Network


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

Hotspot


Dive into the research topics where Sharon Heath is active.

Publication


Featured researches published by Sharon Heath.


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.


The New England Journal of Medicine | 2012

Clonal Architecture of Secondary Acute Myeloid Leukemia

Matthew J. Walter; Dong Shen; Li Ding; Jin Shao; Daniel C. Koboldt; Ken Chen; David E. Larson; Michael D. McLellan; David J. Dooling; Rachel Abbott; Robert S. Fulton; Vincent Magrini; Heather K. Schmidt; Joelle Kalicki-Veizer; Michelle O'Laughlin; Xian Fan; Marcus Grillot; Sarah Witowski; Sharon Heath; John L. Frater; William C. Eades; Michael H. Tomasson; Peter Westervelt; John F. DiPersio; Daniel C. Link; Elaine R. Mardis; Timothy J. Ley; Richard Wilson; Timothy A. Graubert

BACKGROUND The myelodysplastic syndromes are a group of hematologic disorders that often evolve into secondary acute myeloid leukemia (AML). The genetic changes that underlie progression from the myelodysplastic syndromes to secondary AML are not well understood. METHODS We performed whole-genome sequencing of seven paired samples of skin and bone marrow in seven subjects with secondary AML to identify somatic mutations specific to secondary AML. We then genotyped a bone marrow sample obtained during the antecedent myelodysplastic-syndrome stage from each subject to determine the presence or absence of the specific somatic mutations. We identified recurrent mutations in coding genes and defined the clonal architecture of each pair of samples from the myelodysplastic-syndrome stage and the secondary-AML stage, using the allele burden of hundreds of mutations. RESULTS Approximately 85% of bone marrow cells were clonal in the myelodysplastic-syndrome and secondary-AML samples, regardless of the myeloblast count. The secondary-AML samples contained mutations in 11 recurrently mutated genes, including 4 genes that have not been previously implicated in the myelodysplastic syndromes or AML. In every case, progression to acute leukemia was defined by the persistence of an antecedent founding clone containing 182 to 660 somatic mutations and the outgrowth or emergence of at least one subclone, harboring dozens to hundreds of new mutations. All founding clones and subclones contained at least one mutation in a coding gene. CONCLUSIONS Nearly all the bone marrow cells in patients with myelodysplastic syndromes and secondary AML are clonally derived. Genetic evolution of secondary AML is a dynamic process shaped by multiple cycles of mutation acquisition and clonal selection. Recurrent gene mutations are found in both founding clones and daughter subclones. (Funded by the National Institutes of Health and others.).


Nature Genetics | 2012

RECURRENT MUTATIONS IN THE U2AF1 SPLICING FACTOR IN MYELODYSPLASTIC SYNDROMES

Timothy A. Graubert; Dong Shen; Li Ding; Theresa Okeyo-Owuor; Cara L Lunn; Jin Shao; Kilannin Krysiak; Christopher C. Harris; Daniel C. Koboldt; David E. Larson; Michael D. McLellan; David J. Dooling; Rachel Abbott; Robert S. Fulton; Heather K. Schmidt; Joelle Kalicki-Veizer; Michelle O'Laughlin; Marcus Grillot; Jack Baty; Sharon Heath; John L. Frater; Talat Nasim; Daniel C. Link; Michael H. Tomasson; Peter Westervelt; John F. DiPersio; Elaine R. Mardis; Timothy J. Ley; Richard Wilson; Matthew J. Walter

Myelodysplastic syndromes (MDS) are hematopoietic stem cell disorders that often progress to chemotherapy-resistant secondary acute myeloid leukemia (sAML). We used whole-genome sequencing to perform an unbiased comprehensive screen to discover the somatic mutations in a sample from an individual with sAML and genotyped the loci containing these mutations in the matched MDS sample. Here we show that a missense mutation affecting the serine at codon 34 (Ser34) in U2AF1 was recurrently present in 13 out of 150 (8.7%) subjects with de novo MDS, and we found suggestive evidence of an increased risk of progression to sAML associated with this mutation. U2AF1 is a U2 auxiliary factor protein that recognizes the AG splice acceptor dinucleotide at the 3′ end of introns, and the alterations in U2AF1 are located in highly conserved zinc fingers of this protein. Mutant U2AF1 promotes enhanced splicing and exon skipping in reporter assays in vitro. This previously unidentified, recurrent mutation in U2AF1 implicates altered pre-mRNA splicing as a potential mechanism for MDS pathogenesis.


Nature | 2015

Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia

Terrence N. Wong; Giridharan Ramsingh; Andrew L. Young; Christopher A. Miller; Waseem Touma; John S. Welch; Tamara Lamprecht; Dong Shen; Jasreet Hundal; Robert S. Fulton; Sharon Heath; Jack Baty; Jeffery M. Klco; Li Ding; Elaine R. Mardis; Peter Westervelt; John F. DiPersio; Matthew J. Walter; Timothy A. Graubert; Timothy J. Ley; Todd E. Druley; Daniel C. Link; Richard Wilson

Therapy-related acute myeloid leukaemia (t-AML) and therapy-related myelodysplastic syndrome (t-MDS) are well-recognized complications of cytotoxic chemotherapy and/or radiotherapy. There are several features that distinguish t-AML from de novo AML, including a higher incidence of TP53 mutations, abnormalities of chromosomes 5 or 7, complex cytogenetics and a reduced response to chemotherapy. However, it is not clear how prior exposure to cytotoxic therapy influences leukaemogenesis. In particular, the mechanism by which TP53 mutations are selectively enriched in t-AML/t-MDS is unknown. Here, by sequencing the genomes of 22 patients with t-AML, we show that the total number of somatic single-nucleotide variants and the percentage of chemotherapy-related transversions are similar in t-AML and de novo AML, indicating that previous chemotherapy does not induce genome-wide DNA damage. We identified four cases of t-AML/t-MDS in which the exact TP53 mutation found at diagnosis was also present at low frequencies (0.003–0.7%) in mobilized blood leukocytes or bone marrow 3–6 years before the development of t-AML/t-MDS, including two cases in which the relevant TP53 mutation was detected before any chemotherapy. Moreover, functional TP53 mutations were identified in small populations of peripheral blood cells of healthy chemotherapy-naive elderly individuals. Finally, in mouse bone marrow chimaeras containing both wild-type and Tp53+/− haematopoietic stem/progenitor cells (HSPCs), the Tp53+/− HSPCs preferentially expanded after exposure to chemotherapy. These data suggest that cytotoxic therapy does not directly induce TP53 mutations. Rather, they support a model in which rare HSPCs carrying age-related TP53 mutations are resistant to chemotherapy and expand preferentially after treatment. The early acquisition of TP53 mutations in the founding HSPC clone probably contributes to the frequent cytogenetic abnormalities and poor responses to chemotherapy that are typical of patients with t-AML/t-MDS.


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 New England Journal of Medicine | 2016

TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes

John S. Welch; Allegra A. Petti; Christopher A. Miller; Catrina C. Fronick; Michelle O’Laughlin; Robert S. Fulton; Richard Wilson; Jack Baty; Eric J. Duncavage; Bevan Tandon; Yi-Shan Lee; Lukas D. Wartman; Geoffrey L. Uy; Armin Ghobadi; Michael H. Tomasson; Iskra Pusic; Rizwan Romee; Todd A. Fehniger; Keith Stockerl-Goldstein; Ravi Vij; Stephen T. Oh; Camille N. Abboud; Amanda F. Cashen; Mark A. Schroeder; Meagan A. Jacoby; Sharon Heath; Kierstin Luber; M R Janke; Andrew Hantel; Niloufer Khan

BACKGROUND The molecular determinants of clinical responses to decitabine therapy in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) are unclear. METHODS We enrolled 84 adult patients with AML or MDS in a single-institution trial of decitabine to identify somatic mutations and their relationships to clinical responses. Decitabine was administered at a dose of 20 mg per square meter of body-surface area per day for 10 consecutive days in monthly cycles. We performed enhanced exome or gene-panel sequencing in 67 of these patients and serial sequencing at multiple time points to evaluate patterns of mutation clearance in 54 patients. An extension cohort included 32 additional patients who received decitabine in different protocols. RESULTS Of the 116 patients, 53 (46%) had bone marrow blast clearance (<5% blasts). Response rates were higher among patients with an unfavorable-risk cytogenetic profile than among patients with an intermediate-risk or favorable-risk cytogenetic profile (29 of 43 patients [67%] vs. 24 of 71 patients [34%], P<0.001) and among patients with TP53 mutations than among patients with wild-type TP53 (21 of 21 [100%] vs. 32 of 78 [41%], P<0.001). Previous studies have consistently shown that patients with an unfavorable-risk cytogenetic profile and TP53 mutations who receive conventional chemotherapy have poor outcomes. However, in this study of 10-day courses of decitabine, neither of these risk factors was associated with a lower rate of overall survival than the rate of survival among study patients with intermediate-risk cytogenetic profiles. CONCLUSIONS Patients with AML and MDS who had cytogenetic abnormalities associated with unfavorable risk, TP53 mutations, or both had favorable clinical responses and robust (but incomplete) mutation clearance after receiving serial 10-day courses of decitabine. Although these responses were not durable, they resulted in rates of overall survival that were similar to those among patients with AML who had an intermediate-risk cytogenetic profile and who also received serial 10-day courses of decitabine. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT01687400 .).


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.

Collaboration


Dive into the Sharon Heath's collaboration.

Top Co-Authors

Avatar

Daniel C. Link

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Timothy J. Ley

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

Robert S. Fulton

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Walter

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Peter Westervelt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Richard Wilson

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

John S. Welch

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge