Comprehensive Genomic and Transcript Profiling of CBL Gene in Childhood AML: A Report from Children's Oncology Group Studies AAML03P1, AAML0531 and COG/NCI Target AML Initiative

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 170-170
Author(s):  
Diana Chin ◽  
Matthew A. Kutny ◽  
Jonathan Grim ◽  
Robert B. Gerbing ◽  
Kristen Miller ◽  
...  

Abstract The Casitas B-Lineage Lymphoma (CBL) gene encodes for an E3 ubiquitin ligase that targets activated receptor tyrosine kinases for degradation. Mutations of the CBL gene have been described in juvenile myelomonocytic leukemia (JMML) but less is known about mutations and variants of CBL in de novo AML. We previously reported that somatic mutations of CBL are rare in pediatric AML. In this report we present a comprehensive evaluation of genomic and transcript variants of CBL including novel deletion events as well as transcript variants which, in combination with somatic mutations, account for over 6% of pediatric AML with extreme association with inv(16) and favorable outcome. Initial assessment of CBL transcript in a cohort of 100 patients identified previously reported deletion of exon 8 (CBL ΔE8, N=2) associated with CBL splice mutations as well as a novel whole exon 8 and 9 deletion variant (CBL ΔE8+9, N=3) without identifiable underlying somatic alterations. Long distance PCR, as well as custom Nanostring CNV array evaluation revealed a genomic deletion underlying this transcript variant. Subsequent whole genome sequencing as part of COG/NCI TARGET AML initiative, identified discrete genomic deletions of 1998, 3588 and 6189 bp across exon 8 and 9, leading to the generation of this novel variant. We evaluated the functional consequence of the novel CBL ΔE8+9 deletion variant by expressing it in IL3-dependent Ba/F3 cell line. Compared to control cells, Ba/F3 cells expressing CBL ΔE8+9 demonstrated cytokine independent growth. A comprehensive profiling of CBL variants was conducted in 796 pediatric de novo AML patients by transcript profiling (transcript variants) or by exome capture sequencing (somatic mutations including point mutations and smaller indels). All patients were treated on Children's Oncology Group studies AAML03P1 (N=167) and AAML0531 (N=629) and presence of CBL variants was correlated with disease characteristics and clinical outcome. Of the 796 patient specimens tested, 50 patients (6.3%) had one of 3 distinct CBL variants; transcript variant (N=28), somatic mutation (N=14), or dual transcript variant and somatic mutation (N=8). All cases of CBL ΔE8+9 were associated with a corresponding genomic deletion. Out of 14 cases of CBL ΔE8 and 1 case of CBL ΔE9, only 4 cases (27%) had a splice site mutation identified as the underlying mechanism of splice variant. Presence of CBL variants was correlated with clinical characteristics and outcome. Those with CBL variants had a significantly higher prevalence of inv(16) compared with CBL wild type (WT) (37% vs. 13%, p<0.001). This association differed by CBL variant type; 44% transcript variants and 50% dual variants had inv(16) compared to 14% somatic mutations and 13% CBL WT (p<0.001). NPMc+ was more prevalent in those with CBL somatic mutations (29%) than transcript variant (4%), dual variant (0%) or CBL WT (8%) (p=0.035). Similarly, genetic risk groups differed between CBL variants vs. WT (Low risk 70% vs 39%, p=<0.001; Standard risk 22% vs. 46%, p=0.001; High risk 8% vs. 15%, p=0.196). Clinical characteristics including gender, age, race and ethnicity were not significantly different. FAB morphologic assessment revealed an enrichment for the M4 subtype in CBL variant vs. WT (53% vs. 23%, P<.001) which is likely accounted for by the association of inv(16) with this morphologic group. Patients with CBL variants had a 100% clinical remission rate by end of induction II compared to 89% for CBL WT patients (p=0.014). Survival from study entry was similar between CBL mutant vs. WT patients (5 year OS 72% vs. 66%, p=0.24; 5 year EFS 61% vs. 50%, p=0.11). Due to the strong association of CBL mutation with core binding factor leukemia, we assessed whether CBL variant was prognostic of outcome within this favorable risk group, but there was no significant difference in outcomes. Variants of the CBL gene in pediatric AML include genetic mutations with and without whole exon deletions. These CBL variants are highly associated with low risk AML but do not provide independent risk prognosis. The cooperating events of CBL variants in core binding factor leukemia deserve greater study. Our initial analysis of the transcript variants in a cell line model suggest that these large exon 8+9 deletions represent important oncogenic events. The authors would like to gratefully acknowledge the important contributions of the late Dr. Robert Arceci to the AML TARGET initiative. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2584-2584
Author(s):  
Tomohiko Taki ◽  
Hee-Je Kim ◽  
Shigeki Ohtake ◽  
Byung-Sik Cho ◽  
Hitoshi Kiyoi ◽  
...  

Abstract Abstract 2584 Background: Core binding factor (CBF) acute myeloid leukemia (AML) is a specific subgroup of AML, which is commonly related with rearrangements of genes encoding subunits of CBF, AML1 /RUNX1 (AML1-ETO) in t(8;21)(q22;q22) or CBFB (CBFB-MYH11) in inv(16)(p13q22)/t(16;16)(p13;q22), and both clinical outcomes are usually superior in de novo AML. According to literatures, the frequency of patients with t(8;21)(q22;q22) among AML is higher in Japan and Korea than Western countries. However, the accurate incidence and clinical features of CBF-AML are unclear in a large cohort. Patients and methods: We have analyzed 1,536 de novo AML patients from 2 clinical trials of Japan Adult Leukemia Study Group (JALSG, AML97 and AML201 studies, registered from December 1997 to December 2005) and 876 from 3 institutes in Korea (diagnosed from January 2007 to December 2011). Patients from 15 to 64 years old were included in this study, but those with FAB-M3/t(15;17)(q22;q12–21) were excluded. Results: The incidence of CBF-AML was similar in both Japan and Korea (23.3% and 21.2%, P=0.2), however, that of t(8;21) and inv(16) each was a little different between both countries. t(8;21) and inv(16) were observed in 18.6% and 4.9% in Japan, and in 15.0% and 6.3% in Korea, respectively. Of these, the incidence of t(8;21) in Korea was lower than that in Japan (P=0.02), but higher than those reported from Western countries [4.9–8.6% in MRC, CALGB, and SWOG/ECOG except t(15;17), P<0.001]. The prognoses of patients with t(8;21) and inv(16) were similar in complete remission (CR) rate between both countries, but different in 5-year overall survival (OS) and disease-free survival (DFS) rates. CR rate was 91.3% (Japan) and 93.9% (Korea) in t(8;21) (p=0.3), and 98.7% and 100% in inv(16) (P=0.6), respectively. In t(8;21), OS was relatively better in Japan than in Korea (64.6% versus 54.9%, P=0.055), however, DFS tended to be worse in Japan than in Korea (47.6% versus 56.1%, P=0.47). In inv(16), almost the same results with t(8;21) were observed (72.6% versus 65.7% in OS, P=0.2, and 47.7% versus 62.2% in DFS, P=0.06). The white blood cell (WBC) counts and WBC index were prognostic factors in patients with t(8;21), and significantly related to clinical outcome in Japanese patients, however, no significant differences were observed in Korean patients. Patients with t(8;21) having high WBC count (>=10×109/L) were found to have a poor outcome in Japan (44.8% versus 74.5% in OS and 32.4% versus 57.5% in DFS; P<0.001 each), but not in Korea (53.0% versus 57.1% in OS, P=0.8, and 54.0% versus 58.2% in DFS; P=0.6). Additional chromosome abnormalities found in t(8;21) and inv(16) were similar in both countries. In t(8;21), a sole t(8;21) was observed in 25.5% and 25.2%, a loss of a sex chromosome (-X/-Y) in 60.8% and 60.3%, del(9q) in 8.4% and 6.1%, and other abnormalities including 7q- in 23.8% and 29.0% in Japan and Korea, respectively. However, correlations of additional chromosome abnormalities and clinical outcome were different in each country. In Japan, patients with -X/-Y had relatively favorable OS than those with sole t(8;21) [69.1% versus 57.5%, P=0.08]. However, opposite correlation was observed in Korea [48.1% in -X/-Y versus 59.6% in sole t(8;21), P=0.03], although sole t(8;21) had identical OS in both counties [57.5% versus 59.6%, P=0.4] Conclusions: High incidence of t(8;21) in AML was observed in both countries, however, prognostic significance of various factors, such as WBC counts, WBC index, and additional chromosome abnormalities, were different in each country. One of the reasons for these differences may be dependent on the different treatment protocols in each country, and further analysis of factors associated with prognosis in each country is needed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2333-2333
Author(s):  
Maya Thangavelu ◽  
Ryan Olson ◽  
Li Li ◽  
Wanlong Ma ◽  
Steve Brodie ◽  
...  

Abstract Background: Refining risk stratification of acute myeloid leukemia (AML) using molecular profiling, especially those with intermediate cytogentic risk, is becoming standard of care. However, current WHO and ELN classifications are focused on few markers, mainly FLT3, NPM1, and CEBPA. While these abnormalities are relatively common, not all patients with AML and intermediate or normal cytogenetics will have abnormalities in these genes leaving large percentage of patients without refined risk stratification. We demonstrate that using 8 different AML-related genes are adequate to provide one or more molecular markers to further risk stratify patients with de novo AML. Method: Using direct sequencing we analyzed 211 samples referred from community practice with the diagnosis AML for molecular analysis. All samples were evaluated prospectively for mutations in FLT3, NPM1, IDH1, IDH2, CEBPA, WT1, RUNX1, and TP53 using direct sequencing. Fragment length analysis was used in addition to sequencing for FLT3 and NPM1. Available morphology, cytogenetics, and clinical data along with history were reviewed. Results: Of the 211 samples tested 103 (49%) had at least one or more molecular abnormality adequate for refining the risk classification. The mutations detected in these 103 patients were as follows: 27 (26%) FLT-ITD, 10 (10%) FLT3-TKD, 30 (29%) NPM1, 7 (7%) CEBPA, 14 (14%) IDH1, 13 (13%) IDH2, 10 (10%) WT1, 38 (37%) RUNX1, and 2 (2%) TP53. There was significant overlap and most patients had more than one mutation as illustrated in the graph below. However, if the testing was restricted to FLT3, NPM1, CEBPA and DNMT3A, only 56 (54%) would have had refined risk classification and 46% of patients would have remained without subclassification. The most striking finding was that all the remaining patients, who had no molecular abnormality detected in any of these 8 genes, had either history of MDS evolved to AML, therapy-related AML, or cytogenetic abnormalities other than intermediate (multiplex cytogenetic abnormalities or core-binding factor abnormality). Conclusion: Using FLT3, NPM1, CEBPA, and DNMT3A is inadequate for the molecular characterization of patients with AML. Patients with de novo AML and intermediate risk cytogenetics can be adequately prognostically subclassified and molecularly studied by testing only 8 genes. More importantly, this data confirms that the molecular biology driving de novo AML is significantly different from that driving MDS, AML with myelodysplasia-related changes, therapy-related AML, or AML with core binding factor or multiplex cytogenetics. Unlike de novo AML, these entities should be molecularly studied using MDS-specific driver genes. Furthermore, this data suggests that different therapeutic approaches should be developed for MDS and MDS-related AML. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 39-39
Author(s):  
Lisa Eidenschink Brodersen ◽  
Chad A. Hudson ◽  
Todd A. Alonzo ◽  
Robert B. Gerbing ◽  
Laura Pardo ◽  
...  

Introduction: CD74 is a type II transmembrane protein expressed on antigen-presenting cells and an MHC class II chaperone. It has been associated with tumor progression and metastasis in solid tumors, and its expression has been suggested to serve as a prognostic factor in many cancers, with higher relative expression associated with oncogenesis. As the expression of CD74 has been associated with response to bortezomib in multiple myeloma, we inquired whether such correlation might be seen in pediatric AML. We prospectively evaluated CD74 expression by difference from normal (ΔN) flow cytometry and correlated expression with clinical characteristics and outcome as part of Children's Oncology Group protocol AAML1031 that randomized patients younger than 30 years of age with de novo AML to standard treatment with (Arm B) or without (Arm A) bortezomib. The addition of bortezomib to standard chemotherapy increased toxicity but did not improve survival. Given the association of CD74 with B-lymphoid neoplasms and bortezomib's known efficacy in B-cell neoplasms and multiple myeloma, we hypothesized that within Arm B, the patients with CD74 expression would have a more favorable outcome. Methods: In total, 1,139 newly diagnosed pediatric patients with de novo AML were randomized to standard chemotherapy (n=561, Arm A) or standard chemotherapy with bortezomib (n=578, Arm B). All patients received the identical chemotherapy backbone with either four intensive chemotherapy courses or three courses followed by allogeneic hematopoietic stem cell transplantation for high-risk patients. To qualify for this correlative study, 991 patients satisfied 2 criteria: (1) submitting a bone marrow aspirate for ΔN at diagnosis and (2) providing consent for correlative biology studies. All diagnostic specimens were centrally and prospectively evaluated for the expression of CD74 by ΔN. AML was considered to be CD74-positive if the MFI was more than two times above background autofluorescence and more than 40% of the leukemia was above background autofluorescence. Results: Among 991 patients, 263 were CD74-positive (26.5%) by ΔN, with similar prevalence in Arm A (27.9%) and Arm B (25.2%). Correlation of CD74 expression with clinical characteristics showed that those with CD74 expression had higher median age (p&lt;0.001), lower median WBC (p&lt;0.001), higher prevalence of low risk protocol status (p=0.039), lower frequency of CEBPA mutation (p=0.039), inv(16) (p=0.001), and KMT2A rearrangements (p=0.002), and were enriched for t(8;21) (p&lt;0.001) and t(6;9) (p=0.014) fusions. All these features retained significance when patients were sub analyzed by respective treatment arms. CD74-positive patients had a higher morphologic CR rate (p=0.016), however, measurable residual disease by flow cytometry was not significant in the entire cohort or in the sub analysis of treatment arms (p=0.155). CD74-positive patients showed superior 5-year OS (70.6% vs 61.8%, p=0.003) and EFS survival (51.2% vs 43.1%, p=0.007) compared to those who were CD74-negative. For patients in Arm A (no bortezomib), the differences in OS (66.1% vs 61.0%, p=0.138) and EFS (48.9% vs 41.7% p=0.088) were not significant between those that were CD74-positive and those that were CD74-negative (Figure 1). However, patients in Arm B receiving bortezomib that were CD74-positive showed a significant improvement in OS (75.3% vs 62.5%, p=0.006) and EFS (53.6% vs 44.3%, p=0.028) compared to those who were CD74-negative (Figure 1). Comparison of the outcomes for CD74-positive patients with and without bortezomib exposure showed a difference in OS of 66.1% vs. 75.3% for those in Arm A vs. Arm B but did not reach significance (p=0.155). Multivariable analysis for OS yielded a hazard ratio of 0.67 (95% CI: 0.44 - 1.02) and p=0.061, approaching, but not reaching, statistical significance. Conclusions: These data demonstrate that CD74 expression is associated with more favorable disease characteristics and survival. Patients receiving bortezomib that were CD74-positive showed a superior response to therapy compared to patients who did not express CD74, by both OS and EFS, suggesting that CD74-positive childhood AML patients stand to benefit from bortezomib therapy. Bortezomib may induce a mechanistic response in CD74-positive AMLs similar to that in bortezomib-treated B-cell neoplasms and/or multiple myeloma, where bortezomib has proven to be beneficial. Disclosures Cooper: Celgene: Other: Spouse was an employee of Celgene (through August 2019). Pollard:Syndax Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees.


2022 ◽  
Vol 11 ◽  
Author(s):  
Min Yang ◽  
Bide Zhao ◽  
Jinghan Wang ◽  
Yi Zhang ◽  
Chao Hu ◽  
...  

Core Binding Factor (CBF)-AML is one of the most common somatic mutations in acute myeloid leukemia (AML). t(8;21)/AML1-ETO-positive acute myeloid leukemia accounts for 5-10% of all AMLs. In this study, we consecutively included 254 AML1-ETO patients diagnosed and treated at our institute from December 2009 to March 2020, and evaluated molecular mutations by 185-gene NGS platform to explore genetic co-occurrences with clinical outcomes. Our results showed that high KIT VAF(≥15%) correlated with shortened overall survival compared to other cases with no KIT mutation (3-year OS rate 26.6% vs 59.0% vs 69.6%, HR 1.50, 95%CI 0.78-2.89, P=0.0005). However, no difference was found in patients’ OS whether they have KIT mutation in two or three sites. Additionally, we constructed a risk model by combining clinical and molecular factors; this model was validated in other independent cohorts. In summary, our study showed that c-kit other than any other mutations would influence the OS in AML1-ETO patients. A proposed predictor combining both clinical and genetic factors is applicable to prognostic prediction in AML1-ETO patients.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 414-414 ◽  
Author(s):  
Krzysztof Mrózek ◽  
Guido Marcucci ◽  
Kati Maharry ◽  
Deedra Nicolet ◽  
Heiko Becker ◽  
...  

Abstract Abstract 414 The ELN standardized system for reporting cytogenetic and select molecular findings was created to enable comparisons of studies correlating treatment outcome with genetic data in AML (Döhner et al. Blood 2010;115:453). The ELN classification is based on published reports on prognostic import of cytogenetic and molecular alterations, but has not been tested in large cohorts of similarly treated AML pts, except for 1 study comprising ∼20% secondary AML pts (Röllig et al. JCO 2011;29:2758). Thus our goal was to assess the prognostic utility of the ELN classification in a relatively large cohort lacking the confounding effects of AML type (de novo v secondary) or different therapies [chemotherapy v allogeneic stem cell transplant (SCT)]. We analyzed 1,550 de novo AML pts (excluding AML M3, which is not part of the ELN classification) treated with cytarabine-daunorubicin-based chemotherapy on Cancer and Leukemia Group B frontline protocols; no pt received SCT in 1st complete remission (CR). Since consolidation therapy of younger [<60 years (y)] and older ('60 y) pts differed in intensity, we analyzed younger (n=818; median age 44 y, range 17–59) and older (n=732, median age 69 y, range 60–86) pts separately. The pts were classified into the 4 ELN Genetic Groups: Favorable (Fav), Intermediate I (Int I), Intermediate II (Int II) or Adverse (Adv). Younger pts were classified in the Fav Group twice as often and in the Int II and Adv Groups ∼30% less often than older pts (Table 1). Within the Fav Group, core binding factor AML [t(8;21), inv(16) or t(16;16)] was twice as common (P<.001) and NPM1-mut/FLT3-ITD– half as common in younger than in older pts. In the Int I Group, NPM1-mut/FLT3-ITD+ was more common in younger and NPM1-wt/FLT3-ITD– in older pts, whereas in the Int II Group, t(9;11) was twice more common in younger pts. In the Adv Group, pts with balanced cytogenetic abnormalities (abns), ie, inv(3) or t(3;3), t(6;9) and t(v;11)(v;q23), were mainly younger, and pts with –7 mainly older (Table 1).Table 1.Distribution of ELN Groups and subsets in 818 younger and 732 older ptsGenetic Group SubsetYounger Pts %*Older Pts %*PFav4120<.001    t(8;21)2213.01    inv(16) or t(16;16)3312<.001    NPM1-mut, no FLT3-ITD†2854<.001    CEBPA-mut†1720.44Int I1819.64    NPM1-mut & FLT3-ITD†5538.006    NPM1-wt & FLT3-ITD†1012.85    NPM1-wt, no FLT3-ITD†3550.01Int II1930<.001    t(9;11)115.05    Cytogenetic abns not classified as Fav or Adv8995.05Adv2231<.001    inv(3) or t(3;3)83.03    t(6;9)41.02    t(v;11)(v;q23)156.01    −5 or del(5q)33.78    −7512.02    abn(17p)00—    Complex karyotype (≥3 abns)6575.04*For genetic subsets, % are calculated within each Genetic Group†Pts with cytogenetically normal AML After a median follow-up for younger and older pts alive of, respectively, 7.6 y (range, 0.6–19.1) and 6.1 y (range, 2.3–16.4), CR rates, disease-free (DFS) and overall (OS) survival differed significantly across the ELN Groups in both age groups (Table 2, Fig. 1). Pairwise comparisons between individual ELN Groups also yielded significant differences for all of them, except for the comparison between the Int I and Int II Groups, which did not show significant differences for any outcome endpoint in older pts nor for CR rates or DFS in younger pts; OS of younger Int II pts was longer than OS of Int I pts (P=.02; Fig. 2). For each ELN Group, the outcome of older pts was worse than that of younger pts. We conclude that despite differences in frequencies of younger and older pts classified into Fav, Int II and Adv ELN Groups and subsets within these Groups, the ELN classification allows prognostic grouping of both younger and older de novo AML pts. It is hoped that new markers (eg, TET2, ASXL1 mut) will improve pt classification to ELN Int I and Fav Groups.Table 2.Outcomes according to the ELN GroupsOutcomeFavInt IInt IIAdvPYounger pts, n=818n=339n=144n=156n=179    CR rate, %96767950<.001    DFS, median, y5.50.81.20.6.001    OS, median, y11.51.22.10.8<.001Older pts, n=732n=145n=136n=222n=229    CR rate, %83616339<.001    DFS, median, y1.10.60.70.5<.001    OS, median, y1.60.90.90.5<.001 Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4899-4899
Author(s):  
Akitoshi Kinoshita ◽  
Hayato Miyachi ◽  
Hiromichi Matsushita ◽  
Tomohiko Taki ◽  
Miharu Yabe ◽  
...  

Abstract Abstract 4899 [Background] The WHO classification has been widely accepted among physicians who are engaged in treating pediatric AML patients. In 2008, the revised WHO classification has expanded the two categories in AML; AML with recurrent genetic abnormalities and AML with myelodysplasia-related changes. The epidemiology and prognostic significance of these refined categories remains to be explored in children. [Methods] JPLSG AML-05 is a nationwide clinical trial for children with de novo AML, excluding acute promyelocytic leukemia and myeloid leukemia with Down syndrome, which was conducted between November 2006 and December 2010 in Japan. A central review of diagnosis based on the WHO classification was prospectively performed on each case soon after morphological, cytogenetical and immunological data were submitted to data center. Regarding the cases with discrepant results among these parameters, further diagnostic tests including FISH and chimera gene analyses were underwent to confirm the diagnoses. [Results] Four hundred and eighty four patients were enrolled in the study. Thirty patients did not meet the criteria of AML. We could not collected suitable data for diagnosis in 6 patients. Regarding the rest 448 patients, diagnoses based on the WHO classification 2001 and 2008 were determined. According to the 2001 version, 227 (50.6%) had AML with recurrent genetic abnormalities:124 (27.7%) of AML with t(8;21)(q22;q22);(AML1/ETO ), 32 (7.1%) of AML with inv(16)(p13q22); (CBFβ/MYH11), 38 (8.5%) of AML with t(9;11)(p22;q23), and 33 (7.4%) of AML with the other11q23 (MLL) abnormalities, 36 (8.0%) had AML with multilineage dysplasia, and 185 (41.3%) had AML, not otherwise categorized. According to 2008 version, 235 (52.5%) had AML with recurrent genetic abnormalities: 124 (27.7%) of t(8;21)(q22;q22);(AML1/ETO ), 32 (7.1%) of AML with inv(16)(p13q22); (CBFβ/MYH11), 38 (8.5%) of AML with t(9;11)(p22;q23), 33 (7.4%) of AML with the other11q23 (MLL) abnormalities,4 of AML with t(6;9)(p23;q34);DEK-NUP214,2 of AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2);RPN1-EVI13, and 2 of AML with t(1;22)(p13;q13);RBM15-MKL, 88 (19.6.7%) had AML with myelodysplasia-related changes (29 from morphological features of myelodysplasia and 59 from myelodysplasia-related cytogenetic abnormalities), 119 (26.6%) had AML, not otherwise categorized and 7(1.6%) had mixed phenotype acute leukemia (6 of T/myeloid and 1 of B/myeloid). [Discussion] Our comprehensive approach for diagnosis was a useful modality for precise diagnosis of uncertain cases, which might have been assigned to the category of AML, with not otherwise categorized, previously. As a result, the present study shows an increased prevalence of AML with recurrent genetic abnormalities or AML with myeloid dysplasia-related changes among pediatric patients with previously untreated AML. Analysis of the AML-05 trial will elucidate the prognostic impact of these categories. Disclosures: No relevant conflicts of interest to declare.


Cancer ◽  
2009 ◽  
Vol 115 (14) ◽  
pp. 3217-3221 ◽  
Author(s):  
Gautam Borthakur ◽  
E Lin ◽  
Nitin Jain ◽  
Elihu E. Estey ◽  
Jorge E. Cortes ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (20) ◽  
pp. 5409-5415 ◽  
Author(s):  
Ursula Creutzig ◽  
Martin Zimmermann ◽  
Jean-Pierre Bourquin ◽  
Michael N. Dworzak ◽  
Christine von Neuhoff ◽  
...  

Abstract Patients with core binding factor acute myeloid leukemia (CBF-AML) benefit from more intensive chemotherapy, but whether both the t(8;21) and inv(16)/t (16;16) subtypes requires intensification remained to be determined. In the 2 successive studies (AML-BFM-1998 and AML-BFM-2004), 220 CBF-AML patients were treated using the same chemotherapy backbone, whereby reinduction with high-dose cytarabine and mitoxantrone (HAM) was scheduled for these cohorts only in study AML-BFM-1998 but not in AML-BFM-2004 against the background to minimize overtreatment. Five-year overall survival (OS) and event-free survival (EFS) were significantly higher and the cumulative incidence of relapse (CIR) lower in t(8;21) patients treated with HAM (n = 78) compared with without HAM (n = 53): OS 92% ± 3% versus 80% ± 6%, plogrank0.047, EFS 84% ± 4% versus 59% ± 7%, plogrank0.001, and CIR 14% ± 4% versus 34% ± 7%, p(gray)0.006. These differences were not seen for inv(16) (n = 43 and 46, respectively): OS 93% ± 4% versus 94% ± 4%, EFS 75% ± 7% versus 71% ± 9% and CIR 15% ± 6% versus 23% ± 8% (not significant). The subtype t(8;21), but not inv(16), was an independent predictor of worse outcome without HAM reinduction. Based on our data, a 5-year OS of > 90% can be expected for CBF-AML, when stratifying t(8;21), but not inv(16), patients to high-risk chemotherapy, including HAM reinduction.


Sign in / Sign up

Export Citation Format

Share Document