21 results on '"Koschmieder S"'
Search Results
2. Effect of treatment with amifostine used as a single agent in patients with refractory anemia on clinical outcome and serum tumor necrosis factor α levels
- Author
-
Hofmann, W.-K., Seipelt, G., Ottmann, O. G., Kalina, U., Koschmieder, S., Brücher, J., Frickhofen, N., Klausmann, M., Mitrou, P. S., and Hoelzer, D.
- Published
- 2000
- Full Text
- View/download PDF
3. Effect of treatment with amifostine used as a single agent in patients with refractory anemia on clinical outcome and serum tumor necrosis factor alpha levels.
- Author
-
Hofmann, W.-K., Seipelt, G., Ottmann, O. G., Kalina, U., Koschmieder, S., Brücher, J., Frickhofen, N., Klausmann, M., Mitrou, P. S., Hoelzer, D., and Brücher, J
- Subjects
BLOOD ,BLOOD platelets ,BONE marrow ,APLASTIC anemia ,BLOOD diseases ,TUMOR necrosis factors - Abstract
Amifostine increases in vitro burst-forming unit-erythroid and colony-forming unit-granulocyte/granulocyte–macrophage cultured from bone-marrow cells from patients with myelodysplastic syndrome (MDS). Several small clinical studies give divergent informations about the potential of amifostine as single agent to improve hematopoiesis in MDS patients. In these studies, patients with refractory anemia (RA), RA with excess of blasts (RAEB), and RAEB in transformation (RAEB-T) were analyzed together, resulting in response rates varying from 8% to 30%. The present multi-center study evaluated whether treatment with amifostine is of clinical benefit in patients with RA who are transfusion dependent. The effect on transfusion frequency as well as on platelets and absolute neutrophil count (ANC) was examined in 14 patients with RA [median age 67 years (55–72 years), male :female 9:5]. Four treatment cycles were planned, each consisting of intravenous amifostine at 200 mg/m
2 /day three times per week followed by a 2-week interval. Since tumor necrosis factor (TNF)α is a main suppressive cytokine for hematopoiesis in RA patients, serum samples for analyzing endogenous levels of TNFα were collected prior to the study and after four treatment cycles. In three patients (21%), reduced transfusion requirement with prolongation of the transfusion interval from 4 weeks to 8 weeks (two patients) and 4 weeks to 6 weeks was seen. An increase in ANC from 400/μl to 2600/μl and 200/μl to 3400/μl was observed in two patients. Platelets increased from 129,000/μl to 277,000/μl in an additional patient. In one patient, disease progression from RA to RAEB was observed. Serum TNFα levels were increased in MDS patients compared with normal controls (18.8 pg/ml vs 9.1 pg/ml), and there was no change during the treatment with amifostine (17.5 pg/ml). In conclusion, treatment with amifostine as a single agent was of limited benefit in patients with RA. The serum TNFα levels were unchanged during treatment with amifostine in RA patients. [ABSTRACT FROM AUTHOR]- Published
- 2000
- Full Text
- View/download PDF
4. How I diagnose and treat patients in the pre-fibrotic phase of primary myelofibrosis (pre-PMF) - practical approaches of a German expert panel discussion in 2024.
- Author
-
Griesshammer M, Al-Ali HK, Eckardt JN, Fiegl M, Göthert J, Jentsch-Ullrich K, Koschmieder S, Kvasnicka HM, Reiter A, Schmidt B, and Heidel FH
- Abstract
The prefibrotic phase of primary myelofibrosis (pre-PMF) represents a distinct subentity within the spectrum of myeloproliferative neoplasms (MPNs), recognized by the World Health Organization (WHO) and the International Consensus Classification (ICC). Pre-PMF is characterized by unique morphological, clinical, and molecular features, distinguishing it from essential thrombocythemia (ET) and overt myelofibrosis (overt-PMF). The diagnostic process for pre-PMF relies on bone marrow histology, identification of molecular mutations and exclusion of other myeloid neoplasms. Misclassification remains a significant challenge due to overlapping phenotypes and the heterogeneity of clinical presentations, which range from asymptomatic cases to severe cytopenias and a high thrombotic risk. Management strategies for pre-PMF focus on mitigating symptom burden, reducing thromboembolic events, and preventing disease progression. Low-risk patients often benefit from observational approaches or low-dose aspirin, while cytoreductive therapies, such as hydroxyurea or interferon-alpha, are utilized in symptomatic or high-risk cases. JAK inhibitors like ruxolitinib have shown promise in addressing splenomegaly and systemic symptoms, although their role in pre-PMF requires further investigation. Advances in artificial intelligence are enhancing diagnostic precision by refining bone marrow histopathological analysis, paving the way for more accurate disease classification and tailored therapeutic strategies. This position paper integrates insights from a German expert panel discussion, underscoring the need for interdisciplinary collaboration, adherence to updated WHO/ICC diagnostic criteria, and personalized treatment approaches. By addressing diagnostic challenges and therapeutic nuances, it seeks to improve outcomes and quality of life for patients navigating the complexities of pre-PMF., Competing Interests: Declarations. Competing interests: MG reports consultancy and honoraria from Amgen, AOP Pharma, Novartis, BMS, AbbVie, Pfizer, Roche, Janssen, Gilead, AstraZeneca, Sierra, Lilly, GSK. JNE received research funding from Novartis; advisory boards or consulting fees from AstraZeneca, Johnson&Johnson, and Novartis; received honoraria from Amgen, AstraZeneca, Novartis, and Pfizer; received travel support from AstraZeneca, Johnson&Johnson, and Merck; is a co-owner of Cancilico; had a patent issued for artificial intelligence-based image classification software. SK received research funding from Geron, Janssen, AOP Pharma, and Novartis; received advisory board or consulting fees from Pfizer, Incyte, Ariad, Novartis, AOP Pharma, Bristol Myers Squibb, Celgene, Geron, Janssen, CTI BioPharma, Roche, Bayer, GSK, Sierra Oncology, PharmaEssentia, and MSD; received payment or honoraria from Novartis, BMS/Celgene, Pfizer; received travel/accommodation support from Alexion, Novartis, Bristol Myers Squibb, Incyte, AOP Pharma, CTI BioPharma, Pfizer, Celgene, Janssen, Geron, Roche, AbbVie, GSK, Sierra Oncology, Kartos, and MSD; had a patent issued for a BET inhibitor at RWTH Aachen University. BS received advisory board or consulting fees from AbbVie, AOP Pharma, AstraZeneca, BeiGene, Bristol Myers Squibb, Hexal, Incyte, Janssen-Cilag, Novartis, Oncopeptides, Pfizer, Sanofi and SoBi. FHH served as an advisor for Novartis, CTI, Celgene/BMS, Janssen, Abbvie, GSK, Merck and AOP and received research funding from Novartis, Celgene/BMS and CTI., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
5. ROP-ET: a prospective phase III trial investigating the efficacy and safety of ropeginterferon alfa-2b in essential thrombocythemia patients with limited treatment options.
- Author
-
Kiladjian JJ, Marin FF, Al-Ali HK, Alvarez-Larrán A, Beggiato E, Bieniaszewska M, Breccia M, Buxhofer-Ausch V, Cerna O, Crisan AM, Danaila CD, De Stefano V, Döhner K, Empson V, Gora-Tybor J, Griesshammer M, Grosicki S, Guglielmelli P, García-Gutierrez V, Heidel FH, Illés A, Tomuleasa C, James C, Koschmieder S, Krauth MT, Krejcy K, Lazaroiu MC, Mayer J, Nagy ZG, Nicolini FE, Palandri F, Pappa V, Reiter AJ, Sacha T, Schlager S, Schmidt S, Terpos E, Unger M, Wölfler A, Cirici BX, and Klade C
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Prospective Studies, Treatment Outcome, Multicenter Studies as Topic, Clinical Trials, Phase III as Topic, Interferon alpha-2 therapeutic use, Interferon alpha-2 adverse effects, Interferon-alpha therapeutic use, Interferon-alpha adverse effects, Polyethylene Glycols therapeutic use, Polyethylene Glycols adverse effects, Polyethylene Glycols administration & dosage, Recombinant Proteins therapeutic use, Recombinant Proteins adverse effects, Recombinant Proteins administration & dosage, Thrombocythemia, Essential drug therapy
- Abstract
Interferon-based therapies, such as ropeginterferon alfa-2b have emerged as promising disease-modifying agents for myeloproliferative neoplasms (MPNs), including essential thrombocythemia (ET). Current ET treatments aim to normalize hematological parameters and reduce the thrombotic risk, but they do not modify the natural history of the disease and hence, have no impact on disease progression. Ropeginterferon alfa-2b (trade name BESREMi®), a novel, monopegylated interferon alfa-2b with an extended administration interval, has demonstrated a robust and sustained efficacy in polycythemia vera (PV) patients. Given the similarities in disease pathophysiology and treatment goals, ropeginterferon alfa-2b holds promise as a treatment option for ET. The ROP-ET trial is a prospective, multicenter, single-arm phase III study that includes patients with ET who are intolerant or resistant to, and/or are ineligible for current therapies, such as hydroxyurea (HU), anagrelide (ANA), busulfan (BUS) and pipobroman, leaving these patients with limited treatment options. The primary endpoint is a composite response of hematologic parameters and disease-related symptoms, according to modified European LeukemiaNet (ELN) criteria. Secondary endpoints include improvements in symptoms and quality of life, molecular response and the safety profile of ropeginterferon alfa-2b. Over a 3-year period the trial assesses longer term outcomes, particularly the effects on allele burden and clinical outcomes, such as disease-related symptoms, vascular events and disease progression. No prospective clinical trial data exist for ropeginterferon alfa-2b in the planned ET study population and this study will provide new findings that may contribute to advancing the treatment landscape for ET patients with limited alternatives. TRIAL REGISTRATION: EU Clinical Trials Register; EudraCT, 2023-505160-12-00; Registered on October 30, 2023., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Bone marrow biopsy in geriatric patients above the age of 85 years: invaluable or unnecessary? A retrospective analysis.
- Author
-
Zhang KD, Jost E, Panse J, Herwartz R, Lindemann-Docter K, Jonigk D, Kricheldorf K, Köchel A, Sauerbrunn N, Brümmendorf TH, Koschmieder S, and Isfort S
- Subjects
- Humans, Aged, Retrospective Studies, Biopsy, Fluorodeoxyglucose F18, Neoplasm Staging, Bone Marrow pathology, Hodgkin Disease pathology
- Abstract
Bone marrow biopsy (BMB) is a well-established diagnostic tool for various hematological, oncological, and other medical conditions. However, treatment options for geriatric patients (pts) facing these diseases are often constrained. In this single-center, retrospective analysis we assessed the diagnostic value of BMB in geriatric pts aged ≥ 85 years and examined its impact on therapeutic decisions. We examined 156 BMB procedures in 129 pts, extracting data from the electronic patient records and applying descriptive statistical methods. Nearly half of the primary diagnostic procedures (26; 44.1%) resulted in a modification of the initially suspected diagnosis. Notably, 15 (25.4%) of these procedures, led to changes in both the diagnosis and planned interventional treatment. Among the 15 follow-up procedures (36.6%), disease progression was initially suspected based on symptoms, but BMB results excluded such progression. In lymphoma staging biopsies, only 2 (3.6%) prompted a change in therapeutic intervention. Importantly, no BMB-related complications, such as bleeding, infection or nerve damage, were reported. Median survival after BMB was 16.1 months across all pts, yet it varied based on the diagnosis and comorbidity score. The survival of pts with a change in therapy based on BMB results did not significantly differ from those who did not undergo a therapy change. In conclusion, BMB proved to be generally safe and beneficial in this geriatric cancer patient cohort beyond the age of 85 years. However, the advantages of lymphoma staging in this patient population warrant further consideration., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Real-world analysis of ruxolitinib in myelofibrosis: interim results focusing on patients who were naïve to JAK inhibitor therapy treated within the JAKoMo non-interventional, phase IV trial.
- Author
-
Koschmieder S, Isfort S, Schulte C, Jacobasch L, Geer T, Reiser M, Koenigsmann M, Heinrich B, Wehmeyer J, von der Heyde E, Tesch H, Gröschl B, Bachhuber P, Großer S, and Pahl HL
- Subjects
- Adult, Humans, Aged, Splenomegaly drug therapy, Prospective Studies, Nitriles, Treatment Outcome, Janus Kinase Inhibitors, Primary Myelofibrosis diagnosis, Primary Myelofibrosis drug therapy
- Abstract
Ruxolitinib (RUX) is a Janus kinase 1/2 inhibitor (JAKi) approved in the EU for treating disease‑related splenomegaly or symptoms in adults patients with myelofibrosis (MF). This is an interim analysis of JAKoMo, a prospective, non‑interventional, phase IV study in MF. Between 2012-2019 (cutoff March 2021), 928 patients (JAKi-naïve and -pretreated) enrolled from 122 German centers. This analysis focuses on JAKi-naïve patients. RUX was administered according to the Summary of Product Characteristics. Compared to the COMFORT-I, -II, and JUMP trials, patients in JAKoMo were older (median 73 years), had poorer Eastern Cooperative Oncology Group (ECOG) performance statuses (16.5% had ECOG ≥ 2), and were more transfusion dependent (48.5%). JAKoMo represents the more challenging patients with MF encountered outside of interventional studies. However, patients with low-risk International Prognostic Scoring System (IPSS) scores or without palpable splenomegaly were also included. Following RUX treatment, 82.5% of patients experienced rapid (≤ 1 month), significant decreases in palpable spleen size, which remained durable for 24 months (60% patients). Symptom assessment scores improved significantly in Month 1 (median -5.2) up to Month 12 (-6.2). Common adverse events (AEs) were anemia (31.2%) and thrombocytopenia (28.6%). At cutoff, 54.3% of patients had terminated the study due to, death, AEs, or deterioration of health. No new safety signals were observed. Interim analysis of the JAKoMo study confirms RUX safety and efficacy in a representative cohort of real-world, elderly, JAKi-naïve patients with MF. Risk scores were used in less than half of the patients to initiate RUX treatment.Trial registration: NCT05044026; September 14, 2021., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. Step-in dosing of bosutinib in pts with chronic phase chronic myeloid leukemia (CML) after second-generation tyrosine kinase inhibitor (TKI) therapy: results of the Bosutinib Dose Optimization (BODO) Study.
- Author
-
Isfort S, Manz K, Teichmann LL, Crysandt M, Burchert A, Hochhaus A, Saussele S, Kiani A, Göthert JR, Illmer T, Schafhausen P, Al-Ali HK, Stegelmann F, Hänel M, Pfeiffer T, Giagounidis A, Franke GN, Koschmieder S, Fabarius A, Ernst T, Warnken-Uhlich M, Wolber U, Kohn D, Pfirrmann M, Wolf D, and Brümmendorf TH
- Subjects
- Humans, Prospective Studies, Aniline Compounds adverse effects, Tyrosine Kinase Inhibitors, Leukemia, Myeloid, Chronic-Phase drug therapy
- Abstract
The approved dose of bosutinib in chronic phase CML is 400 mg QD in first-line and 500 mg QD in later-line treatment. However, given that gastrointestinal (GI) toxicity typically occurs early after treatment initiation, physicians often tend to start therapy with lower doses although this has never been tested systematically in prospective trials in the Western world. The Bosutinib Dose Optimization (BODO) Study, a multicenter phase II study, investigated the tolerability and efficacy of a step-in dosing concept of bosutinib (starting at 300 mg QD) in chronic phase CML patients in 2
nd or 3rd line who were intolerant and/or refractory to previous TKI treatment. Of 57 patients included until premature closure of the study due to slow recruitment, 34 (60%) reached the targeted dose level of 500 mg QD following the 2-weekly step-in dosing regimen. While the dosing-in concept failed to reduce GI toxicity (grade II-IV, primary study endpoint) to < 40% (overall rate of 60%; 95% CI: 45-74%), bosutinib treatment (mean dosage: 403 mg/day) showed remarkable efficacy with a cumulative major molecular remission (MMR) rate of 79% (95% CI: 66 to 88%) at month 24. Of thirty patients refractory to previous therapy and not in MMR at baseline, 19 (64%) achieved an MMR during treatment. GI toxicity did not significantly impact on patient-reported outcomes (PRO) and led to treatment discontinuation in only one patient. Overall, the results of our trial support the efficacy and safety of bosutinib after failure of second-generation TKI pre-treatment. Trial registration: NCT02577926., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
9. Efficacy and safety of ruxolitinib in patients with newly-diagnosed polycythemia vera: futility analysis of the RuxoBEAT clinical trial of the GSG-MPN study group.
- Author
-
Koschmieder S, Isfort S, Wolf D, Heidel FH, Hochhaus A, Schafhausen P, Griesshammer M, Wolleschak D, Platzbecker U, Döhner K, Jost PJ, Parmentier S, Schaich M, von Bubnoff N, Stegelmann F, Maurer A, Crysandt M, Gezer D, Kortmann M, Franklin J, Frank J, Hellmich M, and Brümmendorf TH
- Subjects
- Humans, Hydroxyurea therapeutic use, Medical Futility, Nitriles therapeutic use, Pyrimidines therapeutic use, Polycythemia Vera diagnosis, Polycythemia Vera drug therapy, Polycythemia Vera genetics, Janus Kinases therapeutic use
- Abstract
Patients (pts) with polycythemia vera (PV) suffer from pruritus, night sweats, and other symptoms, as well as from thromboembolic complications and progression to post-PV myelofibrosis. Ruxolitinib (RUX) is approved for second-line therapy in high-risk PV pts with hydroxyurea intolerance or resistance. The RuxoBEAT trial (NCT02577926, registered on October 1, 2015, at clinicaltrials.gov) is a multicenter, open-label, two-arm phase-IIb trial with a target population of 380 pts with PV or ET, randomized to receive RUX or best available therapy. This pre-specified futility analysis assesses the early clinical benefit and tolerability of RUX in previously untreated PV pts (6-week cytoreduction was allowed). Twenty-eight patients were randomly assigned to receive RUX. Compared to baseline, after 6 months of treatment, there was a significant reduction of median hematocrit (46 to 41%), the median number of phlebotomies per year (4.0 to 0), and median patient-reported pruritus scores (2 to 1), and a trend for reduced night sweat scores (1.5 to 0). JAK2V617F allele burden, as part of the scientific research program, also significantly decreased. One hundred nine adverse events (AEs) occurred in 24/28 patients (all grade 1 to 3), and no pt permanently discontinued treatment because of AEs. Thus, treatment with ruxolitinib in untreated PV pts is feasible, well-tolerated, and efficient regarding the above-mentioned endpoints., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. Clonogenic assays improve determination of variant allele frequency of driver mutations in myeloproliferative neoplasms.
- Author
-
Kalmer M, Pannen K, Lemanzyk R, Wirths C, Baumeister J, Maurer A, Kricheldorf K, Schifflers J, Gezer D, Isfort S, Brümmendorf TH, Koschmieder S, and Chatain N
- Subjects
- Humans, Calreticulin genetics, Calreticulin metabolism, Gene Frequency, Mutation, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders genetics, Neoplasms
- Abstract
Molecular diagnostics moves more into focus as technology advances. In patients with myeloproliferative neoplasms (MPN), identification and monitoring of the driver mutations have become an integral part of diagnosis and monitoring of the disease. In some patients, none of the known driver mutations (JAK2V617F, CALR, MPL) is found, and they are termed "triple negative" (TN). Also, whole-blood variant allele frequency (VAF) of driver mutations may not adequately reflect the VAF in the stem cells driving the disease. We reasoned that colony forming unit (CFU) assay-derived clonogenic cells may be better suited than next-generation sequencing (NGS) of whole blood to detect driver mutations in TN patients and to provide a VAF of disease-driving cells. We have included 59 patients carrying the most common driver mutations in the establishment or our model. Interestingly, cloning efficiency correlated with whole blood VAF (p = 0.0048), suggesting that the number of disease-driving cells correlated with VAF. Furthermore, the clonogenic VAF correlated significantly with the NGS VAF (p < 0.0001). This correlation was lost in patients with an NGS VAF <15%. Further analysis showed that in patients with a VAF <15% by NGS, clonogenic VAF was higher than NGS VAF (p = 0.003), suggesting an enrichment of low numbers of disease-driving cells in CFU assays. However, our approach did not enhance the identification of driver mutations in 5 TN patients. A significant correlation of lactate dehydrogenase (LDH) serum levels with both CFU- and NGS-derived VAF was found. Our results demonstrate that enrichment for clonogenic cells can improve the detection of MPN driver mutations in patients with low VAF and that LDH levels correlate with VAF., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Towards personalized medicine with iPS cell technology: a case report of advanced systemic mastocytosis with associated eosinophilia.
- Author
-
Atakhanov S, Christen D, Rolles B, Schüler HM, Panse J, Chatain N, Koschmieder S, Brümmendorf TH, Toledo MAS, and Zenke M
- Subjects
- Humans, Precision Medicine, Proto-Oncogene Proteins c-kit, Eosinophilia complications, Induced Pluripotent Stem Cells, Mastocytosis, Systemic complications
- Published
- 2022
- Full Text
- View/download PDF
12. Early and late stage MPN patients show distinct gene expression profiles in CD34 + cells.
- Author
-
Baumeister J, Maié T, Chatain N, Gan L, Weinbergerova B, de Toledo MAS, Eschweiler J, Maurer A, Mayer J, Kubesova B, Racil Z, Schuppert A, Costa I, Koschmieder S, Brümmendorf TH, and Gezer D
- Subjects
- Gene Expression Regulation, Neoplastic, Humans, Polycythemia Vera genetics, Primary Myelofibrosis genetics, Thrombocythemia, Essential genetics, Antigens, CD34 genetics, Myeloproliferative Disorders genetics, Transcriptome
- Abstract
Myeloproliferative neoplasms (MPN), comprising essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), are hematological disorders of the myeloid lineage characterized by hyperproliferation of mature blood cells. The prediction of the clinical course and progression remains difficult and new therapeutic modalities are required. We conducted a CD34
+ gene expression study to identify signatures and potential biomarkers in the different MPN subtypes with the aim to improve treatment and prevent the transformation from the rather benign chronic state to a more malignant aggressive state. We report here on a systematic gene expression analysis (GEA) of CD34+ peripheral blood or bone marrow cells derived from 30 patients with MPN including all subtypes (ET (n = 6), PV (n = 11), PMF (n = 9), secondary MF (SMF; post-ET-/post-PV-MF; n = 4)) and six healthy donors. GEA revealed a variety of differentially regulated genes in the different MPN subtypes vs. controls, with a higher number in PMF/SMF (200/272 genes) than in ET/PV (132/121). PROGENγ analysis revealed significant induction of TNFα/NF-κB signaling (particularly in SMF) and reduction of estrogen signaling (PMF and SMF). Consistently, inflammatory GO terms were enriched in PMF/SMF, whereas RNA splicing-associated biological processes were downregulated in PMF. Differentially regulated genes that might be utilized as diagnostic/prognostic markers were identified, such as AREG, CYBB, DNTT, TIMD4, VCAM1, and S100 family members (S100A4/8/9/10/12). Additionally, 98 genes (including CLEC1B, CMTM5, CXCL8, DACH1, and RADX) were deregulated solely in SMF and may be used to predict progression from early to late stage MPN., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
13. Macrophage frequency in the bone marrow correlates with morphologic subtype of myeloproliferative neoplasm.
- Author
-
Molitor DCA, Boor P, Buness A, Schneider RK, Teichmann LL, Körber RM, Horvath GL, Koschmieder S, and Gütgemann I
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cohort Studies, Humans, Middle Aged, Neoplasm Grading methods, Polycythemia Vera pathology, Primary Myelofibrosis pathology, Thrombocythemia, Essential pathology, Young Adult, Bone Marrow pathology, Macrophages pathology, Myeloproliferative Disorders pathology
- Abstract
Bone marrow (BM) fibrosis in myeloproliferative neoplasms (MPNs) is associated with a poor prognosis. The development of myelofibrosis and differentiation of mesenchymal stromal cells to profibrotic myofibroblasts depends on macrophages. Here, we compared macrophage frequencies in BM biopsies of MPN patients and controls (patients with non-neoplastic processes), including primary myelofibrosis (PMF, n = 18), essential thrombocythemia (ET, n = 14), polycythemia vera (PV, n = 12), and Philadelphia chromosome-positive chronic myeloid leukemia (CML, n = 9). In PMF, CD68-positive macrophages were greatly increased compared to CML (p = 0.017) and control BM (p < 0.001). Similar findings were observed by CD163 staining (PMF vs. CML: p = 0.017; PMF vs. control: p < 0.001). Moreover, CD68-positive macrophages were increased in PV compared with ET (p = 0.009) and reactive cases (p < 0.001). PMF had higher frequencies of macrophages than PV (CD68: p < 0.001; CD163: p < 0.001) and ET (CD68: p < 0.001; CD163: p < 0.001). CD163 and CD68 were often co-expressed in macrophages with stellate morphology in Philadelphia chromosome-negative MPN, resulting in a sponge-like reticular network that may be a key regulator of unbalanced hematopoiesis in the BM space and may explain differences in cellularity and clinical course.
- Published
- 2021
- Full Text
- View/download PDF
14. Transcriptional alteration of DNA repair genes in Philadelphia chromosome negative myeloproliferative neoplasms.
- Author
-
Kirschner M, Bornemann A, Schubert C, Gezer D, Kricheldorf K, Isfort S, Brümmendorf TH, Schemionek M, Chatain N, Skorski T, and Koschmieder S
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Mutation, Philadelphia Chromosome, DNA Repair, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Hematologic Neoplasms pathology, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Transcription, Genetic
- Abstract
Philadelphia negative (Ph-neg) myeloproliferative neoplasms (MPN) are a heterogenous group of clonal stem cell disorders. Approved treatment options include hydroxyurea, anagrelide, and ruxolitinib, which are not curative. The concept of synthetic lethality may become an additional therapeutic strategy in these diseases. In our study, we show that DNA repair is altered in classical Ph-neg MPN, as analyzed by gene expression analysis of 11 genes involved in the homologous recombination repair pathway (HRR), the non-homologous end-joining pathway (NHEJ), and the single-strand break repair pathway (SSB). Altogether, peripheral blood-derived cells from 57 patients with classical Ph-neg MPN and 13 healthy controls were analyzed. LIG3 as an essential part of the SSB was significantly lower expressed compared to controls in all three entities (essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF)). In addition, while genes of other DNA-repair pathways showed-possibly compensatory-increased expression in ET (HRR, NHEJ) and PV (NHEJ), MF samples displayed downregulation of all genes involved in NHEJ. With regard to the JAK2 mutational status (analyzed in ET and MF only), no upregulation of the HRR was detected. Though further studies are needed, based on these findings, we conclude that synthetic lethality may become a promising strategy in treating patients with Ph-neg MPN.
- Published
- 2019
- Full Text
- View/download PDF
15. Phase II clinical trial of pazopanib in patients with acute myeloid leukemia (AML), relapsed or refractory or at initial diagnosis without an intensive treatment option (PazoAML).
- Author
-
Kessler T, Koschmieder S, Schliemann C, Crysandt M, Mikesch JH, von Stillfried S, Stelljes M, Pohlen M, Lenz G, Kirsch A, Vehring K, Wardelmann E, Hartmann W, Bormann E, Gerss J, Brümmendorf TH, Müller-Tidow C, and Berdel WE
- Subjects
- Aged, Aged, 80 and over, Angiogenesis Inhibitors adverse effects, Antineoplastic Agents adverse effects, Bone Marrow blood supply, Female, Gastrointestinal Diseases chemically induced, Humans, Indazoles, Kaplan-Meier Estimate, Male, Microvessels drug effects, Middle Aged, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Pyrimidines adverse effects, Recurrence, Salvage Therapy, Sulfonamides adverse effects, Treatment Outcome, Tumor Microenvironment drug effects, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, Sulfonamides therapeutic use
- Abstract
We evaluated pazopanib (800 mg orally QD) in patients not eligible for intensive treatment with relapsed/refractory AML or at initial diagnosis. Patients receiving pazopanib for > 14 days were analyzed for safety, tolerability, and efficacy. Co-primary endpoints were cumulative response rate and reduction of bone marrow microvessel density. Twenty patients (median age 76 years, range 52-86) were treated. Fifteen had relapsed/refractory and five had newly diagnosed AML. Median ECOG performance status was 1 (range 1-3). Four patients had adverse, 15 intermediate, and 1 patient favorable cytogenetic/molecular risk (ELN 2010 criteria). The safety profile of pazopanib was as reported. The most common adverse events of any grade were gastrointestinal. Two patients achieved PR (blast reduction > 50%), 14 stable disease (SD), and 4 progressive disease. Median PFS was 65 days (95% CI 29-105). After the end of the study, 1 CRi and 1 CRp occurred on demethylating agents, and 1 CR upon alloSCT. In these patients, SD and improved general condition on pazopanib allowed therapy escalation. Median OS for the overall study population was 191 days (95% CI 87-435) and 1-year survival was 35%. There was no significant change in microvessel density. Clinical trial information: NCT01361334.
- Published
- 2019
- Full Text
- View/download PDF
16. The impact of myeloproliferative neoplasms (MPNs) on patient quality of life and productivity: results from the international MPN Landmark survey.
- Author
-
Harrison CN, Koschmieder S, Foltz L, Guglielmelli P, Flindt T, Koehler M, Mathias J, Komatsu N, Boothroyd RN, Spierer A, Perez Ronco J, Taylor-Stokes G, Waller J, and Mesa RA
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Quality of Life, Cost of Illness, Myeloproliferative Disorders therapy, Physician-Patient Relations
- Abstract
Myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPNs) associated with high disease burden, reduced quality of life (QOL), and shortened survival. To assess how MPNs affect patients, we conducted a global MPN Landmark survey. This online survey of patients with MPNs and physicians was conducted in Australia, Canada, Germany, Japan, Italy, and the United Kingdom. The survey measured MPN-related symptoms and the impact of MPNs on QOL and the ability to work as well as disease-management strategies. Overall, 219 physicians and 699 patients (MF, n = 174; PV, n = 223; ET, n = 302) completed the survey; 90% of patients experienced MPN-related symptoms. The most frequent and severe symptom was fatigue. Most patients experienced a reduction in QOL, including those with low symptom burden or low-risk scores. A substantial proportion of patients reported impairment at work and in overall activity. Interestingly, physician feedback and blood counts were the most important indicators of treatment success among patients, with improvements in symptoms and QOL being less important. Regarding disease management, our study revealed a lack of alignment between physician and patient perceptions relating to communication and disease management, with patients often having different treatment goals than physicians. Overall, our study suggested that therapies that reduce symptom burden and improve QOL in patients with MPNs are crucial in minimizing disease impact on patient daily lives. Additionally, our findings showed a need for improved patient-physician communication, standardized monitoring of symptoms, and agreement on treatment goals.
- Published
- 2017
- Full Text
- View/download PDF
17. Health care setting and severity, symptom burden, and complications in patients with Philadelphia-negative myeloproliferative neoplasms (MPN): a comparison between university hospitals, community hospitals, and office-based physicians.
- Author
-
Kaifie A, Isfort S, Gattermann N, Hollburg W, Klausmann M, Wolf D, Maintz C, Hänel M, Goekkurt E, Göthert JR, Platzbecker U, Geer T, Parmentier S, Jost E, Serve H, Ehninger G, Berdel WE, Brümmendorf TH, and Koschmieder S
- Subjects
- Aged, Aged, 80 and over, Chi-Square Distribution, Delivery of Health Care methods, Female, Hospitals, Community statistics & numerical data, Hospitals, University statistics & numerical data, Humans, Male, Middle Aged, Myeloproliferative Disorders complications, Myeloproliferative Disorders genetics, Outcome Assessment, Health Care methods, Outcome Assessment, Health Care statistics & numerical data, Philadelphia Chromosome, Physicians statistics & numerical data, Physicians' Offices statistics & numerical data, Prospective Studies, Registries statistics & numerical data, Risk Factors, Symptom Assessment methods, Delivery of Health Care statistics & numerical data, Myeloproliferative Disorders therapy, Severity of Illness Index, Symptom Assessment statistics & numerical data
- Abstract
Philadelphia-negative myeloproliferative neoplasms (MPN) comprise a heterogeneous group of chronic hematological malignancies with significant variations in clinical characteristics. Due to the long survival and the feasibility of oral or subcutaneous therapy, these patients are frequently treated outside of larger academic centers. This analysis was performed to elucidate differences in MPN patients in three different health care settings: university hospitals (UH), community hospitals (CH), and office-based physicians (OBP). The MPN registry of the Study Alliance Leukemia is a non-interventional prospective study including adult patients with an MPN according to WHO criteria (2008). For statistical analysis, descriptive methods and tests for significant differences were used. Besides a different distribution of MPN subtypes between the settings, patients contributed by UH showed an impaired medical condition, a higher comorbidity burden, and more vascular complications. In the risk group analyses, the majority of polycythemia vera (PV) and essential thrombocythemia (ET) patients from UH were classified into the high-risk category due to previous vascular events, while for PV and ET patients in the CH and OBP settings, age was the major parameter for a high-risk categorization. Regarding MPN-directed therapy, PV patients from the UH setting were more likely to receive ruxolitinib within the framework of a clinical trial. In summary, the characteristics and management of patients differed significantly between the three health care settings with a higher burden of vascular events and comorbidities in patients contributed by UH. These differences need to be taken into account for further analyses and design of clinical trials.
- Published
- 2016
- Full Text
- View/download PDF
18. Diagnosis, prevention, and management of bleeding episodes in Philadelphia-negative myeloproliferative neoplasms: recommendations by the Hemostasis Working Party of the German Society of Hematology and Medical Oncology (DGHO) and the Society of Thrombosis and Hemostasis Research (GTH).
- Author
-
Appelmann I, Kreher S, Parmentier S, Wolf HH, Bisping G, Kirschner M, Bergmann F, Schilling K, Brümmendorf TH, Petrides PE, Tiede A, Matzdorff A, Griesshammer M, Riess H, and Koschmieder S
- Subjects
- Anticoagulants adverse effects, Anticoagulants therapeutic use, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Blood Coagulation Factors therapeutic use, Blood Loss, Surgical prevention & control, Blood Transfusion, Blood Vessels drug effects, Blood Vessels pathology, Clinical Trials as Topic, Contraindications, Deamino Arginine Vasopressin therapeutic use, Disease Management, Elective Surgical Procedures, Hemorrhage diagnosis, Hemorrhage prevention & control, Hemorrhage therapy, Humans, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative complications, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative drug therapy, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative genetics, Liver physiopathology, Multicenter Studies as Topic, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Platelet Aggregation Inhibitors adverse effects, Platelet Aggregation Inhibitors therapeutic use, Platelet Function Tests, Platelet Transfusion, Postoperative Hemorrhage etiology, Postoperative Hemorrhage prevention & control, Postoperative Hemorrhage therapy, Thrombophilia drug therapy, Thrombophilia etiology, Tranexamic Acid therapeutic use, von Willebrand Diseases etiology, von Willebrand Factor analysis, Hemorrhage etiology, Hemostatic Techniques, Myeloproliferative Disorders complications
- Abstract
Philadelphia-negative myeloproliferative neoplasms (Ph-negative MPN) comprise a heterogeneous group of chronic hematologic malignancies. The quality of life, morbidity, and mortality of patients with MPN are primarily affected by disease-related symptoms, thromboembolic and hemorrhagic complications, and progression to myelofibrosis and acute leukemia. Major bleeding represents a common and important complication in MPN, and the incidence of such bleeding events will become even more relevant in the future due to the increasing disease prevalence and survival of MPN patients. This review discusses the causes, differential diagnoses, prevention, and management of bleeding episodes in patients with MPN, aiming at defining updated standards of care in these often challenging situations.
- Published
- 2016
- Full Text
- View/download PDF
19. Prophylaxis and management of venous thromboembolism in patients with myeloproliferative neoplasms: consensus statement of the Haemostasis Working Party of the German Society of Hematology and Oncology (DGHO), the Austrian Society of Hematology and Oncology (ÖGHO) and Society of Thrombosis and Haemostasis Research (GTH e.V.).
- Author
-
Kreher S, Ochsenreither S, Trappe RU, Pabinger I, Bergmann F, Petrides PE, Koschmieder S, Matzdorff A, Tiede A, Griesshammer M, and Riess H
- Subjects
- Anticoagulants administration & dosage, Anticoagulants adverse effects, Anticoagulants pharmacokinetics, Disease Susceptibility, Drug Interactions, Female, Hemorrhage chemically induced, Hemorrhage prevention & control, Heparin, Low-Molecular-Weight administration & dosage, Heparin, Low-Molecular-Weight adverse effects, Heparin, Low-Molecular-Weight therapeutic use, Humans, Hydroxyurea therapeutic use, Incidence, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative blood, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative complications, Male, Myeloproliferative Disorders blood, Myeloproliferative Disorders therapy, Phlebotomy, Platelet Aggregation Inhibitors administration & dosage, Platelet Aggregation Inhibitors adverse effects, Platelet Aggregation Inhibitors therapeutic use, Postoperative Complications prevention & control, Pregnancy, Pregnancy Complications, Hematologic drug therapy, Pregnancy Complications, Hematologic prevention & control, Preoperative Care, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors therapeutic use, Quinazolines therapeutic use, Secondary Prevention, Thrombophilia etiology, Venous Thromboembolism epidemiology, von Willebrand Diseases etiology, von Willebrand Diseases physiopathology, Anticoagulants therapeutic use, Myeloproliferative Disorders complications, Thrombophilia drug therapy, Venous Thromboembolism prevention & control
- Abstract
Patients with Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) like polycythemia vera and essential thrombocythemia are at increased risk of arterial and venous thrombosis. Strategies of prevention may consist of platelet aggregation inhibitors and/or cytoreductive agents depending on the underlying disease and the individual risk. Clinical evidence for management of acute venous thromboembolic events in MPN patients is limited. Modality and duration of therapeutic anticoagulation after venous thrombosis has to be evaluated critically with special regard to the increased risk for spontaneous bleeding events associated with the underlying diseases. Both for therapy of the acute event and for secondary prophylaxis, low-molecular-weight heparins should preferentially be used. A prolongation of the therapeutic anticoagulation beyond the usual 3 to 6 months can only be recommended in high-risk settings and after careful evaluation of potential risks and benefits for the individual patient. New direct oral anticoagulants (NOAC) should not preferentially be used due to lack of clinical experience in patients with MPN and potential drug interactions (e.g. with JAK inhibitors). Consequent treatment of the underlying myeloproliferative disease and periodical evaluation of the response to therapy is crucial for optimal secondary prophylaxis of thromboembolic events in those patients.
- Published
- 2014
- Full Text
- View/download PDF
20. Long-term results of a prospective randomized trial evaluating G-CSF priming in intensive induction chemotherapy followed by autologous stem cell transplantation in elderly patients with acute myeloid leukemia.
- Author
-
Bug G, Koschmieder S, Krauter J, Heuser M, Thol F, Wiebe S, Hofmann WK, Klein SA, Wegener G, Göhring G, Heit W, Hoelzer D, Ganser A, and Ottmann OG
- Subjects
- Aged, Aged, 80 and over, Autografts, Disease-Free Survival, Female, Follow-Up Studies, Granulocyte Colony-Stimulating Factor administration & dosage, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Mutation, Prospective Studies, Survival Rate, Time Factors, fms-Like Tyrosine Kinase 3 genetics, Granulocyte Colony-Stimulating Factor adverse effects, Hematopoietic Stem Cell Transplantation, Induction Chemotherapy adverse effects, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute therapy
- Abstract
Few studies have evaluated granulocyte colony-stimulating factor (G-CSF) priming in elderly patients with intensively treated acute myeloid leukemia (AML), and no data are available for genetically defined AML subgroups. We provide long-term results (median follow-up 7.6 years) of a randomized trial in which 183 patients (median age 67 years) received G-CSF prior to (G-CSF priming) or after two cycles of induction chemotherapy. CR rates with G-CSF priming and G-CSF post-chemotherapy were comparable (57 vs. 67 %, p = 0.153), with overall survival (OS) probabilities of 14 vs. 17 % at 10 years. Induction mortality was significantly higher with G-CSF priming (23 vs. 10 %, p = 0.015), primarily in normal karyotype (NK) AML. In this subgroup, a trend for better relapse-free survival (RFS) was observed with G-CSF priming (44 vs. 22 % at 10 years, p = 0.074) but did not translate into an OS benefit. G-CSF priming had no impact on AML with FLT3-ITD and NPM mutations and did not improve outcome in patients with adverse cytogenetics. In a landmark analysis, late consolidation with autologous stem cell transplantation or a second consolidation cycle significantly improved RFS compared with one consolidation cycle (21.0 vs. 12.8 months, p = 0.046). Future studies on G-CSF priming should be restricted to NK AML and used only in post-remission therapy.
- Published
- 2014
- Full Text
- View/download PDF
21. POEMS syndrome treated with melphalan high-dose therapy and autologous blood stem cell transplantation: a single-institution experience.
- Author
-
Thoennissen GB, Thoennissen NH, Fritz F, Hilbig A, Kerkhoff A, Liersch R, Krug U, Koschmieder S, Müller-Tidow C, Mesters R, Kropff M, and Berdel WE
- Subjects
- Adult, Anthracyclines therapeutic use, Combined Modality Therapy, Dexamethasone therapeutic use, Female, Humans, Male, Melphalan administration & dosage, Middle Aged, Myeloablative Agonists administration & dosage, Neurologic Examination, POEMS Syndrome blood, POEMS Syndrome surgery, Transplantation, Autologous, Treatment Outcome, Vascular Endothelial Growth Factor A blood, Melphalan therapeutic use, Myeloablative Agonists therapeutic use, POEMS Syndrome drug therapy, Peripheral Blood Stem Cell Transplantation
- Abstract
The acronym POEMS syndrome stands for a rare multi-system disorder, comprised of polyneuropathy, organomegaly, endocrinopathy, M protein, and skin changes. Here, we present a single-center report of a series of five POEMS patients treated with melphalan high-dose therapy (HDT) with subsequent autologous blood stem cell transplantation (ABSCT). After a median follow-up of 52 months from time of diagnosis (range, 15-192) and a median follow-up of 18 months after ABSCT (range, 11-120), all patients were alive. Overall, no severe transplantation-associated complications such as engraftment syndrome or peri- or post-transplant death were noted. In two cases, HDT followed by ABSCT resulted in a complete hematologic response; in the additional three cases, partial responses (PR) were achieved including one very good hematologic PR. Only one patient with initial PR developed progressive disease nearly 2.5 years after transplantation. Consequently, a second HDT with ABSCT was successfully applied resulting in clinical improvement and hematologic PR. In line with previous single-center reports, melphalan HDT followed by ABSCT proved to be a first-line treatment option with tolerable side effects in severely affected POEMS patients with progressing symptoms.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.