Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Leukemia ; 34(7): 1775-1786, 2020 07.
Article in English | MEDLINE | ID: mdl-31925317

ABSTRACT

In chronic-phase chronic myeloid leukaemia (CP-CML), residual BCR-ABL1+ leukaemia stem cells are responsible for disease persistence despite TKI. Based on in vitro data, CHOICES (CHlorOquine and Imatinib Combination to Eliminate Stem cells) was an international, randomised phase II trial designed to study the safety and efficacy of imatinib (IM) and hydroxychloroquine (HCQ) compared with IM alone in CP-CML patients in major cytogenetic remission with residual disease detectable by qPCR. Sixty-two patients were randomly assigned to either arm. Treatment 'successes' was the primary end point, defined as ≥0.5 log reduction in 12-month qPCR level from trial entry. Selected secondary study end points were 24-month treatment 'successes', molecular response and progression at 12 and 24 months, comparison of IM levels, and achievement of blood HCQ levels >2000 ng/ml. At 12 months, there was no difference in 'success' rate (p = 0.58); MMR was achieved in 80% (IM) vs 92% (IM/HCQ) (p = 0.21). At 24 months, the 'success' rate was 20.8% higher with IM/HCQ (p = 0.059). No patients progressed. Seventeen serious adverse events, including four serious adverse reactions, were reported; diarrhoea occurred more frequently with combination. IM/HCQ is tolerable in CP-CML, with modest improvement in qPCR levels at 12 and 24 months, suggesting autophagy inhibition maybe of clinical value in CP-CML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytogenetic Analysis/methods , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Aged , Female , Follow-Up Studies , Humans , Hydroxychloroquine/administration & dosage , Imatinib Mesylate/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate
2.
Pathologe ; 39(4): 303-307, 2018 Jul.
Article in German | MEDLINE | ID: mdl-29623404

ABSTRACT

The requirements for optimal biobanking from the point of view of the clinical partner can be highly variable. Depending on the material, processing, storage conditions, clinical data, and involvement of external partners, there will be special requirements for the participating clinician and specialist areas. What they all have in common is that the goal of any biobanking must be to improve clinical, translational, and basic research. While in the past biomaterials often had to be individually stored for each research project, modern biobanking offers decisive advantages: a comprehensive ethics vote fulfilling state-of-the-art data safety requirements, standardized processing and storage protocols, specialized biobank software for pseudonymization and localization, protection against power failures and defects of the equipment, centralized and sustainable storage, easy localization and return of samples, and their destruction or anonymization after completion of an individual project. In addition to this important pure storage function, central biobanking can provide a link to clinical data as well as the anonymous use of samples for project-independent research. Both biobank functions serve different purposes, are associated with specific requirements, and should be pursued in parallel. If successful, central biomaterial management can achieve a sustainable improvement of academic and non-academic biomedical research and the optimal use of resources. The close collaboration between clinicians and non-clinicians is a crucial prerequisite for this.


Subject(s)
Biological Specimen Banks , Biomedical Research , Hematology
3.
Leukemia ; 32(3): 774-787, 2018 03.
Article in English | MEDLINE | ID: mdl-28804127

ABSTRACT

T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive neoplasm of mature T-cells with an urgent need for rationally designed therapies to address its notoriously chemo-refractory behavior. The median survival of T-PLL patients is <2 years and clinical trials are difficult to execute. Here we systematically explored the diversity of drug responses in T-PLL patient samples using an ex vivo drug sensitivity and resistance testing platform and correlated the findings with somatic mutations and gene expression profiles. Intriguingly, all T-PLL samples were sensitive to the cyclin-dependent kinase inhibitor SNS-032, which overcame stromal-cell-mediated protection and elicited robust p53-activation and apoptosis. Across all patients, the most effective classes of compounds were histone deacetylase, phosphoinositide-3 kinase/AKT/mammalian target of rapamycin, heat-shock protein 90 and BH3-family protein inhibitors as well as p53 activators, indicating previously unexplored, novel targeted approaches for treating T-PLL. Although Janus-activated kinase-signal transducer and activator of transcription factor (JAK-STAT) pathway mutations were common in T-PLL (71% of patients), JAK-STAT inhibitor responses were not directly linked to those or other T-PLL-specific lesions. Overall, we found that genetic markers do not readily translate into novel effective therapeutic vulnerabilities. In conclusion, novel classes of compounds with high efficacy in T-PLL were discovered with the comprehensive ex vivo drug screening platform warranting further studies of synergisms and clinical testing.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Leukemia, Prolymphocytic, T-Cell/genetics , Mutation , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Cell Cycle/genetics , Cell Line, Tumor , Chromosome Aberrations , Female , Gene Expression , Gene Expression Profiling , Humans , Janus Kinases/metabolism , Leukemia, Prolymphocytic, T-Cell/drug therapy , Leukemia, Prolymphocytic, T-Cell/metabolism , Male , Middle Aged , Molecular Targeted Therapy , Oxazoles/pharmacology , Phenotype , Protein Kinase Inhibitors/pharmacology , STAT Transcription Factors/metabolism , Thiazoles/pharmacology
7.
J Hematol Oncol ; 9: 18, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26944254

ABSTRACT

BACKGROUND: Patients with Ph-negative myeloproliferative neoplasms (MPN), such as polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are at increased risk for thrombosis/thromboembolism and major bleeding. Due to the morbidity and mortality of these events, antiplatelet and/or anticoagulant agents are commonly employed as primary and/or secondary prophylaxis. On the other hand, disease-related bleeding complications (i.e., from esophageal varices) are common in patients with MPN. This analysis was performed to define the frequency of such events, identify risk factors, and assess antiplatelet/anticoagulant therapy in a cohort of patients with MPN. METHODS: The MPN registry of the Study Alliance Leukemia is a non-interventional prospective study including adult patients with an MPN according to WHO criteria (2008). For statistical analysis, descriptive methods and tests for significant differences as well as contingency tables were used to identify the odds of potential risk factors for vascular events. RESULTS: MPN subgroups significantly differed in sex distribution, age at diagnosis, blood counts, LDH levels, JAK2V617F positivity, and spleen size (length). While most thromboembolic events occurred around the time of MPN diagnosis, one third of these events occurred after that date. Splanchnic vein thrombosis was most frequent in post-PV-MF and MPN-U patients. The chance of developing a thromboembolic event was significantly elevated if patients suffered from post-PV-MF (OR 3.43; 95% CI = 1.39-8.48) and splenomegaly (OR 1.76; 95% CI = 1.15-2.71). Significant odds for major bleeding were previous thromboembolic events (OR = 2.71; 95% CI = 1.36-5.40), splenomegaly (OR = 2.22; 95% CI 1.01-4.89), and the administration of heparin (OR = 5.64; 95% CI = 1.84-17.34). Major bleeding episodes were significantly less frequent in ET patients compared to other MPN subgroups. CONCLUSIONS: Together, this report on an unselected "real-world" cohort of German MPN patients reveals important data on the prevalence, diagnosis, and treatment of thromboembolic and major bleeding complications of MPN.


Subject(s)
Blood Coagulation/physiology , Hemorrhage/physiopathology , Myeloproliferative Disorders/physiopathology , Registries/statistics & numerical data , Thrombosis/physiopathology , Adult , Aged , Aged, 80 and over , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Female , Germany/epidemiology , Hemorrhage/diagnosis , Hemorrhage/prevention & control , Humans , Logistic Models , Male , Middle Aged , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/epidemiology , Prevalence , Prospective Studies , Splenomegaly/diagnosis , Splenomegaly/physiopathology , Thrombosis/diagnosis , Thrombosis/prevention & control
8.
Leukemia ; 30(5): 1018-24, 2016 05.
Article in English | MEDLINE | ID: mdl-26854026

ABSTRACT

The Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal disorders involving hematopoietic stem and progenitor cells and are associated with myeloproliferation, splenomegaly and constitutional symptoms. Similar signs and symptoms can also be found in patients with chronic inflammatory diseases, and inflammatory processes have been found to play an important role in the pathogenesis and progression of MPNs. Signal transduction pathways involving JAK1, JAK2, STAT3 and STAT5 are causally involved in driving both the malignant cells and the inflammatory process. Moreover, anti-inflammatory and immune-modulating drugs have been used successfully in the treatment of MPNs. However, to date, many unresoved issues remain. These include the role of somatic mutations that are present in addition to JAK2V617F, CALR and MPL W515 mutations, the interdependency of malignant and nonmalignant cells and the means to eradicate MPN-initiating and -maintaining cells. It is imperative for successful therapeutic approaches to define whether the malignant clone or the inflammatory cells or both should be targeted. The present review will cover three aspects of the role of inflammation in MPNs: inflammatory states as important differential diagnoses in cases of suspected MPN (that is, in the absence of a clonal marker), the role of inflammation in MPN pathogenesis and progression and the use of anti-inflammatory drugs for MPNs. The findings emphasize the need to separate the inflammatory processes from the malignancy in order to improve our understanding of the pathogenesis, diagnosis and treatment of patients with Philadelphia-negative MPNs.


Subject(s)
Inflammation/drug therapy , Myeloproliferative Disorders/drug therapy , Neoplasms/pathology , Anti-Inflammatory Agents/therapeutic use , Clone Cells/pathology , Humans , Myeloproliferative Disorders/pathology
9.
Leukemia ; 30(3): 555-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26522083

ABSTRACT

DNA methylation changes are a constant feature of acute myeloid leukemia. Hypomethylating drugs such as azacitidine are active in acute myeloid leukemia (AML) as monotherapy. Azacitidine monotherapy is not curative. The AML-AZA trial tested the hypothesis that DNA methyltransferase inhibitors such as azacitidine can improve chemotherapy outcome in AML. This randomized, controlled trial compared the efficacy of azacitidine applied before each cycle of intensive chemotherapy with chemotherapy alone in older patients with untreated AML. Event-free survival (EFS) was the primary end point. In total, 214 patients with a median age of 70 years were randomized to azacitidine/chemotherapy (arm-A) or chemotherapy (arm-B). More arm-A patients (39/105; 37%) than arm-B (25/109; 23%) showed adverse cytogenetics (P=0.057). Adverse events were more frequent in arm-A (15.44) versus 13.52 in arm-B, (P=0.26), but early death rates did not differ significantly (30-day mortality: 6% versus 5%, P=0.76). Median EFS was 6 months in both arms (P=0.96). Median overall survival was 15 months for patients in arm-A compared with 21 months in arm-B (P=0.35). Azacitidine added to standard chemotherapy increases toxicity in older patients with AML, but provides no additional benefit for unselected patients.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/therapeutic use , Induction Chemotherapy/methods , Leukemia, Myeloid, Acute/drug therapy , Aged , Cytarabine/therapeutic use , Cytogenetic Analysis , Daunorubicin/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Remission Induction , Survival Analysis
10.
Leukemia ; 30(4): 823-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26621336

ABSTRACT

Chronic myeloid leukemia (CML) is driven by malignant stem cells that can persist despite therapy. We have identified Metastasis suppressor 1 (Mtss1/MIM) to be downregulated in hematopoietic stem and progenitor cells from leukemic transgenic SCLtTA/Bcr-Abl mice and in patients with CML at diagnosis, and Mtss1 was restored when patients achieved complete remission. Forced expression of Mtss1 decreased clonogenic capacity and motility of murine myeloid progenitor cells and reduced tumor growth. Viral transduction of Mtss1 into lineage-depleted SCLtTA/Bcr-Abl bone marrow cells decreased leukemic cell burden in recipients, and leukemogenesis was reduced upon injection of Mtss1-overexpressing murine myeloid 32D cells. Tyrosine kinase inhibitor (TKI) therapy and reversion of Bcr-Abl expression increased Mtss1 expression but failed to restore it to control levels. CML patient samples revealed higher DNA methylation of specific Mtss1 promoter CpG sites that contain binding sites for Kaiso and Rest transcription factors. In summary, we identified a novel tumor suppressor in CML stem cells that is downregulated by both Bcr-Abl kinase-dependent and -independent mechanisms. Restored Mtss1 expression markedly inhibits primitive leukemic cell biology in vivo, providing a therapeutic rationale for the Bcr-Abl-Mtss1 axis to target TKI-resistant CML stem cells in patients.


Subject(s)
Cell Movement , Cell Proliferation , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Chromatin Immunoprecipitation , Gene Expression Regulation, Leukemic , Humans , Mice , Mice, Inbred C3H , Mice, Transgenic , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Leukemia ; 28(10): 2028-39, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24603533

ABSTRACT

Telomere biology is frequently associated with disease evolution in human cancer and dysfunctional telomeres have been demonstrated to contribute to genetic instability. In BCR-ABL(+) chronic myeloid leukemia (CML), accelerated telomere shortening has been shown to correlate with leukemia progression, risk score and response to treatment. Here, we demonstrate that proliferation of murine CML-like bone marrow cells strongly depends on telomere maintenance. CML-like cells of telomerase knockout mice with critically short telomeres (CML-iG4) are growth retarded and proliferation is terminally stalled by a robust senescent cell cycle arrest. In sharp contrast, CML-like cells with pre-shortened, but not critically short telomere lengths (CML-G2) grew most rapidly and were found to express a specific 'telomere-associated secretory phenotype', comprising secretion of chemokines, interleukins and other growth factors, thereby potentiating oncogene-driven growth. Moreover, conditioned supernatant of CML-G2 cells markedly enhanced proliferation of CML-WT and pre-senescent CML-iG4 cells. Strikingly, a similar inflammatory mRNA expression pattern was found with disease progression from chronic phase to accelerated phase in CML patients. These findings demonstrate that telomere-induced senescence needs to be bypassed by leukemic cells in order to progress to blast crisis and provide a novel mechanism by which telomere shortening may contribute to disease evolution in CML.


Subject(s)
Cell Proliferation , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Leukemia/pathology , Telomere/ultrastructure , Animals , Apoptosis , Bone Marrow Cells/cytology , Cell Cycle , Cell Line, Tumor , Cellular Senescence , Chemokines/metabolism , Cytokines/metabolism , Disease Progression , Humans , Inflammation/metabolism , Leukemia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
14.
Leukemia ; 28(6): 1227-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24280869

ABSTRACT

Mutations in the genetic sequence of the DNA de novo methyltransferase DNMT3A (DNA methyltransferase 3A) are found in many patients with acute myeloid leukemia (AML). They lead to dysfunction of DNMT3A protein and represent a marker for poor prognosis. Effects of genetic mutations can be mimicked by epigenetic modifications in the DNA methylation (DNAm) pattern. Using DNAm profiles of the Cancer Genome Atlas Research Network (TCGA), we identified aberrant hypermethylation at an internal promoter region of DNMT3A, which occurred in about 40% of AML patients. Bisulfite pyrosequencing assays designed for this genomic region validated hypermethylation specifically in a subset of our AML samples. High DNAm levels at this site are particularly observed in samples without genetic mutations in DNMT3A. Epimutations and mutations of DNMT3A were associated with related gene expression changes such as upregulation of the homeobox genes in HOXA and HOXB clusters. Furthermore, epimutations in DNMT3A were enriched in patients with poor or intermediate cytogenetic risk, and in patients with shorter event-free survival and overall survival (OS). Taken together, aberrant DNA hypermethylation within the DNMT3A gene, in analogy to DNMT3A mutations, is frequently observed in AML and both modifications seem to be useful for risk stratification or choice of therapeutic regimen.


Subject(s)
Biomarkers, Tumor/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Epigenesis, Genetic/genetics , Genomics , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , DNA Methyltransferase 3A , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/mortality , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
16.
Leukemia ; 26(3): 465-74, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21904381

ABSTRACT

Although BCR-ABL+ stem cells in chronic myeloid leukemia (CML) resist elimination by targeted pharmacotherapy in most patients, immunological graft-versus-leukemia effects can cure the disease. Besides cytotoxic T cells, natural killer (NK) cells may have a role in immune control of CML. Here, we explored the functionality of NK cells in CML patients and in a transgenic inducible BCR-ABL mouse model. Compared with controls, NK-cell proportions among lymphocytes were decreased at diagnosis of CML and did not recover during imatinib-induced remission for 10-34 months. Functional experiments revealed limited in vitro expansion of NK cells from CML patients and a reduced degranulation response to K562 target cells both at diagnosis and during imatinib therapy. Consistent with the results in human CML, relative numbers of NK1.1+ NK cells were reduced following induction of BCR-ABL expression in mice, and the defects persisted after BCR-ABL reversion. Moreover, target-induced degranulation by expanded BCR-ABL+ NK cells was compromised. We conclude that CML is associated with quantitative and functional defects within the NK-cell compartment, which is reproduced by induced BCR-ABL expression in mice. Further work will aim at identifying the mechanisms of NK-cell deficiency in CML and at developing strategies to exploit NK cells for immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Fusion Proteins, bcr-abl/genetics , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Degranulation/genetics , Cell Degranulation/immunology , Child , Disease Models, Animal , Humans , Imatinib Mesylate , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Transgenic , Middle Aged , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Young Adult
17.
Ann Oncol ; 23(4): 990-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21810729

ABSTRACT

BACKGROUND: Chemotherapy for elderly patients with acute myeloid leukemia (AML) results in a median overall survival (OS) of ≤ 1 year. Elderly patients often present with cardiac comorbidity. Gemtuzumab ozogamicin (GO) is active in elderly (≥ 60 years) patients with relapsed AML with low cardiac toxicity. PATIENTS AND METHODS: This randomized phase II study compared a standard combination of ara-C and daunorubicin (DNR; 7+3) versus ara-C plus gemtuzumab ozogamicin (7+GO) as the first course of induction therapy. Primary objectives were comparison of blast clearance on day 16, event-free survival (EFS), and remission duration. OS, complete remission (CR), and tolerability were secondary objectives. RESULTS: One hundred and nineteen patients with de novo AML, treatment-related AML, AML with a history of myelodysplastic syndrome (MDS), or high-risk MDS entered the study. Median age of 115 patients (intent-to-treat population) was 69 years. Protocol outlined a second course 7+3 for patients without blast clearance and two courses of high-dose ara-C consolidation upon CR. Both treatments were equally effective in blast clearance, CR, EFS, remission duration, or OS (median: 7+3, 9 months; 7+GO, 10 months). Induction death rate was higher in the GO group due to veno-occlusive disease. CONCLUSION: The study did not show significant superiority of 7+GO over standard 7+3.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Induction Chemotherapy , Leukemia, Myeloid, Acute/drug therapy , Aged , Aged, 80 and over , Aminoglycosides/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Disease-Free Survival , Female , Gemtuzumab , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Survival Analysis , Treatment Outcome
18.
Leukemia ; 26(5): 1030-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22193968

ABSTRACT

Spleen size ranks among the most important risk factors in chronic myeloid leukemia (CML), but the pathogenic mechanisms of splenic hematopoiesis in CML remain poorly defined. Here, we studied the biology of Bcr-Abl positive leukemia-initiating cells in the spleen, using an inducible transgenic mouse model of CML. Disease kinetics showed greater increases of immature leukemic cells in spleen vs bone marrow (BM). To assess how Bcr-Abl alters the behavior of spleen-derived CML cells, we transplanted these cells either before ('pre-uninduced') or 44 days after ('pre-induced') expression of the oncogene. Mice transplanted with pre-induced spleen cells showed significantly increased neutrophilia and splenomegaly compared with mice receiving pre-uninduced spleen cells, suggesting that Bcr-Abl expression in the donors had increased splenic tumor burden. However, pre-induction also altered the biology of these cells, as shown by a striking increase in erythropoietic potential. These results differ from those of BM-derived CML stem cells where pre-induction of Bcr-Abl had previously been shown to decrease disease transplantability. Moreover, splenic cells were less sensitive to imatinib than BM cells. In conclusion, Bcr-Abl alters the biology of splenic leukemic stem cells by a cell-autonomous mechanism, but the disease phenotype is also influenced by the microenvironment of these cells.


Subject(s)
Bone Marrow Cells/pathology , Disease Models, Animal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Spleen/pathology , Animals , Disease Progression , Flow Cytometry , Fusion Proteins, bcr-abl/metabolism , Mice , Mice, Transgenic , Splenomegaly
20.
Leukemia ; 22(1): 78-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17943165

ABSTRACT

Pim2 is a serine/threonine kinase expressed at high levels in several malignancies including acute leukemia. Pim2 protein is induced by oncogenic Fms-like tyrosine kinase-3 (Flt3)-internal tandem duplications (ITD), but not by Flt3 wild-type receptor (Flt3-Wt) in response to Flt3 ligand (FL). Here we show that Pim2 can complement Flt3-Wt signaling and induce transformation similar to Flt3-ITD in myeloid cells. Our data demonstrate that Pim2 is necessary but not sufficient for Flt3-ITD-induced transformation of 32D cells and primary bone marrow cells as assessed by colony assays. Pim2-induced clonogenic growth of FL-treated 32D-Flt3-Wt cells. Proliferation of 32D-Flt3-Wt cells was significantly enhanced in FL-treated Pim2-overexpressing cells. This increase was associated with enhanced S-phase cell cycle progression. Pim2-overexpressing cells were resistant to apoptosis induced by growth factor deprivation or treatment with tyrosine kinase inhibitor (PKC412). The Flt3 point mutant D835Y, which is not able to support colony growth of myeloid cells, also induced clonogenic growth in the presence of Pim2. In conclusion, Pim2 is an important target of Flt3-ITD-induced transformation, and overexpression of Pim2 together with Flt3-Wt or D835Y receptor mimics Flt3-ITD-mediated transformation. Pim2 complements with Flt3-Wt signaling to induce proliferation by enhancing G(1)/S-phase progression of the cell cycle.


Subject(s)
Cell Transformation, Neoplastic , Hematopoietic Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/metabolism , Mice , Myeloid Cells/metabolism , RNA, Small Interfering/pharmacology , S Phase/physiology , STAT5 Transcription Factor/metabolism , Signal Transduction , Tandem Repeat Sequences/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...