GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Pediatric Blood & Cancer, Wiley, Vol. 68, No. 3 ( 2021-03)
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4177-4177
    Abstract: Abstract 4177 The BCL11B transcription factor, previously identified as a central player in normal α/β T-cell development, has recently been found to play critical roles in the maintenance of T-cell identity, with biallelic Bcl11b inactivation in T-cell precursors severely impairing their continued development into the T-cell lineage. Previous work has demonstrated that Bcl11b haploinsufficiency accelerates the onset of thymic lymphomas in p53-mutant mice, as well as T-lymphoid blast crisis in a mouse model of chronic myeloid leukemia induced by BCR-ABL. Furthermore, we have recently identified recurrent monoallelic Bcl11b deletions in 91% of T-cell acute lymphoblastic leukemias (T-ALL) arising in Atm-deficient mice, further supporting a role for Bcl11b haploinsufficiency in T-cell leukemogenesis. In order to determine whether BCL11B inactivation plays a role in the molecular pathogenesis of human T-ALL, we analyzed BCL11B status in primary T-ALL patient samples by array CGH and sequencing analysis. Monoallelic BCL11B deletions were identified in 6% of cases (n = 3 of 47) analyzed by array CGH, including one microdeletion within the BCL11B locus, one small deletion involving BCL11B and 6 additional genes, and one large 26 Mbp deletion of the distal arm of chromosome 14. BCL11B sequencing revealed heterozygous missense mutations in an additional 9% (n = 4 of 43) of primary T-ALL patient samples and in 19% (n = 3 of 16) of T-ALL cell lines. Structural homology modeling revealed that many of the mutations identified, including 3 of the 4 in the primary patient samples, disrupted key amino acids within BCL11B zinc finger domains that are involved in DNA recognition or structural stabilization required for zinc finger domain-mediated transcriptional activity. Analysis of TCRγ rearrangement status and gene expression data revealed that most cases with BCL11B inactivation were characterized by biallelic TCRγ rearrangements together with an early thymocyte precursor (ETP) gene expression signature, indicating developmental arrest at a prethymocyte stage of T-cell development, which we and others have shown confers an increased risk of treatment failure (Coustan-Smith et al. Lancet Oncol 2009; Gutierrez et al. J Clin Oncol 2010). Given that BCL11B expression during normal T-cell development increases markedly at the prethymocyte stage, our findings suggest that BCL11B inactivation may be directly responsible for developmental arrest at a prethymocyte stage during thymocyte transformation. Our findings provide compelling evidence that BCL11B is a tumor suppressor in a subset of human T-ALLs with a high risk of treatment failure. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 123, No. 7 ( 2014-02-13), p. 1040-1050
    Abstract: c-Myc is required for leukemia-initiating cell maintenance in murine models of T-ALL. c-Myc inhibition prevents the growth of treatment-resistant primary T-ALL patient samples in vitro.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 910-910
    Abstract: Abstract 910 Despite modern intensive therapy, 25% of children with T-ALL will develop treatment-resistant disease, which carries a dismal prognosis. However, no clinical or biologic features have been found to predict prognosis robustly enough to be incorporated into current clinical protocols. The inability to accurately identify patients at high risk of treatment failure at the time of diagnosis is a major impediment to further improvements in outcome in T-ALL, as patients at high risk of treatment failure, who would probably benefit from the introduction of novel therapeutic agents, cannot be differentiated from those who might be cured with reduced-intensity regimens. In an effort to identify genomic alterations present at the time of diagnosis that would predict response to therapy, we performed array comparative genomic hybridization (CGH) on DNA extracted from diagnostic specimens from 47 children with T-ALL treated on either Children's Oncology Group Protocol 9404 or DFCI ALL Consortium Protocol 00-001, which are very similar regimens. The samples analyzed included all of the specimens available from cases in which therapy failed, comprising 9 induction failure and 13 relapse cases, and a control group of 25 long-term event-free survivors. The absence of deletions of genomic DNA at the TCRg, TCRb, and TCRa/TCRd loci at the time of diagnosis, indicating that V(D)J recombination had not occurred, was associated with induction failure, as was deletion of the CDKN2A tumor suppressor locus. The absence of TCRg and TCRb deletions were also associated with inferior event-free and overall survival. The lack of T cell receptor rearrangements in these cases indicates that PCR-based minimal residual disease testing may not be useful in these high-risk cases. The most robust marker of treatment failure identified, lack of homozygous TCRg rearrangement, strongly predicted induction failure (75% vs. 8%,P = 0.0002), inferior event-free survival (P = 0.0009) and inferior overall survival (P 〈 0.0001) in these patients. Our findings were validated using quantitative DNA PCR, an assay that is readily suited to clinical applications. Analysis of gene expression data demonstrated that the group of cases identified by lack of TCR rearrangement showed significant biologic overlap with the high-risk “early T-cell precursor” subtype of T-ALL recently identified using gene expression profiling. Taken together, these data indicate that a subset of T-ALL patients with a very poor response to contemporary therapy can be accurately identified at the time of diagnosis, raising the possibility that alternative therapies can be administered early in treatment to this high group of patients in order to improve their outcome. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 3802-3802
    Abstract: In an effort to further unravel the molecular pathogenesis of T cell acute lymphoblastic leukemia (T-ALL), we performed array CGH on diagnostic specimens from 47 pediatric patients with T-ALL. The LEF1 transcription factor is best known as a positive mediator of oncogenic β-catenin signaling, and it is required for the expression of MYC, Cyclin D1 and Survivin in some contexts, including some normal hematopoietic progenitors. Additionally, Lef1 has been shown to be required for the survival of murine Notch-dependent T cell lymphoma cells. We were thus surprised to find highly focal deletions of LEF1 in 10.6 % (n = 5 of 47) of primary T-ALL patient samples, with 3 cases harboring homozygous deletions and 2 harboring heterozygous deletions of this locus. These deletions involved no other known genes. We then sequenced this gene in 45 of these cases and found heterozygous mutations in 3 additional T-ALL samples, including 2 cases with frameshift mutations predicted to truncate the protein prior to its context-dependent activation and HMG box domains, and one case with an Asp85Asn substitution. Seven of the eight samples harboring LEF1 abnormalities also had mutations in the heterodimerization or PEST domains of NOTCH1. In an effort to characterize the biologic consequences of LEF1 loss in T-ALL, we took advantage of expression microarrays that were previously performed on most of these samples. LEF1 loss defines a novel T-ALL subtype characterized by arrest at the CD1-positive early cortical stage, and whose gene expression profile resembles that of HOX11-positive cases, although expression of HOX11 was generally low in these samples. Loss of LEF1 was mutually exclusive to overexpression of TAL1 and of the HOXA/MEIS1 cluster. Interestingly, LEF1-negative T-ALL cases were associated with increased expression of MYC, and gene set enrichment analysis identified a significant association between LEF1 loss and upregulation of MYC target genes. In addition to its role as a transcriptional activator in the setting of active WNT/β-catenin signaling, LEF1 can also act as a transcriptional repressor in some cellular contexts. Experiments are currently underway to establish the mechanism mediating the tumor suppressor activity of LEF1 in T-ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 28, No. 24 ( 2010-08-20), p. 3816-3823
    Abstract: To identify children with T-cell acute lymphoblastic leukemia (T-ALL) at high risk of induction chemotherapy failure by using DNA copy number analysis of leukemic cells collected at diagnosis. Patients and Methods Array comparative genomic hybridization (CGH) was performed on genomic DNA extracted from diagnostic lymphoblasts from 47 children with T-ALL treated on Children's Oncology Group Study P9404 or Dana-Farber Cancer Institute Protocol 00-01. These samples represented nine patients who did not achieve an initial complete remission, 13 who relapsed, and 25 who became long-term, event-free survivors. The findings were confirmed in an independent cohort of patients by quantitative DNA polymerase chain reaction (DNA-PCR), an assay that is well suited for clinical application. Results Analysis of the CGH findings in patients in whom induction chemotherapy failed compared with those in whom induction chemotherapy was successful identified the absence of biallelic TCRγ locus deletion (ABD), a characteristic of early thymocyte precursors before V(D)J recombination, as the most robust predictor of induction failure (P 〈 .001). This feature was also associated with markedly inferior event-free (P = .002) and overall survival (P 〈 .001) rates: 25% versus 58% and 25% versus 72%, respectively. Using a rapid and inexpensive quantitative DNA-PCR assay, we validated ABD as a predictor of a poor response to induction chemotherapy in an independent series of patients. Conclusion Lymphoblasts from children with T-ALL should be evaluated at diagnosis for deletion within the TCRγ locus. Patients lacking biallelic deletion, which confers a high probability of induction failure with contemporary therapy, should be assigned to alternative therapy in the context of a prospective clinical trial.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2010
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 7, No. 11 ( 2017-11-01), p. 1336-1353
    Abstract: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes. Using a transgenic screen in zebrafish, thymocyte selection–associated high mobility group box protein (TOX) was uncovered as a collaborating oncogenic driver that accelerated T-ALL onset by expanding the initiating pool of transformed clones and elevating genomic instability. TOX is highly expressed in a majority of human T-ALL and is required for proliferation and continued xenograft growth in mice. Using a wide array of functional analyses, we uncovered that TOX binds directly to KU70/80 and suppresses recruitment of this complex to DNA breaks to inhibit nonhomologous end joining (NHEJ) repair. Impaired NHEJ is well known to cause genomic instability, including development of T-cell malignancies in KU70- and KU80-deficient mice. Collectively, our work has uncovered important roles for TOX in regulating NHEJ by elevating genomic instability during leukemia initiation and sustaining leukemic cell proliferation following transformation. Significance: TOX is an HMG box–containing protein that has important roles in T-ALL initiation and maintenance. TOX inhibits the recruitment of KU70/KU80 to DNA breaks, thereby inhibiting NHEJ repair. Thus, TOX is likely a dominant oncogenic driver in a large fraction of human T-ALL and enhances genomic instability. Cancer Discov; 7(11); 1336–53. ©2017 AACR. This article is highlighted in the In This Issue feature, p. 1201
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 889-889
    Abstract: The tendency of mitochondria to undergo or resist BCL2-controlled apoptosis (so-called mitochondrial priming) is a powerful predictor of response to cytotoxic chemotherapy. Fully exploiting this finding will require unraveling the molecular genetics underlying phenotypic variability in mitochondrial priming. We analyzed pre-treatment T-ALL clinical specimens from a cohort of 47 patients (enriched for treatment failure, but with sufficient controls) treated on the COG AALL0434 or DFCI 05001 clinical trials using BH3 profiling analysis to assess mitochondrial apoptotic priming. We found that there was a strong association between resistance to mitochondrial apoptosis and a poor response to induction chemotherapy (P = 0.008). Furthermore, mitochondrial apoptosis resistance predicted significantly inferior event-free survival (65% vs. 91% at 5 years; P = 0.0376). To define the molecular determinants of this mitochondrial apoptosis resistance, we performed targeted exon sequencing and array CGH copy number analysis. This revealed that loss-of-function mutations in the polycomb repressive complex 2 (PRC2) core subunits (EZH2, EED or SUZ12) were associated with mitochondrial apoptosis resistance (P = 0.007) in clinical specimens. PRC2 is a chromatin modifying complex best known for its role in transcriptional repression, which functions as a tumor suppressor in T-ALL, but whether PRC2 regulates mitochondrial apoptosis is unknown. Using shRNA knockdown in human T-ALL cells, we found that depletion of PRC2 subunits in T-ALL cells induced mitochondrial apoptosis resistance, as assessed by BH3 profiling analysis (P 〈 0.001). PRC2 inactivation also induced resistance to chemotherapy-induced apoptosis (P 〈 0.0001), and increased T-ALL fitness following treatment with the antileukemic drug vincristine (P = 0.0001). Apoptosis resistance upon inactivation of EZH2 (a PRC2 catalytic subunit) was reversed by transduction of wild-type EZH2, but not by an EZH2 point mutant with impaired methyltransferase activity, indicating that this effect is mediated by the enzymatic activity of PRC2. In normal mouse thymocytes, heterozygous deletion of the PRC2 subunits Ezh2 or Eed was sufficient to induce apoptosis resistance in non-transformed double-negative T-cell progenitors (P 〈 0.010), indicating that apoptosis resistance can arise prior to oncogenic transformation. The best-known regulators of mitochondrial apoptosis are BCL2-family genes, but RNA-seq analysis of shRNA knockdown of the PRC2 subunits in a T-ALL cell line revealed that PRC2 did not regulate expression of any of the known BCL2 family members. Instead, PRC2 loss led to upregulation of TRAP1, a mitochondrially localized chaperone of the HSP90 family. TRAP1 upregulation was necessary for induction of apoptosis resistance following PRC2 inactivation, because shRNA knockdown of TRAP1 in the human CCRF-CEM cell line completely blocked induction of apoptosis resistance following PRC2 inactivation (P 〈 0.0001). Moreover, pharmacologic TRAP1 inhibition synergized with the antileukemic drugs dexamethasone and doxorubicin (combination index = 0.37 and 0.42, respectively). To define how PRC2 regulates TRAP1, we performed ChIP-seq analysis, which revealed that TRAP1 regulation by PRC2 is indirect. Combined ChIP-seq and RNA-seq analysis revealed a number of direct targets of PRC2, all of which were tested for their ability to upregulate TRAP1 and induce apoptosis resistance. This showed that the LIM domain transcription factor CRIP2 is a direct target of PRC2 that is necessary and sufficient for regulation of TRAP1, and for induction of apoptosis resistance downstream of PRC2 inactivation. To confirm the relevance of our findings, we used the EZH2 inhibitor GSK126 to inhibit enzymatic activity of PRC2, which revealed that EZH2 normally represses CRIP2 and TRAP1 expression in primary patient-derived xenografts. Finally, we found that increased TRAP1 expression correlates with treatment failure in T-ALL clinical specimens (P = 0.028). Taken together, our findings support a model in which loss of PRC2 induces transcriptional upregulation of its direct target CRIP2, which subsequently activates expression of TRAP1, leading to resistance to chemotherapy-induced mitochondrial apoptosis. Disclosures Aries: Pfizer: Employment. Teachey:Amgen: Consultancy; La Roche: Consultancy. Letai:AstraZeneca: Consultancy, Other: Lab research report; AbbVie: Consultancy, Other: Lab research report; Flash Therapeutics: Equity Ownership; Novartis: Consultancy, Other: Lab research report; Vivid Biosciences: Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 604-604
    Abstract: Although contemporary combination chemotherapy can cure a substantial fraction of patients with T-cell acute lymphoblastic leukemia (T-ALL), front-line therapy fails in 15-20% of children and 50-70% of adults, and these patients have a poor prognosis. Strikingly, half of treatment failure events in childhood T-ALL are induction failure, suggesting pre-existing resistance to chemotherapeutics with distinct molecular targets. The molecular basis for induction failure remains poorly understood. Recent work has shown that mitochondrial apoptosis resistance is a cellular phenotype that predicts chemotherapy failure in some tumor types. However, the molecular mechanisms responsible for the striking variability in chemotherapy response among different patients with seemingly identical tumors remain largely unknown. Using a technique known as BH3 profiling, we analyzed mitochondrial apoptosis sensitivity or resistance in pre-treatment clinical specimens from a cohort of 47 children and adolescents treated on the COG AALL0434 or DFCI 05001 clinical trials. We found that mitochondrial apoptosis resistance was strongly associated with a poor response to induction chemotherapy (P = 0.008), as well as inferior 5-year event-free survival (65% vs 88%; P = 0.036 by log-rank test). Apoptosis resistance was weakly associated with the early T-cell precursor (ETP) immunophenotype (P = 0.08), but univariate and multivariable Cox regression analysis including both revealed that apoptosis resistance predicts clinical outcome more strongly than ETP status. To identify molecular lesions underlying mitochondrial apoptosis resistance, we applied targeted exome sequencing and array CGH to this cohort. We found that loss-of-function mutations in genes encoding core components of the polycomb repressive complex 2 (PRC2), including EZH2, EED or SUZ12, are associated with resistance to mitochondrial apoptosis (P = 0.015). PRC2 is a chromatin-modifying complex best known for its role in transcriptional repression. The PRC2 complex has been implicated as a tumor suppressor in T-ALL, but whether PRC2 plays a direct role in chemotherapy response is unknown. To test whether PRC2 regulates mitochondrial apoptosis in human T-ALL, we performed shRNA knockdown of the PRC2 core components EZH2, EED or SUZ12 in human T-ALL cell lines. Knockdown of each of these genes significantly induced mitochondrial apoptosis resistance, as assessed by BH3-profiling. This effect was dependent on the lysine methyltransferase activity of the PRC2 complex, because the effect of EZH2 knock-down was rescued by expression of wild-type EZH2, but not a point mutant that is methyltransferase-defective (P 〈 0.001). PRC2 knockdown also induced significant resistance to apoptosis induction (assessed using caspase 3/7 activation or annexin V/PI staining) in response to various chemotherapeutics with distinct molecular targets, including vincristine, dexamethasone, asparaginase, methotrexate, mercaptopurine, nelarabine, cytarabine and etoposide. To test whether PRC2 regulates mitochondrial apoptosis during normal T-cell development, we took advantage of mice heterozygous for a floxed Ezh2 or Eed allele, and induced deletion of one allele in hematopoietic cells using Mx-Cre activation by pIpC. Controls were Ezh2 and Eed wild-type mice with Mx-Cre activation. BH3 profiling analysis revealed that loss of one Ezh2 or Eed allele is sufficient to induce apoptosis resistance in non-transformed double-negative thymocytes (P = 0.003 for Ezh2 and P = 0.008 for Eed), suggesting that chemotherapy resistance can develop prior to oncogenic transformation. To define the transcriptional consequences of PRC2 inhibition in T-ALL, we performed RNA sequencing of T-ALL cells infected with shRNAs targeting EZH2, EED or SUZ12 (2 independent hairpins for each gene), or two control shRNAs. RNA sequencing analysis revealed a number of candidate transcriptional targets linking PRC2 to the mitochondrial apoptotic machinery, which are currently being investigated using functional genetics and small molecule inhibitors. Collectively, these data implicate polycomb repressive complex 2 function as a key determinant of chemotherapy response in childhood T-ALL. Defining the mechanism linking PRC2 to the mitochondria will provide a rational target for therapeutic intervention. Disclosures Teachey: Novartis: Research Funding. Letai:AbbVie: Consultancy, Research Funding; Tetralogic: Consultancy, Research Funding; Astra-Zeneca: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...