124 results on '"Justus Duyster"'
Search Results
2. Phase I Study of the LSD1 Inhibitor Tranylcypromine (TCP) in Combination with All-Trans Retinoic Acid (ATRA) and Low-Dose Cytarabine (LDAC) in Elderly, Medically Non-Fit Patients with AML or High-Risk MDS (TRANSATRA trial)
- Author
-
Michael Lübbert, Claudia Schmoor, Tobias Berg, Michael Kruszewski, Marcus Matthias Schittenhelm, Katharina Götze, Caroline Pabst, Andrea Kündgen, Tobias Ma, Anna Frey, Kevin Moschallski, Usama-Ur Rehman, Johannes Jung, Justus Duyster, Manfred Jung, Roland Schüle, Ralph Wäsch, and Olga Grishina
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
3. Establishment and Characterization of Mouse Models with Primary and Acquired BTK Inhibitor Resistance
- Author
-
Laura Polcik, Svenja Dannewitz Prosseda, Chingiz Underbayev, Geoffroy Andrieux, Lixia Li, Danielle-Justine Danner, Driti Ashok, Sandra Kissel, Sara Parsa, Natalie Koehler, Melanie Börries, Justus Duyster, Elena Bibikova, Adrian Wiestner, and Tanja Nicole Hartmann
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
4. First-in-Human Study of WT1 Recombinant Protein Vaccination in Elderly Patients with AML in Remission: A Single-Center Experience
- Author
-
Michael Schmitt, Dietmar Pfeifer, Miguel Waterhouse, Justus Duyster, Konstanze Döhner, Michael Lübbert, Stefanie Kreutmair, Anna Frey, and Anna Lena Illert
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,First in human ,Single Center ,Biochemistry ,law.invention ,Vaccination ,law ,Internal medicine ,medicine ,Recombinant DNA ,business - Abstract
Background Vaccination (vac) strategies to maintain remissions in AML have been pursued for decades. The usage of recombinant proteins instead of peptides allows a potential immune response to multiple epitopes, hence could be offered to all patients (pts) independent of HLA expression. Wilms' tumor 1 (WT1) protein is highly immunogenic and frequently overexpressed in AML, thus ranked as a very promising target for novel immunotherapies. Here we report a single-center experience of a phase I/II clinical trial (NCT01051063) of a first-in-human vac strategy based on WT1 recombinant protein (WT1-A10) together with vaccine adjuvant AS01 B in 5 elderly AML pts. Patients and Methods Key eligibility criteria: overexpression of WT1 transcripts in AML blasts at diagnosis (qRT-PCR); 1 or 2 induction chemotherapies, with partial remission (PR) or morphologic complete remission with incomplete blood count recovery (CRi). The vaccine consisted of WT1-A10, a truncated WT-1 protein retaining the N-terminus (amino acids 2-281) of full length WT1 protein (429 aa) linked to the first 11 amino acids of trimethylamine N-oxide reductase signal peptide via one histidine residue combined with the liquid AS01 B adjuvant. AS01 B is an Adjuvant System containing MPL (3-O-desacyl-4´-monophosphoryl lipid A), QS-21 (Quillaja saponaria Molina, fraction 21) and liposome (50µg MPL and 50µg QS-21). One human dose of WT1-A10 + AS01 B contained 200 μg of WT1-A10 antigen. Pts received the vaccine by i.m. injection. To assess cellular response, antigen-specific stimulation of cultured PBMCs was performed with a pool of 123 15mer peptides covering the entire WT1 (1μg/ml/peptide), together with irrelevant re-stimulation plus negative control peptide. CD4 + and CD8 + T cells were serially assessed by intracellular flow cytometry for their ability to produce both IFN-γ and TNF upon antigen stimulation. Results A total of 5 pts (median age 69, range 63-75) were enrolled on the WT1 protein-based vac study at our institution (Table 1), receiving a total of 62 vac after a median of 3 courses (1-5) of standard chemotherapy. The repeated vac had an acceptable safety profile and were thus well tolerated. 2/5 pts experienced therapy-related toxicity, injection site pain (CTCAE v.3, grade 2) and injection site inflammation (CTCAE v.3, grade 1). Symptoms were of mild / moderate severity and resolved completely. No hematologic toxicity was noted. With a median progression-free survival of 28.8 mths (range 1-59) and median overall survival (OS) of 35.4 mths (range 3-75) from the 1st vac, this older patient cohort showed above-average clinical outcome (Table 1), pointing to a potential clinical efficacy of WT1-based vac therapy. All vaccinated pts showed highly elevated WT1 ratios before WT1-based vac therapy and normal levels after vac (Fig. 1A). Two pts demonstrated early relapse after 3 WT1 protein-based vac, and clinical benefit was observed in 3 pts: one achieved complete and sustained measurable residual disease clearance (NPM1 ratios) during WT1 vac, resulting in molecular CR at the 18th vac. The pt died from unrelated reasons 5.5 years after initial diagnosis of AML, 3.5 years after the last WT1 vac, with continued molecular CR. One pt maintained long-term hematological and molecular remission over 59 mths, until molecular relapse occurred 11 mths after the final, 21 st vac. Interestingly, in one case, a complete clonal switch occurred at hematologic relapse following 18 vac, with loss of WT1 overexpression: while the clone at initial diagnosis harbored FLT3, NPM1 and SRSF2 mutations, BRAF, KRAS and STAG2 mutations were detected at relapse (Fig. 1B), pointing to an ongoing suppression of the WT1 expressing AML clone. Flow cytometry studies were conducted in one pt to elucidate specific cellular immune responses. We noted CD4 + T cell immune responses by strong IFN-γ and TNF expression (Fig. 1C), suggestive of efficient immune stimulation post-vac, while CD8 + T cells failed to upregulate these key cytokines. Conclusions This vac strategy showed good feasibility, with a very acceptable safety profile, and appeared to extend remissions beyond the expected duration, together with MRD clearance. Thus, our data provide evidence of potential clinical efficacy of WT1 protein-based vac therapy in AML pts, making this maintenance approach an attractive alternative to more complex strategies, particularly in elderly pts with comorbidities. Figure 1 Figure 1. Disclosures Döhner: Abbvie: Consultancy, Honoraria; Jazz Roche: Consultancy, Honoraria; Daiichi Sankyo: Honoraria, Other: Advisory Board; Janssen: Honoraria, Other: Advisory Board; Celgene/BMS: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Astellas: Research Funding; Agios and Astex: Research Funding. Schmitt: MSD: Membership on an entity's Board of Directors or advisory committees; TolerogenixX: Current holder of individual stocks in a privately-held company; Bluebird Bio: Other: Travel grants; Hexal: Other: Travel grants, Research Funding; Novartis: Other: Travel grants, Research Funding; Kite Gilead: Other: Travel grants; Apogenix: Research Funding. Lübbert: Teva: Research Funding; Janssen: Research Funding; Cheplapharm: Research Funding; Aristopharm: Research Funding; Syros: Honoraria; Pfizer: Honoraria; Janssen: Honoraria, Research Funding; Imago BioSciences: Honoraria; Hexal: Honoraria; Astex: Honoraria; Abbvie: Honoraria.
- Published
- 2021
5. Profiling of Circulating Tumor DNA for Noninvasive Disease Detection, Risk Stratification, and MRD Monitoring in Patients with CNS Lymphoma
- Author
-
Jurik Andreas Mutter, Stefan Alig, Eliza Maria Lauer, Mohammad Shahrokh Esfahani, Jan Mitschke, David M. Kurtz, Julia Kühn, Sabine Bleul, Mari Olsen, Chih Long Liu, Michael C. Jin, Charles Macaulay, Nicolas Noel Neidert, Timo Volk, Sebastian Rauer, Dieter Henrik Heiland, Jürgen Finke, Justus Duyster, Julius Wehrle, Marco Prinz, Gerald Illerhaus, Peter C. Reinacher, Elisabeth Schorb, Maximilian Diehn, Ash A. Alizadeh, and Florian Scherer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Introduction: Clinical outcomes for patients with central nervous system lymphoma (CNSL) are remarkably heterogeneous, yet identification of patients at high risk for treatment failure remains challenging with existing methods. In addition, diagnosis of CNSL requires invasive neurosurgical biopsies that carry procedural risks and often cannot be performed in frail or elderly patients. Circulating tumor DNA (ctDNA) has shown great potential as a noninvasive biomarker in systemic lymphomas. Yet, previous studies revealed low ctDNA detection rates in blood plasma of CNSL patients. In this study, we utilized ultrasensitive targeted high-throughput sequencing technologies to explore the role of ctDNA for disease classification, MRD detection, and early prediction of clinical outcomes in patients with CNSL. Methods: We applied Cancer Personalized Profiling by Deep Sequencing (CAPP-Seq) and Phased Variant Enrichment and Detection Sequencing (PhasED-Seq, Kurtz et al, Nat Biotech 2021) to 85 tumor biopsies, 131 plasma samples, and 62 CSF specimens from 92 CNSL patients and 44 patients with other brain cancers or inflammatory cerebral diseases, targeting 794 distinct genetic regions. Concentrations of ctDNA were correlated with radiological measures of tumor burden and tested for associations with clinical outcomes at distinct clinical time points. We further developed a novel classifier to noninvasively distinguish CNS lymphomas from other CNS tumors based on their mutational landscapes in plasma and CSF, using supervised training of a machine learning approach from tumor whole genome sequencing data and own genotyping analyses, followed by its independent validation. Results: We identified genetic aberrations in 100% of CNSL tumor biopsies (n=63), with a median of 262 mutations per patient. Pretreatment plasma ctDNA was detectable in 78% of plasma samples and in 100% of CSF specimens (Fig. 1a), with ctDNA concentrations ranging from 0.0004 - 5.94% allele frequency (AF, median: 0.01%) in plasma and 0.0049 - 50.47% AF (median: 0.62%) in CSF (Fig. 1b). Compared to ctDNA concentrations in patients with systemic diffuse large B-cell lymphoma (DLBCL, data from Kurtz et al., J Clin Oncol, 2018), plasma ctDNA levels in CNSL were in median more than 200-fold lower (Fig. 1b). We observed a significant correlation of ctDNA concentrations with total radiographic tumor volumes (TRTV) measured by MRI (Fig. 1c,d), but no association with clinical risk scores (i.e., MSKCC score) or concurrent steroid treatment. Assessment of ctDNA at pretreatment time points predicted progression-free survival (PFS) and overall survival (OS), both as continuous and binary variable (Fig. 1e,f). Notably, patients could be stratified into risk groups with particularly favorable or poor prognoses by combining ctDNA and TRTV as pretreatment biomarkers (Fig. 1g). Furthermore, ctDNA positivity during curative-intent induction therapy was significantly associated with clinical outcomes, both PFS and OS (Fig. 1h). Finally, we applied our novel machine learning classifier to 207 specimens from an independent validation cohort of CNSL and Non-CNSL patients. We observed high specificity (100%) and positive predictive value (100%) for noninvasive diagnosis of CNSL, with a sensitivity of 57% for CSF and 21% for plasma, suggesting that a significant subset of CNSL patients might be able to forego invasive surgical biopsies. Conclusions: We demonstrate robust and ultrasensitive detection of ctDNA at various disease milestones in CNSL. Our findings suggest that ctDNA accurately mirrors tumor burden and serves as a valuable clinical biomarker for risk stratification, outcome prediction, and surgery-free lymphoma classification in CNSL. We foresee an important potential future role of ctDNA as a decision-making tool to guide treatment in patients with CNSL. Figure 1 Figure 1. Disclosures Esfahani: Foresight Diagnostics: Current holder of stock options in a privately-held company. Kurtz: Genentech: Consultancy; Roche: Consultancy; Foresight Diagnostics: Consultancy, Current holder of stock options in a privately-held company. Schorb: Riemser Pharma GmbH: Honoraria, Research Funding; Roche: Research Funding; AbbVie: Research Funding. Diehn: BioNTech: Consultancy; RefleXion: Consultancy; Roche: Consultancy; AstraZeneca: Consultancy; Foresight Diagnostics: Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company; CiberMed: Current holder of stock options in a privately-held company, Patents & Royalties; Illumina: Research Funding; Varian Medical Systems: Research Funding. Alizadeh: Foresight Diagnostics: Consultancy, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company; Gilead: Consultancy; Roche: Consultancy, Honoraria; Celgene: Consultancy, Research Funding; Janssen Oncology: Honoraria; CAPP Medical: Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company; Forty Seven: Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company; Cibermed: Consultancy, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company; Bristol Myers Squibb: Research Funding.
- Published
- 2021
6. Randomized Phase II Study of All-Trans Retinoic Acid and Valproic Acid Added to Decitabine in Newly Diagnosed Elderly AML Patients (DECIDER trial): Predictive Impact of TP53 Status
- Author
-
Ralph Wäsch, Richard F. Schlenk, Heiko Becker, Katharina Goetze, Sebastian Scholl, Carsten Schwaenen, Carsten Müller-Tidow, Dennis Zimmer, Hartmut Döhner, Aristoteles Giagounidis, Andreas Neubauer, Michael Lübbert, Edgar Jost, Martina Crysandt, Gerhard Heil, Annette M. May, Björn Hackanson, Felicitas Thol, Ulrich Germing, Claudia Schmoor, Michael Heuser, Helmut R. Salih, Justus Duyster, Marcus M. Schittenhelm, Jürgen Krauter, Konstanze Döhner, Maike de Wit, Andrea Kündgen, Gesine Bug, Dietmar Pfeifer, Arnold Ganser, Wolfram Brugger, and Olga Grishina
- Subjects
Oncology ,Valproic Acid ,medicine.medical_specialty ,business.industry ,Immunology ,All trans ,Retinoic acid ,Decitabine ,Phases of clinical research ,Cell Biology ,Hematology ,Newly diagnosed ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Background TP53 mutations are associated with adverse outcome of AML treated with cytarabine-based regimens. Interestingly, DNA-hypomethylating agents (HMAs) induce a higher response rate in TP53-mutated (MUT) compared to TP53 wildtype (WT) AML (Welch et al., N. Engl. J. Med. 2016, Döhner et al., Leukemia 2018). We conducted a randomized phase II trial (NCT00867672, 2x2 factorial design) asking whether the in vitro cooperativity of DAC with VPA or ATRA translates into clinical benefit. While VPA added to DAC affected neither objective response rate (ORR) nor overall survival (OS), ATRA significantly improved ORR and OS, without added toxicity (Lübbert et al., J. Clin. Oncol. 2020). Preclinical data suggest that HMAs and ATRA have cooperative effects also in TP53 MUT AML. We therefore performed a post-hoc analysis to determine the predictive impact of TP53 status. Patients and Methods Key inclusion criteria: newly diagnosed AML pts >60 yr (non-M3) unfit for induction, ECOG performance status (PS) 0-2. Treatment: DAC 20 mg/m 2 day 1-5 (arms A/B/C/D), VPA p.o. from day 6 (arms B/D), ATRA p.o. day 6-28 (arms C/D) of each 28-day course. For TP53 mutation analyses, the Illumina TruSight Myeloid Sequencing Panel was used for library preparation and an Illumina MiSeq device for sequencing. Key endpoints: ORR (CR/CRi/PR, ELN 2010 criteria) and OS. Original sample size calculation of a total of 200 patients (pts) was based on the primary endpoint ORR (Lübbert et al., Haematologica 2012). ORR was analyzed with logistic regression, OS with Cox regression. Odds ratios (OR) for the effect on ORR and hazard ratios (HR) for the effect on death with 95% confidence intervals (CI) are presented in the genetic subgroups TP53 MUT and TP53 WT including tests for interactions (TFI) between treatment and TP53. These are post-hoc exploratory analyses, hence p-values have to be interpreted in a descriptive sense. Results Between 12/2011 and 2/2015, 200 pts were randomized and treated. Information on TP53 status was available for 168 of 200 pts (84%); 155 of them (92%) had died at last follow-up (June 2021). 61% of pts were aged >75 years (range 61-92), ECOG PS 0/1/2: 19/62/19% (a single pt had a PS of 3); 53% had an HCT-CI >3, 19% WBC >30.000/µl, 30% adverse genetics (ELN 2010), 51% an antecedent hematologic disorder. TP53 aberrations were detected in 42 pts (25%), with 1 (n=27) or 2 mutations (n=12, median variant allele frequency 44%, range, 1.3-99%) in 39 pts, and TP53 deletions in 3 additional pts. The 42 pts with TP53 MUT showed a higher ORR (23.8%) than the 126 pts with TP53 WT (ORR 15.1%), with an OR of 2.04 (95% CI 0.83-4.98), p=0.12. OS (irrespective of treatment) in the TP53 MUT v WT pts was not different (HR, adjusted for treatment: 1.14 [95% CI 0.78-1.66], p=0.51; Fig. A). In both genetic groups, the addition of ATRA had a favorable effect on ORR (ATRA v no ATRA in TP53 MUT: 31.3% v 19.2%, OR 1.91 [95% CI 0.45-8.03]; ATRA v no ATRA in TP53 WT: 18.8% v 10.5%, OR 1.98 [95% CI 0.70-5.61]), TFI p=0.97 (Fig. B). A positive effect of ATRA on OS in both genetic groups was reflected by a median OS of 6.0 v 4.5 months (ATRA v no ATRA in TP53 MUT: HR 0.75 [95% CI 0.38-1.48]), and a median OS of 8.9 v 4.7 months (ATRA v no ATRA in TP53 WT: HR 0.58 [95% CI 0.39-0.86], all results adjusted for VPA, ECOG, HCT-CI, sLDH, Hb), TFI p=0.49 (Fig. C). VPA did not affect ORR in either of the 2 genetic groups (VPA v no VPA in TP53 MUT: 21.7% v 26.3%, OR 0.76 [95% CI 0.18-3.21]; VPA v no VPA in TP53 WT: 16.7% v 13.3%, OR 1.34 [95% CI 0.5-3.61]), TFI p=0.53. The impact of VPA on OS differed between TP53 MUT pts (VPA v no VPA: median OS of 4.2 v 5.3 months, HR 1.31 [95% CI 0.69-2.48], and TP53 WT pts (VPA v no VPA: median OS of 8.4 v 4.8 months, HR 0.67 [95% CI 0.46-0.99], all results adjusted for ATRA, ECOG, HCT-CI, sLDH, Hb; TFI p=0.08, Fig. C). Conclusions Our results confirm the reported higher response rate to DAC in pts with TP53 MUT compared to TP53 WT; the addition of ATRA led to a higher ORR. Improved OS with ATRA was observed particularly in TP53 WT pts. In contrast, VPA did not affect the ORR in either genetic group; TP53 WT pts may benefit from VPA regarding OS. Our exploratory post-hoc results need confirmation in other trials, e.g. in the DECIDER-2 study (adding ATRA or placebo to the recently approved dual treatment of a HMA combined with venetoclax). Cooperative effects of HMAs and retinoids deserve a deeper mechanistic understanding, which may have implications not only for AML but also for other malignancies with impaired TP53. Figure 1 Figure 1. Disclosures Becker: BMS: Honoraria; Pierre Fabre Pharma: Honoraria; Servier: Honoraria; MSD: Honoraria; Novartis: Honoraria. Crysandt: Incyte: Honoraria; Pfizer: Membership on an entity's Board of Directors or advisory committees. Thol: Abbvie: Honoraria; Pfizer: Honoraria; Astellas: Honoraria; Novartis: Honoraria; BMS/Celgene: Honoraria, Research Funding; Jazz: Honoraria. Heuser: Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding; Tolremo: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Research Funding; Jazz: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Membership on an entity's Board of Directors or advisory committees, Research Funding; BergenBio: Research Funding; BMS/Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer Pharma AG: Research Funding; Astellas: Research Funding. Goetze: Abbvie: Other: Advisory Board; BMS/Celgene: Other: Advisory Board, Research Funding. Schlenk: Agios: Honoraria; Astellas: Honoraria, Research Funding, Speakers Bureau; Celgene: Honoraria; Daiichi Sankyo: Honoraria, Research Funding; Abbvie: Honoraria; Hexal: Honoraria; Neovio Biotech: Honoraria; Novartis: Honoraria; Pfizer: Honoraria, Research Funding, Speakers Bureau; Roche: Honoraria, Research Funding; AstraZeneca: Research Funding; Boehringer Ingelheim: Research Funding. Salih: Synimmune GmbH: Honoraria; Pfizer: Honoraria; Novartis: Honoraria; Celgene: Honoraria; BMS: Honoraria. Schittenhelm: Takeda: Other: advisory board; Astellas: Other: advisory board; BMS: Other: advisory board; University of Tuebingen: Patents & Royalties: patent for ASPP2k. Müller-Tidow: Pfizer: Research Funding; Janssen: Consultancy, Research Funding; Bioline: Research Funding. Germing: Novartis: Honoraria, Research Funding; Jazz Pharmaceuticals: Honoraria; Janssen: Honoraria; Celgene: Honoraria; Bristol-Myers Squibb: Honoraria, Other: advisory activity, Research Funding. Giagounidis: Novartis: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees. Wäsch: Amgen: Consultancy, Honoraria; Pfizer: Consultancy; Sanofi: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Novartis: Consultancy; BMS/Celgene: Consultancy; Gilead: Consultancy. Döhner: Jazz Roche: Consultancy, Honoraria; Celgene/BMS: Consultancy, Honoraria, Research Funding; Daiichi Sankyo: Honoraria, Other: Advisory Board; Agios and Astex: Research Funding; Abbvie: Consultancy, Honoraria; Janssen: Honoraria, Other: Advisory Board; Astellas: Research Funding; Novartis: Consultancy, Honoraria, Research Funding. Ganser: Novartis: Honoraria; Jazz Pharmaceuticals: Honoraria; Celgene: Honoraria. Döhner: Astellas: Consultancy, Honoraria, Research Funding; Bristol Myers Squibb: Consultancy, Honoraria, Research Funding; Oxford Biomedicals: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria, Research Funding; Helsinn: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; AstraZeneca: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; Berlin-Chemie: Consultancy, Honoraria; Astex: Consultancy, Honoraria; Agios: Consultancy, Honoraria, Research Funding; Ulm University Hospital: Current Employment; Jazz: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Roche: Consultancy, Honoraria; GEMoaB: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Research Funding; Pfizer: Research Funding. Hackanson: Roche: Consultancy, Honoraria; Astra Zeneca: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Boehringer-Ingelheim: Consultancy, Honoraria; MSD: Consultancy, Honoraria. Lübbert: Cheplapharm: Other: study drug (ATRA); TEVA: Other: study drug (valproic acid); Janssen: Consultancy, Other: study drug (decitabine), Research Funding; Syros: Consultancy, Honoraria; Aristopharm: Other: study drug ; Imago BioSciences: Consultancy, Other: study support with study drug; AbbVie: Consultancy, Honoraria; Astex: Consultancy, Honoraria. OffLabel Disclosure: ATRA, in non-M3 AML valproic acid, in non-M3 AML
- Published
- 2021
7. Activity of Decitabine (DAC) Combined with All-Trans Retinoic Acid (ATRA) in Oligoblastic AML: Subgroup Analysis of a Randomized 2x2 Phase II Trial
- Author
-
Ulrich Germing, Andreas Neubauer, Carsten Mueller-Tidow, Justus Duyster, Aristoteles Giagounidis, Gerhard Heil, Claudia Schmoor, Björn Hackanson, Felicitas Thol, Sebastian Scholl, Hartmut Döhner, Ralph Wäsch, Jürgen Krauter, Carsten Schwaenen, Annette M. May, Maike de Wit, Richard F. Schlenk, Andrea Kündgen, Christoph Rummelt, Gesine Bug, Martina Crysandt, Michael Luebbert, Konstanze Döhner, Heiko Becker, Michael Heuser, Stephan Kremers, Marcus M. Schittenhelm, Arnold Ganser, Wolfram Brugger, Helmut R. Salih, Edgar Jost, Olga Grishina, and Katharina Götze
- Subjects
Chemistry ,Immunology ,Retinoic acid ,All trans ,Decitabine ,Subgroup analysis ,Cell Biology ,Hematology ,Biochemistry ,chemistry.chemical_compound ,Phase (matter) ,Cancer research ,medicine ,medicine.drug - Abstract
Background: DNA-hypomethylating agents are providing a very well-accepted backbone for non-intensive combination treatment of AML/MDS patients (pts), and an in vivo synergism has been demonstrated for the azacitidine+venetoclax combination in the VIALE-A trial (DiNardo et al., EHA 2020). The DAC+ATRA combination also resulted in an improved response rate and survival compared to DAC without ATRA (DECIDER trial, Lübbert et al., J. Clin. Oncol. 2020), also in pts with prior hematologic disorder (mostly MDS); no benefit was seen when valproic acid (VPA) was added to DAC (2x2 factorial design). In a previous study, we had investigated the outcome of elderly pts with oligoblastic AML (i.e. with 20-30% bone marrow blasts, defined as MDS RAEBt according to the French-American-British classification) treated with either DAC or best supportive care within the EORTC 06011 phase III trial (Becker et al., Ann. Hematol. 2015), observing a median overall survival (OS) of 8.0 months (mths) in DAC-treated RAEBt pts. We now hypothesized that the outcome of pts with oligoblastic AML may be improved by the addition of ATRA to DAC. Therefore, in the present exploratory subgroup analysis, pts from the DECIDER cohort with 20-30% bone marrow blasts were analyzed for clinical outcome. Patients and Methods: Key inclusion criteria: newly diagnosed pts >60 years (yr), unfit for induction with non-M3 AML (WHO, de novo or after antecedent hematologic disorder [AHD], therapy-associated [t]AML), ECOG performance status (PS) 0-2. Treatment: DAC 20 mg/m2 day 1-5 (treatment arms A/B/C/D), ATRA p.o. day 6-28 (arms C/D), VPA p.o. continuously from day 6 (arms B/D), of each 28-day course (repeated until relapse/progression, prohibitive toxicity, withdrawal or death). Key endpoints: objective response rate (ORR): CR/CRi/PR, overall (OS) and event-free survival (EFS). Sample size calculation was based on the primary endpoint ORR, assuming an ORR of 25% in arm A (Lübbert et al., Haematologica 2012). For a power of 80% (test in this phase II study at 1-sided alpha=0.1) for an increase of ORR to 40% with ATRA or VPA, 176 pts were necessary, planned sample size 200. Between 12/2011 and 2/2015, 200 pts were randomized and treated. Efficacy analyses were performed in the intention-to-treat (ITT) population. ATRA was investigated by comparing arms C+D vs arms A+B, VPA by comparing arms B+D vs arms A+C, ORR was analyzed with logistic regression estimating odds ratios (OR), OS/EFS with Cox regression estimating hazard ratios (HR), each with 95% confidence intervals (CI), and presented with descriptive two-sided p values of the tests of no treatment effect. Central hematopathologic review (blinded as to treatment arms) was conducted by an independent morphologist. Results: In 56/200 pts of the DECIDER cohort, bone marrow blasts were 20-30% (median, 25%). The number of pts in the randomized arms were: 13 in arm A, 21 in arm B, 9 in arm C, 13 in arm D. Baseline pt characteristics were as follows: male 77%, median age: 75 yr (range 61-88), median WBC: 3400/µl (range 500-52,600), adverse genetics (ELN 2010) present in 25%, ECOG 2 in 13%, comorbidities (HCT-CI) ≥ 3 in 48%, AHD in 68%, tAML in 11% (only slight random imbalances across randomized treatment arms). A median of 5 DAC courses were administered (per arm: 2/5/11/4). Six pts attained a CR, 7 pts a CRi, and 1 pt a PR, resulting in an ORR of 25% (arm A: 7.7%, arm B: 28.6%, arm C: 33.3%, arm D: 30.8%, respectively). Effect on ORR of ATRA vs no ATRA (31.8 vs 20.6%): OR 1.85, CI [0.54,6.37], p=0.33; and of VPA vs no VPA (29.4 vs 18.2%): OR 1.93, CI [0.51,7.24], p=0.33. With 40 deaths out of 56 pts, median OS was 9.5 mths (arm A: 7.6 mths, arm B: 8.9 mths, arm C: 37.2 mths, arm D: 11.2 mths, respectively). Effect on OS of ATRA vs no ATRA (12.5 vs 7.6 mths median OS): HR 0.47, CI [0.24,0.94], p=0.032 (after adjustment for PS, HCT-CI, WBC, LDH, genetic risk: HR 0.42, CI [0.19,0.90], p=0.025); and of VPA vs no VPA (10.0 vs 8.4 mths median OS): HR 0.99, CI [0.51,1.92], p=0.98: A comparable benefit on EFS of ATRA vs no ATRA (but not VPA vs no VPA) was observed. Conclusion: In elderly pts with oligoblastic AML ineligible for induction chemotherapy, the addition of ATRA, but not VPA, to DAC resulted in a clinically meaningful survival benefit; OS of pts receiving DAC without ATRA was very similar to that observed in a previous study. It is tempting to speculate that the combination of an HMA with a retinoid such as ATRA may also be active in MDS pts with excess of blasts. Disclosures Jost: JAZZ: Other: travel support; Roche: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Celgene: Other: travel support. Thol:Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Heuser:Amgen: Research Funding; Bayer: Consultancy, Research Funding; BerGenBio ASA: Research Funding; Daiichi Sankyo: Consultancy, Research Funding; Stemline Therapeutics: Consultancy; Janssen: Consultancy; PriME Oncology: Honoraria; Karyopharm: Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy; Astellas: Research Funding; Roche: Research Funding; Novartis: Consultancy, Honoraria, Research Funding. Götze:Celgene: Research Funding. Schlenk:Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accomodations, Expenses, Research Funding, Speakers Bureau; Novartis: Speakers Bureau; Roche: Research Funding; AstraZeneca: Research Funding; PharmaMar: Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Döhner:Sunesis Pharmaceuticals: Research Funding; Abbvie: Consultancy; Agios: Consultancy; Amgen: Consultancy, Research Funding; Astellas Pharma: Consultancy; Bristol-Myers Squibb: Research Funding; Pfizer: Research Funding; Arog: Research Funding; Roche: Consultancy; Jazz Pharmaceuticals: Consultancy, Honoraria, Research Funding; Astex Pharmaceuticals: Consultancy; Janssen: Consultancy, Honoraria; Daiichi Sankyo: Honoraria; Celgene: Consultancy, Honoraria; Novartis: Honoraria, Research Funding. Salih:Synimmune: Consultancy, Research Funding; Philogen: Consultancy; Medigene: Consultancy; Novartis: Consultancy; Pfizer: Consultancy. Schittenhelm:Pfizer: Consultancy; Astellas: Consultancy. Mueller-Tidow:Jose-Carreras-Siftung: Research Funding; Wilhelm-Sander-Stiftung: Research Funding; BMBF: Research Funding; Deutsche Krebshilfe: Research Funding; Deutsche Forschungsgemeinschaft: Research Funding; Janssen-Cilag Gmbh: Membership on an entity's Board of Directors or advisory committees; BiolineRx: Research Funding; Daiichi Sankyo: Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer AG: Research Funding. Brugger:MorphoSys: Current Employment. Bug:Jazz: Honoraria; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel; Hexal: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; Eurocept: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: Travel; Sanofi: Other: Travel; Neovii: Other: Travel. Wäsch:Pfizer: Consultancy; Amgen: Consultancy; Janssen: Consultancy. Ganser:Celgene: Consultancy; Novartis: Consultancy. Döhner:AstraZeneca: Consultancy, Honoraria; Sunesis: Research Funding; Roche: Consultancy, Honoraria; Pfizer: Research Funding; Oxford Biomedicals: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; Helsinn: Consultancy, Honoraria; Jazz: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Bristol Myers Squibb: Consultancy, Honoraria, Research Funding; Astex: Consultancy, Honoraria; Astellas: Consultancy, Honoraria, Research Funding; AROG: Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Agios: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria; GEMoaB: Consultancy, Honoraria. OffLabel Disclosure: ATRA is approved for APL treatment but not for non-APL AML
- Published
- 2020
8. MicroRNA-155–deficient dendritic cells cause less severe GVHD through reduced migration and defective inflammasome activation
- Author
-
Annette Schmitt-Graeff, Robert Zeiser, Joseena Iype, Gabriele Prinz, Jürgen Finke, Benjamin A. H. Smith, Alessandro Prestipino, Dietmar Pfeifer, Sophia Chen, Yvonne Beck, Marco Idzko, Justus Duyster, and Sebastian Grundmann
- Subjects
Male ,Inflammasomes ,Immunology ,Graft vs Host Disease ,Allopurinol ,chemical and pharmacologic phenomena ,Disease ,Severity of Illness Index ,Biochemistry ,Mice ,Cell Movement ,immune system diseases ,hemic and lymphatic diseases ,microRNA ,medicine ,Animals ,Transplantation, Homologous ,Cells, Cultured ,Mice, Knockout ,Mice, Inbred BALB C ,Innate immune system ,business.industry ,Hematopoietic Stem Cell Transplantation ,Inflammasome ,Dendritic Cells ,Cell Biology ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Transplantation ,MicroRNAs ,surgical procedures, operative ,Graft-versus-host disease ,Female ,business ,Function (biology) ,medicine.drug - Abstract
The successful treatment of acute leukemias with allogeneic hematopoietic cell transplantation (allo-HCT) is limited by acute graft-versus-host disease (GVHD). Because microRNA-155 (miR-155) regulates activation of the innate immune system, we aimed to determine its function in dendritic cells (DCs) during GVHD in an experimental model. We observed that miR-155 deficiency of the recipient led to improved survival, reduced serum levels of proinflammatory cytokines, and lower GVHD histopathology scores. In addition, miR-155(-/-) bone marrow chimeric mice receiving allo-HCT and miR-155(-/-) DCs showed that miR-155 deficiency in the DC compartment was responsible for protection from GVHD. Activated miR-155(-/-) DCs displayed lower expression of various purinergic receptors and impaired migration toward adenosine triphosphate (ATP). Microarray analysis of lipopolysaccharide/ATP-stimulated miR-155(-/-) DCs revealed mitogen-activated protein kinase pathway dysregulation and reduced inflammasome-associated gene expression. Consistent with this gene expression data, we observed reduced ERK activation, caspase-1 cleavage, and IL-1β production in miR-155(-/-) DCs. The connection between miR-155 and inflammasome activation was supported by the fact that Nlrp3/miR-155 double-knockout allo-HCT recipient mice had no increased protection from GVHD compared with Nlrp3(-/-) recipients. This study indicates that during GVHD, miR-155 promotes DC migration toward sites of ATP release accompanied by inflammasome activation. Inhibiting proinflammatory miR-155 by antagomir treatment could help reduce this complication of allo-HCT.
- Published
- 2015
9. MiR-146a regulates the TRAF6/TNF-axis in donor T cells during GVHD
- Author
-
Robert Thimme, Gabriele Prinz, Natalie Stickel, Justus Duyster, Marie Follo, Peter Hasselblatt, Jürgen Finke, Annette Schmitt-Graeff, Robert Zeiser, Ulrich Salzer, and Dietmar Pfeifer
- Subjects
T-Lymphocytes ,Immunology ,Graft vs Host Disease ,Single-nucleotide polymorphism ,Stimulation ,Polymorphism, Single Nucleotide ,Biochemistry ,Mice ,Downregulation and upregulation ,Transcription (biology) ,Genotype ,Animals ,Humans ,Medicine ,TNF Receptor-Associated Factor 6 ,Mice, Inbred BALB C ,Tumor Necrosis Factor-alpha ,business.industry ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Transplantation ,MicroRNAs ,surgical procedures, operative ,Graft-versus-host disease ,Tumor necrosis factor alpha ,business ,Gene Deletion - Abstract
Acute graft-versus-host disease (GVHD) limits the success of allogeneic hematopoietic cell transplantation (allo-HCT); therefore, a better understanding of its biology may improve therapeutic options. We observed miR-146a up-regulation in T cells of mice developing acute GVHD compared with untreated mice. Transplanting miR-146a(-/-) T cells caused increased GVHD severity, elevated tumor necrosis factor (TNF) serum levels, and reduced survival. TNF receptor-associated factor 6 (TRAF6), a verified target of miR-146a, was up-regulated in miR-146a(-/-) T cells following alloantigen stimulation. Higher TRAF6 levels translated into increased nuclear factor-κB activity and TNF production in miR-146a(-/-) T cells. Conversely, the detrimental effect of miR-146a deficiency in T cells was antagonized by TNF blockade, whereas phytochemical induction of miR-146a or its overexpression using a miR-146a mimic reduced GVHD severity. In humans, the minor genotype of the single nucleotide polymorphism rs2910164 in HCT donors, which reduces expression of miR-146a, was associated with severe acute GVHD (grade III/IV). We show that miR-146a functions as a negative regulator of donor T cells in GVHD by targeting TRAF6, leading to reduced TNF transcription. Because miR-146a expression can be exogenously enhanced, our results provide a novel targeted molecular approach to mitigate GVHD.
- Published
- 2014
10. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease
- Author
-
Jürgen Finke, Tony A. Mueller, Sophia Chen, Mareike Verbeek, Julius C. Fischer, Michael Bscheider, Annette Schmitt-Graeff, Nikolas von Bubnoff, Hendrik Poeck, Christian Peschel, Vera Otten, Silvia Spoerl, Martina Schmickl, Justus Duyster, Nimitha R. Mathew, Kristina Maas-Bauer, and Robert Zeiser
- Subjects
Male ,Ruxolitinib ,medicine.medical_treatment ,Immunology ,Drug Evaluation, Preclinical ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Severity of Illness Index ,Biochemistry ,Proinflammatory cytokine ,Mice ,immune system diseases ,Nitriles ,medicine ,Animals ,Humans ,Transplantation, Homologous ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Cells, Cultured ,Mice, Inbred BALB C ,Janus kinase 2 ,Janus kinase 1 ,biology ,business.industry ,Hematopoietic Stem Cell Transplantation ,FOXP3 ,Cell Biology ,Hematology ,Janus Kinase 2 ,medicine.disease ,Mice, Inbred C57BL ,Transplantation ,Disease Models, Animal ,Pyrimidines ,Treatment Outcome ,surgical procedures, operative ,Graft-versus-host disease ,Cytokine ,Blood Group Incompatibility ,biology.protein ,Pyrazoles ,Female ,business ,medicine.drug - Abstract
Graft-versus-host-disease (GVHD) is a severe complication of allogeneic hematopoietic cell transplantation (allo-HCT) characterized by the production of high levels of proinflammatory cytokines. Activated Janus kinases (JAKs) are required for T-effector cell responses in different inflammatory diseases, and their blockade could potently reduce acute GVHD. We observed that inhibition of JAK1/2 signaling resulted in reduced proliferation of effector T cells and suppression of proinflammatory cytokine production in response to alloantigen in mice. In vivo JAK 1/2 inhibition improved survival of mice developing acute GVHD and reduced histopathological GVHD grading, serum levels of proinflammatory cytokines, and expansion of alloreactive luc-transgenic T cells. Mechanistically, we could show that ruxolitinib impaired differentiation of CD4(+) T cells into IFN-γ- and IL17A-producing cells, and that both T-cell phenotypes are linked to GVHD. Conversely, ruxolitinib treatment in allo-HCT recipients increased FoxP3(+) regulatory T cells, which are linked to immunologic tolerance. Based on these results, we treated 6 patients with steroid-refractory GVHD with ruxolitinib. All patients responded with respect to clinical GVHD symptoms and serum levels of proinflammatory cytokines. In summary, ruxolitinib represents a novel targeted approach in GVHD by suppression of proinflammatory signaling that mediates tissue damage and by promotion of tolerogenic Treg cells.
- Published
- 2014
11. Azacytidine Sensitizes AML Cells for Effective Elimination By CD123 CAR T-Cells
- Author
-
Jade Clarson, Deborah L. White, Nadia El Khawanky, Agnes S. M. Yong, Sanaz Taromi, Robert Zeiser, Angel F. Lopez, Amy Hughes, Timothy P. Hughes, Justus Duyster, Michael P. Brown, and Wenbo Yu
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Azacitidine ,Biochemistry ,CD19 ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,Medicine ,biology ,business.industry ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Interleukin-3 receptor ,Bone marrow ,business ,CD8 ,030215 immunology ,medicine.drug - Abstract
Chimeric antigen receptor T-cells (CAR Tc) have yielded impressive remission rates in treatment-refractory B-cell malignancies (B-ALL and B-lymphomas) by targeting CD19, resulting in the first FDA approved CAR Tc therapies, Kymriah and Yescarta. However, the translation of these results for other cancer entities remains a challenge. Pre-clinical studies using second-generation CAR Tc against the interleukin-3 receptor alpha chain (CD123) engendered strong anti-leukemic activity. CD123 CAR Tc clinical studies resulted in transient responses, or complete remission but at the expense of on-target off-tumor toxicities. Our studies employing third-generation anti-CD123 CAR Tc demonstrate strong anti-leukemic activity with no adverse effects in vivo. However, the leukemia was not completely eradicated. Combining anti-CD123 CAR Tc with DNA hypomethylating (HMA) agents may enhance the anti-leukemic effect and survival. HMAs such as azacytidine (Aza) activate key epigenetically silenced pathways in AML cells, inhibiting cell proliferation while enhancing cell immunogenicity. We hypothesized that Aza will increase the expression of CD123 on AML cells resulting in long-term disease eradication by anti-CD123 CAR Tc. The anti-leukemic efficacy, survival advantage, safety and feasibility of the combination treatment with Aza and anti-CD123 CAR Tc were evaluated in vivo. HL-60 (CD123med), MLL-2 (CD123lo), MOLM-13 (CD123hi), primary de novo and relapsed/refractory (r/r) AML cells were cultured for 0-8 days in the presence of Aza (0µM-5µM) and analysed for their CD123 expression by flow cytometry, quantitative western blot and RNAseq. The anti-CD123 CAR was constructed with the humanized CSL362-based ScFv and the CD28-OX40-CD3ζ signaling domain, encoded in a third-generation lentiviral vector and expressed in CD3+ Tc from healthy donors. Rag2γc-/- mice (n=12-16/ group) were engrafted with 1x105 MOLM13/ffLuc AML cells and treated with PBS, 5x106 Non-transduced (NTD) Tc orCAR Tc, 4x 2.5mg/kg Aza, or 5x106 CAR Tc following 4x Aza (2.5mg/kg). Leukemic burden was assessed weekly by bioluminescence imaging. Tc activity and immunophenotyping was performed using flow cytometry at day 35 post engraftment, and survival was monitored. HL-60, MLL-2 and MOLM-13 cells showed significant increases in HLA-DR, PD-L1, STAT1 and IRF7 expression, as well as CD123 when exposed to Aza (Fig 1A,B). Interestingly, the increased effect was seen from day one regardless of concentration. This was similarly reflected in AML patient cells. Aza treatment also arrested cell proliferation and decreased viability in both cell lines and patient cells suggesting Aza can aid in the anti-leukemic effect. Rag2γc-/- mice engrafted with MOLM-13 and treated with Aza and CD123 CAR Tc demonstrated suppressed growth, and eradication of MOLM-13 cells compared to mice treated with CD123 CAR Tc or Aza alone. Additionally, a significant decrease in residual CD123+ cells in the bone marrow (BM) of dual treated mice was seen (Fig 1C). A higher frequency of residual CD8+ T-cells in the BM, and CD4+ Tc in the peripheral blood (PB) and BM of dual treated mice was observed compared to CAR Tc only treated mice. Most prominently, we found a significantly higher mean number of stem cell-like and central memory CD8+ Tc in the BM of dual treated mice (232 cells/µl and 208cells/µl, respectively) compared to the CAR Tc only group (55 cells/µl and 23 cells/µl, respectively). Assessment of immune checkpoint markers on residual CAR Tc of dual treated mice revealed significantly decreased levels of CTLA-4, PD-1 and TIM-3 in the BM, and CTLA-4 in the PB compared to the CAR Tc only group. While CAR Tc treatment alone demonstrated a survival advantage compared to PBS, NTD or Aza treated mice, Aza and CAR Tc treatment had a significantly higher survival rate compared to the CAR Tc only group (92% vs. 46% at day 50, p Our findings indicate that Aza increases immunogenicity and augments the cell surface expression of CD123 on AML cells, allowing enhanced recognition and elimination of malignant cells by CD123 CAR Tc. This is the first demonstration that HMAs and CAR Tc immunotherapy can be used synergistically to treat AML. Considering HMAs are currently under clinical investigation in AML, our data encourage further clinical evaluation of this dual treatment in r/r AML, including high-risk patients that are chemotherapy or allogeneic transplantation ineligible. Disclosures Hughes: Novartis, Bristol-Myers Squibb, Celgene: Research Funding; Novartis, Bristol-Myers Squibb: Consultancy, Other: Travel. White:BMS: Honoraria, Research Funding; AMGEN: Honoraria, Speakers Bureau. Yong:Novartis: Honoraria, Research Funding; Celgene: Research Funding; BMS: Honoraria, Research Funding.
- Published
- 2019
12. A Novel 10-Fluorochrome Multiparameter Flow Cytometry (MFC) Panel for Precise Plasma Cell (PC) and MRD Assessment in Clinical Routine of Multiple Myeloma (MM) Patients and throughout the Disease Course
- Author
-
Stefan J. Mueller, Ralph Wäsch, Cornelius Miething, Sandra M. Woerner, Dagmar Wider, Monika Engelhardt, Mihaela Zlei, Justus Duyster, Marie Follo, and Veronika Riebl
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Leukapheresis ,Plasma cell ,medicine.disease ,Biochemistry ,Transplantation ,Clinical trial ,medicine.anatomical_structure ,EuroFlow ,Internal medicine ,medicine ,business ,Multiple myeloma ,Monoclonal gammopathy of undetermined significance - Abstract
Introduction: Since the introduction of novel agents into the treatment of Multiple Myeloma (MM), much deeper remissions can be achieved. This leads to a demand for disease evaluation tools with higher sensitivity and clinical practicability. In various clinical trials the assessment of the MRD status has been proven to have strongest prognostic value for MM patients (pts). The Euroflow Consortium has pioneered a highly standardized next generation flow (NGF) approach with the establishment of two 8-color tubes tests (Flores-Montero 2017). However, due to cost-intensive and time-consuming preconditions, this MRD approach might not be feasible in every center. Thus, other readily available, validated MRD tools may also guarantee highly sensitive MRD evaluation which allows for early relapse detection and to steer treatment, such as decision upon need for consolidation, duration of maintenance, therapy pauses or even earlier FDA/EMA approval of novel antimyeloma agents. Methods: Our 10 color single tube MFC panel consisting of the antigens CD38, CD138, CD19, CD45, CD27, CD56, CD28, CD81, CD117 and CD200, was systematically validated using Fluorescence-minus-one controls, consecutive spike-in-controls, consistency analyses and measurement of 9 MM cell lines. Thereafter, we assessed 131 Bone Marrow (BM), peripheral blood (PB), and leukapheresis (LA) samples from 97 different MM, MGUS and SMM pts and 13 healthy individuals (HI) as controls. Samples were processed within 6-20 hours of aspiration and a bulk lysis was performed. Subsequently, >3x106 viable nucleated cells were acquired on a BD LRSFortessa Cytometer. Data analysis was performed using the BeckmanCoulter software Kaluza. BM was sampled at initial diagnosis (ID) or at progression (PD), after standard therapy (ST) or stem cell transplantation (SCT). The study was performed with written consent from all pts and approval from the ethics committee. Results: Our MFC approach confirmed the antigen expression in all MM cell lines (RPMI 8226; U266; IM-9; MM1.S; MM1.R; L363; Karpas620; NCI-H929; OPM-2) as previously reported. The spike-in-controls determined the limit of detection (LOD) at 10-5. This LOD was achieved in 89% of our MRD samples. An easy-to-adapt gating strategy to identify abnormal plasma cells (aPC) vs. normal plasma cells (nPC) was also established in our MM pt samples. In this cohort, we identified 10 distinct aPC and nPC subpopulations that differed in their expression pattern, suggesting clonal subtypes of MM. Moreover, in 19% of BM samples, two malignant subpopulations coexisting in the same pt, were identified. Since our BM cohort consisted of 46% samples at ID or PD vs. 54% posttreatment samples after ST and SCT, clonality variation in the former, differences of aPCs vs. nPCs ratio and to the latter group could be determined. Moreover, significant differences of much higher percentage of aPC in symptomatic MM vs SMM/MGUS pts were readily verified with our panel (p=0.0087). With a median time of 51 days from treatment to MRD evaluation, 24% of post treatment samples were MRD negative (MRD-) at the 10-5 level ( Since we also assessed pre- and post-treatment samples, antibody expression levels on PC from ID to post treatment demonstrated a significant normalization in Median Fluorescence Intensity (MFI) of 5 antibodies (CD56, CD81, CD45, CD200, CD27). These may thus serve as informative markers for the MRD assessment of these samples. Conversely, with progression from remission to PD, CD81 and CD45 antibodies showed significant decreases (p=0.0007 and p=0.0026, respectively) in expression, which suggest that both may serve as early relapse markers. Of the assessed LA samples at harvest before ASCT, 44% were MRD-, warranting future investigations into possible implications for pts' prognosis, treatment and PFS/OS data. Conclusion: Here we present a readily available, cost-effective, quick and highly validated MFC panel with an easy to adapt gating strategy that allows for precise aPC vs. nPC assessment in- and outside clinical trials. Our 10 color MFC panel is applicable in BM samples of MM pts and precursor diseases, as well as in LA samples. With the additional MFC information, ideally at ID and repeatedly after treatment, readily available individual treatment decisions seem possible to obtain for every MM patient. Disclosures Wäsch: Amgen: Other: travel, Research Funding; Sanofi: Other: Travel, Research Funding; Jazz: Other: travel, Research Funding; Celgene: Other: travel, Research Funding; Gilead: Other: travel, Research Funding; Takeda: Consultancy; Gilead: Consultancy; Sanofi: Consultancy; Amgen: Consultancy; Novartis: Consultancy; Pfizer: Consultancy.
- Published
- 2019
13. Vaccination Against Poly-N-Acetylglucosamine Decreases Neutrophil Activation and Gvhd While Maintaining Microbial Diversity
- Author
-
Eileen Haring, Ozlem Oyardi, Sandra Duquesne, Burkhard Becher, Gerald B. Pier, Jan Hülsdünker, Georg Häcker, Oliver Pabst, Robert Zeiser, Sonia Tugues, Colette Cywes-Bentley, Oliver S. Thomas, Nicolás Gonzalo Núñez, Schmitt-Graeff, Susanne Unger, Christian Koenecke, Martin J. Blaser, Justus Duyster, Susanne Kirschnek, Zhan Ghao, and Petya Apostolova
- Subjects
biology ,medicine.drug_class ,Chemistry ,Immunology ,Antibiotics ,Inflammation ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Microbiology ,Acetylglucosamine ,Vaccination ,Graft-versus-host disease ,Immunization ,Intestinal mucosa ,biology.protein ,medicine ,Antibody ,medicine.symptom - Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a curative therapy for different malignant and non-malignant hematologic diseases. Major life-threatening complications after allo-HCT are acute graft-versus-host disease (aGVHD) and severe infections. The incidence of GVHD after allo-HCT remains high, despite prophylactic immunosuppressive medication. According to the CIBMTR database, 60% of patients undergoing allo-HCT develop at least grade II-IV aGVHD. Microbial invasion into the intestinal mucosa after allo-HCT triggers the activation of neutrophil granulocytes (neutrophils) and requires antibiotic treatment to prevent sepsis. However, antibiotics lead to a loss of microbial diversity, which is connected to a higher incidence of aGVHD. Anti-microbial therapies which eliminate invading bacteria and diminish neutrophil-mediated damage without reducing the diversity of the microbiota are therefore highly desirable. A potential solution for this could be the use of anti-microbial antibodies that target and mark invading pathogens resulting in their elimination by innate immune cells. Therefore we investigated the potency of both active and passive immunization against the conserved microbial surface polysaccharide Poly-N-acetylglucosamine (PNAG) which is expressed on various pathogens in a murine aGVHD model. We could detect reduced aGVHD related mortality in mice which underwent treatment with an antibody against PNAG (anti-PNAG) or were actively immunized against PNAG. Sequencing analysis of the microbiota before and after allo-HCT showed that anti-PNAG treatment did not have any effect luminal microbial diversity. Mechanistically, we could show that anti-PNAG antibody treatment caused more abundant neutrophil recruitment to the intestinal submucosa. This supports the concept that neutrophils are able to effectively eliminate any opsonized bacteria in this tissue, leading to reduced local inflammation. In agreement with this we observed lower intestinal inflammation and reduced myeloperoxidase activity as well as proliferation of neutrophils in the small intestine of mice treated with anti-PNAG antibody. We demonstrate the potency of targeting PNAG as a novel antimicrobial treatment option in aGVHD by reducing uncontrolled neutrophil activation and thereby interfering with aGVHD without affecting commensal intestinal microbial diversity. Anti-PNAG-antibodies enhance anti-microbial immunity, while reducing the activation of neutrophil-mediated downstream immunopathology which is a novel approach to reduce the severity of GVHD. This could be translated into a clinical application given the modest toxicity profile of vaccination and the availability of fully human anti-PNAG antibodies, both of which have been tested in phase 1 trials in humans (ClinicalTrials.gov Identifier: NCT02853617). Disclosures Cywes-Bentley: OneBiopharma, Inc.: Honoraria. Koenecke:Novartis: Other: none. Pier:OneBiopharma, Inc.: Equity Ownership, Honoraria; Alopexx Vaccine, LLC: Equity Ownership, Honoraria.
- Published
- 2019
14. Prognostic Factors for Survival after Allogeneic Transplantation in Acute Lymphoblastic Leukemia
- Author
-
Hartmut Bertz, Reinhard Marks, Monika Engelhardt, Christine Greil, Jürgen Finke, Ralph Wäsch, Justus Duyster, Robert Zeiser, and Gabriele Ihorst
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Total body irradiation ,medicine.disease ,Biochemistry ,Minimal residual disease ,Transplantation ,Median follow-up ,Acute lymphocytic leukemia ,Internal medicine ,Cohort ,medicine ,Cumulative incidence ,Progression-free survival ,business - Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous disease and treatment guidelines are still evolving. Allogeneic stem cell transplantation (allo-SCT) offers a potentially curative option and is recommended in first relapse and for high-risk patients in first complete remission (CR). Survival after allo-SCT could be substantially improved due to better risk stratification, patient selection and adapted treatment protocols leading to reduced non-relapse mortality (NRM). Prognostic factors for survival after allo-SCT still need to be defined: pheno- and genotype, patients´ age, conditioning regimens and remission status prior to allo-SCT are under discussion. We analyzed the outcome of 180 consecutive ALL patients who received allo-SCT at the Freiburg University Medical Center between 1995 and 2018 with regard to treatment response, survival, adverse reactions, and performed subgroup analyses to identify prognostic factors. The median age in our cohort was 37 years (ys), 19% were older than 55 ys. 27% were diagnosed with Philadelphia (Ph)-positive ALL, 24% with T-ALL. 36% were treated with relapsed/refractory disease. 48% of allo-SCTs were conducted with a HLA-matched, 19% with a HLA-mismatched unrelated and 33% with a related donor. In 61% the conditioning regimen included total body irradiation (TBI). In 48% no minimal residual disease (MRD) was detected prior to allo-SCT, 20% were transplanted in MRD-positive CR. The overall response rate was 86%, with MRD-negativity in 78%. With a median follow up of 10 ys, we observed a median overall survival (OS) of 23 months and a median progression free survival (PFS) of 11 months. The 10ys-OS was 33%, the 10ys-PFS 31%. The cumulative incidence of relapse was 68% at 10 ys, the cumulative incidence of NRM 12%. Acute graft-versus-host disease (GvHD) III-IV° occurred in 17%, severe chronic GvHD in 9%. Survival was significantly better in patients reaching MRD-negative CR before allo-SCT (10ys-OS 48% vs. 19%, p With a very long follow-up and high rate of MRD-assessment, we observed a high response rate and a low rate of severe GvHD. Our data confirm that allo-SCT enables long-term survival in high-risk ALL, suggest that, in certain subgroups, survival may be best in patients transplanted in CR and receiving TBI for conditioning, including the relevant observation that allo-SCT can be performed in carefully selected elderly patients. Disclosures Finke: Medac: Honoraria, Other: research support, Speakers Bureau; Neovii: Honoraria, Other: research support, Speakers Bureau; Riemser: Honoraria, Other: research support, Speakers Bureau. Wäsch:Pfizer: Consultancy; Sanofi: Other: Travel, Research Funding; Celgene: Other: travel, Research Funding; Jazz: Other: travel, Research Funding; Amgen: Consultancy; Gilead: Consultancy; Sanofi: Consultancy; Novartis: Consultancy; Takeda: Consultancy; Amgen: Other: travel, Research Funding; Gilead: Other: travel, Research Funding.
- Published
- 2019
15. The Fanconi Anemia-Associated Protein NIPA Is Essential for the Nuclear Abundance of FANCD2
- Author
-
Marie Follo, Melanie Boerries, Melissa Zwick, Justus Duyster, Stefanie Kreutmair, Annette Schmitt-Graeff, Anna Lena Illert, Milena Pantic, Cathrin Klingeberg, Irith Baumann, Bernhard Kuster, Hiroyuki Kawaguchi, Detlev Schindler, Khalid Shoumariyeh, Michal Kulinski, Geoffroy Andrieux, Nina Cabezas-Wallscheid, Dietmar Pfeifer, Rouzanna Istvanffy, Robert Zeiser, Martina Rudelius, Alina Rudorf, Miriam Erlacher, Christian Peschel, Michael D. Cleary, Tony Andreas Müller, Christine Dierks, Robert A.J. Oostendorp, Jesus Duque-Afonso, Teresa Poggio, Simone Lemeer, Charlotte M. Niemeyer, Marcin W. Wlodarski, and Tamina Rückert
- Subjects
Myeloid ,DNA repair ,Immunology ,Bone marrow failure ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Fanconi anemia ,hemic and lymphatic diseases ,FANCD2 ,medicine ,Cancer research ,Stem cell - Abstract
Inherited bone marrow failure syndromes (IBMFS) are a heterogeneous group of disorders characterized by impaired stem cell function resulting in pancytopenia. Diagnosis of IBMFS presents a major challenge due to limited diagnostic tests and overlapping phenotypes. For that reason, novel and clinical relevant biomarkers and possible targets are urgently needed. Our study defines NIPA as an IBMFS gene, which is significantly downregulated in a distinct subset of MDS-type refractory cytopenia of childhood patients. Mechanistically, NIPA binds FANCD2 and regulates its nuclear abundance. The stabilization of both non- and monoubiquitinated FANCD2 identifies NIPA as an essential player in the Fanconi Anemia (FA) pathway. NIPA thereby prevents MMC hypersensitivity visualized by increased numbers of chromosome radials and reduced cell survival after induction of interstrand crosslinks. To provide proof of principle, re-expression of FANCD2 in Nipa deficient cells restores MMC sensitivity. In a knockout mouse model, Nipa deficiency leads to major cell intrinsic long-term repopulation defects of hematopoietic stem cells (HSCs), with impaired self-renewal in serial transplantations and a bias towards myeloid differentiation. Unresolved DNA damage in Nipa deficient HSCs causes increased sensitivity to cell death and leads to progressive, age-related loss of the HSC pool. Induction of replication stress triggers the phenotypic reduction and functional decline of murine HSCs, resulting in complete bone marrow failure and death of the mice thereby mimicing Fanconi Anemia. Taken together, our study adds NIPA to the short list of FA-associated proteins being essential for a functional DNA repair/FA/BRCA axis and thereby emphasizing its impact as potential diagnostic marker and/or possible target in bone marrow failure syndromes. Disclosures Niemeyer: Celgene: Consultancy.
- Published
- 2019
16. Examining the Role of CD30 in an Anaplastic Large Cell Lymphoma Mouse Model
- Author
-
Melanie Boerries, Geoffroy Andrieux, Dietmar Pfeifer, Robert Zeiser, Stefanie Kreutmair, Teresa Poggio, Anna Lena Illert, Marie Follo, Linda Graessel, Justus Duyster, Cornelius Miething, and Suzanne D. Turner
- Subjects
integumentary system ,CD30 ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,medicine.disease ,Biochemistry ,Lymphoma ,Lymphatic system ,immune system diseases ,hemic and lymphatic diseases ,Cancer research ,medicine ,Anaplastic lymphoma kinase ,T-cell lymphoma ,Kinase activity ,Anaplastic large-cell lymphoma - Abstract
Anaplastic large cell lymphoma (ALCL) represents a heterogeneous group of T-cell non-Hodgkin lymphoma (NHL) mainly affecting children and young adults. About 70% of systemic ALCLs are associated with a characteristic chromosomal translocation, t(2;5)(p23;35) which fuses the anaplastic lymphoma kinase (ALK) gene on chromosome 2 with the nucleophosmin (NPM) gene on chromosome 5, resulting in the NPM-ALK fusion gene, its over-expression and constitutive kinase activity. Immunophenotypic characterization of human ALCL revealed highly CD30-positive cells of T- or Null-Cell-origin and resulted in promising clinical trials with CD30-coupled antibodies. However, the impact of CD30 on disease development remains unclear and the relationship between NPM-ALK and CD30 has been investigated mostly using cell line models. Syngeneic mouse models of cancer can serve as useful models since the tumors develop in situ where the contribution made by the immune system and the extracellular matrix can be investigated. Here, we focus on the involvement of CD30 in a retroviral murine bone marrow transplantation model of ALCL. In this model, the BM of Lck-Cre-transgenic mice is infected with a MSCV-Stop-NPM-ALK-IRES-EGFP vector leading to expression of NPM-ALK in early T-cells. With a latency of 3-4 months, mice develop lymphomas and die from neoplastic T-cell-infiltration of BM and lymphatic organs. To investigate the impact of abrogation of CD30 signals on the development of NPM-ALK+ ALCL in our model, CD30 knockout mice were crossed with Lck-Cre mice. Both Lck-Cre NPM-ALK CD30 wt and Lck-Cre NPM-ALK CD30 ko recipients develop a human ALCL-like lymphoma with a pure T-cell phenotype characterized by Thy1.2+ cells infiltrating the thymus, lymph nodes, spleen and BM. First results from Lck-Cre NPM-ALK CD30 ko transplanted mice showed impaired disease induction and prolonged survival compared to CD30 wt animals. Moreover, secondary transplantation of NPM-ALK thymic lymphomas led to distinct deceleration of disease development upon CD30 deletion. Microarray analyses have shed some light on the mechanisms underlying the delayed lymphoma progression of CD30 deleted tumors with an upregulation of inflammatory pathways and proteins that are master players in inflammation and immune responses. Further characterization of the role of CD30-mediated immune response in disease progression using this mouse model and immunocompromised mice is ongoing. An improved understanding of how the immune system affects tumor progression will extend the rationale in translational strategies to use immunotherapies for patients with T-NHLs. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
17. SRC is a signaling mediator in FLT3-ITD– but not in FLT3-TKD–positive AML
- Author
-
Christian Peschel, Rebekka Grundler, Lars Rönnstrand, Katharina Götze, Elena Razumovskaya, Karsten Spiekermann, Hannes Leischner, Justus Duyster, Stefan K. Bohlander, and Corinna Albers
- Subjects
Blotting, Western ,Immunology ,SH2 domain ,Biochemistry ,Receptor tyrosine kinase ,SH3 domain ,src Homology Domains ,Mice ,fluids and secretions ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Animals ,Humans ,Immunoprecipitation ,Phosphorylation ,RNA, Small Interfering ,Cells, Cultured ,Cell Proliferation ,Mice, Inbred C3H ,Tyrosine-protein kinase CSK ,biology ,Chemistry ,hemic and immune systems ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,Flow Cytometry ,Leukemia, Myeloid, Acute ,src-Family Kinases ,fms-Like Tyrosine Kinase 3 ,Tandem Repeat Sequences ,embryonic structures ,NIH 3T3 Cells ,biology.protein ,Cancer research ,Signal transduction ,Tyrosine kinase ,psychological phenomena and processes ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Mutations of Fms-like tyrosine kinase 3 (FLT3) are among the most frequently detected molecular abnormalities in AML patients. Internal tandem duplications (ITDs) are found in approximately 25% and point mutations within the second tyrosine kinase domain (TKD) in approximately 7% of AML patients. Patients carrying the FLT3-ITD but not the FLT3-TKD mutation have a significantly worse prognosis. Therefore, both FLT3 mutations seem to exert different biologic functions. FLT3-ITD but not FLT3-TKD has been shown to induce robust activation of the STAT5 signaling pathway. In the present study, we investigated the mechanisms leading to differential STAT5 activation and show that FLT3-ITD but not FLT3-TKD uses SRC to activate STAT5. Coimmunoprecipitation and pull-down experiments revealed an exclusive interaction between SRC but not other Src family kinases and FLT3-ITD, which is mediated by the SRC SH2 domain. We identified tyrosines 589 and 591 of FLT3-ITD to be essential for SRC binding and subsequent STAT5 activation. Using site-specific Abs, we found that both residues were significantly more strongly phosphorylated in FLT3-ITD compared with FLT3-TKD. SRC inhibition and knock-down blocked STAT5 activation and proliferation induced by FLT3-ITD but not by FLT3-TKD. We conclude that SRC might be a therapeutic target in FLT3-ITD+ AML.
- Published
- 2012
18. An RNAi-based system for loss-of-function analysis identifies Raf1 as a crucial mediator of BCR-ABL–driven leukemogenesis
- Author
-
Hannes Leischner, Justus Duyster, Anna Lena Illert, Christian Peschel, Cornelius Miething, Corinna Albers, and Melanie Thiede
- Subjects
Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,Genetic Vectors ,Immunology ,Gene Expression ,Genes, abl ,Biology ,Biochemistry ,Viral vector ,Small hairpin RNA ,Mice ,RNA interference ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Gene Knockdown Techniques ,Animals ,DNA Primers ,Mice, Inbred BALB C ,Gene knockdown ,ABL ,Base Sequence ,Oncogene ,Cell Biology ,Hematology ,Proto-Oncogene Proteins c-raf ,Transplantation ,Cell Transformation, Neoplastic ,Cancer research ,Female ,RNA Interference - Abstract
Genetic loss-of-function studies in murine tumor models have been essential in the analysis of downstream mediators of oncogenic transformation. Unfortunately, these studies are frequently limited by the availability of genetically modified mouse strains. Here we describe a versatile method allowing the efficient expression of an oncogene and simultaneous knockdown of targets of interest (TOI) from a single retroviral vector. Both oncogene and TOI-specific miR30-based shRNA are under the control of the strong viral long terminal repeat promoter, resulting in a single shared RNA transcript. Using this vector in a murine syngeneic BM transplantation model for BCR-ABL–induced chronic myeloid leukemia, we find that oncogene expression and target knockdown in primary hematopoietic cells with this vector is efficient both in vitro and in vivo, and demonstrate that Raf1, but not BRAF, modulates BCR-ABL–dependent ERK activation and transformation of hematopoietic cells. This expression system could facilitate genetic loss-of-function studies and allow the rapid validation of potential drug targets in a broad range of oncogene-driven murine tumor models.
- Published
- 2011
19. The Pan-PIM Kinase Inhibitor LGB321 Strongly Increases Ibrutinib Effects in CLL, By Blocking Anti-Apoptotic Pathways and Microenvironmental Interactions
- Author
-
Sarah Decker, Shifa Saleem, Anabel Zwick, Justus Duyster, Sandra Kissel, Katja Zirlik, Konrad Aumann, and Christine Dierks
- Subjects
0301 basic medicine ,Chemistry ,Kinase ,Blocking (radio) ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Apoptosis ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Ibrutinib ,Cancer research - Abstract
Despite remarkable anti-tumor activity of ibrutinib in CLL, some patients develop resistance due to acquired mutations. By adding drugs which target survival pathways and the CLL protective microenvironmental interactions, we aim to increase ibrutinib efficacy and to prevent ibrutinib resistance. The three PIM kinases are involved in important disease mechanisms in CLL, with PIM1 regulating CXCR4 surface expression impacting the interaction with the protective microenvironment, and PIM2/3 affecting the apoptotic machinery by regulating BAD. Here, we investigated the effect of the Pan-PIM kinase inhibitor LGB321 in CLL in vitro and in vivo and its cooperative effect with ibrutinib. LGB321 was highly effective in inducing apoptosis in primary CLL cells, independent of prognostic factors or microenvironmental interactions, and reduced pBAD and total BAD protein levels in CLL. In vitro combination of ibrutinib and LGB321 demonstrated strong additive effects with doubled apoptosis rates compared to single treatment. Further, LGB321 treatment blocked the CXCR4/CXCL12 axis by dephosphorylating the CXCR4 receptor on Ser339, by reducing total CXCR4 protein levels, and by blocking the externalization of the CXCR4 receptor. Concordantly, LGB321 blocked CXCR4 functions regarding migration towards a CXCL12 gradient and homing of LGB321-pretreated primary CLL cells towards the bone marrow of NOG mice. In vivo experiments confirmed the efficacy of LGB321 in 5 different CLL xenograft studies. Transplantation of primary CLL cells into NOG mice and treatment with LGB321 for 2 weeks strongly reduced WBC counts, spleen size and spleen infiltration with human CLL cells in all 5 CLL cases, and blocked BAD as well as CXCR4 phosphorylation also in vivo. Xenograft studies combining ibrutinib with LGB321 demonstrated much faster reduction of WBC counts than ibrutinib single treatment and strongly reduced disease burden in the double treated mice, confirming the cooperative effects of those two inhibitors also in vivo. Our results demonstrate that LGB321 might be an effective treatment option for CLL patients by impairing PIM2/3 mediated CLL-cell survival, and by blocking the PIM1/CXCR4-mediated interaction of CLL cells with the protective microenvironment. Furthermore, LGB321 enhances ibrutinib treatment effects and might help to increase its efficacy and to avoid the development of ibrutinib resistance. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
20. Towards Improved Models of NPM-ALK Induced T-Cell Lymphoma
- Author
-
Robin Khan, Cathrin Klingeberg, Justus Duyster, Cornelius Miething, Khalid Shoumariyeh, Sophia Ehrenfeld, Stefanie Kreutmair, Anna Lena Illert, Desiree Melanie Redhaber, and Teresa Poggio
- Subjects
Chemistry ,Immunology ,Ki-1+ Anaplastic Large Cell Lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Transplantation ,Blood cell depletion therapy ,Antigen ,Cell culture ,hemic and lymphatic diseases ,medicine ,Cancer research ,T-cell lymphoma ,Anaplastic lymphoma kinase ,Interleukin 3 - Abstract
Time- and tissue-specific expression of transgenes is essential for the accurate representation of human disease in in vivo models. To improve flexibility but also fidelity of ALK+ ALCL models, we developed new approaches enabling lineage specific expression of NPM-ALK cDNAs, as well as a novel system for CRISPR/Cas induced Npm-Alk recombination. First, we have designed a new system for lineage-restricted expression of transgenes based on a retroviral vector incorporating a translational stop-cassette flanked by loxP recombination sites. Conditional transgene expression in chimeric mice is rapidly achieved through retroviral infection and subsequent transplantation of hematopoietic stem cells (HSC) derived from transgenic mice expressing Cre-recombinase from a lineage specific promoter. To validate the model, we directed expression of NPM-ALK, the fusion oncogene present in anaplastic large cell lymphoma (ALCL), to T-cells by infecting hematopoietic stem cells from Lck-Cre transgenic mice with a retroviral construct containing the Npm-Alk cDNA preceded by a translational stop cassette. The approach efficiently induced T-cell lymphomas within 12-16 weeks closely resembling the human disease including the expression of the ALCL hallmark antigen CD30. Since NPM-ALK overexpressed from a cDNA acts as a very strong oncogene transforming a range of cell types, we were interested to develop a more physiologic model based on chromosomal recombination, enabling NPM-ALK expression from the endogenous Npm promoter. To achieve this, we have designed guide RNAs (gRNAs) directed to either the intron between exon 4 and 5 for Npm1 on mouse chr. 11, or the intron between exons 19 and 20 for Alk on mouse chr. 17., enabling targeted translocation between the two chromosomes. For further analysis, the IL-3 dependent murine pro-B cell line Ba/F3 stably expressing the Cas9 recombinase was transduced with the respective gRNAs and subsequently grown in the absence of IL-3 to allow positive selection of cells transformed by productive t(11;17) NA recombination. A PCR reaction on genomic DNA using primers covering the translocation breakpoint resulted in a product and Sanger sequencing of the amplicon confirmed the intended recombination at the targeted genomic positions. The translocation was also detectable by fluorescence in-situ hybridization (FISH), and Western blot analysis demonstrated expression of a highly phosphorylated Npm-Alk fusion protein. To further probe for oncogene dependency, we treated Npm-Alk translocated cells and control cells with a specific Alk inhibitor, resulting in rapid cell depletion of the Npm-Alk translocated cells, but not controls. The described Cre/loxP-based system represents a versatile tool for the rapid functional analysis of gene function in a defined lineage or in a developmental stage in vivo, and faithfully recapitulates many features of ALCL. Furthermore, using Crispr/Cas to induce targeted double-strand breaks, we have been able to generate specific Npm-Alk translocations in murine cells, paving the way for novel models which may help to further define the initial pathogenetic event underlying lymphomagenesis in ALCL. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
21. Metabolic Reprogramming Overcomes T Cell Inhibition By AML Cells
- Author
-
Jörg Büscher, Sophia Chen, Robert Zeiser, Michael D. Buck, Khalid Shoumariyeh, David O’Sullivan, Jürgen Finke, Melanie Börries, Franziska M. Uhl, Gesa Richter, Dietmar Pfeifer, Hartmut Bertz, Tobias Madl, Erika L. Pearce, and Justus Duyster
- Subjects
business.industry ,Lymphocyte ,medicine.medical_treatment ,T cell ,Immunology ,Priming (immunology) ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,Leukemia ,medicine.anatomical_structure ,medicine ,Cancer research ,Cytotoxic T cell ,business ,CD8 - Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is the treatment strategy of choice for the majority of patients with acute myeloid leukemia (AML). However, about 40% of patients relapse, which is associated with a poor prognosis. Donor lymphocyte infusions (DLIs) induce remission only in a minority of relapsed AML patients. In order to understand the impact of AML cells on T cell bioenergetics, we investigated the metabolic phenotype and immunological profile of CD8+ T cells isolated from AML patients at primary diagnosis, during remission and relapse after allo-HCT. At diagnosis, T cells exhibited robust metabolic activity and strong immune activation. In contrast, during relapse, T cells demonstrated reduced glycolytic and respiratory activity compared to their status at remission. The metabolic phenotype was accompanied by reduced cytotoxic ability. Metabolomics revealed lactate accumulation and substantial arginine depletion in the serum of these patients. The results observed in the human setting could be reproduced in a murine graft versus leukemia (GVL) model where T cells isolated from AML bearing mice exhibited lower metabolic activity compared to control animals. In vitro co-culture experiments enabled tightly controlled conditions for mechanistic studies. The metabolic impairment induced by AML cells was mediated by soluble factors, mirroring the major changes in serum metabolites of patients, and interfered with the crucial metabolic reprogramming in the priming phase of T cells. Well-known mechanisms such as nutrient deprivation could be excluded. Extensive transcriptomic and metabolomic analyses have been undertaken in order to identify the molecules responsible for the diminished metabolic activation. Influencing mitochondrial architecture thereby boosting respiratory activity prior to infusion led to enhanced anti-leukemic activity of T cells in vivo. The pre-treatment favored an immunological memory phenotype essential for a potent GVL effect. Using metabolic stimuli as pre-treatment of DLIs could promote T cell resistance against tumor evasion and increase anti-tumor immunity. This could greatly improve the success of DLIs and other adoptive T cell transfer strategies in the treatment of AML relapse. Disclosures Finke: Neovii: Consultancy, Honoraria, Other: travel grants, Research Funding; Medac: Consultancy, Honoraria, Other: travel grants, Research Funding; Novartis: Consultancy, Honoraria, Other: travel grants, Research Funding; Riemser: Consultancy, Honoraria, Research Funding.
- Published
- 2018
22. Oncogenic KRASG12D in the Hematopoietic System Causes NLRP3 Inflammasome Activation Leading to Myeloproliferative Syndrome
- Author
-
Tilman Brummer, Shaima’a Hamarsheh, Olaf Groß, Claudia Lengerke, Oliver Gorka, Lena Osswald, Justus Duyster, Melanie Börries, Miriam Erlacher, and Robert Zeiser
- Subjects
Myeloid ,Juvenile myelomonocytic leukemia ,business.industry ,Immunology ,Caspase 1 ,Inflammasome ,Inflammation ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Cancer research ,Bone marrow ,medicine.symptom ,Progenitor cell ,business ,medicine.drug - Abstract
Oncogenic Ras mutations occur frequently in myelodysplastic (MDS) and myeloproliferative syndromes (MPN). It was unclear if besides the direct transforming effect via constant RAS/MEK/ERK signaling, an inflammation related effect of KRAS contributes to the development of the disease. To address this question we performed a microarray based analysis of bone marrow derived from Rosa26CreERT2; LSL-KrasG12D mice after induction of KrasG12D with tamoxifen. We found that the NLRP3 inflammasome and related genes were upregulated. In agreement with increased NLRP3 sensitivity, KrasG12D bone marrow derived dendritic cells (BMDCs) showed increased inflammasome activation upon stimulation with LPS and ATP as quantified by intracellular levels of IL-1β and caspase-1 p20 subunit. To validate the functional role of the NLRP3 inflammasome for the MDS phenotype in vivo we generated Rosa26CreERT2; LSL-KrasG12D; NLRP3-/- mice. We transferred BM (CD45.2) from WT mice, Rosa26CreERT2; LSL-KrasG12D or Rosa26CreERT2; LSL-KrasG12D; NLRP3-/- mice into C57BL/6 WT recipients (CD45.1) after myeloablative conditioning and induced KrasG12D by tamoxifen treatment. Rosa26CreERT2; LSL-KrasG12D BM recipient mice developed MPN stigmata including anemia, leukopenia and thrombocytopenia. The mice also presented weight loss, splenomegaly, and an increase in myeloid cells in peripheral blood, spleen and BM, as well as increased CD11b+ckit+ immature myeloid progenitors. In contrast, Rosa26CreERT2; LSL-KrasG12D; NLRP3-/- recipient mice developed no disease phenotype. BMDCs isolated from Rosa26CreERT2; LSL-KrasG12D recipient mice exhibited strong NLRP3 inflammasome activation and high IL-1β levels which was not found in BMDCs of Rosa26CreERT2; LSL-KrasG12D; NLRP3-/- mice. CD11b+ cells of a Juvenile myelomonocytic leukemia (JMML) patient with an activating KRAS mutation showed increased inflammasome activation as compared to a non-mutant patient, which is in agreement with our observations in mouse models. Our findings support the concept that oncogenic KrasG12D does not only act via its oncogenic driver function, but also enhances activation of the NLRP3/IL-1β axis. This could lead to novel therapeutic approaches combining inhibition of oncogenic signaling and immune modulation via IL-1β blockade. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
23. Checkpoint Inhibition in CSF3R Mutated Chronic Neutrophilic Leukemia
- Author
-
Chuanjiang Yu, Dietmar Pfeifer, Anna Lena Illert, Justus Duyster, Stefanie Kreutmair, Khalid Shoumariyeh, Karsten Spiekermann, Michael Lübbert, Robert Zeiser, Teresa Poggio, and Sivahari Prasad Gorantla
- Subjects
Mutation ,Cell cycle checkpoint ,business.industry ,Immunology ,Chronic neutrophilic leukemia ,Cell Biology ,Hematology ,Colony-stimulating factor ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Transplantation ,Leukemia ,medicine.anatomical_structure ,Granulocyte macrophage colony-stimulating factor ,Cancer research ,Medicine ,Bone marrow ,business ,medicine.drug - Abstract
Mutations in the colony-stimulating factor 3 receptor (CSF3R) have been identified in 90% of chronic neutrophilic leukemia (CNL) and lower frequency in atypical chronic myeloid leukemia (aCML). It was shown recently, that signaling of the different types of CSF3R mutations varies, but a central role of the JAK/STAT pathway activation by oncogenic CSF3R membrane proximal point mutations has been anticipated. However, the exact downstream signaling pathways mediated by CSF3R mutations are still not completely understood. Recent studies in JAK mutated myeloproliferative diseases uncovered the vital importance of JAK/STAT signaling in PD-L1 mediated immune escape, thereby suggesting a possible role and mechanistic link for PD-1/PD-L1 immune checkpoint regulation in CNL and aCML. To investigate the dependence of the oncogenic CSF3R signaling cascade on an activated JAK/STAT axis, we performed Ruxolitinib treatment in MTT assays with BaF3 cells retrovirally infected with pMIG-R1 vectors harboring either CSF3R point or truncation mutations. Our data indicate, that both, CSF3R point and truncation mutations induced cell growth could be inhibited by Ruxolitinib treatment. To narrow down the involvement of the different JAK family kinases, we included CSF3R mutation transduced JAK1 deficient U4C, JAK2 deficient y2A and TYK2 deficient U1A cell lines in combination with different JAK inhibitors. Interestingly, here we were able to show that CSF3R point mutations require functional JAK1/TYK2-STAT3 signaling for oncogenic potential, whereas truncation mutations depend on all JAK kinases through STAT3/5 for activation. To further study potential therapeutic strategies, we examined checkpoint inhibition in CSF3R mutated disease. In this regard we demonstrate a significant induction of PD-L1 expression in murine cell lines upon expression of CSF3R point mutations T615A or T618I. To investigate the impact of PD-L1 upregulation in vivo, we used a retroviral murine bone marrow (BM) transplantation model where we infected BM of Balb/c mice with a vector carrying CSF3R T618I point mutation and transplanted it into lethally irradiated recipients. Transplanted mice developed a CSF3R driven leukemia and died by leukocytosis and organ infiltration, whereas mice transplanted with BM harboring CSF3RT618I and additional ADA mutation failed to induce leukemia. To test checkpoint inhibition as a potential therapeutic strategy in vivo, CSF3RT618I transplanted mice were treated regularly with anti-PD-L1-antibody or isotype control. Interestingly, checkpoint inhibition led to reduced EGFP burden and prolonged survival of responder mice suggesting a treatment response of immunooncologic agents in these rare diseases. In immunophenotypic analyses of primary patient material, we were able to show increased PD-L1 expression in primary diseased myeloid cells isolated from peripheral blood of CNL and aCML patients, thereby defining this structure as a possible new therapeutic target. To summarize, our data expand the knowledge of oncogenic CSF3R signaling pathways and implicate an immunotherapeutic strategy by checkpoint inhibition in CSF3R driven CNL and atypical CML. Disclosures Lubbert: Teva: Other: Study drug; Celgene: Other: Travel Grant; Janssen: Honoraria, Research Funding.
- Published
- 2018
24. Minimal Residual Disease (MRD) Analysis Using Multiparametric Flow Cytometry (MFC) and Identification of Differences in Subpopulations Using Next Generation Sequencing (NGS) in the Bone Marrow (BM) of Multiple Myeloma (MM) Patients (pts)
- Author
-
Milena Pantic, Ingrid Bartsch, Dagmar Wider, Stefan J. Mueller, Veronika Riebl, Marie Follo, Ralph Wäsch, Mascha Binder, Sandra Maria Dold, Justyna Rawluk, Monika Engelhardt, Cornelius Miething, K. Martin Kortüm, Mihaela Zlei, Chrissoula Kiote-Schmidt, Justus Duyster, Robert Zeiser, and Abdel Kareem Azab
- Subjects
Oncology ,medicine.medical_specialty ,medicine.diagnostic_test ,biology ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Leukapheresis ,Biochemistry ,Minimal residual disease ,CD19 ,Flow cytometry ,Transplantation ,EuroFlow ,Internal medicine ,medicine ,biology.protein ,Progression-free survival ,business - Abstract
Introduction: MM is characterized by the accumulation of aberrant BM plasma cells (aPCs). The use of novel agents for anti-MM treatment has enabled the achievement of deeper responses and prolonged progression free survival (PFS) and overall survival (OS). These advances have created the need for sensitive means to detect residual PCs. MFC allows aPC detection with high sensitivity and is used with NGS in clinical trials (CTs) (Paiva, 2015-2018). It allows the detection of residual cells after anti-MM therapies and is a risk denominator of pt subgroups and post-therapeutic outcome. The incorporation of cost-effective, readily available and standardized MRD tools into CTs and clinical practice seems a key requisite. It may allow the improvement of clinical decisions: 1) timing of stem cell transplantation (SCT), 2) need for consolidation, 3) duration of maintenance, 4) therapy modulations and 5) early relapse detection. Since MRD gains importance for novel agents' potency to achieve MRD negativity, including FDA/EMA-approvals, we assessed MFC for detection of aPCs, their change to treatment and over time. Methods: We systematically validated a 6-, 8- and 10-color panel using CD38, CD138, CD19, CD45, CD27, CD56, CD28, CD81, CD117, CD200 and kappa/lambda (k/l) in 9 different MM cell lines (MMCLs) and 213 BM, PB, leukapheresis (LA), extramedullary (EM) samples of 122 different MM pts and 14 healthy individuals (HI). These were immediately analyzed after sampling, using a bulk-lysis protocol. To reach a high sensitivity, >3x106 viable nucleated cells were analyzed. MFC validation included fluorescence-minus-one-(FMO), isotype- and spike-in-controls to evaluate the reliability and sensitivity of our panels. Spike-in controls were performed using a dilution assay to recover defined MMCL cells in the PB of HI. Data analysis was accomplished using the BeckmanCoulter software Kaluza®. We compared MFC using BM and PB samples of HI, MM pts at initial diagnosis (ID) or with progression (PD), after standard therapy (ST) and SCT. Additionally, BM samples at ID were sorted into each 4 aPC- and normal PC- (nPC) subpopulations for DNA and RNA sequencing and follow-up. The study was performed with written consent of all pts and HIs and approval of the ethics committee. Results: The antigen expression levels in 9 MMCLs using our 6-, 8- and 10-color panel could be confirmed as reported. The expression levels of antigens included in all panels were similar in all MMCLs (RPMI 8226; U266; IM-9; MM1.S; MM1.R; L363; Karpas620; NCI-H929; OPM-2). For all panels, we established easy to adapt gating strategies, using commercially available software to identify aPCs vs. nPCs in all analyzed samples. Using the 6-color panel, differentiation into up to 4 phenotypic subpopulations of aPCs and nPCs in MM pts and up to 4 subpopulations of nPCs in HI was achieved, depending on the heterogeneous phenotype the individual was expressing. Moreover, both DNA and RNA of obtained subpopulations were collected and analyzed via sequencing. Since in 1% of our samples, aPCs remained undetected via 6-color panel using surface antigens exclusively, we added k/l and did detect aPCs in all pts. The 10-color panel identified aPC- and nPC-subpopulations in even more detail. We determined a MRD sensitivity of 10-5, confirmed via independent dilution assays. In 48% of BM and PB samples, aPCs were determined at ID and PD; 52% were assessed for MRD after ST and SCT. Of the latter, 55% were BM samples, of which 5% showed MRD negativity of Conclusion: Our 6-, 8-, and 10-color panels allow highly sensitive detection of aPCs vs. nPCs in MM samples. These panels have been validated using various controls, dilution assays, 9 MMCLs, MM pt samples and HI and can be implemented into routine diagnostics. The analysis of DNA and RNA expression patterns in distinct subpopulations will generate relevant insight, which subclones contribute to response vs. resistance mechanisms. The EuroFlow Consortium have established a two 8-color tubes test for identification of aPCs, which - albeit used worldwide - may not be applicable for every center and every pt outside CTs. Therefore, meticulously validated alternatives as shown here are of interest. Our 8-color panel has been included into our CT portfolio. Disclosures Azab: Cellatrix LLC: Equity Ownership, Other: Founder and owner; Targeted Therapeutics LLC: Equity Ownership, Other: Founder and owner; Ach Oncology: Research Funding; Glycomimetics: Research Funding. Zeiser:Jazz Pharmaceuticals: Research Funding. Wäsch:Pfizer: Honoraria.
- Published
- 2018
25. Functional Characterization of Epigenetic Modifiers of Demethylating Therapy in AML
- Author
-
Cornelius Miething, Pia Veratti, Khalid Shoumariyeh, Desiree Melanie Redhaber, Justus Duyster, Jessica Beckert, Silvia Schäfer, Sophia Ehrenfeld, Jan Mitschke, and Laurent Phely
- Subjects
Immunology ,Cancer research ,Cell Biology ,Hematology ,Epigenetics ,Biology ,Biochemistry - Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults, and remains a difficult to treat disease, especially in elderly patients. AML arises in hematopoietic stem and progenitor cells and frequently carries alterations in genes involved in epigenetic regulation. Furthermore, azacitidine (AZA) and decitabine (DAC), two nucleoside analogs inhibiting DNA methylation (DNMTi), have shown clinical efficacy in the therapy of myelodysplastic syndrome (MDS) and AML, highlighting the importance of epigenetic regulation in AML. Up to now, the mechanism of action of AZA and DAC in AML has not been fully elucidated, and reliable biomarkers of therapy response to these drugs do not yet exist. To identify epigenetic response modifiers towards AZA and DAC treatment, we conducted a pooled shRNA-based screen in three human leukemia cell lines using an inducible custom shRNA library targeting more than 660 different genes involved in epigenetic pathways. After selection of retrovirally infected cells, shRNA expression was induced and a fraction of cells were treated with either AZA, DAC or left untreated. 10 days after shRNA induction, genomic DNA was extracted and deep-sequenced to identify genes conferring a selective advantage/disadvantage under these conditions. By comparing shRNA representation changes between the differentially treated groups, we identified multiple candidate genes sensitizing or protecting leukemic cells to/from AZA and DAC treatment. Notably, the DNMT1 gene itself expectedly emerged as an important sensitizer to DNMTi, as did other CXXC domain-containing genes binding to (un-)methylated CpG domains. Another prominent set of genes in the pooled analysis belonged to the DNA damage pathway, including genes such as PARP1 and BRCA1. The identified candidate genes were validated and confirmed in more than 80% when tested in single shRNAs competition assays. Their impact on DNA methylation and gene expression is currently being assessed, with ongoing experiments focusing on the mechanistic role of some of the validated target, as well as the clinical potential for combinatorial therapies. Thus, using an unbiased functional genetic approach, we identified multiple genes including CpG binding proteins and members of the DNA damage pathway as important modulators of DNMTi response. Our results will increase our understanding of the molecular mechanisms driving DNMTi efficacy, and may help to identify new biomarkers and novel targets for combinational therapy in AML. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
26. Oncogenic JAK2V617F requires an intact SH2-like domain for constitutive activation and induction of a myeloproliferative disease in mice
- Author
-
Christian Meyer zum Büschenfelde, Christian Peschel, Justus Duyster, Marcus Kremer, Anna Lena Illert, Sivahari P. Gorantla, Tobias Dechow, and Rebekka Grundler
- Subjects
medicine.medical_treatment ,Immunology ,Mutant ,Biology ,medicine.disease_cause ,SH2 domain ,Biochemistry ,src Homology Domains ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Phosphorylation ,Receptor ,Cell Proliferation ,Mutation ,Myeloproliferative Disorders ,Oncogene ,Cell Biology ,Hematology ,Janus Kinase 2 ,Cytokine ,Cancer research ,Cytokine receptor - Abstract
The oncogenic JAK2V617F mutation is found in myeloproliferative neoplasms (MPNs) and is believed to be critical for leukemogenesis. Here we show that JAK2V617F requires an intact SH2 domain for constitutive activation of downstream signaling pathways. In addition, there is a strict requirement of cytokine receptor expression for the activation of this oncogene. Further analysis showed that the SH2 domain mutation did not interfere with JAK2 membrane distribution. However, coimmunoprecipitated experiments revealed a role for the SH2 domain in the aggregation and cross-phosphorylation of JAK2V617F at the cell membrane. Forced overexpression of cytokine receptors could rescue the JAK2V617F SH2 mutant supporting a critical role of JAK2V617F abundance for constitutive activation. However, under physiologic cytokine receptor expression the SH2 domain is absolutely necessary for oncogenic JAK2V617F activation. This is demonstrated in a bone marrow transplantation model, in which an intact SH2 domain in JAK2V617F is required for the induction of an MPN-like disease. Thus, our results points to an indispensable role of the SH2 domain in JAK2V617F-induced MPNs.
- Published
- 2010
27. Aurora kinases A and B are up-regulated by Myc and are essential for maintenance of the malignant state
- Author
-
Ulrich Keller, Sara Rimpi, Jürgen den Hollander, Martina Rudelius, Justus Duyster, Markus Scheerer, Marcus Kremer, John L. Cleveland, Christian Peschel, Nikolas von Bubnoff, Jonas Nilsson, Andrei Goga, Joanne R. Doherty, Nikolas Graf, Andreas K. Buck, Alexander Hoellein, and Mark A. Hall
- Subjects
Chromatin Immunoprecipitation ,BALB 3T3 Cells ,Lymphoma, B-Cell ,Blotting, Western ,Immunology ,Aurora B kinase ,Aurora inhibitor ,Apoptosis ,Electrophoretic Mobility Shift Assay ,Mice, Transgenic ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Immunoenzyme Techniques ,Proto-Oncogene Proteins c-myc ,Mice ,Aurora kinase ,Aurora Kinases ,Biomarkers, Tumor ,Animals ,Aurora Kinase B ,Humans ,RNA, Messenger ,RNA, Small Interfering ,Kinase activity ,Luciferases ,Cells, Cultured ,Aurora Kinase A ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,B-Lymphocytes ,Lymphoid Neoplasia ,Reverse Transcriptase Polymerase Chain Reaction ,Kinase ,Gene Expression Profiling ,Cell Biology ,Hematology ,Cell cycle ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,Cancer research - Abstract
Myc oncoproteins promote continuous cell growth, in part by controlling the transcription of key cell cycle regulators. Here, we report that c-Myc regulates the expression of Aurora A and B kinases (Aurka and Aurkb), and that Aurka and Aurkb transcripts and protein levels are highly elevated in Myc-driven B-cell lymphomas in both mice and humans. The induction of Aurka by Myc is transcriptional and is directly mediated via E-boxes, whereas Aurkb is regulated indirectly. Blocking Aurka/b kinase activity with a selective Aurora kinase inhibitor triggers transient mitotic arrest, polyploidization, and apoptosis of Myc-induced lymphomas. These phenotypes are selectively bypassed by a kinase inhibitor-resistant Aurkb mutant, demonstrating that Aurkb is the primary therapeutic target in the context of Myc. Importantly, apoptosis provoked by Aurk inhibition was p53 independent, suggesting that Aurka/Aurkb inhibitors will show efficacy in treating primary or relapsed malignancies having Myc involvement and/or loss of p53 function.
- Published
- 2010
28. NPM-ALK–dependent expression of the transcription factor CCAAT/enhancer binding protein β in ALK-positive anaplastic large cell lymphoma
- Author
-
Natasa Anastasov, Antonio Martinez, Mark Raffeld, Falko Fend, Justus Duyster, Cornelius Miething, Leticia Quintanilla-Martinez, Stefania Pittaluga, Angelica Vivero, Elaine S. Jaffe, Martina Rudelius, Theresa Davies-Hill, and Margit Klier
- Subjects
Lymphoid Tissue ,Immunology ,Gene Expression ,Biology ,Transfection ,Biochemistry ,Cell Line, Tumor ,hemic and lymphatic diseases ,Enhancer binding ,medicine ,Humans ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,Kinase activity ,Transcription factor ,Anaplastic large-cell lymphoma ,Ccaat-enhancer-binding proteins ,CCAAT-Enhancer-Binding Protein-beta ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Immunohistochemistry ,Molecular biology ,BCL10 ,Lymphoma ,Cancer research ,Lymphoma, Large B-Cell, Diffuse - Abstract
CCAAT/enhancer binding protein β (C/EBPβ) is one of a 6-member family of C/EBPs. These transcription factors are involved in the regulation of various aspects of cellular growth and differentiation. Although C/EBPβ has important functions in B- and T-cell differentiation, its expression has not been well studied in lymphoid tissues. We, therefore, analyzed its expression by immunohistochemistry and Western blot in normal lymphoid tissues and in 248 well-characterized lymphomas and lymphoma cell lines. Nonneoplastic lymphoid tissues and most B-cell, T-cell, and Hodgkin lymphomas lacked detectable levels of C/EBPβ. In contrast, most (40 of 45; 88%) cases of ALK-positive anaplastic large cell lymphoma (ALCL) strongly expressed C/EBPβ. Western blot analysis confirmed C/EBPβ expression in the ALK-positive ALCLs and demonstrated elevated levels of the LIP isoform, which has been associated with increased proliferation and aggressiveness in carcinomas. Transfection of Ba/F3 and 32D cells with NPM-ALK and a kinase-inhibitable modified NPM-ALK resulted in the induction of C/EBPβ and demonstrated dependence on NPM-ALK kinase activity. In conclusion, we report the constitutive expression of C/EBPβ in ALK-positive ALCL and show its relationship to NPM-ALK. We suggest that C/EBPβ is likely to play an important role in the pathogenesis and unique phenotype of this lymphoma.
- Published
- 2006
29. Bcr-Abl resistance screening predicts a limited spectrum of point mutations to be associated with clinical resistance to the Abl kinase inhibitor nilotinib (AMN107)
- Author
-
Christian Peschel, Paul W. Manley, Jürgen Mestan, Nikolas von Bubnoff, Justus Duyster, and Jana Sänger
- Subjects
Immunology ,Fusion Proteins, bcr-abl ,Biology ,Philadelphia chromosome ,Biochemistry ,Piperazines ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Humans ,Point Mutation ,Genetic Testing ,Protein Kinase Inhibitors ,ABL ,Dose-Response Relationship, Drug ,Point mutation ,Myeloid leukemia ,Imatinib ,Cell Biology ,Hematology ,medicine.disease ,Protein Structure, Tertiary ,Leukemia ,Pyrimidines ,Imatinib mesylate ,Nilotinib ,Drug Resistance, Neoplasm ,Benzamides ,Imatinib Mesylate ,Cancer research ,medicine.drug - Abstract
In advanced-phase chronic myeloid leukemia (CML), resistance to imatinib mesylate is associated with point mutations in the BCR-ABL kinase domain. A new generation of potent ABL kinase inhibitors is undergoing clinical evaluation. It is important to generate specific resistance profiles for each of these compounds, which could translate into combinatorial and sequential treatment strategies. Having characterized nilotinib (AMN107) against a large panel of imatinib mesylate–resistant Bcr-Abl mutants, we investigated which mutants might arise under nilotinib therapy using a cell-based resistance screen. In contrast to imatinib mesylate, resistance to nilotinib was associated with a limited spectrum of Bcr-Abl kinase mutations. Among these were mutations affecting the P-loop and T315I. Rarely emerging resistant colonies at a concentration of 400 nM nilotinib exclusively expressed the T315I mutation. With the exception of T315I, all of the mutations that were identified were effectively suppressed when the nilotinib concentration was increased to 2000 nM, which falls within the peak-trough range in plasma levels (3.6-1.7 μM) measured in patients treated with 400 mg twice daily. Our findings suggest that nilotinib might be superior to imatinib mesylate in terms of the development of resistance. However, our study indicates that clinical resistance to nilotinib may be associated with the predominant emergence of T315I.
- Published
- 2006
30. A cell-based screen for resistance of Bcr-Abl-positive leukemia identifies the mutation pattern for PD166326, an alternative Abl kinase inhibitor
- Author
-
Christian Peschel, William G. Bornmann, Darren R. Veach, Nikolas von Bubnoff, Walter E. Aulitzky, Jana Sänger, Bayard D. Clarkson, Heiko van der Kuip, Petra Seipel, and Justus Duyster
- Subjects
Pyridines ,Immunology ,Cell Culture Techniques ,Fusion Proteins, bcr-abl ,Genes, abl ,Biology ,Transfection ,medicine.disease_cause ,Philadelphia chromosome ,Biochemistry ,Piperazines ,Cell Line ,Mice ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Point Mutation ,Protein kinase A ,Protein Kinase Inhibitors ,Cells, Cultured ,Cell Line, Transformed ,Mutation ,ABL ,Point mutation ,Imatinib ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Molecular biology ,Protein Structure, Tertiary ,Leukemia ,Pyrimidines ,Imatinib mesylate ,Drug Resistance, Neoplasm ,Benzamides ,Imatinib Mesylate ,Cancer research ,medicine.drug - Abstract
In Philadelphia-positive (Ph(+)) leukemia, point mutations within the Bcr-Abl kinase domain emerged as a major mechanism of resistance to imatinib mesylate. We established a cell-based screening strategy for detection of clinically relevant point mutations using Bcr-Abl-transformed Ba/F3 cells. We identified 32 different single-point mutations within the kinase domain of Bcr-Abl. The pattern and frequency of mutations in this cell culture-based screen resembled the pattern and frequency observed in resistant patients. We then applied this screen to an alternative Abl kinase inhibitor. Using PD166326, the frequency of resistant colonies emerging at 5 to 10 times the median growth inhibition (IC50) of PD166326 was significantly lower than with imatinib. In addition, PD166326 produced a distinct pattern of Bcr-Abl mutations. The majority of mutations that came up with both imatinib and PD166326 could effectively be suppressed by increasing the dose of PD166326 to 50 to 500 nM. In contrast, only a few mutations could be suppressed by increasing the imatinib dose to 5 to 10 microM. However, 3 mutations affecting F317 displayed complete resistance to PD166326, but could be effectively inhibited by standard concentrations of imatinib. Thus, this robust and simple screening system provides a rational basis for combinatorial and sequential treatment strategies in targeted cancer therapy.
- Published
- 2005
31. Efficacy and safety of imatinib in adult patients with c-kit–positive acute myeloid leukemia
- Author
-
Thomas M. Fischer, Frank Breitenbuecher, Thomas Kindler, Andreas H. Marx, Georg Hess, Joachim Beck, Birgit Weinkauf, Christoph Huber, Matthias Theobald, Harald Gschaidmeier, Charles James Kirkpatrick, Justus Duyster, and Christian Peschel
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,DNA Mutational Analysis ,Immunology ,Antineoplastic Agents ,Cell Count ,Pilot Projects ,Biochemistry ,Piperazines ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Phosphorylation ,neoplasms ,Aged ,Salvage Therapy ,Chemotherapy ,business.industry ,Remission Induction ,Myeloid leukemia ,Imatinib ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Clinical trial ,Proto-Oncogene Proteins c-kit ,Leukemia ,Pyrimidines ,Treatment Outcome ,Imatinib mesylate ,medicine.anatomical_structure ,Leukemia, Myeloid ,Acute Disease ,Benzamides ,Imatinib Mesylate ,Female ,Bone marrow ,Blast Crisis ,business ,medicine.drug - Abstract
This phase 2 pilot study was conducted to determine the efficacy and safety of imatinib mesylate in patients with c-kit–positive acute myeloid leukemia (AML) refractory to or not eligible for chemotherapy. Twenty-one patients were enrolled and received imatinib 600 mg orally once daily. Five responses were seen primarily in patients, starting with relatively low blast counts in bone marrow (BM) and peripheral blood (PB): 2 patients who were considered refractory on chemotherapy on the basis of persistence of blasts in PB and BM met the criteria for complete hematologic remission, 1 patient had no evidence of leukemia, and 2 patients achieved a minor response. Treatment with imatinib demonstrated a good safety profile and was well tolerated. Western blot analysis and immunohistochemistry demonstrated c-Kit activation in primary AML cells. Further, imatinib treatment of primary AML cells inhibited c-Kit tyrosine-phosphorylation. Genomic DNA-sequencing of c-KIT showed no mutations in exons 2, 8, 10, 11, 12, and 17. Although some of the responses derived from relatively small reductions in leukemic blasts and may be attributable, in part, to prior chemotherapy, these cases suggest that imatinib has interesting clinical activity in a subset of patients with c-kit–positive AML. Further clinical trials are warranted to explore the clinical potential of imatinib in AML and to identify the underlying molecular mechanism.
- Published
- 2004
32. Interferon consensus sequence binding protein (ICSBP; IRF-8) antagonizes BCR/ABL and down-regulates bcl-2
- Author
-
Justus Duyster, Rolf Müller, Emily P. Slater, Andreas Neubauer, Manuel Schmidt, Markus Ritter, Andreas Burchert, Andreas Hochhaus, Martin Eilers, Lorenz C. Hofbauer, Magnus K. R. Samuelsson, and Dali Cai
- Subjects
Male ,Immunology ,Fusion Proteins, bcr-abl ,Down-Regulation ,Apoptosis ,Biology ,Transfection ,Biochemistry ,Piperazines ,Cell Line ,Mice ,Interferon ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,neoplasms ,Psychological repression ,Transcription factor ,Cell Line, Transformed ,Reporter gene ,ABL ,Multipotent Stem Cells ,breakpoint cluster region ,Drug Synergism ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Repressor Proteins ,Cell Transformation, Neoplastic ,Pyrimidines ,Proto-Oncogene Proteins c-bcl-2 ,Drug Resistance, Neoplasm ,Benzamides ,Interferon Regulatory Factors ,Imatinib Mesylate ,Cancer research ,Chronic myelogenous leukemia ,medicine.drug ,Interferon regulatory factors - Abstract
BCR/ABL is the causative genetic aberration in chronic myelogenous leukemia (CML). Mice lacking expression of the interferon (IFN) consensus sequence binding protein (ICSBP), an IFN gamma-inducible transcription factor of the interferon regulatory factor (IRF) family, develop a disease similar to human CML. Mounting evidence suggests a role for ICSBP in the pathogenesis of CML. However, the underlying mechanisms are largely unknown. By stable and conditional expression of ICSBP in wild-type and BCR/ABL-transformed 32D cells (32D/wt and 32D/BA), we found that ICSBP inhibited BCR/ABL-mediated leukemogenesis in vivo. Moreover, ICSBP also overrode BCR/ABL-mediated morphology changes, chemotherapy, and imatinib resistance, as well as BCR/ABL-induced repression of differentiation. Some of these ICSBP effects may be explained in part by an ICSBP-mediated repression of bcl-2, a major antiapoptotic target of BCR/ABL, on transcriptional and protein level. Using reporter gene assays and electrophoretic mobility shift assays we identified that the bcl-2 promoter activity was inhibited by ICSBP by way of a fragment containing 2 characteristic ICSBP-responsive elements. An inverse correlation between ICSBP and bcl-2 expression was confirmed in vivo. Collectively, our findings suggest that ICSBP antagonizes BCR/ABL by down-regulation of bcl-2 and implicates a central role for ICSBP in the pathogenesis of CML, as well as a therapeutic target to overcome drug resistance in bcl-2-dependent tumors.
- Published
- 2004
33. Nucleophosmin-anaplastic lymphoma kinase of anaplastic large-cell lymphoma recruits, activates, and uses pp60c-src to mediate its mitogenicity
- Author
-
Stephan W. Morris, Serge Roche, Catherine Greenland, Bernard Payrastre, Georges Delsol, Ren Yuan Bai, Justus Duyster, Daniel Cussac, and Michèle Allouche
- Subjects
Proto-Oncogene Proteins pp60(c-src) ,Immunology ,Biology ,Biochemistry ,Translocation, Genetic ,Jurkat Cells ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Anaplastic lymphoma kinase ,Anaplastic Lymphoma Kinase ,Phosphorylation ,Kinase activity ,Tyrosine ,Nucleoplasmins ,Anaplastic large-cell lymphoma ,Nucleophosmin ,integumentary system ,Nuclear Proteins ,Receptor Protein-Tyrosine Kinases ,Blood Proteins ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,Phosphoproteins ,medicine.disease ,Fusion protein ,Chromosomes, Human, Pair 2 ,Cancer research ,Chromosomes, Human, Pair 5 ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Tyrosine kinase ,Cell Division - Abstract
Anaplastic large-cell lymphomas (ALCLs) are lymphomas of T or null phenotype often associated with a chromosomal translocation, t(2;5)(p23;q35). This translocation leads to the expression of a hybrid protein consisting of the N-terminal portion of nucleophosmin (NPM) and the intracellular domain of the anaplastic lymphoma kinase (ALK). NPM-ALK possesses a constitutive tyrosine kinase activity responsible for its oncogenic property through activation of downstream effectors such as phospholipase C gamma (PLC-gamma) and the type IA phosphoinositide 3-kinase. Here, we show that the Src-kinases, particularly pp60(c-src), associate with and are activated by NPM-ALK expression in various cells, and in cell lines established from patients with ALCL. The kinase activity and the tyrosine 418 of NPM-ALK are required for its association with Src-kinases. Y418F mutation of NPM-ALK impaired its association with Src-kinases and strongly reduced the proliferation rate of Ba/F3 cells. In agreement, Src-kinase inhibitors or pp60(c-src) siRNA significantly decreased the proliferation rate of NPM-ALK-positive ALCL cell lines. Moreover, using active or inactive forms of pp60(c-src) and NPM-ALK, we provide evidence that NPM-ALK is a potential substrate of pp60(c-src). Overall, our data place Src-kinases as new important downstream effectors of NPM-ALK and as attractive potential therapeutic targets for new ALCL treatment.
- Published
- 2003
34. Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor
- Author
-
Christian Thiede, Justus Duyster, Christine Steudel, Christian Peschel, Cornelius Miething, and Rebekka Grundler
- Subjects
Indoles ,Drug Resistance ,Mitogen-activated protein kinase kinase ,medicine.disease_cause ,Biochemistry ,Tropomyosin receptor kinase C ,Mice ,fluids and secretions ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Enzyme Inhibitors ,Phosphorylation ,Sequence Deletion ,Mutation ,hemic and immune systems ,Hematology ,Tyrphostins ,Milk Proteins ,Recombinant Proteins ,Neoplasm Proteins ,DNA-Binding Proteins ,Leukemia, Myeloid ,Acute Disease ,embryonic structures ,Tyrosine kinase ,Cell Division ,Immunology ,Mutation, Missense ,Biology ,Transfection ,Cell Line ,Structure-Activity Relationship ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Kinase activity ,Codon ,MAP kinase kinase kinase ,Point mutation ,Membrane Proteins ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Hematopoietic Stem Cells ,Staurosporine ,Molecular biology ,Enzyme Activation ,Mutagenesis, Insertional ,Amino Acid Substitution ,fms-Like Tyrosine Kinase 3 ,Trans-Activators ,Cancer research ,Cyclin-dependent kinase 9 ,Protein Processing, Post-Translational - Abstract
Activating mutations of FLT3 have been detected in patients with acute myeloid leukemia (AML). Two distinct types of FLT3 mutations are most common: internal tandem duplication (ITD) of sequences coding for the juxtamembrane domain and point mutations at codon 835 (Asp835) within the kinase domain. Both types of mutations constitutively activate the tyrosine kinase activity of FLT3 in experimental systems and result in factor-independent proliferation of Ba/F3 and 32D cells. Recently, novel mutations within the activation loop were identified in patients with AML: deletion of isoleucine 836 (Ile836del) and an exchange of isoleucine 836 to methionine plus an arginine insertion (Ile836Met+Arg). To examine whether the Ile836 mutations result in constitutive activation of the FLT3 receptor, we introduced both mutant FLT3 cDNAs transiently into HEK 293 cells. Both mutant FLT3 receptors were constitutively autophosphorylated in the absence of ligand and kinase activity led to constitutive activation of downstream signaling cascades as determined by activation of the STAT5 (signal transducer and activator of transcription 5) pathway. When stably expressed in the growth factor–dependent cell lines Ba/F3 and 32D, both deletion and insertion mutants led to factor-independent proliferation, indicating that both mutants have transforming capabilities. We then examined the sensitivity of the FLT3 ITD, FLT3 Asp835Tyr, and the novel FLT3 receptor mutants toward the kinase inhibitors AG1296, PKC412, and SU5614. We show that these FLT3 kinase inhibitors have distinct inhibitory potencies against different activating FLT3 receptor mutants. These results suggest that it may be useful to determine the exact kind of FLT3 mutation when applying receptor kinase inhibitors in clinical trials.
- Published
- 2003
35. The IL-33/ST2 axis augments effector T-cell responses during acute GVHD
- Author
-
Brent H. Koehn, Tobias Wertheimer, James L.M. Ferrara, Vincent Schwarze, Patricia A. Taylor, Lukas Schwab, Yvonne Beck, Robert Zeiser, Natalie Stickel, Annette Schmitt-Graeff, Max Warncke, Susumu Nakae, Benjamin M. Matta, Dawn K. Reichenbach, Jason Devlin, Victor Tkachev, Marie Follo, Heth R. Turnquist, Bruce R. Blazar, Justus Duyster, Dietmar Pfeifer, Tobias Junt, Elisabeth Lieberknecht, Heide Dierbach, Quan Liu, Gabriele Prinz, and Simon C. Watkins
- Subjects
Isoantigens ,Transplantation Conditioning ,T cell ,T-Lymphocytes ,Immunology ,Interleukin-1 Receptor-Like 1 Protein ,Gene Expression ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Receptors, Cell Surface ,Biochemistry ,Severity of Illness Index ,Interferon-gamma ,Mice ,Downregulation and upregulation ,immune system diseases ,medicine ,Animals ,Cluster Analysis ,Humans ,Transplantation, Homologous ,Intestinal Mucosa ,Receptor ,Mice, Knockout ,business.industry ,Gene Expression Profiling ,Interleukins ,Hematopoietic Stem Cell Transplantation ,Interleukin ,Cell Biology ,Hematology ,Interleukin-33 ,Tissue Donors ,Transplantation ,Interleukin 33 ,Intestines ,Disease Models, Animal ,surgical procedures, operative ,medicine.anatomical_structure ,Acute Disease ,Cancer research ,Tumor necrosis factor alpha ,business - Abstract
Interleukin (IL)-33 binding to the receptor suppression of tumorigenicity 2 (ST2) produces pro-inflammatory and anti-inflammatory effects. Increased levels of soluble ST2 (sST2) are a biomarker for steroid-refractory graft-versus-host disease (GVHD) and mortality. However, whether sST2 has a role as an immune modulator or only as a biomarker during GVHD was unclear. We show increased IL-33 production by nonhematopoietic cells in the gastrointestinal (GI) tract in mice post-conditioning and patients during GVHD. Exogenous IL-33 administration during the peak inflammatory response worsened GVHD. Conversely, GVHD lethality and tumor necrosis factor-α production was significantly reduced in il33(-/-) recipients. ST2 was upregulated on murine and human alloreactive T cells and sST2 increased as experimental GVHD progressed. Concordantly, st2(-/-) vs wild-type (WT) donor T cells had a marked reduction in GVHD lethality and GI histopathology. Alloantigen-induced IL-18 receptor upregulation was lower in st2(-/-) T cells, and linked to reduced interferon-γ production by st2(-/-) vs WT T cells during GVHD. Blockade of IL-33/ST2 interactions during allogeneic-hematopoietic cell transplantation by exogenous ST2-Fc infusions had a marked reduction in GVHD lethality, indicating a role of ST2 as a decoy receptor modulating GVHD. Together, these studies point to the IL-33/ST2 axis as a novel and potent target for GVHD therapy.
- Published
- 2014
36. Adhesion to fibronectin selectively protects Bcr-Abl+cells from DNA damage–induced apoptosis
- Author
-
Walter E. Aulitzky, Heiko van der Kuip, Alexander W. Goetz, Cornelius Miething, and Justus Duyster
- Subjects
Integrins ,Recombinant Fusion Proteins ,Immunology ,Integrin ,Fusion Proteins, bcr-abl ,bcl-X Protein ,Apoptosis ,Protein Serine-Threonine Kinases ,Transfection ,Biochemistry ,Piperazines ,Cell Line ,Mice ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Cell Adhesion ,In Situ Nick-End Labeling ,Animals ,Lymphocytes ,Enzyme Inhibitors ,Protein kinase B ,Cell Line, Transformed ,Microscopy, Confocal ,biology ,Cell adhesion molecule ,Cell Cycle ,DNA ,Cell Biology ,Hematology ,Molecular biology ,Fibronectins ,Mitochondria ,Enzyme Activation ,Fibronectin ,Pyrimidines ,Imatinib mesylate ,Gene Expression Regulation ,Proto-Oncogene Proteins c-bcl-2 ,Gamma Rays ,Benzamides ,Imatinib Mesylate ,biology.protein ,Interleukin-3 ,Neural cell adhesion molecule ,Proto-Oncogene Proteins c-akt ,Cell Division ,DNA Damage - Abstract
The phenotype of Bcr-Abl–transformed cells is characterized by a growth factor-independent survival and a reduced susceptibility to apoptosis. Furthermore, Bcr-Abl kinase alters adhesion features by phosphorylating cytoskeletal and/or signaling proteins important for integrin function. Integrin-mediated adhesion to extracellular matrix molecules is critical for the regulation of growth and apoptosis. However, effects of integrin signaling on regulation of apoptosis in cells expressing Bcr-Abl are largely unknown. The influence of adhesion on survival and apoptosis in Bcr-Abl+ and Bcr-Abl− BaF3 cells was investigated. p185bcr-abl–transfected BaF3 cells preadhered to immobilized fibronectin had a significant survival advantage and reduced susceptibility to apoptosis following γ-irradiation when compared with the same cells grown on laminin, on polylysin, or in suspension. Both inhibition of Bcr-Abl kinase by STI571 and inhibition of specific adhesion reversed the fibronectin-mediated antiapoptotic effect in BaF3p185. The DNA damage response of Bcr-Abl−BaF3 cells was not affected by adhesion to fibronectin. In contrast to parental BaF3 cells, BaF3p185 adherent to fibronectin did not release cytochrome c to the cytosol following irradiation. The fibronectin-mediated antiapoptotic mechanism in Bcr-Abl–active cells was not mediated by overexpression of Bcl-XL or Bcl-2 but required an active phosphatidylinositol 3-kinase (PI-3K). Kinase-active Bcr-Abl in combination with fibronectin-induced integrin signaling led to a hyperphosphorylation of AKT. Thus, cooperative activation of PI-3K/AKT by Bcr-Abl and integrins causes synergistic protection of Bcr-Abl+ cells from DNA damage–induced apoptosis.
- Published
- 2001
37. Evaluation of the Risk of Relapse and Survival Using a Conditional Survival Approach in 815 Multiple Myeloma Patients
- Author
-
Justus Duyster, Roland Mertelsmann, Martin Schumacher, Maximilian Schinke, Johannes M. Waldschmidt, Ralph Waesch, Gabriele Ihorst, Inga Promny, and Monika Engelhardt
- Subjects
Oncology ,medicine.medical_specialty ,Relative survival ,business.industry ,Proportional hazards model ,Immunology ,Cancer ,Context (language use) ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Surgery ,Clinical trial ,Internal medicine ,Cohort ,medicine ,Stage (cooking) ,business ,Multiple myeloma - Abstract
Introduction: Disease monitoring based on molecular markers obtained by noninvasive or minimally invasive methods will potentially allow the early detection of treatment response or disease progression in cancer patients (pts). Investigations in order to identify prognostic factors, e.g. pt baseline characteristics or molecular markers, contributing to long-term survival potentially provide important information for pts with multiple myeloma (MM). However, overall survival (OS) is not very informative for pts who already survived 5 or 10 more years (yrs). To better characterize long-term survival, conditional survival (CS) analyses are useful. CS describes probabilities of surviving t additional yrs given they survived s yrs and provides information, how prognosis evolves over time. We evaluated relative survival using a conditional approach and have described initial results in a large data set of MM pts with long-term survival, which is mandatory for the calculation of CS (Hieke et al., Clin Cancer Res 2015). The results were further refined here by accumulating time-dependent laboratory and MM-disease specific risks, including cytogenetics and response to treatment over time. Methods: We evaluated 815 consecutive MM pts treated at our department from 1997 to 2011, with follow-up until 6/2016. We assessed >20 variables and risks, including gender, age, stage, admission period, response and relevant MM-related risk factors over time. We calculated 5-yr CS and stratified 5-yr CS according to disease- and host-related risks. Component-wise likelihood-based boosting and variables selected by boosting were investigated in a multivariable Cox model. The median follow-up time was 10.3 yrs and the median OS in the entire cohort 5.1 yrs. Results: Our pts showed typical characteristics for referral centers as previously described (Engelhardt et al. 2014-2016), e.g. pt frequencies of 70 yrs were 42%, 34% and 24%, respectively. The OS probabilities at 5- and 10-years were 50% and 25%, respectively. The 5-yr CS probabilities remained almost constant over the yrs, if a pt had already survived after initial diagnosis (~50%). According to baseline variables, CS estimates showed no gender difference. The estimated 5-yr survival probabilities varied substantially, from 25% for pts aged >70 to 65% for pts Conclusions: CS has attracted attention in recent yrs either in an absolute or relative form where the latter is based on a comparison with an age-adjusted normal population being highly relevant from a public health perspective. In its absolute form, CS constitutes the quantity of major interest in a clinical context. We defined CS by using the fact that the pt is alive at the prediction time s as the conditioning event. Our CS data have considerable clinical implications: because the 5-yr CS remains stable, post treatment surveillance in MM is continuously justified and end points for clinical trial designs should adjust their follow-up accordingly. Even in the modern treatment era, this is different to e.g. Hodgkin lymphoma, where a low risk of relapse has been demonstrated if pts were event free at 2 yrs in a conditional survival approach (Hapgood et al. JCO 2016). CS data in various hematological tumors therefore vary substantially and remain warranted to clearly determine. Our analysis of time-dependent risk variables from diagnosis to prediction time s and use of response, response duration and other MM-specific variables should refine CS towards an even more specifically determined prognosis. Disclosures Engelhardt: MSD: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Other: Travel/Accommodation/Expenses , Research Funding; Janssen: Consultancy, Honoraria.
- Published
- 2016
38. Time from First Symptom Onset to the Final Diagnosis of Multiple Myeloma - Possible Risks and Future Solutions: Large Retrospective and Confirmatory Prospective Analysis
- Author
-
Karl Henne, Werner Vach, Georg W. Herget, Justus Duyster, Giulia Graziani, Gabriele Ihorst, Ralph Wäsch, Annette M. May, Johannes M. Waldschmidt, and Monika Engelhardt
- Subjects
Pediatrics ,medicine.medical_specialty ,Referral ,business.industry ,Immunology ,Cell Biology ,Hematology ,Disease ,Malignancy ,medicine.disease ,Biochemistry ,Informed consent ,Orthopedic surgery ,Medicine ,medicine.symptom ,business ,Bone pain ,Prospective cohort study ,Multiple myeloma - Abstract
Introduction:Multiple Myeloma (MM) has seen major advances in the understanding and management, among them revised disease definitions and recognition of early diagnosis-defining events. Early MM diagnosis and treatment initiation of symptomatic patients (pts) have been revised via IMWG criteria. In line, early recognition, especially of symptomatic MM, is important. However, MM represents an indolent malignancy with often unspecific initial symptoms. This may complicate diagnosis finding and result in substantial delays until the correct diagnosis is made. Objective literature and larger retro- or prospective analyses are lacking and possible risk factors, which might lead to a delayed diagnosis, remain undetermined. This project addressed this important aspect with the aim to optimize earlier diagnosis finding in MM and as a consequence avoid CRAB-defining events. Methods:We performed an initial retrospective analysis in 101 MM pts, followed by a prospective analysis in 176 pts using a structured MM-specific questionnaire, which was developed within our research group, Comprehensive Cancer Center (CCC) and in close collaboration with our biometrics team. The questionnaire consisted of 9 items and was tested and modified for utmost comprehensibility in 5 successive rounds in 10 healthy volunteers. For both analyses all available medical reports on the duration and type of the initial symptoms leading to the diagnosis of MM were assessed and putative risks for possible delays were determined. The subsequent prospective analysis was complemented with the MM-pt-questionnaire. It also assessed pts' satisfaction with the diagnostic process and inquired suggestions how this could possibly be improved. The trial protocol and MM-questionnaire were approved by the ethics committees. All pts provided written informed consent and all procedures were conducted in accordance with the Declaration of Helsinki and GCP Guidelines. Results:Both retro- and prospective analyses showed comparable pt characteristics with typical age and MM characteristics for tertiary/referral centers as previously described (Engelhardt et al. 2014, 2015, 2016). The median time from pts' first symptoms to the final MM diagnosis was 4 months (ms;0.5-120) in the retrospective and 6ms (0.5-60ms) in the prospective cohort, both analyses showing large variations of very prompt to extremely long diagnosis latencies. The frequency of pts with earlier vs. late diagnosis determination, defined in 4 groups as 12ms, was 38%, 28%, 14% and 20% within the retrospective and similar with 25%, 33%, 7% and 35% in the prospective analysis, respectively (Fig.1A). The prospectively assessed median time from the pts' first contact to a physician to the final MM diagnosis was 3.5ms (0-60ms). Actual CRAB-symptoms at diagnosis were perceived in 12%, 15%, 54% and 80% in the retrospective analysis, confirmed with 15%, 23%, 54% and 74% in the prospective analysis, respectively (Fig.1B). The definite diagnosis of MM was most often made by tertiary centers/hospitals in 81%, followed by outpts' hematologist/oncologist practices in 10% and rarer by general practitioners in 5% or nephrologists practices in 4%. Of interest, 61% of pts were completely or rather satisfied, whereas 39% were less satisfied with the diagnostic process (Fig.1C); 58% believed that their disease could have been diagnosed more expeditiously (Fig.1C). Those pts, who criticized a longer diagnosis latency vs. those that did not showed indeed a longer median time interval from symptom onset to the final MM diagnosis of 9 vs. 3ms, respectively. Risk factors for delays in diagnosis finding were light-chain-, oligo- or asecretory MM types, prior orthopedic and rheumatologic comorbidities disguising MM, unspecific disease symptoms, longer distance to referral centers and pts' residence in smaller rural areas. Conclusions:Considerable latencies in diagnosis finding of ≥12ms occur especially in MM pts suffering from unspecific symptoms like bone pain or anemia-related symptoms. Longer time intervals from the onset of first symptoms to the final MM diagnosis substantially affected pts' satisfaction and illness processing. In times of pt-centered care, this analysis aims to generate relevant insight, how earlier diagnosis can possibly be achieved in the future; these ongoing analyses being presented at the meeting. Figure Figure. Disclosures Engelhardt: Janssen: Research Funding; Celgene: Research Funding; Amgen: Research Funding; MSD: Research Funding.
- Published
- 2016
39. Testing Novel Anti - Multiple Myeloma (MM) Agents in a Suitable Three-Dimensional (3D) Co-Culture Platform
- Author
-
Monika Engelhardt, Manfred Jung, Johanna Senger, Dagmar Wider, Johannes M. Waldschmidt, Martin J. Hug, Justus Duyster, Ralph Wäsch, Gabriele Ihorst, Andreas R. Thomsen, and Stefan J. Müller
- Subjects
Auranofin ,Stromal cell ,medicine.diagnostic_test ,medicine.drug_class ,Bortezomib ,Immunology ,Histone deacetylase inhibitor ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Flow cytometry ,Cell biology ,chemistry.chemical_compound ,chemistry ,In vivo ,Cell culture ,Panobinostat ,medicine ,medicine.drug - Abstract
Introduction: In the past several years significant progress has been made in the understanding of MM pathogenesis, drug resistance and the related role of the bone marrow microenvironment (BMM). Hence various treatment strategies are currently in (pre-) clinical use, targeting both malignant plasma cells and the BM niche. In February 2015 the first epigenetic modifying pan histone deacetylase inhibitor (HDACi), panobinostat, granted approval for relapsed or refractory MM. However there is insufficient data so far to conclusively unravel the complex interactions between "epi-drugs" and the rapid increasing number of antimyeloma agents. Thus, we assess the impact of novel drug combinations (NOX-A12, auranofin, class selective HDACis, Sirt-2 inhibitors) on the BMM in a suitable matrix-based 3D co-culture platform to best mimic in vivo conditions whilst ensuring easy handling. Methods: The innovative 3D co-culture platform is based on transparent agarose matrices with 2 mm deep conical micro-cavities, wherein semi-adherent cell populations can be either seeded in contact or distance co-culture (Fig. 1). Due to the uniquely defined conical shape of the cavities, proliferation as well as treatment response could be monitored up to several weeks by determining the circular area of the cell clusters via transmitted light scanner. To further compare common flat 2D versus 3D culturing with regard to treatment response and growing behavior, FACS analysis and viability assays were performed. In addition, we examined altered expression levels of adhesion molecules (CXCR4, CD138, CD38, Slam-F7) in terms of culture conditions and epigenetic modulations (class-selective HDACis) using multi-panel flow cytometry. Qualitative and quantitative distribution of fluorescence dyes (apoptosis, proliferation) inside the cell agglomerations was visualized using mCherry transduced RPMI-8226 cells combined with confocal imaging and ScanR-analysis. Results: Human MM cell lines (MMCLs; U266 and RPMI-8226) exhibited substantial differences concerning growth and therapeutic responses under 2D versus agarose-based 3D culture conditions. MMCLs and especially primary MM patient samples could be cultured without reconditioning steps for more than three weeks under moderate logarithmic growth conditions inside the agarose matrices. Due to simple media and/or stromal exchange a loss or dying of cells could be consistently avoided. In contrast semi-adherent MMCLs (initial concentration 105 cells/ml) showed exponential proliferation in common flat culture until nutrient starvation induced apoptosis after approx. six days. Furthermore, malignant plasma cells became significantly resistant to proteasome inhibitors (PI) bortezomib (BTZ; 6nM, 48h) and auranofin (AUR; 3 µM, 48h) by forming solid 3D cell-agglomerations and almost remained unaffected if co-cultured additionally with HS-5 BM stromal cells (BMSCs). To exclude uneven distribution of agents, we determined the equal penetration of fluorescent dyes (CellTrace Violet/CFSE) into cell-clusters via confocal imaging. Further data suggest a great impact of 3D co-culture conditions on the expression pattern of modern target structures per se, leading e.g. to a decrease in CXCR4 but persistence of CD138 levels if compared to 2D mono-culture. Moreover we demonstrated a significant decrease of CD138 in low toxic concentrations of panobinostat (8nM, 48h) indicating a intriguing modulation of the BMM by epigenetic modifying agents. For subsequent steps we already verified synergistic antimyeloma effects of the novel HDAC6-selctive inhibitor JS28 in combination with BTZ and aim to further elucidate the influences of various selective deacetylase inhibitors on the BM niche. Conclusions and Ouline: Our recent data underline the importance of accurate 3D co-culture models to mimic in vivo proliferation and drug resistance and to predict later clinical treatment success more precisely. Based on this suitable platform, we are currently assessing the impact of cluster formation, stromal support and "epi-drugs" on the expression pattern of adhesion molecules to derive most rational drug combinations with modern target structures, such as CXCR4, CD138, CD38 and SlamF7 and to allow more effective targeting of the BMM. Disclosures Engelhardt: Janssen: Research Funding; MSD: Research Funding; Amgen: Research Funding; Celgene: Research Funding.
- Published
- 2016
40. Long-Term Follow-up of Patients with Corticosteroid-Refractory Graft-Versus-Host Disease Treated with Ruxolitinib
- Author
-
Ralph Wäsch, Joerg Halter, Reinhard Marks, Claudia Lengerke, Gabriele Ihorst, Nicolaus Kröger, Bruce R. Blazar, Stephan Mielke, Matyas Ecsedi, Francis Ayuk, Il-Kang Na, Jing Du, Friedrich Stölzel, Dietrich W. Beelen, Goetz Ulrich Grigoleit, Rainer Ordemann, Mats Brune, Markus Ditschkowski, Esther Schuler, Christof Scheid, Andreas Neubauer, Gerwin Huls, Ajib Salem, Jakob Passweg, Michael Lübbert, Omid Shah, Jürgen Finke, Robert S. Negrin, Gesine Bug, Andreas Burchert, Silvia Spoerl, Annette Schmitt-Graeff, Alexandros Spyridonidis, Robert Zeiser, Hartmut Bertz, Kristina Maas-Bauer, Petya Apostolova, Udo Holtick, Katja Sockel, Flore Sicre de Fontbrune, Walter J.F.M. van der Velden, Jürgen Kuball, Guido Kobbe, Christian Peschel, Renate Arnold, Justus Duyster, Penter Livius, S K Metzelder, Dominik Wolf, Gérard Socié, Everett Meyer, Ryan Flynn, Nikolas von Bubnoff, and Mareike Verbeek
- Subjects
medicine.medical_specialty ,Ruxolitinib ,Cytopenia ,Study drug ,business.industry ,medicine.drug_class ,Long term follow up ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Graft-versus-host disease ,Refractory ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Overall survival ,Corticosteroid ,business ,030215 immunology ,medicine.drug - Abstract
We have previously reported on the efficacy of the JAK1/2 inhibitor ruxolitinib in corticosteroid-refractory (SR) acute (a) and chronic (c) graft-versus-host disease (GVHD) in 95 patients (pts) (Leukemia 2015;29(10):2062-8). To assess long-term follow-up results, we collected data from the same pts treated in 19 centers in Europe and the US. Pts were classified as SR-aGVHD (n=54, all grade III or IV) or SR-cGvHD (n=41, all moderate or severe). Median numbers of pre-ruxolitinib GVHD treatment lines were 3 (1-7) and 3 (1-10) for SR-aGVHD and SR-cGvHD, respectively. The median follow-up was 19 and 24 months for aGVHD and cGVHD, respectively. The 1-year overall survival (OS) from was 62.4% (CI: 49.4%-75.4%) and 92.7% (CI: 84.7%-100%) for SR-aGVHD and SR-cGvHD, respectively. The estimated median OS (50% death) was 18 months for aGVHD and not reached for cGVHD patients. The median duration of ruxolitinib treatment was 5 and 10 months for patients with SR-aGVHD and SR-cGVHD, respectively reflecting the different biology of the diseases. At follow-up, 22/54 (41%) of SR-aGVHD patients and 10/41 (24%) of SR-cGVHD patients have an ongoing response and are free of any immunosuppression. GVHD relapse or progression after achieved PR/CR was observed in 14/45 (31%) and 13/36 (36%) patients with SR-aGVHD and SR-cGVHD, respectively. Response to re-treatment with Ruxolitinib or any immunosupressive therapy was seen in 11/14 (78%) and 11/13 (86%) patients with SR-aGVHD and SR-cGVHD, respectively. Cytopenia (any grade) and CMV-reactivation were observed during ruxolitinib-treatment in both SR-aGVHD (30/54, 55.6% and 18/54, 33.3%) and SR-cGVHD (7/41, 17.1% and 6/41, 14.6%) patients. These findings extend our previous report by showing that patients with SR-aGVHD and SR-cGVHD may benefit long-term from ruxolitinib treatment with an OS that is relatively high for steroid-refractory GVHD. GVHD-relapse or GVHD-progression rates were moderate and more than 75% of the relapse/progression patients responded to re-treatment with ruxolitinib or other immunosuppression. Disclosures Meyer: Stanford University: Patents & Royalties. Marks:Pfizer: Honoraria. Lübbert:Ratiopharm: Other: Study drug valproic acid; Celgene: Other: Travel Funding; Janssen-Cilag: Other: Travel Funding, Research Funding. Scheid:Novartis: Other: funding outside this work; Celgene: Other: funding outside this work; Janssen: Other: funding outside this work. Kobbe:Celgene: Honoraria, Other: travel support, Research Funding; Jansen: Honoraria, Other: travel support. Negrin:Stanford University: Patents & Royalties. Brune:Meda Pharma: Consultancy. Mielke:JAZZ Pharma: Speakers Bureau; Novartis: Consultancy; MSD: Consultancy, Other: Travel grants; Gilead: Other: Travel grants; Celgene: Other: Travel grants, Speakers Bureau. Kuball:Gadeta B.V,: Membership on an entity's Board of Directors or advisory committees. Kröger:Sanofi: Honoraria, Research Funding; Neovii: Honoraria, Research Funding; Riemser: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Peschel:MophoSys: Honoraria. von Bubnoff:BMS: Honoraria; Amgen: Honoraria; Novartis: Honoraria, Research Funding.
- Published
- 2016
41. The APC/C Coactivator Cdh1 Controls Self-Renewal and Differentiation of Human and Murine HSPCs
- Author
-
Daniel Ewerth, Anna Lena Illert, Monika Engelhardt, Marie Follo, Julia Felthaus, Justus Duyster, Dagmar Wider, Andrea Schmidts, Julia Schueler, Wäsch Ralph, and Stefanie Kreutmair
- Subjects
Cadherin ,medicine.medical_treatment ,Immunology ,Colony-Forming Units Assay ,Recombinant Granulocyte Colony-Stimulating Factor ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,CDH1 ,Cytokine ,Macrophage-1 antigen ,Coactivator ,medicine ,biology.protein ,Stem cell - Abstract
Introduction: The balance between differentiation and self-renewal in hematopoietic stem and progenitor cells (HSPCs) is crucial for homeostasis and lifelong blood cell production. Differentiation is predominantly initiated in the G1 phase of the cell cycle when the E3 ligase anaphase-promoting complex or cyclosome (APC/C) is highly active. Its coactivator Cdh1 determines substrate specificity and mediates proteasomal degradation. Relevant target proteins are associated with cell fate decisions in G1/G0, and there is growing evidence that Cdh1 is an important regulator of differentiation. While this has already been demonstrated in neurons, muscle cells or osteoblasts, little is known about the role of APC/CCdh1 in hematopoiesis. Here we report on the function of Cdh1 in human and murine HSPCs in vitro and in vivo. Methods: Human CD34+ cells from the peripheral blood of G-CSF mobilized donors were exposed to different cytokine combinations and gains or losses of surface marker expression during cell division were determined. By using the established culture conditions Cdh1 expression was detected in distinct hematopoietic lineages and developmental states. CD34+ cells were transduced with a lentivirus to deplete Cdh1 by stably expressing shRNA and was then used for in vitro differentiation in liquid culture or CFU assay. In a second miR-based RNAi approach murine BM cells were depleted of Cdh1 and used for competitive transplantation assays. Complementary xenotransplantation of human Cdh1-depleted CD34+cells was carried out with NSG mice. Results: The stimulation of freshly thawed CD34+ cells with cytokines led to cell cycle entry and proliferation. Self-renewing cells preserved CD34 expression for up to 7 cell divisions with a low proliferation rate. In contrast, during granulopoiesis and erythropoiesis cells divided more frequently with rapid down-regulation of CD34. Cdh1 expression was tightly connected to differentiation status and proliferation properties. In vitro cultured CD34+ cellsand those from BM of healthy human donors showed the highest Cdh1 level compared to moderate or low expression in lymphoid and myeloid cells. Cdh1 is highly expressed at the transcriptional and translational level during both self-renewal and also when cells were directed toward erythroid differentiation. Therefore, high Cdh1 expression is characteristic of immature hematopoietic cells and differentiating precursors. The knockdown of Cdh1 (Cdh1-kd) did not affect proliferation or viability as detected by CFSE staining and measuring the cell cycle length via live-cell imaging. However, Cdh1-kd cells showed a significant maintenance of CD34+ cells under self-renewal conditions and during erythropoiesis with a lower frequency of glycophorin A+ cells. The functional relevance of Cdh1 depletion was verified in CFU assays. Cells with Cdh1-kd formed fewer primary colonies but significantly more secondary colonies, indicating a preference for self-renewal over differentiation. After competitive transplantation Cdh1-depleted murine BM cells showed a significant enhancement in the repopulation of PB, BM and spleen at week 3, while there was no change in cell cycle properties. However, after 8 weeks chimerism in each of the compartments was reduced to that of the control cells. Accordingly, higher LK and LSK frequencies supported the engraftment of Cdh1-depleted cells at week 3, but there was a significant decrease at week 8 compared to control cells, suggestive of stem cell exhaustion. The Cdh1 level also affected cell differentiation in vivo. After 8 weeks the population of B cells (B220+) was increased in transplanted Cdh1-kd cells and the frequency of mature granulocytes (CD11b+ Gr1high) was reduced. Consistently, human Cdh1-depleted CD34+ cells engrafted to a much higher degree in the murine BM 8 and 12 weeks after xenotransplantation, as shown by a higher frequency of human CD45+ cells. Moreover, the increase of human CD19+ B cells with Cdh1-kd confirmed the results of the competitive transplantation. Conclusions: Loss of the APC/C coactivator Cdh1 supports repopulation of murine HSPCs after transplantation with a lymphoid-biased differentiation, and was confirmed in xenotranplantation experiments. In the long-term, Cdh1 loss led to exhaustion of primitive LK and LSK population, highlighting the role of Cdh1 as a critical regulator of HSPC self-renewal and differentiation. Disclosures Engelhardt: Janssen: Research Funding; Amgen: Research Funding; MSD: Research Funding; Celgene: Research Funding.
- Published
- 2016
42. Functional Geriatric Assessment (F-GA) in Multiple Myeloma Patients: Results from a Prospective Multicenter Study Group (DSMM) Trial and Changes from Baseline to Follow-up Assessment
- Author
-
Stefan J. Müller, Wolfram Pönisch, Justus Duyster, Stefan Knop, Christian Langer, Ralph Waesch, Monika Engelhardt, Gabriele Ihorst, Sophia Scheubeck, Sandra Maria Dold, Alexander Zober, Martin Schumacher, and Lars-Olof Mügge
- Subjects
medicine.medical_specialty ,Activities of daily living ,Performance status ,business.industry ,Immunology ,Cell Biology ,Hematology ,Timed Up and Go test ,Biochemistry ,Pulmonary function testing ,Transplantation ,Multicenter trial ,Internal medicine ,Cohort ,Medicine ,business ,Depression (differential diagnoses) - Abstract
Introduction: Due to continuous research and novel agents, today´s treatment choices for MM patients (pts) are numerous. In order to provide best tolerable and adjusted treatment, it is desirable to objectively assess individuals' biological fitness and comorbidities (CM), rather than using pts' chronological age alone (Bron et al. Haematologica 2016). Therefore, it is necessary to define new functional and geriatric test tools that are suitable to rate CM and therapy-related risks. Methods: This prospective multicenter functional geriatric assessment (F-GA) was attentively performed in newly diagnosed (ND) MM pts within German DSMM study centers prior to initiation of antimyeloma treatment, which reflected pts´ baseline health status. Moreover, a follow-up F-GA was conducted ~12 months after this baseline assessment. The F-GA included: 1. rating of fitness by a) physicians and b) pts, 2.Karnofsky Performance Status (KPS), 3.pain scale, 4.Instrumental activity of daily living (IADL), 5.Activity of daily living (ADL), 6.SF12-QoL questionnaire, 7.malnutrition, 8.Mini-Mental status (MMS), 9.geriatric depression scale (GDS) and 10. Timed Up and Go Test (TUGT). In addition, established CM scores were determined: a) Initial-Myeloma Comorbidity Index (I-MCI) and b) Revised-MCI (R-MCI), c) International Myeloma working group (IMWG) score, d) Charlson Comorbidity Index (CCI), e) Hematopoietic Cell Transplantation Specific-Comorbidity Index (HCT-CI), f) Kaplan Feinstein (KF), g) eGFR/ß2MG. The trial protocol was approved by the ethics committees. All pts provided written informed consent and all procedures were conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization and GCP Guidelines. Results: A total of 289 newly diagnosed MM pts of all age groups have already been included in this prospective multicenter trial. Characteristics of pts were typical for tertiary centers with a median age of 62 years (range: 27-85), hemoglobin of 11.0g/dl (5-17), eGFR of 70ml/min/1.73qm (7-163), ß2-MG of 3 mg/l (1-38) and bone marrow plasma cell infiltration of 40% (0-100). Pts estimated their fitness with a median of 3 (1-6) and pain level of 2 (0-10). In line, their frailty assessment revealed a median KPS of 80% (30-100) and TUGT of 10 (4-80). Median functional results were for the ADL 5 (2-6), for the IADL 8 (1-8) and for malnutrition 3 (0-14). The MMS revealed only a slight cognitive deficiency of 28 (15-30) and GDS of 2 (0-13). CM scores showed a median I-MCI of 0 (0-3), R-MCI of 4 (0-9), IMWG of 1 (0-4), CCI of 2 (0-8), HCT-CI of 2 (0-9), KF of 1 (0-3), eGFR/ß2MG score of 1 (0-2). Most relevant F-GA-tools for PFS and OS - according to our current results - seem to be the TUGT, R-MCI and IMWG scores. Those 122 pts that have already received both baseline (T0) and follow-up assessments (T1) showed notable and significant improvement in some of these tests (Tab. 1), e.g. ADL, R-MCI, IMWG, CCI, HCT-CI scores, whereas others, such as the IADL and KF remained almost constant. Pain, depression and malnutrition also improved, albeit to as yet insignificant extends. For 66 pts the second assessment has to be done within the next month, 60 patients died since the start of this trial, 40 of those within the first 12 months and 85 pts dropped out without a second assessment. Conclusion: To the best of our knowledge, this is the first extensive prospective, multicenter F-GA that determines most valuable disease risks, functional tests and CM scores in MM pts. Our results suggest that relevant parameters in MM pts are the KPS, fitness rating by physicians and pts, ADL, TUGT, frailty, malnutrition, pain due to osteolyses, and renal function, whereas the IADL, MMS, GDS and lung function seem less revealing. Most predictive test tools are the R-MCI- and IMWG-scores that distinguish fit, intermediate-fit and frail pts most precisely. The results of our follow-up-assessment demonstrate that pts´ health status may significantly improve upon treatment. We continue this F-GA in an even larger prospective multicenter cohort to consolidate these results which will be presented at the meeting. Disclosures Langer: Celgene: Consultancy; Takeda: Consultancy; Janssen: Consultancy; Amgen: Consultancy; Novartis: Consultancy. Knop:Takeda: Consultancy. Engelhardt:Janssen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Other: Travel/Accommodation/Expenses , Research Funding; MSD: Consultancy, Honoraria.
- Published
- 2016
43. The Pan-PIM Kinase Inhibitor LGB321 Affects Apoptotic Pathways and Microenvironmental Interactions in CLL
- Author
-
Sarah Decker, Katja Zirlik, Konrad Aumann, Thorsten Zenz, Sandra Kissel, Rainer Claus, Justus Duyster, and Christine Dierks
- Subjects
0301 basic medicine ,Stromal cell ,Kinase ,Immunology ,PIM1 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 ,Transplantation ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,In vivo ,hemic and lymphatic diseases ,Ibrutinib ,Cancer research ,medicine ,Bone marrow - Abstract
Introduction The emergence of kinase inhibitors like Ibrutinib has drastically altered treatment strategies and improved outcomes in CLL patients, but lack of cure and resistance to therapy still remain serious problems. The three PIM kinases are involved in various important disease mechanisms in CLL, with PIM1 regulating CXCR4 surface expression impacting its interaction with the microenvironment, and PIM2/3 affecting the apoptotic machinery by regulating BAD. The Pan-PIM kinase inhibitor LGB321 (Novartis) targets all three PIM kinases and therefore affects both, CLL apoposis and its interaction with the microenvironment. Results In the study presented here, we investigated the effect of the Pan-PIM kinase inhibitor LGB321 on CLL in vitro and in vivo. LGB321 was highly effective in inducing apoptosis in primary human CLL cells, independent of risk factors or the mutation status. Apoptosis induction correlated with reduced pBAD and BAD levels. LGB321 was also effective in the presence of protective stromal cells and could completely overcome the stroma protective effects. Furthermore, we found that Pan-PIM inhibitor treatment blocked the CXCR4/CXCL12 axis by dephosphorylating the CXCR4 receptor on Ser339, by reducing total CXCR4 protein levels, and by blocking the externalization of the CXCR4 receptor. Concordantly, LGB321 blocked CXCR4 functions like migration towards a CXCL12 gradient (P Conclusion Our results demonstrate, that the Pan-PIM kinase inhibitor LGB321 might be an effective treatment option for CLL patients by impairing PIM2/3 mediated CLL-cell survival, and by blocking the PIM1/CXCR4-mediated interaction of CLL cells with their protective microenvironment in vitro and in vivo. Future clinical trials should be performed to validate its efficacy in human CLL. Disclosures Claus: Roche: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Janssen-Cilag: Consultancy, Honoraria, Other: Travel Funding.
- Published
- 2016
44. PIM1 Inhibition Enhances and Prolongs Plerixafor-Induced Mobilization of HSCs
- Author
-
Sandra Kissel, Ralph Wäsch, Tony Andreas Müller, Claudius Klein, Anabel Zwick, Marie Follo, Anna Lena Illert, Sarah Decker, Justus Duyster, Claudia Waskow, Christine Dierks, Benjamin Rister, and Alina Rudorf
- Subjects
Stromal cell ,Plerixafor ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CXCR4 ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,biology.protein ,Stromal cell-derived factor 1 ,Bone marrow ,Stem cell ,medicine.drug - Abstract
Introduction The CXCL12/CXCR4 axis was shown to be a major regulator for the interaction of hematopoietic stem cells (HSCs) with the niche and interruption of this pathway mobilizes HSCs from the bone marrow (BM).Therefore CXCR4 antagonists like AMD3100 (Plerixafor) are used in the clinic for the collection of HSCs from patients who fail to mobilize HSCs in response to G-CSF.In our previous studies, we could show that the serine/threonine kinase PIM1 is regulating the surface expression of the CXCR4 receptor on HSCs. In addition, PIM1-deficient HSCs fail to home to a wild type (WT) BM niche. Based on these results, we aimed to improve HSC mobilization by combining CXCR4 and PIM inhibition. Methods In order to study the mobilization efficiency in the murine model, mice were treated with AMD3100 alone or in combination with LGB321, a novel pan-PIM inhibitor. The mice were sacrificed at various timepoints and peripheral blood (PB) was isolated. The percentage of HSCs was then determined by flow cytometry. The mechanism of HSC mobilization was studied in isolated HSC and stroma populations by analyzing mRNA levels and surface expression of CXCR4, its ligand CXCL-12 and other factors, which are crucial for the interaction of HSCs and stromal cells. Results We found that CXCR4 inhibition using AMD3100 leads to a compensatory upregulation of CXCR4 surface expression on total BM cells as well as HSCs. This effect can be reverted by deficiency or inhibition of PIM1 (Figure 1A). As a consequence, HSC mobilization using AMD3100 is strongly enhanced and prolonged in Pim1-deficient mice compared to WT animals (Figure 1B). Likewise, treatment of WT animals with AMD3100 in combination with LGB321 leads to increased and prolonged HSC mobilization compared to animals treated only with AMD3100 (Figure 1C). Besides the downregulation of CXCR4 on HSCs, we found that the Cxcl-12 expression as well as CXCR4 surface expression in CXCL12-abundant reticular (CAR) cells is dramatically decreased in Pim1-deficient mice, which even further increases the mobilization capacity of Pim1-deficient mice. Conclusion Our findings indicate, that PIM1 inhibition counteracts the compensatory upregulation of the CXCR4 receptor on HSCs after Plerixafor treatment and decreases CXCL12 levels within the bone marrow niche. Therefore, targeting PIM kinases in combination with CXCR4 inhibition could improve the collection of stem cells in patients at risk for poor mobilization. Figure 1. PIM1-deficiency or -inhibition enhances HSC mobilization in combination with AMD3100-treatment. (A) Statistical analysis of CXCR4 surface expression on Pim1-/- or WT LSK cells after AMD3100-treatment for 24h. PIM1-deficiency reverts the increased CXCR4 surface expression after AMD3100-treatment. (B) Time course of total LSK cells per ml PB of AMD3100-treated mice. Pim1-/- mice show significantly higher LSK counts. (C) Statistical analysis of total LSK cells per ml of LGB321+AMD3100 treated mice compared to AMD3100-only treated WT animals. PIM-inhibition significantly increases mobilization efficiency of AMD3100. Figure 1. PIM1-deficiency or -inhibition enhances HSC mobilization in combination with AMD3100-treatment. (A) Statistical analysis of CXCR4 surface expression on Pim1-/- or WT LSK cells after AMD3100-treatment for 24h. PIM1-deficiency reverts the increased CXCR4 surface expression after AMD3100-treatment. (B) Time course of total LSK cells per ml PB of AMD3100-treated mice. Pim1-/- mice show significantly higher LSK counts. (C) Statistical analysis of total LSK cells per ml of LGB321+AMD3100 treated mice compared to AMD3100-only treated WT animals. PIM-inhibition significantly increases mobilization efficiency of AMD3100. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
45. Impact of Distinct Genetic and Epigenetic Aberrations on Survival and Response in Acute Myeloid Leukemia Patients Receiving Epigenetic Therapy
- Author
-
Jan K. Hiller, Arzu Yalcin, Verena I. Gaidzik, Michael Lübbert, Lars Bullinger, Mahmoud Abdelkarim, Claudia Schmoor, Charlotte Schmidt-Salzmann, Konstanze Döhner, Nadja Blagitko-Dorfs, Björn Hackanson, and Justus Duyster
- Subjects
Oncology ,medicine.medical_specialty ,NPM1 ,Performance status ,business.industry ,Immunology ,Hazard ratio ,Decitabine ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Leukemia ,Internal medicine ,DNA methylation ,medicine ,business ,Epigenetic therapy ,medicine.drug - Abstract
Background: Treatment with hypomethylating agents such as decitabine, which results in complete and partial remission rates of up to 50%, has become standard of care in older patients with acute myeloid leukemia (AML) who are no candidates for intensive chemotherapy. While clinical parameters such as reduced performance status, high leukocyte counts, elevated lactate dehydrogenase (LDH) levels as well as poor-risk cytogenetics are associated with lower response rates and shorter overall survival, there exists only limited data on molecular biomarkers that enables for selection of AML patients who are likely to benefit from epigenetic therapy. Shen et al. (JCO 2010) could show that distinct DNA methylation changes are prognostic for overall survival in myelodysplastic syndrome patients, however, response to decitabine therapy could not be predicted. Additionally, mutations in the epigenetic-modifier gene DNMT3A were recently reported to be associated with response and survival in AML patients treated with hypomethylating agents, however, while some of these studies revealed a positive association with outcome, other studies showed no association with response and overall survival (Metzeler et al., Leukemia 2012, DiNardo et al., Leuk Lymph 2014, Coombs et al., Blood 2015). Results: In order to contribute further knowledge regarding prognosis and response to hypomethylating therapy in AML patients, we investigated distinct genetic (FLT3-ITD, NPM1, DNMT3A) and epigenetic (estrogen receptor alpha (ERα), C/EBPα and OLIG2) aberrations in 87 AML patients from the recently published phase II decitabine trial (AML00331, Lübbert et al. Haematologica 2012) to identify potential molecular biomarkers. While FLT3-ITD and NPM1 mutational status were not associated with survival or response to therapy, in our cohort patients harboring DNMT3A R882 mutations showed shorter overall survival (hazard ratio (HR): 2.15, 95%-confidence interval (CI): 0.91-5.12, p=0.08). Promoter DNA methylation analyses using pyrosequencing revealed shorter overall survival of patients with higher levels of methylation of ERα (HR: 1.50, CI: 0.97-2.32, p=0.07) and OLIG2 CpG4 (HR: 1.52, CI: 0.96-2.41, p=0.08), while DNA methylation of C/EBPα showed no association with outcome. Importantly, in multivariate analyses adjusted for clinical baseline parameters, the impact of ERα and OLIG2 CpG4 methylation was conserved (HR: 1.74, CI: 1.03-2.96, p=0.04 and HR: 1.61, CI: 0.96-2.71, p=0.07, respectively) whereas the effect of DNMT3A R882 mutations was reduced to HR: 1.94 (CI: 0.79-4.73, p=0.15). In contrast, none of the investigated molecular markers was associated with response to treatment. Conclusion: In addition to the well established adverse prognostic clinical parameters such as patients' age, reduced performance status and elevated LDH levels, we provide further evidence that pretreatment genetic and especially epigenetic analyses including DNMT3A R882 mutation status, as well as ERα and OLIG2 CpG4 DNA methylation status may be potential molecular biomarkers in AML patients undergoing epigenetic therapy. Disclosures Lübbert: Celgene: Other: Travel Funding; Ratiopharm: Other: Study drug valproic acid; Janssen-Cilag: Other: Travel Funding, Research Funding.
- Published
- 2016
46. PIM1-Mediated CXCR4 Phosphorylation: A Potentially Class-Distinct Therapeutic Target of Next Generation Proteasome Inhibitors in Multiple Myeloma
- Author
-
Dagmar Wider, Stefan J. Müller, Monika Engelhardt, Ralph Wäsch, Marie Follo, Johannes M. Waldschmidt, Anna Simon, Sarah Decker, and Justus Duyster
- Subjects
Bortezomib ,Chemistry ,Kinase ,Immunology ,Cell Biology ,Hematology ,Pomalidomide ,medicine.disease ,Biochemistry ,Carfilzomib ,Ixazomib ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Proteasome inhibitor ,medicine ,Cancer research ,Propidium iodide ,Multiple myeloma ,medicine.drug - Abstract
Introduction: The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment is fundamental to MM pathogenesis. Cell adhesion-mediated drug resistance (CAM-DR) is regulated by adhesion receptors on MM cells such as CXCR4, CXCR7, CD49d and CD44. We and others have previously reported that CAM-DR towards drugs like bortezomib, pomalidomide or vorinostat may be dissolved by combining these novel agents with the CXCR4 inhibitor plerixafor. Different than expected, additional treatment with plerixafor in corresponding experiment however did not rescue the cytotoxic effects of the second generation proteasome inhibitor carfilzomib. We hypothesized that carfilzomib itself interferes with the CXCR4-CXCL12 axis in myeloma. Prior reports in AML and CLL indicate that PIM1-mediated CXCR4 phosphorylation at the position S339 is an essential step for CXCR4 recirculation to the cell surface and its function as CXCL12 receptor (Grundler et al. 2009, Decker et al. 2014). In this project, we therefore examined the effects of carfilzomib on the PIM1-CXCR4 axis as a not yet described, potentially class-distinct mechanism of action of this second generation proteasome inhibitor. Methods: U266, RPMI-8226, L363, MOLT-4, NCI-H929 and the stromal cell line M2-10B4 were utilized. Bortezomib (1, 10, 20, 50, 100nM), carfilzomib (20, 50, 100nM) and plerixafor (10, 50, 100μM) were used based on previous studies and are well comparable to clinically relevant doses. CXCL12 stimulation was performed with human recombinant CXCL12 (30nM). For combination studies, cells were preincubated with plerixafor (50µM). Viability was quantified by propidium iodide and annexinV-FITC using flow cytometry. For quantitative real-time PCR and Western blots, U266 monocultured cells were treated with a carfilzomib pulse (t=1h), were allowed to recover for 20 hours, starved for 4 hours and stimulated with CXCL12 for 15 minutes (n=4). PIM-1 mRNA transcript levels were assessed in U266 control vs. U266 treated with a carfilzomib pulse (100nM, t=1h) by qPCR. Data was analyzed according to the "delta-delta-CT method" based on the relative expression of PIM-1 vs. GAPDH. Results were normalized to the mean of the control samples. Results: FACS analyses determined a substantial decrease of CD138 and CXCR4 surface expression in a dose-dependent manner after 1h carfilzomib treatment of U266 cells. Further assessment of downstream signaling revealed that carfilzomib treatment significantly reduces CXCR4 phosphorylation at S339 without changing total levels of CXCR4 (Figure A) or total levels of ERK or pERK (not shown), excluding a general inhibition of phosphorylation or protein synthesis by carfilzomib. Following the hypothesis that CXCR4 is potentially phosphorylated by PIM1 kinase, we assessed the impact of carfilzomib on PIM-1 protein levels: PIM-1 kinase protein was significantly reduced in a dose-dependent manner along with the levels of pCXCR4 in response to increasing doses of carfilzomib (0-100nM, Figure B). To further investigate a possible direct interference at the mRNA level, we evaluated PIM-1 mRNA levels after 1h carfilzomib, confirming substantially reduced PIM-1 RNA transcripts (Figure C). Different from carfilzomib and in line with prior observations (Shay et al. 2005), bortezomib was shown to increase protein levels of PIM-1 (data not shown). Side-by-side comparative assays of bortezomib vs. carfilzomib in terms of reduced CXCR4 expression, decreased CXCR4 phosphorylation and PIM-1 levels on mRNA and protein level are currently ongoing and will be presented at the meeting. Conclusions: Similar to previous reports on ixazomib reducing PIM-1 on protein and mRNA levels by inhibiting the tumor-suppressive microRNA miR33b (Tian et al. 2012), this work provides a potentially distinct mechanism of action of the second generation proteasome inhibitor carfilzomib on the PIM1-CXCR4 axis and identifies PIM-1 as a valid target to overcome CAM-DR in multiple myeloma. Figure Carfilzomib overcomes stroma protection due to PIM-1 kinase inhibition. Figure. Carfilzomib overcomes stroma protection due to PIM-1 kinase inhibition. Disclosures Engelhardt: Janssen: Research Funding; Amgen: Research Funding; MSD: Research Funding; Celgene: Research Funding.
- Published
- 2016
47. Identification of a Novel Chromosomal Translocation t(11;16)(q23;q22) Fusing MLL to Enhancer of mRNA Decapping (EDC)-4 in Smoldering Acute Myeloid Leukemia
- Author
-
Michael Lübbert, Michael L. Cleary, Tobias Ma, Milena Pantic, Heiko Becker, Keisuke Kataoka, Justus Duyster, Gabriele Greve, Julia Schüler, Sabine Bleul, Seishi Ogawa, and Jesús Duque Afonso
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Immunology ,Decitabine ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Transplantation ,Leukemia ,Exon ,hemic and lymphatic diseases ,medicine ,Exome ,Exome sequencing ,medicine.drug - Abstract
Background: Translocations involving the MLL gene located on chromosome 11q23 are usually found in de novo acute myeloid leukemia (AML) and generally confer a dismal prognosis unless the AF9 gene is involved (Döhner et al., Blood 2010;115:453-74). MLL can be fused to multiple different genes, resulting in the large and growing "MLL recombinome" (Meyer et al., Leukemia 2013;27:2165-76). Thus far, only two genes encoding proteins that are part of the mRNA decapping protein complex (i.e., DCP1A, DCPS) have been described as rarely being fused to MLL. Here, we describe an AML with an indolent disease course arising from myelodysplastic syndrome (MDS) that disclosed a unique MLL fusion with another component of the mRNA decapping complex, i.e., EDC4. Patient and Methods: Briefly, a 55 year-old female patient presented with an MDS [timepoint (t) -1] that within 10 months progressed to an AML with 2% blood and 40% bone marrow myeloblasts (t0). The patient refused treatment beyond supportive care. Six months later, a marked blast expansion of 80% was detectable in the blood (t1). The patient received 5 cycles of decitabine (t2, cycle 5), followed by 3 months of hydroxyurea (t3). Samples were depleted from CD3+ cells via MACS; CD3+ cells served as germline control. RNA sequencing libraries were prepared using the NEBNext Ultra RNA Library Prep Kit for Illumina (New England Biolabs), and the SureSelect Human All Exon v5 kit (Agilent Technologies) was used for exome capturing from gDNA. Next generation sequencing was performed on an Illumina Hiseq 2500. The analyses were performed as previously described (Kataoka et al. Nature Genetics 2015;47:1304-15, Becker et al. Blood 2014;123:1883-6). NOD scid gamma mice were used as hosts for patient derived tumor xenografts (PDX). Results: Standard metaphase cytogenetics at the diagnosis of AML (t0) revealed a previously undescribed translocation involving the MLL gene, i.e., t(11;16)(q23;q22), as the sole cytogenetic abnormality. The unknown fusion partner on chromosome 16 was identified by RNA-sequencing as the EDC4 gene (a.k.a. Ge-1), which encodes a key scaffold protein of the mRNA decapping complex; the fusion was confirmed by PCR on cDNA. The translocation led to the in-frame fusion of MLL exon 13 to EDC4 exon 6 which was linked by 19 nucleotides from EDC4 intron 5. The predicted amino acid sequence of the linker was ALNTLLR. Further analyses including exome sequencing on the samples collected over the disease course demonstrated STAG2 as a potential founder mutation that was already present during the MDS (t-1) and persisted throughout the disease course at variant allele frequencies (VAFs) of approximately 45-50%. At the time of transformation to AML (t0), the MLL-EDC4 and two RAS mutations (KRAS p.G13D, NRAS p.G12C) were detectable. Towards the terminal phase (t3), the RAS mutations disappeared and a clone that acquired a mutation in the FLT3 tyrosine kinase domain (TKD; p.D835V; VAF 43%) expanded. Primary blasts from the patient engrafted in NOD scid gamma mice and established a stable PDX serial transplantation mouse model used for drug testing. Conclusions: This report provides the first demonstration of an MLL-EDC4 in-frame fusion, with potential cooperativity with a founder mutation in the STAG2 splicing factor gene during the transformation of MDS to AML and the additional acquisition of a FLT3-TKD mutation during disease progression. RNA sequencing proved to be a very feasible approach to identify novel fusion partners of known oncogenes such as MLL. Disclosures Becker: BMS: Honoraria; Novartis: Honoraria. Kataoka:Yakult: Honoraria; Boehringer Ingelheim: Honoraria; Kyowa Hakko Kirin: Honoraria. Schüler:Oncotest GmbH: Employment. Ogawa:Kan research institute: Consultancy, Research Funding; Sumitomo Dainippon Pharma: Research Funding; Takeda Pharmaceuticals: Consultancy, Research Funding. Lübbert:Janssen-Cilag: Other: Travel Funding, Research Funding; Celgene: Other: Travel Funding; Ratiopharm: Other: Study drug valproic acid.
- Published
- 2016
48. Implementation of a Molecular Tumor Board in Clinical Decision Making at the Medical Center University of Freiburg
- Author
-
Silke Lassmann, Nikolas von Bubnoff, Melanie Börries, Volker Brass, Lisa Lutz, Tilman Brummer, Philipp Demmer, Simone Hettmer, Martin Werner, Christoph Peters, Hauke Busch, Rainer Claus, Justus Duyster, Julius Wehrle, Cornelius Miething, Britta Weddeling, Leman Mehmed, Ralph Fritsch, Oliver Oehlke, Agnes Csanadi, Frank Meiss, and Thalia Erbes
- Subjects
medicine.medical_specialty ,Hematology ,business.industry ,Standard treatment ,Immunology ,Cancer ,Cell Biology ,Disease ,medicine.disease ,Off-label use ,Biochemistry ,Clinical decision making ,Internal medicine ,medicine ,Tumor board ,business ,Exome - Abstract
Introduction: In-depth knowledge about molecular pathogenesis of malignant diseases and rapidly increasing availability of targeted treatment options enables molecularly guided decision-making. We have established a Molecular Tumor Board (MTB) that focuses on patient management based on specific molecular data at the individual patient level. Methods: The MTB has its main focus on hematologic and solid neoplasias progressing during standard treatment, on rare entities and on patients with treatment resistance. The biweekly MTB supports the work of organ-specific boards and external cooperation partners. The MTB multidisciplinary team consists of expert physicians from Hematology, Medical Oncology, Gynecology, Dermatology, Pediatrics and Radiation Oncology as well as Pathology, Molecular biology, Computational Biology and Genetics. Diagnostic and therapeutic recommendations are based on customized diagnostics and a case-by-case literature review. Recommendations are communicated back to the treating physician. Results: In the first year after implementation of the MTB, a total of 92 pts have been discussed in 155 case discussions during 25 MTB meetings. Referred patient cases covered the entire range of malignancies seen by the organ-specific boards including hematologic malignancies. 132 diagnostic recommendations were made in 80/92 (87%) pts, including IHC, ISH or panel sequencing with diagnostic reporting (n=96/72 pts) and exome, genome, transcriptome and/or methylome analysis (n=24/22 pts.). 43 treatment recommendations were made in 39/92 (42%) pts with an implementation rate of 47% (20/43 recommendations in 19/39 pts). Treatment recommendations mainly comprised off-label antibody and tyrosine kinase inhibitor (TKI) therapy (40%) and trial inclusions (28%). Major reasons for non-adherence to recommendations included patient will, death of pts and medical reasons. Objective responses were observed in 5/19 (26%) pts to TKI in- and off-label and antibody off-label treatments. Disease stabilization was achieved in 3/19 (16%) pts. Specifically, the use of PD-(L)1 inhibiting antibodies was suggested in 13 cases (11 off-label) and implemented in 6 cases. Here, 2/6 pts responded or exhibited stable disease upon PD-(L1) blockage. Conclusion: Implementation of a Molecular Tumor Board serves as an interdisciplinary platform for integrating comprehensive molecular data sets as predictive biomarkers in molecular guided, individualized patient care. Our experience demonstrates that individualized treatment recommendation is feasible and effective for a substantial proportion of patients in challenging clinical situations. Disclosures Claus: Roche: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Janssen-Cilag: Consultancy, Honoraria, Other: Travel Funding.
- Published
- 2016
49. The Microenvironmental Stromal Cells Abrogate NF-κb Inhibitor Induced Apoptosis in Chronic Lymphocytic Leumkemia
- Author
-
Katharina Foerster, Katja Zirlik, Carl Philipp Simon-Gabriel, Dorothee Bleckmann, Marco Benkisser-Petersen, Nicolas Thornton, Rainer Claus, and Justus Duyster
- Subjects
Stromal cell ,CD40 ,medicine.diagnostic_test ,biology ,Chemistry ,Kinase ,Immunology ,Cell Biology ,Hematology ,Transfection ,Biochemistry ,Flow cytometry ,Apoptosis ,medicine ,Cancer research ,biology.protein ,Viability assay ,B-cell activating factor - Abstract
Introduction: In recent years, the emergence of kinase inhibitors has drastically altered treatment strategies and improved outcomes in CLL patients, but lack of cure and resistance to therapy still remain serious issues. The transcription factor NF-κB influences several cellular functions such as proliferation, apoptosis and inflammation and is known as a key factor contributing to CLL development and progression. NF-κB is constitutively active in CLL and the NF-κB subunit RELA has been proposed as a prognostic marker in CLL with high RELA DNA-binding activity being predictive of short time to first treatment and overall survival. Therefore, NF-κB has gained attention as a promising therapeutic target. NF-kB inhibition induces apoptosis in CLL cells in vitro. However, whether this effect pertains in vivoremains unclear. Since the microenvironment is crucial for CLL cell viability circumventing apoptosis, we tested whether NF-κB inhibition modulates CLL viability in the presence of the microenvironment. Methods: The specific NF-κB inhibitor Dehydroxymethylepoxyquinomicin (DHMEQ) was used alone (2-5 µg/ml) or combined with fludarabine (10 µM), rhBAFF (50 ng/ml), rhAPRIL (500 ng/ml), rhSDF-1a (100 ng/ml) or CD40 ligand (1 µg/ml) on primary CLL cells cultured alone (monoculture) or on bone marrow stromal cells (BMSC) (co-culture with a ratio of 20 CLL cells per stromal cell) for 48-144 h. Viability and apoptosis were measured by flow cytometry using AnnexinV/PI stainings. Protein expression was analyzed by western blot using standard protocols. NF-κB DNA-binding activity after DHMEQ treatment (5 µg/ml) for 6 h was measured by ELISA for all subunits using 1 µg of protein lysate for the NF-κB1 subunit and 10 µg protein lysate for the subunits RELA, NF-κB2, RELB and c-REL. RELA gene knockdown was performed by siRNA transfection (2 µM targeting and non-targeting siRNA). Results: NF-κB inhibition using DHMEQ led to apoptosis in monocultured CLL cells (viability 74% vs. 24%, n=17, p Conclusion: NF-κB inhibition in primary CLL cells shows great discrepancy between in vitro and in vivo scenarios. While DHMEQ treatment leads to apoptosis in mono-cultured cells by BAX upregulation and increased PARP cleavage, CLL cell viability is not affected in the presence of microenvironment, suggesting that the NF-κB pathway can be bypassed in vivo. Soluble ligands, especially BAFF, appear to be involved in mediating this protective effect. However, the combination of NF-κB inhibition with standard chemotherapy might represent a promising approach and warrants further clinical assessment. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
50. Nucleophosmin–anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic signaling pathway
- Author
-
Cornelius Miething, Stephan W. Morris, Christian Peschel, Justus Duyster, Ren Yuan Bai, and Tao Ouyang
- Subjects
integumentary system ,Akt/PKB signaling pathway ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Wortmannin ,chemistry.chemical_compound ,chemistry ,hemic and lymphatic diseases ,Cancer research ,medicine ,Anaplastic lymphoma kinase ,Signal transduction ,Protein kinase A ,Anaplastic large-cell lymphoma ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
More than half of anaplastic large-cell lymphomas (ALCLs) have a chromosomal translocation t(2;5) that leads to the expression of a hybrid protein composed of the nucleolar phosphoprotein nucleophosmin (NPM) and the anaplastic lymphoma kinase (ALK) that exhibits an unregulated tyrosine kinase activity. We have previously identified PLC-γ as a crucial downstream signaling molecule of NPM-ALK that contributes to its mitogenic potential. Here, we show that NPM-ALK recruits the C-terminal SH2 domain of the phosphatidylinositol 3-kinase (PI 3kinase) p85 subunit. PI 3-kinase assays revealed that the kinase is activated by NPM-ALK in vivo, in turn activating PKB/Akt in NPM-ALK–expressing cells. The use of 2 specific PI 3-kinase inhibitors, wortmannin and LY294002, demonstrated the requirement of PI 3-kinase for the growth of NPM-ALK–transformed cell lines, as well as a cell line established from a patient with ALCL. Primary murine bone marrow retrovirally transduced with NPM-ALK showed a transformed phenotype that was reversible on treatment with PI 3-kinase inhibitors. Flow cytometric analysis revealed that wortmannin-treated NPM-ALK–transformed cell lines underwent apoptosis. Furthermore, apoptosis induced by overexpression of the proapoptotic molecule Bad could be partially blocked by the overexpression of NPM-ALK. Thus, NPM-ALK activates the antiapoptotic PI 3-kinase/Akt pathway, which likely contributes to the molecular pathogenesis of ALCL.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.