scholarly journals Oncogenetic landscape and clinical impact of IDH1 and IDH2 mutations in T-ALL

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Mathieu Simonin ◽  
Aline Schmidt ◽  
Christophe Bontoux ◽  
Marie-Émilie Dourthe ◽  
Etienne Lengliné ◽  
...  

AbstractIDH1 and IDH2 mutations (IDH1/2Mut) are recognized as recurrent genetic alterations in acute myeloid leukemia (AML) and associated with both clinical impact and therapeutic opportunity due to the recent development of specific IDH1/2Mut inhibitors. In T-cell acute lymphoblastic leukemia (T-ALL), their incidence and prognostic implications remain poorly reported. Our targeted next-generation sequencing approach allowed comprehensive assessment of genotype across the entire IDH1 and IDH2 locus in 1085 consecutive unselected and newly diagnosed patients with T-ALL and identified 4% of, virtually exclusive (47 of 49 patients), IDH1/2Mut. Mutational patterns of IDH1/2Mut in T-ALL present some specific features compared to AML. Whereas IDH2R140Q mutation was frequent in T-ALL (25 of 51 mutations), the IDH2R172 AML hotspot was absent. IDH2 mutations were associated with older age, an immature phenotype, more frequent RAS gain-of-function mutations and epigenetic regulator loss-of-function alterations (DNMT3A and TET2). IDH2 mutations, contrary to IDH1 mutations, appeared to be an independent prognostic factor in multivariate analysis with the NOTCH1/FBXW7/RAS/PTEN classifier. IDH2Mut were significantly associated with a high cumulative incidence of relapse and very dismal outcome, suggesting that IDH2-mutated T-ALL cases should be identified at diagnosis in order to benefit from therapeutic intensification and/or specific IDH2 inhibitors.

2021 ◽  
Vol 162 ◽  
pp. S48
Author(s):  
Kathryn Miller ◽  
Herman Chui ◽  
Sarah Chiang ◽  
Lora Ellenson ◽  
Britta Weigelt ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 470-470
Author(s):  
Ana Rita Fragoso ◽  
Tin Mao ◽  
Song Wang ◽  
Steven Schaffert ◽  
Hyeyoung Min ◽  
...  

Abstract Abstract 470 MiRNA-mediated gene regulation represents a fundamental layer of post-transcriptional control of gene expression with diverse functional roles in normal development and tumorigenesis. Whereas some studies have shown that over-expression of miRNA genes may contribute to cancer development and progression, it is yet to be rigorously tested by the loss-of-function genetic approaches whether miRNA genes are required for cancer development and maintenance in mice. Here we show that mir-181a1/b1 coordinates Notch and pre-TCR signals during normal thymocyte differentiation and plays an essential role in development and onset of T-cell acute lymphoblastic leukemia (T-ALL) induced by some Notch mutations. Using gain-of-function and loss-of-function approaches, we demonstrated that mir-181a1/b1 controls Notch and pre-TCR receptor signals during the early stages of T cell development in the thymus by repressing multiple negative regulators of both pathways, including Nrarp, PTPN-22, SHP2, DUSP5, and DUSP6. These results illustrate that a single miRNA can coordinate multiple signaling pathways by modulating the timing and strength of signaling at different stages. Intriguingly, synergistic signaling between Notch and pre-TCR pathways is necessary for the development of T-ALL, and miR-181 family miRNAs are aberrantly expressed in T-ALL patients. These observations raise the possibility that mir-181a1/b1 might contribute to the onset or maintenance of T-ALL by targeting similar pathways in tumor cells as it does in normal thymic progenitor cells. In support of this notion, we found that loss of mir-181a1/b1 significantly delayed the onset and development of T-ALL induced by intracellular domain of Notch1 (ICN1) and caused a 32% increase in the median survival time from 41 days to 54 days in T-ALL mice. Importantly, we noted that loss of mir-181a1/b1 more efficiently repressed the leukemogeneic potential of cells with lower levels of ICN1 expression, suggesting that mir-181a1/b1 may be more effective in inhibiting T-ALL development induced by a Notch mutant with weaker signal strength. Indeed, we demonstrated that loss of mir-181a1b1 essentially blocked T-ALL development induced by the weaker Notch mutant and dramatically decreased mortality from 60% to 10% in these T-ALL mice. Since human Notch mutations identified in T-ALL patients generally have weaker signaling strength and lower oncogenic potential than that of ICN1, our findings indicate that mir-181a1/b1 may play an essential role in development of normal thymic progenitors and Notch-induced T-ALL and may be targeted to treat T-ALL patients harboring Notch mutations. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 837-837
Author(s):  
Hamza Celik ◽  
Cates Mallaney ◽  
Alok Kothari ◽  
Christopher A Miller ◽  
Jasreet Hundal ◽  
...  

Abstract Genome sequencing studies of patient samples have implicated the involvement of various components of the epigenetic machinery in myeloid diseases, including the de novo DNA methyltransferase DNMT3A (Cancer Genome Atlas Research, N Engl J Med, 2013). We have recently shown that Dnmt3a is essential for normal hematopoietic stem cell (HSC) differentiation. Genetic ablation of Dnmt3a resulted in HSCs that showed diminished capacity for peripheral blood generation after serial transplantation (on a per-HSC basis), while phenotypically-defined HSCs accumulated in the bone marrow (Challen et al., Nature Genetics, 2012). Although this differentiation arrest was insufficient to cause overt disease, in these competitive transplants the presence of wild-type whole bone marrow may have suppressed malignant transformation of the mutant HSCs. Dnmt3a-null HSCs were less proliferative than counterpart control HSCs in this transplantation setting, suggesting that the cellular turnover threshold necessary to generate additional genetic and/or epigenetic lesions required for leukemogenesis was not achieved. To further understand the contribution of Dnmt3a loss-of-function in hematopoiesis, we performed non-competitive transplantation of Dnmt3a-null bone marrow. This forces the mutant HSCs to divide in vivoto regenerate the hematopoietic system following lethal irradiation, and should uncover any predispositions to transformation. Mice transplanted with Dnmt3a-null bone marrow in the absence of wild-type support cells succumbed principally to bone marrow failure (median survival 328 days) characteristic of myelodysplastic syndromes (MDS) with symptoms including anemia, neutropenia, bone marrow hypercellularity and splenomegaly with myeloid infiltration. 2/25 mice developed myeloid leukemia with >20% blasts in the blood and bone marrow. 4/25 primary mice succumbed to myeloproliferative disorders, some of which progressed to secondary leukemia after long latency. Exome sequencing was performed to identify co-operating mutations that drove leukemic transformation, and revealed c-Kit mutations found only in the Dnmt3a-null AML samples. As DNMT3A and KIT mutations can co-occur in AML and mastocytosis, we tested whether these two pathways could co-operate in vivo by ectopic introduction of c-Kit variants into hematopoietic progenitors followed by bone marrow transplantation (Figure 1). As previously reported, expression of c-KitD814V in wild-type cells lead to development of B-cell acute lymphoblastic leukemia (B-ALL). However, expression of c-KitD814V in a Dnmt3a-null background lead to acute leukemia with a much shorter latency (median survival 67 days), implicating a synergism between these pathways in vivo. Moreover, the absence of Dnmt3a also distorted the spectrum of leukemia resulting from enforced c-Kit signaling. While some of the mice transplanted with Dnmt3a-null c-KitD814V cells also succumbed to a B-ALL, 4/13 (31%) developed mastocytosis with involvement of myeloid blasts, and 4/13 (31%) mice developed a T-cell acute lymphoblastic leukemia (T-ALL). We show for the first time that these pathways can co-operate to accelerate transformation in vivo. This Dnmt3a/c-Kit disease model resembles the classical “two-hit” model of leukemogenesis in which one mutation in a hematopoietic progenitor cell inhibits differentiation (Dnmt3a loss-of-function), whilst another drives proliferation (c-Kit gain-of-function). Such mouse models present a unique opportunity to study the sequence of early events leading to HSC transformation following Dnmt3a-inactivation. Figure 1 Kaplan-Meier survival curve of mice transplanted with control or Dnmt3a-KO bone marrow progenitor cells transduced with a lentivirus expressing c-KitD814V. *** p <0.001. Figure 1. Kaplan-Meier survival curve of mice transplanted with control or Dnmt3a-KO bone marrow progenitor cells transduced with a lentivirus expressing c-KitD814V. *** p <0.001. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4076-4076
Author(s):  
Wei-Na Zhang ◽  
Ming Zhang ◽  
Jin-yan Huang ◽  
Zhu Chen ◽  
Sai-Juan Chen

Abstract Background. In previous study of adult and childhood B-cell acute lymphoblastic leukemia, through whole exome and transcriptome sequencing we identified three new categories of fusion genes with prognostic significance and much more mutations of epigenetic regulators in adult versus pediatric B-ALL cases. To address how these gene alterations contribute to leukemogenesis and affect prognosis, we performed detailed data analysis and functional study of the three new categories of B-ALL. In addition, the changes of epigenetic regulator were scrutinized in a systematic way. Methods. Differentially expressed genes analysis and gene set enrichment analysis (GSEA) were performed. The regulation of target genes by distinct fusions was further verified through lentivirus mediated overexpression, shRNA induced knockdown, quantitative real-time PCR as well as transcriptional reporter assays. The modification of histone acetylation was evaluated by Western bloting. To observe the effect of fusion genes in vivo, murine retroviral bone marrow transplantation models were established. Results. Through in-depth data analysis, differentially expressed genes and signaling pathways related to fusion genes were identified. Alternative splicing of transcription factor ERG was seen in most of the cases with DUX4 fusions. In some of the cases, both ERG deletion leading to frameshift of the open reading frame and an abnormal short transcript of ERG were also observed. This group can also be characterized by some outlier genes (AGAP1, STAP1, PTPRM1, PCDH17 and PLEKHA6). Overexpression of DUX4-IGH in human B-ALL cell line induced the appearance of the short form of ERG and upregulation of these outlier genes. In mouse models, both DUX4-IGH and the short ERG inhibited the development of B-cell lineage. NT5E, also known as CD73, which has been reported to promote anthracycline resistance, was significantly downregulated in this group, which could explain the favorable prognosis of this type of B-ALL. Pre-BCR signaling pathway maintaining the survival of leukemic cells at the pre-B stage was activated in patients with MEF2D fusions. Genes related to protein amimo acid phosphorylation and cell morphogenesis involved in differentiation were significantly upregulated in this group. Notably, this group exhibited rather poor survival rate, and the expression of CD73 in this group was as high as that in BCR-ABL1 and BCR-ABL1 like group. When juxtaposed to the upstream of ZNF384, EP300 and CREBBP lost the histone acetylation domain, which could reduce the histone acetylation level and be involved in the leukemogenesis. Indeed, EP300-ZNF384 induced the rapid emergence of acute leukemia in mouse with a median survival of 100 days. In addition, we checked the sequence variations in our whole exome/genome dataset containing 203 cases. When mutations of different gene categories were scrutinized, abnormalities of epigenetic regulators (ER) drew our attention. Using the ER database that St. Jude Children's Research Hospital established, we found a total of 140 somatic mutations in 80 ER classified into eight classes. Forty two percent of the 203 patients (56.5% of adult and 30.6% of children) harbored at least one gene mutation among the eight classes of ER. The fact that abnormalities of MLL2 and SETD2, the two ER genes with highest mutation rates, were mutually exclusive in most affected cases suggests that they should bear somehow functional resemblance. Conclusions. Detailed study of MEF2D, ZNF384 and DUX4-IGH gene fusions expands the extant knowledge about molecular subtypes of B-ALL, and provides more potential targets as well as promotes further personalized treatment in both childhood and adult B-ALL. The overview of ER alterations in adult and childhood B-ALL proves the importance of these genes in the pathogenesis of B-ALL, especially in adult cases. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Guillaume P Andrieu ◽  
Milena Kohn ◽  
Mathieu Simonin ◽  
Charlotte Smith ◽  
Agata Cieslak ◽  
...  

T-cell acute lymphoblastic leukemia (T-ALL) are aggressive hematological cancers with dismal outcomes, and are in need of new therapeutic options. Polycomb Repressor Complex 2 (PRC2) loss-of-function alterations were reported in pediatric T-ALL; yet their clinical relevance and functional consequences remain elusive. Here, we extensively analyzed PRC2 alterations in a large series of 218 adult T-ALL patients. We found that PRC2 genetic lesions are frequent events in T-ALL and are not restricted to ETP-ALL. PRC2 loss of function associates with activating mutations of the IL7R/JAK/STAT pathway. PRC2-altered T-ALL patients poorly respond to prednisone, have low bone marrow blast clearance, and persistent minimal residual disease. Furthermore, we identified that PRC2 loss of function profoundly reshapes the genetic and epigenetic landscapes, leading to the reactivation of stem cell programs that cooperate with Bromodomain and Extraterminal (BET) proteins to sustain T-ALL. This study identifies BET proteins as key mediators of the PRC2 loss of function-induced remodeling. Our data has uncovered a targetable vulnerability to BET inhibition that can be exploited to treat PRC2-altered T-ALL patients.


Blood ◽  
2009 ◽  
Vol 114 (5) ◽  
pp. 1038-1045 ◽  
Author(s):  
Valeria Tosello ◽  
Marc R. Mansour ◽  
Kelly Barnes ◽  
Maddalena Paganin ◽  
Maria Luisa Sulis ◽  
...  

The molecular mechanisms involved in disease progression and relapse in T-cell acute lymphoblastic leukemia (T-ALL) are poorly understood. We used single nucleotide polymorphism array analysis to analyze paired diagnostic and relapsed T-ALL samples to identify recurrent genetic alterations in T-ALL. This analysis showed that diagnosis and relapsed cases have common genetic alterations, but also that relapsed samples frequently lose chromosomal markers present at diagnosis, suggesting that relapsed T-ALL emerges from an ancestral clone different from the major leukemic population at diagnosis. In addition, we identified deletions and associated mutations in the WT1 tumor suppressor gene in 2 of 9 samples. Subsequent analysis showed WT1 mutations in 28 of 211 (13.2%) of pediatric and 10 of 85 (11.7%) of adult T-ALL cases. WT1 mutations present in T-ALL are predominantly heterozygous frameshift mutations resulting in truncation of the C-terminal zinc finger domains of this transcription factor. WT1 mutations are most prominently found in T-ALL cases with aberrant rearrangements of the oncogenic TLX1, TLX3, and HOXA transcription factor oncogenes. Survival analysis demonstrated that WT1 mutations do not confer adverse prognosis in pediatric and adult T-ALL. Overall, these results identify the presence of WT1 mutations as a recurrent genetic alteration in T-ALL.


2018 ◽  
Vol 66 (1) ◽  
pp. e27496 ◽  
Author(s):  
Ting-Chi Yeh ◽  
Der-Cherng Liang ◽  
Hsi-Che Liu ◽  
Tang-Her Jaing ◽  
Shih-Hsiang Chen ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-5
Author(s):  
Salah Aref ◽  
Mohamed El Agdar ◽  
Nada Khaled ◽  
Lamyaa Ibrahim ◽  
Mohamed S. El-Ghonemy

This study aimed to determine the clinical impact of CD25+/CD123+ coexpression in adult B-cell acute lymphoblastic leukemia (B-ALL) cases. One hundred and twenty newly diagnosed B-ALL patients (≤60 years old) were included in this study. CD123 and CD25 expression on leukemic blast cells were assessed using flow cytometry. CD25+/CD123+ coexpression was detected in 40/120 B-ALL patients (33.3%). All B-ALL patients showed CD25+/CD123+ coexpression had lower induction of remission response and shorter overall survival as compared to B-ALL cases lacking coexpression. In conclusion, CD25+/CD123+ positive coexpression is a reliable flow cytometry marker for prediction of the outcome of adult B-ALL patients and could be used as a novel parameter for risk stratification of adult B-ALL cases.


2020 ◽  
Vol 48 (7) ◽  
pp. 3496-3512 ◽  
Author(s):  
Francesca Ferrante ◽  
Benedetto Daniele Giaimo ◽  
Marek Bartkuhn ◽  
Tobias Zimmermann ◽  
Viola Close ◽  
...  

Abstract Aberrant Notch signaling plays a pivotal role in T-cell acute lymphoblastic leukemia (T-ALL) and chronic lymphocytic leukemia (CLL). Amplitude and duration of the Notch response is controlled by ubiquitin-dependent proteasomal degradation of the Notch1 intracellular domain (NICD1), a hallmark of the leukemogenic process. Here, we show that HDAC3 controls NICD1 acetylation levels directly affecting NICD1 protein stability. Either genetic loss-of-function of HDAC3 or nanomolar concentrations of HDAC inhibitor apicidin lead to downregulation of Notch target genes accompanied by a local reduction of histone acetylation. Importantly, an HDAC3-insensitive NICD1 mutant is more stable but biologically less active. Collectively, these data show a new HDAC3- and acetylation-dependent mechanism that may be exploited to treat Notch1-dependent leukemias.


Blood ◽  
2020 ◽  
Vol 136 (13) ◽  
pp. 1520-1534 ◽  
Author(s):  
Chunhua Song ◽  
Zheng Ge ◽  
Yali Ding ◽  
Bi-Hua Tan ◽  
Dhimant Desai ◽  
...  

Abstract High-risk B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive disease, often characterized by resistance to chemotherapy. A frequent feature of high-risk B-ALL is loss of function of the IKAROS (encoded by the IKZF1 gene) tumor suppressor. Here, we report that IKAROS regulates expression of the BCL2L1 gene (encodes the BCL-XL protein) in human B-ALL. Gain-of-function and loss-of-function experiments demonstrate that IKAROS binds to the BCL2L1 promoter, recruits histone deacetylase HDAC1, and represses BCL2L1 expression via chromatin remodeling. In leukemia, IKAROS’ function is impaired by oncogenic casein kinase II (CK2), which is overexpressed in B-ALL. Phosphorylation by CK2 reduces IKAROS binding and recruitment of HDAC1 to the BCL2L1 promoter. This results in a loss of IKAROS-mediated repression of BCL2L1 and increased expression of BCL-XL. Increased expression of BCL-XL and/or CK2, as well as reduced IKAROS expression, are associated with resistance to doxorubicin treatment. Molecular and pharmacological inhibition of CK2 with a specific inhibitor CX-4945, increases binding of IKAROS to the BCL2L1 promoter and enhances IKAROS-mediated repression of BCL2L1 in B-ALL. Treatment with CX-4945 increases sensitivity to doxorubicin in B-ALL, and reverses resistance to doxorubicin in multidrug-resistant B-ALL. Combination treatment with CX-4945 and doxorubicin show synergistic therapeutic effects in vitro and in preclinical models of high-risk B-ALL. Results reveal a novel signaling network that regulates chemoresistance in leukemia. These data lay the groundwork for clinical testing of a rationally designed, targeted therapy that combines the CK2 inhibitor, CX-4945, with doxorubicin for the treatment of hematopoietic malignancies.


Sign in / Sign up

Export Citation Format

Share Document