The first mobilization resulted in a PBPC harvest with enough CD34+ cells in 179/200 patients (90%). High‐dose cyclophosphamide, with or without VP16, did not mobilize a higher progenitor cell yield than standard chemotherapy. When performing multiple regression analysis in the 190 patients who received chemotherapy‐containing mobilization, only the number of previous chemotherapy regimens and the exposure to fludarabine predicted for a failure to collect a minimal PBPC count (P = 0.06 and 0.0008 respectively). The target to collect a high CD34+ cell count was negatively associated with the number of previous chemotherapy regimens (P = 0.002). When only non‐Hodgkins lymphoma patients were considered for multivariate analysis, low‐grade histology with fludarabine appeared to be associated with poor PBPC cell yield (P = 0.08 and 0.005 respectively). This data confirms that PBPC harvest should be planned early in the disease course in transplant candidates, and can be obtained after a standard course of chemotherapy.
Journal of Clinical Oncology | 2012
Laurent Garderet; Simona Iacobelli; Philippe Moreau; Mamoun Dib; Ingrid Lafon; Dietger Niederwieser; Tamas Masszi; Jean Fontan; Mauricette Michallet; Alois Gratwohl; Giuseppe Milone; Chantal Doyen; Brigitte Pégourié; Roman Hájek; Philippe Casassus; Brigitte Kolb; Carine Chaleteix; Bernd Hertenstein; Francesco Onida; Heinz Ludwig; Nicolas Ketterer; Christian Koenecke; Marleen van Os; Mohamad Mohty; Andrew Cakana; Norbert Claude Gorin; Theo de Witte; Jean Luc Harousseau; Curly Morris; Gösta Gahrton
PURPOSE This prospective multicenter phase III study compared the efficacy and safety of a triple combination (bortezomib-thalidomide-dexamethasone [VTD]) versus a dual combination (thalidomide-dexamethasone [TD]) in patients with multiple myeloma (MM) progressing or relapsing after autologous stem-cell transplantation (ASCT). PATIENTS AND METHODS Overall, 269 patients were randomly assigned to receive bortezomib (1.3 mg/m(2) intravenous bolus) or no bortezomib for 1 year, in combination with thalidomide (200 mg per day orally) and dexamethasone (40 mg orally once a day on 4 days once every 3 weeks). Bortezomib was administered on days 1, 4, 8, and 11 with a 10-day rest period (day 12 to day 21) for eight cycles (6 months), and then on days 1, 8, 15, and 22 with a 20-day rest period (day 23 to day 42) for four cycles (6 months). RESULTS Median time to progression (primary end point) was significantly longer with VTD than TD (19.5 v13.8 months; hazard ratio, 0.59; 95% CI, 0.44 to 0.80; P = .001), the complete response plus near-complete response rate was higher (45% v 21%; P 0.001), and the median duration of response was longer (17.9 v 13.4 months; P.04) [corrected].The 24-month survival rate was in favor of VTD (71% v 65%; P = .093). Grade 3 peripheral neuropathy was more frequent with VTD (29% v 12%; P = .001) as were the rates of grades 3 and 4 infection and thrombocytopenia. CONCLUSION VTD was more effective than TD in the treatment of patients with MM with progressive or relapsing disease post-ASCT but was associated with a higher incidence of grade 3 neurotoxicity.
Journal of Clinical Oncology | 2012
Christian Gisselbrecht; Norbert Schmitz; Nicolas Mounier; Devinder Gill; David C. Linch; Marek Trneny; André Bosly; Noel Milpied; John Radford; Nicolas Ketterer; Ofer Shpilberg; Ulrich Dührsen; Hans Hagberg; David Ma; Andreas Viardot; Rm Lowenthal; Josette Briere; Gilles Salles; Craig H. Moskowitz; Bertram Glass
PURPOSE The standard treatment for relapsed diffuse large B-cell lymphoma (DLBCL) is salvage chemotherapy followed by high-dose therapy and autologous stem-cell transplantation (ASCT). The impact of maintenance rituximab after ASCT is not known. PATIENTS AND METHODS In total, 477 patients with CD20(+) DLBCL who were in their first relapse or refractory to initial therapy were randomly assigned to one of two salvage regimens. After three cycles of salvage chemotherapy, the responding patients received high-dose chemotherapy followed by ASCT. Then, 242 patients were randomly assigned to either rituximab every 2 months for 1 year or observation. RESULTS After ASCT, 122 patients received rituximab, and 120 patients were observed only. The median follow-up time was 44 months. The 4-year event-free survival (EFS) rates after ASCT were 52% and 53% for the rituximab and observation groups, respectively (P = .7). Treatment with rituximab was associated with a 15% attributable risk of serious adverse events after day 100, with more deaths (six deaths v three deaths in the observation arm). Several factors affected EFS after ASCT (P < .05), including relapsed disease within 12 months (EFS: 46% v 56% for relapsed disease after 12 months), secondary age-adjusted International Prognostic Index (saaIPI) more than 1 (EFS: 37% v 61% for saaIPI < 1), and prior treatment with rituximab (EFS: 47% v 59% for no prior rituximab). A significant difference in EFS between women (63%) and men (46%) was also observed in the rituximab group. In the Cox model for maintenance, the saaIPI was a significant prognostic factor (P < .001), as was male sex (P = .01). CONCLUSION In relapsed DLBCL, we observed no difference between the control group and the rituximab maintenance group and do not recommend rituximab after ASCT.
Journal of Clinical Oncology | 2009
Jean-Luc Harousseau; Hervé Avet-Loiseau; Michel Attal; Catherine Charbonnel; Frederic Garban; Cyrille Hulin; Mauricette Michallet; Thierry Facon; Laurent Garderet; Gerald Marit; Nicolas Ketterer; Thierry Lamy; Laurent Voillat; François Guilhot; Chantal Doyen; Claire Mathiot; Philippe Moreau
PURPOSE The prognostic impact of complete response (CR) achievement in multiple myeloma (MM) has been shown mostly in the context of autologous stem-cell transplantation. Other levels of response have been defined because, even with high-dose therapy, CR is a relatively rare event. The purpose of this study was to analyze the prognostic impact of very good partial response (VGPR) in patients treated with high-dose therapy. PATIENTS AND METHODS All patients were included in the Intergroupe Francophone du Myelome 99-02 and 99-04 trials and treated with vincristine, doxorubicin, and dexamethasone (VAD) induction therapy followed by double autologous stem-cell transplantation (ASCT). Best post-ASCT response assessment was available for 802 patients. RESULTS With a median follow-up of 67 months, median event-free survival (EFS) and 5-year EFS were 42 months and 34%, respectively, for 405 patients who achieved at least VGPR after ASCT versus 32 months and 26% in 288 patients who achieved only partial remission (P = .005). Five-year overall survival (OS) was significantly superior in patients achieving at least VGPR (74% v 61% P = .0017). In multivariate analysis, achievement of less than VGPR was an independent factor predicting shorter EFS and OS. Response to VAD had no impact on EFS and OS. The impact of VGPR achievement on EFS and OS was significant in patients with International Staging System stages 2 to 3 and for patients with poor-risk cytogenetics t(4;14) or del(17p). CONCLUSION In the context of ASCT, achievement of at least VGPR is a simple prognostic factor that has importance in intermediate and high-risk MM and can be informative in more patients than CR.
Blood | 2011
Philippe Moreau; Michel Attal; Brigitte Pegourie; Lucie Planche; Cyrille Hulin; Thierry Facon; Anne-Marie Stoppa; Jean-Gabriel Fuzibet; Bernard Grosbois; Chantal Doyen; Nicolas Ketterer; Catherine Sebban; Brigitte Kolb; Carine Chaleteix; Mamoun Dib; Laurent Voillat; Jean Fontan; Laurent Garderet; Jerome Jaubert; Claire Mathiot; Dixie Lee Esseltine; Hervé Avet-Loiseau; Jean-Luc Harousseau
In the 2005-01 trial, we have demonstrated that bortezomib-dexamethasone as induction therapy before autologous stem cell transplantation was superior to vincristine-adriamycin-dexamethasone. We conducted a post-hoc analysis to assess the prognostic impact of initial characteristics as well as response to therapy in patients enrolled in this study. Multivariate analysis showed that ISS stages 2 and 3 and achievement of response less than very good partial response (VGPR) both after induction therapy and after autologous stem cell transplantation were adverse prognostic factors for progression-free survival, the most important one being achievement of response less than VGPR after induction. Progression-free survival was significantly improved with bortezomib-dexamethasone induction therapy in patients with poor-risk cytogenetics and ISS stages 2 and 3 compared with vincristine-adriamycin-dexamethasone. In these 2 groups of patients, achievement of at least VGPR after induction was of major importance. This study is registered with EudraCT (https://eudract.ema.europa.eu; EUDRACT 2005-000537-38) and http://clinicaltrials.gov (NCT00200681).
Oncologist | 2010
Heinz Ludwig; Meral Beksac; Joan Bladé; Mario Boccadoro; Jamie Cavenagh; Michele Cavo; Meletios A. Dimopoulos; Johannes Drach; Hermann Einsele; Thierry Facon; Hartmut Goldschmidt; Jean-Luc Harousseau; Urs Hess; Nicolas Ketterer; Martin Kropff; Larisa Mendeleeva; Gareth J. Morgan; Antonio Palumbo; Torben Plesner; Jesús F. San Miguel; Ofer Shpilberg; Pia Sondergeld; Pieter Sonneveld; Sonja Zweegman
This review presents an overview of the most recent data using the novel agents thalidomide, bortezomib, and lenalidomide in the treatment of multiple myeloma and summarizes European treatment practices incorporating these novel agents.
Annals of Oncology | 1998
I. Moullet; Gilles Salles; Nicolas Ketterer; C. Dumontet; F. Bouafia; E.-M. Neidhart-Berard; Catherine Thieblemont; Pascale Felman; Bertrand Coiffier
OBJECTIVES Retrospective evaluation of anemia frequency and its prognostic value in patients with different subtypes of non-Hodgkins lymphoma and comparison with other clinical characteristics. PATIENTS AND METHODS Anemia was defined as a hemoglobin value less than or equal to 12 g/dl for all men and women over 50 years of age, and less than or equal to 11 g/dl for women under 50 years of age. The study included 1077 adult lymphoma patients treated between 1980 and 1995 with the following histologic subtypes: 127 patients with small lymphocytic or lymphoplasmacytoid, 62 with marginal zone, 50 with mantle-cell, 208 with follicular, 104 with T-cell lymphoma, 426 with diffuse large-cell and, finally, 73 patients with other high-grade lymphomas. RESULTS Anemia was present in 341 patients (32%). It was an adverse prognostic factor (P < 0.0001) for overall survival (OS) and progression-free survival (PFS) but not for relapse-free survival (RFS). When patients with and those without bone marrow involvement were considered separately, anemia remained an adverse factor. Anemia was significantly associated with shorter PFS in small lymphocytic or lymphoplasmacytoid, mantle cell, diffuse large cell and high-grade lymphomas and with shorter OS in all histologic subgroups except marginal zone lymphoma. In multivariate analysis, anemia was a significant prognostic factor for OS and PFS for the population as a whole (P = 0.0001 and P = 0.0048, respectively) and in patients with bone marrow involvement (P = 0.007 and P = 0.005, respectively) but not in patients without bone marrow involvement. Finally, the addition of anemia to the International Prognostic Index led to an improvement for OS (P = 0.0004) and PFS (P = 0.0004). CONCLUSIONS Anemia is an important adverse prognostic factor for the outcome of lymphoma patients, particularly in some histologic subgroups and in patients with bone marrow involvement.