13 results on '"Kühn MWM"'
Search Results
2. Measurable residual disease monitoring in AML with FLT3-ITD treated with intensive chemotherapy plus midostaurin.
- Author
-
Rücker FG, Bullinger L, Cocciardi S, Skambraks S, Luck TJ, Weber D, Krzykalla J, Pozek E, Schneider I, Corbacioglu A, Gaidzik VI, Meid A, Aicher S, Stegelmann F, Schrade A, Theis F, Fiedler W, Salih HR, Wulf G, Salwender H, Schroeder T, Götze KS, Kühn MWM, Lübbert M, Schlenk RF, Benner A, Thol F, Heuser M, Ganser A, Döhner H, and Döhner K
- Subjects
- Humans, Male, Middle Aged, Female, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Prognosis, Mutation, High-Throughput Nucleotide Sequencing, Tandem Repeat Sequences, Young Adult, Hematopoietic Stem Cell Transplantation, Neoplasm, Residual, Staurosporine analogs & derivatives, Staurosporine therapeutic use, Staurosporine administration & dosage, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality
- Abstract
Abstract: Measurable residual disease (MRD) monitoring in acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITDpos) has been hampered by the broad heterogeneity of ITD mutations. Using our recently developed FLT3-ITD paired-end next-generation sequencing (NGS)-based MRD assay (limit of detection 10-4 to 10-5), we evaluated the prognostic impact of MRD at different time points in 157 patients with FLT3-ITDpos AML who were enrolled in the German-Austrian Acute Myeloid Leukemia Study Group 16-10 trial and who were treated with a combination of intensive chemotherapy and midostaurin, followed by midostaurin maintenance. MRD negativity (MRDneg) after 2 cycles of chemotherapy (Cy2), which was observed in 111 of 142 (78%) patients, was predictive of superior 4-year rates of cumulative incidence of relapse (CIR) (4y-CIR; 26% vs 46%; P = .001) and overall survival (OS) (4y-OS; 70% vs 44%; P = .012). This survival advantage was also seen among patients who underwent allogeneic hematopoietic-cell transplantation during first complete remission (4y-CIR, 14% vs 39%; P = .001; 4y-OS, 71% vs 49%; P = .029). Multivariate models for CIR and OS after Cy2 revealed FLT3-ITD MRDneg as the only consistent favorable variable for CIR (hazard ratio [HR], 0.29; P = .006) and OS (HR, 0.39; P = .018). During follow-up, conversion from MRDneg to MRD positivity (MRDpos) was a strong, independent factor for inferior CIR (HR, 16.64; P < .001) and OS (HR, 4.05; P < .001). NGS-based FLT3-ITD MRD monitoring identifies patients at high risk for relapse and death following treatment with intensive chemotherapy and midostaurin. Using NGS-based technology., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. The Menin story in acute myeloid leukaemia-The road to success.
- Author
-
Kühn MWM and Ganser A
- Subjects
- Humans, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacology, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Molecular Targeted Therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute drug therapy, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Nucleophosmin
- Abstract
The treatment of acute myeloid leukaemia (AML) has changed fundamentally in the last decade with many new targeted therapies entering clinics. Some of the most interesting agents under development are Menin inhibitors which interfere with the interaction of Menin with wild-type (wt) KMT2A or a KMT2A-fusion protein and thereby downregulate the leukaemic gene expression (MEIS1, PBX3, HOX) in NPM1 mutant or KMT2A-rearranged leukaemia. Other HOX and MEIS1 expressing leukaemias may also be sensitive to Menin inhibition. Following the encouraging results as monotherapy in refractory and relapsed AML, the combination of Menin inhibitors with chemotherapeutic agents and other targeted drugs is being investigated clinically., (© 2024 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
4. Refinement of the prognostic impact of somatic CEBPA bZIP domain mutations in acute myeloid leukemia: Results of the AML Study Group (AMLSG).
- Author
-
Rücker FG, Corbacioglu A, Krzykalla J, Cocciardi S, Lengerke C, Germing U, Wulf G, Samra MA, Teichmann LL, Lübbert M, Kühn MWM, Bentz M, Westermann J, Bullinger L, Gaidzik VI, Meid A, Aicher S, Stegelmann F, Weber D, Schrade A, Thol F, Heuser M, Ganser A, Benner A, Döhner H, and Döhner K
- Abstract
Competing Interests: Frank G. Rücker reports honoraria from and consultancy for Jazz Pharmaceuticals, Novartis, and BMS/Celgene; travel support from Jazz Pharmaceuticals. Michael Lübbert reports an advisory role for Abbvie, Astex Pharmaceuticals, Imago BioSciences, Janssen, Otsuka, and Syros; research support from Janssen and Cheplapharm. Michael W. M. Kühn reports honoraria from and consultancy for Pfizer, Kura Oncology, Jazz Pharmaceuticals, BMS/Celgene, and Abbvie; speakers bureau of Gilead. Lars Bullinger reports honoraria from Abbvie, Amgen, Astellas, BMS/Celgene, Daiichi Sankyo, Gilead, Janssen, Jazz Pharmaceuticals, Menarini, Novartis, Pfizer, Roche, and Sanofi; research support from Bayer and Jazz Pharmaceuticals. Verena I. Gaidzik reports an advisory role for Jazz Pharmaceuticals, Abbvie, and Boehringer‐Ingelheim; speakers bureau of Pfizer, Janssen, and Abbvie; and travel support from Abbvie. Frank Stegelmann reports honoraria from and consultancy for AOP Pharma, MorphoSys, BMS/Celgene, Incyte, Novartis, and Pfizer. Felicitas Thol reports an advisory role for Novartis, BMS, Abbvie, Menarini, and Rigel. Michael Heuser reports honoraria from Certara, Jazz Pharmaceuticals, Janssen, Novartis, and Sobi; paid consultancy for Abbvie, Amgen, BMS/Celgene, Glycostem, LabDelbert, Pfizer, PinotBio, and Servier; and research funding to his institution from Abbvie, Agios, Astellas, BMS/Celgene, Glycostem, Jazz Pharmaceuticals, Karyopharm, Loxo Oncology, and PinotBio. Hartmut Döhner declares being in an advisory role for Abbvie, Agios, Amgen, Astellas, AstraZeneca, Berlin Chemie, BMS/Celgene, Daiichi Sankyo, GEMoaB, Gilead, Janssen, Jazz Pharmaceuticals, Novartis, Servier, Stemline, and Syndax; research funding from Abbvie, Agios, Amgen, Astellas, BMS/Celgene, Jazz Pharmaceuticals, Kronos Bio, Novartis, and Pfizer. Konstanze Döhner reports an advisory role for Amgen, BMS/Celgene, Daiichi Sankyo, Janssen, Jazz Pharmaceuticals, Novartis, and Roche; research funding from Agios, Astex, Astellas, BMS/Celgene, and Novartis. All other authors declare no competing interest. The remaining authors declared no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
5. Immunoproteasome function maintains oncogenic gene expression in KMT2A-complex driven leukemia.
- Author
-
Tubío-Santamaría N, Jayavelu AK, Schnoeder TM, Eifert T, Hsu CJ, Perner F, Zhang Q, Wenge DV, Hansen FM, Kirkpatrick JM, Jyotsana N, Lane SW, von Eyss B, Deshpande AJ, Kühn MWM, Schwaller J, Cammann C, Seifert U, Ebstein F, Krüger E, Hochhaus A, Heuser M, Ori A, Mann M, Armstrong SA, and Heidel FH
- Subjects
- Humans, Mice, Animals, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Transcription Factors genetics, Mutation, Gene Expression, Proteomics, Leukemia, Myeloid, Acute metabolism
- Abstract
Pharmacologic targeting of chromatin-associated protein complexes has shown significant responses in KMT2A-rearranged (KMT2A-r) acute myeloid leukemia (AML) but resistance frequently develops to single agents. This points to a need for therapeutic combinations that target multiple mechanisms. To enhance our understanding of functional dependencies in KMT2A-r AML, we have used a proteomic approach to identify the catalytic immunoproteasome subunit PSMB8 as a specific vulnerability. Genetic and pharmacologic inactivation of PSMB8 results in impaired proliferation of murine and human leukemic cells while normal hematopoietic cells remain unaffected. Disruption of immunoproteasome function drives an increase in transcription factor BASP1 which in turn represses KMT2A-fusion protein target genes. Pharmacologic targeting of PSMB8 improves efficacy of Menin-inhibitors, synergistically reduces leukemia in human xenografts and shows preserved activity against Menin-inhibitor resistance mutations. This identifies and validates a cell-intrinsic mechanism whereby selective disruption of proteostasis results in altered transcription factor abundance and repression of oncogene-specific transcriptional networks. These data demonstrate that the immunoproteasome is a relevant therapeutic target in AML and that targeting the immunoproteasome in combination with Menin-inhibition could be a novel approach for treatment of KMT2A-r AML., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
6. Menin inhibitor ziftomenib (KO-539) synergizes with drugs targeting chromatin regulation or apoptosis and sensitizes acute myeloid leukemia with MLL rearrangement or NPM1 mutation to venetoclax.
- Author
-
Rausch J, Dzama MM, Dolgikh N, Stiller HL, Bohl SR, Lahrmann C, Kunz K, Kessler L, Echchannaoui H, Chen CW, Kindler T, Döhner K, Burrows F, Theobald M, Sasca D, and Kühn MWM
- Subjects
- Humans, Myeloid-Lymphoid Leukemia Protein genetics, Nuclear Proteins genetics, Mutation, Apoptosis, Chromatin, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2023
- Full Text
- View/download PDF
7. Epigenetic targeting to enhance acute myeloid leukemia-directed immunotherapy.
- Author
-
Rausch J, Ullrich E, and Kühn MWM
- Subjects
- Humans, Immunotherapy, Azacitidine therapeutic use, Treatment Outcome, Epigenesis, Genetic, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
AML is a malignant disease of hematopoietic progenitor cells with unsatisfactory treatment outcome, especially in patients that are ineligible for intensive chemotherapy. Immunotherapy, comprising checkpoint inhibition, T-cell engaging antibody constructs, and cellular therapies, has dramatically improved the outcome of patients with solid tumors and lymphatic neoplasms. In AML, these approaches have been far less successful. Discussed reasons are the relatively low mutational burden of AML blasts and the difficulty in defining AML-specific antigens not expressed on hematopoietic progenitor cells. On the other hand, epigenetic dysregulation is an essential driver of leukemogenesis, and non-selective hypomethylating agents (HMAs) are the current backbone of non-intensive treatment. The first clinical trials that evaluated whether HMAs may improve immune checkpoint inhibitors' efficacy showed modest efficacy except for the anti-CD47 antibody that was substantially more efficient against AML when combined with azacitidine. Combining bispecific antibodies or cellular treatments with HMAs is subject to ongoing clinical investigation, and efficacy data are awaited shortly. More selective second-generation inhibitors targeting specific chromatin regulators have demonstrated promising preclinical activity against AML and are currently evaluated in clinical trials. These drugs that commonly cause leukemia cell differentiation potentially sensitize AML to immune-based treatments by co-regulating immune checkpoints, providing a pro-inflammatory environment, and inducing (neo)-antigen expression. Combining selective targeted epigenetic drugs with (cellular) immunotherapy is, therefore, a promising approach to avoid unintended effects and augment efficacy. Future studies will provide detailed information on how these compounds influence specific immune functions that may enable translation into clinical assessment., Competing Interests: MWMK receives honoraria and is a consultant for Pfizer, Kura Oncology, Jazz Pharmaceuticals, Bristol-Myers Squibb/Celgene Abbvie, and Servier; is on the speakers bureau of Gilead and receives travel support from Daiichi Sankyo. JR has received travel support from Abbvie. EU has a sponsored research project with Gilead and BMS. EU declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2023 Rausch, Ullrich and Kühn.)
- Published
- 2023
- Full Text
- View/download PDF
8. Intensive chemotherapy with or without gemtuzumab ozogamicin in patients with NPM1-mutated acute myeloid leukaemia (AMLSG 09-09): a randomised, open-label, multicentre, phase 3 trial.
- Author
-
Döhner H, Weber D, Krzykalla J, Fiedler W, Kühn MWM, Schroeder T, Mayer K, Lübbert M, Wattad M, Götze K, Fransecky L, Koller E, Wulf G, Schleicher J, Ringhoffer M, Greil R, Hertenstein B, Krauter J, Martens UM, Nachbaur D, Samra MA, Machherndl-Spandl S, Basara N, Leis C, Schrade A, Kapp-Schwoerer S, Cocciardi S, Bullinger L, Thol F, Heuser M, Paschka P, Gaidzik VI, Saadati M, Benner A, Schlenk RF, Döhner K, and Ganser A
- Subjects
- Adolescent, Adult, Female, Humans, Male, Middle Aged, Young Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cytarabine therapeutic use, Gemtuzumab therapeutic use, Nuclear Proteins genetics, Treatment Outcome, Tretinoin therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute diagnosis, Neoplasm Recurrence, Local drug therapy
- Abstract
Background: Acute myeloid leukaemia with mutated NPM1 is associated with high CD33 expression and intermediate-risk cytogenetics. The aim of this study was to evaluate intensive chemotherapy with or without the anti-CD33 antibody-drug conjugate gemtuzumab ozogamicin in participants with newly diagnosed, NPM1-mutated acute myeloid leukaemia., Methods: This open-label, phase 3 trial was conducted at 56 hospitals in Germany and Austria. Eligible participants were 18 years or older and had newly diagnosed NPM1-mutated acute myeloid leukaemia and an Eastern Cooperative Oncology Group performance status of 0-2. Participants were randomly assigned, using age as a stratification factor (18-60 years vs >60 years), 1:1 to the two treatment groups using allocation concealment; there was no masking of participants and investigators to treatment groups. Participants received two cycles of induction therapy (idarubicin, cytarabine, and etoposide) plus all-trans retinoic acid (ATRA) followed by three consolidation cycles of high-dose cytarabine (or an intermediate dose for those older than 60 years) and ATRA, without or with gemtuzumab ozogamicin (3 mg/m
2 administered intravenously on day 1 of induction cycles 1 and 2, and consolidation cycle 1). The primary endpoints were short-term event-free survival and overall survival in the intention-to-treat population (overall survival was added as a co-primary endpoint after amendment four of the protocol on Oct 13, 2013). The secondary endpoints were event-free survival with long-term follow-up, rates of complete remission, complete remission with partial haematological recovery (CRh), and complete remission with incomplete haematological recovery (CRi), cumulative incidences of relapse and death, and number of days in hospital. This trial is registered with ClinicalTrials.gov (NCT00893399) and has been completed., Findings: Between May 12, 2010, and Sept 1, 2017, 600 participants were enrolled, of which 588 (315 women and 273 men) were randomly assigned (296 to the standard group and 292 to the gemtuzumab ozogamicin group). No difference was found in short-term event-free survival (short-term event-free survival at 6-month follow-up, 53% [95% CI 47-59] in the standard group and 58% [53-64] in the gemtuzumab ozogamicin group; hazard ratio [HR] 0·83; 95% CI 0·65-1·04; p=0·10) and overall survival between treatment groups (2-year overall survival, 69% [63-74] in the standard group and 73% [68-78] in the gemtuzumab ozogamicin group; 0·90; 0·70-1·16; p=0·43). There was no difference in complete remission or CRi rates (n=267 [90%] in the standard group vs n=251 [86%] in the gemtuzumab ozogamicin group; odds ratio [OR] 0·67; 95% CI 0·40-1·11; p=0·15) and complete remission or CRh rates (n=214 [72%] vs n=195 [67%]; OR 0·77; 0·54-1·10; p=0·18), whereas the complete remission rate was lower with gemtuzumab ozogamicin (n=172 [58%] vs n=136 [47%]; OR 0·63; 0·45-0·80; p=0·0068). Cumulative incidence of relapse was significantly reduced by gemtuzumab ozogamicin (2-year cumulative incidence of relapse, 37% [95% CI 31-43] in the standard group and 25% [20-30] in the gemtuzumab ozogamicin group; cause-specific HR 0·65; 0·49-0·86; p=0·0028), and there was no difference in the cumulative incidence of death (2-year cumulative incidence of death 6% [4-10] in the standard group and 7% [5-11] in the gemtuzumab ozogamicin group; HR 1·03; 0·59-1·81; p=0·91). There were no differences in the number of days in hospital across all cycles between treatment groups. The most common treatment-related grade 3-4 adverse events were febrile neutropenia (n=135 [47%] in the gemtuzumab ozogamicin group vs n=122 [41%] in the standard group), thrombocytopenia (n=261 [90%] vs n=265 [90%]), pneumonia (n=71 [25%] vs n=64 [22%]), sepsis (n=85 [29%] vs n=73 [25%]). Treatment-related deaths were documented in 25 participants (4%; n=8 [3%] in the standard group and n=17 [6%] in the gemtuzumab ozogamicin group), mostly due to sepsis and infections., Interpretation: The primary endpoints of the trial of event-free survival and overall survival were not met. However, an anti-leukaemic efficacy of gemtuzumab ozogamicin in participants with NPM1-mutated acute myeloid leukaemia is shown by a significantly lower cumulative incidence of relapse rate, suggesting that the addition of gemtuzumab ozogamicin might reduce the need for salvage therapy in these participants. The results from this study provide further evidence that gemtuzumab ozogamicin should be added in the standard of care treatment in adults with NPM1-mutated acute myeloid leukaemia., Funding: Pfizer and Amgen., Competing Interests: Declaration of interests HD declares being in an advisory role for Abbvie, Agios, Amgen, Astellas, AstraZeneca, Berlin-Chemie, Bristol Myers Squibb, Celgene, Daiichi Sankyo, GEMoaB, Gilead, Janssen, Jazz Pharmaceuticals, Novartis, Servier, Stemline, and Syndax; and research funding from Abbvie, Agios, Amgen, Astellas, Bristol Myers Squibb, Jazz Pharmaceuticals, Kronos-Bio, Novartis, and Pfizer. WF declares a membership on an entity's board of directors or advisory committee for AbbVie, Amgen, ARIAD/Incyte, Celgene, Jazz Pharmaceuticals, MorphoSys, Stemline, Clinigen, Novartis, and Pfizer; patents and royalties from Amgen; support for meeting attendance from Amgen, Daiichi Sankyo, Gilead, Jazz Pharmaceuticals, and Servier; and research funding from Amgen and Pfizer. MWMK declares being in an advisory role for Abbvie, Bristol Myers Squibb, Jazz Pharmaceuticals, Kura-Oncology, and Pfizer; speakers honoraria from Abbvie and Gilead; travel support from Abbvie, Celgene, and Daiichi Sankyo; and research funding from Kura-Oncology. TS declares being in an advisory role for Abbvie, Astellas, Celgene, Janssen, Jazz Pharmaceuticals, Novartis, Takeda, Pfizer Bristol Myers Squibb, and Telix Pharma; speakers honoraria from Abbvie, Astellas, Celgene, Janssen, Jazz Pharmaceuticals, and Novartis; research funding from Jazz Pharmaceuticals and Bristol Myers Squibb; and travel support from Jazz Pharmaceuticals and Medac. KM declares being on an advisory role and paid for lectures for Bristol Myers Squibb; and travel support from Amgen, Astellas, Bristol Myers Squibb, Celgene, Jazz Pharmaceuticals, Novartis, Pfizer, and Roche. ML declares being in an advisory role for AbbVie, Astex Pharmaceuticals, Janssen, Pfizer, Otsuka, and Syros; and research funding from Aristopharm, Cheplapharm, Janssen, and TEVA. KG declares being in an advisory role for Abbvie, BMS, and Servier. LF declares being in an advisory role for AbbVie, Amgen, Medac, Novartis, and Takeda; and research funding from Abbvie, Kite, and Speaker's Bureau for Celgene. EK reports speaker honoraria from Abbvie, Astellas, Celgene/BMS, Jazz, Novartis, and Servier; and being in an advisory or consultancy role for Abbvie, Astellas, Celgene/BMS, Jazz, and Servier. GW reports consultancy for Clinigen and Novartis; and honoraria from Novartis, Gilead, and Takeda. JS reports being in an advisory role and honoraria from AbbVie, Bayer, BMS, Janssen, Novartis, Pfizer, and Sanofi. RG reports being in an advisory role for Abbvie, Astra Zeneca, BMS, Celgene, Daiichi Sankyo, Gilead, Janssen, Merck, MSD, Novartis, Roche, Sanofi, and Takeda. BH reports being in an advisory role for Bristol Myers Squibb, Celgene, Novartis, and Sanofi. UMM reports being in an advisory role for BMS and Sanofi. SM-S reports being in an advisory role for Amgen, BMS/Celgene, Jazz Pharmaceuticals, and Novartis. SK-S reports consultancy for Abbvie, Jazz Pharmaceuticlas, and Pfizer. LB reports being in an advisory role for Abbvie, Amgen, Astellas, Bristol-Myers Squibb, Celgene, Daiichi Sankyo, Gilead, Hexal, Janssen, Jazz Pharmaceuticals, Menarini, Novartis, Pfizer, Sanofi, and Seattle Genetics; and research funding from Bayer and Jazz Pharmaceuticals. FT reports being in an advisory role for Abbvie, Astellas, Bristol Myers Squibb/Celgene, Jazz Pharmaceuticals, Novartis, and Pfizer. MH reports being in an advisory role for Abbvie, BMS/Celgene, Daiichi Sankyo, Jazz Pharmaceuticals, Novartis, Pfizer, Roche, and Tolremo; honoraria from Jazz Pharmaceuticals, Janssen, and Novartis; and research funding to institution from Astellas, Bayer Pharma, BergenBio, Daiichi Sankyo, Jazz Pharmaceuticals, Karyopharm, Novartis, Pfizer, and Roche. PP reports being in an advisory role for Abbvie, Agios, Astellas, Astex Pharmaceuticals, Bristol Myers Squibb, Celgene, Jazz Pharmaceuticals, Novartis, Otsuka Pharma, Pfizer, and Sunesis; speakers bureau for Abbvie, Agios, Astellas, Bristol Myers Squibb, Celgene, Jazz Pharmaceuticals, Novartis, and Pfizer; and travel support for Abbvie, Bristol Myers Squibb, Celgene, Janssen, Novartis, and Takeda. VIG reports being in an advisory role for Abbvie and Pfizer; and speakers bureau for Pfizer and Janssen. RFS reports consulting for or an advisory board membership with Astellas, Daiichi Sankyo, Novartis, and Pfizer; research funding from AstraZeneca, Boehringer Ingelheim, Daiichi Sankyo, Pfizer, PharmaMar, and Roche; and travel, accommodations, and expenses covered by Daiichi Sankyo. KD reports being in an advisory role for Amgen, Bristol Myers Squibb, Celgene, Daiichi Sankyo, Janssen, Jazz Pharmaceuticals, Novartis, and Roche; and research funding from Agios, Astex, Astellas, Bristol Myers Squibb, Celgene, and Novartis. AG reports being in an advisory role for Celgene, JAZZ Pharmaceuticals, and Novartis. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
9. Midostaurin plus intensive chemotherapy for younger and older patients with AML and FLT3 internal tandem duplications.
- Author
-
Döhner H, Weber D, Krzykalla J, Fiedler W, Wulf G, Salih H, Lübbert M, Kühn MWM, Schroeder T, Salwender H, Götze K, Westermann J, Fransecky L, Mayer K, Hertenstein B, Ringhoffer M, Tischler HJ, Machherndl-Spandl S, Schrade A, Paschka P, Gaidzik VI, Theis F, Thol F, Heuser M, Schlenk RF, Bullinger L, Saadati M, Benner A, Larson R, Stone R, Döhner K, and Ganser A
- Subjects
- Adolescent, Adult, Aged, Humans, Middle Aged, Protein-Tyrosine Kinases, Staurosporine adverse effects, Staurosporine analogs & derivatives, Young Adult, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 therapeutic use, Hematopoietic Stem Cell Transplantation methods, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
We conducted a single-arm, phase 2 trial (German-Austrian Acute Myeloid Leukemia Study Group [AMLSG] 16-10) to evaluate midostaurin with intensive chemotherapy followed by allogeneic hematopoietic-cell transplantation (HCT) and a 1-year midosta urin maintenance therapy in adult patients with acute myeloid leukemia (AML) and fms-related tyrosine kinase 3 (FLT3) internal tandem duplication (ITD). Patients 18 to 70 years of age with newly diagnosed FLT3-ITD-positive AML were eligible. Primary and key secondary endpoints were event-free survival (EFS) and overall survival (OS). Results were compared with a historical cohort of 415 patients treated on 5 prior AMLSG trials; statistical analysis was performed using a double-robust adjustment with propensity score weighting and covariate adjustment. Results were also compared with patients (18-59 years) treated on the placebo arm of the Cancer and Leukemia Group B (CALGB) 10603/RATIFY trial. The trial accrued 440 patients (18-60 years, n = 312; 61-70 years, n = 128). In multivariate analysis, EFS was significantly in favor of patients treated within the AMLSG 16-10 trial compared with the AMLSG control (hazard ratio [HR], 0.55; P < .001); both in younger (HR, 0.59; P < .001) and older patients (HR, 0.42; P < .001). Multivariate analysis also showed a significant beneficial effect on OS compared with the AMLSG control (HR, 0.57; P < .001) as well as to the CALGB 10603/RATIFY trial (HR, 0.71; P = .005). The treatment effect of midostaurin remained significant in sensitivity analysis including allogeneic HCT as a time-dependent covariate. Addition of midostaurin to chemotherapy was safe in younger and older patients. In comparison with historical controls, the addition of midostaurin to intensive therapy led to a significant improvement in outcome in younger and older patients with AML and FLT3-ITD. This trial is registered at clinicaltrialsregistry.eu as Eudra-CT number 2011-003168-63 and at clinicaltrials.gov as NCT01477606., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
10. Primary CD33-targeting CAR-NK cells for the treatment of acute myeloid leukemia.
- Author
-
Albinger N, Pfeifer R, Nitsche M, Mertlitz S, Campe J, Stein K, Kreyenberg H, Schubert R, Quadflieg M, Schneider D, Kühn MWM, Penack O, Zhang C, Möker N, and Ullrich E
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Hematopoiesis, Humans, Immunotherapy, Adoptive, Mice, Sialic Acid Binding Ig-like Lectin 3 genetics, Killer Cells, Natural, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
Acute myeloid leukemia (AML) is a malignant disorder derived from neoplastic myeloid progenitor cells characterized by abnormal proliferation and differentiation. Although novel therapeutics have recently been introduced, AML remains a therapeutic challenge with insufficient cure rates. In the last years, immune-directed therapies such as chimeric antigen receptor (CAR)-T cells were introduced, which showed outstanding clinical activity against B-cell malignancies including acute lymphoblastic leukemia (ALL). However, the application of CAR-T cells appears to be challenging due to the enormous molecular heterogeneity of the disease and potential long-term suppression of hematopoiesis. Here we report on the generation of CD33-targeted CAR-modified natural killer (NK) cells by transduction of blood-derived primary NK cells using baboon envelope pseudotyped lentiviral vectors (BaEV-LVs). Transduced cells displayed stable CAR-expression, unimpeded proliferation, and increased cytotoxic activity against CD33-positive OCI-AML2 and primary AML cells in vitro. Furthermore, CD33-CAR-NK cells strongly reduced leukemic burden and prevented bone marrow engraftment of leukemic cells in OCI-AML2 xenograft mouse models without observable side effects., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Cluster of differentiation 33 single nucleotide polymorphism rs12459419 is a predictive factor in patients with nucleophosmin1 -mutated acute myeloid leukemia receiving gemtuzumab ozogamicin.
- Author
-
Teich K, Krzykalla J, Kapp-Schwoerer S, Gaidzik VI, Schlenk RF, Paschka P, Weber D, Fiedler W, Kühn MWM, Schroeder T, Mayer K, Lübbert M, Ramachandran D, Benner A, Ganser A, Döhner H, Heuser M, Döhner K, and Thol F
- Subjects
- Aminoglycosides therapeutic use, Gemtuzumab, Humans, Sialic Acid Binding Ig-like Lectin 3 genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Polymorphism, Single Nucleotide
- Published
- 2021
- Full Text
- View/download PDF
12. Next generation epigenetic modulators to target myeloid neoplasms.
- Author
-
Sasca D, Guezguez B, and Kühn MWM
- Subjects
- Humans, Nucleosomes genetics, Nucleosomes metabolism, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Epigenesis, Genetic drug effects, Gene Expression Regulation, Neoplastic drug effects, Hematologic Neoplasms drug therapy, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Transcription, Genetic drug effects
- Abstract
Purpose of Review: Comprehensive sequencing studies aimed at determining the genetic landscape of myeloid neoplasms have identified epigenetic regulators to be among the most commonly mutated genes. Detailed studies have also revealed a number of epigenetic vulnerabilities. The purpose of this review is to outline these vulnerabilities and to discuss the new generation of drugs that exploit them., Recent Findings: In addition to deoxyribonucleic acid-methylation, novel epigenetic dependencies have recently been discovered in various myeloid neoplasms and many of them can be targeted pharmacologically. These include not only chromatin writers, readers, and erasers but also chromatin movers that shift nucleosomes to allow access for transcription. Inhibitors of protein-protein interactions represent a novel promising class of drugs that allow disassembly of oncogenic multiprotein complexes., Summary: An improved understanding of disease-specific epigenetic vulnerabilities has led to the development of second-generation mechanism-based epigenetic drugs against myeloid neoplasms. Many of these drugs have been introduced into clinical trials and synergistic drug combination regimens have been shown to enhance efficacy and potentially prevent drug resistance., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition.
- Author
-
Dzama MM, Steiner M, Rausch J, Sasca D, Schönfeld J, Kunz K, Taubert MC, McGeehan GM, Chen CW, Mupo A, Hähnel P, Theobald M, Kindler T, Koche RP, Vassiliou GS, Armstrong SA, and Kühn MWM
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Cell Line, Tumor, Coculture Techniques, Drug Synergism, Humans, Leukemia, Myeloid, Acute genetics, Mice, Mice, Inbred NOD, Myeloid Ecotropic Viral Integration Site 1 Protein biosynthesis, Myeloid Ecotropic Viral Integration Site 1 Protein genetics, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Nuclear Proteins genetics, Nucleophosmin, Phosphorylation, Protein Kinase Inhibitors pharmacology, Protein Processing, Post-Translational, Random Allocation, Transcription, Genetic drug effects, fms-Like Tyrosine Kinase 3 biosynthesis, fms-Like Tyrosine Kinase 3 genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Gene Expression Regulation, Leukemic drug effects, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Leukemia, Myeloid, Acute drug therapy, Myeloid-Lymphoid Leukemia Protein antagonists & inhibitors, Neoplasm Proteins antagonists & inhibitors, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins antagonists & inhibitors, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
The interaction of menin (MEN1) and MLL (MLL1, KMT2A) is a dependency and provides a potential opportunity for treatment of NPM1-mutant (NPM1mut) and MLL-rearranged (MLL-r) leukemias. Concomitant activating driver mutations in the gene encoding the tyrosine kinase FLT3 occur in both leukemias and are particularly common in the NPM1mut subtype. In this study, transcriptional profiling after pharmacological inhibition of the menin-MLL complex revealed specific changes in gene expression, with downregulation of the MEIS1 transcription factor and its transcriptional target gene FLT3 being the most pronounced. Combining menin-MLL inhibition with specific small-molecule kinase inhibitors of FLT3 phosphorylation resulted in a significantly superior reduction of phosphorylated FLT3 and transcriptional suppression of genes downstream of FLT3 signaling. The drug combination induced synergistic inhibition of proliferation, as well as enhanced apoptosis, compared with single-drug treatment in models of human and murine NPM1mut and MLL-r leukemias harboring an FLT3 mutation. Primary acute myeloid leukemia (AML) cells harvested from patients with NPM1mutFLT3mut AML showed significantly better responses to combined menin and FLT3 inhibition than to single-drug or vehicle control treatment, whereas AML cells with wild-type NPM1, MLL, and FLT3 were not affected by either of the 2 drugs. In vivo treatment of leukemic animals with MLL-r FLT3mut leukemia reduced leukemia burden significantly and prolonged survival compared with results in the single-drug and vehicle control groups. Our data suggest that combined menin-MLL and FLT3 inhibition represents a novel and promising therapeutic strategy for patients with NPM1mut or MLL-r leukemia and concurrent FLT3 mutation., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.