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. 118, No. 21 ( 2011-11-18), p. 1461-1461
    Abstract: Abstract 1461 Background: Increased expression of EVI1 was reported to be associated with poor outcome in AML. The main mechanism of increased EVI1 expression is based on chromosomal rearrangements involving 3q26, where the EVI1 gene is located. The most frequently observed EVI1 rearrangements are inv(3)(q21q26) and t(3;3)(q21;q26). In addition, a variety of additional partner chromosomes and respective fusion partner genes were identified: 1q41 (DUSP10), 2p15–23, 3p25, 7q21 (CDK6), 7q34 (TCRB), 8q24, 12p13 (ETV6), or 21q22 (RUNX1). In addition, increased EVI1 expression has been reported in subsets of AML with normal karyotype or −7/7q-. Aim: We asked the question whether these AML subsets harbor cytogenetically cryptic EVI1 rearrangements. Patients and Methods: 606 AML and 377 MDS cases with normal karyotype or −7/7q- were screened using FISH technology with probes flanking breakpoints occurring in the EVI1 region (Kreatech, Amsterdam, The Netherlands). Results:EVI1 rearrangements were detected in twenty-seven patients with cytogenetically normal chromosomes 3. By further characterization using FISH analyses on metaphases three new and distinct EVI1 rearrangements were identified in these 27 cases. In detail, ten (2 MDS, 8 AML) cases demonstrated an inv(3)(p24q26), nine (5 MDS, 4 AML) cases showed a t(3;21)(q26;q11), and another eight (1 MDS, 7 AML) cases had a thus far not known der(7)t(3;7)(q26;q21). Moreover, EVI1 expression was measured by quantitative RT-PCR in 22/27 cases with material available (all values given as %EVI1/ABL1). In all investigated cases EVI1 expression was elevated. In 7 cases with inv(3)(p24q26) median EVI1 expression was 92.8 (range: 29.8–146.1), in 8 patients with t(3;21)(q26;q11) 104.9 (range: 41.4–176.3), and in 7 cases with der(7)t(3;7)(q26;q21) 101.8 (range: 4.4–210.4). For comparison, in 56 cases with inv(3)(q21q26)/t(3;3)(q21;q26) median EVI1 expression was 73.9 (range: 7.3–585.6), while EVI1 expression in normal bone marrow was 0.84 (range 0.75–1.28). We next aimed at investigating the novel partner genes deciphered here by using SNP array analyses (Cytogenetics Whole-Genome 2.7M array, Affymetrix, Santa Clara, CA). In 4 cases with der(7)t(3;7)(q26;q21) the high-resolution SNP microarrays revealed breakpoints in the CDK6 gene (breakpoints between 92,399,507 and 92,458,111; range: 59 kb) and centromeric of the EVI1 gene (breakpoints between 168,623,118 and 168,801,200; range: 178 kb). In 3 cases the EVI1-CDK6 rearrangements were confirmed by Sanger sequencing. The t(3;21)(q26;q11) was resolved as follows: in one case with t(3;21)(q26;q11) a duplication of the derivative chromosome 21 was present allowing the identification of breakpoints on chromosomes 3 and 21 by SNP microarray analysis. On chromosome 3 the breakpoint was located within the EVI1 gene (intron 2, breakpoint at 169,011,622) and on chromosome 21 within the NRIP1 gene (intron 3, breakpoint at 16,368,545). This rearrangement was confirmed by Sanger sequencing. In 7 additional cases with t(3;21)(q26;q11) an NRIP1-EVI1 fusion was detected by PCR. In 3 further cases the NRIP1 -EVI1 fusion was characterized on the DNA level by Sanger sequencing. Breakpoints in the EVI1 gene were located in intron 2 and 4 and in the NRIP1 gene in intron 1, 2 and 3, respectively. Finally, we performed an outcome analysis taking available clinical information into account. In cases with the novel EVI1 -rearrangements, median overall survival (OS) was 11.0 months and thus comparable to the median OS of AML with other described EVI1 -rearrangements (cohort: n=80 inv(3)(q21q26)/t(3;3)(q21;q26), n=24 other EVI1 -rearrangements). Conclusions: FISH screening with loci-specific probes for the detection of EVI1 rearrangements identifies a subgroup of patients with cryptic rearrangements and poor outcome, which cannot be detected by chromosome banding analysis. Thus, screening for EVI1 rearrangements enhances diagnostic accuracy and this method is particularly appropriate in AML with normal karyotype and AML with chromosome 7 abnormalities. Disclosures: Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Grossmann:MLL Munich Leukemia Laboratory: Employment. Zenger:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Kohlmann:MLL Munich Leukemia Laboratory: Employment. 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 ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 28, No. 24 ( 2010-08-20), p. 3858-3865
    Abstract: Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic malignancy that is characterized by features of both a myeloproliferative neoplasm and a myelodysplastic syndrome. Thus far, data on a comprehensive cytogenetic or molecular genetic characterization are limited. Patients and Methods Here, we analyzed 81 thoroughly characterized patients with CMML (CMML type 1, n = 45; CMML type 2, n = 36) by applying next-generation sequencing (NGS) technology to investigate CBL, JAK2, MPL, NRAS, and KRAS at known mutational hotspot regions. In addition, complete coding regions were analyzed for RUNX1 (β isoform) and TET2 aberrations. Results Cytogenetic aberrations were found in 18.2% of patients (14 of 77 patients). In contrast, at least one molecular mutation was observed in 72.8% of patients (59 of 81 patients). A mean of 1.6 mutations per patient was observed by this unprecedented screening. In total, 105 variances were detected by this comprehensive molecular screening. After excluding known polymorphisms or silent mutations, 82 distinct mutations remained (CBL, n = 15; JAK2V617F, n = 8; MPL, n = 0; NRAS, n = 10; KRAS, n = 12; RUNX1, n = 7; and TET2, n = 41). With respect to clinical data, a better outcome was seen for patients carrying TET2 mutations (P = .013). Conclusion The number of molecular markers used to categorize myeloid neoplasms is constantly increasing. Here, NGS screening has been demonstrated to support a comprehensive characterization of the molecular background in CMML. A pattern of molecular mutations translates into different biologic and prognostic categories of CMML.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2010
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Genes, Chromosomes and Cancer, Wiley, Vol. 52, No. 4 ( 2013-04), p. 410-422
    Type of Medium: Online Resource
    ISSN: 1045-2257
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 1018988-9
    detail.hit.zdb_id: 1492641-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 23 ( 2011-12-01), p. 6153-6163
    Abstract: Among acute myeloid leukemia (AML) patients with a normal karyotype (CN-AML), NPM1 and CEBPA mutations define World Health Organization 2008 provisional entities accounting for approximately 60% of patients, but the remaining 40% are molecularly poorly characterized. Using whole-exome sequencing of one CN-AML patient lacking mutations in NPM1, CEBPA, FLT3-ITD, IDH1, and MLL-PTD, we newly identified a clonal somatic mutation in BCOR (BCL6 corepressor), a gene located on chromosome Xp11.4. Further analyses of 553 AML patients showed that BCOR mutations occurred in 3.8% of unselected CN-AML patients and represented a substantial fraction (17.1%) of CN-AML patients showing the same genotype as the AML index patient subjected to whole-exome sequencing. BCOR somatic mutations were: (1) disruptive events similar to the germline BCOR mutations causing the oculo-facio-cardio-dental genetic syndrome; (2) associated with decreased BCOR mRNA levels, absence of full-length BCOR, and absent or low expression of a truncated BCOR protein; (3) virtually mutually exclusive with NPM1 mutations; and (4) frequently associated with DNMT3A mutations, suggesting cooperativity among these genetic alterations. Finally, BCOR mutations tended to be associated with an inferior outcome in a cohort of 422 CN-AML patients (25.6% vs 56.7% overall survival at 2 years; P = .032). Our results for the first time implicate BCOR in CN-AML pathogenesis.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1691-1691
    Abstract: Abstract 1691 RUNX1 (runt-related transcription factor 1) mutations constitute a disease-defining molecular aberration in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Mechanistically, deregulations occur either through balanced translocations or molecular mutations. Importantly, patient-specific RUNX1 mutations have been proposed to represent clinically useful biomarkers to follow disease progression from MDS to s-AML, as well as to monitor minimal residual disease (MRD) during AML treatment. As such, unbiased methodologies are warranted to provide necessary diagnostic sensitivity and throughput. Here, we investigated 116 samples from 25 patients (18 AML and 7 MDS) using next-generation amplicon deep-sequencing. For a longitudinal analysis starting at diagnosis and following the course of treatment peripheral blood (n=20) or bone marrow specimens (n=96) were obtained between 11/2005 and 6/2010. PCR assays targeting the RUNX1 beta isoform were performed with 60 ng of genomic DNA, obtained from mononuclear cells. In median, 5 time points per patient were analyzed with a median time span of 14 months (range: 5 – 34 months). The median sampling interval was 3.2 months. For each patient, one or more molecular mutations were known from standard testing at diagnosis using a combination of denaturing high-performance liquid chromatography and direct Sanger sequencing. In 166 amplicons covering the full spectrum of RUNX1 mutations we applied the 454 small volume Titanium chemistry assay to perform ultra-deep sequencing of specific PCR products (454 Life Sciences, Branford, CT). In median, 3346 reads per amplicon were generated, thereby allowing a highly sensitive assessment of RUNX1 mutational burden in these patients. As such, at 5% diagnostic sensitivity, 167 reads would cover a certain molecular mutation. At a cut-off of 0.5% sensitivity in median 17 reads were remaining for evaluation. First, we evaluated the concordance of NGS and conventional methods for the samples being taken at initial diagnosis. In all 25 patients deep-sequencing analyses concordantly detected the mutations known from conventional methods, i.e. in total 9 missense mutations, 1 nonsense mutation, 2 in-frame alterations, and 13 frameshift alterations. At initial diagnosis, deep-sequencing detected in AML cases the mutations with a median burden of 44% sequencing reads, whereas in MDS cases in median 35% sequencing reads harbored the mutations, respectively. In 2/25 (8%) cases, deep-sequencing detected additional low-level mutations (0.9% and 3.2%) that were not observed by standard techniques. Secondly, we investigated whether the technique of ultra-deep sequencing would be superior to current routine testing methods during follow-up and in detecting MRD. In 7/25 (28%) patients, an increasing clone size was detectable earlier than by conventional methods. Clone sizes with mutations as low as 0.2% - 7.0% of reads were detectable by NGS up to 9 months earlier during course of disease than by conventional methods. In no case did NGS miss mutations known by conventional methods. Overall, in 12/25 (48%) patients, ultra-deep sequencing revealed additional subclones and enabled the quantitative assessment of their respective clone size. In 6/25 (24%) cases this ultra-deep sequencing approach allowed to then quantitatively monitor the changing composition of parallel subclones per patient during treatment and disease progression. In particular, in two MDS patients dominant clones were proven to disappear during course of the disease and existing low-level or novel clones were emerging at s-AML stage. Similarly, in two AML patients dominant clones were suppressed during chemotherapy. Previously existing low-level mutations, already observed at the stage of initial diagnosis, were then detected at relapse with much greater mutational burden. Finally, in 2/25 cases with mutations concomitantly occurring in the same amplicon deep-sequencing was able to delineate monoallelic or biallelic status of the mutation. In conclusion, RUNX1 mutations are useful biomarkers with clinical utility for the detection of MRD in patients with hematological malignancies. We here demonstrated that amplicon-based NGS is a suitable method to accurately detect and quantify the variety of RUNX1 aberrations with high sensitivity and enables an individualized monitoring of disease progression and treatment efficacy. Disclosures: Kohlmann: MLL Munich Leukemia Laboratory: Employment. Grossmann:MLL Munich Leukemia Laboratory: Employment. Schindela: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.
    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 ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 752-752
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 752-752
    Abstract: Abstract 752 To evaluate the role of CEBPA mutations (CEBPAmut) in the context of other molecular mutations and cytogenetic aberrations we have analyzed 1567 AML cases for CEBPAmut. The patients were selected according to cytogenetics excluding the following karyotypes: t(15;17)/PML-RARA, t(8;21)/AML1-ETO, inv(16)/t(16;16)/CBFB-MYH11, inv(3)/t(3;3)/EVI1, t(6;9)/DEK-CAN and 11(q23)/MLL, and complex aberrations. The cohort was composed of 697 females and 870 males. Age ranges from 16.7 to 88.3 years (y) (median: 71.0 y). CEBPAmut were detected in 126/1567 cases (8.0%). The biologic characteristics of the CEBPAmut patients (age range 16.7 to 87.6 y, median: 64.8 y) were further investigated. Three different CEBPAmut patterns were observed: 1) in 50/126 cases (39.7%) one mutation and one wildtype allele were detected (monoallelic pattern), 2) 61 cases (48.4%) had two different mutations (biallelic pattern), 3) 15 cases (11.9%) had one mutation without detectable wildtype allele due to loss of heterozygosity (LOH). Overall we found 186 different mutations of following types: 1) 108 led to a premature N-terminal stop of the protein (6 due to a nonsense and 102 due to a frameshift mutation), 2) 60 were inframe mutations in the b-ZIP region, 3) 8 were frameshifts in the b-ZIP region and, 4) 2 were frameshifts 3`of the b-ZIP region, and 8 were C-terminal point mutations. Correlation to cytogenetics shows a normal karyotype (NK) in 86 (68.3%) of the 126 CEBPAmut patients whereas in 40 pts (31.7%) at least one cytogenetic aberration was detected (-7: n=7; +8: n=7, 9q-: n=2; 11q-: n=3, other trisomies: n=11; other non recurrent translocations: n=4, all others: n=6). Cytogenetic aberrations were more frequent in the monoallelic group (55%) compared to the biallelic (35%) (p=0.001) and to cases with LOH (10%) (p=0.047). Interestingly in the total cohort of 13 pts with monosomy 7 seven pts (53.8%) were CEBPA mutated (53.8%) and of these 6 were in the monoallelic group. Additional mutations were detected in 48 cases (RUNX1: n=11, NPM1: n=10, FLT3-ITD: n=20, FLT3-TKD: n=3, MLL-PTD: n=5, NRAS: n=9, IDH1: n=2, IDH2: n=7; 15 pts showed 2 and 2 pts 3 of these mutations). Similar to the cytogenetic aberrations the molecular mutations were more frequent in the monoallelic group (61.9%) compared to the biallelic (31.0%) and the LOH group (7.1%) (p=0.001). NPM1 mutations were mutually exclusive of biallelic CEBPAmut. As previously described we also detected a significantly higher expression of CD7 in the CEBPAmut compared to the CEBPAwt group (71.2% vs. 18.9%, p 〈 0.001). Furthermore, CD7 was higher expressed in biallelic cases as compared to the monoallelic ones (86.2% vs. 43.8%, p=0.011). It was similar in the LOH group (71.4%) compared to the biallelic group. There was no influence of cytogenetic aberrations or any additional mutation on EFS and OS. Solely the presence of high FLT3-ITD load ( 〉 0.5 FLT3-ITD/FLT3wt) was correlated with a shorter EFS (EFS at 2 y: 20.3% vs. 44.8%; p=0.020) and OS (OS at 2 y 54% vs 68%, p=0.047) when compared to the combined group of FLT3wt and those with an FLT3-ITD load of 〈 0.5. Regarding the different CEBPA groups the biallelic cases had a slightly better OS compared to monoallelic cases (OS at 2 y: 75.0% vs. 60.8%; n.s.). The 2-year OS in the LOH group was significantly lower (33.8%; p=0.023, compared to the biallelic group; and p=0.043 compared to 69.7% in the combined biallelic + monoallelic group). In addition, the different functional mutation types were analyzed. Out of frame mutations in b-ZIP had no specific impact on survival within the CEBPAmut cohort. N-terminal stop mutations and in frame mutations in b-ZIP were associated with favourable outcome (OS at 2 y: 72.1% vs. 43.9% all others mutations, p=0.007 and 2 y OS at 2 y: 76.3% vs. 46.9%; p=0.043, respectively), whereas all other mutations were extremely unfavourable (OS at 2 y: 0% vs. 70.5% compared to N-terminal and b-ZIP mutations, p 〈 0.001). In summary, 1) the biology and prognostic impact varies depending on distinct CEBPAmut patterns. 2) Cytogenetic and molecular alterations had no prognostic impact with the exception of FLT3-ITD with a mutation load of 〉 0.5 FLT3-ITD/FLT3wt. 3) Our data for the first time demonstrate that CEPBAmut with LOH are associated with an even inferior outcome than monoallelic mutations. In conclusion, these data show that CEBPA should be analyzed in detail in all NPM1wt NK AML and in those with unfavourable but non-complex karyotypes. Disclosures: Schnittger: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Alpermann:MLL Munich Leukemia Laboratory: Employment. Eder:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Grossmann: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.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2517-2517
    Abstract: Abstract 2517 Introduction: CEBPA (CCAAT/enhancer binding protein alpha) encodes a member of the basic region leucine zipper (bZIP) transcription factor family essential for myeloid differentiation. CEBPA mutations occur predominantly in AML with a normal karyotype and CEBPA mutated AML has been included as provisional entity in the WHO classification. Cases with biallelic mutations were reported as being associated with a favorable clinical outcome, thus patients are spared from allogeneic transplantation in first CR. Screening for CEBPA mutations in patients with AML is often performed applying a combination of fragment length analysis, DHPLC and subsequent direct sequencing using Sanger technique (conventional methods). Study Design: Next-generation amplicon deep-sequencing (454 Life Sciences, Branford, CT) is a more sensitive quantitative detection method than Sanger sequencing and thus was used to analyze 144 samples from 29 CEBPA mutated AML patients with a normal karyotype. For a longitudinal analysis starting at diagnosis and following the course of treatment bone marrow (n=134) or peripheral blood (n=10) samples were obtained between 5/2006 and 6/2011. The sequencing assay targeted the complete coding region of CEBPA, covered with 4 amplicons, and was performed using genomic DNA extracted from mononuclear cells. In median, 711 reads per amplicon were generated using the NGS assay, thereby allowing a sensitive quantitative assessment of the CEBPA mutational burden in order to monitoring minimal residual disease (MRD). In median, 4 time points per patient (range: 2–9) were included with a median time span of 9.5 months (range: 1–45 months). The median sampling interval was 2 months (range: 0.3–45 months). Results: First, we evaluated the concordance of mutation detection by comparing data from NGS and conventional methods using the samples at initial diagnosis. In all 29 AML patients NGS concordantly detected the mutations known from conventional methods, i.e. in total 26 frame-shifts, 15 in-frame alterations, 8 missense, and 2 nonsense mutations. Further, at initial diagnosis, deep-sequencing detected the mutations with a median burden of 44% sequencing reads (range 3%–88%) and thus already allowed a quantitative assessment of the mutational load. There was no difference observed for 6 patients with monoallelic vs. 21 cases with biallelic mutations (excluding 2 cases with homozygous alterations). We next investigated the distribution of clones and their underlying kinetics of clone size reduction during subsequent high-dose chemotherapy cycles. Overall, 26/29 cases were evaluable and the clone size was assessed by NGS at the second analysis point during course of disease–in median 63 days from time of diagnosis (range 10–215 days): (i) In 16/26 cases, deep-sequencing was not able anymore to detect the mutations as observed at diagnosis. 14 of these 16 negative cases stayed in complete molecular remission till the end of follow-up (median follow-up 6.5 months, range 1–34.2; 2/14 cases with allogeneic stem cell transplantation). (ii) Interestingly, in 4/26 cases residual disease with clones ranging from 8%–50% was indicative of non-response to treatment. In this subgroup 3/4 patients were characterized by resistant disease or early relapse (1 case excluded due to short follow-up). (iii) In the remainder group of 6/26 patients with mutations still detectable in a range of 0.12%–3.7%, complete molecular remission status was achieved at subsequent time points. However, in this group also 3 relapses were observed including 2 cases with allogeneic stem cell transplantation. Of note, in 3/6 cases from the latter group, NGS had outperformed conventional methods and was able to still detect residual clones enabling a superior monitoring of therapy response. In all cases with biallelic mutations both clones responded in parallel with similar kinetics. Moreover, 5 patients were investigated following relapse of AML or non-response to therapy. In all 5/5 analyses including 2 monoallelic and 3 biallelic alterations the same mutations as harbored at initial diagnosis remained detectable. Conclusion:CEBPA mutations provide increasing clinical utility for the detection of MRD. We here demonstrated that deep-sequencing is a suitable unbiased and robust method to accurately detect and quantify CEBPA aberrations enabling an individualized monitoring of disease status and treatment efficacy. Disclosures: Kohlmann: MLL Munich Leukemia Laboratory: Employment; Roche Diagnostics: Honoraria. Grossmann:MLL Munich Leukemia Laboratory: Employment. Fasan:MLL Munich Leukemia Laboratory: Employment. Stopp:MLL Munich Leukemia Laboratory: Employment. Alpermann:MLL Munich Leukemia Laboratory: Employment. Schindela: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.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 144-144
    Abstract: Abstract 144 PicoTiterPlate (PTP) pyrosequencing allows the detection of low-abundance oncogene aberrations in complex samples even with low tumor content. Here, we compared deep sequencing data of two Next-Generation Sequencing (NGS) assays to detect molecular mutations using a PCR-based strategy and, in addition, to uncover inversions, translocations, and insertions in a targeted sequence enrichment workflow (454 Life Sciences, Roche Diagnostics Corporation, Branford, CT). First, we studied 95 patients (CMML, n=81; AML, n=6; MDS, n=3; MPS, n=3; ET, n=2) using the amplicon approach and investigated seven candidate genes with relevance in oncogenesis of myeloid malignancies: TET2, RUNX1, JAK2, MPL, KRAS, NRAS, and CBL. 43 primer pairs were designed to cover the complete coding regions of TET2, RUNX1 (beta isoform), and hotspot regions of the latter genes. In total, 4128 individual PCR reactions were performed with DNA isolated from bone marrow mononuclear cells, followed by product purification, fluorometric quantitation, and equimolar pooling of the corresponding 43 amplicon products to generate one single sequence library per patient. For sequencing, a 454 8-lane PTP was used applying standard FLX chemistry and representing one patient per lane. The median number of base pairs sequenced per patient was 9.23 Mb. For each amplicon a median of 840 reads was generated (coverage range: 485–1929 reads). As initial proof-of-concept analysis 27 of the 95 patients with known mutations (n=32) as detected by conventional sequencing or melting curve analyses were investigated (range of cells carrying the respective mutation: 1.1% for JAK2 V617F to 98.14% for TET2 C1464X). In all cases, 454 NGS confirmed results from routine diagnostic methods (GS Amplicon Variant Analyzer software version 2.0.01). We then investigated the remaining 69 CMML patients: In median, 2 variances (range 1–8 variances), i.e. differences in comparison to the reference sequence, per patient were detected. These variances included both point mutations in all candidate genes and large deletions (12-19 bp) in CBL, RUNX1, and TET2. Only 20/81 patients of the CMML-cohort (24.69%) were without any detectable mutation. Secondly, in a cohort of six AML bone marrow specimens a custom NimbleGen array (385K format; Madison, WI) was used to perform a targeted DNA sequence enrichment procedure. In total, capture probes spanning 1.91 Mb were designed to represent all coding regions of 92 target genes (1559 exons) with relevance in hematological malignancies (e.g. KIT, NF1, TP53, BCR, ABL1, NPM1, or FLT3). In addition, the complete genomic regions were targeted for RUNX1, CBFB, and MLL. For sequencing, 454 Titanium chemistry was applied, loading three patients per lane on a 2-lane PTP including three molecular identifiers (MIDs) each. Data analysis was performed using the GS Reference Mapper software version 2.0.01. For the enrichment assay, the median enrichment of the targeted genomic loci was 207-fold, as assessed by ligation-mediated LM-PCR. Overall, 1,098,132 reads were generated in the two lanes, yielding a total sequence length of 386,097,740 bases. In median, 96.52% of the sequenced bases mapped against the human genome, and 66.0% were derived from the customized NimbleGen array capture probes, resulting in a median coverage of 18.7-fold . With this method it was possible to detect and confirm point mutations (KIT, FLT3-TKD, and KRAS) and insertions (FLT3-ITD). Moreover, by capturing chimeric DNA fragments and generating reads mapping to both fusion partners this approach detected balanced aberrations, i.e. inv(16)(p13q22) and the translocations t(8;21)(q22;q22) or t(9;11)(p22;q23). In conclusion, both assays to specifically sequence targeted regions with oncogenic relevance on a NGS platform demonstrated promising results and are feasible. The amplicon approach is more suitable for detection of mutations in a routine setting and is ideally suited for large genes such as TET2, ATM, and NF1, which are labor-intensive to sequence conventionally. The array-based capturing assay is characterized by a complex and time-consuming workflow with low-throughput. However, the ability to detect balanced genomic aberrations which are detectable thus far only by cytogenetics and FISH has the potential to become an important diagnostic assay, especially in tumors in which cytogenetics can not be applied successfully. Disclosures: Grossmann: MLL Munich Leukemia Laboratory: Employment. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Dicker:MLL Munich Leukemia Laboratory: Employment. Kazak:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Kern:MLL Munich Leukemia Laboratory: 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 ...
  • 9
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 417-417
    Abstract: Abstract 417 Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic malignancy that is characterized by features of both a myeloproliferative neoplasm and a myelodysplastic syndrome. Here, we analyzed 81 CMML cases (45 CMML-1, 36 CMML-2). In chromosome banding analysis 59/76 (77.6%) patients showed a normal karyotype (data not availabel in 5 cases). Recurrent chromosome aberrations were trisomy 8 (n=6; 7.9%), monosomy 7 (n=3; 3.9%), and loss of the Y-chromosome (n=5; 6.6%). Fluorescence in situ hybridization (FISH) detected the deletion of one allele of the TET2 gene in 4/71 cases (5.6%). Thus, the majority of cases can not be genetically characterized by these techniques. Therefore, we applied next-generation sequencing (NGS) technology to investigate 7 candidate genes, represented by 43 PCR-products, at known mutational hotspot regions, i.e. CBL (exons 8 and 9), JAK2 (exons 12 and 14), MPL (exon 10), NRAS (exons 2 and 3), and KRAS (exons 2 and 3). In addition, complete coding regions were analyzed for RUNX1 (beta isoform) and TET2. NGS was performed using 454 FLX amplicon chemistry (Roche Diagnostics Corporation, Branford, CT). The median number of base pairs sequenced per patient was 9.24 Mb. For each target gene a median of 911 reads was generated (coverage range: 736-fold to 1606-fold). This approach allowed a high-sensitive detection of molecular mutations, e.g. detecting the JAK2 V617F mutation down to 1.16% of reads. In total, 146 variances were detected by this comprehensive molecular mutation screening (GS Amplicon Variant Analyzer software version 2.0.01). In 80.4% of variances consistent results were obtained after confirming NGS mutations with melting curve analysis and conventional sequencing. In the remaining discrepant variances (19.6%) NGS deep-sequencing outperformed conventional methods due to the higher sensitivity of the platform. After excluding 19 polymorphisms or silent mutations 127 distinct mutations in 61/81 patients (75.3%) were detected: CBL: n=21 point mutations and one deletion (18 bp) found in 20 cases (24%); JAK2: n=8 mutations (V617F) found in 8 cases (9.8%); MPL: no mutations found; NRAS: n=23 mutations found in 18 cases (22.2%); KRAS: n=12 mutations found in 10 cases (12.3%); RUNX1: n=6 point mutations and one deletion (14 bp) found in 7 cases (8.6%); and TET2: n=49 point mutations and 6 deletions (2-19 bp; 5/6 out-of-frame) found in 41 cases (50.6%). Furthermore, in 21 TET2-mutated cases 11 mutations previously described in the literature were detectable, whereas 28 cases carried novel mutations (n=28). In the cohort of TET2-mutated cases 17/41 (41.3%) patients harbored TET2 abnormalities as sole aberration. Interestingly, CBL mutations were found to be significantly associated with TET2 mutations (Fisher's exact test, p=0.008). In 17 of 20 (85.0%) CBL-mutated cases TET2 abnormalities were concomitantly observed. In contrast, no significant associations were found between any of the point mutations or deletions and the karyotype. There were also no associations observed between molecular aberrations and the diagnostic categories CMML-1 and CMML-2. With respect to clinical data a trend for better outcome was seen for patients that carried either or both TET2 and CBL mutations (median OS 130.4 vs. 17.3 months, alive at 2 yrs: 72.0% vs. 43.9%; p=0.13). In conclusion, 75.3% of CMMLs harbored at least one molecular aberration. In median 2 mutations per case were observed. Compared to limited data from the literature we detected not only a higher frequency of CBL mutations, but also add data on novel TET2 mutations. In particular, comprehensive NGS screening here for the first time has demonstrated its strength to further genetically characterize and delineate prognostic groups within this type of hematological malignancy. Disclosures: Kohlmann: MLL Munich Leukemia Laboratory: Employment. Grossmann:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Kazak:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Weiss:MLL Munich Leukemia Laboratory: Employment. Dicker:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Kern:MLL Munich Leukemia Laboratory: 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...