scholarly journals Cryptic genomic lesions in adverse-risk acute myeloid leukemia identified by integrated whole genome and transcriptome sequencing

Leukemia ◽  
2019 ◽  
Vol 34 (1) ◽  
pp. 306-311 ◽  
Author(s):  
Jaeseung C. Kim ◽  
Philip C. Zuzarte ◽  
Tracy Murphy ◽  
Michelle Chan-Seng-Yue ◽  
Andrew M. K. Brown ◽  
...  
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1665-1665
Author(s):  
Elisabeth Mack ◽  
Danny Langer ◽  
André Marquardt ◽  
Alfred Ultsch ◽  
Michael G Kiehl ◽  
...  

Abstract Background Acute Myeloid Leukemia (AML) is the most common acute leukemia in adults with a poor overall prognosis. Although the disease has been extensively characterized on the molecular level, this knowledge is translating only slowly into the clinic, particularly with regard to novel therapeutic concepts. Presumably, this striking imbalance substantially is due to the long time required to complete genetic analyses so that results are not available when treatment has to be initiated. Specifically, cytogenetic examinations to determine the karyotype of the malignant blasts, which has been the most important parameter for risk stratification for more than thirty years, take up to two weeks. Next generation sequencing (NGS) technology essentially catalyzed efforts to dissect the genomic landscape of AML, leading to the identification of a large variety of AML driver genes and distinct molecular risk groups. However, these emerging molecular classes of AML do not cover all patients, implying that karyotyping is not dispensable for AML diagnostics at this point. Here we present an integrated approach to AML diagnostics that incorporates these complementary genetic examinations - focused mutational screening of AML-related genes and karyotyping - in one NGS assay. Methods We combined targeted resequencing of DNA and RNA using commercially available panels (TruSigth Myeloid, Illumina and FusionPlex Heme, ArcherDx) to detect AML-associated short sequence variants and gene fusions with low coverage whole genome sequencing for copy number variation analysis. Sequencing was performed on an Illumina MiSeq instrument with a read length of 2x150 bp and a coverage of 3.75 M reads for the TruSight Myeloid panel, 2.25 M reads for the FusionPlex panel and 1.5 M reads for the whole genome library. Variants and fusions were called using the manufacturers' analysis software and a previously published algorithm to identify ITDs (ITD-seek, Au et al., 2016). CNV analysis was performed by comparing read distribution in an AML whole genome library to in silico randomly sampled reads from the reference genome using an in house-developed algorithm. Results Initial testing of our approach on leukemia cell lines and peripheral blood leukocytes from healthy donors revealed sensitivities of 2% and 1-25% for the detection of DNA variants and fusions, respectively. Applying stringent filter criteria, we recovered 75% of verified COSMIC variants and 100% of known fusions in undiluted AML samples without false positives. Chromosomal gains and losses were detected with high confidence with a sensitivity of 10%. We were able to reliably distinguish between normal and complex karyotypes, although NGS-karyotyping based on known fusions and CNV-analysis missed some details of highly aberrant karyotypes such as derivative chromosomes and chromosomal translocations that did not involve genes included in the FusionPlex panel. Our preliminary experience on our method in a diagnostic setting confirms high correlation with reference laboratory results and no relevant differences with regard to treatment decisions. Moreover, we find that NGS considerably accelerates genetic diagnostics of AML as the entire workflow from sample to report including three parallel library preparations, sequencing and data analysis can be completed within 5 days. Operational costs amount approximately 1,700 USD (1,500 EUR) per sample with the low throughput equipment used in this work, which is in the range of expenses for currently established AML diagnostics. Conclusions NGS allows for comprehensive translocation and mutation screening, however, some technical and bioinformatics optimization is required to achieve consistently high sensitivity and specificity for all target genes. CNV analysis of low coverage whole genome sequencing data adds valuable information on numerical chromosomal aberrations, thus allowing construction of a virtual karyotype to substitute for difficult and time-consuming cytogenetics. In summary, we present a reliable, fast and cost-effective strategy to combine molecular and cytogenetics for AML diagnostics in a single NGS run in order to pave the way for a more differentiated clinical management of AML patients in the near future. Disclosures Kiehl: Roche: Consultancy, Other: Travel grants, Speakers Bureau.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 306-306 ◽  
Author(s):  
Francine E. Garrett-Bakelman ◽  
Sheng Li ◽  
Stephen S. Chung ◽  
Todd Hricik ◽  
Rapaport Franck ◽  
...  

Abstract Acute Myeloid Leukemia (AML) remains a clinical challenge, with most patients dying of relapsed disease. The complete biological basis of relapse remains unclear. Genetic lesions and heterogeneity have been proposed as key drivers of clinical outcome, yet do not fully explain leukemia relapse. Epigenomic dysregulation is a hallmark of newly diagnosed AML. Plasticity is a core property of the epigenome, enabling cells to adapt to stressful conditions, independent of genetic alterations. Hence we asked whether epigenomic plasticity might contribute to AML progression, have functional consequences and be independent of genetic influences in AML (a question that has not been addressed for any tumor type). Methods. We formed an international consortium to collect and profile paired diagnosis and relapse AML specimens. We extracted DNA and RNA from 138 clinically annotated AML patient samples. We obtained matched germline DNA as genetic controls, and fourteen normal CD34+ specimens as DNA methylation and transcriptome controls. We performed methylome sequencing (ERRBS), genomic sequencing (exomes and targeted resequencing) and transcriptomic (RNA-seq) profiling. For a single patient, more intensive multi-layer profiling (whole genome sequencing, ERRBS, RNA-seq and single cell RNA-seq) was performed at five serial time points. We quantified epigenetic allelic heterogeneity (epialleles) using a novel approach that employs entropy equations (MethClone), and validated epiallele composition using orthogonal methods. Some of the major conclusions are: 1) Epigenetic allelic diversity is an independent variable linked to clinical outcome. Statistically significant epiallele shift (ΔS <-90) was detected at thousands of genomic loci (eloci) at diagnosis. High eloci burden correlated (Wilcoxon test) with a shorter relapse free probability in the entire cohort (p = 0.043) and in intermediate-risk patients based on the Medical Research Council (p= 0.016) and European Leukemia Net (p=0.057) criteria. Multivariate analysis using Cox proportional hazards regression model revealed that the epiallele burden was an independent variable correlated with relapse free survival (p = 0.021). 2) Promoter epialleles are linked to hypervariable transcriptional regulation. We observed substantial change in epiallele burden at relapse versus diagnosis. A subset of the eloci localized to gene promoters. High promoter epiallele variance was significantly associated with high transcriptional variance (p<0.001) based on RNA-seq, including genes that were significantly differentially expressed at relapse. Deconvolution of leukemia blast populations using Single Cell RNA-seq confirmed that the presence of promoter epialleles was linked to hypervariable transcriptional states (p<0.001). 3) AML patients can be classified according to epigenetic allele progression at relapse. K-means clustering based on epiallele shift at diagnosis versus relapse distributed patients into three classes: those with reduced, increasing or stable epiallele burden. Strikingly, there was no correlation between epiallele changes and the patterns of genomic evolution. Furthermore, there was no correlation between epiallele patterns acquired with mutations in epigenetic modifiers or other recurrently mutated genes in AML. 4) Epigenetic heterogeneity upon disease relapse is divergent from the genetic landscape. Integrating whole genome sequencing and methylome analysis we observed that a) significant increases in epigenetic heterogeneity precede significant changes in the abundance of somatic mutations; b) whereas a high number of somatic mutations were shared across all time points, epialleles exhibited dominance of distinct and unique eloci at each time point; and c) the variant epiallele frequency decreased earlier in progression than somatic mutation variant allele frequency, suggesting that epigenetic clonal diversification can precede genetic clonal evolution. Summary. Based on our results we propose that epigenetic allele diversity allows populations of leukemia cells to sample transcriptional states more freely thus creating the potential for greater evolutionary fitness. This provides an additional independent mechanism of plasticity that can explain the resilient nature of AML to adapt and survive exposure to chemotherapy drugs, independent of genetic heterogeneity. Disclosures Perl: Actinium Pharmaceuticals: Consultancy; Asana Biosciences: Consultancy; Arog Pharmaceuticals: Consultancy; Ambit/Daichi Sankyo: Consultancy; Astellas US Pharma Inc.: Consultancy. Becker:Millenium: Research Funding. Lewis:Roche: Honoraria, Other: Travel; Amgen: Other: Travel. Levine:Loxo Oncology: Membership on an entity's Board of Directors or advisory committees; CTI BioPharma: Membership on an entity's Board of Directors or advisory committees; Foundation Medicine: Consultancy.


Leukemia ◽  
2011 ◽  
Vol 25 (5) ◽  
pp. 821-827 ◽  
Author(s):  
P A Greif ◽  
S H Eck ◽  
N P Konstandin ◽  
A Benet-Pagès ◽  
B Ksienzyk ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document