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. 120, No. 21 ( 2012-11-16), p. 3542-3542
    Abstract: Abstract 3542 The prognosis of acute promyelocytic leukemia (APL) has improved markedly over the last two decades. Clinical trials have demonstrated excellent survival of patients receiving anthracycline-based chemotherapy in combination with all-trans-retinoid acid (ATRA). It is not clear whether the excellent survival results demonstrated in clinical trials during recent years, also do apply to unselected patients including elderly and frail patients for whom a clinical trial may not be available. In order to investigate survival and death rates in APL over the course of treatment, we conducted a retrospective analysis of survival in 105 consecutive APL-patients diagnosed in Denmark January 2000 through June 2012. Data were retrieved from the Danish National Acute Leukemia Registry which covers 95 – 100 % of all acute leukemia cases diagnosed in Denmark since January 2000 (not including children). A diagnosis of APL was confirmed in 105 (3.9 %) of 2726 adult patients (age ≥ 15 years) with acute myeloid leukemia (AML) diagnosed in Denmark during the 12½ year period. This corresponds to an incidence rate of 1.53 APL cases per million inhabitants per year. The diagnosis of APL was based upon cytogenetic analysis (performed in 96 (91 %) of cases), and/or interphase FISH (iFISH), and/or RT-PCR in a total of 100 cases. In 5 cases clinical signs and morphological changes in bone marrow were highly suggestive of APL and these cases were, thus, deemed to have APL. Median age at diagnosis of APL was 50 years (range 15 to 83 years) and median leukocyte count was 2.65 (range 0.2 to 99.0 × 109/L). In 104 patients with data available for analysis, very early death (VED, within the first week from the date of diagnostic bone marrow) occurred in 10 cases (9.6 %), and early death (ED, death within 30 day from diagnosis) occurred in 22 (21 %) cases. Death between day 30 and 5 years from diagnosis occurred in 11 cases. No deaths were seen after 5 years from diagnosis. For all patients, estimated overall survival at 10 years was 65 % (Figure 1). Patients surviving beyond day 30 from day of diagnosis (82 cases) had an excellent overall survival of greater than 80 % at 10 years (Figure 2). Survival of patients were strongly correlated with WHO-performance status whereas age over 50 years and presenting leukocyte count greater than 10 × 109/L could not be definitely associated with inferior survival (Table 1 and Figure 3). Table 1. Factors of importance to survival in unselected APL-patients Probability of overall survival (Univariate Cox Regression, nevaluable= 104) Probability of overall survival (multivariate Cox Regression, nevaluable= 101) Variable Hazard ratio 95% CI of HR P value Hazard ratio 95% CI of HR P value Age (≤50 vs. 〉 50 years) 2.17 1.07–4.42 0.03 1.44 0.67–3.09 NS WHO performance status (0–1 vs. ≥2) 5.20 2.57–10.5 〈 10−4 4.06 1.88–8.78 〈 10−4 WBC (≤10 × vs. 〉 10 × 109/L) 1.76 0.86–3.61 NS 1.62 0.79–3.33 NS From this population-based analysis we conclude that early death continues to be the predominant hazard to patients with APL. Two thirds of deaths in APL-patients do occur during the first month from diagnosis. Patients with a poor performance status at disease presentation carry a particularly dismal prognosis. For all APL-patients, every possible effort should be made to facilitate prompt diagnosis and speedy initiation of treatment with ATRA and anthracycline-based chemotherapy. When timely applied, currently available treatments are highly effective and result in cure in a high proportion of patients including elderly patients and patients with high leukocyte count at time of disease presentation. 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: 2012
    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. 120, No. 22 ( 2012-11-22), p. 4311-4316
    Abstract: Some women suffering from leukemia require bone marrow transplantation to be cured. Bone marrow transplantation is associated with a high risk of sterility, and some patients are offered fertility preservation by cryopreservation of the ovarian cortex. Transplantation of the ovarian cortex to women cured of leukemia who became menopausal is currently not performed because of the risk of introducing the disease. In this study, individual pieces of ovarian cortex intended for reimplantation from 25 patients with leukemia were transplanted to each of 25 nude mice for 20 weeks. The ovarian cortex was examined before and after transplantation by histology and immunohistochemistry, and RT–quantitative PCR (in the 7 patients with a known marker). Seventeen patients had the ovarian cortex retrieved when they were in complete remission. Before transplantation, 4 of 7 pieces (2 from patients in complete remission) of ovarian cortex had a positive RT–quantitative PCR. After transplantation, none of the mice revealed any sign of disease, neither in the pieces of ovarian cortex transplanted nor in any of the murine organs evaluated. Thus, the ovaries from patients in complete remission do not appear to contain viable malignant cells contrasting ovarian tissue retrieved before treatment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    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. 118, No. 21 ( 2011-11-18), p. 3514-3514
    Abstract: Abstract 3514 Isocitrate dehydrogenase (IDH) is a metabolic enzyme that catalyzes a reaction in the tricarboxylic acid cycle. Gain of function mutations in the IDH1/2 genes have been reported in different malignancies and are observed in 15–30% of de novo AML with association to a normal karyotype and to NPM1 mutations. The exact role of IDH1/2 mutations in leukemogenesis remains to be determined. IDH mutations have not previously been studied in a cohort of therapy-related myelodysplasia (t-MDS) and therapy-related acute myeloid leukemia (t-AML). To evaluate the frequency of IDH1/2 mutations in t-MDS and t-AML, and their possible association to type of previous therapy and to other genetic abnormalities, DNA from 140 well-characterized patients with t-MDS (n=89) and t-AML (n=51) were analyzed with high-resolution melting followed by sequencing. All patients have previously been examined cytogenetically and investigated for mutations in 12 other genes: FLT3(ITD, TKD), KIT, JAK2, KRAS, NRAS, BRAF, PTPN11, RUNX1, MLL(ITD), CEBPA, NPM1, and TP53. In total, IDH mutations were detected in 12 of 140 patients (9%). 3 patients had a mutation in IDH1 and 9 patients had a mutation in IDH2 (Table 1), all mutations previously reported in de novo AML. No patients had concurrent IDH1 and IDH2 mutations. IDH mutations were not related to previous therapy with alkylating agents, topoisomerase II inhibitors or radiotherapy, but were significantly associated with other types of therapy not firmly established to be leukemogenic (p=0.004). The latency period to development of t-MDS/t-AML was not different between IDH1/2 positive (+) cases and cases with IDH (wt) (64 and 48 months, respectively, p=0.118). 4/5 cases with t-MDS and IDH+ progressed to AML compared to 27/84 t-MDS cases with IDHwt (p=0.048).Table 1:Characteristics of 12 patients with t-MDS/t-AML and mutations in IDH1/2CaseAge/sext-AML/t-MDSPrevious therapyKaryotypeOther mutationsIDH Mutation1974/FAMLAlk45,XX,-7/48,XX,der(1;7)(q10;p10),+11, +13/46,XX–IDH1 R132G2963/FAMLRT46, XXNPM1 FLT3-ITDIDH1 R132G3663/FAMLAlk46,XX,+2,+8/47,XX,der(6)t(1;6) (q?25;p21),+8N-RASIDH2 R172K4472/MMDSAlk46,XY,+1,der(1;7)(q10;p10)/46,XY–IDH2 R140Q5562/FMDS→AMLRT46, XXRUNX1IDH2 R140L7272/FMDS→AMLAlk, T II, RT46,XX,+1,der(1;7)(q10;p10)/50,XX,idem, +8,+9,14+21RUNX1IDH2 R140Q8178/MMDS→AMLAlk46,XY,der(17)t(11;17)(q13;p13),i(13) (q10)/47,idem,+der(13)t(11;13) (q13;p11)IDH2 R172K10443/FMDS→AMLAlk47,XX,+1,der(1;7)(q10;p10),+8RUNX1IDH1 R132C10944/FAMLMtx, Aza46, XXIDH2 R140Q11952/FAMLAlk, T II, RT46, XXNPM1IDH2 R140Q13325/MAMLVCR, MTX, Asp,6-MP46, XXIDH2 R140Q18060/MMDS→AMLMtx46, XXMLL-ITDIDH2 R140Q6-MP, 6 mercaptopurine; Alk, alkylating agent; Asp, l-asparaginase; Aza, azathioprine; Mtx, methotrexate; RT, radiotherapy, T II, topoisomerase inhibitor, VCR, vincristine. IDH mutations were significantly associated with a normal karyotype (6/12 cases with IDH+ vs. 18/128 with IDHwt, p=0.006) and der(1;7)(q10;p10) resulting in trisomi 1q and loss of 7q (4/12 cases with IDH+ vs. 7/128 with IDHwt, p=0.008), but was inversely correlated to other chromosome 7 abnormalities (1/12 cases with IDH+ vs. 54/128 with IDHwt, p=0.03). No patient with mutated IDH had chromosome 5 abnormalities, TP53 mutations or recurrent balanced translocations. 7/12 patients with mutated IDH1/2 had other gene mutations characteristic of AML (Table 1). The frequency of each of these other mutations were not different from patients with wildtype IDH1/2 (RUNX1, p=0.4; NPM1, p=0.2; FLT3, p=1.0; MLL, p=0.165; N-RAS, p=1.0). In conclusion, mutations of IDH1/2 were observed in 9% of patients with t-MDS/t-AML. They were not related to any specific type of therapy but perhaps associated with transformation from MDS to AML. IDH mutations clustered in the genetic pathway characterized by a normal karyotype and mutations of NPM1, and the pathway characterized by 7q−/−7 and RUNX1 point mutations. The significant association observed between IDH1/2 mutations and der(1;7)(q10;p10) may indicate that this cytogenetic aberration represents a specific entity, biologically distinct from other chromosome 7 abnormalities. This is also supported by the different clinical outcome between cases with der(1;7) and other cases with -7/7q- (Sanada et al, Leukemia 2007). Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 109, No. 11 ( 2007-06-01), p. 4973-4979
    Abstract: We recently identified a disease-specific gene CLLU1 in chronic lymphocytic leukemia (CLL) and also demonstrated that high CLLU1 expression levels predict poor clinical outcome. To validate this finding, we measured CLLU1 mRNA expression levels by real-time reverse transcriptase–polymerase chain reaction (RT-PCR) in 175 patients with CLL. Analyses of IgVH mutational status, ZAP-70 expression, CD38 expression, and chromosomal aberrations were also performed. High levels of CLLU1 expression were associated with shorter overall survival (P 〈 .001), with a 7% increase in risk of early death by each doubling of the CLLU1 expression level. Stratification for age at diagnosis demonstrated a strong prognostic significance of CLLU1 expression in patients younger than 70 years (P 〈 .001), but not in patients aged 70 or older (P = .61). The prognostic significance of IgVH mutational status and ZAP-70 expression had a similar age-dependent variation. Multivariate analysis in the younger age group showed that CLLU1 expression analysis added further prognostic information within all prognostic subgroups, with the exception of patients with unmutated IgVH CLL. Only CLLU1 expression and IgVH mutational status had independent predictive power. Thus, analysis of CLLU1 expression is highly applicable in risk prediction in CLL for patients of an age eligible for risk stratification.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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. 106, No. 11 ( 2005-11-16), p. 709-709
    Abstract: We have recently identified and cloned a novel disease specific gene CLLU1, with a strong prognostic significance in a small test cohort of CLL patients (ASH 2004, #770). To validate this finding in a larger series, 176 newly diagnosed, previously untreated CLL patients referred to Rigshospitalet, Copenhagen in 1991–1999, were investigated. Clinical characteristics of the population were: 116 stage A patients (66 %), median age 69.9 years, 98 males (56%), median follow up 59 months. The end points for the statistical analysis were overall survival and time to initiation of treatment. At time of analysis 91 (52%) patients had died and 104 (59%) had commenced treatment. Frozen patient samples, taken at time of diagnosis, were investigated for CLLU1 expression, as well as for mutational status, CD38 expression, ZAP-70 expression and cytogenetic aberrations. CLLU1 expression was assayed by QRT-PCR using the cDNA1 splice variant, and expressed as fold upregulation above the CLLU1 level in normal B-cells. CD38 and ZAP-70 analysis was performed by flow cytometry, using a 20 % cut-off level. Cytogenetic analysis was performed by FISH. The previously reported (ASH 2004, #770) median upregulation of CLLU1 in CLL cells was accurately reproduced and confirmed (25.5-fold (N = 176) vs. 27.7-fold (N = 59) in test population). Also in accordance with the pilot study, segregation of the patients based on the median CLLU1 expression level divided the population in two groups with significantly different times to initiation of treatment and borderline significant difference in overall survival. However, further analysis found an optimal cut-off level at 40-fold upregulation of CLLU1 expression; use of this cut-off demonstrated a highly significant difference in overall survival for patients with upregulated expression of CLLU1 (p = 0.0023). The median overall survival was 60.2 months for patients with CLLU1 expression above 40-fold compared to 100.8 months for the group with lower expression level. Likewise the time to initiation of first treatment was significantly shorter in the high-level expression group (p = 0.0018, median time to first treatment 9.3 months vs. 54.9 months). Furthermore, CLLU1 expression was upregulated in all poor prognostic subgroups investigated: Binet stage B or C, IgVH unmutated, unfavorable cytogenetics, high CD38 expression and high ZAP-70 expression. Our new study confirms that CLLU1 is a strong prognostic factor in CLL, comparable to other established prognostic markers. The exclusive upregulation of CLLU1 expression in CLL suggests a putative role for CLLU1 in the pathogenesis of CLL, and makes this gene a strong candidate for future gene-targeted treatment strategies. Figure Figure
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    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. 120, No. 21 ( 2012-11-16), p. 2394-2394
    Abstract: Abstract 2394 Introduction: We have recently shown that the allelic methylation level of the non-coding RNA, VTRNA2-1, predict outcome in acute myeloid leukemia (AML) (Treppendahl et al., 2012). VTRNA2-1 is located on chromosome 5q31.1, in the commonly deleted region for higher risk myelodysplastic syndrome (MDS) and de novo AML. Around 75% of the healthy Danish population carries a monoallelically methylated VTRNA2-1 promoter while the rest carries 2 unmethylated promoters. A hypermethylated promoter was only observed in patients. These interindividual differences in the methylation pattern are intriguing, and our data suggest that the gene dosage of this particular type of ncRNA may play an important role in tumor progression or response to therapy, since AML patients with hypomethylation of both alleles of the VTRNA2-1 promoter have a significantly better prognosis, while those with hypermethylation or loss of the second VTRNA2-1 copy have a poorer outcome(Treppendahl et al., 2012). Accordingly, we speculated if the allelic methylation levels of VTRNA2-1 also predict outcome in higher risk MDS patients. Methods: Bone marrow mononuclear cells from primary higher risk MDS patients (IPSS category INT2 and HIGH risk) and peripheral blood mononuclear cells, sampled during treatment with azacytidine, were analyzed for promoter methylation by pyrosequencing and methylation specific melting curve analysis. Results: Bone mononuclear cells from 57 higher risk MDS patients, never treated with azacytidine, were examined for VTRNA2-1 promoter methylation. 18 (32%) cases carried an unmethylated promoter (less than 15% methylation), 31 (54%) cases carried an intermediate methylated promoter (15–41% methylation) and 8 (14%) cases carried a hypermethylated promoter (more than 41% methylation). Patients with hypomethylation of the VTRNA2-1 promoter have a considerable better prognosis than those with intermediate or hypermethylation of the VTRNA2-1 promoter (P=0.026). Interestingly, in an azacytidine treated cohort the survival benefit of having an unmethylated promoter disappeared and a tendency towards a better survival of the methylated cases were observed (N=28, P=0.180).The in vivo effect of azacytidine on VTRNA2-1 methylation were examined in a small cohort of MDS patients (N=6), showing that azacytidine can induce demethylation of the VTRNA2-1 promoter in peripheral blood mononuclear cells. Discussion: Our studies show, that the allelic methylation of VTRNA2-1 can predict outcome, not only in AML patients, but also in higher risk MDS patients not treated with azacytidine. Patients with hypomethylation of the VTRNA2-1 promoter have a considerable better outcome than those with intermediate methylation or hypermethylation of the promoter. Interestingly, in the group of azacytidine treated patients there was no difference in survival between cases with and without methylation of the VTRNA2-1 promoter These data could indicate that patients with methylation of the VTRNA2-1 promoter might have a survival benefit of the azacytidine treatment. Thus we suggest that constitutive interindividual differences in the methylation of VTRNA2-1 potentially can be used as a pretreatment marker for selecting patients who will have a survival benefit of azacytidine treatment. This is to our best knowledge the first study identifying a potential epigenetic marker for selecting patients with benefit of treatment with azacytidine before treatment start. Our study is conducted in a quite heterogeneous and small patient cohort, and it has to be verified in a more homogenously treated larger group of MDS patients, ideally in a prospective clinical trial. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Hematology Vol. 2007, No. 1 ( 2007-01-01), p. 392-397
    In: Hematology, American Society of Hematology, Vol. 2007, No. 1 ( 2007-01-01), p. 392-397
    Abstract: In therapy-related myelodysplasia (t-MDS) and acute myeloid leukemia (t-AML), at least eight alternative genetic pathways have been defined based on characteristic recurrent chromosome abnormalities. Patients presenting as t-MDS and patients presenting as overt t-AML cluster differently in these pathways. The cytogenetic pattern depends on the type of leukemogenic therapy received: alkylating agents, topoisomerase II inhibitors, or radiotherapy. Three types of gene mutations are observed in MDS and AML: (1) Activating mutations of genes in the tyrosine kinase–RAS/BRAF signal transduction pathway, leading to increased cell proliferation (Class I mutations); (2) Inactivating mutations of genes encoding hematopoietic transcription factors, resulting in disturbed cell differentiation (Class II mutations); and (3) Inactivating mutations of the tumor suppressor gene p53. At least 14 different genes have been identified as mutated in t-MDS and t-AML, clustering differently and characteristically in the eight genetic pathways. Class I and Class II mutations are significantly associated, indicating their cooperation in leukemogenesis The chromosome aberrations and gene mutations detected in the therapy-related and in the de novo subsets of MDS and AML are identical, although the frequencies with which they are observed may differ. Hence, therapy-related and de novo MDS and AML are identical diseases and should be subclassified and treated similarly.
    Type of Medium: Online Resource
    ISSN: 1520-4391 , 1520-4383
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 2084287-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 2585-2585
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2585-2585
    Abstract: The majority of patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) has acquired cytogenetic and molecular abnormalities of diagnostic and prognostic importance. Lately, focus has been on genes involved in epigenetic regulation, such as IDH, TET2, ASXL1 and DNMT3A, in which mutations have been shown to affect prognosis. The subgroup of MDS/AML with chromosome 7 abnormalities, are associated with somatic mutations of the RUNX1 gene, and so far monosomy 7 and der(1;7)(q10;p10) have been regarded as similar cytogenetic entities because they both result in loss of 7q. However, we have recently shown that mutations of the IDH gene are significantly associated with der(1;7)(q10;p10), but are inversely correlated with other chromosome 7 abnormalities in therapy-related MDS (t-MDS) and AML (t-AML) (Westman et.al. Leukemia 2013; 27(4):957-9). The aim of the study was to further molecularly characterize a larger cohort of patients with der(1;7)(q10;p10) and to compare the frequency of IDH and other mutations to MDS/AML cases with monosomy 7 as the sole abnormality. Methods Genomic DNA from 19 de novo and therapy-related MDS/AML cases with der(1;7)(q10;p10) was analyzed for mutations of FLT3, NPM1, IDH1/2, RUNX1 and DNMT3A genes by Sanger sequencing. For comparison 22 cases with monosomy 7 were investigated for mutations of the same genes. Additional investigations of possible mutations of ASXL1 and TET2 are ongoing and will be presented. Statistical evaluations were performed using Fisherxs exact test (two-tailed) or Wilcoxonxs two-sample test. Results There was no difference between patients with der(1;7) and monosomy 7 in clinical characteristics such as sex, age, presentation as MDS or AML, or de novo or therapy-related disease (Table 1). In total, 14 of 19 patients with der(1;7)(q10;p10) had mutations of IDH, RUNX1 or DNMT3A. Seven patients had a mutation in only one of these 5 genes, while the remaining 7 patients had mutations in 2 of the genes (Table 1). Nine patients had RUNX1 mutations (47%), 7 patients had IDH mutations (37%), and 3 patients had DNMT3A mutations (16%). As for patients with monsomy 7, nine of 22 patients had mutations of IDH, RUNX1 or DNMT3A but only one of the patients had mutations in more than one gene (Table 1). Five patients had RUNX1 mutations (23%), 3 patients had DNMT3A mutations (14%), and one patient had a mutation of IDH1 (5%). No mutations were detected in NPM1 or FLT3 in any of the patients. When comparing molecular characteristics, mutations of RUNX1, IDH, and DNMT3A were significantly more common in patients with der(1;7) compared to patients with monosomy 7 (p=0.03). IDH mutations were significantly associated with der(1;7) (p=0.02), whereas there was no difference in the distribution of RUNX1 and DNMT3A mutations between patients with der(1;7) and patients with monosomy 7 (p= 0.1 and 0.7, respectively). There was no difference in mutation frequency between patients with de novo and therapy-related MDS/AML (p=0.3). Conclusions In MDS/AML with chromosome 7 abnormalities, IDH mutations are significantly associated with der(1;7) compared to cases with monosomy 7, whereas mutations of RUNX1 and DNMT3A are equally distributed between the two cytogenetic subgroups. This difference in mutation status of IDH supports that der(1;7) and monosomy 7 should not be regarded as similar entities and suggests that der(1;7) has a specific biological effect in leukemogenesis different from that of other chromosome 7 defects. Our findings are in line with a recent multicenter study showing different clinical outcomes for patients with der(1;7) compared to patients with -7/7q- (Ganster et.al., Prognostic impact of der(1;7) in MDS is different from del(7q), EHA 2013). More studies are needed to determine if der(1;7) and monosomy 7 show other molecular differences than IDH1/2 mutation status. 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2688-2688
    Abstract: Introduction: Several recent publications have advanced our knowledge of the prognostic significance of clonal cytogenetic abnormalities in MDS, yet the genetic risk assessment of the rare karyotypic aberrations in MDS patients (pts) remains unknown. Using the German-Austrian (G-A) Cytogenetics Database, we previously defined 24 cytogenetic prognostic subgroups; however, 12 subgroups characterized by non-complex (isolated or one additional abnormality only) karyotypes with del(9q), del(15q), t(15q), del(12p), −X, t(1q), t(7q), t(17q), −21, t(11q23), +19, t(5q) were observed infrequently ( & lt;10 pts) and considered too few for an informative risk assessment. To increase the number of informative pts and expand/validate the statistical robustness of these rare cytogenetic subgroups, an international collaboration was initiated and promoted under the auspices of the MDS Foundation. Patients and Methods: A total of 90 new MDS pts with rare recurring abnormalities was collected from 12 MDS Foundation Centers of Excellence, of which 66 pts fulfilled the non-complex requirement. Survival was estimated by Kaplan-Meier analysis and restricted to pts treated by supportive care only. The final analysis included 108 pts: del(9q)(n=10); del(12p)=17; −X=10; t(1q)=13; t(7q)=14; −21=12; t(11q23)=12; +19=10; t(5q)=10. The frequency of non-complex del(15q), t(15q) and t(17q) remained below ten pts and deemed ineligible for further analysis at this time. Results: The pooled international data showed an excellent correlation with the G-A data set in 6 of 10 cytogenetic subgroups: t(7q): 34.7 months (mo.) median survival (G-A) vs. 34.7 mo. (new data), del(9q): not reached vs. 63.1 mo., t(11q23): 20.0 vs. 28.0 mo, del(12p): 108.0 mo. vs. not reached, +19: 19.8 vs. 21.7 mo., −21: 32.0 vs. 35.0 mo. A moderate correlation was found for t(1q): 34.7 mo. vs. not reached and for del(15q): not reached vs. 26.7 mo. Discordant median survival was observed for t(5q): 4.4 mo. vs. not reached and −X: 56.4 mo. vs. 15.7 mo. Conclusions: Through an international collaboration, we were able to define the prognostic impact of nine distinct yet infrequent, recurring cytogenetic aberrations observed in MDS pts. The discordant median survival differences observed in two subgroups may be attributed to various translocation partners and/or secondary abnormalities. Even though data collection remains ongoing, the current results expand and refine the cytogenetic prognostic classification system for pts suffering from MDS and underscore the need for standardized cytogenetic testing in MDS for further international collaborations.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3629-3629
    Abstract: Abstract 3629 Introduction: A tumor suppressor has long been sought for in the commonly deleted region on chromosome 5q in myelodysplastic syndrome and acute myeloid leukaemia (MDS/AML). MicroRNAs (miRs) may have tumor suppressor function since they may repress the expression of proto-oncogenes through post-transcriptional regulation by binding to the 3′UTR of mRNA. It has recently been shown that several types of haematological malignancies have globally altered expression of miRs. DNA methylation cause targeted down regulation of individual tumor suppressor genes that may be directly involved in the pathogenesis of MDS/AML and we speculated whether epigenetic downregulation of miRs also plays a role in MDS/AML pathogenesis. Methods: Cell lines were treated with the hypomethylating agent 5-azacytidine. Upregulated miRs were identified by microarray (Exiqon) analysis and confirmed by RT qPCR analysis. Transcription start site was determined by 5′RACE and promoter methylation by methylation specific melting curve analysis. MiR expression and processing were analysed by qPCR, Northern blotting, and si-RNA mediated knock down of Drosha. Clinical data were collected from patient files. Results: We focused on one upregulated miR that locates to the commonly deleted region of AML/MDS. We found that the processing of this “miR” was independent of Drosha, and northern blotting showed that this was in fact a different type of non-coding RNA (ncRNA). The gene that encodes this ncRNA has a CpG island at the transcription start site. This CpG island is unmethylated in normal CD34+ cells, normal peripheral blood lymphocytes and methylated in three different myeloid cells lines (HL60, NB4, F-36P). Normal CD34+ cells and normal peripheral blood lymphocytes expressed the ncRNA, while no expression was seen in the cell lines. However, treatment by 5-azacytidine derepressed expression of this ncRNA. Bone marrow mononuclear cells from 101 high-risk MDS and AML patients, isolated at the time of diagnosis, were examined for methylation of the ncRNA promoter. Methylation was detected in 48% of the patients. Patients with no methylation have a significant better survival as compared to patients with methylation of the promotor. Discussion: This is the first example of targeted methylation of this type of RNA that has not previously been implicated in cancer. The preliminary results indicate that methylation of this ncRNA may be used as a prognostic marker for survival in high-risk MDS- and AML patients. We are currently investigating the function of this ncRNA by performing stable transfections into MDS/AML cell lines. Disclosures: Jones: Lilly: Consultancy; Millipore: Consultancy.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...