scispace - formally typeset
Search or ask a question
Author

Barbara A. Weir

Bio: Barbara A. Weir is an academic researcher from Broad Institute. The author has contributed to research in topics: Cancer & Lung cancer. The author has an hindex of 51, co-authored 75 publications receiving 37727 citations. Previous affiliations of Barbara A. Weir include Harvard University & Queen's University.


Papers
More filters
Journal ArticleDOI
23 Oct 2008-Nature
TL;DR: The interim integrative analysis of DNA copy number, gene expression and DNA methylation aberrations in 206 glioblastomas reveals a link between MGMT promoter methylation and a hypermutator phenotype consequent to mismatch repair deficiency in treated gliobeasts, demonstrating that it can rapidly expand knowledge of the molecular basis of cancer.
Abstract: Human cancer cells typically harbour multiple chromosomal aberrations, nucleotide substitutions and epigenetic modifications that drive malignant transformation. The Cancer Genome Atlas ( TCGA) pilot project aims to assess the value of large- scale multi- dimensional analysis of these molecular characteristics in human cancer and to provide the data rapidly to the research community. Here we report the interim integrative analysis of DNA copy number, gene expression and DNA methylation aberrations in 206 glioblastomas - the most common type of primary adult brain cancer - and nucleotide sequence aberrations in 91 of the 206 glioblastomas. This analysis provides new insights into the roles of ERBB2, NF1 and TP53, uncovers frequent mutations of the phosphatidylinositol- 3- OH kinase regulatory subunit gene PIK3R1, and provides a network view of the pathways altered in the development of glioblastoma. Furthermore, integration of mutation, DNA methylation and clinical treatment data reveals a link between MGMT promoter methylation and a hypermutator phenotype consequent to mismatch repair deficiency in treated glioblastomas, an observation with potential clinical implications. Together, these findings establish the feasibility and power of TCGA, demonstrating that it can rapidly expand knowledge of the molecular basis of cancer.

6,761 citations

Journal ArticleDOI
TL;DR: A robust gene expression-based molecular classification of GBM into Proneural, Neural, Classical, and Mesenchymal subtypes is described and multidimensional genomic data is integrated to establish patterns of somatic mutations and DNA copy number.

5,764 citations

01 Jan 2010
TL;DR: The Cancer Genome Atlas Network recently cataloged recurrent genomic abnormalities in glioblastoma multiforme (GBM) and proposed a robust gene expression-based molecular classification of GBM into Proneural, Neural, Classical, and Mesenchymal subtypes as discussed by the authors.
Abstract: The Cancer Genome Atlas Network recently cataloged recurrent genomic abnormalities in glioblastoma multiforme (GBM). We describe a robust gene expression-based molecular classification of GBM into Proneural, Neural, Classical, and Mesenchymal subtypes and integrate multidimensional genomic data to establish patterns of somatic mutations and DNA copy number. Aberrations and gene expression of EGFR, NF1, and PDGFRA/IDH1 each define the Classical, Mesenchymal, and Proneural subtypes, respectively. Gene signatures of normal brain cell types show a strong relationship between subtypes and different neural lineages. Additionally, response to aggressive therapy differs by subtype, with the greatest benefit in the Classical subtype and no benefit in the Proneural subtype. We provide a framework that unifies transcriptomic and genomic dimensions for GBM molecular stratification with important implications for future studies.

4,464 citations

Journal ArticleDOI
Rameen Beroukhim, Craig H. Mermel1, Craig H. Mermel2, Dale Porter3, Guo Wei2, Soumya Raychaudhuri4, Soumya Raychaudhuri2, Jerry Donovan3, Jordi Barretina2, Jordi Barretina1, Jesse S. Boehm2, Jennifer Dobson1, Jennifer Dobson2, Mitsuyoshi Urashima5, Kevin T. Mc Henry3, Reid M. Pinchback2, Azra H. Ligon4, Yoon Jae Cho6, Leila Haery2, Leila Haery1, Heidi Greulich, Michael R. Reich2, Wendy Winckler2, Michael S. Lawrence2, Barbara A. Weir1, Barbara A. Weir2, Kumiko E. Tanaka1, Kumiko E. Tanaka2, Derek Y. Chiang1, Derek Y. Chiang7, Derek Y. Chiang2, Adam J. Bass4, Adam J. Bass1, Adam J. Bass2, Alice Loo3, Carter Hoffman1, Carter Hoffman2, John R. Prensner1, John R. Prensner2, Ted Liefeld2, Qing Gao2, Derek Yecies1, Sabina Signoretti1, Sabina Signoretti4, Elizabeth A. Maher8, Frederic J. Kaye, Hidefumi Sasaki9, Joel E. Tepper7, Jonathan A. Fletcher4, Josep Tabernero10, José Baselga10, Ming-Sound Tsao11, Francesca Demichelis12, Mark A. Rubin12, Pasi A. Jänne4, Pasi A. Jänne1, Mark J. Daly1, Mark J. Daly2, Carmelo Nucera13, Ross L. Levine14, Benjamin L. Ebert4, Benjamin L. Ebert1, Benjamin L. Ebert2, Stacey Gabriel2, Anil K. Rustgi15, Cristina R. Antonescu14, Marc Ladanyi14, Anthony Letai1, Levi A. Garraway1, Levi A. Garraway2, Massimo Loda4, Massimo Loda1, David G. Beer16, Lawrence D. True17, Aikou Okamoto5, Scott L. Pomeroy6, Samuel Singer14, Todd R. Golub2, Todd R. Golub18, Todd R. Golub1, Eric S. Lander2, Eric S. Lander1, Eric S. Lander19, Gad Getz2, William R. Sellers3, Matthew Meyerson2, Matthew Meyerson1 
18 Feb 2010-Nature
TL;DR: It is demonstrated that cancer cells containing amplifications surrounding the MCL1 and BCL2L1 anti-apoptotic genes depend on the expression of these genes for survival, and a large majority of SCNAs identified in individual cancer types are present in several cancer types.
Abstract: A powerful way to discover key genes with causal roles in oncogenesis is to identify genomic regions that undergo frequent alteration in human cancers. Here we present high-resolution analyses of somatic copy-number alterations (SCNAs) from 3,131 cancer specimens, belonging largely to 26 histological types. We identify 158 regions of focal SCNA that are altered at significant frequency across several cancer types, of which 122 cannot be explained by the presence of a known cancer target gene located within these regions. Several gene families are enriched among these regions of focal SCNA, including the BCL2 family of apoptosis regulators and the NF-kappaBeta pathway. We show that cancer cells containing amplifications surrounding the MCL1 and BCL2L1 anti-apoptotic genes depend on the expression of these genes for survival. Finally, we demonstrate that a large majority of SCNAs identified in individual cancer types are present in several cancer types.

3,375 citations

Journal ArticleDOI
Li Ding1, Gad Getz2, David A. Wheeler3, Elaine R. Mardis1, Michael D. McLellan1, Kristian Cibulskis2, Carrie Sougnez2, Heidi Greulich2, Heidi Greulich4, Donna M. Muzny3, Margaret Morgan3, Lucinda Fulton1, Robert S. Fulton1, Qunyuan Zhang1, Michael C. Wendl1, Michael S. Lawrence2, David E. Larson1, Ken Chen1, David J. Dooling1, Aniko Sabo3, Alicia Hawes3, Hua Shen3, Shalini N. Jhangiani3, Lora Lewis3, Otis Hall3, Yiming Zhu3, Tittu Mathew3, Yanru Ren3, Jiqiang Yao3, Steven E. Scherer3, Kerstin Clerc3, Ginger A. Metcalf3, Brian Ng3, Aleksandar Milosavljevic3, Manuel L. Gonzalez-Garay3, John R. Osborne1, Rick Meyer1, Xiaoqi Shi1, Yuzhu Tang1, Daniel C. Koboldt1, Ling Lin1, Rachel Abbott1, Tracie L. Miner1, Craig Pohl1, Ginger A. Fewell1, Carrie A. Haipek1, Heather Schmidt1, Brian H. Dunford-Shore1, Aldi T. Kraja1, Seth D. Crosby1, Christopher S. Sawyer1, Tammi L. Vickery1, Sacha N. Sander1, Jody S. Robinson1, Wendy Winckler2, Wendy Winckler4, Jennifer Baldwin2, Lucian R. Chirieac4, Amit Dutt4, Amit Dutt2, Timothy Fennell2, Megan Hanna2, Megan Hanna4, Bruce E. Johnson4, Robert C. Onofrio2, Roman K. Thomas5, Giovanni Tonon4, Barbara A. Weir4, Barbara A. Weir2, Xiaojun Zhao4, Xiaojun Zhao2, Liuda Ziaugra2, Michael C. Zody2, Thomas J. Giordano6, Mark B. Orringer6, Jack A. Roth, Margaret R. Spitz7, Ignacio I. Wistuba, Bradley A. Ozenberger8, Peter J. Good8, Andrew C. Chang6, David G. Beer6, Mark A. Watson1, Marc Ladanyi9, Stephen R. Broderick9, Akihiko Yoshizawa9, William D. Travis9, William Pao9, Michael A. Province1, George M. Weinstock1, Harold E. Varmus9, Stacey Gabriel2, Eric S. Lander2, Richard A. Gibbs3, Matthew Meyerson4, Matthew Meyerson2, Richard K. Wilson1 
23 Oct 2008-Nature
TL;DR: Somatic mutations in primary lung adenocarcinoma for several tumour suppressor genes involved in other cancers and for sequence changes in PTPRD as well as the frequently deleted gene LRP1B are found.
Abstract: Determining the genetic basis of cancer requires comprehensive analyses of large collections of histopathologically well-classified primary tumours. Here we report the results of a collaborative study to discover somatic mutations in 188 human lung adenocarcinomas. DNA sequencing of 623 genes with known or potential relationships to cancer revealed more than 1,000 somatic mutations across the samples. Our analysis identified 26 genes that are mutated at significantly high frequencies and thus are probably involved in carcinogenesis. The frequently mutated genes include tyrosine kinases, among them the EGFR homologue ERBB4; multiple ephrin receptor genes, notably EPHA3; vascular endothelial growth factor receptor KDR; and NTRK genes. These data provide evidence of somatic mutations in primary lung adenocarcinoma for several tumour suppressor genes involved in other cancers--including NF1, APC, RB1 and ATM--and for sequence changes in PTPRD as well as the frequently deleted gene LRP1B. The observed mutational profiles correlate with clinical features, smoking status and DNA repair defects. These results are reinforced by data integration including single nucleotide polymorphism array and gene expression array. Our findings shed further light on several important signalling pathways involved in lung adenocarcinoma, and suggest new molecular targets for treatment.

2,615 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.
Abstract: The cBio Cancer Genomics Portal (http://cbioportal.org) is an open-access resource for interactive exploration of multidimensional cancer genomics data sets, currently providing access to data from more than 5,000 tumor samples from 20 cancer studies. The cBio Cancer Genomics Portal significantly lowers the barriers between complex genomic data and cancer researchers who want rapid, intuitive, and high-quality access to molecular profiles and clinical attributes from large-scale cancer genomics projects and empowers researchers to translate these rich data sets into biologic insights and clinical applications.

11,912 citations

Journal ArticleDOI
TL;DR: A practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics, which makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries.
Abstract: The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.

10,947 citations

Journal ArticleDOI
TL;DR: In this article, the authors present an approach for efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data.
Abstract: Rapid improvements in sequencing and array-based platforms are resulting in a flood of diverse genome-wide data, including data from exome and whole-genome sequencing, epigenetic surveys, expression profiling of coding and noncoding RNAs, single nucleotide polymorphism (SNP) and copy number profiling, and functional assays. Analysis of these large, diverse data sets holds the promise of a more comprehensive understanding of the genome and its relation to human disease. Experienced and knowledgeable human review is an essential component of this process, complementing computational approaches. This calls for efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data. However, the sheer volume and scope of data pose a significant challenge to the development of such tools.

10,798 citations

Journal ArticleDOI
TL;DR: A unified analytic framework to discover and genotype variation among multiple samples simultaneously that achieves sensitive and specific results across five sequencing technologies and three distinct, canonical experimental designs is presented.
Abstract: Recent advances in sequencing technology make it possible to comprehensively catalogue genetic variation in population samples, creating a foundation for understanding human disease, ancestry and evolution. The amounts of raw data produced are prodigious and many computational steps are required to translate this output into high-quality variant calls. We present a unified analytic framework to discover and genotype variation among multiple samples simultaneously that achieves sensitive and specific results across five sequencing technologies and three distinct, canonical experimental designs. Our process includes (1) initial read mapping; (2) local realignment around indels; (3) base quality score recalibration; (4) SNP discovery and genotyping to find all potential variants; and (5) machine learning to separate true segregating variation from machine artifacts common to next-generation sequencing technologies. We discuss the application of these tools, instantiated in the Genome Analysis Toolkit (GATK), to deep whole-genome, whole-exome capture, and multi-sample low-pass (~4×) 1000 Genomes Project datasets.

10,056 citations

Journal ArticleDOI
04 Oct 2012-Nature
TL;DR: The ability to integrate information across platforms provided key insights into previously defined gene expression subtypes and demonstrated the existence of four main breast cancer classes when combining data from five platforms, each of which shows significant molecular heterogeneity.
Abstract: We analysed primary breast cancers by genomic DNA copy number arrays, DNA methylation, exome sequencing, messenger RNA arrays, microRNA sequencing and reverse-phase protein arrays. Our ability to integrate information across platforms provided key insights into previously defined gene expression subtypes and demonstrated the existence of four main breast cancer classes when combining data from five platforms, each of which shows significant molecular heterogeneity. Somatic mutations in only three genes (TP53, PIK3CA and GATA3) occurred at >10% incidence across all breast cancers; however, there were numerous subtype-associated and novel gene mutations including the enrichment of specific mutations in GATA3, PIK3CA and MAP3K1 with the luminal A subtype. We identified two novel protein-expression-defined subgroups, possibly produced by stromal/microenvironmental elements, and integrated analyses identified specific signalling pathways dominant in each molecular subtype including a HER2/phosphorylated HER2/EGFR/phosphorylated EGFR signature within the HER2-enriched expression subtype. Comparison of basal-like breast tumours with high-grade serous ovarian tumours showed many molecular commonalities, indicating a related aetiology and similar therapeutic opportunities. The biological finding of the four main breast cancer subtypes caused by different subsets of genetic and epigenetic abnormalities raises the hypothesis that much of the clinically observable plasticity and heterogeneity occurs within, and not across, these major biological subtypes of breast cancer.

9,355 citations