Wilms' Tumor 1 (WT1) Gene Mutations in Pediatric T-Acute Lymphoblastic Leukemia.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3075-3075
Author(s):  
Aline Renneville ◽  
Sophie Kaltenbach ◽  
Emmanuelle Clappier ◽  
Sandra Collette ◽  
Jean-Baptiste Micol ◽  
...  

Abstract Abstract 3075 Poster Board III-12 The Wilms' tumor 1 (WT1) gene, located at chromosome band 11p13, encodes a transcriptional regulator involved in normal hematopoietic development. WT1 mutations have been identified in approximately 10 % of acute myeloid leukemia (AML), where it has recently been found to predict poor outcome, but also in T-cell acute lymphoblastic leukemias (T-ALL). Our aim was to evaluate the frequency, the main associated features and the prognostic significance of WT1 mutations in a cohort of pediatric patients with T-ALL treated according to EORTC-CLG trials. A total of 146 children, aged 7 months to 17 years, with newly diagnosed T-ALL were included in this study. Patients were treated according to 2 consecutive EORTC trials: 58 881 and 58 951. Immunophenotypic subtypes were classified according to the EGIL. Standard karyotype as well as molecular screening of HOX11/TLX1, HOX11L2/TLX3 and HOXA10 overexpression, SIL-TAL, NUP214-ABL, CALM-AF10 fusions were performed at diagnosis. WT1 transcript level was quantified by real-time PCR (RQ-PCR). Mutations of NOTCH1 exons 26, 27, 34, FBXW7 exons 9, 10, and WT1 exon 7, 9 were screened by direct sequencing. At least one WT1 mutation was found at diagnosis in 15 out of 146 (10%) T-ALL. WT1 mutations were predominantly exon 7 frameshift mutations (14/15 cases), consisting of small duplications, deletions or combined insertions/deletions, and were predicted to result in the production of a truncated protein missing the normal zinc finger domain. The remaining mutated patient harbored a somatically acquired missense mutation in exon 9 (C388Y), previously described in Denys-Drash syndrome. Only 4 out of 15 (27%) patients had 2 WT1 mutations and all WT1 mutations identified showed retention of the wild-type allele. Clonal evolution was investigated by analysis of 12 diagnostic-relapse pairs. Identical WT1 mutation was found at relapse in 3/4 mutated patients whereas 1/4 patients acquired an additional WT1 exon 7 mutation at relapse. One of the 8 patients with WT1 wild-type T-ALL at diagnosis acquired a WT1 exon 7 mutation at relapse. WT1 mutated and wild-type patients did not significantly differ in terms of age, gender, white blood cell count, or mediastinal involvement. Interestingly, WT1 mutated patients had significantly higher WT1 mRNA expression levels (median: 84% [25-837] for WT1 mutants vs 17% [0.007-657] for WT1 wild-type cases, p=0.005). This is in line with the trend for earlier developmental stage arrest observed in our WT1 mutated T-ALL as compared with WT1 wild-type T-ALL. Indeed, WT1 is preferentially expressed in immature hematopoietic progenitors and down-regulated in more differentiated cells. No association was found between the presence of WT1 mutations and NOTCH1 activating lesions. WT1 mutation was associated with HOX genes deregulation. HOX11 or HOX11L2 were overexpressed in 10/15 (67%) WT1 mutated ALL versus 29/123 (24%) WT1 wild-type ALL (p=0.001). In addition, HOXA overexpression and MLL-AF6 were found in one WT1 mutated T-ALL each. Overall, HOX deregulation was demonstrated in 12/15 (80%) WT1 mutated ALL at diagnosis and was also found in the T-ALL that acquired WT1 mutation at relapse. Despite being subclonal lesions strongly associated with HOX11 and HOX11L2 overexpression in T-ALL, WT1 mutations and NUP214-ABL fusion were found independent. A possible impact of WT1 mutation on outcome was investigated. The incidence of very high risk features was similar for patients with WT1 mutated and wild-type T-ALL. No significant differences were found between the WT1 mutated and wild-type group regarding 5-year event free survival (71.6% vs 74.1%; Wald test stratified for protocol: p=0.8) and overall survival (81.8% vs 81.3%; p=0.9). Notably, HOX112 overexpression, which is found in half of WT1 mutated T-ALL, has no pejorative impact either on outcome in EORTC trials. In conclusion, our study confirms that the type and incidence of WT1 mutations are very similar in pediatric T-ALL and AML, although the frequency of bi-allelic alterations may be lower in T-ALL. However, in contrast with AML, no pejorative outcome was associated with WT1 mutation. Moreover, we found that WT1 mutations are highly associated with direct or indirect aberrant HOX genes expression. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5291-5291
Author(s):  
Paulo Vidal Campregher ◽  
Roberta Cardoso Petroni ◽  
Nair Muto ◽  
Juliana Nogueira M. Rodrigues ◽  
Roberta Sitnik ◽  
...  

Abstract NOTCH1 is a proto-oncogene with activating mutations described in a variety of malignancies, including acute lymphoblastic leukemia (ALL), mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL). While the prognostic significance of NOTCH1 mutations remains controversial in ALL, recent data suggest that NOTCH1 PEST domain mutations are associated with adverse prognosis in patients with CLL. NOTCH1 mutations are found in around 8% of CLL patients at diagnosis, and since this disease has a heterogeneous clinical course and few prognostic markers, we aimed at designing a fast, cost effective and robust assay to detect NOTCH1 PEST domain mutations in patients with CLL. While 92% of the mutations in NOTCH1 PEST domain found in CLL are insertions or deletions, only 8% are represented by point mutations. Therefore we decided to use a fragment analysis approach in our assay. Given that a single mutation (c.7544_7545delCT), represents roughly 75% of all PEST domain mutations in CLL we designed a test that can, at the same time, detect the presence of this mutation specifically and also any insertion or deletion in exon 34. We designed a PCR reaction using one FAM-labeled forward primer anchored at codon 2407 and two reverse primers. One specific for the c.7544_7545delCT mutation anchored at codon 2414 yielding a product of 356 base pairs (bp) and one anchored at codon 2425, yielding a product of 391 bp, comprising the hot spot for mutations in the NOTCH1 PEST domain. Primers were designed with Primer3 software (http://frodo.wi.mit.edu/) and the specificity of the reaction evaluated using the tool “PCR in silico” (http://genome.ucsc.edu/cgi-bin/hgPcr?command=start). The test yields three possible outputs: a) A single 391 bp peak: wild type samples b) Three peaks (391 bp, 389 bp and 356 bp): heterozygous for c.7544_7545delCT c) Two peaks (391 bp and another bigger or smaller, depending on the size of insertion / deletion): another insertion or deletion, but not c.7544_7545delCT. We have studied 91 blood samples from CLL patients in diverse disease stages and found NOTCH1 mutations in 16 patients, (17.5%). When only patients with trisomy 12, isolated or with other cytogenetic abnormalities, were analyzed (N=16), we found an incidence of NOTCH1 mutations of 43% (7/16). At the same time, the frequency of NOTCH1 mutations in patients without trisomy 12 was 12% (9/75), in agreement with previous reports. All mutated cases (N=13) had the mutation c.7544_7545delCT. In conclusion, we have designed a robust, fast and cost effective assay for routine identification of NOTCH1 PEST domain mutations, suitable for implementation in the clinical setting. Additionally we have found that the incidence and distribution among cytogenetic groups of NOTCH1 mutation in Brazilian patients with CLL is similar to the incidence described in other cohorts. A – Wild Type NOTCH1 revealed by the presence of a single 391 bp peak. B – Presence of heterozygous c.7544_7545delCT mutation evidenced by the presence of a 356 bp peak,corresponding to the allele specific pcr peak; and a double peak at 391 bp and 389 bp positions, corresponding to the wild type product (391 bp) and to the mutated product (389 bp) detected with the wild type primers. Figure 1 Assay Results for NOTCH1 PEST Domain Mutations Figure 1. Assay Results for NOTCH1 PEST Domain Mutations Disclosures: No relevant conflicts of interest to declare.


Haematologica ◽  
2010 ◽  
Vol 95 (6) ◽  
pp. 942-949 ◽  
Author(s):  
S. Heesch ◽  
N. Goekbuget ◽  
A. Stroux ◽  
J. O. Tanchez ◽  
C. Schlee ◽  
...  

Blood ◽  
1998 ◽  
Vol 91 (8) ◽  
pp. 2961-2968 ◽  
Author(s):  
L. King-Underwood ◽  
K. Pritchard-Jones

In a previous study of acute leukemia, we have shown thatWT1 gene mutations occur in both myeloid and biphenotypic subtypes, where they are associated with refractoriness to standard induction chemotherapy. We have now extended this study to a total of 67 cases (34 acute myeloid leukemia [AML], 23 acute lymphoblastic leukemia [ALL], 10 acute undifferentiated leukemia [AUL]/biphenotypic) and find that WT1 mutations occur in 14% of AML and 20% of biphenotypic leukemia, but are rare in ALL (one case). In contrast to the findings in Wilms' tumor, where mutations in the WT1 gene usually behave according to Knudson's two hit model for tumor suppressor genes, seven of eight leukemia-associated WT1 mutations are heterozygous, implying a dominant or dominant-negative mode of action in hematopoietic cells. In AML, the presence of a WT1 mutation is associated with failure to achieve complete remission and a lower survival rate. These data (1) confirm that WT1 mutations underlie a similar proportion of cases of AML to that seen in Wilms' tumors and (2) show for the first time that WT1 mutations can contribute to leukemogenesis of lymphoid as well as myeloid origin, suggesting that its normal role in hematopoiesis lies at a very early progenitor stage. The relationship of WT1 mutation to chemoresistance merits further investigation.


2011 ◽  
Vol 59 (8) ◽  
pp. 1258-1262 ◽  
Author(s):  
Hoda Ali Sadek ◽  
Wafaa Hassan El-Metnawey ◽  
Iman Abdel-Mohsen Shaheen ◽  
Mervat Mamdouh Korshied ◽  
Azza Sobieh Mohamed

Blood ◽  
1998 ◽  
Vol 91 (8) ◽  
pp. 2969-2976 ◽  
Author(s):  
Kazushi Inoue ◽  
Hiroya Tamaki ◽  
Hiroyasu Ogawa ◽  
Yoshihiro Oka ◽  
Toshihiro Soma ◽  
...  

The WT1 gene is a tumor-suppressor gene that was isolated as a gene responsible for Wilms' tumor, a childhood kidney neoplasm. We have previously reported that the WT1 gene is strongly expressed in leukemia cells with an increase in its expression levels at relapse and an inverse correlation between its expression levels and prognosis, thus making it a novel tumor marker for leukemic blast cells. Furthermore, WT1 antisense oligomers have been found to inhibit the growth of leukemic cells. These results strongly suggested the involvement of the WT1 gene in human leukemogenesis. The present study was performed to prove our hypothesis that the WT1 gene plays a key role in leukemogenesis and performs an oncogenic function in hematopoietic progenitor cells, rather than a tumor-suppressor gene function. 32D cl3, an interleukin-3–dependent myeloid progenitor cell line, differentiates into mature neutrophils in response to granulocyte colony-stimulating factor (G-CSF). However, when transfected wild-type WT1 gene was constitutively expressed in 32D cl3, the cells stopped differentiating and continued to proliferate in response to G-CSF. As for signal transduction mediated by G-CSF receptor (G-CSFR), Stat3α was constitutively activated in wild-type WT1-infected 32D cl3 in response to G-CSF, whereas, in WT1-uninfected 32D cl3, activation of Stat3α was only transient. However, most interesting was the fact that G-CSF stimulation resulted in constitutive activation of Stat3β only in wild-type WT1-infected 32D cl3, but not in WT1-uninfected 32D cl3. Thus, WT1 expression constitutively activated both Stat3α and Stat3β. A transient activation of Stat1 was detected in both wild-type WT1-infected and uninfected 32D cl3 after G-CSF stimulation, but no difference in its activation was found. No activation of MAP kinase was detected in both wild-type WT1-infected and uninfected 32D cl3 after G-CSF stimulation. These results demonstrated that WT1 expression competed with the differentiation-inducing signal mediated by G-CSFR and constitutively activated Stat3, resulting in the blocking of differentiation and subsequent proliferation. Therefore, the data presented here support our hypothesis that the WT1 gene plays an essential role in leukemogenesis and performs an oncogenic function in hematopoietic progenitor cells and represent the first demonstration of an important role of the WT1 gene in signal transduction in hematopoietic progenitor cells.


2011 ◽  
Vol 170 (12) ◽  
pp. 1529-1534 ◽  
Author(s):  
Spyridon Megremis ◽  
Andromachi Mitsioni ◽  
Irene Fylaktou ◽  
Sofia Kitsiou Tzeli ◽  
Filadelfia Komianou ◽  
...  

2016 ◽  
Vol 17 (1) ◽  
pp. 21-32 ◽  
Author(s):  
Sumiko Kobayashi ◽  
Yasunori Ueda ◽  
Yasuhito Nannya ◽  
Hirohiko Shibayama ◽  
Hideto Tamura ◽  
...  

2016 ◽  
Vol 16 (3) ◽  
pp. e21-e26 ◽  
Author(s):  
Gholamreza Toogeh ◽  
Mani Ramzi ◽  
Mohammad Faranoush ◽  
Naser Amirizadeh ◽  
Sezaneh Haghpanah ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-28
Author(s):  
Hassan Awada ◽  
Arda Durmaz ◽  
Carmelo Gurnari ◽  
Misam Zawit ◽  
Sunisa Kongkiatkamon ◽  
...  

Mutations in tumor suppressor genes and oncogenes are both potentially therapeutically actionable in acute myeloid leukemia (AML). The Wilms' Tumor 1 (WT1) gene is located on 11p13 and encodes a zinc finger transcription factor which has been found to be overexpressed and mutated in AML. In normal development, WT1 is only expressed in a small subset of hematopoietic stem cells. While its overexpression suggests an oncogenic role, the invariable presence of mutations in the cysteine-histidine zinc finger domains indicates a tumor suppressor function, similar to that in WAGR syndrome/11p deletion syndrome in which it was first discovered. Like its unknown function in AML, the clinical significance and genetic associations of WT1 mutations have been also controversial. Although studies of WT1 mutations in AML have been conducted, the lack of solid clinical and molecular characterization of large WT1-mutant (WT1MT) AML cohort has hampered its definition. In this study, we took advantage of a compendia of genomic results from Cleveland Clinic and publicly available data of 2188 AML patients (primary (p)AML, n= 1636; secondary (s)AML, n= 433; therapy-related (t)AML, n= 119, excluding cases with acute promyelocytic leukemia, MLL-rearrangement, and core-binding factor AML). While several reports only focused on cytogenetic normal AML (CN-AML), which represented 61% of our cohort, we additionally included all other cytogenetic risk groups. In total, WT1 mutations were detected in 5% (114/2188) of patients. WT1 mutations were enriched in pAML (85%) compared to sAML (11%) and tAML (4%). Thirty-nine patients (13%) carried more than 1 WT1 mutation. WT1MT were younger [59 vs 64 years, P=0.0002] and more often females (55% vs 45%, P=0.03) as compared to WT1 wild type (WT1WT) patients. Univariate analyses of baseline parameters showed that WT1MT AML had a more proliferative phenotype with a higher WBC [15.1 vs 9.5 x109/L, P=0.03] and bone marrow blast percentages [73 vs 59%, P=0.002] and with lower platelet counts [44 vs 56 x109/L, P=0.008] compared to WT1WT cases. In the WT1MT cohort, 70% had a normal karyotype, with complex karyotype being significantly less frequent vsWT1WT patients [4 vs 16%, P=0.001]. The most common cytogenetic abnormalities in WT1MT patients included +8 (8%) followed by -9/del(9q) (3%) and -7/del(7q) (3%). Only 1 patient carried inv(3)/t(3;3) or -17/del(17p). In sum, no statistical differences in cytogenetics were found between WT1MTvsWT1WT AML patients. Next, identified mutational signatures of WT1MT patients. A panel of 44 myeloid genes and their hotspot configurations were selected according to their relevance in AML. In comparison to WT1WT AML patients, multivariate analyses showed that WT1MT patients had higher odds of biallelic CEBPA (12 vs 3%; P=0.009) and FLT3 internal tandem duplication mutations (FLT3ITD, 31 vs 16%; P=0.01) but lower odds of SRSF2 mutations (2 vs 9%, P=0.04). Since FLT3ITD has been previously described to be associated with WT1 mutations, we also focused on investigating whether mutations in the tyrosine kinase domain (TKD) were frequent in WT1MT as well. Although we found increased percentages of FLT3TKD (11%) among the WT1MT patients compared to WT1WT cohort (8%), this difference did not reach statistical significance. To uncover multifactor lesions (cytogenetic and/ or additional molecular lesions) of prognostic importance, we performed survival analyses. Although the combination of WT1 mutations and FLT3TKD shortened overall survival (OS) by 2-times in WT1MT patients vsWT1WT cases with FLT3TKD (23.7 vs 45.9 months), this result was not significant (P=0.1). In addition, the concurrent presence of other cytogenetic and molecular features didn't reveal significant impact on OS. In sum, using an adequately powered cohort, our study of the genomic landscape of WT1MT AML patients identified its genomic associations and their clinical and prognostic inferences. The application of advanced machine learning methods to large datasets of WT1MT AML patients might be crucial to capture the complex genomic interactions of WT1 gene in AML. Disclosures Carraway: BMS: Consultancy, Other: Research support, Speakers Bureau; Stemline: Consultancy, Speakers Bureau; Takeda: Other: Independent Advisory Committe (IRC); ASTEX: Other: Independent Advisory Committe (IRC); Abbvie: Other: Independent Advisory Committe (IRC); Novartis: Consultancy, Speakers Bureau; Jazz: Consultancy, Speakers Bureau. Nazha:MEI: Other: Data monitoring Committee; Novartis: Speakers Bureau; Incyte: Speakers Bureau; Jazz: Research Funding. Sekeres:Pfizer: Consultancy; BMS: Consultancy; Takeda/Millenium: Consultancy. Maciejewski:Alexion, BMS: Speakers Bureau; Novartis, Roche: Consultancy, Honoraria.


Sign in / Sign up

Export Citation Format

Share Document