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. 3515-3515
    Abstract: Abstract 3515 Background: The core binding factor (CBF) AML can be achieved long-term remission with high dose cytarabine-based chemotherapy alone. However, those with C-KIT gene mutation (esp. D816 C-KIT mutation) showed worse treatment outcomes compared to those with wild type C-KIT gene. The remaining cases without D816 C-KIT mutation is around 75% of CBF AML, which implies requirement of more sophisticated dissection of the patients according to their prognosis. Single nucleotide polymorphism (SNP) array (SNP-A) could detect cryptic abnormal genomic lesions, not identified by metaphase cytogenetics(MC). In this study, we analyzed the prognostic value of SNP-A based karyotyping combined with MC and its association with C-KIT mutation to facilitate further stratification of CBF AML patients. Methods and Materials: A total of 98 CBF AML patients were included and of whom, 63 (64%) and 35 patients (36%) were t(8;21) and inv(16)/t(16;16), respectively. Genome-Wide Human SNP 6.0 Array (Affymetrix, CA, USA) was performed using DNAs from marrow samples taken at diagnosis. Results: A total of 40 abnormal genomic lesions in 25 patients (26%) were detected by SNP-A karyotyping analysis, with a mean of 1.6 lesions per affected case (median size 33.6 Mb; range 0.4–145.9 Mb), including 3 CN-LOH lesions, 17 gain lesions, and 20 loss lesions. Survival of the patients with abnormal lesion(s) detected by SNP-A or/and MC was worse than those without any lesions in terms of 2 years' overall survival (OS; 57.5% vs 76.4%, p=0.028), event-free (EFS; 45.7% vs 66.2%, p=0.072) and leukemia free survival (LFS; 49.0% vs 77.4%, p=0.015). In contrast, MC alone could not stratify patients according to their long-term prognosis. Especially, in the subgroup with inv(16)/t(16;16), survival of patients with abnormal SNP-A/MC lesion showed worse than that of those without lesion (40.9±12.7% vs 80.2±10.4% at 2 yrs, p=0.040), but not in the subgroup with t(8;21) (66.85±9.1% vs 74.4±7.8% at 2 yrs, p=0.240). As for the subgroup with D816 C-KIT mutation, there were no differences of OS (p=0.417), EFS (p=0.380) and LFS (p=0.218) according to the presence of abnormal lesions detected by either SNP-A or MC. However, in the subgroup without D816 C-KIT mutation, those with abnormal lesions detected by either SNP-A or MC showed worse survival compared to those without abnormal lesions with respect to OS (61.6±8.7% vs 82.7±5.6% at 2 yrs, p=0.038). Multivariate analysis confirmed prognostic impact of abnormal SNP/MC lesions on OS (HR 2.743, p=0.020), EFS (HR 2.434, p=0.025), and LFS (HR 3.350, p=0.012). Conclusion: This study suggests that combined use of SNP-A with MC in the initial evaluation of CBF AML can provide an important prognostic value, especially in the inv(16)/t(16;16) subgroup or in the patients without having D816 C-KIT mutation. 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. 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 ...
  • 3
    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 ...
  • 4
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 4862-4862
    Abstract: Background: Allogeneic hematopoietic cell transplantation (HSCT) is the only curative potential for patients with MDS. HSCT following reduced-intensity conditioning (RIST) have enabled the extension of transplantation to include older or medically infirm patients with myeloid malignancies. However, it is very difficult to determine the value of outcomes of RIST for myelodysplastic syndrome (MDS) because of the heterogeneity of diseases included in most trials and the small number of MDS. Method: Twenty-two patients with de novo MDS classified by WHO criteria received an allograft using a Fludarabine (30 mg/m2/day for 5 days)/Busulfex (3.2 mg/kg/day for 2 days) (14 patients) or Fludarabine (30 mg/m2/day for 5 days)/Busulfex (3.2 mg/kg/day for 2 days)/ATG (2.5 mg/kg/day for 2 days) (8 patients). The median age of patients was 43 years (range, 16–55). Donors were HLA-compatible sibling (n = 19) or matched unrelated (n = 3). Result: Nineteen patients (86.4%) achieved engraftment. At a median follow-up of 18.9 months (range, 13.1–24.8 months), the estimated 2-year overall survival (OS), event-free survival (EFS), transplantation-related mortality and relapse incidence were 78.7%, 67.7%, 12.6%, and 22.5%, respectively. Acute GVHD greater than grade II was developed in 3 patients (15.8%). Chronic GVHD were developed in 10 patients (55.6%) and all of them did not receive ATG as conditioning. Variables influencing EFS were chronic GVHD (None/Limited [20.8%] versus Extensive [100%] , P = 0.001), marrow blasts before transplantation ( & lt;10% [85.1%] versus & gt;10% [20.8%], P = 0.006), and WHO criteria (RA [100%] versus RCMD [75%] versus RAEB-1 [80%] versus RAEB-2 [20.8%], P = 0.043). Conclusion: These observations indicate that high level of pretransplant leukemic blasts and the absence of chronic graft-versus-host disease provide negative effects on clinical outcome of transplantation with Fludarabine-Busulfex based reduced intensity conditioning for patients with the de novo myelodysplastic syndrome according to WHO criteria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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. 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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3832-3832
    Abstract: Background: Stem cells display remarkably high levels of 5-hydroxymethylcytosine (5hmC) and, both TET2 or IDH1/2 mutations can impair 5hmC generation. However, the implications of 5hmC have not been evaluated comprehensively in patients with normal karyotype (NK)-AML, especially in aspect of prognostic value in survivals. Methods: A total of 407 patients were included in the present study, and all met the following eligibility criteria: 1) age ≥ 15 years; 2) a diagnosis of NK-AML confirmed by conventional cytogenetic analysis; and, 3) treatment with induction chemotherapy using a standard protocol (a 3-day course of anthracycline with a 7-day course of cytosine arabinoside). NK-AML patients were diagnosed from October 1998 to September 2012 in seven participating institutes. Among 407 patients with NK-AML who received induction therapy, we selected the 376 patients available the analysis of 5-hmC, retrospectively. For quantitation of 5hmC, Quest 5-hmC DNA ELISA kits (Zymo Research) were used following the manufacturer's protocol. TET2 or IDH1/2 mutation analysis were performed using direct sequencing. We analyzed for 5hmC levels in patients with TET2 or IDH1/2 mutations and, to know the correlation of 5hmC levels with mutant alleles. Results: The prevalence rates for the mutations were 13.0% in TET2mut, 7.2% in IDH1 and,14.1% in IDH2mut. Mutation rates of TET2 or IDH1/2 was 34.6% (130/376). We examined whether the range of 5hmC values correlated with each mutations. TET2, IDH1/2 mutated patients had significant lower levels of 5hmC compared with patients without any TET2 or IDH1/2 mutations (all, p 〈 0.001). The median value of 5hmC level were: TET2mut (median: 0.051%, range: 0.002%-0.120%), IDH1mut (median: 0.044%, range: 0.004%-0.641%), IDH2mut (median: 0.050%, range: 0.001%-0.457%), any mutation of TET2 or IDH1/2 (median: 0.048% , range: 0.001%-0.641%) and, TET2 wild-type and IDH1/2 wild type (median: 0.084%, range: 0.0003%-0.999%). In control group (TET2 wild-type and IDH1/2 wild type), 5hmC levels distributed with broad range but, 5hmC levels were tightly clustered in patients with TET2, IDH1 or IDH2 mutations. With a median follow-up duration of 55.5 months (range, 0.7-179.8 months), there was no significant difference in overall survival (OS), event free survival (EFS) and relapse risk according to TET2mut or IDH1/2mut (all, p 〉 0.05). To identify the role of 5hmC levels in clinical significances, we sub-classified this group with tertile category for 5hmC values. However, we could not find the clinical significant in OS, EFS and relapse risk according to the 5hmC levels (all, p 〉 0.05). Conclusion: TET2 or IDH1/2 mutated patients had lower levels of 5hmC. 5hmC levels distributed with wide range in patients with TET2 and IDH1/2 wild type and tightly clustered in patients with TET2, IDH1 or IDH2 mutations. Apart from affecting the methylation status of the DNA, other processes may be influenced by 5hmC levels in patients with NK-AML with TET2 wild-type and IDH1/2 wild type. In addition, 5hmC may not be a prognostic value to predict the survivals of relapse risk in NK-AML. Disclosures Jang: Alexion Pharmaceuticals: Research Funding. Kim:Novartis Pharmaceuticals: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding.
    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, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1098-1098
    Abstract: Purpose: Previously, we demonstrated the positive impact of first-line imatinib interim therapy on the outcome of allogeneic stem cell transplantation (SCT) in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph-positive ALL) (Blood2005;105:3449). Here, we analyzed for risk factors that affect transplantation outcome, and focused particularly on the prognostic relevance of minimal residual disease (MRD) during treatment course. Patients and Methods: Fifty-two consecutive adults with Ph-positive ALL who completed SCT following imatinib therapy were enrolled in this prospective study. MRD assessment was performed using real-time quantitative polymerase chain reaction. Results: Forty-three (87.8%) of the 49 evaluable patients showed a decrease in MRD after imatinib therapy. Molecular remission rates were 18.4% and 44.4% after the first and second imatinib cycles, respectively. Forty-eight (92.3%) of the 52 patients received SCT during first complete remission. With a median follow-up of 42 months after SCT, the actuarial 3-year relapse and disease-free survival (DFS) rates were 22.9% ± 6.6% and 67.3% ± 7.2%, respectively. An MRD level of ≥ 10−3 after the first imatinib cycle was found to be the most powerful predictor of relapse (47.5% ± 14.3% versus 11.4% ± 6.4%, P = .009) and DFS (45.0% ± 13.2% versus 80.9% ± 8.0%, P = .016). The presence of chronic graft-versus-host disease was also found to be associated with a lower relapse (5.3% ± 5.1% versus 37.6% ± 10.8%, P = .029) and better DFS (82.0% ± 9.5% versus 62.4% ± 10.8%, P = .039). Conclusion: In the setting of allogeneic SCT following imatinib therapy, prospective MRD monitoring may allow us to identify subgroups of Ph-positive ALL patients at high risk of relapse at an earlier treatment stage.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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. 136, No. Supplement 1 ( 2020-11-5), p. 1-2
    Abstract: Only a few studies investigated prognostic values of next-generation sequencing-based measurable residual disease (NGS-MRD) detection in AML before or after allogeneic hematopoietic stem cell transplantation (Allo-HSCT). Moreover, NGS-MRD assessments were mostly performed only at pre-HSCT, and results were discordant. The current study longitudinally collected samples before and after Allo-HSCT and clinical data from two independent prospective cohorts (n=132) registered at ClinicalTrials.gov (NCT01751997) and CRIS (Clinical Research Information Service, KCT0002261), and investigated the role of NGS-MRD assessment. Donor groups consisted of matched sibling (23%), matched-unrelated (35%), and haploidentical familial donors (42%). Enrolled patients received myeloablative (MAC; 44%) or reduced-intensity conditioning (RIC; 56%). Customized amplicon-based targeted NGS including 67 genes (41 entire coding regions and 26 hot spots) was used and mean coverage was over 2000. With 33 months of median follow-up of survivors, 24 patients experienced post-transplant relapse. Persistent mutations were detectable at pre-HSCT (57/132, 43%) and at 1 month (23/114, 20%) after HSCT. Mutant allelic burdens at pre-HSCT and at 1 month after HSCT in relapsed patients were higher than in non-relapsed patients. Any persistent mutations at pre-HSCT and at 1 month after HSCT were significantly associated with post-transplant relapse (34.8% vs. 6.7%, p & lt;0.001 at pre-HSCT; 43.5% vs. 13.0%, p & lt;0.0001 at 1 month after HSCT) and worse overall survival (54.4% vs. 78.7%, p=0.010 at pre-HSCT; 44.9% vs. 76.8%, p=0.002 at 1 month after HSCT). NGS-MRD positivity was determined as complete mutational clearance by comparisons with various cutoffs of variant allele frequencies. Multivariate analysis confirmed that MRD positivity was an adverse prognostic factor for relapse and overall survival. Of note, optimal time points of NGS-MRD assay were different according to conditioning intensity. NGS-MRD detection at pre-HSCT was significantly associated with higher relapse in those who received MAC, while NGS-MRD detection at 1 month after HSCT was in those who received RIC. We also found that MRD positivity in genes related with clonal hematopoiesis were also significantly associated with post-transplant relapse. Serial NGS-MRD monitoring after HSCT revealed that most residual clones of false positive patients at pre-HSCT and at 1 month after HSCT were disappeared within 3 months after HSCT. At relapse, NGS showed not only clonal expansion or reappearing but also evolution of new clones. The current study demonstrated that NGS-MRD assessment both at pre-HSCT and at 1 month after HSCT were useful for predicting post-transplant relapse and there were no differences according to type of mutations. Optimal time points of NGS-MRD assessment depend on conditioning intensity. Disclosures Kim: Chugai: Consultancy, Honoraria; Daiichi Sankyo: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Hanmi: Consultancy, Honoraria; BL & H: Research Funding; AbbVie: Honoraria; Amgen Inc.: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; SL VaxiGen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; AML Global Portal: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Yuhan: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi-Genzyme: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    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. 112, No. 11 ( 2008-11-16), p. 1179-1179
    Abstract: Background : Transplantation-associated thrombotic microangiopathy (TMA) is infrequent but devastating complications after allogeneic hematopoietic stem cell transplantation (HSCT). However, there were no widely accepted criteria for the definition of TMA. Recently, the Blood and Marrow Transplants Clinical Trials Network (BMT CTN) and International Working Group (IWG) proposed a definition for TMA with some differences. The BMT CTN defined TMA as an increased number of schistocytes (3 2/high power field (HPF)) and elevated lactate dehydrogenase (LDH), concurrent renal and/or neurologic dysfunction, and negative Coombs’ test results. On the other hand, the IWG defined TMA by adding prolonged or progressive thrombocytopenia, a decrease in hemoglobin and serum haptoglobin concentration instead of concurrent renal and/or neurologic dysfunction and negative Coombs’ test results. In addition, the value of an increased number of schistocytes (34% or 3 8/HPF) for diagnosis of TMA by IWG is higher than that by BMT CTN (3 2/HPF). Until now, there has been no report to validate both criteria for TMA. Patients and Methods : We retrospectively examined 672 consecutive patients (median age 34 years, range 15–68) who underwent allogeneic HSCT from matched sibling donor (n = 442) and alternative donor (n = 230) at Catholic HSCT Center between January 2002 and December 2006 to analyze the role of some predicting factors for the incidence and outcome of TMA after HSCT and to validate two recently proposed criteria for TMA. We defined TMA as an increased number of schistocytes (3 2/HPF), elevated LDH, prolonged or progressive thrombocytopenia, a decrease in hemoglobin and serum haptoglobin concentration, and negative Coombs’ test results (O-TMA). O-TMA with renal and/or neurologic dysfunction was compatible with TMA defined by BMT CTN (B-TMA), and O-TMA with increased number of schistocytes over 8/HPF was compatible with TMA defined by IWG (I-TMA). TMA-associated death was defined as the interval from the onset of TMA to death which was associated with TMA without a relapse from underlying diseases after the onset of TMA. Results : Among 672 patients, 85 patients (13%) developed O-TMA including 41 patients (6%) with B-TMA and 17 patients (3%) with I-TMA. On multivariate analysis, TMA occurrence was higher in patients with older age (P = 0.001), mismatched human leukocyte antigen (P & lt; 0.001), and acute graft-versus-host disease (GVHD) 3 grade II (P & lt; 0.001) for O-TMA, in patients with older age (P = 0.002), advanced disease status at HSCT (P = 0.005), and acute GVHD 3 grade II (P & lt; 0.001) for B-TMA, and advanced disease status at HSCT (P = 0.017) and acute GVHD 3 grade III (P & lt; 0.001) for I-TMA. In patients with O-TMA, patients with renal dysfunction (hazard ratio 4.03, 95% CI 2.38–6.83, P & lt; 0.001) and/or concurrent veno-occlusive disease (hazard ratio 3.16, 95% CI 1.50–6.62, P = 0.002) proved to have significantly decreased overall survival by multivariate analysis. In addition, the presence of renal dysfunction was the only significant factor associated with increased TMA-associated death (hazard ratio 11.61, 95% CI 4.79–28.16, P & lt; 0.001). Maximal schistocytes count over 8/HPF and other factors including sex mismatch, acute GVHD 3 grade III, advanced disease status at HSCT, maximal LDH over 1298 IU/l, neurologic dysfunction, and concurrent cytomegalo virus infection that were significant on univariate analysis failed to show the significance relationship on multivariate analysis. Conclusions : This study identifies which patients are more prone to developing TMA after allogeneic HSCT according to three definitions for TMA (O-TMA, B-TMA and I-TMA). Notably, using multivariate analysis in patients with O-TMA, this retrospective study shows the importance of renal dysfunction compared to the maximal number of schistocytes count, which suggests that there are possible shortcomings of the definition by I-TMA that can miss out those with schistocytes count less than 8/HPF who may need early detection as a TMA. The proposed definitions of TMA need to be modified and be validated with prospective studies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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...