Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Immunobiology, Elsevier BV, Vol. 180, No. 4-5 ( 1990-6), p. 385-394
    Type of Medium: Online Resource
    ISSN: 0171-2985
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1990
    detail.hit.zdb_id: 2060227-3
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Annals of Hematology, Springer Science and Business Media LLC, Vol. 62, No. 1 ( 1991-2), p. 25-31
    Type of Medium: Online Resource
    ISSN: 0939-5555 , 1432-0584
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 1991
    detail.hit.zdb_id: 1458429-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 12-12
    Abstract: Background: We recently showed that the combination of ATRA and arsenic trioxide (ATO) is at least not inferior and possibly superior to standard ATRA and chemotherapy (CHT) in the front-line management of low/intermediate risk APL (Italian-German APL 0406 trial; Lo-Coco et al., NEJM 2013). We report herein on the extended and final series of 276 patients (162 were in the previous report) with the last case being enrolled into the study in January 2013. Methods: The APL0406 study was a prospective, open-label, randomized intergroup trial conducted by the Italian GIMEMA and the German SAL and AMLSG study groups. Eligible patients were adults aged 18- 〈 71 years with newly diagnosed, genetically confirmed, non-high-risk (WBC≤10x109/L) APL. Patients were randomized to receive the ATO+ATRA combination originally developed by the MD Anderson Cancer Center group, or the Italian AIDA2000 protocol (Estey et al., Blood 2006 and Lo-Coco et al., Blood 2010). Patients in the ATRA-ATO arm received ATO 0.15/kg plus ATRA 45 mg/m2 daily until CR, then ATO 5 days/week, 4 weeks on 4 weeks off, for a total of 4 courses and ATRA 2 weeks on and 2 weeks off for a total of 7 courses. Patients in the ATRA-CHT arm received the standard AIDA (ATRA+Idarubicin) induction followed by 3 cycles of anthracycline-based consolidation together with ATRA and low dose CHT and ATRA for maintenance. The primary study objective was EFS at 2 years. Results: A total of 254 patients were evaluable for response to induction. CR was achieved in 122/122 (100%) in the ATRA-ATO versus 128/132 (97%) in the ATRA-CHT arm (P=0.12). Four patients died during induction in the ATRA-CHT arm. After a median follow-up of 36 months (range 1-75 months), the 2-year EFS was 98% and 84.9% in the ATRA-ATO and ATRA-CHT groups respectively (P= 0.0002), The 2-year cumulative incidence of relapse (CIR) rate was 1.1% and 9.4%, respectively (P=0.005) and, finally, the 2-year overall survival (OS) rate was 99.1% vs. 94.4% (P=0.01) for ATRA-ATO vs ATRA-CHT, respectively. Conclusions: The data on this extended cohort demonstrate a significantly augmented survival benefit coupled to a higher antileukemic efficacy provided by ATRA-ATO as compared to ATRA-CHT, in low/intermediate risk APL. These results further support ATRA-ATO as the new standard of care in this clinical setting. Figure 1 Figure 1. Disclosures Platzbecker: Teva: Honoraria. Off Label Use: Presentation includes off-label use of arsenic trioxide (ATO) in front-line management of APL. ATO is currently approved in treatment of relapsed APL in the US and Europe. Breccia:Bristol Myers Squibb: Consultancy; Novartis: Consultancy. Fiedler:TEVA: Travel reimbursement for meeting attendance Other. Lübbert:Cephalon / TEVA: Travel support Other. Link:TEVA: Consultancy, Speakers Bureau. Radsak:Celgene: Research Funding. Döhner:TEVA: Research Funding. Schlenk:TEVA: Research Funding, Speakers Bureau. Lo-Coco:TEVA: Honoraria; Lundbeck: Honoraria.
    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 ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 31, No. 22 ( 2013-08-01), p. 2819-2824
    Abstract: The introduction of BEACOPP escalated (escalated-dose bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone) has significantly improved tumor control and overall survival in patients with advanced-stage Hodgkin lymphoma. However, this regimen has also been associated with higher treatment-related mortality (TRM). Thus, we analyzed clinical course and risk factors associated with TRM during treatment with BEACOPP escalated . Patients and Methods In this retrospective analysis, we investigated incidence, clinical features, and risk factors for BEACOPP escalated -associated TRM in the German Hodgkin Study Group trials HD9, HD12, and HD15. Results Among a total of 3,402 patients, TRM of 1.9% (64 of 3,402) was mainly related to neutropenic infections (n = 56; 87.5%). Twenty of 64 events occurred during the first course of BEACOPP escalated (31.3%). Higher risk of TRM was seen in patients age ≥ 40 years with poor performance status (PS) and in patients age ≥ 50 years. PS and age were then used to construct a new risk score; those with a score ≥ 2 had TRM of 7.1%, whereas patients who scored 0 or 1 had TRM of 0.9%. Conclusion The individual risk of TRM associated with BEACOPP escalated can be predicted by a simple algorithm based on age and PS. High-risk patients should receive special clinical attention.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2013
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 493-493
    Abstract: Abstract 493 Background: According to retrospective analyses, the presence of a mutated Nucleophosmin-1 gene (NPM1+) in acute myeloid leukemia (AML) is associated with a favorable prognosis, particularly in the absence of an FLT3-ITD mutation (FLT3-ITD-). Therefore, AML with NPM1+/FLT3-ITD- and normal karyotype has been classified as favorable risk in current prognostic classifications. In order to assess the predictive value with regard to allogeneic stem cell transplantation (allo SCT), we compared the clinical course of 309 NPM1+ AML patients eligible for allo SCT in a donor versus no-donor analysis. Patients and Methods: Patients diagnosed with AML, aged 18–60 years, and treated in the AML 2003 trial of the Study Alliance Leukemia (SAL) were analyzed. According to the risk-adapted treatment strategy of the trial, cytogenetically intermediate-risk (IR) and adverse-risk (AR) patients should receive an allo SCT as consolidation treatment if an HLA-identical-sibling donor (IR) or HLA-matched related or unrelated donor (AR) was available. Patients with no available donor received high-dose cytarabine-based consolidation or autologous SCT. In order to avoid selection bias in an as-treated analysis of transplanted versus non-transplanted patients, we compared relapse-free survival (RFS) and overall survival (OS) depending on the availability of a suitable donor in a donor-no-donor analysis. Survival analyses were performed using the Kaplan-Meier method including log-rank tests for significance testing. Cox regression models and Wald tests were used for multivariate analyses on the influence of potential prognostic variables on the outcomes. Results: Of 1182 patients enrolled in the AML 2003 trial between December 2003 and November 2009, 375 were NPM1+ (32%), and 309 patients were eligible for evaluation for the donor vs. no-donor analysis. Their median age was 49 years, 304 patients had an intermediate-risk karyotype according to MRC criteria (98%), and amongst them there were 277 patients with a normal karyotype (90%). The FLT3-ITD mutation was present in 144 patients (37%). A donor was identified for 77 patients (25%), of whom 57 actually received allo SCT as first consolidation (74%). The no-donor group consisted of 232 patients. Age, disease status, cytogenetic profile, and FLT3-ITD incidence were equally distributed between the two groups. Median follow up was 41 months (3.4 years). The 3-year RFS in the donor and no-donor groups was 72% (95%–CI 61%–82%) and 47% (95%–CI 40%–55%), respectively. (p=0.007). The OS in the donor and no-donor groups were 70% (95%–CI 59%–81%) versus 60% (95%–CI 54%–67%) after 3 years, and 70% (95%–CI 59%–81%) versus 53% (95%–CI 45%–61%) after 5 years (p=0.138). In multivariate analyses, the presence of a donor as a prerequisite for allo SCT retained its statistically significant favorable influence on RFS (HR=0.56) even after adjustment for established risk factors such as FLT3-ITD, cytogenetic risk, WBC, LDH, age, and disease status. In patients with normal karyotype and NPM1+/FLT3-ITD- (n=152), the 3-year RFS in the donor and no-donor groups was 87% (95%–CI 77%–97%) and 53% (95%–CI 42%–63%), respectively (p=0.001). Conclusions: According to our results, allo SCT leads to a significantly prolonged RFS in NPM1+ AML patients with a pronounced effect even in NPM1+/FLT3-ITD- patients. The absence of a statistically significant difference in OS is most likely due to the fact that relapsed NPM1+ patients responded well to salvage treatment, particularly to allo SCT from an unrelated donor. Our data suggest that patients with NPM1+ AML who have a well-matched donor benefit from allo SCT in first remission. This hypothesis is currently being tested prospectively in a randomized controlled trial (“ETAL-1”, NCT01246752) evaluating allo SCT in all intermediate-risk AML patients with a well-matched sibling or unrelated donor identified until the achievement of first CR. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 6, No. 1 ( 2006-12)
    Abstract: Sensitivity of breast tumors to anticancer drugs depends upon dynamic interactions between epithelial tumor cells and their microenvironment including stromal cells and extracellular matrix. To study drug-sensitivity within different compartments of an individual tumor ex vivo , culture models directly established from fresh tumor tissues are absolutely essential. Methods We prepared 0.2 mm thick tissue slices from freshly excised tumor samples and cultivated them individually in the presence or absence of taxol for 4 days. To visualize viability, cell death, and expression of surface molecules in different compartments of non-fixed primary breast cancer tissues we established a method based on confocal imaging using mitochondria- and DNA-selective dyes and fluorescent-conjugated antibodies. Proliferation and apoptosis was assessed by immunohistochemistry in sections from paraffin-embedded slices. Overall viability was also analyzed in homogenized tissue slices by a combined ATP/DNA quantification assay. Results We obtained a mean of 49 tissue slices from 22 breast cancer specimens allowing a wide range of experiments in each individual tumor. In our culture system, cells remained viable and proliferated for at least 4 days within their tissue environment. Viability of tissue slices decreased significantly in the presence of taxol in a dose-dependent manner. A three-color fluorescence viability assay enabled a rapid and authentic estimation of cell viability in the different tumor compartments within non-fixed tissue slices. Conclusion We describe a tissue culture method combined with a novel read out system for both tissue cultivation and rapid assessment of drug efficacy together with the simultaneous identification of different cell types within non-fixed breast cancer tissues. This method has potential significance for studying tumor responses to anticancer drugs in the complex environment of a primary cancer tissue.
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 2041352-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 96, No. 5 ( 2000-09-01), p. 1933-1939
    Abstract: Chronic myeloid leukemia (CML) is a malignant stem cell disease characterized by an expansion of myeloid progenitor cells expressing the constitutively activated Bcr-Abl kinase. This oncogenic event causes a deregulation of apoptosis and cell cycle progression. Although the molecular mechanisms protecting from apoptosis in CML cells are well characterized, the cell cycle regulatory event is poorly understood. An inhibitor of the cyclin-dependent kinases, p27, plays a central role in the regulation of growth factor dependent proliferation of hematopoietic cells. Therefore, we have analyzed the influence of Bcr-Abl in the regulation of p27 expression in various hematopoietic cell systems. An active Bcr-Abl kinase causes down-regulation of p27 expression in murine Ba/F3 cells and human M07 cells. Bcr-Abl blocks up-regulation of p27 after growth factor withdrawal and serum reduction. In addition, p27 induction by transforming growth factor-beta (TGF-β) is completely blocked in Bcr-Abl positive M07/p210 cells. This deregulation is directly mediated by the activity of the Bcr-Abl kinase. A Bcr-Abl kinase inhibitor completely abolishes p27 down-regulation by Bcr-Abl in both Ba/F3 cells transfected either with a constitutively active Bcr-Abl or with a temperature sensitive mutant. The down-regulation of p27 by Bcr-Abl depends on proteasomal degradation and can be blocked by lactacystin. Overexpression of wild-type p27 partially antagonizes Bcr-Abl–induced proliferation in Ba/F3 cells. We conclude that Bcr-Abl promotes cell cycle progression and activation of cyclin-dependent kinases by interfering with the regulation of the cell cycle inhibitory protein p27.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2000
    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. 114, No. 22 ( 2009-11-20), p. 20-20
    Abstract: Abstract 20 Introduction We have shown in a monocenter study (ASH 2005, abstract # 428) that routine prophylactic platelet (plt) transfusions are not necessary in patients (pts) with acute myeloid leukemia (AML). Therefore, we started a multicenter randomized study comparing the routine prophylactic (morning trigger: plts ≤ 10/nl, arm P) with a therapeutic platelet transfusion strategy (arm T). Methods Diagnosis of AML and feasability of intensive chemotherapy within standard protocols was required (M3 only in CR). In arm T transfusions were given only when clinically relevant bleeding (more than petechiae) happened regardless of the morning plt count. For safety reasons prophalyctic plts were transfused in case of invasive aspergillosis, sepsis syndrom and repeated or continous headache. In case of headache pts in arm T received a ct-scan. The experimental transfusion strategy startet at day 8 of induction and at day 1 of consolidation. Primary endpoint was the reduction of plt transfusions by 20%; secondary endpoints were bleeding complications, red blood cell transfusions, side effects, duration of thrombocytopenia and hospital stay. Results Until July 2009 175 pts have been randomized. Full data were evaluable for 161 pts (79 arm P and 82 arm T). Median age was 52 years (20-78). We registered 251 induction and 112 consolidation cycles. Mean duration of thrombocytopenia ( 〈 20/nl or 〈 10/nl) was 17 and 8 days, respectively. Duration of thrombocytopenia was significantly longer in arm T. Depite the longer duration, the number of platelet transfusions in arm T was significantly reduced, by about 25% (p 〈 0.001). Number of red blood cell transfusions were not different (p = 0.95). Days in hospital and side effects were not different in both groups. Bleeding complications (more than petechial) were significantly increased in arm T showing a 2.3 times higher risk compared to arm P. This is clearly correlated with bleeding being the trigger for plt transfusion in arm T. Majority of hemorrhages were of minor or moderate significance that could all be treated effectively by plt transfusion. Most important was the observation that we registered 5 minor and 2 major cerebral hemorrhages in arm T and none in arm P. Minor cerebral hemorrhages with headache were not registered in arm P since we did not perform ct-scans routinely in arm P in case of headache. Minor cerebral complications were 2 sub-/epidural bleedings after vasovagal collapses of pts at plt counts of 53 and 96/nl, respectively, and 3 spontanuous minor subarachnidal/ intracerebral hemorrhages at plt counts of between 15 and 〈 5/nl. All 5 minor cerebral complications were detected by ct-scan performed due to headache. All pts recovered after plt transfusion without any sequelae.The two other pts died following fatal cerebral bleeding. One pt. had repeated headache since several days. At a plt count of 11/nl fatal bleeding happened. A ct-scan revealed intracerebral mass bleeding. The pt was randomized to the experimental strategy but even following standard transfusion strategy the pt would not have been transfused prophylactically. The only fatal cerebral bleeding that must be clearly attributed to the therapeutic strategy was a female pt with a pulmonary fungal infection that improved under antifungal treatment. She had a morning platelet count of 〈 5/nl. During the following night she woke up with severe headache. Despite timely platelet transfusions she died of progressive mass bleeding. Unfortunately, we could not perform an autopsy to see whether the patient had fungal involvement of the brain. 1-2 % of fatal cerebral bleedings are reported in AML trials despite a prophylactic strategy. Whether these two complications happened by chance in arm T or were the consequence of the experimental strategy cannot be answered by our study. To give proof of this important question one would need about 2000 pts in both arms for statistical reasons. Conclusions A therapeutic platelet transfusion strategy according to our protocol is feasible in AML pts during intensive chemotherapy and can reduce the number of platelet transfusions significantly. An increased risk of fatal cerebral bleeding cannot be excluded by this study. An international collaboration would be necessary to set up a study large enough to determine the final safety. 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 329-329
    Abstract: Abstract 329 Background: Acute Myeloid Leukemia (AML) is a life threatening disease requiring intensive induction treatment for long-term survival. Since the majority of patients are older than 60 years at diagnosis, balancing pros and cons of intensive chemotherapy is difficult. In this context prognostic factors are important tools for clinical judgement, decision making and patient information. We present here our analysis of potential prognostic factors derived from the updated data of a large multicenter trial for patients with AML older than 60 years, who were treated according to the same protocol. Patients and Methods: The prospective AML96 trial of the Study Alliance Leukemia (former DSIL) included 909 patients with a median age of 67 years (range, 61-87). Protocol treatment included two cycles of induction therapy containing daunorubicin and cytarabine in a 7+3 fashion (DA). Consolidation treatment consisted of m-amsacrine and intermediate dose cytarabine (10g/m2). Univariate and multivariate analyses using potential prognostic factors were performed for the following outcomes: complete remission (CR), overall survival (OS), disease-free survival (DFS) and cumulative incidence of relapse (CIR) / time to progression (TTP). Results: The median follow-up time is 68 months (5.66 years). Of all treated patients, 414 (45%) completed two cycles of DA induction according to the protocol, 181 (20%) received only one DA, and 111 patients (12%) died during the first induction cycle. The remaining 203 patients (23%) received a second induction cycle outside the protocol due to insufficient response to the first induction. 203 patients received a consolidation treatment after two cycles of induction. 454 of all 909 patients reached a CR (50%). The independent prognostic factors for achieving CR were karyotype, NPM mutation status, white blood cell count (WBC), blast count at day 15 after initiation of first induction, CD34 expression and secondary versus primary AML. The median OS, DFS and TTP were 0.76, 0.76 and 0.88 years, respectively. Five-year OS, DFS and CIR were 9.7%, 14% and 79.1% respectively. Multivariate analyses revealed that karyotype, NPM mutation status, white blood cell count (WBC) and LDH were of independent prognostic significance for OS, DFS and CIR. Even though NPM-FLT3 grouping showed a significant influence on all outcomes, it seems that in the age group above 60, the negative impact of FLT3 was less influential compared to the positive impact of NPM mutational status. Favourable and high cytogenetic risk groups displayed markedly different OS times irrespective of the other identified prognostic factors. However, the largest group of patients with intermediate-risk karyotype could be subdivided into two groups with distinct prognostic profiles: Based on hazard ratios from the Cox model, we established an additive risk model using the following relative points (in brackets): high CD34 expression (1 point), high WBC (1 point), higher age (2 points), high LDH (3 points), and mutated NPM (-2 points). Applying this model results in the following four groups: favourable cytogenetics, intermediate cytogenetics with favourable risk features (score ≤3), intermediate.cytogenetics with adverse risk features (score 〉 3) and high risk cytogenetics. The 3-year OS of these groups were 39.5%, 30%, 10.6% and 3.3%, respectively. Conclusion: Results of a large cohort enable us to demonstrate that, depending on prognostic factors, elderly AML patients with an intermediate-risk cytogenetic profile can be subdivided into a more favourable group with an OS time span similar to CBF AML, and a group with a distinctively worse prognosis. The proposed risk-stratification score can serve as a tool for patient information and decision making with regard to treatment strategy. Whilst patients with CBF AML or with intermediate cytogenetics and favourable risk features potentially benefit from intensive induction treatment, patients with high-risk cytogenetics or intermediate risk with adverse prognostic features have very poor OS rates despite intensive chemotherapy. From our study we conclude that this group of patients could be spared the potential side effects of intensive induction and should instead be offered best supportive care or alternative treatment approaches. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 144-144
    Abstract: Abstract 144 Background: Sorafenib is a multi-kinase inhibitor with activity against several oncogenic kinases, which may play a role in the pathogenesis of acute myeloid leukemia (AML). In-vitro data and results from non-randomized clinical trials suggest that sorafenib might be an effective drug for the treatment of AML. So far, no randomized-controlled data are available for treatment of newly diagnosed AML patients up to the age of 60 years. We present the first results from the randomized placebo-controlled SORAML trial of the Study Alliance Leukemia (SAL). Patients and Methods: Between March 2009 and October 2011, 276 patients from 25 centers were enrolled in the SORAML trial (NCT00893373). The main eligibility criteria were: newly diagnosed AML, age from 18 to 60 years and suitability for intensive therapy. The treatment plan for all patients included two cycles of induction with DA (daunorubicin 60 mg/m2 days 3–5 plus cytarabine 100 mg/m2 cont. inf. days 1–7), followed by three cycles of high-dose cytarabine consolidation (3 g/m2 b.i.d. days 1, 3, 5). Patients without response after DA I received second induction with HAM (cytarabine 3 g/m2 b.i.d. days 1–3 plus mitoxantrone 10 mg/m2 days 3–5). Allogeneic stem cell transplantation was scheduled for all intermediate-risk patients in first complete remission with a family donor and for all high-risk patients with a matched donor. At study inclusion, patients were randomized to receive either sorafenib (800 mg/day) or placebo as add-on to standard treatment. Block randomization at a ratio of 1:1 was performed within cytogenetic and molecular risk strata, allocation was concealed and treatment was double blinded. Study medication was given on days 10–19 of DA I+II or HAM, from day 8 of each consolidation until 3 days before the start of the next consolidation and as maintenance for 12 months after the end of consolidation. The primary endpoint of the trial is event-free survival (EFS) with an event being defined as either failure to achieve a complete remission (CR) after induction, relapse or death. Secondary endpoints were overall survival (OS), CR rate and incidence of adverse events (AE). We present the results of the planned interim analysis (intent to treat) after the occurrence of 50% of EFS events. The O'Brien/Fleming adjusted significance level was set at p=0.0052. Results: Out of 276 randomized patients, 264 were evaluable for EFS, 132 in each arm. Demographic and disease characteristics were equally distributed between the two arms; the FLT3-ITD incidence was 16%. The median cumulative dose of administered study medication was equal in both arms. The CR rates were 56% versus 60% in the placebo versus sorafenib arm (p=0.622). By the time of analysis, a total number of 100 events had occurred. After a median observation time of 18 months, the median EFS was 12.2 months in the placebo arm and was not reached in the sorafenib arm, corresponding to a 1-year EFS of 50% versus 64% (p=0.023). The median OS had not been reached in both arms, the 2-year OS was 66% versus 72% in placebo and sorafenib arms, respectively (p=0.367). The most common reported AEs CTC Grade ≥3 were infectious complications including fever and pneumonia, followed by bleeding events, cardiac and hepatic toxicity, hypertension, skin toxicity and headache. The risk for hepatic toxicity (relative risk 6.2, p=0.025) and bleeding events (relative risk 3.6, p=0.016) was significantly higher in the sorafenib arm while the incidence of all other AEs showed no significant differences. Conclusions: In younger AML patients, the addition of sorafenib to standard chemotherapy is feasible but associated with a higher risk of liver toxicity and bleeding events. Sorafenib treatment resulted in a marked EFS prolongation; this difference is not significant according to the adjusted significance level of this interim analysis. Results from the final analysis including post-hoc exploration of molecularly defined subgroups are necessary for drawing final conclusions on the efficacy of sorafenib. Disclosures: Off Label Use: sorafenib for the treatment of acute myeloid leukemia. Serve:Bayer: Research Funding. Ehninger:Bayer: 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages