GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 732-732
    Abstract: Introduction: Despite progress in understanding acute myeloid leukemia (AML) biology, most patients (pts) still have a poor prognosis. MicroRNAs (miR) have emerged as important players in AML biology. High expression of miR-181a has been associated with improved outcomes in AML pts. Mature miR-181a originates from two precursor molecules pri-miR-181a-1 & pri-miR-181a-2 which have been associated with distinct biological features & are derived from chromosome 1q32.1 & 9q33.3, respectively. Allogeneic stem cell transplantation (HCT) offers a potential curative treatment option for AML pts. With the introduction of reduced intensity conditioning (RIC) regimens, this procedure also became available for older or comorbid individuals. Here, we measured pri-miR-181a-1 & pri-miR-181a-2 and analyzed their impact on survival in RIC-HCT treated AML pts. Patients and Methods: 139 AML pts (median age 64 years [y], range 22-75y) who received RIC-HCT between 2000 and 2012 with pretreatment material available were included in our analysis. The conditioning regimens were based on Fludarabine 30mg/m2 on day -4 till -1 followed by 2 Gy total body irradiation. Disease status at RIC-HCT was first (n=79, 57%) or second complete remission (CR; n=23, 16%) or more advanced disease (n=37, 27%). Donors were human leukocyte antigen (HLA) matched related (n=22, 16%) or HLA matched (n=83, 60%) or mismatched (n=34, 24%) unrelated. The mutation status of the NPM1 & CEBPA genes & the FLT3-ITD status were assessed at diagnosis. Pts were grouped according to the European LeukemiaNet (ELN) classification in favorable (26%), intermediate-I (26%), intermediate-II (21%) or adverse (27%). Pri-miR-181a-1 & pri-miR-181a-2 expressions were measured by RT-PCR, normalized to 18S & the median normalized gene expression was used to define high & low expressers. Median follow up was 4.3y for pts alive. Results: The expression of both precursor molecules correlated well (Pearson Correlation 0.78), but 23% had a discordant expression status. At diagnosis pts with high pri-miR-181a-1 expression were younger at time of RIC-HCT (P=.01), had higher % peripheral blood (P=.05) & bone marrow (P=.04) blasts & by trend fewer NPM1 mutations (P=.054). Pts with high pri-miR-181a-2 expression had higher white blood count (WBC, P=.04). Combining the expression status information for both precursors, pts with high pri-miR-181a-1 and/or high pri-miR-181a-2 expression were younger at time of RIC-HCT (P=.04).Pts with high pri-miR-181a-1 and/or high pri-miR-181a-2 expression had longer overall survival (OS; P=.004, Figure 1) & event-free survival (EFS; P=.002). Interestingly a strong impact on outcome was observed in the ELN favorable group (OS: P=.001 & EFS: P=.008, Figure 1) and in the ELN intermediate II group (OS: P 〈 .001 & EFS: P 〈 .001, Figure 1), while no significant impact of high pri-miR-181a-1 and/or high pri-miR-181a-2 expression on outcome was observed in the intermediate I & adverse ELN groups.In multivariate analyses pri-miR-181a-1 and/or high pri-miR-181a-2 expression status remained a strong prognostic factor for OS (hazard ratio [HR] 0.53; 95% confidence interval [CI] 0.36-0.79; P=.002) and EFS (HR 0.52 CI 0.36-0.75; P 〈 .001) in the entire group of pts. Conclusion: In conclusion, pri-miR-181a-1 & pri-miR-181a-2 expression correlated well, but were not fully concordant & associated with distinct clinical characteristics. The expression status of pri-miR-181a-1 & pri-miR-181a-2 at diagnosis is a strong independent prognostic factor for AML pts undergoing RIC-HCT. The prognostic impact was strongest in the ELN favorable and intermediate II groups. Pri-miR-181a-1 & pri-miR-181a-2 expression levels may contribute to improved risk stratification for AML pts undergoing RIC-HCT, refining the ELN favorable and intermediate II group. Furthermore, pretherapeutic miR elevation either pharmacologically or by miR-replacement therapies may improve outcomes in AML pts undergoing RIC-HCT. Figure 1 Figure 1. Disclosures Lange: Novartis: 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3266-3266
    Abstract: Abstract 3266 Poster Board III-1 Introduction: NmE2 (Nm23-H2, NDP kinase B) is one of a family of proteins that catalyze the transfer of gamma-phosphate between nucleoside-triphosphates and diphosphates. The two major family members, NmE1 and NmE2 are strongly implicated in the control of differentiation, proliferation, migration and apoptosis via interactions which are often independent of their kinase activity, NmE2 being a transcriptional activator of the c-myc gene. We recently identified NmE2 as a tumour associated, HLA-A32+ restricted, antigen in a patient with CML and found the protein (but not the mRNA) to be generally over expressed in CML but not in other haematological malignancies. We also detected a specific T-cell response in peripheral blood cells of a patient 5 years after transplantation. This identifies NmE2 as a potential target for both molecular and immunotherapy of CML. However, the development of immunotherapeutic approaches will depend on the ability of NmE2 to function as a tumour antigen in common HLA backgrounds. The aims of this study were firstly to investigate the antigenicity of NmE2 in the HLA-A2 background (which accounts for more than 50% of the Caucasian population), and secondly to characterise the regulatory relationship between Bcr/Abl and NmE2 using a cell line model of CML. Materials and Methods: 5 nonameric NmE2 peptides with predicted anchor amino acids for HLA-A2 were loaded at concentrations of 10μM separately onto HLA-A2 expressing antigen presenting cells. Elispot Assays were carried out with CD8+ MLLCs (for the identification of antigenic peptides) or CD8+ cells isolated directly from a CML patient at different time points after HCT. Ba/F3 cells stably expressing wild type and mutant forms of Bcr/Abl were treated with imatinib and nilotinib (0 – 10 μM) for 48h. Bcr/Abl activity was assessed by FACS using antibodies specific for the phosphorylated forms of CrkL and Stat5. NmE2 and c-Myc protein were detected by immunocytochemistry and Western blotting with specific antibodies [Santa Cruz, clones L-16 and 9E10 respectively]. Levels of nme2 and c-myc mRNA were determined by quantitative real time PCR. Results: Full length NmE2 protein and 2 of 5 HLA-A2 anchor-containing peptides tested (NmE2132–140 and NmE2112–120) were specifically recognized by the HLA-A2+ CD8+ MLLC, demonstrating the antigenicity of NmE2 in the HLA-A2 background in vitro. Furthermore, while CD8+ T-cells from a transplanted HLA-A2+ CML patient showed little or no specific reactivity in the first 10 months after HCT, a distinct reactivity (up to 0.6 % NmE2 reactive CD8+ T cells) became apparent at later stages, consistent with the development of an immune response against NmE2-expressing cells in vivo. The patient remained negative for bcr/abl transcripts throughout this period. BA/F3 Bcr/Abl cells expressed increased levels of NmE2 protein (but not mRNA) compared to the parent BA/F3 line. Interestingly, treatment with imatinib or nilotinib reduced NmE2 protein expression in BA/F3 Bcr/Abl, but not in cells expressing Bcr/Abl mutants resistant to the respective inhibitors. Treatment of BA/F3 Bcr/Abl cells with the PI3K inhibitor Ly294002 resulted in reduced Bcr/Abl activity and a corresponding reduction in both c-Myc and NmE2 protein levels, without affecting mRNA levels. Conclusion: The over expression of NmE2 is closely linked to Bcr/Abl kinase activity, the predominant level of regulation being post-transcriptional and dependent on PI-3K activity. The NmE2 protein is restricted by HLA-A2 as well as by HLA-A32. The development of an NmE2-specific T-cell response in a CML patient after stem cell transplantation suggests that NmE2 functions as a tumour antigen in HLA-A2+ patients in vivo and may be relevant to the long term immune control of CML. NmE2 is therefore a promising candidate for the development of new immunotherapeutic strategies for the treatment of CML. Disclosures: Lange: BMS: Honoraria; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Niederwieser:BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1037-1037
    Abstract: Background: Most acute myeloid leukemia (AML) patients (pts) stilldo not achieve long-term survival. Better risk stratification and novel therapeutic avenues are needed to improve pts outcomes. The expression of microRNAs has been demonstrated to be altered in AML & miR-based therapies are entering clinical trials. MiR-320a, maps to chromosome 8p21.3 & is known to play a role in several tumors; e.g. higher miR-320a expression suppresses the progression of colorectal cancer. In AML miR-320a has been shown to inhibit cell proliferation, likely by targeting the transferrin receptor 1. Objective: The objective of this study was to investigate whether a differential expression of pri-miR-320a associated with outcome in AML pts. Methods: We assessed the expression levels of pri-miR-320a, a precursor molecule of mature miR-320a. The pri-miR-320a expression levels in 129 AML pts were assessed by quantitative reverse transcription polymerase chain reaction & normalized to a housekeeping gene (18S). The 75th percentile was chosen as a cut-off discriminating between high & low pri-miR-320a expressers. We analyzed 129 AML pts (median age at HCT 64 years [y]; range 22–74 y) who received reduced intensity conditioning (RIC; Fludarabine 30mg/m^2 at day -4 to -2 & 2 Gy total body irradiation at day 0)-hematopoietic cell transplantation (HCT) at the University of Leipzig, with pretreatment bone marrow available. The median follow-up was 4.5 y for pts alive. European LeukemiaNet (ELN) genetic classification was: favorable (n=33; 25.6%), intermediate I (n=33; 25.6%), intermediate II (n=29; 22.5%) or adverse (n=30; 23.3%). The pts were also characterized for FLT3-ITD status, CEBPA, IDH1, IDH2 and NPM1 mutations. Results: At diagnosis high pri-miR-320a expression associated by trend with lower hemoglobin levels (P=.094), lower white blood cell counts (P=.079) & lower peripheral blast counts (P=.096). High pri-miR-320a expressers less frequently had NPM1 (P=.038) or CEBPA (P=.025) mutations. Interestingly, pri-miR-320a expression was significantly lower in pts with trisomy 8 (P=.018) compared to non-trisomy 8 pts & in the ELN intermediate II group none of the trisomy 8 pts was in the high pri-miR-320a expressing group. Analysis of our AML pts, showed a significant association between pri-miR-320a expression status & clinical outcome. In the entire group of pts high pri-miR-320a expressers had a longer overall survival (OS; P=.086; Figure 1) by trend & a significantly longer event-free survival (EFS; P=.032). The strongest impact of pri-miR-320a was found in the ELN intermediate II group, where high pri-miR-320a expression was associated by trend with longer OS (P=.059; Figure 1) & a significantly longer EFS (P=.034). In multivariate analysis, the prognostic impact of pri-miR-320a expression status was confirmed in the entire group of pts (OS: P 〈 .01, hazard ratio (HR) 0.45, 95% confidence interval (CI) 0.27-0.75; EFS: P 〈 .01, HR 0.45, 95% CI 0.28-0.71). Conclusion: High expression of pri-miR-320a associates with distinct clinical & molecular features. Interestingly, in AML pts with trisomy 8 pri-miR-320a that maps to chromosome 8 has a significantly lower expression, indicating a possible leukemogenic link to trisomy 8 associated AML. High pri-miR-320a expression significantly associates with better outcome especially in the ELN intermediate II group. These results suggest that pri-miR-320a could be used as a marker to refine current AML risk stratifications. Increasing miR-320a, by e.g. miR-replacement therapies, might improve outcomes of AML pts. Figure 1: Figure 1:. OS (A) and EFS (B) in AML pts according to pri-miRNA-320a expression status, OS (C) and EFS (D) in AML pts in the ELN intermediate II group according to pri-miRNA-320a expression status Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    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. 124, No. 21 ( 2014-12-06), p. 1860-1860
    Abstract: In retrospective studies, CALR mutation is found to be a favorable prognostic variable in myelofibrosis (MF) compared with JAK2, or MPL mutations. With the availability of the JAK1/JAK2 inhibitor ruxolitinib (RUX) for treatment of MF, it is not yet known whether response varies across mutational subgroups and whether the prognostic implication of the driver mutations in terms of survival (OS) changes under RUX. We studied the prevalence and prognostic weight of clinical parameters in the IPSS and DIPSS-plus and the impact of RUX on OS in the context of mutation status. Patient and Methods: As of November 2009, RUX for treatment of splenomegaly and/or constitutional symptoms became an option at the University of Leipzig. All patients (pt) with MF seen since then were included (n=127; f=43%, median age 58 years) with the exception of pt with MPL mut+ because of small number. Screening for the JAK2V617F and CALR mutations was performed as previously published. Cytogenetic-risk categorization was conducted as published (Caramazza et al. Leukemia 2011). Results: JAK2 mut+ (group A; 60.6%) constituted the largest group, followed by CALR mut+ (group B; 19.7%), and triple-negative (group C; 19.7%). The median duration of MF was 22 months with no difference in the 3 groups. Higher-risk IPSS was present in the majority [Int-II 34.4%; high-risk 42.4%]. Constitutional symptoms were present across all groups, although more frequent in group A which was also characterized by prominent splenomegaly (Table). Group B pt were younger with low WBC in contrast to goup C pt who were older, anemic, with high WBC and low platelets, unfavorable cytogenetics and high-risk IPSS. Hb 〈 100 g/L (54%), and transfusion-dependency (32%) were frequently present in group B and was similar to group A. RUX was given to 72 (57%) pt [graoup A (66%), group B (56%), group C (29%)] with a median exposure of 8 months. The response of 80% was not influenced by the mutation status, cytogenetics, or IPSS variables. Leukemic transformation was documented in 22 pt [14 (64%) pt had no RUX exposure] after a median of 18 months after diagnosis. The incidence was highest in group C (36%) and lowest in group B (4%) (p=0.004). Unfavorable cytogenetics were 75%, and 27% in group C and A respectively. After a median follow-up of 31 months, OS at 3-years was 82% and worst in group C (72%) vs 80% for group A (p=0.05) vs 96% in goup B (p=0.003). In univariate analysis, non-mutated status (p=0.02), Int-2/high-risk IPSS (p=0.06), anemia (p=0.03), transfusion dependency (p=0.04), and platelets 〈 100 x109/L (p=0.06) but not unfavorable cytogenetics were associated with an inferior OS. In multivariate analysis, advanced IPSS only (p=0.04) but not the mutation status (p=0.4) retained its negative impact on OS. Generally, in group A pt treated with RUX (n=50), OS was only marginally inferior to group B (p=0.08). In pt with int-2/high-risk IPSS, OS in group A with RUX (n=41) was similar to that of group B (p=0.4). Conclusions: The mutation status in MF bestows distinct clinical phenotypes and has crucial prognostic and therapeutic implications. Patients with non-mutated MF had the worst prognosis, unfavourable cytogenetics, and the highest rate of leukemic transformation. Although the IPSS retains its value, the prognostic power of certain factors such as anemia and constitutional symptoms in CALR-mutated pt needs to be re-evaluated. Response to ruxolitinib seems to be independent of the mutation status. More importantly, a JAK1/JAK2 inhibition appears to be capable of attenuating the prognostic implication of the different mutation profiles by improving the less-favorable survival associated with non-CALR-mutated MF through a disease-modifying effect. The full potential of a sustained JAK1/JAK2 inhibition in modifying survival in the context of the various mutational profiles needs to be further studied in a larger cohort of patients. Table Clinical variables in patients with different driver mutations Variable JAK2 mut+ CALR mut+ Triple-negative p N (%) 77 (60.6) 25 (19.7) 25 (19.7) Median age (years) 59 51 61 0.02 Constitutional symptoms (%) 69 52 46 0.08 Median palpable spleen (cm) 10 4 3 0.001 Int-2/high-risk IPSS (%) 79 52 96 0.01 WBC 〉 25 x109/L (%) 21 11.5 44 0.008 Median peripheral blasts (%) 1 0.5 2 ns Median Hb (g/L) 106 99 86 0.008 Hb 〈 100 g/L (%) 42 54 80 0.03 Transfusion dependency (%) 33 32 67 0.009 Platelets 〈 100 x109/L (%) 30 8 52 0.04 Unfavorable cytogenetics (%) 31 14 48 0.06 Disclosures Al-Ali: Novartis: Honoraria, Research Funding. Jaekel:Novartis: Honoraria. Lange:Novartis: Honoraria. Roskos:Novartis: Honoraria. Niederwieser:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 396-396
    Abstract: Mastocytosis is a hematopoietic disorder characterized by abnormal growth and accumulation of neoplastic mast cells (MC) in various organ systems. Using updated WHO criteria and the proposal of the consensus group, the disease can be divided into cutaneous mastocytosis (CM), indolent systemic mastocytosis (ISM), smouldering SM (SSM), SM with an associated hematologic neoplasm (SM-AHN), aggressive SM (ASM), and mast cell leukemia (MCL). In adult patients with skin involvement but unknown/unavailable bone marrow (BM) studies, the provisional diagnosis ´mastocytosis in the skin (MIS)´ is appropriate. Although this classification has been validated repeatedly and is of prognostic significance, additional prognostic parameters have been identified in recent years (yrs). We have established a patient-registry in the ECNM where over 1,000 cases with confirmed mastocytosis are included. The aim of this study was to identify and validate new prognostic variables predicting survival in patients with mastocytosis and to prepare a simple prognostic scoring system applicable in daily practice. Using the data set of the ECNM registry we analyzed overall survival (OS) and event-free survival (EFS; i.e. until death or progression) in 1,088 patients with mastocytosis (median age: 45.7 yrs; range: 0.1-83.3 yrs, f:m ratio, 1:0.79), including CM (n=152), MIS (n=126), ISM (n=650), SSM (n=26), SM-AHN (n=89), ASM (n=35), and MCL (n=10). The median observation period was 3.5 yrs (75-25% percentile: 1.5-6.9 yrs, maximum 34.1 yrs). In the entire cohort, the median OS was not reached. The probability to be alive after 5, 10, and 20 yrs was 89%, 83%, and 70%, respectively. As expected, the WHO classification turned out to be of utmost predictive significance (Figure 1A; p 〈 0.005). In patients with non-advanced disease, namely CM, MIS, ISM, and SSM, the median survival was not reached, and the survival at 5 yrs was 100%, 97%, 98%, and 85%, respectively, whereas in advanced SM, namely SM-AHN, ASM, and MCL, the median survival was 2.8, 4.1, and 0.8 yrs, respectively. Patients with advanced SM were found to be older, to have higher serum tryptase- and alkaline phosphatase (aPhos) levels, higher white blood counts (WBC), lower hemoglobin (Hb) and platelet (PLT) counts, and more frequently presented with organomegaly (hepatomegaly, splenomegaly, or lymphadenopathy). Moreover, the male/female ratio was higher in advanced mastocytosis. To define the relative impact of the identified risk factors we randomly divided the total cohort (50:50) into a learning set and a validation set, performed uni- and multivariate analyses, and subsequently calculated cut off values for optimal prognostication. In these studies, the poor prognosis of patients with advanced SM was confirmed. In patients with non-advanced disease, all variables tested were significant concerning OS in univariate analyses. However, WBC, Hb, and lactate dehydrogenase had to be excluded due to variance inflation. In multivariate analyses, age 〉 70 yrs, PLT 〈 80 G/L (not related to SM), and aPhos ≥240 U/L were significant predictors concerning OS. Based on these parameters, we established a simple prognostic scoring system. In this score, patients with non-advanced disease (CM, MIS, ISM, SSM) without additional risk factors comprised the low risk group, those with non-advanced disease and presence of one or more risk factors (age 〉 70 yrs, PLT 〈 80 G/L, aPhos ≥240 U/L) the intermediate risk group, and those with advanced disease (with or without additional risk factors) the high risk group. The median OS in the low risk group was not reached, in the intermediate risk group it was 13.5 yrs, and in the high risk group 3.5 yrs (p 〈 0.005; Figure 1B). Significant differences were also observed regarding EFS (p 〈 0.005, Figure 1C). In conclusion, the WHO classification remains the gold-standard of prognostication in patients with mastocytosis, but additional factors, namely age, PLT, and aPhos, are powerful additional variables predicting OS in these patients. Based on in-depth analyses of the ECNM registry data-set, a simple prognostic scoring system for mastocytosis was established and is recommended to define the probability of OS and EFS in patients with mastocytosis in daily practice. Disclosures Sperr: Amgen: Honoraria, Research Funding; Novartis: Honoraria. Gotlib:Incyte Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Valent:Amgen: Honoraria; Celgene: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Deciphera Pharmaceuticals: Research Funding; Ariad: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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. 124, No. 21 ( 2014-12-06), p. 3143-3143
    Abstract: The presence of BCR-ABL kinase domain mutations below the detection limit of sanger sequencing (low level mutations, LLM) predicts the outcome of subsequent therapy in patients with imatinib resistance (Parker et al. JCO 2011, Blood 2012). We have evaluated LLM in the context of the ENEST1st trial, which addresses the frequency of deep molecular response (MR4) after 18 months on nilotinib 300mg BID (NI) in newly diagnosed patients with chronic myeloid leukemia (CML) in chronic phase (CP). We have investigated the incidence of detectable LLM in the CD34+ progenitor cell compartment and correlated the mutation status (LLM vs. no LLM) with the primary endpoint of the ENEST1st trial, MR4at 18 months. Additionally, we evaluated MMR at 18 months. Seventy eight ENEST1st study patients (pts) provided 10ml of peripheral blood or 2ml bone marrow after written informed consent. CD34+ selection was carried out by MACS® (Miltenyi Biotec) and the CD34+purity was subsequently determined by fluorescent activated cell sorting (FACS). Aliquots of 105 CD34+ were used for RNA extraction, cDNA synthesis and BCR-ABL amplification followed by Ligation PCR (L-PCR) for mutations T315I, Y253H, E255K/V, and F359V. This method has previously been shown to achieve a dynamic detection range of 100% to 〈 0.1% mutant allele (3–3.5 log). No patient showed BCR-ABL kinase domain mutations detected by Sanger sequencing spanning ABL exons 4–9. Fifty one of 78 pts (65%) with 105 CD34+ cells and a documented CD34+ purity of 〉 50% were available for BCR-ABL amplification. Amplification was successful from 41 (52%) of these CD34+samples. A total of 205 L-PCR assays of CD34+ cells identified 29 (14%) mutations (T315Ix12, Y253Hx7, E255Kx8/Vx1 and F359Vx1) from 21/41 pts (51%). The median quantitative level of mutant alleles in CD34+ cells was 0.13 (range 0.06–0.54) BCR-ABLmutant/ BCR-ABLunmutated. One pt was taken off the study because of adverse events before the 3 months evaluation, 2 pts between 3 and 6 months and 1 after the 6 months evaluation (9.7%). Two pts were screened for the trial and did not fulfil the inclusion criteria (4.9%). One pt lost complete hematological response after 6 months and was switched to a different TKI and one pt had a persistent suboptimal response and switched TKI after 12 months on NI (4.9%). These two pts were considered as non-optimal responders at all time points after the change of treatment. All patients except one had typical transcripts (b2/a2 or b3/a2). One pt had an atypical transcript and was not available for MR4 assessment. At 18 months, 31 of 41 patients were evaluable for BCR-ABL levels. Thirteen out of 31 (42%) pts had MR4 or higher at 18 months (6 pts in the LLM group). Twenty six (84%) pts had MMR (10 in the LLM group), while five pts did not reach MMR at 18 months (4 pts with LLM). No statistically significant differences in the proportion of patients achieving MR4 or MMR could be detected between the two groups. In conclusion, T315I, Y253H, E255K or F359V mutations in CD34 selected cells below the detection limit of sanger sequencing do not predict the achievement of MR4 or higher in patients on nilotinib in the ENEST1st study. Therefore, screening for low level BCR-ABL mutations at diagnosis of CML is currently not recommended. Disclosures Giles: Novartis: Honoraria, Research Funding. Hochhaus:Novartis: Research Funding; BMS: Research Funding; MSD: Research Funding; Ariad: Research Funding; Pfizer: Research Funding. Frank:Novartis: Employment. Lange:Novartis: 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: 2014
    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. 126, No. 23 ( 2015-12-03), p. 2032-2032
    Abstract: Introduction: Outcomes of most acute myeloid leukemia (AML) patients (pts) remain poor. Enhanced risk-stratification and individualized treatment approaches are urgently needed. The Hedgehog (Hh) signaling pathway is important in embryonic development & stem cell biology & converges into activation of the transcription factor GLI1. In mice, loss of Gli1 increased the number of quiescent long-term hematopoietic stem cells with enhanced engraftment capacities & impaired myeloid development. Nevertheless, the biological role & prognostic impact of GLI1 expression in human AML remains to be fully elucidated. Non-myeloablative conditioning regimens (NMA) are increasingly used in AML pts undergoing hematopoietic stem cell transplantation (HCT) ineligible for conventional conditioning. In NMA-HCT the therapeutic approach is mainly based on an immunological graft-versus-leukemia (GvL) effect. Here, we tested whether the expression of GLI1 is associated with outcome in AML pts undergoing NMA-HCT. Patients & Methods: We analyzed 135 pts with diagnostic bone marrow (BM) available who were treated at our institution between January 2000 & June 2012. Median age at diagnosis was 64 years (y; range 38-75y). We included pts with de novo (n=84; 62.2%), secondary (n=37; 27.4%) or therapy-related (n=14; 10.4%) AML. All pts received NMA-HCT consisting of fludarabine (30mg/m² at days -4 to -2) & 2Gy total body irradiation (day 0). All pts received granulocyte colony stimulating factor (G-CSF)-mobilized peripheral blood stem cells on day 0. Donors were HLA-matched related (n=22; 16.3%) or HLA-matched (n=80; 59.3%) or mismatched (≥1 antigen; n=33; 24.4%) unrelated. Median follow-up was 4.3 y for pts alive. At diagnosis, cytogenetics were determined using standard techniques for banding & fluorescence in-situ hybridization in BM of all pts. The presence of FLT3 -ITD, FLT3 -TKD & expression status of EVI1, BAALC, ERG, MN1, mir-9 & mir-181a as well as the mutation status of the NPM1, CEBPA, IDH1, IDH2 & DNMT3A genes were determined. All mononuclear cells in pts' BM were assessed for presence of CD7, CD14, CD34, CD38, CD34+/CD38-, CD45, CD56, CD117, CD56 & glycophorin A. GLI1 expression was measured by a Taqman-probe based quantitative reverse transcription polymerase chain reaction & normalized to 18S as internal control. The third quartile of the normalized gene expression was used to define high & low GLI1 expressers. Results: At diagnosis, pts with high GLI1 expression had increased % of blasts in BM (p 〈 .01) & in peripheral blood by trend (p =.10) & were more likely to have de novo AML by trend (p =.06). Pts with high GLI1 expression were less likely to have a monosomal karyotype by trend (p =.07) & high GLI1 expression associated with higher % of CD14 (p =.05) & lower % of CD45 (p =.01) positive cells. High GLI1 expressers more frequently had IDH2 gene mutations (p =.01) & were lower EVI1 expressers (p =.05). High GLI1 expression was associated with significantly increased MN1 (p 〈 .01), BAALC (p =.02) & ERG (p 〈 .01) expression as well as lower expression of mir-9 (p =.01) & mir-181a (p 〈 .01). High GLI1 expressers also had a significantly longer overall survival (OS; p 〈 .01, Figure 1A) & longer event-free survival (EFS; p =.10; Figure 1B) by trend. In multivariable analysis a high GLI1 expression associated with longer OS (Hazard Ratio 0.42; 95% Confidence Interval 0.23-0.79; p 〈 .01). None of the analyzed clinical or biological parameters were significantly associated with EFS in multivariable analysis. Conclusion: High GLI1 expression associated with distinct clinical and biological characteristics. The observation that elevated GLI1 expression was linked to IDH2 mutations suggests that - similar to glioblastoma - IDH mutations may be associated with an activated hedgehog signaling pathway in AML. Moreover, we found that high GLI1 expression was an independent prognostic factor for longer OS in AML pts receiving NMA-HCT. An effect that may be mediated by a decreased number of quiescent long-term AML stem cells in high GLI1 expressers, which may be especially beneficial in NMA-HCT treated AML pts. Further mechanistic studies are needed to explore the observed results biologically & clinical validation studies are necessary to confirm our finding of an improved outcome of high GLI1 expressers in AML receiving NMA-HCT. Figure 1. Figure 1. Disclosures Franke: Novartis: Other: Travel Costs; MSD: Other: Travel Costs; BMS: Honoraria. Niederwieser:Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
    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 ...
  • 8
    In: Blood Advances, American Society of Hematology, Vol. 8, No. 11 ( 2024-06-11), p. 2890-2900
    Abstract: Certain laboratory abnormalities correlate with subvariants of systemic mastocytosis (SM) and are often prognostically relevant. To assess the diagnostic and prognostic value of individual serum chemistry parameters in SM, 2607 patients enrolled within the European Competence Network on Mastocytosis and 575 patients enrolled within the German Registry on Eosinophils and Mast Cells were analyzed. For screening and diagnosis of SM, tryptase was identified as the most specific serum parameter. For differentiation between indolent and advanced SM (AdvSM), the following serum parameters were most relevant: tryptase, alkaline phosphatase, β2-microglobulin, lactate dehydrogenase (LDH), albumin, vitamin B12, and C-reactive protein (P & lt; .001). With regard to subvariants of AdvSM, an elevated LDH of ≥260 U/L was associated with multilineage expansion (leukocytosis, r = 0.37, P & lt; .001; monocytosis, r = 0.26, P  & lt; .001) and the presence of an associated myeloid neoplasm (P & lt; .001), whereas tryptase levels were highest in mast cell leukemia (MCL) vs non-MCL (308μg/L vs 146μg/L, P = .003). Based on multivariable analysis, the hazard-risk weighted assignment of 1 point to LDH (hazard ratio [HR], 2.1; 95% confidence interval [CI] , 1.1-4.0; P = .018) and 1.5 points each to β2-microglobulin (HR, 2.7; 95% CI, 1.4-5.4; P = .004) and albumin (HR, 3.3; 95% CI, 1.7-6.5; P = .001) delineated a highly predictive 3-tier risk classification system (0 points, 8.1 years vs 1 point, 2.5 years; ≥1.5 points, 1.7 years; P & lt; .001). Moreover, serum chemistry parameters enabled further stratification of patients classified as having an International Prognostic Scoring System for Mastocytosis–AdvSM1/2 risk score (P = .027). In conclusion, serum chemistry profiling is a crucial tool in the clinical practice supporting diagnosis and prognostication of SM and its subvariants.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2024
    detail.hit.zdb_id: 2876449-3
    detail.hit.zdb_id: 2915908-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1575-1575
    Abstract: In acute myeloid leukemia (AML) high expression of the transcription factor ERG (ETS related gene) is associated with dismal outcome. The mechanisms that regulate differential ERG expression remain to be fully elucidated. MicroRNAs (miRs), small RNAs that are able to regulate gene expression, have emerged as important players in AML. We hypothesized that part of the differential expression of ERG is mediated by miRs. In silico prediction tools identified three putative miR-9 binding sites (BS) in the 3'-untranslated region (UTR) of ERG. First, we determined the expression levels of ERG & miR-9 in eight leukemia cell lines (i.e. KG1a, K562, THP-1, MV4-11, EOL1, NB4, OCI-AML3 & ME1) & found an inverse correlation between ERG & miR-9 expression (rank correlation -0.90). The cell line KG1a had the highest ERG & low miR-9 expression, and was therefore used for miR-9 overexpression experiments. In these cells miR-9 overexpression decreased ERG expression at the mRNA level to 82±7% (P=.079) & at the protein level to 72±14% (P=.005) after 12 hours (h) compared to empty vector control transfected cells. Next, we tested the activity of the three putative miR-9 BS in the 3'-UTR of ERG using luciferase assays. 12 h after cotransfection of HEK-293T cells with a miR-9 overexpression vector & an appropriate luciferase vector containing two of the putative BS (BS1 & BS2) from the 3'-UTR of ERG, we found the luciferase activity reduced to 52±4% (P=.023). Mutation experiments showed BS1, but not BS2 to be essential for this activity. The insertion of BS3 into the luciferase vector had no effect on reporter gene expression. Thus miR-9 most likely regulates expression of the transcription factor ERG by directly binding to BS1 in its 3'-UTR. To test if a differential expression of miR-9 is also of functional significance in AML, we first analyzed its impact on cell proliferation. Overexpression of miR-9 led to decreased proliferation rates in KG1a cells compared to control vector treated cells. After 5 days, the relative cell count was 133±21% vs. 241±67% in the miR-9 overexpressing cells compared to empty vector expressing cells, respectively. Next, we determined if this difference was based on a higher apoptosis rate. An Annexin V staining revealed no significant difference between the apoptotic threshold of miR-9 overexpressing (21%) and empty vector cells (21%) after 24 h. However, a subsequent cell cycle analysis demonstrated a higher percentage of miR-9 overexpressing cells in the G2/M phase, (39±2%) compared to the empty vector control treated cells (31±3%) after 24 h (P=.084), indicating that the cell cycle is slowed or stopped during cell division. Since miR-9 targets the poor prognostic marker ERG & higher miR-9 expression led to decreased proliferation & reduced cycling in AML cells in vitro we speculated that the differential miR-9 expression would also impact the outcome of AML patients (pts). Mature miR-9 is derived from three precursor molecules of which pre-miR-9-1 & pre-miR-9-3 are known to be expressed in hematopoietic cells. We assessed the pre-miR-9-1 expression of bone marrow by real-time PCR in 131 AML pts (median age 64 [range 22 – 75] years) with favorable (n=4, 3%), intermediate (n=90, 69%), adverse (n=33, 25%), or unknown (n=4, 3%) cytogenetic risk (according to the Medical Research Council [MRC] Classification) who received a RIC-HCT. The median follow-up was 4 years. The pre-miR-9-1 expression levels were normalized to ABL to define high & low pre-miR-9-1 expressers by the median expression of all AML pts. At diagnosis, high pre-miR-9-1 expresser status associated with a lower white blood count (P=.065) and lower % of peripheral blasts (P=.108) by trend. Furthermore, pts with high pre-miR-9-1 expression were more likely to be NPM1 wild-type (P=.047) & FLT3-ITD negative (P=.020). Pts with high pre-miR-9-1 had a lower probability of relapse (P=.048). In conclusion, miR-9 targets & regulates expression of the poor outcome predictor ERG. Overexpression of miR-9 led to decreased proliferation and a pause in AML cell cycling. Furthermore, high pre-miR-9-1 expression associated with reduced relapse rates in AML. Thus a pharmacologically induced expression of miR-9 in AML blasts may improve outcomes of AML pts. 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: 2014
    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. 128, No. 22 ( 2016-12-02), p. 517-517
    Abstract: Allogeneic hematopoietic cell transplantation (HSCT) is a powerful consolidation option for acute myeloid leukemia (AML) patients (pts) in hematologic complete remission (CR). Disease recurrence after HSCT remains a major clinical problem & early identification of AML pts at risk of relapse is crucial to improve outcomes. High expression of the AML associated gene BAALC (Brain and acute leukemia, cytoplasmic) at diagnosis adversely impacts on outcomes in AML pts. Little is known about its prognostic capacity during disease course & as a marker of residual disease. Here we adopted digital droplet polymerase chain reaction (ddPCR) for absolute quantification of BAALC copy numbers in peripheral blood (PB) prior to HSCT in AML pts in hematologic CR. We identified 82 AML pts with PB in first (60%) or second CR (23%) or CRi (17%) up to 28 days prior to HSCT available. Median age at HSCT was 63.9 (range 50.8-76.2) years (y). All pts received non-myeloablative (NMA) conditioning (fludarabine 3x30 mg & 2 Gy total body irradiation). At diagnosis, mutation status (mut) of the NPM1, CEBPA, IDH1, IDH2, & DNMT3A gene & presence of FLT3-ITD or FLT3-TKD were assessed. In pre-HSCT PB, absolute quantification of BAALC copy numbers was performed by ddPCR & results were normalized to ABL1 copy numbers.Additionally, absolute BAALC copy numbers wereassessedin PB of healthy controls (n=7) with a median age of 62.7 (range 39.6-82.0) y. Pts were grouped according to the European LeukemiaNet (ELN) classification in 21% favorable, 23% intermediate-I, 24% intermediate-II, 23% adverse & 9% unknown. Pts & healthy control were evenly matched in age (P=1) & sex (P=1). BAALC/ABL1 copy numbers did not differ between AML pts at HSCT (median 0.03 [range 0.01-2.48]) & the healthy controls (median 0.04 [range 0.03-0.10], P=.34, Figure 1). A cut-off point of 0.14absolute BAALC/ABL1 copies was determined using the R package 'OptimalCutpoints' & used to define pts with high (26%) & low (74%) pre-HSCT BAALC/ABL1 copy numbers. The copy number at this cut-off point was higher than the two-fold standard deviation over the median of the healthy controls (0.10 BAALC/ABL1). Pts with high & low pre-HSCT BAALC/ABL1 copy numbers did not differ significantly in pre-treatment characteristics (i.e. hemoglobin, white blood count, platelets, blasts in bone marrow or PB, ELN genetic group, FLT3-ITD, FLT3-TKD, NPM1, CEBPA, DNMT3A, IDH1 or IDH2 mut) or remission status at HSCT (CR1 vs. CR2 vs. CRi). However, pts with high pre-HSCT BAALC/ABL1 copy numbers had a significantly higher cumulative incidence of relapse (CIR, P=.02, Figure 2a) & shorter overall survival (OS, P=.02, Figure 2b). High pre-HSCT BAALC/ABL1 copy numbers especially impacted on CIR when we restricted our analysis to pts with normal cytogenetics (P=.003). In multivariate analysis for the entire cohort, high pre-HSCT BAALC/ABL1 copy numbers retained the prognostic impact on CIR (Hazard Ratio [HR] 3.6, Confidence Interval [CI] 1.6-8.2, P=.002) after adjustment for disease status at HSCT (P=.006) & the prognostic impact on OS (HR 2.2, CI 1.1-4.3, P=.02). In conclusion, ddPCR is a feasible method for absolute quantification of BAALC copy numbers in PB, which may indicate residual disease burden in AML pts. High PB BAALC/ABL1 copy numbers ( 〉 0.14) in AML pts in hematologic CR at HSCT associated with higher CIR & shorter OS in univariate & multivariate models. AML pts with high PB BAALC/ABL1 copy numbers at HSCT should be closely monitored for relapse in the post-transplant period. In the future prospective studies will be required to validate the absolute PB BAALC/ABL1 copy number cut-off point & to evaluate whether AML pts with high BAALC/ABL1 copy numbersmight benefit from additional treatment before HSCT. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures Poenisch: Mundipharma: Research Funding. Niederwieser:Amgen: Speakers Bureau; Novartis Oncology Europe: Research Funding, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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...