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. 412-412
    Abstract: Abstract 412 Background: Somatic mutations of the ASXL1 (Additional Sex Comb-Like 1) gene on chromosome 20q11.1 were identified in various myeloid malignancies with the highest incidence reported in CMML (∼40%) and lower frequencies in MDS, AML, CML, and myeloproliferative neoplasia. The ASXL1 protein has been suggested to act as chromatin modifier and the highly conserved C-terminal plant homeo-domain (PHD) finger is presumably critical for its function. ASXL1 mutations cluster in exon 12 and are mainly frameshift mutations predicted to remove the PHD domain. We and others (Paschka et al., Haematologica 2011;96(s2);425; Chou et al., Blood 2010;116:4086–94) have recently reported first results on the unfavorable prognostic impact of ASXL1 mutations in AML. However, the clinical relevance of these mutations still needs to be elucidated in larger AML cohorts. Methods: Mutational analyses of ASXL1 were performed on diagnostic samples from 1429 patients with AML aged 18 to 61 years. All patients were intensively treated on one of two AMLSG trials [AML HD98A (n=745), Schlenk et al., J Clin Oncol. 2010;28:4642–8; AMLSG 07–04(n=684), NCT00151242]. GeneScan-based fragment analysis of several amplicons spanning ASXL1 exon 12 was used to screen for ASXL1 mutations. Samples showing altered GeneScan profiles were amplified in a second PCR reaction, and the amplicons were sequenced to confirm the mutation and to determine the mutation type. Patients were also assessed for the presence of NPM1, FLT3 (ITD and TKD), CEBPA, IDH1/2, RUNX1, and DNMT3A mutations by standard PCR-based methods. Results: ASXL1 mutations were detected in 90 (5.9%) of 1429 patients. All mutations were heterozygous frameshifts predicted to cause loss of the PHD finger. The most common mutation was a duplication of guanine at position 1934 (c.1934dup) identified in 59% (53/90) of the mutated cases. Three other ASXL1 mutations were detected in more than one patient: c.1900_1922del (n=16), c.1934del (n=5), c.1960dup (n=3). The majority of mutations (91%) clustered within or around a glycine-rich protein domain spanning amino acids 642–685. Patients with ASXL1 mutations were older (P 〈 .0001), more frequently males (P=.03), and they more frequently had secondary AML (P=.02). They also showed lower values for WBC (P=.01), bone marrow (P=.0002) and circulating (P=.001) blasts, and LDH serum levels (P=.007). ASXL1 mutations were more frequent in patients exhibiting intermediate-risk cytogenetics (P=.06). Among the ASXL1 mutated cases, 42% were cytogenetically normal (CN), 23% had other intermediate-risk cytogenetics, 26% had high-risk, and 9% low-risk [t(8;21) n=7, t(15;17) n=1, no inv(16)/t(16;16)] cytogenetics. ASXL1 mutations were associated with RUNX1 (P 〈 .001) and IDH2R140 mutations (P=.006). In contrast, ASXL1 mutations were less frequent in patients with NPM1 (P 〈 .001), FLT3-ITD (P 〈 .001), and DNMT3A (P=.02) mutations. The median follow-up for survival was 4.8 years. Patients with an ASXL1 mutation had an inferior complete remission (CR) rate compared with ASXL1 wildtype patients (57% vs 74%; P 〈 .001); the same was true for the subset of CN-AML (58% vs 77%; P=.04). In both, the entire cohort (P=.04) and in CN-AML (P=.07) ASXL1 mutations were associated with a worse event-free-survival, whereas no impact of these mutations was observed on relapse-free-survival. Patients with ASXL1 mutation had a shorter overall survival (OS) compared to those with ASXL1 wildtype (P=.002; 4-year OS rates, 32% vs 47%). The adverse impact of ASXL1 mutations on OS was also present in the subgroup of CN-AML (P=.009; 4-year OS rates, 33% vs 48%). In multivariable analysis, ASXL1 mutation was in trend an unfavorable factor for OS in CN-AML (hazard ratio: 1.46; P=.087). Conclusions: ASXL1 mutations were identified in ∼6% of younger AML patients and were associated with intermediate-risk cytogenetics. ASXL1 mutations frequently occurred with RUNX1 and IDH2R140 mutations, and were less frequent in NPM1 and FLT3-ITD mutated AML. ASXL1 mutations were associated with lower CR rate and inferior OS. The exact role of ASXL1 in normal hematopoiesis still needs to be defined in functional studies as well as the potential role of mutated ASXL1 protein in mediating resistance to chemotherapy. Disclosures: Kündgen: Celgene: Honoraria; Novartis: Honoraria.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1747-1747
    Abstract: Abstract 1747 Poster Board I-773 Introduction Patients with MDS, especially those with a lower-risk type of disease, are prone to develop iron overload, partly due to increased duodenal iron resorption triggered by ineffective erythropoiesis, but mainly as a consequence of chronic transfusion therapy. Transfusion dependency is clearly associated with a decreased likelyhood of survival. It is less clear how far that association is attributable to the severity of the underlying bone marrow disease as opposed to toxic effects of iron overload. A large retrospective study from Spain recently showed that iron overload (serum ferritin 〉 1000 ng/μl) was a significant prognostic factor for overall survival and leukemia-free survival. On multivariate analysis, the impact of iron overload was independent of transfusion requirement. If iron overload itself has an adverse effect on survival, iron chelation (IC) should do the opposite. This was indeed suggested by a partly retrospective and partly prospective observational study from France which showed a strong survival benefit from chelation therapy in patients with lower-risk MDS. We gathered that short of a prospective phase III trial, a retrospective matched-pair analysis might provide the best available data to identify a survival effect of iron chelation therapy in MDS. Methods Our matched-pair analysis included 93 patients with various types of MDS undergoing long-term chelation therapy, for whom we were able to identify 93 matched partners in the Düsseldorf MDS Registry (n=3552) who received supportive care only (SC, excluding iron chelation). Pairs were matched according to age at diagnosis, gender, MDS type according to WHO classification, and IPSS score. All 186 patients had iron overload, defined as serum ferritin of at least 500ng μl. Patients were followed up until death or until June 30, 2009. Results Median age was 63 yrs in the iron chelation group and 67 yrs in the supportive care group (p=n.s.). In both groups, the distribution among WHO types was as follows: 4 RA, 7 RARS, 41 RCMD, 22 MDS with del(5q), 8 RAEB I, 7 RAEB-II, 4 CMML-I. Both groups showed the following IPSS risk profile distribution: 37% low risk, 46% intermediate-I, 14% intermediate-II, and 3% high risk. Median ferritin level in the chlelated group was 1954 (498-7580), median ferritin level in the non-chelated group was 945 (508-5800). In the IC group, patients received the following chelators: deferoxamine (n=54), deferiprone (n=5), deferasirox (n=32), deferoxamine or followed by deferasirox (n=12), and deferoxamine plus deferiprone (n=4). The mean duration of the iron chelation was 28 months for Deferasirox and 39 months for Deferoxamine. Among patients receiving iron chelation therapy, 52% died in the observation period, as compared with 58% in the SC group. Median survival time in the IC group was 74 months vs. 49 months in the SC group (p=0.002). There was no significant difference regarding the risk of evolution into acute myeloid leukemia (AML). The cumulative risk of AML transformation between the IC group and the SC group was 10% vs. 12% two years after diagnosis, and 19% vs. 18% five years after diagnosis (p=0.73). Conclusions Our data support the idea that iron chelation therapy results in improved overall survival of MDS patients, mainly by reducing the risk of non-leukemic death. Since this was a retrospective analysis of registry data, we cannot determine exactly which clinical complications (e.g. cardiac events or infections) were diminished. This goal must be achieved by a well-designed prospective clinical trial. Our results do not seem to confirm that iron chelation therapy decreases the risk of leukemic transformation. However, such an effect may require elimination of labile plasma iron in order to suppress oxidative stress. LPI levels were not available in this retrospective matched-pair analysis but should be included in prospective studies. Disclosures Fox: Novartis: Consultancy, Honoraria, Participation in advisory boards in desferasirox trials, Speakers Bureau. Germing:Novartis: Honoraria, Research Funding, Speakers Bureau. Gattermann:Novartis: Honoraria, Participation in Advisory Boards on deferasirox clinical trials.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 598-598
    Abstract: Abstract 598 Thrombocytopenia is prevalent in up to 65% of patients with myelodysplastic syndrome (MDS) at the time of diagnosis and thrombocytopenic hemorrhage is a significant clinical problem that is often complicated by platelet aggregation defects. Little is known about the pathophysiology of this insufficient platelet function. Here, we delineate a reduced expression of critical platelet aggregation-related proteins by analyzing the platelet proteome of 7 patients with MDS and 7 normal donors. Patients' median platelet count was 60 × 10E9/L (range 37–109 × 109/L) and none of the patients examined had received prior anticoagulant treatment, chemotherapy or platelet transfusions. Differential 2-dimensional in-gel electrophoresis coupled with a time-of-flight Ultraflex-Tof/Tof mass spectrometer enabled the discovery of 120 differential protein spots. Among these, we identified a total of 35 proteins including 26 proteins that are integral part of the integrin aIIbβ3 receptor (GPIIb/IIIa, Fibrinogen receptor) signaling such as Talin-1 and Vinculin. In resting platelets the integrin aIIbβ3 receptor exhibits a low-affinity (inactive) state which is shifted to a high-affinity (active) state following inside-out activation. Talin-1 expression has been shown to be essential for this inside-out activation of the integrin aIIbβ3 receptor and consecutive platelet aggregation in an in-vivo model. We hypothesized that the reduced expression of Talin-1 and its co-factor Vinculin may inhibit activation of the integrin aIIbβ3 receptor and thereby contribute to the platelet aggregation defect seen in patients with MDS. Therefore, we performed further functional studies on integrin aIIbβ3 receptor activation and platelet spreading/aggregation with platelets derived from an independent cohort of 7 patients with MDS and 7 normal donors. In this new cohort, patients' median platelet count was 94 × 109/L (range 60–120 × 109/L) and again all patients had never received prior platelet transfusions or anti-coagulant treatment. When we looked at the surface expression of the integrin aIIbβ3 receptor on resting platelets by means of flow-cytometry, we did not detect any differences between platelets from patients with MDS and normal donors. Then, we activated platelets from normal donors and patients with MDS with 0.01U/μl and 0.001U/μl thrombin and measured binding of PAC-1, which is highly specific for detection of the active form of the integrin aIIbβ3 receptor. Here, we found a significantly lower shift from the inactive to the active form in platelets derived from patients with MDS dropping from 92.15% and 91.46% in normal donors to 41.97% and 48.45% (p = 0.01 and p = 0.006), respectively. We confirmed this suggested lack of adhesion and aggregation capacities in MDS platelets by confocal microscopy and single platelet imaging of washed platelets stimulated with 0.01U/μl thrombin which were adhered to immobilized fibrinogen. Consecutive platelet aggregation assays also revealed an insufficient response to stimuli like Collagen and ADP. Our findings provide for the first-time insight into the molecular pathology of defective platelet aggregation in MDS and suggest a mechanism of defective inside-out signaling caused by a reduced expression of proteins required for integrin aIIbβ3 receptor activation. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 110, No. 13 ( 2007-12-15), p. 4385-4395
    Abstract: We have generated a large, unique database that includes morphologic, clinical, cytogenetic, and follow-up data from 2124 patients with myelodysplastic syndromes (MDSs) at 4 institutions in Austria and 4 in Germany. Cytogenetic analyses were successfully performed in 2072 (97.6%) patients, revealing clonal abnormalities in 1084 (52.3%) patients. Numeric and structural chromosomal abnormalities were documented for each patient and subdivided further according to the number of additional abnormalities. Thus, 684 different cytogenetic categories were identified. The impact of the karyotype on the natural course of the disease was studied in 1286 patients treated with supportive care only. Median survival was 53.4 months for patients with normal karyotypes (n = 612) and 8.7 months for those with complex anomalies (n = 166). A total of 13 rare abnormalities were identified with good (+1/+1q, t(1q), t(7q), del(9q), del(12p), chromosome 15 anomalies, t(17q), monosomy 21, trisomy 21, and −X), intermediate (del(11q), chromosome 19 anomalies), or poor (t(5q)) prognostic impact, respectively. The prognostic relevance of additional abnormalities varied considerably depending on the chromosomes affected. For all World Health Organization (WHO) and French-American-British (FAB) classification system subtypes, the karyotype provided additional prognostic information. Our analyses offer new insights into the prognostic significance of rare chromosomal abnormalities and specific karyotypic combinations in MDS.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3363-3363
    Abstract: Abstract 3363 Poster Board III-251 The introduction of reduced-intensity conditioning regimens and the reduction of therapy-related complications have increased the upper age limit of the recipient and encouraging results of allogeneic stem cell transplantation in MDS-patients have been reported up to the age of 70 years. However, whether elderly patients with MDS should undergo allogeneic stem cell transplantation is a matter of debate. We used a multi-state approach to compare the outcome in elderly advanced MDS patients (aged 55 – 69 years) with RAEB or RAEB-t who received only best supportive care and were reported to the Düsseldorf registry (n = 137), to those who received allogeneic stem cell transplantation and were reported to the European Group for Blood and Marrow Transplantation (EBMT) (n = 246). A simple direct comparison is biased by non-observable patients who die before transplant. We used left-truncation and modelling of transition probabilities to obtain estimates of survival after diagnosis; moreover scenario analyses were performed to quantify sensitivity for untestable assumptions and to estimate survival for various strategies depending on the time between diagnosis and transplant. In the non-transplant cohort, diagnosis was RAEB in 100 (73%) and RAEB-t in 37 patients (27%). The median age of the 83 male and 54 female patients was 62 years. Cytogenetic data were available in 79 patients and reported to be abnormal in 60 %. In the transplant cohort 168 were male and 78 female. Disease status at diagnosis was RAEB (70%) and RAEB-t (30%). At time of transplantation, diagnosis was RAEB (54%), RAEB-t (34%) and transformed secondary acute leukemia (12%). Transplantation from HLA-identical sibling (n = 175) or unrelated donor (n = 71) was performed after standard myeloablative (n = 76) or reduced-intensity conditioning (n = 170). Cytogenetic data were available in 88 patients and reported to be abnormal in 60 %. The median follow-up of the non-transplant cohort was 21 and of the transplant cohort measured from date of transplant was 64 months. Median time between date of diagnosis and date of transplant was 7 months. The cumulative incidence of non-relapse mortality of the transplant cohort at 1 year and 3 years was 15% and 31%, respectively. The cumulative incidence of relapse was 14% and 29% at the same time points. Univariately, after accounting for left truncation and modelling of the death rate before transplant, the hazard ratio for survival is estimated as 0.72 in favour of transplant (95 % CI: 0.53 – 0.99; p = 0.04). After adjustment for year of diagnosis, the hazard ratio remained unchanged (the year of diagnosis was neither a confounder nor it showed interaction). In a multivariate analysis, adjusting for age at diagnosis, sub-diagnosis (RAEB vs. RAEB-t), cytogenetics (abnormal vs. normal), the hazard ratio for transplantation versus no-transplantation: 0.77 (95 % CI: 0.44 – 0.99; p = 0.1) while the other hazard ratio's were: 1.4 for “age 〉 60” (95 % CI: 1.10 – 1.70; p = 0.02); 1.20 for “RAEB-t” (95 % CI: 0.92 – 1.48; p = 0.2) and 1.46 for “abnormal cytogenetics” (95 % CI: 1.16 – 1.76; p = 0.01). Despite of an estimated benefit of allogeneic SCT in comparison to best supportive care in elderly MDS patients with RAEB or RAEB-t, this conclusion evidently relies on several statistical assumptions, notably about the selection mechanism among patients scheduled for transplant but never reaching that treatment. Only a prospective trial comparing hypomethylating agents with a reduced-intensity transplant approach according to donor availability is capable of answering this question appropriately but our analysis may provide sufficient indication that such a randomization is both needed and ethically justified. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 945-945
    Abstract: Abstract 945 In 2001, the WHO defined the category MDS with del(5q) due to unique cytogenetic, morphologic, hematologic, clinical, prognostic and therapeutic features. The survival of these patients, as well as patients with refractory cytopenia with unilineage dysplasia (RCUD) and refractory anemia with ring sideroblasts is favorable in comparison to other MDS types. Data on disease progression to a more advanced MDS category or to acute leukemia (AML) are sparse and have not been examined in detail. In order to address this issue we collated data of all patients with MDS and del(5q) characterized by low or intermediate-1 IPSS risk score that had been included into various collaborating MDS registries. Patients were followed from diagnosis and data on cell counts, transfusion dependency, and MDS progression were documented. No patients received treatment other than best supportive care. The status of 62 patients was censored at the time of the initiation of Lenalidomide therapy. AML progression was defined as 〉 20% marrow blasts. Estimates of survival probability were calculated with the Kaplan-Meier method. The cumulative incidence of progression to AML was calculated both with the Kaplan-Meier method and with the competing risk method where “death without progression to AML” is considered as competing event, not as censoring. For both events the cumulative incidences are estimated simultaneously. This method has the advantage that it takes into account that there is a difference between end of follow-up and death. Depending on the number of competing events, the curves are lower than those calculated with the Kaplan-Meier estimator. We identified 303 patients, median age at diagnosis 65 years (28-91), 71% were females. Median follow up time was 3 years. Median survival was 71.5 months. Patients with del(5q) as a sole chromosomal aberration had a median survival of 73 months as compared to 19.3 months in patients with more than 1 additional aberrations. Patients who had red cell transfusion need at diagnosis had a median survival of 39 months vs. 97 months in transfusion independent patients (p=0.00005). Transfusion need at diagnosis was the most important parameter for survival. Patients in the WPSS very low risk group had a median survival of 107 months, as compared to 73 and 56 months in the low and intermediate risk group and 37 months in the high risk group. 44 of the 303 patients (15%) progressed to AML ( 〉 20% marrow blasts). The cumulative AML progression rate calculated with the Kaplan-Meier method was 7% at 2 years and 18.2% at 5 years. The cumulative risk of AML progression calculated with the competing risk method was 6.6% at 2 years and 15.1% at 5 years. Factors associated with the risk of AML transformation were intermediate-I IPSS risk and high risk WPSS score, marrow blast count 〉 5%, and red-cell transfusion need at diagnosis. Survival and progression rates did not differ among the participating centers. In conclusion, survival of patients with MDS and del(5q) is high and is comparable to patients with RCUD and RARS, but is associated with a risk of AML-transformation similar to RCMD without del(5q). Further cytogenetic and molecular studies are warranted in order to identify patients at greater risk of progression. Disclosures: Germing: Novartis, Celgene: Honoraria, Research Funding. Lauseker:Celgene: Research Funding. Hildebrandt:Celgene: Research Funding. Symeonidis:Celgene: Research Funding. Cermak:Celgene: Research Funding. Pfeilstöcker:Celgene: Research Funding. Nösslinger:Celgene: Research Funding. Sekeres:Celgene: Research Funding. Maciejewski:Celgene: Research Funding. Haase:Celgene: Research Funding. Schanz:Celgene: Research Funding. Seymour:Celgene: Research Funding. Weide:Celgene: Research Funding. Lübbert:Celgene: Research Funding. Platzbecker:Celgene: Research Funding. Valent:Celgene: Research Funding. Götze:Celgene: Research Funding. Stauder:Celgene: Research Funding. Blum:Celgene: Research Funding. Kreuzer:Celgene: Research Funding. Schlenk:Celgene: Research Funding. Aul:Celgene: Research Funding. Kündgen:Celgene: Research Funding. Hasford:Celgene: Research Funding. Giagounidis:Celgene: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 4926-4926
    Abstract: Abstract 4926 Introduction: Chromosomal banding analysis (CBA) of bone marrow metaphases is the gold standard to identify chromosomal abnormalities in myelodysplastic syndromes (MDS). We aim to comprehensively detect and follow chromosomal abnormalities during the course of the disease without the need of repeated bone marrow biopsies. In ongoing studies we attempt to achieve this goal by performing serial fluorescence in situ hybridization (FISH) analysis on CD34+ peripheral blood cells (PBC). The aim of this pilot study was to establish SNP-array-analysis (SNP-A) on CD34+ PBC to complement genetic analysis on peripheral blood by identifying chromosomal abnormalities not detectable by FISH and/or CBA. Methods: We immunomagnetically enriched CD34+ PBC of 20 patients (pts) with MDS (16 pts), suspected MDS (1 pts) and secondary acute myeloid leukemia (sAML, 3 pts). SNP-A was performed with arrays from Affymetrix (3x SNP 6. 0, 4x Cyto 2. 7, 13x CytoScanHD). Fresh or frozen CD34+ PBC of 10 pts and in methanol/acetic acid fixed CD34+ PBC of 9 patients were successfully processed. One whole genome amplified sample was included. CBA and FISH-A was done for all patients. Results: By CBA, 3 pts had no chromosomal abnormalities, 8 pts had one abnormality, 6 pts had 2–4 abnormalities and 3 pts had more than 6 abnormalities. By SNP-A on CD34+ PBC, additional abnormalities could be revealed in 13/20 pts. In two pts they were also confirmed by FISH-A. Most of them were micro-deletions not detectable by CBA. In addition, SNP-A revealed uniparental disomies (UPD) in 5/20 pts. Of the 3 pts with no detectable abnormalities in CBA, one had a micro-deletion in 4q24 (TET2). The other two had an insufficient number of metaphases. One of them showed a highly complex karyotype by FISH-A and SNP-A on CD34+ PBC. The other one had suspected MDS and did not show any abnormalities by SNP-A. The 17 pts with ≤ 6 abnormalities in CBA showed 55 abnormalities by CBA, FISH-A and SNP-A altogether. 34/55 (62%) abnormalities could be detected by SNP-A and/or FISH-A, but not by CBA. 24/55 (44%) abnormalities could only be detected by SNP-A. 4/55 (7%) of abnormalities were structural abnormalities or small clones and were only detected by CBA. Serial analysis indicated clonal evolution: A patient with 16 abnormalities detectable by CBA and additional three by FISH and SNP-A developed two further micro-deletions (del(2)(q31q32), del(4)(q24q26)) within four months. When a MDS patient with a known 20q-deletion (isolated by CBA and FISH) progressed to AML 25 months after first diagnosis we detected 3 micro-deletions by SNP-A of peripheral blood (0. 98 Mb on 4q, 1. 31 Mb on 12q, 2. 55 Mb on 12q) thus resulting in 4 cytogenetic alterations fulfilling the criteria of complex and prognostically unfavorable abnormalities. Conclusions: Recently it was shown that abnormalities detectable by SNP-A, but not by CBA, could worsen prognosis of MDS patients. We succeeded in detecting these additional abnormalities without the need of bone marrow biopsies out of peripheral blood. Nevertheless, by parallel FISH and SNP-A of CD34+ PBC, most abnormalities detectable by CBA of bone marrow metaphases could be detected. Comprehensive genetic analysis at close intervals thus is possible without the need of bone marrow biopsies to study clonal evolution. The information gained could be used for therapy decisions, to improve prognostication and to unravel genetic evolutionary steps towards acute leukemia. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 412-412
    Abstract: Abstract 412 Background: A large proportion of patients are currently not eligible for genotype-adapted strategies in acute myeloid leukemia (AML), in particular those lacking specific genetic aberrations such as PML-RARA, CBFB-MYH11, RUNX1-RUNX1T1, NPM1 or activating FLT3 mutations. This subgroup of patients accounts for about one-third of all AML patients and mainly includes the large group of AML with myelodysplasia-related changes, AML with recurrent cytogenetic abnormalities [inv(3) or t(3;3), t(9;11), t(v;11q23)] and cytogenetically normal AML (CN-AML) with wild-type NPM1 and FLT3. Prognosis in this subgroup of patients is generally poor. Azacitidine has been shown to be active in AML with low blast counts frequently observed in AML with myelodysplasia-related changes and in CN-AML in the absence of specific gene mutations. Aims: To evaluate clinical efficacy of azacitidine in combination with intensive induction chemotherapy and in maintenance for two years as single agent in patients with AML who are not candidates for genotype-adapted treatment approaches. Methods: Patients with AML in the absence of specific genetic aberrations (PML-RARA, CBFB-MYH11, RUNX1-RUNX1T1, NPM1 mutation, activating FLT3 mutations) who are fit for intensive chemotherapy were eligible. Patients were up-front randomized for induction therapy into one standard arm and three experimental arms; i) ICE (standard arm), idarubicin (12 mg/m2/day, iv, days 1,3,5), cytarabine (100 mg/m2/day, cont. infusion, days 1–7), etoposide (100 mg/m2/day, iv, days 1,2,3); ii) AZA-prior, azacitidine (100 mg/m2/day, sc, days 1–5), idarubicin (12 mg/m2/day, iv, days 6, 8, 10), etoposide 100 mg/m2/day, iv, days 6,7,8); iii) AZA-concurrent, azacitidine (100 mg/m2/day, sc, days 1–5), idarubicin (12 mg/m2/day, iv, days 1,3,5), etoposide 100 mg/m2/day, iv, days 1,2,3); iv) AZA-after, idarubicin (12 mg/m2/day, iv, days 1,3,5), etoposide 100 mg/m2/day, iv, days 1,2,3), azacitidine (100 mg/m2/day, sc, days 4–8). After two induction cycles for patients achieving complete remission (CR), consolidation therapy was prioritized; first priority) allogeneic hematopoietic blood stem cell transplantation (HSCT) from matched related as well as unrelated donors, second priority) 3 courses of high-dose cytarabine followed by two-year maintenance therapy with azacitidine as single agent (50 mg/m2/day, sc, days 1–5, every 4 weeks) in patients initially randomized to experimental treatment. The primary endpoint was achievement of CR. The statistical design of the study was based on the Simon's optimal two-stage design applied for each arm separately. The null hypothesis was CR-rate equal or below 0.40 whereas the alternative hypothesis was a CR rate of at least 0.55 with a power of 80% and a level of significance of 5%. Thus, in each arm at least 12 of 26 patients with response to induction therapy were necessary after the first to proceed to the second stage. Results: During the first stage of the study 104 patients were randomized; median age was 62.5 years (range 18–82), 46% were female. Data on cytogenetics showed intermediate risk karyotype in 67% (n=50) including CN-AML (n=31) and high-risk karyotype in 33% (n=25). The most frequent serious adverse events were grade 3/4 infection with an overall incidence of 25% and ranging from 20 to 34% in the different treatment arms. The number of responding patients in the treatment arms AZA-prior and AZA-concurrent after the first stage of the study were 11 of 26 (42%) and 10 of 26 (38%)Both arms, AZA-prior and were terminated accordingly. In contrast, the treatment arms ICE and AZA-after were carried forward to the second stage of patient recruitment since responding patients at that time were 14 of 26 (54%) in both arms. In total, 100 patients each have been enrolled in both treatment arms, ICE and AZA-after, with CR-rates of 59% and 52%, respectively (p=0.39). To date, 60 patients received an allogeneic HSCT (n=36 matched unrelated donors, n=23 matched related donors, n=1 haploidentical family donor). Maintenance treatment was started in 12 patients. Conclusion: Induction therapy with ICE or idarubicin, etoposide followed by azacitidine (AZA-after) appears equally effective in producing CR in patients with AML who are not candidates for genotype-adapted treatment approaches. An amendment perpetuating the treatment arms ICE and AZA-after within a phase-III concept is planned. Disclosures: Schlenk: Celgene: Research Funding. Off Label Use: Azacitidine combined with intensive chemotherapy.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1693-1693
    Abstract: Abstract 1693 Background: The WHO 2008 classification of hematological malignancies defines distinct acute myeloid leukemia (AML) entities based on cytogenetic and molecular characteristics. However, immunophenotypotyping and morphology are still essential for rapid and correct diagnosis and for guidance of initial treatment, which in the future will be more and more genotype-specific. Although associations between phenotype and genotype have been described, correlations were evaluated mostly in retrospective studies not considering more recently identified gene mutations. Aims: To identify correlations between genotype and phenotype in adult AML. Methods: Immunophenotyping, cytogenetics and molecular analyses were performed centrally within the diagnostic screening procedure of a prospective multicenter phase III treatment trial for patients with AML aged 18 to 59 years. A total of 270 samples were analysed. The antibody panel for immunophenotyping was based on the recommendations of the German network of competence chronic and acute leukemias and the WHO-2008 classification. Results: AML with NPM1 mutation (NPM1mut) were characterized by higher expression of membrane-bound (m) CD33 (98% vs. 83%, p 〈 0.0001), mCD11b (90% vs. 36%, p 〈 0.0001) and mCD184 (90% vs. 37%, p 〈 0.0001), whereas mCD34 (14% vs. 82%, p 〈 0.0001), cytoplasmatic (c) CD34 (25% vs. 85%, p 〈 0.0001), mHLA-DR (70% vs. 85%, p=0.001), mCD117 (66% vs. 86%, p 〈 0.0001), and mCD133 (8% vs. 40%, p 〈 0.0001) were significantly lower expressed compared with NPMwt AML. In AML with NPM1mut, the presence of an activating FLT3 mutation was associated with higher mCD34 (22% vs. 3% p=0.014), cCD34 (33% vs. 13% p=0.03) and lower mCD61 (11% vs. 31% p=0.03) and mCD42b (0% vs. 14% p=0.04) expression. AML carrying a CEBPA double-allelic mutation (double-CEBPAmut) was characterized by higher expression of mCD34 (100% vs. 56%, p 〈 0.0001), cCD34 (100% vs. 62%, p=0.001), mCD117 (100% vs. 79%, p=0.003) and of the lymphoid-associated antigen mCD7 (93% vs. 27% p 〈 0.0001) compared to single-CEBPAmut and CEBPAwt. This phenotype was strongly related to double-CEBPAmut (chi square test p 〈 0.0001) but not to AML with single-CEBPAmut. AML with t(8;21) presented with a characteristic phenotype consisting of high cCD34 (100% vs. 62%, p=0.0009), mCD34 (100% vs.56% p=0.0001), mHLA-DR (100% vs.63% p=0.02), mCD117 (100 vs. 78%, p=0.03), and lymphoid-associated antigens mCD19 (76% vs. 2% p 〈 0.00001) and mCD56 (64% vs.11% p 〈 0.00001) expression. AML with inv(16) or t(16;16) had higher expression of cMPO (100% vs. 88%, p=0.02), mCD34 (90% vs 60%, p=0.0002), cCD34 (86% vs. 65%, p=0.01) and mHLA-DR (96% vs. 81%, p=0.01), whereas lymphoid antigens and platelet-associated antigens were significantly lower expressed (mCD56, 0% vs. 17%, p=0.01; mCD7 3% vs. 35%, p=0.0002; mCD61, 0% vs. 20%, p=0.003; mCD41, 9% vs. 38%, p=0.008; mCD42b, 0% vs. 9%, p=0.04). The associations of specific phenotypes to genotypes were as follows: i) cMPOpos/mCD19pos/cCD34pos/mCD34pos/mCD117pos/mHLA-DRpos to t(8;21) (p 〈 0.0001; sensitivity, 76%; specificity, 99%); ii) cMPOpos/mCD7pos/mCD34pos/cCD34pos/mCD117pos to double-CEBPAmut (p 〈 0.0001; sensitivity, 94%; specificity, 90%); iii) cMPOpos/mCD33pos/mCD34neg/mHLA-DRneg to NPM1mut or acute promyelocytic leukemia exhibiting a t(15;17) (p 〈 0.0001; sensitivity, 20%; specificity, 98%); iv) cMPOpos/cCD34pos/mCD34pos/mHLA-DRpos/mCD56neg/mCD7neg/mCD61neg/mCD41neg to inv(16) (p 〈 0.0001; sensitivity, 75%; specificity, 91%). Conclusions: The present study shows strong associations between specific immunophenotype patterns and genotypes in AML, which might be useful for daily clinical practice and future genotype-specific treatment. Disclosures: Salih: Pfizer: Research Funding. Döhner:Pfizer: Research Funding. Schlenk:Celgene, Pfizer, Novartis, Amgen, Cephalon: 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages