Network


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

Hotspot


Dive into the research topics where Katja Weisel is active.

Publication


Featured researches published by Katja Weisel.


Journal of Clinical Oncology | 2012

Bortezomib Induction and Maintenance Treatment in Patients With Newly Diagnosed Multiple Myeloma: Results of the Randomized Phase III HOVON-65/ GMMG-HD4 Trial

Pieter Sonneveld; Ingo G.H. Schmidt-Wolf; Bronno van der Holt; Laila el Jarari; Uta Bertsch; Hans Salwender; Sonja Zweegman; Edo Vellenga; Annemiek Broyl; Igor Wolfgang Blau; Katja Weisel; Shulamiet Wittebol; Gerard M. J. Bos; Marian Stevens-Kroef; Christof Scheid; Michael Pfreundschuh; Dirk Hose; Anna Jauch; Helgi van der Velde; Reinier Raymakers; Martijn R. Schaafsma; M. J. Kersten; Marinus van Marwijk-Kooy; Ulrich Duehrsen; Walter Lindemann; Pierre W. Wijermans; Henk M. Lokhorst; Hartmut Goldschmidt

PURPOSE We investigated whether bortezomib during induction and maintenance improves survival in newly diagnosed multiple myeloma (MM). PATIENTS AND METHODS In all, 827 eligible patients with newly diagnosed symptomatic MM were randomly assigned to receive induction therapy with vincristine, doxorubicin, and dexamethasone (VAD) or bortezomib, doxorubicin, and dexamethasone (PAD) followed by high-dose melphalan and autologous stem-cell transplantation. Maintenance consisted of thalidomide 50 mg (VAD) once per day or bortezomib 1.3 mg/m(2) (PAD) once every 2 weeks for 2 years. The primary analysis was progression-free survival (PFS) adjusted for International Staging System (ISS) stage. RESULTS Complete response (CR), including near CR, was superior after PAD induction (15% v 31%; P < .001) and bortezomib maintenance (34% v 49%; P < .001). After a median follow-up of 41 months, PFS was superior in the PAD arm (median of 28 months v 35 months; hazard ratio [HR], 0.75; 95% CI, 0.62 to 0.90; P = .002). In multivariate analysis, overall survival (OS) was better in the PAD arm (HR, 0.77; 95% CI, 0.60 to 1.00; P = .049). In high-risk patients presenting with increased creatinine more than 2 mg/dL, bortezomib significantly improved PFS from a median of 13 months to 30 months (HR, 0.45; 95% CI, 0.26 to 0.78; P = .004) and OS from a median of 21 months to 54 months (HR, 0.33; 95% CI, 0.16 to 0.65; P < .001). A benefit was also observed in patients with deletion 17p13 (median PFS, 12 v 22 months; HR, 0.47; 95% CI, 0.26 to 0.86; P = .01; median OS, 24 months v not reached at 54 months; HR, 0.36; 95% CI, 0.18 to 0.74; P = .003). CONCLUSION Bortezomib during induction and maintenance improves CR and achieves superior PFS and OS.


The New England Journal of Medicine | 2012

Continuous Lenalidomide Treatment for Newly Diagnosed Multiple Myeloma

Antonio Palumbo; Roman Hájek; Michel Delforge; Martin Kropff; Maria Teresa Petrucci; John Catalano; Heinz Gisslinger; Wieslaw Wiktor-Jedrzejczak; Mamia Zodelava; Katja Weisel; Nicola Cascavilla; Genadi Iosava; Michele Cavo; Janusz Kloczko; Joan Bladé; Meral Beksac; Ivan Spicka; Torben Plesner; Joergen Radke; Christian Langer; Dina Ben Yehuda; Alessandro Corso; Lindsay Herbein; Zhinuan Yu; Jay Mei; Christian Jacques; Meletios A. Dimopoulos

BACKGROUND Lenalidomide has tumoricidal and immunomodulatory activity against multiple myeloma. This double-blind, multicenter, randomized study compared melphalan-prednisone-lenalidomide induction followed by lenalidomide maintenance (MPR-R) with melphalan-prednisone-lenalidomide (MPR) or melphalan-prednisone (MP) followed by placebo in patients 65 years of age or older with newly diagnosed multiple myeloma. METHODS We randomly assigned patients who were ineligible for transplantation to receive MPR-R (nine 4-week cycles of MPR followed by lenalidomide maintenance therapy until a relapse or disease progression occurred [152 patients]) or to receive MPR (153 patients) or MP (154 patients) without maintenance therapy. The primary end point was progression-free survival. RESULTS The median follow-up period was 30 months. The median progression-free survival was significantly longer with MPR-R (31 months) than with MPR (14 months; hazard ratio, 0.49; P<0.001) or MP (13 months; hazard ratio, 0.40; P<0.001). Response rates were superior with MPR-R and MPR (77% and 68%, respectively, vs. 50% with MP; P<0.001 and P=0.002, respectively, for the comparison with MP). The progression-free survival benefit associated with MPR-R was noted in patients 65 to 75 years of age but not in those older than 75 years of age (P=0.001 for treatment-by-age interaction). After induction therapy, a landmark analysis showed a 66% reduction in the rate of progression with MPR-R (hazard ratio for the comparison with MPR, 0.34; P<0.001) that was age-independent. During induction therapy, the most frequent adverse events were hematologic; grade 4 neutropenia was reported in 35%, 32%, and 8% of the patients in the MPR-R, MPR, and MP groups, respectively. The 3-year rate of second primary tumors was 7% with MPR-R, 7% with MPR, and 3% with MP. CONCLUSIONS MPR-R significantly prolonged progression-free survival in patients with newly diagnosed multiple myeloma who were ineligible for transplantation, with the greatest benefit observed in patients 65 to 75 years of age. (Funded by Celgene; MM-015 ClinicalTrials.gov number, NCT00405756.).


Lancet Oncology | 2013

Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial

Jesús F. San Miguel; Katja Weisel; Philippe Moreau; Martha Q. Lacy; Kijoung Song; Michel Delforge; Lionel Karlin; Hartmut Goldschmidt; Anne Banos; Albert Oriol; Adrian Alegre; Christopher Chen; Michele Cavo; Laurent Garderet; Valentina Ivanova; Joaquin Martinez-Lopez; Andrew R. Belch; Antonio Palumbo; Steve Schey; Pieter Sonneveld; Xiaoyan Yu; Lars Sternas; Christian Jacques; Mohamed H. Zaki; Meletios A. Dimopoulos

BACKGROUND Few effective treatments exist for patients with refractory or relapsed and refractory multiple myeloma not responding to treatment with bortezomib and lenalidomide. Pomalidomide alone has shown limited efficacy in patients with relapsed multiple myeloma, but synergistic effects have been noted when combined with dexamethasone. We compared the efficacy and safety of pomalidomide plus low-dose dexamethasone with high-dose dexamethasone alone in these patients. METHODS This multicentre, open-label, randomised phase 3 trial was undertaken in Australia, Canada, Europe, Russia, and the USA. Patients were eligible if they had been diagnosed with refractory or relapsed and refractory multiple myeloma, and had failed at least two previous treatments of bortezomib and lenalidomide. They were assigned in a 2:1 ratio with a validated interactive voice and internet response system to either 28 day cycles of pomalidomide (4 mg/day on days 1-21, orally) plus low-dose dexamethasone (40 mg/day on days 1, 8, 15, and 22, orally) or high-dose dexamethasone (40 mg/day on days 1-4, 9-12, and 17-20, orally) until disease progression or unacceptable toxicity. Stratification factors were age (≤75 years vs >75 years), disease population (refractory vs relapsed and refractory vs bortezomib intolerant), and number of previous treatments (two vs more than two). The primary endpoint was progression-free survival (PFS). Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT01311687, and with EudraCT, number 2010-019820-30. FINDINGS The accrual for the study has been completed and the analyses are presented. 302 patients were randomly assigned to receive pomalidomide plus low-dose dexamethasone and 153 high-dose dexamethasone. After a median follow-up of 10·0 months (IQR 7·2-13·2), median PFS with pomalidomide plus low-dose dexamethasone was 4·0 months (95% CI 3·6-4·7) versus 1·9 months (1·9-2·2) with high-dose dexamethasone (hazard ratio 0·48 [95% CI 0·39-0·60]; p<0·0001). The most common grade 3-4 haematological adverse events in the pomalidomide plus low-dose dexamethasone and high-dose dexamethasone groups were neutropenia (143 [48%] of 300 vs 24 [16%] of 150, respectively), anaemia (99 [33%] vs 55 [37%], respectively), and thrombocytopenia (67 [22%] vs 39 [26%], respectively). Grade 3-4 non-haematological adverse events in the pomalidomide plus low-dose dexamethasone and high-dose dexamethasone groups included pneumonia (38 [13%] vs 12 [8%], respectively), bone pain (21 [7%] vs seven [5%], respectively), and fatigue (16 [5%] vs nine [6%], respectively). There were 11 (4%) treatment-related adverse events leading to death in the pomalidomide plus low-dose dexamethasone group and seven (5%) in the high-dose dexamethasone group. INTERPRETATION Pomalidomide plus low-dose dexamethasone, an oral regimen, could be considered a new treatment option in patients with refractory or relapsed and refractory multiple myeloma. FUNDING Celgene Corporation.


The New England Journal of Medicine | 2014

Lenalidomide and Dexamethasone in Transplant-Ineligible Patients with Myeloma

Lotfi Benboubker; Meletios A. Dimopoulos; Angela Dispenzieri; John Catalano; Andrew R. Belch; Michele Cavo; Antonello Pinto; Katja Weisel; Heinz Ludwig; Nizar J. Bahlis; Anne Banos; Mourad Tiab; Michel Delforge; Jamie Cavenagh; Catarina Geraldes; Je Jung Lee; Christine Chen; Albert Oriol; Javier de la Rubia; Lugui Qiu; Darrell White; Daniel Binder; Kenneth C. Anderson; Jean Paul Fermand; Philippe Moreau; Michel Attal; Robert Knight; Guang Chen; Jason Van Oostendorp; Christian Jacques

BACKGROUND The combination melphalan-prednisone-thalidomide (MPT) is considered a standard therapy for patients with myeloma who are ineligible for stem-cell transplantation. However, emerging data on the use of lenalidomide and low-dose dexamethasone warrant a prospective comparison of the two approaches. METHODS We randomly assigned 1623 patients to lenalidomide and dexamethasone in 28-day cycles until disease progression (535 patients), to the same combination for 72 weeks (18 cycles; 541 patients), or to MPT for 72 weeks (547 patients). The primary end point was progression-free survival with continuous lenalidomide-dexamethasone versus MPT. RESULTS The median progression-free survival was 25.5 months with continuous lenalidomide-dexamethasone, 20.7 months with 18 cycles of lenalidomide-dexamethasone, and 21.2 months with MPT (hazard ratio for the risk of progression or death, 0.72 for continuous lenalidomide-dexamethasone vs. MPT and 0.70 for continuous lenalidomide-dexamethasone vs. 18 cycles of lenalidomide-dexamethasone; P<0.001 for both comparisons). Continuous lenalidomide-dexamethasone was superior to MPT for all secondary efficacy end points, including overall survival (at the interim analysis). Overall survival at 4 years was 59% with continuous lenalidomide-dexamethasone, 56% with 18 cycles of lenalidomide-dexamethasone, and 51% with MPT. Grade 3 or 4 adverse events were somewhat less frequent with continuous lenalidomide-dexamethasone than with MPT (70% vs. 78%). As compared with MPT, continuous lenalidomide-dexamethasone was associated with fewer hematologic and neurologic toxic events, a moderate increase in infections, and fewer second primary hematologic cancers. CONCLUSIONS As compared with MPT, continuous lenalidomide-dexamethasone given until disease progression was associated with a significant improvement in progression-free survival, with an overall survival benefit at the interim analysis, among patients with newly diagnosed multiple myeloma who were ineligible for stem-cell transplantation. (Funded by Intergroupe, Francophone du Myélome and Celgene; FIRST ClinicalTrials.gov number, NCT00689936; European Union Drug Regulating Authorities Clinical Trials number, 2007-004823-39.).


Blood | 2012

Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p

Kai Neben; Henk M. Lokhorst; Anna Jauch; Uta Bertsch; Thomas Hielscher; Bronno van der Holt; Hans Salwender; Igor Wolfgang Blau; Katja Weisel; Michael Pfreundschuh; Christof Scheid; Ulrich Dührsen; Walter Lindemann; Ingo G.H. Schmidt-Wolf; Norma Peter; Christian Teschendorf; Hans Martin; Mathias Haenel; Hans Günter Derigs; Marc S. Raab; Anthony D. Ho; Helgi van de Velde; Dirk Hose; Pieter Sonneveld; Hartmut Goldschmidt

In patients with multiple myeloma (MM), risk stratification by chromosomal abnormalities may enable a more rational selection of therapeutic approaches. In the present study, we analyzed the prognostic value of 12 chromosomal abnormalities in a series of 354 MM patients treated within the HOVON-65/GMMG-HD4 trial. Because of the 2-arm design of the study, we were able to analyze the effect of a bortezomib-based treatment before and after autologous stem cell transplantation (arm B) compared with standard treatment without bortezomib (arm A). For allanalyzed chromosomal aberrations, progression-free survival (PFS) and overall survival (OS) were at least equal or superior in the bortezomib arm compared with the standard arm. Strikingly, patients with del(17p13) benefited the most from the bortezomib-containing treatment: the median PFS in arm A was 12.0 months and in arm B it was 26.2 months (P = .024); the 3 year-OS for arm A was 17% and for arm B it was 69% (P = .028). After multivariate analysis, del(17p13) was an independent predictor for PFS (P < .0001) and OS (P < .0001) in arm A, whereas no statistically significant effect on PFS (P = .28) or OS (P = .12) was seen in arm B. In conclusion, the adverse impact of del(17p13) on PFS and OS could be significantly reduced by bortezomib-based treatment, suggesting that long-term administration of bortezomib should be recommended for patients carrying del(17p13).


Lancet Oncology | 2016

Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study

Meletios A. Dimopoulos; Philippe Moreau; Antonio Palumbo; Douglas E. Joshua; Ludek Pour; Roman Hájek; Thierry Facon; Heinz Ludwig; Albert Oriol; Hartmut Goldschmidt; Laura Rosiñol; Jan Straub; Aleksandr Suvorov; Carla Araujo; Elena Rimashevskaya; Tomas Pika; Gianluca Gaidano; Katja Weisel; Vesselina Goranova-Marinova; Anthony P. Schwarer; Leonard Minuk; Tamas Masszi; Ievgenii Karamanesht; Massimo Offidani; Vania Tietsche de Moraes Hungria; Andrew Spencer; Robert Z. Orlowski; Heidi H. Gillenwater; Nehal Mohamed; Shibao Feng

BACKGROUND Bortezomib with dexamethasone is a standard treatment option for relapsed or refractory multiple myeloma. Carfilzomib with dexamethasone has shown promising activity in patients in this disease setting. The aim of this study was to compare the combination of carfilzomib and dexamethasone with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma. METHODS In this randomised, phase 3, open-label, multicentre study, patients with relapsed or refractory multiple myeloma who had one to three previous treatments were randomly assigned (1:1) using a blocked randomisation scheme (block size of four) to receive carfilzomib with dexamethasone (carfilzomib group) or bortezomib with dexamethasone (bortezomib group). Randomisation was stratified by previous proteasome inhibitor therapy, previous lines of treatment, International Staging System stage, and planned route of bortezomib administration if randomly assigned to bortezomib with dexamethasone. Patients received treatment until progression with carfilzomib (20 mg/m(2) on days 1 and 2 of cycle 1; 56 mg/m(2) thereafter; 30 min intravenous infusion) and dexamethasone (20 mg oral or intravenous infusion) or bortezomib (1·3 mg/m(2); intravenous bolus or subcutaneous injection) and dexamethasone (20 mg oral or intravenous infusion). The primary endpoint was progression-free survival in the intention-to-treat population. All participants who received at least one dose of study drug were included in the safety analyses. The study is ongoing but not enrolling participants; results for the interim analysis of the primary endpoint are presented. The trial is registered at ClinicalTrials.gov, number NCT01568866. FINDINGS Between June 20, 2012, and June 30, 2014, 929 patients were randomly assigned (464 to the carfilzomib group; 465 to the bortezomib group). Median follow-up was 11·9 months (IQR 9·3-16·1) in the carfilzomib group and 11·1 months (8·2-14·3) in the bortezomib group. Median progression-free survival was 18·7 months (95% CI 15·6-not estimable) in the carfilzomib group versus 9·4 months (8·4-10·4) in the bortezomib group at a preplanned interim analysis (hazard ratio [HR] 0·53 [95% CI 0·44-0·65]; p<0·0001). On-study death due to adverse events occurred in 18 (4%) of 464 patients in the carfilzomib group and in 16 (3%) of 465 patients in the bortezomib group. Serious adverse events were reported in 224 (48%) of 463 patients in the carfilzomib group and in 162 (36%) of 456 patients in the bortezomib group. The most frequent grade 3 or higher adverse events were anaemia (67 [14%] of 463 patients in the carfilzomib group vs 45 [10%] of 456 patients in the bortezomib group), hypertension (41 [9%] vs 12 [3%]), thrombocytopenia (39 [8%] vs 43 [9%]), and pneumonia (32 [7%] vs 36 [8%]). INTERPRETATION For patients with relapsed or refractory multiple myeloma, carfilzomib with dexamethasone could be considered in cases in which bortezomib with dexamethasone is a potential treatment option. FUNDING Onyx Pharmaceuticals, Inc., an Amgen subsidiary.


American Journal of Roentgenology | 2011

Whole-body diffusion-weighted MRI with apparent diffusion coefficient mapping for early response monitoring in multiple myeloma: preliminary results.

Marius Horger; Katja Weisel; Wilhelm Horger; Ali Mroue; Michael Fenchel; Matthias P. Lichy

OBJECTIVE The purpose of our study was to prospectively assess the feasibility of whole-body diffusion-weighted imaging (DWI) for short-term evaluation of response to treatment in multiple myeloma patients using a single-shot echo-planar imaging DWI sequence with a Stejskal-Tanner diffusion encoding scheme and spectral fat suppression. SUBJECTS AND METHODS Twelve consecutive patients (nine men and three women; mean age, 61.4 years; age range, 54-79 years) underwent whole-body DWI (b = 50, 400, and 800 s/mm(2)) both at baseline and 3 weeks (mean, 23 days) after onset of therapy. Bone marrow and extramedullary manifestations were evaluated by quantitative image analysis using measurements of the mean apparent diffusion coefficient (ADC). These parameters were correlated with myeloma response according to standard criteria and were evaluated parallel to MRI and continuously for more than 6 months afterward. RESULTS Fifty-three myeloma lesions, 50 medullary (28 axial and 22 appendicular skeleton) and three extramedullary, were analyzed. Eleven patients were classified as responders and one as a nonresponder. DWI results accurately (100%) correlated with disease course according to standard clinical and laboratory criteria. All involved lesions showed restricted diffusion at baseline. ADC quantification yielded an increase of 63.9% (range, 8.7-211.3%) in responders and a decrease of 7.8% in the sole nonresponding patient during therapy. In parallel, M-gradient measurement showed a mean decrease of 45.1% (range, 19.6-88.8%) in responders and an increase of 21.8% in the nonresponder. Amplitude of response measured by the course of ADC values proved higher in the appendicular skeleton (99.8%) compared with the axial skeleton (54.3%) (p = 0.037). CONCLUSION Whole-body DWI with ADC analysis represents a feasible diagnostic tool for assessment of short-term treatment response in myeloma patients.


Haematologica | 2014

Bortezomib before and after autologous stem cell transplantation overcomes the negative prognostic impact of renal impairment in newly diagnosed multiple myeloma: a subgroup analysis from the HOVON-65/GMMG-HD4 trial

Christof Scheid; Pieter Sonneveld; Ingo G.H. Schmidt-Wolf; Bronno van der Holt; Laila el Jarari; Uta Bertsch; Hans Salwender; Sonja Zweegman; Igor Wolfgang Blau; Edo Vellenga; Katja Weisel; Michael Pfreundschuh; Kon-Siong G. Jie; Kai Neben; Helgi van de Velde; Ulrich Duehrsen; M. Ron Schaafsma; Walter Lindemann; Marie José Kersten; Norma Peter; Mathias Hänel; Sandra Croockewit; Hans Martin; Shulamiet Wittebol; Gerard M. J. Bos; Marinus van Marwijk-Kooy; Pierre W. Wijermans; Hartmut Goldschmidt; Henk M. Lokhorst

Renal impairment is frequent in patients with multiple myeloma and is correlated with an inferior prognosis. This analysis evaluates the prognostic role of renal impairment in patients with myeloma treated with bortezomib before and after autologous stem cell transplantation within a prospective randomized phase III trial. Eight hundred and twenty-seven newly diagnosed myeloma patients in the HOVON-65/GMMG-HD4 trial were randomized to receive three cycles of vincristine, adriamycin, dexamethasone (VAD) or bortezomib, adriamycin, dexamethasone (PAD) followed by autologous stem cell transplantation and maintenance with thalidomide 50 mg daily (VAD-arm) or bortezomib 1.3 mg/m2 every 2 weeks (PAD-arm). Baseline serum creatinine was less than 2 mg/dL (Durie-Salmon-stage A) in 746 patients and 2 mg/dL or higher (stage B) in 81. In myeloma patients with a baseline creatinine ≥2 mg/dL the renal response rate was 63% in the VAD-arm and 81% in the PAD-arm (P=0.31). The overall myeloma response rate was 64% in the VAD-arm versus 89% in the PAD-arm with 13% complete responses in the VAD-arm versus 36% in the PAD-arm (P=0.01). Overall survival at 3 years for patients with a baseline creatinine ≥2 mg/dL was 34% in the VAD-arm versus 74% in the PAD-arm (P<0.001) with a progression-free survival rate at 3 years of 16% in the VAD-arm versus 48% in the PAD-arm (P=0.004). Overall and progression-free survival rates in the PAD-arm were similar in patients with a baseline creatinine ≥2 mg/dL or <2 mg/dL. We conclude that a bortezomib-containing treatment before and after autologous stem cell transplantation overcomes the negative prognostic impact of renal impairment in patients with newly diagnosed multiple myeloma. The trial was registered at www.trialregister.nl as NTR213 and at www.controlled-trials.com as ISRCTN 64455289


Leukemia | 2014

Expert panel consensus statement on the optimal use of pomalidomide in relapsed and refractory multiple myeloma

Meletios A. Dimopoulos; Xavier Leleu; Antonio Palumbo; P. Moreau; Michel Delforge; Michele Cavo; Heinz Ludwig; Gareth J. Morgan; Faith E. Davies; Pieter Sonneveld; Steve Schey; Sonja Zweegman; Markus Hansson; Katja Weisel; M.V. Mateos; Thierry Facon; Jesús F. San Miguel

In this report, a panel of European myeloma experts discuss the role of pomalidomide in the treatment of relapsed and refractory multiple myeloma (RRMM). Based on the available evidence, the combination of pomalidomide and low-dose dexamethasone is a well-tolerated and effective treatment option for patients with RRMM who have exhausted treatment with lenalidomide and bortezomib. The optimal starting dose of pomalidomide is 4 mg given on days 1–21 of each 28-day cycle, whereas dexamethasone is administered at a dose of 40 mg weekly (reduced to 20 mg for patients aged >75 years). The treatment should continue until evidence of disease progression or unacceptable toxicity. Dose-modification schemes have been established for patients who develop neutropenia, thrombocytopaenia and other grade 3–4 adverse events during pomalidomide therapy. Guidance on the prevention and management of infections and venous thromboembolism is provided, based on the available clinical evidence and the experience of panel members. The use of pomalidomide in special populations, such as patients with advanced age, renal impairment or unfavourable cytogenetic features, is also discussed.


Leukemia | 2015

Phase III trial of bortezomib, cyclophosphamide and dexamethasone (VCD) versus bortezomib, doxorubicin and dexamethasone (PAd) in newly diagnosed myeloma

Elias K. Mai; Uta Bertsch; Jan Dürig; Christina Kunz; Mathias Haenel; Igor Wolfgang Blau; Markus Munder; Anna Jauch; B. Schurich; Thomas Hielscher; Maximilian Merz; B. Huegle-Doerr; Anja Seckinger; Dirk Hose; Jens Hillengass; Marc-Steffen Raab; Kai Neben; Hans-Walter Lindemann; M. Zeis; Christian Gerecke; Ingo G.H. Schmidt-Wolf; Katja Weisel; C Scheid; Hans Salwender; H. Goldschmidt

We aimed at demonstrating non-inferiority of bortezomib/cyclophosphamide/dexamethasone (VCD) compared to bortezomib/doxorubicin/dexamethasone (PAd) induction therapy with respect to very good partial response rates or better (⩾VGPR) in 504 newly diagnosed, transplant-eligible multiple myeloma patients. VCD was found to be non-inferior to PAd with respect to ⩾VGPR rates (37.0 versus 34.3%, P=0.001). The rates of progressive disease (PD) were 0.4% (VCD) versus 4.8% (PAd; P=0.003). In the PAd arm, 11 of 12 patients with PD had either renal impairment (creatinine ⩾2 mg/dl) at diagnosis or the cytogenetic abnormality gain 1q21, whereas no PD was observed in these subgroups in the VCD arm. Leukocytopenia/neutropenia (⩾3°) occurred more frequently in the VCD arm (35.2% versus 11.3%, P<0.001). Neuropathy rates (⩾2°) were higher in the PAd group (14.9 versus 8.4%, P=0.03). Serious adverse events, both overall and those related to thromboembolic events, were higher in the PAd group (32.7 versus 24.0%, P=0.04 and 2.8 versus 0.4%, P=0.04). Stem cell collection was not impeded by VCD. VCD is as effective as PAd in terms of achieving ⩾VGPR rates with fewer PD and has a favorable toxicity profile. Therefore, VCD is preferable to PAd as induction therapy.

Collaboration


Dive into the Katja Weisel's collaboration.

Top Co-Authors

Avatar

Meletios A. Dimopoulos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Michel Delforge

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Albert Oriol

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge