33 results on '"Katja C, Weisel"'
Search Results
2. Implications and prognostic impact of mass spectrometry in patients with newly-diagnosed multiple myeloma
- Author
-
Elias K. Mai, Stefanie Huhn, Kaya Miah, Alexandra M. Poos, Christof Scheid, Katja C. Weisel, Uta Bertsch, Markus Munder, Oscar Berlanga, Dirk Hose, Anja Seckinger, Anna Jauch, Igor W. Blau, Mathias Hänel, Hans J. Salwender, Axel Benner, Marc S. Raab, Hartmut Goldschmidt, and Niels Weinhold
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Mass spectrometry (MS) is a promising tool for monitoring monoclonal protein in plasma cell dyscrasias. We included 480 transplant-eligible newly-diagnosed multiple myeloma (MM) patients from the GMMG-MM5 trial (EudraCT No. 2010-019173-16) and performed a retrospective MS analysis at baseline (480 patients) and at the pre-defined, consecutive time points after induction (444 patients), prior to maintenance (305 patients) and after one year of maintenance (227 patients). We found that MS negativity was significantly associated with improved progression-free survival (PFS) even in patients with complete response (CR) at all investigated follow-up time points. The prognostic impact was independent of established risk factors, such as the revised International Staging System. Combining MS and baseline cytogenetics improved the prediction of outcome: MS-positive patients with high-risk cytogenetics had a dismal PFS of 1.9 years (95% confidence interval [CI]: 1.6–2.3 years) from the start of maintenance. Testing the value of sequential MS prior to and after one year of maintenance, patients converting from MS positivity to negativity had an excellent PFS (median not reached) while patients converting from MS negativity to positivity progressed early (median 0.6 years, 95% CI: 0.3-not reached). Among patients with sustained MS positivity, the baseline high-risk cytogenetic status had a significant impact and defined a group with poor PFS. Combining minimal residual disease (MRD) in the bone marrow and MS allowed the identification of double negative patients with a favorable PFS (median 3.33 years, 95% CI: 3.08-not reached) and no overall survival events. Our study provides strong evidence that MS is superior to conventional response monitoring, highlighting the potential of MS to become a new standard. Our data indicate that MS should be performed sequentially and combined with baseline disease features and MRD to improve its clinical value. Clinical Trials Register: EudraCT No. 2010-019173-16
- Published
- 2023
- Full Text
- View/download PDF
3. Management of patients with multiple myeloma beyond the clinical-trial setting: understanding the balance between efficacy, safety and tolerability, and quality of life
- Author
-
Evangelos Terpos, Joseph Mikhael, Roman Hajek, Ajai Chari, Sonja Zweegman, Hans C. Lee, María-Victoria Mateos, Alessandra Larocca, Karthik Ramasamy, Martin Kaiser, Gordon Cook, Katja C. Weisel, Caitlin L. Costello, Jennifer Elliott, Antonio Palumbo, and Saad Z. Usmani
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Treatment options in multiple myeloma (MM) are increasing with the introduction of complex multi-novel-agent-based regimens investigated in randomized clinical trials. However, application in the real-world setting, including feasibility of and adherence to these regimens, may be limited due to varying patient-, treatment-, and disease-related factors. Furthermore, approximately 40% of real-world MM patients do not meet the criteria for phase 3 studies on which approvals are based, resulting in a lack of representative phase 3 data for these patients. Therefore, treatment decisions must be tailored based on additional considerations beyond clinical trial efficacy and safety, such as treatment feasibility (including frequency of clinic/hospital attendance), tolerability, effects on quality of life (QoL), and impact of comorbidities. There are multiple factors of importance to real-world MM patients, including disease symptoms, treatment burden and toxicities, ability to participate in daily activities, financial burden, access to treatment and treatment centers, and convenience of treatment. All of these factors are drivers of QoL and treatment satisfaction/compliance. Importantly, given the heterogeneity of MM, individual patients may have different perspectives regarding the most relevant considerations and goals of their treatment. Patient perspectives/goals may also change as they move through their treatment course. Thus, the ‘efficacy’ of treatment means different things to different patients, and treatment decision-making in the context of personalized medicine must be guided by an individual’s composite definition of what constitutes the best treatment choice. This review summarizes the various factors of importance and practical issues that must be considered when determining real-world treatment choices. It assesses the current instruments, methodologies, and recent initiatives for analyzing the MM patient experience. Finally, it suggests options for enhancing data collection on patients and treatments to provide a more holistic definition of the effectiveness of a regimen in the real-world setting.
- Published
- 2021
- Full Text
- View/download PDF
4. Lenalidomide versus bortezomib maintenance after frontline autologous stem cell transplantation for multiple myeloma
- Author
-
Marc-Andrea Baertsch, Elias K. Mai, Thomas Hielscher, Uta Bertsch, Hans J. Salwender, Markus Munder, Stephan Fuhrmann, Ulrich Dührsen, Peter Brossart, Kai Neben, Jana Schlenzka, Christina Kunz, Marc S. Raab, Jens Hillengaß, Anna Jauch, Anja Seckinger, Dirk Hose, Steffen Luntz, Pieter Sonneveld, Henk Lokhorst, Hans Martin, Martin Goerner, Martin Hoffmann, Hans-Walter Lindemann, Helga Bernhard, Igor W. Blau, Christof Scheid, Britta Besemer, Katja C. Weisel, Mathias Hänel, Jan Dürig, Hartmut Goldschmidt, and German-Speaking Myeloma Multicenter Group (GMMG)
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Lenalidomide (LEN) maintenance (MT) post autologous stem cell transplantation (ASCT) is standard of care in newly diagnosed multiple myeloma (MM) but has not been compared to other agents in clinical trials. We retrospectively compared bortezomib (BTZ; n = 138) or LEN (n = 183) MT from two subsequent GMMG phase III trials. All patients received three cycles of BTZ-based triplet induction and post-ASCT MT. BTZ MT (1.3 mg/m2 i.v.) was administered every 2 weeks for 2 years. LEN MT included two consolidation cycles (25 mg p.o., days 1–21 of 28 day cycles) followed by 10–15 mg/day for 2 years. The BTZ cohort more frequently received tandem ASCT (91% vs. 33%) due to different tandem ASCT strategies. In the LEN and BTZ cohort, 43% and 46% of patients completed 2 years of MT as intended (p = 0.57). Progression-free survival (PFS; HR = 0.83, p = 0.18) and overall survival (OS; HR = 0.70, p = 0.15) did not differ significantly with LEN vs. BTZ MT. Patients with
- Published
- 2021
- Full Text
- View/download PDF
5. Rates of Influenza and Pneumococcal Vaccination and Correlation With Survival in Multiple Myeloma Patients
- Author
-
Michael A. Thompson, Mario Boccadoro, Xavier Leleu, Jorge Vela-Ojeda, Frits van Rhee, Katja C. Weisel, Robert M. Rifkin, Saad Z. Usmani, Roman Hájek, Gordon Cook, Rafat Abonour, Mira Armour, Kathryn E. Morgan, Su-Peng Yeh, Caitlin L. Costello, Jesus G. Berdeja, Faith E. Davies, Jeffrey A. Zonder, Hans C. Lee, Jim Omel, Andrew Spencer, Evangelos Terpos, Vania T.M. Hungria, Noemi Puig, Chengcheng Fu, Renda H. Ferrari, Kaili Ren, Dawn Marie Stull, and Ajai Chari
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2023
6. Prognostic significance of cytogenetic heterogeneity in patients with newly diagnosed multiple myeloma
- Author
-
Maximilian Merz, Anna Jauch, Thomas Hielscher, Tilmann Bochtler, Stefan Olaf Schönland, Anja Seckinger, Dirk Hose, Uta Bertsch, Kai Neben, Marc Steffen Raab, Jens Hillengass, Hans Salwender, Igor Wolfgang Blau, Hans-Walter Lindemann, Ingo G.H. Schmidt-Wolf, Christof Scheid, Mathias Haenel, Katja C. Weisel, and Hartmut Goldschmidt
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: We investigated subclonal cytogenetic aberrations (CA) detected by interphase fluorescence in situ hybridization (iFISH) in patients with newly diagnosed multiple myeloma (MM) enrolled in the Haemato Oncology Foundation for Adults in the Netherlands (HOVON)–65/German-Speaking MM Group (GMMG)–HD4 phase 3 trial. Patients were either treated with 3 cycles of vincristine, Adriamycin, and dexamethasone or bortezomib, Adriamycin, and dexamethasone and then thalidomide or bortezomib maintenance after tandem autologous transplantation. Subclones were defined either by presence of different copy numbers of the same chromosome loci and/or CA present in at least 30% less and maximally 2/3 of cells compared with the main clone CA. Patients with subclones harbored more frequently high risk (31.0%) or hyperdiploid main clone aberrations (24.8%) than patients with t(11;14) in the main clone (10.1%). Gains and deletions of c-MYC were the only CA that occurred more frequently as subclone (8.1%/20.5%) than main clone (6.2%/3.9%, respectively). Treatment with bortezomib completely overcame the negative prognosis of high-risk CA in patients without subclones, but not in patients with additional subclonal CA. High-risk patients treated without bortezomib showed dismal outcome whether subclones were present or not. Cytogenetic heterogeneity defined by subclonal CA is of major prognostic significance in newly diagnosed MM patients treated with bortezomib within the HOVON-65/GMMG-HD4 trial.
- Published
- 2018
- Full Text
- View/download PDF
7. Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial
- Author
-
Hartmut Goldschmidt, Elias K Mai, Uta Bertsch, Roland Fenk, Eva Nievergall, Diana Tichy, Britta Besemer, Jan Dürig, Roland Schroers, Ivana von Metzler, Mathias Hänel, Christoph Mann, Anne M Asemissen, Bernhard Heilmeier, Niels Weinhold, Stefanie Huhn, Katharina Kriegsmann, Steffen P Luntz, Tobias A W Holderried, Karolin Trautmann-Grill, Deniz Gezer, Maika Klaiber-Hakimi, Martin Müller, Cyrus Khandanpour, Wolfgang Knauf, Christof Scheid, Markus Munder, Thomas Geer, Hendrik Riesenberg, Jörg Thomalla, Martin Hoffmann, Marc S Raab, Hans J Salwender, Katja C Weisel, Joachim Behringer, Helga Bernhard, Christiane Bernhardt, Igor W Blau, Claus Bolling, Daniel Debatin, Gerrit Dingeldein, Barbara Ferstl, Claudia Fest, Stefan Fronhoffs, Stephan Fuhrmann, Tobias Gaska, Martin Görner, Ullrich Graeven, Jochen Grassinger, Michael Heinsch, Gerhard Held, Olaf Hopfer, Peter Immenschuh, Dominic Kaddu-Mulindwa, Martine Klausmann, Stefan Klein, Yon-Dschun Ko, Georg Köchling, Michael Koenigsmann, Philippe Kostrewa, Doris Maria Kraemer, Stephan Kremers, Martin Kropff, Paul La Rosée, Rolf Mahlberg, Uwe Martens, Michael Neise, Holger Nückel, Wolfram Pönisch, Maria Procaccianti, Mohammed R Rafiyan, Peter Reimer, Armin Riecke, Mathias Rummel, Volker Runde, Markus Schaich, Christoph Scheid, Martin Schmidt-Hieber, Stefan Schmitt, Daniel Schöndube, Andreas Schwarzer, Peter Staib, Heike Steiniger, Dirk Sturmberg, Hans-Joachim Tischler, Arne Trummer, Barbara Tschechne, Walter Verbeek, Bettina Whitlock, Maike de Wit, Matthias Zaiß, and Carsten Ziske
- Subjects
Male ,Bortezomib ,Antineoplastic Combined Chemotherapy Protocols ,Medizin ,Humans ,Female ,Induction Chemotherapy ,Hematology ,Middle Aged ,Multiple Myeloma ,Lenalidomide ,Dexamethasone - Abstract
Anti-CD38 monoclonal antibodies have consistently shown increased efficacy when added to standard of care for patients with multiple myeloma. We aimed to assess the efficacy of isatuximab in addition to lenalidomide, bortezomib, and dexamethasone in patients with newly diagnosed transplantation-eligible multiple myeloma.This open-label, multicentre, randomised, active-controlled, phase 3 trial was done at 67 academic and oncology practice centres in Germany. This study is ongoing and divided into two parts; herein, we report results from part 1. Eligible patients were aged 18-70 years; had a confirmed diagnosis of untreated multiple myeloma requiring systemic treatment and a WHO performance status of 0-2; and were eligible for induction therapy, high-dose melphalan and autologous haematopoietic stem-cell transplantation, and maintenance treatment. Patients were randomly assigned (1:1) to receive three 42-day cycles of induction therapy either with isatuximab plus lenalidomide, bortezomib, and dexamethasone (isatuximab group) or lenalidomide, bortezomib, and dexamethasone alone (control group) using a web-based system and permuted blocks. Patients in both groups received lenalidomide (25 mg orally on days 1-14 and 22-35), bortezomib (1·3 mg/mBetween Oct 23, 2018, and Sep 22, 2020, 660 patients were included in the ITT analysis (331 in the isatuximab group and 329 in the control group). 654 (99%) patients were White, two were African, one was Arabic, and three were Asian. 250 (38%) were women and 410 (62%) were men. The median age was 59 years (IQR 54-64). MRD negativity after induction therapy was reached in 166 (50%) patients in the isatuximab group versus 117 (36%) in the control group (OR 1·82 [95% CI 1·33-2·48]; p=0·00017). Median follow-up time from start to end of induction therapy was 125 days (IQR 125-131) versus 125 days (125-132). At least one grade 3 or 4 adverse event occurred in 208 (63%) of 330 patients versus 199 (61%) of 328 patients. Neutropenia of grade 3 or 4 occurred in 77 (23%) versus 23 (7%) patients and infections of grade 3 or 4 occurred in 40 (12%) versus 32 (10%) patients. Among 12 deaths during induction therapy, one death due to septic shock in the isatuximab group and four deaths (one cardiac decompensation, one hepatic and renal failure, one cardiac arrest, and one drug-induced enteritis) in the control group were considered treatment-related.Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone for induction therapy improved rates of MRD negativity with no new safety signals in patients with newly diagnosed transplantation-eligible multiple myeloma.Sanofi and Bristol Myers Squibb (Celgene).
- Published
- 2022
8. Final Analysis of Carfilzomib, Dexamethasone, and Daratumumab Versus Carfilzomib and Dexamethasone in the CANDOR Study
- Author
-
Saad Z. Usmani, Hang Quach, Maria-Victoria Mateos, Ola Landgren, Xavier Leleu, David S. Siegel, Katja C. Weisel, Xiaomei Shu, Chuang Li, and Meletios A. Dimopoulos
- Subjects
Hematology - Abstract
CANDOR (NCT03158688) is a phase 3, randomized, open-label trial comparing carfilzomib, daratumumab, and dexamethasone (KdD) versus carfilzomib and dexamethasone (Kd) in adults with relapsed/refectory multiple myeloma (RRMM) with 1-3 prior therapies. The CANDOR study met its primary endpoint of progression-free survival (PFS) in the primary analysis. Here we report the final analysis of the study, including secondary endpoints and subgroup analyses thereof. Median follow-up was 50 months. Patients treated with KdD had higher minimal residual disease-negative (MRD-) achievement rates (28% vs 9%; odds ratio [OR], 4.22 [95% CI, 2.28-7.83]) and MRD- complete response rates (22% vs 8%; OR, 3.55 [1.83-6.88]) than those treated with Kd. Median PFS was 28.4 months for KdD versus 15.2 months for Kd (hazard ratio [HR], 0.64 [95% CI, 0.49-0.83]). Median overall survival (OS) was 50.8 months versus 43.6 months, respectively (HR, 0.78 [0.60-1.03]; P=0.042). Trends toward improved OS occurred in predefined subgroups, including patients refractory to lenalidomide (KdD, not reached; Kd, 38.2 months; HR, 0.69 [0.43-1.11]) and refractory to proteasome inhibitor (KdD, 43.2 months; Kd, 30.0 months; HR, 0.70 [0.45-1.09]), and there was significant improvement in patients with high-risk cytogenetics (KdD, 34.3 months; Kd: 17.1 months; HR, 0.52 [0.29-0.94]). No new safety signals were identified. In summary, the final analysis of CANDOR confirmed the PFS benefit and showed a trend in OS benefit with KdD versus Kd. These findings reinforce KdD as a standard of care for RRMM, especially in clinically relevant patient subgroups.
- Published
- 2023
9. Impfempfehlungen beim Multiplen Myelom: ein Konsensbericht des European Myeloma Network
- Author
-
Heinz Ludwig, Mario Boccadoro, Philippe Moreau, Jesus San-Miguel, Michele Cavo, Charlotte Pawlyn, Sonja Zweegman, Thierry Facon, Christoph Driessen, Roman Hajek, Melitios A. Dimopoulos, Francesca Gay, Herve Avet-Loiseau, Evangelos Terpos, Niklas Zojer, Mohamad Mohty, Maria-Victoria Mateos, Hermann Einsele, Michel Delforge, Jo Caers, Katja C. Weisel, Graham Jackson, Laurent Garderet, Monika Engelhardt, Niels van de Donk, Xavier Leleu, Hartmut Goldschmidt, Meral Beksac, Inger Nijhof, Niels Abildgaard, Sara Bringhen, and Pieter Sonneveld
- Abstract
Bei der Impfung handelt es sich um eine der erfolgreichsten medizinischen Maßnahmen, die bereits Millionen von Menschen das Leben gerettet hat. Die Impfung ist besonders wichtig bei Patienten mit einem Multiplen Myelom, deren Infektionsrisiko aufgrund der krankheitsinhärenten Immunsuppression sowie der immunsuppressiven Wirkung der Therapie erhöht ist. Daher sollten alle geeigneten Maßnahmen ergriffen werden, um eine wirksame Immunantwort gegen weitverbreitete Krankheitserreger wie Influenza, Pneumokokken, das Varicella-Zoster-Virus sowie gegen Bakterien und Viren (Haemophilus influenzae, Meningokokken und Hepatitis), die häufig ein erhebliches Risiko für Patienten mit einem Multiplen Myelom darstellen, anzuregen. Patienten nach autologer und insbesondere nach allogener Transplantation haben stark reduzierte Antikörpertiter und benötigen daher ein breiteres Impfspektrum. Die Impfreaktion ist bei Myelom-Patienten oft weniger stark ausgeprägt als in der Allgemeinbevölkerung, sodass entweder die Messung der Antikörpertiter nach der Impfung und/oder eine Wiederholungsimpfung erforderlich ist. In dieser Arbeit stellen wir die Daten zusammen, die zur Impfung von Patienten mit Multiplem Myelom existieren, und geben Empfehlungen für die klinische Praxis.
- Published
- 2022
10. A phase II clinical trial of combined BRAF/MEK inhibition for BRAFV600E-mutated multiple myeloma
- Author
-
Nicola, Giesen, Manik, Chatterjee, Christof, Scheid, Alexandra M, Poos, Britta, Besemer, Kaya, Miah, Axel, Benner, Nicole, Becker, Thomas, Moehler, Ivana, Metzler, Cyrus, Khandanpour, Andrea, Seidel-Glaetzer, Karolin, Trautmann-Grill, K Martin, Kortüm, Carsten, Müller-Tidow, Gunhild, Mechtersheimer, Benjamin, Goeppert, Albrecht, Stenzinger, Niels, Weinhold, Hartmut, Goldschmidt, Katja C, Weisel, and Marc S, Raab
- Abstract
Activating BRAF mutations are found in a small subset of patients with newly diagnosed multiple myeloma but prevalence increases in late stage, refractory disease, and are associated with adverse outcome. This prospective single-arm, open-label, multicenter phase II trial assessed the efficacy and safety of combined BRAF/MEK inhibition, using encorafenib and binimetinib, in patients with relapsed/refractory multiple myeloma (RRMM) carrying a BRAFV600E mutation (ClinicalTrials.gov identifier: NCT02834364). Patients received 450 mg encorafenib once daily and binimetinib 45 mg twice daily. The primary end point was the overall response rate achieved within the first year after start of treatment according to IMWG criteria. Twelve RRMM patients with a median of 5 prior lines of therapy were enrolled. The overall response rate was 83.3% with 10 patients achieving at least a partial response. The median progression-free survival (PFS) was 5.6 months and overall survival was 55% at 24 months. Emerging resistance to therapy was driven by RAS mutations and structural variants involving the BRAF locus. This is the first prospective clinical trial to demonstrate that combined BRAF/MEK inhibition is highly effective in patients with BRAFV600E mutated RRMM and represents a successful targeted precision medicine approach in this disease.
- Published
- 2022
11. Implications and prognostic impact of mass spectrometry in patients with newly-diagnosed multiple myeloma
- Author
-
Elias K. Mai, Stefanie Huhn, Kaya Miah, Alexandra M. Poos, Christof Scheid, Katja C. Weisel, Uta Bertsch, Markus Munder, Oscar Berlanga, Dirk Hose, Anja Seckinger, Anna Jauch, Igor W. Blau, Mathias Hänel, Hans J. Salwender, Axel Benner, Marc S. Raab, Hartmut Goldschmidt, Niels Weinhold, Hematology, and Basic (bio-) Medical Sciences
- Subjects
bone marrow ,hematology ,Immunology ,Neoplasm, Residual/diagnosis ,Cell Biology ,Prognosis ,Biochemistry ,Treatment Outcome ,oncology ,Internal Medicine ,Humans ,Multiple Myeloma/diagnosis ,Retrospective Studies ,mass spectrometry ,transplantation - Abstract
Mass spectrometry (MS) is a promising tool for monitoring monoclonal protein in plasma cell dyscrasias. We included 480 transplant-eligible newly-diagnosed multiple myeloma (MM) patients from the GMMG-MM5 trial (EudraCT No. 2010-019173-16) and performed a retrospective MS analysis at baseline (480 patients) and at the pre-defined, consecutive time points after induction (444 patients), prior to maintenance (305 patients) and after one year of maintenance (227 patients). We found that MS negativity was significantly associated with improved progression-free survival (PFS) even in patients with complete response (CR) at all investigated follow-up time points. The prognostic impact was independent of established risk factors, such as the revised International Staging System. Combining MS and baseline cytogenetics improved the prediction of outcome: MS-positive patients with high-risk cytogenetics had a dismal PFS of 1.9 years (95% confidence interval [CI]: 1.6–2.3 years) from the start of maintenance. Testing the value of sequential MS prior to and after one year of maintenance, patients converting from MS positivity to negativity had an excellent PFS (median not reached) while patients converting from MS negativity to positivity progressed early (median 0.6 years, 95% CI: 0.3-not reached). Among patients with sustained MS positivity, the baseline high-risk cytogenetic status had a significant impact and defined a group with poor PFS. Combining minimal residual disease (MRD) in the bone marrow and MS allowed the identification of double negative patients with a favorable PFS (median 3.33 years, 95% CI: 3.08-not reached) and no overall survival events. Our study provides strong evidence that MS is superior to conventional response monitoring, highlighting the potential of MS to become a new standard. Our data indicate that MS should be performed sequentially and combined with baseline disease features and MRD to improve its clinical value.Clinical Trials Register: EudraCT No. 2010-019173-16
- Published
- 2022
12. Risk factors for poor humoral response to primary and booster SARS-CoV-2 vaccination in hematologic and oncological outpatients-COVIDOUT study
- Author
-
Martin Schönlein, Victoria Wrage, Susanne Ghandili, Sibylle C. Mellinghoff, Thomas Theo Brehm, Lisa B. Leypoldt, Nils Utz, Roland M. Schrader, Winfried Alsdorf, Niklas Börschel, Lara Bußmann, Martin Schönrock, Dorothea Perlick, Gerhard Schön, Karl Verpoort, Marc Lütgehetmann, Julian Schulze zur Wiesch, Katja C. Weisel, Carsten Bokemeyer, Philippe Schafhausen, and Marianne Sinn
- Subjects
Cancer Research ,COVID-19 Vaccines ,Oncology ,Risk Factors ,SARS-CoV-2 ,Outpatients ,Vaccination ,COVID-19 ,Humans - Published
- 2022
13. Analysis of renal impairment in MM-003, a phase III study of pomalidomide + low - dose dexamethasone versus high - dose dexamethasone in refractory or relapsed and refractory multiple myeloma
- Author
-
Katja C. Weisel, Meletios A. Dimopoulos, Philippe Moreau, Martha Q. Lacy, Kevin W. Song, Michel Delforge, Lionel Karlin, Hartmut Goldschmidt, Anne Banos, Albert Oriol, Adrian Alegre, Christine Chen, Michele Cavo, Laurent Garderet, Valentina Ivanova, Joaquin Martinez-Lopez, Stefan Knop, Xin Yu, Kevin Hong, Lars Sternas, Christian Jacques, Mohamed H. Zaki, and Jesus San Miguel
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Pomalidomide + low-dose dexamethasone is effective and well tolerated for refractory or relapsed and refractory multiple myeloma after bortezomib and lenalidomide failure. The phase III trial MM-003 compared pomalidomide + low-dose dexamethasone with high-dose dexamethasone. This subanalysis grouped patients by baseline creatinine clearance ≥ 30 − < 60 mL/min (n=93, pomalidomide + low-dose dexamethasone; n=56, high-dose dexamethasone) or ≥ 60 mL/min (n=205, pomalidomide + low-dose dexamethasone; n=93, high-dose dexamethasone). Median progression-free survival was similar for both subgroups and favored pomalidomide + low-dose dexamethasone versus high-dose dexamethasone: 4.0 versus 1.9 months in the group with baseline creatinine clearance ≥ 30 − < 60 mL/min (P
- Published
- 2016
- Full Text
- View/download PDF
14. Impact of prior treatment and depth of response on survival in MM-003, a randomized phase 3 study comparing pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone in relapsed/refractory multiple myeloma
- Author
-
Jesus F. San Miguel, Katja C. Weisel, Kevin W. Song, Michel Delforge, Lionel Karlin, Hartmut Goldschmidt, Philippe Moreau, Anne Banos, Albert Oriol, Laurent Garderet, Michele Cavo, Valentina Ivanova, Adrian Alegre, Joaquin Martinez-Lopez, Christine Chen, Christoph Renner, Nizar Jacques Bahlis, Xin Yu, Terri Teasdale, Lars Sternas, Christian Jacques, Mohamed H. Zaki, and Meletios A. Dimopoulos
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Pomalidomide is a distinct oral IMiD® immunomodulatory agent with direct antimyeloma, stromal-support inhibitory, and immunomodulatory effects. The pivotal, multicenter, open-label, randomized phase 3 trial MM-003 compared pomalidomide + low-dose dexamethasone vs high-dose dexamethasone in 455 patients with refractory or relapsed and refractory multiple myeloma after failure of bortezomib and lenalidomide treatment. Initial results demonstrated significantly longer progression-free survival and overall survival with an acceptable tolerability profile for pomalidomide + low-dose dexamethasone vs high-dose dexamethasone. This secondary analysis describes patient outcomes by treatment history and depth of response. Pomalidomide + low-dose dexamethasone significantly prolonged progression-free survival and favored overall survival vs high-dose dexamethasone for all subgroups analyzed, regardless of prior treatments or refractory status. Both univariate and multivariate analyses showed that no variable relating to either the number (≤ or > 3) or type of prior treatment was a significant predictor of progression-free survival or overall survival. No cross-resistance with prior lenalidomide or thalidomide treatment was observed. Patients achieving a minimal response or better to pomalidomide + low-dose dexamethasone treatment experienced a survival benefit, which was even higher in those achieving at least a partial response (17.2 and 19.9 months, respectively, as compared with 7.5 months for patients with less than minimal response). These data suggest that pomalidomide + low-dose dexamethasone should be considered a standard of care in patients with refractory or relapsed and refractory multiple myeloma regardless of prior treatment. ClinicalTrials.gov: NCT01311687; EudraCT: 2010-019820-30.
- Published
- 2015
- Full Text
- View/download PDF
15. Cytogenetics and long-term survival of patients with refractory or relapsed and refractory multiple myeloma treated with pomalidomide and low-dose dexamethasone
- Author
-
Meletios A. Dimopoulos, Katja C. Weisel, Kevin W. Song, Michel Delforge, Lionel Karlin, Hartmut Goldschmidt, Philippe Moreau, Anne Banos, Albert Oriol, Laurent Garderet, Michele Cavo, Valentina Ivanova, Adrian Alegre, Joaquin Martinez-Lopez, Christine Chen, Andrew Spencer, Stefan Knop, Nizar J. Bahlis, Christoph Renner, Xin Yu, Kevin Hong, Lars Sternas, Christian Jacques, Mohamed H. Zaki, and Jesus F. San Miguel
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Patients with refractory or relapsed and refractory multiple myeloma who no longer receive benefit from novel agents have limited treatment options and short expected survival. del(17p) and t(4;14) are correlated with shortened survival. The phase 3 MM-003 trial demonstrated significant progression-free and overall survival benefits from treatment with pomalidomide plus low-dose dexamethasone compared to high-dose dexamethasone among patients in whom bortezomib and lenalidomide treatment had failed. At an updated median follow-up of 15.4 months, the progression-free survival was 4.0 versus 1.9 months (HR, 0.50; P
- Published
- 2015
- Full Text
- View/download PDF
16. Health-related quality of life from the MM-003 trial of pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone in relapsed and/or refractory multiple myeloma
- Author
-
Kevin W. Song, Meletios A. Dimopoulos, Katja C. Weisel, Philippe Moreau, Antonio Palumbo, Andrew Belch, Stephen Schey, Pieter Sonneveld, Lars Sternas, Xin Yu, Ramesh Amatya, Mara S. Monzini, Mohamed Zaki, Christian Jacques, and Jesus San Miguel
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2015
- Full Text
- View/download PDF
17. Multiples Myelom: Der Innovationsfluss in der Myelomtherapie reißt nicht ab
- Author
-
Katja C. Weisel
- Published
- 2022
18. Depth of Response to Isatuximab, Carfilzomib, Lenalidomide and Dexamethasone (Isa-KRd) in Front-Line Treatment of High-Risk Multiple Myeloma: Interim Analysis of the GMMG-CONCEPT Trial
- Author
-
Lisa B. Leypoldt, Anne Marie Asemissen, Britta Besemer, Mathias Haenel, Igor Wolfgang Blau, Martin Görner, Yon-Dschun Ko, Hans Christian Reinhardt, Peter Staib, Christoph Mann, Raphael Lutz, Markus Munder, Ullrich Graeven, Rudolf Peceny, Hans Salwender, Anna Jauch, Manola Zago, Axel Benner, Diana Tichy, Carsten Bokemeyer, Hartmut Goldschmidt, Katja C. Weisel, and German Speaking Myeloma Multicenter Group (GMMG)
- Subjects
medicine.medical_specialty ,education.field_of_study ,Study drug ,Population ,Ethics committee ,Front line ,Interim analysis ,Safety profile ,Informed consent ,Family medicine ,Induction therapy ,medicine ,Business ,education - Abstract
Background: High-risk (HR) multiple myeloma (MM) has an impaired prognostic outcome. Addition of anti-CD38 monoclonal antibodies to standard-of-care regimens improved response rates and depth of response. Here, we report a prespecified interim analysis (IA) following induction therapy with the quadruplet regimen isatuximab, carfilzomib, lenalidomide and dexamethasone (Isa-KRd) in newly diagnosed (ND) HR MM patients (pts) in the phase II multicentric GMMG-CONCEPT trial (NCT03104842). Methods: 153 pts with HR NDMM were enrolled into the trial, the IA reports on the first 50 pts evaluable for IA. Pts receive Isa-KRd in induction, consolidation and Isa-KR maintenance. Transplant eligible pts undergo high-dose therapy. This IA reports on overall response rates (ORR) during induction. Findings: 50 pts were included in the IA population for ORR. HR MM was defined by del17p in 52%, t(4;14) in 38%, t(14;16) in 12% and > 3 copies 1q21 in 42% and ISS stage 2/3 disease. 46/50 pts completed induction treatment. ORR was 100%, with 5 pts (10·0%) showing partial response (PR), 22 (44·0%; including 4 in arm B) very good partial response (VGPR) and 23 (46·0%) complete response (CR). Hematologic grade 3/4 treatment-emergent adverse events (≥ 10%) were neutropenia (34·0%), leukopenia (26·0%) and thrombocytopenia (14·0%). Main non-hematologic toxicities grade 3/4 were hypertension (12·0%) and infection (8·0%). Interpretation: We report for the first time on a trial investigating Isa-KRd quadruplet treatment in solely HR NDMM. Isa-KRd induction induces rapid and deep responses. The overall safety profile is consistent with previous reports. Trial Registration: The trial is registered at clinicaltrials.gov (NCT03104842). Funding Statement: Study drug and financial support by Amgen, Celgene-BMS and Sanofi. Declaration of Interests: Dr. Leypoldt reports grants and non-financial support from Celgene-BMS, grants and non-financial support from Sanofi, grants and non-financial support from Amgen, during the conduct of the study; non-financial support from GSK, non-financial support from Abbvie, outside the submitted work; Dr. Asemissen has nothing to disclose. Dr. Besemer has nothing to disclose. Dr. Hanel reports personal fees from Celgene, personal fees from Novartis, personal fees from Takeda, personal fees from Amgen, during the conduct of the study; Dr. Blau has nothing to disclose. Dr. Gorner has nothing to disclose. Dr. Ko has nothing to disclose. Dr. Reinhardt reports personal fees from Abbvie, grants from Gilead, personal fees from Merck, other from CDL Therapeutics GmbH, outside the submitted work; Dr. Staib reports grants, personal fees, non-financial support and other from Abbvie, grants, personal fees, non-financial support and other from Amgen, grants, personal fees, non-financial support and other from Celgene, grants, personal fees, non-financial support and other from Janssen-Cilag, grants, personal fees, non-financial support and other from Novartis, grants, personal fees, non-financial support and other from Gilead, grants, personal fees, non-financial support and other from Pfizer, grants, personal fees, non-financial support and other from Roche, outside the submitted work; Dr. Mann has nothing to disclose. Dr. Lutz has nothing to disclose. Dr. Munder reports personal fees and non-financial support from Janssen, personal fees and non-financial support from Amgen, grants from Incyte, personal fees and non-financial support from BMS, personal fees from Abbvie, personal fees from Sanofi, personal fees from GSK, personal fees from Takeda, outside the submitted work; Dr. Graeven reports personal fees from Amgen, personal fees and non-financial support from Boehringer Ingelheim, personal fees from Daichi Sankyo, personal fees from Servier, personal fees from Celgene, personal fees from Astra Zeneca, personal fees from Johnson Johnson, non-financial support from Merck, personal fees from MSD, personal fees from BMS, during the conduct of the study; Dr. Peceny reports grants and personal fees from Sanofi Genzyme, grants from Novartis, grants from DRK Blutspendedienst NSTOB, grants from Boehringer Ingelheim Pharma GmbH & Co KG, grants from Celgene, outside the submitted work; Dr. Salwender reports personal fees from Bristol-Myers Squibb/Celgene, personal fees from Janssen Cilag, personal fees from Glaxo Smith Kline, personal fees from Oncopeptides, personal fees from Takeda, personal fees from Sanofi, personal fees from AbbVie, personal fees from Amgen, outside the submitted work; Dr. Jauch has nothing to disclose. Dr. Zago has nothing to disclose. Axel Benner has nothing to disclose. Dr. Tichy has nothing to disclose. Dr. Bokemeyer reports personal fees from Sanofi Aventis, personal fees from Merck KgA, personal fees from Bristol-Myers Squibb, personal fees from Merck Sharp & Dohme, personal fees from Lilly Imclone, personal fees from Bayer Healthcare, personal fees from GSO Contract research, personal fees from AOK-Rheinland-Hamburg, personal fees from Novartis, outside the submitted work; Dr. Goldschmidt reports grants, personal fees, non-financial support and other from Amgen, grants, personal fees, non-financial support and other from BMS, grants, personal fees, non-financial support and other from Celgene, grants, personal fees, and other from Chugai, grants, personal fees, non-financial support and other from Janssen, grants, personal fees, non-financial support and other from Sanofi, other from Incyte, other from Molecular Partners, other from Merck Sharp and Dohme (MSD), other from Mundipharma, grants, personal fees, non-financial support and other from Takeda, personal fees and other from Novartis, personal fees from Adaptive Biotechnology, personal fees from GlaxoSmithKline (GSK), outside the submitted work. Dr. Weisel reports grants from AMGEN, grants from Celgene, grants from Sanofi, during the conduct of the study; grants, personal fees and non-financial support from Amgen, personal fees and non-financial support from BMS, grants, personal fees and non-financial support from Celgene, personal fees from Adaptive Biotech, grants, personal fees and non-financial support from Janssen, personal fees and non-financial support from GSK, personal fees from Karyopharm, grants, personal fees and non-financial support from Sanofi, personal fees and non-financial support from Takeda, personal fees from Oncopeptides, personal fees from Roche, outside the submitted work. Ethics Approval Statement: All patients provided written informed consent. The trial was approved by the competent authorities and the Tuebingen University Ethics Committee.
- Published
- 2021
19. Comparison of NGS and MFC Methods: Key Metrics in Multiple Myeloma MRD Assessment
- Author
-
Katharina Kriegsmann, Michael Hundemer, Nicole Hofmeister-Mielke, Philipp Reichert, Calin-Petru Manta, Mohamed H.S. Awwad, Sandra Sauer, Uta Bertsch, Britta Besemer, Roland Fenk, Mathias Hänel, Markus Munder, Katja C. Weisel, Igor W. Blau, Andreas Neubauer, Carsten Müller-Tidow, Marc S. Raab, Hartmut Goldschmidt, Stefanie Huhn, and for the German-speaking Myeloma Multicenter Group (GMMG)
- Subjects
Cancer Research ,Correlation coefficient ,MRD Negativity ,Concordance ,EuroFlow ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,multiple myeloma (MM) ,next-generation sequencing (NGS) ,Multiple myeloma ,Tumor Load ,Mathematics ,Very Good Partial Response ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Minimal residual disease ,minimal residual disease (MRD) ,Oncology ,030220 oncology & carcinogenesis ,multicolor flow cytometry (MFC) ,Nuclear medicine ,business ,030215 immunology - Abstract
In order to meet the challenges in data evaluation and comparability between studies in multiple myeloma (MM) minimal residual disease (MRD) assessment, the goal of the current study was to provide a step-by-step evaluation of next-generation sequencing (NGS) and multicolor flow cytometry (MFC) data. Bone marrow (BM) sample pairs from 125 MM patients were analyzed by NGS and MFC MM MRD methods. Tumor load (TL) and limit of detection (LOD) and quantification (LOQ) were calculated. The best-fit MRD cut-off was chosen as 1 × 10−5, resulting in an overall 9.6% (n overall = 12 (NGS n = 2, MFC n = 10)) nonassessable cases. The overall concordance rate between NGS and MFC was 68.0% (n = 85), discordant results were found in 22.4% (11.2% (n = 14) of cases in each direction. Overall, 55.1% (n = 60/109) and 49.5% (n = 54/109) of patients with a serological response ≥ very good partial response (VGPR) showed BM MRD negativity by NGS and MFC, respectively. A good correlation in the TL assessed by both techniques was found (correlation coefficient = 0.8, n = 40, p <, 0.001). Overall, our study shows good concordance between MM BM MRD status and TL when comparing NGS and MFC at a threshold of 10–5. However, a sufficient number of analyzed events and calculation of MRD key metrics are essential for the comparison of methods and evaluability of data at a specific MRD cut-off.
- Published
- 2020
20. Modulation of CXC Chemokine Receptor Expression and Function in Human Neutrophils during Aging In Vitro Suggests a Role in Their Clearance from Circulation
- Author
-
Katja C. Weisel, Frank Bautz, Gabriele Seitz, Sedat Yildirim, Lothar Kanz, and Robert Möhle
- Subjects
Pathology ,RB1-214 - Abstract
In mice, differential regulation of CXC chemokine receptor expression in circulating polymorphonuclear neutrophils (PMNs) undergoing senescence results in homing to the bone marrow. However, the role of this compartment and of the chemokine receptor CXCR4 is still under discussion, and only scarce data exist about CXCR4 function in human PMN. In our study, we provide evidence that also in human neutrophils, expression (cell surface and mRNA), chemotactic and signaling functions of the homing-related chemokine receptor CXCR4 are upregulated during aging in vitro, independent of addition of stimulatory cytokines (TNF, IL-1, IL-8, G-CSF). In contrast, interleukin-8 receptors are downmodulated (CXCR2) or remain unchanged (CXCR1), suggesting that human PMNs undergoing senescence acquire a phenotype that impairs inflammatory extravasation and favors homing to the bone marrow or other tissues involved in sequestration. Partially retained responsiveness to interleukin-8 may be important for neutrophil function when senescence occurs after extravasation in inflamed tissues.
- Published
- 2009
- Full Text
- View/download PDF
21. Prognostic Impact of Serum Free Light Chain Ratio Normalization in Patients with Multiple Myeloma Treated within the GMMG-MM5 Trial
- Author
-
Eva-Maria Klein, Diana Tichy, Hans J. Salwender, Elias K. Mai, Jan Duerig, Katja C. Weisel, Axel Benner, Uta Bertsch, Mabast Akhavanpoor, Britta Besemer, Markus Munder, Hans-Walter Lindemann, Dirk Hose, Anja Seckinger, Steffen Luntz, Anna Jauch, Ahmet Elmaagacli, Stephan Fuhrmann, Peter Brossart, Martin Goerner, Helga Bernhard, Marc S. Raab, Igor W. Blau, Mathias Haenel, Christof Scheid, Hartmut Goldschmidt, on behalf of the German-Speaking Myeloma Multicenter Group (GMMG), Hematology, and Basic (bio-) Medical Sciences
- Subjects
Normalization (statistics) ,Cancer Research ,medicine.medical_specialty ,Every Three Months ,Medizin ,Urology ,Article ,time-dependent analysis ,Internal Medicine ,medicine ,RC254-282 ,Multiple myeloma ,biology ,hematology ,Proportional hazards model ,business.industry ,Hazard ratio ,prognostic factors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,immune reconstitution ,medicine.disease ,Confidence interval ,multiple myeloma ,Transplantation ,Oncology ,biology.protein ,Antibody ,business ,serum free light chain ratio normalization - Abstract
We investigated the prognostic impact of time-dependent serum free light chain ratio (FLCr) normalization in 590 patients with secretory multiple myeloma (MM) during first-line treatment within the German-Speaking Myeloma Multicenter Group MM5 trial. Serum free light chains (sFLC) were assessed by the Freelite test at baseline, after induction, mobilization, autologous blood stem cell transplantation, consolidation and every three months during maintenance or follow up within two years after the start of maintenance. The proportion of patients with a normal or normalized FLCr increased from 3.6% at baseline to 23.2% after induction and 64.7% after consolidation. The achievement of FLCr normalization at any one time before the start of maintenance was associated with significantly prolonged progression-free survival (PFS) (p <, 0.01, hazard ratio (HR) = 0.61, 95% confidence interval (95% CI) = 0.47–0.79) and overall survival (OS) (p = 0.02, HR = 0.67, 95% CI = 0.48–0.93) in multivariable time-dependent Cox regression analyses. Furthermore, reaching immune reconstitution, defined as the normalization of uninvolved immunoglobulins, before maintenance was associated with superior PFS (p = 0.04, HR = 0.77, 95% CI = 0.60–0.99) and OS (p = 0.01, HR = 0.59, 95% CI = 0.41–0.86). We conclude that FLCr normalization during therapy is an important favorable prognostic factor in MM. Therefore, we recommend serial measurements of sFLC during therapy until achieving FLCr normalization, even in patients with secretory MM.
- Published
- 2021
22. RETRACTED ARTICLE: Immuntherapie des multiplen Myeloms
- Author
-
Lothar Kanz and Katja C. Weisel
- Subjects
Gynecology ,03 medical and health sciences ,medicine.medical_specialty ,0302 clinical medicine ,business.industry ,030220 oncology & carcinogenesis ,medicine ,business ,030215 immunology - Abstract
Die Zulassung von zwei monoklonalen Antikorpern fur die Behandlung des multiplen Myeloms (MM) im Jahr 2016 durch die europaische Arzneimittel-Agentur (EMA) hat eine neue Ara in der zielgerichteten Immuntherapie des MM eingeleitet. Mittlerweile stehen verschiedene neue Ansatze in der Immuntherapie des MM zur Verfugung, die auf unterschiedlichen Therapiestrategien basieren.
- Published
- 2017
23. Retraction Note: Immuntherapie des multiplen Myeloms
- Author
-
Katja C. Weisel and Lothar Kanz
- Subjects
Philosophy ,Humanities - Abstract
Die Autoren haben den Artikel „Immuntherapie des multiplen Myeloms“ [Weisel K, Kanz L. InFo Onkologie. 2017;20(Suppl 1):49-53] zuruckgezogen, weil dieser in wesentlichen Teilen auf dem englischsprachigen Review „Immunologic approaches for the treatment of multiple myeloma“ von Leo Rasche, Niels Weinhold, Gareth J. Morgan, Frits van Rhee und Faith E. Davies [Rasche L et al. Cancer Treat Rev. 2017;55:190-9] basiert. Diese Retraction ersetzt das fruhere Erratum [Weisel K, Kanz L. InFo Onkologie. 2018;21(2):54]. Beide Autoren haben dieser Retraction zugestimmt.
- Published
- 2018
24. Erratum zu: RETRACTED ARTICLE: Immuntherapie des multiplen Myeloms
- Author
-
Lothar Kanz and Katja C. Weisel
- Subjects
Gynecology ,medicine.medical_specialty ,business.industry ,medicine ,business - Published
- 2018
25. Stromal cell lines from the aorta-gonado-mesonephros region are potent supporters of murine and human hematopoiesis
- Author
-
Ying Gao, Katja C. Weisel, Malcolm A.S. Moore, and Jae-Hung Shieh
- Subjects
Cancer Research ,Stromal cell ,Antigens, CD34 ,Bone Marrow Cells ,Stem cell factor ,Biology ,Cell Line ,Mice ,Genetics ,medicine ,Animals ,Humans ,Cloning, Molecular ,Progenitor cell ,Gonads ,Molecular Biology ,Aorta ,Cell Proliferation ,Stem Cells ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Fetal Blood ,Embryonic stem cell ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cord blood ,Mesonephros ,Immunology ,Stromal Cells ,Stem cell ,Adult stem cell - Abstract
Objective The hematopoietic system is nurtured by a supportive stroma environment allowing maintenance and differentiation of hematopoietic stem cells (HSC). However, only a limited number of these stromal cell clones support hematopoiesis in the absence of cytokine supplementation. So far, only two bone marrow–derived stromal cell lines (OP9 and S17) are capable of inducing hematopoietic differentiation of totipotent murine and human embryonic stem cells (ESC). Here, the potential of more than 100 stromal cell lines developed from the aorta-gonado-mesonephros (AGM) region was investigated in supporting adult and embryonic hematopoiesis. In addition, extensive phenotypic analysis should elucidate possible mechanisms involved in maintenance of hematopoietic stem cell function. Methods More than 100 stromal cell clones derived from the AGM region of E10.5 mouse embryos were isolated. Hematopoietic stem cell support was tested for adult murine and human cord blood hematopoietic stem cells and hematopoietic cells derived from murine ESC. Genotypic and phenotypic characterization was performed including gene array analysis. Results It was demonstrated that multiple clones showed high efficiency in supporting maintenance and expansion of primitive murine and human hematopoietic progenitors. In addition, we demonstrated for the first time that AGM stromal cell lines are also potent inducers of hematopoietic differentiation of murine ESC. Microarray analysis of AGM lines revealed a characteristic genotype with expression of genes involved in regulating hematopoiesis as well as mesodermal and early B cell development. Conclusion These AGM stromal cell lines may be of value in elucidating molecular mechanisms regulating early stem cell development and hematopoietic differentiation from ES-derived mesoderm.
- Published
- 2006
26. Meeting summary: International Symposium and Workshop on Hematopoietic Stem Cells V, University of Tübingen, Germany, September 16–18, 2004
- Author
-
Katja C. Weisel, Tim H. Brümmendorf, Donald Orlic, Willem E. Fibbe, and Lothar Kanz
- Subjects
Cancer Research ,Genetics ,Cell Biology ,Hematology ,Molecular Biology - Published
- 2005
27. Phase I/IIa Study of the Human Anti-CD38 Antibody MOR202 (MOR03087) in Relapsed or Refractory Multiple Myeloma
- Author
-
Marc S Raab, Manik Chatterjee, Hartmut Goldschmidt, Hermine Agis, Igor W Blau, Hermann Einsele, Monika Engelhardt, Barbara Ferstl, Martin Gramatzki, Christoph Röllig, Katja C. Weisel, Pia Klöpfer, Dominika Weinelt, Stefan Härtle, and Christian Peschel
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry ,health care economics and organizations - Abstract
Background: CD38 is a type II transmembrane glycoprotein that is expressed at high levels on multiple myeloma cells. MOR202 is a HuCAL-derived, human, IgG1 anti-CD38 monoclonal antibody showing effective antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and high activity in preclinical models of multiple myeloma. Patients and Methods: Here we reportinterimsafety and preliminary efficacy data from this ongoing, multicenter, MOR202 dose-escalation, phase I/IIa study in patients with relapsed or refractory disease who had failed ≥2 prior therapies for multiple myeloma, including an immunomodulatory drug and a proteasome inhibitor. The objectives are to evaluate the safety, maximum tolerated dose (MTD)/recommended dose and preliminary efficacy of MOR202 when administered as monotherapy or in combination with dexamethasone (DEX); pomalidomide (POM) + DEX; and lenalidomide (LEN) + DEX. Patients received MOR202 as a 2-hour intravenous infusion every 2 weeks (q2w; dose levels 0.01-16 mg/kg), 4 mg/kg weekly (q1w) and 4, 8 and 16 mg/kg q1w + DEX. The combination cohorts receiving MOR202 8 mg/kg with LEN + DEX and POM + DEX have been opened, and the 16 mg/kg q1w with LEN + DEX or POM + DEX, as well as confirmation cohorts, are planned. Results: As of 26 June 2015, 44 patients have been treated; 31 and 13 patients in the q2w and q1w dose levels, respectively. Median age was 69 years (range 44-80); median number of prior therapy lines was 4 (2-11). The MTD has not been reached. The most common treatment-emergent adverse events (TEAEs) at any grade were anemia (15 patients, 34%), fatigue (14 patients, 32%), infusion-related reactions (IRRs) and leukopenia (13 patients, 30% each), lymphopenia and nausea (11 patients, 25% each). Grade ≥3 TEAEs were reported for 28 patients (64%); the most common included lymphopenia (8 patients, 18%), leukopenia (5 patients, 11%) and hypertension (4 patients, 9%). IRRs arose mainly during the first infusion; all were grade 1-2 except for one patient (grade 3); no IRRs occurred in patients receiving MOR202 in combination with DEX. Infections were commonly reported (26 patients, 59%) but in the majority of the cases were not considered to be treatment-related. There have been no treatment-related deaths. Pharmacokinetic (PK) data demonstrated a significant target-mediated drug disposition effect for most patients treated q2w. By contrast, patients treated q1w (4 or 8 mg/kg) showed constant or slightly accumulating MOR202 trough levels, suggesting the potential for full target occupancy at 16 mg/kg. Long-lasting tumor control has already been observed in early monotherapy cohorts, including one partial response and one very good partial response in the weekly cohorts; efficacy analyses are ongoing. First data from the dose escalation of the weekly cohorts with DEX and the combination cohorts with LEN + DEX and POM + DEX will be presented. Conclusions: At doses up to 16 mg/kg,MOR202 was safe and well tolerated. Encouraging preliminary activity of MOR202 was observed, especially with the weekly regimen. PK data show the potential for full target occupancy in patients receiving MOR202 16 mg/kg q1w. This dosing schedule of MOR202 is currently being tested in combination with DEX, LEN + DEX, and POM + DEX. Disclosures Raab: MorphoSys: Research Funding; Novartis: Consultancy, Research Funding, Speakers Bureau; Celgene: Consultancy; BMS: Consultancy. Goldschmidt:Chugai: Honoraria, Research Funding, Speakers Bureau; Onyx: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Millenium: Honoraria, Research Funding, Speakers Bureau; Janssen-Cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Bristol-Myers Squibb: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Einsele:Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen/Onyx: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau. Ferstl:Bristol-Myers Squibb: Other: Advisory board; Novartis: Other: Case report presentation. Weisel:Celgene: Consultancy, Honoraria, Other: Travel Support, Research Funding; Amgen: Consultancy, Honoraria, Other: Travel Support; Novartis: Other: Travel Support; Janssen Pharmaceuticals: Consultancy, Honoraria, Other: Travel Support, Research Funding; BMS: Consultancy, Honoraria, Other: Travel Support; Onyx: Consultancy, Honoraria; Noxxon: Consultancy. Klöpfer:MorphoSys: Employment. Weinelt:MorphoSys: Employment. Härtle:MorphoSys: Employment.
- Published
- 2015
28. Safety of Treatment (Tx) with Pomalidomide (POM) and Low-Dose Dexamethasone (LoDEX) in Patients (Pts) with Relapsed or Refractory Multiple Myeloma (RRMM) and Renal Impairment (RI), Including Those on Dialysis
- Author
-
Karthik Ramasamy, Meletios A. Dimopoulos, Niels W.C.J. van de Donk, Barbara Gamberi, Frank Bridoux, Matthew Streetly, Elisabeth Kueenburg, Barbara Rosettani, Shona Collins, Frederik Lersch, Pamela Bacon, Katja C. Weisel, and Pieter Sonneveld
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Background: RI occurs in ≈ 20%-30% of newly diagnosed MM pts and is associated with poor prognosis (Knudsen et al. Eur J Haematol. 2000; Kyle et al. Mayo Clin Proc. 2003). Data from 2 pivotal trials (MM-002, MM-003) suggested comparable efficacy and tolerability of POM + LoDEX in pts with or without moderate RI (Siegel ASH 2012; Weisel ASCO 2013). However, these trials excluded pts with severe RI. MM-013 (NCT02045017) is a European multicenter, open-label phase 2 study designed to assess the efficacy, safety, and pharmacokinetics of POM + LoDEX in RRMM pts with moderate or severe RI, including those on dialysis. Methods: The trial is enrolling RRMM pts (N = 80) across 3 cohorts: cohort A (moderate RI, estimated glomerular filtration rate [eGFR] ≥ 30 to < 45 mL/min/1.73 m2, n = 33), cohort B (severe RI without dialysis, eGFR < 30 mL/min/1.73 m2, n = 33), and cohort C (severe RI requiring dialysis, n = 14). Pts must have MM-related RI and have received ≥ 1 prior Tx (including lenalidomide). POM 4 mg is administered on days 1-21 of a 28-day cycle and LoDEX 40 mg/day (20 mg for pts aged > 75 yrs) on days 1, 8, 15, and 22 until progressive disease (PD) or unacceptable toxicity. At the time of submission of this abstract, 17 pts terminated Tx; this abstract focuses on tolerability in these pts. Results: This trial is still recruiting; at the time of data cutoff for this abstract, 39 pts were enrolled. Data are included for 17 pts who discontinued Tx. Of all 39 pts, 12 were assigned to cohort A, 18 to cohort B, and 9 to cohort C. The median age of the total population was 72 yrs (range, 52-86 yrs), with 67.7% being male. The median number of prior lines of therapy was 4.0 (3.5 in cohort A, 5.0 in cohort B, and 3.0 in cohort C). This distribution was similar in the 17 pts who discontinued Tx so far (4, 7, and 6 in cohorts A, B, and C, respectively), with a median age of 72 yrs and 58.8% being male. Reasons for discontinuation of Tx were PD (7 pts), adverse events (AEs; 3 pts), death (5 pts: 2 pts due to PD, 2 pts due to infections, 1 pt due to hyperkalemia), and other reasons (2 pts: 1 pt aged 86 yrs with general health problems, 1 pt with increasing RI). Median Tx duration in these pts was 6.9 weeks in cohort A, 12.6 weeks in cohort B, and 12.9 weeks in cohort C. The dosage of POM was reduced to 3 mg in 3 pts (1 patient in each cohort), in all cases due to an AE (thrombocytopenia in 2 pts, pneumonia in 1 pt). However, no further Tx reductions occurred. The most frequent toxicity of any grade in the pts who discontinued was hematologic (82.4% [14 pts]), notably neutropenia in 58.8% (50% in cohort A, 42.9% in cohort B, 83.3% in cohort C), anemia in 52.9% (50% in cohort A, 28.6% in cohort B, 83.3% in cohort C), and thrombocytopenia in 52.9% (75% in cohort A, 14.3% in cohort B, 83.3% in cohort C). Grade 3/4 neutropenia occurred in 47.1%; grade 3/4 thrombocytopenia occurred in 35.3%. Notably, febrile neutropenia was reported in only 1 pt in cohort A. Granulocyte colony-stimulating factor was used in 52.9% of pts. Non-hematologic AEs were less frequent. Infections occurred in 7 pts (41.2%), all of which were pulmonary infections, with the exception of 1 case of nasopharyngitis. Asthenia (23.5%) and fatigue (23.5%) occurred predominantly in cohort C. No thromboembolic events or secondary primary malignancies have been reported to date. Conclusions: These data suggest that the combination of POM and LoDEX can be safely administered in pts with RI. A starting dose of POM 4 mg can be used throughout all stages of RI, and the side effects seen in this population have been previously reported with POM use (ie, mainly hematologic events and infections). Rates of neutropenia and thrombocytopenia are similar to reports in a non-RI population. Dose modifications should be considered in pts who develop neutropenia and thrombocytopenia; in pts showing signs of infections, dose interruptions may be considered. Disclosures Off Label Use: Pomalidomide in MM patients with renal insufficiency.. Dimopoulos:Janssen: Honoraria; Celgene: Honoraria; Janssen-Cilag: Honoraria; Genesis: Honoraria; Onyx: Honoraria; Novartis: Honoraria; Amgen: Honoraria. van de Donk:Janssen Pharmaceuticals: Research Funding; Amgen: Research Funding; Celgene: Research Funding. Gamberi:Celgene Corporation: Membership on an entity's Board of Directors or advisory committees; Mundipharma: Membership on an entity's Board of Directors or advisory committees; Janssen Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Kueenburg:Celgene Corporation: Consultancy, Honoraria. Rosettani:Celgene Corporation: Employment. Collins:Celgene Corporation: Employment. Lersch:Celgene Corporation: Employment. Bacon:Celgene Corporation: Employment, Equity Ownership. Weisel:Noxxon: Consultancy; Celgene: Consultancy, Honoraria, Other: Travel Support, Research Funding; BMS: Consultancy, Honoraria, Other: Travel Support; Novartis: Other: Travel Support; Janssen Pharmaceuticals: Consultancy, Honoraria, Other: Travel Support, Research Funding; Onyx: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Other: Travel Support. Sonneveld:Amgen: Honoraria, Research Funding; Karyopharm: Research Funding; SkylineDx: Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Research Funding; Janssen: Honoraria, Research Funding.
- Published
- 2015
29. Meeting summary: International Symposium and Workshop on Hematopoietic Stem Cells VI, University of Tübingen, Germany, September 14-16, 2006
- Author
-
Katja C. Weisel, Willem E. Fibbe, and Lothar Kanz
- Subjects
Cancer Research ,Genetics ,Animals ,Humans ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Mesenchymal Stem Cell Transplantation ,Molecular Biology ,Embryonic Stem Cells ,Hematopoiesis - Published
- 2007
30. Introduction
- Author
-
DONALD ORLIC, KATJA C. WEISEL, WILLEM E. FIBBE, and LOTHAR KANZ
- Subjects
History and Philosophy of Science ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Published
- 2005
31. Long-Term Coculture of Adult Human Healthy Donor CD34+ Cells Shows Elevation of Telomeres in a Subgroup of Donors Independent of Age, Stem Cell Function and Telomerase Activity
- Author
-
Lothar Kanz, Katja C. Weisel, Malcolm A.S. Moore, and Kaida Wu
- Subjects
Telomerase ,Immunology ,CD34 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Telomere ,Haematopoiesis ,medicine.anatomical_structure ,Cord blood ,medicine ,Bone marrow ,Stem cell ,Progenitor cell - Abstract
Long-term cytokine-supplemented or stromal cocultures of human CD34+ cells, particularly from cord blood (CB), show expansion of hematopoietic progenitors and stem cells. Ultimately, however cultures decline and terminally differentiate. Despite upregulation of telomerase activity in proliferating primitive hematopoietic cells, telomere shortening has generally been reported in long-term cultures of CB, bone marrow or G-CSF mobilized peripheral blood CD34+ cells. In earlier reports, we described a long-term culture of hematopoietic stem cells on a murine OP9 bone marrow stroma cell line transfected with an adenovector expressing thrombopoietin, which allowed an extensive proliferation and self-renewal of CB CD34+ cells for 4–5 months with sustained elevation of telomerase activity and without concomitant significant telomere shortening (Blood, 2004). Here, we evaluated adult healthy donor peripheral blood (PB) CD34+ cells in the same OP9/Tpo coculture system. To determine progenitor and stem cell production, standard CFC and 2ndry cobblestone area-forming cell assays (CAFC assayed at 5 weeks on MS5 stroma) were undertaken weekly with suspension cells. In addition telomere length was measured by telomere restriction fragment (TRF) assay, and telomerase activity by TRAP assay on input CD34+ cells, and weekly on culture suspension cells. Maximum total cell, CFC and CAFC production was seen in the first 4 weeks with up to 80-fold expansion in cell count, up to 4-fold expansion in CFC and up to 13-fold expansion in CAFC. Thereafter a continuous decrease in production of cells, CFC 2ndry CAFC was observed and cultures terminated at week 8. Mean telomere length of input PB CD34+ cells was 9,5 ± 0,5 kbp. After 4 weeks in culture, telomere length remained stable (9,6 ± 0,5 kbp). In 3/6 cultures terminated, cultures showed only a slightly decrease of telomere length compared to the input population (0,55 ± 0,1 kbp loss). However, in 2/6 cultures we could demonstrate an elevation of telomeres (+ 0,3 kbp) independent of a rapid loss of telomerase activity in all cultures during the culture period. Furthermore, the elevation of telomeres did not correlate with an enhanced stem/progenitor cell activity. These data confirm earlier results of granulocyte telomere change in myeloma patients following chemotherapy and tandem transplantation, where 154/193 patients showed an expected loss of telomeres during the treatment period, whereas 39/193 patients had an unexpected elevation of telomeres. We could now show in healthy donors that this phenomenon is independent of bone marrow stress due to chemotherapeutic treatment. In conclusion, we could show that the stromal coculture system with OP9/Tpo is highly effective in stem/progenitor cell expansion not only in CB but also in PB CD34+ cells. It is the first culture system, which allows expansion of hematpoietic cells without significant telomere erosion. We furthermore describe for the first time an age-independent healthy donor population which shows telomerase-independent, significant telomere elevation. Further studies have to demonstrate, if this phenomenon is potentially linked to a higher susceptibility for cancer disease.
- Published
- 2004
32. Wnt1 Overexpression Leads to Enforced Cardiomyogenesis and Inhibition of Hematopoiesis in Murine Embryonic Stem Cells.
- Author
-
Katja C. Weisel, Hans-Georg Kopp, Malcolm A.S. Moore, Lorenz Studer, and Tiziano Barberi
- Subjects
- *
HEMATOPOIESIS , *EMBRYONIC stem cells , *GENE expression , *CELLULAR signal transduction , *CELL differentiation , *EMBRYOLOGY , *LABORATORY mice - Abstract
Recent findings emphasized a critical role for the Wnt signaling pathway during the early steps of embryogenesis, including the development of the hematopoietic system and cardiac development. To date, the role of Wnt in promoting or inhibiting development of both tissues was discussed controversially, dependent on species and time point of expression. Differentiation of embryonic stem cells (ESC) recapitulates early stages of mammalian development. In the present study, we generated murine ESC lines overexpressing Wnt1 (Wnt1 ES). When induced to differentiate toward the cardiomyocytic lineage, Wnt1 ES showed a significant increased ability to generate cardiomyocytes when compared with a control ESC (control ES) line. In addition, Wnt1 ES cells were unable to form hematopoietic cells, whereas development of endothelial cells, a cell type closely associated with blood during embryogenesis, was comparable to control ES. Finally, cardiac differentiation was markedly decreased by the addition of the Wnt antagonist Dkk-1 to the culture medium. These findings suggest that Wnt1 may regulate differentiation of immature mesodermal cells in a tissue-specific manner. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
33. Extended In Vitro Expansion of Adult, Mobilized CD34Cells Without Significant Cell Senescence Using a Stromal Cell Coculture System with Single Cytokine Support.
- Author
-
Katja C. Weisel, Malcolm A.S. Moore, Lothar Kanz, and Robert Möhle
- Subjects
- *
MACROPHAGES , *CYTOKINES , *CELLULAR immunity , *IMMUNOREGULATION - Abstract
The macrophage colony-stimulating factor–deficient bone marrow stromal cell line OP9, derived from osteopetrotic mice, is known to support hematopoietic stem cell (HSC) expansion as well as hematopoietic differentiation of embryonic stem cells. Coculture of HSC in the OP9 system requires cytokine support to achieve significant cell expansion. Recently, we reported extensive expansion without cell senescence of cord blood (CB)-derived HSC cocultured with OP9 stromal cells for more than 18 weeks with a single cytokine support using human thrombopoietin (TPO). In this study, we evaluated the efficiency of the OP9/TPO coculture system to sustain long-term hematopoiesis of adult, granulocyte colony-stimulating factor mobilized human peripheral blood (PB) CD34cells. Maximum cell expansion was attained during the first 4 weeks of coculture. At the same time, the maximum progenitor cell expansion was demonstrated by the production of colony-forming cells and cobblestone area–forming cells. In contrast to the expansion of CB CD34cells, PB CD34cells showed termination of cultures after 8 weeks, independent of the cell expansion rates attained. The evaluation of cell senescence by assessing the telomere length in most cultures showed no relevant telomere shortening, despite rapid decrease in telomerase activity. Interestingly, increases in telomere length were demonstrated. In conclusion, OP9/TPO system provides extensive stem cell expansion without concomitant telomere erosion for both CB and adult CD34cells. Termination of adult CD34cell cocultures seems to be independent of telomere length. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.