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: Leukemia, Springer Science and Business Media LLC, Vol. 34, No. 3 ( 2020-03), p. 914-918
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2008023-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1836-1836
    Abstract: Introduction: Therapy using high-dose cytarabine (HiDAC) according to the CALGB scheme (3g/m2 bid. days 1,3,5) is recognized as a standard consolidation treatment for younger adult patients (pts) with AML. Pegfilgrastim (PF) has been shown to be effective in reducing the duration of neutropenia in treatment of solid tumors and it seems to be even more effective than Filgrastim in reducing the incidence of infection. Methods: The AMLSG 07-04 trial (NCT00151242) was initiated in September 2004 (age 18–60 yrs). Consolidation therapy for cytogenetic favorable- and intermediate-risk groups consists of 3 cycles of HiDAC (3g/m2 bid. days 1,3,5) with PF 6mg given at day 10 (1-3-5 schedule) or after amendment no. 2 of HiDAC (3g/m2 bid. days 1,2,3) with PF 6mg given at day 8 (1-2-3 schedule). As a control group, pts randomized from AMLSG into the German AML Intergroup protocol using the standard 1-3-5 schedule for consolidation therapy with allowed interventional application of G-CSF were used. Results: Data from 251 pts and a total of 584 cycles are available (137 pts and 324 cycles, 1-3-5 schedule; 78 pts and 185 cycles, 1-2-3 schedule; 36 pts and 75 cycles, German AML Intergroup standard arm). Data from all three consolidation cycles were pooled for the comparison between the AMLSG 07-04 1-3-5 schedule and the German AML Intergroup standard arm. The duration of leukopenia (LP) and neutropenia (NP) were significantly shorter in pts receiving PF within the AMLSG 07-04 trial compared to pts within the German AML Intergroup standard arm (intention-to-treat, p=0.08 and p=0.03; as-treated, p=0.01 and 0.008, respectively). This beneficial effect of PF on LP and NP increased with the number of cycles. This was paralleled by a lower incidence of infection ≥CTC grade 3 with 40% in the 1-3-5 schedule and 67% in the German AML Intergroup standard arm (p & lt;0.0001). The comparison of the 1-2-3 schedule with the 1-3-5 schedule within the AMLSG 07-04 protocol revealed significantly shorter LP and NP in favor for the 1-2-3 schedule (p=0.03 and p=0.004, respectively). In median, pts after the 1-3-5 and the 1-2-3 schedule achieved a leukocyte count above 1.0/μl and a neutrophil count above 0.5/μl at day 20 and day 22 as well as 16 and 17, respectively. In a single center experience using out patient platelet and red blood cell support, the median time of hospitalization for the 1-3-5 (n=32 cycles) and the 1-2-3 (n=25) schedule could be reduced to 7.5 and 5 days with incidences for readmission of 33% and 12.5%, respectively. Conclusion: The administration of PF after HiDAC-based consolidation therapy in AML significantly shortened the duration of leuko- and neutropenia, reduced the rate of severe infections, and reduced the period of hospitalization.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 2360-2360
    Abstract: Aberrations of the MLL gene on chromosome 11q23 occur in about 5–10% of adult patients with acute myeloid leukemia (AML). Most commonly, fusion of the MLL gene to the genes AF9 on chromosome 9 or AF6 on chromosome 6 are found resulting in the translocation t(9;11) and t(6;11), respectively. The remaining patients with aberrations of chromosome 11q23 constitute a heterogeneous group with numerous fusion partners. This genetic heterogeneity hampers risk stratification of these patients. Accordingly, AML patients with 11q23 aberrations have varyingly been stratified as intermediate or high risk patients by different study groups. To analyze the impact of different 11q23 aberrations on the prognosis of AML patients up to 60 years, a pooled data analysis of 5 consecutive studies for the treatment of adult AML patients was performed. All patients received double induction treatment with araC and an anthracycline followed by an intensive consolidation with either a high dose araC based chemotherapy regimen or an autologous or allogeneic stem cell transplantation. In total, 137 patients with 11q23 aberrations were identified by cytogenetics and/or molecular techniques. 51 patients (37%) had a t(9;11), 19 patients (14%) a t(6;11), 8 patients (6%) a t(11;19), 6 patients (4%) a t(11;17) and 5 patients (4%) a t(10;11). 48 patients (35%) hold other fusion partners or deletions of 11q23. For further evaluation, patients harbouring other 11q23 aberrations than t(9;11) and t(6;11) were grouped together. Median age of all patients was 39 years (range 16–60). Overall complete remission rate was 75% with no significant difference between the groups (82% for t(9;11), 74% for t(6;11) and 70% for others). Altogether, 9% of the patients had treatment failure and 8% died during induction. Relapse-free survival (RFS) and overall survival (OS) at 5 years for the entire group was 34% and 28%, respectively. RFS and OS of the t(6;11) group was 0% and 11% respectively and thus was significantly inferior to patients with t(9;11) (RFS: 48%, OS: 33%) and patients with other 11q23 aberrations (RFS: 32%, OS: 28%). Within the t(9;11) group there was no difference in RFS or OS between patients who had a t(9;11) as a sole aberration (n=35) and patients with additional aberrations (n=16). Moreover, in t(9;11) no difference in RFS was observed between treatment with high dose araC, autologous or allogeneic stem cell transplantation as late consolidation. In conclusion, these data demonstrate that prognosis of patients with 11q23 is heterogeneous. Patients with t(9;11) have a relatively good outcome independent of the consolidation therapy used. In contrast, the prognosis of patients with t(6;11) is extremely poor. Therefore, patients with t(6;11) should be regarded as high risk and alternative treatment strategies for this subgroup are required.
    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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 2353-2353
    Abstract: Internal tandem duplications (ITD) of the FLT3 gene have been identified as negative prognostic marker in patients with acute myeloid leukemia (AML) exhibiting a normal karyotype. To evaluate the impact of different postremission strategies, such as high-dose cytarabine based chemotherapy (chemo), autologous (auto-SCT) or allogeneic stem cell transplantation (allo-SCT), in patients with normal karyotype and FLT3-ITD, we initiated a pooled data analysis within the prospective treatment trials AML-2/95, AML-1/99, AMLHD93, and AMLHD98A. Methods: All patients (age 16–60 years) received two cycles of induction therapy with standard-dose cytarabine combined with etoposide and idarubicin. After a first consolidation therapy, patients were assigned to allo-SCT if an HLA-identical sibling donor was available in all four trials. In the AML-2/95 and AMLHD93 trials, all other patients were assigned to chemo, whereas in the AML-1/99 and AMLHD98A trials patients were randomized between chemo and auto-SCT. Patients were analyzed for the presence of FLT3-ITD by genomic DNA PCR and conventional gel electrophoresis. All statistical tests were stratified for the variable “study group”. Results: Between 1993 and 2004 872 patients exhibiting a normal karyotype were registered. To date, results of FLT3 mutation analysis are available for 620 patients, and 182 (29%) exhibited an ITD. Response to induction therapy was 72% and 79% in the FLT3-ITD positive group and the FLT3-ITD negative group, respectively. After a median follow-up of 41 months, relapse-free (RFS) and overall survival (OS) were 33% versus 51% (p & lt;0.0001) and 27% versus 46% (p & lt;0.0001) in the FLT3-ITD positive and the FLT3-ITD negative groups, respectively. Analysis based on the availability of an HLA-matched family donor revealed a significantly better RFS (p=0.007) of 60% for patients with a donor (n=26) versus 21% for patients without a donor (n=102). Conclusion: In AML patients with normal karyotype exhibiting a FLT3-ITD mutation who achieve CR after induction therapy, consolidation with allo-SCT seems to overcome the negative impact of FLT3-ITD on outcome.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 2352-2352
    Abstract: Recently, mutations in the CEBPA gene, encoding CCAAT/enhancer binding protein alpha have been identified in 10 to 15% of patients with acute myeloid leukemia (AML) exhibiting a normal karyotype and mutant CEBPA was shown to predict favorable outcome. However, the impact of different postremission strategies such as high-dose cytarabine based chemotherapy (chemo), autologous (auto-SCT) or allogeneic stem cell transplantation (allo-SCT) in subgroups defined by these molecular markers is under discussion. Therefore, we initiated a pooled data analysis on patients exhibiting a normal karyotype at diagnosis treated within the prospective treatment trials AML-2/95, AML-1/99, AMLHD93 and AMLHD98A. All patients (age 16–60 years) received two cycles of induction therapy with standard dose cytarabine combined with etoposide and idarubicin. After a first consolidation therapy, patients were assigned to an allo-SCT if an HLA-identical sibling donor was available in all four trials. In the AML-2/95 and AMLHD93 trials all other patients were assigned to chemo whereas in the AML-1/99 and AMLHD98A trials patients were randomised between auto-SCT and chemo. All patients were analyzed for CEBPA-mutations in the N-terminal transactivation domains and the C-terminal basic region-leucine zipper domain by direct sequencing of genomic DNA. Between 1993 and 2004 a total of n=872 patients exhibiting a normal karyotype had been registered. Actually in 413 patients results of CEBPA analyses are available and 54 (13%) exhibited a mutation (46% N-terminal, 15% C-terminal, 39% N- and C-terminal). Response to induction therapy was 85% (46/54) and 74% (259/352) in the CEBPA mutated group and the CEBPA wild-type group, respectively. RFS and OS were 59% and 38% (p=0.01) as well as 66% and 41% (p=0.002) in the CEBPA mutated group and the CEBPA wild-type group, respectively after a median follow-up of 42 months. The distribution of postremission therapy of the 46 patients in the CEBPA mutated group was n=22 Chemo, n=13 allo-tpl, n=9 auto-tpl, n=2 allo-tpl from a matched unrelated donor. Analysis based on the availability of a matched related donor revealed a significant better RFS (p=0.03) and OS (p=0.03) for patients with an matched related donor. Conclusion: Although AML patients with normal karyotype and a CEBPA mutation comprises a group with favorable prognosis allogeneic transplantation from an HLA-matched family donor seems to improve long term outcome. Confirmation of these preliminary data on a larger series of patients are necessary to improve a risk-adapted decision on postremission strategies.
    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. 116, No. 21 ( 2010-11-19), p. 4179-4179
    Abstract: Abstract 4179 In acute myeloid leukemia (AML), complex karyotype is defined as the presence of three or more chromosome abnormalities in the absence of one of the recurrent genetic abnormalities as defined by the recent WHO classification. AML with complex karyotype (CK-AML) account for approximately 10 to 15% of all cases and are associated with preceding myelodysplasia (MDS) or exposure to toxic agents; the prognosis of patients is very poor. So far, little is known about the molecular mechanisms underlying initiation or progression of CK-AML. To identify genomic regions of potential pathogenic relevance, we used microarray-based techniques [array-comparative genomic hybridization (CGH) and single-nucleotide polymorphism (SNP) analysis] for high-resolution genome-wide analysis in 242 cases, including 171 (71%) cases enrolled on clinical protocols using intensive chemotherapy. Among other genomic imbalances, we identified loss of chromosome band 17q11.2 encompassing the NF1 locus in 55 (23%) of the 242 cases. Interestingly, three of these cases exhibited homozygous loss of NF1. Based on these findings and the fact that NF1 is recurrently altered in myeloid malignancies, we further investigated its role in CK-AML. Therefore, we analyzed 11 cases with heterozygous microdeletions of NF1 for mutations in the remaining allele by direct sequencing of exons 1 to 60 and identified 5 mutations in 4 cases; all of these mutations resulted in a premature stop codon (3 frameshift mutations, 2 nonsense mutations); one frameshift mutation (c.2033dupC) was recurrent. Combining the findings from array-based and mutation analyses, we so far identified 7 patients with biallelic NF1 gene alterations, i.e. homozygous loss or loss of one allele and at least one mutation in the remaining allele. Since correlation of NF1 alteration with data from array-based genomic profiling revealed a significant correlation with loss of chromosome band 17p13 encompassing TP53 (P 〈 .001), we correlated NF1 alteration with the TP53 status (mutation and/or loss), which was available for all 242 cases, and found a positive correlation with both TP53 alteration (mutation and/or loss) and TP53 mutation (P 〈 .001 each). In addition, NF1 alteration was significantly correlated with biallelic TP53 alterations (loss and mutation or homozygous mutations) (P 〈 .001). We than further investigated the two genotypes NF1alteration/TP53alteration (n=50) and NF1no alteration/TP53alteration (n=109) with regard to their association with other genomic imbalances. The genotype NF1alteration/TP53alteration was significantly correlated to the total number of deletions (median 9 vs 7; P = .025), the genomic complexity as measured by the total number of aberrations per case (median 13 vs 11; P = .039), and the presence of 16q loss (50% [25/50] vs 29% [32/109], P = .014) when compared with the NF1no alteration/TP53alteration genotype. Notably, in a recently published murine model deficiency of ICSBP, located on 16q24, was shown to synergize with NF1 haplo-insufficiency in leukemogenesis. In conclusion, the NF1 gene is found to be recurrently altered in CK-AML. Being associated with specific genomic aberrations, NF1 alteration is likely cooperating in myeloid leukemogenesis or disease progression. One important co-player might be TP53 that has an important role in genomic stability. The exact mechanism of interaction between NF1 and TP53 or other concurrent genetic alterations have to be further investigated. Disclosures: Döhner: Pfizer: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1441-1441
    Abstract: Abstract 1441 Introduction: AML with translocations involving the mixed-lineage leukemia gene (MLL) on chromosome band 11q23 occur in 5–10% of adult cases. Outcome of patients with t(11q23) is generally poor, except for AMLs with t(9;11) that are associated with an intermediate prognosis. Clinical risk factors have been proposed, however, due to the rarity of these AML subtypes and to the great heterogeneity of MLL fusion partners, validation of such risk factors remains a challenge. In two previous studies (Lugthart S et al., Blood 2008; Gröschel S, Lugthart S et al., J Clin Oncol 2010) that included all AML subsets, it has been shown that high expression of the ecotropic viral integration 1 gene (EVI1+ ) is associated with t(11q23) AML; furthermore, EVI1+ appeared to be associated with inferior prognosis in this subset of AML. The aim of this study was to evaluate the prognostic impact of EVI1+ in a large cohort of AMLs with t(11q23) in the context of clinical features and cytogenetic and molecular genetic markers. Patients and Methods: In the AMLSG data base we identified 147 AMLs with t(11q23). In 109 cases material was available for EVI1 expression analysis. Patients were treated on seven different prospective treatment protocols of the German-Austrian AMLSG (HD93, HD98A, HD98B, 07–04, 06–04, SHG295, SHG0199). Results: The 147 MLL -rearranged AMLs were divided into three subgroups: t(9;11)(p22;q23), n=70 (47.6%); t(6;11)(q27;q23), n=26 (17.7%); and t(v; 11q23), n=51 (34.7%). Most common translocation partners within t(v; 11q23) were t(11;19)(q23;p13) (n=15, 29.4%) and t(10;11)(p13;q23) (n=6, 11.8%). Comparison of clinical characteristics between the three subgroups revealed significantly higher LDH levels (median 550 U/L, p=.049), higher rate of therapy-related AML (33.8%, p=.002), and higher incidence of trisomy 8 (25.7%, p=.05) in AML with t(9;11). Deregulated expression of EVI1 was found in 55/109 (50.5%) of all t(11q23) cases; AMLs with t(6;11) showed the highest frequency of EVI1+ (85.7%), followed by AMLs with t(9;11) (44.2%), and t(v; 11q23) (38.9%) (p=.001). Concurrent gene mutations were virtually absent in t(9;11) AMLs; only rare FLT3 -ITDs were identified (3/70 cases; 4.3%), whereas no sequence variations were found for all other genes analyzed (CEBPA, NPM1, RUNX1, IDH1/2, ASXL1, TET2, DNMT3A). There was no difference in achievement of complete remission (CR) between EVI1− and EVI1+ t(11q23) AMLs. In multivariable analysis of the entire t(11q23) cohort, EVI1+ was the only prognostic factor predicting for inferior overall survival (OS) (p=.017, HR 1.88, 95%-CI: 1.12–3.16) and relapse-free survival (RFS) (p=.013, HR 1.96, 95%-CI: 1.15–3.32). EVI1+ AMLs with t(11q23) in first CR had significantly better outcome (OS, p=.01; RFS, p=.002) after allogeneic stem-cell transplantation compared with other consolidation therapies; the 5-year OS rate of EVI1+ AML was 53 % and 0 % after allogeneic SCT and other consolidation therapies, respectively. We also separately analyzed the subset of t(9;11) AML: compared with EVI1− patients, EVI1+ patients had higher white blood cell counts (p=.045); additional presence of trisomy 8 was only found in EVI1- t(9;11) AMLs (11/29, 37.9%; p=.001). In multivariable analysis, EVI1+ again was the sole independent adverse prognostic factor for OS (p=.017, HR 2.55, 95%-CI: 1.18–5.51) and RFS (p=.02, HR 2.39, 95%-CI: 1.14–4.97). Conclusion: Deregulated expression of EVI1 is found in about half of t(11q23) AMLs. EVI1+ is the strongest prognostic factor in all t(11q23) AMLs and in the subset of t(9;11) AMLs. Patients with EVI1+MLL -rearranged AML appear to benefit from allogeneic SCT in first CR. Disclosures: Germing: Celgene: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2613-2613
    Abstract: Abstract 2613 Poster Board II-589 Cytogenetically normal acute myeloid leukemia (CN-AML) represents a biologically and clinically heterogeneous group. During recent years novel molecular markers like FLT3, CEBPA and NPM1 gene mutations as well as deregulated expression of single genes such as EVI1, MN1, ERG and BAALC have been identified that provide important prognostic information in CN-AML. Furthermore, DNA microarray-based gene expression profiling (GEP) has been shown to capture the molecular heterogeneity of leukemia and has been applied to build clinical outcome predictors in CN-AML. In this study, we wanted to assess whether GEP based outcome prediction using novel biostatistical approaches applied to large gene expression data sets could refine previous findings. First we profiled gene expression in a large set of clinically well annotated CN-AML (entered on a multicenter trial for patients 〈 60 years, AMLSG 07-04, n=154 cases) using Affymetrix Human Genome U133plus2.0 microarrays. Then, we applied L1-penalized Cox proportional hazards regression to develop a sparse prognostic model for overall survival. Using this algorithm we were able to define a gene expression signature correlated with outcome in CN-AML. Interestingly, our model resulted in a signature that only comprised one probe set corresponding to the HBG1 gene (hemoglobin, gamma A). While quantitative RT-PCR validated correct measurement of HBG1 expression (correlation r=0.96, n=15), we next evaluated our finding in independent cohorts of CN-AML cases. We applied our “HBG1 gene signature” to previously published CN-AML data (Metzeler et al. Blood 2008) comprising 163 patient samples on Affymetrix U133A/B (data set I) and 79 patient samples on Affymetrix U133plus2.0 microarrays (data set II). Univariate Cox PH regression showed that our gene expression predictor was highly significant for overall survival in both data sets (P=0.002, HR 1.71, 95%CI 1.23–2.39; and P 〈 0.001, HR 3.03, 95%CI 1.84–5.02, respectively). Adjusted for age, NPM1, and FLT3-ITD mutational status HBG1 retained its prognostic relevance in multivariate analysis (P=0.007 and P 〈 0.001, respectively). As HBG1 expression might be used as novel prognostic marker in AML, the role of HBG1 expression in AML remains to be determined. HBG1 expression might represent a surrogate marker indicating the activation of distinct oncoproteins such as MYB, which recently has been shown to transcriptionally repress the level of gamma-globulin. On the other hand HBG1 expression has been shown to be induced upon exposure to azacytidine. Thus, HBG1 expression could also reflect underlying epigenetic profiles of prognostic relevance. While studying HBG1 expression in larger CN-AML cohorts as well as in the context of other molecular markers might help to further determine the prognostic impact of HBG1, integrative analyses combining GEP with genomics and epigenomics data sets will provide novel insights into the mechanisms underlying HBG1 expression in CN-AML. 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2185-2185
    Abstract: Abstract 2185 Background: Secondary (s)AML from MDS is more frequent in older AML patients, and associated with an overall worse outcome with standard chemotherapy than de novo AML, particularly after MDS of longer duration (1). The azanucleoside hypomethylating agents 5-azacytidine (Vidaza) and 5-aza-2′-deoxycytidine (Decitabine, DAC) are active in MDS and, as recently shown, also AML. Compared to other predictors of response to these drugs, MDS duration prior to treatment thus far has received only limited attention, with two recent publications reporting conflicting results (2, 3). To independently validate our finding that shorter duration of MDS prior to DAC treatment may be a novel predictor of poor outcome (2, 4), we now applied this parameter to a large trial of low-dose DAC in AML pts (aged 〉 60 years and judged ineligible for standard induction chemotherapy), about half of them with sAML from MDS with variable disease duration. Patients and Methods: Comparisons of response rate (RR, i.e. CR or PR) and overall survival (OS) from start of treatment according to MDS duration (pre-specified categorization according to quartiles) were performed post-hoc in 109 patients (pts) with previously untreated sAML (median age 72 years) treated with DAC (given over 72 hours, every 6 weeks, for up to 4 courses, followed by “maintenance” with 3 daily 1-hour infusions of DAC 20 mg/m2 every 4–6-weeks). Median WBC prior to treatment was 5.200/μl, median serum LDH 279U/l, 31.2% of pts had adverse cytogenetics, 82.6% had a performance status 〉 1, and 80.7% had a comorbidity index (HCT-CI) 〉 =1. Comparisons by logistic regression and Cox regression (univariate and multivariate, adjusted for other prognostic factors showing an effect in this population of sAML pts) were performed. Results: Of the 227 AML patients treated within the 00331 trial, 109 (48%) had prior MDS with known MDS duration, with a median duration of 8 (25% quartile 3, 75% quartile 25, range 1–101) mths. The overall RR in these pts was 26/109 (24%), the overall 1 yr OS rate was 31% (94 deaths). A comparison of RR according to MDS duration revealed a trend to an increase in RR with longer duration of MDS [ 〈 3: 4/25 (16%), 3–8: 5/29 (17%), 8–25: 7/27 (26%), 〉 =25 mths: 10/28 (36%), test for heterogeneity p=0.29, test for trend p=0.06]. Similarly, when OS from start of DAC was analyzed according to this parameter, for pts with previous MDS of longer duration there was a trend to better outcome [ 〈 3: 1 yr OS rate 23%, 3–8: 28%, 8–25: 26%, 〉 =25 mths: 46%, test for heterogeneity p=0.17, test for trend p=0.16]. When these analyses were adjusted for other prognostic factors showing an effect in this population of sAML pts (comorbidity index, sLDH with respect to RR, and performance status, comorbidity index, and white blood count with respect to OS), the results were similar (effect of MDS duration with respect to RR: test for heterogeneity p=0.35, test for trend p=0.06, and effect of MDS duration with respect to OS: test for heterogeneity p=0.04, test for trend p=0.11). Conclusion: In this large cohort of uniformly treated pts with sAML, MDS of longer duration appeared to be associated with a better outcome, even after adjusting for important other prognostic factors. These results are supported by a similar analysis of MDS pts randomized in the 06011 EORTC intergroup trial (which compares DAC to Best Supportive Care), where MDS patients with longer ( 〉 =3 mths) disease duration prior to treatment also had better outcome (4). They warrant application of this discriminator in the evaluation also of other non-intensive AML treatment modalities. References 1. Estey et al., Blood 90:2969-77, 1997 2. Wijermans et al., Ann. Hematol. 84 Suppl 1:9-14, 2005 3. Kantarjian et al., Cancer 109:265-73, 2007 4. Lübbert, Suciu et al., Abstract submitted, ASH 2010 Disclosures: Off Label Use: decitabine is FDA-approved for treatment of MDS and AML with up to 30% blasts. In the present study, patients with AML and higher blast percentage were treated. Platzbecker: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Döhner: Pfizer: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 803-803
    Abstract: For many years, clinical management of Acute Myeloid Leukemia (AML) has relied on patient classification into molecular groups, mostly defined by fusion genes. Recent insights of AML genomes have uncovered extended heterogeneity implicating 〉 100 recurrently mutated genes, many of which are infrequently mutated. In each patient, multiple mutations are present defining unique genetic and clonal constellations. This genetic diversity significantly complicates the translation of molecular findings into routine clinical practice. We present our full analysis on the genomic characterization of 1540 AML patients enrolled in clinical trials of the German-Austrian AML Study Group. Together with cytogenetic profiling we map 5234 pathogenic lesions across 77 genomic loci. Amongst these, we characterise a cluster of hotspot mutations in the MYC oncogene. Overall we find ≥1 driver mutation in 96% patients, and ≥2 in 85%. The earliest mutations in AML evolution implicate genes mutated in age-related clonal hematopoiesis (DNMT3A, ASXL1, TET2) or fusion genes, followed by ordered acquisition of mutations in transcription, chromatin or splicing regulators. RTK/RAS mutations frequently represent late events with evidence of parallel evolution in 14% of AML. We formally model genomic structure and find that AML is subdivided in at least 11 molecular and clinically distinct classes defined by t(15;17), t(8;21), inv(16)/t(16;16), t(6;9), inv(3)/t(3;3), AML defined by MLL- rearrangements, CEBPAbi-allelic, NPM1, TP53/complex karyotype, AML with chromatin/splicing factor mutations, and provisionally AML with 〈 3 aneuploidies. ~87% of patients with acquired mutations are molecularly classified. Each class is defined by a distinct subset of genetic lesions, with evidence of preferred order in mutation acquisition, thus guiding future development of minimal residual disease and combination therapy protocols. 19% (n=291) of patients were classified in the chromatin/spliceosome class. In this group, mutations in splicing factor genes and/or RUNX1 cluster with mutations in chromatin modifiers (ASXL1, EZH2, STAG2, MLLPTD). Patients in this group mostly represented Intermediate risk AML (ELN recommendations), were older, with lower WBC/blasts, inferior response rates to induction chemotherapy, poor long-term clinical outlook, higher rates of secondary AML and MDS-related morphology. Compared to classes defined by fusion genes, classes defined by genes are considerably more complex. We explore whether variability of clinical response (complete remission, relapse, relapse related mortality and overall survival) is at least in part accounted for by the extended genomic landscape. We find that the recurrent secondary and tertiary genotypes (often implicating rare genes/mutation-hotspots) markedly redefine clinical response and long-term curability beyond those predicted by single classifier lesions. To this effect, we apply global statistical models to calculate the contributions of genomic variables to overall risk whilst taking into account demographic, diagnostic and treatment factors. We find that gene-by-gene interactions are associated with additive as well as epistatic effects to patients risk, and contribute ~10% of relapse related mortality risk. We build prognostication models tailored to individual patients molecular, demographic and clinical variables at time of diagnosis and deliver more accurate risk predictions. For example, on the basis of the composite genomic and clinical profiles subsets of patients categorized as Favorable/Intermediate risk AML show risk estimates associated with adverse prognosis. Such patients are evaluated for therapeutic protocol selection tailored to higher risk groups (transplant at first CR instead of relapse), and ascertained for overall survival benefit. We apply same approaches for high-risk patients associated with favorable profiles and collectively deliver a paradigm of personally tailored risk assessment coupled with appropriate selection of therapeutic intervention. Taken together comprehensive genome profiling shows that genetic heterogeneity in AML is not random. Characterization of the extended genetic framework beyond single classifier lesions, informs future strategies for personalized prognostication, minimal residual disease monitoring and combination therapy protocols. Disclosures Schlenk: Janssen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Honoraria, Research Funding; Boehringer-Ingelheim: Honoraria; Novartis: Honoraria, Research Funding; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Arog: Honoraria, Research Funding; Teva: Honoraria, Research Funding. Campbell:14M genomics: Other: Co-founder and consultant.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...