Network


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

Hotspot


Dive into the research topics where Barry S. Taylor is active.

Publication


Featured researches published by Barry S. Taylor.


Cancer Cell | 2010

Integrative Genomic Profiling of Human Prostate Cancer

Barry S. Taylor; Nikolaus Schultz; Haley Hieronymus; Anuradha Gopalan; Yonghong Xiao; Brett S. Carver; Vivek K. Arora; Poorvi Kaushik; Ethan Cerami; Boris Reva; Yevgeniy Antipin; Nicholas Mitsiades; Thomas Landers; Igor Dolgalev; John Major; Manda Wilson; Nicholas D. Socci; Alex E. Lash; Adriana Heguy; James A. Eastham; Howard I. Scher; Victor E. Reuter; Peter T. Scardino; Chris Sander; Charles L. Sawyers; William L. Gerald

Annotation of prostate cancer genomes provides a foundation for discoveries that can impact disease understanding and treatment. Concordant assessment of DNA copy number, mRNA expression, and focused exon resequencing in 218 prostate cancer tumors identified the nuclear receptor coactivator NCOA2 as an oncogene in approximately 11% of tumors. Additionally, the androgen-driven TMPRSS2-ERG fusion was associated with a previously unrecognized, prostate-specific deletion at chromosome 3p14 that implicates FOXP1, RYBP, and SHQ1 as potential cooperative tumor suppressors. DNA copy-number data from primary tumors revealed that copy-number alterations robustly define clusters of low- and high-risk disease beyond that achieved by Gleason score. The genomic and clinical outcome data from these patients are now made available as a public resource.


Science | 2014

Mutational Analysis Reveals the Origin and Therapy-driven Evolution of Recurrent Glioma

Brett E. Johnson; Tali Mazor; Chibo Hong; Michael Barnes; Koki Aihara; Cory Y. McLean; Shaun D. Fouse; Shogo Yamamoto; Hiroki R. Ueda; Kenji Tatsuno; Saurabh Asthana; Llewellyn E. Jalbert; Sarah J. Nelson; Andrew W. Bollen; W. Clay Gustafson; Elise Charron; William A. Weiss; Ivan Smirnov; Jun S. Song; Adam B. Olshen; Soonmee Cha; Yongjun Zhao; Richard A. Moore; Andrew J. Mungall; Steven J.M. Jones; Martin Hirst; Marco A. Marra; Nobuhito Saito; Hiroyuki Aburatani; Akitake Mukasa

Back with a Vengeance After surgery, gliomas (a type of brain tumor) recur in nearly all patients and often in a more aggressive form. Johnson et al. (p. 189, published online 12 December 2013) used exome sequencing to explore whether recurrent tumors harbor different mutations than the primary tumors and whether the mutational profile in the recurrences is influenced by postsurgical treatment of patients with temozolomide (TMZ), a chemotherapeutic drug known to damage DNA. In more than 40% of cases, at least half of the mutations in the initial glioma were undetected at recurrence. The recurrent tumors in many of the TMZ-treated patients bore the signature of TMZ-induced mutagenesis and appeared to follow an evolutionary path to high-grade glioma distinct from that in untreated patients. Primary brain tumors and their recurrences can exhibit vastly different mutational profiles. Tumor recurrence is a leading cause of cancer mortality. Therapies for recurrent disease may fail, at least in part, because the genomic alterations driving the growth of recurrences are distinct from those in the initial tumor. To explore this hypothesis, we sequenced the exomes of 23 initial low-grade gliomas and recurrent tumors resected from the same patients. In 43% of cases, at least half of the mutations in the initial tumor were undetected at recurrence, including driver mutations in TP53, ATRX, SMARCA4, and BRAF; this suggests that recurrent tumors are often seeded by cells derived from the initial tumor at a very early stage of their evolution. Notably, tumors from 6 of 10 patients treated with the chemotherapeutic drug temozolomide (TMZ) followed an alternative evolutionary path to high-grade glioma. At recurrence, these tumors were hypermutated and harbored driver mutations in the RB (retinoblastoma) and Akt-mTOR (mammalian target of rapamycin) pathways that bore the signature of TMZ-induced mutagenesis.


Science | 2012

Genome Sequencing Identifies a Basis for Everolimus Sensitivity

Gopa Iyer; Aphrothiti J. Hanrahan; Matthew I. Milowsky; Hikmat Al-Ahmadie; Sasinya N. Scott; Manickam Janakiraman; Mono Pirun; Chris Sander; Nicholas D. Socci; Irina Ostrovnaya; Agnes Viale; Adriana Heguy; Luke Peng; Timothy A. Chan; Bernard H. Bochner; Dean F. Bajorin; Michael F. Berger; Barry S. Taylor; David B. Solit

Tumor genome sequencing reveals the molecular basis of a patient’s unexpected and dramatic response to a cancer drug. Cancer drugs often induce dramatic responses in a small minority of patients. We used whole-genome sequencing to investigate the genetic basis of a durable remission of metastatic bladder cancer in a patient treated with everolimus, a drug that inhibits the mTOR (mammalian target of rapamycin) signaling pathway. Among the somatic mutations was a loss-of-function mutation in TSC1 (tuberous sclerosis complex 1), a regulator of mTOR pathway activation. Targeted sequencing revealed TSC1 mutations in about 8% of 109 additional bladder cancers examined, and TSC1 mutation correlated with everolimus sensitivity. These results demonstrate the feasibility of using whole-genome sequencing in the clinical setting to identify previously occult biomarkers of drug sensitivity that can aid in the identification of patients most likely to respond to targeted anticancer drugs.


Nature Genetics | 2010

Subtype-specific genomic alterations define new targets for soft tissue sarcoma therapy

Jordi Barretina; Barry S. Taylor; Shantanu Banerji; Alexis Ramos; Mariana Lagos-Quintana; Penelope DeCarolis; Kinjal Shah; Nicholas D. Socci; Barbara A. Weir; Alan Ho; Derek Y. Chiang; Boris Reva; Craig H. Mermel; Gad Getz; Yevgenyi Antipin; Rameen Beroukhim; John Major; Charles Hatton; Richard Nicoletti; Megan Hanna; Ted Sharpe; Timothy Fennell; Kristian Cibulskis; Robert C. Onofrio; Tsuyoshi Saito; Neerav Shukla; Christopher Lau; Sven Nelander; Serena J. Silver; Carrie Sougnez

Soft-tissue sarcomas, which result in approximately 10,700 diagnoses and 3,800 deaths per year in the United States, show remarkable histologic diversity, with more than 50 recognized subtypes. However, knowledge of their genomic alterations is limited. We describe an integrative analysis of DNA sequence, copy number and mRNA expression in 207 samples encompassing seven major subtypes. Frequently mutated genes included TP53 (17% of pleomorphic liposarcomas), NF1 (10.5% of myxofibrosarcomas and 8% of pleomorphic liposarcomas) and PIK3CA (18% of myxoid/round-cell liposarcomas, or MRCs). PIK3CA mutations in MRCs were associated with Akt activation and poor clinical outcomes. In myxofibrosarcomas and pleomorphic liposarcomas, we found both point mutations and genomic deletions affecting the tumor suppressor NF1. Finally, we found that short hairpin RNA (shRNA)-based knockdown of several genes amplified in dedifferentiated liposarcoma, including CDK4 and YEATS4, decreased cell proliferation. Our study yields a detailed map of molecular alterations across diverse sarcoma subtypes and suggests potential subtype-specific targets for therapy.


Nature Genetics | 2009

Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis

Jennifer C King; Jin Xu; John Wongvipat; Haley Hieronymus; Brett S. Carver; David H Leung; Barry S. Taylor; Chris Sander; Robert D. Cardiff; Suzana S. Couto; William L. Gerald; Charles L. Sawyers

The TMPRSS2-ERG fusion, present in approximately 50% of prostate cancers, is less common in prostatic intraepithelial neoplasia (PIN), raising questions about whether TMPRSS2-ERG contributes to disease initiation. We identified the translational start site of a common TMPRSS2-ERG fusion and showed that transgenic TMPRSS2-ERG mice develop PIN, but only in the context of PI3-kinase pathway activation. TMPRSS2-ERG–positive human tumors are also enriched for PTEN loss, suggesting cooperation in prostate tumorigenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2010

The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner

Eric W. Joseph; Christine A. Pratilas; Poulikos I. Poulikakos; Madhavi Tadi; Weiqing Wang; Barry S. Taylor; Ensar Halilovic; Yogindra Persaud; Feng Xing; Agnes Viale; James H. Tsai; Paul B. Chapman; Gideon Bollag; David B. Solit; Neal Rosen

Tumors with mutant BRAF and some with mutant RAS are dependent upon ERK signaling for proliferation, and their growth is suppressed by MAPK/ERK kinase (MEK) inhibitors. In contrast, tumor cells with human EGF receptor (HER) kinase activation proliferate in a MEK-independent manner. These findings have led to the development of RAF and MEK inhibitors as anticancer agents. Like MEK inhibitors, the RAF inhibitor PLX4032 inhibits the proliferation of BRAFV600E tumor cells but not that of HER kinase-dependent tumors. However, tumors with RAS mutation that are sensitive to MEK inhibition are insensitive to PLX4032. MEK inhibitors inhibit ERK phosphorylation in all normal and tumor cells, whereas PLX4032 inhibits ERK signaling only in tumor cells expressing BRAFV600E. In contrast, the drug activates MEK and ERK phosphorylation in cells with wild-type BRAF. In BRAFV600E tumor cells, MEK and RAF inhibitors affect the expression of a common set of genes. PLX4032 inhibits ERK signaling output in mutant BRAF cells, whereas it transiently activates the expression of these genes in tumor cells with wild-type RAF. Thus, PLX4032 inhibits ERK signaling output in a mutant BRAF-selective manner. These data explain why the drug selectively inhibits the growth of mutant BRAF tumors and suggest that it will not cause toxicity resulting from the inhibition of ERK signaling in normal cells. This selectivity may lead to a broader therapeutic index and help explain the greater antitumor activity observed with this drug than with MEK inhibitors.


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

V600EBRAF is associated with disabled feedback inhibition of RAF–MEK signaling and elevated transcriptional output of the pathway

Christine A. Pratilas; Barry S. Taylor; Qing Ye; Agnes Viale; Chris Sander; David B. Solit; Neal Rosen

Tumors with mutant BRAF and those with receptor tyrosine kinase (RTK) activation have similar levels of phosphorylated ERK, but only the former depend on ERK signaling for proliferation. The mitogen-activated protein kinase, extracellular signal-regulated kinase kinase (MEK)/ERK-dependent transcriptional output was defined as the genes whose expression changes significantly 8 h after MEK inhibition. In V600EBRAF cells, this output is comprised of 52 genes, including transcription factors that regulate transformation and members of the dual specificity phosphatase and Sprouty gene families, feedback inhibitors of ERK signaling. No such genes were identified in RTK tumor cells, suggesting that ERK pathway signaling output is selectively activated in BRAF mutant tumors. We find that RAF signaling is feedback down-regulated in RTK cells, but is insensitive to this feedback in BRAF mutant tumors. Physiologic feedback inhibition of RAF/MEK signaling down-regulates ERK output in RTK cells; evasion of this feedback in mutant BRAF cells is associated with increased transcriptional output and MEK/ERK-dependent transformation.


Nature Genetics | 2011

The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma.

Matthew Bott; Marie Brevet; Barry S. Taylor; Shigeki Shimizu; Tatsuo Ito; Lu Wang; Jenette Creaney; Richard A. Lake; Maureen F. Zakowski; Boris Reva; Chris Sander; Robert Delsite; Simon N. Powell; Qin Zhou; Ronglai Shen; Adam B. Olshen; Valerie W. Rusch; Marc Ladanyi

Malignant pleural mesotheliomas (MPMs) often show CDKN2A and NF2 inactivation, but other highly recurrent mutations have not been described. To identify additional driver genes, we used an integrated genomic analysis of 53 MPM tumor samples to guide a focused sequencing effort that uncovered somatic inactivating mutations in BAP1 in 23% of MPMs. The BAP1 nuclear deubiquitinase is known to target histones (together with ASXL1 as a Polycomb repressor subunit) and the HCF1 transcriptional co-factor, and we show that BAP1 knockdown in MPM cell lines affects E2F and Polycomb target genes. These findings implicate transcriptional deregulation in the pathogenesis of MPM.


PLOS ONE | 2010

Automated network analysis identifies core pathways in glioblastoma.

Ethan Cerami; Emek Demir; Nikolaus Schultz; Barry S. Taylor; Chris Sander

Background Glioblastoma multiforme (GBM) is the most common and aggressive type of brain tumor in humans and the first cancer with comprehensive genomic profiles mapped by The Cancer Genome Atlas (TCGA) project. A central challenge in large-scale genome projects, such as the TCGA GBM project, is the ability to distinguish cancer-causing “driver” mutations from passively selected “passenger” mutations. Principal Findings In contrast to a purely frequency based approach to identifying driver mutations in cancer, we propose an automated network-based approach for identifying candidate oncogenic processes and driver genes. The approach is based on the hypothesis that cellular networks contain functional modules, and that tumors target specific modules critical to their growth. Key elements in the approach include combined analysis of sequence mutations and DNA copy number alterations; use of a unified molecular interaction network consisting of both protein-protein interactions and signaling pathways; and identification and statistical assessment of network modules, i.e. cohesive groups of genes of interest with a higher density of interactions within groups than between groups. Conclusions We confirm and extend the observation that GBM alterations tend to occur within specific functional modules, in spite of considerable patient-to-patient variation, and that two of the largest modules involve signaling via p53, Rb, PI3K and receptor protein kinases. We also identify new candidate drivers in GBM, including AGAP2/CENTG1, a putative oncogene and an activator of the PI3K pathway; and, three additional significantly altered modules, including one involved in microtubule organization. To facilitate the application of our network-based approach to additional cancer types, we make the method freely available as part of a software tool called NetBox.


Nature Genetics | 2010

Somatic mutations of the Parkinson's disease–associated gene PARK2 in glioblastoma and other human malignancies

Selvaraju Veeriah; Barry S. Taylor; Shasha Meng; Fang Fang; Emrullah Yilmaz; Igor Vivanco; Manickam Janakiraman; Nikolaus Schultz; Aphrothiti J. Hanrahan; William Pao; Marc Ladanyi; Chris Sander; Adriana Heguy; Eric C. Holland; Philip B. Paty; Paul S. Mischel; Linda M. Liau; Timothy F. Cloughesy; Ingo K. Mellinghoff; David B. Solit; Timothy A. Chan

Mutation of the gene PARK2, which encodes an E3 ubiquitin ligase, is the most common cause of early-onset Parkinsons disease. In a search for multisite tumor suppressors, we identified PARK2 as a frequently targeted gene on chromosome 6q25.2–q27 in cancer. Here we describe inactivating somatic mutations and frequent intragenic deletions of PARK2 in human malignancies. The PARK2 mutations in cancer occur in the same domains, and sometimes at the same residues, as the germline mutations causing familial Parkinsons disease. Cancer-specific mutations abrogate the growth-suppressive effects of the PARK2 protein. PARK2 mutations in cancer decrease PARK2s E3 ligase activity, compromising its ability to ubiquitinate cyclin E and resulting in mitotic instability. These data strongly point to PARK2 as a tumor suppressor on 6q25.2–q27. Thus, PARK2, a gene that causes neuronal dysfunction when mutated in the germline, may instead contribute to oncogenesis when altered in non-neuronal somatic cells.

Collaboration


Dive into the Barry S. Taylor's collaboration.

Top Co-Authors

Avatar

David B. Solit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nikolaus Schultz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael F. Berger

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc Ladanyi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Chang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Agnes Viale

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nicholas D. Socci

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David M. Hyman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Philip Jonsson

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge