Mutational Spectrum Analysis of Interesting Correlation and Interrelationship Between RNA Splicing Pathway and Commonly Targeted Genes in Myelodysplastic Syndrome

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 273-273 ◽  
Author(s):  
Yasunobu Nagata ◽  
Masashi Sanada ◽  
Ayana Kon ◽  
Kenichi Yoshida ◽  
Yuichi Shiraishi ◽  
...  

Abstract Abstract 273 Myelodysplastic syndromes (MDS) are a heterogeneous group of myeloid neoplasms showing a frequent transition to acute myeloid leukemia. Although they are discriminated from de novo AML by the presence of a preleukemic period and dysplastic cell morphology, the difference in molecular genetics between both neoplasms has not been fully elucidated because of the similar spectrum of gene mutations. In this regards, the recent discovery of frequent pathway mutations (45∼90%) involving the RNA splicing machinery in MDS and related myeloid neoplasm with their rare mutation rate in de novo AML provided a novel insight into the distinct molecular pathogenesis of both neoplasms. Thus far, eight components of the RNA splicing machinery have been identified as the targets of gene mutations, among which U2AF35, SF3B1, SRSF2 and ZRSR2 show the highest mutation rates in MDS and CMML. Meanwhile, the frequency of mutations shows a substantial variation among disease subtypes, although the genetic/biological basis for these differences has not been clarified; SF3B1 mutations explain >90% of the spliceosome gene mutations in RARS and RCMD-RS, while mutations of U2AF35 and ZRSR2 are rare in these categories (< 5%) but common in CMML (16%) and MDS without increased ring sideroblasts (20%). On the other hand, SRSF2 mutations are most frequent in CMML (30%), compared with other subtypes (<10 %) (p<0.001) (Yoshida K, et al, unpublished data). So to obtain an insight into the genetic basis for these difference, we extensively explored spectrums of gene mutations in a set of 161 samples with MDS and related myeloid neoplasms, in which mutations of 10 genes thus far identified as major targets in MDS were examined and their frequencies were compared with regard to the species of mutated components of the splicing machinery. The mutation status of the 161 specimens was determined using the target exon enrichment followed by massively parallel sequencing. In total, 86 mutations were identified in 81(50%) in the 8 components of the splicing machinery. The mutations among 4 genes, U2AF35 (N = 20), SRSF2 (N = 31), SF3B1 (N = 15) and ZRSR2 (N = 10), explained most of the mutations with a much lower mutational rate for SF3A1 (N = 3), PRPF40B (N = 3), U2AF65 (N = 3) and SF1 (N = 1). Conspicuously, higher frequency 4 components of the splicing machinery were mutated in 76 out of the 161 cases (47.2%) in a mutually exclusive manner. On the other hand, 172 mutations of the 10 common targets were identified among 117, including 41 TET2 (25%), 32 RUNX1 (20%), 26 ASXL1 (16%), 24 RAS (NRAS/KRAS) (15%), 22 TP53 (14%), 17 IDH1/2 (10%), 10 CBL (6%) and 10 EZH2 (6%) mutations. We examined the difference between the major spliceosome mutations in terms of the number of the accompanying mutations in the 10 common gene targets. The possible bias from the difference in disease subtypes was compensated by multiple regressions. The SRSF2 mutations are more frequently associated with accompanying gene mutations with a significantly higher number of those mutations (N=29; OR 6.2; 95%CI 1.1–35) compared with that of the U2AF35 mutations (N=14) (p=0.038). Commonly involving the E/A splicing complexes, these splicing pathway mutations lead to compromised 3' splice site recognition. However, individual mutations may still have different impacts on cell functions, which could contribute to the determination of discrete disease phenotypes. It was demonstrated that SRSF2 was involved in the regulation of DNA stability and that depletion of SRSF2 can lead to DNA hypermutability, which may explain the higher number of accompanying gene mutation in SRSF2-mutated cases than cases with other spliceosome gene mutations. In conclusion, it may help to disclosing the genetic basis of MDS and related myeloid neoplasms that highly paralleled resequencing was confirmed SRSF2 mutated case significantly overlapped common mutations. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 18-18
Author(s):  
R. Coleman Lindsley ◽  
Brenton G. Mar ◽  
Emanuele Mazzola ◽  
Peter Grauman ◽  
Sarah Shareef ◽  
...  

Abstract Acute myeloid leukemia (AML) is a biologically heterogeneous disease with marked variability in treatment sensitivity and long-term clinical outcomes. Along with age and cytogenetics, disease ontogeny is a powerful predictor of prognosis in AML patients, reflected by reduced likelihood of remission and shorter overall survival in patients who develop AML after exposure to leukemogenic therapies (therapy-related AML; t-AML) or after antecedent MDS (secondary AML; s-AML). We performed targeted deep sequencing of 82 genes on diagnostic samples from 194 patients enrolled on the ACCEDE trial, a phase 3 study of induction chemotherapy in s-AML and t-AML, the largest reported prospective data set in this patient population. Mutations in TP53 (HR 1.86; 1.21, 2.86) and RAS pathway genes (HR 1.63; 1.13, 2.33) were associated with reduced overall survival. To evaluate the genetic basis of distinct AML ontogenies, we identified mutations in this rigorously-defined s-AML cohort and compared them to mutations identified in 180 cases of non-M3 de novo AML reported in The Cancer Genome Atlas. Mutations in eight genes were >95% specific for s-AML, including SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, and STAG2 (termed “secondary-type” mutations); three alterations were >95% specific for de novo AML, including NPM1 mutation, CBF rearrangement, and 11q23 rearrangement. All other mutations were not specific to either AML subtype (termed “pan-AML” mutations). We asked whether an ontogeny-based genetic classification of AML could define subgroups among patients within the heterogeneous category of t-AML. Among subjects with clinically defined t-AML, 33% (34/101) harbored secondary-type mutations. Characteristics of this genetically defined subset of t-AML patients (age, sex, number of mutations, percentage with chromosome 5 or 7 abnormalities) were indistinguishable from patients with clinically defined s-AML without prior exposure to leukemogenic therapy. The proportion of t-AML patients with secondary-type mutations increased with age, comprising 8% of patients under 40 years old, 23% of those between 40 and 70 years old, and 61% of those over 70 years old. 47% of patients with clinically defined t-AML had only de novo/pan-AML alterations. Compared to those with secondary-type mutations, these patients were younger, had a female predominance, and had fewer mutations. In this t-AML subgroup, the frequency of mutations in NPM1 (28%), FLT3 (23%), DNMT3A (30%), TET2 (13%), and IDH1/2 (13%) was similar to that of de novo AML without exposure to leukemogenic therapy; RAS pathway mutations (41% vs. 19%) were more common. TP53 mutations were present in 23% of t-AML cases and associated with complex and monosomal karyotypes. To evaluate the genetic basis of disease progression and treatment resistance, we analyzed serial samples from a subset of s-AML subjects. At AML transformation, 59% acquired new driver mutations not present in a paired MDS sample. Of these new mutations, 74% involved genes encoding regulators of transcription or signal transduction; mutations in TP53 and genes involved in epigenome modification or RNA splicing were rarely gained. After induction chemotherapy, 69% of patients who achieved complete remission still had detectable persistence of disease-driving mutations. In nearly half of these cases, we observed a dichotomous pattern at remission, whereby some mutations became undetectable and others persisted at high allele fraction (mean = 26%), suggesting selective clearance of chemosensitive, blast-associated subclones and persistence of a chemoresistant founder clone. In this context, mutations in genes involved in signaling and transcriptional regulation were preferentially lost, while mutations in TP53 and genes involved in epigenome modification and RNA splicing were preferentially retained. In conclusion, by performing genetic analysis across a spectrum of AML subtypes, we uncover ontogeny-defining alterations in AML that enable objective classification of individual AMLs into distinct genetic subgroups. Application of this ontogeny-based genetic classification resolves t-AML into distinct clinical categories and raises questions about the pathogenesis of therapy-related leukemogenesis. Lastly, through serial genetic assessment at key clinical timepoints we identify associations between genetic pathways and disease evolution. Disclosures Stone: Celator: Consultancy; Sunesis: Steering Committee, Steering Committee Other; Celgene: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 458-458 ◽  
Author(s):  
Kenichi Yoshida ◽  
Masashi Sanada ◽  
Yuichi Shiraishi ◽  
Daniel Nowak ◽  
Yasunobu Nagata ◽  
...  

Abstract Abstract 458 MDS and related disorders comprise a group of myeloid neoplasms characterized by deregulated blood cell production and a predisposition to AML. Although currently, a number of gene alterations have been implicated in the pathogenesis of MDS, they do not fully explain the pathogenesis of MDS, because many of them are also found in other myeloid malignancies and roughly 20% of MDS cases have no known genetic changes. So, in order to clarify a complete registry of gene mutations in MDS and identify those discriminate MDS from other myeloid neoplasms, we performed whole-exome sequencing of 29 cases showing myelodysplasia. A total of 268 somatic mutations or 9.2 mutations per sample were identified. Among these 41 occurred in recurrent gene targets, which not only included a spectrum of known gene targets in MDS, such as TET2, EZH2, NRAS/KRAS, RUNX1, TP53 and DNMT3A, but also affected previously unknown genes that are commonly mapped to the RNA splicing pathway, including U2AF35, SRSF2 and ZRSR2. Together with additional three (SF3A1, SF3B1 and PRPF40B) found in single cases, 16 (55.2%) of the 29 discovery cases carried a mutation affecting the component of the splicing machinery. To confirm the observation, we examined 9 spliceosome genes for mutations in a large set of myeloid neoplasms (N=582) using a high-throughput mutation screen of pooled DNA followed by confirmation/identification of candidate mutations. In total, 219 mutations were identified in 209 out of the 582 specimens of myeloid neoplasms. Mutations of the splicing machinery were highly specific to diseases showing myelodysplastic features, including 19 of 23 (83%) cases with RARS, 43 of 50 (86%) RCMD-RS, 68 of 155 (44%) other MDS, 48 of 88 (55%) CMML, and 16 of 62 (26%) secondary AML with MDS features with a string preference of SF3B1 mutations to RARS and RCMD-RS and of SRSF2 to CMML, while they were rare in cases with de novo AML (N=151) and MPD (N=53). The mutations among 4 genes, U2AF35 (N = 37), SRSF2 (N = 56), SF3B1 (N = 79) and ZRSR2 (N = 23), explained most of the mutations with a much lower mutational rate for SF3A1 (N = 8), PRPF40B (N = 7), U2AF65 (N = 4) and SF1 (N = 5). Interestingly, mutations in the former three genes showed clear hot spots, indicating a gain-of-function nature of these mutations. On the other hand, two thirds of the ZRSR2 mutations are nonsense or frameshift changes causing premature truncation of the protein. Significantly, these mutations occurred in an almost completely mutually exclusive manner among mutated cases, and commonly affected those components of the splicing complex that are engaged in the 3' splice site recognition during RNA splicing, strongly indicating production of unspliced or aberrantly spliced RNA species are incriminated for the pathogenesis of MDS. In fact, when transduced into HeLa cells, the recurrent S34F U2AF35 mutant induced the increase in the production of unspliced RNA species and elicited the activation of the nonsense mediated decay pathway. Functionally, the U2AF35 mutants seemed to cause deregulated stem cell functions, because CD34(−) KSL cells transduced with various U2AF35 mutants invariably showed reduced chimerism in competitive reconstitution assay. In accordance with this, the S34F U2AF35 mutant lead to suppression of cell growth in a variety of cell types, including HeLa cells, in which expression of the mutant induced a G2/M cell cycle arrest and increased apoptosis. In conclusion, whole-exome sequencing unexpectedly revealed the high frequency of the splicing pathway mutations in MDS and related myeloid neoplasms, providing the first evidence indicating that compromised RNA splicing by gene mutations are responsible for human pathogenesis. Disclosures: Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4132-4132
Author(s):  
SuXia Geng ◽  
Xin Du ◽  
Jianyu Weng ◽  
Liye Zhong ◽  
Rong Guo ◽  
...  

Abstract Abstract 4132 The objective was to definite the expression level of aven mRNA of white blood cells from peripheral blood(PB)of de novo acute myeloid leukemia and preliminary analyze its clinical significance, providing a experimental basis for evaluating prognosis. Aven mRNA levels in PB samples from 69 AML patients were detected by using real-time quantitative PCR. The relation of aven mRNA level with clinical and hematological characteristics (age, sex, WBC, Hb, Plt, LDH, Blast% in PB and BM,FAB subtype) and treatment outcome (complete remission rate and relapse rate)were analyzed. Twenty-one normal individuals served as controls. The level of aven mRNA was between 11.72% and 178.93 %(median 37.2%) in de novo AML and between 10.81% and 50.98 %(median 28.81%) in normal individuals. Aven mRNA level was higher in the AML group than in the controls (p=0.006). When we compared aven mRNA with other clinical and hematological parameters, there were significant correlations between aven mRNA and age(r=0.25,p=0.039),aven mRNA and hemoglobin level (r=0.29,p=0.019),aven mRNA and FAB subtype(r=0.253,p=0.036). We found that median level of aven mRNA in group whose age older than median age was higher than group whose age younger than median level(p=0.018).The complete remission rate after two cycles chemotherapy in group with lower aven mRNA level(25/30,83.33%)was higher than group with higher aven mRNA level(21/30,70%). But the difference was not significant(p=0.22).The difference of aven mRNA expression level between AML patients with relapse and that without relapse was not significant (p=0.076). In conclusions, the level of aven mRNA in de novo AML is overexpression. The overexpression of aven mRNA is likely to play an important role in tumorigenesis of AML. Association of aven mRNA expression with treatment outcome and relapse was not observed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 289-289 ◽  
Author(s):  
Marius Bill ◽  
Deedra Nicolet ◽  
Ann-Kathrin Eisfeld ◽  
Krzysztof Mrózek ◽  
Christopher J. Walker ◽  
...  

Abstract Introduction: Prognosis of AML pts is still poor mainly because of refractoriness to or relapse after intensive chemotherapy. High rates of relapse are also attributed to LSCs, which are a small subset of cells with acquired abnormal self-renewal capacity and increased resistance to chemotherapy. A better understanding of LSCs is critical to improve outcomes of pts with AML. Ng et al. (Nature 2016;540:433) defined a 17 stemness-associated gene score that was highly prognostic. Aims: The aim of this study was to validate the prognostic relevance of the 17-gene LSC score and explore its utility in the context of the ELN classification. We also examined gene mutations associated with the 17-gene LSC score. Methods: We analyzed a total of 934 pts [729 aged <60 years (y) and 205 aged ≥60 y] with de novo AML. We used whole transcriptome expression data (RNAseq) to calculate the aforementioned 17-gene LSC score for each pt in our cohort. Similar to Ng et al., we used the median of the whole cohort to discriminate between pts with LSChigh and LSClow scores. The mutational status of 80 cancer- and leukemia-associated genes (Eisfeld et al. Leukemia 2017;31:2211) were determined using a targeted next-generation sequencing panel, CEBPA mutations using Sanger sequencing, and an internal tandem duplication (ITD) of the FLT3 gene using fragment analysis in pretreatment bone marrow or blood samples. All pts were treated on frontline Cancer and Leukemia Group B/Alliance protocols. Results: A comparison of pretreatment clinical and genetic features revealed that LSChigh pts were older (P<.001; median age, 53 vs 46 y) and had higher platelet counts (P<.001; median, 63 vs 50x109/L) than LSClow pts. Pts with a LSChigh score more frequently had FLT3-ITD (P<.001) and mutations in the ASXL1 (P=.001), DNMT3A (P<.001), RUNX1 (P=.002), SRSF2 (P=.02), STAG2 (P=.009), TET2 (P=.008) and TP53 (P<.001) genes. Conversely, these pts had a lower frequency of biallelic CEBPA (P<.001), GATA2 (P=.008) and KIT (P<.001) mutations. Because of differences in treatment intensity, we analyzed outcomes of younger and older pts separately. Younger pts with a LSChigh score had a lower complete remission (CR) rate (P<.001; 63% vs 87%), shorter disease-free survival (DFS; P<.001; 3-y rates, 26% vs 48%; Figure 1A) and overall survival (OS; P<.001; 3-y rates, 27% vs 59%; Figure 1B) compared to those of LSClow pts. In multivariable analyses including clinical and genetic factors that impact on outcome, a LSChigh score associated with lower remission rates (P<.001; HR: 0.36), shorter DFS (P<.001; HR: 1.67) and OS (P<.001; HR: 1.88) after adjusting for other co-variates. We also analyzed the prognostic impact of the LSC score with respect to the 2017 ELN classification. We found that LSC score associated with different ELN groups (P<.001), with LSChigh pts being more often classified in the Adverse or Intermediate group and less often in the Favorable group. Within the ELN Favorable and Adverse groups, LSChigh score retained its prognostic impact and identified pts with a lower CR rate and shorter DFS and OS (Table1). In older pts, a LSChigh score also associated with lower CR rate (P=.004; 50% vs 72%), shorter DFS (P=.04; 3-y rates, 6% vs 17%; Figure 1C) and OS (P<.001; 3-y rates, 9% vs 27%; Figure 1D). In multivariable analyses, LSC score remained significant only for OS (P<.003; HR: 1.70) after adjusting for other co-variates. Regarding the ELN classification, pts with LSChigh score in the Favorable group had shorter OS (P=.05; 3-y rates, 17% vs 50%) and, by trend, shorter DFS (P=.09; 3-y rates, 17% vs 39%); no significant differences were found in Intermediate or Adverse groups. Conclusions: We used RNAseq expression data and applied the previously established 17-gene LSC signature to score 934 de novo AML pts. We detected distinct mutational differences between LSChigh and LSClow pts, with LSChigh pts more often carrying gene mutations associated with age-related clonal hematopoiesis (i.e., ASXL1, DNMT3A, TET2, SRSF2 and TP53 mutations). Moreover, this score, derived from the expression of stemness-associated genes, has not only a prognostic impact on its own but also in the context of the current 2017 ELN classification. Disclosures Kolitz: Magellan Health: Consultancy, Honoraria. Powell:Rafael Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4608-4608
Author(s):  
June-Won Cheong ◽  
Yuri Kim ◽  
Sun Young Park ◽  
In Hae Park ◽  
Jin Seok Kim ◽  
...  

Abstract The incidence of acute myeloid leukemia (AML) increases with age. Because of poor performance status, co-morbidity and treatment-related side effects, a conventional dose chemotherapy containing anthracyclins may be toxic to the majority of elderly patients. In contrast, the administration of suboptimal dose of myelosuppressive chemotherapy could lead to an unsuccessful clinical outcome including lower complete remission (CR) rate. To evaluate the effect of attenuated dose of idarubicin, compared to the standard dose, on the clinical outcome and chemotherapy-related complications, we analyze the consecutive 32 elderly de novo AML patients (range, 60 – 74 years) with normal karyotype. Eleven patients received one cycle of conventional-dose remission induction chemotherapy (idarubicin, 12 mg/m2/day on days 1–3 and cytarabine 100mg/m2/day on days 1–7) (Group 1) and 21 patients received attenuated-dose idarubicn (8 mg/m2/day on days 1–3) and cytarabine (100mg/m2/day on days 1–7) (Group 2). Six cases (54.5%) in Group 1 and 12 cases (57.1%) in Group 2 had CR. The difference of CR between the two groups was not significant (P = 0.59). The intervals from the chemotherapy-starting date to the date of CR documentation were not also different between two groups (median 31.5 days on Group 1 vs 27.0 days on Group 2) (P = 0.29). The median number of transfusion requirement during the induction therapy was not different in the red blood cells (10 units, each) and platelets (16.5 units in Group 1 vs 18.0 units in Group 2; P > 0.05). Thirty patients received the recombinant human granulocyte colony-stimulating factor (G-CSF) three days after termination of chemotherapy. The duration of G-CSF administration was not different between two groups (P = 0.86). However, the frequency of septicemia and septic shock after induction chemotherapy was statistically significantly higher in Group 1 (54.5% and 9.5%, respectively) compared to that in Group 2 (36.3% and 0.5%, respectively) (P < 0.01). We also observed a higher incidence of clinically-documented invasive fungal infection in Group 1 (45.5%) compared to Group 2 (15.0%), although the difference was not statistically significant (P = 0.095). The incidence of other regimen-related toxicities including renal dysfunction, hepatic dysfunction and heart failure was not different between two groups. Overall survival and disease-free survival also were not different between the groups. In conclusion, the attenuated dose of idarubicin can be recommended for the remission induction chemotherapy for the elderly de novo AML patients with normal karyotype since it is associated with lower incidence of sepsis and septic shock with comparable CR rate.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 318-318
Author(s):  
Thorsten Klampfl ◽  
Ashot Harutyunyan ◽  
Tiina Berg ◽  
Bettina Gisslinger ◽  
Francesco Passamonti ◽  
...  

Abstract Abstract 318 The classical myeloproliferative neoplasms (MPNs) comprise of three entities: polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). Despite distinct phenotypic features of MPN entities they share characteristics like clonal hematopoiesis, risk for thrombosis and bleeding and tendency to transform to secondary acute myeloid leukemia (post-MPN AML). In order to investigate the genetic lesions associated with MPN a large single-center cohort of 311 MPN patients was analyzed for chromosomal aberrations using high resolution Affymetrix SNP 6.0 arrays. The cohort included 150 patients with PV, 90 with ET, 68 with PMF and 3 with post-MPN AML. Of the 311 patients, 144 (46%) had a normal karyotype and 167 (54%) harbored 1 to 8 detectable chromosomal aberrations. We found 51 gains, 102 deletions and 143 uniparental disomies (UPDs). A total of 13 recurrent chromosomal defects (more than three events) were detected. We investigated if either the number of chromosomal aberrations in a patient or specific types of lesions associate with a certain patient group defined by clinical criteria. Chromosomal aberrations were equally distributed among the three MPN entities and only 9pUPD showed significant clustering with PV. We did not detect an association between the number of chromosomal aberrations and disease duration. Patients positive or negative for JAK2 mutations did not differ significantly in the frequency of chromosomal aberrations (except of the association of 9pUPD with JAK2 positive MPN). Patients with complex karyotype were significantly older than patients with normal karyotype (P<0.001). Transformation to post-MPN AML is an important complication in MPN. To investigate associations between chromosomal changes and transformation, we included additional 19 post-MPN AML patients from another center into the study (total N=22). Patients in the post-MPN AML group harbored significantly more chromosomal lesions (P<0.001). Recurrent aberrations of chromosomes 1q, 7p, 7q, 5q, and 3q strongly associated with post-MPN AML. When we reviewed the clinical data of patients in chronic phase MPN harboring the leukemia-associated aberrations, they showed features of disease progression, and some transformed to AML at a later follow-up. We were able to map a common deleted region (CDR) on chromosome 4 to the tet oncogene family member 2 (TET2), a gene frequently deleted in myeloid disorders. On chromosome 7p we mapped a CDR to the Ikaros transcription factor (IKZF1) and a 7q CDR mapped to a novel putative tumor suppressor, the cut-like homeobox 1 gene (CUX1). Interestingly, in one patient who carried a UPD of chromosome 7q we did not detect a mutation in the CUX1 gene but an R288Q mutation was found in the EZH2 gene. Chromosome 7 aberrations in our cohort were strongly linked to post-MPN AML. Our results show that at least three chromosome 7 genes (IKZF1, CUX1, and EZH2) are relevant in leukemic transformation. In addition to chromosome 7, we found gains of chromosome 1q equally relevant in post-MPN AML. We mapped the common 1q amplification to a 3.5 Mbp region that contained the MDM4 gene. Mdm4 is a known negative regulator of p53 and was frequently shown amplified in various cancers. This result prompted us to investigate the relevance of the p53 pathway in post-MPN AML and we sequenced TP53 in all 22 leukemic patients and found mutations in 6 cases (27.3%). Interestingly, none of the patients with TP53 mutation carried an MDM4 amplification. Taken together, 10 out of 22 post-MPN AML cases (45.5%) had evidence of a p53-related defect. To gain deeper insight into the pathways involved in transformation to post-MPN AML we sequenced genes commonly affected in de novo AML, and found two patients with mutations in FLT3, two patients with RUNX1 mutations, two patients with either IDH1 or IDH2 mutations. We conclude that lesions known to play an important role in de novo AML are present only in a fraction of post-MPN AML patients. In this study we show that aberrations of the p53 pathway together with the chromosome 7 lesions affecting IKZF1 and CUX1 are present in 64% of all post-MPN AML patients. Our data give insight into the genetic complexity and heterogeneity of MPN patients in chronic phase as well as in post-MPN AML. The marked genetic heterogeneity of MPN patients will render targeted therapies challenging and underlines the requirement of personalized treatments. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2168-2168
Author(s):  
Yuho Najima ◽  
Daichi Sadato ◽  
Yuka Harada ◽  
Chizuko Hirama ◽  
Keisuke Oboki ◽  
...  

Abstract Background Although the outcome after allogeneic hematopoietic stem cell transplantation (Allo-HSCT) for the patients with acute myeloid leukemia (AML) in complete remission has been improved, the prognosis of patients with active disease is still dismal, generally with 20 - 30% of overall survival (OS) at 2 years. Prognostic value of gene mutations detected by the next generation sequencing (NGS) for this extremely poor group remains to be evaluated. Methods A total of 120 patients with AML not in hematological remission who received the first allo-HSCT at our institute between April 2005 and December 2017 were enrolled. For each patient, genomic DNA was extracted from the frozen bone marrow sample harboring leukemic blasts which was preserved at the nearest available date before the initiation of conditioning regimen. Sequencing was performed using TruSight Myeloid Sequencing Panel® on the MiniSeq system (Illumina). Gene variants were detected by in-house analysis pipeline. Charts were retrospectively reviewed on survival, relapse, and non-relapse mortality (NRM). The Kaplan-Meier method was used to assess OS using the log-rank test. Univariate and multivariate analysis were performed to identify potential prognostic factors. The Cox proportional hazards method was used for the multivariate analysis to assess OS. Gray's test and the Fine-Gray test were used to assess the cumulative incidence of relapse (CIR). Competing risks were relapse and NRM. Results Median follow-up of survivors was 1345 days (235 - 4888 days). Median age at transplant was 51 (range 21 - 71). Grafts were from bone marrow (n = 67, 55.8%), peripheral blood (n=42, 35.0%) and cord blood (n=11, 9.2%). Refined disease risk index (Blood. 2014;123:3664-71) scored high (n=61, 51.3%) and very high (n=58, 48.7%). OS at 2 years of the whole cohort was 27.3% (95% confidence interval [CI], 19.4% - 35.7%). There was no significant difference in OS between patients in primary induction failure and in relapse (OS at 2 years: 26.5% [n=50] vs 28.7% [n=70], p= 0.293). NGS analysis revealed TP53 loss-of-function mutation in 23 (19.2%) patients. Among all detected gene mutations, TP53 mutation was the most powerful predictor of poor OS after allo-HSCT (OS at 2 years: 13.5% vs 30.5% for TP53+ [n=23] vs TP53- [n=97], p= 0.0184). Consistent with previous reports, monosomal karyotype (MK, J Clin Ocnol 2010:26;4791-7) was significantly associated with positive TP53 mutation (13.3% of non-MK vs 37.9% of MK, p=0.006). Of note, all the patients (n=11) positive for both prognostic factors died within 1 year after allo-HSCT, whereas OS of the patients without either factor (n=78) was 33.6% at 2 years. Multivariate analysis on OS revealed MK, TP53 mutation, de novo AML (no prior history of MDS), ECOG performance status score 2 or more, C reactive protein 1.0 mg/dL or more, peripheral blood blast frequency of 1% or more at the initiation of conditioning regimen were independent prognostic factors for poor OS. Among them, to determine the prognostic factors critical for deciding the indication of allo-HSCT, we chose pre-transplant factors which were available around one months before transplant. From this point of view, multivariate analysis revealed independent prognostic factors for poor OS after allo-HSCT including MK (Hazard ratio [HR] 2.05; 95% confidence interval [CI] 1.30 - 3.25, p=0.00217), TP53 mutation (HR 1.72; 95% CI 1.04 - 2.86, p=0.035), and de novo AML (HR 1.67; 95% CI 1.04 - 2.86, p=0.036). Because the HR of these three factors were comparable, a score of 1 was assigned to each factor. OS at 2 years was 49.7%, 26.5% and 13.1% for patients with low (score 0, n=23), intermediate (score 1, n=63) and high (score 2 or 3, n=34) risk, respectively (p<0.001, Figure 1A). CIR was significantly lower in low risk group compared to intermediate and high risk groups (9.0%, 49.7% and 48.0% at 2 years, p=0.0128, Figure 1B), whereas NRM was comparable (41.3%, 32.5% and 41.7% at 2 years, p =0.515). Conclusions NGS-analysis revealed the importance of TP53 mutation on transplant outcome of AML in non-remission. The combined score with the information on MK, de novo AML, and TP53 mutation stratified the patients into three groups, including low risk with 50% survival rate, and high risk with shorter survival after allo-HSCT for whom the other strategy might be optimal. Our results would require further validation in larger cohorts. Disclosures Harada: Celgene: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1008-1008 ◽  
Author(s):  
Der-Cherng Liang ◽  
Lee-Yung Shih ◽  
Ming-Chung Kuo ◽  
Chao-Ping Yang ◽  
Hsi-Che Liu ◽  
...  

Abstract Background and purpose: The Mixed-lineage leukemia (MLL) gene located at 11q23 can be fused to a variety of partner genes through chromosomal translocations (MLL-T) in acute leukemia. The co-existence of commonly known mutated genes has not been comprehensively studied in MLL-T AML. We aimed to determine (1) the prevalence and clinical relevance of gene mutations at diagnosis, and (2) the genetic evolution profile at relapse in de novo AML with MLL-T. Materials and methods: Bone marrow samples from 80 de novo AML with MLL-T patients were analyzed on 20 gene mutations involving activating signaling pathway (class I), transcription pathway (class II), epigenetic regulators (class III), and tumor suppressors, TP53 and WT1 (class IV). MLL-T was all confirmed by FISH analysis. The common MLL fusion transcripts were detected by RT-PCR, including 26 MLLT3, 13 MLLT10, 12 MLLT4, 11 ELL, 3 MLLT1, 3 AFF1, and 2 EPS1. Another 6 infrequent or rare partner genes (one each of MLLT6,CBL, ARHGEF12, TET1, SEPT6, and SEPT9) were identified by cDNA panhandle PCR. In the remaining 4 cases, the partner genes were not identified. Mutational analyses were performed with PCR-based assays followed by direct sequencing. Twenty-two of 26 patients who relapsed had relapse samples for comparative analysis. Results: The frequencies of gene mutations of class I, II, III, and IV, in 80 de novo MLL-T AML patients were 49.4%, 3.8%, 10.3% and 1.3%, respectively. Together, 53.8% of patients with MLL-T had at least one mutated gene. KRAS (17.7%), FLT3-TKD (11.5%), and NRAS (11.4%) mutations occurred most frequently. Among the epigenetic regulators, 7.9% of patients had DNMT3A, 2.7% TET2, 1.4% ASXL1, and none had IDH1/2 or EZH2 mutations. Three patients had 3 co-existing mutations and 9 patients had two mutations, with 6 of the 12 carrying FLT3-TKD mutations. Gene mutations of transcription pathway and tumor suppressors rarely occurred, only involving NPM1 in two cases and one each for RUNX1 and WT1 mutation. The patients with DNMT3A mutations were significantly associated with older age (P=0.005), FAB AML-M4 (P<0.0001), and higher circulating monocytes (P=0.022). Patients with NRAS mutations had lower circulating monocytes (P<0.0001). KRAS-mutated patients had a higher percentage of marrow leukemic cells (P=0.002). No differences were observed between clinico-hematological features and mutation status of other genetic subtypes. Of the 65 MLL-T AML patients who received standard chemotherapy, 53 achieved complete remission. There were no differences in the outcomes between AML patients with MLLT3-MLL and MLL-MLLT10; patients with both subtypes had a superior event-free survival compared to those with other MLL-fusion transcripts (P=0.036) and a trend towards favorable overall survival (P=0.066). We failed to find significant differences between outcomes and the mutation status of each functional class. All the 3 patients carrying FLT3-ITD had an overall survival less than 6 months. Of the 22 paired diagnosis and relapse samples, 3 patients retained the same NRAS mutations at relapse whereas another one changed from G12D to G12C at relapse. Another 3 relapsed with identical KRAS mutations; while one acquired KRAS mutation at relapse. Two patients had stable DNMT3A mutations at both phases. All the 4 patients who harbored FLT3-TKD mutations at diagnosis lost the mutations at relapse. PTPN11 mutation was found in only one case at diagnosis, while none at relapse. One each had identical mutated gene of CBL and WT1 at both diagnosis and relapse. One each acquired TP53 and WT1 at relapse. Conclusions: Our results showed that co-existing mutations of RAS and FLT3-TKD were most frequently detected at diagnosis in de novo MLL-T AML. Gene mutations, except FLT3-ITD, had no impact on outcomes in MLL-T AML. FLT3-TKD mutations were absent in all relapse samples while clonal evolution of WT1, TP53 or KRAS mutations might contribute to the relapse of leukemia in a subset of MLL-T AML patients. Grant support: NHRI-EX93-9011SL, NHRI-EX96-9434SI, NSC95-2314-B-195-001, NSC96-2314-B-195-006-MY3, NSC97-2314-B-182-011-MY3, MMH-E-101-09 and OMRPG3C0021. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3592-3592
Author(s):  
Satoshi Wakita ◽  
Hiroki Yamaguchi ◽  
Yoshio Mitamura ◽  
Fumiko Kosaka ◽  
Takashi Shimada ◽  
...  

Abstract Abstract 3592 Introductions: Gene mutationswere found in acute myeloid leukemia (AML) and their importance has been noted. Flt3, NPM1 and CEBPA were detected in “intermediate cytogenetic risk” group, and are becoming possible to distinguish subsets of patients with different outcomes. Moreover, several groups have reported that these mutations would be useful for not only predictive markers, but also minimal residual disease (MRD) markers in AML. Several recent studiesprovided compelling evidence that mutations in epigenetics modifying genes contribute to AML pathogenesis. DNMT3a mutations were common (about 20% frequency) in de novo AML and associated with poor prognosis. Furthermore, mutations of IDH1/2 and TET2 that also seem to be collaborating on DNA methylation modifying are detected, too. To clarify the importance and dynamics of these mutations in clinical course, we examined Flt3, NPM1, CEBPA, DNMT3a and IDH1/2 gene mutations in paired samples at initial presentation and relapse of AML patients. Materials and Methods: We analyzed the samples from adultpatients with de novo AML diagnosed at Nippon Medical School Hospital from 2000 to 2010. Mutation analyses were performed for Flt3 ITD by PCR amplification, Flt3 TKD by PCR-RFLP, and NPM1, CEBPA, IDH1/2 and DNMT3a mutations by direct sequence. Results: The 31 AML patients were enrolled. In chromosomal analysis at initial presentation, 19 with normal karyotype (NK-) AML, 2 with trisomy8, 4 with 11q23 associated, 1 with monosomy7, 2 with complex karyotypes and 3 with non-specific aberrations were observed. 15 cases were comparable for paired samples at diagnosis and relapse. 13 of them (86.7%) showed additional chromosomal aberrations at relapse. Gene mutations were detected more frequently in cytogenetic intermediate risk group (83.3%) than poor risk group (0%). There were 11 patients with Flt3 ITD at initial presentation, but 3 of them had no detectable mutation at relapse. Flt3 TKD were found in 3 patients at initial presentation, but all of them were lost at relapse. Among 12 patients with NPM1 mutation at initial presentation, 3 of them lost their mutation at relapse. CEBPA mutation was detected in only one paired sample at diagnosis and relapse. DNMT3a mutations were detected in 8 patients both at initial presentation and relapse. IDH2 mutations were detected in two patients at initial presentation, but 1 of them was lost at relapse. In summary, of the 37 gene mutations at initial presentation, 10 gene mutations were lost at relapse, and only 1 acquired gene mutation was detected at relapse. Flt3 ITD, NPM1, DNMT3a and IDH2 mutations frequently coexisted with another mutation. Discussion: This study is the first report of consecutive analyses on the major gene mutations in AML. Newly acquired gene mutations at relapse are rare compared to frequent additional chromosomal aberrations at relapse. Flt3 ITD mutations at initial presentation were detected also at relapse. This finding indicates that Flt3 ITD are responsible for relapse and refractoriness. On the other hand, all 3 cases with Flt3 TKD lost the mutation at relapse, suggesting that Flt3 TKD mutation does not contribute to their relapse. Some of Flt3 ITD, Flt3 TKD and NPM1 mutations could not be detected at relapse, indicating that these mutations should be used carefully for MRD marker. DNMT3a mutations were detected both at diagnosis and relapse in all 8 cases. This finding suggests that DNMT3a mutations might be a useful MRD marker. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2537-2537
Author(s):  
Takeshi Ryotokuji ◽  
Hiroki Yamaguchi ◽  
Ikuko Omori ◽  
Tuneaki Hirakawa ◽  
Satoshi Wakita ◽  
...  

Abstract Abstract 2537 Background and Aims: Several gene mutations were found in acute myeloid leukemia (AML) and were expected to be used as prognostic factors. The nucleophosmin 1 (NPM1) and CCAAT/enhancer-binding protein alpha (CEBPA) gene mutations are associated with a favorable outcome and lack of a transplant benefit, but FMS-like tyrosine kinase 3 (FLT3) mutations leading to an internal tandem duplication (ITD) are associated with adverse outcome. Recently the DNA methyltransferase 3A (DNMT3A) gene mutation was identified by whole-genome sequencing in patients with AML. DNMT3A encodes for the enzyme DNA (cytosine-5)-methyltransferase 3A and belongs to the family of other methyltransferases like DNMT1 and DNMT3B. These enzymes are involved in adding methyl groups to the cytosine residue of CpG dinucleotides and thus play an important role in epigenetic regulation of genes, but the mechanism of DNMT3A mutation -associated leukemogenesis was still unknown. About clinical impact of DNMT3A mutation, several groups reported that DNMT3A mutation have been associated with adverse outcome. We analyzed clinical features and prognostic impact of DNMT3A mutation in patients with Japanese patients with AML. Methods: We retrospectively analyzed 188 cases of de novo AML treated at Nippon Medical School Hospital and its affiliated facilities from 2000 to 2010. We analyzed 188 samples at initial presentation and 70 samples at relapse. Mutation analyses were performed for FLT3ITD by PCR amplification, and NPM1, CEBPA, and DNMT3A mutations by direct sequence. To validate sequencing results, PCR products were inserted into the pCR2.1-TOPO vector using a TOPO TA cloning kit (Invitrogen, Carlsbad, CA). Recombinant plasmids isolated from 8 to 12 white colonies were sequenced. Results: The median age was 53.0 years (range, 17–87 years) with 62.1% being males. Patients were followed-up for 0.03–126.1 months after initial presentation, with a median of 19.9 months. DNMT3A mutations were detected in 35 cases (18.6%) at initial presentation, and 12 cases (17.1%) at relapse. Most frequently, codon R882 located in exon 23 was mutated, and 32 cases (91.4%) of these mutations were located within methyltrasferase domain. We evaluated the correlation of clinical and genetic characteristics with DNMT3A mutations. Age (p=0.2884), sex (p=0.6964), and platelet count (p=0.9940) were not significantly different in AML patients with and without DNMT3A mutations. However the frequency of higher white blood cell count (p=0.0001), M5 in FAB classification (p=0.0018), and intermediate risk karyotype (p=0.0032) in patients with DNMT3A mutations were significantly higher than those in patients without them. Also, patients with DNMT3A mutations had a mutation in NPM1 (p<0.0001) and FLT3ITD (p=0.009) more frequently. We next assessed the prognostic impact of DNMT3A mutations. For total cohort of patients with AML, complete remission rate and rates of relapse free survival (RFS) at 5 years were not statistically different between AML patients with and without DNMT3A mutations. However the overall survival (OS) at 5 years in patients with DNMT3A mutation (11.0%) was significantly lower than those in patients without them (28.9%) (p=0.0005). Among the intermediate risk karyotype or FLT3ITD negative AML patients, RFS at 5 years were not statistically different between AML patients with and without DNMT3A mutations, but OS at 5 years in patients with DNMT3A mutation (intermediate risk karyotype: 12.6%, Flt3ITD negative: 11.6%) was significantly lower than those in patients without them (intermediate risk karyotype: 23.9%, p=0.0231, FLT3ITD negative: 30.9%, p=0.0046). Conclusions: This study shows that DNMT3A mutation is an important adverse prognostic factor among intermediate risk karyotype or FLT3ITD negative AML patients. Recently TET2 and Isocitrate Dehydrogenase (IDH) 1/2 gene mutations in de novo AML were reported. These genes including DNMT3A play an important role in epigenetic regulation of genes such as methylation etc, and mutations of these genes may be associated with leukemogenesis of AML. Now we are analyzing TET2 and IDH1/2 mutations among same our AML cohort, and we will show the prognostic impact of TET2, IDH1/2 and DNMT3A mutations in patients with de novo AML. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document