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: American Journal of Hematology, Wiley, Vol. 89, No. 6 ( 2014-06), p. 610-615
    Abstract: Although acute myeloid leukemia (AML) with t (8;21) belongs to the favorable risk AML subset, relapse incidence may reach 30% in those patients. RUNX1‐RUNX1T1 fusion transcript is a well‐established marker for minimal residual disease (MRD) monitoring. In this study, we investigated the feasibility and performances of RUNX1‐RUNX1T1 DNA as MRD marker in AML with t (8;21). In 17/22 patients with t (8;21)‐positive AML treated in the French CBF‐2006 trial, breakpoints in RUNX1 and RUNX1T1 were identified using long‐range PCR followed by next‐generation sequencing. RUNX1‐RUNX1T1 DNA quantification was performed by real‐time quantitative PCR using patient‐specific primers and probe. MRD levels were evaluated in 71 follow‐up samples from 16 patients, with a median of four samples [range 2–7] per patient. RUNX1 breakpoints were located in intron 5 in all cases. RUNX1T1 breakpoints were located in intron 1b in 15 cases and in intron 1a in two cases. RUNX1‐RUNX1T1 MRD levels measured on DNA and RNA were strongly correlated ( r  = 0.8, P   〈  0.0001). Discordant MRD results were observed in 10/71 (14%) of the samples: in three samples from two patients who relapsed, RUNX1‐RUNX1T1 was detectable only on DNA, while RUNX1‐RUNX1T1 was detectable only on RNA in seven samples. MRD monitoring on genomic DNA is feasible, but with sensitivity variations depending on the patient breakpoint sequence and the qPCR assay efficiency. Although interpretation of the results is easier because it is closely related to the number of leukemic cells, this method greatly increases time, cost and complexity, which limits its interest in routine practice. Am. J. Hematol. 89:610–615, 2014. © 2014 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 0361-8609 , 1096-8652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 1492749-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood Advances, American Society of Hematology, Vol. 5, No. 23 ( 2021-12-14), p. 5258-5268
    Abstract: WT1 overexpression is frequently identified in acute myeloid leukemia (AML) and has been reported to be a potential marker for monitoring measurable residual disease (MRD). We evaluated the use of postinduction WT1 MRD level as a prognostic factor, as well as the interaction between postinduction WT1 MRD response and the effect of allogeneic stem cell transplantation (allo-SCT) in the first complete remission (CR). In the ALFA-0702 trial, patients with AML, aged 18 to 59, had a prospective quantification of WT1 MRD. The occurrence of a WT1 MRD ratio & gt;2.5% in bone marrow or & gt;0.5% in peripheral blood was defined as MRDhigh, and ratios below these thresholds were defined as MRDlow. The prognostic value of MRD after induction chemotherapy was assessed in 314 patients in first CR by comparing the risk of relapse, the relapse-free survival (RFS), and the overall survival (OS). Interaction between MRD response and the allo-SCT effect was evaluated in patients by comparing the influence of allo-SCT on the outcomes of patients with MRDhigh with those with MRDlow. The results showed that patients with MRDhigh after induction had a higher risk of relapse and a shorter RFS and OS. The MRD response remained of strong prognostic value in the subset of 225 patients with intermediate-/unfavorable-risk AML who were eligible for allo-SCT, because patients with MRDhigh had a significantly higher risk of relapse resulting in worse RFS and OS. The effect of allo-SCT was higher in patients with MRDlow than in those with MRDhigh, but not significantly different. The early WT1 MRD response highlights a population of high-risk patients in need of additional therapy.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 127, No. 20 ( 2016-05-19), p. 2451-2459
    Abstract: Recurrent mutations in chromatin modifiers and cohesin were observed in t(8;21) AML, but not inv(16) AML. t(8;21) AML patients with mutations in kinase signaling plus chromatin modifiers or cohesin members had the highest risk of relapse.
    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: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2327-2327
    Abstract: Background: Acute myeloid leukemia (AML) is a heterogeneous disease even within the same genetic subgroup, e.g. within the NPM1 mutated subgroup, some patients have additional mutations in FLT3, IDH1/2, DNMT3A or TET2. Recent reports have shown that minimal residual disease (MRD) in AML, during or after treatment has prognostic impact. However, the clinical assessment of MRD in AML faces many questions. Firstly, which of the potential molecular and/or cellular markers should be included? Secondly, what type of biological sample should be analyzed? Thirdly, what is the sensitivity threshold to set and what are the relevant time frames for the MRD evaluation. The current technology development next-generation sequencing (NGS), opens new perspectives for monitoring MRD in AML, with the opportunity to expand and multiplex various markers. In this context, we tested the NGS technology for MRD monitoring IDH1/2 and DNMT3A in AML cases with mutated NPM1. We validated our results against MRD evaluated by RTqPCR. Patients and methods: In this study, we included 94 patients (pts) from the ALFA 0701 trial. NPM1 monitoring by RTqPCR was performed as previously described (Lambert et al, Oncotarget july 2014). IDH1/2 and DNMT3A monitoring was done by NGS using the Ion Torrent Proton instrument. In order to obtain very high coverage (approximately 2 Millions reads by sample), 24 samples were analyzed per run. Bioinformatic analysis was performed according to our previous work (Boyer et al, Am JHematol 2013). Results. 94 samples from 31 NPM1 mutated pts were analyzed. (17pts were positive for IDH1/2 and 15 were positive for DNMT3A). Sequencing data showed an excellent depth with a median of 2012459 reads (range 102,657 to 5,160,118 reads) and 966,298 reads (range 565,152 to 2,700,349 reads) for IDH1/2 and DNMT3A, respectively. Unfortunately, despite of this excellent coverage, a median of 520 reads were found positive in the negative controls due to a multi steps cross contaminations (OT2, Clonal amplification, purification). Thus, limiting the sensitivity to 0.07% (0.001-0.097%, p 〈 0.001, Fisher’s exact test) and 0.1% (0.011-0.426%, p 〈 0.0001, Fisher’s exact test) for IDH1/2 and DNMT3A, respectively. The correlation with NPM1 MRD was 78% (r = 0.68183, p 〈 0.0001) and 78.6% (r = 0.55514, p 〈 0.0001) for IDH1/2 and DNMT3A, respectively. For the 17 IDH1/2 positive patients, we found concordant MRD results between IDH1/2 mutation and NPM1 RTqPCR in 13 cases: (4 pos/pos), which was associated to disease relapse, (9 neg/neg), which was associated to maintenance of CR1. For the four remaining patients, we have observed a discrepancy between some NPM1 results and IDH1/2 results. Indeed, these 4 patients presented one or more MRD negative points using the NPM1 marker, while rates of the IDH1/2 marker ranged from 0.1% to 47%. Three of them, have relapsed after 504,395,158 days, and all of them with the same NPM1 mutation detected at diagnosis. The one patient, who has not relapsed, did evolve to a myelodysplastic syndrome which was NPM1 negative. For the 15 DNMT3A positive patients, we found co-occurrence of the NPM1 mutation and IDH1/2 mutations in 9 cases (8 pos/pos), which was associated to disease relapse and we found concordant MRD results between DNMT3A mutation and NPM1 RTqPCR, (1 neg/neg), which was associated to maintenance of CR1. For the six remaining patients, we have observed a discrepancy between NPM1 results and DNMT3A results. Indeed, those 6 patients presented a great majority of negative results using MRD monitoring by NPM1, while rates of MRD by DNMT3A ranged from 5% to 45%. All of these 6 pts are still in first CR after a median follow up of 4 years. Bone marrow and PBMC were analyzed for 3 of them. A positive result was observed in all cell fractions except for the T cells compartment. Conclusion. Monitoring of MRD by NGS mutation detection in two genes IDH1/2 is feasible and valuable, because it enabled us to predict relapse in additional patients, with an area under the curve of 0.7971 (95% CI: 0.6693 - 0.9250), and 100% if we include the patient who evolved to MDS. Furthermore, this category of patients may benefit from new targeted therapies such as inhibitors of mutated IDH proteins. On the contrary, DNMT3A is not a good marker for MRD monitoring, because we detected persistence of a preleukemic clone in 40% of patients remaining in CR, reflecting the molecular heterogeneity clonal hematopoiesis. 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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1062-1062
    Abstract: Introduction.The Ecotropic viral integration site 1 (EVI1) gene is located on chromosome band 3q26.2, and is composed of 16 exons. Upon diagnosis, EVI1 overexpression has been implicated in 10% of patients with acute myeloid leukemia (AML) and confers a poor prognosis. It occurs with or without chromosomal rearrangements involving 3q26.2 and is often associated with adverse cytogenetic abnormalities, but can also be found in normal karyotypes. At this point, expression of the EVI1 mRNA relative to the housekeeping gene Abelson (ABL) is determined by real time quantitative PCR (RQ-PCR) with ΔCt method. Although this technique gives good results upon diagnosis, it is not powerful enough to monitor patients in minimal residual disease (MRD). We therefore decided to develop a technique of absolute quantification of EVI1 expression in RQ-PCR with plasmid synthesis in order to determine overexpression thresholds in bone marrow (BM) and peripheral blood (PB), to follow up on changes in AML disease, and to look for differences of overexpression according to cytogenetics abnormalities. Methods. This study focuses on 71 patients with AML overexpressing EVI1 included in the ALFA-0701 and ALFA-0702 trials for whom frozen material was available. BM and PB samples were collected at AML diagnosis (n=71) and during follow-up (n=152) as defined in the ALFA-0701 and ALFA-0702 trials. Threshold values in PB and BM were established by analyzing 31 normal PB and normal 19 BM samples. To achieve quantification, we cloned a sequence of EVI1 localized on exons 14 and 15, framing forward primer, reverse primer and TaqMan probe used by Groschel et al in 2010. The sequence was integrated into plasmids pCR2.1 TOPO (Invitrogen) which were used to determine the standard curve. RQ-PCR was performed on ABI7900 (Applied Biosystems). In parallel, the expression of the housekeeping gene ABL was evaluated by RQ-PCR for each sample. The results were expressed as a percentage of EVI1 copies by ABL copies. For every patient, Karyotype and follow-up data were confronted with the percentage of EVI1 expression. Results. To establish our overexpression threshold, we analyzed EVl1 expression in the population of negative witnesses. On PB, the median value of EVl1 expression was 0,11% [0,02 – 0,44], and the maximal value was 0,49%. On BM, the median value was 0,36% [0,01 – 0,96] , and the maximal value was 0,96%. Thus, the overexpression threshold was fixed at 0,5% for PB and at 1% for BM. Upon diagnosis, we confirmed EVl1 overexpression for 62 samples out of 71. Mismatches were observed for 9 samples of PB which were ranked in weak positive in relative quantification. EVl1 median expression were 23,26% [4,29 – 516,1] on BM and 3,61% [0,63 – 34,3] on PB. We analyzed EVI1 expression upon diagnosis and MRD1 for 20 patients. In BM, expression varied from 21,61% [4,18 – 592,70] to 3,56% [0,48 – 85,63]. In PB, expression varied from 4% [0,61 – 58,71] to 0,22% [0,01 – 11,51]. Although reduction was significant (p 〈 0,001), it remained insufficient for MRD follow-up on this parameter.Otherwise, upon diagnosis, we found a statistically significant difference between patients with intermediate cytogenetic (n=21) and patients with adverse cytogenetic (n=41) (p 〈 0,01) with an EVl1 median expression of 2.75% [0,54 – 24,07] against 10,88% [1,63 – 218,7] (fig 1). Strongest EVI1 overexpression were found in the MLL rearrangement sub-group (n=11) (p 〈 0,01) : 49,18% [1,7 – 614,1] vs 6,93% [0,69 – 109,9] .We did not reveal a strong link between rates and variations of EVI1 expression on the survival of patients, but a trend towards a less pejorative survival seems to appear when EVI1 overexpression is associated with a normal karyotype versus a complex karyotype (fig 2). Fig 1) EVI1 expression in diagnosis 2) Overall survival according to cytogenetic. Fig. 1) EVI1 expression in diagnosis 2) Overall survival according to cytogenetic. Conclusions. We developed a model of absolute quantification of EVI1 expression based on plasmids containing a target sequence located in exons 14 and 15. Nevertheless, the basal expression of EVI1 is high and variations are weak between diagnosis and follow-up. However, we observed higher expression in patients with adverse cytogenetics abnormalities. But, it seems that cytogenetic abnormalities associated with EVI1 overexpression confer a poor prognosis, rather than overexpression in itself. 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 ...
  • 6
    In: Oncotarget, Impact Journals, LLC, Vol. 6, No. 39 ( 2015-12-08), p. 42345-42353
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancers, MDPI AG, Vol. 12, No. 1 ( 2019-12-30), p. 88-
    Abstract: Acute myeloid leukemia (AML) encompasses heterogeneous entities with dismal outcomes. Intermediate and unfavorable-risk AML represent the most difficult-to-treat entities. We recently reported the benefit of the clofarabine-based consolidation (CLARA) regimen compared to the standard high-dose cytarabine (HDAC) regimen in younger AML patients. Here, we aimed at assessing the clinical significance of single-nucleotide polymorphism (SNP)-array alterations and their interactions with chemotherapy regimens. A SNP-array was successfully performed in 187 out of the 221 intent-to-treat patients (CLARA arm: n = 92 patients, HDAC arm: n = 95 patients). The CLARA regimen did not significantly improve relapse-free survival (RFS) among patients who displayed a complex karyotype when compared to the HDAC regimen (4-year RFS (4y-RFS): 36.4% vs. 18.8%, respectively; p = 0.134). Defining micro-complex karyotypes from at least four SNP-array lesions enabled us to refine and enlarge the subset of adverse patients. In such patients, the CLARA regimen significantly improved RFS compared to the HDAC regimen (4y-RFS: 44.4% vs. 13.8%, respectively; p = 0.004). From our study cohort, 8% of patients displayed TP53 mutations, which were associated with an impaired RFS (4y-RFS: 20.0% vs 43.7%; p = 0.029). In a multivariate analysis, micro-complex karyotypes remained the sole poor prognostic factor in the HDAC arm (hazard ratio (HR) = 2.324 (95% confidence interval (CI) = 1.337–4.041), p = 0.003). The SNP array represents a powerful and reproductive approach to refine adverse AML patients that may benefit from alternative consolidation regimens.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2019
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 103, No. 5 ( 2018-05), p. 822-829
    Type of Medium: Online Resource
    ISSN: 0390-6078 , 1592-8721
    Language: English
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2018
    detail.hit.zdb_id: 2186022-1
    detail.hit.zdb_id: 2030158-3
    detail.hit.zdb_id: 2805244-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1353-1353
    Abstract: Acute myeloid leukemia (AML) with t(8;21) chromosomal translocation, leading to the RUNX1-RUNX1T1 fusion, belong to the favorable risk AML subset. However, relapse incidence may reach 30-40% in these patients. Minimal residual disease monitoring (MRD) based on the quantification of RUNX1-RUNX1T1 fusion transcript by real-time quantitative PCR (RQ-PCR) has been reported to be an independent prognostic factor for the risk of relapse. The specificity of the RUNX1-RUNX1T1 fusion and the high sensitivity of RQ-PCR techniques have made RUNX1-RUNX1T1 an ideal marker to assess treatment response in t(8;21) AML. Undetectable MRD could mean either that tumor cells persist in a latent state without RNA expression or that MRD level is below the sensitivity threshold. Studies in chronic myeloid leukemia showed that BCR-ABL DNA was still detectable in patients in long-term complete molecular response with undetectable BCR-ABL fusion transcript. Using a similar approach, we investigated the use of RUNX1-RUNX1T1 DNA as a MRD marker in t(8;21) AML, instead of RUNX1-RUNX1T1 mRNA. This approach allows linking results directly to the amount of leukemic cells, since each leukemic cell contains one copy of the RUNX1-RUNX1T1 sequence, while the level of RUNX1-RUNX1T1 mRNA may vary from a patient to another. Methods This study focuses on 17 patients with t(8;21) AML included in the CBF-2006 trial and for whom frozen material was available for further molecular analysis. Bone marrow and blood samples were collected at AML diagnosis and during follow-up, as defined in the CBF-2006 trial. Eight patients relapsed during follow-up and 9 were still in complete remission at the end of the study. Interestingly, 3 patients relapsed with a previously undetectable MRD (in blood and bone marrow samples). First, we identified the breakpoints in the RUNX1 and RUNX1T1 genes for each patient using long-range PCR approaches, coupled with next-generation sequencing (NGS) on Personal Genome Machine™ (PGM). The stability of the RUNX1-RUNX1T1 rearrangement at relapse was checked by Sanger sequencing. Then, we performed quantification of RUNX1-RUNX1T1 DNA by RQ-PCR using Taqman technology. For each patient, a primer pair and a probe were designed using the patient's unique RUNX1-RUNX1T1 breakpoint sequence. The forward and reverse primers were located in RUNX1 and RUNX1T1 genes, respectively, and the probe was located at the RUNX1-RUNX1T1 junction. Calibration curves were established using 10-fold dilutions of the diagnostic DNA of each patient in normal control DNA. Results were given as a ratio of chimeric DNA amount in the follow-up sample to chimeric DNA amount at diagnosis. Results Chromosomal breakpoints were located in RUNX1 intron 5 for all patients. RUNX1T1 breakpoints were located in intron 1b for 15 patients, and in intron 1a for 2 patients (Fig. 1). Quantification failed for 1 patient which was further leave up. Between 2 and 7 follow-up samples were studied for the other patients (median 4.5). DNA monitoring was strongly correlated with RNA monitoring (Fig. 2). Sensitivity threshold, determined by the lowest diagnostic sample dilution which gives a signal, was 10-5 for 7 patients, 10-4 for 6 patients, and only 5.10-4 for 3 patients. MRD was detectable in 31 samples and undetectable in 30 samples by both methods, whereas MRD was detectable only on RNA in 7 samples, probably because of a lack of sensitivity of the RQ-PCR assay. Interestingly, RUNX1T1-RUNX1 DNA was detected in 3 samples from 2 patients who relapsed and for whom RUNX1T1-RUNX1 transcript was undetectable, despite a good RNA quality. Conclusions Overall, RUNX1-RUNX1T1 MRD levels on DNA and RNA were quite similar. The level of mRNA expression did not seem to change during follow-up when compared with the amount of DNA. MRD monitoring on genomic DNA is a useful method, but with sensitivity variations depending on the patient's breakpoint sequence and the efficiency of the RQ-PCR assay. DNA has potential advantages: it is more stable than RNA and a best substrate for collection, processing, transport and storage. Additionally, interpretation of the results is easier because it is closely related to the number of leukemic cells. However, this method greatly increases complexity, time of implementation, and cost of monitoring MRD, which limits its interest in routine practice. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 993-993
    Abstract: Context. The prognostic value of gene mutations in older AML patients (pts) treated intensively remains unclear. Only one study has explored the role of mutation patterns determined by NGS in older AML pts prospectively treated with various chemotherapies in years 2000-2010 (Eisfeld Leukemia 2018). Methods. Pts older than 60y enrolled in the ALFA-1200 trial (NCT01966497) between 09/2012 and 06/2016 were sequenced with a 37-gene myeloid panel. Pts received one 7+3 course followed by 2 intermediate-dose cytarabine courses. Pts with non-favorable risk were eligible for allogeneic stem cell transplantation (SCT). Variable selection for multivariate analyses was performed by lasso penalized regression including age, gender and log(WBC) as covariates. Results. Sequencing was done in 471 (93%) of the 509 enrolled pts. Median age and WBC count were 68y and 5.3x109/L, respectively (resp). CR (including CRp) was achieved in 341 (72%) pts and 90 underwent RIC-SCT in first CR. With a median follow-up of 25.4 months, median OS was 20.7 months. Pts had a median of 3 mutations (range 1-10). The 17 mostly frequently mutated genes (≥5% of pts, by decreasing frequency: DNMT3A, NPM1, TET2, ASXL1, FLT3, SRSF2, IDH2, RUNX1, NRAS, IDH1, STAG2, BCOR, TP53, PTPN11, U2AF1, EZH2 and KRAS) were retained for prognostic analyses. Genes belonging to a common pathway (eg. NRAS and KRAS) may have divergent prognostic values, preventing biology-informed grouping of mutations. Cytogenetic risk (derived from ELN 2017, Döhner Blood 2017, not considering gene mutations) was favorable (fav), intermediate (int), adverse (adv) and missing in 3%, 72%, 18% and 7% resp. Because of the few pts with fav cytogenetics in our cohort, pts were further grouped into non-adv and adv cytogenetics. CR rates and median OS were 75.6% vs 56.6% and 24.8 vs 9.5 months in pts with non-adv and adv cytogenetics, resp (both p 〈 0.0001). Because of difference in mutational patterns and gene-gene interactions, the prognostic role of mutations was considered independently in these two non-adv and adv subgroups. In the 388 pts with non-adv cytogenetics, NPM1 mutations independently predicted improved CR rate (Odds Ratio [OR]=2.3, p=0.014), while mutations in ASXL1 (OR=0.46, p=0.012), RUNX1 (OR=0.46, p=0.013) and NRAS (OR=0.49, p=0.04) had independent adverse predictive value. In univariate analysis the shorter OS of FLT3-ITD pts was confined to allele ratios≥ 0.5 (FLT3-ITDhigh, p=0.02). In a multivariate analysis accounting for clinical covariates, mutations in NPM1 (Hazard Ratio [HR] =0.45, p 〈 0.0001) and in SRSF2 (HR=0.64, p=0.03) predicted improved outcome, while FLT3-ITDhigh (HR=2.00, p=0.03), mutations in DNMT3A (HR=1.74, p=0.001), ASXL1 (HR=1.84, p=0.002) and NRAS (HR=1.70, p=0.009), but not RUNX1 or TP53, independently predicted worse OS. Significant interactions (eg. NPM1 - SRSF2, p=0.009, NPM1 - DNMT3A, p=0.03) precluded a simple NPM1-based stratification of pts with non-adv cytogenetics. This led to define a new prognostic hierarchy (Figure). The 49 NPM1mut pts with SRSF2 mutation and/or without adverse co-mutations (FLT3-ITDhighDNMT3A, ASXL1 and NRAS) had a median OS of 49.7 months, defining very low risk. NPM1wt pts without adverse co-mutations (n=114) had a median OS of 30.7 months and were considered at low risk. Among pts with ≥1 adverse co-mutation, NPM1 status had no significant prognostic influence (p=0.18). Regardless of NPM1 status, pts with a single (n=187) or ≥2 (n=38) adverse co-mutations (FLT3-ITDhighDNMT3A, ASXL1 or NRAS) had a median OS of 21.0 and 12.0 months, resp, and were considered at intermediate and high risk, resp. In the 83 pts with adv cytogenetics, TP53 mutations predicted shorter OS (p=0.004). Among pts with adv cytogenetics, those without TP53 mutation had a median OS of 12.6 months and were thus classified as high risk while the median OS of the 30 pts with adv cytogenetics and TP53 mutations was only 5.4 months, defining very high risk disease. This stratification resulted in improved OS prediction compared to the full molecular ELN 2017 (C-index 0.63 vs 0.58, resp). This stratification also predicted Relapse-Free Survival (RFS, Figure, p 〈 0.0001). Censoring at SCT did not affect these results. Conclusion. In AML patients older than 60y treated intensively, mutations in 7 genes (NPM1, SRSF2, FLT3, DNMT3A, ASLX1, NRAS and TP53) can refine the prognosis of cytogenetic sub-groups. Figure Figure. Disclosures Cluzeau: MENARINI: Consultancy; CELGENE: Consultancy; JAZZ PHARMA: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    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...