Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 612(7939): 301-309, 2022 12.
Article in English | MEDLINE | ID: mdl-36450978

ABSTRACT

Clonal haematopoiesis involves the expansion of certain blood cell lineages and has been associated with ageing and adverse health outcomes1-5. Here we use exome sequence data on 628,388 individuals to identify 40,208 carriers of clonal haematopoiesis of indeterminate potential (CHIP). Using genome-wide and exome-wide association analyses, we identify 24 loci (21 of which are novel) where germline genetic variation influences predisposition to CHIP, including missense variants in the lymphocytic antigen coding gene LY75, which are associated with reduced incidence of CHIP. We also identify novel rare variant associations with clonal haematopoiesis and telomere length. Analysis of 5,041 health traits from the UK Biobank (UKB) found relationships between CHIP and severe COVID-19 outcomes, cardiovascular disease, haematologic traits, malignancy, smoking, obesity, infection and all-cause mortality. Longitudinal and Mendelian randomization analyses revealed that CHIP is associated with solid cancers, including non-melanoma skin cancer and lung cancer, and that CHIP linked to DNMT3A is associated with the subsequent development of myeloid but not lymphoid leukaemias. Additionally, contrary to previous findings from the initial 50,000 UKB exomes6, our results in the full sample do not support a role for IL-6 inhibition in reducing the risk of cardiovascular disease among CHIP carriers. Our findings demonstrate that CHIP represents a complex set of heterogeneous phenotypes with shared and unique germline genetic causes and varied clinical implications.


Subject(s)
COVID-19 , Cardiovascular Diseases , Humans , Clonal Hematopoiesis/genetics , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics
2.
Blood ; 139(16): 2483-2498, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35020836

ABSTRACT

NOTCH1 is a well-established lineage specifier for T cells and among the most frequently mutated genes throughout all subclasses of T cell acute lymphoblastic leukemia (T-ALL). How oncogenic NOTCH1 signaling launches a leukemia-prone chromatin landscape during T-ALL initiation is unknown. Here we demonstrate an essential role for the high-mobility-group transcription factor Tcf1 in orchestrating chromatin accessibility and topology, allowing aberrant Notch1 signaling to convey its oncogenic function. Although essential, Tcf1 is not sufficient to initiate leukemia. The formation of a leukemia-prone epigenetic landscape at the distal Notch1-regulated Myc enhancer, which is fundamental to this disease, is Tcf1-dependent and occurs within the earliest progenitor stage even before cells adopt a T lymphocyte or leukemic fate. Moreover, we discovered a unique evolutionarily conserved Tcf1-regulated enhancer element in the distal Myc-enhancer, which is important for the transition of preleukemic cells to full-blown disease.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Carcinogenesis/genetics , Cell Line, Tumor , Chromatin/genetics , Humans , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics
3.
Trends Immunol ; 42(5): 418-431, 2021 05.
Article in English | MEDLINE | ID: mdl-33858773

ABSTRACT

Enhancers control dynamic changes in gene expression and orchestrate the tightly controlled transcriptional circuitries that direct and coordinate cell growth, proliferation, survival, lineage commitment, and differentiation during lymphoid development. Enhancer hijacking and neoenhancer formation at oncogene loci, as well as aberrant activation of oncogene-associated enhancers, can induce constitutive activation of self-perpetuating oncogenic transcriptional circuitries, and contribute to the malignant transformation of immature lymphoid progenitors in acute lymphoblastic leukemia (ALL). In this review, we present recent discoveries of the role of enhancer dynamics in mouse and human lymphoid development, and discuss how genetic and epigenetic alterations of enhancer function can promote leukemogenesis, and potential strategies for targeting the enhancer machinery in the treatment of ALL.


Subject(s)
Enhancer Elements, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Carcinogenesis/genetics , Cell Differentiation , Enhancer Elements, Genetic/genetics , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
4.
Nature ; 553(7689): 511-514, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342136

ABSTRACT

Relapsed acute lymphoblastic leukaemia (ALL) is associated with resistance to chemotherapy and poor prognosis. Gain-of-function mutations in the 5'-nucleotidase, cytosolic II (NT5C2) gene induce resistance to 6-mercaptopurine and are selectively present in relapsed ALL. Yet, the mechanisms involved in NT5C2 mutation-driven clonal evolution during the initiation of leukaemia, disease progression and relapse remain unknown. Here we use a conditional-and-inducible leukaemia model to demonstrate that expression of NT5C2(R367Q), a highly prevalent relapsed-ALL NT5C2 mutation, induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukaemia cell growth and leukaemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2+/R367Q mutant cells is associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukaemia lymphoblasts. These results identify the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and support a role for IMPDH inhibition in the treatment of ALL.


Subject(s)
5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Clonal Evolution , Drug Resistance, Neoplasm/genetics , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Proliferation , Disease Models, Animal , Female , Gain of Function Mutation/genetics , Guanosine/biosynthesis , HEK293 Cells , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Male , Mercaptopurine/pharmacology , Mercaptopurine/therapeutic use , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Purines/metabolism , Receptor, Notch1/metabolism , Recurrence , Xenograft Model Antitumor Assays
6.
Nat Immunol ; 11(8): 717-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622884

ABSTRACT

ZFP36L1 and ZFP36L2 are RNA-binding proteins (RBPs) that interact with AU-rich elements in the 3' untranslated region of mRNA, which leads to mRNA degradation and translational repression. Here we show that mice that lacked ZFP36L1 and ZFP36L2 during thymopoiesis developed a T cell acute lymphoblastic leukemia (T-ALL) dependent on the oncogenic transcription factor Notch1. Before the onset of T-ALL, thymic development was perturbed, with accumulation of cells that had passed through the beta-selection checkpoint without first expressing the T cell antigen receptor beta-chain (TCRbeta). Notch1 expression was higher in untransformed thymocytes in the absence of ZFP36L1 and ZFP36L2. Both RBPs interacted with evolutionarily conserved AU-rich elements in the 3' untranslated region of Notch1 and suppressed its expression. Our data establish a role for ZFP36L1 and ZFP36L2 during thymocyte development and in the prevention of malignant transformation.


Subject(s)
Nuclear Proteins/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Tristetraprolin/deficiency , Amino Acid Sequence , Animals , Butyrate Response Factor 1 , Conserved Sequence , Humans , Immunophenotyping , Kaplan-Meier Estimate , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Alignment , Thymus Gland/growth & development , Transcription, Genetic , Tristetraprolin/genetics , Tristetraprolin/immunology
7.
Blood ; 135(12): 921-933, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31971569

ABSTRACT

Activating mutations in cytosolic 5'-nucleotidase II (NT5C2) are considered to drive relapse formation in acute lymphoblastic leukemia (ALL) by conferring purine analog resistance. To examine the clinical effects of NT5C2 mutations in relapsed ALL, we analyzed NT5C2 in 455 relapsed B-cell precursor ALL patients treated within the ALL-REZ BFM 2002 relapse trial using sequencing and sensitive allele-specific real-time polymerase chain reaction. We detected 110 NT5C2 mutations in 75 (16.5%) of 455 B-cell precursor ALL relapses. Two-thirds of relapses harbored subclonal mutations and only one-third harbored clonal mutations. Event-free survival after relapse was inferior in patients with relapses with clonal and subclonal NT5C2 mutations compared with those without (19% and 25% vs 53%, P < .001). However, subclonal, but not clonal, NT5C2 mutations were associated with reduced event-free survival in multivariable analysis (hazard ratio, 1.89; 95% confidence interval, 1.28-2.69; P = .001) and with an increased rate of nonresponse to relapse treatment (subclonal 32%, clonal 12%, wild type 9%, P < .001). Nevertheless, 27 (82%) of 33 subclonal NT5C2 mutations became undetectable at the time of nonresponse or second relapse, and in 10 (71%) of 14 patients subclonal NT5C2 mutations were undetectable already after relapse induction treatment. These results show that subclonal NT5C2 mutations define relapses associated with high risk of treatment failure in patients and at the same time emphasize that their role in outcome is complex and goes beyond mutant NT5C2 acting as a targetable driver during relapse progression. Sensitive, prospective identification of NT5C2 mutations is warranted to improve the understanding and treatment of this aggressive ALL relapse subtype.


Subject(s)
5'-Nucleotidase/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Adolescent , Alleles , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Frequency , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Recurrence , Young Adult
8.
Nature ; 526(7574): 519-24, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26200345

ABSTRACT

Chronic lymphocytic leukaemia (CLL) is a frequent disease in which the genetic alterations determining the clinicobiological behaviour are not fully understood. Here we describe a comprehensive evaluation of the genomic landscape of 452 CLL cases and 54 patients with monoclonal B-lymphocytosis, a precursor disorder. We extend the number of CLL driver alterations, including changes in ZNF292, ZMYM3, ARID1A and PTPN11. We also identify novel recurrent mutations in non-coding regions, including the 3' region of NOTCH1, which cause aberrant splicing events, increase NOTCH1 activity and result in a more aggressive disease. In addition, mutations in an enhancer located on chromosome 9p13 result in reduced expression of the B-cell-specific transcription factor PAX5. The accumulative number of driver alterations (0 to ≥4) discriminated between patients with differences in clinical behaviour. This study provides an integrated portrait of the CLL genomic landscape, identifies new recurrent driver mutations of the disease, and suggests clinical interventions that may improve the management of this neoplasia.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/genetics , 3' Untranslated Regions/genetics , Alternative Splicing/genetics , B-Lymphocytes/metabolism , Carrier Proteins/genetics , Chromosomes, Human, Pair 9/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins , Enhancer Elements, Genetic/genetics , Genomics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PAX5 Transcription Factor/biosynthesis , PAX5 Transcription Factor/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Transcription Factors/genetics
9.
Nature ; 514(7523): 513-7, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25132549

ABSTRACT

T-cell acute lymphoblastic leukaemia (T-ALL) is a haematological malignancy with a dismal overall prognosis, including a relapse rate of up to 25%, mainly because of the lack of non-cytotoxic targeted therapy options. Drugs that target the function of key epigenetic factors have been approved in the context of haematopoietic disorders, and mutations that affect chromatin modulators in a variety of leukaemias have recently been identified; however, 'epigenetic' drugs are not currently used for T-ALL treatment. Recently, we described that the polycomb repressive complex 2 (PRC2) has a tumour-suppressor role in T-ALL. Here we delineated the role of the histone 3 lysine 27 (H3K27) demethylases JMJD3 and UTX in T-ALL. We show that JMJD3 is essential for the initiation and maintenance of T-ALL, as it controls important oncogenic gene targets by modulating H3K27 methylation. By contrast, we found that UTX functions as a tumour suppressor and is frequently genetically inactivated in T-ALL. Moreover, we demonstrated that the small molecule inhibitor GSKJ4 (ref. 5) affects T-ALL growth, by targeting JMJD3 activity. These findings show that two proteins with a similar enzymatic function can have opposing roles in the context of the same disease, paving the way for treating haematopoietic malignancies with a new category of epigenetic inhibitors.


Subject(s)
Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Animals , Benzazepines/pharmacology , Epigenesis, Genetic/drug effects , Histone Demethylases/genetics , Histones/chemistry , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Lysine/metabolism , Methylation/drug effects , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 114(8): 2006-2011, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28174276

ABSTRACT

The Notch1 gene is a major oncogenic driver and therapeutic target in T-cell acute lymphoblastic leukemia (T-ALL). However, inhibition of NOTCH signaling with γ-secretase inhibitors (GSIs) has shown limited antileukemic activity in clinical trials. Here we performed an expression-based virtual screening to identify highly active antileukemic drugs that synergize with NOTCH1 inhibition in T-ALL. Among these, withaferin A demonstrated the strongest cytotoxic and GSI-synergistic antileukemic effects in vitro and in vivo. Mechanistically, network perturbation analyses showed eIF2A-phosphorylation-mediated inhibition of protein translation as a critical mediator of the antileukemic effects of withaferin A and its interaction with NOTCH1 inhibition. Overall, these results support a role for anti-NOTCH1 therapies and protein translation inhibitor combinations in the treatment of T-ALL.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Biosynthesis/drug effects , Receptor, Notch1/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/therapeutic use , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy/methods , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Withanolides/pharmacology , Xenograft Model Antitumor Assays , eIF-2 Kinase/metabolism
11.
Proc Natl Acad Sci U S A ; 114(14): E2911-E2919, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28314854

ABSTRACT

Activating mutations of NOTCH1 (a well-known oncogene in T-cell acute lymphoblastic leukemia) are present in ∼4-13% of chronic lymphocytic leukemia (CLL) cases, where they are associated with disease progression and chemorefractoriness. However, the specific role of NOTCH1 in leukemogenesis remains to be established. Here, we report that the active intracellular portion of NOTCH1 (ICN1) is detectable in ∼50% of peripheral blood CLL cases lacking gene mutations. We identify a "NOTCH1 gene-expression signature" in CLL cells, and show that this signature is significantly enriched in primary CLL cases expressing ICN1, independent of NOTCH1 mutation. NOTCH1 target genes include key regulators of B-cell proliferation, survival, and signal transduction. In particular, we show that NOTCH1 transactivates MYC via binding to B-cell-specific regulatory elements, thus implicating this oncogene in CLL development. These results significantly extend the role of NOTCH1 in CLL pathogenesis, and have direct implications for specific therapeutic targeting.


Subject(s)
B-Lymphocytes/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Receptor, Notch1/genetics , B-Lymphocytes/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Leukemic , Genes, myc , Humans , Mutation , Receptor, Notch1/blood
12.
Proc Natl Acad Sci U S A ; 114(4): 764-769, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28062691

ABSTRACT

Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of non-Hodgkin lymphomas frequently associated with poor prognosis and for which genetic mechanisms of transformation remain incompletely understood. Using RNA sequencing and targeted sequencing, here we identify a recurrent in-frame deletion (VAV1 Δ778-786) generated by a focal deletion-driven alternative splicing mechanism as well as novel VAV1 gene fusions (VAV1-THAP4, VAV1-MYO1F, and VAV1-S100A7) in PTCL. Mechanistically these genetic lesions result in increased activation of VAV1 catalytic-dependent (MAPK, JNK) and non-catalytic-dependent (nuclear factor of activated T cells, NFAT) VAV1 effector pathways. These results support a driver oncogenic role for VAV1 signaling in the pathogenesis of PTCL.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Guanine/metabolism , Lymphoma, T-Cell, Peripheral/genetics , Mutation/genetics , Proto-Oncogene Proteins c-vav/genetics , Translocation, Genetic/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Humans , Jurkat Cells , Sequence Deletion/genetics
13.
Proc Natl Acad Sci U S A ; 113(40): 11306-11311, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27655895

ABSTRACT

Although multiagent combination chemotherapy is curative in a significant fraction of childhood acute lymphoblastic leukemia (ALL) patients, 20% of cases relapse and most die because of chemorefractory disease. Here we used whole-exome and whole-genome sequencing to analyze the mutational landscape at relapse in pediatric ALL cases. These analyses identified numerous relapse-associated mutated genes intertwined in chemotherapy resistance-related protein complexes. In this context, RAS-MAPK pathway-activating mutations in the neuroblastoma RAS viral oncogene homolog (NRAS), kirsten rat sarcoma viral oncogene homolog (KRAS), and protein tyrosine phosphatase, nonreceptor type 11 (PTPN11) genes were present in 24 of 55 (44%) cases in our series. Interestingly, some leukemias showed retention or emergence of RAS mutant clones at relapse, whereas in others RAS mutant clones present at diagnosis were replaced by RAS wild-type populations, supporting a role for both positive and negative selection evolutionary pressures in clonal evolution of RAS-mutant leukemia. Consistently, functional dissection of mouse and human wild-type and mutant RAS isogenic leukemia cells demonstrated induction of methotrexate resistance but also improved the response to vincristine in mutant RAS-expressing lymphoblasts. These results highlight the central role of chemotherapy-driven selection as a central mechanism of leukemia clonal evolution in relapsed ALL, and demonstrate a previously unrecognized dual role of RAS mutations as drivers of both sensitivity and resistance to chemotherapy.


Subject(s)
Clonal Evolution/genetics , Genes, ras , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Base Sequence , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Methotrexate/pharmacology , Methotrexate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology , Vincristine/therapeutic use
14.
J Immunol ; 197(1): 296-302, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27259858

ABSTRACT

Matrix metalloproteases (MMPs) regulate innate immunity acting over proinflammatory cytokines, chemokines, and other immune-related proteins. MMP-25 (membrane-type 6-MMP) is a membrane-bound enzyme predominantly expressed in leukocytes whose biological function has remained largely unknown. We have generated Mmp25-deficient mice to elucidate the in vivo function of this protease. These mutant mice are viable and fertile and do not show any spontaneous phenotype. However, Mmp25-null mice exhibit a defective innate immune response characterized by low sensitivity to bacterial LPS, hypergammaglobulinemia, and reduced secretion of proinflammatory molecules. Moreover, these immune defects can be tracked to a defective NF-κB activation observed in Mmp25-deficient leukocytes. Globally, our findings provide new mechanistic insights into innate immunity through the activity of MMP-25, suggesting that this proteinase could be a potential therapeutic target for immune-related diseases.


Subject(s)
Hypergammaglobulinemia/immunology , Leukocytes/immunology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Matrix Metalloproteinases, Membrane-Associated/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Protein Binding , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 112(25): 7773-8, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056302

ABSTRACT

The transcription factor T-helper-inducing POZ/Krueppel-like factor (ThPOK, encoded by the Zbtb7b gene) plays widespread and critical roles in T-cell development, particularly as the master regulator of CD4 commitment. Here we show that mice expressing a constitutive T-cell-specific ThPOK transgene (ThPOK(const) mice) develop thymic lymphomas. These tumors resemble human T-cell acute lymphoblastic leukemia (T-ALL), in that they predominantly exhibit activating Notch1 mutations. Lymphomagenesis is prevented if thymocyte development is arrested at the DN3 stage by recombination-activating gene (RAG) deficiency, but restored by introduction of a T-cell receptor (TCR) transgene or by a single injection of anti-αßTCR antibody into ThPOK(const) RAG-deficient mice, which promotes development to the CD4(+)8(+) (DP) stage. Hence, TCR signals and/or traversal of the DN (double negative) > DP (double positive) checkpoint are required for ThPOK-mediated lymphomagenesis. These results demonstrate a novel link between ThPOK, TCR signaling, and lymphomagenesis. Finally, we present evidence that ectopic ThPOK expression gives rise to a preleukemic and self-perpetuating DN4 lymphoma precursor population. Our results collectively define a novel role for ThPOK as an oncogene and precisely map the stage in thymopoiesis susceptible to ThPOK-dependent tumor initiation.


Subject(s)
Gene Expression Regulation , Lymphoma, T-Cell/pathology , T-Lymphocytes/cytology , Transcription Factors/genetics , Animals , Incidence , Lymphoma, T-Cell/genetics , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Notch/metabolism , Signal Transduction , Transgenes
16.
Blood ; 126(7): 833-41, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-25966987

ABSTRACT

T-cell immunophenotype of acute lymphoblastic leukemia (T-ALL) is an uncommon aggressive leukemia that can present with leukemic and/or lymphomatous manifestations. Molecular studies are enhancing our understanding of the pathogenesis of T-ALL, and the discovery of activating mutations of NOTCH1 and FBXW7 in a majority of patients has been a seminal observation. The use of pediatric intensive combination chemotherapy regimens in adolescents and young adults has significantly improved the outcome of patients with T-ALL. The use of nelarabine for relapsed and refractory T-ALL results in responses in a substantial minority of patients. Allogeneic hematopoietic cell transplantation (HCT) still plays a key role in patients with high-risk or relapsed/refractory disease. γ-Secretase inhibitors hold promise for the treatment of patients with NOTCH1 mutations, and the results of clinical trials with these agents are eagerly awaited. It is recommended that younger patients receive a pediatric-intensive regimen. Older and unfit patients can receive suitable multiagent chemotherapy and be allocated to HCT based on their response, risk factors, and comorbidities. Although advances in the treatment of T-ALL have lagged behind those of B-cell ALL, it is hoped that the molecular revolution will enhance our understanding of the pathogenesis and treatment of this aggressive lymphoid malignancy.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Age Factors , Algorithms , Antineoplastic Combined Chemotherapy Protocols , Child , Disease-Free Survival , Drugs, Investigational/therapeutic use , Female , Hematopoietic Stem Cell Transplantation , Humans , Male , Middle Aged , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Prognosis , Young Adult
17.
Blood ; 125(18): 2806-14, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25784680

ABSTRACT

Oncogenic activation of NOTCH1 signaling plays a central role in the pathogenesis of T-cell acute lymphoblastic leukemia, with mutations on this signaling pathway affecting more than 60% of patients at diagnosis. However, the transcriptional regulatory circuitries driving T-cell transformation downstream of NOTCH1 remain incompletely understood. Here we identify Hairy and Enhancer of Split 1 (HES1), a transcriptional repressor controlled by NOTCH1, as a critical mediator of NOTCH1-induced leukemogenesis strictly required for tumor cell survival. Mechanistically, we demonstrate that HES1 directly downregulates the expression of BBC3, the gene encoding the PUMA BH3-only proapoptotic factor in T-cell acute lymphoblastic leukemia. Finally, we identify perhexiline, a small-molecule inhibitor of mitochondrial carnitine palmitoyltransferase-1, as a HES1-signature antagonist drug with robust antileukemic activity against NOTCH1-induced leukemias in vitro and in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Leukemic , Gene Targeting/methods , Homeodomain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic/drug effects , Gene Silencing , HEK293 Cells , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/physiology , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Molecular Targeted Therapy , Perhexiline/therapeutic use , Receptor, Notch1/genetics , Transcription Factor HES-1
18.
Blood ; 126(22): 2479-83, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26443624

ABSTRACT

The development of the dual Janus kinase 1/2 (JAK1/2) inhibitor ruxolitinib for the treatment of myeloproliferative neoplasms (MPNs) has led to studies of ruxolitinib in other clinical contexts, including JAK-mutated acute lymphoblastic leukemia (ALL). However, the limited ability of JAK inhibition to induce molecular or clinicopathological responses in MPNs suggests a need for development of better therapies for JAK kinase-dependent malignancies. Here, we demonstrate that heat shock protein 90 (HSP90) inhibition using a purine-scaffold HSP90 inhibitor in early clinical development is an effective therapeutic approach in JAK-dependent ALL and can overcome persistence to JAK-inhibitor therapy in ALL cells.


Subject(s)
Benzodioxoles/pharmacology , HSP90 Heat-Shock Proteins , Janus Kinase 1 , Janus Kinase 2 , Neoplasm Proteins , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Purines/pharmacology , Animals , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mice , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Xenograft Model Antitumor Assays
19.
Pediatr Blood Cancer ; 63(9): 1660-3, 2016 09.
Article in English | MEDLINE | ID: mdl-27149388

ABSTRACT

A 4-year-old male with the diagnosis of T-cell acute lymphoblastic leukemia (T-ALL) relapsed after 19 months with an acute myeloid leukemia (AML). Immunoglobulin and T-cell receptor gene rearrangements analyses reveal that both leukemias were rearranged with a clonal relationship between them. Comparative genomic hybridization (Array-CGH) and whole-exome sequencing analyses of both samples suggest that this leukemia may have originated from a common T/myeloid progenitor. The presence of homozygous deletion of p16/INK4A, p14/ARF, p15/INK4B, and heterozygous deletion of WT1 locus remained stable in the leukemia throughout phenotypic switch, revealing that this AML can be genetically associated to T-ALL.


Subject(s)
Leukemia, Myeloid, Acute/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Child, Preschool , Gene Deletion , Gene Rearrangement, T-Lymphocyte , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recurrence
20.
Blood ; 122(1): 74-82, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23687089

ABSTRACT

Adult T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic tumor associated with poor outcome. In this study, we analyzed the prognostic relevance of genetic alterations, immunophenotypic markers, and microarray gene expression signatures in a panel of 53 adult T-ALL patients treated in the Eastern Cooperative Oncology Group E2993 clinical trial. An early immature gene expression signature, the absence of bi-allelic TCRG deletion, CD13 surface expression, heterozygous deletions of the short arm of chromosome 17, and mutations in IDH1/IDH2 and DNMT3A genes are associated with poor prognosis in this series. In contrast, expression of CD8 or CD62L, homozygous deletion of CDKN2A/CDKN2B, NOTCH1 and/or FBXW7 mutations, and mutations or deletions in the BCL11B tumor suppressor gene were associated with improved overall survival. Importantly, the prognostic relevance of CD13 expression and homozygous CDKN2A/CDKN2B deletions was restricted to cortical and mature T-ALLs. Conversely, mutations in IDH1/IDH2 and DNMT3A were specifically associated with poor outcome in early immature adult T-ALLs. This trial was registered at www.clinicaltrials.gov as #NCT00002514.


Subject(s)
Gene Expression Regulation, Leukemic , Genomics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Adult , Female , Gene Dosage/genetics , Humans , Immunophenotyping , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microarray Analysis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Risk Factors , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL