Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 134(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38618957

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive immature T cell cancer. Mutations in IL7R have been analyzed genetically, but downstream effector functions such as STAT5A and STAT5B hyperactivation are poorly understood. Here, we studied the most frequent and clinically challenging STAT5BN642H driver in T cell development and immature T cell cancer onset and compared it with STAT5A hyperactive variants in transgenic mice. Enhanced STAT5 activity caused disrupted T cell development and promoted an early T cell progenitor-ALL phenotype, with upregulation of genes involved in T cell receptor (TCR) signaling, even in absence of surface TCR. Importantly, TCR pathway genes were overexpressed in human T-ALL and mature T cell cancers and activation of TCR pathway kinases was STAT5 dependent. We confirmed STAT5 binding to these genes using ChIP-Seq analysis in human T-ALL cells, which were sensitive to pharmacologic inhibition by dual STAT3/5 degraders or ZAP70 tyrosine kinase blockers in vitro and in vivo. We provide genetic and biochemical proof that STAT5A and STAT5B hyperactivation can initiate T-ALL through TCR pathway hijacking and suggest similar mechanisms for other T cell cancers. Thus, STAT5 or TCR component blockade are targeted therapy options, particularly in patients with chemoresistant clones carrying STAT5BN642H.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Humans , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein-Tyrosine Kinases , Receptors, Antigen, T-Cell/genetics , Signal Transduction , STAT5 Transcription Factor/genetics
2.
Leuk Lymphoma ; 62(14): 3440-3451, 2021 12.
Article in English | MEDLINE | ID: mdl-34414843

ABSTRACT

Patients with relapsed/refractory chronic lymphocytic leukemia (R/R CLL) whose treatment failed with a Bruton's tyrosine kinase inhibitor have poor outcomes. We investigated tafasitamab plus idelalisib (cohort A) or venetoclax (cohort B) in this patient population in a phase II study (NCT02639910). In total, 24 patients were enrolled (cohort A: n = 11, median time on study, 7.4 months; cohort B: n = 13, median time on study, 15.6 months). The most common treatment-emergent adverse event (TEAE) in cohort A was anemia (63.6%) and in cohort B was infusion-related reaction (53.8%). The most common severe TEAE was neutropenia (cohort A: 45.5%; cohort B: 46.2%). The best overall response rate was 90.9% (cohort A) and 76.9% (cohort B). Undetectable minimal residual disease in peripheral blood was achieved in 2/8 patients (cohort A) and 6/7 patients (cohort B). Overall, these results suggest that anti-CD19 antibody-based combinations may be important in the treatment of patients with CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, B-Cell , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein Kinase Inhibitors/adverse effects , Purines , Quinazolinones , Sulfonamides
3.
Int J Mol Sci ; 20(2)2019 Jan 16.
Article in English | MEDLINE | ID: mdl-30654457

ABSTRACT

Oncogenic fusion protein RUNX1-ETO is the product of the t(8;21) translocation, responsible for the most common cytogenetic subtype of acute myeloid leukemia. RUNX1, a critical transcription factor in hematopoietic development, is fused with almost the entire ETO sequence with the ability to recruit a wide range of repressors. Past efforts in providing a comprehensive picture of the genome-wide localization and the target genes of RUNX1-ETO have been inconclusive in understanding the underlying mechanism by which it deregulates native RUNX1. In this review; we dissect the current data on the epigenetic impact of RUNX1 and RUNX1-ETO. Both share similarities however, in recent years, research focused on epigenetic factors to explain their differences. RUNX1-ETO impairs DNA repair mechanisms which compromises genomic stability and favors a mutator phenotype. Among an increasing pool of mutated factors, regulators of DNA methylation are frequently found in t(8;21) AML. Together with the alteration of both, histone markers and distal enhancer regulation, RUNX1-ETO might specifically disrupt normal chromatin structure. Epigenetic studies on the fusion protein uncovered new mechanisms contributing to leukemogenesis and hopefully will translate into clinical applications.


Subject(s)
Epigenesis, Genetic , Genomic Instability , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Animals , Chromatin/metabolism , DNA Methylation/genetics , Humans , Oncogene Proteins, Fusion/chemistry
4.
Blood ; 132(7): 694-706, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29907599

ABSTRACT

Inhibition of Janus-kinase 1/2 (JAK1/2) is a mainstay to treat myeloproliferative neoplasms (MPN). Sporadic observations reported the co-incidence of B-cell non-Hodgkin lymphomas during treatment of MPN with JAK1/2 inhibitors. We assessed 626 patients with MPN, including 69 with myelofibrosis receiving JAK1/2 inhibitors for lymphoma development. B-cell lymphomas evolved in 4 (5.8%) of 69 patients receiving JAK1/2 inhibition compared with 2 (0.36%) of 557 with conventional treatment (16-fold increased risk). A similar 15-fold increase was observed in an independent cohort of 929 patients with MPN. Considering primary myelofibrosis only (N = 216), 3 lymphomas were observed in 31 inhibitor-treated patients (9.7%) vs 1 (0.54%) of 185 control patients. Lymphomas were of aggressive B-cell type, extranodal, or leukemic with high MYC expression in the absence of JAK2 V617F or other MPN-associated mutations. Median time from initiation of inhibitor therapy to lymphoma diagnosis was 25 months. Clonal immunoglobulin gene rearrangements were already detected in the bone marrow during myelofibrosis in 16.3% of patients. Lymphomas occurring during JAK1/2 inhibitor treatment were preceded by a preexisting B-cell clone in all 3 patients tested. Sequencing verified clonal identity in 2 patients. The effects of JAK1/2 inhibition were mirrored in Stat1-/- mice: 16 of 24 mice developed a spontaneous myeloid hyperplasia with the concomitant presence of aberrant B cells. Transplantations of bone marrow from diseased mice unmasked the outgrowth of a malignant B-cell clone evolving into aggressive B-cell leukemia-lymphoma. We conclude that JAK/STAT1 pathway inhibition in myelofibrosis is associated with an elevated frequency of aggressive B-cell lymphomas. Detection of a preexisting B-cell clone may identify individuals at risk.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Lymphoma, B-Cell/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Primary Myelofibrosis/enzymology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Retrospective Studies
5.
J Pathol ; 236(4): 445-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25820993

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is a rare, aggressive, non-Hodgkin's lymphoma that is characterized by CD30 expression and disease onset in young patients. About half of ALCL patients bear the t(2;5)(p23;q35) translocation, which results in the formation of the nucleophosmin-anaplastic lymphoma tyrosine kinase (NPM-ALK) fusion protein (ALCL ALK(+)). However, little is known about the molecular features and tumour drivers in ALK-negative ALCL (ALCL ALK(-)), which is characterized by a worse prognosis. We found that ALCL ALK(-), in contrast to ALCL ALK(+), lymphomas display high miR-155 expression. Consistent with this, we observed an inverse correlation between miR-155 promoter methylation and miR-155 expression in ALCL. However, no direct effect of the ALK kinase on miR-155 levels was observed. Ago2 immunoprecipitation revealed miR-155 as the most abundant miRNA, and enrichment of target mRNAs C/EBPß and SOCS1. To investigate its function, we over-expressed miR-155 in ALCL ALK(+) cell lines and demonstrated reduced levels of C/EBPß and SOCS1. In murine engraftment models of ALCL ALK(-), we showed that anti-miR-155 mimics are able to reduce tumour growth. This goes hand-in-hand with increased levels of cleaved caspase-3 and high SOCS1 in these tumours, which leads to suppression of STAT3 signalling. Moreover, miR-155 induces IL-22 expression and suppresses the C/EBPß target IL-8. These data suggest that miR-155 can act as a tumour driver in ALCL ALK(-) and blocking miR-155 could be therapeutically relevant. Original miRNA array data are to be found in the supplementary material (Table S1).


Subject(s)
Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 5 , Lymphoma, Large-Cell, Anaplastic/genetics , MicroRNAs/genetics , Translocation, Genetic , Anaplastic Lymphoma Kinase , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Case-Control Studies , Caspase 3/metabolism , Cell Line, Tumor , DNA Methylation , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Humans , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Lymphoma, Large-Cell, Anaplastic/therapy , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transfection , Xenograft Model Antitumor Assays
6.
Mutat Res ; 682(1): 7-12, 2009.
Article in English | MEDLINE | ID: mdl-19167516

ABSTRACT

The activator protein 1 (AP-1) transcription factor is assembled from jun-jun, jun-fos, or jun-atf family protein homo- or heterodimers. AP-1 belongs to the class of basic leucine zipper (bZIP) transcription factors. It binds to promoters of its target genes in a sequence-specific manner, and transactivates or represses them. AP-1 proteins are implicated in the regulation of a variety of cellular processes including proliferation and survival, differentiation, growth, apoptosis, cell migration, and transformation. The decision if a given AP-1 factor is positively or negatively regulating a specific target gene is made upon abundance of dimerization partners, dimer-composition, post-translational regulation, and interaction with accessory proteins. In this review we describe translational control mechanisms that can regulate the abundance of AP-1 proteins. The Atf4/5, and JunD (mRNAs) are regulated by upORF dependent mechanisms. JUNB (mRNA) translation is controlled via mTOR. Translation efficiency of the unstable c-Fos (mRNA) can be decreased by the miRNA mir7B, while its perinuclear translation might facilitate efficient nuclear c-fos protein import. c-Jun (mRNA) appears to be regulated by both, m7G cap (CAP)-dependent and CAP-independent translational control mechanisms, via putative internal ribosome entry segments (IRES). IRES elements were also proposed to play a role in the regulation of JunD (mRNA). We conclude that in addition to transcriptional and post-translational control mechanisms translational regulation contributes to the balanced production of AP-1 proteins, in order to maintain physiological cellular conditions.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis , Transcription Factor AP-1/physiology , Humans , MicroRNAs/metabolism , RNA Cap-Binding Proteins/metabolism , Transcription Factor AP-1/genetics , Transcription Factors/physiology
7.
Oncogene ; 23(4): 894-904, 2004 Jan 29.
Article in English | MEDLINE | ID: mdl-14749762

ABSTRACT

Recurrent disease following high-dose chemotherapy is a major problem in patients with acute myeloid leukemia (AML). To identify its characteristics, we performed expression profiling in blasts from untreated AML and relapse, using a specific cDNA microarray comprising 4128 genes generated by cDNA subtraction supplemented with cancer-associated genes. Expression analysis of 18 AML bone marrow specimens showed that recurrent AML is commonly associated with the mRNA expression changes in a set of 58 genes. Increased cellular proliferation was indicated by the overexpression of the transferrin receptor, proliferating cell nuclear antigen, and G1 cyclins. An immunohistochemical study for Ki-67-positive blasts in 18 paired bone marrow biopsy samples confirmed a highly significant (P<0.0001) increase in the proliferation fraction at relapse. In addition, we found enhanced activation of the RAF/MEK/ERK cascade as mRNAs of MKP-1, c-jun, c-fos, and egr-1 were significantly increased at relapse. Immunohistochemistry and immunoblotting analyses for biphosphorylated ERK1/2 protein provide additional evidence for enhanced activation of the RAF/MEK/ERK pathway. The degree of increase is significantly correlated with the increased proliferation. Furthermore, the genes identified provide a rationale for further studies on predictive diagnosis and therapeutic intervention.


Subject(s)
Cell Division , Gene Expression , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Acute Disease , Adult , Aged , Blotting, Western , DNA, Complementary , Humans , Immunohistochemistry , Leukemia, Myeloid/enzymology , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Recurrence , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...