Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jacqueline E. Payton is active.

Publication


Featured researches published by Jacqueline E. Payton.


The New England Journal of Medicine | 2009

Recurring Mutations Found by Sequencing an Acute Myeloid Leukemia Genome

Elaine R. Mardis; Li Ding; David J. Dooling; David E. Larson; Michael D. McLellan; Ken Chen; Daniel C. Koboldt; Robert S. Fulton; Kim D. Delehaunty; Sean McGrath; Lucinda A. Fulton; Devin P. Locke; Vincent Magrini; Rachel Abbott; Tammi L. Vickery; Jerry S. Reed; Jody S. Robinson; Todd Wylie; Scott M. Smith; Lynn K. Carmichael; James M. Eldred; Christopher C. Harris; Jason Walker; Joshua B. Peck; Feiyu Du; Adam F. Dukes; Gabriel E. Sanderson; Anthony M. Brummett; Eric Clark; Joshua F. McMichael

BACKGROUND The full complement of DNA mutations that are responsible for the pathogenesis of acute myeloid leukemia (AML) is not yet known. METHODS We used massively parallel DNA sequencing to obtain a very high level of coverage (approximately 98%) of a primary, cytogenetically normal, de novo genome for AML with minimal maturation (AML-M1) and a matched normal skin genome. RESULTS We identified 12 acquired (somatic) mutations within the coding sequences of genes and 52 somatic point mutations in conserved or regulatory portions of the genome. All mutations appeared to be heterozygous and present in nearly all cells in the tumor sample. Four of the 64 mutations occurred in at least 1 additional AML sample in 188 samples that were tested. Mutations in NRAS and NPM1 had been identified previously in patients with AML, but two other mutations had not been identified. One of these mutations, in the IDH1 gene, was present in 15 of 187 additional AML genomes tested and was strongly associated with normal cytogenetic status; it was present in 13 of 80 cytogenetically normal samples (16%). The other was a nongenic mutation in a genomic region with regulatory potential and conservation in higher mammals; we detected it in one additional AML tumor. The AML genome that we sequenced contains approximately 750 point mutations, of which only a small fraction are likely to be relevant to pathogenesis. CONCLUSIONS By comparing the sequences of tumor and skin genomes of a patient with AML-M1, we have identified recurring mutations that may be relevant for pathogenesis.


Nature | 2012

Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing

Li Ding; Timothy J. Ley; David E. Larson; Christopher A. Miller; Daniel C. Koboldt; John S. Welch; Julie Ritchey; Margaret A. Young; Tamara Lamprecht; Michael D. McLellan; Joshua F. McMichael; John W. Wallis; Charles Lu; Dong Shen; Christopher C. Harris; David J. Dooling; Robert S. Fulton; Lucinda Fulton; Ken Chen; Heather K. Schmidt; Joelle Kalicki-Veizer; Vincent Magrini; Lisa Cook; Sean McGrath; Tammi L. Vickery; Michael C. Wendl; Sharon Heath; Mark A. Watson; Daniel C. Link; Michael H. Tomasson

Most patients with acute myeloid leukaemia (AML) die from progressive disease after relapse, which is associated with clonal evolution at the cytogenetic level. To determine the mutational spectrum associated with relapse, we sequenced the primary tumour and relapse genomes from eight AML patients, and validated hundreds of somatic mutations using deep sequencing; this allowed us to define clonality and clonal evolution patterns precisely at relapse. In addition to discovering novel, recurrently mutated genes (for example, WAC, SMC3, DIS3, DDX41 and DAXX) in AML, we also found two major clonal evolution patterns during AML relapse: (1) the founding clone in the primary tumour gained mutations and evolved into the relapse clone, or (2) a subclone of the founding clone survived initial therapy, gained additional mutations and expanded at relapse. In all cases, chemotherapy failed to eradicate the founding clone. The comparison of relapse-specific versus primary tumour mutations in all eight cases revealed an increase in transversions, probably due to DNA damage caused by cytotoxic chemotherapy. These data demonstrate that AML relapse is associated with the addition of new mutations and clonal evolution, which is shaped, in part, by the chemotherapy that the patients receive to establish and maintain remissions.


Nature | 2008

DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome

Timothy J. Ley; Elaine R. Mardis; Li Ding; Bob Fulton; Michael D. McLellan; Ken Chen; David J. Dooling; Brian H. Dunford-Shore; Sean McGrath; Matthew Hickenbotham; Lisa Cook; Rachel Abbott; David E. Larson; Dan Koboldt; Craig S. Pohl; Scott M. Smith; Amy Hawkins; Scott Abbott; Devin P. Locke; LaDeana W. Hillier; Tracie L. Miner; Lucinda Fulton; Vincent Magrini; Todd Wylie; Jarret Glasscock; Joshua J. Conyers; Nathan Sander; Xiaoqi Shi; John R. Osborne; Patrick Minx

Acute myeloid leukaemia is a highly malignant haematopoietic tumour that affects about 13,000 adults in the United States each year. The treatment of this disease has changed little in the past two decades, because most of the genetic events that initiate the disease remain undiscovered. Whole-genome sequencing is now possible at a reasonable cost and timeframe to use this approach for the unbiased discovery of tumour-specific somatic mutations that alter the protein-coding genes. Here we present the results obtained from sequencing a typical acute myeloid leukaemia genome, and its matched normal counterpart obtained from the same patient’s skin. We discovered ten genes with acquired mutations; two were previously described mutations that are thought to contribute to tumour progression, and eight were new mutations present in virtually all tumour cells at presentation and relapse, the function of which is not yet known. Our study establishes whole-genome sequencing as an unbiased method for discovering cancer-initiating mutations in previously unidentified genes that may respond to targeted therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Acquired copy number alterations in adult acute myeloid leukemia genomes

Matthew J. Walter; Jacqueline E. Payton; Rhonda E. Ries; William D. Shannon; Hrishikesh Deshmukh; Yu Zhao; Jack Baty; Sharon Heath; Peter Westervelt; Mark A. Watson; Michael H. Tomasson; Rakesh Nagarajan; Brian P. O'Gara; Clara D. Bloomfield; Krzysztof Mrózek; Rebecca R. Selzer; Todd Richmond; Jacob Kitzman; Joel Geoghegan; Peggy S. Eis; Rachel Maupin; Robert S. Fulton; Michael D. McLellan; Richard Wilson; Elaine R. Mardis; Daniel C. Link; Timothy A. Graubert; John F. DiPersio; Timothy J. Ley

Cytogenetic analysis of acute myeloid leukemia (AML) cells has accelerated the identification of genes important for AML pathogenesis. To complement cytogenetic studies and to identify genes altered in AML genomes, we performed genome-wide copy number analysis with paired normal and tumor DNA obtained from 86 adult patients with de novo AML using 1.85 million feature SNP arrays. Acquired copy number alterations (CNAs) were confirmed using an ultra-dense array comparative genomic hybridization platform. A total of 201 somatic CNAs were found in the 86 AML genomes (mean, 2.34 CNAs per genome), with French-American-British system M6 and M7 genomes containing the most changes (10–29 CNAs per genome). Twenty-four percent of AML patients with normal cytogenetics had CNA, whereas 40% of patients with an abnormal karyotype had additional CNA detected by SNP array, and several CNA regions were recurrent. The mRNA expression levels of 57 genes were significantly altered in 27 of 50 recurrent CNA regions <5 megabases in size. A total of 8 uniparental disomy (UPD) segments were identified in the 86 genomes; 6 of 8 UPD calls occurred in samples with a normal karyotype. Collectively, 34 of 86 AML genomes (40%) contained alterations not found with cytogenetics, and 98% of these regions contained genes. Of 86 genomes, 43 (50%) had no CNA or UPD at this level of resolution. In this study of 86 adult AML genomes, the use of an unbiased high-resolution genomic screen identified many genes not previously implicated in AML that may be relevant for pathogenesis, along with many known oncogenes and tumor suppressor genes.


JAMA | 2011

Identification of a Novel TP53 Cancer Susceptibility Mutation Through Whole-Genome Sequencing of a Patient With Therapy-Related AML

Daniel C. Link; Laura G. Schuettpelz; Dong Shen; Jinling Wang; Matthew J. Walter; Shashikant Kulkarni; Jacqueline E. Payton; Jennifer Ivanovich; Paul J. Goodfellow; Michelle M. Le Beau; Daniel C. Koboldt; David J. Dooling; Robert S. Fulton; R. Hugh F. Bender; Lucinda Fulton; Kimberly D. Delehaunty; Catrina C. Fronick; Elizabeth L. Appelbaum; Heather K. Schmidt; Rachel Abbott; Michelle O'Laughlin; Ken Chen; Michael D. McLellan; Nobish Varghese; Rakesh Nagarajan; Sharon Heath; Timothy A. Graubert; Li Ding; Timothy J. Ley; Gerard P. Zambetti

CONTEXT The identification of patients with inherited cancer susceptibility syndromes facilitates early diagnosis, prevention, and treatment. However, in many cases of suspected cancer susceptibility, the family history is unclear and genetic testing of common cancer susceptibility genes is unrevealing. OBJECTIVE To apply whole-genome sequencing to a patient without any significant family history of cancer but with suspected increased cancer susceptibility because of multiple primary tumors to identify rare or novel germline variants in cancer susceptibility genes. DESIGN, SETTING, AND PARTICIPANT: Skin (normal) and bone marrow (leukemia) DNA were obtained from a patient with early-onset breast and ovarian cancer (negative for BRCA1 and BRCA2 mutations) and therapy-related acute myeloid leukemia (t-AML) and analyzed with the following: whole-genome sequencing using paired-end reads, single-nucleotide polymorphism (SNP) genotyping, RNA expression profiling, and spectral karyotyping. MAIN OUTCOME MEASURES Structural variants, copy number alterations, single-nucleotide variants, and small insertions and deletions (indels) were detected and validated using the described platforms. RESULTS; Whole-genome sequencing revealed a novel, heterozygous 3-kilobase deletion removing exons 7-9 of TP53 in the patients normal skin DNA, which was homozygous in the leukemia DNA as a result of uniparental disomy. In addition, a total of 28 validated somatic single-nucleotide variations or indels in coding genes, 8 somatic structural variants, and 12 somatic copy number alterations were detected in the patients leukemia genome. CONCLUSION Whole-genome sequencing can identify novel, cryptic variants in cancer susceptibility genes in addition to providing unbiased information on the spectrum of mutations in a cancer genome.


The Journal of Molecular Diagnostics | 2014

Validation of a Next-Generation Sequencing Assay for Clinical Molecular Oncology

Catherine E. Cottrell; Hussam Al-Kateb; Andrew J. Bredemeyer; Eric J. Duncavage; David H. Spencer; Haley J. Abel; Christina M. Lockwood; Ian S. Hagemann; Stephanie M. O’Guin; Lauren C. Burcea; Christopher S. Sawyer; Dayna M. Oschwald; Jennifer L. Stratman; Dorie A. Sher; Mark R. Johnson; Justin T. Brown; Paul F. Cliften; Bijoy George; Leslie McIntosh; Savita Shrivastava; TuDung T. Nguyen; Jacqueline E. Payton; Mark A. Watson; Seth D. Crosby; Richard D. Head; Robi D. Mitra; Rakesh Nagarajan; Shashikant Kulkarni; Karen Seibert; Herbert W. Virgin

Currently, oncology testing includes molecular studies and cytogenetic analysis to detect genetic aberrations of clinical significance. Next-generation sequencing (NGS) allows rapid analysis of multiple genes for clinically actionable somatic variants. The WUCaMP assay uses targeted capture for NGS analysis of 25 cancer-associated genes to detect mutations at actionable loci. We present clinical validation of the assay and a detailed framework for design and validation of similar clinical assays. Deep sequencing of 78 tumor specimens (≥ 1000× average unique coverage across the capture region) achieved high sensitivity for detecting somatic variants at low allele fraction (AF). Validation revealed sensitivities and specificities of 100% for detection of single-nucleotide variants (SNVs) within coding regions, compared with SNP array sequence data (95% CI = 83.4-100.0 for sensitivity and 94.2-100.0 for specificity) or whole-genome sequencing (95% CI = 89.1-100.0 for sensitivity and 99.9-100.0 for specificity) of HapMap samples. Sensitivity for detecting variants at an observed 10% AF was 100% (95% CI = 93.2-100.0) in HapMap mixes. Analysis of 15 masked specimens harboring clinically reported variants yielded concordant calls for 13/13 variants at AF of ≥ 15%. The WUCaMP assay is a robust and sensitive method to detect somatic variants of clinical significance in molecular oncology laboratories, with reduced time and cost of genetic analysis allowing for strategic patient management.


JAMA | 2015

Association Between Mutation Clearance After Induction Therapy and Outcomes in Acute Myeloid Leukemia

Jeffery M. Klco; Christopher A. Miller; Malachi Griffith; Allegra A. Petti; David H. Spencer; Shamika Ketkar-Kulkarni; Lukas D. Wartman; Matthew J. Christopher; Tamara Lamprecht; Nicole M. Helton; Eric J. Duncavage; Jacqueline E. Payton; Jack Baty; Sharon Heath; Obi L. Griffith; Dong Shen; Jasreet Hundal; Gue Su Chang; Robert S. Fulton; Michelle O'Laughlin; Catrina C. Fronick; Vincent Magrini; Ryan Demeter; David E. Larson; Shashikant Kulkarni; Bradley A. Ozenberger; John S. Welch; Matthew J. Walter; Timothy A. Graubert; Peter Westervelt

IMPORTANCE Tests that predict outcomes for patients with acute myeloid leukemia (AML) are imprecise, especially for those with intermediate risk AML. OBJECTIVES To determine whether genomic approaches can provide novel prognostic information for adult patients with de novo AML. DESIGN, SETTING, AND PARTICIPANTS Whole-genome or exome sequencing was performed on samples obtained at disease presentation from 71 patients with AML (mean age, 50.8 years) treated with standard induction chemotherapy at a single site starting in March 2002, with follow-up through January 2015. In addition, deep digital sequencing was performed on paired diagnosis and remission samples from 50 patients (including 32 with intermediate-risk AML), approximately 30 days after successful induction therapy. Twenty-five of the 50 were from the cohort of 71 patients, and 25 were new, additional cases. EXPOSURES Whole-genome or exome sequencing and targeted deep sequencing. Risk of identification based on genetic data. MAIN OUTCOMES AND MEASURES Mutation patterns (including clearance of leukemia-associated variants after chemotherapy) and their association with event-free survival and overall survival. RESULTS Analysis of comprehensive genomic data from the 71 patients did not improve outcome assessment over current standard-of-care metrics. In an analysis of 50 patients with both presentation and documented remission samples, 24 (48%) had persistent leukemia-associated mutations in at least 5% of bone marrow cells at remission. The 24 with persistent mutations had significantly reduced event-free and overall survival vs the 26 who cleared all mutations. Patients with intermediate cytogenetic risk profiles had similar findings. [table: see text]. CONCLUSIONS AND RELEVANCE The detection of persistent leukemia-associated mutations in at least 5% of bone marrow cells in day 30 remission samples was associated with a significantly increased risk of relapse, and reduced overall survival. These data suggest that this genomic approach may improve risk stratification for patients with AML.


Journal of Clinical Investigation | 2009

High throughput digital quantification of mRNA abundance in primary human acute myeloid leukemia samples

Jacqueline E. Payton; Nicole R. Grieselhuber; Li-Wei Chang; Mark Murakami; Gary K. Geiss; Daniel C. Link; Rakesh Nagarajan; Mark A. Watson; Timothy J. Ley

Acute promyelocytic leukemia (APL) is characterized by the t(15;17) chromosomal translocation, which results in fusion of the retinoic acid receptor alpha (RARA) gene to another gene, most commonly promyelocytic leukemia (PML). The resulting fusion protein, PML-RARA, initiates APL, which is a subtype (M3) of acute myeloid leukemia (AML). In this report, we identify a gene expression signature that is specific to M3 samples; it was not found in other AML subtypes and did not simply represent the normal gene expression pattern of primary promyelocytes. To validate this signature for a large number of genes, we tested a recently developed high throughput digital technology (NanoString nCounter). Nearly all of the genes tested demonstrated highly significant concordance with our microarray data (P < 0.05). The validated gene signature reliably identified M3 samples in 2 other AML datasets, and the validated genes were substantially enriched in our mouse model of APL, but not in a cell line that inducibly expressed PML-RARA. These results demonstrate that nCounter is a highly reproducible, customizable system for mRNA quantification using limited amounts of clinical material, which provides a valuable tool for biomarker measurement in low-abundance patient samples.


Journal of Clinical Investigation | 2011

Sequencing a mouse acute promyelocytic leukemia genome reveals genetic events relevant for disease progression

Lukas D. Wartman; David E. Larson; Zhifu Xiang; Li Ding; Ken Chen; Ling Lin; Patrick Cahan; Jeffery M. Klco; John S. Welch; Cheng Li; Jacqueline E. Payton; Geoffrey L. Uy; Nobish Varghese; Rhonda E. Ries; Mieke Hoock; Daniel C. Koboldt; Michael D. McLellan; Heather K. Schmidt; Robert S. Fulton; Rachel Abbott; Lisa Cook; Sean McGrath; Xian Fan; Adam F. Dukes; Tammi L. Vickery; Joelle Kalicki; Tamara Lamprecht; Timothy A. Graubert; Michael H. Tomasson; Elaine R. Mardis

Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML). It is characterized by the t(15;17)(q22;q11.2) chromosomal translocation that creates the promyelocytic leukemia-retinoic acid receptor α (PML-RARA) fusion oncogene. Although this fusion oncogene is known to initiate APL in mice, other cooperating mutations, as yet ill defined, are important for disease pathogenesis. To identify these, we used a mouse model of APL, whereby PML-RARA expressed in myeloid cells leads to a myeloproliferative disease that ultimately evolves into APL. Sequencing of a mouse APL genome revealed 3 somatic, nonsynonymous mutations relevant to APL pathogenesis, of which 1 (Jak1 V657F) was found to be recurrent in other affected mice. This mutation was identical to the JAK1 V658F mutation previously found in human APL and acute lymphoblastic leukemia samples. Further analysis showed that JAK1 V658F cooperated in vivo with PML-RARA, causing a rapidly fatal leukemia in mice. We also discovered a somatic 150-kb deletion involving the lysine (K)-specific demethylase 6A (Kdm6a, also known as Utx) gene, in the mouse APL genome. Similar deletions were observed in 3 out of 14 additional mouse APL samples and 1 out of 150 human AML samples. In conclusion, whole genome sequencing of mouse cancer genomes can provide an unbiased and comprehensive approach for discovering functionally relevant mutations that are also present in human leukemias.


The Journal of Molecular Diagnostics | 2013

Detection of FLT3 Internal Tandem Duplication in Targeted, Short-Read-Length, Next-Generation Sequencing Data

David H. Spencer; Haley J. Abel; Christina M. Lockwood; Jacqueline E. Payton; Philippe Szankasi; Todd W. Kelley; Shashikant Kulkarni; John D. Pfeifer; Eric J. Duncavage

A recurrent somatic mutation frequently found in cytogenetically normal acute myeloid leukemia (AML) is internal tandem duplication (ITD) in the fms-related tyrosine kinase 3 gene (FLT3). This mutation is generally detected in the clinical laboratory by PCR and electrophoresis-based product sizing. As the number of clinically relevant somatic mutations in AML increases, it becomes increasingly attractive to incorporate FLT3 ITD testing into multiplex assays for many somatic mutations simultaneously, using next-generation sequencing (NGS). However, the performance of most NGS analysis tools for identifying medium-size insertions such as FLT3 ITD mutations is largely unknown. We used a multigene, targeted NGS assay to obtain deep sequence coverage (>1000-fold) of FLT3 and 26 other genes from 22 FLT3 ITD-positive and 29 ITD-negative specimens to examine the performance of several commonly used NGS analysis tools for identifying FLT3 ITD mutations. ITD mutations were present in hybridization-capture sequencing data, and Pindel was the only tool out of the seven tested that reliably detected these insertions. Pindel had 100% sensitivity (95% CI = 83% to 100%) and 100% specificity (95% CI = 88% to 100%) in our samples; Pindel provided accurate ITD insertion sizes and was able to detect ITD alleles present at estimated frequencies as low as 1%. These data demonstrate that FLT3 ITDs can be reliably detected in panel-based, next-generation sequencing assays.

Collaboration


Dive into the Jacqueline E. Payton's collaboration.

Top Co-Authors

Avatar

Timothy J. Ley

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Daniel C. Link

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Mark A. Watson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Michael D. McLellan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sharon Heath

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John F. DiPersio

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Elaine R. Mardis

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rakesh Nagarajan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert S. Fulton

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge