Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 56, No. 8 ( 2015-08-03), p. 2289-2295
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2015
    detail.hit.zdb_id: 2030637-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: European Journal of Haematology, Wiley, Vol. 100, No. 2 ( 2018-02), p. 154-162
    Abstract: Randomized comparison of two treatment strategies in frontline therapy of acute promyelocytic leukemia ( APL ): all‐trans retinoic acid ( ATRA ) and double induction intensified by high‐dose cytosine arabinoside ( HD ara‐C) (German AMLCG ) and therapy with ATRA and anthracyclines (Spanish PETHEMA , LPA 99). Patients and results Eighty of 87 adult patients with genetically confirmed APL of all risk groups were eligible. The outcome of both arms was similar: AMLCG vs PETHEMA : hematological complete remission 87% vs 83%, early death 13% vs 17% ( P  = .76), overall survival, event‐free survival, leukemia‐free survival, cumulative incidence of relapse at 6 years 75% vs 78% ( P  = .92); 75% vs 68% ( P  = .29); 86% vs 81% ( P  = .28); and 0% vs 12% ( P  = .04, no relapse vs four relapses), respectively. The median time to achieve molecular remission ( RT ‐ PCR negativity of PML ‐ RARA ) was 60 days in both arms ( P  = .12). The AMLCG regimen was associated with a longer duration of neutropenia ( P  = .02) and a higher rate of WHO grade ≥3 infections. Conclusions The small number of patients limits the reliability of conclusions. With these restrictions, the outcomes of both approaches were similar and show the limitations of ATRA and chemotherapy. The HD ara‐C–containing regimen was associated with a lower relapse rate in high‐risk APL .
    Type of Medium: Online Resource
    ISSN: 0902-4441 , 1600-0609
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2018
    detail.hit.zdb_id: 2027114-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Leukemia, Springer Science and Business Media LLC, Vol. 33, No. 5 ( 2019-5), p. 1124-1134
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2008023-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1382-1382
    Abstract: Introduction: Insulin-like growth factor binding proteins (IGFBP) are carriers of insulin-like growth factors (IGFs). They prolong their half-life and modulate their availability and activity. The IGF-signaling system has been shown to have an important role in various solid cancers and hematologic malignancies. High levels of IGFBP2 and IGFBP7 have been associated with chemoresistance, relapse and inferior survival in different leukemias. Acute promyelocytic leukemia (APL) is a distinct subtype of acute myeloid leukemia (AML), which has been reported to have increased expression of IGFBP2. However, there is no data on its impact on prognosis. In addition, no data exist on IGFBP7 in APL. The aim of this study was to elucidate the influence on prognosis of both candidate genes in APL patients. Methods: Expression levels of IGFBP2 und IGFBP7 were retrospectively analysed in bone marrow (BM) samples at the time of initial diagnosis from 69 APL patients (42 female, 27 male) after informed consent. Median age of patients was 46 years (range 19 to 82 years). All patients were diagnosed and treated in the German AML Cooperative Group (AMLCG) studies. Treatment consisted of simultaneous ATRA and double induction chemotherapy including high dose ara-C, one cycle of consolidation chemotherapy and 3 years monthly maintenance chemotherapy. In patients older than 60 years, the second induction cycle was at the discretion of the treating physician. Three patients (4%) received an induction of ATRA and an anthracycline without ara-C. BM samples of 22 healthy volunteers served as a control group. Multiplex reverse transcriptase quantitative real-time PCR (qRT-PCR) was performed on a LightCycler® 480 (Roche, Mannheim, Germany) PCR system. Glucose-6-phosphat isomerase was used as a housekeeping gene. For quantification of relative expression values a modified delta-delta CT calculation model according to Pfaffl was used after determination of PCR efficiencies. cDNA from the cell line K562 served as a calibrator in each run. All reactions were performed in triplicates. IGFBP2 expression groups were defined as follows: Patients with IGFBP2 expression below or equal the 25th percentile (IGFBP2low) were compared to patients with higher IGFBP2 expression (IGFBP2high). Overall survival (OS), relapse free survival (RFS) and the cumulative incidence of relapse (CIR) were calculated using the Kaplan-Meier method and a log-rank test was used to compare differences between the groups (p 〈 0.05). Results: Expression levels of IGFBP2 did not differ between APL patients and healthy controls. However, there was a significantly higher relapse rate in APL patients with low IGFBP2 expression (CIR: 25% in the IGFBP2low group vs. 5% in the IGFBP2high group; p=0.04). Accordingly, RFS of patients in the IGFBP2low group was also inferior (41% vs. 81% in the IGFBP2high group; p=0.0002; Fig. 1). The OS of patients who had responded to induction therapy was also influenced by IGFBP2 expression (OS of responders: 61% for IGFBP2low vs. 83% for IGFBP2high; p=0.02). However, there was no significant difference in the analysis of OS of all patients including patients who suffered early death. In contrast to these findings, IGFBP7 was expressed significantly lower in APL patients compared to healthy controls (p 〈 0.0001) but there was no association with outcome of APL patients. Conclusion: Of the two analysed IGFBP family members only IGFBP2 showed impact on the prognosis of APL patients. Remarkably, IGFBP2 was not overexpressed compared to healthy controls in our patient cohort. The reason might be that whole BM samples of healthy controls were used instead of subpopulations. Still, among the APL patients cohort its expression showed a strong influence on prognosis especially on relapse rate and RFS. To find low expression as a negative prognostic marker is surprising as for leukemias only high expression has been reported as a negative factor. However, IGFBP2 has been proposed to suppress tumor development through binding IGFs and preventing IGF-receptor driven tumorigenesis. This is supported by the fact that the IGFBP2 promotor is hypermethylated in various types of cancer. In summary, we identified low IGFBP2 expression as a novel prognostic marker for APL. Further investigations are warranted to clarify its possible role in leukemia. Figure 1. Figure 1. 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: 2015
    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. 122, No. 21 ( 2013-11-15), p. 1323-1323
    Abstract: With current therapy regimens over 75% of patients with de novo acute promyelocytic leukemia (APL) can be cured. Approaches to further improve patient outcome by stratifying patients at the time of initial diagnosis according to their individual risk and to adjust therapy accordingly have been based on clinical features only. Molecular markers have not been established for risk stratification as yet. Recently, we have shown that high expression levels of the genes brain and acute leukemia, cytoplasmic (BAALC) and ets related gene (ERG) are associated with inferior outcome in APL patients. In addition, data indicate that aberrant expression of the gene Wilms’ tumor 1 (WT1) is a negative prognostic factor with regard to overall survival (OS) after complete remission (CR) and relapse free survival (RFS) in APL. In this study we evaluated the prognostic relevance of a combined score integrating the expression levels of the above mentioned genes to further improve risk stratification in APL patients. Methods Expression levels of BAALC, ERG and WT1 of 62 patients with newly diagnosed APL were retrospectively analyzed in bone marrow mononuclear cells using multiplex reverse transcriptase quantitative real-time PCR (qRT-PCR). Median age of patients was 47 years (range: 19 to 82y). All patients gave informed consent. Patients were diagnosed and treated in the German AML Cooperative Group (AMLCG) study with a treatment of simultaneous ATRA and double induction chemotherapy including high-dose ara-C, consolidation and maintenance chemotherapy. The following gene expression levels were identified as negative risk factors in preceding studies: BAALC expression ≥25th percentile (BAALChigh), ERG expression 〉 75th percentile (ERGhigh) and WT1 expression ≤25th percentile or ≥75th percentile (WT1low/high). A risk score was developed as follows: for the presence of one of the mentioned risk factors one scoring point was assigned to a respective patient, i.e. a maximum of 3 points (one point for BAALChigh, ERGhigh and WT1low/high, respectively) and a minimum of 0 points (i.e. presenting with none of the aforementioned risk factors) could be allocated to one patient. Accordingly, patients were divided into four risk groups: 7 patients scored 0 points (= low risk), 27 patients scored 1 point (= intermediate 1 risk), 19 patients scored 2 points (= intermediate 2 risk) and 9 patients scored 3 points (= high risk). Subsequently, OS, RFS and relapse free interval (RFI) were calculated using the Kaplan-Meier method and a log-rank test was used to compare differences between the four risk groups (p 〈 0.05). Results The integrative risk score divided patients into four groups with significantly different outcome. The low risk group showed a RFS of 100% at 10 years of follow-up compared to the intermediate 1 risk group with 81%, the intermediate 2 risk group with 58% and the high risk group with a RFS of 42% only (median survival: 4.6y) (p=0.02). In accordance, the RFI differed significantly between the four groups: low risk 100%, intermediate 1 risk 100%, intermediate 2 risk 89% and high risk 71% (p=0.049). There was no statistically significant difference between the 4 groups with regard to OS in the entire patient cohort. However, there was a clear trend towards a difference in OS in patients who achieved a CR after induction therapy: low risk 100%, intermediate 1 risk 81%, intermediate 2 risk 68% and high risk 53% survival at 10 years of follow-up (p=0.09). Conclusion Integration of expression levels of the genes BAALC, ERG and WT1 into a scoring system identifies 4 risk groups with significantly different outcome with regard to RFS and RFI. It might be a promising approach to guide therapeutic decisions in patients with APL. However, multivariate analyses and validation of these data in an independent patient cohort is warranted. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 4008-4008
    Abstract: Depth of molecular remission on tyrosine kinase inhibitor (TKI) treatment is of rising importance for chronic myeloid leukemia (CML) patients (pts) with regard to possible treatment discontinuation and competing TKIs available to improve molecular response. At present, it is unknown which level of deep molecular response is necessary for optimal prognosis and for successfully stopping therapy. The aim of this work is both to evaluate the technical feasibility of molecular monitoring at the mentioned level and to search for factors allowing to predict MR5.0 in pts on imatinib (IM)-based treatment. Methods Real-time quantitative PCR on mRNA BCR-ABL transcripts in addition to total ABL transcripts as internal control has been performed on a LightCycler platform in 1,442 pts within the randomized CML-Study IV and adapted according to the International Scale (IS). In order to qualify for MR5.0 the BCR-ABLIS expression should meet one of the following criteria: a positive result ≤0.001% or a negative result with a minimum sample quality of 100,000 ABL copies (Cross et al., Leukemia 2012). Calculating cumulative incidences of remission or progression, the competing risks progression and/or death before possible progression were considered. Cox models were estimated for the multivariate analysis. Results In 1,198 of the 1,442 molecularly examined pts at least one sample fulfilled the sensitivity criteria for a MR5.0 (8,266 of 24,101 samples, 34.3%). Cumulative incidence of MR5.0 was 51% at 8 years. The median time to MR5.0 according to randomized treatment arms differed as follows: IM 800mg 79.7 months (mos), IM 400mg 95.0 mos, IM 400mg + IFNα 98.0 mos, IM 400mg + AraC 103.3 mos, IM 400mg after IFN failure 112.9 mos. A Cox model examining the different treatment arms compared to IM 400mg revealed a significantly higher chance for MR5.0 in the IM 800mg arm (HR 1.305, 95% CI 1.003-1.698, p=0.048). Baseline factors like thrombocytosis 〉 450/nl were associated with better responses (HR 1.701 compared to 〈 450/nl, 95% CI 1.405-2.059, p 〈 0.001) and higher leukocyte counts 〉 100/nl (HR 0.503 compared to 〈 50/nl, 95% CI 0.400-0.632, p 〈 0.001) and 50-100/nl (HR 0.746 compared to 〈 50/nl, 95% CI 0.591-0.942, p=0.014) with unfavorable responses. Other upfront factors like age, gender, blasts, eosinophils, hemoglobin, and EUTOS score did not significantly influence the probability for MR5.0. Taken all treatment arms together, our analyses have shown that the chance of achieving a MR5.0 by 8 years was considerably reduced if the pts had a BCR-ABLIS 〉 10% at 3 mos (40.2% vs 58.0%), 〉 1% at 6 mos (40.3% vs 68.7%), 〉 0.1% at 12 mos (37.7% vs 72.0%), and 〉 0.1% at 24 mos (21.5% vs 60.5%). Conclusion This evaluation of a large randomized trial reveals feasibility of MR5.0 detection in the majority of pts underlining the benefits of standardized molecular monitoring on the IS with optimized highly sensitive technologies. Baseline low leukocyte count, high thrombocyte count and high dose IM treatment are predictors of future MR5.0. Further, early molecular landmarks qualify for excellent outcome giving hope to a rising number of pts to successfully discontinue treatment and avoid possible side effects or comorbidities. Disclosures: Müller: Novartis: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Honoraria, Research Funding; Ariad: Consultancy, Honoraria, Research Funding. Hehlmann:BMS: Consultancy, Research Funding; Novartis: Research Funding. Hochhaus:Novartis: Consultancy, Honoraria, Research Funding, Travel Other; BMS: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Ariad: Consultancy, Honoraria. Saussele:Novartis: Honoraria, Research Funding, Travel Other; BMS: Honoraria, Research Funding, Travel, Travel Other; Pfizer: Honoraria.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3996-3996
    Abstract: Current evidence indicates that acquired genetic instability in chronic myeloid leukemia (CML) as a consequence of the balanced reciprocal translocation t(9;22)(q34;q11) or the variant translocation t(v;22) and the resulting BCR-ABL fusion causes the continuous acquisition of additional chromosomal aberrations (ACA) and mutations and thereby progression to accelerated phase and blast crisis (BC). At least 10% of patients in chronic phase (CP) CML show ACA already at diagnosis and more than 80% of patients acquire ACA during the transformation process into BC. Therefore, alterations at diagnosis as well as acquisition of chromosomal changes during treatment are considered as a poor prognostic factor. Differences in progression-free survival (PFS) and overall survival (OS) have been detected depending on the type of ACA. Patients with major route ACA (+8, i(17)(q10), +19, +der(22)t(9;22)(q34;q11)) and with other alterations like -X, del(1)(q21), del(5)(q11q14), +10, -21 at diagnosis resulting in an unbalanced karyotype have a worse outcome. Patients with minor route ACA (for example reciprocal translocations other than the t(9;22)(q34;q11) (e.g. t(1;21), t(2;16), t(3;12), t(4;6), t(5;8), t(15;20)) resulting in a balanced karyotype show no differences in OS and PFS compared to patients with the standard translocation, a variant translocation or the loss of the Y chromosome (Fabarius et al., Blood 2011). Here we compare the type of chromosomal changes (i.e. balanced vs. unbalanced karyotypes) during the course of the disease from CP to BC aiming to provide a valid parameter for future risk stratification. Patients and Methods Clinical and cytogenetic data available from 1,346 out of 1,524 patients at diagnosis (40% females vs. 60% males; median age 53 years (range, 16-88)) with Philadelphia and BCR-ABL positive CP CML included until March 2012 in the German CML-Study IV (a randomized 5-arm trial to optimize imatinib therapy) were investigated. ACA were comparatively analyzed in CP and in BC. Results At diagnosis 1,174/1,346 patients (87%) had the standard t(9;22)(q34;q11) only and 75 patients (6%) had a variant t(v;22). Ninety-seven patients (7%) had additional cytogenetic aberrations. Of these, 44 patients (3%) lacked the Y chromosome (-Y) and 53 patients (4%) had ACA. Regarding the patients with ACA thirty-six of the 53 patients (68%) had an unbalanced karyotype and 17/53 patients (32%) a balanced karyotype. During the course of the disease 73 patients (out of 1,524 patients) developed a BC during the observation time (5%). Cytogenetic data were available in 52 patients with BC (21 patients with BC had no cytogenetic analysis). Three patients had a normal male or female karyotype after stem cell transplantation. Nine patients showed the translocation t(9;22)(q34;q11) or a variant translocation t(v;22) (six and three patients, respectively) only and in 40 patients ACA could be observed in BC (40/49 (82%)). Out of these 40 patients with ACA, 90% showed an unbalanced karyotype whereas only 10% of patients had a balanced karyotype. No male patient in BC showed the loss of the Y chromosome pointing to a minor effect of this numerical alteration on disease progression. Conclusion We conclude that patients with CML and unbalanced karyotype at diagnosis are under higher risk to develop CML BC compared to patients with balanced karyotypes or compared to patients without ACA. In BC, 90% of CML patients showed unbalanced karyotypes (only 68% of CML patients at diagnosis have unbalanced karyotypes) supporting the hypothesis that the imbalance of chromosomal material is a hallmark of disease progression, representing the natural history of the disease from CP to BC and indicating therefore a strong prognostic impact. Consequently, different therapeutic options (such as intensive therapy or stem cell transplantation) should be considered for patients with unbalanced karyotypes in CP CML at diagnosis. Disclosures: Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Hehlmann:BMS: Consultancy, Research Funding; Novartis: Research Funding. Hochhaus:Novartis: Consultancy, Honoraria, Research Funding, travel Other; BMS: Consultancy, Honoraria, Research Funding; Pfizer : Consultancy, Honoraria; Ariad : Consultancy, Honoraria. Müller:Ariad: Honoraria; BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding, Speakers Bureau. Saussele:Pfizer: Honoraria; BMS: Honoraria, Research Funding, Travel, Travel Other; Novartis: Honoraria, Research Funding, Travel Other.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3138-3138
    Abstract: Introduction: The clonal selection of a mutant BCR-ABL positive clone can be observed in about one of two patients with imatinib-resistant chronic myeloid leukemia (CML). The early detection of BCR-ABL kinase domain mutations is crucial, since it allows to change the tyrosine kinase inhibitor (TKI) regimen in a timely manner and may therefore prevent disease progression and the accumulation of further genetic lesions. European LeukemiaNet (ELN) recommendations suggest a mutation analysis if optimal response criteria are not achieved at 3, 6, 12 or 18 months, or whenever a loss of optimal response occurs (Soverini et al., Blood 2011). Several attempts have been made to derive this indication from a specific increase of BCR-ABL levels. Here we report on the correlation of a rise in BCR-ABL transcript levels and the prevalence of BCR-ABL kinase domain mutations in imatinib-treated patients of the CML-Study IV. Methods: A total of 1,173 patients were enrolled until 2009 and randomized to one of four imatinib-based treatment arms. BCR-ABLIS of 988 patients was determined in 7,876 samples by quantitative RT-PCR in the central laboratory (median sample number per patient: 8.4, range 1-37; median follow up: 34 months, range 0-86), representing the eligible patients for the study. Thereby, the estimated intra-laboratory variance is assumed to be about 20%. A first rise of BCR-ABLIS to at least two-fold and 〉 0.1% between two samples of a patient's molecular course defined a sample suspected of bearing a mutant BCR-ABL positive clone. A mutation analysis was performed on this critical sample by direct sequencing of ABL exons 4 to 10. Results: A critical rise in BCR-ABLIS was observed in 231 of 988 patients (23%) after a median of 15.2 months on treatment (range 2.8-59.4). In the corresponding sample 33 mutant clones could be detected in 31 patients (13%). Thereby a steeper rise of BCR-ABLIS was correlated with a higher incidence of BCR-ABL mutations in the respective group (table). A total of 18 different mutations could be detected, the most frequent were: M244V, n=7 (21%); E255K, n=4 (12%); T315I, n=3 (9%); L248V, G250E, L387M and F486S, n=2 (6%), respectively. Mutations occur in a substantial proportion (8%) of patients with an only 2 to 3-fold rise of BCR-ABLIS transcript levels (table). Therefore, the most sensitive cut-off should be applied and mutation analysis may be triggered by a doubling of BCR-ABL transcripts at levels 〉 0.1% IS. Conclusion: BCR-ABL kinase domain mutations occur already in a substantial proportion of patients with a doubling of BCR-ABL transcript levels, which should determine mutation analysis. Table 1. Rise of BCR-ABL expression Patients (n) Patients with BCR-ABL mutations (n) Patients with BCR-ABL mutations (%) Inter-sample interval(median, days) 2 to 3-fold 72 6 8.3 98 3 to 5-fold 50 3 6.0 100 5 to 10-fold 39 4 10.3 99 10 to 100-fold 49 10 20.4 98 〉 100-fold 21 8 38.1 125 〉 2-fold (total) 231 31 13.4 101 Disclosures Hanfstein: Novartis: Research Funding; Bristol-Myers Squibb: Honoraria. Hehlmann:Novartis: Research Funding; Bristol-Myers Squibb: Research Funding. Saussele:Novartis: Honoraria, Research Funding, Travel Other; Bristol-Myers Squibb: Honoraria, Research Funding, Travel, Travel Other; Pfizer: Honoraria, Travel, Travel Other. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Neubauer:MedUpdate: Honoraria, Speakers Bureau. Kneba:Novartis: Consultancy, Equity Ownership, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Pfirrmann:Novartis: Consultancy; Bristol-Myers Squibb: Honoraria. Hochhaus:Pfizer: Consultancy, Research Funding; ARIAD: Honoraria, Research Funding; Bristol-Myers Squibb: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding. Müller:Novartis: Honoraria, Research Funding; Bristol Myers Squibb: Honoraria, Research Funding; ARIAD: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3761-3761
    Abstract: Abstract 3761 Introduction: Early assessment of molecular and cytogenetic response at 3 months of imatinib treatment has been shown to predict survival and might trigger treatment intensification in slow responders who are supposed to harbor a BCR-ABL positive clone with inferior susceptibility to tyrosine kinase inhibition (Hanfstein et al., Leukemia 2012). BCR-ABL transcript levels at 3 months depend on levels at diagnosis and the subsequent decline under treatment. Which of both parameters determines the clinical course and allows for prediction of survival is unclear. The BCR-ABL/ABL ratio is supposed to be skewed for high values, e.g. 〉 10%, due to the fact that ABL transcripts are also amplified from the fusion gene and in fact BCR-ABL/(ABL + BCR-ABL) is determined. Therefore, Beta-glucuronidase (GUS) was used as reference gene to determine high transcript levels at diagnosis. In addition, the linearity of the BCR-ABL/GUS scale allowed for an optimization of prognostic cut-off levels. We compared the significance of 1) BCR-ABL/GUS at diagnosis, 2) BCR-ABL/GUS at 3 months, 3) the individual reduction of transcripts given by (BCR-ABL/GUS at 3 months)/(BCR-ABL/GUS at diagnosis), and 4) the established 10% BCR-ABL/ABL landmark expressed on the international scale (BCR-ABLIS). Patients and methods: A total of 337 patients (pts) were investigated. According to the protocol of the German CML study IV pts could have been pre-treated with imatinib up to 6 weeks before randomization. 56 pts with imatinib onset before initial blood sampling within the study were excluded from the analysis. A total of 281 evaluable patients (median age 51 years, range 17–85, 42% female) were treated with an imatinib-based therapy consisting of imatinib 400 mg/d (n=76), imatinib 800 mg/d (n=110) and combinations of standard dose imatinib with interferon alpha (n=84) and low-dose cytarabine (n=11). Median follow-up was 4.8 years (range 1–10). Transcript levels of BCR-ABL, ABL, and GUS were determined by quantitative RT-PCR from samples taken before imatinib onset (“at diagnosis”) and 3 month samples. Only patients expressing typical BCR-ABL transcripts (b2a2 and/or b3a2) were considered. Disease progression was defined by the incidence of accelerated phase, blastic phase or death from any reason. A landmark analysis was performed for progression free survival (PFS) and overall survival (OS) after dichotomizing patients by a cut-off optimized by the cumulative martingale residuals method. Results: The median BCR-ABL/GUS ratio was 15.5% at diagnosis (0.07–271) and 0.62% at 3 months (0–34.7) reflecting a decline by 1.4 log. Disease progression was observed in 17 patients (6.0%), 14 of them died (5.0%). With regard to the above described parameters the following findings were observed: 1) at diagnosis no cut-off level could be identified for BCR-ABL/GUS ratios to separate two prognostic groups according to long-term PFS or OS. 2) At 3 months an optimized 2.8% BCR-ABL/GUS cut-off separated a high-risk group of 61 pts (22% of pts, 8-year PFS 78%, 8-year OS 81%) from a good-risk group of 220 pts (78% of pts, 8-year PFS 94%, 8-year OS 94%, p 〈 0.001, respectively). 3) At 3 months an individual reduction of BCR-ABL transcripts to at least 40% (0.4 log) of the initial level separated best and divided a high-risk group of 33 pts (12% of pts, 8-year PFS 74%, 8-year OS 80%) from a good-risk group of 248 pts (88% of pts, 8-year PFS 93%, 8-year OS 93%, p 〈 0.001, respectively). 4) When the established 10% BCR-ABLIS at 3 months was investigated, 63 pts were high-risk (22% of pts, 8-year PFS 82%, 8-year OS 85%) and 218 good-risk (78% of pts, 8-year PFS 91%, 8-year OS 93%, p=0.002 for PFS, p=0.011 for OS). Conclusions: Initial BCR-ABL transcript levels at diagnosis did not show prognostic significance. To predict survival at 3 months of treatment the absolute transcript level normalized by ABL or GUS can be used. Disclosures: Schnittger: MLL Munich Leukemia Laboratory: Equity Ownership. Hochhaus:Novartis, BMS, MSD, Ariad, Pfizer: Consultancy Other, Honoraria, Research Funding. Müller:Novartis, BMS: Consultancy, Honoraria, Research Funding.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2773-2773
    Abstract: Introduction: With many treatment options for chronic myeloid leukemia (CML), endpoints like health-related quality of life (HRQoL) move into focus and might be essential for deciding on treatment strategies. We sought to evaluate HRQoL in CML patients who had been registered in four consecutive studies of the German CML study group. Methods: The EORTC QLQ-C30 questionnaire was used to assess HRQoL of CML patients. Functional scales and global health status were calculated in accordance with Aaronson (1993) and Fayers (2001). With scales ranging from 0 to 100, 8 points are regarded as a minimally important difference (Efficace et al., 2013). Baseline data of responders (R) and non-responders (NR) were compared. Associations between two variables were assessed by the Fisher or Mann-Whitney tests, as appropriate. The global health status and the functioning scores were compared between groups with the van Elteren test, if the groups were stratified for another variable. Furthermore, results of the global health status and the functioning scores in our sample were standardized in accordance with the age (18-29, 30-39, 40-49, 50-59, 60-69, ≥70 years) and sex distribution of the 2448 participants of the German HRQoL outcome study reported by Hinz et al. 2014. The outcome of our sample was then compared with the outcome of these 2448 patients representing QoL of the German population in general. Comparison was performed using a t test. Results: A questionnaire was sent to 1634 patients. During January to April 2011, 858 questionnaires (53%) were sent back. Compared to NR, R were older (median age: 55 vs. 58, p=0.0426); years since diagnosis (median 6.5 vs. 7.4) and the percentage that had been transplanted were lower (24%vs.18%). No differences were observed regarding sex, Euro score, or time after allogeneic hematopoietic stem cell transplantation (HSCT). When answering the questionnaire, 517 (60%) patients received imatinib 400mg (IM400) and 102 (12%) were off therapy after HSCT. Less than 10% of patients received imatinib 800mg, imatinib+AraC or interferon alpha, nilotinib, or dasatinib. Time since diagnosis was ≤3 years in 156 (18%), 〉 3 and ≤7 years in 309 (36%), and 〉 7 years in 393 (46%) of the patients. Women (352, 41%) perceived a significant reduction in global health status (mean: 62.7, p 〈 0.001), role (65.4, p=0.0016), emotional (60.3, p=0.0002), and physical functioning (74.9, p 〈 0.0001) when compared to males (68.9, 71.5, 67.6, and 82.7, respectively). In the latter two cases, this perception met the definition of a clinical relevance. Results on significance did not change with adjustment for age. Compared to the German population, the 858 CML patients had significantly lower scores for global health status (mean: 67.9, p 〈 0.0001), role (70.8, p 〈 0.0001), social (69.2, p 〈 0.0001), emotional (64.6, p 〈 0.0001), physical (81.0, p 〈 0.0001) and cognitive functioning (77.3, p 〈 0.0001). Only for global health status, the difference was below 8. To evaluate HRQoL in patients with long standing disease, 100 patients with diagnosis 〉 7 years off therapy after HSCT and 203 patients receiving IM400 were analyzed. Adjusted for age group and sex, CML patients receiving IM400 for more than 7 years had lower scores for global health status (mean: 63.8, p 〈 0.0001 ), role (66.7, p 〈 0.0001), social (68.8, p 〈 0.0001), emotional (64.0, p 〈 0.0001), physical (75.2, p 〈 0.0001) and cognitive functioning (68.0, p 〈 0.0001) than the German control population. With respect to all six HRQoL scores, significantly lower scores than from the German population were also observed for the CML patients being seven years without treatment after HSCT: global health status (mean: 69.2, p 〈 0.0001 ), role (68.6, p 〈 0.0001), social (67.5, p 〈 0.0001), emotional (68.1, p 〈 0.0001), physical (83.1, p 〈 0.0001) and cognitive functioning (71.2, p=0.0053). Conclusions: In this cross-sectional study, women showed an impaired global health status, role, emotional, and physical functioning compared to males. Considering all 858 CML patients, the HRQoL was significantly impaired in all scales when compared to the German population. The same results were observed for the subgroups of patients either receiving IM400 for at least 7 years or being off therapy 7 years after HSCT. Reduced HRQoL remains an issue for all patients after long-term TKI treatment or after HSCT. These data may serve as a basis to evaluate HRQoL in stopping studies in CML. Disclosures Saussele: BMS: Honoraria, Other: Travel grant, Research Funding; Novartis Pharma: Honoraria, Other: Travel grant, Research Funding; ARIAD: Honoraria; Pfizer: Honoraria, Other: Travel grant. Kremers:Novartis: Honoraria; Bristol Myers Squibb: Other: Travel costs, supporting educational meeting; Novartis: Other: supporting educational meeting. Hochhaus:Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; ARIAD: Honoraria, Research Funding. Müller:BMS: Honoraria, Other: Consulting or Advisory Role, Research Funding; Novartis: Honoraria, Other: CONSULTING OR ADVISORY ROLE, Research Funding; ARIAD Pharmaceuticals Inc.: Honoraria, Other: Consulting & Advisory Role, Research Funding. Hehlmann:Novartis Pharma: Research Funding; BMS: Consultancy. Pfirrmann:BMS: Consultancy, Honoraria; Novartis Pharma: Consultancy, Honoraria.
    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
    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