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: Annals of Hematology, Springer Science and Business Media LLC, Vol. 94, No. 11 ( 2015-11), p. 1927-1928
    Type of Medium: Online Resource
    ISSN: 0939-5555 , 1432-0584
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 1458429-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 111, No. 3 ( 2008-02-01), p. 1686-1689
    Abstract: We searched for JAK2 exon 12 mutations in patients with JAK2 (V617F)-negative myeloproliferative disorders. Seventeen patients with polycythemia vera (PV), including 15 sporadic cases and 2 familial cases, carried deletions or duplications of exon 12 in circulating granulocytes but not in T lymphocytes. Two of the 8 mutations detected were novel, and the most frequent ones were N542-E543del and E543-D544del. Most patients with PV carrying an exon 12 mutation had isolated erythrocytosis at clinical onset, unlike patients with JAK2 (V617F)-positive PV, most of whom had also elevations in white blood cell and/or platelet counts. Both patients with familial PV carrying an exon 12 mutation had an affected sibling with JAK2 (V617F)-positive PV. Thus, several somatic mutations of JAK2 exon 12 can be found in a myeloproliferative disorder that is mainly characterized by erythrocytosis. Moreover, a genetic predisposition to acquisition of different JAK2 mutations is inherited in families with myeloproliferative disorders.
    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. 126, No. 23 ( 2015-12-03), p. 4093-4093
    Abstract: Fusion oncogenes resulting from chromosomal aberrations are common disease drivers in myeloid malignancies. The most prominent example is BCR-ABL1 fusion present in chronic myeloid leukemia, which together with essential thromobocythemia (ET), primary myelofibrosis (PMF) and polycythemia vera (PV) belongs to the classic myeloproliferative neoplasms (MPN). The BCR-ABL1 negative MPNs are driven by somatic mutations in JAK2, MPL and CALR. MPN patients can progress to acute myeloid leukemia (AML) but the transformation process is not well understood. Studies using standard karyotyping and SNP microarrays have shown that disease progression is characterized by an increase in karyotype complexity. We aimed to identify novel fusion oncogenes in patients with BCR-ABL1 negative MPN during chronic phase and disease progression in high-throughput and cost-efficient manner using RNA-seq technology. In addition this approach enabled us to perform RNA-seq variant calling for identification of gene mutations on the same cohort of patients. Whole transcriptome sequencing was performed on 121 patients (112 chronic phase MPN and 9 secondary AML samples) and 23 healthy controls in a 100 base pair paired-end manner. The cohort consisted of 44% PMF, 22% ET, 12% PV and 6% secondary AML patients. The output of three fusion detection tools (Defuse, Tophat-fusion and SOAPfuse) was combined in order to increase sensitivity. Extensive filtering steps were applied in order to enrich for cancer specific fusion events, including filtering for fusions appearing in healthy individuals, filtering for read-throughs and false positives with external databases and manual inspection of sequencing reads. The outcome of analysis for Defuse, Tophat-fusion and SOAPfuse resulted in the total of 52, 54 and 38 candidate fusions, respectively. Candidate fusions were Sanger-sequenced and for Tophat-fusion and Defuse the validation rate was 60%, while for SOAPfuse only 20% could be validated. Approximately 70% of the fusion candidates were not shared among the 3 tools which underlines the importance of selecting the union of all calls from each tool rather than the intersect. We did not observe clustering of breakpoints along the genome. Most fusion candidates could be detected in PMF which corresponds to the disease entity that was most represented in the cohort (44% of patients). No enrichment for fusions was found in 7 triple negative (no JAK2, CALR, MPL mutations) cases. 42% of chromosomal aberrations were translocations, followed by duplication (31%), inversion (14%) and deletion events (11%). Among the intragenic fusions, approximately half had genomic breakpoints less than 1 Mb apart. 70% of validated fusions were out of frame, while 28% were in frame. In the leukemic samples a higher abundance of fusions was found (4/9). Typical fusions for de novo AML were not detected within secondary AML (sAML) samples. We did not detect a recurrent fusion oncogene in our patient cohort. In a PMF patient with JAK2-V617F mutation we identified a BCR-ABL1 fusion, indicating a clonal exchange which was consistent with patient's phenotype. Another PMF patient exhibited an inversion event involving the first exon of CUX1, causing a CUX1 loss of function. Other fusions in chronic MPN patients affected genes involved in histone modifications (SMYD3-AHCTF1, KDM4B-CYHR1). In post-MPN AML patients we identified a somatic in frame-fusion involving INO80D and GPR1 and a fusion truncating the first 3 exons of RUNX2 (XPO5-RUNX2). The high quality of RNA sequencing data, allowed us to set up a variant detection workflow that will be compared with matched samples that have been exome sequenced. Preliminary results could demonstrate that mutations in the JAK2 gene in a cohort of 96 patients were all correctly recalled, emphasizing its sensitivity. Fusion events among patients in chronic phase MPN are rare and the majority of these events imply loss of function of both fusion gene partners. This approach adds valuable information on the true frequency of inactivation of genes such as CUX1 in patients, as small inversions like the one described above would not be detectable by other methods. Detection of a subclone with BCR-ABL1 fusion underlines the strength of the fusion detection workflow for diagnostic purposes. Typical de novo AML fusions were not found in sAML and further suggests that de novo AML and sAML are distinct disease entities on a genetic level. Disclosures Gisslinger: Janssen Cilag: Honoraria, Speakers Bureau; Sanofi Aventis: Consultancy; AOP ORPHAN: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau; Geron: Consultancy. Kralovics:AOP Orphan: Research Funding; Qiagen: Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 4088-4088
    Abstract: Inherent tendency for thrombosis is a major complication in myeloproliferative neoplasms (MPN). The molecular basis of thrombosis in MPN is not well understood, however, genetic factors have been proposed to play a role. To agnostically investigate the role of common germline variation in MPN thrombophilia, we performed genome-wide association studies (GWAS) in MPN patient cohorts characterized for arterial thrombosis (AT) events. In our discovery cohort (n=383) from Vienna, Austria, 18% of patients have suffered from AT after MPN diagnosis, and 33% of patients showed records for AT events at any time. Of this discovery cohort, a subset of patients (n=302) selected independently from thrombotic status was genotyped on the Affymetrix Genome-Wide SNP 6.0 array platform. After assessment of case-control setup and genotyping quality as standardly implemented in GWAS, confidently genotyped single nucleotide polymorphisms (SNPs) were tested for allelic association with occurrence of AT after MPN diagnosis. We observed an association signal beyond genome-wide statistical significance (P 〈 5x10-8) at chromosome 6q25.3, tagged by rs6455579 and six additional closely related tag-SNPs. The 6q25.3 risk haplotype is common with a minor allele frequency (MAF) of ~8% in the Vienna MPN cohort. The minor allele rs6455579_C correlated with increased risk for AT. We next genotyped the full discovery cohort from Vienna (n=383) and a replication cohort from Pavia, Italy (n=505) using qPCR-based SNP genotyping for rs6455579 and computed odds ratios (ORs) to estimate the effect sizes of the association. In the Vienna cohort, rs6455579 genotype was significantly associated with both "AT after diagnosis" (P =1.90x10-8, ORhet=6.93) and "AT at any time" (P =6.96x10-4, ORhet=3.07). In the Pavia cohort, an association with "AT after diagnosis" could not be observed (P =0.14), however, the association of rs6455579 with "AT at any time" could be reproduced at formal statistical significance (P =7.92x10-3, ORhet=2.55). Moreover, Kaplan-Meier statistics on the Pavia cohort revealed a significant difference in AT-free survival after diagnosis upon rs6455579 genotype (log-rank-test P =0.009; rs6455579 homozygous major vs. heterozygous), underpinning the relevance of disease duration for the impact of the haplotype on MPN-related AT. To gain insight into the physiological mechanisms behind the 6q25.3 risk haplotype, we next evaluated the discovery cohort for correlations of the haplotype with clinical parameters other than AT. We could observe a significant trend for increased white blood cell (WBC) count in CALR mutated (n=90, P =2.50x10-3) but not in JAK2 mutated (n=182, P =0.52) ET and PMF patients carrying the risk allele. CALR mutated patients have been previously reported to exhibit significantly prolonged thrombosis-free survival as compared to JAK2 mutated patients. Thus, our observation indicates that the 6q25.3 germline risk haplotype might impact on WBC count most strongly in a subgroup of patients considered low risk for thrombosis based on the somatic mutational status. The 6q25.3 core haplotype (R2 〉 0.3) spans ~300 kilobases, covering intergenic sequence as well as promoter and 5' exons of the TULP4 gene. To test for the possibility of a rare coding variant in TULP4 or other more distant genes underlying the association through long-range linkage disequilibrium, we used genotype imputation in conjunction with the 1000 genomes reference panel to infer genotypes on all untyped variants (MAF 〉 1%) in a 5 megabase region centered on the core haplotype. We did not detect any coding variants reflecting the association, and the tag-SNPs from the initial GWAS remained the most strongly associated variants. Causative non-coding genetic variation identified in GWAS is thought to exert its function through differential regulation of specific target genes. Therefore, we evaluated a potential influence of the 6q25.3 risk haplotype on TULP4 gene expression in peripheral blood (The Cancer Genome Atlas (TCGA) LAML dataset, RNA-Seq on 173 acute myeloid leukemia patients). Indeed we detected significantly decreased TULP4 expression in risk haplotype carriers (P =0.029), providing indirect evidence for reduced TULP4 transcript levels impacting on elevated risk for AT in MPN. Further studies will be required to functionally assess the potential role of TULP4 in MPN-related AT. Disclosures Gisslinger: Novartis: Honoraria, Research Funding, Speakers Bureau; Geron: Consultancy; Sanofi Aventis: Consultancy; Janssen Cilag: Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; AOP ORPHAN: Consultancy, Honoraria, Research Funding, Speakers Bureau. Kralovics:AOP Orphan: Research Funding; Qiagen: Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 318-318
    Abstract: Abstract 318 The classical myeloproliferative neoplasms (MPNs) comprise of three entities: polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). Despite distinct phenotypic features of MPN entities they share characteristics like clonal hematopoiesis, risk for thrombosis and bleeding and tendency to transform to secondary acute myeloid leukemia (post-MPN AML). In order to investigate the genetic lesions associated with MPN a large single-center cohort of 311 MPN patients was analyzed for chromosomal aberrations using high resolution Affymetrix SNP 6.0 arrays. The cohort included 150 patients with PV, 90 with ET, 68 with PMF and 3 with post-MPN AML. Of the 311 patients, 144 (46%) had a normal karyotype and 167 (54%) harbored 1 to 8 detectable chromosomal aberrations. We found 51 gains, 102 deletions and 143 uniparental disomies (UPDs). A total of 13 recurrent chromosomal defects (more than three events) were detected. We investigated if either the number of chromosomal aberrations in a patient or specific types of lesions associate with a certain patient group defined by clinical criteria. Chromosomal aberrations were equally distributed among the three MPN entities and only 9pUPD showed significant clustering with PV. We did not detect an association between the number of chromosomal aberrations and disease duration. Patients positive or negative for JAK2 mutations did not differ significantly in the frequency of chromosomal aberrations (except of the association of 9pUPD with JAK2 positive MPN). Patients with complex karyotype were significantly older than patients with normal karyotype (P 〈 0.001). Transformation to post-MPN AML is an important complication in MPN. To investigate associations between chromosomal changes and transformation, we included additional 19 post-MPN AML patients from another center into the study (total N=22). Patients in the post-MPN AML group harbored significantly more chromosomal lesions (P 〈 0.001). Recurrent aberrations of chromosomes 1q, 7p, 7q, 5q, and 3q strongly associated with post-MPN AML. When we reviewed the clinical data of patients in chronic phase MPN harboring the leukemia-associated aberrations, they showed features of disease progression, and some transformed to AML at a later follow-up. We were able to map a common deleted region (CDR) on chromosome 4 to the tet oncogene family member 2 (TET2), a gene frequently deleted in myeloid disorders. On chromosome 7p we mapped a CDR to the Ikaros transcription factor (IKZF1) and a 7q CDR mapped to a novel putative tumor suppressor, the cut-like homeobox 1 gene (CUX1). Interestingly, in one patient who carried a UPD of chromosome 7q we did not detect a mutation in the CUX1 gene but an R288Q mutation was found in the EZH2 gene. Chromosome 7 aberrations in our cohort were strongly linked to post-MPN AML. Our results show that at least three chromosome 7 genes (IKZF1, CUX1, and EZH2) are relevant in leukemic transformation. In addition to chromosome 7, we found gains of chromosome 1q equally relevant in post-MPN AML. We mapped the common 1q amplification to a 3.5 Mbp region that contained the MDM4 gene. Mdm4 is a known negative regulator of p53 and was frequently shown amplified in various cancers. This result prompted us to investigate the relevance of the p53 pathway in post-MPN AML and we sequenced TP53 in all 22 leukemic patients and found mutations in 6 cases (27.3%). Interestingly, none of the patients with TP53 mutation carried an MDM4 amplification. Taken together, 10 out of 22 post-MPN AML cases (45.5%) had evidence of a p53-related defect. To gain deeper insight into the pathways involved in transformation to post-MPN AML we sequenced genes commonly affected in de novo AML, and found two patients with mutations in FLT3, two patients with RUNX1 mutations, two patients with either IDH1 or IDH2 mutations. We conclude that lesions known to play an important role in de novo AML are present only in a fraction of post-MPN AML patients. In this study we show that aberrations of the p53 pathway together with the chromosome 7 lesions affecting IKZF1 and CUX1 are present in 64% of all post-MPN AML patients. Our data give insight into the genetic complexity and heterogeneity of MPN patients in chronic phase as well as in post-MPN AML. The marked genetic heterogeneity of MPN patients will render targeted therapies challenging and underlines the requirement of personalized treatments. 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: 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
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 4101-4101
    Abstract: Allogeneic bone marrow transplantation (ABMT) is so far the only curative treatment option for patients with myelofibrosis (MF), although it carries risk of treatment related mortality and relapse. The effect of somatic mutations present in the clone causing the disease on the outcome of the ABMT is unknown. The aim of the study was to investigate the genetic basis of clonal evolution of MF patients undergoing ABMT and identify candidate prognostic markers of relapse. We performed high-resolution genetic analysis on DNA samples from five patients with MF who received ABMT. The analysis of acquired chromosomal deletions, gains and losses of heterozygosity was performed using SNP 6.0 microarrays. Somatic mutations in the genomes of the patients were identified using whole exome sequencing (WES) of tumor (granulocytes) and matched control tissue (T-lymphocytes). The 51-bp paired-end next generation sequencing was performed using HiSeq2000 system. The somatic origin of mutations was validated by Sanger sequencing. In the three relapsed cases we analyzed both the samples prior to ABMT and at relapse. For the two non-relapse cases only the baseline sample was analyzed, as the chimerism level at last follow up (3 and 5 years after ABMT) was 100% of the donor. We selected one relapse and one non-relapse case who did not carry mutations in JAK2 or MPL, as well as one relapse and one non-relapse case who had close to 100% JAK2-V617F mutational burden. Patient 1 was a 49 year-old diagnosed with MPL-W515A positive post essential thrombocythemia (ET) MF. Following ABMT the patient achieved complete clinical and molecular remission. Three years later the patient developed myelodysplastic syndrome with re-appearance of the MPL-W515A positive clone and within three months refractory anemia with excess blasts II phenotype. The clonal evolution analysis revealed that the clone causing the relapse was the same one present at the initial post-ET MF diagnosis, which acquired additional somatic mutations and chromosomal aberrations. The initial clone causing post-ET MF carried 12 somatic mutations including MPL, DNMT3A, U2AF1, ASXL1, SIRT2, RAD50 and other genes, while no chromosomal aberrations were detectable. The relapsed clone acquired additional 9 somatic mutations in genes TP53, MYO18B, CDYL and others, as well as deletion (del) 7p, del7q, chromothripsis of chromosomes 11 and 16, and del22q. Patient 2 is a 35 year-old diagnosed with post-ET MF. Mutations in JAK2 or MPL were not detected. Prior to ABMT a small clone with a deletion on chromosome 12q targeting SOCS2 gene among 3 other, could be detected by SNP array. The relapsed sample showed a full clone with 2 deletions targeting 2.5Mb region on chromosome 12 containing the SOCS2 gene, as well as a single gene ARID1B on chromosome 6. ARID1B is a member of SWI/SNF chromatin remodeling complex and a putative tumor suppressor. WES identified only 3 somatic mutations in this patient (MTUS2, SP3 and PCDH12). MTUS2 and SP3 mutations arose in the relapsed clone. Patient 3 is a 57 year-old diagnosed with post polycythemia vera MF. The patient carried JAK2-V617F mutation coupled with a 9p UPD, therefore having a 99.2% mutational burden. Within 4 months after BMT the JAK2-V617F clone was detectable with 32% mutational burden, reaching 98% one month later. Apart from 9p UPD no other chromosomal aberration was detectable before BMT or at relapse. Preliminary WES data revealed 7 somatic mutations at relapse, including APBB1 and DOCK4. Patient 4 was diagnosed with JAK2/MPL negative post-ET MF, while patient 5 had primary MF. In patient 4 we identified 10 somatic mutations and no chromosomal aberrations, while patient 5 carried the 9p UPD and a deletion of 20q, 11 somatic mutations and a germline variant in TET2 gene. It seems that the sheer number of somatic mutations does not predict ABMT outcome. As the validation of WES hits is still ongoing, we expect to see other gene mutations that may serve as positive or negative predictors of the ABMT outcome in patients with MF. They will reflect the genomic adaptation that facilitates the escape from the graft-versus-myelofibrosis effect of the donor cells and allows the clonal dominance leading in some cases to disease progression. Further analysis of possible presence of subclones carrying relapse associated mutations before ABMT will have important implications for estimating success of ABMT in MF 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: 2013
    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. 127, No. 3 ( 2016-01-21), p. 362-365
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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. 126, No. 23 ( 2015-12-03), p. 606-606
    Abstract: Essential thrombocythemia (ET) and primary myelofibrosis (PMF) are chronic myeloproliferative neoplasms (MPN) characterized by clonal hematopoiesis and hyperproliferation of terminally differentiated myeloid cells. Most of the cases are sporadic and driven by somatic mutations, although familial clustering is observed. The most common mutation affecting 50-60% of the cases is JAK2-V617F, while 25-30% of the patients carry somatic mutations in exon 9 of CALR. MPL exon 10 mutations affect ~5% of the cases. JAK2, CALR and MPL mutations are mutually exclusive and account for 〉 90% of ET and PMF cases. In 12% of ET and 5% of PMF cases the disease drivers remain unknown. These patients are termed as triple negative. The mutational analysis for diagnostic purposes is limited to exons 14 of JAK2, exon 10 of MPL and exon 9 of CALR. The aim of this study was to identify disease causing mutation in triple negative cases of ET and PMF. To identify the somatic mutations that are potential disease drivers in triple negative MPN we performed whole exome sequencing (WES) on paired samples from the tumor and control tissue of 4 patients with ET and 4 patients with PMF. We identified somatic mutations in 3/8 analyzed cases. In two PMF cases we identified somatic mutations in genes relevant for MPN- TET2, ASXL1, CBL, SRSF2 and a mutation in MPL-S204P. We did not identify a novel recurrent mutation. In the 5 cases without somatic mutations, we looked for germline mutations in genes relevant for MPN. We identified germline mutations MPL-V285E and JAK2-G571S in one PMF case and one case of ET, respectively. SNP microarray analysis for presence of chromosomal aberrations revealed a uniparental disomy of chromosome 6p in the case with MPL -V285E mutation, suggesting clonal hematopoiesis. To determine the frequency of MPL and JAK2 mutations outside exons 10 and 14 in triple negative MPN, we performed Sanger sequencing of all coding exons of MPL in 62 patients and of JAK2 in 49 patients. We detected variants outside exon 10 of MPL in 6/62 cases (9.7%). MPL-T119I, MPL-S204F, MPL-E230G and MPL-Y591D were somatic mutations, while MPL-R321W was germline. We identified an additional patient with MPL-S204P mutation, however the control tissue was not available. JAK2 variants were found in 4/49 cases (8.1%). JAK2-G335D and JAK2-V625F were germline mutations, while for the patients with JAK2-F556V and JAK2-G571S the control tissue was unavailable. In total, we identified non-canonical MPL mutations in 8/70 (11.4%) and JAK2 mutations in 5/57 (8.8%) triple negative cases of ET and PMF. All mutations were heterozygous. The mutations in MPL and JAK2 were mutually exclusive in our patient cohort. The expression of identified MPL mutants did not induce cytokine independent growth of Ba/F3 cells, but the MPL-Y591D expressing cells showed marked hypersensitivity to TPO compared to the wild type. Using a luciferase reporter assay in JAK2-deficient gamma 2A cells, where we transiently expressed the wild type or mutant MPL cDNAs, JAK2, STAT5, STAT5 reporter Spi-Luc, and pRL-TK for transfection control, we could demonstrate that all identified MPL mutations lead to constitutive activation of JAK/STAT signaling. As the detection of activity required longer times (48h) than for the MPL-W515K (24h), we concluded that the identified mutations have a milder effect of the function of MPL. By Western immunodetection we could demonstrate that expression of JAK2-F556V and JAK2-V625F in Ba/F3-MPL cells, lead to the increased phosphorylation of STAT5 in the absence of cytokines. We also observed increased sensitivity to TPO in the Ba/F3 MPL cell lines expressing JAK2-F556V and JAK2-V625F. JAK2-V625F and JAK2-F556V are mild gain-of-function mutations, while JAK2-G335D and JAK2-G571S do not seem to alter the function on the JAK2 protein. The results of our study suggest that sequencing of all coding exons of MPL and JAK2 is recommended for the diagnostic work-up of the ET and PMF patients who do not carry other more common mutations. The lack of evidence for clonal disease in 50% of the triple negative cases and presence of germline mutations suggests that a proportion of cases are likely to be hereditary MPN-like disorders. Application of whole genome sequencing or RNA sequencing for fusion oncogene detection will likely fill in the gap of the remaining triple negative MPN cases with clonal hematopoiesis in which we did not identify a recurrent driving mutation using WES. Disclosures Gisslinger: AOP ORPHAN: Consultancy, Honoraria, Research Funding, Speakers Bureau; Sanofi Aventis: Consultancy; Geron: Consultancy; Janssen Cilag: Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau. Kralovics:AOP Orphan: Research Funding; Qiagen: Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 123, No. 15 ( 2014-04-10), p. 2416-2419
    Abstract: Somatic indels of CALR exon 9 are present in about 20% to 25% of sporadic patients with essential thrombocythemia or primary myelofibrosis. These mutations are found also in familial cases of essential thrombocythemia or primary myelofibrosis as somatically acquired events.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    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. 435-435
    Abstract: Abstract 435 Transformation to acute myeloid leukemia (AML) is a major complication of myeloproliferative neoplasms (MPNs). Although clinical prognostic factors have been defined such as age, severe anemia, elevated white blood cell count with a high proportion of immature myeloid cells and a high number of circulating CD34+ cells, there is little knowledge of the genetic changes leading to leukemic transformation in MPN. To gain insight into these genetic changes, we studied 12 patients with post-MPN leukemia using Affymetrix SNP 6.0 microarrays. Acquired genetic lesions were detected in 10 patients. Beyond well described frequent events like 9pUDP, chromosome 7p deletions (del7p) emerged as a novel recurrent defect in this patient group. Mapping of the minimal deleted region of del7p by combining microarray, copy number and loss of heterozygosity analysis resulted in a minimal deleted region of 0.25 Mb in size, restricted to the IKZF1 gene. IKZF1 is encoding the transcription factor Ikaros, which is known to have a pleiotropic function in the regulation of hematopoiesis, as well as crucial involvement in hematopoietic malignancies, primarily of the lymphoid lineages. Based on the observation of 16.7% (2 out of 12) of post-MPN leukemia patients carrying an IKZF1 deletion, we further screened a chronic phase MPN patient cohort from Vienna for the defect and found 1 out of 235 informative patients carrying an IKZF1 deletion. Thus, deletion of IKZF1 exhibited statistically significant clustering with post-MPN leukemia when compared to chronic phase MPN (P=0.0063). To validate these results in an independent patient cohort, we screened 20 post-MPN leukemic and 156 chronic phase MPN patients from Pavia for loss of IKZF1, and found 4 and 1 positive patients, respectively (P=0.0006). Taking these two cohorts together, IKZF1 deletions could be found in 0.5% of chronic MPN patients and 18.8% of post-MPN leukemic patients, showing statistically significant association with post-MPN leukemia (P 〈 0.0001). We could define haploinsufficiency as mechanism of IKZF1 tumor suppressor inactivation, since IKZF1 exon sequencing of del7p patients did not show any mutations on the non-deleted allele of IKZF1 and IKZF1 mRNA could be detected in del7p patients. In order to characterize the role of IKZF1 deletion in clonal expansion, we monitored oncogenic mutation and deletion burdens in granulocytes of two del7p patients in serial samples over time. In both patients, one being positive for JAK2-V617F and the other for MPL-W515L, increased IKZF1 deletion burden was detected as a late event after the acquisition of oncogenic mutations. We also examined the hematopoietic progenitors of a del7p patient by genotyping individual BFU-E and CFU-GM colonies for defects detected in granulocytes. IKZF1 deletion was found to be a late defect in the clonal evolution of MPN, occurring after oncogenic mutations such as JAK2-V617F and chromosomal aberrations such as del13q. In murine hematopoietic progenitor cells, sh-RNA induced deficiency for Ikzf1 resulted in cytokine hypersensitivity in vitro. Based on these data we can conclude that IKZF1 deletion does not induce arrest in myeloid differentiation, it is a late genetic event in the clonal evolution of MPN and represents an important step in the leukemic transformation of a subpopulation of MPN 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: 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...