David S. DeLuca
Broad Institute
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by David S. DeLuca.
The New England Journal of Medicine | 2011
Lili Wang; Michael S. Lawrence; Youzhong Wan; Petar Stojanov; Carrie Sougnez; Kristen E. Stevenson; Lillian Werner; Andrey Sivachenko; David S. DeLuca; Li Zhang; Wandi Zhang; Alexander R. Vartanov; Stacey M. Fernandes; Natalie R. Goldstein; Eric G. Folco; Kristian Cibulskis; Bethany Tesar; Quinlan L. Sievers; Erica Shefler; Stacey B Gabriel; Nir Hacohen; Robin Reed; Matthew Meyerson; Todd R. Golub; Eric S. Lander; Donna Neuberg; Jennifer R. Brown; Gad Getz; Catherine J. Wu
BACKGROUND The somatic genetic basis of chronic lymphocytic leukemia, a common and clinically heterogeneous leukemia occurring in adults, remains poorly understood. METHODS We obtained DNA samples from leukemia cells in 91 patients with chronic lymphocytic leukemia and performed massively parallel sequencing of 88 whole exomes and whole genomes, together with sequencing of matched germline DNA, to characterize the spectrum of somatic mutations in this disease. RESULTS Nine genes that are mutated at significant frequencies were identified, including four with established roles in chronic lymphocytic leukemia (TP53 in 15% of patients, ATM in 9%, MYD88 in 10%, and NOTCH1 in 4%) and five with unestablished roles (SF3B1, ZMYM3, MAPK1, FBXW7, and DDX3X). SF3B1, which functions at the catalytic core of the spliceosome, was the second most frequently mutated gene (with mutations occurring in 15% of patients). SF3B1 mutations occurred primarily in tumors with deletions in chromosome 11q, which are associated with a poor prognosis in patients with chronic lymphocytic leukemia. We further discovered that tumor samples with mutations in SF3B1 had alterations in pre-messenger RNA (mRNA) splicing. CONCLUSIONS Our study defines the landscape of somatic mutations in chronic lymphocytic leukemia and highlights pre-mRNA splicing as a critical cellular process contributing to chronic lymphocytic leukemia.
Science | 2015
Marta Melé; Pedro G. Ferreira; Ferran Reverter; David S. DeLuca; Jean Monlong; Michael Sammeth; Taylor R. Young; Jakob M. Goldmann; Dmitri D. Pervouchine; Timothy J. Sullivan; Rory Johnson; Ayellet V. Segrè; Sarah Djebali; Anastasia Niarchou; Fred A. Wright; Tuuli Lappalainen; Miquel Calvo; Gad Getz; Emmanouil T. Dermitzakis; Kristin Ardlie; Roderic Guigó
Expression, genetic variation, and tissues Human genomes show extensive genetic variation across individuals, but we have only just started documenting the effects of this variation on the regulation of gene expression. Furthermore, only a few tissues have been examined per genetic variant. In order to examine how genetic expression varies among tissues within individuals, the Genotype-Tissue Expression (GTEx) Consortium collected 1641 postmortem samples covering 54 body sites from 175 individuals. They identified quantitative genetic traits that affect gene expression and determined which of these exhibit tissue-specific expression patterns. Melé et al. measured how transcription varies among tissues, and Rivas et al. looked at how truncated protein variants affect expression across tissues. Science, this issue p. 648, p. 660, p. 666; see also p. 640 RNA expression documents patterns of human transcriptome variation across individuals and tissues. [Also see Perspective by Gibson] Transcriptional regulation and posttranscriptional processing underlie many cellular and organismal phenotypes. We used RNA sequence data generated by Genotype-Tissue Expression (GTEx) project to investigate the patterns of transcriptome variation across individuals and tissues. Tissues exhibit characteristic transcriptional signatures that show stability in postmortem samples. These signatures are dominated by a relatively small number of genes—which is most clearly seen in blood—though few are exclusive to a particular tissue and vary more across tissues than individuals. Genes exhibiting high interindividual expression variation include disease candidates associated with sex, ethnicity, and age. Primary transcription is the major driver of cellular specificity, with splicing playing mostly a complementary role; except for the brain, which exhibits a more divergent splicing program. Variation in splicing, despite its stochasticity, may play in contrast a comparatively greater role in defining individual phenotypes.
Bioinformatics | 2012
David S. DeLuca; Joshua Z. Levin; Andrey Sivachenko; Timothy Fennell; Marc-Danie Nazaire; Chris Williams; Michael Reich; Wendy Winckler; Gad Getz
Summary: RNA-seq, the application of next-generation sequencing to RNA, provides transcriptome-wide characterization of cellular activity. Assessment of sequencing performance and library quality is critical to the interpretation of RNA-seq data, yet few tools exist to address this issue. We introduce RNA-SeQC, a program which provides key measures of data quality. These metrics include yield, alignment and duplication rates; GC bias, rRNA content, regions of alignment (exon, intron and intragenic), continuity of coverage, 3′/5′ bias and count of detectable transcripts, among others. The software provides multi-sample evaluation of library construction protocols, input materials and other experimental parameters. The modularity of the software enables pipeline integration and the routine monitoring of key measures of data quality such as the number of alignable reads, duplication rates and rRNA contamination. RNA-SeQC allows investigators to make informed decisions about sample inclusion in downstream analysis. In summary, RNA-SeQC provides quality control measures critical to experiment design, process optimization and downstream computational analysis. Availability and implementation: See www.genepattern.org to run online, or www.broadinstitute.org/rna-seqc/ for a command line tool. Contact: [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.
Blood | 2014
Mohini Rajasagi; Sachet A. Shukla; Edward F. Fritsch; Derin B. Keskin; David S. DeLuca; Ellese M. Carmona; Wandi Zhang; Carrie Sougnez; Kristian Cibulskis; John Sidney; Kristen E. Stevenson; Jerome Ritz; Donna Neuberg; Vladimir Brusic; Stacey Gabriel; Eric S. Lander; Gad Getz; Nir Hacohen; Catherine J. Wu
Genome sequencing has revealed a large number of shared and personal somatic mutations across human cancers. In principle, any genetic alteration affecting a protein-coding region has the potential to generate mutated peptides that are presented by surface HLA class I proteins that might be recognized by cytotoxic T cells. To test this possibility, we implemented a streamlined approach for the prediction and validation of such neoantigens derived from individual tumors and presented by patient-specific HLA alleles. We applied our computational pipeline to 91 chronic lymphocytic leukemias (CLLs) that underwent whole-exome sequencing (WES). We predicted ∼22 mutated HLA-binding peptides per leukemia (derived from ∼16 missense mutations) and experimentally confirmed HLA binding for ∼55% of such peptides. Two CLL patients that achieved long-term remission following allogeneic hematopoietic stem cell transplantation were monitored for CD8(+) T-cell responses against predicted or confirmed HLA-binding peptides. Long-lived cytotoxic T-cell responses were detected against peptides generated from personal tumor mutations in ALMS1, C6ORF89, and FNDC3B presented on tumor cells. Finally, we applied our computational pipeline to WES data (N = 2488 samples) across 13 different cancer types and estimated dozens to thousands of predicted neoantigens per individual tumor, suggesting that neoantigens are frequent in most tumors.
Proceedings of the National Academy of Sciences of the United States of America | 2014
Jennifer A. Perry; Adam Kiezun; Peter Tonzi; Eliezer M. Van Allen; Scott L. Carter; Sylvan C. Baca; Glenn S. Cowley; Ami S. Bhatt; Esther Rheinbay; Chandra Sekhar Pedamallu; Elena Helman; Amaro Taylor-Weiner; Aaron McKenna; David S. DeLuca; Michael S. Lawrence; Lauren Ambrogio; Carrie Sougnez; Andrey Sivachenko; Loren D. Walensky; Nikhil Wagle; Jaume Mora; Carmen Torres; Cinzia Lavarino; Simone dos Santos Aguiar; José Andrés Yunes; Silvia Regina Brandalise; Gabriela Elisa Mercado-Celis; Jorge Melendez-Zajgla; Rocio Cardenas-Cardos; Liliana Velasco-Hidalgo
Significance We present, to our knowledge, the first comprehensive next-generation sequencing of osteosarcoma in combination with a functional genomic screen in a genetically defined mouse model of osteosarcoma. Our data provide a strong rationale for targeting the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway in osteosarcoma and a foundation for rational clinical trial design. These findings present an immediate clinical opportunity because multiple inhibitors of this pathway are currently in clinical trials. Osteosarcoma is the most common primary bone tumor, yet there have been no substantial advances in treatment or survival in three decades. We examined 59 tumor/normal pairs by whole-exome, whole-genome, and RNA-sequencing. Only the TP53 gene was mutated at significant frequency across all samples. The mean nonsilent somatic mutation rate was 1.2 mutations per megabase, and there was a median of 230 somatic rearrangements per tumor. Complex chains of rearrangements and localized hypermutation were detected in almost all cases. Given the intertumor heterogeneity, the extent of genomic instability, and the difficulty in acquiring a large sample size in a rare tumor, we used several methods to identify genomic events contributing to osteosarcoma survival. Pathway analysis, a heuristic analytic algorithm, a comparative oncology approach, and an shRNA screen converged on the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway as a central vulnerability for therapeutic exploitation in osteosarcoma. Osteosarcoma cell lines are responsive to pharmacologic and genetic inhibition of the PI3K/mTOR pathway both in vitro and in vivo.
Science | 2015
Manuel A. Rivas; Matti Pirinen; Donald F. Conrad; Monkol Lek; Emily K. Tsang; Konrad J. Karczewski; Julian Maller; Kimberly R. Kukurba; David S. DeLuca; Menachem Fromer; Pedro G. Ferreira; Kevin S. Smith; Rui Zhang; Fengmei Zhao; Eric Banks; Ryan Poplin; Douglas M. Ruderfer; Shaun Purcell; Taru Tukiainen; Eric Vallabh Minikel; Peter D. Stenson; David Neil Cooper; Katharine H. Huang; Timothy J. Sullivan; Jared L. Nedzel; Carlos Bustamante; Jin Billy Li; Mark J. Daly; Roderic Guigó; Peter Donnelly
Expression, genetic variation, and tissues Human genomes show extensive genetic variation across individuals, but we have only just started documenting the effects of this variation on the regulation of gene expression. Furthermore, only a few tissues have been examined per genetic variant. In order to examine how genetic expression varies among tissues within individuals, the Genotype-Tissue Expression (GTEx) Consortium collected 1641 postmortem samples covering 54 body sites from 175 individuals. They identified quantitative genetic traits that affect gene expression and determined which of these exhibit tissue-specific expression patterns. Melé et al. measured how transcription varies among tissues, and Rivas et al. looked at how truncated protein variants affect expression across tissues. Science, this issue p. 648, p. 660, p. 666; see also p. 640 Protein-truncated variants impact gene expression levels and splicing across human tissues. [Also see Perspective by Gibson] Accurate prediction of the functional effect of genetic variation is critical for clinical genome interpretation. We systematically characterized the transcriptome effects of protein-truncating variants, a class of variants expected to have profound effects on gene function, using data from the Genotype-Tissue Expression (GTEx) and Geuvadis projects. We quantitated tissue-specific and positional effects on nonsense-mediated transcript decay and present an improved predictive model for this decay. We directly measured the effect of variants both proximal and distal to splice junctions. Furthermore, we found that robustness to heterozygous gene inactivation is not due to dosage compensation. Our results illustrate the value of transcriptome data in the functional interpretation of genetic variants.
PLOS ONE | 2014
Angela N. Brooks; Peter S. Choi; Luc de Waal; Tanaz Sharifnia; Marcin Imielinski; Gordon Saksena; Chandra Sekhar Pedamallu; Andrey Sivachenko; Mara Rosenberg; Juliann Chmielecki; Michael S. Lawrence; David S. DeLuca; Gad Getz; Matthew Meyerson
Although recurrent somatic mutations in the splicing factor U2AF1 (also known as U2AF35) have been identified in multiple cancer types, the effects of these mutations on the cancer transcriptome have yet to be fully elucidated. Here, we identified splicing alterations associated with U2AF1 mutations across distinct cancers using DNA and RNA sequencing data from The Cancer Genome Atlas (TCGA). Using RNA-Seq data from 182 lung adenocarcinomas and 167 acute myeloid leukemias (AML), in which U2AF1 is somatically mutated in 3–4% of cases, we identified 131 and 369 splicing alterations, respectively, that were significantly associated with U2AF1 mutation. Of these, 30 splicing alterations were statistically significant in both lung adenocarcinoma and AML, including three genes in the Cancer Gene Census, CTNNB1, CHCHD7, and PICALM. Cell line experiments expressing U2AF1 S34F in HeLa cells and in 293T cells provide further support that these altered splicing events are caused by U2AF1 mutation. Consistent with the function of U2AF1 in 3′ splice site recognition, we found that S34F/Y mutations cause preferences for CAG over UAG 3′ splice site sequences. This report demonstrates consistent effects of U2AF1 mutation on splicing in distinct cancer cell types.
Journal of Clinical Investigation | 2014
Hideo Watanabe; Qiuping Ma; Shouyong Peng; Guillaume Adelmant; Danielle Swain; Wenyu Song; Cameron Fox; Joshua M. Francis; Chandra Sekhar Pedamallu; David S. DeLuca; Angela N. Brooks; Su Wang; Jianwen Que; Anil K. Rustgi; Kwok-Kin Wong; Keith L. Ligon; X. Shirley Liu; Jarrod A. Marto; Matthew Meyerson; Adam J. Bass
The transcription factor SOX2 is an essential regulator of pluripotent stem cells and promotes development and maintenance of squamous epithelia. We previously reported that SOX2 is an oncogene and subject to highly recurrent genomic amplification in squamous cell carcinomas (SCCs). Here, we have further characterized the function of SOX2 in SCC. Using ChIP-seq analysis, we compared SOX2-regulated gene profiles in multiple SCC cell lines to ES cell profiles and determined that SOX2 binds to distinct genomic loci in SCCs. In SCCs, SOX2 preferentially interacts with the transcription factor p63, as opposed to the transcription factor OCT4, which is the preferred SOX2 binding partner in ES cells. SOX2 and p63 exhibited overlapping genomic occupancy at a large number of loci in SCCs; however, coordinate binding of SOX2 and p63 was absent in ES cells. We further demonstrated that SOX2 and p63 jointly regulate gene expression, including the oncogene ETV4, which was essential for SOX2-amplified SCC cell survival. Together, these findings demonstrate that the action of SOX2 in SCC differs substantially from its role in pluripotency. The identification of the SCC-associated interaction between SOX2 and p63 will enable deeper characterization the downstream targets of this interaction in SCC and normal squamous epithelial physiology.
Journal of Immunological Methods | 2011
Guang Lan Zhang; David S. DeLuca; Derin B. Keskin; Lou Chitkushev; Tanya Zlateva; Ole Lund; Ellis L. Reinherz; Vladimir Brusic
Abstract MULTIPRED2 is a computational system for facile prediction of peptide binding to multiple alleles belonging to human leukocyte antigen (HLA) class I and class II DR molecules. It enables prediction of peptide binding to products of individual HLA alleles, combination of alleles, or HLA supertypes. NetMHCpan and NetMHCIIpan are used as prediction engines. The 13 HLA Class I supertypes are A1, A2, A3, A24, B7, B8, B27, B44, B58, B62, C1, and C4. The 13 HLA Class II DR supertypes are DR1, DR3, DR4, DR6, DR7, DR8, DR9, DR11, DR12, DR13, DR14, DR15, and DR16. In total, MULTIPRED2 enables prediction of peptide binding to 1077 variants representing 26 HLA supertypes. MULTIPRED2 has visualization modules for mapping promiscuous T-cell epitopes as well as those regions of high target concentration – referred to as T-cell epitope hotspots. Novel graphic representations are employed to display the predicted binding peptides and immunological hotspots in an intuitive manner and also to provide a global view of results as heat maps. Another function of MULTIPRED2, which has direct relevance to vaccine design, is the calculation of population coverage. Currently it calculates population coverage in five major groups in North America. MULTIPRED2 is an important tool to complement wet-lab experimental methods for identification of T-cell epitopes. It is available at http://cvc.dfci.harvard.edu/multipred2/.
Cancer Cell | 2016
Lili Wang; Angela N. Brooks; Jean Fan; Youzhong Wan; Rutendo Gambe; Shuqiang Li; Sarah Hergert; Shanye Yin; Samuel S. Freeman; Joshua Z. Levin; Lin Fan; Michael Seiler; Silvia Buonamici; Peter G. Smith; Kevin F. Chau; Carrie Cibulskis; Wandi Zhang; Laura Z. Rassenti; Emanuela M. Ghia; Thomas J. Kipps; Stacey M. Fernandes; Donald B. Bloch; Dylan Kotliar; Dan A. Landau; Sachet A. Shukla; Robin Reed; David S. DeLuca; Jennifer R. Brown; Donna Neuberg; Gad Getz
Mutations in SF3B1, which encodes a spliceosome component, are associated with poor outcome in chronic lymphocytic leukemia (CLL), but how these contribute to CLL progression remains poorly understood. We undertook a transcriptomic characterization of primary human CLL cells to identify transcripts and pathways affected by SF3B1 mutation. Splicing alterations, identified in the analysis of bulk cells, were confirmed in single SF3B1-mutated CLL cells and also found in cell lines ectopically expressing mutant SF3B1. SF3B1 mutation was found to dysregulate multiple cellular functions including DNA damage response, telomere maintenance, and Notch signaling (mediated through KLF8 upregulation, increased TERC and TERT expression, or altered splicing of DVL2 transcript, respectively). SF3B1 mutation leads to diverse changes in CLL-related pathways.