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
Filter
  • American Society of Hematology  (16)
  • Zhang, Zhaolei  (16)
Material
Publisher
  • American Society of Hematology  (16)
Language
Years
Subjects(RVK)
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4281-4281
    Abstract: Introduction: The discoveries of JAK2-V617F as well as MPL and CARL mutations have greatly clarified the underlying genetics of Philadelphia negative myeloproliferative disease (MPN). Mutation status on these three genes, especially JAK2-V617F, can characterize over 90% of MPN patients. However, the heterogeneity of MPN in terms of its AML transformation and treatment response remains unclear. To assess the difference in mutational status between MPN patients who progress to secondary AML and those who do not, we aim to examine longitudinal samples taken from multiple time points using next generation sequencing. Patients and Methods: Bone-marrow (BM) samples were collected from 19 MPNpatientsfrom2003 to 2012atChonnamNational UniversityHwasunHospital.The diagnosis of MPN was established according to the revised criteria of the World Health Organization.Longitudinal samples were taken at the time of diagnosis and at a follow-up as well as its T-cell fractions (CD+3) isolated from the peripheral blood using MAC separation column.Targeted sequencing was performed using an Agilent custom probe set of a panel of 84 myeloid genes. We multiplexed and sequenced the samples using an IlluminaHiseq2000. Results: The mean on-target coverage for the 57 sequenced samples was 861.4x. We detected a total of 48 somatic mutations in 25 genes in 17 patients (89%) throughout the course of the disease. Five of the 25 genes were recurrently mutated (JAK2, IDH2, ASXL1, SRSF2 and, TP53). As expected, JAK2-V617F was the most commonly observed (15/17 patients). One of the patients without JAK2-V617F carried a MPL mutation and another was triple negative.At the time of follow-up, 12 patients had chronic MPN (11 stable disease and 1 spleen response) withRuxolitinib treatment for a median duration of 373 days (range 255 - 729). Among 7 progressed patients, 5 patients had additional mutation at diagnosis of MPN other than JAK2 or MPL: MPN-13 (DNMT3A and ASXL1), MPN-14 (SRSF2 and IDH2), MPN-15 (IDH2), MPN16 (U2AF1), MPN-17 (IDH2 and SRSF2) as shown in figures. On the other hand, 4/12 non-progressed patients carried additional mutations: MPN-01 (ZRSR2), MPN-05 (ASXL1, CBL, FGR, KMT2D and TET2), MPN-10 (ASXL1, CDH13, EED, EZH2, MN1, NF1 and TRRAP), MPN-11 (FOXP1). In summary, only ASXL1 and JAK2-V617F were recurrently mutated among the non-progressed group. At the time of leukemic transformation, 6 out of 7 patients acquired new mutations: MPN-13 (SETBP1 and TP53), MPN-14 (RUNX1, ASXL1, and IDH1), MPN-15 (TP53 and CASP8), MPN-16 (TP53), MPN-18 (CEBPA), and MPN-20 (ASXL1). In the remaining case (MPN-17), increase of JAK2-V617F VAF was also observed. In summary, TP53 mutation was the most common mutation to acquire by the time of leukemic transformation (n=3). Other mutations acquired by this stage were in SETBP1, RUNX1, IDH1, CEBPA, and ASXL1. We did not observe any significant difference in the allelic burden increase of JAK2-V617F from diagnosis to follow-up between the non-progressed and progressed groups (Figure A). Conclusion: There is no significant difference in mutation burden increase of JAK2-V617F between patients who progressed to secondary AML and patients that did not. Acquisition of mutations other than JAK2-V617F at both diagnosis and at follow-up is associated with the risk of transformation to secondary AML. Mutation profiling using a myeloid gene panel at timed follow-up after MPN diagnosis can be more helpful than monitoring JAK2-V617F status in these patients. Figure Figure. 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: 2016
    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. 132, No. Supplement 1 ( 2018-11-29), p. 4361-4361
    Abstract: Introduction Myelodysplastic syndrome (MDS) is a heterogeneous group of myeloid disorder characterized by defective bone marrow (BM) hematopoiesis with peripheral blood cytopenias and risk for progression to acute myeloid leukemia. Accurate determination of prognosis is critical to select an appropriate therapy and to detect any case progressing to leukemic transformation which brings ominous prognosis in patients with MDS. Despite clinical risk models, additional molecular data are needed to enhance the prediction of patients' clinical courses and to aid disease management. Therefore, the present study attempted to classify the higher risk MDS (HR-MDS) patients according to their molecular risk through targeted RNA-sequencing, to correlate it with clinical risk models, and analyzed molecular risk grouping for prognostic stratification power, especially for leukemic transformation in higher-risk patients with MDS treated with hypomethylating agents (HMA) including azacitidine or decitabine. Patients and Methods A total of 30 patients were included with HR-MDS by International Prognostic Scoring System (IPSS). Overall, 60 bone marrow samples (30 diagnosis and follow-up pairs) were subject for targeted RNA-seq using Illumina TruSight Pan-Cancer panel. After read mapping by Tophat2, gene count was measured using HTSeq followed by DEseq2 for differential gene expression quantification. All 60 samples as well as 30 samples from T-cell fraction (CD3+, as a control) were also subjected for DNA-seq targeting a panel of 84 commonly mutated genes in myeloid malignancies (Agilent SureSelect). All downstream computational and statistical analyses were performed using R and Python. Results The median age was 65 years (range 40-84 years) with 16 male patients (53%). Twenty-seven (90%) and 3 (10%) patients were intermediate-2 and high risk by IPSS, respectively. According to revised IPSS (IPSS-R), the distribution of risk groups was as follows: low (n=5, 17%), intermediate (n=8, 27%), high (n=11, 37%), and very high (n=6, 20%). A total of 56 mutations were detected in the diagnostic samples from 30 patients. Frequently mutated genes were DDX41 (n=5) and TP53 (n=4). Best response to HMA (16 azacitidine and 14 decitabine) was achieved in median 4 cycles (range 3-8). Complete response (CR) including marrow CR was achieved in 18 patients (60%), and 10 patients (33%) received allogeneic hematopoietic cell transplantation. Overall survival (OS) rate was not well correlated with IPSS-R risk groups. With median follow-up duration of 28.2 months (range 3.8-95), 3-years' OS rate showed 40%, 75%, 36%, and 67% in low, intermediate, high, and very high risk, respectively. Unsupervised clustering using top 100 genes with highest variance revealed 3 distinct clusters (n=8, 9, and 13 in group 1, 2, and 3), 3-years' OS rate of which showed 73%, 57%, and 35% in group 1, 2, and 3, respectively (p=0.004 between group 3 vs group 1/2). Despite inferior long-term outcomes in the group 3, the baseline clinical variables of some patients were classified as favorable implying that clinical factor does not reflect adverse long-term outcomes: 4 out of 13 patients with low risk by IPSS-R eventually experienced adverse outcome. The 3-years' leukemic transformation rate was 0%, 33% and 57% in group 1, 2, and 3 (p=0.039 between group 3 vs group 1/2). In the multivariate analyses, besides achievement of CR, the risk group 3 by RNA-seq were identified as independent adverse prognostic factors for OS (p=0.007, HR 6.75 [1.68-27.17]) as well as leukemic transformation (p=0.013, HR 6.91 [1.49-31.95] ). In the gene set enrichment analysis using MSigDB, hematopoietic stem cell genes were enriched in RNA-seq group 3, suggesting that the high-risk signature on RNA-seq is linked with stemness of hematopoietic stem cells. Conclusion RNA-seq can be utilized to identify the higher risk patients with MDS. The higher risk group by RNA-seq enriched with genes with hematopoietic stem cells, which suggests that stemness in hematopoietic stem cells is linked with resistance to HMA therapy and increasing risk of leukemic transformation in HR-MDS. Figure. Figure. 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: 2018
    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. 134, No. Supplement_1 ( 2019-11-13), p. 4263-4263
    Abstract: Background: Hypomethylating agents (HMAs) are used to treat patients with lower-risk myelodysplastic syndrome (LR-MDS) relapsing after the use of hematopoietic cytokines or presenting initially with more than two lineages of cytopenias. However, the significance of underlying genetics and allelic burden changes after HMAs are still under investigation. This study investigated the effects of allelic burden changes on the long-term outcomes in LR-MDS patients treated with HMAs. Methods: This study included 61 patients with LR-MDS treated with azacitidine. Bone marrow samples were taken at diagnosis and follow-up after median 4 cycles. Targeted deep sequencing on a custom myeloid gene panel of 84 genes (Agilent SureSelect) was performed on trios of T-cell, pre-HMA, and post-HMA samples on 61 LR-MDS patients. Patients were divided into groups according to the post-HMA variant allele frequency (VAF): low-VAF ( 〈 2%), high-VAF (≥ 2%), and no-mutation group (absence of mutations at diagnosis and follow-up). Overall survival (OS) was defined as the time from the azacitidine treatment until death from any cause, which was analyzed using the Kaplan-Meier method and the groups were compared using the log-rank test. The cumulative incidence of AML was calculated using the Gray method, considering death without AML as a competing risk. Fine-Gray proportional hazard regression with a competing event was used to identify risk factors for the incidence of AML. Results: Median age was 67 years (range 31-81), and 41 patients (67%) were male. IPSS risk group were low in 2 patients (3%) and intermediate-1 in 59 (97%). At diagnosis, 38 patients harbored at least one mutation. Most frequently mutated genes were ASXL1 (n=11, 18%), TET2 (n=10, 16%), and SRSF2 (n=7, 11%) followed by RUNX1, SF3B1, U2AF1, IDH2, and DNMT3A. Azacitidine was administered median 8 cycles (range 2-44). The overall response (CR, PR, HI) was achieved in 18 patients (30%). With median follow-up duration of 31 months (range 4.7-135 months), leukemic transformation occurred in 11 patients (18%). Mutational allelic burdens were decreased from median 20.9% (range 0.1-67.2) to 11.0% (range 0.0-74.9%). At follow-up, 5 patients were low-VAF group and 33 were high-VAF group. OS rate was not different between the low-VAF and high-VAF group (50% vs 46% at 3 years; p=0.80). Three-year cumulative incidence of AML was higher in high-VAF group compared to low-VAF (0%) and no-mutation group (4.8%, p=0.02). However, non-leukemic mortality was higher in low-VAF group than no-mutation group (60% vs 23%, p=0.09), which explains similar OS rate between low-VAF and high-VAF group. In the multivariate analysis, high-VAF was an independent predictive factor for an AML transformation in LR-MDS patients treated with azacitidine (HR 5.20, p=0.04). Conclusion: The current study showed that the high residual allelic burden is associated with an increased AML transformation in LR-MDS patients treated with azacitidine, irrespective of the clinical response. The higher non-leukemic mortality explains inferior OS in low-VAF group compared to no-mutation group. Figure 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: 2019
    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. 128, No. 22 ( 2016-12-02), p. 5253-5253
    Abstract: Background and Objectives Acute Myeloid Leukemia (AML) is a cytogenetically and molecularly heterogeneous disease. In the recent decades, many genetic mutations and their clinical significances in AML have been identified with the development of new genomics technology. Based on these advances, new 2 entities were added to the WHO 2008 classification : AML with mutated NPM1 and AML with mutated CEBPA. Likewise, AML with RUNX1 mutation are now considered as a new provisional entity in the next update of WHO classification. In this work, we characterized patients with cytogenetically normal AML according to RUNX1 mutational status and analyzed several co-mutations by next generation sequencing. Patients and Methods A total of 419 patients were included in the present study who met the following eligibility criteria: 1) age ≥ 15 years; 2) a diagnosis of AML with normal karyotype confirmed by conventional cytogenetic analysis. Analysis of genetic mutations were performed using targeted resequencing by Illumina Hiseq 2000 (Sureselect custom probe set targeting 94 myeloid gene panel including RUNX1 mutation). Samples for the confirmation of first complete response were also analyzed in 163 patients. The majority of patients (97%) received '3+7' standard induction chemotherapy. Median age was 53(range 15-84). Results Overall, most common mutations for this cohort were NPM1(33.9%), DNMT3A(30.3%), NRAS(20.2%), IDH2(15.0%), FLT3(12.2%), CEBPA(11.1%). RUNX1 mutations were found in 22 of 419 (5.4%) patients. 7 of 13 available samples in complete remission still had RUNX1 mutation. The patients with RUNX1 mutations were older than those with wild-type RUNX1. (p=0.006) and RUNX1 mutation had a trend of male preponderance. The WBC count and blast percentage of peripheral blood and bone marrow were not different according to RUNX1 mutational status. The complete response rate was significantly lower in RUNX1 mutated group compared with wild-type group. (57% vs. 84%, p=0.005) In univariable survival analysis, RUNX1 mutations were significantly associated with inferior event-free survival (EFS) (p 〈 0.001), relapse-free survival (RFS) (p=0.009) and overall survival (OS) (p=0.002). However, in multivariable analysis, RUNX1 mutation was not an independent prognostic factor for inferior EFS (hazard ratio(HR) 1.48, p=0.286), RFS (HR 2.15, p=0.057) OS (HR 1.14, p=0.716). Co-mutation analysis revealed that ASXL1 (26%,p=0.001), KRAS (26%, p=0.009), BCOR (16%, p=0.032) were correlated with RUNX1 mutation. None of the patients with RUNX1 mutation had NPM1 mutation and only one patient had CEBPA mutation. Conclusion In cytogenetically normal AML, RUNX1 mutation is observed in 5.4% and is mutually exclusive of the NPM1 and CEBPA mutation. Older age and lower complete response rate is correlated with RUNX1 mutation. In univariable survival analysis, RUNX1 mutation is associated with poor clinical outcomes. 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: 2016
    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. 140, No. Supplement 1 ( 2022-11-15), p. 1018-1019
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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. 132, No. 15 ( 2018-10-11), p. 1604-1613
    Abstract: Higher allelic burden at day 21 of post-HCT is associated with higher risk of relapse and mortality. Longitudinal tracking of AML patients receiving HCT is feasible and provides clinically relevant information.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1391-1391
    Abstract: Introduction NGS-based detection of minimal residual disease (MRD) has been successfully demonstrated for its correlation with relapse risk in AML. However, in a subtype of AML, CBF-AML, its clinical relevance of residual allelic burden at complete remission (CR) has not been fully explored. The standard MRD detection method in CBF-AML is quantitative PCR (qPCR). In this study, we aimed to explore applicability and feasibility of NGS-based MRD detection in CBF-AML taking various approaches, rather than simply using residual allele burden at CR. Patients and Methods Fifty-three patients (pts) diagnosed with CBF-AML were enrolled in this study (31 pts with RUNX1-RUNX1T1 and 22 pts with CBFB-MYH11). All 53 patients achieved complete remission (CR). We performed targeted deep sequencing on 84 genes in 106 samples collected at diagnosis and at CR as well as T-cell (n = 53, CD3+) fraction as a control using Illumina Hiseq 2500. Mean on-target coverage for 159 sequenced was 1,572x. The level of RUNX1-RUNX1T1 was measured at diagnosis and at CR for 29 patients using qPCR. Results At diagnosis, 99 mutations from 49 pts (n = 49/53, 92%) were detected, where median number of mutations for 49 pts was 2 (range 1-6). Consistent with previous studies, KIT (36%), NRAS (32%), KRAS (17%), ASXL2 (15%) were commonly mutated. Among mutations detected at diagnosis, cKIT-D816 mutation and mutations in genes in DNA methylation pathway (DNMT3A and TET2) were associated with higher risk of relapse (5.29, [1.89 - 14.87], p = 0.002 and 3.15 [1.07 - 9.26] , p= 0.037, respectively). In CR samples, 46 mutations from 32 pts were still detectable (46/99, 46%, mean VAF: 0.60%, range 0.04%-6.28%, Fig A). Only 4 mutations from 2 pts were over 2.5% (2 in TET2, 1 in ASXL1, and 1 in U2AF1). When tracing back at diagnosis, allelic burden of 46 mutations detected at remission were higher than 53 cleared mutations (p 〈 0.002), indicating clonal mutations are more likely to be detected at CR (Fig A). They were mostly in genes associated with activated signaling (32/46, 70%). When considering complete clearance rate, mutations in genes associated with activated signaling, DNA methylation, and spliceosome tended to be persistent at CR (32/64, 4/5, and 1/1), whereas mutations in cohesin complex and chromatin modifiers were mostly completely cleared (0/8 and 4/13). We then assessed clinical relevance of mutation clearance from various perspectives. We did not find association of mutation clearance at 0.3% (MC03) with OS (p = 0.43) or with relapse risk (p = 0.8, Fig B). Complete mutation clearance also did not show significant association with OS and relapse risk. Among 29 pts with RUNX1-RUNX1T1 with available qPCR data, 20 pts were MRD-positive by qPCR at CR. Nine pts who achieved MRD-negative also achieved MC03 as well. When considering only MRD-positive pts, achievement of MC03 did not affect OS (p = 0.69) and relapse incidence (p = 0.86, Fig C). Lastly, we assessed whether persistence of high risk mutations at CR (cKIT-D816, DNMT3A, and TET2) is associated with higher risk of relapse, but complete clearance of KIT-D816 mutation also did not affect OS and relapse incidence (p = 0.94 and p = 0.40, respectively, Fig D). We were not able to analyze DNMT3A and TET2 mutations as only 1/5 mutation was cleared. Conclusion Current study demonstrates that low residual allelic burden measured by NGS at CR does not provide additional clinically relevant information in addition to baseline mutation profile nor qPCR-based MRD in CBF-AML Figure. Figure. 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: 2018
    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, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2876-2876
    Abstract: Introduction Acute myeloid leukemia (AML) is a genetically heterogeneous disease. A recent study (NEJM, 2016) classified 1540 patients into 14 subgroups using mutation information from targeted next generation sequencing data as well as cytogenetic information [1]. The classification criteria of 7 of these subgroups rely solely on mutation information. NK-AML is characterized by its lack of cytogenetic abnormalities. In this study, we attempted to replicate the prognostic stratification in an independent set of NK-AML patients using the NEJM study's genomic classification criteria. Patients and Methods This study included a total of 393 patients who met the following eligibility criteria: 1) age ≥ 15 years; 2) a diagnosis of NK-AML confirmed by conventional cytogenetic analysis; 3) treatment with induction chemotherapy using a standard protocol (a 3-day course of anthracycline with a 7-day course of cytosine arabinoside). The median follow-up duration was 55.1 months (range, 0.7-182.9). Analysis of genetic mutations were performed using targeted sequencing by Illumina Hiseq 2000 (Agilent custom probe set targeting entire exon regions of a myeloid panel consisting of 94 genes). Results We identified driver mutations across 28 genes or genomic regions, with 2 or more driver mutations identified in 15/393 patients (3.8%). Based on the genomic classification criteria, the patients were classified as follows: 136 patients (34.6%) with NPM1 mutations, 42 patients (10.7%) with mutated chromatin modifiers and/or RNA-splicing genes, 6 patients (1.5%) with TP53 mutations, 40 patients (10.2%) with biallelic CEBPA mutations, 8 patients (2.0%) with IDH2-R172 mutations and no other class-defining lesions, 108 patients (27.5%) with driver mutations but no detected class-defining lesions, 38 patients (9.7%) with no detected driver mutations, and 15 patients (3.8%) who met the criteria of more than one genomic subgroup. Of the 393 patients, 325 patients (82.7%) achieved complete remission (CR). CR rates vary depending on the genomic subgroup (75.9%-97.4%). The CR rate for each subgroup was as follows: 86.8% (118/136) of patients with NPM1 mutations61.9% (26/42) of patients with mutated chromatin and/or RNA-splicing genes83.3% (5/6) of patients with TP53 mutations97.5% (38/40) of patients with biallelic CEBPA mutations87.5% (7/8) of patients with IDH2-R172 mutations and no other class-defining lesions75.9% (82/108) of patients with driver mutations but no detected class-defining lesions97.3% (37/38) of patients with no detected driver mutations80.0% (12/15) of patients meeting criteria of more than one subgroup 5-year OS and 5-year relapse incidence (RI) for each subgroup was as follows: 49.3% (95% CI, 40.1-58.5) and 39.8% (95% CI, 30.1-49.2) of patients with NPM1 mutations11.6% (95% CI, 1.4-21.8) and 71.4% (95% CI, 45.7-86.5) of patients with mutated chromatin and/or RNA-splicing genes50.0% (95% CI, 10.0-90.0) and 20.0% (95% CI, 0.4-61.2) of patients with TP53 mutations68.3% (95% CI, 53.4-83.2) and 19.7% (95% CI, 8.5-34.4) of patients with biallelic CEBPA mutations56.3% (95% CI, 17.3-95.3) and 21.4% (95% CI, 0.3-67.3) of patients with IDH2-R172 mutations and no other class-defining lesions26.6% (95% CI, 17.4-35.8) and 53.2% (95% CI, 40.7-64.3) of patients with driver mutations but no detected class-defining lesions29.1% (95% CI, 14.2-44.0) and 43.8% (95% CI, 27.1-59.3) of patients with no detected driver mutations40.0% (95% CI, 15.3-64.7) and 33.3% (95% CI, 9.2-60.3) of patients that meet the criteria of more than one subgroup. The CR rates of the subgroup with mutated chromatin and/or RNA-splicing genes was significantly lower than the rest of the cohort (61.9% vs. 85.2%, p=0.00016). The 5-year OS and 5-year RI of the subgroup were also poorer than the others [61.9% vs. 85.2% in OS (p=0.00016), 71.4% vs. 40.1% in RI (p 〈 0.0001)]. Conclusion Our NK-AML cohort showed similar survival patterns to the cohort in Papaemmanuil et al (NEJM 2016). The subgroup in AML with mutated chromatin and/or RNA-Splicing genes had the poorest prognosis with respect to CR rate and overall survival. This analysis replicates the result of recently published genomic classification and supports its use for categorizing NK-AML patients. Reference [1] Genomic Classification and Prognosis in Acute Myeloid Leukemia. Papaemmanuil E et al. N Engl J Med, 2016 vol. 374 (23) pp. 2209-2221. Figure Figure. Disclosures Jang: Kyowa Hakko Kirin Co., Ltd.: 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 52-52
    Abstract: Introduction: Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders characterized by dysplastic changes in one or more cellular lineages causing impaired bone marrow function. One third of patients diagnosed with MDS progress to secondary acute myeloid leukemia (sAML). These patients have significantly worse prognoses than de novo AML patients. Azacitidine (AZA), a hypomethylating agent is commonly used to treat MDS patients as a frontline therapy. Although its survival benefits over supportive care in a randomized trial has been demonstrated, the underlying genetics and clonal dynamics upon AZA response/AML progression have not been well examined. Using next generation sequencing (NGS) technology, we attempted to assess the clinical relevance of somatic mutations and their dynamics as they relate to AZA treatment in MDS patients using longitudinal samples. Patients and Methods: Ninety-five MDS patients (56 lower risk and 39 higher risk MDS based on the revised IPSS scoring system) were enrolled in this study. The median age of the 95 patients is 67 years (range of 31 Ð 84) and median follow-up duration was 747 days (range of 137-3328 days). We performed targeted deep sequencing (entire exon region of a panel of 84 myeloid genes, Agilent custom probe set) on 285 bone-marrow samples including the longitudinal samples taken at diagnosis (n=95) and post-AZA treatment, (median 4 cycles) as well as T-cell fraction (CD3+). We multiplexed and sequenced the samples using an Illumina Hiseq 2000. After read mapping and variant calling, hierarchical clustering, pathway and survival analyses were performed in R. Results: Targeted sequencing on the myeloid gene panel revealed 176 mutations in 68 patients (68/95, 71.6%) with a median of 2 mutations per patient (ranges 2-6). The average on-target coverage for 285 sequenced samples was 1205x. Twenty-five of 44 mutated genes were recurrently mutated. ASXL1 was the most frequently mutated in the cohort (21%), followed by TET2 (15%), DNMT3A (11%), and SRSF2 (11%). Mutated genes were then grouped into 8 biological pathways, defined in The Cancer Genome Atlas (TCGA) AML study. The most frequent biological pathway with mutated genes at diagnosis was DNA methylation (28.4%), followed by spliceosome (25.2%), chromatin modifiers (22.1%), myeloid transcription factors (TFs) (11.6%), activated signaling (11.6%), tumor suppressors (12.6%), and cohesin complex (6.3%). When assessing the differences in patterns of variant allele frequency (VAF), we found significant VAF reduction in responders compared to non-responders (p = 0.007, repeated measures using general linear model, Figure A). Multivariate analyses revealed that mutation burden in different genes and biological pathways have distinct impact on AZA response, AML transformation, and overall survival. Higher bone marrow blast percentage (5%) was associated with all three measures (Figure B). Most significantly, mutations in activated signaling pathway genes are associated with AML progression (p=0.002). In addition, we could not detect decreased VAFs in activated signalling pathway genes even in responders (Figure C-D). Patients with SRSF2 mutations tend to respond to AZA (OR 14.084, p=0.003). Mutations in tumor suppressors (HR 4.825, p 〈 0.001) and myeloid TFs (HR 3.070, p=0.020) were adverse prognostic factors in overall survival. Of interest, mutations in DNA methylation pathway were not independent prognostic factor for AZA response, AML transformation, or overall survival. Conclusion: These data and analyses show that reduction in mutation burden is correlated with AZA response. Mutations in different genes and biological pathways are associated with distinct clinical measures that tumor suppressors and myeloid TFs were identified as poor prognostic factors in terms of OS. Persistent mutation burden in activated signaling pathways is a strong predictor for AML transformation. In summary, longitudinal tracking of MDS patients using NGS may improve criteria for AZA response and early detection of AML progression. Figure 1. Figure 1. Disclosures Jang: Alexion Pharmaceuticals, Inc: 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: 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. 132, No. Supplement 1 ( 2018-11-29), p. 2669-2669
    Abstract: Introduction Recent studies utilizing NGS demonstrated that residual allelic burden at complete remission (CR) is associated with worse overall survival (OS) and relapse incidence in AML. Quantitative PCR (qPCR) based disease monitoring is current practice in CBF-AML. However, qPCR requires standardization and the result for the same sample may vary depending on several factors. Also, other known prognostic factors such as cKIT mutation require an additional test. As RNA-seq can detect gene rearrangement as well as somatic mutations, we hypothesized that RNA-seq on samples taken at diagnosis and at remission can be used to monitor these genetic alterations simultaneously and can be utilized for minimal residual disease (MRD) monitoring in CBF-AML. Patients and Methods This study included 42 CBF-AML patients (23 RUNX1-RUNX1T1 and 19 CBFB-MYH11 AML). Overall, 84 bone marrow samples (42 diagnosis-CR pairs) were subjected to targeted RNA-seq using Illumina TruSight Pan-Cancer panel. After read mapping, gene count was measured using HTSeq followed by DEseq2 for gene expression quantification. Average number of sequenced reads was 3.5M reads with 87% overall mapping rate. Gene fusions in diagnostic samples were detected using EricScript. All 84 samples as well as 42 samples from T-cell fraction (CD3+, as a control) were also subjected to DNA sequencing, targeting a panel of 84 genes (Agilent SureSelect custom gene panel). Average on-target coverage was 1,606x. All other computational analyses were done using R and python. Results In diagnostic samples, class-defining gene fusion events were detected in all 42 patients. In CR samples, we tracked identical junctions identified in corresponding diagnostic samples. As expected, both CBFB-MYH11 and RUNX1-RUNX1T1 showed significant reduction in all CR samples compared to their corresponding diagnostic samples (p 〈 2.2e-13 and p 〈 6.3e-05, Fig A and B). CBFB-MYH11 was detectable in 6/19 CR samples (32%) and RUNX1-RUNX1T1 was detectable in 15/23 CR samples (65%). Reduction level of RUNX1-RUNX1T1 measured by RNA-seq showed positive correlation with the reduction level measured by qPCR (Pearson's Rho = 0.74, p 〈 5.4e-05, Fig C). As per mutational profile at diagnosis, we detected 74 mutations in 38 samples (n=38/42, 90%). NRAS (36%), KIT (36%), KRAS (17%) and, ASXL2 (17%) were commonly mutated. Survival analyses on each gene and each protein locus identified cKIT-D816 mutation as an adverse prognostic factor (HR = 3.57, [1.15 - 11.11], p = 0.028). We were able to detect all cKIT-D816 mutations in RNA-seq. Using information from NGS, we built a prognostic model for RUNX1-RUNX1T1 AML (n = 23). Decision tree analysis identified three distinct subgroups of RUNX1-RUNX1T1 AML on the basis of reduction level of RUNX1-RUNX1T1 and mutation profile (Fig D). Consistent with previous studies, 3-log or deeper reduction of RUNX1-RUNX1T1 transcript level was the most significant prognostic factor (low risk group). The algorithm further divided the patients who failed to achieve 3-log reduction according to the presence of cKIT-D816 mutation at diagnosis (intermediate and high risk group). For three defined groups, 2-year OS rates were 87%, 74%, and 33% (p = 0.08, Fig E) and 2-year relapse incidence rates were 13%, 42%, and 67% (p = 0.048, Fig F). Conclusion RNA-seq can be utilized to quantify RUNX1-RUNX1T1 and CBFB-MYH11 transcripts on diagnostic and CR samples in CBF-AML. We also showed that RNA-seq can stratify RUNX1-RUNX1T1 AML patients into three risk groups according to their long-term prognosis. Figure. Figure. 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: 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...