109 results on '"Martin Bornhaeuser"'
Search Results
2. Author Reply to Peer Reviews of CD38 promotes hematopoietic stem cell dormancy via c-Fos
- Author
-
Liliia Ibneeva, Sumeet Pal Singh, Anupam Sinha, Sema Elif Eski, Rebekka Wehner, Luise Rupp, Juan Alberto Perez-Valencia, Alexander Gerbaulet, Susanne Reinhardt, Manja Wobus, Malte Bonin, Jaime Sancho, Frances E Lund, Andreas Dahl, Marc Schmitz, Martin Bornhaeuser, Triantafyllos Chavakis, Ben Wielockx, Tatyana Grinenko, and Iryna Kovtun
- Published
- 2023
3. Tandem Duplications of the UBTF gene in Adult AML: A Rare but Recurrent Alteration Associated with Myelodysplasia and Poor Outcome
- Author
-
Julia-Annabell Georgi, Sebastian Stasik, Sven Zukunft, Marita Hartwig, Christoph Röllig, Uta Oelschlaegel, Utz Krug, Tim Sauer, Sebastian Scholl, Andreas Hochhaus, Tim H Brümmendorf, Ralph Naumann, Björn Steffen, Hermann Einsele, Markus Schaich, Andreas Burchert, Andreas Neubauer, Kerstin Schaefer-Eckart, Christoph Schliemann, Stefan W. Krause, Mathias Haenel, Richard Noppeney, Ulrich Kaiser, Claudia D. Baldus, Martin Kaufmann, Carsten Müller-Tidow, Uwe Platzbecker, Wolfgang E. Berdel, Hubert Serve, Gerhard Ehninger, Martin Bornhaeuser, Johannes Schetelig, Frank P. Kroschinsky, and Christian Thiede
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
4. Fast-Track Measurable Residual Disease Detection By Multiparametric Flow Cytometry in Acute Myeloid Leukemia before Allogeneic Hematopoietic Cell Transplantation
- Author
-
Maximilian Alexander Röhnert, Michael Kramer, Jonas Schadt, Philipp Ensel, Christoph Röllig, Johannes Schetelig, Friedrich Stölzel, Martin Bornhaeuser, Uta Oelschlägel, and Malte von Bonin
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
5. Single versus double induction with '7+3' containing 60 versus 90 mg Daunorubicin for newly diagnosed AML: results from the randomized controlled SAL Dauno-double trial [Abstract]
- Author
-
Christoph Röllig, Björn Steffen, Christoph Schliemann, Jan-Henrik Mikesch, Nael Alakel, Regina Herbst, Mathias Haenel, Richard Noppeney, Maher Hanoun, Martin Kaufmann, Zdenek Racil, Kerstin Schäfer-Eckart, Tim Sauer, Andreas Neubauer, Claudia D. Baldus, Jolana Mertova, Edgar Jost, Dirk Niemann, Jan Novak, Stefan W. Krause, Sebastian Scholl, Andreas Hochhaus, Gerhard Held, Tomáš Szotkowski, Christoph Schmid, Andreas Rank, Lars Fransecky, Michael Kramer, Frank Fiebig, Annett Haake, Friedrich Stoelzel, Johannes Schetelig, Jan Moritz Middeke, Uwe Platzbecker, Christian Thiede, Carsten Müller-Tidow, Wolfgang E. Berdel, Hubert Serve, Gerhard Ehninger, Jiří Mayer, and Martin Bornhaeuser
- Subjects
Immunology ,Cell Biology ,Hematology ,ddc:610 ,Biochemistry - Published
- 2022
6. Venetoclax Plus High-Dose Cytarabine and Mitoxantrone As Feasible and Effective Novel Treatment for Relapsed AML: Results of the Phase-I SAL Relax Trial
- Author
-
Christoph Röllig, Lars Fransecky, Maher Hanoun, Björn Steffen, Sabrina Kraus, Christoph Schliemann, Annett Haake, Frank Fiebig, Sven Zukunft, Nael Alakel, Jan Moritz Middeke, Martin Bornhaeuser, Friedrich Stoelzel, Johannes Schetelig, Leo Ruhnke, Michael Kramer, Malte Von Bonin, Maximilian Alexander Röhnert, Uta Oelschlägel, Claudia D. Baldus, Hubert Serve, and Martin Wermke
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
7. Analysis of Patient-Level Data from 3 Cooperative Group Trials Confirms a Survival Advantage for NPM1m Patients Achieving MRD-Negative CR after Intensive Induction
- Author
-
Konstanze Döhner, Hartmut Döhner, Daniela Weber, Silke Kapp-Schwoerer, Amanda Gilkes, Ian Thomas, Sean Johnson, Nicola Potter, Yana Bevan, Jad Othman, Nigel H. Russell, Christoph Röllig, Christian Thiede, Martin Bornhaeuser, Thomas Oellerich, Jenna Elder, Luis A. Carvajal, Zung To, Jorge DiMartino, and Richard Dillon
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
8. Gut Microbiome, Metabolome and Immune Changes in Myelodysplastic Syndromes
- Author
-
Ekaterina Balaian, Nikolai Karcher, Katja Sockel, Uta Oelschlägel, Alexander Funk, Manja Wobus, Maria Uhlig, Matthias W. Groß, Sebastian Zeissig, Martin Bornhaeuser, Triantafyllos Chavakis, and Georg Zeller
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
9. Allogeneic hematopoietic cell transplantation in patients ≤ 60 years with intermediate-risk acute myeloid leukemia in first remission - results of the randomized etal-1 trial
- Author
-
Hubert Serve, Mathias Haenel, Johanna Tischer, Martin Bornhaeuser, Christoph Schliemann, Karsten Spiekermann, Kerstin Schaefer-Eckart, Wolfgang E. Berdel, Lutz P. Mueller, Gesine Bug, Stefan Klein, Georg Lenz, Christoph Schmid, Dietrich W. Beelen, Edgar Jost, Nael Alakel, Markus Pfirrmann, Gerhard Ehninger, Matthias Stelljes, Wolf Roesler, Friedrich Stoelzel, Michael Kramer, Uwe Platzbecker, Christoph Röllig, Johannes Schetelig, Bertram Glass, and Andreas Burchert
- Subjects
Oncology ,medicine.medical_specialty ,Hematopoietic cell ,business.industry ,Immunology ,First remission ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,Transplantation ,Internal medicine ,medicine ,In patient ,business ,Intermediate risk - Abstract
Allogeneic hematopoietic cell transplantation (HCT) offers the highest chance for cure in patients with adverse-risk acute myeloid leukemia (AML) when performed in first remission (CR1). In contrast, patients in CR1 with favorable risk do not seem to benefit from allogeneic HCT due to the inherent risk of transplant-related mortality. Donor vs. no donor comparisons as well as prospective matched-pair analyses have suggested that allogeneic HCT performed in intermediate-risk AML may provide a higher probability of overall survival or relapse-free survival in patients ≤ 60 years of age with an acceptable risk for transplant-related mortality. On the other hand, many intermediate-risk patients relapsing after conventional chemotherapy may be successfully salvaged by allogeneic HCT. The role of allogeneic HCT in cytogenetically defined intermediate-risk AML patients in CR1 was addressed by a prospective randomized trial performed in 16 centers in Germany. Key inclusion criteria were: AML with intermediate-risk cytogenetics, first CR or CRi after conventional induction therapy, age of 18-60 years, and availability of an HLA-matched sibling or unrelated donor. For unrelated donors, a 9 out of 10 HLA allelic match was acceptable except for patients with an NPM1 mutation, for whom full 10/10 allele matching was required. Randomization was stratified according to age (< 40 vs. 40-60), NPM1/FLT3, and CEBP-alpha mutational status and unrelated vs. related donor availability. Endpoints included overall-survival as primary outcome and relapse-free survival (RFS), cumulative incidence of relapse, treatment-related mortality, and quality of life measured according to the short form (36) health status. From 2010 - 2018, 143 patients in CR1 were randomized into Arm A (n=76, allogeneic HCT) and Arm B (n=67, conventional consolidation and allo-HCT only in case of relapse). In July 2018, the trial was stopped prematurely due to slow accrual (143 out of 356 pts. randomized). Median age of the trial cohort was 51 years (range, 19-60), with 42% exhibiting an NPM1 and 25% a FLT3 mutation. A normal karyotype was reported in 84% of the included patients. All mentioned characteristics did not differ between both treatment arms. Sibling donors were available for 44 (31% of patients), matched unrelated donors for 99 (69%) patients. According to the intent-to-treat analysis, the probability of survival at 2 years was 71% (95% CI 60-81%) and 84% (95% CI 73-92%) in Arm A (Transplant) and Arm B (conventional consolidation), respectively (p=0.120, Figure 1A). RFS after allogeneic HCT was 69% (95% CI 57-80%) compared to 41% (95% CI 29-54%) after conventional consolidation (p=0.001, Figure 1B). Primary allogeneic HCT reduced the cumulative incidence of relapse at 2 years from 57% [95%-CI 46-71%] after conventional consolidation to 20% [95%-CI 13-31%] after HCT (p SF (36) scoring suggested a trend towards a lower physical functioning throughout the first 3 months after randomization in the primary HCT arm. No significant differences in vitality, mental health, social and emotional functioning could be documented between both treatment arms. In summary, the results of this first prospective randomized trial did not show that allogeneic HCT performed immediately after achievement of CR1 in patients with cytogenetically defined intermediate-risk AML ≤ 60 years of age conveys an overall survival advantage. However, allogeneic HCT in CR1 significantly reduced the relapse risk and was not associated with relevant impairments in quality of life. Although the limited statistical power of the trial does not allow definitive conclusions, delayed allogeneic transplantation seems to be a potential treatment algorithm in CR1 intermediate-risk AML with an available donor. Figure 1 Figure 1. Disclosures Schliemann: Jazz Pharmaceuticals: Consultancy, Research Funding; Roche: Consultancy; Philogen S.p.A.: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy; AstraZeneca: Consultancy; Boehringer-Ingelheim: Research Funding; Novartis: Consultancy; Abbvie: Consultancy, Other: travel grants; Astellas: Consultancy; BMS: Consultancy, Other: travel grants. Schetelig: Roche: Honoraria, Other: lecture fees; Novartis: Honoraria, Other: lecture fees; BMS: Honoraria, Other: lecture fees; Abbvie: Honoraria, Other: lecture fees; AstraZeneca: Honoraria, Other: lecture fees; Gilead: Honoraria, Other: lecture fees; Janssen: Honoraria, Other: lecture fees . Glass: Riemser: Research Funding; Roche: Consultancy, Research Funding, Speakers Bureau; Kite: Consultancy; BMS: Consultancy; Novartis: Consultancy; Helios Klinik Berlin-Buch: Current Employment. Platzbecker: Janssen: Honoraria; Celgene/BMS: Honoraria; Novartis: Honoraria; Takeda: Honoraria; Geron: Honoraria; AbbVie: Honoraria. Burchert: Novartis: Honoraria, Research Funding; AOP Orphan: Honoraria, Research Funding; Pfizer: Honoraria; Incyte: Honoraria; Gilead: Honoraria; BMS: Honoraria. Haenel: Jazz: Consultancy, Honoraria; GSK: Consultancy; Bayer Vital: Honoraria; Takeda: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Amgen: Consultancy; Celgene: Consultancy, Honoraria. Mueller: Amgen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Other: Travel Support; CTI: Membership on an entity's Board of Directors or advisory committees; Gentium: Other: Travel Support; Gilead: Other: Travel Support; Janssen: Other: Travel Support; Novartis: Other: Travel Support; Pfizer: Other: Travel Support; Sanofi: Other: Travel Support. Berdel: Philogen S.p.A.: Consultancy, Current equity holder in publicly-traded company, Honoraria, Membership on an entity's Board of Directors or advisory committees. Stelljes: Novartis: Consultancy, Speakers Bureau; MSD: Consultancy, Speakers Bureau; Pfizer: Consultancy, Research Funding, Speakers Bureau; Kite/Gilead: Consultancy, Speakers Bureau; Celgene/BMS: Consultancy, Speakers Bureau; Medac: Speakers Bureau; Amgen: Consultancy, Speakers Bureau.
- Published
- 2021
10. Targeting of radioactive platinum-bisphosphonate anticancer drugs to bone of high metabolic activity
- Author
-
Otto C. Boerman, Sandra Heskamp, Uwe Karst, Nicola Margiotta, Michele Iafisco, Michael Brand, Martin Bornhaeuser, Karlijn Codee-van der Schilden, Nandini Asokan, Barbara Crone, Robin A. Nadar, Sander C.G. Leeuwenburgh, Kambiz Farbod, Alessandra Curci, Jeroen J.J.P. van den Beucken, and Lukas Schlatt
- Subjects
0301 basic medicine ,Male ,Magnetic Resonance Spectroscopy ,theranostic ,medicine.medical_treatment ,lcsh:Medicine ,Platinum Compounds ,Pharmacology ,Mice ,0302 clinical medicine ,Drug Delivery Systems ,bone-targeting ,Bone cancer ,lcsh:Science ,Zebrafish ,Multidisciplinary ,Molecular medicine ,Diphosphonates ,Chemistry ,Reconstructive and regenerative medicine Radboud Institute for Molecular Life Sciences [Radboudumc 10] ,030220 oncology & carcinogenesis ,Drug delivery ,Toxicity ,Injections, Intravenous ,Systemic administration ,chemistry.chemical_element ,Antineoplastic Agents ,Bone Neoplasms ,Calcium ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Article ,Bone and Bones ,03 medical and health sciences ,Medical research ,In vivo ,medicine ,Bisphosphonate ,Animals ,Radioisotopes ,Tibia ,Bone metastases ,lcsh:R ,In vitro ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Q ,195mPlatinum ,Platinum ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] - Abstract
Platinum-based chemotherapeutics exhibit excellent antitumor properties. However, these drugs cause severe side effects including toxicity, drug resistance, and lack of tumor selectivity. Tumor-targeted drug delivery has demonstrated great potential to overcome these drawbacks. Herein, we aimed to design radioactive bisphosphonate-functionalized platinum (195mPt-BP) complexes to confirm preferential accumulation of these Pt-based drugs in metabolically active bone. In vitro NMR studies revealed that release of Pt from Pt BP complexes increased with decreasing pH. Upon systemic administration to mice, Pt-BP exhibited a 4.5-fold higher affinity to bone compared to platinum complexes lacking the bone-seeking bisphosphonate moiety. These Pt-BP complexes formed less Pt-DNA adducts compared to bisphosphonate-free platinum complexes, indicating that in vivo release of Pt from Pt-BP complexes proceeded relatively slow. Subsequently, radioactive 195mPt-BP complexes were synthesized using 195mPt(NO3)2(en) as precursor and injected intravenously into mice. Specific accumulation of 195mPt-BP was observed at skeletal sites with high metabolic activity using micro-SPECT/CT imaging. Furthermore, laser ablation-ICP-MS imaging of proximal tibia sections confirmed that 195mPt BP co-localized with calcium in the trabeculae of mice tibia.
- Published
- 2020
11. Intensified Cytarabine Dose during Consolidation Therapy in AML Patients Under 65 Years Is Not Associated with Survival Benefit
- Author
-
Maher Hanoun, Johannes Kullmer, Christoph Schliemann, Andreas Neubauer, Mathias Haenel, Ulrich Kaiser, Sabrina Kraus, Gerhard Held, Hermann Einsele, Kerstin Schäfer-Eckhard, Tim H. Brümmendorf, Carsten Mueller-Tidow, Claudia D. Baldus, Christoph Röllig, Sebastian Scholl, Martin Goerner, Dirk Niemann, Michael Kramer, Hubert Serve, Uwe Platzbecker, Björn Steffen, Hans Christian Reinhardt, Martin Bornhaeuser, Stefan W. Krause, Tim Sauer, Martin Kaufmann, Ruth Seggewiss-Bernhard, Lars Fransecky, Leo Ruhnke, and Edgar Jost
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Consolidation therapy ,Survival benefit ,Internal medicine ,Cytarabine ,Medicine ,business ,medicine.drug - Abstract
Background: Acute myeloid leukemia (AML) is characterized by a high relapse rate, indicating insufficient clearance of leukemia-initiating cells. Depending on genetic risk stratification, consolidating chemotherapy proves to significantly reduce the risk of relapse. In particular, in younger AML patients higher dosage of cytarabine appears to improve long-term outcome, while there is no apparent benefit of multiagent combination, compared to cytarabine monotherapy. However, to this end the optimal dosage of single agent cytarabine in consolidation therapy after 7+3 remission induction remains elusive. Methods: Here, we retrospectively assessed the impact of different dosages of cytarabine consolidation on outcome in a large real-world data set from the German Study Alliance Leukemia-Acute Myeloid Leukemia (SAL-AML) registry. Patients below 65 years of age, registered between April 2005 and September 2020 with non-acute promyelocytic leukemia, who attained complete remission after intensive induction and received at least one consolidation cycle with intermediate (IDAC) or high dose cytarabine (HiDAC) were selected. To account for differences in patient and disease characteristics between both groups, the average treatment effect was estimated by propensity score weighting. Results: 642 patients received HiDAC consolidation with a median dosage of 5794.88 (IQR, 4745.48-5971.56) mg/m 2/d with a median number of 3 cycles (IQR, 2-3), whereas 178 patients received IDAC consolidation with 1946.16 (IQR, 1869.51-2469.15) mg/m 2/d with a median of 2 cycles (IQR, 1-3). IDAC-treated patients showed in average a higher age (median (IQR) 58.5 (49-62) years vs. 50 (41-56) years) and more comorbidities with 43.8% having an HCT-CI score of 2-4, compared to 22.3% among HiDAC-treated patients. Alongside, significantly more secondary (5.1% vs. 3.1%) and therapy-related (12.4% vs. 4.1%) AML as well as more adverse (14.5% vs. 6.5%) and less favorable (40.6% vs. 56%) genetic risk features according to ELN 2017 risk classification were found among IDAC-treated patients. After propensity score weighting for differences in patient and disease characteristics, overall survival after 5 years was comparable between HiDAC-treated (71.1 %) and IDAC-treated (67.7%) patients. Moreover, no significant differences in relapse-free survival were observed after 5 years (47.4 vs. 45.2%). Notably, more patients treated with IDAC received allogeneic stem cell transplantation in first remission (37.6 vs. 19.8%) while significantly more HiDAC-treated patients underwent allogeneic stem cell transplantation in relapse (30.8 vs. 20.2%). Censoring for allogeneic stem cell transplantation in first remission revealed no significant survival difference with regard to cytarabine dosage. Considering only ELN favorable risk AML patients, there was no difference in 5-years overall (80.5% vs. 83.9%) nor relapse-free (57.7% vs. 56.8%) survival. Of note, significantly more patients treated with HiDAC suffered from ≥3 CTCAE infectious complications (56.7 vs. 44.1%), which was more striking in patients above 50 years of age. The rate of other ≥3 CTCAE non-hematological toxicities and secondary malignancies was comparable in both treatment groups. Conclusion: This retrospective analysis suggests no significant benefit of high dose cytarabine compared to intermediate dosages in consolidation for AML patients under 65 years of age, independent of ELN risk group. Disclosures Krause: Siemens: Research Funding; Takeda: Honoraria; Pfizer: Honoraria; art-tempi: Honoraria; Kosmas: Honoraria; Gilead: Other: travel support; Abbvie: Other: travel support. Schliemann: Philogen S.p.A.: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Other: travel grants; Astellas: Consultancy; AstraZeneca: Consultancy; Boehringer-Ingelheim: Research Funding; BMS: Consultancy, Other: travel grants; Jazz Pharmaceuticals: Consultancy, Research Funding; Novartis: Consultancy; Roche: Consultancy; Pfizer: Consultancy. Haenel: Jazz: Consultancy, Honoraria; GSK: Consultancy; Bayer Vital: Honoraria; Takeda: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Amgen: Consultancy; Celgene: Consultancy, Honoraria. Brummendorf: Takepart Media: Honoraria; Repeat Diagnostics: Research Funding; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Patents & Royalties, Research Funding; Janssen: Honoraria; Bristol Myers: Research Funding. Fransecky: Abbvie: Honoraria, Research Funding; Medac: Honoraria; Novartis: Honoraria; Amgen: Honoraria; Takeda: Honoraria. Einsele: Janssen, Celgene/BMS, Amgen, GSK, Sanofi: Consultancy, Honoraria, Research Funding. Held: MSD: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Research Funding; Roche: Research Funding; Acortech Biopharma: Research Funding; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees. Platzbecker: Janssen: Honoraria; Celgene/BMS: Honoraria; AbbVie: Honoraria; Geron: Honoraria; Takeda: Honoraria; Novartis: Honoraria. Baldus: Amgen: Honoraria; Celgene/BMS: Honoraria; Novartis: Honoraria; Jazz: Honoraria. Mueller-Tidow: Janssen Cilag: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Bioline: Consultancy, Research Funding.
- Published
- 2021
12. Prediction of Complete Remission and Survival in Acute Myeloid Leukemia Using Supervised Machine Learning
- Author
-
Claudia D. Baldus, Tim Sauer, Hubert Serve, Carsten Müller-Tidow, Maher Hanoun, Martin Bornhaeuser, Christian Thiede, Karsten Wendt, Martin Kaufmann, Christoph Schliemann, Kerstin Schaefer-Eckart, Michael Kramer, Julia-Annabell Georgi, Stefan W. Krause, Peter Heisig, Frank Kroschinsky, Sebastian Stasik, Johannes Schetelig, Mathias Haenel, Uwe Platzbecker, Christoph Röllig, Jan Moritz Middeke, and Jan-Niklas Eckardt
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Immunology ,medicine ,Complete remission ,Myeloid leukemia ,Cell Biology ,Hematology ,business ,Biochemistry - Abstract
Achievement of complete remission (CR) signifies a crucial milestone in the therapy of acute myeloid leukemia (AML) while refractory disease is associated with dismal outcomes. Hence, accurately identifying patients at risk is essential to tailor treatment concepts individually to disease biology. Machine Learning (ML) is a branch of computer science that can process large data sets for a plethora of purposes. The underlying mechanism does not necessarily begin with a manually drafted hypothesis model. Rather the ML algorithms can detect patterns in pre-processed data and derive abstract information. We used ML to predict CR and 2-year overall survival (OS) in a large multi-center cohort of 1383 AML patients who received intensive induction therapy using clinical, laboratory, cytogenetic and molecular genetic data. To enable a customizable and reusable technological approach and achieve optimal results, we designed a data-driven platform with an embedded, automated ML pipeline integrating state-of-the-art software technology for data management and ML models. The platform consists of five scalable modules for data import and modelling, data transformation, model refinement, machine learning algorithms, feature support and performance feedback that are executed in an iterative manner to approach step-wisely the optimal configuration. To reduce dimensionality and the the risk of overfitting, dynamic feature selection was used, i.e. features were selected according to their support by feature selection algorithms. To be included in an ML model, a feature had to pass a pre-determined threshold of overall predictive power determined by summing the normalized scores of the feature selection algorithms. Features below the threshold were automatically excluded from the ML models for the respective iteration. In that way, features of high redundancy or low entropy were automatically filtered out. Our classification algorithms were completely agnostic of pre-existing risk classifications and autonomously selected predictive features both including established markers of favorable or adverse risk as well as identifying markers of so-far controversial relevance. De novo AML, extramedullary AML, double-mutated (dm) CEBPA, mutations of CEBPA-bZIP, NPM1, FLT3-ITD, ASXL1, RUNX1, SF3B1, IKZF1, TP53, U2AF1, t(8;21), inv(16)/t(16;16), del5/del5q, del17, normal or complex karyotypes, age and hemoglobin at initial diagnosis were statistically significant markers predictive of CR while t(8;21), del5/del5q, inv(16)/t(16;16), del17, dm CEBPA, CEBPA-bZIP, NPM1, FLT3-ITD , DNMT3A, SF3B1, U2AF1, TP53, age, white blood cell count, peripheral blast count, serum LDH and Hb at initial diagnosis as well as extramedullary manifestations were predictive for 2-year OS. For prediction of CR and 2-year OS, AUROCs ranged between 0.77 - 0.86 and 0.63 - 0.74, respectively. We provide a method to automatically select predictive features from different data types, cope with gaps and redundancies, apply and optimize different ML models, and evaluate optimal configurations in a scalable and reusable ML platform. In a proof-of-concept manner, our algorithms utilize both established markers of favorable or adverse risk and also provide further evidence for the roles of U2AF1, IKZF1, SF3B1, DNMT3A and bZIP mutations of CEBPA in AML risk prediction. Our study serves as a fundament for prospective validation and data-driven ML-guided risk assessment in AML at initial diagnosis for the individual patient. Image caption: Patient features were automatically selected by machine learning to predict complete remission (CR) and 2-year overall survival (OS) after intensive induction therapy. Based on a continuous feature support metric with a predefined cut-off of 0.5 (determined by optimal classification performance), 27 and 25 features were automatically selected for prediction of CR (A) and 2-year OS (C), respectively. For each of these features predicted by machine learning, odds ratios and 95% confidence intervals (CI) were calculated for CR (B) and 2 year OS (D). BMB: bone marrow blast count; FLT3h/low: FLT3-ITD ratio, h=high>0.5; Hb: hemoglobin; karyotype, c: complex aberrant karyotype (≥ 3 aberrations); karyotype, n: normal karyotype (no aberrations); LDH: lactate dehydrogenase; PBB: peripheral blood blast count; PLT: platelet count; WBC: white blood cell count. Figure 1 Figure 1. Disclosures Schetelig: Roche: Honoraria, Other: lecture fees; Novartis: Honoraria, Other: lecture fees; BMS: Honoraria, Other: lecture fees; Abbvie: Honoraria, Other: lecture fees; AstraZeneca: Honoraria, Other: lecture fees; Gilead: Honoraria, Other: lecture fees; Janssen: Honoraria, Other: lecture fees . Platzbecker: Janssen: Honoraria; Celgene/BMS: Honoraria; AbbVie: Honoraria; Novartis: Honoraria; Takeda: Honoraria; Geron: Honoraria. Müller-Tidow: Pfizer: Research Funding; Janssen: Consultancy, Research Funding; Bioline: Research Funding. Baldus: Celgene/BMS: Honoraria; Amgen: Honoraria; Novartis: Honoraria; Jazz: Honoraria. Krause: Siemens: Research Funding; Takeda: Honoraria; Pfizer: Honoraria; art-tempi: Honoraria; Kosmas: Honoraria; Gilead: Other: travel support; Abbvie: Other: travel support. Haenel: Bayer Vital: Honoraria; Jazz: Consultancy, Honoraria; GSK: Consultancy; Takeda: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Amgen: Consultancy; Celgene: Consultancy, Honoraria. Schliemann: Philogen S.p.A.: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Other: travel grants; Astellas: Consultancy; AstraZeneca: Consultancy; Boehringer-Ingelheim: Research Funding; BMS: Consultancy, Other: travel grants; Jazz Pharmaceuticals: Consultancy, Research Funding; Novartis: Consultancy; Roche: Consultancy; Pfizer: Consultancy. Middeke: Roche: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Jazz: Consultancy; Astellas: Consultancy, Honoraria; Sanofi: Honoraria, Research Funding; Novartis: Consultancy; Gilead: Consultancy; Glycostem: Consultancy; UCB: Honoraria.
- Published
- 2021
13. Trends in Allogeneic Stem Cell Transplantation for Myelofibrosis in Europe between 1995-2018: An EBMT Retrospective Analysis
- Author
-
Patrice Chevallier, Nicolaus Kröger, Igor Wolfgang Blau, Uwe Platzbecker, Dragana Stamatovic, Marie Robin, Dietrich W. Beelen, D. J. Eikema, Joaquin Martinez-Lopez, Ibrahim Yakoub-Agha, Micha Srour, Linda Koster, Patrick Hayden, Jakob Passweg, Jan J. Cornelissen, Donal P. McLornan, Emanuele Angelucci, Tomasz Czerw, Martin Bornhaeuser, Liesbeth C. de Wreede, Jürgen Finke, Grant McQuaker, Riitta Niittyvuopio, Antonin Vitek, and Juan Carlos Hernandez Boluda
- Subjects
medicine.medical_specialty ,Karnofsky Performance Status ,Marrow transplantation ,business.industry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Transplantation ,Family medicine ,Retrospective analysis ,medicine ,In patient ,business ,Bristol-Myers - Abstract
Aim: Dynamic assessment of trends over time in patient- and transplant-specific characteristics and outcomes for patients undergoing 1st allogeneic haematopoietic cell transplant (allo-HCT) for Myelofibrosis (MF). Methods and Results: A total of 4142 MF patients were analysed who underwent allo-HCT between 1995-2018 (24-year period) across 278 centres based on data reported to the European Society for Blood and Marrow Transplantation. For analysis, 4 cohorts were considered based on year of allo-HCT: 60 years accounted for 8.7% of adults undergoing allo-HCT whereas for 2015-2018 this was 47%. Over time, increasing number of patients with a Karnofsky performance status (KPS) Conclusions: Despite a marked increase over this 24-year period in recipient age, RIC regimen utilisation and use of both URD and MMRD, this comprehensive analysis demonstrates stable OS and EFS rates. However, rates of GVHD have decreased over time, in particular extensive cGVHD. Further work is required to improve both the considerable NRM and relapse rates which remain significant. Disclosures McLornan: JAZZ PHARMA: Honoraria, Speakers Bureau; CELGENE: Honoraria, Speakers Bureau; NOVARTIS: Honoraria, Speakers Bureau. Platzbecker:Amgen: Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Geron: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding. Chevallier:Incyte Corporation: Honoraria. Martínez-Lopez:Altum, Hosea: Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; Vivia Biotech: Honoraria; Amgen: Speakers Bureau; Takeda: Speakers Bureau; Roche: Speakers Bureau; Janssen: Speakers Bureau; Incyte: Research Funding, Speakers Bureau; Bristol Myers Squibb: Research Funding, Speakers Bureau; Novartis: Research Funding. Yakoub-Agha:Celgene: Honoraria; Jazz Pharmaceuticals: Honoraria; Novartis: Honoraria; Gilead/Kite: Honoraria, Other: travel support; Janssen: Honoraria.
- Published
- 2020
14. Reconstruction of human AML reveals stem cell origin and therapeutic targets for treatment resistant CD34(-/Lo) MLL-rearranged leukemia
- Author
-
Bernd B. Zeisig, Anskar Y.H. Leung, Boban Stanojevic, Chi Wai Eric So, Malte von Bonin, Chiou Tsun Tsai, Magdalena Zarowiecki, Tsz Kan Fung, Jan Zuna, Boris Lenhard, Suming Huang, Ghulam J. Mufti, Martin Bornhaeuser, Claire Lynn, Marketa Zaliova, Huacheng Luo, and Wellcome Trust
- Subjects
Severe combined immunodeficiency ,Science & Technology ,Immunology ,CD34 ,Myeloid leukemia ,1103 Clinical Sciences ,Cell Biology ,Hematology ,Biology ,CD38 ,medicine.disease ,Biochemistry ,Leukemia ,Haematopoiesis ,hemic and lymphatic diseases ,medicine ,Cancer research ,1114 Paediatrics and Reproductive Medicine ,Progenitor cell ,Stem cell ,neoplasms ,Life Sciences & Biomedicine ,1102 Cardiorespiratory Medicine and Haematology - Abstract
Identification of the origins of acute myeloid leukemia (AML) stem cells has been a holy grail for a better understanding of the developmental biology of the disease and the design of effective treatments. Studies using primary AML samples on patient-derived xenograft models identified AML stem cells in multiple different CD34/CD38 cellular fractions, which shared similar immunophenotypes of hematopoietic stem/progenitor cells including hematopoietic stem cells (HSCs), lymphoid-primed multipotent progenitors (LMPPs) and granulocyte-macrophage progenitors (GMPs). However inference of cells-of-origin based on the retrospective approach has major limitations as the reported HSC/LMPP/GMP-like AML stem cells are phenotypically different from their normal counterparts; and AML stem cells identified in late developmental stages do not necessarily maintain the same immunophenotypes of the disease initiating (pre-leukemic) cells, which can retain a relatively normal differentiation potential, and only their descendants acquire additional events becoming AML stem cells. While prospective disease modelling using mouse cells has provided unique insights into the potential origins of AML stem cells, human and mouse cells have different transformation requirements, distinct telomere biology, and a significant degree divergence of transcriptional regulation, chromatin state and gene regulatory networks that can profoundly affect their transformation potential and associated cancer biology. Therefore we reason that deconstruction of AML stem cell hierarchy from primary human samples followed by reconstruction of the corresponding human disease using candidate cell populations will give novel insights into this issue. Given that AML is a highly heterogeneous disease, our study initially focused on genetically well-defined MLL-rearranged AML frequently found in both infant and adult leukemia. By analyzing primary human AML patient samples, the current study unexpectedly revealed that AML stem cells driven by MLL fusions almost exclusively resided in immunophenotypically mature CD34-/loCD38+ compartments, as demonstrated by in vitro long-term culture initiating cells and in vivo limiting dilution xeno-transplantation assays into immunodeficient NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice. Using highly purified populations of hematopoietic stem/progenitor cells from normal human umbilical cord blood (UCB) to reconstruct of the human disease, we found that only human HSCs and common myeloid progenitors (CMPs), but not developmentally branched LMPPs and GMPs, could be disease initiating cells, transformed by MLL-fusions and induced leukemia in vivo, suggesting a key difference between human and mouse leukemia. As revealed by RNA-sequencing, LMPPs and GMPs failed to activate stem cell transcriptional programs and genes essential for MLL-leukemia including MEIS1 and HMGA2 in the early phase of transformation. MLL-fusions transformed HSCs and CMPs were immunophenotypically indistinguishable to leukemic cells of human MLL-AML patients, and also had an enrichment of AML stem cells in CD34-/loCD38+ compartment. Using machine learning, a specific gene signature could stratify patients into HSCs- and CMPs-derived AML, in which HSCs-derived AML showed a significantly poorer prognosis. To further investigate the biology of HSCs/CMPs-derived MLL-AML related to treatment responses, we subjected HSC/CMP MLL-AML leukemia with chemotherapeutic drugs and inhibitor of Bromodomain and Extra-Terminal motif (BET) currently used in AML trial. Interestingly, CMP-derived MLL-AML was treatment sensitive and PDX models transplanted with CMP-like AML samples could be largely cured by chemotherapy or BET inhibitor targeted therapy. In contrast, human HSCs-derived AML was highly resistant to the treatments. Strikingly, shRNA-mediated knockdown or pharmacological inhibition by fidaxomicin targeting ATP-binding cassette (ABC) transporters, ABCC3 that is highly expressed in HSCs-derived MLL-AML could re-sensitize the cells to the current chemotherapy. Together, the current study not only for the first time functionally identifies the origin of human MLL-AML stem cells, but also provides a new actionable venue for overcoming stem cells-associated treatment resistance by repositioning an anti-diarrhea drug, fidaxomicin currently available in the clinics. Disclosures Mufti: Celgene Corporation: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Cellectis: Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2019
15. Targeting the FMS-like Tyrosin Kinase 3 with the Unicar System: Preclinical Comparison of Murine and Humanized Single-Chain Variable Fragment-Based Targeting Modules
- Author
-
Frederick Fasslrinner, Marc Schmitz, Liliana Raquel Loureiro, Martin Bornhaeuser, Stefanie Koristka, Claudia Arndt, Anja Feldmann, Gundram Jung, and Michael Bachmann
- Subjects
Myeloid ,business.industry ,T cell ,Immunology ,CD33 ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,Epitope ,Chimeric antigen receptor ,medicine.anatomical_structure ,Antigen ,hemic and lymphatic diseases ,medicine ,Cancer research ,business ,B cell - Abstract
Clinical translation of chimeric antigen receptor (CAR) T cell therapy in myeloid malignancies is progressing slowly compared to its success in treatment of B cell malignancies. Clinical experiences with CAR T cell therapies against the currently investigated tumor-associated antigens (TAA) (e.g. CD33, CD123 and FMS-like tyrosine kinase 3 (FLT3)) were discouraging and severe side effects occurred (cytokine release syndrome, neurotoxicity and myeloid aplasia) (Hoffmann et al. Journal of Clinical Medicine 2019). Probably targeting a single TAA is insufficient to treat high risk myeloid malignancies with CAR T cell therapies. Therefore, combined targeting of two or even more TAAs seems to be a promising approach. In order to implement such a multiple tumor targeting strategy, we developed a modular CAR T cell system termed UniCAR. The system consists of a universal CAR (UniCAR) directed against the La peptide epitope E5B9 combined with single-chain variable fragment (scFv) -based target modules (TM). In contrast to conventional CARs, anti-tumor activity of UniCAR T cells is only turned on in the presence of the TMs. Thus, this approach will allow UniCAR T cell control due to the short half-life of the TM and therefore has a favorable safety profile. Furthermore, different TMs against several TAAs can be administered both sequentially or in parallel to increase the anti-tumor efficacy or face disease relapse due to antigen escape mechanisms. In the field of myeloid malignancies our group developed retargeting strategies against the TAAs CD33 and CD123 (Cartellieri et al. Blood Cancer Journal 2016). In addition, we have developed a new TM for the UniCAR system that is directed against the TAA FLT3. FLT3 is highly expressed on acute myeloid leukemia (AML) cells and also present on CD123low AML samples (Riccioni et al. British Journal of Haematology 2011). The novel FLT3 TM was constructed by fusion of the variable domain of the heavy and the light chain of the murine anti-FLT3 monoclonal antibody (4G8) to the E5B9 UniCAR epitope. In light of a potential clinical application, we in parallel generated a humanized FLT3 TM to further decrease its immunogenicity. Both FLT3 TMs were tested in vitro against different AML cell lines, by using flow cytometry based killing assays as described elsewhere (Fasslrinner et al. British Journal of Haematology 2019). The functionality of the FLT3 TMs in vitro was highly effective. Both FLT3 TMs were able to redirect UniCAR T cells for AML cell lysis already in the picomolar range and were moreover comparable effective than the previously developed CD123 TM. Thus, humanization of the FLT3 TM did not lead to a decrease in anti-tumor efficacy. In summary, we could show that both the novel murine FLT3 TM and the humanized counterpart redirected UniCAR T cells and induced highly effective elimination of AML cells in vitro. Thus, the flexible application of the FLT3-based UniCAR system seems to be a promising tool for cell-based AML therapy alone or even in combination with other AML-specific TMs (e.g. CD33, CD123). Disclosures Koristka: Intellia Therapeutics: Employment. Jung:Synimmune: Other: shareholder interest. Bachmann:GEMoaB Monoclonals: Equity Ownership, Patents & Royalties.
- Published
- 2019
16. Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation with Venetoclax, Hypomethylating Agents and DLI - a Retrospective Multi Center Study
- Author
-
Edgar Jost, Oliver Kriege, Thomas Schroeder, Lambros Kordelas, Daniela Heidenreich, Christoph Schmid, Lutz P. Mueller, Judith Schaffrath, Gesine Bug, Daniel Wolff, Matthias Hoepting, Andreas Hausmann, Sabine Dressler, Christina Rautenberg, Jennifer Kaivers, Martin Bornhaeuser, Guido Kobbe, Eva-Maria Wagner-Drouet, Stefan Klein, Salem Ajib, Esther Schuler, Martina Crysandt, and Klaus Hirschbühl
- Subjects
Oncology ,medicine.medical_specialty ,Venetoclax ,business.industry ,medicine.medical_treatment ,Immunology ,Azacitidine ,Medizin ,Decitabine ,Salvage therapy ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,Tumor lysis syndrome ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,business ,Lenalidomide ,medicine.drug - Abstract
Introduction: The most common cause of treatment failure after allogeneic hematopoetic stem cell transplantation (aHSCT) is relapse. The combination of venetoclax and the hypomethylating agents (HMA) azacitidine (AZA) or decitabine (DAC) have shown promising efficacy in elderly patients with AML. We here present clinical data on 32 patients, who were treated with an HMA/venetoclax combination therapy (HMAClax) for relapse of a myeloid malignancy after aHSCT, collected retrospectively from 11 German centers. Results: Sixteen patients (50%) were male, median age was 54 years (30.8-71.5). Diagnoses at aHSCT were 25 AML (17 primary, 8 emerging from MDS, CMML or OMF), 5 MDS, 1 CMML and one atypical CML. Twenty six patients were treated for relapse after their 1st and 6 after their 2nd aHSCT. Only 9 patients were in CR at aHSCT. The majority received a graft of a matched unrelated donor (21), 4 from an HLA-identical sibling and 7 from a haploidentical relative. Conditioning was myeloablative in 15 and RIC in 17patients. Median time from aHSCT to last relapse was 5.7ms (1.1-67.8). Five patients had molecular (MR) and 23 had hematologic relapses (HR), 4 patients had extramedullary manifestations 3 with concurrent HR and 1 with MR. Twenty-one patients were treated for 1st and 5 for 2nd relapse after 1st aHSCT. Four patients were treated for 1st and 2 for 2nd relapse after 2nd aHSCT. HMAClax was first line therapy for relapse in 8, 2nd line in 22, 3rd line 1 and 4th line in 1 patient. In 21 patients relapse had been refractory to HMA (+/- DLI, +/- lenalidomide). Median time from relapse to HMAClax was 1.8 ms (0.3-42.9). Twelve patients received AZA and 19 DAC with venetoclax. One patient was switched from AZAClax to DAClax because of rising MRD after 6 cycles and back to AZAClax after another 7 cycles. Six patients received DLI. Median number of cycles was 2 (1-15). Six patients are still on therapy. In total 75 cycles were given. Three patients had non-fatal tumor lysis syndrome. All but one patient had grade 3/4 neutropenia and 25 patients (78%) had grade 3/4 thrombocytopenia. Hospital admission for grade 3/4 infections was necessary in 23 patients (72%), 5 of these infections (22%) were fatal. Overall response rate was 43% (12/28, 2 CR MRD-, 4 CR, 2 CRi, 3 PR, 1 MLFS). Two patients died of infection before first response evaluation and in another 2 response has not been evaluated yet. ORR for patients who received first line HMAClax was 80% (4/5) and 35% (8/23) for salvage treatment. Three of 5 patients with MR reached CR, 2 received HMAClax first line. Time to best response was 1.2ms (0.7-3.8). Six patients lost best response after 1ms (0.4-3.3.) 2 underwent second transplant in remission, 4 have ongoing responses (0.4, 0.7, 3.1 and 8.8ms at last follow up). On July 25th 2019, median follow up was 3.3 ms (0.9-17.3), 20 patients (63%) had died and 12 were alive. Six were continuing HMAClax. One patient developed cGvHD and 4 underwent second aHSCT (2 in remission). Estimated median overall survival was 3.7ms (CI 2.9-4.7). Four responders are continuing treatment with HMAClax. Patients, who responded had an estimated OS of 11.1ms (2 underwent second aHSCT in remission). Median survival of patients with HMAClax first line therapy was 5.8ms and of patients with HMAClax salvage therapy 3.7ms. Conclusion: For patients relapsing after aHSCT, venetoclax plus AZA or DAC seems to be an effective, but also highly hematotoxic therapy. Responses occurred fast and were more frequently seen during 1st line treatment for relapse. Duration of response was short, especially in patients receiving HMAClax as 2nd, 3rd or 4th line therapy. Therefore HMAClax should be explored as 1st line therapy for relapse after aHSCT in combination with DLI or as a bridge to 2nd transplant. Disclosures Schuler: Celgene: Other: travel grants; Novartis: Honoraria, Other: travel grants; Alexion: Other: travel grants. Bug:Gilead Sciences: Membership on an entity's Board of Directors or advisory committees, Other: Travel grants; Hexal: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Honoraria; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel grants; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; Sanofi: Other: travel grants; Celgene Neovii: Other: travel grant. Crysandt:Incyte: Membership on an entity's Board of Directors or advisory committees; Amgem: Other: travel grant; celgene: Other: travel grant; Pfizer: Other: travel grant; Gilead: Other: travel grant. Jost:Jazz Pharmaceuticals: Honoraria; Sanofi: Honoraria; Gilead: Other: travel grants; Daiichi: Honoraria. Kaivers:Jazz Pharmaceuticals: Other: Travel Support. Mueller:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; CTI Life Sciences: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees, Other: Financing of Scientific Research; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astellas: Honoraria; Gentium: Honoraria; Gilead: Honoraria; Janssen: Honoraria; Jazz: Honoraria; Pharmaceuticals: Honoraria; Neovii: Honoraria; Novartis: Honoraria; Sanofi: Honoraria. Rautenberg:Jazz Pharmaceuticals: Other: Travel Support; Celgene: Honoraria, Other: Travel Support. Wolff:Takeda: Honoraria; Mallinckrodt: Honoraria; Novartis: Honoraria; Neovi: Honoraria. Schroeder:Celgene Corporation: Consultancy, Honoraria, Research Funding. Off Label Use: Venetoclax was used in combination with azacitidine or decitabine. The combination is not approved in the EU so far.. Kobbe:Pfizer: Honoraria, Other: Travel support; Abbvie: Honoraria, Other: Travel support; Biotest: Honoraria, Other: Travel support; Roche: Honoraria, Other: Travel support; Jazz: Honoraria, Other: Travel support; MSD: Honoraria, Other: Travel support; Celgene: Honoraria, Other: Travel support, Research Funding; Takeda: Honoraria, Other: Travel support; Amgen: Honoraria, Other: Travel support, Research Funding; Neovii: Honoraria, Other: Travel support; Medac: Honoraria, Other: Travel support; Novartis: Honoraria, Other: Travel support. OffLabel Disclosure: Venetoclax was used in combination with azacitidine or decitabine. The combination is not approved in the EU so far.
- Published
- 2019
17. Prognostic Value of Cpss Cytogenetic Risk Classification in Patients with CMML after Allogeneic Hematopoietic Stem Cell Transplantation: A Retrospective Multicenter Study of the Chronic Malignancies Working Party of the EBMT
- Author
-
Marie Robin, Victoria Potter, Henrik Sengeloev, Guido Kobbe, Didier Blaise, Ellen Meijer, Sheree Hazelaar, Arnold Ganser, Gérard Socié, Yves Chalandon, Peter Dreger, Ibrahim Yakoub-Agha, Dietger Niederwieser, Jürgen Finke, Dirk-Jan Eikema, Francesco Onida, Johan Maertens, Hendrik Veelken, Noel Milpied, Christian Koenecke, Nicolaus Kröger, Dietrich W. Beelen, Martin Bornhaeuser, Per Ljungman, and Maija Itälä-Remes
- Subjects
Oncology ,medicine.medical_specialty ,Univariate analysis ,Multivariate analysis ,business.industry ,Myelodysplastic syndromes ,medicine.medical_treatment ,Immunology ,Cytogenetics ,Medizin ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Surgery ,Transplantation ,Regimen ,Internal medicine ,medicine ,Chromosome abnormality ,ComputingMethodologies_GENERAL ,business - Abstract
Introduction The only curative treatment approach for patients with myelodysplastic syndromes (MDS) is allogeneic hematopoietic stem cell transplantation (HSCT), but disease relapse after transplantation is a major concern. Predictors for disease outcome after HSCT are limited. However, unfavorable cytogenetic abnormalities have been shown to serve as predictors for MDS-relapse after transplantation. Similar to the data available in MDS-patients not undergoing HSCT (Schanz et al. J Clin Oncol 2012), there is evidence that the novel 5-group cytogenetic classification has a better predictive value for outcome after HSCT than standard IPSS cytogenetics (Deeg et al. Blood 2012). The aim of this large multicentric, international study was to retrospectively determine the impact of the new 5-group cytogenetic MDS classification on outcome after HSCT. Patients and Methods Patients were selected from the EBMT database who had received HSCT for the treatment of MDS between 1982 and 2010 and for whom sufficient cytogenetic information was available. In total, 903 patients were included into the study. At time of HSCT, 97 (10.7%) patients had untreated MDS, 218 (24.1%) patients had advanced MDS or AML evolving from MDS in complete remission, and 227 (25.1%) patients were not in remission after treatment (in 12.3% information on stage of the disease was not available). Median time between diagnosis and transplant was 6.6 months (range 0.2-359.3). Matched related donor HSCT was performed in 574 patients (63.6%), and matched unrelated donor HSCT in 329 patients (36.4%). Bone marrow (35.4%) or peripheral blood (64.6%) served as stem cell graft. Myeloablative preparative regimens were used in 582 patients (64.5%), and a non-myeloablative regimen was given to 320 patients (35.4%). Impact of cytogenetic classification was analyzed in uni- and multivariate models regarding overall survival (OS) and relapse free survival (RFS) after HSCT. Predictive performance of the 2 classifications was compared by means of the cross-validated log partial likelihood. Results Estimated 5-year RFS and OS were 32% and 36% respectively. According to the 5-group cytogenetic classification 19 (2.1%) patients had very good risk cytogenetics, 204 (22.6%) normal risk cytogenetics, 438 (48.5%) intermediate risk cytogenetics, 178 (19.7%) poor risk cytogenetics, and 64 (7.1%) very poor risk cytogenetics. Good, intermediate, and poor risk cytogenetics according to IPSS were found in 192 (38.0%), 500 (40.2%), and 211 (23.7%) patients, respectively. In univariate analysis 5-group cytogenetic information was found to be strongly associated with OS and RFS (OS: log-rank test P
- Published
- 2018
18. Continuous high-resolution in vivo imaging reveals tumor-specific dissemination in an embryonic zebrafish xenograft model
- Author
-
Christopher Schmied, Steffen Volger, Michael Brand, Martin Bornhaeuser, Martin Wermke, Stephan Daetwyler, Stefanie N. Bernas, Manja Wobus, Katrin Lambert, Gerd Kempermann, Nandini Asokan, and Jan Huisken
- Subjects
Innate immune system ,Cell ,Biology ,medicine.disease ,biology.organism_classification ,Embryonic stem cell ,Metastasis ,medicine.anatomical_structure ,Immunology ,Cancer cell ,medicine ,Cancer research ,ROCK1 ,Zebrafish ,Preclinical imaging - Abstract
Mechanisms mediating tumor metastasis are crucial for diagnostic and therapeutic targeting. Here, we take advantage of a transparent embryonic zebrafish xenograft model (eZXM) to visualize and track injected human leukemic and breast cancer cells in real time using selective plane illumination microscopy (SPIM) for up to 30 hours. Injected cells exhibited disease-specific patterns of intravascular distribution with leukemic cells moving faster than breast cancer cells. While breast cancer cells predominantly adhered to nearby regions, about 30% invaded the avascularized tissue, reminiscent of their metastatic phenotype. Survival of the injected tumor cells was partly inhibited by the cellular innate immune system of the recipient embryos and leukemic cell dissemination was effectively inhibited by pharmacological ROCK1 blockade. These observations, and the ability to image several embryos simultaneously, support the use of eZXM and SPIM imaging as a functional screening platform to identify compounds that restricts cancer cell spread and invasion.
- Published
- 2017
- Full Text
- View/download PDF
19. Development of Novel Anti-CD10 Target Modules for Redirection of Universal CAR T Cells Against CD10-Positive Malignancies
- Author
-
Nicola Mitwasi, Ralf Bergmann, Václav Hořejší, Anja Hoffmann, Liliana Raquel Loureiro, Anja Feldmann, Martin Bornhaeuser, Claudia Arndt, Stefanie Koristka, Justyna Jureczek, Michael Bachmann, and Nicole Berndt
- Subjects
Chemistry ,T cell ,Melanoma ,Immunology ,Cell ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chimeric antigen receptor ,Epitope ,Cytokine release syndrome ,medicine.anatomical_structure ,Antigen ,hemic and lymphatic diseases ,medicine ,Cancer research ,B cell - Abstract
The common acute lymphoblastic leukemia antigen CD10 is a marker for several hematological malignancies, including acute lymphoblastic leukemia as well as T and B cell lymphomas, Burkitt lymphomas, and some solid tumors like renal cell carcinomas, pancreatic tumors and melanomas. Because of its tumor related expression pattern, CD10 is an attractive target for adoptively transferred T cells that are genetically modified to express chimeric antigen receptors (CARs). Recently, conventional CAR T cell therapy targeting CD19-positive hematological malignancies was clinically approved because of its impressive effectiveness in patients. However, CAR T cells can also cause severe side effects like on-target, off-tumor reactions, tumor lysis syndrome and cytokine release syndrome. Most critically, activity of conventional CAR T cells cannot be controlled, once they are applied in patients. As CD10 is also widely expressed on normal tissues, CAR T cell reactivity has to be controllable in order to stop CAR T cell therapy in case of on-target, off-tumor toxicities occur. Especially for this purpose, we have recently established a switchable, modular and universal CAR platform technology, named UniCAR system, which can be repeatedly turned on and off. In contrast to conventional CARs, that directly recognize a tumor-associated antigen (TAA) on the tumor cell surface via their extracellular single-chain variable fragment (scFv), the UniCAR system is structured in a modular manner of two components. The first component are T cells genetically engineered to express UniCARs and the second component are target modules (TMs). Most importantly, UniCARs cannot directly bind to a TAA because their extracellular scFv is directed against the peptide epitope E5B9 which is not present on the surface of living cells. Consequently, UniCAR armed T cells are per se inert. They can be redirected towards tumor cells only via a TM. TMs consist of a scFv targeting a TAA and the epitope E5B9 recognized by UniCARs allowing a cross-linkage of UniCAR T cells with tumor cells which results in T cell activation. As TMs have a very short half-life, UniCAR T cell activity can be controlled by dosing of the TM. Once the TM is administered, UniCAR T cells can be switched on, but once the TM injection is stopped and the TM is eliminated, UniCAR T cells are switched off immediately. Here, we show proof of concept for functionality of the UniCAR system targeting CD10-positive malignancies. Therefor, a novel anti-CD10 TM was constructed which is able to redirect UniCAR T cells to eliminate CD10-expressing tumor cells. In summary, we have established a universal, switchable, modular UniCAR platform technology that can be used to target CD10-positive malignancies. Disclosures Koristka: Intellia Therapeutics: Employment. Bachmann:GEMoaB Monoclonals: Equity Ownership, Patents & Royalties.
- Published
- 2019
20. Altered Structure and Function of Mesenchymal Stromal Cell-Derived Extracellular Matrix in MDS Can be Restored By Luspatercept
- Author
-
Susann Winter, Cindy Welzel, Manja Wobus, Carsten Werner, Uwe Platzbecker, Valentina Magno, Martin Bornhaeuser, Anna Mies, and Friedrich Stoelzel
- Subjects
Stromal cell ,Decellularization ,biology ,Chemistry ,Immunology ,Mesenchymal stem cell ,Integrin ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,Extracellular matrix ,Fibronectin ,Luspatercept ,biology.protein ,Progenitor cell - Abstract
Introduction The involvement of the bone marrow microenvironment (BMME) into disease progression and therapeutic response of myelodysplastic syndromes (MDS) is indisputable. Hereby, mesenchymal stromal cells (MSCs) play an important role for both the support of the leukemic clone and the remaining healthy hematopoietic stem and progenitor cells (HSPCs). The extracellular matrix (ECM) secreted by MSCs regulates stem cell fate through the modulation of cytokine and growth factor delivery and may also be targeted by clinically available drugs such as luspatercept, a novel recombinant fusion protein containing modified extracellular domain of activin receptor IIB. Luspatercept is a first-in-class erythroid maturation agent with promising results in lower-risk MDS patients with red blood cell transfusion dependency. Aim To shed light on the largely unknown composition and function of the MSC-derived ECM, we have characterized ECM from MDS patients vs. healthy controls and elucidate how luspatercept may modulate their functional characteristics. Methods Bone marrow-derived MSCs from patients with lower-risk MDS and age-matched healthy donors (HD) were treated with RAP-536, a murine homologue of luspatercept harboring the same activin receptor IIB domain. MSCs of three patients were treated with RAP-536 and RNA sequencing was carried out. Gene expression and pathway analyses were performed using the Reactome tool (https://reactome.org). Candidate genes were validated by quantitative real-time PCR (qPCR). For the generation of ECM, MSCs were seeded on poly-octadecene-alt-maleic anhydride and human fibronectin coated glass slides in the presence or absence of RAP-536. To yield cell-free ECM structures, cultures were decellularized at day 10 and analyzed by scanning electron microscopy (SEM), sulfated glycosaminoglycan (GAG), fibronectin and collagen staining as well as GAG quantification (Blyscan assay). Moreover, purified HD CD34+ HSPCs were cultured on ECM scaffolds for 6 and 9 days, respectively. Subsequently, expansion of adherent and supernatant cells was determined and the phenotype was analyzed by flow cytometry. Results RNA sequencing of MDS MSCs after six days of RAP-536 treatment revealed a total of 58 significantly regulated genes, thereof 24 up- and 34 down-regulated genes. Gene enrichment and pathway analyses revealed a striking involvement in ECM organization, collagen biosynthesis and formation. Moreover, integrin cell surface interaction genes showed significantly differential expression. Focusing on collagens as important ECM components, we identified Col7A1 and Col4A2 to be down-regulated. Indeed, both collagen mRNAs were significantly decreased by 46% and 25%, respectively, in MSCs after RAP-536 treatment compared to untreated controls. SEM characterization and immunofluorescence staining of the ECM showed a more compact fiber network produced by MDS MSCs. Moreover, MDS ECM contained higher levels of collagen and GAGs. Blyscan assay confirmed the latter observation, showing significantly higher sulfated GAG concentrations in MDS ECM. Interestingly, trapping of TGFβ superfamily ligands, such as GDF-11, by RAP-536 clearly reduced Col4 staining intensity in MDS MSC ECM. Structural and compositional ECM differences had functional impact on the expansion of HSPCs cultured on the matrices. Significant higher total cell numbers were detected on healthy ECM (18.3-fold vs. 12.1-fold expansion, *p< 0.05) but not on MDS ECM (12.9-fold) after 9 days of culture. The number of adherent cells increased 8.5-fold on healthy and 4.3-fold on MDS ECM and could be further increased after RAP-536 treatment of MSCs. Using flow cytometry, we found a 3.1-fold increased proportion of CD90+ HSPCs in the adherent fraction on healthy but only 1.8-fold on MDS MSC ECM. Integrin αIIb (CD41), αV (CD51) and β3 (CD61) were found to be significantly higher expressed in the adherent HSPC fraction. RAP-536 treatment resulted in up to 20% higher expression of both CD90 and integrin subunits. Summary We demonstrate an association between induced collagen abundance and reduced hematopoietic support in ECM derived from MDS MSCs and conclude that compact MDS ECM structure induced by TGFβ superfamily members may alter the cytokine environment for HSPCs. Consequently, TGFβ ligand trapping by RAP-536/luspatercept leads to ECM re-organization and thus an improved hematopoietic support. Disclosures Stoelzel: Shire: Consultancy, Other: Travel funding; Neovii: Other: Travel funding; JAZZ Pharmaceuticals: Consultancy. Platzbecker:Celgene: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria; Novartis: Consultancy, Honoraria.
- Published
- 2019
21. Development of Target Modules for Early and Late Stage Cancer Treatment Using Switchable Unicar T Cell Therapy
- Author
-
Nicola Mitwasi, Stefanie Koristka, Anja Feldmann, Ralf Bergmann, Claudia Arndt, Liliana Raquel Loureiro, Justyna Jureczek, Michael Bachmann, Anja Hoffmann, Nicole Berndt, and Martin Bornhaeuser
- Subjects
business.industry ,T cell ,Immunology ,Late-stage cancer ,Cancer therapy ,Cell Biology ,Hematology ,Hematologic Neoplasms ,medicine.disease ,Antigen binding ,Biochemistry ,Cytokine release syndrome ,medicine.anatomical_structure ,Immunoglobulin g4 ,Cancer research ,Medicine ,business ,Bodily secretions - Abstract
The clinical efficacy of CAR T cell therapies has been widely recognized, particularly in the treatment of hematologic malignancies. Nevertheless, CAR T cells also have the capability to elicit undesired effects such as on-target/off-tumor recognition and cytokine release syndrome. To increase clinical safety of CAR T cell therapy, a novel modular universal CAR platform termed UniCAR was developed by our group. In the UniCAR system, antigen-binding specificity and signaling features are two distinct moieties, in which the antigen specificity is provided by targeting modules (TMs) to redirect UniCAR T cells in an individualized time- and target-dependent manner. In this way, UniCAR T-cells acquire killing potential only in the presence of a tumour-specific TM. Given the reduced size of such molecules, they are rapidly eliminated and therefore, need to be continuously infused. Thus, possible side effects and activation of UniCAR T cells can be easily monitored and controlled by TM dosing. During the onset of therapy, tumor burden and the risk for severe side effects are high and regulation of CAR T cell activity is particularly important at this stage. For this reason, TMs with extended half-life may play an important role by improving eradication of residual tumor cells in late phases of treatment and further expedite clinical application. In this line of thought, a set of novel short-lived and longer-lasting TMs directed against several tumor-associated antigens was developed. Short-lived TMs are composed of a tumor-specific binding moiety fused to the La peptide epitope (E5B9) which is recognized by UniCAR T cells. In order to generate extended half-life TMs, these two components are fused via an Fc domain derived from the human IgG4 molecule. In vitro and in vivo assays have shown that both short-lived and longer-lasting TMs efficiently redirect UniCAR T cells to cancer cells in a highly target-specific manner, thereby promoting the secretion of pro-inflammatory cytokines and tumor cell lysis. Further assays using PET-imaging, demonstrated that all TM formats specifically enriched at the tumor site presenting either short or prolonged serum half-lives. From a clinical point of view, after the initial reduction of tumor burden promoted by the small TMs, IgG4-based TMs could be subsequently administrated allowing a more convenient and personalized treatment of the patients avoiding the continuous infusion of the short-lived TMs. Furthermore, the specific accumulation of such IgG4-based TMs at the tumor site sets these molecules as attractive candidates for in vivo imaging and endoradiotherapy. Taken together, combination of switchable UniCAR T cells and TMs with different sizes, specificities and half-lives represent a flexible and individualized approach at different stages of cancer treatment. Disclosures Koristka: Intellia Therapeutics: Employment. Bachmann:GEMoaB Monoclonals: Equity Ownership, Patents & Royalties.
- Published
- 2019
22. A Novel Revcar Platform for Switchable and Gated Tumor Targeting
- Author
-
Martin Bornhaeuser, Ralf Bergmann, Nicola Mitwasi, Anja Feldmann, Anja Hoffmann, Justyna Jureczek, Michael Bachmann, Liliana Raquel Loureiro, Nicole Berndt, Claudia Arndt, Enrico Kittel-Boselli, and Stefanie Koristka
- Subjects
Chemistry ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Chimeric antigen receptor ,Tumor antigen ,Epitope ,Cell biology ,Cytokine ,medicine.anatomical_structure ,Antigen ,Tumor Escape ,medicine ,Single-chain variable fragment - Abstract
Hematological malignancies are successfully treated with chimeric antigen receptor (CAR) armed T cells. Despite the clinical success, CAR T cell therapy struggles still with some problems including the selection of tumor escape variants and on-target, off-tumor side reactions as well as massive cytokine release and uncontrollability of CAR T cell activity in the patients. In order to enable controllability of CAR T cells and to avoid unspecific side effects, we established a novel switchable, split and adaptable CAR platform technology, termed RevCAR system. The novel RevCARs lack the single chain variable fragment (scFv) commonly used as extracellular domain in conventional CARs. Instead of the scFv, RevCARs contain only a small peptide epitope as extracellular portion. This design reduces the CAR size, avoids unspecific antigen binding and prevents antigen independent tonic signaling caused by scFv dimerization. As RevCAR T cells do not recognize anything, they are per se inert. Only in the presence of a corresponding bispecific antibody based target module (RevTM) they can be specifically redirected to tumor cells. Therefor RevTMs consist of two scFvs. One recognizes the RevCAR peptide epitope and the other one simultaneously binds to a tumor associated antigen (TAA). By dosing of the RevTM, which has a very short half-life, the reactivity of RevCAR T cells can be switched on and off reversibly. Another advantage is that the RevCAR system can be flexibly adapted to any tumor antigen simply by exchanging the RevTM. Furthermore, the small RevCAR size is favorable for inserting more than one RevCAR in the same T cell thus facilitating the mode of gated targeting which is a highly attractive approach to minimize the risk for on-target, off-tumor toxicities against healthy tissues and to increase tumor specificity of conventional CAR T cells. For 'AND' gate targeting via the RevCAR system, two different RevCARs were constructed and expressed simultaneously in the same T cell. The two RevCARs differed with respect to the extracellular peptide epitope and the intracellular signaling domain. Moreover, the respective transmembrane domain was selected to isolate the respective RevCAR signal. The first RevCAR is designed to transmit the activation signal, the second RevCAR to deliver a costimulatory signal. For efficient RevCAR T cell activation, both RevCARs must be engaged via their respective RevTM which on the one hand binds to one of the two RevCAR epitopes and on the other hand to one of two TAAs expressed on the same target cell. Here, we present two RevCAR/RevTM systems for retargeting of AML cells as well as solid tumor cells including via gated targeting. In summary, we show proof of concept for a novel switchable RevCAR system that can be used for retargeting of AML cells as well as solid tumors. The novel modular RevCAR platform is characterized by small size, lacks unwanted tonic signaling effects, allows the control of RevCAR T cell activity, enables gated targeting strategies, and can be adapted to any tumor antigen and tumor type. Disclosures Koristka: Intellia Therapeutics: Employment. Bachmann:GEMoaB Monoclonals: Equity Ownership, Patents & Royalties.
- Published
- 2019
23. Effects of spleen status on early outcomes after hematopoietic cell transplantation
- Author
-
Marcelo C. Pasquini, Manza A. Agovi, David I. Marks, Vikas Gupta, Hillard M. Lazarus, Gorgun Akpek, Richard T. Maziarz, J D Rizzo, Uday R. Popat, O Ringdén, Kenneth R. Cooke, Martin Bornhaeuser, P.L. McCarthy, Karen K. Ballen, Brent R. Logan, Brian J. Bolwell, Vincent T. Ho, and Dipnarine Maharaj
- Subjects
Adult ,Male ,medicine.medical_specialty ,Time Factors ,Adolescent ,Platelet Engraftment ,medicine.medical_treatment ,Splenectomy ,Spleen ,Hematopoietic stem cell transplantation ,stem cell transplantation ,Gastroenterology ,Article ,splenectomy ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,myeloproliferative disease ,Survival rate ,Retrospective Studies ,Transplantation ,Neutrophil Engraftment ,business.industry ,Hematopoietic Stem Cell Transplantation ,Engraftment ,Hematology ,Middle Aged ,Allografts ,medicine.disease ,3. Good health ,Survival Rate ,surgical procedures, operative ,medicine.anatomical_structure ,Graft-versus-host disease ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,Immunology ,spleen ,Female ,business ,Follow-Up Studies ,030215 immunology - Abstract
To assess the impact of spleen status on engraftment and early morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT), we analyzed 9,683 myeloablative allograft recipients from 1990 to 2006; 472 had prior splenectomy (SP), 300 splenic irradiation (SI), 1,471 with splenomegaly (SM), and 7,440 with normal spleen (NS). Median times to neutrophil and platelet engraftment were 15 vs. 18 days and 22 vs. 24 days for the SP and NS groups, respectively (p5.7x106/kg improved platelet engraftment at day+28. After adjusting variables by Cox regression, the incidence of graft-versus-host disease (GVHD) and overall survival were not different among groups. Splenomegaly is associated with delayed engraftment while splenectomy prior to HCT facilitates early engraftment without impact on survival.
- Published
- 2012
24. Dissociation of Osteogenic and Immunological Effects by the Selective Glucocorticoid Receptor Agonist, Compound A, in Human Bone Marrow Stromal Cells
- Author
-
Martin Bornhaeuser, Lorenz C. Hofbauer, Martina Rauner, Jan Tuckermann, Guy Haegeman, Claudia Goettsch, Nicola Stein, Sylvia Thiele, and Karolien De Bosscher
- Subjects
Adult ,Male ,medicine.medical_specialty ,Stromal cell ,medicine.medical_treatment ,Osteoclasts ,Tyramine ,Apoptosis ,Bone Marrow Cells ,Acetates ,Monocytes ,Bone remodeling ,Young Adult ,Paracrine signalling ,Receptors, Glucocorticoid ,Endocrinology ,Glucocorticoid receptor ,Osteoprotegerin ,Osteogenesis ,Internal medicine ,Ethylamines ,medicine ,Humans ,RNA, Messenger ,Cells, Cultured ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Chemistry ,Osteoblast ,Middle Aged ,Coculture Techniques ,Cytokine ,medicine.anatomical_structure ,RANKL ,biology.protein ,Female ,Interleukin-1 - Abstract
Glucocorticoids (GCs) regulate various physiological processes, including bone remodeling. Whereas physiological amounts of GCs are required for proper human osteoblast differentiation, prolonged exposure to GCs leads to substantial bone loss in vivo predominantly by inhibiting osteoblast functions. Compound A (CpdA) is a novel GC receptor modulator with the potential of an improved benefit/risk profile. Here we tested the osteoimmunological effects of CpdA on primary human osteoblasts and their paracrine interactions with osteoclasts. To assess the antiinflammatory potential of CpdA in human bone marrow stromal cell (BMSC)-derived osteoblasts, cells were stimulated with lipopolysaccharide and cytokine expression was determined. Similar to dexamethasone (DEX), CpdA profoundly suppressed lipopolysaccharide-induced TNF-α (−63%), IL-1β (−38%), and IL-6 (−36%) (P < 0.05) mRNA levels. Of note, CpdA failed to induce osteogenic differentiation of BMSCs, whereas DEX and budesonide enhanced matrix mineralization an d increased runt-related transcription factor 2 and alkaline phosphatase mRNA levels up to 5-fold in a dose-dependent manner. Interestingly, each substance promoted cell proliferation by 7–10% and suppressed apoptosis by 25–30% at low concentrations and early differentiation stages, whereas high concentrations (1 μm) suppressed proliferation and stimulated apoptosis in mature osteoblasts. Finally, CpdA did not increase the receptor activator of nuclear factor-κB ligand to osteoprotegerin mRNA ratio as compared with DEX and did not stimulate the formation of osteoclasts in coculture with BMSCs. In summary, CpdA displays dissociated osteogenic and immunological effects in human BMSCs that are distinct from those of conventional GCs. Whether the specific osteoimmunological profile of CpdA translates into a relevant in vivo effect needs to be further explored.
- Published
- 2011
25. Differences Between CEBPA bZIP and TAD Mutations and Their Effect on Outcome-an Analysis in 4578 Patients with Acute Myeloid Leukemia
- Author
-
Gerhard Ehninger, Michael Kramer, Jan Moritz Middeke, Franziska Taube, Sylvia Herold, Norbert Schmitz, Martin Bornhaeuser, Uwe Platzbecker, Wolfgang E. Berdel, Sebastian Stasik, Johannes Schetelig, Walter E. Aulitzky, Hubert Serve, Alwin Kraemer, Mathias Haenel, Wolf Roesler, Kerstin Schaefer-Eckart, Hermann Einsele, Claudia D. Baldus, Julia-Annabell Georgi, Christian Eberlein, Christian Thiede, and Christoph Roellig
- Subjects
Genetics ,Oncology ,Mutation ,medicine.medical_specialty ,NPM1 ,Myeloid ,Immunology ,Myeloid leukemia ,Context (language use) ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Enhancer binding ,Internal medicine ,CEBPA ,medicine ,Missense mutation ,030215 immunology - Abstract
Mutations of the key myeloid transcription factor CCAAT/enhancer binding protein alpha (C/EBPa) are found in 5-10% of patients with acute myeloid leukemia (AML). Two mutational clusters exist, in the aminoterminal transcription activation domains (TAD1 or 2) and in the basic leucine zipper domain (bZIP) located at the carboxyterminal-part of the protein. Biallelic mutations (biCEBPA) have been found to be associated with improved outcome and are now included as an independent entity in the WHO-classification. In contrast, monoallelic CEBPA-mutations (moCEBPA) do not appear to provide prognostic information. We characterized a large cohort of AML patients for CEBPA mutations and further analyzed the mutational spectrum of mono- and biallelic CEBPA-mutant AML patients to better understand potential differences in the biology of these groups. Patients and Methods: Patients (including all age groups) analyzed had a newly diagnosed AML and were registered in clinical protocols of the Study Alliance Leukemia (SAL)(AML96, AML2003 or AML60+, SORAML) or the SAL-register. Screening for CEBPA mutations was done using PCR and capillary electrophoresis. All identified CEBPA mutations were confirmed using conventional Sanger sequencing and the samples were further analyzed using next generation sequencing (Trusight Myeloid Panel, Illumina) for the presence of associated alterations. Results: In the 4578 patients analyzed, 228 (5%) with CEBPA-mutations were identified. An initial analysis revealed substantial clinical differences between the different mutation subtypes. Patients with biCEBPA (n=111) were significantly younger (median age 46 yrs) than wt-CEBPA patients (median 57 yrs; p Disclosures Middeke: Sanofi: Honoraria. Platzbecker:Janssen-Cilag: Honoraria, Research Funding; Celgene Corporation: Honoraria, Research Funding; TEVA Pharmaceutical Industries: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Thiede:AgenDix: Employment, Other: Ownership.
- Published
- 2016
26. Histone Deacetylase Inhibitors As Enhancers of Human Hematopoietic Stem Cell Activity
- Author
-
Manja Wobus, Martin Stoeter, Martin Kraeter, Jens Friedrichs, Guruchandar Arulmozhivarman, and Martin Bornhaeuser
- Subjects
Immunology ,Mesenchymal stem cell ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,CD146 ,CD90 ,Stem cell ,Progenitor cell - Abstract
Introduction The identification of compounds which increase the number but also keep or enhance the activity of hematopoietic stem and progenitor cells (HSPCs) could improve the clinical outcome after autologous and allogeneic hematopoietic stem cell transplantation (HSCT). So far, most attempts to increase HSPC numbers ex vivo have been unsuccessful because of either inadequate cell numbers and/or loss of engraftment capacity and HSPC quality during expansion. Executing drug discovery screens in vertebrate systems is generally expensive, technically challenging and time consuming. Therefore, the zebrafish represents a versatile vertebrate model allowing HSPC regulation and development studies during embryogenesis and adulthood. Methods We used a semi-automated chemical screen to identify modulators of HSPC activity by transgenic (cmyb:EGFP) zebrafish embryos. Verification of identified histone deacetylase (HDAC) inhibitor candidates was carried out in vitro using human CD34+ HSPCs which were isolated from apharesis samples of healthy donors after mobilization with G-CSF by anti-CD34 coupled magnetic beads. The influence of HDAC inhibitors on HSPC phenotype, gene expression pattern as well as adhesion and migration capacity was analyzed after 5 days of treatment either in single or in co-culture with bone marrow-derived mesenchymal stromal cells (MSCs). Results The HDAC inhibitors valproic acid (VPA), resminostat and entinostat were shown to significantly amplify the number of hematopoietic precursors in a chemical in vivo zebrafish embryo screen (Arulmozhivarman et al. 2016). Treatment of human CD34+ HSPCs with these compounds in vitro resulted in a significantly increased percentage of CD34+CD90+ cells up to 60% compared to controls which showed only 2% of double positive cells as well as in 3-fold higher CD34+ and about 12-fold higher CD34+CD90+ absolute cell numbers. CD34 is a well-known surface marker for human immature HSPCs and in combination with CD90 it defines a potentially pluripotent subpopulation. In a co-culture setting, we found that VPA treated cells showed 2 to 3-fold higher attachment capacity on MSCs compared to the control cells. This finding led us to quantify the adhesive capacity of cells using static adhesion assay and atomic force microscopy based single-cell force spectroscopy (AFM-SCFS). Interestingly, detachment forces of VPA treated HSPCs were 3 times increased on MSCs compared to control cells and a similar phenotype was observed by static adhesion assay. Accordingly, the chemokine-mediated migration of VPA treated HSPCs towards SDF-1/CXCL12 was inhibited. To reveal underlying downstream molecules and mechanisms mediating the modified cellular characteristics, a whole genome expression array was carried out for HSPCs treated with VPA in comparison to untreated controls. Amongst a panel of regulated genes, the melanoma cell adhesion molecule (MCAM/CD146), Notch 3 and its downstream effector Hes-1 as well as the SDF-1 receptor CXCR-4 were found to be significantly changed. Whereas the decreased expression of CXCR4 correlates with the inhibited migration potential of VPA-treated HSPCs and Notch-3/Hes-1 have a known role in normal and malignant hematopoiesis (Gu et al. 2016), the induced expression of MCAM on HSPCs was not described so far. The result was confirmed by flow cytometry which revealed a 40% MCAM-positive cell population when treated with VPA, whereas the control showed only negative cells. Additionally, significant higher transcript levels were detected for MCAM by quantitative real-time PCR in VPA expanded cells. Recently, we described a role of MCAM in MSCs for the hematopoietic support (Stopp et al. 2013). The inducible expression in HSPCs may reflect homotypic interactions which preserve a more immature subpopulation with high stem cell activity. Conclusion We describe for the first time the ability of the HDAC inhibitors VPA, resminostat and entinostat to efficiently expand CD34+ HSPCs ex vivo especially supporting a CD34+CD90+ subpopulation with potentially high stem cell activity. Moreover, a potential role of MCAM in this context may offer new perspectives of the HSPC expansion ex vivo for the improvement of HSCT. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
27. Birth order and transplant outcome in HLA-identical sibling stem cell transplantation – an analysis on behalf of the Center for International Blood and Marrow Transplantation (CIBMTR)
- Author
-
David B. Miklos, Arnold Ganser, Susan R. Marino, Marcelo Fernandez-Vina, Matthias Eder, Vikas Gupta, Edmund K. Waller, Michael Haagenson, Mahmoud Aljurf, Stephanie J. Lee, Christiane Dobbelstein, Martin Bornhaeuser, Jon J. van Rood, Marilyn S. Pollack, Vijay Reddy, Kwang Woo Ahn, Stephen R. Spellman, and Gregory A. Hale
- Subjects
Male ,HLA-DP Antigens ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Cohort Studies ,0302 clinical medicine ,Young adult ,Child ,0303 health sciences ,Hematopoietic Stem Cell Transplantation ,Microchimerism ,Hematology ,Middle Aged ,Tissue Donors ,3. Good health ,surgical procedures, operative ,Treatment Outcome ,030220 oncology & carcinogenesis ,Child, Preschool ,Hematologic Neoplasms ,Cohort ,Female ,Donor ,Cohort study ,Adult ,medicine.medical_specialty ,Adolescent ,Hematopoietic stem cell transplantation graft-versus-host-disease ,Chimerism ,Article ,03 medical and health sciences ,Young Adult ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Sibling ,030304 developmental biology ,Aged ,Pregnancy ,Transplantation ,business.industry ,Siblings ,medicine.disease ,Immunology ,Birth Order ,business - Abstract
Allogeneic stem cell transplantation (SCT) is the most effective treatment option for many hematologic malignancies, but graft-versus-host disease (GVHD) remains a major cause of treatment failure. Along with well-established risk factors for transplantation outcomes, recent single-center studies have identified a birth order effect in HLA-identical sibling SCT, with lower rates of acute and chronic GVHD and improved overall survival when the donor is younger than the recipient. One hypothesized mechanism for this effect is microchimerism due to fetomaternal and transmaternal sibling cell trafficking during pregnancy as the donor is exposed to recipient antigens in utero. The aim of the present study was to validate previously reported single-center data in a large, multicenter cohort provided by the Center for International Blood and Marrow Transplantation. All adult and pediatric patients (n = 11,365) with a hematologic malignancy who underwent allogeneic SCT with a graft from an HLA-identical sibling donor between 1990 and 2007 were included. When donors were younger than recipients, there was a significantly lower rate of acute GVHD grade II-IV and chronic GVHD in children, as well as a lower rate of chronic GVHD in adolescents. However, the hypothesized overall positive effect of lower relapse and better survival when donors are younger than recipients was not observed. Our data suggest that if otherwise equally matched, a graft from a younger sibling may be superior to a graft from an older sibling for children and adolescents undergoing SCT.
- Published
- 2013
28. High Prevalence of Functional Laa Specific Cytotoxic T Lymphocytes in Healthy Individuals-Implications for Strategies in Adoptive T Cell Therapies of Relapsed Leukemia
- Author
-
Torsten Tonn, Marcus Odendahl, Sarah Matko, and Martin Bornhaeuser
- Subjects
PRAME ,T cell ,Immunology ,Cell Biology ,Hematology ,Tumor Specific Peptide ,Biology ,Major histocompatibility complex ,Biochemistry ,Epitope ,medicine.anatomical_structure ,Antigen ,medicine ,biology.protein ,Cytotoxic T cell ,CD8 - Abstract
While adoptive transfer of virus antigen specific T cells has shown to be effective in therapy of resistant recurrent viremia which is frequently associated with the lack of protective immunity following hematopoietic stem cell transplantation, the transfer of leukemia associated antigen specific (LAA) T cells is less implemented and appears to depend on factors that hamper a successful translation into the clinic. Among them are low frequencies and low antigen affinity of LAA specific T cells which currently mandate laborious in vitro expansion protocols. Moreover, screening of healthy individuals with regard to the presence of LAA specific T cells revealed contradictory results. Since we failed to detect LAA specific T cells in healthy donors using single peptide specificities to known LAA epitopes coupled to MHC Streptamers, here we asked if the use of peptide mixes comprising 15mers overlapping by 11 amino acids and spanning the entire LAA protein could elicit in vitro T cell responses in healthy donors, otherwise undetectable by single peptide staining. A cohort of 48 HLA A*0201 healthy individuals was screened using intracellular cytokine staining (ICS) after stimulation with tumor specific peptide mixes representing well known LAAs (WT1, PRAME, NY-ESO, Survivin and p53). While distinct T-helper cell responses were not observed in either of the specimen tested, cytotoxic T lymphocytes could be elicited and measured after incubation with peptide mixes for 5 hours and subsequent CD8+ IFNγ+ staining in 12 out of 48 healthy subjects. Only one individual displayed specifies against multiple antigens (WT1:0,1%; PRAME:0,5%; NY-ESO:0,1%; p53:0.06%), while the remaining responses were directed to one single antigen per individual. Most prevalent and highest T cell frequencies were found against PRAME in 5 out of all screened subjects (mean 0.4±0.3%; max. 0.8%), followed by WT1 in 4 (mean 0.07±0.03%; max. 0.1%) and NY-ESO in 3 individuals (mean 0,07±0,04%; max. 0,1%); one showed CD8 T cells specific against Survivin (0,03%) and 2 individuals had CD8 frequencies specific against p53 (0,05±0,01; max. 0,06%), respectively. The calculated limit of detection (LOD) for the enumeration of LAA specific T cells was 0,02%. In contrary, testing LAA positive individuals with according MHC Streptamers presenting single peptides of previously described epitopes showed no frequencies exceeding LOD. Further analysis showed LAA specific CD8+ IFNγ+ T cells exhibit mainly a less differentiated phenotype (CD45RA+, CCR7+/-, TNFα+, IL-2+/-) and could be immune-magnetically isolated to purities of 94.5±0.7% using a PRAME-specific IFN-γ capture assay yielding 1*104 antigen specific T cells out of 4*107 PBMCs. Simultaneous enrichment of helper T cells to a purity of 73.0±7.6% proofed their existence, despite no CD4+ response could be detected via ICS in the first place. The cytotoxic potential of the cell product was confirmed in an Europium assay using T2 cells loaded with PRAME peptide mix. The specific lysis accounted to 19.3% at an E:T ratio of 1:1 after 90 minutes of co-incubation. In conclusion, using LAA specific peptide mixes in combination with ICS we were able to show a relatively high prevalence of LAA specific T cells, especially for PRAME, in healthy donors. These LAA specific T cells can be enriched without the need of in vitro expansion culturing ex vivo using the IFN-γ capture assay with regard to achieving a functional LAA specific T cell product for adoptive T cell transfer. Furthermore, a less differentiated phenotype exhibited by a large proportion of LAA specific T cells might contribute to their long term survival in a patient after transplantation. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
29. Allogeneic hematopoietic SCT in patients with AML following treosulfan/fludarabine conditioning
- Author
-
Dietrich W. Beelen, Jochen Casper, Rudolf Trenschel, I. W. Blau, Kajsa Larsson, Hannes Wandt, Jerzy Holowiecki, Martin Bornhaeuser, Joachim Baumgart, Axel R. Zander, Lutz Uharek, H. Einsele, Sebastian Giebel, Tomasz Kruzel, Tapani Ruutu, Mathias Freund, Kerstin Schaefer-Eckart, Heidrun A. Mylius, Uwe Pichlmeier, Miroslaw Markiewicz, Liisa Volin, and Gernot Stuhler
- Subjects
Adult ,Male ,medicine.medical_specialty ,Allogeneic transplantation ,Transplantation Conditioning ,medicine.drug_class ,Medizin ,Treosulfan ,Antimetabolite ,Gastroenterology ,Young Adult ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Transplantation, Homologous ,Prospective Studies ,Adverse effect ,Busulfan ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,Total body irradiation ,Middle Aged ,Surgery ,Fludarabine ,Regimen ,Leukemia, Myeloid, Acute ,Female ,business ,Vidarabine ,medicine.drug - Abstract
An alternative reduced-toxicity conditioning regimen for allogeneic transplantation, based on treosulfan and fludarabine, has recently been identified. The safety and efficacy of this new conditioning regimen has been investigated prospectively in patients with AML. A total number of 75 patients with AML in CR were treated with 3 × 14 g/m(2) treosulfan and 5 × 30 mg/m(2) fludarabine, followed by matched sibling or unrelated SCT. Patients were evaluated for engraftment, adverse events, GVHD, and for non-relapse mortality, relapse incidence, overall and disease-free survival (DFS). All patients showed primary engraftment of neutrophils after a median of 20 days. Non-hematological adverse events grade III-IV in severity included mainly infections (59%) and gastrointestinal symptoms (7%). Acute GVHD grade II-IV occurred in 21% and extensive chronic GVHD occurred in 16% of the patients. After a median follow-up of 715 days, the 2-year overall and DFS estimates were 61% and 55%, respectively. The 2-year incidences of relapse and non-relapse mortality reached 34% and 11%, respectively. In summary, our data confirm promising safety and efficacy of the treosulfan-based conditioning therapy in AML patients, ClinicalTrials.gov Identifier: NCT01063660.
- Published
- 2011
30. Treatment of high risk AML and MDS with Ara-C-Containing standard chemotherapy followed by reduced-intensity conditioning allogeneic stem cell transplantation (RIC-SCT) during Aplasia: a survey of the German Cooperative Transplant Study Group (GCTSG)
- Author
-
Peter Dreger, Gerhard Ehninger, Christoph Schmid, Anthony D. Ho, Cathrin Theuser, Martin Bornhaeuser, Joachim Kienast, Jolanta Dengler, Matthias Stelljes, and Ute Hegenbart
- Subjects
Melphalan ,medicine.medical_specialty ,business.industry ,Immunology ,Induction chemotherapy ,Cell Biology ,Hematology ,Total body irradiation ,Biochemistry ,Gastroenterology ,Chemotherapy regimen ,Surgery ,Fludarabine ,Transplantation ,Regimen ,hemic and lymphatic diseases ,Internal medicine ,Cyclosporin a ,medicine ,business ,medicine.drug - Abstract
Prognosis of patients (pts.) with refractory or relapsed acute myeloid leukaemia (AML) or advanced myelodysplasia (MDS) is poor. Preliminary reports suggest that induction of aplasia by a standard AML regimen followed by reduced intensity conditioned stem cell transplantation (RIC-SCT) in aplasia may be an effective strategy in this situation. A survey was conducted by the German Cooperative Transplant Study Group (GCTSG) to investigate the outcome after such an approach. In April 2006, GCTSG centers were asked if they had performed RIC-SCT in aplasia. Centers with positive response received a questionnaire including details on patient and donor characteristics, disease status, induction chemotherapy, transplant procedure and outcome. Data on 44 pts. (24 male, 20 female; median age 49y, range 19 – 66) with high-risk MDS (n=9), relapsed (n=16) or primary refractory AML (n=19) were reported by 3 centers. Pts. received aplasia-inducing chemotherapy with the following regimens: idarubicine / fludarabine / ara-C (n=16), daunorubicine / ara-C (n=15), fludarabine / ara-C / amsacrine (n=5), or other ara-C-based schedules (n=8). Subsequent transplant conditioning therapy consisted of melphalan (150 mg/m2) plus fludarabine (150mg/m2) in n=28, TBI (total body irradiation, 8Gy) plus fludarabine (150mg/m2) in n=11, and TBI (4Gy) plus cyclophosphamide (80 – 120 mg/kg) in n=5 pts. and was commenced after a median of 15 days (8 – 40) from the start of the preceding chemotherapy. Allogeneic peripheral blood (n=41) or bone marrow (n=3) stem cells from sibling (n=16), matched unrelated donors (n=18) or mismatched donors (n=10) were infused thereafter. Graft versus host disease (GVHD) prophylaxis was performed with cyclosporin A monotherapy (n=33) or by combination of a calcineurine inhibitor and methotrexate (n=11), n=31 received additional anti-thymocyte globuline. 43 pts. achieved stable engraftment and 41 showed > 90% donor chimerism after 28 – 90 days. Grade 2–3 acute GVHD occurred in 34%, whereas limited or extensive chronic GVHD was seen in 26% of the evaluable pts. The 100-day mortality rate was 24%: 5 pts. succumbed to treatment related complications, 4 to disease relapse. The median follow up is 183 days (19–1453). Actuarial survival rates estimated by Kaplan-Meier analysis are 56% and 30% at 6 and 12 months, whilst actuarial progression free survival rates are 42% and 30% at 6 and 12 months, respectively. This multi-center retrospective analysis suggests feasibility of the approach combining leukemia burden reduction by standard AML induction chemotherapy followed by RIC-SCT performed in aplasia. Treatment-related complications were comparable to those of a conventional transplant setting, thus disease control was achievable in a substantial proportion of patients with poor prognosis AML and MDS. Prospective studies are needed to assess whether durable remissions are achievable in patients with these otherwise fatal conditions.
- Published
- 2006
31. High Dose Therapy and Autologous Stem Cell Transplantation in Marginal Zone Lymphoma : An EBMT-FIL-Gimeto Retrospective Study
- Author
-
Catherine Thieblemont, Ariane Boumendil, Mohammed Wattad, Maurizio Musso, Annarita Conconi, Cristiana Pascutto, Virginia Valeria Ferretti, Herve Finel, Anette Haenel, Irit Avivi, Per Ljungman, Peter Dreger, Denis Caillot, Christian Berthou, Pavel Jindra, Martin Bornhaeuser, Martin Gramatzki, Silvia Montoto, Shannon Haenel, Gerhard Held, Luca Arcaini, Norbert Ifrah, Blaise Didier, David Pohlreich, Francesco Zaja, Christof Scheid, Gilles Salles, Giuseppe Milone, Jean-Henri Bourhis, Emmanuelle Nicolas-Virelizier, Devizzi Liliana, and François Guilhot
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,MALT lymphoma ,Cell Biology ,Hematology ,Total body irradiation ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Gastroenterology ,Surgery ,Log-rank test ,Transplantation ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Rituximab ,Cumulative incidence ,business ,medicine.drug - Abstract
Introduction:The role of autologous stem cell transplantation (ASCT) in patients with marginal zone lymphomas (MZL) is not fully elucidated. The aim of the present study was to determine the outcome of patients undergoing ASCT for relapsed/refractory MZL, and define prognostic factors affecting that outcome. Methods: Eligible for this study were patients with nodal, extra-nodal (MALT) or splenic MZL , aged ≥18 ears, who underwent a first ASCT between July 1994 and February 2013, and reported to the European Society for Blood and Marrow Transplantation (EBMT) registry, and/or the Fondazione Italiana Linfomi (FIL) and the Gruppo Italiano Trapianto di Midollo Osseo (GITMO) networks. Patients with a history of MZL transformation were excluded. Review of written diagnostic reports was mandatory for inclusion. Log rank tests were used to assess the impact of baseline characteristics on overall survival (OS) and event-free survival (EFS). In multivariate analysis, the effect of prognostic factors was evaluated using Cox regression models. Cumulative incidence of relapse (IR) and cumulative incidence of non-relapse mortality (NRM) were estimated with a competing-risk approach. Risk factors for IR and NRM were estimated by Pepe & Mori test or by Fine & Gray model. Results: The study included 199 patients, 111 patients (56%) with MALT lymphoma, 55 patients with nodal MZL (28%) and 33 patients (16%) with splenic MZL.. Median age at transplantation was 56 years (range, 25-71 years). Median time from diagnosis to ASCT was 2 years (0.1-28.0). Median number of prior therapies was 1 , (range 1-8) , including rituximab in 74%. 96% were transplanted with chemosensitive disease; 70 (37%) in CR1/PR1 ,113(59%) in CR/PR >1 and 7 in SD (4%) Median calendar year of ASCT was 2006, with 17,1% of transplants being performed before 2001. Total body irradiation-based high-dose regimen was used in 16 patients (8%), whilst 92% of the patients received high-dose chemotherapy only. Median follow-up was 4.1 years (0.1-19.1). Five-year cumulative incidence of relapse/progression (IR) and non-relapse mortality (NRM) were 38% (95%CI 30-45%) and 9 %( 95%CI 6-14%) respectively. Five-year event-free survival (EFS) and overall survival (OS) were 53% (95%CI 45-61%) and 73% (95%CI 65-79%), respectively. Multivariate analysis revealed age >65 years to be associated with shorter EFS and a shorter OS (HR=8.6, 95%CI 2.8-26.2, p Additionally, MZL predicted a shorter OS than MALT (HR=3.2, 95%CI 1.2-8.7, p=0.023). Notably, rituximab had no statistically significant effect on transplant outcome.Risk of secondary malignancies approached 6.8%. Conclusions: ASCT is a feasible and effective procedure when offered to MZL patients younger than 65 years, even in those previously exposed to rituximab. Disclosures Zaja: MedImmune: Research Funding.
- Published
- 2014
32. Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with acute lymphoblastic leukemia: no difference in related compared with unrelated transplant in first complete remission
- Author
-
Olle Ringdén, Volker Diehl, Martin Bornhaeuser, Axel R. Zander, Christoph Scheid, Bernd Hertenstein, Nicolaus Kroeger, Arnold Ganser, Harts Martin, Mats Remberger, Gerhard Ehninger, Dieter Hoelzer, Michael G. Kiehl, Axel A. Fauser, Rainer Schwerdtfeger, J. Kienast, Ludwig Kraut, and Mathias Stelljes
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Histocompatibility Testing ,Internal medicine ,Acute lymphocytic leukemia ,medicine ,Humans ,Transplantation, Homologous ,Family ,Survival analysis ,Chemotherapy ,business.industry ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Survival Analysis ,Tissue Donors ,Surgery ,Transplantation ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Female ,Bone marrow ,Stem cell ,business - Abstract
Purpose The role of unrelated allogeneic stem-cell transplantation in acute lymphoblastic leukemia (ALL) patients is still not clear, and only limited data are available from the literature. We analyzed factors affecting clinical outcome of ALL patients receiving a related or unrelated stem-cell graft from matched donors. Patients and Methods The total study population was 264 adult patients receiving a myeloablative allogeneic stem-cell transplant for ALL at nine bone marrow transplantation centers between 1990 and 2002. Of these, 221 patients receiving a matched related or unrelated graft were analyzed. One hundred forty-eight patients received transplantation in complete remission; 62 patients were in relapse; and 11 patients were refractory to chemotherapy before transplant. Fifty percent of patients received bone marrow, and 50% received peripheral blood stem cell from a human leukocyte antigen–identical related (n = 103), or matched unrelated (n = 118) donor. Results Disease-free survival (DFS) at 5 years was 28%, with 76 patients (34%) still alive (2.2 to 103 months post-transplantation), and 145 deceased (65 relapses, transplant-related mortality, 45%). We observed an advantage regarding DFS in favor of patients receiving transplantation during their first complete remission (CR) in comparison with patients receiving transplantation in or after second CR (P = .014) or who relapsed (P < .001). We observed a clear trend toward improved survival in favor of B-lineage ALL patients compared with T-lineage ALL patients (P = .052), and Philadelphia chromosome–positive patients had no poorer outcome than Philadelphia chromosome–negative patients. Total-body irradiation–based conditioning improved DFS in comparison with busulfan (P = .041). Conclusion Myeloablative matched related or matched unrelated allogeneic hematopoietic stem-cell transplantation in ALL patients should be performed in first CR.
- Published
- 2004
33. HPC enumeration with the Sysmex XE-2100 can guide further flow cytometric CD34(+) measurements and timing of leukaphereses
- Author
-
Christian Thiede, Uta Oelschlaegel, Martin Bornhaeuser, Kristina Hoelig, and G. Ehninger
- Subjects
Cancer Research ,medicine.medical_specialty ,Cd34 cells ,Immunology ,Urology ,CD34 ,Antigens, CD34 ,Cell Count ,Poor mobilizers ,Hematology analyzer ,Enumeration ,Immunology and Allergy ,Medicine ,Humans ,In patient ,Leukapheresis ,Genetics (clinical) ,Transplantation ,Sysmex XE-2100 ,business.industry ,Hematopoietic Stem Cell Transplantation ,Reproducibility of Results ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Peripheral blood ,Oncology ,business - Abstract
Background The aim of this study was to evaluate whether HPC counts measured with the hematology analyzer can predict CD34 + levels in peripheral blood and in the apheresis product, as detected by standard flow cytometry. The main focus was the evaluation of HPC counts in poor mobilizers. Methods Progenitor cell quantification was performed measuring HPC counts provided by the Sysmex XE-2100 hematology analyzer and CD34 + counts obtained in parallel by flow cytometry. Peripheral blood of patients who had received chemotherapy and G-CSF (142 measurements) and healthy donors mobilized with G-CSF alone (106 measurements) was investigated. HPC counts in peripheral blood were also correlated with apheresis yield. Results HPC counts were significantly higher than CD34 + counts (3.5 fold in patients and 1.7 fold in healthy donors, p=0.0015). Our data indicate that HPC counts ≤10/μL in pretreated patients predict a low probability of adequate CD34 + counts in peripheral blood and yields 6 /kg in subsequent aphereses. Furthermore, repetitive low HPC enumerations in an individual were followed by insufficient CD34 + counts in peripheral blood or aphereses in 81% of investigations. In healthy donors low HPC counts (≤10/μL; 12/106 measurements) did not exclusively predict low CD34 + counts (median 23/μL). Discussion HPC counts can be used to schedule the start of CD34 + measurements (threshold>10 HPC/μL) in patients mobilized after chemotherapy for autologous donation. Thus, expensive and time-consuming CD34 + enumerations can perhaps be minimized. HPC measurements cannot completely replace flow cytometric CD34 + enumeration. In particular, healthy stem-cell donors should be monitored with both methods to exclude false negative HPC measurements.
- Published
- 2003
34. Lenalidomide maintenance after allogeneic HSCT seems to trigger acute graft-versus-host disease in patients with high-risk myelodysplastic syndromes or acute myeloid leukemia and del(5q): results of the LENAMAINT trial
- Author
-
Ulrich Germing, Rudolf Trenschel, Gerhard Ehninger, Katja Sockel, Eva Mischak-Weissinger, Jürgen Finke, Dietrich W. Beelen, Brigitte Mohr, Jochen Greiner, Uwe Platzbecker, Christian Thiede, Christian Unzicker, Guido Kobbe, Martin Wermke, and Martin Bornhaeuser
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Myeloid ,Medizin ,Antineoplastic Agents ,Oncogene Protein p21(ras) ,hemic and lymphatic diseases ,Internal medicine ,Biomarkers, Tumor ,Humans ,Medicine ,WT1 Proteins ,Lenalidomide ,Aged ,Aged, 80 and over ,Chromosome Aberrations ,Base Sequence ,Gene Expression Regulation, Leukemic ,business.industry ,Myelodysplastic syndromes ,Myeloid leukemia ,Hematology ,Middle Aged ,medicine.disease ,Thalidomide ,Transplantation ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,medicine.anatomical_structure ,Online-Only Articles ,Myelodysplastic Syndromes ,Mutation ,Immunology ,Chromosomes, Human, Pair 5 ,Female ,Tumor Suppressor Protein p53 ,Stem cell ,business ,medicine.drug - Abstract
Patients with chromosome 5 abnormalities and high-risk myelodysplastic syndromes or acute myeloid leukemia have a poor outcome. We hypothesized that increasing doses of lenalidomide may benefit this group of patients by inhibiting the tumor clone, as assessed by fluorescence in situ hybridization for del(5q31).Twenty-eight patients at diagnosis or with relapsed disease and not eligible for standard therapy (16 with acute myeloid leukemia, 12 with intermediate-risk 2 or high-risk myelodysplastic syndrome) were enrolled in this prospective phase II multicenter trial and treated with lenalidomide up to 30 mg daily for 16 weeks. Three patients had isolated del(5q), six had del(5q) plus one additional aberration, 14 had del(5q) and a complex karyotype, four had monosomy 5, and one had del(5q) identified by fluorescence in situ hybridization only.Major and minor cytogenetic responses, assessed by fluorescence in situ hybridization, were achieved in 5/26 (19%) and 2/26 (8%) patients, respectively, who received one or more dose of lenalidomide, while two patients achieved only a bone marrow response. Nine of all 26 patients (35%) and nine of the ten who completed the 16 weeks of trial responded to treatment. Using the International Working Group criteria for acute myeloid leukemia and myelodysplastic syndrome the overall response rate in treated patients with acute myeloid leukemia was 20% (3/15), while that for patients with myelodysplastic syndrome was 36% (4/11). Seven patients stopped therapy due to progressive disease and nine because of complications, most of which were disease-related. Response rates were similar in patients with isolated del(5q) and in those with additional aberrations. Interestingly, patients with TP53 mutations responded less well than those without mutations (2/13 versus 5/9, respectively; P=0.047). No responses were observed among 11 cases with deleterious TP53 mutations.Our data support a role for higher doses of lenalidomide in poor prognosis patients with myelodysplastic syndrome and acute myeloid leukemia with deletion 5q. (Clinicaltrials.gov identifier NCT00761449).
- Published
- 2012
35. Outcome and prognostic features of intensive care unit treatment in patients with hematological malignancies
- Author
-
Mathias Haenel, Gerhard Ehninger, Frank Kroschinsky, Martin Bornhaeuser, Matthias Weise, Ulrich Schuler, Gert Hoeffken, and Thomas Illmer
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Population ,Critical Care and Intensive Care Medicine ,law.invention ,Cohort Studies ,law ,Intensive care ,Internal medicine ,Germany ,Hemofiltration ,medicine ,Humans ,Intensive care medicine ,education ,Aged ,Retrospective Studies ,Mechanical ventilation ,education.field_of_study ,business.industry ,Retrospective cohort study ,Middle Aged ,Prognosis ,Intensive care unit ,Patient Discharge ,Survival Rate ,Regimen ,Intensive Care Units ,Logistic Models ,Treatment Outcome ,SAPS II ,Hematologic Neoplasms ,Female ,business - Abstract
Objective. To assess the outcome of intensive care unit (ICU) treatment in patients with hematological malignancies. Design and setting. Retrospective cohort study in the medical ICU of a university hospital. Patients. 104 critically ill patients after receiving conventional chemotherapy or autologous hematopoietic stem cell transplantation. Interventions. We analyzed demographic data, underlying disease, intensity of antineoplastic regimen, cause of admission, need for mechanical ventilation, and hemofiltration, ICU survival, and survival after discharge, furthermore neutrophil count, C-reactive protein (≤150 vs. >150 mg/l), antithrombin III, prothrombin time, and SAPS II (≤50 vs. >50) at ICU admission. All recorded variables were evaluated for prognostic relevance by univariate and multivariate analyses. Measurements and results. Overall ICU mortality was 44%, with significantly higher mortality in ventilated patients (74% vs. 12% in nonventilated patients, p
- Published
- 2001
36. Allogeneic Hematopoietic Cell Transplantation Is Effective In Patients With Advanced Systemic Mastocytosis: A Multicenter Retrospective Analysis
- Author
-
Andrew L. Gilman, Cem Akin, Martin Bornhaeuser, Tsiporah B. Shore, Eva Wagner, Christoph Schmid, H. Joachim Deeg, Daniel J. Weisdorf, Peter Valent, Bernd Gruhn, Hans Hägglund, Miguel-Angel Perales, Esperanza B. Papadopoulos, William J. Hogan, Uday R. Popat, Alexandra Boehm, Tanja Gromke, Robert K. Stuart, Herrad Baurmann, Andreas Reiter, Werner Rabitsch, Vinod Pullarkat, Bart L. Scott, A. John Barrett, Gregory M. Vercellotti, Sebastian Kreil, Tor Shwayder, Michael Doubek, Eleni Tholouli, Ryan Shanley, Jack W. Hsu, Wolfgang R. Sperr, Steven M. Devine, Damaj Gandhi, Celalettin Ustun, Ryotaro Nakamura, Maria Theresa Van Lint, Lucy A. Godley, Olivier Hermine, and Livio Pagano
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Hematopoietic stem cell transplantation ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Systemic mastocytosis ,Cladribine ,030304 developmental biology ,0303 health sciences ,Chemotherapy ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Mast cell leukemia ,3. Good health ,Surgery ,Transplantation ,Leukemia ,medicine.anatomical_structure ,Bone marrow ,business ,030215 immunology ,medicine.drug - Abstract
Systemic mastocytosis (SM) is a rare hematologic neoplasm characterized by abnormal growth and accumulation of tissue mast cells (MC) in various organ systems, including bone marrow (BM). Indolent and advanced forms of SM have been described. Whereas patients with ISM have a normal or near normal life-expectancy, patients with advanced SM, including those suffering from mast cell leukemia (MCL) have a poor prognosis. In these patients, neoplastic MC are usually resistant against conventional drugs and various targeted drugs. In rapidly progressive aggressive SM (ASM) and MCL, polychemotherapy followed by allogeneic hematopoietic stem cell transplantation (alloHCT) has been proposed. However, outcome of alloHCT in advanced SM is unknown, and it also remains uncertain whether clinically relevant graft-versus-SM (GVSM) effects may occur in these patients, as only sporadic case reports have been published. We performed a retrospective multi-center analysis to evaluate the outcome of alloHCT in patients with advanced SM. Fifty-four advanced SM patients receiving SCT in 32 transplantation centers in Europe and America were identified between 1990 and 2013. The median patient age was 45 years. Donors were: HLA identical siblings (31), unrelated donors (URD) (15), umbilical cord blood donors (UCB) (2), and haploidentical donors (1). In 5 patients, stem cell source was not defined (5). Thirty-four patients received myeloablative conditioning (MAC) and 18 received reduced intensity conditioning regimens (RIC). In 2 patients, conditioning regimen was not specified. Indications for alloHCT were SM with an associated clonal hematologic non-mast cell lineage disease (SM-AHNMD) (n=32), MCL (n=13, including one with MCL-AHNMD), 8 with ASM and 1 with myelomastocytic leukemia (MML). The most prevalent AHNMD was acute myeloid leukemia (AML, n=16). With follow-up of 35-6180 (median 365) days, SM responses (defined as ≥50% decrease in BM mast cells ± decrease in serum tryptase ± regression of other organ manifestations) were observed in 39 patients (72%), including complete responses (CR) documented in 12 patients (22%). Eleven patients had stable disease, whereas 4 patients (7%) progressed immediately after alloHCT (primary resistance). In addition, 10 patients progressed (5 of them within 100 days) after an initial response. Progression was most frequently seen in MCL patients (n=6, 50%). In the AHNMD group, only 8 patients relapsed/progressed (25%). The overall survival (OS) and SM progression-free survival (PFS) at 1 year were 63% and 50% for all patients, 77% and 68% for SM-AHNMD, 63% and 50% for ASM, and 25% and 17% for MCL, respectively. The strongest predictive variable associated with inferior survival was a diagnosis of MCL. Other factors associated with poor outcome were: Karnofsky performance status ≤70%, ≥2 SM regimens given before alloHCT (e.g., steroids, cladribine, chemotherapy, tyrosine kinase inhibitor), donor source (alternative donors-UCB and haploidentical compared to sibling or URD), SM progression within the first 100 days, normal cytogenetics (compared to t(8;21) (q22;q22), and RIC (compared to MAC). The following variables were not associated with poor outcome: patient and donor age, recipient-donor sex match status, graft source (BM vs. peripheral stem cells), BM mast cell percentage at time of alloHCT, and CR status of AML or SM response at time of alloHCT. This largest multi-center analysis of results in advanced SM provides evidence for clinical efficacy of alloHCT, presumably because of a GVSM effect of alloHCT (achieving CR, and response to donor-lymphocyte infusions and RIC alloHCT). However, responses varied among different SM categories: while patients with SM-AHNMD enjoyed excellent outcomes, the OS for MCL patients in general, was poor. Nevertheless it is remarkable that 3 of 13 patients with MCL – an otherwise fatal disease with a median survival of Disclosures: Vercellotti: Sangart Inc.: Research Funding; Seattle Genetics: Research Funding. Akin:Novartis: Consultancy. Valent:Novartis: Consultancy, Honoraria, Research Funding.
- Published
- 2013
37. Multidrug-Related Protein 1 (MRP1) Polymorphisms rs129081, rs212090, and rs212091 Predict Survival In Acute Myeloid Leukemia
- Author
-
Desiree Kunadt, Christian Dransfeld, Maria Schmiedgen, Michael Kramer, Christoph Röllig, Christian Thiede, Martin Bornhaeuser, Ulrich Mahlknecht, Gerhard Ehninger, Markus Schaich, and Friedrich Stölzel
- Subjects
Oncology ,NPM1 ,medicine.medical_specialty ,dbSNP ,Anthracycline ,Daunorubicin ,Immunology ,Induction chemotherapy ,Single-nucleotide polymorphism ,Cell Biology ,Hematology ,Biology ,Bioinformatics ,Biochemistry ,Internal medicine ,Cytarabine ,medicine ,biology.protein ,ABCC11 ,medicine.drug - Abstract
Background ABCB1 (=MDR1, multidrug resistance protein 1) single nucleotide polymorphisms (SNPs) were shown to have a significant impact on therapy outcome in patients with acute myeloid leukemia (AML). Furthermore, an independent significant impact on treatment response and patient survival of SNPs in the genes for ABCC4 (MRP4), ABCC5 (MRP5) and ABCC11 (MRP8) related SNPs has also been demonstrated. In contrast, therapeutic strategies trying to modulate the anthracycline efflux of these transporters have failed in most clinical trials so far. Recently, higher dosages of daunorubicin used during induction chemotherapy have been associated with a better outcome in certain subgroups of AML patients. Hence, in times of individual diagnostic genetic analyses available as point-of-care diagnostics, the goal of this study was to further investigate whether SNPs in ABC-transporter genes, which are responsible for anthracycline efflux, have an independent impact on treatment outcome. Patients and Methods DNA samples were obtained from bone marrow aspirates of 160 Caucasian patients with newly diagnosed AML as part of the prospective AML2003 trial (NCT00180102). The cohort solely consisted of patients with a normal karyotype, based on conventional G-banding, minimizing false results in case of gain or loss of chromosomal material. All patients received double induction chemotherapy with daunorubicin and cytarabine. After DNA extraction, quantitative real time PCR was performed, using a total of 49 SNP assays investigating SNPs of seven different ABC genes. The identification of the corresponding SNPs was performed in an in silico analysis using the NIH dbSNP database and HapMap while statistical univariate and multivariate analyses were performed using SPSS. Results We detected three ABCC1 (MRP1) SNPs: rs129081 (CACCCC[C/G]ACTCCA), rs212090 (TTACTG[A/T]TCCCAC), and rs212091 (ACCTTA[A/G]AGAACA) with a significant influence on disease-free survival (DFS) or overall survival (OS), respectively. Patients carrying the homozygous rs129081 GG-SNP had a significant longer 5-year OS and 5-year DFS compared to the homozygous wildtype CC and heterozygous CG patients (OS: 68% [GG] vs. 40% [CC] vs. 64%, [CG], p=.035; DFS: 64% vs. 35% vs. 50%, p=.01). SNP rs212090 revealed a statistically significant difference in DFS when comparing homozygous alleles TT and AA (wildtype), 40% vs. 68%, p=.021. SNP rs212091 showed a significant difference concerning OS, with homozygous SNP GG leading to worse OS (0% vs. wildtype AA 64% vs. heterozygous AG 59%, p=.006). Again, there was a significant difference in DFS between both homozygous alleles AA (wildtype) and GG (55% vs. 0%, p=.018). Furthermore, there were no significant differences of standard clinical and laboratory baseline characteristics, FLT3-ITD mutation, or NPM1-mutation status, or chemotherapeutic toxicities. In order to exclude false positive findings of SNPs conferred as a result of leukemic transformation, we obtained saliva germline DNA from patients in complete remission who were treated by chemoconsolidation and performed a confirmatory analysis with the investigated SNPs, including rs129081, rs212090, and rs212091. Here, all SNPs were shown to be expressed in germline DNA in remission and bone marrow samples at diagnosis alike. The multivariate models for rs129081, rs212090 (TT), rs212091(AG), and rs212091(AA) revealed significances of p=.024, p=.029, p=.042, and p=.017 respectively for DFS but not for OS (except for rs212091[AA]). After adjustment for a false discovery rate of 5% still a trend towards the association of the SNPs and DFS could be seen. Therefore, more research is necessary to strengthen this evidence. Conclusion In this study we found a significant influence of rs129081, rs212090, and rs212091 SNPs (ABCC1, MRP1) on survival in AML in univariate analyses. Interestingly, these polymorphisms were not associated with other AML specific characteristics at diagnosis and were shown to be expressed in germline DNA and AML DNA alike. Hence, we suggest a prognostic effect of these SNPs which might be responsible for differential anthracycline susceptibility. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
38. The Immunologic Composition of the Bone Marrow Changes Rapidly During the Harvest Procedure
- Author
-
Claudia Schönefeldt, Martin Bornhaeuser, Uwe Platzbecker, Malte von Bonin, Sebastian Tuve, Martin Wermke, Cathrin Wegner, Kristina Hölig, Uta Oelschlegel, and Rainer Ordemann
- Subjects
Peanut butter ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Andrology ,Transplantation ,Graft-versus-host disease ,medicine.anatomical_structure ,Immunophenotyping ,Harvest Procedure ,medicine ,Bone marrow ,CD8 - Abstract
Abstract 1915 Bone marrow (BM) has been associated with a decreased rate of Graft-versus-Host-Disease (GvHD) when compared to peripheral blood stem cells (PBSC) in the context of allogeneic stem cell transplantation (allo-SCT). It has been reasoned that this may relate in part to the differential quantitative and qualitative immunologic composition of both stem cell sources. It has, for example, been noted that BM contains a higher percentage of CD8+ T-cells, natural killer (NK) cells and recently also more regulatory T-cells than PBSC. It is further known that the total nucleated cell concentration (TNC) correlates negatively with the BM harvest volume, which could argue for an increasing dilution of the transplant with peripheral blood (pB) during the BM harvest. How this impacts on the different immunological compartments and how the BM at different time points of the harvest procedure compares to PBSC products has not been determined yet. During routine BM harvest procedures (n = 24) we subjected a part of the aspirate drawn at the beginning, after withdrawing half of the prescribed volume and at the end of the harvest procedure to detailed immunophenotyping using multicolor flowcytometry including intracellular staining for FoxP3 and IL17. The PBSC products of an age matched group (n = 20) were analyzed in parallel with the same flowcytometry protocols and used as a comparison. During the BM harvest the median TNC dropped rapidly from 53.2 Gpt/l in the beginning to 17.0 Gpt/l after half the volume had been collected and finally reached 6.2 Gpt/l at the end of the procedure. As expected, the first BM specimen contained a higher concentration of NK- (6.7 vs. 4.6 %, p = 0.001) and B-cells (14.1 vs. 10.2 %, p < 0.001) but less T-helper-(Th)-cells (36.0 vs. 53.7 %, p < 0.001) than pB. After collection half of the prescribed volume the NK-cell concentration had already dropped to 4.5 %, which was not significantly different from pB (p = 0.412) anymore. The B-cell concentration, however, remained at levels comparable to the first specimen (14.5 %) and significantly differed from pB (p < 0.001). The last BM specimen drawn still had a higher B-cell concentration than pB (10.9 vs. 10.2 %), although this was not statistically significant (p = 0.265). Th-cells demonstrated a steady increase in concentration during the harvest with a median concentration of 43.4 and 47.0 % in the middle and at the end of the procedure. Both the halfway value and the end of harvest concentration were significantly different from pB (p < 0.001 and p = 0.004, respectively). An overview on the NK-, B- and Th-cell-concentrations at the different time points analyzed can be found in Figure 1. Within the Th compartment we were unable to see a clear trend for Treg which had a concentration of 7.6 % in the beginning, 7.0% halfway and 7.1 % after all BM had been collected. None of these concentrations was significantly different from that found in pB (7.6 %). The same applied to Th17 cells which made up 0.51 % of Th in the beginning, 0.59 % in the middle and 0.50 % at the end of the procedure and 0.84 % in the pB. Moreover there was no significant difference between the first BM specimen and PBSC products with respect to Treg (7.6 vs. 6.7 %, p = 0.181) or Th17 (0.51 vs. 0.61 %, p = 0.100). In contrast, the NK- and Th-cell concentration in PBSC products was significantly higher than in the first BM specimen (NK: 10.7 % vs. 6.7 %, p =0.005; Th: 40.3 % vs. 36.0 %, p =0.038). We conclude that the composition of immune cells within the BM changes significantly during the harvest procedure probably due to an increasing dilution with peripheral blood. These changes variably affect different compartments and may have an impact on post-transplant immunological function and complications. Therefore BM harvest volumes may need to be considered when comparing BM and PBSC with respect to clinical outcomes. In contrast to previous reports, we found no indication that BM, no matter at which time point analyzed, contained a higher concentration of Treg when compared to PBSC. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
39. Functional Influence of CXCL12 Regulating miRNAs in Mesenchymal Stem Cells (MSCs)
- Author
-
Gerhard Ehninger, Martin Bornhaeuser, Friedrich Stölzel, Ruth H. Strasser, Fernando A. Fierro, Laleh S. Arabanian, Thomas Illmer, and David M. Poitz
- Subjects
Chemokine ,biology ,Immunology ,Mesenchymal stem cell ,Cell Biology ,Hematology ,Biochemistry ,Proinflammatory cytokine ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,microRNA ,biology.protein ,medicine ,Stromal cell-derived factor 1 ,Bone marrow ,Stem cell - Abstract
Abstract 2350 CXCL12 is a chemokine known to be critical for the regulation of the interaction between hematopoietic stem cells (HSCs) and their niche in the bone marrow, e.g. mesenchymal stem cells (MSCs). MicroRNAs (miRNAs) are post-transcriptional regulators recently shown to mediate a variety of cellular processes in the bone marrow niche. However, identification of specific miRNAs and their regulatory role in the crosstalk between HSCs and MSCs are still poorly understood. From a library of 470 miRNAs, 26 miRNAs were shown to downregulate the levels of CXCL12 in the supernatant of the human MSC line SCP-1. Eight of them (miR-23, 130b, 135, 200b, 200c, 216, 222, 602) were chosen for further investigation according to their significant interaction with the 3'UTR of CXCL12 as determined by luciferase assay. Among them, miR-23a,130 and 222 were expressed in 46 human primary MSCs, whereas the other miRs show negligible expression in resting MSCs. However, we observed, that MSCs that underwent adipogenic and osteogenic differentiation showed strongly decreased CXCL12 protein values early (day 5) and at later stages (day 14). The later drop in CXCL12 expression was clearly associated with an increased expression of miR-23a and miR-200. We furthermore tested a subset of stimuli (proinflammatory cytokines, cytotoxic drugs, chemokines) for their ability to modulate the described miRNAs. Amongst them, exclusively the application of transforming growth factor ß1 (TGF-ß1), resulted in the induction of miR-23a and at the same time reduction of CXCL12. The effect was counteracted by transfection of anti-miR-23 molecules. Taken together, we have shown for the first time that CXCL12-targeting miRNAs (in particular miR23a) have a significant potential to regulate the properties of the stem cell niche. Moreover, miR-23 is implicated in the signalling pathway of TGF-ß1 in human MSCs. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
40. A Multicenter Retrospective Analysis on Pentostatin As Salvage Therapy of Severe Steroid Refractory Intestinal Acute Graft Versus Host Disease
- Author
-
Stefan Klein, Martin Bornhaeuser, Wolf-Karsten Hofmann, Gesine Bug, Kerstin Schaefer-Eckart, Hans Martin, Peter Dreger, Johannes Schetelig, Christoph Schmid, Thomas Schmitt, Rainer Schwerdtfeger, and Hannes Wandt
- Subjects
medicine.medical_specialty ,Basiliximab ,business.industry ,Immunology ,Salvage therapy ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Surgery ,Transplantation ,Graft-versus-host disease ,Median follow-up ,Internal medicine ,Prednisolone ,medicine ,Pentostatin ,Alemtuzumab ,business ,medicine.drug - Abstract
Abstract 3048 Acute graft-versus-host disease (aGvHD) of the gastrointestinal (GI) tract is still a major clinical challenge after allogeneic stem cell transplantation. Patients with steroid-refractory disease have a poor prognosis. Pentostatin, an inhibitor of adenosine deaminase, has shown efficacy as salvage therapy in steroid-refractory aGvHD of the GI tract in small single center studies. Here we report on the experience with pentostatin in severe steroid-refractory aGvHD of the GI tract at seven German transplant centers. PATIENTS: A total number of 123 patients who had been treated with pentostatin due to intestinal steroid-refractory aGvHD between 2000 and 2011 were retrospectively analyzed. Steroid-refractory aGvHD was defined as progression or no improvement of diarrhea despite treatment with prednisolone (≥ 2mg/kg/d) for ≥ 3 days. Pentostatin was infused at a dose of 1mg/m2 for 3 consecutive days. In patients with impaired renal function the dose of pentostatin was reduced. Patients received 1–4 cycles. Steroids and calcineurin inhibitors (CNI) were continued. Response after therapy with pentostatin was classified as complete (CR, no ongoing symptoms of GvHD), very good partial (VGPR, residual symptoms only) or no response (NR). 50 females and 73 males with a median age of 50 (range: 19–70) years were included. The underlying diseases were AML (n=71), ALL (n=15), CML/MPS (n=6), lymphoma (n=12), MDS (n=10), and multiple myeloma (n=9). 85 patients received reduced intensity and 38 myeloablative conditioning. Patients had been transplanted from matched related (n=38), matched unrelated (n=53) or mismatched donors (n=32). All patients suffered from severe steroid-refractory intestinal aGvHD overall grade III (n=59) or IV (n=64). Patients received pentostatin as first line salvage (n=109) or beyond first line salvage therapy (n=14). Results: 52 patients (43%) responded after salvage therapy with pentostatin. 39 patients (32%) achieved CR, 13 patients (11%) VGPR. Median survival was 104 days; 2-year and long term survival rates were 26 and 19% with a median follow up of 45 months. Among 109 patients who received pentostatin as first line salvage therapy 49 (45%) responded (37 × CR [34%] and 12 × VGPR [11%]). Median survival, 2-year and long term survival were essentially the same as in the total cohort of patients. After the first infusion of pentostatin clinical improvement occurred within a median of 14 (range: 1–58) days. 71 patients (57%) did not respond. Responding patients had a significantly (p Conclusions: The outcome after salvage therapy of III/IV° steroid-refractory intestinal aGvHD with pentostatin is at least within the range as reported for other salvage approaches. In this critical clinical situation pentostatin has some superior characteristics: a sustainable effect, moderate toxicity, easy application and cost-effectiveness. Moreover, this analysis suggests that the outcome of steroid-refractory aGvHD cannot be improved by the application of more than one immunosuppressive salvage drug in addition to steroids and CNI or by second line salvage approaches. Disclosures: Klein: Hospira: Honoraria, Research Funding. Off Label Use: pentostatin is not licensed for use in acute GvHD.
- Published
- 2011
41. Early Lenalidomide Maintenance to Prevent Relapse of High-Risk MDS and AML Patients with Del(5q) Following Allogeneic HCT - Results of the 'LENAMAINT' Trial
- Author
-
Rudolf Trenschel, Guido Kobbe, Katja Sockel, Martin Bornhaeuser, Brigitte Mohr, Gerhard Ehninger, Uwe Platzbecker, Jochen Greiner, Ulrich Germing, Jürgen Finke, Dietrich W. Beelen, and Christian Unzicker
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Alpha interferon ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Neutropenia ,medicine.disease ,Biochemistry ,Surgery ,Clinical trial ,Graft-versus-host disease ,Maintenance therapy ,Internal medicine ,medicine ,business ,Adverse effect ,Lenalidomide ,medicine.drug - Abstract
Abstract 3060 Background: Chromosome 5 abnormalities in patients (pts) with acute myeloid leukemia (AML) or advanced myelodysplastic syndrome (MDS) are mostly associated with a poor outcome. Although allogeneic hematopoietic stem cell transplantation (HCT) is the treatment approach with the highest curative potential, early relapse rates following HCT are still considerably high. Since therapeutic options in these pts are often limited, the prevention of relapse represents a major challenge. Lenalidomide (LEN) has been successfully used in MDS pts with del(5q) cytogenetic abnormalities. Besides a direct anti-proliferative effect on del(5q) progenitors, its immunmodulatory function might also enhance T-or NK cell mediated GVL effects. Therefore it could be an effective maintenance drug to prevent relapse after allogeneic HCT in pts with MDS or AML and del(5q) abnormalities. Methods: We report results of a prospective multicenter phase II clinical trial evaluating the efficacy to prevent relapse as well as safety of LEN maintenance following HCT in pts with MDS or AML and cytogenetic abnormalities including del(5q). Only pts achieving a complete hematological remission (CR) after HCT were eligible. In the absence of toxicity or relapse pts could receive up to 12 cycles of LEN maintenance at a dose of 10 mg/day orally, for 21 days, with 7 days rest (28 day cycle). Results: Ten pts with either MDS (n=1, RAEB-1) or AML (n=9) with a median age of 65 years (range 40–72) were included. The disease status prior to allogeneic HCT were CR in 4 pts, relapse/refractory disease in 3 pts, unknown in 2 pts while the MDS pt had not received any prior therapy. A complex aberrant karyotype including del(5q) was documented in 5 pts. Two pts had an additional cytogenetic abnormality besides del(5q) and 3 pts displayed single del(5q). While three pts underwent allogeneic HCT from a matched sibling donor, four pts were transplanted from a matched unrelated and three from a mismatched unrelated donor (single allele or antigen mismatch). Conditioning was of reduced intensity and was followed by the infusion of peripheral blood stem cells in all pts. LEN maintenance therapy was started after complete hematopoietic recovery with documented complete remission at a median of 2.5 months (range 2–4 months) following HCT. After a median of 4 cycles 8 of 10 pts (80%) had to discontinue LEN treatment due to relapse (n=4), development of severe GvHD grade 2–4 (n=2) or other adverse events (n=2). Altogether, 6 of 10 pts (60%) developed severe acute GvHD grade 3–4 within the first 2 cycles of LEN maintenance. All pts were still under systemic immunosuppression at the time of GvHD appearance. Other common adverse events were gastrointestinal side effects (nausea) and myelotoxicity. Reversible neutropenia grade 3/4 was documented in 3 of 10 (30%) of the pts while thrombocytopenia grade 3/4 occurred in 4 of 10 (40%) of them. During the treatment course specific T cell responses against different tumor/leukemia-associated antigens (TAAs/LAAs) using ELISpot analysis for Interferon alpha and granzyme B were measured. Sufficient T cells before and during LEN treatment were only available in one pt, who showed an increase of specific T cell responses against the TAAs/LAAs. With a median follow-up of 254 days (range 32–677) from the start of LEN maintenance, 5 of 10 pts (50%) are currently alive with four pts in continuous CR since the time of HCT. The study was stopped prematurely because of suspected induction of GVHD by LEN. Conclusions: Early LEN maintenance to prevent relapse following HCT in pts with MDS or AML and del(5q) may be associated with the induction of severe acute GVHD. Disclosures: Kobbe: Celgene: Consultancy, Research Funding; Ortho Biotec: Consultancy. Germing:Celgene: Consultancy, Research Funding. Bornhaeuser:Celgene: Honoraria. Platzbecker:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2011
42. 66 Allogeneic stem-cell transplantation in chronic myelomonocytic leukemia (CMML): Factors affecting transplantation outcome. A study of the CLWP of the EBMT
- Author
-
D. W. Beelen, N. Kroeger, T. de Witte, Hildegard T. Greinix, Martin Bornhaeuser, Jean-Pierre Jouet, Rainer Schwerdtfeger, Jürgen Finke, Ghulam J. Mufti, A. van Bietsen, Liisa Volin, and Argyris Symeonidis
- Subjects
Cancer Research ,medicine.medical_specialty ,Hematology ,business.industry ,Anemia ,Myelodysplastic syndromes ,Chronic myelomonocytic leukemia ,Total body irradiation ,medicine.disease ,Transplantation ,Leukemia ,medicine.anatomical_structure ,Oncology ,Internal medicine ,medicine ,Bone marrow ,business - Abstract
s of the 11th International Symposium on Myelodysplastic Syndromes (MDS) 18 May 2011 21 May 2011 Oral Presentations / Leukemia Research 35 (2011) S14–S26 S25 66 Allogeneic stem-cell transplantation in chronic myelomonocytic leukemia (CMML): Factors affecting transplantation outcome. A study of the CLWP of the EBMT A. Symeonidis, A. van Bietsen, G. Mufti, J. Finke, D. Beelen, M. Bornhaeuser, H. Greinix, J.P. Jouet, L. Volin, R. Schwerdtfeger, T. de Witte, N. Kroeger. Hematology Division, Dept of Internal Medicine, University of Patras Medical School, Patras, Greece; Dept of Medical Statistics and Bioinformatics, Chronic Leukemias Working Party of the EBMT, Leiden, The Netherlands; Dept of Hematological Medicine, GKT School of Medicine, London, UK; Dept of Medicine, Hematology, Oncology, University of Freiburg, Freiburg, Dept of Bone Marrow Transplantation, University Hospital of Essen, Essen, Medizinische Klinik und Polyklinik I, Universitaetsklinikum Dresden, Dresden, Germany; Klinik fuer Innere Medizin I, Medizinische Universitaet Wien, Vienna, Austria; Service des Maladies du Sang, Hopital Claude Huriez, Lille Cedex, Lille, France; Division of Hematology, Dept of Medicine, Helsinki University Central Hospital, Helsinki, Finland; KMT Zentrum, Deutsche Klinik fuer Diagnostik, Wiesbaden, Germany; Dept of Hematology, Radboud University Nijmegen, Nijmegen, The Netherlands; Bone Marrow Transplantation Centre, University Hospital Eppendorf, Hamburg, Germany According to WHO classification CMML is characterized, first, as dysplastic or proliferative and second, as type-1 or type-2. Results of allogeneic stem-cell transplantation (allo-SCT) in CMML are usually pooled together with other MDS. We retrospectively analyzed transplantation outcome in 489 patients with CMML from the EBMT database (females 160, males 329, median age 52.7 years, range 19.5–75.4 years). Among 145 patients, 67 had CMML-dysplastic and 78 CMML-proliferative, whereas among 214 patients with known subtype, 87 had type-1 (40.6%), 32 had type-2 (15%) and 95 had evolved to AML (44.4%). Conditioning was standard in 249 (55.7%) and reduced-intensity in 198 (44.3%). Donors were HLA-identical siblings (245~50.1%), other related (23~4.7%) or unrelated ones (221~45.2%). Disease status at allo-SCT was complete remission (CR) in 108 patients (22.1%), no-CR in 331 (67.7%) and unknown in 50 (10.2%). Stem cell source was bone marrow in 125 (25.6%) and peripheral blood (PBSC) in 365 (74.4%). Engraftment was successful in 432/460 patients (93.9%) and unknown in 29. Grades 0–1 acute GVHD was reported in 291/448 evaluable patients (65%), grades 2–4 in 149 (33.3%) and was unknown in 8. Chronic GVHD was reported in 102/197 patients (limited 49, extensive 53). At the time of this analysis 224 patients were alive (45.8%), of whom 198 (40.5%) were disease-free and 120 (24.5%) had relapsed. Among 265 deaths, 95 were disease-related (35.8%), 145 transplant-related (54.7%) and 25 other cause-related (9.4%). The probability of nonrelapse mortality at 4 years was 35% and it was lower in female to female allo-SCT (p =0.069). Patients with abnormal cytogenetics had higher risk for relapse (p = 0.017) and for death after relapse (p = 0.03). Patients transplanted in CR had lower probability for non-relapse death (p = 0.006) and longer RFS and OS (p =0.010 and p=0.018 respectively). Kaplan–Meier estimates for OS of the whole group was 46.7 months (95% Confidence Interval 38.1– 55.2 months). Kaplan-Meier estimates for RFS was longer in PBSC recipients (p = 0.006). In multivariate analysis the only significant prognostic factor for OS was disease status at SCT (CR vs noCR, p = 0.010). Analysis also included interval from diagnosis to transplant, dysplastic vs proliferative and type-1 vs type-2 disease, intensity of the conditioning, donor matching, cytogenetics, stemcell source, T-cell depletion, administration of total body irradiation, grading of acute and chronic GVHD and the year of allo-SCT. Allo-SCT remains a promising treatment option for patients with CMML and patients should preferably be referred to allo-SCT after achieving the best possible remission status. 67 Response to ESA treatment in patients with MDS: Determination of a predictive score, from a retrospective analysis of 669 patients A. Symeonidis, P. Zikos, A. Galanopoulos, I. Kotsianidis, A. Kouraklis, E. Terpos, M. Protopapa, H. Papadaki, V. Lambropoulou, A. Aktypi, P. Bakarakos, S. Michalopoulou, A. Anastasiadis, E. Michalis, N. Zoumbos. Hematology Division, Dept of Internal Medicine, University of Patras Medical School, Hematology, St Andreas Hospital of Patras, Patras, Dept of Hematology, G. Gennimatas General Hospital of Athens, Athinai, Dept of Hematology, Democritus Thrace University, Alexandroupolis, Dept of Medical Therapeutics, National and Kapodistrian University of Athens, Athinai, Dept of Hematology, General Hospital of Serres, Serres, Dept of Hematology, University of Crete School of Medicine, Heraklion, Olympion Hospital of Patras, Patras, Greece To investigate factors predicting for a favorable response to treatment with ESA in patients with MDS, 669 patients with a complete follow-up, treated for at least 12 weeks, were retrospectively analyzed. Patients (441 males, 228 females, median age 74 years) were classified as RA (n =257), RARS (n =119), RAEB (n =210), CMML (n =71) and unclassified MDS (n =12). IPSS [available in 618 patients (92.4%)] was Low in 232 (37.5%), Int-1 in 249 (40.3%), Int-2 in 98 (15.9%) and High in 39 patients (6.3%). Age, gender, baseline hemoglobin and platelet count, serum Epo, Response to Anemia Index (RAI), defined as the Hb× log(Epo) component, WHO classification, IPSS, WPSS, cytogenetics, marrow blasts, marrow cellularity, previous transfusions and time from diagnosis to treatment were tested as predictors. At 12 weeks 136 patients (20.3%) achieved a complete (CR) and 150 (22.4%) a partial response (PR). Overall, 212 patients (31.7%) achieved CR and 123 PR [18.4%, overall response (OR) 50.1%]. OR was 66% in FAB-RA, 51.3% in FAB-RARS, 33.6% in RAEB, 46.5% in CMML, 77.6% in WHORA, 66.2% in WHO-RARS, 60.6% in CMML-D, 58.2% in RCMD, 48.9% in del-5q, 43.3% in RAEB-1, 32.6% in RCMD-RS, 31.7% in CMML-P, and 20.9% in RAEB-2. In multivariate analysis WPSS was the most important predictor, with a probability of CR at 12 weeks 59.6%, 37.2%, 34.4%, 9% and 0% for the very low, low, intermediate, high and very high risk group respectively (p 21 vs 26.5% for those with Epo > 200). Other factors affecting response were RAI (p < 0.001), interval from diagnosis to treatment (p< 0.001), bone marrow cellularity (p< 0.001), previous transfusions (p < 0.001), hemoglobin (p < 0.001), platelet count (p < 0.01), serum Epo (p < 0.01) and cytogenetic group (p < 0.02). By using the 4 main factors (WPSS, RAI, marrow cellularity and interval from diagnosis to treatment) we have created a score, which could predict 85%, 47%, 35%, 15% and 0% probability for response in patients exhibiting 1, 2, 3, 4 and 5 of these factors. Therefore, WPSS, RAI, marrow cellularity and interval from diagnosis to treatment constitute a score, highly predictive for response to ESA treatment in patients with MDS. 68 ASXL1 is frequently mutated and associated with a negative prognosis in MDS F. Thol, I. Friesen, F. Damm, H. Yun, E.M. Weissinger, J. Krauter, K. Wagner, A. Chaturvedi, A. Sharma, M. Wichmann, G. Gohring, C. Schumann, G. Bug, O. Ottmann, W.-K. Hofmann, B. Schlegelberger, M. Heuser, A. Ganser. Medizinische Hochschule Hannover, Hannover, University Hospital Mannheim, Mannheim, University of Frankfurt, Frankfurt/Main, Germany Myelodysplastic syndrome (MDS) is a heterogeneous disease of the hematopoetic stem cell caused by mutations, deregulated gene expression and epigenetic modifications of genes leading
- Published
- 2011
43. Appearance of Mature 6-Sulfo LacNAc+ Dendritic Cells In Early and Late Engraftment After Allogeneic Stem Cell Transplantation
- Author
-
Konrad Mager, Jörgen Radke, Nona Shayegi, Rebekka Wehner, Conrad Heuchel, Gerhard Ehninger, Jan Moritz Middeke, Christoph Röllig, Uta Ölschlegel, Felix Bahr, Sebastian Tuve, Marc Schmitz, Martin Wermke, Christian Thiede, Martin Bornhaeuser, Uwe Platzbecker, and Johannes Schetelig
- Subjects
CD86 ,Myeloid ,T cell ,Immunology ,hemic and immune systems ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,Interleukin 12 ,medicine ,Stem cell ,Antigen-presenting cell ,CD8 - Abstract
Abstract 3720 Background: Dendritic cells (DCs) are professional antigen-presenting cells which display an extraordinary capacity to induce, sustain and regulate T cell responses. Recently, 6-sulfo LacNAc+ (slan) DCs (formerly termed M-DC8+ DCs) have been described as a major subpopulation of proinflammatory human blood DCs which are principal producers of tumor necrosis factor-alpha and interleukin-12. In addition, it has been demonstrated that slanDCs efficiently induce antigen-specific CD4+ and CD8+ T cells and direct the polarization of naïve CD4+ T lymphocytes into Th1 cells. In the present study, we investigated the reconstitution kinetics of slanDCs after allogeneic stem cell transplantation (aSCT) in comparision to CD1c+ myeloid DCs and plasmacytoid DCs representing two additional major human blood DC subsets. Material and Methods: The frequency of slanDCs, CD1c+ myeloid DCs and plasmacytoid DCs in the peripheral blood was quantified by flow cytometry in 70 patients following aSCT at different time points in early engraftment ( Results: (1) Early engraftment (30 days post transplantation): Interestingly, in the late phase post transplantation, the frequency of slanDCs steadily increases and these DCs represent the most abundant DC subpopulation in the second and third month post transplantation. The frequency of CD1c+ myeloid DCs and plasmacytoid DCs remains unchanged. Again, the majority of slanDCs show a mature phenotype in contrast to CD1c+ myeloid DCs and plasmacytoid DCs. Conclusion: Whereas the early engraftment phase after aSCT is dominated by CD1c+ myeloid DCs and plasmacytoid DCs, slanDCs represent the most abundant DC subset in the late engraftment phase. Furthermore, in both engraftment phases the majority of slanDCs display a mature phenotype in contrast to CD1c+ myeloid DCs and plasmacytoid DCs. Current studies are focused on functional assays and the role of individual DC populations in acute graft-versus-host disease and graft-versus-leukemia responses in the early and late phase following aSCT. Disclosures: Platzbecker: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2010
44. Spleen Status and Engraftment After Allogeneic Hematopoietic Stem Cell Transplantation (HCT)
- Author
-
Matthew Carabasi, J. Douglas Rizzo, Vikas Gupta, Gregory A. Hale, Vincent T. Ho, David I. Marks, Brian J. Bolwell, Philip L. McCarthy, Hillard M. Lazarus, Marcelo C. Pasquini, Dipnarine Maharaj, Martin Bornhaeuser, Kenneth R. Cooke, Richard T. Maziarz, Uday R. Popat, Olle Ringdén, Manza A. Agovi, Gorgun Akpek, Brent R. Logan, and Karen K. Ballen
- Subjects
medicine.medical_specialty ,Myeloid ,business.industry ,medicine.medical_treatment ,Immunology ,Splenectomy ,Spleen ,Cell Biology ,Hematology ,Disease ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Gastroenterology ,medicine.anatomical_structure ,Graft-versus-host disease ,Internal medicine ,Cord blood ,medicine ,Bone marrow ,business - Abstract
Abstract 3486 Delayed myeloid engraftment after HCT is a risk for increased morbidity and mortality, especially in patients with splenomegaly (SM) at time of transplant. Time to engraftment and overall survival after HCT have not been well analyzed in patients with prior splenectomy (SP) or splenic irradiation (SI), when compared to patients with normal spleens (NS) or with SM. A total of 9,683 recipients with myeloproliferative diseases and/or myelodysplasia who were reported to CIBMTR after receiving a myeloablative allogeneic HCT between 1990 to 2006 were compared according to the spleen status at transplant: 472 SP; 300 SI; 1,471 SM and 7,440 NS. Recipients of cord blood grafts were excluded. The median age was 39 years for all groups, the SP group had a higher proportion of patients with Karnofsky performance score OR d21 WBC1 (95% CI2) p-value OR d28 Platelet3 (95% CI) p-value RR4 Overall Mortality (95% CI) p-value Normal spleen (NS) 1 - 1 - 1 - Splenectomy (SP) 2.25 (1.76–2.89) Disclosures: Maziarz: Millenium: Speakers Bureau; Genzyme: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.
- Published
- 2010
45. Characterization of Extramedullary Acute Myeloid Leukemia – Results of the AML96 Trial
- Author
-
Friedrich Stölzel, Michael Kramer, Markus Schaich, Gerhard Ehninger, Christoph Röllig, Uta Oelschlägel, Martin Bornhaeuser, Brigitte Mohr, Jörgen Radke, and Christian Thiede
- Subjects
medicine.medical_specialty ,Myeloid ,business.industry ,medicine.medical_treatment ,Immunology ,Induction chemotherapy ,Myeloid leukemia ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Gastroenterology ,Surgery ,Leukemia ,medicine.anatomical_structure ,Median follow-up ,Internal medicine ,medicine ,Myeloid sarcoma ,business ,Survival analysis - Abstract
Abstract 2154 Background: Myeloid Sarcoma (MS) is defined as an extramedullary mass composed of myeloid blasts occurring at an anatomical site other than the bone marrow. Furthermore, the term extramedullary manifestation (EM) is applied if it accompanies overt acute myeloid leukemia (AML) and represents non-effacing tissue infiltration. EM is reported to correspond often to the skin but can affect almost every site of the body. The prognosis of MS or EM has been discussed controversially in the past. EM at diagnosis of AML is generally thought to be a rare event. However, data defining the prevalence of EM at diagnosis of AML and its prognostic value are missing. The aim of this analysis was to provide data for estimating the prevalence of EM at diagnosis of AML and to determine its relevance by including clinical and laboratory data from patients being treated in the prospective AML96 trial of the Study Alliance Leukemia (SAL) study group. Patients and Methods: A total of 326 patients with AML (age 17 – 83 years) and EM were treated within the AML96 trial with a median follow up of 8.8 years (95% CI, 8.4 to 9.3 years). All patients received double induction chemotherapy. Consolidation therapy contained high-dose cytosine arabinoside and for patients ≤ 60 years of age the option of autologous or allogeneic hematopoietic stem cell transplantation (HSCT). Logistic regression analyses were used to identify prognostic variables for CR rates. The method of Kaplan-Meier was used to estimate OS and EFS. Confidence interval (CI) estimation for the survival curves was based on the cumulative hazard function using the Greenwood's formula for the SE estimation. Survival distributions were compared using the log rank test. Results: 17% of the AML patients entered into the AML96 trial were diagnosed with EM. In 313 of the 326 patients (96%) EM was evident at diagnosis. The majority of patients with EM were diagnosed with de novo AML (84%, n=273), whereas gingival infiltration (51%, n=166) displayed the main EM of AML with CNS involvement being less common (4%, n=14). The majority of patients had a cytogenetic intermediate risk profile (71%, n=221) with a total of 172 patients (56%) harboring a normal karyotype. Patients with EM had a statistically significant lower median CD34-positivity of bone marrow blasts, higher percentage of FAB subtypes M4 and M5, higher WBC counts and LDH at diagnosis and higher percentage of NPM1 mutations compared to those patients without EM (all p Conclusions: This analysis represents the largest study so far investigating the impact of EM AML. Patients with EM AML have distinct differences from AML patients without EM regarding their clinical and molecular characteristics at diagnosis. However these differences do not translate into differences in response to induction chemotherapy. Compared to patients without EM, survival analysis revealed differences according to the NPM1/FLT3-ITD mutation status which is also described for patients without EM AML. However, the prognosis for patients with EM who harbor a mutated NPM1 the prognosis at relapse seems to be dismal. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
46. H1N1-09 Infections In Patients with Hematologic or Oncologic Malignancies: A Single-Centre Experience
- Author
-
Katja Sockel, Michael Laniado, Gerhard Ehninger, Martin Bornhaeuser, Johannes Schetelig, Christoph Pöhlmann, and Nona Shayegi
- Subjects
medicine.medical_specialty ,Oseltamivir ,business.industry ,Pleural effusion ,Immunology ,Cell Biology ,Hematology ,Lung injury ,medicine.disease ,Biochemistry ,Asymptomatic ,Surgery ,Transplantation ,Pneumonia ,chemistry.chemical_compound ,chemistry ,Respiratory failure ,Atypical pneumonia ,Internal medicine ,medicine ,medicine.symptom ,business - Abstract
Abstract 4866 Introduction The first pandemic of the 21st century was caused by the novel influenza A (H1N1) virus, now known as pandemic H1N1-09 virus. Its first wave reached Germany in autumn 2009. Though for the German public health sector this pandemic was a challenge, a state of national emergency could not be recognized. A popular current believe is that the threat by this pandemic had been overestimated by national and international health care experts. Here, we report our experience with the pandemic H1N1-09 virus in hematologic patients when the first wave hit Germany between December 2009 and February 2010. Methods Viral diagnostic for all patients was performed in a central virologic laboratory. All patients with at least one sample tested positive by PCR for H1N1-09 between October 2009 and March 2010 were included into the analysis. Samples were obtained by nasal wash, nasopharyngeal swab or broncho-alveolary lavage. The medical charts of all patients with H1N1-09 infection were reviewed systematically. Results 15 patients with underlying hematologic diseases (10 male and 5 female) with a median age of 52 years (range 18–70 years) were tested positive for H1N1-09 virus by PCR in our department. Notably, in 12 Patients H1N1-09 was nosocomially acquired after a median of 16 days hospitalization (range 6 – 42 days). 13 patients (87%) got infected between December 2 and December 29. At the time of infection all patients were immunocompromised with 11 patients being cytopenic after chemotherapy and four patients after allogeneic hematopoietic cell transplantation (HCT) among the latter two patients were in aplasia after allogeneic HCT (day +5 and day +11 after HCT). CT scan was performed in 8 patients at the time of diagnosis. Seven patients presented with signs of atypical pneumonia on CT scan. Ground glass opacity, consolidation, airway wall thickening, airway dilatation, pleural effusion and lymphadenopathy were common findings. In 10 patients viral clearance was monitored by RT-PCR. The median duration of viral shedding was 10 days (range 4 – 41 days). Prolonged viral persistence was associated with severe lung injury. All patients received Oseltamivir as first-line therapy, except three patients, who died prior to the confirmation of the diagnosis by RT-PCR. Simultaneously broad spectrum antibiotics and antimycotics were administered. 5 patients (33%) with respiratory failure needed invasive mechanical ventilation (MV) at the time of the H1N1-09 infection. Three out of these patients died. Within a follow up of 6 months eight patients have died. Six patients (40%) have died from the infection. Among these three patients died from fulminant pulmonary failure whereas three patients died several weeks after H1N1-09 infection from subsequent respiratory or multiorgan failure. The impact on indirect mortality cannot yet be fully assessed, since in some patients the infection caused significant delay of anti-leukemic therapy and acquired comorbidities resulted in dose-reductions of chemotherapy. Conclusions In contrast to largely mild infections in the healthy German population pandemic H1N1-09 pneumonia represented a life-threatening infection for hematologic patients associated with a high mortality due to acute respiratory failure, late pulmonary complications and delay of antitumor treatment. One alarming finding was the frequency of nosocomial infections. This observation points to the possibility of transmission of the virus from patient to patient, visitors to patient or even from medical staff to patient. With this observation in our institution patients admitted to hospital were put under quarantine before they were allowed to be accommodated in a double room in the hematologic unit. Transmission of the virus from asymptomatic staff or visitors to patient is another major concern. The suspected vaccination rate of medical staff in Germany was less than 20%. Especially, when asymptomatic or mild H1N1 infections occur - as it was the case in Germany – the medical staff and visitors could become important vectors of infection. The most effective measure against this threat is active immunization of visitors prior to patient contact and medical staff in hematologic units. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
47. Shift to a New Donor Does Not Improve the Outcome After Second Allogeneic Stem Cell Transplantation (alloSCT) in Acute Leukemia Relapse After a First Allosct – a Risk Factor Analysis by the German Stem Cell Registry (DRST)
- Author
-
Ralph Mayer, Rainer Schwerdtfeger, Juergen Finke, Hans Martin, Christof Scheid, Stefan Schoenland, Arnold Ganser, Dietrich W. Beelen, Maximilian Christopeit, Ulrike Feldmann, Martin Gramatzki, Herbert G. Sayer, Gerhard Behre, Hellmut Ottinger, Christoph Faul, Lutz Uharek, Guido Kobbe, Axel A. Fauser, Hermann Einsele, Martin Bornhaeuser, Joachim Kienast, Axel R. Zander, Hubert Schrezenmeier, Christoph Schmid, Donald Bunjes, Hans-Jochem Kolb, and Ernst Holler
- Subjects
Oncology ,medicine.medical_specialty ,Acute leukemia ,Univariate analysis ,business.industry ,medicine.medical_treatment ,Standard treatment ,Immunology ,Hazard ratio ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Biochemistry ,Surgery ,Transplantation ,Log-rank test ,Regimen ,Internal medicine ,medicine ,business - Abstract
Abstract 3328 Poster Board III-216 Introduction Relapse is a major cause of treatment failure after alloSCT against acute leukaemia, and no standard treatment has been established in this challenging situation. The introduction of reduced conditioning regimens, and the broader availability of alternative donors have increased the possibilities to perform a second alloSCT as salvage treatment, using different preparative regimen and/or different stem cell donors. Methods To evaluate the role of a second alloSCT (tx2) for the treatment of relapse after first alloSCT (tx1), we performed a nationwide retrospective analysis based on the German registry for stem cell transplantation (DRST). Datasets were completed by the reporting centres on request, following a specifically designed questionnaire. Results 212 patients (69% AML, 31% ALL), from 23 centres were included. Median age at tx1 was 37y. Donor at tx1 were HLA identical siblings (41%), matched unrelated (39%), mismatched family or unrelated (17%) or syngeneic donors (3%). Conditioning intensity at tx1 was standard (SIC, 62%), intermediate (intC, 25%) or reduced (RIC, 13%). Median remission after tx1 was 7 months, median time from relapse to tx2 was 74d. At tx2, patients were aplastic (4%), in CR (20%) or showed active disease (76%). In 59%, the same donor was used for tx1 and tx2, whereas a different donor was chosen in 41%. Conditioning at tx1/tx2 were SIC/SIC (14%), intC/intC (10%), (RIC/RIC (10%), less intensive at tx2 (mostly intC or RIC after SIC, 58%), or more intensive at tx2 (SIC after RIC or intC, 8%). Following tx2, CR was achieved in 56% of patients, out of which 81% relapsed again. Hence, leukemia was the most frequent cause of death. With a median FU of 23 months after tx2, median OS after tx2 is 117d. In a univariate analysis (log rank), OS after tx2 depended on stage at tx1 (CR vs. active disease, p12m (31%), p Conclusion Survival of acute leukemia after second allogeneic SCT is determined by the duration of remission after tx1. Using an alternative donor for tx2 did not improve the results in our series. Further analysis is required to evaluate the role of RIC regimen for tx2. Disclosures No relevant conflicts of interest to declare.
- Published
- 2009
48. T-Cell Depletion in Allogeneic Hematopoietic Cell Transplantation for Chronic Lymphocytic Leukemia: A Retrospective EBMT Analysis
- Author
-
Michel van Gelder, Johan Maertens, Liisa Volin, Donald Milligan, Peter Dreger, Aolis Gratwohl, Dietger Niederwieser, T. de Witte, Ronald Brand, Johannes Schetelig, and Martin Bornhaeuser
- Subjects
medicine.medical_specialty ,business.industry ,Proportional hazards model ,medicine.medical_treatment ,Incidence (epidemiology) ,Chronic lymphocytic leukemia ,Immunology ,Hazard ratio ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Gastroenterology ,Transplantation ,Internal medicine ,medicine ,Alemtuzumab ,Cumulative incidence ,business ,medicine.drug - Abstract
Abstract 2307 Poster Board II-284 Objectives: T-cell depletion (TCD) is controversial in patients with chronic lymphocytic leukemia (CLL). While TCD is a powerful tool to prevent graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (HCT) concern is raised about an increased incidence of relapse. The aim of this retrospective analysis was to study the impact of in vivo TCD in patients with CLL registered with the EBMT database. Patients and Methods: Patients with CLL who received allogeneic HCT from a matched sibling (SIB) or unrelated donor (UD) and cyclosporine-based GVHD prophylaxis between 2001 and 2008 were eligible. Patients who received ex vivo T-cell depleted grafts were excluded. The outcome of three major groups of patients was compared: Patients who were transplanted without TCD, those who received anti-thymocyte globulin (ATG) and patients who received alemtuzumab (CAM). Baseline and follow-up data were downloaded from the EBMT database. Results: 413 patients were eligible. 73% of patients had SIB donors and 27% matched UD. Reduced intensity conditioning regimens were applied for the majority of patients (82%). No TCD was used in 234 patients, while 100 patients received CAM and 79 patients ATG. The median follow-up after HCT was 22 months (1 to 92 months). Univariate comparisons of GVHD and the rates of DLI are reported for patients with matched sibling donors only. After SIB-HCT the cumulative incidence of acute GVHD II-IV was 7% with CAM, 22% with ATG and 34% without TCD (gray test, p=0.0001). Subsequently, 28% of patients with CAM received prophylactic donor lymphocyte infusions (DLI) compared to 5% of patients with ATG and 9% without TCD (p=0.03). DLI for any reason was given to 45% of patients with CAM, 24% with ATG and 15% without TCD (p=0.009). As a result of late-onset GVHD the incidence of chronic GVHD after TCD with CAM increased from 20% at 1 year to 60% at 4 years after SIB-HCT. At 4 years after HCT comparable cumulative incidences of chronic GVHD were observed (70% without TCD, 59% after ATG and 60% after CAM; p=0.1). For the whole cohort of patients 4-year overall survival (OS) and progression-free survival (PFS) were 57% (95% CI, 50% to 64%) and 44% (95% CI, 37% to 51%). At 4 years the cumulative incidence of relapse was 28% (95% CI, 17% to 39%) and the incidence of non-relapse mortality was 28% (95% CI 18% to 38%). In multivariate Cox regression analysis of PFS when GVHD prophylaxis without TCD was used as reference category the adjusted hazard ratio for TCD with CAM was 1.4 (95% CI, 0.9 to 2.2, p=0.2) and for TCD with ATG 1.4 (95% CI, 0.9 to 2.3, p=0.1). For relapse incidence the adjusted hazard ratio for TCD with CAM was 1.4 (95% CI, 0.7 to 2.6, p=0.3) and for TCD with ATG 1.0 (95% CI, 0.5 to 2.1, p=0.9) and for non-relapse mortality the adjusted hazard ratio for TCD with CAM was 1.4 (95% CI, 0.8 to 2.8, p=0.3) and for TCD with ATG 1.9 (95% CI, 1.0 to 3.5, p=0.06). Conclusion: In patients with CLL the combination of in vivo TCD and DLI appears to result in comparable rates of chronic GVHD and PFS compared to T-cell replete HCT. Disclosures: Schetelig: Bayer Schering: Research Funding.
- Published
- 2009
49. Early Treatment Intensification in High Risk (HR)-AML Patients Using Allogeneic Hematopoietic Stem Cell Transplantion in a Randomized Multicenter Setting.
- Author
-
Illmer, Thomas, primary, Schaich, Markus A., additional, Thiede, Christian, additional, Platzbecker, Uwe, additional, Hannes, Wandt, additional, Anthony, Ho D., additional, Eckhard, Thiel, additional, Norbert, Schmitz, additional, Hermann, Einsele, additional, Andreas, Neubauer, additional, Reinhard, Andresen, additional, Walter, Aulitzky, additional, Matthias, Haehnel, additional, Hubert, Serve, additional, Wolfgang, Berdel, additional, Martin, Bornhaeuser, additional, and Gerhard, Ehninger, additional
- Published
- 2007
- Full Text
- View/download PDF
50. Localization of Hematopoietic Stem Cells in Coculture with Mesenchymal Stromal Cells Impacts on Phenotype and Cell Cycle Status
- Author
-
Nael Alakel, Fernando A. Fierro, Rainer Ordemann, Martin Bornhaeuser, Gerhard Ehninger, Katrin Mueller, and Duohui Jing
- Subjects
Stromal cell ,Cell division ,Immunology ,Mesenchymal stem cell ,CD34 ,Cell Biology ,Hematology ,Cell cycle ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,Stem cell ,Mitosis - Abstract
Hematopoietic stem cells (HSC) are defined by their capacity of self-renewal and differentiation. In recent years it became clear that cell to cell contact mediated communication between mesenchymal stromal cells (MSC) and HSC is important for homeostasis of hematopoiesis. MSC play a crucial role in the so called bone marrow niche giving rise to the majority of marrow stromal cell lineages. In vitro we investigated the impact of MSC on CD34 purified HSC expansion and differentiation demonstrating a promoting impact of MSC on adherent HSC in comparison to non adherent HSC in terms of phenotype, migration capacity and clonogenicity. Performing phase contrast microscopy and confocal microscopy we are able to distinguish HSC which are located on the surface of a MSC monolayer (phase-bright cells) and HSC which are covered by MSC monolayer (phase-dim cells). Both HSC fractions and the non-adherent cells were isolated separately by performing serial washing steps. All three fractions were analyzed at fixed time points during the first week of co-culture in term of cell cycle progression, proliferation, maturation and cell division accompanied differentiation. First we performed propidium iodide (PI) staining for cell cycle analysis revealing that the phase-bright cells contained the highest percentage of G2 cells in comparison to the non adherent cells and the phase-dim cells; 13.9 ±1.0% vs 1.3 ±1.2% vs 2.7 ±2.0%, p
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.