Network


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

Hotspot


Dive into the research topics where Jaime M. Guidry Auvil is active.

Publication


Featured researches published by Jaime M. Guidry Auvil.


Nature Genetics | 2013

The genetic landscape of high-risk neuroblastoma

Trevor J. Pugh; Olena Morozova; Edward F. Attiyeh; Shahab Asgharzadeh; Jun S. Wei; Daniel Auclair; Scott L. Carter; Kristian Cibulskis; Megan Hanna; Adam Kiezun; Jaegil Kim; Michael S. Lawrence; Lee Lichenstein; Aaron McKenna; Chandra Sekhar Pedamallu; Alex H. Ramos; Erica Shefler; Andrey Sivachenko; Carrie Sougnez; Chip Stewart; Adrian Ally; Inanc Birol; Readman Chiu; Richard Corbett; Martin Hirst; Shaun D. Jackman; Baljit Kamoh; Alireza Hadj Khodabakshi; Martin Krzywinski; Allan Lo

Neuroblastoma is a malignancy of the developing sympathetic nervous system that often presents with widespread metastatic disease, resulting in survival rates of less than 50%. To determine the spectrum of somatic mutation in high-risk neuroblastoma, we studied 240 affected individuals (cases) using a combination of whole-exome, genome and transcriptome sequencing as part of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative. Here we report a low median exonic mutation frequency of 0.60 per Mb (0.48 nonsilent) and notably few recurrently mutated genes in these tumors. Genes with significant somatic mutation frequencies included ALK (9.2% of cases), PTPN11 (2.9%), ATRX (2.5%, and an additional 7.1% had focal deletions), MYCN (1.7%, causing a recurrent p.Pro44Leu alteration) and NRAS (0.83%). Rare, potentially pathogenic germline variants were significantly enriched in ALK, CHEK2, PINK1 and BARD1. The relative paucity of recurrent somatic mutations in neuroblastoma challenges current therapeutic strategies that rely on frequently altered oncogenic drivers.


Nature Genetics | 2015

Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations

Thomas F. Eleveld; Derek A. Oldridge; Virginie Bernard; Jan Koster; Leo Colmet Daage; Sharon J. Diskin; Linda Schild; Nadia Bessoltane Bentahar; Angela Bellini; Mathieu Chicard; Eve Lapouble; Valérie Combaret; Patricia Legoix-Né; Jean Michon; Trevor J. Pugh; Lori S. Hart; JulieAnn Rader; Edward F. Attiyeh; Jun S. Wei; Shile Zhang; Arlene Naranjo; Julie M. Gastier-Foster; Michael D. Hogarty; Shahab Asgharzadeh; Malcolm A. Smith; Jaime M. Guidry Auvil; Thomas B. K. Watkins; Danny A. Zwijnenburg; Marli E. Ebus; Peter van Sluis

The majority of patients with neuroblastoma have tumors that initially respond to chemotherapy, but a large proportion will experience therapy-resistant relapses. The molecular basis of this aggressive phenotype is unknown. Whole-genome sequencing of 23 paired diagnostic and relapse neuroblastomas showed clonal evolution from the diagnostic tumor, with a median of 29 somatic mutations unique to the relapse sample. Eighteen of the 23 relapse tumors (78%) showed mutations predicted to activate the RAS-MAPK pathway. Seven of these events were detected only in the relapse tumor, whereas the others showed clonal enrichment. In neuroblastoma cell lines, we also detected a high frequency of activating mutations in the RAS-MAPK pathway (11/18; 61%), and these lesions predicted sensitivity to MEK inhibition in vitro and in vivo. Our findings provide a rationale for genetic characterization of relapse neuroblastomas and show that RAS-MAPK pathway mutations may function as a biomarker for new therapeutic approaches to refractory disease.


Nature Communications | 2015

Rise and fall of subclones from diagnosis to relapse in pediatric B-acute lymphoblastic leukaemia

Xiaotu Ma; Michael Edmonson; Donald Yergeau; Donna M. Muzny; Oliver A. Hampton; Michael Rusch; Guangchun Song; John Easton; Richard C. Harvey; David A. Wheeler; Jing Ma; HarshaVardhan Doddapaneni; Bhavin Vadodaria; Gang Wu; Panduka Nagahawatte; William L. Carroll; I-Ming Chen; Julie M. Gastier-Foster; Mary V. Relling; Malcolm A. Smith; Meenakshi Devidas; Jaime M. Guidry Auvil; James R. Downing; Mignon L. Loh; Cheryl L. Willman; Daniela S. Gerhard; Charles G. Mullighan; Stephen P. Hunger; Jinghui Zhang

There is incomplete understanding of genetic heterogeneity and clonal evolution during cancer progression. Here we use deep whole-exome sequencing to describe the clonal architecture and evolution of 20 pediatric B-acute lymphoblastic leukaemias from diagnosis to relapse. We show that clonal diversity is comparable at diagnosis and relapse and clonal survival from diagnosis to relapse is not associated with mutation burden. Six pathways were frequently mutated, with NT5C2, CREBBP, WHSC1, TP53, USH2A, NRAS and IKZF1 mutations enriched at relapse. Half of the leukaemias had multiple subclonal mutations in a pathway or gene at diagnosis, but mostly with only one, usually minor clone, surviving therapy to acquire additional mutations and become the relapse founder clone. Relapse-specific mutations in NT5C2 were found in nine cases, with mutations in four cases being in descendants of the relapse founder clone. These results provide important insights into the genetic basis of treatment failure in ALL and have implications for the early detection of mutations driving relapse.


Nature Genetics | 2017

The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia

Yu Liu; John Easton; Ying Shao; Jamie L. Maciaszek; Zhaoming Wang; Mark R. Wilkinson; Kelly McCastlain; Michael Edmonson; Stanley Pounds; Lei Shi; Xin Zhou; Xiaotu Ma; Edgar Sioson; Yongjin Li; Michael Rusch; Pankaj Gupta; Deqing Pei; Cheng Cheng; Malcolm A. Smith; Jaime M. Guidry Auvil; Daniela S. Gerhard; Mary V. Relling; Naomi J. Winick; Andrew J. Carroll; Nyla A. Heerema; Elizabeth A. Raetz; Meenakshi Devidas; Cheryl L. Willman; Richard C. Harvey; William L. Carroll

Genetic alterations that activate NOTCH1 signaling and T cell transcription factors, coupled with inactivation of the INK4/ARF tumor suppressors, are hallmarks of T-lineage acute lymphoblastic leukemia (T-ALL), but detailed genome-wide sequencing of large T-ALL cohorts has not been carried out. Using integrated genomic analysis of 264 T-ALL cases, we identified 106 putative driver genes, half of which had not previously been described in childhood T-ALL (for example, CCND3, CTCF, MYB, SMARCA4, ZFP36L2 and MYCN). We describe new mechanisms of coding and noncoding alteration and identify ten recurrently altered pathways, with associations between mutated genes and pathways, and stage or subtype of T-ALL. For example, NRAS/FLT3 mutations were associated with immature T-ALL, JAK3/STAT5B mutations in HOXA1 deregulated ALL, PTPN2 mutations in TLX1 deregulated T-ALL, and PIK3R1/PTEN mutations in TAL1 deregulated ALL, which suggests that different signaling pathways have distinct roles according to maturational stage. This genomic landscape provides a logical framework for the development of faithful genetic models and new therapeutic approaches.


Cancer Research | 2016

Genomic profiling of pediatric acute myeloid leukemia reveals a changing mutational landscape from disease diagnosis to relapse

Jason E. Farrar; Heather L. Schuback; Rhonda E. Ries; Daniel Wai; Oliver A. Hampton; Lisa R. Trevino; Todd A. Alonzo; Jaime M. Guidry Auvil; Tanja M. Davidsen; Patee Gesuwan; Leandro C. Hermida; Donna M. Muzny; Ninad Dewal; Navin Rustagi; Lora Lewis; Alan S. Gamis; David A. Wheeler; Malcolm A. Smith; Daniela S. Gerhard; Soheil Meshinchi

The genomic and clinical information used to develop and implement therapeutic approaches for acute myelogenous leukemia (AML) originated primarily from adult patients and has been generalized to patients with pediatric AML. However, age-specific molecular alterations are becoming more evident and may signify the need to age-stratify treatment regimens. The NCI/COG TARGET-AML initiative used whole exome capture sequencing (WXS) to interrogate the genomic landscape of matched trios representing specimens collected upon diagnosis, remission, and relapse from 20 cases of de novo childhood AML. One hundred forty-five somatic variants at diagnosis (median 6 mutations/patient) and 149 variants at relapse (median 6.5 mutations) were identified and verified by orthogonal methodologies. Recurrent somatic variants [in (greater than or equal to) 2 patients] were identified for 10 genes (FLT3, NRAS, PTPN11, WT1, TET2, DHX15, DHX30, KIT, ETV6, KRAS), with variable persistence at relapse. The variant allele fraction (VAF), used to measure the prevalence of somatic mutations, varied widely at diagnosis. Mutations that persisted from diagnosis to relapse had a significantly higher diagnostic VAF compared with those that resolved at relapse (median VAF 0.43 vs. 0.24, P < 0.001). Further analysis revealed that 90% of the diagnostic variants with VAF >0.4 persisted to relapse compared with 28% with VAF <0.2 (P < 0.001). This study demonstrates significant variability in the mutational profile and clonal evolution of pediatric AML from diagnosis to relapse. Furthermore, mutations with high VAF at diagnosis, representing variants shared across a leukemic clonal structure, may constrain the genomic landscape at relapse and help to define key pathways for therapeutic targeting. Cancer Res; 76(8); 2197-205. ©2016 AACR.


Nature Genetics | 2017

A Children's Oncology Group and TARGET initiative exploring the genetic landscape of Wilms tumor

Samantha Gadd; Vicki Huff; Amy L. Walz; Ariadne H. A. G. Ooms; Amy E. Armstrong; Daniela S. Gerhard; Malcolm A. Smith; Jaime M. Guidry Auvil; Daoud Meerzaman; Qing Rong Chen; Chih Hao Hsu; Chunhua Yan; Cu Nguyen; Ying Hu; Leandro C. Hermida; Tanja M. Davidsen; Patee Gesuwan; Yussanne Ma; Zusheng Zong; Andrew J. Mungall; Richard A. Moore; Marco A. Marra; Jeffrey S. Dome; Charles G. Mullighan; Jing Ma; David A. Wheeler; Oliver A. Hampton; Nicole Ross; Julie M. Gastier-Foster; Stefan T. Arold

We performed genome-wide sequencing and analyzed mRNA and miRNA expression, DNA copy number, and DNA methylation in 117 Wilms tumors, followed by targeted sequencing of 651 Wilms tumors. In addition to genes previously implicated in Wilms tumors (WT1, CTNNB1, AMER1, DROSHA, DGCR8, XPO5, DICER1, SIX1, SIX2, MLLT1, MYCN, and TP53), we identified mutations in genes not previously recognized as recurrently involved in Wilms tumors, the most frequent being BCOR, BCORL1, NONO, MAX, COL6A3, ASXL1, MAP3K4, and ARID1A. DNA copy number changes resulted in recurrent 1q gain, MYCN amplification, LIN28B gain, and MIRLET7A loss. Unexpected germline variants involved PALB2 and CHEK2. Integrated analyses support two major classes of genetic changes that preserve the progenitor state and/or interrupt normal development.


Nature Medicine | 2017

The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions

Hamid Bolouri; Jason E. Farrar; Timothy J. Triche; Rhonda E. Ries; Emilia L. Lim; Todd A. Alonzo; Yussanne Ma; Richard G. Moore; Andrew J. Mungall; Marco A. Marra; Jinghui Zhang; Xiaotu Ma; Yu Liu; Yanling Liu; Jaime M. Guidry Auvil; Tanja M. Davidsen; Patee Gesuwan; Leandro C. Hermida; Bodour Salhia; Stephen Capone; Giridharan Ramsingh; Christian M. Zwaan; Sanne Noort; Stephen R. Piccolo; E. Anders Kolb; Alan S. Gamis; Malcolm A. Smith; Daniela S. Gerhard; Soheil Meshinchi

We present the molecular landscape of pediatric acute myeloid leukemia (AML) and characterize nearly 1,000 participants in Childrens Oncology Group (COG) AML trials. The COG–National Cancer Institute (NCI) TARGET AML initiative assessed cases by whole-genome, targeted DNA, mRNA and microRNA sequencing and CpG methylation profiling. Validated DNA variants corresponded to diverse, infrequent mutations, with fewer than 40 genes mutated in >2% of cases. In contrast, somatic structural variants, including new gene fusions and focal deletions of MBNL1, ZEB2 and ELF1, were disproportionately prevalent in young individuals as compared to adults. Conversely, mutations in DNMT3A and TP53, which were common in adults, were conspicuously absent from virtually all pediatric cases. New mutations in GATA2, FLT3 and CBL and recurrent mutations in MYC-ITD, NRAS, KRAS and WT1 were frequent in pediatric AML. Deletions, mutations and promoter DNA hypermethylation convergently impacted Wnt signaling, Polycomb repression, innate immune cell interactions and a cluster of zinc finger–encoding genes associated with KMT2A rearrangements. These results highlight the need for and facilitate the development of age-tailored targeted therapies for the treatment of pediatric AML.


Nature Communications | 2015

MLLT1 YEATS domain mutations in clinically distinctive Favourable Histology Wilms tumours.

Elizabeth J. Perlman; Samantha Gadd; Stefan T. Arold; Anand Radhakrishnan; Daniela S. Gerhard; Lawrence J. Jennings; Vicki Huff; Jaime M. Guidry Auvil; Tanja M. Davidsen; Jeffrey S. Dome; Daoud Meerzaman; Chih Hao Hsu; Cu Nguyen; James M. Anderson; Yussanne Ma; Andrew J. Mungall; Richard A. Moore; Marco A. Marra; Charles G. Mullighan; Jing Ma; David A. Wheeler; Oliver A. Hampton; Julie M. Gastier-Foster; Nicole Ross; Malcolm A. Smith

Wilms tumour is an embryonal tumour of childhood that closely resembles the developing kidney. Genomic changes responsible for the development of the majority of Wilms tumours remain largely unknown. Here we identify recurrent mutations within Wilms tumours that involve the highly conserved YEATS domain of MLLT1 (ENL), a gene known to be involved in transcriptional elongation during early development. The mutant MLLT1 protein shows altered binding to acetylated histone tails. Moreover, MLLT1-mutant tumours show an increase in MYC gene expression and HOX dysregulation. Patients with MLLT1-mutant tumours present at a younger age and have a high prevalence of precursor intralobar nephrogenic rests. These data support a model whereby activating MLLT1 mutations early in renal development result in the development of Wilms tumour.


Nature | 2018

Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours

Xiaotu Ma; Yu Liu; Yanling Liu; Ludmil B. Alexandrov; Michael Edmonson; Charles Gawad; Xin Zhou; Yongjin Li; Michael Rusch; John Easton; Robert Huether; Veronica Gonzalez-Pena; Mark R. Wilkinson; Leandro C. Hermida; Sean Davis; Edgar Sioson; Stanley Pounds; Xueyuan Cao; Rhonda E. Ries; Zhaoming Wang; Xiang Chen; Li Dong; Sharon J. Diskin; Malcolm A. Smith; Jaime M. Guidry Auvil; Paul S. Meltzer; Ching C. Lau; Elizabeth J. Perlman; John M. Maris; Soheil Meshinchi

Analysis of molecular aberrations across multiple cancer types, known as pan-cancer analysis, identifies commonalities and differences in key biological processes that are dysregulated in cancer cells from diverse lineages. Pan-cancer analyses have been performed for adult but not paediatric cancers, which commonly occur in developing mesodermic rather than adult epithelial tissues. Here we present a pan-cancer study of somatic alterations, including single nucleotide variants, small insertions or deletions, structural variations, copy number alterations, gene fusions and internal tandem duplications in 1,699 paediatric leukaemias and solid tumours across six histotypes, with whole-genome, whole-exome and transcriptome sequencing data processed under a uniform analytical framework. We report 142 driver genes in paediatric cancers, of which only 45% match those found in adult pan-cancer studies; copy number alterations and structural variants constituted the majority (62%) of events. Eleven genome-wide mutational signatures were identified, including one attributed to ultraviolet-light exposure in eight aneuploid leukaemias. Transcription of the mutant allele was detectable for 34% of protein-coding mutations, and 20% exhibited allele-specific expression. These data provide a comprehensive genomic architecture for paediatric cancers and emphasize the need for paediatric cancer-specific development of precision therapies.


Blood | 2016

CSF3R mutations have a high degree of overlap with CEBPA mutations in pediatric AML.

Julia E. Maxson; Rhonda E. Ries; Yi Cheng Wang; Robert B. Gerbing; E. Anders Kolb; Sarah L Thompson; Jaime M. Guidry Auvil; Marco A. Marra; Yussanne Ma; Zusheng Zong; Andrew J. Mungall; Richard G. Moore; William Long; Patee Gesuwan; Tanja M. Davidsen; Leandro C. Hermida; Seamus B Hughes; Jason E. Farrar; Jerald P. Radich; Malcolm A. Smith; Daniela S. Gerhard; Alan S. Gamis; Todd A. Alonzo; Soheil Meshinchi

Publishers Note: There is an [Inside Blood Commentary][1] on this article in this issue. To the editor: Childhood cancers represent distinct clinical entities, often with unique genomic alterations and therapeutic responses that differ from cancers arising in adults. Pediatric acute myeloid

Collaboration


Dive into the Jaime M. Guidry Auvil's collaboration.

Top Co-Authors

Avatar

Daniela S. Gerhard

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Malcolm A. Smith

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tanja M. Davidsen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Patee Gesuwan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Leandro C. Hermida

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Soheil Meshinchi

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Alan S. Gamis

Children's Mercy Hospital

View shared research outputs
Top Co-Authors

Avatar

Jason E. Farrar

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Rhonda E. Ries

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Todd A. Alonzo

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge