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. 118, No. 21 ( 2011-11-18), p. 425-425
    Abstract: Abstract 425 The introduction of all-trans-retinoic acid (ATRA) in front line therapy of acute promyelocytic leukemia (APL) has improved the outcome of all age groups. In the elderly patients (pts), multi-morbidity and higher vulnerability to chemotherapy-related toxicity are the main problems reducing the chance of cure. This has led to recommendations to reduce the intensity of therapy in elderly APL pts. We report on the long-term outcome of pts with newly diagnosed APL registered in two prospective studies of the German AML Cooperative Group (AMLCG) from December 1994 until June 2011. The therapy consisted of ATRA and anthracycline/ara-C-based induction and consolidation therapy (TAD/HAM–TAD) followed by maintenance therapy as reported previously in younger APL pts (Lengfelder et al. Leukemia 2009;23:2248–2258). In pts ≥60 years (y), the administration of the second induction cycle (HAM with an age adapted cumulative ara-C dose of 6g/sqm) was at the discretion of the treating physician. After December 2005, the pts were included in the ongoing APL protocol and randomized between the AMLCG strategy and the protocols of the Spanish PETHEMA. Among 295 adult pts with newly diagnosed APL, 83 pts (28%) were ≥60y of age. Seventeen elderly pts (20%) were not enrolled in the study, due to death before start of therapy, contraindications against chemotherapy or concomitant other malignancy. Eleven pts randomized in the PETHEMA arm were excluded from the present analysis to cover homogeneity of therapy. In 53 of 55 pts treated according to the AMLCG protocol, results are available. Median age was 67 y (range 60 to 83); 58% were male, 42% female; 68% had low/intermediate and 32% high risk according to Sanz score. Morphology was FAB M3 in 62%, M3v in 38%. Cytogenetics showed t(15;17) alone in 52%, and combination with other abnormalities in 48% of pts. The bcr1/bcr2 transcript of PML/RARA was found in 41% and the bcr3 transcript in 59% of pts. Forty-four pts (83%) achieved complete remission (CR). Early death (ED) occurred in 9 pts (17%). Median time to ED was 12 days (range 2 to 19) after start of therapy. Causes of ED were bleeding, multi-organ failure and sepsis. Manifest APL differentiation syndrome occurred in 25% of pts and WHO grade '3 bleeding, fever/infection or cardiac failure in 8%, 43% and 17% of pts, respectively. After consolidation therapy, 96% of pts were in molecular remission. After a median follow up of 5.3 y (1 day to 12.8 y), the 6-year overall (OS), event free (EFS) and relapse free survival (RFS) and the cumulative incidence of relapse (CIR) were 45%, 41%, 50% and 26%, respectively. The outcome was further analyzed according to risk group, number of induction cycles, and age ≥60y to 69y and ≥70y. Pts with pretreatment white blood cell (WBC) count 〈 10 × 109/L (low/intermediate risk; n=36) had a significantly superior outcome compared to pts with high WBC counts (high risk; n=17) resulting in a CR rate of 92% vs. 65% and ED rate of 8% vs. 35%, respectively (p=0.02). The 6-year OS, EFS, RFS and CIR of the low/intermediate risk pts was 56%, 53%, 60% and 14%, respectively, compared to 25%, 15%, 23% and 58% in high risk pts (p=0.006, p=0.0004; p=0.008; p 〈 0.01). All 12 pts, who had received two induction cycles achieved CR, and no relapse occurred so far resulting in a significantly superior outcome compared to patients, who had received only one induction cycle (OS: p=0.007; EFS: p=0.0002; RFS: p=0.01). In the pts '70y (n=17), 71% entered CR and 29% died from ED. Separated according to low and high WBC counts, the CR rate of this group was 83% vs. 40% and the ED rate 17% vs. 60% (p=0.07), respectively, resulting in an inferior OS (p=0.08) of the pts with high WBC count. Conclusions: Among our elderly pts, we found a high proportion of pts with high risk profile and a high rate of exclusion from the study due to death before the start of therapy or poor condition. In patients included in our study, high WBC count and advanced age are associated with a high risk of ED. The inferior outcome in pts, who received only one induction cycle, suggests that intensification of chemotherapy should be considered in elderly APL pts, if possible. The incorporation of arsenic trioxide might be an alternative, which could be investigated in future trials. 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 101, No. 1 ( 2003-01-01), p. 64-70
    Abstract: Risk assessment in acute myeloid leukemia (AML) using pretreatment characteristics may be improved by incorporating parameters of early response to therapy. In the 1992 trial of the German AML Cooperative Group (AMLCG), the amount of residual leukemic blasts in bone marrow was assessed one week after the first induction course (day 16 blasts). A total of 449 patients 16 to 76 years of age (median, 53 years) with de novo AML entered the trial and were evaluable. Treatment included TAD/HAM (thioguanine, cytosine arabinoside, and daunorubicin/high-dose cytosine arabinoside and mitoxantrone) double induction, TAD consolidation, and randomly either maintenance therapy or S-HAM consolidation. Cytogenetics were favorable, intermediate, unfavorable and not available in 10.0%, 48.3%, 13.1%, and 28.5%, respectively. Day 16 blasts ranged from 0% to 100% (median, 5%, mean ± SD, 18.6 ± 28.5%). Complete remission (CR) rate was 72.6%, 17.6% had persistent leukemia (PL), and 9.8% succumbed to hypoplastic death. Median overall survival (OS), event-free survival (EFS), and relapse-free survival (RFS) were 18, 9, and 15 months with 28.4%, 21.6%, and 30.1% at 5 years, respectively. As a continuous variable, day 16 blasts were related to CR rate (P  & lt; 0.0001), PL rate (P  & lt; 0.0001), OS (P  & lt; 0.0001), EFS (P  & lt; 0.0001), and RFS (P = 0.0049). Multivariate analyses identified the following parameters to be associated with the respective end points. CR rate: day 16 blasts (P  & lt; .0001), age (P = .0036), and LDH (P = .0072); OS: unfavorable cytogenetics (P  & lt; .0001), day 16 blasts (P  & lt; .0001), age (P  & lt; .0001), and LDH (P = .0040); EFS: unfavorable cytogenetics (P  & lt; .0001), LDH (P  & lt; .0001), day 16 blasts (P  & lt; .0001), and age (P = .0061); RFS: unfavorable cytogenetics (P  & lt; .0001), LDH (P  & lt; .0001), and day 16 blasts (P = .0359). The prognostic significance of day 16 blasts is independent of pretherapeutic parameters and predicts outcome even in patients achieving a CR.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2003
    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. 128, No. 22 ( 2016-12-02), p. 1066-1066
    Abstract: Despite recent advances, treatment of elderly patients with AML remains a challenge because of adverse disease biology, comorbidities and therapy related toxicities. The balance between effectivity and toxicity of treatment strategies play a key role. Since comparative studies are lacking, a prospective randomized trial was designed among German AML study groups with different treatment strategies to compare outcome. Patients ≥60 years with all AML subtypes except M3 were randomized up-front to a common standard arm (CSA) (10%) and to study specific arms (90%) of the AMLCG or the OSHO. The CSA consisted of one or two inductions of araC 100 mg/m2/d continuous IV (CI) d 1-7 d and daunorubicin (dauno) 60 mg/m2/d IV d 3, 4, 5 and two courses of araC 1 g/m2/d BID IV d 1, 3 and 5 as consolidation (Mayer RJ et al, NEJM 1994). The AMLCG study arm randomized TAD (araC 100 mg/m2/d CI d1-2 followed by BID d 3-8, dauno 60 mg/m2/d IV d 3-5 and 6-thioguanine 100 mg/m2/d po BID d 3-9) and HAM [araC 1 mg/m2/d IV BID d 1-3 and mitoxantrone (mito) 10 mg/m2/d IV d 3-5] versus two courses of HAM with any 2nd course only given if blasts persisted ± G-CSF. Two courses of TAD were given as consolidation followed by maintenance chemotherapy over three years. The OSHO study arm included araC 1 g/m²/d BID IV d 1 + 3 + 5 + 7 and mito 10 mg/m2/d IV d 1 - 3 for one or two induction courses and ara-C 500 mg/m² BID 1h IV d 1 + 3 + 5 in combination with mito10 mg/m2/d IV d 1 + 2 as consolidation. Pegfilgrastim 6 mg s.c. was applied on day 10 of induction and on d 8 of consolidation. The study was approved by the IRB and registered at clinicaltrials.gov (NCT01497002 and NCT00266136). Written informed consent was obtained from all patients prior to randomization. Between April 1st, 2005 and May 26th, 2015 1286 patients were assigned randomly to the CSA (n=132) or to the study groups arm (n=1154). After excluding 139 patients (10.8%), 1147 patients were eligible for analysis, 1120 with follow-up for overall survival (OS) and 1079 for complete remission (CR) analysis. Baseline characteristics of all eligible patients showed median ages of 68 (60-82) years for the CSA and 69 (60-87) and 70 (60-85) years in the study arms A and B, respectively (p=0.05). Proportions of patients with secondary AML differed significantly between study arms (A: 42%, B: 30%, CSA: 36%; p=0.003). The CSA had less flt3 wildtype/npm1 wildtype patients (31%) vs. arm A (51% p=0.040) and arm B (58%, p=0.0455). No differences were observed with respect to cytogenetic risk groups, white blood cell counts, LDH, and npm1 mutant/ flt3-wildtype or mutant. The primary endpoint event free survival (EFS) did not differ between the CSA and study group strategies. Three-year EFS was 12.4% (95% CI: 6.7 - 19.9%) in the CSA, 15.6% (95% CI: 13.1 - 18.3%) in group A and 11.4% (95% CI, 7.4% to 16.4%) in group B (n.s.;Fig.1). With a median follow-up of 67 months, OS did not differ significantly between CSA and study group regimens. The 3-year survival probability was 22.3% (95% CI: 14.7-30.9%) in the CSA, 24.7% (95% CI: 21.6-27.9%) in group A and 22.4% (95% CI, 16.7% - 18.3%) in group B (Fig.2). CR status after 90 days of therapy was evaluated as secondary endpoint. The proportion of patients in CR in the CSA [51% (95% CI: 42-61%)] was comparable to the 50% (95% CI: 47-54%) and 48% (95% CI: 41-55%) of the study group arms (p=n.s.). Persistent leukemia was seen in 16% (95% CI: 10-24%) in the CSA vs 17% (95% CI: 14-19%) and 12% (95% CI: 8-17%) in groups A and B, respectively (both p= n.s.). A total of 226 patients died within 90 days of treatment, 24% (95% CI: 17-33%) in CSA, 19% in group A (95% CI: 16-22%) and 27% (95% CI: 21-33%) in group B; CSA vs A p=0.1859, CSA vs B p=0.5902). Death without AML was 3% in CSA, 2% in group A and 3% in group B, death with AML was 9% in CSA, 6% in group A and 5% in group B and death from indeterminate cause was 12% in CSA, 11% in group A and 20% in group B. Three-year relapse free survival (RFS) was 21.3% (95% CI: 12.2 - 31.0) in the CSA, 28.9% (95% CI: 24.9 -33.0%) in group A and 24.0% (95% CI: 16.8 - 31.9) in group B (both p=n.s.; Fig.3). In multivariate analysis independent variables for EFS and OS were age, type of disease, cytogenetic group and WBC count, but not the allocation to one of the treatment arms. Age and cytogenetic group were determinants for RFS. Conclusion A strictly prospective comparison of different treatment strategies in patients with AML did not show clinically relevant outcome differences when compared to a common standard arm. Figure 1. Figure 1. Figure 2. Figure 2. Figure 3. Figure 3. Disclosures Niederwieser: Novartis Oncology Europe: Research Funding, Speakers Bureau; Amgen: Speakers Bureau. Hoffmann:Novartis Oncology Europe: Research Funding. Al-Ali:Celgene: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding. Hegenbart:Pfizer: Other: Travel grant; Janssen: Honoraria, Other: Travel grant. Sayer:Riemser Pharma: Consultancy. Hochhaus:BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; ARIAD: Honoraria, Research Funding. Fischer:Novartis: Consultancy, Honoraria. Dreger:Novartis: Consultancy; Janssen: Consultancy; Gilead: Consultancy; Gilead: Speakers Bureau; Roche: Consultancy; Novartis: Speakers Bureau. Hiddemann:Roche: Membership on an entity's Board of Directors or advisory committees; Genentech: Other: Grants; Roche: Other: Grants.
    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 ...
  • 4
    In: Journal of the American Medical Directors Association, Elsevier BV, Vol. 17, No. 5 ( 2016-05), p. 465.e9-465.e15
    Type of Medium: Online Resource
    ISSN: 1525-8610
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of the American Medical Directors Association, Elsevier BV, Vol. 18, No. 11 ( 2017-11), p. 967-973.e1
    Type of Medium: Online Resource
    ISSN: 1525-8610
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Leukemia, Springer Science and Business Media LLC, Vol. 33, No. 12 ( 2019-12), p. 2830-2841
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Applied Social Psychology, Wiley, Vol. 51, No. 10 ( 2021-10), p. 987-998
    Abstract: To learn more about why people falsely confess without external pressure, we examined voluntary blame‐taking in three experiments. Drawing from theories of prosocial behavior and social identity, we investigated whether participants' blame‐taking is influenced by (a) their relationship with the guilty person (Experiment 1) and (b) the group membership of the person asking to take the blame (Experiments 2a and 2b). In Experiment 1, participants ( N  = 130) considered whether they would take the blame for a small traffic violation for their best friend and a stranger in a vignette‐scenario. As expected, intended blame‐taking rates were higher for their best friend (60.8%) than for a stranger (8.5%). Reported reasons for taking the blame included reciprocity and empathy. In Experiments 2a and 2b ( Ns  = 60; 89), we tested actual blame‐taking behavior in two field experiments, using a new experimental paradigm. An experimenter approached participants and asked them to commit insurance fraud for a broken camera. Participants who shared the same group as the person in need were more willing to take the blame (47%‐48%) than participants who were from a different group (21%‐23%). The most frequently listed reason for taking the blame was empathy. Implications for the occurrence of voluntary blame‐taking behavior to protect someone else are discussed.
    Type of Medium: Online Resource
    ISSN: 0021-9029 , 1559-1816
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 2066531-3
    SSG: 5,2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: PLoS ONE, Public Library of Science (PLoS), Vol. 7, No. 10 ( 2012-10-23), p. e47185-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2012
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2840-2840
    Abstract: The Armadillo repeat-containing X-linked protein 1 (ARMCX1)gene encodes a member of the ALEX family of proteins and is located on the X chromosome. We found this gene aberrantly expressed in acute myeloid leukemia (AML) cells. We analyzed ARMCX1 expression in a cohort of 508 AML patients who received intensive chemotherapy within AMLCG clinical trial. Expression levels of ARMCX1 were determined in pretreatment bone marrow or peripheral blood samples by using oligonucleotide microarrays (GSE37642). High transcript levels (dichotomized at the median expression) of ARMCX1 were predictors for inferior overall survival (OS) (p=0.0027, Log-Rank test) and a lower rate of complete remission (CR) (p=0.0017). Older patients (≥ 60 years) showed an elevated incidence of high ARMCX1 expression compared to younger patients ( 〈 60 years) (56% vs. 44%, p=0.008, Chi-Squared test). In the subgroup analyses we found that for younger AML patients ( 〈 60 years) ARMCX1 expression was not predictive for survival (OS, PFS, EFS) and CR. However, in older patients (≥ 60 years) OS (p=0.002, median survival 11 vs 8 months), event free survival (EFS) (p=0.0006, median 6 vs 2 months) and progression free survival (PFS) (p=0.013, median 10 vs 7 months) were significantly lower among patients with high ARMCX1 expression. CR rate of elderly AML patients with low ARMCX1 expression levels was 60% vs. 41% with high levels (P=0.003). Older AML patients with low ARMCX1 expression had a 5-year OS of 20% (vs. 7% for patients with high expression). Furthermore, prognostic significance of high ARMCX1 expression was confirmed in an independent AML patient cohort of 293 AML patients from the HOVON group (GSE6891). Both, OS (p 〈 0.001) and EFS (p 〈 0.001) were significantly inferior for patients with high ARMCX1expression in this cohort. Multivariate logistic regression did not show any association between ARMCX1 expression and sex, lactatdehydrogenase (LDH) or leukocyte count (WBC). Interestingly, high expression of ARMCX1 was strongly associated with high United Kingdom Medical Research Council (MRC) risk classification (p 〈 0.0001). Evaluation of ARMCX1 expression against a clinical multivariate Cox-Regression model (age, gender) showed that dichotomized ARMCX1 added a significant amount of information to the model and was a risk factor for OS (p=0.0020, HR= 1.377 (95%-CI 1.124, 1.686)). Strikingly, including MRC risk classification into the multivariate Cox-Regression model high ARMCX1 expression remained a significant risk factor for OS (p=0.0413, HR=1.243 (95%-CI 1.009, 1.531)). Taken together, we analyzed ARMCX1 expression levels in a large AML patient cohort. In this study, high ARMCX1 expression levels were identified as a negative prognostic factor for AML patients with respect to survival and remission. Disclosures Hiddemann: Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Research Funding. Stelljes:Pfizer: Consultancy.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1682-1682
    Abstract: Introduction In Acute Myeloid Leukemia (AML) internal tandem duplications (ITD) in the fms-related tyrosine kinase 3 (FLT3) are a frequent event associated with an unfavorable prognosis. At diagnosis, the FLT3-ITD status is routinely assessed by fragment analysis of PCR-amplified cDNA. However, this assay only provides information on the length but not on the position and sequence of the insertion. Therefore, it is attractive to overcome this limitation by the use of high-throughput amplicon sequencing (HTAS) as an alternative strategy for FLT3-ITD detection. To proof the feasibility and accuracy of this approach we performed HTAS on 260 AML patients, of which 250 were FLT3-ITD positive according to routine diagnostics. Patients and Methods All samples were obtained from patients treated on the German Acute Myeloid Leukemia Cooperative Group (AMLCG) trials 1999, 2004 and 2008. All patients received intensive induction therapy with curative intent. At diagnosis, patients had a median age of 60 years (range, 18-80 years). Additional molecular marker were screened routinely: NPM1 mutation (62%), KMT2A-PTD (8%), CEBPA mutations (8%). According to the ELN-classification patients clustered into the following groups: ELN intermediate I (70%), intermediate II (21%) or adverse (9%). A normal karyotype was observed in 71%, while 29% had an aberrant karyotpye. All patient samples were analyzed routinely by FLT3 fragment analysis. For HTAS of FLT3 we used custom FLT3 cDNA primers including barcode and adapter-sequences enabling a one-step PCR-protocol. Sequencing was performed (2x250bp paired end) on a MiSeq (Illumina). Per run, up to 96 samples were sequenced, yielding a median of 79,110 reads per amplicon (range: 31,996 - 259,783). As controls, cDNA from FLT3-ITD positive (Molm-13) and negative (HL60) cell lines were used. Sequencing data were aligned to the FLT3 cDNA reference (NM_004119.2) and FLT3-ITDs were called using Pindel software (version 0.2.5a7). Results Based on the HTAS results obtained from the FLT3 wild-type control HL60, we set the cut-off for ITD detection at a variant allele frequency (VAF) of 0.5%. Using this threshold, all patients which were assessed to be FLT3-ITD negative in diagnostic routine, were also negative in HTAS. FLT3-ITDs were detected by HTAS in 242 out of 250 (97%) patients who were FLT3-ITD positive patients according to routine fragment analysis. For six out of the 8 remaining patients, no valid ITD was detected by HTAS possibly due to one of the following reasons: a low mutational burden resulting in a VAF below the cut-off level (n=4) or a deletion near the ITD (n=2) potentially interfering with ITD-detection. In total, 308 ITDs were detected in 242 patients by HTAS while in these patients 282 ITDs were detected by routine diagnostics. Of note, HTAS missed 13 subclonal ITDs reported in routine, while 39 additional subclonal ITDs were detected by HTAS only. Overall, HTAS detected a higher number of ITDs per patient (mean: 1.27; range: 1-4) compared to fragment analysis (mean: 1.17; range: 1-3). Patients with more than one ITD according to HTAS showed a trend towards shorter overall and relapse free survival (p=0.105, p=0.104 respectively; Figure 1A). The ITD position (i.e. affected FLT3 domain) based on HTAS did not impact on clinical outcome. There was a significant correlation between the FLT3-ITD levels detected by fragment analysis and HTAS (Pearson, R=0.801, p=0.01; Figure 1B). High FLT3-ITD levels measured by fragment analysis had a significant impact on RFS, whereas this effect was not observed for FLT3-ITD levels measured by HTAS. The quantification of FLT3-ITD by HTAS might be hampered by underestimation of the VAFs of long ITDs that are less likely to be mapped correctly compared to shorter ITDs. Conclusion In summary, our study demonstrates the feasibility of HTAS for FLT3-ITD detection in AML. In particular, the identification of subclonal ITDs with high sensitivity provides additional information with potential prognostic value. Thus, HTAS may serve as a robust tool that could be implemented in future diagnostic routines. However, bioinformatic algorithms for ITD detection may need further improvement, e.g. to optimize ITD quantification and to facilitate the detection of ITDs in combination with deletions. Disclosures Hiddemann: Roche: Other: Grants; Genentech: Other: Grants; Roche: Membership on an entity's Board of Directors or advisory committees.
    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...