Functional genomic landscape of acute myeloid leukaemia

Jeffrey W Tyner, Cristina E Tognon, Daniel Bottomly, Beth Wilmot, Stephen E Kurtz, Samantha L Savage, Nicola Long, Anna Reister Schultz, Elie Traer, Melissa Abel, Anupriya Agarwal, Aurora Blucher, Uma Borate, Jade Bryant, Russell Burke, Amy Carlos, Richie Carpenter, Joseph Carroll, Bill H Chang, Cody Coblentz, Amanda d'Almeida, Rachel Cook, Alexey Danilov, Kim-Hien T Dao, Michie Degnin, Deirdre Devine, James Dibb, David K Edwards 5th, Christopher A Eide, Isabel English, Jason Glover, Rachel Henson, Hibery Ho, Abdusebur Jemal, Kara Johnson, Ryan Johnson, Brian Junio, Andy Kaempf, Jessica Leonard, Chenwei Lin, Selina Qiuying Liu, Pierrette Lo, Marc M Loriaux, Samuel Luty, Tara Macey, Jason MacManiman, Jacqueline Martinez, Motomi Mori, Dylan Nelson, Ceilidh Nichols, Jill Peters, Justin Ramsdill, Angela Rofelty, Robert Schuff, Robert Searles, Erik Segerdell, Rebecca L Smith, Stephen E Spurgeon, Tyler Sweeney, Aashis Thapa, Corinne Visser, Jake Wagner, Kevin Watanabe-Smith, Kristen Werth, Joelle Wolf, Libbey White, Amy Yates, Haijiao Zhang, Christopher R Cogle, Robert H Collins, Denise C Connolly, Michael W Deininger, Leylah Drusbosky, Christopher S Hourigan, Craig T Jordan, Patricia Kropf, Tara L Lin, Micaela E Martinez, Bruno C Medeiros, Rachel R Pallapati, Daniel A Pollyea, Ronan T Swords, Justin M Watts, Scott J Weir, David L Wiest, Ryan M Winters, Shannon K McWeeney, Brian J Druker, Jeffrey W Tyner, Cristina E Tognon, Daniel Bottomly, Beth Wilmot, Stephen E Kurtz, Samantha L Savage, Nicola Long, Anna Reister Schultz, Elie Traer, Melissa Abel, Anupriya Agarwal, Aurora Blucher, Uma Borate, Jade Bryant, Russell Burke, Amy Carlos, Richie Carpenter, Joseph Carroll, Bill H Chang, Cody Coblentz, Amanda d'Almeida, Rachel Cook, Alexey Danilov, Kim-Hien T Dao, Michie Degnin, Deirdre Devine, James Dibb, David K Edwards 5th, Christopher A Eide, Isabel English, Jason Glover, Rachel Henson, Hibery Ho, Abdusebur Jemal, Kara Johnson, Ryan Johnson, Brian Junio, Andy Kaempf, Jessica Leonard, Chenwei Lin, Selina Qiuying Liu, Pierrette Lo, Marc M Loriaux, Samuel Luty, Tara Macey, Jason MacManiman, Jacqueline Martinez, Motomi Mori, Dylan Nelson, Ceilidh Nichols, Jill Peters, Justin Ramsdill, Angela Rofelty, Robert Schuff, Robert Searles, Erik Segerdell, Rebecca L Smith, Stephen E Spurgeon, Tyler Sweeney, Aashis Thapa, Corinne Visser, Jake Wagner, Kevin Watanabe-Smith, Kristen Werth, Joelle Wolf, Libbey White, Amy Yates, Haijiao Zhang, Christopher R Cogle, Robert H Collins, Denise C Connolly, Michael W Deininger, Leylah Drusbosky, Christopher S Hourigan, Craig T Jordan, Patricia Kropf, Tara L Lin, Micaela E Martinez, Bruno C Medeiros, Rachel R Pallapati, Daniel A Pollyea, Ronan T Swords, Justin M Watts, Scott J Weir, David L Wiest, Ryan M Winters, Shannon K McWeeney, Brian J Druker

Abstract

The implementation of targeted therapies for acute myeloid leukaemia (AML) has been challenging because of the complex mutational patterns within and across patients as well as a dearth of pharmacologic agents for most mutational events. Here we report initial findings from the Beat AML programme on a cohort of 672 tumour specimens collected from 562 patients. We assessed these specimens using whole-exome sequencing, RNA sequencing and analyses of ex vivo drug sensitivity. Our data reveal mutational events that have not previously been detected in AML. We show that the response to drugs is associated with mutational status, including instances of drug sensitivity that are specific to combinatorial mutational events. Integration with RNA sequencing also revealed gene expression signatures, which predict a role for specific gene networks in the drug response. Collectively, we have generated a dataset-accessible through the Beat AML data viewer (Vizome)-that can be leveraged to address clinical, genomic, transcriptomic and functional analyses of the biology of AML.

Figures

Extended Data Fig. 1.. Genomic Landscape of…
Extended Data Fig. 1.. Genomic Landscape of the Beat AML Cohort.
622 specimens from 531 unique patients were subjected to whole exome sequencing. Automated and manual curation steps (described in the Methods, Supplementary Information, and at http://vizome.org/additional_figures_BeatAML.html) were used to arrive at a final set of high confidence variants (annotated in Supplementary Information, Table S7) and the earliest sample for each unique patient was used in this analysis. Clinical cytogenetics and gene fusion calls from RNA-sequencing were used to curate recurrent gene rearrangements (Supplementary Information). The mutational profile for each unique patient is shown with frequency ranked mutational events in the upper portion and frequency ranked gene rearrangements in the lower portion. The mosaic plot is annotated with clinical features of each case, such as diagnosis/relapse and de novo/transformed disease states and the first bar chart to the right summarizes the cohort frequencies of mutational and gene rearrangement events. The last bar chart on the right summarizes the frequency of significant drug/mutation associations for the given gene across the cohort with drug sensitivity displayed in red and drug resistance displayed in blue. Eleven genes not previously reported to be somatically mutated in cancer were observed with mutations at ~1% cohort frequency: CUB and Sushi multiple domains 2 (CSMD2), NAC alpha domain containing (NACAD), teneurin transmembrane protein 2 (TENM2), aggrecan (ACAN), ADAM metallopeptidase with thrombospondin type 1 motif 7 (ADAMTS7), immunoglobulin-like and fibronectin type III domain containing 1 (IGFN1), neurobeachin like 2 (NBEAL2), poly(U) binding splicing factor 60 (PUF60), zinc finger protein 687 (ZNF687), cadherin EGF LAG seven-pass G-type receptor 2 (CELSR2), and glutamate ionotropic receptor NMDA type subunit 2B (GRIN2B). For the n used to correlate each drug with mutations, please see Supplementary Information (Table S17).
Extended Data Fig. 2.. Transcriptomic Landscape of…
Extended Data Fig. 2.. Transcriptomic Landscape of the Beat AML Cohort.
451 specimens from 411 unique AML patients were subjected to RNA-sequencing. The 2,000 genes with greatest differential expression across these unique AML patients are displayed on the heatmap. The heatmap is annotated with disease type, ELN risk stratification groups, genetic, and cytogenetic features of disease as noted in the figure legend.
Extended Data Fig. 3.. Functional Drug Sensitivity…
Extended Data Fig. 3.. Functional Drug Sensitivity Landscape of the Beat AML Cohort.
409 specimens from 363 unique AML patients were subjected to an ex vivo drug sensitivity assay whereby freshly isolated mononuclear cells from blood or bone marrow of patient specimens were incubated with graded concentrations of 122 small-molecule inhibitors (7 dose points in addition to the no drug control). Probit curve fits were used to compute drug response metrics, and the z-score of area under the dose response curve is plotted for each unique patient specimen against each drug. Drug sensitivity (blue) and resistance (red) are annotated by a color gradient, with grey indicating no drug data available. The heatmap is annotated at the top and bottom with major clinical, cytogenetic, and genetic features of disease as noted in the figure legend.
Extended Data Fig. 4.. Drug response in…
Extended Data Fig. 4.. Drug response in de novo versus transformed AML cases.
The average inhibitor response AUCs for all cases that were de novo (n=288) versus all cases that transformed from a background of MDS (n=111) were compared for every inhibitor having at least 3 cases with evaluable data in each group. The middle point represents the average difference in AUC between the two groups with the bars representing the 95% confidence interval. For the sample size and statistical results of each drug/sample group correlation please see Supplementary Information (Table S20).
Extended Data Fig. 5.. Pairwise Drug Sensitivity…
Extended Data Fig. 5.. Pairwise Drug Sensitivity Correlations and Association with Drug Family.
To understand patterns of small-molecule sensitivity against prior annotation of each drug’s gene and pathway targets, drugs were placed into drug families according to target genes and/or pathways and the Pearson’s correlation value of each drug was plotted onto a clustered heatmap showing drugs with similar or dissimilar patterns of sensitivity across the patient cohort. Prior knowledge annotation of the drug families to which each drug could be assigned is shown to the right of the heatmap with alternating black and grey boxes/labels used to aid in tracking. Descriptions of each drug family as well as the n used to calculate each pairwise drug correlation are found in Supplementary Information (Table S11,21).
Extended Data Fig. 6. Binary Drug Response…
Extended Data Fig. 6. Binary Drug Response Calls and Correlation with Clinical Subsets.
A. For the intersect of every specimen with evaluable response data for each inhibitor, we created a threshold for binary sensitive/resistant calls based on whether the individual specimen response fell within the most sensitive 20% of all specimens tested against that drug. A matrix plot showing the unsupervised clustering of the binary calls can be found at http://vizome.org/additional_figures_BeatAML.html. The binary drug resistance calls for each specimen were rolled up into a single value representing the proportion of drugs to which an individual specimen was sensitive (left) or resistant (right). Specimens were divided into Favorable and Adverse groups based on ELN 2017 criteria to determine whether overall drug sensitivity/resistance correlated with prognostic features of disease (n=233 patients). B. The binary drug resistance calls for each specimen as in panel B. Specimens were divided into diagnostic groups based on WHO 2016 categories to determine whether overall drug sensitivity/resistance correlated with cytogenetic or morphologic features of disease (n=340 patients). For both A and B the upper and lower points of the boxplots show 1.5 times the interquartile range (IQR) from the upper/lower lines while the upper, middle and bottom lines indicate the 75th, median and 25th percentile of the data with the notches extending 1.58 * IQR / sqrt(n). Specific sample sizes of each group are reported in Supplementary Information (Table S22).
Extended Data Fig. 7.. Integration of Genetic…
Extended Data Fig. 7.. Integration of Genetic Events with Drug Sensitivity.
(A) Circos plot showing AML rearrangements in the center, mutational events in the next concentric ring, and gene expression events in the outer ring. The size/width indicates frequency of the event and FDR-corrected q-value of association with drug sensitivity is color-coded (sensitivity (red); resistance (blue)). For each gene, tests involving expression were two-sided t-tests (linear model) of the differences between sensitive and resistant samples. For mutational events, the average difference in AUC between mutant/wild type samples was determined using two-sided t-tests from a linear model as shown in Fig 2A. FDR was computed using the Benjamini-Hochberg over all the drugs. The n used to correlate each mutational event with drug sensitivity is reported in the Supplementary Information (Table S17). (B) As in Fig. 2A, the average difference in AUC drug response between mutant and wild type cases was determined using a two-sided t-test from a linear model fit (plotted on the horizontal axis and FDR-corrected q-value is plotted on the vertical axis). The analysis here represents only FLT3-ITD negative cases. FDR was computed using the Benjamini-Hochberg (BH) over all the drugs. The n used to correlate each mutational event with drug sensitivity is reported in the Supplementary Information (Table S17). Expanded and interactive plots are available in our online data browser (www.vizome.org) and (http://vizome.org/additional_figures_BeatAML.html).
Extended Data Fig. 8.. Integration of Drug…
Extended Data Fig. 8.. Integration of Drug Sensitivity with Genetic Events.
Correlation of drug sensitivity with mutational events. The average difference in AUC drug response between mutant and wild type cases was determined using a two-sided t-test from a linear model fit. FDR was computed using the BH over all the drugs. The degree of significance is represented on the vertical axis (sensitivity (red); resistance (blue)). The n used to correlate each mutational event with drug sensitivity is reported in the Supplementary Information (Table S17).
Extended Data Fig. 9.. Functional Drug Sensitivity…
Extended Data Fig. 9.. Functional Drug Sensitivity Landscape of the Beat AML Cohort.
A. Co-occurrences with regard to WGCNA gene expression clusters and/or mutational events (coefficients) were detected by multivariate modeling with respect to ibrutinib response (resistance (blue); sensitivity (red)) with degree of correlation quantified in top track stacked barplot. All coefficients that appear in 25% of the bootstrap sample sets are shown as segments of the circle. Segment width (the colored ring) corresponds to the percentage of bootstrap samples in which that coefficient appears (quantified above the dotted line). The variables appear in descending order clockwise starting at 12 o’clock. Each link indicates pairwise co-occurrence of mutational events and gene expression clusters (width represents frequency of the co-occurrence). The largest co-occurrence for each coefficient is quantified. B. The capacity of differential gene expression to distinguish the 20% most ibrutinib sensitive (n=46) from 20% most resistant (n=44) specimens is shown on a principle component plot (n=239 patient samples total tested for ibrutinib sensitivity and RNA-Sequencing). For the n used to correlate each drug with gene expression and perform Lasso regression, please see Supplementary Information (Table S17).
Fig. 1.. Comparative Genomic Landscape of AML.
Fig. 1.. Comparative Genomic Landscape of AML.
(A) Frequency of the 33 mutational events that were cumulatively most frequent in Beat AML (n=531 patients) and TCGA (n=200 patients). Top row represents the full Beat AML cohort and the middle bar represents only the de novo Beat AML cases. Mutations were summarized by gene as was done by TCGA whereas the FLT3-ITD mutations were kept separate in the rest of this manuscript. (B) Mutational events at 2% frequency or less in the de novo cases of Beat AML and TCGA were compared for overlap. Venn diagram displays the overlap with the small circles within each compartment representing a size-scaled frequency of each mutational event. (C) Analysis as in panel B with only the non-de novo Beat AML cases versus TCGA. (D) Co-occurrence or exclusivity of the most recurrent mutational events in the Beat AML cohort (n=531 patients) were assessed using the DISCOVER method with a dot plot indicating the odds ratio of co-occurrence (blue) or exclusivity (red) using color-coding and circle size as well as asterisks that indicate FDR-corrected statistical significance.
Fig. 2.. Integration of Genetic Events with…
Fig. 2.. Integration of Genetic Events with Drug Sensitivity.
(A) Average difference in AUC drug response between mutant and wild type cases was determined using a two-sided t-test from a linear model fit (plotted on the horizontal axis and FDR-corrected q-value is plotted on the vertical axis). FDR was computed using the Benjamini-Hochberg (BH) over all the drugs. The n used to correlate each mutational event with drug sensitivity is reported in the Supplementary Information (Table S17). Expanded and interactive plots are available in our online data browser (www.vizome.org) and (http://vizome.org/additional_figures_BeatAML.html). (B)Area under the curve for ibrutinib or entospletinib (n=277 or 168 patient samples, respectively) was plotted for cases with single, double, or triple mutation of FLT3-ITD, NPM1, and DNMT3A with the mean and one s.d. indicated by center and outer gray bars, respectively. An ANOVA was conducted using the Bonferroni approach (statistical results and sample size for all groups reported in Supplementary Information, Tables S18,19). (C) Inhibitors of JAK family kinases were assessed for activity against cases with BCOR mutation alone or BCOR in combination with SRSF2, RUNX1, or DNMT3A. The AUC values are plotted per case with the mean and one standard deviation indicated by center and outer gray bars, respectively. There was a significant difference in AUC by two-sided t-test (t(42)=−2.489, p=0.0168, 95% CI [−73.018, −7.643]) between BCOR and RUNX1 (n=16) versus the average of BCOR alone (n=16), BCOR and SRSF2 (n=8), and BCOR and DNMT3A (n=4).
Fig. 3.. Integration of Gene Expression and…
Fig. 3.. Integration of Gene Expression and Drug Sensitivity Patterns.
Differential gene expression signature distinguishing the 20% most ibrutinib sensitive (n=46) from 20% most resistant (n=44) specimens. Heatmaps for all other drugs are available in our online data browser (http://vizome.org/additional_figures_BeatAML.html). For the n used to correlate each drug with gene expression, please see Supplementary Information (Table S17).

References

    1. Jemal A, Siegel R, Xu J & Ward E Cancer statistics, 2010. CA: a cancer journal for clinicians 60, 277–300, (2010).
    1. National Cancer Institute: Surveillance, Epidemiology, and End Results Program, <> (2018).
    1. Papaemmanuil E et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med 374, 2209–2221, (2016).
    1. Arber DA et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127, 2391–2405, (2016).
    1. Dohner H et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 129, 424–447, (2017).
    1. TCGA. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 368, 2059–2074, (2013).
    1. Byrd JC et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 100, 4325–4336, (2002).
    1. Patel JP et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med 366, 1079–1089, (2012).
    1. Haferlach T et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28, 241–247, (2014).
    1. Lundberg P et al. Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 123, 2220–2228, (2014).
    1. Deininger MWN, Tyner JW & Solary E Turning the tide in myelodysplastic/myeloproliferative neoplasms. Nat Rev Cancer 17, 425–440, (2017).
    1. Busque L et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet 44, 1179–1181, (2012).
    1. Genovese G et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 371, 2477–2487, (2014).
    1. Jaiswal S et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 371, 2488–2498, (2014).
    1. Xie M et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 20, 1472–1478, (2014).
    1. Huang ME et al. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood 72, 567–572, (1988).
    1. Shen ZX et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood 89, 3354–3360, (1997).
    1. Nakao M et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 10, 1911–1918, (1996).
    1. Tse KF, Mukherjee G & Small D Constitutive activation of FLT3 stimulates multiple intracellular signal transducers and results in transformation. Leukemia 14, 1766–1776, (2000).
    1. Yamamoto Y et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 97, 2434–2439, (2001).
    1. Yokota S et al. Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines. Leukemia 11, 1605–1609, (1997).
    1. Knapper S et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 108, 3262–3270, (2006).
    1. O’Farrell AM et al. An innovative phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients. Clin Cancer Res 9, 5465–5476, (2003).
    1. Smith BD et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 103, 3669–3676, (2004).
    1. DeAngelo DJ et al. Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in patients with acute myelogenous leukemia or high-risk myelodysplastic syndrome: safety, pharmacokinetics, and pharmacodynamics. Blood 108, 3674–3681, (2006).
    1. Stone RM et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med 377, 454–464, (2017).
    1. Mardis ER et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 361, 1058–1066, (2009).
    1. Wang F et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 340, 622–626, (2013).
    1. Rohle D et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 340, 626–630, (2013).
    1. Fiskus W et al. Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood 114, 2733–2743, (2009).
    1. Schenk T et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med 18, 605–611, (2012).
    1. Daigle SR et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65, (2011).
    1. Itzykson R et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia 25, 1147–1152, (2011).
    1. Welch JS et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N Engl J Med 375, 2023–2036, (2016).
    1. Konopleva M et al. Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia. Cancer Discov 6, 1106–1117, (2016).
    1. DiNardo CD et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol, (2018).
    1. Tyner JW et al. Kinase Pathway Dependence in Primary Human Leukemias Determined by Rapid Inhibitor Screening. Cancer Res 73, 285–296, (2013).
    1. Benjamini Y & Hochberg Y Controlling the false discovery rate: A practical and powerful approach to multiple testing. Journal of the Royal Statistical Society, Series B (Methodological) 57, 289–300, (1995).
    1. Puissant A et al. SYK is a critical regulator of FLT3 in acute myeloid leukemia. Cancer Cell 25, 226–242, (2014).
    1. Canisius S, Martens JWM & Wessels LFA A novel independence test for somatic alterations in cancer shows that biology drives mutual exclusivity but chance explains most co-occurrence. Genome biology 17, 261, (2016).
    1. Li H & Durbin R Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760, (2009).
    1. McKenna A et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20, 1297–1303, (2010).
    1. Huntley MA et al. ReportingTools: an automated result processing and presentation toolkit for high-throughput genomic analyses. Bioinformatics 29, 3220–3221, (2013).
    1. Buffalo V qrqc: Quick Read Quality Control. R package version 1.22.0, . (2012).
    1. Cibulskis K et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31, 213–219, (2013).
    1. Koboldt DC et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 22, 568–576, (2012).
    1. McLaren W et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070, (2010).
    1. Kandoth, C. .
    1. varscan: Variant calling and somatic mutation/CNV detection for next-generation sequencing data (2018).
    1. vcf2maf: Convert a VCF into a MAF, where each variant is annotated to only one of all possible gene isoforms (Memorial Sloan Kettering, 2018).
    1. Zhao H et al. CrossMap: a versatile tool for coordinate conversion between genome assemblies. Bioinformatics 30, 1006–1007, (2014).
    1. Lek M et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291, (2016).
    1. Ye K, Schulz MH, Long Q, Apweiler R & Ning Z Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871, (2009).
    1. Costello M et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Research 41, e67, (2013).
    1. Kottaridis PD et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 98, 1752–1759, (2001).
    1. Dohner K et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood 106, 3740–3746, (2005).
    1. Falini B, Nicoletti I, Martelli MF & Mecucci C Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood 109, 874–885, (2007).
    1. Huang Q et al. A rapid, one step assay for simultaneous detection of FLT3/ITD and NPM1 mutations in AML with normal cytogenetics. Br J Haematol 142, 489–492, (2008).
    1. Wouters BJ et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 113, 3088–3091, (2009).
    1. Liao Y, Smyth GK & Shi W The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res 41, e108, (2013).
    1. Liao Y, Smyth GK & Shi W featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930, (2014).
    1. Langfelder P & Horvath S WGCNA: an R package for weighted correlation network analysis. BMC bioinformatics 9, 559, (2008).
    1. Hansen KD, Irizarry RA & Wu Z Removing technical variability in RNA-seq data using conditional quantile normalization. Biostatistics 13, 204–216, (2012).
    1. Kim D & Salzberg SL TopHat-Fusion: an algorithm for discovery of novel fusion transcripts. Genome biology 12, R72, (2011).
    1. Langfelder P & Horvath S Fast R Functions for Robust Correlations and Hierarchical Clustering. Journal of Statistical Software 46, (2012).
    1. Langfelder P, Luo R, Oldham MC & Horvath S Is My Network Module Preserved and Reproducible? PLoS computational biology 7, e1001057, (2011).
    1. Parsana P et al. Addressing confounding artifacts in reconstruction of gene co-expression networks. bioRxiv, 202903, (2017).
    1. Zheng X et al. A high-performance computing toolset for relatedness and principal component analysis of SNP data. Bioinformatics 28, 3326–3328, (2012).
    1. International HapMap C The International HapMap Project. Nature 426, 789–796, (2003).
    1. Zheng X & Weir BS Eigenanalysis of SNP data with an identity by descent interpretation. Theoretical Population Biology 107, 65–76, (2016).
    1. Robinson MD & Oshlack A A scaling normalization method for differential expression analysis of RNA-seq data. Genome biology 11, R25, (2010).
    1. Slovak ML, Theisen A & Shaffer LG in The Principles of Clinical Cytogenetics (eds Gersen Steven L. & Keagle Martha B.) 23–49 (Springer; New York, 2013).
    1. Kurtz SE et al. Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic malignancies. Proc Natl Acad Sci U S A 114, E7554–E7563, (2017).
    1. Davis MI et al. Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol 29, 1046–1051, (2011).
    1. Blucher AS, Choonoo G, Kulesz-Martin M, Wu G & McWeeney SK Evidence-Based Precision Oncology with the Cancer Targetome. Trends in pharmacological sciences 38, 1085–1099, (2017).
    1. Gu Z, Eils R & Schlesner M Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849, (2016).
    1. Ritchie ME et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research 43, e47–e47, (2015).
    1. Leek JT & Storey JD Capturing Heterogeneity in Gene Expression Studies by Surrogate Variable Analysis. PLOS Genetics 3, e161, (2007).
    1. Law CW, Chen Y, Shi W & Smyth GK voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome biology 15, R29, (2014).
    1. Parker HS, Bravo HC & Leek JT Removing batch effects for prediction problems with frozen surrogate variable analysis. PeerJ 2, e561, (2014).
    1. Fraley C & Raftery AE Enhanced Model-Based Clustering, Density Estimation, and Discriminant Analysis Software: MCLUST. Journal of Classification 20, 263–286, (2003).
    1. Pison G, Struyf A & Rousseeuw PJ Displaying a clustering with CLUSPLOT. Computational Statistics & Data Analysis 30, 381–392, (1999).
    1. Package corrplot is for visualizing a correlation matrix (2018).
    1. Tibshirani R Regression Shrinkage and Selection via the Lasso. Journal of the Royal Statistical Society. Series B (Methodological) 58, 267–288, (1996).
    1. Friedman J, Hastie T & Tibshirani R Regularization Paths for Generalized Linear Models via Coordinate Descent. Journal of Statistical Software 33, (2010).
    1. Iorio F et al. A Landscape of Pharmacogenomic Interactions in Cancer. Cell 166, 740–754, (2016).

Source: PubMed

3
購読する