28 results on '"Florian Heidel"'
Search Results
2. PB1774: PHARMACOLOGICAL STRATEGIES TO OVERCOME RESISTANCE IN AML CELL LINE MODELS HARBORING ACTIVATING FLT3-ITD AND PTPN11 MUTATIONS
- Author
-
Maximilian Fleischmann, Nathalie Urban, Ole Hansen, Ulf Schnetzke, Florian Heidel, Andreas Hochhaus, Joerg Mueller, and Sebastian Scholl
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. P1028: MYELOFIBROSIS (MF) TREATED WITH RUXOLITINIB (RUX) MONOTHERAPY: PREDICTORS OF EARLY DISCONTINUATION AND DEATH ON TREATMENT. THE SHORT-TERM RUX PROGNOSTIC MODEL (STR-PM)
- Author
-
Francesca Palandri, Giuseppe Auteri, Alessandra Iurlo, Elena Maria Elli, Simona Paglia, Massimiliano Bonifacio, Mario Tiribelli, Malgorzata Monica Trawinska, Nicola Polverelli, Giulia Benevolo, Alessia Tieghi, Florian Heidel, Fabrizio Cavalca, Giovanni Caocci, Eloise Beggiato, Gianni Binotto, Francesco Cavazzini, Maurizio Miglino, Costanza Bosi, Monica Crugnola, Monica Bocchia, Bruno Martino, Novella Pugliese, Marta Venturi, Camilla Mazzoni, Alessandro Isidori, Daniele Cattaneo, Mauro Krampera, Fabrizio Pane, Daniela Cilloni, Gianpietro Semenzato, Roberto M. Lemoni, Antonio Cuneo, Elisabetta Abruzzese, Nicola Vianelli, Michele Cavo, Giuseppe Palumbo, and Massimo Breccia
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
4. PB2681: GAPS IN THE ASSESSMENT AND MONITORING OF CARDIOVASCULAR RISK AND PSYCHOLOGICAL BURDEN IN POLYCYTHEMIA VERA: LANDMARK 2.0: A WORLDWIDE HEALTH SURVEY
- Author
-
Claire Harrison, David Ross, Laura Fogliatto, Lynda Foltz, Lambert Busque, Zhijian Xiao, Florian Heidel, Michael Koehler, Giuseppe A. Palumbo, Massimo Breccia, Norio Komatsu, Keita Kirito, Blanca Xicoy Cirici, Joaquín Martinez-Lopez, Alicia Rovó, Cheryl Petruk, Catalin Bobirca, Laura Mirams, Abigail Mcmillan, Gavin Harper, and Jean-Jacques Kiladjian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
5. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis
- Author
-
Francesca Palandri, Giuseppe Alberto Palumbo, Elena Maria Elli, Nicola Polverelli, Giulia Benevolo, Bruno Martino, Elisabetta Abruzzese, Mario Tiribelli, Alessia Tieghi, Roberto Latagliata, Francesco Cavazzini, Micaela Bergamaschi, Gianni Binotto, Monica Crugnola, Alessandro Isidori, Giovanni Caocci, Florian Heidel, Novella Pugliese, Costanza Bosi, Daniela Bartoletti, Giuseppe Auteri, Daniele Cattaneo, Luigi Scaffidi, Malgorzata Monica Trawinska, Rossella Stella, Fiorella Ciantia, Fabrizio Pane, Antonio Cuneo, Mauro Krampera, Gianpietro Semenzato, Roberto Massimo Lemoli, Alessandra Iurlo, Nicola Vianelli, Michele Cavo, Massimo Breccia, and Massimiliano Bonifacio
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
6. Intracellular retention of ABL kinase inhibitors determines commitment to apoptosis in CML cells.
- Author
-
Daniel B Lipka, Marie-Christine Wagner, Marek Dziadosz, Tina Schnöder, Florian Heidel, Mirle Schemionek, Junia V Melo, Thomas Kindler, Carsten Müller-Tidow, Steffen Koschmieder, and Thomas Fischer
- Subjects
Medicine ,Science - Abstract
Clinical development of imatinib in CML established continuous target inhibition as a paradigm for successful tyrosine kinase inhibitor (TKI) therapy. However, recent reports suggested that transient potent target inhibition of BCR-ABL by high-dose TKI (HD-TKI) pulse-exposure is sufficient to irreversibly commit cells to apoptosis. Here, we report a novel mechanism of prolonged intracellular TKI activity upon HD-TKI pulse-exposure (imatinib, dasatinib) in BCR-ABL-positive cells. Comprehensive mechanistic exploration revealed dramatic intracellular accumulation of TKIs which closely correlated with induction of apoptosis. Cells were rescued from apoptosis upon HD-TKI pulse either by repetitive drug wash-out or by overexpression of ABC-family drug transporters. Inhibition of ABCB1 restored sensitivity to HD-TKI pulse-exposure. Thus, our data provide evidence that intracellular drug retention crucially determines biological activity of imatinib and dasatinib. These studies may refine our current thinking on critical requirements of TKI dose and duration of target inhibition for biological activity of TKIs.
- Published
- 2012
- Full Text
- View/download PDF
7. Therapiestrategien bei myeloproliferativen Neoplasien
- Author
-
Florian Heidel
- Published
- 2022
8. Data from Mitochondrial BAX Determines the Predisposition to Apoptosis in Human AML
- Author
-
Frank Edlich, Florian Heidel, Jens U. Marquardt, Konstanze Döhner, Denise Wolleschak, Franziska Todt, Peter Scholz-Kreisel, Kathrin Funk, Diana Becker, Enrico Schalk, Cornelius Wiedenmann, and Frank Reichenbach
- Abstract
Purpose: Cell-to-cell variability in apoptosis signaling contributes to heterogenic responses to cytotoxic stress in clinically heterogeneous neoplasia, such as acute myeloid leukemia (AML). The BCL-2 proteins BAX and BAK can commit mammalian cells to apoptosis and are inhibited by retrotranslocation from the mitochondria into the cytosol. The subcellular localization of BAX and BAK could determine the cellular predisposition to apoptotic death.Experimental Design: The relative localization of BAX and BAK was determined by fractionation of AML cell lines and patient samples of a test cohort and a validation cohort.Results: This study shows that relative BAX localization determines the predisposition of different AML cell lines to apoptosis. Human AML displays a surprising variety of relative BAX localizations. In a test cohort of 48 patients with AML, mitochondria-shifted BAX correlated with improved patient survival, FLT3-ITD status, and leukocytosis. Analysis of a validation cohort of 80 elderly patients treated with myelosuppressive chemotherapy confirmed that relative BAX localization correlates with probability of disease progression, FLT3-ITD status, and leukocytosis. Relative BAX localization could therefore be helpful to identify elderly or frail patients who may benefit from cytotoxic therapy.Conclusions: In this retrospective analysis of two independent AML cohorts, our data suggest that Bax localization may predict prognosis of patients with AML and cellular predisposition to apoptosis, combining the actual contribution of known and unknown factors to a final “common path.” Clin Cancer Res; 23(16); 4805–16. ©2017 AACR.
- Published
- 2023
9. Tables S1 - 5 from Mitochondrial BAX Determines the Predisposition to Apoptosis in Human AML
- Author
-
Frank Edlich, Florian Heidel, Jens U. Marquardt, Konstanze Döhner, Denise Wolleschak, Franziska Todt, Peter Scholz-Kreisel, Kathrin Funk, Diana Becker, Enrico Schalk, Cornelius Wiedenmann, and Frank Reichenbach
- Abstract
S1 BAX/BAK localization for the test cohort S2 clinical characterization of patient cohorts S3 GISTIC analysis of the test cohort S4 BAX/BAK localization for the validation cohort S5 Multivariate analysis of variance for human AML survival probablity
- Published
- 2023
10. Adressen
- Author
-
Kurt Possinger, Anne C. Regierer, Jan Eucker, Annette Dieing, Bernd Flath, Gunnar Folprecht, Michael Geißler, Florian Heidel, Erhard Hiller, Andreas Hochhaus, Kai Hübel, Henning Jann, Christian Jehn, Ulrich Keilholz, Konrad Klinghammer, Wolfgang Knauf, Hans-Jochem Kolb, Hannes Kroenlein, Anne Sophie Kubasch, Diana Lüftner, Matthias Möhlig, Ralph Naumann, Helmut Oettle, null Oettle, null Mayer, Ulrich-Frank Pape, Uwe Platzbecker, Andreas Rank, Peter Reichardt, Oliver Rick, Damian Rieke, Hanno Riess, Markus Ruhnke, Markus Schaich, Andreas Schalhorn, Ann-Kristin Schmälter, Alexander Schmittel, Christian Scholz, Hubert Schrezenmeier, Carsten-Oliver Schulz, Dorothee Speiser, and Bertram Wiedenmann
- Published
- 2020
11. Gynäkologische Tumoren
- Author
-
Kurt Possinger, Anne C. Regierer, Jan Eucker, Annette Dieing, Bernd Flath, Gunnar Folprecht, Michael Geißler, Florian Heidel, Erhard Hiller, Andreas Hochhaus, Kai Hübel, Henning Jann, Christian Jehn, Ulrich Keilholz, Konrad Klinghammer, Wolfgang Knauf, Hans-Jochem Kolb, Hannes Kroenlein, Anne Sophie Kubasch, Diana Lüftner, Matthias Möhlig, Ralph Naumann, Helmut Oettle, Ulrich-Frank Pape, Uwe Platzbecker, Andreas Rank, Peter Reichardt, Oliver Rick, Damian Rieke, Hanno Riess, Markus Ruhnke, Markus Schaich, Andreas Schalhorn, Ann-Kristin Schmälter, Alexander Schmittel, Christian Scholz, Hubert Schrezenmeier, Carsten-Oliver Schulz, Dorothee Speiser, and Bertram Wiedenmann
- Published
- 2020
12. Chronische myeloproliferative Neoplasien (CMPN)
- Author
-
Florian Heidel and Andreas Hochhaus
- Published
- 2020
13. 1014 – POST-TRANSLATIONAL MODIFICATION OF SPLICING FACTORS IN JAK2-MUTATED MPN
- Author
-
Florian Heidel
- Subjects
Cancer Research ,Genetics ,Cell Biology ,Hematology ,Molecular Biology - Published
- 2021
14. Dendritic crystallization in hydrous basaltic magmas controls magma mobility within the Earth’s crust
- Author
-
Fabio Arzilli, Margherita Polacci, Giuseppe La Spina, Nolwenn Le Gall, Edward W. Llewellin, Richard A. Brooker, Rafael Torres-Orozco, Danilo Di Genova, David A. Neave, Margaret E. Hartley, Heidy M. Mader, Daniele Giordano, Robert Atwood, Peter D. Lee, Florian Heidelbach, and Mike R. Burton
- Subjects
Science - Abstract
In situ 4D experiments at high temperature and moderate pressure reveal that rapid dendritic crystallization in hydrous basaltic magmas promotes a rheological transition within minutes, controlling magma mobility within the Earth’s crust.
- Published
- 2022
- Full Text
- View/download PDF
15. Novel pathway in Bcr-Abl signal transduction involves Akt-independent, PLC-γ1-driven activation of mTOR/p70S6-kinase pathway
- Author
-
Junia V. Melo, Daniel B. Lipka, Frank Breitenbuecher, Corinna Albers, Florian Heidel, Tim H. Brümmendorf, Justus Duyster, C. Huber, Boyka Markova, and Thomas Fischer
- Subjects
Cancer Research ,Blotting, Western ,Medizin ,Fusion Proteins, bcr-abl ,Apoptosis ,Protein Serine-Threonine Kinases ,Biology ,Piperazines ,Mice ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Genetics ,Animals ,Humans ,RNA, Small Interfering ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,CAMK ,PI3K/AKT/mTOR pathway ,Phospholipase C gamma ,Cell growth ,Kinase ,TOR Serine-Threonine Kinases ,RPTOR ,Intracellular Signaling Peptides and Proteins ,Ribosomal Protein S6 Kinases, 70-kDa ,Cell biology ,Enzyme Activation ,Pyrimidines ,Benzamides ,embryonic structures ,Imatinib Mesylate ,Cancer research ,Phosphorylation ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
In chronic myeloid leukemia, activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway is crucial for survival and proliferation of leukemic cells. Essential downstream molecules involve mammalian target of rapamycin (mTOR) and S6-kinase. Here, we present a comprehensive analysis of the molecular events involved in activation of these key signaling pathways. We provide evidence for a previously unrecognized phospholipase C-gamma1 (PLC-gamma1)-controlled mechanism of mTOR/p70S6-kinase activation, which operates in parallel to the classical Akt-dependent machinery. Short-term imatinib treatment of Bcr-Abl-positive cells caused dephosphorylation of p70S6-K and S6-protein without inactivation of Akt. Suppression of Akt activity alone did not affect phosphorylation of p70-S6K and S6. These results suggested the existence of an alternative mechanism for mTOR/p70S6-K activation. In Bcr-Abl-expressing cells, we detected strong PLC-gamma1 activation, which was suppressed by imatinib. Pharmacological inhibition and siRNA knockdown of PLC-gamma1 blocked p70S6-K and S6 phosphorylation. By inhibiting the Ca-signaling, CaMK and PKCs we demonstrated participation of these molecules in the pathway. Suppression of PLC-gamma1 led to inhibition of cell proliferation and enhanced apoptosis. The novel pathway proved to be essential for survival and proliferation of leukemic cells and almost complete cell death was observed upon combined PLC-gamma1 and Bcr-Abl inhibition. The pivotal role of PLC-gamma1 was further confirmed in a mouse leukemogenesis model.
- Published
- 2009
16. Entwicklung von Tyrosinkinase-Inhibitoren bei hämatologischen Neoplasien. FLT3 und JAK2 als therapeutischeTargets
- Author
-
Daniel B. Lipka, Thomas Fischer, Christoph Huber, and Florian Heidel
- Subjects
Pharmacology ,Gynecology ,medicine.medical_specialty ,business.industry ,medicine ,Pharmaceutical Science ,Pharmacology (medical) ,Hematologic Neoplasms ,business ,Tyrosine kinase - Abstract
Tyrosinkinase-Inhibitoren stellen einen wichtigen Beitrag zur Weiterentwicklung und Verbesserung der Therapie von hamatologischen Neoplasien dar. Zahlreiche Inhibitoren befinden sich bereits in fortgeschrittener klinischer Testung. Wahrend bei einem Teil der Erkrankungen (CML) die Inhibitor-Therapie bereits als Standard etabliert ist, befinden sich die “small molecules” bei Akuter Myeloischer Leukamie und chronisch-myeloproliferativen Syndromen noch in der Etablierungsphase. Die Entwicklung von neuen zielgerichteten Substanzen zahlt zu den grosen praklinischen und klinischen Herausforderungen der nachsten Jahre.
- Published
- 2008
17. Addition of rituximab to standard therapy improves response rate and progression-free survival in relapsed or refractory thrombotic thrombocytopenic purpura and autoimmune haemolytic anaemia
- Author
-
Georg Hess, Daniel B. Lipka, Inge Scharrer, Charis von Auer, Florian Heidel, and Christoph Huber
- Subjects
Adult ,Male ,medicine.medical_specialty ,Time Factors ,Thrombotic thrombocytopenic purpura ,Salvage therapy ,Gastroenterology ,Disease-Free Survival ,Antibodies, Monoclonal, Murine-Derived ,Pharmacotherapy ,Refractory ,Recurrence ,Median follow-up ,Germany ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Immunologic Factors ,Progression-free survival ,Aged ,Retrospective Studies ,Salvage Therapy ,Purpura, Thrombocytopenic, Idiopathic ,business.industry ,Standard treatment ,Antibodies, Monoclonal ,Hematology ,Middle Aged ,medicine.disease ,Surgery ,Treatment Outcome ,Drug Therapy, Combination ,Female ,Rituximab ,Anemia, Hemolytic, Autoimmune ,business ,Follow-Up Studies ,medicine.drug - Abstract
SummaryTreatment of relapsed or refractory autoimmune mediated haemolytic syndromes, such as autoimmune haemolytic anaemia (AIHA) and thrombotic thrombocytopenic purpura (TTP), represents a therapeutic challenge. Here we report on our experience with the monoclonal anti-CD20 antibody rituximab (R) compared to standard treatment in these diseases. Patients with non-familialTTP orAIHA and no underlying malignancy were included in our analysis. Safety and efficacy of R-treatment were compared to results obtained in standard treatment approaches. Altogether, 27 patients were analyzed, comprising 15 patients withTTP and 12 patients with AIHA. The patients’ average age at the time of diagnosis was 54 years. Eleven patients received antibody treatment (8 TTP, 3 AIHA). No acute or late WHO grade III/IV toxicity associated with rituximab was noted. With standard therapy, the overall response rate (ORR) was 66.7% for AIHA and 65.8% for TTP, respectively. For the R-containing regimens the ORR was 100%. In patients with TTP, median progression free survival (PFS) with R-treatment was 3.8 years, as compared to 0.1 years in the standard-treatment group. In patients with AIHA median PFS was not reached for R-containing treatment; all patients are in sustained remissions with a median follow up of 12.5 months. In the absence of prospective trials, our data underline the safety and efficacy of rituximab in relapsed and refractory autoimmune anaemias with favourable response rates and promising long-term progressionfree survival. Therefore, prospective clinical trials evaluating rituximab as salvage- and first-line-therapy are clearly warranted.
- Published
- 2007
18. The kinase inhibitor LS104 induces apoptosis, enhances cytotoxic effects of chemotherapeutic drugs and is targeting the receptor tyrosine kinase FLT3 in acute myeloid leukemia
- Author
-
Thomas Kindler, Daniel B. Lipka, Florian Heidel, Stefan Kasper, Boyka Markova, Christoph Huber, Thomas Fischer, Frank Breitenbuecher, and Yvette Hoehn
- Subjects
Adult ,Male ,Cancer Research ,Daunorubicin ,medicine.drug_class ,Blotting, Western ,Fluorescent Antibody Technique ,Apoptosis ,Pharmacology ,Receptor tyrosine kinase ,Tyrosine-kinase inhibitor ,Styrenes ,Colony-Forming Units Assay ,Mice ,Bone Marrow ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Cells, Cultured ,CD135 ,Animals ,Humans ,Point Mutation ,Tissue Distribution ,Aged ,Cell Proliferation ,Aged, 80 and over ,biology ,Acrylonitrile ,Cytarabine ,Myeloid leukemia ,Cell Differentiation ,Hematology ,Middle Aged ,medicine.disease ,Leukemia ,Leukemia, Myeloid, Acute ,Oncology ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,biology.protein ,Cancer research ,Drug Therapy, Combination ,Female ,medicine.drug ,Signal Transduction - Abstract
Activating mutations of FLT3 are found in approximately one-third of acute myeloid leukemia (AML)-cases and are considered to represent an attractive therapeutic target. In this study, we report that the hydroxystyryl-acrylonitrile compound LS104 inhibits proliferation and induces potent cytotoxic effects in FLT3 expressing leukemic cells in vitro. Immunoblot and phosphoprotein-FACS analysis demonstrated inhibiton of phosphorylation of FLT3-ITD and of its downstream targets. In pharmacokinetic studies, a rapid and dose dependent cellular uptake of LS104 lasting up to 11h could be demonstrated. Combination of LS104 with chemotherapeutic agents markedly enhanced cytotoxic effects. Recently, a phase I clinical trial investigating LS104 in refractory/relapsed hematologic malignancies has been initiated.
- Published
- 2008
19. Focal progression in patients with gastrointestinal stromal tumors after initial response to imatinib mesylate: a three-center-based study of 38 patients
- Author
-
Thomas Fischer, Salah-Eddin Al-Batran, Jacob Robert Izbicki, Jan Stoehlmacher, Elke Jäger, Claudius Dechow, Thomas Kraus, Florian Heidel, Markus Düx, Joerg T. Hartmann, and Eva Wardelmann
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Stromal cell ,Gastrointestinal Stromal Tumors ,Salvage therapy ,Antineoplastic Agents ,Disease-Free Survival ,Piperazines ,Surgical oncology ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Neoplasm ,Humans ,neoplasms ,Survival rate ,Aged ,Retrospective Studies ,Salvage Therapy ,Dose-Response Relationship, Drug ,business.industry ,Gastroenterology ,Retrospective cohort study ,Imatinib ,General Medicine ,Middle Aged ,medicine.disease ,digestive system diseases ,Survival Rate ,Proto-Oncogene Proteins c-kit ,Imatinib mesylate ,Pyrimidines ,Treatment Outcome ,Drug Resistance, Neoplasm ,Benzamides ,Mutation ,Disease Progression ,Imatinib Mesylate ,Female ,business ,medicine.drug ,Follow-Up Studies - Abstract
This study aimed to investigate the outcome of patients with advanced gastrointestinal stromal tumors (GISTs) exhibiting focal disease progression during imatinib therapy, treated by surgical resection and imatinib continuation.A consecutive series of 38 patients with metastatic GISTs who underwent treatment with imatinib at our centers during a defined period of time was evaluated. Patients were evaluated for demographics including tumor-related features, initial response, disease recurrence, and salvage treatment modalities, and were classified as having either focal or generalized progression upon presentation prior to salvage therapy.After a median follow-up of 31.8 months, 25 of the 38 (65.8%) patients had progressed. Nine (36%) patients were classified as having focal and 16 (64%) as having generalized progression. Salvage therapies were: surgical resection and imatinib dose escalation in patients exhibiting focal progression and imatinib dose escalation alone in the majority of patients exhibiting generalized progression. Focal progression was associated with prolonged progression-free survival (PFS) and overall survival (OS) after salvage therapy as compared with generalized progression (median PFS and OS, 11.3 months and not attained, versus 2.5 and 22.8 months, respectively). Six-month PFS was 89% and 39% in patients exhibiting focal and generalized progression, respectively. KIT mutation analysis of controlled and progressive lesions was performed in 4 patients with focal progression. Secondary KIT mutations affected progressive lesions, whereas nonprogressive lesions harbored the original mutations only.Patients with advanced GIST exhibiting focal disease progression during imatinib therapy may benefit from surgical resection and imatinib continuation. Imatinib resistance seems to be partial in these patients.
- Published
- 2007
20. T1162 Analysis of Antiproliferative and Chemosensitizing Effects of Sunitinib on Human Esophagogastric Cancer Cells: Synergistic Interaction With Vandetanib via Inhibition of Multireceptor Tyrosine Kinase Pathways
- Author
-
Ines Gockel, Hauke Lang, Orestis Lyros, Florian Heidel, Markus Moehler, Carl C. Schimanski, Annett Mueller, and Peter R. Galle
- Subjects
Hepatology ,Esophagogastric cancer ,Chemistry ,Sunitinib ,Gastroenterology ,medicine ,Cancer research ,Vandetanib ,Tyrosine kinase ,medicine.drug - Published
- 2010
21. 3,4-Diarylmaleimides Effectively Inhibit Proliferation of FLT3-ITD-Positive Leukemic Cells, Induce Apoptosis and Show Additive Effects in Combination with Chemotherapeutic Drugs
- Author
-
Florian Heidel, Christoph Huber, Christian Peifer, Boyka Markova, Thomas Fischer, Jan-Peter Kramp, Gerd Dannhardt, Fian K. Mirea, Daniel B. Lipka, and Stanislav Plutizki
- Subjects
Sorafenib ,Kinase ,Angiogenesis ,Immunology ,Cell Biology ,Hematology ,Pharmacology ,Biology ,Biochemistry ,Chemotherapy regimen ,Mechanism of action ,Apoptosis ,hemic and lymphatic diseases ,medicine ,Cytarabine ,medicine.symptom ,Protein kinase B ,medicine.drug - Abstract
Internal tandem duplication (ITD) mutations of FLT3 are present in leukemic blasts of approximately 30% of AML patients. ITD-mutations of FLT3 confer a worse prognosis and decreased overall survival. Therefore, FLT3-tyrosine kinase is considered an attractive drug target in AML and several FLT3-tyrosine kinase inhibitors (TKIs) are currently being tested in clinical trials (CEP701, MLN518, Sorafenib, PKC412). However, using these drugs as monotherapy, against the setting of remarkable efficacy has emerged the problem of short duration of remission indicating rapid development of secondary resistance. In addition, up to 30% of patients may show primary resistance to currently available FLT3-TKIs. Therefore, to overcome these limitations further clinical development requires novel approaches (e.g. combination with chemotherapy) and development of more efficacious FLT3-TKI. Here, we investigated two novel TKIs (3,4-Diarylmaleimides, compounds 53 and 80) previously described as angiogenesis inhibitors (Peifer, J. Med. Chem. 2006), to determine their mechanism of action and efficacy in FLT3-ITD transfected 32D cells as well as in primary ITD-positive AML blasts. Western Blotting experiments of 32D-FLT3-ITD cells confirmed dose dependent inhibition/dephosphorylation of downstream targets as STAT5 (pTyr694/699), ERK (pThr202/pTyr204) and AKT (pSer473) upon incubation with either compound using nanomolar concentrations. Both compounds (cpd.) showed induction of apoptosis with an IC50 of 350nM (cpd. 53) and 800nM (cpd. 80), respectively, as detected by sub G1-fraction in cell cycle analysis. Using colony assays we defined a possible therapeutic window of these inhibitors: Bone-marrow cells of healthy donors were incubated with increasing concentrations of both inhibitors revealing no significant inhibition of colony formation up to a dose of 5 μM. As clinical studies in AML will evaluate FLT3 inhibitors in combination with chemotherapy, we tested both cpds. in vitro in combination with cytosine arabinosid (Ara-C) and daunorubicine (DNR). Within a range of 1–50nM DNR and 0.3–10μM Ara-C, both cpds. showed additive effects in combination with chemotherapy in 32D-FLT3-ITD cells as revealed by apoptosis assays. Primary AML blasts harboring the FLT3-ITD length mutation were also investigated to determine the efficacy in patient material. These experiments revealed apoptosis rates of 10–30% upon incubation with 3,4-Diarylmaleimides for 72 hours. Cellular up-take of cpd. 53 in primary AML blasts could be demonstrated within 5 minutes of incubation using FACS analysis. Increased fluorescence (200–400% of baseline values) could be detected for more than 24 hours suggesting stable intracellular concentration of the inhibitor in AML blasts within this time period. To evaluate signaling targets of 3,4-Diarylmaleimides in detail, we applied a kinomics peptide chip containing a set of 960 kinase substrates (human sequences only) in triplicate on a slide. Phosphorylation of peptide substrates was detected by autoradiography. Results of evaluation will be presented. In conclusion, 3,4-Diarylmaleimides show efficacy in FLT3-ITD-positive cells alone as well as in combination with chemotherapy. Currently, experiments evaluating toxicity in murine models are planned to explore a possible clinical application.
- Published
- 2007
22. The JAK2 Kinase Inhibitor LS104 Induces Growth-Arrest and Apoptosis in JAK2V617F Positive Cells
- Author
-
Linda S. Hoffmann, Stefan Kasper, Daniel B. Lipka, Boyka Markova, Joseph Elliot, Thomas Kindler, Frank Breitenbuecher, Christoph Huber, Ross L. Levine, Thomas Fischer, and Florian Heidel
- Subjects
MAPK/ERK pathway ,Kinase ,medicine.medical_treatment ,Immunology ,Autophosphorylation ,food and beverages ,Cell Biology ,Hematology ,Transfection ,Biology ,Biochemistry ,Molecular biology ,Erythropoietin receptor ,Cytokine ,hemic and lymphatic diseases ,medicine ,Cancer research ,Kinase activity ,Protein kinase B - Abstract
The JAK2V617F-mutation (V617F) is a novel, highly prevalent molecular marker in Ph-negative myeloproliferative disease (MPD). In vitro, the V617F mutation confers cytokine independent growth of Ba/F3 cells expressing erythropoietin receptor (EpoR) and constitutive activation of the JAK2 kinase and of the JAK-STAT pathway. In a murine bone-marrow transplant model the V617F-mutation alone is sufficient to induce a polycythemia vera-like phenotype. Therefore, mutant JAK2 kinase is a promising target for kinase inhibitor development. In this report, we characterize the small molecule LS104 (previously CR4; Grunberger et al., Blood 2003) as a novel non-ATP-competitive JAK2V617F kinase inhibitor. Ba/F3 cells stably transfected with EpoR and either the V617F-mutant (Ba/F3-EpoR-VF) or wildtype JAK2 (Ba/F3-EpoR-WT) were treated with LS104. Apoptosis assays as well as immunoblotting of JAK2 and of downstream signalling pathways were performed. The effects of LS104 on kinase activity were determined in an in vitro JAK2 kinase assay. A combination of LS104 with JAK-inhibitor I, which acts via the ATP-binding site, was tested in apoptosis assays. Finally, growth of endogenous erythroid colonies (EECs) obtained from patients with V617F-positive MPD was tested upon addition of LS104. LS104 selectively and dose dependently induced apoptosis in Ba/F3-EpoR-VF cells as compared to Ba/F3-EpoR-WT control cells. By immunoblotting we found inhibition of JAK2 autophosphorylation and of downstream targets as STAT5, AKT and ERK upon treatment with LS104. Activation of these targets by JAK2 was confirmed in experiments using JAK2 siRNA. The IC50 for JAK2 in an in vitro kinase assay using LS104 was
- Published
- 2007
23. LS104, a Novel Kinase Inhibitor, Induces Apoptosis, Synergizes with Cytostatic Drugs and Is Targeting the Receptor Tyrosine Kinase FLT3
- Author
-
Frank Breitenbuecher, Yvette Hoehn, Daniel B. Lipka, Thomas Kindler, Florian Heidel, Stefan Kasper, Christoph Huber, Thomas Fischer, and Helmut Thomas
- Subjects
Kinase ,Cell growth ,Immunology ,Tyrosine phosphorylation ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Receptor tyrosine kinase ,Leukemia ,chemistry.chemical_compound ,chemistry ,Annexin ,hemic and lymphatic diseases ,embryonic structures ,medicine ,biology.protein ,Kinase activity ,Tyrosine - Abstract
Fms-like tyrosine kinase 3 (FLT3), a member of the class III tyrosine kinase receptor family, is expressed in up to 90% of acute myeloid leukemia (AML). Activating mutations like internal tandem duplication (ITD) of the juxtamembrane domain and kinase domain point mutations are found in approximately 35% of AML-cases and are considered to represent an attractive therapeutic target. In this study, we report that the novel hydroxystyryl-acrylonitrile compound LS104 induces potent cytotoxic effects in FLT3 ITD-positive leukemic cells. As a cellular model to investigate FLT3-ITD specific effects we used 32D myeloid cells stably transfected with FLT3-ITD and wt-FLT3, respectively. In MTS assays, pronounced inhibition of cell growth was seen at nanomolar concentration (IC50=50nM) which could be partially rescued by addition of IL3. LS104 at a concentration ranging from 3–10μM readily induced apoptosis as evaluated by cell cycle analysis, annexin-V assays and PARP cleavage. Throughout the experiments, FLT3-ITD expressing cells showed significantly higher sensitivity towards LS104 than cells expressing wt-FLT3. Similar results were observed evaluating the cytotoxic effects of LS104 in the human myeloid-leukemia derived FLT3-ITD harbouring MV4;11 and the wt-FLT3 expressing lymphoid-leukemia derived RS4;11 cell line. Immunoblot analysis demonstrated that LS104 inhibits tyrosine phosphorylation of FLT3-ITD and of its downstream target STAT5. This points out that FLT3-ITD is a molecular target of LS104. Interestingly, efficacy of LS104 to induce apoptosis was significantly reduced in 32D cells transfected with a FLT3-ITD isoform (N676K) previously reported to be associated with clinical resistance of FLT3-ITD to the kinase inhibitor PKC412 (Heidel et al., ASH 2004). This result strongly suggests that the mechanism of action of LS104 is indeed inhibition of FLT3 kinase activity rather than inhibition of other kinases targeted by LS104. As clinical development of FLT3 kinase inhibitors in AML likely will be in combination with chemotherapy, we evaluated the in vitro effects of combining LS104 with the cytostatic drugs cytosine arabinosid (Ara-C) and daunorubicin (DNR). In 32D FLT3-ITD cells, cell cycle analysis showed strong synergy in induction of apoptosis upon incubation with LS104 (1–3μM) plus Ara-C (1μM) and DNR (10nM), respectively. Using primary blasts from AML patients, we further evaluated cytotoxicity of LS104 at various concentrations and in combination with Ara-C and DNR, respectively. LS104 as a single agent was shown to induce apoptosis in primary AML blasts and combination of LS104 with cytostatic drugs resulted in additive and synergistic effects in leukemic blasts from 4 out of 6 patients investigated. Western-blotting of primary AML blasts revealed inhibition of FLT3-ITD and STAT5 tyrosine phosphorylation by LS104 in a dose-dependent way (3–50μM). Interestingly, LS104 showed intrinsic fluorescence activity. Using FACS analysis and fluorescence microscopy we could demonstrate a rapid and dose-dependent cellular uptake of LS104 in leukemic cell lines and in primary blasts. After removal of LS104 from medium, enhanced fluorescence corresponding to intracellular LS104 was detectable up to 24 hours. This property could be used to monitor drug uptake of primary AML blasts in vivo. In conclusion, our data provide a preclinical framework for clinical trials of LS104 in FLT3-ITD positive leukemia.
- Published
- 2005
24. Mechanisms of Resistance to the FLT3-Tyrosine Kinase Inhibitor PKC412 in Patients with AML
- Author
-
Johannes Roesel, Pamela Cohen, Florian Heidel, C. Huber, Thomas Kindler, Virginia M. Klimek, Eli Estey, Thomas Fischer, Richard Stone, Eric J. Feldman, D. DeAngelo, H.-M. Thiede, Frank Breitenbuecher, Fian K. Solem, Birgit Carius, Gary J. Schiller, Francis J. Giles, Stephen D. Nimer, and G. Ehninger
- Subjects
biology ,medicine.drug_class ,Kinase ,Cell growth ,Immunology ,Autophosphorylation ,Clone (cell biology) ,Cell Biology ,Hematology ,Biochemistry ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,hemic and lymphatic diseases ,medicine ,biology.protein ,Cancer research ,Phosphorylation ,Tyrosine kinase - Abstract
The FLT3 receptor tyrosine kinase is expressed in 70-90% of cases of AML. Up to 35% of patients with AML show mutations in the JM-region or kinase domain of FLT3. These lead to autophosphorylation promoting ligand-independent cell proliferation and inhibition of apoptosis. Treatment with FLT3 tyrosine kinase inhibitors (TKI) is a promising tool in therapy of AML. Preliminary results investigating the FLT3-TKI PKC412 in patients with relapsed/refractory AML revealed that 11/15 patients (73%) with mutated FLT3 and 16/46 patients (35%) with WT FLT3 showed a >50% blast response in peripheral blood (Estey E et al. Blood.2003; 102:919a). Despite its remarkable efficacy in reducing the leukemic clone, remissions upon single agent therapy using PKC412 tend to be short and secondary resistance occurs rather rapidly. So far, very little is known on the mechanisms of resistance to therapy using FLT3-TKIs. Therefore, we sought to characterize the molecular mechanisms involved. Overall PB and BM samples from 5 patients undergoing single agent therapy with PKC412 were analyzed. Three patients revealed reactivation of FLT3-kinase at relapse. Unchanged protein levels of Flt3 as detected by Western blotting before therapy and at relapse showed that amplification of FLT3 does not appear to play a role in development of secondary resistance. PKC412 plasma levels, as measured by HPLC, were similar in patients with and without reactivation of the kinase. Therefore, we examined the bioactivity of plasma obtained at relapse to inhibit phosphorylation of FLT3 in MV4-11 cells harboring FLT3-ITD. In two patients, plasma inhibited FLT3-tyrosine phosphorylation at the time of remission as well as at relapse, while in one patient, plasma was not able to inhibit FLT3-phosphorylation. Therefore, in this patient, an inhibitory activity in serum appeared to be operating. A potential candidate for this inhibitory activity is alpha1-glycoprotein (AGP) that was shown previously to mediate resistance to STI571 treatment. Our experiments showed that AGP is indeed able to inhibit bioactivity of PKC412 in-vitro. However, AGP-levels monitored in PB samples from patients included in this analysis did not show elevated plasma levels at relapse. Ex-vivo treatment of primary blasts with PKC412 at relapse showed that in one patient, blasts were still sensitive to PKC412. In two other patients, blasts showed resistance using a wide dose range of PKC412. Therefore, in these patients, cDNA sequencing of FLT3 was performed. In one patient, we identified a point mutation leading to an aminoacid exchange within the N-lobe of the FLT3-kinase domain at relapse. This mutation was not present at start of PKC412 therapy. Experiments analyzing the functional role of the novel mutation in the hematopoietic cell line 32D are in progress and will be presented. In conclusion, we have preliminarily identified examples of kinase-dependent and independent mechanisms operating in clinical resistance to PKC412.
- Published
- 2004
25. Results of a multicenter phase II trial for older patients with c‐Kit‐positive acute myeloid leukemia (AML) and high‐risk myelodysplastic syndrome (HR‐MDS) using low‐dose Ara‐C and Imatinib.
- Author
-
Florian Heidel, Jorge Cortes, Frank G. Rücker, Walter Aulitzky, Laurie Letvak, Thomas Kindler, Christoph Huber, Hartmut Döhner, Hagop Kantarjian, and Thomas Fischer
- Subjects
- *
MYELODYSPLASTIC syndromes , *MYELOID leukemia , *IMATINIB , *ANTINEOPLASTIC agents , *DISEASES in older people - Abstract
Imatinib (IM) is a potent tyrosine kinase inhibitor of c‐Kit. c‐Kit is expressed in the majority of patients with acute myeloid leukemia (AML). Whereas clinical trials evaluating monotherapy with IM in AML revealed low response rates, Ara‐C and IM showed synergistic effects in vitro. This suggested evaluation of a combination treatment.Low‐dose Ara‐C (LDAC) combined with IM was tested to determine the efficacy and safety of this regimen. Forty patients from 4 centers with c‐Kit‐positive AML (n = 34) and high‐risk myelodysplastic syndrome (HR‐MDS) (n = 6) with a median age of 73 years were enrolled. They were either not eligible for myelosuppressive therapy and/or had recurring/refractory disease.Thirty‐eight patients were evaluable for analysis. In 6 of 38 patients a blast response was observed. Eight of 38 patients showed stable disease for more than 2 months. The objective hematologic response rate was low (11%), with 2 patients showing hematologic improvement and 1 each with a partial response (PR) or complete response (CR). Median overall survival was 138 days, with 20% of patients alive after an observation period of 600 days. Study medication was applied in an ambulatory setting with minimal hospitalization time, an early mortality rate of only 18.9%, and a low toxicity rate.LDAC plus IM does not appear to be inferior in older AML patients incomparison with historic controls receiving myelosuppressive therapy. However, this trial also shows that LDAC/IM does not appear to be more effective than LDAC monotherapy in a patient population not selected for appropriate molecular markers. Cancer 2007 © 2007 American Cancer Society. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
26. Analysis of Antiproliferative and Chemosensitizing Effects of Sunitinib on Human Esophagogastric Cancer Cells: Synergistic Interaction With Vandetanib via Inhibition of Multireceptor Tyrosine Kinase Pathways
- Author
-
Florian Heidel, Ines Gockel, Carl C. Schimanski, Annett Mueller, Peter R. Galle, Hauke Lang, Markus Moehler, and Orestis Lyros
- Subjects
Cancer Research ,Umbilical Veins ,Indoles ,Esophageal Neoplasms ,Apoptosis ,Vandetanib ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Piperidines ,Sunitinib ,Medicine ,Drug Interactions ,Epidermal growth factor receptor ,Phosphorylation ,Cells, Cultured ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Drug Synergism ,Flow Cytometry ,ErbB Receptors ,Oncology ,Drug Therapy, Combination ,medicine.drug ,Signal Transduction ,medicine.medical_specialty ,medicine.drug_class ,Blotting, Western ,Antineoplastic Agents ,Stomach Neoplasms ,Internal medicine ,Humans ,Pyrroles ,Propidium iodide ,RNA, Messenger ,Protein Kinase Inhibitors ,Cell Proliferation ,Vascular Endothelial Growth Factor Receptor-1 ,business.industry ,Cancer ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-3 ,Vascular Endothelial Growth Factor Receptor-2 ,Endocrinology ,chemistry ,Cancer research ,biology.protein ,Quinazolines ,Endothelium, Vascular ,business ,Proto-Oncogene Proteins c-akt - Abstract
The receptor tyrosine kinases (RTKs), epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor 1-3 (VEGFR1-3), are frequently expressed in gastric cancer and are putative therapeutic targets in this disease. We have investigated the anti-proliferative and chemosensitizing properties of the multitargeted small-molecule RTK inhibitors sunitinib and vandetanib in a panel of 4 human gastric and esophageal cancer cell lines. In the 1st instance, the expression of potential targets of these small-molecule inhibitors was examined by reverse transcriptase-polymerase chain reaction, western blotting, and flow cytometry. EGFR mRNA and protein was detected in all cases, with VEGFR2 expression noted in all but 1 line. Both EGF and VEGF were shown to stimulate tumor cell growth, and both sunitinib and vandetanib were found to be associated with significant dose-dependent inhibition of proliferation and an enhancement of apoptosis, as determined by MTT and propidium iodide/Annexin V labeling assays, respectively. The addition of sunitinib to VEGF-stimulated NCI-N87 cells was associated with a reduction in MAPK phosphorylation (pMAPK) but not Akt phosphorylation (pAkt), whereas the addition of vandetanib was associated with reductions in both VEGF- and EGF-mediated VEGFR2 phosphorylation, pMAPK and pAkt. Co-administration of sunitinib significantly enhanced the sensitivity of MKN-45 cells to cisplatin and irinotecan. In addition, vandetanib synergistically enhanced the sunitinib-associated inhibition of gastric cancer cell growth. In conclusion, these preliminary data confirm the importance of EGFR and VEGFR signaling in gastric cancer and suggest that the simultaneous inhibition of RTK-pathways through sunitinib and vandetanib may provide therapeutic benefit in this disease.
27. Impact of comorbidities and body mass index in patients with myelofibrosis treated with ruxolitinib
- Author
-
Daniele Cattaneo, Nicola Sgherza, Roberto Latagliata, Francesco Cavazzini, Renato Fanin, Antonio Cuneo, Giuseppe A. Palumbo, Alessia Tieghi, Nicola Vianelli, Gianpietro Semenzato, Gianni Binotto, Robin Foà, Matteo Molica, Mariella D'Adda, Roberto M. Lemoli, Francesca Palandri, Michele Cavo, Daniela Bartoletti, Alessandra Iurlo, Massimo Breccia, Elisabetta Abruzzese, Nicola Polverelli, Domenico Russo, Giuseppe Auteri, Mario Tiribelli, Florian H. Heidel, Lucia Catani, Costanza Bosi, Alessandro Isidori, Luigi Scaffidi, Franco Aversa, Micaela Bergamaschi, Massimiliano Bonifacio, Monica Crugnola, Massimo Breccia, Daniela Bartoletti, Massimiliano Bonifacio, Giuseppe A. Palumbo, Nicola Polverelli, Elisabetta Abruzzese, Micaela Bergamaschi, Alessia Tieghi, Mario Tiribelli, Alessandra Iurlo, Francesco Cavazzini, Nicola Sgherza, Gianni Binotto, Alessandro Isidori, Mariella D’Adda, Monica Crugnola, Costanza Bosi, Florian Heidel, Matteo Molica, Luigi Scaffidi, Daniele Cattaneo, Roberto Latagliata, Giuseppe Auteri, Roberto M. Lemoli, Renato Fanin, Domenico Russo, Franco Aversa, Antonio Cuneo, Gianpietro Semenzato, Lucia Catani, Michele Cavo, Nicola Vianelli, Robin Foà, and Francesca Palandri
- Subjects
Male ,Ruxolitinib ,Time Factors ,Myelofibrosis ,BMI ,CCI ,Comorbidities ,Adult ,Age Factors ,Aged ,Aged, 80 and over ,Disease-Free Survival ,Female ,Humans ,Middle Aged ,Primary Myelofibrosis ,Pyrazoles ,Sex ,Sex Factors ,Survival Rate ,Body Mass Index ,Gastroenterology ,0302 clinical medicine ,80 and over ,Hematology ,Myelofibrosi ,General Medicine ,030220 oncology & carcinogenesis ,Cohort ,BMI, CCI, Comorbidities, Myelofibrosis, Ruxolitinib ,Comorbiditie ,Underweight ,medicine.symptom ,medicine.drug ,medicine.medical_specialty ,Anemia ,NO ,03 medical and health sciences ,Internal medicine ,Nitriles ,medicine ,Survival rate ,business.industry ,medicine.disease ,Pyrimidines ,business ,Body mass index ,030215 immunology - Abstract
Comorbidities defined by the Charlson comorbidity index (CCI) and body mass index (BMI) are significantly associated with outcome in patients who receive continuous treatment with tyrosine kinase inhibitors. We evaluated the impact of CCI and BMI on responses, drug-related toxicities, and outcome in a cohort of 402 patients with myelofibrosis (MF) treated with ruxolitinib in 23 European Hematology Centers. Comorbidities were evaluable in all 402 patients. A higher (≥ 3) CCI did not correlate with a lower spleen reduction at any time (p = 0.68) or symptoms' response (p = 0.11), but influenced the onset of anemia during the first 3months of treatment and later (p = 0.02 and p = 0.03, respectively) in patients without anemia baseline. BMI was evaluable in 380 patients and did not correlate with differences in spleen and symptoms response (p = 0.57 and p = 0.49, respectively). A higher CCI and a lower BMI correlated also with a reduced overall survival (p
- Published
- 2018
28. Ruxolitinib in elderly patients with myelofibrosis: impact of age and genotype. A multicentre study on 291 elderly patients
- Author
-
G. Semenzato, Massimiliano Bonifacio, Mario Tiribelli, Daniela Bartoletti, Lucia Catani, Bruno Martino, Micaela Bergamaschi, Florian H. Heidel, Giuseppe A. Palumbo, Gianni Binotto, Francesca Palandri, Antonio Cuneo, Mariella D'Adda, Michele Cavo, Alessandra Iurlo, Massimo Breccia, Alessandro Isidori, Elena Sabattini, Maria Rosaria Sapienza, Nicola Polverelli, Giulia Benevolo, Roberto M. Lemoli, Nicola Sgherza, Umberto Vitolo, Monica Crugnola, Elisabetta Abruzzese, Adalberto Ibatici, Nicola Vianelli, Costanza Bosi, Franco Aversa, Roberto Latagliata, Francesco Cavazzini, Alessia Tieghi, Giovanni Martinelli, Francesca Palandri, Lucia Catani, Massimiliano Bonifacio, Giulia Benevolo, Florian Heidel, Giuseppe A. Palumbo, Monica Crugnola, Elisabetta Abruzzese, Daniela Bartoletti, Nicola Polverelli, Micaela Bergamaschi, Mario Tiribelli, Alessandra Iurlo, Massimo Breccia, Francesco Cavazzini, Alessia Tieghi, Gianni Binotto, Alessandro Isidori, Bruno Martino, Mariella D'Adda, Costanza Bosi, Elena Sabattini, Umberto Vitolo, Franco Aversa, Adalberto Ibatici, Roberto M. Lemoli, Nicola Sgherza, Antonio Cuneo, Giovanni Martinelli, Giampietro Semenzato, Michele Cavo, Nicola Vianelli, Maria R. Sapienza, and Roberto Latagliata
- Subjects
0301 basic medicine ,elderly ,high molecular risk ,high molecular risk mutations ,myelofibrosis ,ruxolitinib ,Ruxolitinib ,medicine.medical_specialty ,Genotype ,Socio-culturale ,Hematology ,Older population ,03 medical and health sciences ,0302 clinical medicine ,myelofibrosi ,Older patients ,Risk Factors ,Internal medicine ,Nitriles ,Medicine ,Humans ,Myelofibrosis ,Aged ,Janus Kinases ,Retrospective Studies ,business.industry ,Age Factors ,elderly, high molecular risk, high molecular risk mutations, myelofibrosis, ruxolitinib ,medicine.disease ,Mutation ,Primary Myelofibrosis ,Pyrazoles ,Survival Analysis ,Treatment Outcome ,Peripheral blood ,Discontinuation ,030104 developmental biology ,Pyrimidines ,030220 oncology & carcinogenesis ,high molecular risk mutation ,business ,medicine.drug - Abstract
Ruxolitinib is a JAK1/2 inhibitor that may control myelofibrosis (MF)-related splenomegaly and symptoms and can be prescribed regardless of age. While aging is known to correlate with worse prognosis, no specific analysis is available to confirm that ruxolitinib is suitable for use in older populations. A clinical database was created in 23 European Haematology Centres and retrospective data on 291 MF patients treated with ruxolitinib when aged ≥65 years were analysed in order to assess the impact of age and molecular genotype on responses, toxicities and survival. Additional mutations were evaluated by a next generation sequencing (NGS) approach in 69 patients with available peripheral blood samples at the start of ruxolitinib treatment. Compared to older (age 65-74 years) patients, elderly (≥75 years) showed comparable responses to ruxolitinib, but higher rates of drug-induced anaemia and thrombocytopenia and worse survival. Nonetheless, the ruxolitinib discontinuation rate was comparable in the two age groups. Number and types of molecular abnormalities were comparable across age groups. However, the presence of high molecular risk (HMR) mutations significantly affected survival, counterbalancing the effect of aging. Indeed, elderly patients with
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.