Divergent Epigenomes in Pediatric and Adult Acute Myeloid Leukemia Implicate Cell of Origin and Transcriptional Silencing of Immune Responses As Sources of Clinically Relevant Heterogeneity: A Report from the Children's Oncology Group and NCI/COG Therapeutically Applicable Research to Generate Effective Treatments (TARGET) Initiative

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1046-1046
Author(s):  
Timothy Junius Triche ◽  
Jason E Farrar ◽  
Hamid Bolouri ◽  
Rhonda E. Ries ◽  
Emilia L. Lim ◽  
...  

Abstract Acute myeloid leukemia (AML) carries a poor prognosis across age groups. In children, AML has become the leading cause of leukemia mortality, with only 60% of cases securing long-term remission. In adults, outcomes are far worse, with 5 year survival approaching 24%. The mutational and transcriptional characterization of AML1has not yet translated into improved outcomes for most patients. The TARGET AML project is an effort of Children's Oncology Group (COG) and the National Cancer Institute to characterize molecular abnormalities in pediatric AML. 197 cases were selected for whole genome sequencing (WGS) of diagnostic specimens, 284 cases for mRNA sequencing, 289 cases for DNA methylation arrays, and 721 cases for targeted sequencing (182 assayed by WGS). Most patients (93%) were uniformly treated on COG study AAML0531 or AAML03P1. The Cancer Genome Atlas (TCGA) AML project1characterized 177 comparable adult AMLs with identical assays. DNA methylation changes radically during differentiation of blood cells2, and recurrent pre-leukemic mutations in adult AML3affect DNA methylation and chromatin modifiers. We thus investigated whether differences in cell-of-origin, immune signalling, and regulatory aberrations were captured by focal or regional differences in DNA methylation, within or between adult and pediatric AML patients. In cytogenetically similar TARGET and TCGA AML cases, striking differences in DNA methylation emerge (fig. 1). Pediatric FLT3-mutant AMLs dominate a cluster with normal-progenitor-like DNA methylation. Mutant DNMT3A, RUNX1, and TP53, which selectively favor preleukemic hematopoietic stem cells3,4,5 (HSCs), are common in adult AML, rare in pediatric AML, and tend towards HSC-like hypermethylation. Transcriptional & epigenetic signatures of the cell of origin persist even after leukemic transformation6. Thus we sought to identify the most likely cell of origin for each case. Previous studies of mRNA7 and DNA methylation8 differences in HSCs and progenitor cells (HSPCs), leukemic stem cells (LSCs), and AML blasts allowed us to model these differences in TCGA and TARGET AMLs. RNAseq results revealed many LSC-like cases with aberrant β-catenin signaling and TP53 regulation, distinct from blasts and normal HSPCs (fig. 2a). DNA methylation segregated cases resembling granulocyte/monocyte progenitors (GMPs) from those resembling other HSPC subsets (fig. 2b). DNMT3A mutants strongly associated with HSC/LSC-like mRNA expression, as did most MLL-rearranged AMLs. Nearly all TP53 and RUNX1 mutants presented LSC-like mRNA expression and retained HSC-like methylomes. These results suggest that decades of selective HSC attrition enable cooperating adult-specific mutations to initiate leukemia, while the timescales in pediatric AML favor fusion genes capable of transforming progenitors as well as HSCs. With matched mRNA expression & DNA methylation data from 256 TARGET cases and 156 TCGA cases, we found over 100 genes where DNA methylation accompanied loss of transcription (silencing) in AML but not in normal HSPCs (fig. 3a). Many such genes lie in regions affected by recurrent copy number aberrations, most notably chromosome arms 5q and 19q. Recurrently mutated or deleted genes such as DNMT3A, TET2, SPRY4, and CDKN2A/B are silenced, some mutually exclusively with mutations or CNV. Functional enrichment analyses of silenced genes with DAVID9revealed 4 clusters: NK-cell signaling, innate immune response regulation, transcriptional regulation, and (on chromosome 19q) zinc finger genes involved in Toll-like receptor signaling. Some silencing co-occurs with specific molecular features, but no event was perfectly predicted by any molecular or cytogenetic feature (fig. 3b). Drug-gene interaction mining with DGIDb10 suggests silencing may inform treatment. Silencing of the mitotic checkpoint gene CHFR may confer sensitivity to microtubule inhibitors11, silencing of MGMT suggests greater benefit from alkylating agents12, and demethylating agents may benefit cases with silenced immune response13. Biomarker driven clinical trials will be needed to evaluate these and other markers in pediatric and adult AML, but evidence of independent genetic and epigenetic evolution in AML14supports their continued investigation. This work is dedicated to the late Robert J. Arceci, without whom none of this would have been possible. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 24-24
Author(s):  
Anneke D. van Dijk ◽  
Fieke W Hoff ◽  
Yihua Qiu ◽  
Eveline S. de Bont ◽  
Sophia W.M. Bruggeman ◽  
...  

Background: Acute myeloid leukemia (AML) is an epigenetically heterogeneous disease. The intensity of treatment is currently guided by cytogenetic and molecular genetic risk classifications; however these incompletely predict outcomes, requiring additional information for more accurate predictions. We aimed to identify potential prognostic implications of epigenetic modification of histone proteins, with a focus of H3K27 methylation in relation to mutations in chromatin, splicing and transcriptional regulators. Material and methods: Histone methylation mark expressions were evaluated in a cohort of 241 AML bone marrow (BM) and peripheral blood (PB) samples from patients admitted at the MD Anderson Cancer Center relative to their expression in CD34+ BM derived samples from healthy donors. Simultaneous analysis of 230 proteins was performed using the reverse phase protein array - a high-throughput, quantitative proteomic platform that enables identification of aberrant expressed proteins and the pathways they act in. Additional mutational analysis was performed on 65 BM samples. Results:H3K27Me3 was significantly lower in both BM and PB leukemic-derived samples compared to their expression in normal BM (figure 1A). A greater loss of H3K27Me3 associated with increased proliferative potential and shorter overall survival (OS) in the whole patient population (n=241, HR=0.64, 95% CI=0.47-0.87, p<0.01), as well as in subsets, e.g. cytogenetically normal AML (n=110, HR=0.62, 95% CI=0.40-0.97, p=0.03). To study the prognostic impact of H3K27Me3 in the context of cytogenetic aberrations and mutations, multivariate cox regression analysis was performed which identified H3K27Me3 level as an independent favorable prognostic factor in all (HR=0.74, 95%CI=0.57-0.95, p=0.02) as well as in P53 mutated AML (n=54, HR=0.48, 95%CI=0.26-0.87, p=0.02). A total of 78 AML patients had molecular data available for the major methylation affecting genes, i.e. IDH1, IDH2, DNMT3A and TET2. The level of H3K27Me3 was not prognostic in patients without any DNA methylation affecting mutation present, but patients with at least one mutation in any of these had better outcome when H3K27Me3 levels were high (highest tertile, figure 1A) compared to those with lower levels (median OS 7.1 vs. 24.1 months, HR=0.42, 95% CI=0.21-0.83, p=0.01, figure 1B). Mutations in U2AF1 and SRSF2 affect the spliceosome and are frequently found in antecedent hematological disorders (AHD), as well as are mutations in chromatin regulating genes ASXL1 and BCOR. We observed significant decreased H3K27Me3 in patients with these mutations corresponding with observed lower levels of H3K27Me3 in patients with AHD than those without (p=0.035). BCOR, SRSF2, U2AF1 and ASXL1 mutations confer poor prognosis in myeloid malignancies, however, in our cohort of 65 sequenced AML patients; not individual or a combination of these mutations were independent prognostic factors, but the degree of H3K27Me3 in these patients (HR= 0.49, 95% CI=0.25-0.95, p=0.03). To recognize dysregulated pathways in AML patients with the identified loss of H3K27Me3, we examined correlations of H3K27Me3 with the other 229 proteins on the array. H3K27Me3 is catalyzed by the polycomb group protein EZH2 and is linked to transcriptional repression via the formation of heterochromatin regions. To identify upregulated proteins and pathways upon the loss of H3K27Me3, we focused on significant negatively correlated proteins with H3K27Me3 leading us to the identification of 20 total and 6 phospho-proteins that showed increased expression upon decreased H3K27Me3. Functional enrichment analysis of this protein set revealed an upregulated anti-apoptotic phenotype. Conclusion:This study shows that proteomic profiling of epigenetic modifications on the histone level have clinical implications in AML and support the idea that epigenetic patterns contribute to a more accurate picture of the leukemic state complementing cytogenetic and molecular genetic subgrouping. Figure 1. A) Lower H3K27Me3 in BM and PB derived AML samples compared to normal CD34+. **** represents p<0.0001, ns = not significant. B) Overall survival probability in AML patients with any DNA methylation affecting mutation (i.e. IDH1/2, DNMT3A, TET2, n=53) according to H3K27Me3 low (blue) and high (orange) status. Figure 1 Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
xi chen ◽  
Qin Yan ◽  
Xiang Qin ◽  
Jing Liu ◽  
Yan Zeng ◽  
...  

Abstract BackgroundAFF family genes (AFF1, AFF2, AFF3, AFF4, AFFs) are closely related to the occurrence, development and prognosis of a variety of tumors, but the relationship between AFFs and acute myeloid leukemia (AML) is still unclear. This study aims to explore the relationship between AFFs and the prognosis of AML by bioinformatics and real time quantitative PCR. MethodsAnalyze the expression of AFFs in AML patients and AML cell lines through ONCOMINE, GEPIA, EMBL-EBI, and CCLE databases, explore the prognostic value of AFFs in AML through the TCGA database,explore Co-expression and functional enrichment analysis of AFFs through the Linkedomics, DAVID and KOBAS database.We collected peripheral blood samples of 24 cases of AML (non-M3 subtype) and 8 cases of benign individuals, and detected the mRNA expression level of AFFs by real-time fluorescent quantitative PCR (clinical data comes from hospitalized medical records). ResultsThe results of multiple data sets showed that the mRNA transcription level of AFF1, AFF2, AFF3, and AFF4 in AML was significantly higher than that of normal people. Among them, the high expression of AFF1, AFF2, and AFF3 was statistically significant (p<0.05). Our clinical data showed that the mRNA expression levels of AFF1 and AFF2 in AML patients were higher than those in the control group (p<0.05). The high expression of AFF1 was significantly negatively correlated with overall survival (OS) (p<0.05), the high expression of AFF2 was significantly positively correlated with OS (p<0.05), and the high expression of AFF3 was not correlated with OS (p>0.05). ConclusionAFF1 and AFF2 are highly expressed in AML. The high expression of AFF1 is significantly negatively correlated with OS, and the high expression of AFF2 is significantly positively correlated with OS, which may become a new target for treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 593-593
Author(s):  
Jatinder K. Lamba ◽  
Amit Mitra ◽  
Kristine Crews ◽  
Stanley Pounds ◽  
Xueyuan Cao ◽  
...  

Abstract Abstract 593 Cytarabine (ara-C) is one of the most widely used chemotherapeutic agents in the treatment of acute myeloid leukemia (AML). Inter-patient variability in clinical response following ara-C treatment has been correlated with leukemic cell concentration of ara-C triphosphate (ara-CTP), the active metabolite of the drug. NT5C2 is an inactivating enzyme in the activation pathway of ara-C, and catalyzes conversion of ara-C monophosphate to ara-C. We identified genetic variations in NT5C2 and investigated the association of NT5C2 genetic variations with its mRNA expression and with intracellular ara-CTP accumulation. The coding exons of NT5C2 were resequenced using genomic DNA from EBV-transformed B-lymphoblastoid HapMap cell lines derived from 90 CEPH (representing 30 trios with European ancestry) and 90 YRI (30 trios with African ancestry) sample. Thirty-nine genetic variants (one insertion-deletion and 38 SNPs), including three nonsynonymous SNPs (Thr3Ala, Lys47Arg, Gln136Arg), were identified. The strongly linked SNPs (R2 > 0.8) in CEPH and YRI were grouped into ten distinct groups. NT5C2 mRNA expression levels were determined in all samples and was significantly associated with ethnicity (p = 8.5 × 10-6), with subjects of African ancestry having significantly higher NT5C2 mRNA expression as compared to the subjects with European ancestry (Figure A). Several NT5C2 SNPs were associated with its mRNA expression in both the ethnic groups. To determine the clinical implication of NT5C2 SNPs, we investigated selected germline NT5C2 SNPs in pediatric AML patients treated on the St Jude AML 97 protocol (n=55). Patients were randomly assigned to receive ara-C as either a short daily infusion (500 mg/m2/dose intravenously over 2 hrs daily for 5 days) or a continuous infusion (500 mg/m2/day as a continuous infusion over 5 days). Bone marrow was collected at the end of the ara-C infusion on day1 for patients receiving the short daily infusion (n=27), and at 10 hrs after the start of the infusion for those receiving the continuous infusion (n=28). Ara-CTP levels in leukemia cells were analyzed by HPLC. Intracellular accumulation of ara-CTP was significantly higher when given as short daily infusion, as compared to continuous infusion (0.56 ±0.5 vs. 0.33± 0.31 nmol ara-CTP/2×107leukemic cells; p = 0.014). The inter-patient variability for blast ara-CTP concentration was 40-fold in the short infusion arm and 101-fold in continuous infusion arm. Karyotypes, gender, age, and ethnicity had no significant influence on the leukemic ara-CTP levels. NT5C2 Group 1 SNPs were found to be significantly associated with intracellular ara-CTP levels in leukemic blasts from ara-C-treated Pediatric AML Patients (Figure B). These results suggest that genetic variation in NT5C2 could influence its activity and/or expression and may predict the variability observed in intracellular levels of the ara-C active metabolite, ara-CTP, which in turn can influence treatment response. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 687-687 ◽  
Author(s):  
Emilia L. Lim ◽  
Diane L. Trinh ◽  
Rhonda E. Ries ◽  
Yussanne Ma ◽  
James Topham ◽  
...  

Abstract Introduction Induction chemotherapy results in complete remission in 80% of children with acute myeloid leukemia (AML). However, many patients either fail to achieve a remission, or relapse after an initial response and subsequently die of their disease. Although large numbers of somatic karyotypic and molecular alterations have been identified, the majority of them do not indicate a specific target or distinct pathway that can be readily exploited for therapeutic intervention. Materials & Methods As part of a genome-scale approach to identify prognostic markers and therapeutic targets, we provide a comprehensive characterization of the pediatric AML transcriptome, detailing miRNA & mRNA expression patterns and miRNA:mRNA interactions that are characteristic of the disease. A total of 676 patients were considered for this study. Our discovery cohort consisted of miRNA-seq from 259 primary, 22 refractory and 38 relapse samples, and mRNA-seq from 158 primary, 12 refractory and 47 relapse samples. We confirmed our survival analyses on a validation cohort that consisted of miRNA-seq and mRNA-seq from 378 and 87 primary samples, respectively. Unsupervised non-negative matrix factorization (NMF) was used to identify patient subgroups based on miRNA/mRNA expression. To identify miRNA/mRNA expression associated with patient survival, Cox proportional hazards analysis was performed. Wilcoxon tests were performed to identify differentially expressed miRNAs/mRNAs between samples. To screen for functional miRNA:mRNA interactions, we identified miRNA and mRNA pairs with anti-correlated expression profiles and miRNA binding site predictions consistent with miRNA:mRNA interaction. Results Survival analysis of both the discovery and validation cohorts revealed that 6 miRNAs were associated with overall survival (OS) and event free survival (EFS) (p-val<0.05, q-val<0.1): miR-181c-3p and miR-378c were associated with superior OS and EFS (Hazard Ratio (HR): 0.79-0.88), while miR-106a-3p, miR-106a-5p, miR-363-3p and miR-20b-5p were associated with inferior OS and EFS (HR: 1.14-1.36). All 4 of the miRNAs that were associated with inferior survival are members of the polycistronic miR-106a-363 cluster. Differential expression analysis revealed that miR-106a-363 was abundantly expressed in relapse and refractory samples and in primary samples of refractory patients (q-val<0.05). Integrative miRNA:mRNA expression analysis and luciferase reporter assays further revealed that targets of miR-106a-5p include NDUFC2, NDUFA10, UQCRB, ATP5J2-PTCD1 and ATP5S. Interestingly, these genes are involved in oxidative phosphorylation, a process that is suppressed in treatment-resistant leukemic cells[1]. NMF clustering of miRNA expression profiles revealed 2 groups of patients, with each group characterized by particular genomic alterations: Group 1 cases were enriched for NPM1 mutation and FLT3 -ITD, while Group 2 cases were enriched for t(8;21), inv(16), MLL rearrangements and CEBPA mutation (Fisher's exact test p-val<0.05). NMF clustering of mRNA expression revealed 5 groups of patients, in which the group with abundant expression of ribosomal genes was further distinguished by superior OS and EFS (log-rank p-val<0.05). Analysis of the mRNA data showed a decrease in expression of one mRNA isoform of ribosomal protein L28 (RPL28) in relapse samples (q-val<0.05). In addition, survival analyses revealed that abundant expression of ribosomal protein L10 (RPL10) is associated with superior OS in both the discovery and validation cohorts (p-val<0.05, q-val<0.1, HR: 0.78 & 0.47). Conclusions Through a detailed analysis of the transcriptome (Figure A), we identified miRNAs whose expression levels were significantly associated with clinical outcome. In addition, we showed that abundant expression of miR-106a-363 might contribute to treatment resistance by modulating genes involved in energy metabolism. We also demonstrated that reduced expression of ribosomal genes is associated with inferior outcomes, suggesting a dysregulation of protein translation in treatment resistance. Overall, our transcriptome profiles provide clinically meaningful data for risk and response identification and define novel pathways that may be amenable to therapeutic targeting. Figure 1. Summary of the pediatric AML transcriptome Lagadinou ED, et al. Cell Stem Cell. 2013. Figure 1. Summary of the pediatric AML transcriptome. / Lagadinou ED, et al. Cell Stem Cell. 2013. Disclosures No relevant conflicts of interest to declare.


1992 ◽  
Vol 10 (9) ◽  
pp. 1419-1429 ◽  
Author(s):  
S J Kuerbitz ◽  
C I Civin ◽  
J P Krischer ◽  
Y Ravindranath ◽  
C P Steuber ◽  
...  

PURPOSE Although the expression of both myeloid- and lymphoid-associated cell-surface antigens in acute myeloid leukemia (AML) has been described, the clinical significance of such antigen expression remains unknown in the pediatric population. We sought to define an antibody panel for optimal diagnostic antigenic analysis and to test associations among antigen expression and a number of clinical features at presentation and prognosis in pediatric AML. PATIENTS AND METHODS We reviewed the extensive immunophenotypic analysis performed at the time of diagnosis on 132 assessable patients registered on a single Pediatric Oncology Group AML protocol between 1984 and 1988. RESULTS Eighty-eight percent of patients were identified by testing for expression of CD33 and CD13. Overall, 61% of patients expressed at least one lymphoid-associated antigen, most commonly CD4, CD7, or CD19. Expression of CD5, CD10, CD20, or CD22, commonly detected in T- or B-lineage pediatric acute lymphoid leukemia (ALL), was uncommon; coexpression of multiple lymphoid-associated antigens was also uncommon. Expression of the monocyte-associated antigen CD14 correlated with French-American-British (FAB) M4 or M5 morphology. Otherwise, no correlation between antigen expression and FAB classification was noted. None of the myeloid, lymphoid, natural-killer (NK), or progenitor-associated antigens were associated with significant differences in the likelihood of remission induction or event-free survival when expressor versus nonexpressor groups were compared. CONCLUSIONS The distribution of cell-surface antigen expression in pediatric acute leukemia usually permitted the discrimination of AML from ALL by using a limited panel of antibodies. Although the expression of lymphoid-associated antigens was common, such expression did not seem to be associated with an adverse prognosis in pediatric AML.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 595-595
Author(s):  
Jason E Farrar ◽  
Hamid Bolouri ◽  
Rhonda E. Ries ◽  
Timothy Junius Triche ◽  
Emilia L. Lim ◽  
...  

Abstract The age distribution of acute myeloid leukemia is unusual among malignancies, with onset spanning from early infancy until past the 9th decade. Despite similar histology, cytogenetic abnormalities and recent identification of somatic mutations (e.g., DNMT3A mutations) have highlighted differences in the events driving adult compared to childhood de novo AML. However, the full extent of these differences remains unknown and is likely to have relevance to treatment approaches. The TARGET AML initiative is an effort of the Children's Oncology Group (COG) and the National Cancer Institute to comprehensively characterize the molecular abnormalities of pediatric AML. The dataset comprises 1) whole genome sequencing (WGS) of AML and matched remission bone marrow in 197 cases, 2) mRNA transcriptome sequencing of 284 cases, 3) miRNA sequencing of 692 cases, 4) methylation array data on 289 cases, and 5) targeted capture sequencing of 174 candidate genes identified from WGS in 800 diagnostic samples, including 182 with WGS (Figure 1). The majority of patients (93%) studied were uniformly treated on COG study AAML0531 or its pilot safety precursor study, AAML03P1. Relapsed specimen data (not shown) are available for a subset of these cases. All patient samples were obtained by written consent upon enrollment in the clinical trial. Consistent with adult studies, we identified a relatively low mutational burden, with 2206 somatic tier 1 mutations resulting in a coding change in 1682 genes (median 6 per patient) from the WGS discovery data. We successfully verified 70-90% of variant calls by secondary methods. Also as with adult data, there were relatively few recurrently mutated genes, with fewer than 40 genes altered in >2% of samples. However, there were marked differences in somatic mutation frequencies in comparison to adult TCGA data, both by raw frequency and after adjustment for cytogenetic subtypes present among the two cohorts (Figure 2). Mutations in TP53, NPM1, IDH1, IDH2, TET2 and DNMT3A are more frequent in adult compared to pediatric disease; in contrast, mutations in NRAS, KRAS, WT1, FLT3, PTPN11, GATA2, ASXL2, MYC, SETD2, EZH2 and IKZF1 appear more common in pediatric AML. Mutations of several genes, including CEBPA, ASXL2 and KRAS are not only more common in pediatric AML, they show peak prevalence within specific pediatric age groups. In addition, several genes, including FLT3, WT1, and KIT show significant differences in mutational hotspots compared to adults. Pediatric-adult differences in AML were not limited to somatic gene mutations, but extended to focal and chromosomal copy number alterations (CNA), translocations, miRNA expression, and methylation-induced gene silencing. We identified recurrent focal CNAs in multiple regions not reported in adult AML including 15 heterozygous focal deletions impacting ELF1, an ETS-family transcriptional regulator of hematopoiesis and leukemia driver as well as deletions of the splicing regulator MBNL1 in 10 cases, 8 of which co-occurred with focal deletions of the hematopoietic transcriptional regulator, ZEB2. De novo assembly of mRNA sequencing data identified fusion transcripts in 63% of cases compared to 45% of TCGA LAML. In addition to cytogenetically evident fusions with well-described enrichment for MLL translocations in pediatrics, we identified 29 diagnostic samples (10%) with nucleoporin family fusions (NUP98 with NSD1, KDM5A, PHF23, HOXD13, HMGB3, BRWD3, and CLINT; NUP214 with DEK and SET), CBFA2T3-GLIS2 fusions in 5, and rare fusions of ETS transcription factor genes (FUS-FEV, ETV6-INO8D). Comparison of miRNA expression patterns between adult and pediatric specimens similarly showed marked differences in expression of key regulatory miRNAs including let-7 family members. Finally, analysis of mRNA expression and DNA methylation for the identification of epigenetically silenced genes suggested that, although specific events favor silencing in adults or children, an overall pattern of gene silencing was more prevalent in pediatric compared to adult cases. This work extends our understanding of the heterogeneity of AML, demonstrates fundamental differences in the biology of pediatric- and adult-onset disease, and suggests important age-related differences within "pediatric" AML. This rich dataset should provide a foundation for the establishment of biologically-guided treatment in children with AML. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 610-610
Author(s):  
Marijana Vujkovic ◽  
Edward F. Attiyeh ◽  
Rhonda E. Ries ◽  
Michelle Horn ◽  
Elizabeth K. Goodman ◽  
...  

Abstract Background Childhood acute myeloid leukemia (AML) is characterized by chromosomal instability and requires intensive therapy for cure. Here we present a large-scale study from 3 Children's Oncology Group (COG) trials to define the genomic architectural profiles of pediatric AML and to describe whole-arm gains and losses and focal rearrangements using SNP microarrays, accounting for mutation status, non-neoplastic cell infiltration and aneuploidy. We also investigate the association between somatic copy number aberrations (CNAs) and event free survival (EFS). Patients and Methods A total of 505 matched tumor-remission samples from 459 children with de novo AML were obtained from the COG studies AAML-0531, AAML-03P1, and CCG-2961. 254 paired tumor-remission samples were genotyped on the Affymetrix SNP 6.0 chip at the University of Washington, Seattle, WA, and 251 on the Illumina 2.5M OmniQuad at the Children's Hospital of Philadelphia, PA. All genotyping output was converted to log R ratio and B-allele frequency values and annotations updated to hg19, GRCh37. Illumina intensities were additionally corrected for patterns of genomic wave. A matched allele-specific copy number analysis of tumors (ASCAT) was performed using ASCAT 2.2. A total of 246 (98%) Illumina and 247 (97%) Affymetrix samples passed quality control criteria. ASCAT profiles of patients genotyped on both platforms were manually reviewed and those with highest false CNA calls excluded. Samples with a low signal-to-noise ratio were either eliminated or visually annotated. All CNA segments were manually inspected and false positives removed. The resulting 452 ASCAT profiles were stratified into the risk categories 1) favorable, e.g. inv(16), t(16;16), t(8;21), NPM1, and CEBPα, 2) standard, e.g. normal karyotype, +8, +21, +22, del(7q), del(9q), abnormal 11q23, or other structural changes, and 3) poor, e.g. -5, -7, del(5q), or FLT3/ITD+. GISTIC 2.0 was used to identify genomic areas with significant recurrent aberrations, e.g. amplifications, deletions, and copy-neutral loss-of-heterozygosity (CN-LOH) events. Finally, we investigated whether profiles of genomic instability in the AML genome are predictive of 3 year EFS by using the total number of CNAs as a measure for allelic imbalance. Results The inter-platform concordance for samples genotyped on the two platforms was very high. On average leukemic samples acquire 1.14 somatic CNAs, with a mean of 1.1, 1.3, and 0.8 in the favorable, standard, and poor risk groups respectively (Fig 1). CN-LOH events are observed in 14% (n = 64) of the patients, with 28% involving chromosome 13, and others involving the arms of 11p (23%), 1p (11%), 9p (8%), 7q (6%), 19q (6%), and 3q (5%). Known mutations in AML were enriched in recurrent focal CNA regions, as shown by amplifications on 17q24 (*TK1), 1q32, 3q28, 11q23 (*MLL), 6q27 (*DLL1), 2q32.1, and 4q35.2 (Fig 2). Focal CN-LOH regions were confined to 11p15.5 (*NUP98, *PICALM, *WT1), 1p36.3 (*RUNX3, *NRAS), 9p24.3 (*MLLT3), 3q25.3, 6p23 and 7q35 (*MLL3). Deletions include 7q36.1 (*MLL3, *EZH2), 16p13.11 (*MYH11), 9q21.32, 11p13 (*WT1), 2q37.1 (*IDH1, *DNMT3A), 10p12.31 (*MLLT10), 11q23.3 (*MLL), 16q22.1 (*CBFB), and 1p36.3 (*RUNX3). The association between CNA status and 3 year EFS approached statistical significance for all patients (Table 1), and EFS was significantly lower in standard risk patients with CNAs (51% no CNA vs 34% with CNAs, P 0.03). CNA status did not alter the event risk in the other risk groups. Conclusions The number of CNAs occurring in this cohort is lower than previously reported. However, the presence of somatic CNAs in standard risk patients is significantly associated with a worse treatment outcome that is similar to the poor risk group. This study confirms the established regions of CNA enrichment in pediatric AML and identifies novel regions that may involve driving mediators of tumor fitness and/or acquired resistance to targeted therapies. Disclosures: No relevant conflicts of interest to declare.


Genes ◽  
2021 ◽  
Vol 12 (6) ◽  
pp. 895
Author(s):  
Olga Krali ◽  
Josefine Palle ◽  
Christofer L. Bäcklin ◽  
Jonas Abrahamsson ◽  
Ulrika Norén-Nyström ◽  
...  

Pediatric acute myeloid leukemia (AML) is a heterogeneous disease composed of clinically relevant subtypes defined by recurrent cytogenetic aberrations. The majority of the aberrations used in risk grouping for treatment decisions are extensively studied, but still a large proportion of pediatric AML patients remain cytogenetically undefined and would therefore benefit from additional molecular investigation. As aberrant epigenetic regulation has been widely observed during leukemogenesis, we hypothesized that DNA methylation signatures could be used to predict molecular subtypes and identify signatures with prognostic impact in AML. To study genome-wide DNA methylation, we analyzed 123 diagnostic and 19 relapse AML samples on Illumina 450k DNA methylation arrays. We designed and validated DNA methylation-based classifiers for AML cytogenetic subtype, resulting in an overall test accuracy of 91%. Furthermore, we identified methylation signatures associated with outcome in t(8;21)/RUNX1-RUNX1T1, normal karyotype, and MLL/KMT2A-rearranged subgroups (p < 0.01). Overall, these results further underscore the clinical value of DNA methylation analysis in AML.


Sign in / Sign up

Export Citation Format

Share Document