Association of CDKN2A/2B deletion with homozygous deletion of TCR- γ gene in T-ALL.

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e18516-e18516
Author(s):  
Anita Chopra ◽  
Jay Singh ◽  
Deepak Verma ◽  
Nishi Rajput ◽  
Sameer Bakhshi ◽  
...  

e18516 Background: T-lineage acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease that arises in a multistep fashion through acquisition of several genetic aberrations. Absence of biallelic deletion of TCR-γ (ABD), a marker of immaturity, and CDKN2A/2B deletion have been identified as poor predictors of outcome in T-ALL treated with contemporary protocols. The aim of this study was to determine the correlation between ABD and CDKN2A/2B in Indian T-ALL patients. Methods: A total of 31 cases of T-ALL, diagnosed on morphology, cytochemistry and immunophenotyping, were included in the study. The age of the patients ranged from 1-50 years. The patients were investigated for CDKN2A/2B deletion by multiplex ligation dependent probe amplification (MLPA) using the commercially available probe set P383 (MRC Holland). The status of the deletion of TCR- γ was determined using quantitative assay described by Gutierrez et al.1 Results: Absence of biallelic deletion of TCR- γ gene were found in 14 (45.16%) patients. Homozygous deletion was seen in 13 (41.93%) patients while monoallelic deletion was seen in 5 (16.12%) patients. CDKN2A/2B deletions were found in (48.38%) patients. On correlation of CDKN2A/2B deletion and TCR- γ deletion, it was found that CDKN2A/2B deletion was found in 2/14 (14.28%) patients with ABD and 10/13 (76.92%) patients with homozygous deletion. Conclusions: CDKN2A/2B deletion are more common in patients with homozygous deletion of TCR- γ gene.

Blood ◽  
1990 ◽  
Vol 75 (3) ◽  
pp. 730-735
Author(s):  
J Cheng ◽  
P Scully ◽  
JY Shew ◽  
WH Lee ◽  
V Vila ◽  
...  

Human leukemia cell lines were examined for the status of the retinoblastoma (RB) protein by immunoblotting analysis using antibodies raised against the TrpE-RB fusion protein. One of 16 cell lines examined, the T-cell acute lymphoblastic leukemia (ALL) line HSB-2, lacked the 110-Kd RB protein. Southern blot analysis of genomic DNA extracted from HSB-2 cells showed a large homozygous deletion of the RB gene, stretching from exon 18 beyond exon 27. Northern blot analysis showed multiple, abnormal RB transcripts in HSB-2. A truncated protein (72 Kd) was detected with 35S-methionine labeling but not with 32P- orthophosphate labeling of the HSB-2 cells. The genomic deletion of greater than 85 kb DNA at the RB locus (13q14) was not detectable in the karyotype of the HSB-2 cells. Among the 16 human leukemia cell lines examined for the status of the RB gene, only one, the HSB-2 line, showed an abnormal RB protein. Further study of primary leukemia and lymphoma samples appears to be warranted.


Blood ◽  
1990 ◽  
Vol 75 (3) ◽  
pp. 730-735 ◽  
Author(s):  
J Cheng ◽  
P Scully ◽  
JY Shew ◽  
WH Lee ◽  
V Vila ◽  
...  

Abstract Human leukemia cell lines were examined for the status of the retinoblastoma (RB) protein by immunoblotting analysis using antibodies raised against the TrpE-RB fusion protein. One of 16 cell lines examined, the T-cell acute lymphoblastic leukemia (ALL) line HSB-2, lacked the 110-Kd RB protein. Southern blot analysis of genomic DNA extracted from HSB-2 cells showed a large homozygous deletion of the RB gene, stretching from exon 18 beyond exon 27. Northern blot analysis showed multiple, abnormal RB transcripts in HSB-2. A truncated protein (72 Kd) was detected with 35S-methionine labeling but not with 32P- orthophosphate labeling of the HSB-2 cells. The genomic deletion of greater than 85 kb DNA at the RB locus (13q14) was not detectable in the karyotype of the HSB-2 cells. Among the 16 human leukemia cell lines examined for the status of the RB gene, only one, the HSB-2 line, showed an abnormal RB protein. Further study of primary leukemia and lymphoma samples appears to be warranted.


Blood ◽  
1997 ◽  
Vol 89 (11) ◽  
pp. 4161-4166 ◽  
Author(s):  
Ursula R. Kees ◽  
Paul R. Burton ◽  
Changlong Lü ◽  
David L. Baker

Abstract The p16 gene (MTS1, CDKN2, p16INK4A, CDKI) encoding an inhibitor of cyclin-dependent kinase 4 (cdk4) has been found to be deleted in various types of tumors, including leukemia, and is thought to code for a tumor suppressor gene. Our preliminary findings on eight pediatric patients with acute lymphoblastic leukemia (ALL) suggested that the survival of patients carrying a homozygous p16 gene deletion was significantly inferior to that of those without a deletion. The present study on 48 patients tested the hypothesis that the clinical outcome for pediatric ALL patients is correlated with the presence or absence of the p16 gene. Overall, nine of 48 children (18.3%) carried a homozygous p16 deletion. Such deletions were significantly more common (P = .003) among T-ALL patients (five of eight, 62.5%) than among precursor-B-ALL patients (four of 40, 10.0%). Of nine patients exhibiting p16 deletions, eight (88.9%) were classified as high-risk patients by the recognized prognostic factors of age, white blood cell count, and T-cell phenotype. The 4-year event-free survival in the study population as a whole was 72.7%. Without adjustment for other risk factors (univariate model), the presence of a homozygous p16 deletion was associated with a markedly increased probability of both relapse (P = .0003) and death (P = .002). These findings raise the question of whether the p16 deletion itself confers an increased risk of relapse after adjusting for the known risk factors. In this analysis, the estimated risk multiplier factor for relapse in patients carrying the p16 deletion was 14.0 (P = .0004) and for the risk of death 15.6 (P = .0008). We therefore conclude that the presence of a homozygous p16 deletion may well be an important risk factor for both relapse and death in childhood ALL, and that its prognostic effect is not a consequence of confounding by other factors already known to influence outcome in this disease.


2010 ◽  
Vol 88 (3) ◽  
pp. 249-265 ◽  
Author(s):  
Shabnam Shalapour ◽  
Cornelia Eckert ◽  
Karl Seeger ◽  
Madlen Pfau ◽  
Javier Prada ◽  
...  

Blood ◽  
2018 ◽  
Vol 131 (3) ◽  
pp. 289-300 ◽  
Author(s):  
Arnaud Petit ◽  
Amélie Trinquand ◽  
Sylvie Chevret ◽  
Paola Ballerini ◽  
Jean-Michel Cayuela ◽  
...  

Key Points In pediatric T-ALL, oncogenetic markers, MRD, and WBC count are independent predictors of outcome. These factors should be used together for individual treatment stratification.


2014 ◽  
Vol 38 (5) ◽  
pp. 532-536 ◽  
Author(s):  
Joseph M. Brandwein ◽  
Eshetu G. Atenafu ◽  
Andre C. Schuh ◽  
Karen W.L. Yee ◽  
Aaron D. Schimmer ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1485-1485
Author(s):  
Almut Giese ◽  
Reinhard Ullmann ◽  
Cornelia Eckert ◽  
Renate Kirschner-Schwabe ◽  
Guenter Henze ◽  
...  

Abstract The ETV6/RUNX1 fusion, resulting from the cryptic translocation t(12;21)(p13;q22), is the most common genetic rearrangement in B-cell precursor (BCP) acute lymphoblastic leukemia (ALL) with a prevalence of approximately 20–25% at first presentation. Although generally associated with favorable risk features and advantageous long-term survival rates, similar frequencies of ETV6/RUNX1 positivity at relapse question its prognostic significance. At relapse, a second continuous complete remission can again be achieved in the majority of patients with ETV6/RUNX1-positive ALL, however, a substantial proportion (approximately 25%) exhibit a poor response to treatment and eventually experience a subsequent relapse. Currently, critical secondary genetic events are assumed to be pivotal for t(12;21) positive ALL leukemogenesis. At the chromosomal level, additional numeric aberrations may also contribute to differences both in treatment response and course of disease, and may provide additional prognostic information. Aiming at identifying the incidence and the clinical relevance of additional genetic aberrations at the time point of first ALL relapse diagnosis, we performed whole genome high resolution tiling-path bacterial artificial chromosome (BAC) array CGH of leukemic cell DNA from 53 patients with ETV6/RUNX1-positive first relapse enrolled in the relapse trials ALL-REZ BFM of the Berlin-Frankfurt-Münster study group. Additional genetic aberrations were detected in all of the ETV6/RUNX1-positive leukemic bone marrow samples with a mean number of seven aberrations per ALL. Chromosomal losses were observed approximately 2.5 times more frequently than gains. A high proportion of the identified copy number alterations occurred in recurrently affected chromosomal regions. Copy number alterations most commonly detected by high-resolution array CGH were losses of 12p13 (49%, 26/53), 6q21 (34%, 18/53), 15q15.1 (23%, 12/53), 9p21 (21%, 11/53), 3p21 (21%, 11/53), 5q (19%, 10/53), 19q13 (17%, 9/53), 10q (13%, 7/53), 3p14.2 (11%, 6/53) and gains of 21q22 (32%, 17/53) and of 12p (21%, 11/53). Loss of the whole X-chromosome, detected in 17% (9/53) of the analyzed samples, was observed exclusively in females. In contrast, gain of Xq, identified in 13% (7/53) of the samples, was detectable solely in males. Evaluation of the clinical parameters in relation to recurrent copy number alterations revealed that loss of the whole X-chromosome was associated with a molecular good response to treatment (P=0.03), as assessed by sensitive minimal residual disease (MRD) monitoring. In contrast, loss of 5q31.3, detected in 11% (6/53) of the analyzed samples, was associated with a dismal molecular response to therapy (P=0.019) and with a tendency to a worse outcome (P=0.054). Thus, this study demonstrates that relapsed ETV6/RUNX1-positive ALL is characterized by (a.) multiple additional genomic alterations, in particular by a high incidence of chromosomal losses, which occur predominantly in recurrently affected chromosomal regions, some of which (b.) are of significant prognostic relevance. These findings support the notion that several additional chromosomal changes are not only required for the process of malignant transformation in ETV6/RUNX1-positive ALL (leukemogenesis) but also influence therapeutic success. Perspectively, high resolution genomic profiling will provide valuable information enabling a more refined, individualized therapy and an improved risk stratification in relapsed childhood ALL.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2407-2407
Author(s):  
Annette Fasan ◽  
Melanie Zenger ◽  
Madlen Ulke ◽  
Wolfgang Kern ◽  
Torsten Haferlach ◽  
...  

Abstract Introduction: In acute lymphoblastic leukemia (ALL) genetic diversity at diagnosis has been reported to be correlated with an increased likelihood of emerging drug resistance and an increased relapse risk. Aim: To evaluate 1. the frequency of chromosome abnormalities at initial diagnosis and their acquisition during the course of the disease (clonal evolution), 2. the pattern of acquired genetic abnormalities. Patients: We investigated 11 adult ALL patients (age 18.9-76.7 years, median 42.6 years) with paired samples at initial diagnosis and at relapse by chromosome banding analysis (CBA), array-based comparative genomic hybridization (aCGH) and molecular analysis. The cohort consisted of 7 B-ALL and 4 T-ALL cases. CBA was performed in both diagnostic and relapse samples in 10 cases. aCGH was performed in 6 cases (12x270 K microarray slides, Roche Nimblegen, Madison, Wl) both at initial diagnosis and at relapse. Molecular analysis was performed in all cases by 454 amplicon deep-sequencing (Roche, Branford, CT) in the following genes: AKT1, FBXW7, IL7R, JAK1, NOTCH1, PHF6, PTEN and WT1 in T-ALL cases, and in CRLF2, IL7R, IKZF1, JAK2, LEF1 and PAX5 in B-ALL cases at diagnosis and relapse. Results: Median duration from diagnosis to relapse was 10 months (range 4-18 months). Regarding cytogenetics of B-ALL, 2 cases had t(9;22)(q34;q11), 1 case a MYC-rearrangement, 3 cases complex aberrations, 1 case a translocation t(5;16)(p15;q13) and 1 case had a normal karyotype (NK). In T-ALL, 2 cases had NK, 1 case had t(10;14) (q24;q11) and in 1 case no cytogenetic data was available. At initial diagnosis, 8/10 cases (80.0%) with available CBA had one cytogenetic clone, while 2/10 (20.0%) cases had 2 cytogenetically related subclones. At relapse, one case retained both clones already detected at diagnosis. The second case showed loss of genetic aberrations in one subclone (clonal regression), while the second clone was stable. Evolution of genetically related clones was detected by CBA in 5/10 cases. The acquired abnormalities comprized unbalanced (n=8) or balanced (n=3) translocations, gains (n=3) or deletions (n=2) of whole or partial chromosomes. 3 of these 5 cases in addition showed clonal regression which may, however, be due to impaired in vitro proliferation of the subclone at relapse. The lost abnormalities comprized unbalanced (n=6) or balanced (n=1) translocations, gains (n=1) or deletions (n=1) of whole or partial chromosomes. In all 6 cases analyzed by aCGH, clonal evolution was detected. In one case, without clonal evolution detected by CBA, aCGH revealed clonal evolution on the submicroscopic level. In detail, 92 copy number alterations (CNA) were observed (mean 8 per case, range: -33) at initial diagnosis and 105 CNA at relapse (mean 9 per case; range 2-36). Losses were more frequent than gains (79 vs 13 at initial diagnosis; 87 vs 18 at relapse). 53 CNA of initial diagnosis and 33 of relapse were larger than 10 Mbp and were already detected by CBA accordingly. 43 CNA at initial diagnosis (mean: 7 per case; range 4-11) and 40 CNA at relapse (mean 7 per case; range 3-11) were smaller than 10 Mbp and thus below the resolution of CBA. The most frequent CNA were loss of 9p21 (CDKN2A, n=5) and 7p12 (IKZF1, n=3). These stayed stable from initial diagnosis to relapse. Apart from these, no specific pattern of gains or losses of distinct chromosomal regions was observed. In 4/6 cases, clonal regression of 5 CNA was detected on submicroscopic level. By sequencing 17 mutations and 2 variants were observed in 9/11 patients without major changes between initial diagnosis and relapse. In B-ALL cases, mutations were detected in JAK2 (n=2) and IKZF1 (n=3). In 2 cases a variant in CRLF2 was observed, which cannot be assigned as mutation or SNP according to current knowledge. In T-ALL cases, mutations were detected in NOTCH1 (n=3), PHF6 (n=1), PTEN (n=1), WT1 (n=1) and IL7R (n=1). The pattern of mutations was stable in 7/9 cases. One case lost a mutation in NOTCH1 at relapse, while another patient gained a mutation in IKZF1. Two patients without mutations at diagnosis also showed no mutations at relapse. Conclusions: 1. Adult ALL is characterized by multiple genetic aberrations a subset of which can be detected by aCGH only. 2. The genomic heterogeneity of adult ALL increases at relapse. 3. Based on this limited set of patients gene mutations seem to be more stable between diagnosis and relapse compared to cytogenetic aberrations. Disclosures Fasan: MLL Munich Leukemia Laboratory: Employment. Zenger:MLL Munich Leukemia Laboratory: Employment. Ulke:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document