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: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4179-4179
    Abstract: Abstract 4179 In acute myeloid leukemia (AML), complex karyotype is defined as the presence of three or more chromosome abnormalities in the absence of one of the recurrent genetic abnormalities as defined by the recent WHO classification. AML with complex karyotype (CK-AML) account for approximately 10 to 15% of all cases and are associated with preceding myelodysplasia (MDS) or exposure to toxic agents; the prognosis of patients is very poor. So far, little is known about the molecular mechanisms underlying initiation or progression of CK-AML. To identify genomic regions of potential pathogenic relevance, we used microarray-based techniques [array-comparative genomic hybridization (CGH) and single-nucleotide polymorphism (SNP) analysis] for high-resolution genome-wide analysis in 242 cases, including 171 (71%) cases enrolled on clinical protocols using intensive chemotherapy. Among other genomic imbalances, we identified loss of chromosome band 17q11.2 encompassing the NF1 locus in 55 (23%) of the 242 cases. Interestingly, three of these cases exhibited homozygous loss of NF1. Based on these findings and the fact that NF1 is recurrently altered in myeloid malignancies, we further investigated its role in CK-AML. Therefore, we analyzed 11 cases with heterozygous microdeletions of NF1 for mutations in the remaining allele by direct sequencing of exons 1 to 60 and identified 5 mutations in 4 cases; all of these mutations resulted in a premature stop codon (3 frameshift mutations, 2 nonsense mutations); one frameshift mutation (c.2033dupC) was recurrent. Combining the findings from array-based and mutation analyses, we so far identified 7 patients with biallelic NF1 gene alterations, i.e. homozygous loss or loss of one allele and at least one mutation in the remaining allele. Since correlation of NF1 alteration with data from array-based genomic profiling revealed a significant correlation with loss of chromosome band 17p13 encompassing TP53 (P 〈 .001), we correlated NF1 alteration with the TP53 status (mutation and/or loss), which was available for all 242 cases, and found a positive correlation with both TP53 alteration (mutation and/or loss) and TP53 mutation (P 〈 .001 each). In addition, NF1 alteration was significantly correlated with biallelic TP53 alterations (loss and mutation or homozygous mutations) (P 〈 .001). We than further investigated the two genotypes NF1alteration/TP53alteration (n=50) and NF1no alteration/TP53alteration (n=109) with regard to their association with other genomic imbalances. The genotype NF1alteration/TP53alteration was significantly correlated to the total number of deletions (median 9 vs 7; P = .025), the genomic complexity as measured by the total number of aberrations per case (median 13 vs 11; P = .039), and the presence of 16q loss (50% [25/50] vs 29% [32/109], P = .014) when compared with the NF1no alteration/TP53alteration genotype. Notably, in a recently published murine model deficiency of ICSBP, located on 16q24, was shown to synergize with NF1 haplo-insufficiency in leukemogenesis. In conclusion, the NF1 gene is found to be recurrently altered in CK-AML. Being associated with specific genomic aberrations, NF1 alteration is likely cooperating in myeloid leukemogenesis or disease progression. One important co-player might be TP53 that has an important role in genomic stability. The exact mechanism of interaction between NF1 and TP53 or other concurrent genetic alterations have to be further investigated. Disclosures: Döhner: Pfizer: Research Funding.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 237-237
    Abstract: Abstract 237 Mutations in the nucleophosmin 1 (NPM1) gene represent one of the most frequent gene mutations in acute myeloid leukemia (AML), in particular in cytogenetically normal (CN)-AML. NPM1 mutations (NPM1mut) are considered as an early genetic event in the pathogenesis of AML. To address the role of clonal evolution from diagnosis to relapse in NPM1mut AML, we applied high-resolution genome-wide single nucleotide polymorphism (SNP) array analysis using the Affymetrix 6.0 platform to detect copy number alterations (CNAs) and uniparental disomies (UPDs) in paired samples from 42 patients. In addition, we determined NPM1 and FLT3 [internal tandem duplication (ITD) and tyrosine kinase domain (TKD)] mutation status in all samples. Blood or bone marrow samples obtained at complete morphologic remission were available for all patients to exclude germline copy number variations. At diagnosis, 29 cases (69%) had a normal karyotype by cytogenetics and no CNAs and UPDs by SNP analysis. In the 13 remaining cases, we found a total of 10 CNAs in 7 cases (19%), and 6 UPDs in 6 cases (14%): deletions of 9q21 (size range 0.9 to 17 Mb) were detected in 5 cases and were the only recurrent CNA; the only recurrent UPD affected the long arm of chromosome 13 in 4 cases, all resulting in homozygous FLT3-ITD mutations with FLT3-ITD/wildtype ratios 〉 1; heterozygous FLT3-ITD and –TKD mutations were detected in 9 and 7 patients, respectively. At the time of relapse, the number of CNAs increased (34 CNAs in 16 cases, 38%) while the frequency of UPDs remained unchanged (6 UPDs in 6 cases, 14%). Of note, in 6 patients (14%) the NPM1 mutation was no longer detectable at the time of relapse; SNP analysis showed completely distinct CNAs/UPDs in 4 of these patients; 3 of these 4 cases had a small gain at 11q23 corresponding to MLL partial tandem duplications as confirmed by PCR. These findings suggest that these 4 cases were therapy-related AMLs (t-AML) rather than relapsed AML. The median interval from diagnosis to relapse/tAML in these 4 cases was 65 months compared with 9 months for the relapsed cases still having the NPM1 mutation. In the two remaining cases, genetic alterations were neither present at diagnosis nor at relapse. Analysis of other gene mutations (eg, IDH1 and 2, DNMT3A, ASXL1, p53) is currently under way to further elucidate the clonal origin of these cases. Of the 36 NPM1mut positive relapse samples, 15 maintained a “normal karyotype”, and 2 showed the CNAs already present at diagnosis; 19 relapse samples (53%) displayed clonal evolution with acquiring new (n=15) and/or loosing single aberrations (n=4): Acquired recurrent alterations comprised deletions of tumor suppressor genes [ETV6 (n=2), TP53 (n=2), NF1 (n=2), WT1 (n=2)], most of which are uncommon in de novo NPM1mut AML. All 6 UPDs detected in relapse samples affected 13q, of which 3 were already present at diagnosis. One patient with initial heterozygous FLT3-ITD mutation developed a homozygous state by acquiring UPD13q at relapse. Two cases with wild-type FLT3 at diagnosis acquired UPD13q at relapse. Of note, one UPD13q was not present in the corresponding relapse sample anymore. In conclusion, almost half (45%) of NPM1mut AML showed evolution to a more aberrant karyotype at relapse, including acquisition of high-risk genetic changes that may account for the adverse prognosis of relapsed patients. Conversely, other alterations such as UPD13q or del(9q) detected at diagnosis were not always present in relapse samples, implying that relapse had evolved from a more ancestral clone. In addition, our data suggest that in a proportion of cases t-AML rather than relapse had developed. Further analysis, such as gene mutation studies of paired diagnosis/ relapse samples, will provide more detailed information on clonal evolution events in the pathogenesis of NPM1mut AML. 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: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2632-2632
    Abstract: Abstract 2632 Poster Board II-608 In acute myeloid leukemia (AML) complex karyotype is defined as the presence of ≥3 chromosome abnormalities in the absence of one of the chromosomal rearrangements listed in the category “AML with recurrent genetic abnormalities” (WHO 2008). This subset of AML accounts for approximately 10 to 15 % of all AML and it is characterized by a very poor prognosis. To identify novel genomic regions of interest in this AML subgroup, we recently applied microarray-based techniques (array-comparative genomic hybridization [CGH] and single nucleotide polymorphism [SNP] analysis) allowing high-resolution genome-wide screening of genomic imbalances in 245 patients; 171 of the 245 (70%) patients were enrolled in clinical protocols using intensive chemotherapy. Applying these techniques, we identified deletions of 17p13 encompassing the TP53 locus in 94 of 245 (38%) cases. Based on these findings and the fact that TP53 is a frequently affected gene in numerous malignancies, we aimed to investigate its role in complex karyotype AML. Therefore, we screened 188 of the 245 patients forTP53 mutations either by direct sequencing of exon 5 to 9 or by denaturating high-performance liquid chromatography of exon 4 to 10 on a WAVE® system followed by direct sequencing of the positive cases. In total, we identified 137 mutations (exon 4 [e4] n=2; e5 n=41; e6 n=26; e7 n=35; e8 n=26; e10 n=1; splice sites n=6) in 106 patients (56%) with a maximum of four mutations per case. Of these 137 mutations, 108 were missense mutations, 15 frame shift mutations, 8 nonsense mutations, and 6 were located at intronic splice sites. Combining the findings from array-based and mutation analyses, TP53 gene alterations were identified in 129 of 188 (69%) patients. Of note, 29% of these cases showed biallelic TP53 gene alterations with deletion of one allele and at least one mutation in the remaining allele. When correlating TP53 alterations (mutations and/or deletions) with genomic data from array-based analyses, we found positive correlations with the presence of 5q and 7q losses (Chi-square test P 〈 .001 and P = .001, respectively), total number of deletions (P 〈 .001), gains (P = .002), high-level DNA amplifications (P 〈 .001), and genomic complexity as measured by total number of aberrations per case (P 〈 .001). Correlation of TP53 alterations with clinical characteristics revealed that patients with TP53 alterations were older (median age 60 versus 55 years, P = .01) and showed in trend a lower WBC (median, 6.6 versus 14.2 × 109/L, P = .14); there was no difference in bone marrow and blood blast counts, in platelet counts, or in type of AML (de novo vs. secondary vs. treatment-related). Clinical outcome analyses were restricted to patients enrolled in prospective clinical trials (n=171). AML exhibiting a TP53 alteration had significantly lower complete remission (CR) rates [25% (26/104) versus 56% (24/43), P 〈 .001], inferior event-free survival (EFS, P = .009), relapse-free survival (RFS, P = .02), and overall survival (OS, P 〈 .001) compared with TP53 wild type AML. In multivariate models for achievement of CR, RFS, and OS, TP53 alteration retained its significance as an independent risk factor (CR: OR 0.38, P 〈 .001; RFS: HR 1.65, P = .02; OS: HR: 2.15, P = .002). In conclusion, TP53 is the most frequently known affected gene in complex karyotype AML. We found that the TP53 status is associated with specific genomic aberrations as well as the degree of genomic complexity, a finding that fits well into the TP53 pathomechanism of genomic instability. Importantly, among AML with complex karyotype TP53 alterations significantly predicted inferior outcome allowing for a further refinement of this AML subgroup. 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1600-1600
    Abstract: Abstract 1600 Poster Board I-626 Background The V-ets erythroblastosis virus E26 (ETS) oncogene family is one of the largest families of transcription factors. ETS transcription factors are characterized by two major functional domains, a transcription domain and an evolutionarily highly conserved DNA-binding domain, also known as ETS domain that mediates binding to purine-rich DNA sequences. Most ETS family proteins are nuclear targets for activation of the Ras-MAP kinase signalling pathway. Therefore, they play a significant role in regulating cellular functions such as cell growth, apoptosis, development and differentiation. ETS transcription factors have been implicated in leukemia by chromosomal rearrangement, and more commonly by gene amplification and/or overexpression. Moreover, overexpression of ERG was shown to be an adverse predictor for clinical outcome in AML with normal cytogenetics (CN). In our recent study on complex karyotype AML, array-CGH (comparative genomic hybridization) analysis identified small genomic amplifications affecting ERG/ETS2 in 21q22 and ETS1/FLI1 in 11q23 in about 10% of the cases. Correlation with global gene expression profiling showed that ERG and ETS2 as well as ETS1 and FLI1 were overexpressed in these cases. Aims: To evaluate expression levels of ERG, ETS2, ETS1 and FLI1 in a large cohort of younger (16 to 60 years of age) adult CN-AML patients (pts) and their impact on clinical outcome. Methods The expression of ERG, ETS2, ETS1 and FLI1 was determined by quantitative real-time reverse transcriptase polymerase chain reaction (qPCR) assay in 343 CN-AML pts who were entered on 3 AMLSG treatment protocols (AMLHD93, AML HD98-A, AMLSG 07-04). ERG, ETS2, ETS1, and FLI1 were dichotomized into two major groups according to their expression levels. The upper quartile was chosen as the cut point and the set of patients with gene expression above were defined as Q4 group. Univariable as well as multivariable regression models were used to evaluate the influence of ERG, ETS2, ETS1 and FLI1 on induction success, event-free, relapse-free and overall survival. Multivariable analyses were stratified for AMLSG treatment protocols. Results Partial correlation analysis revealed positive correlations of expression levels between ETS2 and ERG (ρ=0.45) being the strongest, followed by ERG and FLI1 (ρ=0.4), as well as ETS1 and FLI1 (ρ=0.31). Correlation of ERG, ETS2, ETS1 and FLI1 with white blood count (WBC) revealed a significant association between high gene expression (Q4) and elevated WBC (ERG, p=0.004; ETS2, p=0.002, FLI1 p 〈 0.001), whereas high expression of ETS1 was associated with a significantly lower WBC (p 〈 0.001). Univariable as well as multivariable analyses on induction success revealed high ETS2 as an unfavourable marker (OR, 0.29, p=0.01). In univariable analysis, there was a significantly inferior relapse-free survival (RFS) and overall survival (OS) for high ERG (p=.01; p=.06, respectively) and high ETS2 (p=.002; p=.03, respectively) that was even more pronounced when both ERG Q4 and ETS2 Q4 (ERG Q4 ∩ ETS2 Q4) (p 〈 0.001; p=.001, respectively) were included as one variable and compared with the rest. In multivariable analysis for the endpoints event-free survival (EFS), RFS and OS, a significant effect was found for RFS for ERG Q4 ∩ ETS2 Q4 (p=.002); the only significant variables that consistently appeared in the model were NPM1mut, FLT3-ITDpos and WBC. In subgroup analysis for the genotypes CEBPAmut, NPM1mut/FLT3-ITDneg, and all others (NPM1mut/FLT3-ITDpos, NPM1wt/FLT3-ITDpos, NPM1wt/FLT3-ITDneg) according to the hierarchical model, ERG Q4 was associated with an inferior EFS (p=.04) and OS (p=.03) in the favorable CEBPAmut genotype and became even more significant for the variable ERG Q4 ∩ ETS2 Q4 (EFS, p=.007, RFS, p=.002; OS, p=.06, respectively). For the NPM1mut/FLT3-ITDneg subgroup, again ERG Q4 ∩ ETS2 Q4 was associated with an adverse RFS (p=.04), but not with OS (p=0.07). Conclusions In our study on a large cohort of homogenously treated CN-AML patients, ERG and ETS2 expression were highly correlated. Overexpression of both genes had a significant impact on clinical outcome of CN-AML patients. Moreover, adverse effects of high ERG and high ETS2 expression on prognosis were also shown for the genetic AML subgroups CEBPAmut and NPM1mut/FLT3-ITDneg. 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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3558-3558
    Abstract: Abstract 3558 Acute myeloid leukemia with complex karyotype (CK-AML, CK+) is defined as ≥3 acquired chromosome abnormalities in the absence of recurrent genetic abnormalities (WHO 2008). CK-AML account for 10–15% of all AML and are characterized by a dismal outcome. To delineate prognostic markers in this unfavorable subgroup, we performed integrative analysis using genomic profiling (array-comparative genomic hybridization [CGH] and/or single-nucleotide polymorphism [SNP] analysis), as well as TP53 mutation screening in 234 CK-AML. TP53 mutations were found in 141/234 (60%) CK-AML comprising 130 missense, 21 insertion/deletion, nine nonsense, and eight splice site mutations; genomic losses of TP53 were identified in 94/234 (40%). Combining these data, TP53 alterations were detected in 70% of patients, and at least 66% of these exhibited biallelic alterations. TP53 alterations (loss and/or mutation in TP53) were characterized by a higher degree of genomic complexity, as measured by total number of copy number alterations per case (mean±SD 14.30±9.41 versus 6.16±5.53, P 〈 .0001), and by the association with specific genomic alterations, that is, monosomy 3 or losses of 3q (-3/3q-) (P=.002), -5/5q- (P 〈 .0001), -7/7q- (P=.001), -16/16q- (P 〈 .0001), -18/18q- (P=.001), and -20/20q- (P=.004); gains of chromosome 1 or 1p (+1/+1p) (P=.001), +11/+11q (P=.0002), +13/+13q (P =.02), and +19/+19p (P =.04); and amplifications in 11q13∼25 [amp(11)(q13∼25)]. The recently described cytogenetic category “monosomal karyotype” (MK), defined as two or more autosomal monosomies or one single autosomal monosomy in the presence of structural abnormalities, for which a prognostic impact could be demonstrated even in CK-AML, was correlated with TP53 alterations (P 〈 .0001). Clinically, TP53altered CK-AML patients were older (median age, 61 versus 54 years, P =.002), had lower bone marrow (BM) blast counts (median 65% versus 78%, P=. 04), and had lower complete remission (CR) rates (28% versus 50%, P =.01). For multivariable analysis, a conditional model was used with an age cut point at 60 years to address the different treatment intensities applied in the different age cohorts. In this model the only significant factors for CR achievement were TP53altered (OR, 0.55; 95%-CI, 0.30 to 1.00; P =.05) and age (OR for a 10 years difference, 0.67; 95%-CI, 0.52 to 0.87; P =.003). TP53 altered predicted for inferior survival; the 3-year estimated survival rates for CK+/TP53altered and CK+/TP53unaltered patients were as follows: event-free survival (EFS), 1% versus 13% (log-rank, P =.0007); relapse-free survival (RFS), 7% versus 30% (P =.01); and overall survival (OS), 3% versus 28% (P 〈 .0001), respectively. Other variables predicting for inferior OS in univariable analyses were age and MK. Among the cohort of CK+/MK+ AML, TP53altered patients had a significantly worse OS (P =.0004). Multivariable analysis (stratified for age at cut point of 60 years) revealed TP53altered (HR, 2.43; 95%-CI, 1.56 to 3.77; P =.0001), logarithm of WBC (HR, 1.62; 95%-CI 1.17 to 2.26; P =.004), and age (HR for 10 years difference, 1.26; 95%-CI, 1.01 to 1.56, P =.04), but not MK as significant variables for OS. In addition, explorative subset analysis suggested that allogeneic hematopoietic stem-cell transplantation in first CR which was performed in 30 CK-AML did not impact outcome in TP53altered CK-AML. In summary, TP53 is the most frequently known altered gene in CK-AML. TP53 alterations are associated with older age, genomic complexity, specific DNA copy number alterations, MK, and dismal outcome. In multivariable analysis, TP53 alteration is the most important prognostic factor in CK-AML, outweighing all other variables, including the MK category. TP53 mutational status should be assessed in clinical trials investigating novel agents in order to identify compounds that may be effective in this subset of patients. 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: 2011
    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...