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. 112, No. 11 ( 2008-11-16), p. 4250-4250
    Abstract: In CML real time PCR for the detection of residual disease usually has been done by the detection of the BCR-ABL-fusion transcript. In addition some reports have shown the usefulness of WT1 as a target for follow up in CML blast crisis. The aim of this study was to evaluate whether WT1 in addition to BCR-ABL assessment in imatinib treated CML can give any new information. Expression of BCR-ABL as well as WT1 (ELN assay) was normalized to ABL and expressed as % to ABL. In total 268 bone marrow samples (spls) of 40 patients (pts) have been analysed. Sixteen spls were newly diagnosed untreated CML. 252 spls were analysed at several timepoint during treatment with imatinib starting with a “near to untreated” sample with a & gt;50% BCR-ABL/ABL ratio. According to standard definitions 6 of the 40 pts showed no response, 16 minor (MR), and 18 major molecular response (MMR) as determined by BCR-ABL expression. Three of the MR and one of the MMR pts relapsed. The 14 pretreatment spls showed a median WT1 expression of 3.36 (range 0.176–14.9) whereas the median BCR-ABL level was 56.6 (range 11.4–149.2). Thus the BCR-ABL level was more than one log above the WT1 level and was more uniform within the cohort. This lower total expression and a background level of 0.04 (as estimated from 7 normal bone marrows) results in 2–3 log lower sensitivity of WT1 compared to BCR-ABL. The correlation of WT1 and BCR-ABL in the total cohort of 268 spls was low (r=0.259). Median WT1 expression in BCR-ABL negative spls (n=15) was 0.055 and thus correlates to that of normal spls. Regarding the total follow up in all 40 pts a good correlation of WT1 and BCR-ABL was found only in 9 cases, all of which had a WT1 expression & gt;10 at the start of follow up. 7 pts had a WT1 expression between 1–7 with good correlation to BCR-ABL but restricted sensitivity. 24 pts had a WT1 expression of & lt;1 and a BCR-ABL ratio & gt;50 at start of follow up. In these pts there was no correlation of BCR-ABL and WT1 during follow up. In two of these 24 pts there was even a negative correlation with increasing WT1 levels during FU even though BCR-ABL remained undetectable. Cytogenetic investigation of these two pts revealed the development of Ph-negative aberrant clones (one with +8 and one with +11). This result raised the hypothesis that increasing WT1 expression in BCR-ABL negative CML during follow up may indicate the development of BCR-ABL negative clones. Therefore we measured the WT1 expression in a new cohort of 26 CML pts that developed Ph-clones during follow up. All these pts had low ( & lt;0.5) or undetectable BCR-ABL. Chromosomal aberrations in these cases were −7 (n=1), +8 (n=12), +11 (n=1), del(20q) (n=1), −Y (n=10)–X (n=1). A high WT1 expression (range 5–177) was detected in the two cases with +11 and −7 and in 10 of 12 cases with +8. A low WT1 expression ( & lt;0.5) was detected in the case with 20q- and in all 10 cases with–Y. An intermediate expression of 1.5 was detected in the case with +X. In summary a high WT1 expression was detected in 12/26 (46%) of all cases with new chromosomal aberrations in a Ph-negative clone and in 12/16 (75%) excluding the 10 cases with -Y. So far, the clinical relevance of Ph-negative clones is unclear. In rare cases the development of secondary MDS has been described. The cases that were followed here were all still in CR of CML with no signs of a secondary disease. However, high WT1 expression was detected in most cases with +8 but never with–Y, possibly indicating unequal biological relevance of these aberrations. The 10–100-times elevated WT1 expression in the +8 and +11 cases suggest that these aberrations may be of adverse impact. However, further clinical follow up is needed to show the outcome in these pts. In contrast, none of the cases with–Y revealed elevated WT1 expression suggestive for the expansion of healthy–Y clones that frequently can occur in a normal male bone marrow. 21 pts with Ph-negative clones could be followed for 13–36 months. There was a good correlation of the WT1 expression level with the number of metaphases carrying the chromosomal aberration (r=0.729). In one case an increase of WT1 expression was measured 11 months before detection of the evolving +8 suggestive of a preexisting cytogenetically undetected clone. In conclusion, BCR-ABL is superior to WT1 as marker for follow up in CML. Increasing WT1 levels in BCRABL negative follow up spls can indicate the development of Ph-negative clones under imatinib treatment. Future studies will show whether the WT1 level in Ph-clones may indicate the relevance of certain aberrations for outcome in these patients.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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. 112, No. 11 ( 2008-11-16), p. 3203-3203
    Abstract: It has previously been shown that imatinib uptake into chronic myeloid leukemia (CML) cells is dependent on human Organic Cation Transporter 1 (hOCT1; SLC22A1). In more recent work on clinical samples it was further shown that low hOCT1 expression of this influx transporter may be an important mechanism of imatinib resistance. To further evaluate this issue we have retrospectively quantified pretreatment hOCT1 mRNA expression in 92 CML patients (pts) that responded with major molecular remission within the first year of treatment and compared these results to 19 pts with primary resistance to imatinib. We found that all 19 resistant pts had low hOCT1 expression (median: 2.032 (expressed as %hOCT1/ABL); range 0.18–4.24). Although the median hOCT1 expression at diagnosis in the responders was higher (median 8.417) the range was very heterogeneous (0.45–188.2) with only 30% of all responders having a significantly higher expression than the resistant pts. As in vitro studies have shown that genetic variants of the SLC22A1 gene that codes for hOCT1 can have a negative effect on the transport of some substrates we hypothesized that not only certain hOTC1 expression levels but also different genetic variants within the SLC22A1 gene may be associated with different efficiencies of imatinib uptake. Using high resoluting melting and subsequent sequencing we have genotyped exons 1, 2, 5, 6, 7, 9, 10, and 11 in 109 responders as well as in 55 resistant pts, thus each 326 alleles were evaluated. We detected 12 different exonic polymorphisms. Two of these, a G38D and a Y404C were so far undescribed variants. Both nonsynonymous variants were detected in heterozygeous forms, the G38D in one responder and the Y404C variant in one resistant pt. All other variants were detected in frequencies similar to those that have already been described (R61C: 0.07, L160F: 0.76, P341L: 0.01, G401S: & lt;0.01, M408V: 0.60, delM420: 0.19, G465R: 0.05, V519I: & lt;0.01). In addition the silent variants S51S and V501V were detected with frequencies of 0.26% and 0.01% respectively. All variants in heterozygous as well as in homozygous form were distributed equally between responders and resistant patients. Thus we did not find any correlation between SLC22A1 genotype and imatinib response. In addition there was also no correlation of any of these polymorphisms to the high expressers. We found that those polymorphisms that have been described to severely affect hOTC1 functions in vitro were very rare (P341L and G401S with & lt;0.01% each) or even never detected (P283L and R287L) in our cohort. Thus, although in vitro studies have shown that hOCT1 polymorphisms may severely affect function with respect to substrat specificity and transport efficiency of imatinib they do not seem to play a major role in response of CML patients to imatinib. In addition, we analyzed the three most frequent polymorphisms in exons 12, 21, and 26 in the multidrug resistence gene (MDR1) that codes for an efflux transporter implicated in imatinib efflux. In total 84 responders and 38 resistant patients were analyzed. We found that the exon12 nt1236t allele is more frequently observerd in resistant patients (p=0.045) whereas there was only a week association for the exon21 nt 2677t allele (p=0.121) and the exon26 nt3435t allele (p=0.139) to resistance. In conclusion, it seems to be unlikely that genetic variants of hOCT1 play a major role in imatinib resistance if at all, also the hOCT1 expression levels account for the response of only a few cases. It remains unclear whether hOCT1 plays a role in influx of imatinib or whether its function may be overwritten by other influx transporters like the very homologous and functionally redundant hOCT3 just in the vicinity of hOCT1. 3) The role of efflux transporters in imatinb resistance may be more important, however we detected only a weak association to certain polymorphisms in MDR1 to resistance in our cohort.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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. 438-438
    Abstract: Abstract 438 TET2 mutations have recently been described in various myeloid malignancies. To further evaluate the role of TET2 mutations in myeloproliferative neoplasms (MPN) we have analysed 96 MPN that have been well characterized by cytomorphology, cytogenetics and molecular genetics. The cohort consisted of 53 males and 43 females with a median age of 64.9 years (range: 16.6-86.3 years). Diagnosis was ET (n=22), HES (n=5), PMF (n=12), PV (n=32), MPN unclassifiable (MPN-u) (n=25). The ET, PMF and MPN-u were mainly selected for unmutated JAK2 status. Cytogenetics was availabel in 94/96 cases (98%). All ET and HES cases had a normal karyotype. In MPN-u 3 of 25 (12%), in OMF 3 of 12 (25%) and in PV 4 of 31(12.9%) revealed chromosomal aberrations. In all cases a BCR-ABL rearrangement was excluded. In addition in all cases mutation analysis for JAK2V617F, JAK2exon12, MPLW515 and CBL was performed. The total cohort was composed of 39 cases with JAK2V617F (3 × ET, 6 × PMF, 27 × PV, 3 × MPN-u), 5 cases with JAK2exon12 (all PV), 4 cases with MPLW515 (3 × ET, 1 × PMF), 2 cases with CBL mutation (both MPN-u). TET2 mutations were analyzed by amplification and sequencing of 21 PCR fragments covering the total coding region. Within the total cohort 20/96 cases (20.8%) revealed a TET2 mutation. Two different TET2 mutations in parallel were detected in three cases: one with MPN-u and two PV with homozygous JAK2V617F mutations. Throughout the gene the mutations were distributed as follows: exon4 (n=11), exon6 (n=4), exon7 (n=3), exon11 (n=5). 14 were missense, 3 nonsense and 6 were frameshift mutations. To analyze a further potential gene defect based on a TET2 deletion 15/20 cases from which methanol/acidic acid fixed cells were availabel were also analyzed by FISH (fluorescence in situ hybridization) for TET2 deletions. No deletion was detected in any of these cases. Thus with the exception of three cases with two different mutations all other mutated cases probably have retained one intact TET2 allele. With respect to diagnostic entities the TET2 mutations were distributed as follows, ET: 2/22 (9.1%), HES: 1/5 (20%), PMF: 4/12 (33.3%), PV: 9/31 (29%) and MPN-u: 4/27 (14.8%). With respect to other molecular genetic markers the TET2 mutations were distributed as follows: JAK2V617F: 10/20 (50%) (PV: n=8; PMF: n=2) from which 7/10 had JAK2V617F with a high mutation load (classified on the absence of a JAK2 wildtype allele) (PV: n=7; PMF: n=1), JAK2exon12: 1/5 (PV), MPLW515: 1/4 (PMF), CBL: 1/2 (MPN-u) , FIP1L1-PDGFRA: 1/5 case. Thus, 14/20 TET2 mutated cases (70%) revealed a detectable second mutation, 7 (50%) of which even with a high JAK2V617F mutation load. Taking also cytogenetics into account three further cases revealed aberrations resulting in a total of 17/20 TET2 mutated cases (85%) that had genetic markers in addition. 8/20 (40%) even had two or more genetic events in addition to the TET2 mutation. 2/3 cases with two TET2 mutation also had a very high JAK2V617F load. And five high load JAK2V617F cases had only a 50% TET2 mutation load, indicating that JAK2V617F was the first mutation in these cases followed by TET2 mutation as a second hit. There was no independent correlation of TET2 mutation with any of the analyzed MPN entities (p=0.359, n.s.). On the other hand TET2 mutations are more frequent in cases with further mutations compared to those without any other mutation irrespective of diagnosis (p=0.059). These data indicate that TET2 mutations 1) occur in all different subtypes of MPN and thus are no markers that indicate a specific entity, 2) are associated with other genetic markers that are more specific for certain MPN entities like FIP1L1-PDGFRA for HES, MPL for ET and PMF, JAK2exon12 for PV , 3) seem to be more likely associated with progression of MPN e.g. accumulation of mutations at least in MPN. Disclosures: Schnittger: MLL Munich Leukemia Lab: Equity Ownership. Tschulik:MLL Munich Leukemia Lab: Employment. Wendland:MLL Munich Leukemia Lab: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Dicker:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Lab: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership.
    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. 112, No. 11 ( 2008-11-16), p. 698-698
    Abstract: The aim of this study was to further evaluate the impact of minimal residual disease (MRD) in NPM1 mutated AML in comparison to other factors like FAB, cytogenetics, FLT3 mutations, NPM1 mutation type and age. In total 1002 samples of 219 NPM1 mutated (NPM1mut) patients (pts) were analysed at diagnosis, during, and after therapy. Pts were treated within different AML trials, and follow-up samples were referred to perform an NPM1 specific RQ-PCR for MRD. The cohort was comprised of 112 females and 107 males, median age was 58.8 years (range: 20–79 years). 207 had de novo AML (M0: n=5; M1: n=49; M2: n=55; M4=57; M5: n=28, M5: n=6; M7: n=1, nd: n=6), 4 s-AML and 5 t-AML. Cytogenetic data was available in 215 pts: 178 with normal (NK) and 37 with aberrant karyotypes (+4: n=4; +8: n=7; +21: n=2, two or more trisomies: n=4; -Y: n=4; del(7q): n=2; del(9q): n=3; del(20q): n=2; rare translocations: n=9). At diagnosis 87/219 pts (39.7%) had FLT3-ITDs in addition to the NPM1mut. FLT3-TKD status was available in 206 cases (14 mutated (6.7%) and 192 WT). The NPM1 mutation types were A (n=174), B (n=13), D (n=14), I: (n=4), L: (n=2), R: (n=4) and 8 with individual rare types. Univariate analysis for overall survival (OS) revealed unfavourable impact for age (p=0.049), and for FLT3-ITD (p=0.002), favourable impact for FLT3-TKD (p=0.046), and no impact for FAB, chromosomal aberrations or NPM1 mutation type. For MRD assessment for all 14 different NPM1 mutation types mRNA based RQ-PCR assays were established with sensitivities of 10,000–1,000,000. For each patient 2–17 samples (spls) were analyzed (median: 4) spanning a median follow up time of 252 days (range: 18–2347 days). Paired samples of diagnosis and relapse were available in 71 pts, in 8 pts also from second relapse. At relapse all cases had high NPM1 levels comparable to those at diagnosis. The FLT3-ITD status was mutated (+/+) at both time points in 25 pts and −/− in another 25 pts. 10 pts gained FLT3-ITD at relapse and 3 lost it. For 48 paired samples cytogenetics was available for both time points. A normal karyotype (NK) at both time points was detected in 36 pts, 7 cases showed a normal or aberrant karyotype (AK) at diagnosis and and AK at relapse (two of these gained additional aberrations at relapse), 2 different AK at both time points in were detected in 3 cases and a regression from AK to NK in 2 cases. These data show that NPM1 seems to be the primary genetic aberration in these cases and detection of NPM1 is more reliable to detect relapse than cytogenetics. To analyse the impact of NPM1 mutation levels on prognosis four different follow-up intervals were defined: interval 1: days 21–60 after start of therapy; interval 2: days 61–120; interval 3: days 121–365, 4: 〉 365 days. First a set of 605 samples referred for analysis during first line treatment were analysed. Using Cox regression analysis a significant impact of MRD levels (as continuous variable) on EFS was detected for interval 2 (128 spls, p=0.008), interval 3 (214 spl; p 〈 0.001), interval 4 (171 spls; p 〈 0.001) but not for the early interval up to day 60 showing that early molecular response is not relevant for long time outcome. A multivariate analysis showed that MRD was the most significant prognostic parameter (p 〈 0.001) (p-values for interval 3), followed by age (p=0.003), and pretreatment FLT3-ITD status (p=0.065). The same analysis was performed for a second set of 183 spls taken from 50 pts during salvage therapy after relapse. The most relevant interval for this group was between days 30–60 (26 spls; p=0.003). In a third set 87 spls from 28 pts after allogeneic bone marrow transplantation were analyzed. A prognostic impact of MRD could be shown for interval 2 (17 spls; p=0.005) and 3 (23 spls; p=0.006) (no samples from later intervals available). Of the total cohort 325 spls were analysed in parallel with RQ-PCR for NPM1 and genescan for the FLT3-ITD. A high correlation of both follow up markers was observed (r=0.807, p 〈 0.001). Although the method for NPM1 detection is 3–4 log ranges more sensitive our data suggest parallel assessment for FLT3-ITD for high risk patients as many of them aquired FLT3-ITD as additional marker during progression. In conclusion, MRD is the most relevant prognostic marker in NPM1 mutated AML and it is a very useful tool to assess therapy response and to guide therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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: British Journal of Haematology, Wiley, Vol. 146, No. 6 ( 2009-09), p. 665-668
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 1475751-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 11 ( 2009-09-10), p. 2220-2231
    Abstract: Nucleophosmin (NPM1)–mutated acute myeloid leukemia (AML), which is recognized as a provisional entity in the World Health Organization 2008 classification of myeloid neoplasms, accounts for 30% of AML. We analyzed 1227 diagnostic and follow-up samples in 252 NPM1-mutated AML patients with 17 different NPM1 mutation–specific real-time quantitative polymerase chain reaction (RQ-PCR) assays. Paired diagnostic/relapse samples of 84 patients revealed stable NPM1 mutations in all cases, suggesting that they are pathogenetically early events and thus applicable for minimal residual disease detection. A total of 47 relapses were predictable because of an NPM1 mutation level (%NPM1/ABL1) increase of at least 1 log or in 15 cases because of NPM1 mutation levels not decreasing less than 3 log ranges. A high prognostic value of NPM1 levels was shown for 4 different intervals after therapy was initiated. Furthermore, thresholds of 0.1 and 0.01%NPM1/ABL1 during/after treatment discriminated between prognostic subgroups. Univariate analyses, including age, white blood cell count, blast count, CD34 positivity, FLT3 mutations status, FAB type, karyotype, NPM1 mutation type, and pretreatment NPM1 mutational level, showed that, besides NPM1 mutation level, only age and FLT3-LM mutation status were prognostically significant for EFS. Multivariate analysis, including age, FLT3-LM status, and NPM1 mutation level at different time points, demonstrated that NPM1 level was the most relevant prognostic factor during first-line treatment. Similar results were obtained in patients undergoing second-line chemotherapy or allogeneic stem cell transplantation.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 884-884
    Abstract: Abstract 884 Blast crisis is the terminal phase of chronic myeloid leukemia (CML) with a short median survival of approximately six months. At present, little is known about molecular mechanisms underlying disease progression. We hypothesized that mutations occurring in other myeloid and lymphatic malignancies are acquired during disease progression from chronic phase to blast crisis. Here, in total 40 blast crisis CML cases (n=25 myeloid, n=10 lymphoid, n=5 not specified) were analyzed, all diagnosed between 9/2005 and 7/2009. First, all cases were investigated for IKZF1 deletions by PCR using specific primer pairs for the common intragenic deletions spanning from exon 2–7, or exon 4–7 as published by Iacobucci et al. (Blood, 114:2159-67, 2009). In total, in 17.5% (7/40) of cases intragenic IKZF1 deletions were detected. Secondly, next-generation deep-sequencing (454 Life Sciences, Branford, CT) was used to investigate 11 candidate genes in all 40 patients for a broad molecular screening. Known hotspot regions were sequenced for CBL (exons 8 and 9), NRAS (exons 2 and 3), KRAS (exons 2 and 3), IDH1 (exon 4), IDH2 (exon 4), and NPM1 (exon 12). Complete coding regions were analyzed for RUNX1, TET2, WT1, and TP53. To perform this comprehensive study, amplicon-based deep-sequencing was applied using the small volume Titanium chemistry assay. To cope with the great number of amplicons, in total 59, 48.48 Access Arrays were applied (Fluidigm, South San Francisco, CA), amplifying and barcode-tagging 48 amplicons across 48 samples in one single array (2,304 reactions). In median, 430 reads per amplicon were obtained, thus yielding sufficient coverage for detection of mutations with high sensitivity. Further, ASXL1 exon 12 aberrations were investigated by Sanger sequencing. In summary, after excluding known polymorphisms and silent mutations in 33/40 patients 53 mutations were identified: RUNX1 (16/40; 40.0%), ASXL1 (12/40; 30.0%), WT1 (6/40; 15.0%), NRAS (2/40; 5.0%), KRAS (2/40; 5.0%), TET2 (3/40; 7.5%), CBL (1/40; 2.5%), TP53 (1/40; 2.5%), IDH1 (3/40; 7.5%), IDH2 (0/40), and NPM1 (0/40). Thus, 82.5% of blast crisis CML patients harbored at least one molecular aberration. In median, one affected gene per patient was observed (range 1–5). In detail, RUNX1 was associated with additional mutations in other genes, i.e. 9/16 cases were harboring additional mutations in combination with RUNX1. Similarly, in 8/12 patients with ASXL1 mutations additional aberrations were detected. With respect to myeloid or lymphoid features ASXL1 mutations (n=11) were exclusively observed in patients with myeloid blast crisis (n=1 not specified), in contrast 5/7 IKZF1 cases were detected in cases with lymphoid features (n=1 myeloid, n=1 not specified). Interestingly, besides IKZF1 (n=5) and RUNX1 (n=3) alterations there was no other mutated gene occurring in lymphoid blast crisis CML. In addition, no aberration was detected in NPM1, and in contrast to published data, in our cohort only one patient harbored a mutation in TP53. Moreover, for 8 patients with mutations in IKZF1 (n=3), RUNX1 (n=3), ASXL1 (n=1), WT1 (n=2), and IDH1 (n=2), matched DNA from the initial diagnosis at chronic state was available. In these specimens respective IKZF1 deletions, RUNX1, and ASXL1 mutations were not detectable indicating that IKZF1, RUNX1, and ASXL1 mutations had been developed during disease progression and act as driver mutations in these cases. WT1 and IDH1 mutations occurred at first diagnosis in one case each, indicating these genes would constitute passenger mutations. In conclusion, this comprehensive study on 12 molecular markers enabled to characterize for the first time that 82.5% of blast crisis CML cases harbor specific molecular mutations. IKZF1 and RUNX1 alterations were identified as important markers of disease progression from chronic state to blast crisis. Moreover, technically, a novel combination of a high-throughput sample preparation assay for targeted PCR-based next-generation deep-sequencing was developed and allowed to broaden our molecular understanding in blast crisis CML. Disclosures: Grossmann: MLL Munich Leukemia Laboratory: Employment. Eder:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Wille:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership, 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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2560-2560
    Abstract: Abstract 2560 Background: Loss of CEBPA function due to mutations is thoroughly explored in AML. Recently epigenetic modifications such as promoter methylation have gained increasing interest as additional mechanisms for transcriptional regulation of cancer related genes. In this context, the clinical impact of aberrant CEBPA promoter methylation (PM) in AML is controversially discussed. The aim of this study was to clarify the frequency and the significance of aberrant CEBPA PM with regard to clinical features in a large cohort of de novo AML patients. The study comprises the CEBPA core promoter as well as the distal and proximal CEBPA promoter region as it has been shown that these upstream located regions also bear promoter activity. Patients:CEBPA PM was analyzed in a cohort of 574 de novo AML patients with normal karyotype (NK-AML) and without CEBPA mutations. The cohort was composed of 268 females and 306 males. Age ranged from 20.0 to 89.6 years (median: 63.7). In addition, methylation status was assessed in 48 patients with biallelic (n=10) or monoallelic (n=38) CEBPA mutations to exclude coincidence with CEBPA PM. As the fusion transcript RUNX1-RUNX1T1 is known to down-regulate CEBPA mRNA and protein levels in AML, we also analyzed CEBPA distal PM status in 44 RUNX1-RUNX1T1 positive AML patients to evaluate a possible correlation between CEBPA distal PM and RUNX1-RUNX1T1 induced down-regulation of CEBPA expression. All patients were intensively treated using standard AML protocols. Methods: Proximal PM and distal PM were analyzed in the total cohort using Sanger sequencing. Core PM was screened by methylation specific PCR in a subcohort of 326 CEBPA unmutated cases. Methylation data was correlated to clinical outcomes and to the presence of FLT3-ITD (n=175/568), NPM1 (n=256/565), RUNX1 (n=91/275), MLL-PTD (n=76/567), IDH1G105 (n=47/353), IDH1R132 (n=38/371), IDH2R140 (n=65/332) and IDH2R172 (n=12/342) molecular mutations. In addition, CEBPA mRNA expression levels were assessed by quantitative real time PCR (Taqman®, Life Technologies, Carlsbad, CA) in 39 cases with CEBPA distal PM and in 8 cases of the NK-AML cohort tested negative for CEBPA distal PM. CEBPA expression was normalized against the expression of the control gene ABL1. Results: The CEBPA distal promoter was methylated in 54/574 cases (9.4%) whereas CEBPA proximal PM was found in none of the 574 cases. Methylation of the CEBPA core promoter was detected in only 8 of 326 cases (2.5%). As CEBPA proximal and core PM seem to be rare events in AML, they were excluded from further analysis. None of the 48 CEBPA mutated cases revealed any PM and thus aberrant CEBPA PM and mutation status were mutually exclusive. Surprisingly, analysis of CEBPA mRNA expression level revealed no difference between CEBPA distal PM positive and CEBPA distal PM negative cases (mean ± SD 145 ± 97.9 and range 2.7–474.2 vs. 141.4 ± 85.3 and 23.1–259.2, n.s.) suggesting that CEBPA distal PM has no influence on CEBPA mRNA expression in NK-AML. In contrast, we observed a significantly higher frequency of CEBPA distal PM in patients with RUNX1-RUNX1T1 positive AML (n=17/44; 38.6%) compared to the NK-AML cohort (n=55/572; 9.4%) (p 〈 0.001) indicating a correlation between RUNX1-RUNX1T1 induced down-regulation of CEBPA expression and CEBPA distal PM. In the NK-AML cohort, there was no correlation between CEBPA distal PM and age, sex, white blood cell count or Hb levels at diagnosis compared to unmethylated cases. We also were not able to detect a significant correlation between the presence of CEBPA distal PM and the other molecular mutations except for the frequency of IDH2R140 mutations which was significantly lower in CEBPA distal PM positive compared to CEBPA distal PM negative cases (21/267; 7.9% vs. 13/65, 20%, p=0.010). In addition, CEBPA distal PM was not related to overall survival, event free survival or incidence of relapse in NK-AML. Also in subcohorts that were defined by specific molecular mutations (FLT3-ITD, NPM1, RUNX1, MLL-PTD, IDH1, or IDH2) no prognostic impact of CEBPA distal PM could be shown. Conclusion:CEBPA PM in NK-AML was detected in 11.9% of all cases and seems to be mainly restricted to the distal promoter (9.4%). In contrast to RUNX1/RUNX1T1 positive AML no impact of CEBPA PM on CEBPA mRNA expression levels was detected in NK AML. We also conclude that the presence of aberrant CEBPA PM has no clinical relevance in NK-AML and therefore is negligible as prognostic marker. Disclosures: Fasan: MLL Munich Leukemia Laboratory: Employment. Alpermann:MLL Munich Leukemia Laboratory: Employment. Grossmann:MLL Munich Leukemia Laboratory: Employment. Eder:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 827-827
    Abstract: TP53 is the most frequently mutated gene in cancer. The prognostic impact of TP53 mutations has been demonstrated in CLL, AML, and MDS. However, data on the frequency and prognostic impact of TP53 mutations in ALL is scarce. Aims We aimed at determining the TP53 mutation frequency, the association with cytogenetic subgroups and age, as well as the impact on survival. Patients and Methods In total, a large cohort of 625 patients with ALL was analyzed for TP53 mutations by deep-sequencing allowing to simultaneously quantify the mutation load. In all patients chromosome banding analyses have been performed. In addition, in 341 patients the copy number state of TP53 was determined by FISH. Results The cohort comprised 353 male and 272 female patients, median age was 49.5 years (range: 0.1-91.4 years). The cohort included the following groups: normal karyotype (n=101; 16.2%), t(9;22)(q34;q11) (n=162; 25.9%), MLL-translocations (n=37; 5.9%), MYC-translocations (n=40; 6.4%), t(12;21)(p13;q22) (n=15; 2.4%), low hypodiploidy ( 〈 40 chromosomes) (n=24; 3.8%), high hyperdiploidy (51-68 chromosomes) (n=38; 6.1%), complex karyotype (n=69; 11.0%), other cytogenetic abnormalities (n=139; 22.2%). Detailed data on immunophenotyping was available for 408 patients (T-lineage: n=105; B-lineage: n=267, Burkitt: n=36). In the total cohort, the frequency of TP53 mutations was 15.7% (98/625). TP53 mutations were most frequent in ALL with low hypodiploidy (22/24; 91.7%) and MYC-translocated ALL (25/40; 62.5%) and also quite frequent in ALL with complex karyotype (16/69; 23.2%), ALL with normal karyotype (13/101; 17.4%), and in MLL-translocated ALL (6/37; 16.2%). TP53 mutations were rare in t(9;22)(q34;q11) (7/162; 4.3%), high hyperdiploidy (3/38; 6.1%), and other cytogenetic abnormalities (6/139; 4.3%) and absent in ALL with t(12;21)(p13;q22) (0/15; 0%). Furthermore, TP53 mutations were less frequent in T-lineage ALL (8/105; 7.6%) as compared to B-lineage and Burkitt ALL (41/267; 15.4% and 21/36; 58.3%). TP53 mutation frequency increased with age (TP53mut 〈 60 years vs ≥ 60 years 10.8% (45/417) vs 25.5% (53/208), p 〈 0.0001). 25/64 (39.1%) TP53 mutated patients with available TP53 deletion status showed a deletion of the second allele, while in 17/319 (5.3%) TP53wt patients with available TP53 deletion status a TP53 deletion was detected (p 〈 0.001). 11/98 (11.2%) TP53 mutated patients showed two TP53 mutations. Median overall survival (OS) was significantly shorter in TP53mut vs TP53wt patients (18.8 months vs 75.5 months, p 〈 0.0001). OS at 4 years in patients 〈 60 years was 80.1% in TP53wt compared to 56.8% in TP53mut (p=0.012) and 59.3% vs 22.6% in patients ≥ 60 years (p 〈 0.0001). Also within the cytogenetic categories MYC-translocated and complex karyotype TP53 mutations had a significant adverse impact on overall survival. Further, patients with either two TP53 mutations or one TP53 mutation and an accompanying TP53 deletion had a significantly shorter OS as compared to patients with only one altered TP53 allele (median OS 11.5 vs 63.1 months, p=0.009). In contrast, OS in patients with a TP53 deletion without a TP53 mutation did not differ from patients without TP53 alterations. In addition, the TP53 mutation load was investigated by next-generation sequencing and varied between 2% and 98% (median: 41%). Within the subset of patients with TP53 mutation, patients with a mutation load 〉 20% showed a significantly shorter OS as compared to patients with a lower mutation load (median OS 11.5 months vs not reached, p=0.003). Interestingly, OS in patients with a TP53 mutation load ≤ 20% did not differ from TP53wt patients. In multivariate Cox regression analysis including parameters significantly associated with shorter OS in univariate analysis the following factors retained an independent adverse impact on OS: age ( 〈 60 years vs ≥ 60 years, HR=2.2; p=0.01), MLL-translocation (HR=2.8; p=0.03), and TP53mut 〉 20% (HR: 3.1, p=0.01). Conclusions 1. TP53 is mutated in 15.7% of ALL with the highest frequency in ALL with low hypodiploidy (91.7%) and MYC-translocated ALL (62.5%). 2. The TP53 mutation frequency increases with age. 3. TP53 mutations are associated with short survival independent of age and specific cytogenetic alterations. 4. TP53 mutations had a significant impact on OS only if the mutation load was 〉 20%. Disclosures: Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Weissmann:MLL Munich Leukemia Laboratory: Employment. Kuznia:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2015
    In:  Blood Vol. 126, No. 23 ( 2015-12-03), p. 2578-2578
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2578-2578
    Abstract: Background: Mutations in RUNX1 have been reported in 5 to 20% of AML. RUNX1 mutated AML is associated with a myeloid rather than monocytic differentiation, shows a typical pattern of cytogenetic abnormalities with a high frequency of trisomy 8 or 13, has a typical pattern of additional molecular mutations with a high frequency of accompanying ASXL1 and SF3B1 mutations and is nearly mutually exclusive of NPM1 and CEBPA double mutations and other entity-defining genetic abnormalities. In a subset of patients with RUNX1 mutations loss of the wild-type allele can be assumed due to a high mutation load. The aim of this study was the detailed analysis of a subset of RUNX1 mutated AML with RUNX1 wild-type loss with respect to accompanying cytogenetic and molecular genetic abnormalities and prognostic impact. Patients and Methods: A cohort of 467 AML with RUNX1 mutations (mut) at diagnosis identified during diagnostic work-up in our laboratory were the basis of this study. Median age was 72 years (yrs) (range 18-91 yrs), and male:female ratio 296: 171. 366 patients had de novo AML, 77 s-AML following MDS, 24 t-AML. For all patients (pts) cytogenetics and for 341 data on FAB subtype was available. Mutation data was available for NPM1 (n=456), MLL-PTD (n=453), CEBPA (n=449), FLT3-ITD (n=457), FLT3-TKD (n=457), WT1 (n=398), ASXL1 (n=313), TP53 (n=231), DNMT3A (n=177), TET2 (n=174), NRAS (n=305), KRAS (n=213) and SF3B1 (n=119). 64 patients with a mutation load of RUNX1 mutation 〉 70% evaluated by sequencing analysis were selected for further analysis. All 64 cases were analysed by genomic arrays (SurePrint G3 ISCA CGH+SNP Microarray, Agilent, Waldbronn, Germany) to determine the copy number state and copy neutral loss of heterozygosity (CN-LOH). Median age was 73 yrs (range 24-87 yrs), and male:female ratio was 27: 37. 50 patients had de novo AML, 11 s-AML following MDS, 3 t-AML. Results: Array CGH revealed a cytogenetically cryptic deletion on the long arm of chromosome 21 encompassing the RUNX1 gene in 5/64 (8%) patients while a CN-LOH on 21q including the RUNX1 gene was observed in 45 cases (70%). Thus in 50 cases (78%) with a high RUNX1 mutation load a RUNX1 wild-type loss (wt-loss) was detected by array CGH. In 43% (6/14) of the remaining cases the high RUNX1 mutation load was caused by amplification of the long arm of chromosome 21 either due to gain of whole chromosomes 21 or to an isochromosome 21q. First we focused on the characterization of RUNX1 mutated cases with RUNX1 wt-loss. In 22/50 cases (44%) an aberrant karyotype was observed with a distinct aberration pattern. 11 cases harbored +13, 5 had +8 and 6 cases a loss of 7q. No other recurrent abnormalities were observed. With respect to concurrent mutations the following frequencies were found: ASXL1 (42%), FLT3 -ITD (34%), TET2 (21%), KRAS (11%), MLL-PTD (8%), NRAS (7%), and FLT3-TKD (6%). No NPM1 mutation or CEBPA double mutations were identified. Comparison of those cases with RUNX1 wt-loss to all other RUNX1 mutated AML (n=417) revealed a significantly higher frequency of +13 (22% vs 9%, p=0.01) and FLT3 -ITD (34% vs 19%, p=0.015). FAB subtypes M0 and M1 were more frequent (46% vs 12%, p 〈 0.001; 35% vs 22%, n.s.) and M2 and M4 less frequent (14% vs 46%, p 〈 0.0001; 5% vs 17%, n.s.). Survival analyses were restricted to 212 de novo AML pts with RUNX1 mut who received intensive chemotherapy (median overall survival (OS): 20 months (mo), median event-free survival (EFS): 12 mo). Median OS and EFS was shorter in patients with RUNX1 wt-loss compared to those without (15 vs 20 mo, n.s., 10 vs 12 mo, p=0.04). In univariate Cox regression analysis a negative impact on OS was observed for RAS mut (relative risk (RR): 2.2, p=0.005), male gender (RR: 1.6, p=0.02), and age (RR: 1.3 per decade, p 〈 0.001). Shorter EFS was associated with RUNX1 wt-loss (RR: 1.7, p=0.04), RAS mut (RR: 1.9, p=0.02) and age (RR: 1.2 per decade, p 〈 0.001). In multivariate analysis RAS mut (OS: RR: 2.4, p=0.002; EFS: RR: 2.0, p=0.008) and age (OS: RR: 1.3 per decade, p 〈 0.001; EFS: RR: 1.2 per decade, p 〈 0.001) had independent prognostic impact. Conclusions: RUNX1 mutated AML with wild-type loss is a distinct AML subset that does not overlap with any of the genetically defined WHO categories and is characterized by an immature phenotype (81% FAB Subtype M0 and M1) and a higher frequency of +13 and FLT3-ITD as compared to RUNX1 mutated AML without wild-type loss. Wild-type loss and RAS mutations are associated with inferior outcome in RUNX1 mutated AML. Disclosures Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Nadarajah:MLL Munich Leukemia Laboratory: Employment. Fasan:MLL Munich Leukemia Laboratory: Employment. Perglerová:MLL2 s.r.o.: Employment. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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...