698 results on '"Stuart, Joshua M."'
Search Results
2. The manatee variational autoencoder model for predicting gene expression alterations caused by transcription factor perturbations
- Author
-
Yang, Ying, Seninge, Lucas, Wang, Ziyuan, Oro, Anthony, Stuart, Joshua M., and Ding, Hongxu
- Published
- 2024
- Full Text
- View/download PDF
3. Analysis of germline-driven ancestry-associated gene expression in cancers
- Author
-
Chambwe, Nyasha, Sayaman, Rosalyn W, Hu, Donglei, Huntsman, Scott, Network, The Cancer Genome Analysis, Carrot-Zhang, Jian, Berger, Ashton C, Han, Seunghun, Meyerson, Matthew, Damrauer, Jeffrey S, Hoadley, Katherine A, Felau, Ina, Demchok, John A, Mensah, Michael KA, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Knijnenburg, Theo A, Robertson, A Gordon, Yau, Christina, Benz, Christopher, Huang, Kuan-lin, Newberg, Justin Y, Frampton, Garrett M, Mashl, R Jay, Ding, Li, Romanel, Alessandro, Demichelis, Francesca, Zhou, Wanding, Laird, Peter W, Shen, Hui, Wong, Christopher K, Stuart, Joshua M, Lazar, Alexander J, Le, Xiuning, Oak, Ninad, Kemal, Anab, Caesar-Johnson, Samantha, Zenklusen, Jean C, Ziv, Elad, Beroukhim, Rameen, and Cherniack, Andrew D
- Subjects
Biological Sciences ,Health Sciences ,Genetics ,Human Genome ,Biotechnology ,Cancer ,Good Health and Well Being ,Gene Expression ,Germ Cells ,Humans ,Neoplasms ,Quantitative Trait Loci ,RNA ,Messenger ,Cancer Genome Analysis Network ,Bioinformatics ,Computer sciences ,Genomics ,RNAseq ,Sequence analysis - Abstract
Differential mRNA expression between ancestry groups can be explained by both genetic and environmental factors. We outline a computational workflow to determine the extent to which germline genetic variation explains cancer-specific molecular differences across ancestry groups. Using multi-omics datasets from The Cancer Genome Atlas (TCGA), we enumerate ancestry-informative markers colocalized with cancer-type-specific expression quantitative trait loci (e-QTLs) at ancestry-associated genes. This approach is generalizable to other settings with paired germline genotyping and mRNA expression data for a multi-ethnic cohort. For complete details on the use and execution of this protocol, please refer to Carrot-Zhang et al. (2020), Robertson et al. (2021), and Sayaman et al. (2021).
- Published
- 2022
4. Dual RNA-Seq analysis of SARS-CoV-2 correlates specific human transcriptional response pathways directly to viral expression
- Author
-
Maulding, Nathan D, Seiler, Spencer, Pearson, Alexander, Kreusser, Nicholas, and Stuart, Joshua M
- Subjects
Genetics ,Pneumonia & Influenza ,Emerging Infectious Diseases ,Infectious Diseases ,Pneumonia ,Lung ,Prevention ,Immunization ,Biotechnology ,Biodefense ,Vaccine Related ,Aetiology ,2.1 Biological and endogenous factors ,Infection ,Good Health and Well Being ,A549 Cells ,COVID-19 ,Gene Expression Regulation ,Viral ,Humans ,RNA-Seq ,SARS-CoV-2 ,Transcriptome - Abstract
The SARS-CoV-2 pandemic has challenged humankind's ability to quickly determine the cascade of health effects caused by a novel infection. Even with the unprecedented speed at which vaccines were developed and introduced into society, identifying therapeutic interventions and drug targets for patients infected with the virus remains important as new strains of the virus evolve, or future coronaviruses may emerge that are resistant to current vaccines. The application of transcriptomic RNA sequencing of infected samples may shed new light on the pathways involved in viral mechanisms and host responses. We describe the application of the previously developed "dual RNA-seq" approach to investigate, for the first time, the co-regulation between the human and SARS-CoV-2 transcriptomes. Together with differential expression analysis, we describe the tissue specificity of SARS-CoV-2 expression, an inferred lipopolysaccharide response, and co-regulation of CXCL's, SPRR's, S100's with SARS-CoV-2 expression. Lipopolysaccharide response pathways in particular offer promise for future therapeutic research and the prospect of subgrouping patients based on chemokine expression that may help explain the vastly different reactions patients have to infection. Taken together these findings highlight unappreciated SARS-CoV-2 expression signatures and emphasize new considerations and mechanisms for SARS-CoV-2 therapeutic intervention.
- Published
- 2022
5. Analytical protocol to identify local ancestry-associated molecular features in cancer
- Author
-
Carrot-Zhang, Jian, Han, Seunghun, Zhou, Wanding, Damrauer, Jeffrey S, Kemal, Anab, Network, Cancer Genome Atlas Analysis, Berger, Ashton C, Meyerson, Matthew, Hoadley, Katherine A, Felau, Ina, Caesar-Johnson, Samantha, Demchok, John A, Mensah, Michael KA, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Zenklusen, Jean C, Chambwe, Nyasha, Knijnenburg, Theo A, Robertson, A Gordon, Yau, Christina, Benz, Christopher, Huang, Kuan-lin, Newberg, Justin, Frampton, Garret, Mashl, R Jay, Ding, Li, Romanel, Alessandro, Demichelis, Francesca, Sayaman, Rosalyn W, Ziv, Elad, Laird, Peter W, Shen, Hui, Wong, Christopher K, Stuart, Joshua M, Lazar, Alexander J, Le, Xiuning, Oak, Ninad, Cherniack, Andrew D, and Beroukhim, Rameen
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Health Sciences ,Oncology and Carcinogenesis ,Health Disparities ,Human Genome ,Cancer ,Cancer Genomics ,Minority Health ,4.1 Discovery and preclinical testing of markers and technologies ,2.1 Biological and endogenous factors ,Genetics ,Population ,Genome ,Human ,Genomics ,Genotyping Techniques ,Humans ,Neoplasms ,Phenotype ,Cancer Genome Atlas Analysis Network ,Bioinformatics - Abstract
People of different ancestries vary in cancer risk and outcome, and their molecular differences may indicate sources of these variations. Determining the "local" ancestry composition at each genetic locus across ancestry-admixed populations can suggest causal associations. We present a protocol to identify local ancestry and detect the associated molecular changes, using data from the Cancer Genome Atlas. This workflow can be applied to cancer cohorts with matched tumor and normal data from admixed patients to examine germline contributions to cancer. For complete details on the use and execution of this protocol, please refer to Carrot-Zhang et al. (2020).
- Published
- 2021
6. A community challenge to evaluate RNA-seq, fusion detection, and isoform quantification methods for cancer discovery
- Author
-
Creason, Allison, Haan, David, Dang, Kristen, Chiotti, Kami E, Inkman, Matthew, Lamb, Andrew, Yu, Thomas, Hu, Yin, Norman, Thea C, Buchanan, Alex, van Baren, Marijke J, Spangler, Ryan, Rollins, M Rick, Spellman, Paul T, Rozanov, Dmitri, Zhang, Jin, Maher, Christopher A, Caloian, Cristian, Watson, John D, Uhrig, Sebastian, Haas, Brian J, Jain, Miten, Akeson, Mark, Ahsen, Mehmet Eren, Participants, SMC-RNA Challenge, Zhang, Hongjiu, Wang, Yifan, Guan, Yuanfang, Nguyen, Cu, Sugai, Christopher, Jha, Alokkumar, Li, Jing Woei, Dobin, Alexander, Stolovitzky, Gustavo, Guinney, Justin, Boutros, Paul C, Stuart, Joshua M, and Ellrott, Kyle
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Cancer ,Cancer Genomics ,Generic health relevance ,Humans ,Neoplasms ,Protein Isoforms ,RNA ,RNA-Seq ,Sequence Analysis ,RNA ,SMC-RNA Challenge Participants ,Cloud compute ,DREAM Challenge ,RNA fusion ,RNA-seq ,benchmark ,crowd-sourced ,evaluation ,isoform quantification ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
The accurate identification and quantitation of RNA isoforms present in the cancer transcriptome is key for analyses ranging from the inference of the impacts of somatic variants to pathway analysis to biomarker development and subtype discovery. The ICGC-TCGA DREAM Somatic Mutation Calling in RNA (SMC-RNA) challenge was a crowd-sourced effort to benchmark methods for RNA isoform quantification and fusion detection from bulk cancer RNA sequencing (RNA-seq) data. It concluded in 2018 with a comparison of 77 fusion detection entries and 65 isoform quantification entries on 51 synthetic tumors and 32 cell lines with spiked-in fusion constructs. We report the entries used to build this benchmark, the leaderboard results, and the experimental features associated with the accurate prediction of RNA species. This challenge required submissions to be in the form of containerized workflows, meaning each of the entries described is easily reusable through CWL and Docker containers at https://github.com/SMC-RNA-challenge. A record of this paper's transparent peer review process is included in the supplemental information.
- Published
- 2021
7. Accurate cancer phenotype prediction with AKLIMATE, a stacked kernel learner integrating multimodal genomic data and pathway knowledge.
- Author
-
Uzunangelov, Vladislav, Wong, Christopher K, and Stuart, Joshua M
- Subjects
Bioinformatics ,Mathematical Sciences ,Biological Sciences ,Information and Computing Sciences - Abstract
Advancements in sequencing have led to the proliferation of multi-omic profiles of human cells under different conditions and perturbations. In addition, many databases have amassed information about pathways and gene "signatures"-patterns of gene expression associated with specific cellular and phenotypic contexts. An important current challenge in systems biology is to leverage such knowledge about gene coordination to maximize the predictive power and generalization of models applied to high-throughput datasets. However, few such integrative approaches exist that also provide interpretable results quantifying the importance of individual genes and pathways to model accuracy. We introduce AKLIMATE, a first kernel-based stacked learner that seamlessly incorporates multi-omics feature data with prior information in the form of pathways for either regression or classification tasks. AKLIMATE uses a novel multiple-kernel learning framework where individual kernels capture the prediction propensities recorded in random forests, each built from a specific pathway gene set that integrates all omics data for its member genes. AKLIMATE has comparable or improved performance relative to state-of-the-art methods on diverse phenotype learning tasks, including predicting microsatellite instability in endometrial and colorectal cancer, survival in breast cancer, and cell line response to gene knockdowns. We show how AKLIMATE is able to connect feature data across data platforms through their common pathways to identify examples of several known and novel contributors of cancer and synthetic lethality.
- Published
- 2021
8. Modeling Human TBX5 Haploinsufficiency Predicts Regulatory Networks for Congenital Heart Disease.
- Author
-
Kathiriya, Irfan S, Rao, Kavitha S, Iacono, Giovanni, Devine, W Patrick, Blair, Andrew P, Hota, Swetansu K, Lai, Michael H, Garay, Bayardo I, Thomas, Reuben, Gong, Henry Z, Wasson, Lauren K, Goyal, Piyush, Sukonnik, Tatyana, Hu, Kevin M, Akgun, Gunes A, Bernard, Laure D, Akerberg, Brynn N, Gu, Fei, Li, Kai, Speir, Matthew L, Haeussler, Maximilian, Pu, William T, Stuart, Joshua M, Seidman, Christine E, Seidman, JG, Heyn, Holger, and Bruneau, Benoit G
- Subjects
Heart Ventricles ,Myocytes ,Cardiac ,Animals ,Humans ,Mice ,Heart Defects ,Congenital ,T-Box Domain Proteins ,Cell Differentiation ,Transcription ,Genetic ,Body Patterning ,Gene Dosage ,Mutation ,Models ,Biological ,Gene Regulatory Networks ,Haploinsufficiency ,MEF2 Transcription Factors ,cardiomyocyte differentiation ,congenital heart disease ,disease modeling ,gene dosage ,gene regulation ,gene regulatory networks ,haploinsufficiency ,human induced pluripotent stem cells ,single cell transcriptomics ,transcription factor ,Heart Disease - Coronary Heart Disease ,Cardiovascular ,Genetics ,Human Genome ,Stem Cell Research ,Heart Disease ,Congenital Structural Anomalies ,Pediatric ,1.1 Normal biological development and functioning ,Aetiology ,2.1 Biological and endogenous factors ,Underpinning research ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Haploinsufficiency of transcriptional regulators causes human congenital heart disease (CHD); however, the underlying CHD gene regulatory network (GRN) imbalances are unknown. Here, we define transcriptional consequences of reduced dosage of the CHD transcription factor, TBX5, in individual cells during cardiomyocyte differentiation from human induced pluripotent stem cells (iPSCs). We discovered highly sensitive dysregulation of TBX5-dependent pathways-including lineage decisions and genes associated with heart development, cardiomyocyte function, and CHD genetics-in discrete subpopulations of cardiomyocytes. Spatial transcriptomic mapping revealed chamber-restricted expression for many TBX5-sensitive transcripts. GRN analysis indicated that cardiac network stability, including vulnerable CHD-linked nodes, is sensitive to TBX5 dosage. A GRN-predicted genetic interaction between Tbx5 and Mef2c, manifesting as ventricular septation defects, was validated in mice. These results demonstrate exquisite and diverse sensitivity to TBX5 dosage in heterogeneous subsets of iPSC-derived cardiomyocytes and predicts candidate GRNs for human CHDs, with implications for quantitative transcriptional regulation in disease.
- Published
- 2021
9. Prioritizing transcriptional factors in gene regulatory networks with PageRank
- Author
-
Ding, Hongxu, Yang, Ying, Xue, Yuanqing, Seninge, Lucas, Gong, Henry, Safavi, Rojin, Califano, Andrea, and Stuart, Joshua M
- Subjects
Stem Cell Research - Nonembryonic - Human ,Genetics ,Stem Cell Research ,Biotechnology ,Generic health relevance ,Gene Network ,Molecular Biology ,Omics - Abstract
Biological states are controlled by orchestrated transcriptional factors (TFs) within gene regulatory networks. Here we show TFs responsible for the dynamic changes of biological states can be prioritized with temporal PageRank. We further show such TF prioritization can be extended by integrating gene regulatory networks reverse engineered from multi-omics profiles, e.g. gene expression, chromatin accessibility, and chromosome conformation assays, using multiplex PageRank.
- Published
- 2021
10. Exploring Integrative Analysis Using the BioMedical Evidence Graph
- Author
-
Struck, Adam, Walsh, Brian, Buchanan, Alexander, Lee, Jordan A, Spangler, Ryan, Stuart, Joshua M, and Ellrott, Kyle
- Subjects
Human Genome ,Genetics ,Generic health relevance ,Cancer ,Antineoplastic Agents ,Biomarkers ,Tumor ,Computational Biology ,Computer Graphics ,Databases ,Factual ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Humans ,Medical Informatics ,Neoplasms ,Signal Transduction - Abstract
PurposeThe analysis of cancer biology data involves extremely heterogeneous data sets, including information from RNA sequencing, genome-wide copy number, DNA methylation data reporting on epigenetic regulation, somatic mutations from whole-exome or whole-genome analyses, pathology estimates from imaging sections or subtyping, drug response or other treatment outcomes, and various other clinical and phenotypic measurements. Bringing these different resources into a common framework, with a data model that allows for complex relationships as well as dense vectors of features, will unlock integrated data set analysis.MethodsWe introduce the BioMedical Evidence Graph (BMEG), a graph database and query engine for discovery and analysis of cancer biology. The BMEG is unique from other biologic data graphs in that sample-level molecular and clinical information is connected to reference knowledge bases. It combines gene expression and mutation data with drug-response experiments, pathway information databases, and literature-derived associations.ResultsThe construction of the BMEG has resulted in a graph containing > 41 million vertices and 57 million edges. The BMEG system provides a graph query-based application programming interface to enable analysis, with client code available for Python, Javascript, and R, and a server online at bmeg.io. Using this system, we have demonstrated several forms of cross-data set analysis to show the utility of the system.ConclusionThe BMEG is an evolving resource dedicated to enabling integrative analysis. We have demonstrated queries on the system that illustrate mutation significance analysis, drug-response machine learning, patient-level knowledge-base queries, and pathway level analysis. We have compared the resulting graph to other available integrated graph systems and demonstrated the former is unique in the scale of the graph and the type of data it makes available.
- Published
- 2020
11. Copy Number Loss of 17q22 Is Associated with Enzalutamide Resistance and Poor Prognosis in Metastatic Castration-Resistant Prostate Cancer
- Author
-
Guan, Xiangnan, Sun, Duanchen, Lu, Eric, Urrutia, Joshua A, Reiter, Robert Evan, Rettig, Matthew, Evans, Christopher P, Lara, Primo, Gleave, Martin, Beer, Tomasz M, Thomas, George V, Huang, Jiaoti, Aggarwal, Rahul R, Quigley, David A, Foye, Adam, Chen, William S, Youngren, Jack, Weinstein, Alana S, Stuart, Joshua M, Feng, Felix Y, Small, Eric J, Xia, Zheng, and Alumkal, Joshi J
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Human Genome ,Urologic Diseases ,Cancer ,Biotechnology ,Prostate Cancer ,Genetics ,Benzamides ,Biomarkers ,Tumor ,Biopsy ,Chromosomes ,Human ,Pair 17 ,DNA Copy Number Variations ,Disease-Free Survival ,Drug Resistance ,Neoplasm ,Humans ,Male ,Nitriles ,Phenylthiohydantoin ,Prostate ,Prostatic Neoplasms ,Castration-Resistant ,RNA-Seq ,Survival Analysis ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeThe purpose of this study was to measure genomic changes that emerge with enzalutamide treatment using analyses of whole-genome sequencing and RNA sequencing.Experimental designOne hundred and one tumors from men with metastatic castration-resistant prostate cancer (mCRPC) who had not been treated with enzalutamide (n = 64) or who had enzalutamide-resistant mCRPC (n = 37) underwent whole genome sequencing. Ninety-nine of these tumors also underwent RNA sequencing. We analyzed the genomes and transcriptomes of these mCRPC tumors.ResultsCopy number loss was more common than gain in enzalutamide-resistant tumors. Specially, we identified 124 protein-coding genes that were more commonly lost in enzalutamide-resistant samples. These 124 genes included eight putative tumor suppressors located at nine distinct genomic regions. We demonstrated that focal deletion of the 17q22 locus that includes RNF43 and SRSF1 was not present in any patient with enzalutamide-naïve mCRPC but was present in 16% (6/37) of patients with enzalutamide-resistant mCRPC. 17q22 loss was associated with lower RNF43 and SRSF1 expression and poor overall survival from time of biopsy [median overall survival of 19.3 months in 17q22 intact vs. 8.9 months in 17q22 loss, HR, 3.44 95% confidence interval (CI), 1.338-8.867, log-rank P = 0.006]. Finally, 17q22 loss was linked with activation of several targetable factors, including CDK1/2, Akt, and PLK1, demonstrating the potential therapeutic relevance of 17q22 loss in mCRPC.ConclusionsCopy number loss is common in enzalutamide-resistant tumors. Focal deletion of chromosome 17q22 defines a previously unappreciated molecular subset of enzalutamide-resistant mCRPC associated with poor clinical outcome.
- Published
- 2020
12. Transcriptional profiling identifies an androgen receptor activity-low, stemness program associated with enzalutamide resistance
- Author
-
Alumkal, Joshi J, Sun, Duanchen, Lu, Eric, Beer, Tomasz M, Thomas, George V, Latour, Emile, Aggarwal, Rahul, Cetnar, Jeremy, Ryan, Charles J, Tabatabaei, Shaadi, Bailey, Shawna, Turina, Claire B, Quigley, David A, Guan, Xiangnan, Foye, Adam, Youngren, Jack F, Urrutia, Joshua, Huang, Jiaoti, Weinstein, Alana S, Friedl, Verena, Rettig, Matthew, Reiter, Robert E, Spratt, Daniel E, Gleave, Martin, Evans, Christopher P, Stuart, Joshua M, Chen, Yiyi, Feng, Felix Y, Small, Eric J, Witte, Owen N, and Xia, Zheng
- Subjects
Urologic Diseases ,Genetics ,Prostate Cancer ,Aging ,Cancer ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Aged ,Aged ,80 and over ,Antineoplastic Agents ,Benzamides ,Drug Resistance ,Neoplasm ,Gene Expression Profiling ,Humans ,Male ,Middle Aged ,Nitriles ,Phenylthiohydantoin ,Prostate-Specific Antigen ,Prostatic Neoplasms ,Castration-Resistant ,Receptors ,Androgen ,enzalutamide ,resistance ,androgen receptor ,stemness - Abstract
The androgen receptor (AR) antagonist enzalutamide is one of the principal treatments for men with castration-resistant prostate cancer (CRPC). However, not all patients respond, and resistance mechanisms are largely unknown. We hypothesized that genomic and transcriptional features from metastatic CRPC biopsies prior to treatment would be predictive of de novo treatment resistance. To this end, we conducted a phase II trial of enzalutamide treatment (160 mg/d) in 36 men with metastatic CRPC. Thirty-four patients were evaluable for the primary end point of a prostate-specific antigen (PSA)50 response (PSA decline ≥50% at 12 wk vs. baseline). Nine patients were classified as nonresponders (PSA decline
- Published
- 2020
13. Comprehensive Analysis of Genetic Ancestry and Its Molecular Correlates in Cancer
- Author
-
Carrot-Zhang, Jian, Chambwe, Nyasha, Damrauer, Jeffrey S, Knijnenburg, Theo A, Robertson, A Gordon, Yau, Christina, Zhou, Wanding, Berger, Ashton C, Huang, Kuan-lin, Newberg, Justin Y, Mashl, R Jay, Romanel, Alessandro, Sayaman, Rosalyn W, Demichelis, Francesca, Felau, Ina, Frampton, Garrett M, Han, Seunghun, Hoadley, Katherine A, Kemal, Anab, Laird, Peter W, Lazar, Alexander J, Le, Xiuning, Oak, Ninad, Shen, Hui, Wong, Christopher K, Zenklusen, Jean C, Ziv, Elad, Network, Cancer Genome Atlas Analysis, Aguet, Francois, Ding, Li, Demchok, John A, Mensah, Michael KA, Caesar-Johnson, Samantha, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Alfoldi, Jessica, Karczewski, Konrad J, MacArthur, Daniel G, Meyerson, Matthew, Benz, Christopher, Stuart, Joshua M, Cherniack, Andrew D, and Beroukhim, Rameen
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Genetics ,Human Genome ,Clinical Research ,Cancer ,Biotechnology ,DNA Methylation ,DNA-Binding Proteins ,Ethnicity ,F-Box-WD Repeat-Containing Protein 7 ,Gene Expression Regulation ,Neoplastic ,Genetic Predisposition to Disease ,Genetics ,Population ,Genome ,Human ,Genomics ,High-Throughput Nucleotide Sequencing ,Humans ,MicroRNAs ,Mutation ,Neoplasm Proteins ,Neoplasms ,Transcription Factors ,Von Hippel-Lindau Tumor Suppressor Protein ,Cancer Genome Atlas Analysis Network ,TCGA ,admixture ,ancestry ,cancer ,eQTL ,genomics ,mRNA ,methylation ,miRNA ,mutation ,Neurosciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
We evaluated ancestry effects on mutation rates, DNA methylation, and mRNA and miRNA expression among 10,678 patients across 33 cancer types from The Cancer Genome Atlas. We demonstrated that cancer subtypes and ancestry-related technical artifacts are important confounders that have been insufficiently accounted for. Once accounted for, ancestry-associated differences spanned all molecular features and hundreds of genes. Biologically significant differences were usually tissue specific but not specific to cancer. However, admixture and pathway analyses suggested some of these differences are causally related to cancer. Specific findings included increased FBXW7 mutations in patients of African origin, decreased VHL and PBRM1 mutations in renal cancer patients of African origin, and decreased immune activity in bladder cancer patients of East Asian origin.
- Published
- 2020
14. Pan-cancer analysis of whole genomes
- Author
-
Campbell, Peter J, Getz, Gad, Korbel, Jan O, Stuart, Joshua M, Jennings, Jennifer L, Stein, Lincoln D, Perry, Marc D, Nahal-Bose, Hardeep K, Ouellette, BF Francis, Li, Constance H, Rheinbay, Esther, Nielsen, G Petur, Sgroi, Dennis C, Wu, Chin-Lee, Faquin, William C, Deshpande, Vikram, Boutros, Paul C, Lazar, Alexander J, Hoadley, Katherine A, Louis, David N, Dursi, L Jonathan, Yung, Christina K, Bailey, Matthew H, Saksena, Gordon, Raine, Keiran M, Buchhalter, Ivo, Kleinheinz, Kortine, Schlesner, Matthias, Zhang, Junjun, Wang, Wenyi, Wheeler, David A, Ding, Li, Simpson, Jared T, O'Connor, Brian D, Yakneen, Sergei, Ellrott, Kyle, Miyoshi, Naoki, Butler, Adam P, Royo, Romina, Shorser, Solomon I, Vazquez, Miguel, Rausch, Tobias, Tiao, Grace, Waszak, Sebastian M, Rodriguez-Martin, Bernardo, Shringarpure, Suyash, Wu, Dai-Ying, Demidov, German M, Delaneau, Olivier, Hayashi, Shuto, Imoto, Seiya, Habermann, Nina, Segre, Ayellet V, Garrison, Erik, Cafferkey, Andy, Alvarez, Eva G, Maria Heredia-Genestar, Jose, Muyas, Francesc, Drechsel, Oliver, Bruzos, Alicia L, Temes, Javier, Zamora, Jorge, Baez-Ortega, Adrian, Kim, Hyung-Lae, Mashl, R Jay, Ye, Kai, DiBiase, Anthony, Huang, Kuan-lin, Letunic, Ivica, McLellan, Michael D, Newhouse, Steven J, Shmaya, Tal, Kumar, Sushant, Wedge, David C, Wright, Mark H, Yellapantula, Venkata D, Gerstein, Mark, Khurana, Ekta, Marques-Bonet, Tomas, Navarro, Arcadi, Bustamante, Carlos D, Siebert, Reiner, Nakagawa, Hidewaki, Easton, Douglas F, Ossowski, Stephan, Tubio, Jose MC, De La Vega, Francisco M, Estivill, Xavier, Yuen, Denis, Mihaiescu, George L, Omberg, Larsson, Ferretti, Vincent, Sabarinathan, Radhakrishnan, Pich, Oriol, Gonzalez-Perez, Abel, Weiner, Amaro Taylor, Fittall, Matthew W, Demeulemeester, Jonas, Tarabichi, Maxime, and Roberts, Nicola D
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Bioinformatics and Computational Biology ,Genetics ,Oncology and Carcinogenesis ,Cancer Genomics ,Biotechnology ,Human Genome ,Cancer ,Prevention ,2.1 Biological and endogenous factors ,Cell Proliferation ,Cellular Senescence ,Chromothripsis ,Cloud Computing ,DNA Mutational Analysis ,Evolution ,Molecular ,Female ,Genome ,Human ,Genomics ,Germ-Line Mutation ,High-Throughput Nucleotide Sequencing ,Humans ,Information Dissemination ,Male ,Mutagenesis ,Mutation ,Neoplasms ,Oncogenes ,Promoter Regions ,Genetic ,RNA Splicing ,Reproducibility of Results ,Telomerase ,Telomere ,ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium ,General Science & Technology - Abstract
Cancer is driven by genetic change, and the advent of massively parallel sequencing has enabled systematic documentation of this variation at the whole-genome scale1-3. Here we report the integrative analysis of 2,658 whole-cancer genomes and their matching normal tissues across 38 tumour types from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). We describe the generation of the PCAWG resource, facilitated by international data sharing using compute clouds. On average, cancer genomes contained 4-5 driver mutations when combining coding and non-coding genomic elements; however, in around 5% of cases no drivers were identified, suggesting that cancer driver discovery is not yet complete. Chromothripsis, in which many clustered structural variants arise in a single catastrophic event, is frequently an early event in tumour evolution; in acral melanoma, for example, these events precede most somatic point mutations and affect several cancer-associated genes simultaneously. Cancers with abnormal telomere maintenance often originate from tissues with low replicative activity and show several mechanisms of preventing telomere attrition to critical levels. Common and rare germline variants affect patterns of somatic mutation, including point mutations, structural variants and somatic retrotransposition. A collection of papers from the PCAWG Consortium describes non-coding mutations that drive cancer beyond those in the TERT promoter4; identifies new signatures of mutational processes that cause base substitutions, small insertions and deletions and structural variation5,6; analyses timings and patterns of tumour evolution7; describes the diverse transcriptional consequences of somatic mutation on splicing, expression levels, fusion genes and promoter activity8,9; and evaluates a range of more-specialized features of cancer genomes8,10-18.
- Published
- 2020
15. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes.
- Author
-
Rheinbay, Esther, Nielsen, Morten Muhlig, Abascal, Federico, Wala, Jeremiah A, Shapira, Ofer, Tiao, Grace, Hornshøj, Henrik, Hess, Julian M, Juul, Randi Istrup, Lin, Ziao, Feuerbach, Lars, Sabarinathan, Radhakrishnan, Madsen, Tobias, Kim, Jaegil, Mularoni, Loris, Shuai, Shimin, Lanzós, Andrés, Herrmann, Carl, Maruvka, Yosef E, Shen, Ciyue, Amin, Samirkumar B, Bandopadhayay, Pratiti, Bertl, Johanna, Boroevich, Keith A, Busanovich, John, Carlevaro-Fita, Joana, Chakravarty, Dimple, Chan, Calvin Wing Yiu, Craft, David, Dhingra, Priyanka, Diamanti, Klev, Fonseca, Nuno A, Gonzalez-Perez, Abel, Guo, Qianyun, Hamilton, Mark P, Haradhvala, Nicholas J, Hong, Chen, Isaev, Keren, Johnson, Todd A, Juul, Malene, Kahles, Andre, Kahraman, Abdullah, Kim, Youngwook, Komorowski, Jan, Kumar, Kiran, Kumar, Sushant, Lee, Donghoon, Lehmann, Kjong-Van, Li, Yilong, Liu, Eric Minwei, Lochovsky, Lucas, Park, Keunchil, Pich, Oriol, Roberts, Nicola D, Saksena, Gordon, Schumacher, Steven E, Sidiropoulos, Nikos, Sieverling, Lina, Sinnott-Armstrong, Nasa, Stewart, Chip, Tamborero, David, Tubio, Jose MC, Umer, Husen M, Uusküla-Reimand, Liis, Wadelius, Claes, Wadi, Lina, Yao, Xiaotong, Zhang, Cheng-Zhong, Zhang, Jing, Haber, James E, Hobolth, Asger, Imielinski, Marcin, Kellis, Manolis, Lawrence, Michael S, von Mering, Christian, Nakagawa, Hidewaki, Raphael, Benjamin J, Rubin, Mark A, Sander, Chris, Stein, Lincoln D, Stuart, Joshua M, Tsunoda, Tatsuhiko, Wheeler, David A, Johnson, Rory, Reimand, Jüri, Gerstein, Mark, Khurana, Ekta, Campbell, Peter J, López-Bigas, Núria, PCAWG Drivers and Functional Interpretation Working Group, PCAWG Structural Variation Working Group, Weischenfeldt, Joachim, Beroukhim, Rameen, Martincorena, Iñigo, Pedersen, Jakob Skou, Getz, Gad, and PCAWG Consortium
- Subjects
PCAWG Drivers and Functional Interpretation Working Group ,PCAWG Structural Variation Working Group ,PCAWG Consortium ,Humans ,Neoplasms ,Gene Expression Regulation ,Neoplastic ,Mutation ,Genome ,Human ,Databases ,Genetic ,DNA Breaks ,INDEL Mutation ,Genome-Wide Association Study ,Gene Expression Regulation ,Neoplastic ,Genome ,Human ,Databases ,Genetic ,General Science & Technology - Abstract
The discovery of drivers of cancer has traditionally focused on protein-coding genes1-4. Here we present analyses of driver point mutations and structural variants in non-coding regions across 2,658 genomes from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium5 of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). For point mutations, we developed a statistically rigorous strategy for combining significance levels from multiple methods of driver discovery that overcomes the limitations of individual methods. For structural variants, we present two methods of driver discovery, and identify regions that are significantly affected by recurrent breakpoints and recurrent somatic juxtapositions. Our analyses confirm previously reported drivers6,7, raise doubts about others and identify novel candidates, including point mutations in the 5' region of TP53, in the 3' untranslated regions of NFKBIZ and TOB1, focal deletions in BRD4 and rearrangements in the loci of AKR1C genes. We show that although point mutations and structural variants that drive cancer are less frequent in non-coding genes and regulatory sequences than in protein-coding genes, additional examples of these drivers will be found as more cancer genomes become available.
- Published
- 2020
16. Pathway and network analysis of more than 2500 whole cancer genomes.
- Author
-
Reyna, Matthew A, Haan, David, Paczkowska, Marta, Verbeke, Lieven PC, Vazquez, Miguel, Kahraman, Abdullah, Pulido-Tamayo, Sergio, Barenboim, Jonathan, Wadi, Lina, Dhingra, Priyanka, Shrestha, Raunak, Getz, Gad, Lawrence, Michael S, Pedersen, Jakob Skou, Rubin, Mark A, Wheeler, David A, Brunak, Søren, Izarzugaza, Jose MG, Khurana, Ekta, Marchal, Kathleen, von Mering, Christian, Sahinalp, S Cenk, Valencia, Alfonso, PCAWG Drivers and Functional Interpretation Working Group, Reimand, Jüri, Stuart, Joshua M, Raphael, Benjamin J, and PCAWG Consortium
- Subjects
PCAWG Drivers and Functional Interpretation Working Group ,PCAWG Consortium ,Humans ,Neoplasms ,Computational Biology ,Chromatin Assembly and Disassembly ,Gene Expression Regulation ,Neoplastic ,RNA Splicing ,Mutation ,Genome ,Human ,Databases ,Genetic ,Metabolic Networks and Pathways ,Promoter Regions ,Genetic - Abstract
The catalog of cancer driver mutations in protein-coding genes has greatly expanded in the past decade. However, non-coding cancer driver mutations are less well-characterized and only a handful of recurrent non-coding mutations, most notably TERT promoter mutations, have been reported. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancer across 38 tumor types, we perform multi-faceted pathway and network analyses of non-coding mutations across 2583 whole cancer genomes from 27 tumor types compiled by the ICGC/TCGA PCAWG project that was motivated by the success of pathway and network analyses in prioritizing rare mutations in protein-coding genes. While few non-coding genomic elements are recurrently mutated in this cohort, we identify 93 genes harboring non-coding mutations that cluster into several modules of interacting proteins. Among these are promoter mutations associated with reduced mRNA expression in TP53, TLE4, and TCF4. We find that biological processes had variable proportions of coding and non-coding mutations, with chromatin remodeling and proliferation pathways altered primarily by coding mutations, while developmental pathways, including Wnt and Notch, altered by both coding and non-coding mutations. RNA splicing is primarily altered by non-coding mutations in this cohort, and samples containing non-coding mutations in well-known RNA splicing factors exhibit similar gene expression signatures as samples with coding mutations in these genes. These analyses contribute a new repertoire of possible cancer genes and mechanisms that are altered by non-coding mutations and offer insights into additional cancer vulnerabilities that can be investigated for potential therapeutic treatments.
- Published
- 2020
17. A community effort to create standards for evaluating tumor subclonal reconstruction.
- Author
-
Salcedo, Adriana, Tarabichi, Maxime, Espiritu, Shadrielle Melijah G, Deshwar, Amit G, David, Matei, Wilson, Nathan M, Dentro, Stefan, Wintersinger, Jeff A, Liu, Lydia Y, Ko, Minjeong, Sivanandan, Srinivasan, Zhang, Hongjiu, Zhu, Kaiyi, Ou Yang, Tai-Hsien, Chilton, John M, Buchanan, Alex, Lalansingh, Christopher M, P'ng, Christine, Anghel, Catalina V, Umar, Imaad, Lo, Bryan, Zou, William, DREAM SMC-Het Participants, Simpson, Jared T, Stuart, Joshua M, Anastassiou, Dimitris, Guan, Yuanfang, Ewing, Adam D, Ellrott, Kyle, Wedge, David C, Morris, Quaid, Van Loo, Peter, and Boutros, Paul C
- Subjects
DREAM SMC-Het Participants ,Clone Cells ,Humans ,Neoplasms ,Gene Dosage ,Mutation ,Polymorphism ,Single Nucleotide ,Genome ,Algorithms ,Reference Standards ,Computer Simulation ,DNA Copy Number Variations ,Human Genome ,Cancer ,Genetics - Abstract
Tumor DNA sequencing data can be interpreted by computational methods that analyze genomic heterogeneity to infer evolutionary dynamics. A growing number of studies have used these approaches to link cancer evolution with clinical progression and response to therapy. Although the inference of tumor phylogenies is rapidly becoming standard practice in cancer genome analyses, standards for evaluating them are lacking. To address this need, we systematically assess methods for reconstructing tumor subclonality. First, we elucidate the main algorithmic problems in subclonal reconstruction and develop quantitative metrics for evaluating them. Then we simulate realistic tumor genomes that harbor all known clonal and subclonal mutation types and processes. Finally, we benchmark 580 tumor reconstructions, varying tumor read depth, tumor type and somatic variant detection. Our analysis provides a baseline for the establishment of gold-standard methods to analyze tumor heterogeneity.
- Published
- 2020
18. Using Transcriptional Signatures to Find Cancer Drivers with LURE.
- Author
-
Haan, David, Tao, Ruikang, Friedl, Verena, Anastopoulos, Ioannis N, Wong, Christopher K, Weinstein, Alana S, and Stuart, Joshua M
- Subjects
Humans ,Neoplasms ,Computational Biology ,Mutation ,Machine Learning ,Cancer ,Genomics ,Drivers ,Gene Expression ,Human Genome ,Genetics - Abstract
Cancer genome projects have produced multidimensional datasets on thousands of samples. Yet, depending on the tumor type, 5-50% of samples have no known driving event. We introduce a semi-supervised method called Learning UnRealized Events (LURE) that uses a progressive label learning framework and minimum spanning analysis to predict cancer drivers based on their altered samples sharing a gene expression signature with the samples of a known event. We demonstrate the utility of the method on the TCGA Pan-Cancer Atlas dataset for which it produced a high-confidence result relating 59 new connections to 18 known mutation events including alterations in the same gene, family, and pathway. We give examples of predicted drivers involved in TP53, telomere maintenance, and MAPK/RTK signaling pathways. LURE identifies connections between genes with no known prior relationship, some of which may offer clues for targeting specific forms of cancer. Code and Supplemental Material are available on the LURE website: https://sysbiowiki.soe.ucsc.edu/lure.
- Published
- 2020
19. Reproducible biomedical benchmarking in the cloud: lessons from crowd-sourced data challenges
- Author
-
Ellrott, Kyle, Buchanan, Alex, Creason, Allison, Mason, Michael, Schaffter, Thomas, Hoff, Bruce, Eddy, James, Chilton, John M, Yu, Thomas, Stuart, Joshua M, Saez-Rodriguez, Julio, Stolovitzky, Gustavo, Boutros, Paul C, and Guinney, Justin
- Subjects
Information and Computing Sciences ,Software Engineering ,Networking and Information Technology R&D (NITRD) ,Bioengineering ,Generic health relevance ,Algorithms ,Benchmarking ,Information Dissemination ,Models ,Biological ,Reproducibility of Results ,Environmental Sciences ,Biological Sciences ,Bioinformatics - Abstract
Challenges are achieving broad acceptance for addressing many biomedical questions and enabling tool assessment. But ensuring that the methods evaluated are reproducible and reusable is complicated by the diversity of software architectures, input and output file formats, and computing environments. To mitigate these problems, some challenges have leveraged new virtualization and compute methods, requiring participants to submit cloud-ready software packages. We review recent data challenges with innovative approaches to model reproducibility and data sharing, and outline key lessons for improving quantitative biomedical data analysis through crowd-sourced benchmarking challenges.
- Published
- 2019
20. Biological process activity transformation of single cell gene expression for cross-species alignment.
- Author
-
Ding, Hongxu, Blair, Andrew, Yang, Ying, and Stuart, Joshua M
- Subjects
Leukocytes ,Mononuclear ,Animals ,Zebrafish ,Humans ,Mice ,Gene Expression Profiling ,Sequence Analysis ,RNA ,Computational Biology ,Systems Biology ,Gene Expression ,Gene Expression Regulation ,Developmental ,Single-Cell Analysis ,Signal-To-Noise Ratio ,Mouse Embryonic Stem Cells ,Gene Expression Regulation ,Developmental ,Leukocytes ,Mononuclear ,Sequence Analysis ,RNA - Abstract
The maintenance and transition of cellular states are controlled by biological processes. Here we present a gene set-based transformation of single cell RNA-Seq data into biological process activities that provides a robust description of cellular states. Moreover, as these activities represent species-independent descriptors, they facilitate the alignment of single cell states across different organisms.
- Published
- 2019
21. PLATYPUS: A Multiple-View Learning Predictive Framework for Cancer Drug Sensitivity Prediction.
- Author
-
Graim, Kiley, Friedl, Verena, Houlahan, Kathleen E, and Stuart, Joshua M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Cancer ,8.4 Research design and methodologies (health services) ,Health and social care services research ,Good Health and Well Being ,Antineoplastic Agents ,Cell Line ,Tumor ,Computational Biology ,Databases ,Factual ,Drug Resistance ,Neoplasm ,Humans ,Information Storage and Retrieval ,Machine Learning ,Neoplasms ,Patient-Specific Modeling ,Pharmacogenomic Variants ,Precision Medicine ,Software ,Supervised Machine Learning ,Pattern Recognition ,Multiple View Learning ,Drug Sensitivity ,Incompleteness ,Unlabeled Data ,Semi-Supervised ,Co-Training ,Integrative Genomics ,Systems Biology ,Multidimensional ,Multi-Omic - Abstract
Cancer is a complex collection of diseases that are to some degree unique to each patient. Precision oncology aims to identify the best drug treatment regime using molecular data on tumor samples. While omics-level data is becoming more widely available for tumor specimens, the datasets upon which computational learning methods can be trained vary in coverage from sample to sample and from data type to data type. Methods that can 'connect the dots' to leverage more of the information provided by these studies could offer major advantages for maximizing predictive potential. We introduce a multi-view machinelearning strategy called PLATYPUS that builds 'views' from multiple data sources that are all used as features for predicting patient outcomes. We show that a learning strategy that finds agreement across the views on unlabeled data increases the performance of the learning methods over any single view. We illustrate the power of the approach by deriving signatures for drug sensitivity in a large cancer cell line database. Code and additional information are available from the PLATYPUS website https://sysbiowiki.soe.ucsc.edu/platypus.
- Published
- 2019
22. Germline contamination and leakage in whole genome somatic single nucleotide variant detection
- Author
-
Sendorek, Dorota H, Caloian, Cristian, Ellrott, Kyle, Bare, J Christopher, Yamaguchi, Takafumi N, Ewing, Adam D, Houlahan, Kathleen E, Norman, Thea C, Margolin, Adam A, Stuart, Joshua M, and Boutros, Paul C
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Cancer ,Generic health relevance ,Good Health and Well Being ,Algorithms ,Genome ,Human ,Germ Cells ,Humans ,Internet ,Neoplasms ,Polymorphism ,Single Nucleotide ,User-Computer Interface ,Whole Genome Sequencing ,Cancer genomics ,Next-generation sequencing ,Mutation calling ,Germline contamination ,Germline leakage ,Patient identifiability ,Single nucleotide variant ,SNV ,Mathematical Sciences ,Information and Computing Sciences ,Bioinformatics ,Biological sciences ,Information and computing sciences ,Mathematical sciences - Abstract
BackgroundThe clinical sequencing of cancer genomes to personalize therapy is becoming routine across the world. However, concerns over patient re-identification from these data lead to questions about how tightly access should be controlled. It is not thought to be possible to re-identify patients from somatic variant data. However, somatic variant detection pipelines can mistakenly identify germline variants as somatic ones, a process called "germline leakage". The rate of germline leakage across different somatic variant detection pipelines is not well-understood, and it is uncertain whether or not somatic variant calls should be considered re-identifiable. To fill this gap, we quantified germline leakage across 259 sets of whole-genome somatic single nucleotide variant (SNVs) predictions made by 21 teams as part of the ICGC-TCGA DREAM Somatic Mutation Calling Challenge.ResultsThe median somatic SNV prediction set contained 4325 somatic SNVs and leaked one germline polymorphism. The level of germline leakage was inversely correlated with somatic SNV prediction accuracy and positively correlated with the amount of infiltrating normal cells. The specific germline variants leaked differed by tumour and algorithm. To aid in quantitation and correction of leakage, we created a tool, called GermlineFilter, for use in public-facing somatic SNV databases.ConclusionsThe potential for patient re-identification from leaked germline variants in somatic SNV predictions has led to divergent open data access policies, based on different assessments of the risks. Indeed, a single, well-publicized re-identification event could reshape public perceptions of the values of genomic data sharing. We find that modern somatic SNV prediction pipelines have low germline-leakage rates, which can be further reduced, especially for cloud-sharing, using pre-filtering software.
- Published
- 2018
23. Valection: design optimization for validation and verification studies
- Author
-
Cooper, Christopher I, Yao, Delia, Sendorek, Dorota H, Yamaguchi, Takafumi N, P’ng, Christine, Houlahan, Kathleen E, Caloian, Cristian, Fraser, Michael, SMC-DNA Challenge Participants, Ellrott, Kyle, Margolin, Adam A, Bristow, Robert G, Stuart, Joshua M, and Boutros, Paul C
- Subjects
Sequence Analysis ,DNA ,Software Validation ,Verification ,Validation ,Candidate-selection ,DNA sequencing ,SMC-DNA Challenge Participants ,Mathematical Sciences ,Biological Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
BackgroundPlatform-specific error profiles necessitate confirmatory studies where predictions made on data generated using one technology are additionally verified by processing the same samples on an orthogonal technology. However, verifying all predictions can be costly and redundant, and testing a subset of findings is often used to estimate the true error profile.ResultsTo determine how to create subsets of predictions for validation that maximize accuracy of global error profile inference, we developed Valection, a software program that implements multiple strategies for the selection of verification candidates. We evaluated these selection strategies on one simulated and two experimental datasets.ConclusionsValection is implemented in multiple programming languages, available at: http://labs.oicr.on.ca/boutros-lab/software/valection.
- Published
- 2018
24. Combining accurate tumor genome simulation with crowdsourcing to benchmark somatic structural variant detection
- Author
-
Lee, Anna Y, Ewing, Adam D, Ellrott, Kyle, Hu, Yin, Houlahan, Kathleen E, Bare, J Christopher, Espiritu, Shadrielle Melijah G, Huang, Vincent, Dang, Kristen, Chong, Zechen, Caloian, Cristian, Yamaguchi, Takafumi N, Kellen, Michael R, Chen, Ken, Norman, Thea C, Friend, Stephen H, Guinney, Justin, Stolovitzky, Gustavo, Haussler, David, Margolin, Adam A, Stuart, Joshua M, and Boutros, Paul C
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Bioinformatics and Computational Biology ,Genetics ,Oncology and Carcinogenesis ,Cancer ,Human Genome ,Generic health relevance ,Algorithms ,Benchmarking ,Computer Simulation ,Crowdsourcing ,Databases ,Genetic ,Genetic Variation ,Genome ,Human ,Genomics ,High-Throughput Nucleotide Sequencing ,Humans ,Neoplasms ,Software ,Somatic mutations ,Simulation ,Structural variants ,Cancer genomics ,Whole-genome sequencing ,ICGC-TCGA DREAM Somatic Mutation Calling Challenge Participants ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
BackgroundThe phenotypes of cancer cells are driven in part by somatic structural variants. Structural variants can initiate tumors, enhance their aggressiveness, and provide unique therapeutic opportunities. Whole-genome sequencing of tumors can allow exhaustive identification of the specific structural variants present in an individual cancer, facilitating both clinical diagnostics and the discovery of novel mutagenic mechanisms. A plethora of somatic structural variant detection algorithms have been created to enable these discoveries; however, there are no systematic benchmarks of them. Rigorous performance evaluation of somatic structural variant detection methods has been challenged by the lack of gold standards, extensive resource requirements, and difficulties arising from the need to share personal genomic information.ResultsTo facilitate structural variant detection algorithm evaluations, we create a robust simulation framework for somatic structural variants by extending the BAMSurgeon algorithm. We then organize and enable a crowdsourced benchmarking within the ICGC-TCGA DREAM Somatic Mutation Calling Challenge (SMC-DNA). We report here the results of structural variant benchmarking on three different tumors, comprising 204 submissions from 15 teams. In addition to ranking methods, we identify characteristic error profiles of individual algorithms and general trends across them. Surprisingly, we find that ensembles of analysis pipelines do not always outperform the best individual method, indicating a need for new ways to aggregate somatic structural variant detection approaches.ConclusionsThe synthetic tumors and somatic structural variant detection leaderboards remain available as a community benchmarking resource, and BAMSurgeon is available at https://github.com/adamewing/bamsurgeon .
- Published
- 2018
25. Author Correction: Pathway and network analysis of more than 2500 whole cancer genomes
- Author
-
Reyna, Matthew A., Haan, David, Paczkowska, Marta, Verbeke, Lieven P. C., Vazquez, Miguel, Kahraman, Abdullah, Pulido-Tamayo, Sergio, Barenboim, Jonathan, Wadi, Lina, Dhingra, Priyanka, Shrestha, Raunak, Getz, Gad, Lawrence, Michael S., Pedersen, Jakob Skou, Rubin, Mark A., Wheeler, David A., Brunak, Søren, Izarzugaza, Jose M. G., Khurana, Ekta, Marchal, Kathleen, von Mering, Christian, Sahinalp, S. Cenk, Valencia, Alfonso, Reimand, Jüri, Stuart, Joshua M., and Raphael, Benjamin J.
- Published
- 2022
- Full Text
- View/download PDF
26. Clinical and Genomic Characterization of Treatment-Emergent Small-Cell Neuroendocrine Prostate Cancer: A Multi-institutional Prospective Study.
- Author
-
Aggarwal, Rahul, Huang, Jiaoti, Alumkal, Joshi J, Zhang, Li, Feng, Felix Y, Thomas, George V, Weinstein, Alana S, Friedl, Verena, Zhang, Can, Witte, Owen N, Lloyd, Paul, Gleave, Martin, Evans, Christopher P, Youngren, Jack, Beer, Tomasz M, Rettig, Matthew, Wong, Christopher K, True, Lawrence, Foye, Adam, Playdle, Denise, Ryan, Charles J, Lara, Primo, Chi, Kim N, Uzunangelov, Vlado, Sokolov, Artem, Newton, Yulia, Beltran, Himisha, Demichelis, Francesca, Rubin, Mark A, Stuart, Joshua M, and Small, Eric J
- Subjects
Prostate Cancer ,Clinical Research ,Human Genome ,Genetics ,Cancer ,Urologic Diseases ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,Aged ,Aged ,80 and over ,Carcinoma ,Neuroendocrine ,DNA Repair ,Humans ,Male ,Middle Aged ,Prospective Studies ,Prostatic Neoplasms ,Castration-Resistant ,Clinical Sciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Purpose The prevalence and features of treatment-emergent small-cell neuroendocrine prostate cancer (t-SCNC) are not well characterized in the era of modern androgen receptor (AR)-targeting therapy. We sought to characterize the clinical and genomic features of t-SCNC in a multi-institutional prospective study. Methods Patients with progressive, metastatic castration-resistant prostate cancer (mCRPC) underwent metastatic tumor biopsy and were followed for survival. Metastatic biopsy specimens underwent independent, blinded pathology review along with RNA/DNA sequencing. Results A total of 202 consecutive patients were enrolled. One hundred forty-eight (73%) had prior disease progression on abiraterone and/or enzalutamide. The biopsy evaluable rate was 79%. The overall incidence of t-SCNC detection was 17%. AR amplification and protein expression were present in 67% and 75%, respectively, of t-SCNC biopsy specimens. t-SCNC was detected at similar proportions in bone, node, and visceral organ biopsy specimens. Genomic alterations in the DNA repair pathway were nearly mutually exclusive with t-SCNC differentiation ( P = .035). Detection of t-SCNC was associated with shortened overall survival among patients with prior AR-targeting therapy for mCRPC (hazard ratio, 2.02; 95% CI, 1.07 to 3.82). Unsupervised hierarchical clustering of the transcriptome identified a small-cell-like cluster that further enriched for adverse survival outcomes (hazard ratio, 3.00; 95% CI, 1.25 to 7.19). A t-SCNC transcriptional signature was developed and validated in multiple external data sets with > 90% accuracy. Multiple transcriptional regulators of t-SCNC were identified, including the pancreatic neuroendocrine marker PDX1. Conclusion t-SCNC is present in nearly one fifth of patients with mCRPC and is associated with shortened survival. The near-mutual exclusivity with DNA repair alterations suggests t-SCNC may be a distinct subset of mCRPC. Transcriptional profiling facilitates the identification of t-SCNC and novel therapeutic targets.
- Published
- 2018
27. Comprehensive Characterization of Cancer Driver Genes and Mutations
- Author
-
Bailey, Matthew H, Tokheim, Collin, Porta-Pardo, Eduard, Sengupta, Sohini, Bertrand, Denis, Weerasinghe, Amila, Colaprico, Antonio, Wendl, Michael C, Kim, Jaegil, Reardon, Brendan, Ng, Patrick Kwok-Shing, Jeong, Kang Jin, Cao, Song, Wang, Zixing, Gao, Jianjiong, Gao, Qingsong, Wang, Fang, Liu, Eric Minwei, Mularoni, Loris, Rubio-Perez, Carlota, Nagarajan, Niranjan, Cortés-Ciriano, Isidro, Zhou, Daniel Cui, Liang, Wen-Wei, Hess, Julian M, Yellapantula, Venkata D, Tamborero, David, Gonzalez-Perez, Abel, Suphavilai, Chayaporn, Ko, Jia Yu, Khurana, Ekta, Park, Peter J, Van Allen, Eliezer M, Liang, Han, Lawrence, Michael S, Godzik, Adam, Lopez-Bigas, Nuria, Stuart, Joshua M, Wheeler, David A, Getz, Gad, Chen, Amy, Lazar, Alexander J, Mills, Gordon B, Karchin, Rachel, Ding, Li, Caesar-Johnson, Samantha J, Demchok, John A, Felau, Ina, Kasapi, Melpomeni, Ferguson, Martin L, Hutter, Carolyn M, Sofia, Heidi J, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Zenklusen, Jean C, Zhang, Jiashan, Chudamani, Sudha, Liu, Jia, Lolla, Laxmi, Naresh, Rashi, Pihl, Todd, Sun, Qiang, Wan, Yunhu, Wu, Ye, Cho, Juok, DeFreitas, Timothy, Frazer, Scott, Gehlenborg, Nils, Heiman, David I, Lin, Pei, Meier, Sam, Noble, Michael S, Saksena, Gordon, Voet, Doug, Zhang, Hailei, Bernard, Brady, Chambwe, Nyasha, Dhankani, Varsha, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Liu, Yuexin, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, Broom, Bradley M, Hegde, Apurva M, Ju, Zhenlin, Kanchi, Rupa S, Korkut, Anil, Li, Jun, and Ling, Shiyun
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,MC3 Working Group ,Cancer Genome Atlas Research Network ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
(Cell 173, 371–385.e1–e9; April 5, 2018) It has come to our attention that we made two errors in preparation of this manuscript. First, in the STAR Methods, under the subheading of “Hypermutators and Immune Infiltrates” within the “Quantification and Statistical Analysis” section, we inadvertently referred to Figures S7A–S7C for data corresponding to sample stratification by hypermutator status alone in the last sentence. It should have referred to Figure S6A–S6C. Second, the lists of highly frequent missense mutations for COAD (colon adenocarcinoma) and READ (rectum adenocarcinoma) displayed in Figure S7 were incorrect because when we ordered the mutations in the initial analysis, we mistakenly combined the two cancer types COAD and READ for analysis, despite the fact that they were listed as two separate cancer types in the x-axis of the figure. After re-ordering the mutations by frequency for COAD and READ independently, information on highly frequent missense mutations for each of these cancer types is different and updated now in the revised Figure S7. These errors don't change the major conclusions of the paper and have been corrected online. We apologize for any confusion they may have caused. [Figure-presented]
- Published
- 2018
28. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer.
- Author
-
Quigley, David A, Dang, Ha X, Zhao, Shuang G, Lloyd, Paul, Aggarwal, Rahul, Alumkal, Joshi J, Foye, Adam, Kothari, Vishal, Perry, Marc D, Bailey, Adina M, Playdle, Denise, Barnard, Travis J, Zhang, Li, Zhang, Jin, Youngren, Jack F, Cieslik, Marcin P, Parolia, Abhijit, Beer, Tomasz M, Thomas, George, Chi, Kim N, Gleave, Martin, Lack, Nathan A, Zoubeidi, Amina, Reiter, Robert E, Rettig, Matthew B, Witte, Owen, Ryan, Charles J, Fong, Lawrence, Kim, Won, Friedlander, Terence, Chou, Jonathan, Li, Haolong, Das, Rajdeep, Li, Hui, Moussavi-Baygi, Ruhollah, Goodarzi, Hani, Gilbert, Luke A, Lara, Primo N, Evans, Christopher P, Goldstein, Theodore C, Stuart, Joshua M, Tomlins, Scott A, Spratt, Daniel E, Cheetham, R Keira, Cheng, Donavan T, Farh, Kyle, Gehring, Julian S, Hakenberg, Jörg, Liao, Arnold, Febbo, Philip G, Shon, John, Sickler, Brad, Batzoglou, Serafim, Knudsen, Karen E, He, Housheng H, Huang, Jiaoti, Wyatt, Alexander W, Dehm, Scott M, Ashworth, Alan, Chinnaiyan, Arul M, Maher, Christopher A, Small, Eric J, and Feng, Felix Y
- Subjects
Humans ,Prostatic Neoplasms ,Neoplasm Metastasis ,Cyclin-Dependent Kinases ,Proto-Oncogene Proteins c-myc ,BRCA2 Protein ,Receptors ,Androgen ,Gene Expression Profiling ,Genomics ,Tandem Repeat Sequences ,Mutation ,Aged ,Aged ,80 and over ,Middle Aged ,Male ,Tumor Suppressor Protein p53 ,Genomic Structural Variation ,DNA Copy Number Variations ,Exome ,Whole Genome Sequencing ,BRCA2 ,androgen receptor ,castration resistant prostate cancer ,chromothripsis ,gene fusion ,genomics ,metastases ,structural variation ,tandem duplication ,whole-genome sequencing ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
While mutations affecting protein-coding regions have been examined across many cancers, structural variants at the genome-wide level are still poorly defined. Through integrative deep whole-genome and -transcriptome analysis of 101 castration-resistant prostate cancer metastases (109X tumor/38X normal coverage), we identified structural variants altering critical regulators of tumorigenesis and progression not detectable by exome approaches. Notably, we observed amplification of an intergenic enhancer region 624 kb upstream of the androgen receptor (AR) in 81% of patients, correlating with increased AR expression. Tandem duplication hotspots also occur near MYC, in lncRNAs associated with post-translational MYC regulation. Classes of structural variations were linked to distinct DNA repair deficiencies, suggesting their etiology, including associations of CDK12 mutation with tandem duplications, TP53 inactivation with inverted rearrangements and chromothripsis, and BRCA2 inactivation with deletions. Together, these observations provide a comprehensive view of how structural variations affect critical regulators in metastatic prostate cancer.
- Published
- 2018
29. Integrated Molecular Characterization of Testicular Germ Cell Tumors
- Author
-
Shen, Hui, Shih, Juliann, Hollern, Daniel P, Wang, Linghua, Bowlby, Reanne, Tickoo, Satish K, Thorsson, Vésteinn, Mungall, Andrew J, Newton, Yulia, Hegde, Apurva M, Armenia, Joshua, Sánchez-Vega, Francisco, Pluta, John, Pyle, Louise C, Mehra, Rohit, Reuter, Victor E, Godoy, Guilherme, Jones, Jeffrey, Shelley, Carl S, Feldman, Darren R, Vidal, Daniel O, Lessel, Davor, Kulis, Tomislav, Cárcano, Flavio M, Leraas, Kristen M, Lichtenberg, Tara M, Brooks, Denise, Cherniack, Andrew D, Cho, Juok, Heiman, David I, Kasaian, Katayoon, Liu, Minwei, Noble, Michael S, Xi, Liu, Zhang, Hailei, Zhou, Wanding, ZenKlusen, Jean C, Hutter, Carolyn M, Felau, Ina, Zhang, Jiashan, Schultz, Nikolaus, Getz, Gad, Meyerson, Matthew, Stuart, Joshua M, Akbani, Rehan, Wheeler, David, Laird, Peter W, Nathanson, Katherine L, Cortessis, Victoria K, Hoadley, Katherine A, Wheeler, David A, Hughes, Daniel, Covington, Kyle, Jayaseelan, Joy C, Korchina, Viktoriya, Lewis, Lora, Hu, Jianhong, Doddapaneni, HarshaVardhan, Muzny, Donna, Gibbs, Richard, Hollern, Daniel, Vincent, Benjamin G, Chai, Shengjie, Smith, Christof C, Auman, J Todd, Shi, Yan, Meng, Shaowu, Skelly, Tara, Tan, Donghui, Veluvolu, Umadevi, Mieczkowski, Piotr A, Jones, Corbin D, Wilkerson, Matthew D, Balu, Saianand, Bodenheimer, Tom, Hoyle, Alan P, Jefferys, Stuart R, Mose, Lisle E, Simons, Janae V, Soloway, Matthew G, Roach, Jeffrey, Parker, Joel S, Hayes, D Neil, Perou, Charles M, Saksena, Gordon, Cibulskis, Carrie, Schumacher, Steven E, Beroukhim, Rameen, Gabriel, Stacey B, and Ally, Adrian
- Subjects
Urologic Diseases ,Rare Diseases ,Human Genome ,Cancer ,Biotechnology ,Genetics ,DNA Copy Number Variations ,DNA Methylation ,Gene Expression Regulation ,Neoplastic ,Humans ,Male ,MicroRNAs ,Neoplasms ,Germ Cell and Embryonal ,Proto-Oncogene Proteins c-kit ,Seminoma ,Testicular Neoplasms ,ras Proteins ,Cancer Genome Atlas Research Network ,DNA methylation ,KIT ,The Cancer Genome Atlas ,copy number ,exome sequencing ,miR-375 ,nonseminoma ,seminoma ,testicular germ cell tumors ,Biochemistry and Cell Biology ,Medical Physiology - Abstract
We studied 137 primary testicular germ cell tumors (TGCTs) using high-dimensional assays of genomic, epigenomic, transcriptomic, and proteomic features. These tumors exhibited high aneuploidy and a paucity of somatic mutations. Somatic mutation of only three genes achieved significance-KIT, KRAS, and NRAS-exclusively in samples with seminoma components. Integrated analyses identified distinct molecular patterns that characterized the major recognized histologic subtypes of TGCT: seminoma, embryonal carcinoma, yolk sac tumor, and teratoma. Striking differences in global DNA methylation and microRNA expression between histology subtypes highlight a likely role of epigenomic processes in determining histologic fates in TGCTs. We also identified a subset of pure seminomas defined by KIT mutations, increased immune infiltration, globally demethylated DNA, and decreased KRAS copy number. We report potential biomarkers for risk stratification, such as miRNA specifically expressed in teratoma, and others with molecular diagnostic potential, such as CpH (CpA/CpC/CpT) methylation identifying embryonal carcinomas.
- Published
- 2018
30. Comparative RNA-Sequencing Analysis Benefits a Pediatric Patient With Relapsed Cancer
- Author
-
Newton, Yulia, Rassekh, S Rod, Deyell, Rebecca J, Shen, Yaoqing, Jones, Martin R, Dunham, Chris, Yip, Stephen, Leelakumari, Sreeja, Zhu, Jingchun, McColl, Duncan, Swatloski, Teresa, Salama, Sofie R, Ng, Tony, Hendson, Glenda, Lee, Anna F, Ma, Yussanne, Moore, Richard, Mungall, Andrew J, Haussler, David, Stuart, Joshua M, Jantzen, Colleen, Laskin, Janessa, Jones, Steven JM, Marra, Marco A, and Morozova, Olena
- Subjects
Genetics ,Pediatric Research Initiative ,Pediatric ,Biotechnology ,Pediatric Cancer ,Human Genome ,Rare Diseases ,Cancer ,Oncology and carcinogenesis - Abstract
Clinical detection of sequence and structural variants in known cancer genes points to viable treatment options for a minority of children with cancer.1 To increase the number of children who benefit from genomic profiling, gene expression information must be considered alongside mutations.2,3 Although high expression has been used to nominate drug targets for pediatric cancers,4,5 its utility has not been evaluated in a systematic way.6 We describe a child with a rare sarcoma that was profiled with whole-genome and RNA sequencing (RNA-Seq) techniques. Although the tumor did not harbor DNA mutations targetable by available therapies, incorporation of gene expression information derived from RNA-Seq analysis led to a therapy that produced a significant clinical response. We use this case to describe a framework for inclusion of gene expression into the clinical genomic evaluation of pediatric tumors.
- Published
- 2018
31. A Pan-Cancer Analysis of Enhancer Expression in Nearly 9000 Patient Samples
- Author
-
Chen, Han, Li, Chunyan, Peng, Xinxin, Zhou, Zhicheng, Weinstein, John N, Network, The Cancer Genome Atlas Research, Caesar-Johnson, Samantha J, Demchok, John A, Felau, Ina, Kasapi, Melpomeni, Ferguson, Martin L, Hutter, Carolyn M, Sofia, Heidi J, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Zenklusen, Jean C, Zhang, Jiashan, Chudamani, Sudha, Liu, Jia, Lolla, Laxmi, Naresh, Rashi, Pihl, Todd, Sun, Qiang, Wan, Yunhu, Wu, Ye, Cho, Juok, DeFreitas, Timothy, Frazer, Scott, Gehlenborg, Nils, Getz, Gad, Heiman, David I, Kim, Jaegil, Lawrence, Michael S, Lin, Pei, Meier, Sam, Noble, Michael S, Saksena, Gordon, Voet, Doug, Zhang, Hailei, Bernard, Brady, Chambwe, Nyasha, Dhankani, Varsha, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Liu, Yuexin, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, Broom, Bradley M, Hegde, Apurva M, Ju, Zhenlin, Kanchi, Rupa S, Korkut, Anil, Li, Jun, Liang, Han, Ling, Shiyun, Liu, Wenbin, Lu, Yiling, Mills, Gordon B, Ng, Kwok-Shing, Rao, Arvind, Ryan, Michael, Wang, Jing, Zhang, Jiexin, Abeshouse, Adam, Armenia, Joshua, Chakravarty, Debyani, Chatila, Walid K, de Bruijn, Ino, Gao, Jianjiong, Gross, Benjamin E, Heins, Zachary J, Kundra, Ritika, La, Konnor, Ladanyi, Marc, Luna, Augustin, Nissan, Moriah G, Ochoa, Angelica, Phillips, Sarah M, Reznik, Ed, Sanchez-Vega, Francisco, Sander, Chris, Schultz, Nikolaus, Sheridan, Robert, Sumer, S Onur, Sun, Yichao, Taylor, Barry S, Wang, Jioajiao, Zhang, Hongxin, Anur, Pavana, Peto, Myron, Spellman, Paul, Benz, Christopher, and Stuart, Joshua M
- Subjects
Genetics ,Cancer ,Human Genome ,2.1 Biological and endogenous factors ,Aetiology ,Aneuploidy ,B7-H1 Antigen ,Chromatin ,Databases ,Genetic ,Enhancer Elements ,Genetic ,Gene Expression Regulation ,Neoplastic ,Humans ,Immunotherapy ,Neoplasms ,Sequence Analysis ,RNA ,Survival Rate ,Cancer Genome Atlas Research Network ,PD-L1 expression ,The Cancer Genome Atlas ,aneuploidy ,chromatin state ,enhancer expression ,mutation burden ,pan-cancer analysis ,prognostic markers ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
The role of enhancers, a key class of non-coding regulatory DNA elements, in cancer development has increasingly been appreciated. Here, we present the detection and characterization of a large number of expressed enhancers in a genome-wide analysis of 8928 tumor samples across 33 cancer types using TCGA RNA-seq data. Compared with matched normal tissues, global enhancer activation was observed in most cancers. Across cancer types, global enhancer activity was positively associated with aneuploidy, but not mutation load, suggesting a hypothesis centered on "chromatin-state" to explain their interplay. Integrating eQTL, mRNA co-expression, and Hi-C data analysis, we developed a computational method to infer causal enhancer-gene interactions, revealing enhancers of clinically actionable genes. Having identified an enhancer ∼140 kb downstream of PD-L1, a major immunotherapy target, we validated it experimentally. This study provides a systematic view of enhancer activity in diverse tumor contexts and suggests the clinical implications of enhancers.
- Published
- 2018
32. Perspective on Oncogenic Processes at the End of the Beginning of Cancer Genomics
- Author
-
Ding, Li, Bailey, Matthew H, Porta-Pardo, Eduard, Thorsson, Vesteinn, Colaprico, Antonio, Bertrand, Denis, Gibbs, David L, Weerasinghe, Amila, Huang, Kuan-lin, Tokheim, Collin, Cortés-Ciriano, Isidro, Jayasinghe, Reyka, Chen, Feng, Yu, Lihua, Sun, Sam, Olsen, Catharina, Kim, Jaegil, Taylor, Alison M, Cherniack, Andrew D, Akbani, Rehan, Suphavilai, Chayaporn, Nagarajan, Niranjan, Stuart, Joshua M, Mills, Gordon B, Wyczalkowski, Matthew A, Vincent, Benjamin G, Hutter, Carolyn M, Zenklusen, Jean Claude, Hoadley, Katherine A, Wendl, Michael C, Shmulevich, llya, Lazar, Alexander J, Wheeler, David A, Getz, Gad, Network, The Cancer Genome Atlas Research, Caesar-Johnson, Samantha J, Demchok, John A, Felau, Ina, Kasapi, Melpomeni, Ferguson, Martin L, Sofia, Heidi J, Tarnuzzer, Roy, Wang, Zhining, Yang, Liming, Zenklusen, Jean C, Zhang, Jiashan, Chudamani, Sudha, Liu, Jia, Lolla, Laxmi, Naresh, Rashi, Pihl, Todd, Sun, Qiang, Wan, Yunhu, Wu, Ye, Cho, Juok, DeFreitas, Timothy, Frazer, Scott, Gehlenborg, Nils, Heiman, David I, Lawrence, Michael S, Lin, Pei, Meier, Sam, Noble, Michael S, Saksena, Gordon, Voet, Doug, Zhang, Hailei, Bernard, Brady, Chambwe, Nyasha, Dhankani, Varsha, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Liu, Yuexin, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Zhang, Wei, Broom, Bradley M, Hegde, Apurva M, Ju, Zhenlin, Kanchi, Rupa S, Korkut, Anil, Li, Jun, Liang, Han, Ling, Shiyun, Liu, Wenbin, Lu, Yiling, Ng, Kwok-Shing, Rao, Arvind, Ryan, Michael, Wang, Jing, Weinstein, John N, Zhang, Jiexin, and Abeshouse, Adam
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Bioinformatics and Computational Biology ,Genetics ,Oncology and Carcinogenesis ,Cancer ,Cancer Genomics ,Human Genome ,Biotechnology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Carcinogenesis ,DNA Repair ,Databases ,Genetic ,Genes ,Neoplasm ,Genomics ,Humans ,Metabolic Networks and Pathways ,Microsatellite Instability ,Mutation ,Neoplasms ,Transcriptome ,Tumor Microenvironment ,Cancer Genome Atlas Research Network ,TCGA ,cancer ,cancer genomics ,omics ,oncogenic process ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The Cancer Genome Atlas (TCGA) has catalyzed systematic characterization of diverse genomic alterations underlying human cancers. At this historic junction marking the completion of genomic characterization of over 11,000 tumors from 33 cancer types, we present our current understanding of the molecular processes governing oncogenesis. We illustrate our insights into cancer through synthesis of the findings of the TCGA PanCancer Atlas project on three facets of oncogenesis: (1) somatic driver mutations, germline pathogenic variants, and their interactions in the tumor; (2) the influence of the tumor genome and epigenome on transcriptome and proteome; and (3) the relationship between tumor and the microenvironment, including implications for drugs targeting driver events and immunotherapies. These results will anchor future characterization of rare and common tumor types, primary and relapsed tumors, and cancers across ancestry groups and will guide the deployment of clinical genomic sequencing.
- Published
- 2018
33. Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer.
- Author
-
Hoadley, Katherine A, Yau, Christina, Hinoue, Toshinori, Wolf, Denise M, Lazar, Alexander J, Drill, Esther, Shen, Ronglai, Taylor, Alison M, Cherniack, Andrew D, Thorsson, Vésteinn, Akbani, Rehan, Bowlby, Reanne, Wong, Christopher K, Wiznerowicz, Maciej, Sanchez-Vega, Francisco, Robertson, A Gordon, Schneider, Barbara G, Lawrence, Michael S, Noushmehr, Houtan, Malta, Tathiane M, Cancer Genome Atlas Network, Stuart, Joshua M, Benz, Christopher C, and Laird, Peter W
- Subjects
Cancer Genome Atlas Network ,Chromosomes ,Humans ,Neoplasms ,Aneuploidy ,Neoplasm Proteins ,MicroRNAs ,RNA ,Messenger ,Cluster Analysis ,DNA Methylation ,CpG Islands ,Mutation ,Databases ,Factual ,TCGA ,cancer ,cell-of-origin ,genome ,methylome ,organs ,proteome ,subtypes ,tissues ,transcriptome ,Genetics ,Cancer ,Human Genome ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences - Abstract
We conducted comprehensive integrative molecular analyses of the complete set of tumors in The Cancer Genome Atlas (TCGA), consisting of approximately 10,000 specimens and representing 33 types of cancer. We performed molecular clustering using data on chromosome-arm-level aneuploidy, DNA hypermethylation, mRNA, and miRNA expression levels and reverse-phase protein arrays, of which all, except for aneuploidy, revealed clustering primarily organized by histology, tissue type, or anatomic origin. The influence of cell type was evident in DNA-methylation-based clustering, even after excluding sites with known preexisting tissue-type-specific methylation. Integrative clustering further emphasized the dominant role of cell-of-origin patterns. Molecular similarities among histologically or anatomically related cancer types provide a basis for focused pan-cancer analyses, such as pan-gastrointestinal, pan-gynecological, pan-kidney, and pan-squamous cancers, and those related by stemness features, which in turn may inform strategies for future therapeutic development.
- Published
- 2018
34. Revealing cancer subtypes with higher-order correlations applied to imaging and omics data
- Author
-
Graim, Kiley, Liu, Tiffany Ting, Achrol, Achal S, Paull, Evan O, Newton, Yulia, Chang, Steven D, Harsh, Griffith R, Cordero, Sergio P, Rubin, Daniel L, and Stuart, Joshua M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Brain Cancer ,Brain Disorders ,Rare Diseases ,Urologic Diseases ,Clinical Research ,Cancer ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,Computational Biology ,DNA Copy Number Variations ,Genotype ,Glioblastoma ,Humans ,Magnetic Resonance Imaging ,Mutation ,Phenotype ,Molecular subtyping ,Community detection ,MRI ,Magnetic resonance imaging ,Clustering ,Genetics ,Medical Biochemistry and Metabolomics ,Genetics & Heredity ,Medical biochemistry and metabolomics - Abstract
BackgroundPatient stratification to identify subtypes with different disease manifestations, severity, and expected survival time is a critical task in cancer diagnosis and treatment. While stratification approaches using various biomarkers (including high-throughput gene expression measurements) for patient-to-patient comparisons have been successful in elucidating previously unseen subtypes, there remains an untapped potential of incorporating various genotypic and phenotypic data to discover novel or improved groupings.MethodsHere, we present HOCUS, a unified analytical framework for patient stratification that uses a community detection technique to extract subtypes out of sparse patient measurements. HOCUS constructs a patient-to-patient network from similarities in the data and iteratively groups and reconstructs the network into higher order clusters. We investigate the merits of using higher-order correlations to cluster samples of cancer patients in terms of their associations with survival outcomes.ResultsIn an initial test of the method, the approach identifies cancer subtypes in mutation data of glioblastoma, ovarian, breast, prostate, and bladder cancers. In several cases, HOCUS provides an improvement over using the molecular features directly to compare samples. Application of HOCUS to glioblastoma images reveals a size and location classification of tumors that improves over human expert-based stratification.ConclusionsSubtypes based on higher order features can reveal comparable or distinct groupings. The distinct solutions can provide biologically- and treatment-relevant solutions that are just as significant as solutions based on the original data.
- Published
- 2017
35. A Community Challenge for Inferring Genetic Predictors of Gene Essentialities through Analysis of a Functional Screen of Cancer Cell Lines
- Author
-
Gönen, Mehmet, Weir, Barbara A, Cowley, Glenn S, Vazquez, Francisca, Guan, Yuanfang, Jaiswal, Alok, Karasuyama, Masayuki, Uzunangelov, Vladislav, Wang, Tao, Tsherniak, Aviad, Howell, Sara, Marbach, Daniel, Hoff, Bruce, Norman, Thea C, Airola, Antti, Bivol, Adrian, Bunte, Kerstin, Carlin, Daniel, Chopra, Sahil, Deran, Alden, Ellrott, Kyle, Gopalacharyulu, Peddinti, Graim, Kiley, Kaski, Samuel, Khan, Suleiman A, Newton, Yulia, Ng, Sam, Pahikkala, Tapio, Paull, Evan, Sokolov, Artem, Tang, Hao, Tang, Jing, Wennerberg, Krister, Xie, Yang, Zhan, Xiaowei, Zhu, Fan, Community, Broad-DREAM, Afsari, Bahman, Aittokallio, Tero, Boehm, Jesse S, Chang, Yu-Chuan, Chen, Tenghui, Chong, Zechen, Elmarakeby, Haitham, Fertig, Elana J, Gonçalves, Emanuel, Gong, Pinghua, Hafemeister, Christoph, Hahn, William C, Heath, Lenwood, Kędziorski, Łukasz, Khemka, Niraj, King, Erh-kan, Lauria, Mario, Liu, Mark, Machado, Daniel, Mamitsuka, Hiroshi, Margolin, Adam A, Mazurkiewicz, Mateusz, Menden, Michael P, Migacz, Szymon, Nie, Zhi, Praveen, Paurush, Priami, Corrado, Rizzetto, Simone, Rocha, Miguel, Root, David E, Rudd, Cameron, Rudnicki, Witold R, Saez-Rodriguez, Julio, Song, Lei, Stolovitzky, Gustavo, Stuart, Joshua M, Sun, Duanchen, and Szalai, Bence
- Subjects
Biological Sciences ,Genetics ,Biotechnology ,Prevention ,Cancer ,Human Genome ,Algorithms ,Cell Line ,Tumor ,Gene Expression ,Genes ,Essential ,Genomics ,Humans ,RNA ,Small Interfering ,Broad-DREAM Community ,cancer genomics ,community challenge ,crowdsourcing ,functional screen ,machine learning ,oncogene ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
We report the results of a DREAM challenge designed to predict relative genetic essentialities based on a novel dataset testing 98,000 shRNAs against 149 molecularly characterized cancer cell lines. We analyzed the results of over 3,000 submissions over a period of 4 months. We found that algorithms combining essentiality data across multiple genes demonstrated increased accuracy; gene expression was the most informative molecular data type; the identity of the gene being predicted was far more important than the modeling strategy; well-predicted genes and selected molecular features showed enrichment in functional categories; and frequently selected expression features correlated with survival in primary tumors. This study establishes benchmarks for gene essentiality prediction, presents a community resource for future comparison with this benchmark, and provides insights into factors influencing the ability to predict gene essentiality from functional genetic screens. This study also demonstrates the value of releasing pre-publication data publicly to engage the community in an open research collaboration.
- Published
- 2017
36. TumorMap: Exploring the Molecular Similarities of Cancer Samples in an Interactive Portal
- Author
-
Newton, Yulia, Novak, Adam M, Swatloski, Teresa, McColl, Duncan C, Chopra, Sahil, Graim, Kiley, Weinstein, Alana S, Baertsch, Robert, Salama, Sofie R, Ellrott, Kyle, Chopra, Manu, Goldstein, Theodore C, Haussler, David, Morozova, Olena, and Stuart, Joshua M
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Genetics ,Biotechnology ,Human Genome ,Chromosome Mapping ,Computational Biology ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Genome ,Human ,Genomics ,Humans ,Mutation ,Neoplasms ,Reproducibility of Results ,Software ,User-Computer Interface ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Vast amounts of molecular data are being collected on tumor samples, which provide unique opportunities for discovering trends within and between cancer subtypes. Such cross-cancer analyses require computational methods that enable intuitive and interactive browsing of thousands of samples based on their molecular similarity. We created a portal called TumorMap to assist in exploration and statistical interrogation of high-dimensional complex "omics" data in an interactive and easily interpretable way. In the TumorMap, samples are arranged on a hexagonal grid based on their similarity to one another in the original genomic space and are rendered with Google's Map technology. While the important feature of this public portal is the ability for the users to build maps from their own data, we pre-built genomic maps from several previously published projects. We demonstrate the utility of this portal by presenting results obtained from The Cancer Genome Atlas project data. Cancer Res; 77(21); e111-4. ©2017 AACR.
- Published
- 2017
37. Common and cell-type specific responses to anti-cancer drugs revealed by high throughput transcript profiling.
- Author
-
Niepel, Mario, Hafner, Marc, Duan, Qiaonan, Wang, Zichen, Paull, Evan O, Chung, Mirra, Lu, Xiaodong, Stuart, Joshua M, Golub, Todd R, Subramanian, Aravind, Ma'ayan, Avi, and Sorger, Peter K
- Subjects
Cell Line ,Tumor ,Humans ,Antineoplastic Agents ,Gene Expression Profiling ,Drug Synergism ,High-Throughput Screening Assays ,Genetics ,Cancer ,Pediatric Research Initiative ,5.1 Pharmaceuticals ,1.1 Normal biological development and functioning ,Cell Line ,Tumor ,MD Multidisciplinary - Abstract
More effective use of targeted anti-cancer drugs depends on elucidating the connection between the molecular states induced by drug treatment and the cellular phenotypes controlled by these states, such as cytostasis and death. This is particularly true when mutation of a single gene is inadequate as a predictor of drug response. The current paper describes a data set of ~600 drug cell line pairs collected as part of the NIH LINCS Program ( http://www.lincsproject.org/ ) in which molecular data (reduced dimensionality transcript L1000 profiles) were recorded across dose and time in parallel with phenotypic data on cellular cytostasis and cytotoxicity. We report that transcriptional and phenotypic responses correlate with each other in general, but whereas inhibitors of chaperones and cell cycle kinases induce similar transcriptional changes across cell lines, changes induced by drugs that inhibit intra-cellular signaling kinases are cell-type specific. In some drug/cell line pairs significant changes in transcription are observed without a change in cell growth or survival; analysis of such pairs identifies drug equivalence classes and, in one case, synergistic drug interactions. In this case, synergy involves cell-type specific suppression of an adaptive drug response.
- Published
- 2017
38. TRACING CO-REGULATORY NETWORK DYNAMICS IN NOISY, SINGLE-CELL TRANSCRIPTOME TRAJECTORIES
- Author
-
Altman, Russ B, Dunker, A Keith, Hunter, Lawrence, Ritchie, Marylyn D, Murray, Tiffany A, Klein, Teri E, CORDERO, PABLO, and STUART, JOSHUA M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biotechnology ,Genetics ,1.1 Normal biological development and functioning ,Underpinning research ,Cell Differentiation ,Computational Biology ,Computer Simulation ,Gene Expression Profiling ,Gene Regulatory Networks ,Humans ,Models ,Genetic ,Models ,Neurological ,Models ,Statistical ,Neural Stem Cells ,Neurogenesis ,Normal Distribution ,Signal-To-Noise Ratio ,Single-Cell Analysis ,Systems Biology ,single-cell measurements ,Gaussian mixtures ,transcriptomics ,single-cell trajectory reconstruction - Abstract
The availability of gene expression data at the single cell level makes it possible to probe the molecular underpinnings of complex biological processes such as differentiation and oncogenesis. Promising new methods have emerged for reconstructing a progression 'trajectory' from static single-cell transcriptome measurements. However, it remains unclear how to adequately model the appreciable level of noise in these data to elucidate gene regulatory network rewiring. Here, we present a framework called Single Cell Inference of MorphIng Trajectories and their Associated Regulation (SCIMITAR) that infers progressions from static single-cell transcriptomes by employing a continuous parametrization of Gaussian mixtures in high-dimensional curves. SCIMITAR yields rich models from the data that highlight genes with expression and co-expression patterns that are associated with the inferred progression. Further, SCIMITAR extracts regulatory states from the implicated trajectory-evolvingco-expression networks. We benchmark the method on simulated data to show that it yields accurate cell ordering and gene network inferences. Applied to the interpretation of a single-cell human fetal neuron dataset, SCIMITAR finds progression-associated genes in cornerstone neural differentiation pathways missed by standard differential expression tests. Finally, by leveraging the rewiring of gene-gene co-expression relations across the progression, the method reveals the rise and fall of co-regulatory states and trajectory-dependent gene modules. These analyses implicate new transcription factors in neural differentiation including putative co-factors for the multi-functional NFAT pathway.
- Published
- 2017
39. Prophetic Granger Causality to infer gene regulatory networks
- Author
-
Carlin, Daniel E, Paull, Evan O, Graim, Kiley, Wong, Christopher K, Bivol, Adrian, Ryabinin, Peter, Ellrott, Kyle, Sokolov, Artem, and Stuart, Joshua M
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Cancer ,Bioengineering ,Biotechnology ,Genetics ,Causality ,Computational Biology ,Gene Regulatory Networks ,Humans ,Machine Learning ,Models ,Theoretical ,Neoplasms ,Systems Biology ,General Science & Technology - Abstract
We introduce a novel method called Prophetic Granger Causality (PGC) for inferring gene regulatory networks (GRNs) from protein-level time series data. The method uses an L1-penalized regression adaptation of Granger Causality to model protein levels as a function of time, stimuli, and other perturbations. When combined with a data-independent network prior, the framework outperformed all other methods submitted to the HPN-DREAM 8 breast cancer network inference challenge. Our investigations reveal that PGC provides complementary information to other approaches, raising the performance of ensemble learners, while on its own achieves moderate performance. Thus, PGC serves as a valuable new tool in the bioinformatics toolkit for analyzing temporal datasets. We investigate the general and cell-specific interactions predicted by our method and find several novel interactions, demonstrating the utility of the approach in charting new tumor wiring.
- Published
- 2017
40. Targeting Adaptive Pathways in Metastatic Treatment-Resistant Prostate Cancer: Update on the Stand Up 2 Cancer/Prostate Cancer Foundation-Supported West Coast Prostate Cancer Dream Team.
- Author
-
Aggarwal, Rahul, Beer, Tomasz M, Gleave, Martin, Stuart, Joshua M, Rettig, Matthew, Evans, Christopher P, Youngren, Jack, Alumkal, Joshi J, Huang, Jiaoti, Thomas, George, Witte, Owen, and Small, Eric J
- Subjects
(AR) pathway inhibitors ,abiraterone ,enzalutamide ,Prostate Cancer ,Clinical Research ,Cancer ,Urologic Diseases ,Good Health and Well Being ,Clinical Sciences - Abstract
The Stand Up 2 Cancer/Prostate Cancer Foundation-funded West Coast Dream Team project is a prospective multi-institutional study focused on acquiring metastatic castration-resistant prostate cancer (mCRPC) biopsy tissue at the time of resistance to abiraterone or enzalutamide. It is the first large-scale study designed to analyze mCRPC tissue specifically in this patient population. Study accrual is on target, with 261 out of a planned 300 metastatic tumor biopsies performed by August 2016. Paired biopsies have been completed in 42 patients, with paired genomic data before and after therapy obtained in 26 cases. Accrual is expected to be complete by December 2016.
- Published
- 2016
41. Inferring causal molecular networks: empirical assessment through a community-based effort
- Author
-
Hill, Steven M., Heiser, Laura M., Cokelaer, Thomas, Unger, Michael, Nesser, Nicole K., Carlin, Daniel E., Zhang, Yang, Sokolov, Artem, Paull, Evan O., Wong, Chris K., Graim, Kiley, Bivol, Adrian, Wang, Haizhou, Zhu, Fan, Afsari, Bahman, Danilova, Ludmila V., Favorov, Alexander V., Lee, Wai Shing, Taylor, Dane, Hu, Chenyue W., Long, Byron L., Noren, David P., Bisberg, Alexander J., Mills, Gordon B., Gray, Joe W., Kellen, Michael, Norman, Thea, Friend, Stephen, Qutub, Amina A., Fertig, Elana J., Guan, Yuanfang, Song, Mingzhou, Stuart, Joshua M., Spellman, Paul T., Koeppl, Heinz, Stolovitzky, Gustavo, Saez-Rodriguez, Julio, Mukherjee, Sach, Hill, Steven M., Heiser, Laura M., Cokelaer, Thomas, Unger, Michael, Nesser, Nicole K., Carlin, Daniel E., Zhang, Yang, Sokolov, Artem, Paull, Evan O., Wong, Chris K., Graim, Kiley, Bivol, Adrian, Wang, Haizhou, Zhu, Fan, Afsari, Bahman, Danilova, Ludmila V., Favorov, Alexander V., Lee, Wai Shing, Taylor, Dane, Hu, Chenyue W., Long, Byron L., Noren, David P., Bisberg, Alexander J., Mills, Gordon B., Gray, Joe W., Kellen, Michael, Norman, Thea, Friend, Stephen, Qutub, Amina A., Fertig, Elana J., Guan, Yuanfang, Song, Mingzhou, Stuart, Joshua M., Spellman, Paul T., Koeppl, Heinz, Stolovitzky, Gustavo, Saez-Rodriguez, Julio, and Mukherjee, Sach
- Abstract
It remains unclear whether causal, rather than merely correlational, relationships in molecular networks can be inferred in complex biological settings. Here we describe the HPN-DREAM network inference challenge, which focused on learning causal influences in signaling networks. We used phosphoprotein data from cancer cell lines as well as in silico data from a nonlinear dynamical model. Using the phosphoprotein data, we scored more than 2,000 networks submitted by challenge participants. The networks spanned 32 biological contexts and were scored in terms of causal validity with respect to unseen interventional data. A number of approaches were effective, and incorporating known biology was generally advantageous. Additional sub-challenges considered time-course prediction and visualization. Our results suggest that learning causal relationships may be feasible in complex settings such as disease states. Furthermore, our scoring approach provides a practical way to empirically assess inferred molecular networks in a causal sense.
- Published
- 2024
42. Clinical and genomic characterization of Low PSA Secretors: a unique subset of metastatic castration resistant prostate cancer
- Author
-
Aggarwal, Rahul, Romero, Gustavo Rubio, Friedl, Verena, Weinstein, Alana, Foye, Adam, Huang, Jiaoti, Feng, Felix, Stuart, Joshua M., and Small, Eric J.
- Published
- 2021
- Full Text
- View/download PDF
43. Author Correction: Analyses of non-coding somatic drivers in 2,658 cancer whole genomes
- Author
-
Rheinbay, Esther, Nielsen, Morten Muhlig, Abascal, Federico, Wala, Jeremiah A., Shapira, Ofer, Tiao, Grace, Hornshøj, Henrik, Hess, Julian M., Juul, Randi Istrup, Lin, Ziao, Feuerbach, Lars, Sabarinathan, Radhakrishnan, Madsen, Tobias, Kim, Jaegil, Mularoni, Loris, Shuai, Shimin, Lanzós, Andrés, Herrmann, Carl, Maruvka, Yosef E., Shen, Ciyue, Amin, Samirkumar B., Bandopadhayay, Pratiti, Bertl, Johanna, Boroevich, Keith A., Busanovich, John, Carlevaro-Fita, Joana, Chakravarty, Dimple, Chan, Calvin Wing Yiu, Craft, David, Dhingra, Priyanka, Diamanti, Klev, Fonseca, Nuno A., Gonzalez-Perez, Abel, Guo, Qianyun, Hamilton, Mark P., Haradhvala, Nicholas J., Hong, Chen, Isaev, Keren, Johnson, Todd A., Juul, Malene, Kahles, Andre, Kahraman, Abdullah, Kim, Youngwook, Komorowski, Jan, Kumar, Kiran, Kumar, Sushant, Lee, Donghoon, Lehmann, Kjong-Van, Li, Yilong, Liu, Eric Minwei, Lochovsky, Lucas, Park, Keunchil, Pich, Oriol, Roberts, Nicola D., Saksena, Gordon, Schumacher, Steven E., Sidiropoulos, Nikos, Sieverling, Lina, Sinnott-Armstrong, Nasa, Stewart, Chip, Tamborero, David, Tubio, Jose M. C., Umer, Husen M., Uusküla-Reimand, Liis, Wadelius, Claes, Wadi, Lina, Yao, Xiaotong, Zhang, Cheng-Zhong, Zhang, Jing, Haber, James E., Hobolth, Asger, Imielinski, Marcin, Kellis, Manolis, Lawrence, Michael S., von Mering, Christian, Nakagawa, Hidewaki, Raphael, Benjamin J., Rubin, Mark A., Sander, Chris, Stein, Lincoln D., Stuart, Joshua M., Tsunoda, Tatsuhiko, Wheeler, David A., Johnson, Rory, Reimand, Jüri, Gerstein, Mark, Khurana, Ekta, Campbell, Peter J., López-Bigas, Núria, Weischenfeldt, Joachim, Beroukhim, Rameen, Martincorena, Iñigo, Pedersen, Jakob Skou, and Getz, Gad
- Published
- 2023
- Full Text
- View/download PDF
44. Phosphoproteome Integration Reveals Patient-Specific Networks in Prostate Cancer.
- Author
-
Drake, Justin M, Paull, Evan O, Graham, Nicholas A, Lee, John K, Smith, Bryan A, Titz, Bjoern, Stoyanova, Tanya, Faltermeier, Claire M, Uzunangelov, Vladislav, Carlin, Daniel E, Fleming, Daniel Teo, Wong, Christopher K, Newton, Yulia, Sudha, Sud, Vashisht, Ajay A, Huang, Jiaoti, Wohlschlegel, James A, Graeber, Thomas G, Witte, Owen N, and Stuart, Joshua M
- Subjects
Humans ,Phosphoproteins ,Proteome ,Signal Transduction ,Algorithms ,Male ,Transcriptome ,Prostatic Neoplasms ,Castration-Resistant ,Precision Medicine ,Prostatic Neoplasms ,Castration-Resistant ,Developmental Biology ,Biological Sciences ,Medical and Health Sciences - Abstract
We used clinical tissue from lethal metastatic castration-resistant prostate cancer (CRPC) patients obtained at rapid autopsy to evaluate diverse genomic, transcriptomic, and phosphoproteomic datasets for pathway analysis. Using Tied Diffusion through Interacting Events (TieDIE), we integrated differentially expressed master transcriptional regulators, functionally mutated genes, and differentially activated kinases in CRPC tissues to synthesize a robust signaling network consisting of druggable kinase pathways. Using MSigDB hallmark gene sets, six major signaling pathways with phosphorylation of several key residues were significantly enriched in CRPC tumors after incorporation of phosphoproteomic data. Individual autopsy profiles developed using these hallmarks revealed clinically relevant pathway information potentially suitable for patient stratification and targeted therapies in late stage prostate cancer. Here, we describe phosphorylation-based cancer hallmarks using integrated personalized signatures (pCHIPS) that shed light on the diversity of activated signaling pathways in metastatic CRPC while providing an integrative, pathway-based reference for drug prioritization in individual patients.
- Published
- 2016
45. Inferring causal molecular networks: empirical assessment through a community-based effort
- Author
-
Hill, Steven M, Heiser, Laura M, Cokelaer, Thomas, Unger, Michael, Nesser, Nicole K, Carlin, Daniel E, Zhang, Yang, Sokolov, Artem, Paull, Evan O, Wong, Chris K, Graim, Kiley, Bivol, Adrian, Wang, Haizhou, Zhu, Fan, Afsari, Bahman, Danilova, Ludmila V, Favorov, Alexander V, Lee, Wai Shing, Taylor, Dane, Hu, Chenyue W, Long, Byron L, Noren, David P, Bisberg, Alexander J, Mills, Gordon B, Gray, Joe W, Kellen, Michael, Norman, Thea, Friend, Stephen, Qutub, Amina A, Fertig, Elana J, Guan, Yuanfang, Song, Mingzhou, Stuart, Joshua M, Spellman, Paul T, Koeppl, Heinz, Stolovitzky, Gustavo, Saez-Rodriguez, Julio, and Mukherjee, Sach
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Networking and Information Technology R&D (NITRD) ,Bioengineering ,Cancer ,2.1 Biological and endogenous factors ,Generic health relevance ,Algorithms ,Causality ,Computational Biology ,Computer Simulation ,Gene Expression Profiling ,Gene Regulatory Networks ,Humans ,Models ,Biological ,Neoplasms ,Protein Interaction Mapping ,Signal Transduction ,Software ,Systems Biology ,Tumor Cells ,Cultured ,HPN-DREAM Consortium ,Technology ,Medical and Health Sciences ,Developmental Biology ,Biological sciences - Abstract
It remains unclear whether causal, rather than merely correlational, relationships in molecular networks can be inferred in complex biological settings. Here we describe the HPN-DREAM network inference challenge, which focused on learning causal influences in signaling networks. We used phosphoprotein data from cancer cell lines as well as in silico data from a nonlinear dynamical model. Using the phosphoprotein data, we scored more than 2,000 networks submitted by challenge participants. The networks spanned 32 biological contexts and were scored in terms of causal validity with respect to unseen interventional data. A number of approaches were effective, and incorporating known biology was generally advantageous. Additional sub-challenges considered time-course prediction and visualization. Our results suggest that learning causal relationships may be feasible in complex settings such as disease states. Furthermore, our scoring approach provides a practical way to empirically assess inferred molecular networks in a causal sense.
- Published
- 2016
46. N-Myc Drives Neuroendocrine Prostate Cancer Initiated from Human Prostate Epithelial Cells.
- Author
-
Lee, John K, Phillips, John W, Smith, Bryan A, Park, Jung Wook, Stoyanova, Tanya, McCaffrey, Erin F, Baertsch, Robert, Sokolov, Artem, Meyerowitz, Justin G, Mathis, Colleen, Cheng, Donghui, Stuart, Joshua M, Shokat, Kevan M, Gustafson, W Clay, Huang, Jiaoti, and Witte, Owen N
- Subjects
Cell Line ,Tumor ,Epithelial Cells ,Animals ,Mice ,Inbred NOD ,Humans ,Mice ,SCID ,Neuroendocrine Tumors ,Adenocarcinoma ,Prostatic Neoplasms ,Cell Transformation ,Neoplastic ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Phenylurea Compounds ,Azepines ,Pyrimidines ,Proto-Oncogene Proteins c-myc ,Neoplasm Proteins ,Recombinant Fusion Proteins ,Antineoplastic Agents ,Protein Kinase Inhibitors ,Orchiectomy ,Xenograft Model Antitumor Assays ,Transduction ,Genetic ,Gene Expression Regulation ,Neoplastic ,Enzyme Activation ,Genes ,myc ,Male ,Proto-Oncogene Proteins c-akt ,Neoplastic Stem Cells ,Molecular Targeted Therapy ,Exome ,Laser Capture Microdissection ,Aurora Kinase A ,Cell Line ,Tumor ,Mice ,Inbred NOD ,SCID ,Cell Transformation ,Neoplastic ,Transduction ,Genetic ,Gene Expression Regulation ,Genes ,myc ,Oncology & Carcinogenesis ,Oncology and Carcinogenesis ,Neurosciences - Abstract
MYCN amplification and overexpression are common in neuroendocrine prostate cancer (NEPC). However, the impact of aberrant N-Myc expression in prostate tumorigenesis and the cellular origin of NEPC have not been established. We define N-Myc and activated AKT1 as oncogenic components sufficient to transform human prostate epithelial cells to prostate adenocarcinoma and NEPC with phenotypic and molecular features of aggressive, late-stage human disease. We directly show that prostate adenocarcinoma and NEPC can arise from a common epithelial clone. Further, N-Myc is required for tumor maintenance, and destabilization of N-Myc through Aurora A kinase inhibition reduces tumor burden. Our findings establish N-Myc as a driver of NEPC and a target for therapeutic intervention.
- Published
- 2016
47. Pathway-Based Genomics Prediction using Generalized Elastic Net.
- Author
-
Sokolov, Artem, Carlin, Daniel E, Paull, Evan O, Baertsch, Robert, and Stuart, Joshua M
- Subjects
Animals ,Humans ,Proteome ,Chromosome Mapping ,Protein Interaction Mapping ,Signal Transduction ,Models ,Genetic ,Computer Simulation ,Pattern Recognition ,Automated ,Models ,Genetic ,Pattern Recognition ,Automated ,Mathematical Sciences ,Biological Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
We present a novel regularization scheme called The Generalized Elastic Net (GELnet) that incorporates gene pathway information into feature selection. The proposed formulation is applicable to a wide variety of problems in which the interpretation of predictive features using known molecular interactions is desired. The method naturally steers solutions toward sets of mechanistically interlinked genes. Using experiments on synthetic data, we demonstrate that pathway-guided results maintain, and often improve, the accuracy of predictors even in cases where the full gene network is unknown. We apply the method to predict the drug response of breast cancer cell lines. GELnet is able to reveal genetic determinants of sensitivity and resistance for several compounds. In particular, for an EGFR/HER2 inhibitor, it finds a possible trans-differentiation resistance mechanism missed by the corresponding pathway agnostic approach.
- Published
- 2016
48. ONE-CLASS DETECTION OF CELL STATES IN TUMOR SUBTYPES
- Author
-
Altman, Russ B, Dunker, A Keith, Hunter, Lawrence, Ritchie, Marylyn D, Murray, Tiffany A, Klein, Teri E, SOKOLOV, ARTEM, PAULL, EVAN O, and STUART, JOSHUA M
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Stem Cell Research ,Human Genome ,Breast Cancer ,Cancer ,Genetics ,Generic health relevance ,Good Health and Well Being ,Breast Neoplasms ,Computational Biology ,Embryonic Stem Cells ,Female ,Gene Expression Profiling ,Humans ,Logistic Models ,Neoplasms ,Neoplastic Stem Cells ,Precision Medicine ,Support Vector Machine ,Urinary Bladder Neoplasms ,One-class models ,Pan-Cancer - Abstract
The cellular composition of a tumor greatly influences the growth, spread, immune activity, drug response, and other aspects of the disease. Tumor cells are usually comprised of a heterogeneous mixture of subclones, each of which could contain their own distinct character. The presence of minor subclones poses a serious health risk for patients as any one of them could harbor a fitness advantage with respect to the current treatment regimen, fueling resistance. It is therefore vital to accurately assess the make-up of cell states within a tumor biopsy. Transcriptome-wide assays from RNA sequencing provide key data from which cell state signatures can be detected. However, the challenge is to find them within samples containing mixtures of cell types of unknown proportions. We propose a novel one-class method based on logistic regression and show that its performance is competitive to two established SVM-based methods for this detection task. We demonstrate that one-class models are able to identify specific cell types in heterogeneous cell populations better than their binary predictor counterparts. We derive one-class predictors for the major breast and bladder subtypes and reaffirm the connection between these two tissues. In addition, we use a one-class predictor to quantitatively associate an embryonic stem cell signature with an aggressive breast cancer subtype that reveals shared stemness pathways potentially important for treatment.
- Published
- 2016
49. Identifying Aspects of the Post-Transcriptional Program Governing the Proteome of the Green Alga Micromonas pusilla.
- Author
-
Waltman, Peter H, Guo, Jian, Reistetter, Emily Nahas, Purvine, Samuel, Ansong, Charles K, van Baren, Marijke J, Wong, Chee-Hong, Wei, Chia-Lin, Smith, Richard D, Callister, Stephen J, Stuart, Joshua M, and Worden, Alexandra Z
- Subjects
Algal Proteins ,RNA ,Algal ,RNA ,Messenger ,Codon ,3' Untranslated Regions ,Sequence Analysis ,RNA ,Proteomics ,Photosynthesis ,Protein Biosynthesis ,Transcription ,Genetic ,Gene Expression Regulation ,Plant ,Photoperiod ,Chlorophyta ,Molecular Sequence Annotation ,Gene Ontology ,Untranslated Regions ,Gene Expression Regulation ,Plant ,RNA ,Algal ,Messenger ,Sequence Analysis ,Transcription ,Genetic ,General Science & Technology - Abstract
Micromonas is a unicellular motile alga within the Prasinophyceae, a green algal group that is related to land plants. This picoeukaryote (
- Published
- 2016
50. A basal stem cell signature identifies aggressive prostate cancer phenotypes
- Author
-
Smith, Bryan A, Sokolov, Artem, Uzunangelov, Vladislav, Baertsch, Robert, Newton, Yulia, Graim, Kiley, Mathis, Colleen, Cheng, Donghui, Stuart, Joshua M, and Witte, Owen N
- Subjects
Cancer ,Aging ,Stem Cell Research - Nonembryonic - Non-Human ,Prostate Cancer ,Stem Cell Research ,Urologic Diseases ,Genetics ,Stem Cell Research - Nonembryonic - Human ,Aetiology ,2.1 Biological and endogenous factors ,Antigens ,CD ,Epithelial Cells ,Female ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Humans ,Male ,Mammary Glands ,Human ,Neoplasm Metastasis ,Neuroendocrine Tumors ,Phenotype ,Prostatic Neoplasms ,Proto-Oncogene Proteins c-myc ,Sequence Analysis ,RNA ,Signal Transduction ,Stem Cells ,Transcription Factors ,RNA-seq ,prostate cancer ,stem cell signature ,basal cell ,neuroendocrine prostate cancer - Abstract
Evidence from numerous cancers suggests that increased aggressiveness is accompanied by up-regulation of signaling pathways and acquisition of properties common to stem cells. It is unclear if different subtypes of late-stage cancer vary in stemness properties and whether or not these subtypes are transcriptionally similar to normal tissue stem cells. We report a gene signature specific for human prostate basal cells that is differentially enriched in various phenotypes of late-stage metastatic prostate cancer. We FACS-purified and transcriptionally profiled basal and luminal epithelial populations from the benign and cancerous regions of primary human prostates. High-throughput RNA sequencing showed the basal population to be defined by genes associated with stem cell signaling programs and invasiveness. Application of a 91-gene basal signature to gene expression datasets from patients with organ-confined or hormone-refractory metastatic prostate cancer revealed that metastatic small cell neuroendocrine carcinoma was molecularly more stem-like than either metastatic adenocarcinoma or organ-confined adenocarcinoma. Bioinformatic analysis of the basal cell and two human small cell gene signatures identified a set of E2F target genes common between prostate small cell neuroendocrine carcinoma and primary prostate basal cells. Taken together, our data suggest that aggressive prostate cancer shares a conserved transcriptional program with normal adult prostate basal stem cells.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.