scholarly journals Haematological Malignancies: Overview of the Recent Progresses in Genetics

2021 ◽  
Author(s):  
Mounia Bendari ◽  
Sofia Sraidi ◽  
Nisrine Khoubila

Genetic defects play a major role in pathogenesis of the most of haematological malignancies, including cytogenetic abnormalities, gene mutations, and abnormal gene expression. Our knowledge about the genetics of haematological disorders has been dramatically improved during the past decade, due to revolution of sequencing technologies which have played a crucial role. In this chapter, we describe the techniques commonly employed for elucidating chromosomal aberrations, prognostic impact of recurrent chromosomal abnormalities, and recently updated risk stratification systems. We will summarise the chromosomal abnormalities recently identified on many of haematological diseases such acute myeloid leukaemia, acute lymphoid leukaemia, myelodysplasic syndrome, multiple myeloma, meyloproliferative disease and clarify their impacts on clinical phenotype and prognosis, as well as their role in the pathogenesis of these diseases. The aim of this chapter is to provide a brief overview of the recent progresses in haematological diseases genetics.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5228-5228
Author(s):  
Genki Yamato ◽  
Hiroki Yamaguchi ◽  
Hiroshi Handa ◽  
Norio Shiba ◽  
Satoshi Wakita ◽  
...  

Abstract Background Acute myeloid leukemia (AML) is a complex disease caused by various genetic alterations. Some prognosis-associated cytogenetic aberrations or gene mutations such as FLT3-internal tandem duplication (ITD), t(8;21)(q22;q22)/RUNX1-RUNX1T1, and inv(16)(p13q22)/CBFB-MYH11 have been found and used to stratify the risk. Numerous gene mutations have been implicated in the pathogenesis of AML, including mutations of DNMT3A, IDH1/2, TET2 and EZH2 in addition to RAS, KIT, NPM1, CEBPA and FLT3in the recent development of massively parallel sequencing technologies. However, even after incorporating these molecular markers, the prognosis is unclear in a subset of AML patients. Recently, NUP98-NSD1 fusion gene was identified as a poor prognostic factor for AML. We have reported that all pediatric AML patients with NUP98-NSD1 fusion showed high expression of the PR domain containing 16 (PRDM16; also known as MEL1) gene, which is a zinc finger transcription factor located near the breakpoint at 1p36. PRDM16 is highly homologous to MDS1/EVI1, which is an alternatively spliced transcript of EVI1. Furthermore, PRDM16 is essential for hematopoietic stem cell maintenance and remarkable as a candidate gene to induce leukemogenesis. Recent reports revealed that high PRDM16 expression was a significant marker to predict poor prognosis in pediatric AML. However, the significance of PRDM16 expression is unclear in adult AML patients. Methods A total of 151 adult AML patients (136 patients with de novo AML and 15 patients with relapsed AML) were analyzed. They were referred to our institution between 2004 and 2015 and our collaborating center between 1996 and 2013. The median length of follow-up for censored patients was 30.6 months. Quantitative RT-PCR analysis was performed using the 7900HT Fast Real Time PCR System with TaqMan Gene Expression Master Mix and TaqMan Gene Expression Assay. In addition to PRDM16, ABL1 was also evaluated as a control gene. We investigated the correlations between PRDM16 gene expression and other genetic alterations, such as FLT3-ITD, NPM1, and DNMT3A, and clarified the prognostic impact of PRDM16 expression in adult AML patients. Mutation analyses were performed by direct sequence analysis, Mutation Biased PCR, and the next-generation sequencer Ion PGM. Results PRDM16 overexpression was identified in 29% (44/151) of adult AML patients. High PRDM16 expression correlated with higher white blood cell counts in peripheral blood and higher blast ratio in bone marrow at diagnosis; higher coincidence of mutation in NPM1 (P = 0.003) and DNMT3A (P = 0.009); and lower coincidence of t(8;21) (P = 0.010), low-risk group (P = 0.008), and mutation in BCOR (P = 0.049). Conversely, there were no significant differences in age at diagnosis and sex distribution. Patients with high PRDM16 expression tended to be low frequency in M2 (P = 0.081) subtype, and the remaining subtype had no significant differences between high and low PRDM16 expression. Remarkably, PRDM16 overexpression patients were frequently observed in non-complete remission (55.8% vs. 26.3%, P = 0.001). Patients with high PRDM16 expression tended to have a cumulative incidence of FLT3-ITD (37% vs. 21%, P = 0.089) and MLL-PTD (15% vs. 5%, P = 0.121). We analyzed the prognosis of 139 patients who were traceable. The overall survival (OS) and median survival time (MST) of patients with high PRDM16 expression were significantly worse than those of patients with low expression (5-year OS, 17% vs. 32%; MST, 287 days vs. 673 days; P = 0.004). This trend was also significant among patients aged <65 years (5-year OS, 25% vs. 48%; MST, 361 days vs. 1565 days, P = 0.013). Moreover, high PRDM16 expression was a significant prognostic factor for FLT3-ITD negative patients aged < 65 years in the intermediate cytogenetic risk group (5-year OS, 29% vs. 58%; MST, 215 days vs. undefined; P = 0.032). Conclusions We investigated the correlations among PRDM16 expression, clinical features, and other genetic alterations to reveal clinical and prognostic significance. High PRDM16 expression was independently associated with non-CR and adverse outcomes in adult AML patients, as well as pediatric AML patients. Our finding indicated that the same pathogenesis may exist in both adult and pediatric AML patients with respect to PRDM16 expression, and measuring PRDM16 expression was a powerful tool to predict the prognosis of adult AML patients. Disclosures Inokuchi: Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria; Celgene: Honoraria; Pfizer: Honoraria.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 539-539
Author(s):  
Vera Grossmann ◽  
Susanne Schnittger ◽  
Alexander Kohlmann ◽  
Christiane Eder ◽  
Annette Fasan ◽  
...  

Abstract Abstract 539 Background: Chromosomal translocations of the MLL gene on chromosome 11q23 are associated with a unique subset of acute lymphoblastic or acute myeloid leukemias (AML). In adults, MLL rearrangements are detected in 3% of de novo AML and in 10% of therapy-related AML (t-AML) cases and are associated with poor prognosis. In addition to disease defining mutations recent high-throughput sequencing studies had shown that almost all myeloid malignancies accumulate a large number of cooperating gene mutations. Aim: Determination of somatic mutations occurring in cases harboring MLL rearrangements and investigation of the prognostic impact of molecular and additional chromosomal aberrations. Patients and Methods: We investigated a cohort of 110 adult AML (80 de novo, 30 t-AML) cases harboring an 11q23 translocation. The cohort was composed of 66 females and 44 males; median age: 55.8 years. MLL translocation partners were as follows: MLLT3 (n=46), MLLT4 (n=15), ELL (n=15); MLLT10 (n=8), others (n=26). Chromosome banding analysis data was available in all cases and survival data in 78 cases (median overall survival (OS) was 10.1 months). Patients were screened for mutations in ASXL1 (n=98), CBL (n=62), CEBPA (n=61), FLT3-ITD (n=103), FLT3-TKD (n=95), IDH1 (n=96), IDH2 (n=84), KRAS (n=107), NPM1 (n=101), NRAS (n=106), PTPN11 (n=99), RUNX1 (n=110), and TP53 (n=110) using amplicon deep-sequencing (454 Roche Life Sciences, Branford, CT), direct Sanger sequencing or melting curve analysis. Results: Overall, mutations were detected in 59/110 (53.6%) cases. We discovered that 42/110 (38.2%) MLL-translocated AML cases harbored mutations within the RAS signalling pathway (KRAS mut: 23/107; 21.5%; NRAS mut: 22/106; 20.8%; PTPN11 mut: 3/99, 3.0%) or alterations in the RAS regulating FLT3 gene (FLT3-ITD: 4/103, 3.9%, and FLT3-TKD: 10/95, 10.5%). Additional mutations were detected in the tumor suppressor gene TP53 (8/110; 7.3%), ASXL1 (6/98; 6.1%), RUNX1 (4/110; 3.6%), and IDH1 (1/96). No mutation was detected in IDH2, CBL, CEBPA, and NPM1. Most cases showed only one mutation (n=39, 66.1%), whereas 17 cases (28.8%) showed two and 3 cases (5.1%) three mutations in different genes. No difference of mutation distribution was seen between de novo and t-AML. In this cohort, no associations amongst gene mutations were observed, however, FLT3-ITD was associated with MLL-ELL (3/14 vs 1/89, P=0.008) and PTPN11 mutations with MLLT10-MLL (2/8 vs 1/91, P=0.017) alterations. In addition, KRAS mut and NRAS mut correlated with high WBC count (KRAS mut: 103.0±79 vs 59.2±67 x109/L, P=0.016; NRAS mut: 94.7±57 vs 60.4±72 x109/L, P=0.080). Further, we were interested in the prognostic impact of single gene mutations. NRAS mut and TP53 mut showed both a non-significant inferior impact on OS, i.e. OS after 2 years: 19.1% vs 46.4%, P=0.62; 0% vs 41.3%, P=0.114. Further, TP53 mutations were correlated with shorter event-free survival (EFS) (EFS after 2 years: 0% vs 20.0%, P=0.029). No associations with prognosis were observed for the remaining genes and translocation partners. In contrast, age was associated with OS and EFS (<60 years, n=59 vs ≥60 years, n=51: OS after 2 years: 51.4% vs 26.3%, P=0.003, EFS after 2 years: 28.0% vs 7.7%, P=0.004). Within the cohort of cases ≥60 years, TP53 mutations (n=5) were associated with worse EFS and OS in comparison to TP53 wild-type cases (n=45) (EFS after 2 years: 8.4% vs 0%, P= 0.006; OS after 2 years: 28.5% vs 0%, P=0.045). Of note, no correlations between mutation frequency and age were observed. We next focused on whether the number of mutations showed any impact on survival. This analysis revealed that cases with more than one mutation (n=20) showed shorter EFS (EFS after 2 years: 10.0% vs 27.3%, P=0.020). Finally, we concentrated on AML with t(9;11)(p22;q23)/MLLT3-MLL, recognized as a distinct WHO-entity. We neither detected an association of MLLT3-MLL (n=46) with OS (P=0.445) or EFS (P=0.644) in comparison to the remaining translocation partners nor a distinct gene mutation profile. However, NRAS mutations correlated with shorter OS and EFS in cases with MLLT3-MLL (after 2 years OS: 17.8% vs 48.3%, P=0.045; after 2 years EFS: 17.8% vs 35.2%, P=0.056). Conclusions: In patients with MLL-translocations a high number of secondary alterations (53.6%), predominantly in RAS pathway components (38.2%), were detected. This may have implication on novel therapeutic options in this unfavorable AML subset. Disclosures: Grossmann: MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Eder:MLL Munich Leukemia Laboratory: Employment. Fasan:MLL Munich Leukemia Laboratory: Employment. Kern:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership.


2008 ◽  
Vol 24 (9) ◽  
pp. 603-610 ◽  
Author(s):  
Y Yang ◽  
XM Jin ◽  
CH Yan ◽  
Y Tian ◽  
JY Tang ◽  
...  

The 8-hydroxy-2′-deoxyguanosine (8-OHdG), an oxidized nucleoside of DNA, not only is a widely used biomarker for the measurement of endogenous oxidative DNA damage but might also be a risk factor for many diseases including cancer. Metal exposure may play an important role in oxidative DNA damage among children. However, few studies on urinary 8-OHdG and metals have been conducted in children with acute leukemia. In the present study, urinary Ni and 8-OHdG were examined in 116 children with acute leukaemia (94 acute lymphoid leukaemia [ALL] and 22 acute myeloid leukaemia [AML]) and 51 healthy child controls. Our result showed that urinary Ni in acute leukaemia patients (ALL: 68.40 ± 133.98, AML: 41.48 ± 76.31 ng/mg creatinine) was significantly higher than that in controls (62.47 ± 124.90 vs 17.63 ± 46.17 ng/mg creatinine, P < 0.05). Similarly, the pretherapy level of urinary 8-OHdG in patients (ALL: 11.83 ± 16.23, AML: 12.36 ± 11.36 ng/mg creatinine) was significantly elevated compared with controls (11.92 ± 15.42 vs 4.03 ± 4.70 ng/mg creatinine, P < 0.05). Moreover, urinary 8-OHdG and urinary Ni showed a weak but significant association with increased risk of childhood leukaemia. The present study suggests that Ni may be an etiologic factor for childhood acute leukaemia by oxidative DNA damage.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2303-2303
Author(s):  
Juan Du ◽  
Richard F. Schlenk ◽  
Andrea Corbacioglu ◽  
Marianne Habdank ◽  
Claudia Scholl ◽  
...  

Abstract Inversion or translocation of chromosome 16 inv(16)/t(16;16) [hereafter abbreviated inv(16)] represent common cytogenetic abnormalities in adult acute myeloid leukemia (AML). At the molecular level inv(16) result in the generation of the CBFb-MYH11 fusion protein that is known to interfere with the heterodimeric transcription factor RUNX1/CBFb and thereby contributes to impaired differentiation of hematopoietic cells. Patients (pts.) with inv(16) are considered to have a favorable outcome, in particular when treated with cytarabine-based consolidation regimens. However, a significant proportion of these pts. relapse and survival after 5 years is about 60%. These findings together with studies from murine models suggest that additional genetic lesions are underlying the clinical heterogeneity of inv(16)-positive AML. The recently described mutations in the signaling molecules FLT3, KIT and RAS represent potential secondary genetic lesions that might contribute to leukemic transformation through constitutive activation. In this study we determined the incidence of KIT (exons 8, 10, 11, and 17), FLT3 (ITD; TKD at D835/I836,) and RAS (NRAS/KRAS exon1, exon2) mutations in 94 adult AML pts. (16 to 60 years; median age 41 years) with inv(16) and evaluated their prognostic impact on clinical outcome. KIT and RAS mutation screening was performed using a sensitive DHPLC-based assay; samples with abnormal profile were confirmed by direct sequencing. FLT3 mutations were identified as previously described. Pts. were entered on 3 AMLSG treatment trials [AML HD93, AML HD98A, AMLSG 07–04]. Postremission therapy implied cytarabine-based (HiDAC n=57) regimens as well as autologous (n=23) or allogeneic (n=13) stem cell transplantation (SCT) in first CR. Mutations were identified in 84% of inv(16) AML with highest frequencies in NRAS (47%) followed by KIT (26%) and FLT3-TKD (15%); 10/24 KIT mutations affected exon17. KRAS and FLT3-ITD mutations were detected in 10% and 3%, respectively. Complete remission (CR) rate was 90% for the whole group. In univariable analyses, FLT3-TKD mutations were associated with a significant inferior relapse-free survival (RFS) (p=0.01). For the other mutations there was no significant difference in RFS when comparing mutated and unmutated pts. Multivariable analysis adjusted for postremission therapy revealed FLT3-TKD (HR 2.39, p=0.04) and in trend KIT exon17 mutations (HR 2.8, p=0.06) as adverse prognostic factors. Therefore, an explorative subgroup analysis was performed for KIT exon17 mutations for the different postremission strategies. In pts. treated with HiDAC, KIT exon17 mutations were associated with a significant inferior RFS (p<0.0001), in contrast to pts. receiving SCT (p=0.70). For overall survival (OS) none of the tested variables were significantly associated with prognosis. KIT, FLT3, or RAS gene mutations can be detected in 84% of inv(16)-positive AML further sustaining the model of cooperating gene mutations. Although the numbers are still quite small, FLT3-TKD and KIT exon17 mutations are of prognostic relevance; the prognostic impact of KIT exon17 mutations seems to be abrogated by SCT strategies. Thus, KIT and FLT3 mutation status might reach clinical importance with regard to the availability of specific inhibitors and the type of postremission therapy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3373-3373
Author(s):  
Sheng-Chieh Chou ◽  
Jih-Luh Tang ◽  
Liang-In Lin ◽  
Hsin-An Hou ◽  
Chien-Yuan Chen ◽  
...  

Abstract Abstract 3373 Poster Board III-261 Purpose Several gene mutations had been found to have clinical implications in patients with acute myeloid leukemia (AML), especially in those with normal karyotype. However, the role of such gene mutations for AML patients receiving allogeneic hematopoietic stem cell transplantation (allo-HSCT) was unclear and inconclusive. We retrospectively evaluated the prognostic impact of 8 gene mutations in adult AML patients undergoing allo-HSCT. Materials & Methods From 1995 to 2007, a total of 463 consecutive adult patients with de novo non-M3 AML had comprehensive gene mutation analyses at the National Taiwan University Hospital. Three hundred and twenty five patients who received conventional induction chemotherapy were enrolled in this study. Those who received only low dose chemotherapy or palliative treatment were excluded. The genetic alterations analyzed included NPM1, FLT3/ITD, FLT3/TKD, CEBPA, AML1/RUNX1, RAS, MLL/PTD, and WT1. The clinical implication of these genetic alterations in the patients receiving allo-HSCT was analyzed, and the result was compared with that in patients without allo-HSCT. Results The clinical characteristics in the patients receiving allo-HSCT (n=100) and those without (n=225) were similar with the exception of age, being younger in the former group (35.4 years vs. 49.5 years p<0.001). In univariate analysis, older age (Age > 45 years), higher initial WBC count (WBC>50K/μL), elevated LDH level, unfavorable karyotype, FLT3/ITD, mutations of AML1/RUNX1 were significantly associated with poorer overall survival (OS) in patients not receiving allo-HSCT; While NPM1mut/FLT3ITDneg and CEBPA mutations served as significantly good prognostic indicators. In multivariate analysis, age, WBC count, karyotype, FLT3/ITD, AML1/RUNX1, CEBPA and NPM1mut/FLT3ITDneg remained to be independent prognostic factors in non-allo-HSCT patients. However, in patients receiving allo-HSCT, only unfavorable karyotype and disease status (refractory or remission) at the time of transplantation were associated with poorer OS both in univariate and multivariate analyses. The similar prognostic impact of FLT3/ITD, CEBPA, AML1/RUNX1 and NPM1 on OS was not seen in patients receiving allo-HSCT. Furthermore, in contrast to its poor prognostic impact in non-allo-HSCT patients, mutation of AML1/RUNX1 was a significant good prognostic factor for relapse free survival (p=0.046), although not for OS, in allo-HSCT group. Conclusion FLT3/ITD, mutations of AML1/RUNX1, CEBPA and NPM1 have great prognostic implication for OS in AML patients not receiving allo-HSCT. However, their impact on OS is ameliorated in patients receiving allo-HSCT. The results need to be confirmed by further studies on more patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2538-2538
Author(s):  
Shunichiro Yamaguchi ◽  
Kenji Tokunaga ◽  
Eisaku Iwanaga ◽  
Tomoko Nanri ◽  
Taizo Shimomura ◽  
...  

Abstract Abstract 2538 Aims: Among acute myeloid leukemia (AML) patients with intermediate-risk cytogenetics, C/EBPa mutations represent a distinct disease entity with a favorable clinical outcome and is adopted in the current WHO classification of AML as a provisional disease entity in the category AML with recurrent genetic abnormalities. CEBPA encodes a transcription factor that is essential for neutrophil development. AML patients with CEBPA mutations can be separated into two subgroups with a single mutation in the CEBPA (CEBPA sm) and double mutations (CEBPA dm). Biallelic mutations consisted of an N-terminal frameshift mutation and a C-terminal inframe bZIP mutation were detected in the majority of CEBPA dm, whereas CEBPA sm occurs in either N-terminal or C-terminal regions. More recent data indicate that favorable outcome is mainly observed in AML patients with CEBPA dm but not with CEBPA sm. In addition, concurrent gene mutations may occur more frequently in AML with CEBPA sm than in CEBPA dm. In contrast, transcription factor GATA2 mutations are frequently identified in AML with CEBPA dm. In this study, we examined incidence, concurrent gene mutations and clinical significance of CEBPA dm and CEBPA sm in Japanese adults with cytogenetically intermediate-risk AML. Methods: To identify the prevalence and prognostic impact of CEBPA dm and CEBPA sm, we examined 111 patients with intermediate-risk AML who were mainly treated with the JALSG protocols. Age ranged from 16 to 86 years, with a median of 58.5 years. DNA was extracted from bone marrow or peripheral blood mononuclear cells at diagnosis and subjected to PCR amplification and direct sequencing of the CEBPA, FLT3, NPM1, IDH1, IDH2, DNMT3A and GATA2 genes. This study was approved by the Institutional Review Boards and informed consent was obtained from each patient according to guidelines based on the revised Declaration of Helsinki. Results: Of 111 cytogenetically intermediate-risk AML, we found 12 (10.8%) CEBPA dm and 7 (6.3%) CEBPA sm. In 7 CEBPA sm, one NPM1 mutation and one FLT3-ITD were detected. Two FLT 3-ITD and no concurrent mutation of NPM1 were found in CEBPA dm. No mutation in the IDH1, IDH2, DNMT3A exon 23 was identified in both patients with CEBPA sm and CEBPA dm. On the other hand, mutations in the GATA2 zinc finger domains were detected in 3 of 12 (25%) patients with CEBPA dm. No GATA2 mutations were found in 7 CEBPA sm. One of 21 patients with wild-type CEBPA (CEBPA wt) had a GATA2 mutation. Patients with CEBPA double or single mutations showed a better 5-year overall survival (OS) compared to CEBPA wt (51.3% vs 16.0%, P=0.0048). CEBPA dm AML was associated with a significant superior clinical outcome compared with CEBPA wt (5-year OS, 55.6% vs 16.0%, P=0.0025). However, no significant difference was identified between CEBPA dm and CEBPA sm AML (5-years OS, 55.6% vs 42.9%, P=0.1375) or between CEBPA sm and CEBPA wt AML (5-year OS, 42.9% vs 16.0%, P=0.4827). In addition, the presence of additional GATA2 mutations did not significantly influence the clinical outcome of AML patients with CEBPA dm. Conclusions: A total of 19 (17.1%) patients with cytogenetically intermediate-risk AML harbored CEBPA mutations. Our study indicates that the presence of the CEBPA dm but not CEBPA sm is associated with favorable outcome in Japanese patients with cytogenetically intermediate-risk AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. SCI-33-SCI-33 ◽  
Author(s):  
Bob Löwenberg ◽  
Peter Valk ◽  
Ivo P. Touw ◽  
Ruud Delwel

Abstract Abstract SCI-33 The treatment of acute myeloid leukemia (AML) remains associated with considerable failure rates. From a therapy viewpoint AML presents as a kaleidoscope of highly diverse diseases for which the (epi)genetic heterogeneity of the disease is the overriding determinant of treatment failure. Diverse coemerging molecular abnormalities (gene mutations, expression abnormalities) related, for instance, to the genes CEBPA, nucleophosmin-1, FLT-3, RUNX1, ASXL1, P53, G-CSF-R, IDH1/IDH2, DNMT3A, and EVI-1 in variable composite configurations, determine the considerable “interindividual” and “intraindividual” variations of AML. As these somatic genetic abnormalities in transformed hematopoietic progenitor cells perturb critical cellular pathways and functions, they confer a profound impact upon the clinical phenotype of the disease—and also may define unique diagnostic subtypes (diagnosis) and therapeutic resistance (treatment response). Using genome-wide approaches and well-defined cohorts, the identified molecular biomarkers provide insights into the variability of treatment outcome, and furnish an informative framework for risk-adapted treatment decisions according to disease risk and therapeutic responsiveness (prognosis and treatment choice). In this presentation, examples of strategies to assess the prognostic impact of genomic abnormalities will be discussed, as well as the challenge to integrate the numerous markers into meaningful decision algorithms that can be used for more individualized therapy choices, for example, regarding the use of particular therapeutic agents and the utility (risk-benefit) of allogeneic stem cell transplantation. Disclosures: Löwenberg: Skyline Diagnostics: Membership on an entity's Board of Directors or advisory committees.


2020 ◽  
Vol 117 (42) ◽  
pp. 26340-26346
Author(s):  
Marius Bill ◽  
Krzysztof Mrózek ◽  
Jessica Kohlschmidt ◽  
Ann-Kathrin Eisfeld ◽  
Christopher J. Walker ◽  
...  

Balanced rearrangements involving theKMT2Agene, located at 11q23, are among the most frequent chromosome aberrations in acute myeloid leukemia (AML). Because of numerous fusion partners, the mutational landscape and prognostic impact of specific 11q23/KMT2Arearrangements are not fully understood. We analyzed clinical features of 172 adults with AML and recurrent 11q23/KMT2Arearrangements, 141 of whom had outcome data available. We compared outcomes of these patients with outcomes of 1,097 patients without an 11q23/KMT2Arearrangement categorized according to the 2017 European LeukemiaNet (ELN) classification. Using targeted next-generation sequencing, we investigated the mutational status of 81 leukemia/cancer-associated genes in 96 patients with 11q23/KMT2Arearrangements with material for molecular studies available. Patients with 11q23/KMT2Arearrangements had a low number of additional gene mutations (median, 1; range 0 to 6), which involved the RAS pathway (KRAS,NRAS, andPTPN11) in 32% of patients.KRASmutations occurred more often in patients with t(6;11)(q27;q23)/KMT2A-AFDNcompared with patients with the other 11q23/KMT2Asubsets. Specific gene mutations were too infrequent in patients with specific 11q23/KMT2Arearrangements to assess their associations with outcomes. We demonstrate that younger (age <60 y) patients with t(9;11)(p22;q23)/KMT2A-MLLT3had better outcomes than patients with other 11q23/KMT2Arearrangements and those without 11q23/KMT2Arearrangements classified in the 2017 ELN intermediate-risk group. Conversely, outcomes of older patients (age ≥60 y) with t(9;11)(p22;q23) were poor and comparable to those of the ELN adverse-risk group patients. Our study shows that patients with an 11q23/KMT2Arearrangement have distinct mutational patterns and outcomes depending on the fusion partner.


Leukemia ◽  
2008 ◽  
Vol 23 (2) ◽  
pp. 262-270 ◽  
Author(s):  
I H I M Hollink ◽  
C M Zwaan ◽  
M Zimmermann ◽  
T C J M Arentsen-Peters ◽  
R Pieters ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document