Genetic Events in Ikaros Gene Family (IKZF1 and IKZF2) in Pediatric T-Cell Lymphoblastic Leukemia/Lymphoma

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4622-4622
Author(s):  
Atsushi Narita ◽  
Hideki Muramatsu ◽  
Nao Yoshida ◽  
Hirotoshi Sakaguchi ◽  
Sayoko Doisaki ◽  
...  

Abstract Abstract 4622 The Ikaros gene family, which includes Ikaros (IKZF1), Helios (IKZF2), and Aiolos (IKZF3), is a primary regulator of lymphocyte differentiation and is involved in the development of acute lymphoblastic leukemia (ALL). In particular, loss of IKZF1 is primarily associated with T-cell ALL (T-ALL) development in mouse models. Moreover, Helios expression has been detected in early T-cell lineage and is involved in human lymphoid proliferative disease. We sought to determine whether the Ikaros gene family is associated with leukemogenesis in T-ALL and T-cell non-Hodgkin lymphoma (T-NHL) in Japanese children. Fifty-eight Japanese children (39 boys and 19 girls; age, <18 years) with T-ALL (n = 50) and T-NHL (n = 8) were evaluated. The median age at diagnosis was 8 years (1–15 years). Samples were collected at Japanese Nagoya First Red Cross Hospital and Nagoya University Hospital. Blasts from bone marrow (n = 40), peripheral blood (n = 9), pleural effusions (n = 6), and lymph nodes (n=3) were subjected to DNA extraction. All coding exons in IKZF1 were amplified by polymerase chain reaction using the genomic DNA of patients with T-ALL/T-NHL. After amplification using a BigDye Terminator Cycle Sequencing kit (Life Technologies, Carlsbad, CA, USA) in the second step, products were analyzed in an ABI/PRISM 3130xl Genetic Analyzer (Life Technologies). Multiplex ligation-dependent probe amplification (MLPA) analyses were performed using a SALSA MLPA reaction mixture according to the manufacturer's protocol [P202 kit; IKZF1(IKAROS); MRC-Holland, Amsterdam, Netherlands]. Amplification products were quantified and identified by capillary electrophoresis on an ABI/PRISM 3130xl Genetic Analyzer. Informed consent was received from parents of the patients. Approval for this study, including molecular analyses, was obtained from the institutional ethics committee of Nagoya University Graduate School of Medicine. An IKZF1 mutation was found in only 1 of 59 (2%) patients (exon 5, c.476 A>G, p.N159S; patient number 41) who was diagnosed with primary immune deficiency after birth. She received regular immunoglobulin replacement therapy just before the development of T-ALL at 13 years of age. Her IKZF1 mutation was confirmed as a germline mutation by sequencing of genomic DNA from her fingernails. In the IKZF1 null mouse model, Ikaros inactivation is closely linked to T-ALL development, which is highly associated with acquisition of a somatic NOTCH1 mutation. As in the mouse model, we could identify a somatic NOTCH1 mutation (exon 26, c.4750, insGAG) in the sample upon T-ALL development. IKZF1 mutations were not found in the other 57 samples. This finding implied that the IKZF1 mutation was a rare genetic event in de novo T-ALL/T-NHL in children. Furthermore, we analyzed all samples by MLPA to determine if monoallelic genomic deletions were present. Although 59 samples exhibited normal biallelic IKZF1 and IKZF3 chromosomal regions, we identified IKZF2 microdeletions in 2 (3.4%) patients (numbers 9 and 49). One of them with an IKZF2 microdeletion died of leukemia relapse after allogeneic bone marrow transplantation from a human leukocyte antigen (HLA)-matched sibling donor. Overexpression of full-length Helios (IKZF2) blocks αβ T-cell differentiation at the CD4-CD8- stage in the thymus, and results in the increased frequencies of γδ T and natural killer (NK) cells in peripheral lymphoid organs. Overexpression of dominant negative Helios leads to increased T-cell proliferation upon T-cell receptor stimulation and the development of T lymphoma. Furthermore, dominant negative Helios isoforms or allelic losses have been detected in some human T-ALL/T-NHL. These results suggest that Helios is an essential regulator of T-cell homeostasis and a tumor suppressor. In general, IKZF1 mutation or deletion is a rare event in pediatric patients with T-ALL/T-NHL. We could identify a secondary T-ALL patient after primary immunodeficiency who shared genetic findings with an IKZF1 null mouse model (germline IKZF1 mutation and somatic NOTCH1 mutation). We found IKZF2 microdeletion in 2 of 59 patients. The genetic events in the Ikaros gene family (IKZF1 and IKZF2) are associated with leukemogenesis in a few pediatric patients T-ALL/T-NHL. Disclosures: No relevant conflicts of interest to declare.

2012 ◽  
Vol 53 (7) ◽  
pp. 1425-1428 ◽  
Author(s):  
Monika D. Kraszewska ◽  
Małgorzata Dawidowska ◽  
Maria Kosmalska ◽  
Łukasz Sędek ◽  
Władysław Grzeszczak ◽  
...  

Blood ◽  
2016 ◽  
Vol 127 (15) ◽  
pp. 1907-1911 ◽  
Author(s):  
Cristina Mirantes ◽  
Maria Alba Dosil ◽  
David Hills ◽  
Jian Yang ◽  
Núria Eritja ◽  
...  

Key Points CD45-driven expression of Cre generates the first mouse model that allows specific and exclusive deletion of Pten in hematopoietic cells. Pten deletion in CD45-expressing cells causes T-cell acute lymphoblastic leukemia, but no other hematologic malignancies.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1835-1835
Author(s):  
Joachim R. Gothert ◽  
Rachael Brake ◽  
C. Glenn Begley ◽  
David J. Izon

Abstract The acquired activation of stem cell leukemia (SCL) during T-lymphopoiesis is a common event in T-cell acute lymphoblastic leukemia. Here, we generated a novel tamoxifen-inducible transgenic mouse model (lck-ERT2-SCL) to study the cellular targets of acquired SCL activation during T-cell development. Upon tamoxifen treatment we observed the thymic emergence of abnormal, non-cycling CD8+TCRβlow and immature CD4+CD8+ (double-positive, DP) cells displaying increased viability. Unexpectedly, fetal thymic organ culture analysis of lck-ERT2-SCL thymi revealed the development of DP cells before the emergence of CD8+TCRβlow cells, which implied the derivation of CD8+TCRβlow cells from DPs rather than immature CD8 single-positive (SP) thymocytes. Interestingly, histone deacetylase (HDAC) inhibition with trichostatin A (TSA) had a divergent effect on SCL perturbed thymopoiesis: TSA increased T-cell receptor surface expression within DP and CD8 SP cells however did not alter the CD8 shifted CD4/CD8-ratio. Furthermore, we studied the expression of NOTCH1 in SCL induced TCRβlow thymocytes. Strikingly, we found that SCL induced NOTCH1-upregulation in DP TCRβlow cells. We therefore conclude that SCL promotes the emergence of abnormal CD8+TCRβlow cells by an only partially HDAC dependent mechanism from DP TCRβlow cells. Moreover, SCL induced DP TCRβlow cells are characterized by upregulated NOTCH1, which in turn might promote the effect of acquired NOTCH1 mutations during T-leukemogenesis.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 7-7
Author(s):  
Mark Y Chiang ◽  
M. Eden Childs ◽  
Candice Romany ◽  
Olga Shestova ◽  
Jon Aster ◽  
...  

Abstract Abstract 7 Notch signaling is activated in ∼70% of human T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) samples and many human and mouse T-ALL cell lines require Notch signals for growth and survival. To gain insight into the role of Notch during induction of T-ALL, we used a fully penetrant, conditional, transgenic KrasG12D mouse model in which ∼80% of T-ALLs acquire activating Notch1 mutations in the endogenous locus. We crossed mice bearing this transgene with Rosa26-DNMAMLf/f mice, which conditionally express the pan-Notch inhibitor DNMAML. T-ALL developed in these mice despite the expression of DNMAML throughout T-cell development. ∼75% of T-ALL tumors acquired activating Notch1 mutations and suppressed expression of DNMAML, which is consistent with frequent “escape” of Notch from inhibition for efficient T-ALL development. We next compared T-ALL cells that lacked DNMAML expression with T-ALL cells that continued to express DNMAML. T-ALL cells lacking DNMAML expressed the direct Notch target c-Myc at higher levels, proliferated at a higher rate, and contained ∼10-fold higher levels of leukemia-initiating cells. Moreover, DNMAML-positive T-ALLs lost DNMAML after transfer into secondary recipients. These data underscore the strong selective pressure for Notch signals during generation and maintenance of T-ALL. We next sought a mechanistic answer for the strong selective pressure for Notch activation. c-Myc and Akt have both been posited to be critical targets of oncogenic Notch signals. To compare the relative contributions of c-Myc and Akt to lymphomagenesis, we overexpressed c-Myc and activated AKT in the KrasG12D-driven mouse model. T-ALLs induced by KrasG12D and Akt acquired activating Notch1 mutations in ∼70% of tumors, which were sensitive to Notch inhibitors (gamma-secretase inhibitors [GSI]). In contrast, T-ALLs induced by KrasG12D and c-Myc did not acquire Notch1 mutations and were resistant to GSI. We conclude that upregulation of c-Myc is sufficient to substitute for Notch in lymphomagenesis, whereas activation of Akt signaling is not. These data identify c-Myc not AKT as the driving force behind Notch-induced lymphomagenesis. These data emphasize the Notch/c-Myc axis as an attractive, rational, therapeutic target in T-ALL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1419-1419
Author(s):  
Kenji Tokunaga ◽  
Shunichiro Yamaguchi ◽  
Eisaku Iwanaga ◽  
Tomoko Nanri ◽  
Taizo Shimomura ◽  
...  

Abstract Abstract 1419 Aims: Molecular pathogenesis of acute lymphoblastic leukemia (ALL) has largely been verified in pediatric patients and the identification of genetic alterations have contributed to stratifying therapeutic applications. In adult patients with ALL, cytogenetic and genetic abnormalities have not sufficiently been elucidated and therapeutic improvement has been hindered. CREB binding protein (CREBBP) is a transcriptional coactivator that interacts with a diverse range of transcription factors and regulates transcription by histone acetylation in hematopoiesis. Mutations of the CREBBP gene are recently found in approximately 2–4% of pediatric patients with ALL. Especially in relapsed cases, the mutations prevail (18–63%) and are possible markers for prediction of relapse in pediatric ALL. In adult patients with ALL, the clinical significance of CREBBP mutations remains to be determined. Here we examined adult ALL patients in an attempt to determine the incidence, clinical characteristics and prognostic impact of the CREBBP mutations. Methods: We investigated 71 adult patients with newly diagnosed ALL treated with JALSG protocols between 1986 and 2010. Age ranged from 15 to 86 years, with a median of 54 years. CREBBP mutations are dominantly identified in histone acetyltransferase (HAT) domain. HAT domain in the CREBBP gene was amplified with RT-PCR using RNA isolated from the peripheral blood or bone marrow mononuclear cells at diagnosis and was subjected to direct sequencing. We compared clinical profiles between patients with and without CREBBPHAT domain mutations. This study was approved by the Institutional Review Boards and informed consent was obtained from each patient according to guidelines based on the revised Declaration of Helsinki. Results: CREBBP HAT domain mutations were detected in 8 of 71 (11.3%) patients: one nonsense mutation, five insertion mutations with frameshifts, and five missense mutations. Two patients harbored biallelic mutations. The mutations at diagnosis in adult patients were seen more frequently than those in pediatric patients ever reported. Such mutations were not completely identical to those detected in pediatric ALL, but were seen in the region within the HAT domain, indicating that such mutations are loss-of-function mutations. The mutations were found in both B-cell (6/53: 11.3%) and T-cell (1/9: 11.1%) ALL, and distributed in patients harboring IKZF1 alterations (3/31: 9.7%) or the BCR-ABL fusion gene (2/19: 10.5%). There were no statistical difference in age, sex, leukocyte, platelet counts and complete remission rate between patients with and without the CREBBP HAT domain mutations. Patients with the mutations had a trend with worse cumulative incidence of relapse (P=0.4637), relapse-free survival (P=0.4195) and OS (P=0.2349) compared to patients lacking the mutations, but statistical significance was not detected in this small cohort. Conclusions: CREBBP HAT domain mutations at diagnosis in adult ALL are found more frequently than in pediatric ALL. This may be one of the mechanisms that adult ALL has been associated with poor OS compared with pediatric ALL. In this study, CREBBP HAT domain mutations were observed in various subtypes of ALL: both B-cell and T-cell ALL, and both Philadelphia chromosome positive and negative ALL. In pediatric ALL, CREBBP mutations were frequently seen in relapsed patients but not in previously untreated patients. These observations suggest that CREBBP mutations play an important role in an additional late event(s) leading to the development and progression of ALL. Our study implies the possibility that mutations of the CREBBP gene are associated with the pathogenesis and prognostic marker of adult ALL and represent specific epigenetic modifiers in adult ALL, serving as potential therapeutic targets. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2418-2418
Author(s):  
Lori A. Ehrlich ◽  
Katherine S. Yang-Iott ◽  
Amy DeMicco ◽  
Craig H. Bassing

Abstract Abstract 2418 Acute lymphoblastic leukemia (ALL) is diagnosed in approximately 2500 children per year. Although high cure rates have been achieved for ALL, these cancers account for the highest number of non-brain tumor cancer-related deaths in children. T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of immature TCRβ−CD4+/CD8+ T-cells that represents ∼15% of pediatric ALL diagnoses, comprises most of the therapy-resistant ALL tumors, and exhibits a high frequency of relapse. The Ataxia Telangiectasia mutated (ATM) protein kinase activates the cellular response to DNA double strand breaks (DSBs) to coordinate DNA repair with cell survival, proliferation, and differentiation. Somatic inactivating ATM mutations occur in 10–20% of T-ALL and T cell lymphoblastic lymphoma (T-LL) tumors and are associated with resistance to genotoxic chemotherapy drugs and therapy relapse, likely driven by increased genomic instability in cells lacking functional ATM. The impaired DSB response of ATM-deficient cells can be exploited to design combinations of genotoxic drugs that specifically kill these cells in vitro. However, the in vivo potential of such drug combinations to treat T-ALL have not been reported. We sought to develop a pre-clinical mouse model that could be used to test effectiveness of such drug combinations to treat T-ALLs and T-LLs with somatic ATM inactivation. Although germline ATM-deficient (Atm−/−) mice succumb by six months of age to immature CD4+/CD8+ T-cell lymphomas containing genomic instability analogous to human T-ALL tumors, we sought a more physiologic model that would avoid potential complications due to ATM-deficiency in thymic epithelial cells. Thus, we generated and characterized VavCre:Atmflox/flox mice with conditional Atm inactivation restricted to hematopoietic cell lineages. These mice contain reduced numbers of TCRβ−CD4+/CD8+, TCRβ+CD4+/CD8−, and TCRβ+CD4−/CD8+ thymocytes and of TCRβ+CD4+ and TCRb+CD8+ splenic T-cells, mirroring the phenotype of Atm−/− mice. We have found that VavCre:Atmflox/flox mice succumb at an average of 95 days (range 53–183 days) to clonal TCRβ−CD4+/CD8+ or TCRβ+CD4−/CD8+ thymic lymphomas. Evaluation of the bone marrow in a subset of these mice indicates that the lymphoma has disseminated and are classified as leukemia. Our initial cytogenetic analyses of these tumors indicate that they contain both clonal translocations involving chromosome 12 and/or chromosome 14 and deletion of one allelic copy of the haploinsufficient Bcl11b tumor suppressor gene. Hemizygous BCL11B inactivation occurs in ∼20% of human T-ALL tumors, indicating the clinical relevance of VavCre:Atmflox/flox mice as a model for human T-ALL. Our ongoing studies include complete cytogenetic and molecular characterization of VavCre:Atmflox/flox tumors and in vivo testing of chemotherapeutics targeting the Atm pathway in this mouse model of T-ALL/T-LL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 91 (3) ◽  
pp. 1016-1020 ◽  
Author(s):  
Betty Gardie ◽  
Jean-Michel Cayuela ◽  
Sophie Martini ◽  
François Sigaux

We have previously shown that the disruption/deletion of the MTS(MTS1-MTS2) locus due to illegitimate V(D)J recombinase activity is a genetic event characteristic of T-cell acute lymphoblastic leukemia (T-ALL). Inactivation of the p16INK4a tumor suppressor protein, encoded by MTS1, is thought to be the major functional consequence of these chromosomal rearrangements. The two other cell cycle inhibitors encoded by genes identified in the locus (p19ARF by MTS1 and p15INK4b by MTS2), also represent possible candidates for inactivating events. By analyzing p16INK4aexpression in three cases in which an identical 36-kb deletion had deleted MTS2 and disrupted the p19ARF, but spared the p16INK4aMTS1 encoding exons, we have excluded p16INK4a and pinpointed p19ARF and/or p15INK4b as the functional target(s) of this rearrangement. Moreover, by the study of the MTS genomic configuration of 149 rearranged alleles from a large series of T-ALL cases, we have shown that p19ARF encoding exons were always disrupted or deleted, whereas p16INK4a and p15INK4b encoding exons were spared in four and 21 cases, respectively. These results suggest that p19ARF may be targeted by the genetic events that occur in the MTS locus in the majority of T-ALLs.


Sign in / Sign up

Export Citation Format

Share Document