3,484 results on '"Lawrence, Michael"'
Search Results
52. Prospective validation of ctHPVDNA for detection of minimal residual disease and prediction of recurrence in patients with HPV-associated head and neck cancer treated with surgery.
- Author
-
Hirayama, Shun, primary, Al-Inaya, Yana, additional, Aye, Ling, additional, Bryan, Michael E., additional, Das, Dipon, additional, Mendel, Julia, additional, Naegele, Saskia, additional, Faquin, William C, additional, Sadow, Peter, additional, Fisch, Adam S., additional, Lin, Derrick, additional, Varvares, Mark A, additional, Feng, Allen, additional, Emerick, Kevin S., additional, Deschler, Daniel G., additional, Lawrence, Michael S., additional, Iafrate, A. John, additional, Chan, Annie, additional, Richmon, Jeremy, additional, and Faden, Daniel, additional
- Published
- 2024
- Full Text
- View/download PDF
53. Multi-feature next-generation liquid biopsy for diagnosis and prognosis in HPV-associated head and neck cancer.
- Author
-
Aye, Ling, primary, Bryan, Michael E., additional, Das, Dipon, additional, Hirayama, Shun, additional, Al-Inaya, Yana, additional, Mendel, Julia, additional, Naegele, Saskia, additional, Fisch, Adam S., additional, Faquin, William C, additional, Sadow, Peter, additional, Chan, Annie, additional, Lawrence, Michael S., additional, Mirabello, Lisa, additional, Iafrate, John, additional, Waterboer, Tim, additional, Wirth, Lori J., additional, Richmon, Jeremy, additional, and Faden, Daniel, additional
- Published
- 2024
- Full Text
- View/download PDF
54. Advocacy in Sports Cardiology
- Author
-
Dineen, Elizabeth H., primary, Lawrence, Michael, additional, Husaini, Mustafa, additional, Danielian, Alfred, additional, Dean, Peter, additional, Davis, Lindsay, additional, Edmonds, Kenneth, additional, Chung, Eugene H., additional, Kim, Jonathan H., additional, and Phelan, Dermot M., additional
- Published
- 2024
- Full Text
- View/download PDF
55. The Immune Landscape of Cancer
- Author
-
Thorsson, Vésteinn, Gibbs, David L, Brown, Scott D, Wolf, Denise, Bortone, Dante S, Yang, Tai-Hsien Ou, Porta-Pardo, Eduard, Gao, Galen F, Plaisier, Christopher L, Eddy, James A, Ziv, Elad, Culhane, Aedin C, Paull, Evan O, Sivakumar, IK Ashok, Gentles, Andrew J, Malhotra, Raunaq, Farshidfar, Farshad, Colaprico, Antonio, Parker, Joel S, Mose, Lisle E, Vo, Nam Sy, Liu, Jianfang, Liu, Yuexin, Rader, Janet, Dhankani, Varsha, Reynolds, Sheila M, Bowlby, Reanne, Califano, Andrea, Cherniack, Andrew D, Anastassiou, Dimitris, Bedognetti, Davide, Mokrab, Younes, Newman, Aaron M, Rao, Arvind, Chen, Ken, Krasnitz, Alexander, Hu, Hai, Malta, Tathiane M, Noushmehr, Houtan, Pedamallu, Chandra Sekhar, Bullman, Susan, Ojesina, Akinyemi I, Lamb, Andrew, Zhou, Wanding, Shen, Hui, Choueiri, Toni K, Weinstein, John N, Guinney, Justin, Saltz, Joel, Holt, Robert A, Rabkin, Charles S, 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, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, and Broom, Bradley M
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Cancer Genome Atlas Research Network - Abstract
(Immunity 48, 812–830.e1–e14; April 17, 2018) In the originally published version of this article, the authors neglected to include Younes Mokrab and Aaron M. Newman as co-authors and misspelled the names of authors Charles S. Rabkin and Ilya Shmulevich. The author names have been corrected here and online. In addition, the concluding sentence of the subsection “Immune Signature Compilation” in the Method Details in the original published article was deemed unclear because it did not specify differences among the gene set scoring methods. The concluding sentences now reads “Gene sets from Bindea et al., Senbabaoglu et al., and the MSigDB C7 collection were scored using single-sample gene set enrichment (ssGSEA) analysis (Barbie et al., 2009), as implemented in the GSVA R package (Hänzelmann et al., 2013). All other signatures were scored using methods found in the associated citations.”
- Published
- 2019
56. Passenger hotspot mutations in cancer driven by APOBEC3A and mesoscale genomic features
- Author
-
Buisson, Rémi, Langenbucher, Adam, Bowen, Danae, Kwan, Eugene E, Benes, Cyril H, Zou, Lee, and Lawrence, Michael S
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Cancer ,Human Genome ,Genetics ,Biotechnology ,Stem Cell Research ,Cell Transformation ,Neoplastic ,Computational Biology ,Cytidine Deaminase ,Genomics ,HEK293 Cells ,Humans ,Mutation ,Neoplasms ,Proteins ,General Science & Technology - Abstract
Cancer drivers require statistical modeling to distinguish them from passenger events, which accumulate during tumorigenesis but provide no fitness advantage to cancer cells. The discovery of driver genes and mutations relies on the assumption that exact positional recurrence is unlikely by chance; thus, the precise sharing of mutations across patients identifies drivers. Examining the mutation landscape in cancer genomes, we found that many recurrent cancer mutations previously designated as drivers are likely passengers. Our integrated bioinformatic and biochemical analyses revealed that these passenger hotspot mutations arise from the preference of APOBEC3A, a cytidine deaminase, for DNA stem-loops. Conversely, recurrent APOBEC-signature mutations not in stem-loops are enriched in well-characterized driver genes and may predict new drivers. This demonstrates that mesoscale genomic features need to be integrated into computational models aimed at identifying mutations linked to diseases.
- Published
- 2019
57. Stromal Microenvironment Shapes the Intratumoral Architecture of Pancreatic Cancer
- Author
-
Ligorio, Matteo, Sil, Srinjoy, Malagon-Lopez, Jose, Nieman, Linda T, Misale, Sandra, Di Pilato, Mauro, Ebright, Richard Y, Karabacak, Murat N, Kulkarni, Anupriya S, Liu, Ann, Vincent Jordan, Nicole, Franses, Joseph W, Philipp, Julia, Kreuzer, Johannes, Desai, Niyati, Arora, Kshitij S, Rajurkar, Mihir, Horwitz, Elad, Neyaz, Azfar, Tai, Eric, Magnus, Neelima KC, Vo, Kevin D, Yashaswini, Chittampalli N, Marangoni, Francesco, Boukhali, Myriam, Fatherree, Jackson P, Damon, Leah J, Xega, Kristina, Desai, Rushil, Choz, Melissa, Bersani, Francesca, Langenbucher, Adam, Thapar, Vishal, Morris, Robert, Wellner, Ulrich F, Schilling, Oliver, Lawrence, Michael S, Liss, Andrew S, Rivera, Miguel N, Deshpande, Vikram, Benes, Cyril H, Maheswaran, Shyamala, Haber, Daniel A, Fernandez-Del-Castillo, Carlos, Ferrone, Cristina R, Haas, Wilhelm, Aryee, Martin J, and Ting, David T
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Biological Sciences ,Rare Diseases ,Genetics ,Digestive Diseases ,Stem Cell Research ,Pancreatic Cancer ,Cancer ,Stem Cell Research - Nonembryonic - Human ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Cancer-Associated Fibroblasts ,Carcinoma ,Pancreatic Ductal ,Cell Proliferation ,Coculture Techniques ,Epithelial-Mesenchymal Transition ,Female ,HEK293 Cells ,Heterografts ,Humans ,Mice ,Mice ,Inbred NOD ,Mice ,SCID ,Mitogen-Activated Protein Kinases ,Pancreatic Neoplasms ,RNA-Seq ,STAT3 Transcription Factor ,Stromal Cells ,Transfection ,Tumor Microenvironment ,mass spectrometry ,pancreatic cancer ,pancreatic ductal adenocarcinoma ,single cell RNA-sequencing ,single cell spatial analysis ,stromal microenvironment ,tumor architecture ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Single-cell technologies have described heterogeneity across tissues, but the spatial distribution and forces that drive single-cell phenotypes have not been well defined. Combining single-cell RNA and protein analytics in studying the role of stromal cancer-associated fibroblasts (CAFs) in modulating heterogeneity in pancreatic cancer (pancreatic ductal adenocarcinoma [PDAC]) model systems, we have identified significant single-cell population shifts toward invasive epithelial-to-mesenchymal transition (EMT) and proliferative (PRO) phenotypes linked with mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) signaling. Using high-content digital imaging of RNA in situ hybridization in 195 PDAC tumors, we quantified these EMT and PRO subpopulations in 319,626 individual cancer cells that can be classified within the context of distinct tumor gland "units." Tumor gland typing provided an additional layer of intratumoral heterogeneity that was associated with differences in stromal abundance and clinical outcomes. This demonstrates the impact of the stroma in shaping tumor architecture by altering inherent patterns of tumor glands in human PDAC.
- Published
- 2019
58. Next-generation characterization of the Cancer Cell Line Encyclopedia
- Author
-
Ghandi, Mahmoud, Huang, Franklin W, Jané-Valbuena, Judit, Kryukov, Gregory V, Lo, Christopher C, McDonald, E Robert, Barretina, Jordi, Gelfand, Ellen T, Bielski, Craig M, Li, Haoxin, Hu, Kevin, Andreev-Drakhlin, Alexander Y, Kim, Jaegil, Hess, Julian M, Haas, Brian J, Aguet, François, Weir, Barbara A, Rothberg, Michael V, Paolella, Brenton R, Lawrence, Michael S, Akbani, Rehan, Lu, Yiling, Tiv, Hong L, Gokhale, Prafulla C, de Weck, Antoine, Mansour, Ali Amin, Oh, Coyin, Shih, Juliann, Hadi, Kevin, Rosen, Yanay, Bistline, Jonathan, Venkatesan, Kavitha, Reddy, Anupama, Sonkin, Dmitriy, Liu, Manway, Lehar, Joseph, Korn, Joshua M, Porter, Dale A, Jones, Michael D, Golji, Javad, Caponigro, Giordano, Taylor, Jordan E, Dunning, Caitlin M, Creech, Amanda L, Warren, Allison C, McFarland, James M, Zamanighomi, Mahdi, Kauffmann, Audrey, Stransky, Nicolas, Imielinski, Marcin, Maruvka, Yosef E, Cherniack, Andrew D, Tsherniak, Aviad, Vazquez, Francisca, Jaffe, Jacob D, Lane, Andrew A, Weinstock, David M, Johannessen, Cory M, Morrissey, Michael P, Stegmeier, Frank, Schlegel, Robert, Hahn, William C, Getz, Gad, Mills, Gordon B, Boehm, Jesse S, Golub, Todd R, Garraway, Levi A, and Sellers, William R
- Subjects
Genetics ,Human Genome ,Lung ,Cancer ,Biotechnology ,Lung Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Antineoplastic Agents ,Biomarkers ,Tumor ,Cell Line ,Tumor ,DNA Methylation ,Drug Resistance ,Neoplasm ,Ethnicity ,Gene Editing ,Histones ,Humans ,MicroRNAs ,Molecular Targeted Therapy ,Neoplasms ,Protein Array Analysis ,RNA Splicing ,General Science & Technology - Abstract
Large panels of comprehensively characterized human cancer models, including the Cancer Cell Line Encyclopedia (CCLE), have provided a rigorous framework with which to study genetic variants, candidate targets, and small-molecule and biological therapeutics and to identify new marker-driven cancer dependencies. To improve our understanding of the molecular features that contribute to cancer phenotypes, including drug responses, here we have expanded the characterizations of cancer cell lines to include genetic, RNA splicing, DNA methylation, histone H3 modification, microRNA expression and reverse-phase protein array data for 1,072 cell lines from individuals of various lineages and ethnicities. Integration of these data with functional characterizations such as drug-sensitivity, short hairpin RNA knockdown and CRISPR-Cas9 knockout data reveals potential targets for cancer drugs and associated biomarkers. Together, this dataset and an accompanying public data portal provide a resource for the acceleration of cancer research using model cancer cell lines.
- Published
- 2019
59. Insertion of a synthetic switch into insulin provides metabolite-dependent regulation of hormone–receptor activation
- Author
-
Chen, Yen-Shan, Gleaton, Jeremy, Yang, Yanwu, Dhayalan, Balamurugan, Phillips, Nelson B., Liu, Yule, Broadwater, Laurie, Jarosinski, Mark A., Chatterjee, Deepak, Lawrence, Michael C., Hattier, Thomas, Michael, M. Dodson, and Weiss, Michael A.
- Published
- 2021
60. Symmetric and asymmetric receptor conformation continuum induced by a new insulin
- Author
-
Xiong, Xiaochun, Blakely, Alan, Kim, Jin Hwan, Menting, John G., Schäfer, Ingmar B., Schubert, Heidi L., Agrawal, Rahul, Gutmann, Theresia, Delaine, Carlie, Zhang, Yi Wolf, Artik, Gizem Olay, Merriman, Allanah, Eckert, Debbie, Lawrence, Michael C., Coskun, Ünal, Fisher, Simon J., Forbes, Briony E., Safavi-Hemami, Helena, Hill, Christopher P., and Chou, Danny Hung-Chieh
- Published
- 2022
- Full Text
- View/download PDF
61. Single-cell transcriptomic profiling for inferring tumor origin and mechanisms of therapeutic resistance
- Author
-
Lin, Maoxuan, Sade-Feldman, Moshe, Wirth, Lori, Lawrence, Michael S., and Faden, Daniel L.
- Published
- 2022
- Full Text
- View/download PDF
62. Activation of the human insulin receptor by non-insulin-related peptides
- Author
-
Kirk, Nicholas S., Chen, Qi, Wu, Yingzhe Ginger, Asante, Anastasia L., Hu, Haitao, Espinosa, Juan F., Martínez-Olid, Francisco, Margetts, Mai B., Mohammed, Faiz A., Kiselyov, Vladislav V., Barrett, David G., and Lawrence, Michael C.
- Published
- 2022
- Full Text
- View/download PDF
63. Interaction of a viral insulin-like peptide with the IGF-1 receptor produces a natural antagonist
- Author
-
Moreau, Francois, Kirk, Nicholas S., Zhang, Fa, Gelfanov, Vasily, List, Edward O., Chrudinová, Martina, Venugopal, Hari, Lawrence, Michael C., Jimenez, Veronica, Bosch, Fatima, Kopchick, John J., DiMarchi, Richard D., Altindis, Emrah, and Ronald Kahn, C.
- Published
- 2022
- Full Text
- View/download PDF
64. 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
65. trackr: A Framework for Enhancing Discoverability and Reproducibility of Data Visualizations and Other Artifacts in R
- Author
-
Becker, Gabriel, Moore, Sara E., and Lawrence, Michael
- Subjects
Statistics - Computation - Abstract
Research is an incremental, iterative process, with new results relying and building upon previous ones. Scientists need to find, retrieve, understand, and verify results in order to confidently extend them, even when the results are their own. We present the trackr framework for organizing, automatically annotating, discovering, and retrieving results. We identify sources of automatically extractable metadata for computational results, and we define an extensible system for organizing, annotating, and searching for results based on these and other metadata. We present an open-source implementation of these concepts for plots, computational artifacts, and woven dynamic reports generated in the R statistical computing language.
- Published
- 2017
- Full Text
- View/download PDF
66. Pancreatic Fibrosis Assessment Using Picosirius Red Staining in Chronic Pancreatitis Patients Undergoing Total Pancreatectomy with Islet Autotransplantation
- Author
-
Liu, Yang, Vasu, Srividya, Kumano, Kenjiro, SoRelle, Jeffrey A., Lawrence, Michael C., and Naziruddin, Bashoo
- Published
- 2022
- Full Text
- View/download PDF
67. A Pan-Cancer Analysis Reveals High-Frequency Genetic Alterations in Mediators of Signaling by the TGF-β Superfamily
- Author
-
Korkut, Anil, Zaidi, Sobia, Kanchi, Rupa S, Rao, Shuyun, Gough, Nancy R, Schultz, Andre, Li, Xubin, Lorenzi, Philip L, Berger, Ashton C, Robertson, Gordon, Kwong, Lawrence N, Datto, Mike, Roszik, Jason, Ling, Shiyun, Ravikumar, Visweswaran, Manyam, Ganiraju, Rao, Arvind, Shelley, Simon, Liu, Yuexin, Ju, Zhenlin, Hansel, Donna, de Velasco, Guillermo, Pennathur, Arjun, Andersen, Jesper B, O'Rourke, Colm J, Ohshiro, Kazufumi, Jogunoori, Wilma, Nguyen, Bao-Ngoc, Li, Shulin, Osmanbeyoglu, Hatice U, Ajani, Jaffer A, Mani, Sendurai A, Houseman, Andres, Wiznerowicz, Maciej, Chen, Jian, Gu, Shoujun, Ma, Wencai, Zhang, Jiexin, Tong, Pan, Cherniack, Andrew D, Deng, Chuxia, Resar, Linda, 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, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, Broom, Bradley M, Hegde, Apurva M, Li, Jun, Liang, Han, Liu, Wenbin, and Lu, Yiling
- Subjects
Biological Sciences ,Genetics ,Cancer ,Biotechnology ,Human Genome ,Cancer Genomics ,2.1 Biological and endogenous factors ,Bone Morphogenetic Protein 5 ,DNA Methylation ,Humans ,MicroRNAs ,Mutation Rate ,Neoplasms ,Receptor ,Transforming Growth Factor-beta Type I ,Signal Transduction ,Smad Proteins ,Transforming Growth Factor beta ,Cancer Genome Atlas Research Network ,DNA methylation ,Pan-Cancer ,TCGA ,TGF-β ,TGF-β pathway ,The Cancer Genome Atlas ,cancer ,microRNA ,mutation hotspot ,transcription ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
We present an integromic analysis of gene alterations that modulate transforming growth factor β (TGF-β)-Smad-mediated signaling in 9,125 tumor samples across 33 cancer types in The Cancer Genome Atlas (TCGA). Focusing on genes that encode mediators and regulators of TGF-β signaling, we found at least one genomic alteration (mutation, homozygous deletion, or amplification) in 39% of samples, with highest frequencies in gastrointestinal cancers. We identified mutation hotspots in genes that encode TGF-β ligands (BMP5), receptors (TGFBR2, AVCR2A, and BMPR2), and Smads (SMAD2 and SMAD4). Alterations in the TGF-β superfamily correlated positively with expression of metastasis-associated genes and with decreased survival. Correlation analyses showed the contributions of mutation, amplification, deletion, DNA methylation, and miRNA expression to transcriptional activity of TGF-β signaling in each cancer type. This study provides a broad molecular perspective relevant for future functional and therapeutic studies of the diverse cancer pathways mediated by the TGF-β superfamily.
- Published
- 2018
68. 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
69. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma
- Author
-
Ricketts, Christopher J, De Cubas, Aguirre A, Fan, Huihui, Smith, Christof C, Lang, Martin, Reznik, Ed, Bowlby, Reanne, Gibb, Ewan A, Akbani, Rehan, Beroukhim, Rameen, Bottaro, Donald P, Choueiri, Toni K, Gibbs, Richard A, Godwin, Andrew K, Haake, Scott, Hakimi, A Ari, Henske, Elizabeth P, Hsieh, James J, Ho, Thai H, Kanchi, Rupa S, Krishnan, Bhavani, Kwiatkowski, David J, Lui, Wembin, Merino, Maria J, Mills, Gordon B, Myers, Jerome, Nickerson, Michael L, Reuter, Victor E, Schmidt, Laura S, Shelley, C Simon, Shen, Hui, Shuch, Brian, Signoretti, Sabina, Srinivasan, Ramaprasad, Tamboli, Pheroze, Thomas, George, Vincent, Benjamin G, Vocke, Cathy D, Wheeler, David A, Yang, Lixing, Kim, William Y, Robertson, A Gordon, Spellman, Paul, Rathmell, W Kimryn, Linehan, W Marston, 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, Broom, Bradley M, Hegde, Apurva M, Ju, Zhenlin, Korkut, Anil, Li, Jun, Liang, Han, and Ling, Shiyun
- Subjects
Biological Sciences ,Cancer Genome Atlas Research Network ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
(Cell Reports 23, 313–326; April 3, 2018) In the originally published version of this article, the author list contained two errors. Specifically, David J. Kwiatkowski was misspelled as David J. Kwaitkowski, and William Y. Kim was inadvertently written as William T. Kim. Both names have been corrected online. The authors regret this error.
- Published
- 2018
70. Pan-Cancer Analysis of lncRNA Regulation Supports Their Targeting of Cancer Genes in Each Tumor Context
- Author
-
Chiu, Hua-Sheng, Somvanshi, Sonal, Patel, Ektaben, Chen, Ting-Wen, Singh, Vivek P, Zorman, Barry, Patil, Sagar L, Pan, Yinghong, Chatterjee, Sujash S, 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, Weinstein, John N, 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, and Anur, Pavana
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Breast Cancer ,Human Genome ,Cancer ,Biotechnology ,Women's Health ,Genetics ,Cancer Genomics ,2.1 Biological and endogenous factors ,Cell Line ,Cell Line ,Tumor ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Genes ,Tumor Suppressor ,Humans ,Neoplasms ,Oncogenes ,RNA ,Long Noncoding ,Cancer Genome Atlas Research Network ,RNA-binding proteins ,cancer gene ,interactome ,lncRNA ,microRNA ,modulation ,noncoding RNA ,pan-cancer ,regulation ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Long noncoding RNAs (lncRNAs) are commonly dysregulated in tumors, but only a handful are known to play pathophysiological roles in cancer. We inferred lncRNAs that dysregulate cancer pathways, oncogenes, and tumor suppressors (cancer genes) by modeling their effects on the activity of transcription factors, RNA-binding proteins, and microRNAs in 5,185 TCGA tumors and 1,019 ENCODE assays. Our predictions included hundreds of candidate onco- and tumor-suppressor lncRNAs (cancer lncRNAs) whose somatic alterations account for the dysregulation of dozens of cancer genes and pathways in each of 14 tumor contexts. To demonstrate proof of concept, we showed that perturbations targeting OIP5-AS1 (an inferred tumor suppressor) and TUG1 and WT1-AS (inferred onco-lncRNAs) dysregulated cancer genes and altered proliferation of breast and gynecologic cancer cells. Our analysis indicates that, although most lncRNAs are dysregulated in a tumor-specific manner, some, including OIP5-AS1, TUG1, NEAT1, MEG3, and TSIX, synergistically dysregulate cancer pathways in multiple tumor contexts.
- Published
- 2018
71. Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences across 33 Cancer Types
- Author
-
Seiler, Michael, Peng, Shouyong, Agrawal, Anant A, Palacino, James, Teng, Teng, Zhu, Ping, Smith, Peter G, 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, Weinstein, John N, 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, and Spellman, Paul
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer Genomics ,Human Genome ,Cancer ,2.1 Biological and endogenous factors ,Cell Line ,Tumor ,Genes ,Tumor Suppressor ,Humans ,Loss of Function Mutation ,Mutation Rate ,Neoplasms ,Oncogenes ,RNA Splicing ,RNA Splicing Factors ,Cancer Genome Atlas Research Network ,FUBP1 ,RBM10 ,SF3B1 ,SRSF2 ,U2AF1 ,cancer ,mutation ,splicing ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Hotspot mutations in splicing factor genes have been recently reported at high frequency in hematological malignancies, suggesting the importance of RNA splicing in cancer. We analyzed whole-exome sequencing data across 33 tumor types in The Cancer Genome Atlas (TCGA), and we identified 119 splicing factor genes with significant non-silent mutation patterns, including mutation over-representation, recurrent loss of function (tumor suppressor-like), or hotspot mutation profile (oncogene-like). Furthermore, RNA sequencing analysis revealed altered splicing events associated with selected splicing factor mutations. In addition, we were able to identify common gene pathway profiles associated with the presence of these mutations. Our analysis suggests that somatic alteration of genes involved in the RNA-splicing process is common in cancer and may represent an underappreciated hallmark of tumorigenesis.
- Published
- 2018
72. Integrated Genomic Analysis of the Ubiquitin Pathway across Cancer Types
- Author
-
Ge, Zhongqi, Leighton, Jake S, Wang, Yumeng, Peng, Xinxin, Chen, Zhongyuan, Chen, Hu, Sun, Yutong, Yao, Fan, Li, Jun, Zhang, Huiwen, Liu, Jianfang, Shriver, Craig D, Hu, Hai, 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, Liang, Han, Ling, Shiyun, Liu, Wenbin, Lu, Yiling, Mills, Gordon B, Ng, Kwok-Shing, Rao, Arvind, Ryan, Michael, Wang, Jing, Weinstein, John N, 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, and Sun, Yichao
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Human Genome ,Genetics ,Biotechnology ,Cancer Genomics ,Cancer ,Good Health and Well Being ,Cell Line ,Tumor ,Gene Expression Regulation ,Neoplastic ,Genome ,Human ,Genomics ,Humans ,Metabolic Networks and Pathways ,Neoplasms ,Oncogene Proteins ,Ubiquitination ,Cancer Genome Atlas Research Network ,FBXW7 ,The Cancer Genome Atlas ,biomarker ,cancer prognosis ,pan-cancer analysis ,therapeutic targets ,tumor subtype ,ubiquitin pathway ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Protein ubiquitination is a dynamic and reversible process of adding single ubiquitin molecules or various ubiquitin chains to target proteins. Here, using multidimensional omic data of 9,125 tumor samples across 33 cancer types from The Cancer Genome Atlas, we perform comprehensive molecular characterization of 929 ubiquitin-related genes and 95 deubiquitinase genes. Among them, we systematically identify top somatic driver candidates, including mutated FBXW7 with cancer-type-specific patterns and amplified MDM2 showing a mutually exclusive pattern with BRAF mutations. Ubiquitin pathway genes tend to be upregulated in cancer mediated by diverse mechanisms. By integrating pan-cancer multiomic data, we identify a group of tumor samples that exhibit worse prognosis. These samples are consistently associated with the upregulation of cell-cycle and DNA repair pathways, characterized by mutated TP53, MYC/TERT amplification, and APC/PTEN deletion. Our analysis highlights the importance of the ubiquitin pathway in cancer development and lays a foundation for developing relevant therapeutic strategies.
- Published
- 2018
73. lncRNA Epigenetic Landscape Analysis Identifies EPIC1 as an Oncogenic lncRNA that Interacts with MYC and Promotes Cell-Cycle Progression in Cancer
- Author
-
Wang, Zehua, Yang, Bo, Zhang, Min, Guo, Weiwei, Wu, Zhiyuan, Wang, Yue, Jia, Lin, Li, Song, 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, Weinstein, John N, Zhang, Jiexin, Abeshouse, Adam, Armenia, Joshua, Chakravarty, Debyani, Chatila, Walid K, Bruijn, Inode, 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, and Peto, Myron
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Biological Sciences ,Cancer Genomics ,Human Genome ,Genetics ,Cancer ,Women's Health ,Breast Cancer ,Animals ,Binding Sites ,Breast Neoplasms ,Cell Cycle ,Cell Line ,Tumor ,CpG Islands ,DNA Methylation ,Epigenesis ,Genetic ,Female ,Gene Expression Regulation ,Neoplastic ,Humans ,Mice ,Neoplasm Transplantation ,Prognosis ,Promoter Regions ,Genetic ,Proto-Oncogene Proteins c-myc ,RNA ,Long Noncoding ,Up-Regulation ,Cancer Genome Atlas Research Network ,CIMP ,ENSG00000224271 ,EPIC1 ,LOC284930 ,MYC ,P21 ,TCGA pan-cancer ,breast cancer ,long noncoding RNA ,Neurosciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
We characterized the epigenetic landscape of genes encoding long noncoding RNAs (lncRNAs) across 6,475 tumors and 455 cancer cell lines. In stark contrast to the CpG island hypermethylation phenotype in cancer, we observed a recurrent hypomethylation of 1,006 lncRNA genes in cancer, including EPIC1 (epigenetically-induced lncRNA1). Overexpression of EPIC1 is associated with poor prognosis in luminal B breast cancer patients and enhances tumor growth in vitro and in vivo. Mechanistically, EPIC1 promotes cell-cycle progression by interacting with MYC through EPIC1's 129-283 nt region. EPIC1 knockdown reduces the occupancy of MYC to its target genes (e.g., CDKN1A, CCNA2, CDC20, and CDC45). MYC depletion abolishes EPIC1's regulation of MYC target and luminal breast cancer tumorigenesis in vitro and in vivo.
- Published
- 2018
74. Comparative Molecular Analysis of Gastrointestinal Adenocarcinomas
- Author
-
Liu, Yang, Sethi, Nilay S, Hinoue, Toshinori, Schneider, Barbara G, Cherniack, Andrew D, Sanchez-Vega, Francisco, Seoane, Jose A, Farshidfar, Farshad, Bowlby, Reanne, Islam, Mirazul, Kim, Jaegil, Chatila, Walid, Akbani, Rehan, Kanchi, Rupa S, Rabkin, Charles S, Willis, Joseph E, Wang, Kenneth K, McCall, Shannon J, Mishra, Lopa, Ojesina, Akinyemi I, Bullman, Susan, Pedamallu, Chandra Sekhar, Lazar, Alexander J, Sakai, Ryo, 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, 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, Broom, Bradley M, Hegde, Apurva M, Ju, Zhenlin, Korkut, Anil, Li, Jun, Liang, Han, Ling, Shiyun, Liu, Wenbin, Lu, Yiling, Mills, Gordon B, Ng, Kwok-Shing, Rao, Arvind, Ryan, Michael, Wang, Jing, Weinstein, John N, Zhang, Jiexin, Abeshouse, Adam, Armenia, Joshua, Chakravarty, Debyani, Chatila, Walid K, Bruijn, Inode, Gao, Jianjiong, Gross, Benjamin E, Heins, Zachary J, Kundra, Ritika, La, Konnor, and Ladanyi, Marc
- Subjects
Biological Sciences ,Genetics ,Cancer ,Human Genome ,Rare Diseases ,Genetic Testing ,Biotechnology ,Cancer Genomics ,Colo-Rectal Cancer ,Digestive Diseases ,Adenocarcinoma ,Aneuploidy ,Chromosomal Instability ,DNA Methylation ,DNA Polymerase II ,DNA-Binding Proteins ,Epigenesis ,Genetic ,Female ,Gastrointestinal Neoplasms ,Gene Regulatory Networks ,Heterogeneous-Nuclear Ribonucleoproteins ,Humans ,Male ,Microsatellite Instability ,MutL Protein Homolog 1 ,Mutation ,Poly-ADP-Ribose Binding Proteins ,Polymorphism ,Single Nucleotide ,Proto-Oncogene Proteins p21(ras) ,RNA-Binding Proteins ,SOX9 Transcription Factor ,Cancer Genome Atlas Research Network ,cancer ,colon ,colorectal ,epigenetic ,esophagus ,genomic ,methylation ,rectum ,stomach ,tumor ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
We analyzed 921 adenocarcinomas of the esophagus, stomach, colon, and rectum to examine shared and distinguishing molecular characteristics of gastrointestinal tract adenocarcinomas (GIACs). Hypermutated tumors were distinct regardless of cancer type and comprised those enriched for insertions/deletions, representing microsatellite instability cases with epigenetic silencing of MLH1 in the context of CpG island methylator phenotype, plus tumors with elevated single-nucleotide variants associated with mutations in POLE. Tumors with chromosomal instability were diverse, with gastroesophageal adenocarcinomas harboring fragmented genomes associated with genomic doubling and distinct mutational signatures. We identified a group of tumors in the colon and rectum lacking hypermutation and aneuploidy termed genome stable and enriched in DNA hypermethylation and mutations in KRAS, SOX9, and PCBP1.
- Published
- 2018
75. 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
76. Genomic and Functional Approaches to Understanding Cancer Aneuploidy
- Author
-
Taylor, Alison M, Shih, Juliann, Ha, Gavin, Gao, Galen F, Zhang, Xiaoyang, Berger, Ashton C, Schumacher, Steven E, Wang, Chen, Hu, Hai, Liu, Jianfang, Lazar, Alexander J, 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, Weinstein, John N, 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, and Wang, Jioajiao
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Bioinformatics and Computational Biology ,Genetics ,Oncology and Carcinogenesis ,Cancer Genomics ,Human Genome ,Cancer ,Lung ,Lung Cancer ,2.1 Biological and endogenous factors ,Aneuploidy ,Carcinoma ,Squamous Cell ,Cell Cycle ,Cell Proliferation ,Chromosome Aberrations ,Chromosome Deletion ,Chromosomes ,Human ,Pair 3 ,Databases ,Genetic ,Genomics ,Humans ,Mutation Rate ,Tumor Suppressor Protein p53 ,Cancer Genome Atlas Research Network ,aneuploidy ,cancer genomics ,genome engineering ,lung squamous cell carcinoma ,Neurosciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Aneuploidy, whole chromosome or chromosome arm imbalance, is a near-universal characteristic of human cancers. In 10,522 cancer genomes from The Cancer Genome Atlas, aneuploidy was correlated with TP53 mutation, somatic mutation rate, and expression of proliferation genes. Aneuploidy was anti-correlated with expression of immune signaling genes, due to decreased leukocyte infiltrates in high-aneuploidy samples. Chromosome arm-level alterations show cancer-specific patterns, including loss of chromosome arm 3p in squamous cancers. We applied genome engineering to delete 3p in lung cells, causing decreased proliferation rescued in part by chromosome 3 duplication. This study defines genomic and phenotypic correlates of cancer aneuploidy and provides an experimental approach to study chromosome arm aneuploidy.
- Published
- 2018
77. Spatial Organization and Molecular Correlation of Tumor-Infiltrating Lymphocytes Using Deep Learning on Pathology Images
- Author
-
Saltz, Joel, Gupta, Rajarsi, Hou, Le, Kurc, Tahsin, Singh, Pankaj, Nguyen, Vu, Samaras, Dimitris, Shroyer, Kenneth R, Zhao, Tianhao, Batiste, Rebecca, Van Arnam, John, 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, Weinstein, John N, 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, and Wang, Jioajiao
- Subjects
Biological Sciences ,Cancer ,Genetics ,Human Genome ,Networking and Information Technology R&D (NITRD) ,Machine Learning and Artificial Intelligence ,Good Health and Well Being ,Deep Learning ,Humans ,Image Interpretation ,Computer-Assisted ,Lymphocytes ,Tumor-Infiltrating ,Neoplasms ,Cancer Genome Atlas Research Network ,artificial intelligence ,bioinformatics ,computer vision ,deep learning ,digital pathology ,immuno-oncology ,lymphocytes ,machine learning ,tumor microenvironment ,tumor-infiltrating lymphocytes ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Beyond sample curation and basic pathologic characterization, the digitized H&E-stained images of TCGA samples remain underutilized. To highlight this resource, we present mappings of tumor-infiltrating lymphocytes (TILs) based on H&E images from 13 TCGA tumor types. These TIL maps are derived through computational staining using a convolutional neural network trained to classify patches of images. Affinity propagation revealed local spatial structure in TIL patterns and correlation with overall survival. TIL map structural patterns were grouped using standard histopathological parameters. These patterns are enriched in particular T cell subpopulations derived from molecular measures. TIL densities and spatial structure were differentially enriched among tumor types, immune subtypes, and tumor molecular subtypes, implying that spatial infiltrate state could reflect particular tumor cell aberration states. Obtaining spatial lymphocytic patterns linked to the rich genomic characterization of TCGA samples demonstrates one use for the TCGA image archives with insights into the tumor-immune microenvironment.
- Published
- 2018
78. Genomic, Pathway Network, and Immunologic Features Distinguishing Squamous Carcinomas
- Author
-
Campbell, Joshua D, Yau, Christina, Bowlby, Reanne, Liu, Yuexin, Brennan, Kevin, Fan, Huihui, Taylor, Alison M, Wang, Chen, Walter, Vonn, Akbani, Rehan, Byers, Lauren Averett, Creighton, Chad J, Coarfa, Cristian, Shih, Juliann, Cherniack, Andrew D, Gevaert, Olivier, Prunello, Marcos, Shen, Hui, Anur, Pavana, Chen, Jianhong, Cheng, Hui, Hayes, D Neil, Bullman, Susan, Pedamallu, Chandra Sekhar, Ojesina, Akinyemi I, Sadeghi, Sara, Mungall, Karen L, Robertson, A Gordon, Benz, Christopher, Schultz, Andre, Kanchi, Rupa S, Gay, Carl M, Hegde, Apurva, Diao, Lixia, Wang, Jing, Ma, Wencai, Sumazin, Pavel, Chiu, Hua-Sheng, Chen, Ting-Wen, Gunaratne, Preethi, Donehower, Larry, Rader, Janet S, Zuna, Rosemary, Al-Ahmadie, Hikmat, Lazar, Alexander J, Flores, Elsa R, Tsai, Kenneth Y, Zhou, Jane H, Rustgi, Anil K, Drill, Esther, Shen, Ronglei, Wong, Christopher K, 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, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, and Zhang, Wei
- Subjects
Biological Sciences ,Sexually Transmitted Infections ,Infectious Diseases ,Human Genome ,Genetics ,Biotechnology ,Cancer Genomics ,Cancer ,2.1 Biological and endogenous factors ,Carcinoma ,Squamous Cell ,Cell Line ,Tumor ,DNA Methylation ,Epithelial-Mesenchymal Transition ,Gene Expression Regulation ,Neoplastic ,Genomics ,Humans ,Metabolic Networks and Pathways ,Polymorphism ,Genetic ,Cancer Genome Atlas Research Network ,bladder carcinoma with squamous differentiation ,cervical squamous cell carcinoma ,esophageal squamous cell carcinoma ,genomics ,head and neck squamous cell carcinoma ,human papillomavirus ,lung squamous cell carcinoma ,proteomics ,transcriptomics ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
This integrated, multiplatform PanCancer Atlas study co-mapped and identified distinguishing molecular features of squamous cell carcinomas (SCCs) from five sites associated with smoking and/or human papillomavirus (HPV). SCCs harbor 3q, 5p, and other recurrent chromosomal copy-number alterations (CNAs), DNA mutations, and/or aberrant methylation of genes and microRNAs, which are correlated with the expression of multi-gene programs linked to squamous cell stemness, epithelial-to-mesenchymal differentiation, growth, genomic integrity, oxidative damage, death, and inflammation. Low-CNA SCCs tended to be HPV(+) and display hypermethylation with repression of TET1 demethylase and FANCF, previously linked to predisposition to SCC, or harbor mutations affecting CASP8, RAS-MAPK pathways, chromatin modifiers, and immunoregulatory molecules. We uncovered hypomethylation of the alternative promoter that drives expression of the ΔNp63 oncogene and embedded miR944. Co-expression of immune checkpoint, T-regulatory, and Myeloid suppressor cells signatures may explain reduced efficacy of immune therapy. These findings support possibilities for molecular classification and therapeutic approaches.
- Published
- 2018
79. The Immune Landscape of Cancer
- Author
-
Thorsson, Vésteinn, Gibbs, David L, Brown, Scott D, Wolf, Denise, Bortone, Dante S, Ou Yang, Tai-Hsien, Porta-Pardo, Eduard, Gao, Galen F, Plaisier, Christopher L, Eddy, James A, Ziv, Elad, Culhane, Aedin C, Paull, Evan O, Sivakumar, IK Ashok, Gentles, Andrew J, Malhotra, Raunaq, Farshidfar, Farshad, Colaprico, Antonio, Parker, Joel S, Mose, Lisle E, Vo, Nam Sy, Liu, Jianfang, Liu, Yuexin, Rader, Janet, Dhankani, Varsha, Reynolds, Sheila M, Bowlby, Reanne, Califano, Andrea, Cherniack, Andrew D, Anastassiou, Dimitris, Bedognetti, Davide, Mokrab, Younes, Newman, Aaron M, Rao, Arvind, Chen, Ken, Krasnitz, Alexander, Hu, Hai, Malta, Tathiane M, Noushmehr, Houtan, Pedamallu, Chandra Sekhar, Bullman, Susan, Ojesina, Akinyemi I, Lamb, Andrew, Zhou, Wanding, Shen, Hui, Choueiri, Toni K, Weinstein, John N, Guinney, Justin, Saltz, Joel, Holt, Robert A, Rabkin, Charles S, Lazar, Alexander J, Serody, Jonathan S, Demicco, Elizabeth G, Disis, Mary L, Vincent, Benjamin G, Shmulevich, Ilya, 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 Julia, 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, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Miller, Michael, and Reynolds, Sheila
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Immunotherapy ,Genetics ,Human Genome ,Cancer ,Cancer Genomics ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Child ,Female ,Genomics ,Humans ,Interferon-gamma ,Macrophages ,Male ,Middle Aged ,Neoplasms ,Prognosis ,Th1-Th2 Balance ,Transforming Growth Factor beta ,Wound Healing ,Young Adult ,Cancer Genome Atlas Research Network ,cancer genomics ,immune subtypes ,immuno-oncology ,immunomodulatory ,immunotherapy ,integrative network analysis ,tumor immunology ,tumor microenvironment - Abstract
We performed an extensive immunogenomic analysis of more than 10,000 tumors comprising 33 diverse cancer types by utilizing data compiled by TCGA. Across cancer types, we identified six immune subtypes-wound healing, IFN-γ dominant, inflammatory, lymphocyte depleted, immunologically quiet, and TGF-β dominant-characterized by differences in macrophage or lymphocyte signatures, Th1:Th2 cell ratio, extent of intratumoral heterogeneity, aneuploidy, extent of neoantigen load, overall cell proliferation, expression of immunomodulatory genes, and prognosis. Specific driver mutations correlated with lower (CTNNB1, NRAS, or IDH1) or higher (BRAF, TP53, or CASP8) leukocyte levels across all cancers. Multiple control modalities of the intracellular and extracellular networks (transcription, microRNAs, copy number, and epigenetic processes) were involved in tumor-immune cell interactions, both across and within immune subtypes. Our immunogenomics pipeline to characterize these heterogeneous tumors and the resulting data are intended to serve as a resource for future targeted studies to further advance the field.
- Published
- 2018
80. Oncogenic Signaling Pathways in The Cancer Genome Atlas
- Author
-
Sanchez-Vega, Francisco, Mina, Marco, Armenia, Joshua, Chatila, Walid K, Luna, Augustin, La, Konnor C, Dimitriadoy, Sofia, Liu, David L, Kantheti, Havish S, Saghafinia, Sadegh, Chakravarty, Debyani, Daian, Foysal, Gao, Qingsong, Bailey, Matthew H, Liang, Wen-Wei, Foltz, Steven M, Shmulevich, Ilya, Ding, Li, Heins, Zachary, Ochoa, Angelica, Gross, Benjamin, Gao, Jianjiong, Zhang, Hongxin, Kundra, Ritika, Kandoth, Cyriac, Bahceci, Istemi, Dervishi, Leonard, Dogrusoz, Ugur, Zhou, Wanding, Shen, Hui, Laird, Peter W, Way, Gregory P, Greene, Casey S, Liang, Han, Xiao, Yonghong, Wang, Chen, Iavarone, Antonio, Berger, Alice H, Bivona, Trever G, Lazar, Alexander J, Hammer, Gary D, Giordano, Thomas, Kwong, Lawrence N, McArthur, Grant, Huang, Chenfei, Tward, Aaron D, Frederick, Mitchell J, McCormick, Frank, Meyerson, Matthew, 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, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, Broom, Bradley M, Hegde, Apurva M, and Ju, Zhenlin
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Oncology and Carcinogenesis ,Cancer Genomics ,Human Genome ,Cancer ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Databases ,Genetic ,Genes ,Neoplasm ,Humans ,Neoplasms ,Phosphatidylinositol 3-Kinases ,Signal Transduction ,Transforming Growth Factor beta ,Tumor Suppressor Protein p53 ,Wnt Proteins ,Cancer Genome Atlas Research Network ,PanCanAtlas ,TCGA ,cancer genome atlas ,cancer genomics ,combination therapy ,pan-cancer ,precision oncology ,signaling pathways ,therapeutics ,whole exome sequencing ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFβ signaling, p53 and β-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.
- Published
- 2018
81. Machine Learning Identifies Stemness Features Associated with Oncogenic Dedifferentiation
- Author
-
Malta, Tathiane M, Sokolov, Artem, Gentles, Andrew J, Burzykowski, Tomasz, Poisson, Laila, Weinstein, John N, Kamińska, Bożena, Huelsken, Joerg, Omberg, Larsson, Gevaert, Olivier, Colaprico, Antonio, Czerwińska, Patrycja, Mazurek, Sylwia, Mishra, Lopa, Heyn, Holger, Krasnitz, Alex, Godwin, Andrew K, Lazar, Alexander J, 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, and Sanchez-Vega, Francisco
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research - Nonembryonic - Non-Human ,Rare Diseases ,Machine Learning and Artificial Intelligence ,Human Genome ,Genetics ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Cancer Genomics ,Cancer ,Good Health and Well Being ,Carcinogenesis ,Cell Dedifferentiation ,DNA Methylation ,Databases ,Genetic ,Epigenesis ,Genetic ,Humans ,Machine Learning ,MicroRNAs ,Neoplasm Metastasis ,Neoplasms ,Stem Cells ,Transcriptome ,Tumor Microenvironment ,Cancer Genome Atlas Research Network ,The Cancer Genome Atlas ,cancer stem cells ,dedifferentiation ,epigenomic ,genomic ,machine learning ,pan-cancer ,stemness ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Cancer progression involves the gradual loss of a differentiated phenotype and acquisition of progenitor and stem-cell-like features. Here, we provide novel stemness indices for assessing the degree of oncogenic dedifferentiation. We used an innovative one-class logistic regression (OCLR) machine-learning algorithm to extract transcriptomic and epigenetic feature sets derived from non-transformed pluripotent stem cells and their differentiated progeny. Using OCLR, we were able to identify previously undiscovered biological mechanisms associated with the dedifferentiated oncogenic state. Analyses of the tumor microenvironment revealed unanticipated correlation of cancer stemness with immune checkpoint expression and infiltrating immune cells. We found that the dedifferentiated oncogenic phenotype was generally most prominent in metastatic tumors. Application of our stemness indices to single-cell data revealed patterns of intra-tumor molecular heterogeneity. Finally, the indices allowed for the identification of novel targets and possible targeted therapies aimed at tumor differentiation.
- Published
- 2018
82. 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 ,Human Genome ,Biotechnology ,Cancer ,Cancer Genomics ,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
83. A Comprehensive Pan-Cancer Molecular Study of Gynecologic and Breast Cancers
- Author
-
Berger, Ashton C, Korkut, Anil, Kanchi, Rupa S, Hegde, Apurva M, Lenoir, Walter, Liu, Wenbin, Liu, Yuexin, Fan, Huihui, Shen, Hui, Ravikumar, Visweswaran, Rao, Arvind, Schultz, Andre, Li, Xubin, Sumazin, Pavel, Williams, Cecilia, Mestdagh, Pieter, Gunaratne, Preethi H, Yau, Christina, Bowlby, Reanne, Robertson, A Gordon, Tiezzi, Daniel G, Wang, Chen, Cherniack, Andrew D, Godwin, Andrew K, Kuderer, Nicole M, Rader, Janet S, Zuna, Rosemary E, Sood, Anil K, Lazar, Alexander J, Ojesina, Akinyemi I, Adebamowo, Clement, Adebamowo, Sally N, Baggerly, Keith A, Chen, Ting-Wen, Chiu, Hua-Sheng, Lefever, Steve, Liu, Liang, MacKenzie, Karen, Orsulic, Sandra, Roszik, Jason, Shelley, Carl Simon, Song, Qianqian, Vellano, Christopher P, Wentzensen, Nicolas, 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, Miller, Michael, Reynolds, Sheila, Shmulevich, Ilya, Thorsson, Vesteinn, Zhang, Wei, Akbani, Rehan, Broom, Bradley M, Ju, Zhenlin, Li, Jun, Liang, Han, and Ling, Shiyun
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Human Genome ,Genetics ,Precision Medicine ,Women's Health ,Cancer Genomics ,Breast Cancer ,4.1 Discovery and preclinical testing of markers and technologies ,Breast Neoplasms ,DNA Copy Number Variations ,Databases ,Genetic ,Female ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Genital Neoplasms ,Female ,Humans ,Mutation ,Organ Specificity ,Prognosis ,RNA ,Long Noncoding ,Receptors ,Estrogen ,Cancer Genome Atlas Research Network ,TCGA ,The Cancer Genome Atlas ,breast cancer ,cervical cancer ,gynecologic cancer ,omics ,ovarian cancer ,pan-gynecologic ,uterine cancer ,uterine carcinosarcoma ,Neurosciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
We analyzed molecular data on 2,579 tumors from The Cancer Genome Atlas (TCGA) of four gynecological types plus breast. Our aims were to identify shared and unique molecular features, clinically significant subtypes, and potential therapeutic targets. We found 61 somatic copy-number alterations (SCNAs) and 46 significantly mutated genes (SMGs). Eleven SCNAs and 11 SMGs had not been identified in previous TCGA studies of the individual tumor types. We found functionally significant estrogen receptor-regulated long non-coding RNAs (lncRNAs) and gene/lncRNA interaction networks. Pathway analysis identified subtypes with high leukocyte infiltration, raising potential implications for immunotherapy. Using 16 key molecular features, we identified five prognostic subtypes and developed a decision tree that classified patients into the subtypes based on just six features that are assessable in clinical laboratories.
- Published
- 2018
84. 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, Network, The Cancer Genome Atlas, 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, 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, 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, Weinstein, John N, 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, and Phillips, Sarah M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Cancer Genomics ,Networking and Information Technology R&D (NITRD) ,Biotechnology ,Cancer ,2.1 Biological and endogenous factors ,Aneuploidy ,Chromosomes ,Cluster Analysis ,CpG Islands ,DNA Methylation ,Databases ,Factual ,Humans ,MicroRNAs ,Mutation ,Neoplasm Proteins ,Neoplasms ,RNA ,Messenger ,Cancer Genome Atlas Network ,TCGA ,cancer ,cell-of-origin ,genome ,methylome ,organs ,proteome ,subtypes ,tissues ,transcriptome ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical 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
85. Pathogenic Germline Variants in 10,389 Adult Cancers
- Author
-
Huang, Kuan-lin, Mashl, R Jay, Wu, Yige, Ritter, Deborah I, Wang, Jiayin, Oh, Clara, Paczkowska, Marta, Reynolds, Sheila, Wyczalkowski, Matthew A, Oak, Ninad, Scott, Adam D, Krassowski, Michal, Cherniack, Andrew D, Houlahan, Kathleen E, Jayasinghe, Reyka, Wang, Liang-Bo, Zhou, Daniel Cui, Liu, Di, Cao, Song, Kim, Young Won, Koire, Amanda, McMichael, Joshua F, Hucthagowder, Vishwanathan, Kim, Tae-Beom, Hahn, Abigail, Wang, Chen, McLellan, Michael D, Al-Mulla, Fahd, Johnson, Kimberly J, 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, 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, Weinstein, John N, Zhang, Jiexin, Abeshouse, Adam, Armenia, Joshua, Chakravarty, Debyani, Chatila, Walid K, de Bruijn, Ino, and Gao, Jianjiong
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Oncology and Carcinogenesis ,Cancer ,Rare Diseases ,Prevention ,Cancer Genomics ,Human Genome ,2.1 Biological and endogenous factors ,DNA Copy Number Variations ,Databases ,Genetic ,Gene Deletion ,Gene Frequency ,Genetic Predisposition to Disease ,Genotype ,Germ Cells ,Germ-Line Mutation ,Humans ,Loss of Heterozygosity ,Mutation ,Missense ,Neoplasms ,Polymorphism ,Single Nucleotide ,Proto-Oncogene Proteins c-met ,Proto-Oncogene Proteins c-ret ,Tumor Suppressor Proteins ,Cancer Genome Atlas Research Network ,LOH ,cancer predisposition ,germline and somatic genomes ,variant pathogenicity ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
We conducted the largest investigation of predisposition variants in cancer to date, discovering 853 pathogenic or likely pathogenic variants in 8% of 10,389 cases from 33 cancer types. Twenty-one genes showed single or cross-cancer associations, including novel associations of SDHA in melanoma and PALB2 in stomach adenocarcinoma. The 659 predisposition variants and 18 additional large deletions in tumor suppressors, including ATM, BRCA1, and NF1, showed low gene expression and frequent (43%) loss of heterozygosity or biallelic two-hit events. We also discovered 33 such variants in oncogenes, including missenses in MET, RET, and PTPN11 associated with high gene expression. We nominated 47 additional predisposition variants from prioritized VUSs supported by multiple evidences involving case-control frequency, loss of heterozygosity, expression effect, and co-localization with mutations and modified residues. Our integrative approach links rare predisposition variants to functional consequences, informing future guidelines of variant classification and germline genetic testing in cancer.
- Published
- 2018
86. Systematic Analysis of Splice-Site-Creating Mutations in Cancer
- Author
-
Jayasinghe, Reyka G, Cao, Song, Gao, Qingsong, Wendl, Michael C, Vo, Nam Sy, Reynolds, Sheila M, Zhao, Yanyan, Climente-González, Héctor, Chai, Shengjie, Wang, Fang, Varghese, Rajees, Huang, Mo, Liang, Wen-Wei, Wyczalkowski, Matthew A, Sengupta, Sohini, Li, Zhi, Payne, Samuel H, Fenyö, David, Miner, Jeffrey H, Walter, Matthew J, 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, Weinstein, John N, 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, and Phillips, Sarah M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Cancer Genomics ,Human Genome ,Rare Diseases ,Cancer ,Immunotherapy ,Genetics ,5.1 Pharmaceuticals ,2.1 Biological and endogenous factors ,BRCA1 Protein ,GATA3 Transcription Factor ,HEK293 Cells ,Humans ,Mutation ,Neoplasms ,Poly (ADP-Ribose) Polymerase-1 ,Programmed Cell Death 1 Receptor ,RNA Splice Sites ,Tumor Suppressor Protein p53 ,X-linked Nuclear Protein ,Cancer Genome Atlas Research Network ,RNA ,mutations of clinical relevance ,splicing ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
For the past decade, cancer genomic studies have focused on mutations leading to splice-site disruption, overlooking those having splice-creating potential. Here, we applied a bioinformatic tool, MiSplice, for the large-scale discovery of splice-site-creating mutations (SCMs) across 8,656 TCGA tumors. We report 1,964 originally mis-annotated mutations having clear evidence of creating alternative splice junctions. TP53 and GATA3 have 26 and 18 SCMs, respectively, and ATRX has 5 from lower-grade gliomas. Mutations in 11 genes, including PARP1, BRCA1, and BAP1, were experimentally validated for splice-site-creating function. Notably, we found that neoantigens induced by SCMs are likely several folds more immunogenic compared to missense mutations, exemplified by the recurrent GATA3 SCM. Further, high expression of PD-1 and PD-L1 was observed in tumors with SCMs, suggesting candidates for immune blockade therapy. Our work highlights the importance of integrating DNA and RNA data for understanding the functional and the clinical implications of mutations in human diseases.
- Published
- 2018
87. Driver Fusions and Their Implications in the Development and Treatment of Human Cancers
- Author
-
Gao, Qingsong, Liang, Wen-Wei, Foltz, Steven M, Mutharasu, Gnanavel, Jayasinghe, Reyka G, Cao, Song, Liao, Wen-Wei, Reynolds, Sheila M, Wyczalkowski, Matthew A, Yao, Lijun, Yu, Lihua, Sun, Sam Q, Group, The Fusion Analysis Working, 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, Weinstein, John N, 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, and Sun, Yichao
- Subjects
Biological Sciences ,Digestive Diseases ,Rare Diseases ,Human Genome ,Genetics ,Biotechnology ,Cancer Genomics ,Cancer ,5.1 Pharmaceuticals ,Good Health and Well Being ,Antineoplastic Agents ,Carcinogenesis ,Cell Line ,Tumor ,Humans ,Molecular Targeted Therapy ,Neoplasms ,Oncogene Fusion ,Oncogene Proteins ,Fusion ,Fusion Analysis Working Group ,Cancer Genome Atlas Research Network ,RNA ,cancer ,fusion ,gene fusions ,translocation ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Gene fusions represent an important class of somatic alterations in cancer. We systematically investigated fusions in 9,624 tumors across 33 cancer types using multiple fusion calling tools. We identified a total of 25,664 fusions, with a 63% validation rate. Integration of gene expression, copy number, and fusion annotation data revealed that fusions involving oncogenes tend to exhibit increased expression, whereas fusions involving tumor suppressors have the opposite effect. For fusions involving kinases, we found 1,275 with an intact kinase domain, the proportion of which varied significantly across cancer types. Our study suggests that fusions drive the development of 16.5% of cancer cases and function as the sole driver in more than 1% of them. Finally, we identified druggable fusions involving genes such as TMPRSS2, RET, FGFR3, ALK, and ESR1 in 6.0% of cases, and we predicted immunogenic peptides, suggesting that fusions may provide leads for targeted drug and immune therapy.
- Published
- 2018
88. Molecular Characterization and Clinical Relevance of Metabolic Expression Subtypes in Human Cancers
- Author
-
Peng, Xinxin, Chen, Zhongyuan, Farshidfar, Farshad, Xu, Xiaoyan, Lorenzi, Philip L, Wang, Yumeng, Cheng, Feixiong, Tan, Lin, Mojumdar, Kamalika, Du, Di, Ge, Zhongqi, Li, Jun, Thomas, George V, Birsoy, Kivanc, Liu, Lingxiang, Zhang, Huiwen, Zhao, Zhongming, Marchand, Calena, 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, 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, and Reznik, Ed
- Subjects
Biological Sciences ,Cancer Genomics ,Human Genome ,Cancer ,Nutrition ,Genetics ,2.1 Biological and endogenous factors ,Cell Line ,Tumor ,Core Binding Factor Alpha 2 Subunit ,Drug Resistance ,Neoplasm ,HEK293 Cells ,Humans ,Metabolic Networks and Pathways ,Neoplasms ,Snail Family Transcription Factors ,Transcriptome ,Cancer Genome Atlas Research Network ,The Cancer Genome Atlas ,carbohydrate metabolism ,drug sensitivity ,master regulator ,prognostic markers ,somatic drivers ,therapeutic targets ,tumor subtypes ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Metabolic reprogramming provides critical information for clinical oncology. Using molecular data of 9,125 patient samples from The Cancer Genome Atlas, we identified tumor subtypes in 33 cancer types based on mRNA expression patterns of seven major metabolic processes and assessed their clinical relevance. Our metabolic expression subtypes correlated extensively with clinical outcome: subtypes with upregulated carbohydrate, nucleotide, and vitamin/cofactor metabolism most consistently correlated with worse prognosis, whereas subtypes with upregulated lipid metabolism showed the opposite. Metabolic subtypes correlated with diverse somatic drivers but exhibited effects convergent on cancer hallmark pathways and were modulated by highly recurrent master regulators across cancer types. As a proof-of-concept example, we demonstrated that knockdown of SNAI1 or RUNX1-master regulators of carbohydrate metabolic subtypes-modulates metabolic activity and drug sensitivity. Our study provides a system-level view of metabolic heterogeneity within and across cancer types and identifies pathway cross-talk, suggesting related prognostic, therapeutic, and predictive utility.
- Published
- 2018
89. Machine Learning Detects Pan-cancer Ras Pathway Activation in The Cancer Genome Atlas
- Author
-
Way, Gregory P, Sanchez-Vega, Francisco, La, Konnor, Armenia, Joshua, Chatila, Walid K, Luna, Augustin, Sander, Chris, Cherniack, Andrew D, Mina, Marco, Ciriello, Giovanni, Schultz, Nikolaus, 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, Weinstein, John N, Zhang, Jiexin, Abeshouse, Adam, Chakravarty, Debyani, de Bruijn, Ino, Gao, Jianjiong, Gross, Benjamin E, Heins, Zachary J, Kundra, Ritika, Ladanyi, Marc, Nissan, Moriah G, Ochoa, Angelica, Phillips, Sarah M, Reznik, Ed, Sheridan, Robert, Sumer, S Onur, Sun, Yichao, Taylor, Barry S, and Wang, Jioajiao
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer ,Networking and Information Technology R&D (NITRD) ,Human Genome ,Cancer Genomics ,Precision Medicine ,Machine Learning and Artificial Intelligence ,Good Health and Well Being ,Cell Line ,Tumor ,Gene Expression Regulation ,Neoplastic ,Genome ,Human ,Humans ,Machine Learning ,Neoplasms ,Signal Transduction ,ras Proteins ,Cancer Genome Atlas Research Network ,Gene expression ,HRAS ,KRAS ,NF1 ,NRAS ,Ras ,TCGA ,drug sensitivity ,machine learning ,pan-cancer ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Precision oncology uses genomic evidence to match patients with treatment but often fails to identify all patients who may respond. The transcriptome of these "hidden responders" may reveal responsive molecular states. We describe and evaluate a machine-learning approach to classify aberrant pathway activity in tumors, which may aid in hidden responder identification. The algorithm integrates RNA-seq, copy number, and mutations from 33 different cancer types across The Cancer Genome Atlas (TCGA) PanCanAtlas project to predict aberrant molecular states in tumors. Applied to the Ras pathway, the method detects Ras activation across cancer types and identifies phenocopying variants. The model, trained on human tumors, can predict response to MEK inhibitors in wild-type Ras cell lines. We also present data that suggest that multiple hits in the Ras pathway confer increased Ras activity. The transcriptome is underused in precision oncology and, combined with machine learning, can aid in the identification of hidden responders.
- Published
- 2018
90. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics
- Author
-
Liu, Jianfang, Lichtenberg, Tara, Hoadley, Katherine A, Poisson, Laila M, Lazar, Alexander J, Cherniack, Andrew D, Kovatich, Albert J, Benz, Christopher C, Levine, Douglas A, Lee, Adrian V, Omberg, Larsson, Wolf, Denise M, Shriver, Craig D, Thorsson, Vesteinn, 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, 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, Weinstein, John N, 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, and Sumer, S Onur
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Precision Medicine ,Women's Health ,Networking and Information Technology R&D (NITRD) ,Human Genome ,Genetics ,Biotechnology ,Cancer Genomics ,Cancer ,Good Health and Well Being ,Databases ,Genetic ,Genomics ,Humans ,Kaplan-Meier Estimate ,Neoplasms ,Proportional Hazards Models ,Cancer Genome Atlas Research Network ,Cox proportional hazards regression model ,TCGA ,The Cancer Genome Atlas ,clinical data resource ,disease-free interval ,disease-specific survival ,follow-up time ,overall survival ,progression-free interval ,translational research ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
For a decade, The Cancer Genome Atlas (TCGA) program collected clinicopathologic annotation data along with multi-platform molecular profiles of more than 11,000 human tumors across 33 different cancer types. TCGA clinical data contain key features representing the democratized nature of the data collection process. To ensure proper use of this large clinical dataset associated with genomic features, we developed a standardized dataset named the TCGA Pan-Cancer Clinical Data Resource (TCGA-CDR), which includes four major clinical outcome endpoints. In addition to detailing major challenges and statistical limitations encountered during the effort of integrating the acquired clinical data, we present a summary that includes endpoint usage recommendations for each cancer type. These TCGA-CDR findings appear to be consistent with cancer genomics studies independent of the TCGA effort and provide opportunities for investigating cancer biology using clinical correlates at an unprecedented scale.
- Published
- 2018
91. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas
- Author
-
Knijnenburg, Theo A, Wang, Linghua, Zimmermann, Michael T, Chambwe, Nyasha, Gao, Galen F, Cherniack, Andrew D, Fan, Huihui, Shen, Hui, Way, Gregory P, Greene, Casey S, Liu, Yuexin, Akbani, Rehan, Feng, Bin, Donehower, Lawrence A, Miller, Chase, Shen, Yang, Karimi, Mostafa, Chen, Haoran, Kim, Pora, Jia, Peilin, Shinbrot, Eve, Zhang, Shaojun, Liu, Jianfang, Hu, Hai, Bailey, Matthew H, Yau, Christina, Wolf, Denise, Zhao, Zhongming, Weinstein, John N, Li, Lei, Ding, Li, Mills, Gordon B, Laird, Peter W, Wheeler, David A, Shmulevich, Ilya, 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, Dhankani, Varsha, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, Miller, Michael, Reynolds, Sheila, Thorsson, Vesteinn, 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, and Zhang, Jiexin
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Women's Health ,Orphan Drug ,Rare Diseases ,Human Genome ,Ovarian Cancer ,Cancer Genomics ,Cancer ,2.1 Biological and endogenous factors ,Cell Line ,Tumor ,DNA Damage ,Gene Silencing ,Genome ,Human ,Humans ,Loss of Heterozygosity ,Machine Learning ,Mutation ,Neoplasms ,Recombinational DNA Repair ,Tumor Suppressor Proteins ,Cancer Genome Atlas Research Network ,DNA damage footprints ,DNA damage repair ,The Cancer Genome Atlas PanCanAtlas project ,epigenetic silencing ,integrative statistical analysis ,mutational signatures ,protein structure analysis ,somatic copy-number alterations ,somatic mutations ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
DNA damage repair (DDR) pathways modulate cancer risk, progression, and therapeutic response. We systematically analyzed somatic alterations to provide a comprehensive view of DDR deficiency across 33 cancer types. Mutations with accompanying loss of heterozygosity were observed in over 1/3 of DDR genes, including TP53 and BRCA1/2. Other prevalent alterations included epigenetic silencing of the direct repair genes EXO5, MGMT, and ALKBH3 in ∼20% of samples. Homologous recombination deficiency (HRD) was present at varying frequency in many cancer types, most notably ovarian cancer. However, in contrast to ovarian cancer, HRD was associated with worse outcomes in several other cancers. Protein structure-based analyses allowed us to predict functional consequences of rare, recurrent DDR mutations. A new machine-learning-based classifier developed from gene expression data allowed us to identify alterations that phenocopy deleterious TP53 mutations. These frequent DDR gene alterations in many human cancers have functional consequences that may determine cancer progression and guide therapy.
- Published
- 2018
92. Scalable Open Science Approach for Mutation Calling of Tumor Exomes Using Multiple Genomic Pipelines
- Author
-
Ellrott, Kyle, Bailey, Matthew H, Saksena, Gordon, Covington, Kyle R, Kandoth, Cyriac, Stewart, Chip, Hess, Julian, Ma, Chiotti, Kami E, McLellan, Michael, Sofia, Heidi J, Hutter, Carolyn, Getz, Gad, Wheeler, David, Ding, Li, Group, MC3 Working, Network, The Cancer Genome Atlas Research, Caesar-Johnson, Samantha J, Demchok, John A, Felau, Ina, Kasapi, Melpomeni, Ferguson, Martin L, Hutter, Carolyn M, 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, Kim, Jaegil, Lawrence, Michael S, Lin, Pei, Meier, Sam, Noble, Michael S, 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, Weinstein, John N, 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, and Schultz, Nikolaus
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer ,Human Genome ,Rare Diseases ,Genetic Testing ,Networking and Information Technology R&D (NITRD) ,Cancer Genomics ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Algorithms ,Exome ,Genomics ,High-Throughput Nucleotide Sequencing ,Humans ,Information Dissemination ,Mutation ,Neoplasms ,Sequence Analysis ,DNA ,Software ,Exome Sequencing ,MC3 Working Group ,Cancer Genome Atlas Research Network ,PanCanAtlas project ,TCGA ,large-scale ,open science ,pan-cancer ,reproducible computing ,somatic mutation calling ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
The Cancer Genome Atlas (TCGA) cancer genomics dataset includes over 10,000 tumor-normal exome pairs across 33 different cancer types, in total >400 TB of raw data files requiring analysis. Here we describe the Multi-Center Mutation Calling in Multiple Cancers project, our effort to generate a comprehensive encyclopedia of somatic mutation calls for the TCGA data to enable robust cross-tumor-type analyses. Our approach accounts for variance and batch effects introduced by the rapid advancement of DNA extraction, hybridization-capture, sequencing, and analysis methods over time. We present best practices for applying an ensemble of seven mutation-calling algorithms with scoring and artifact filtering. The dataset created by this analysis includes 3.5 million somatic variants and forms the basis for PanCan Atlas papers. The results have been made available to the research community along with the methods used to generate them. This project is the result of collaboration from a number of institutes and demonstrates how team science drives extremely large genomics projects.
- Published
- 2018
93. Pan-cancer Alterations of the MYC Oncogene and Its Proximal Network across the Cancer Genome Atlas
- Author
-
Schaub, Franz X, Dhankani, Varsha, Berger, Ashton C, Trivedi, Mihir, Richardson, Anne B, Shaw, Reid, Zhao, Wei, Zhang, Xiaoyang, Ventura, Andrea, Liu, Yuexin, Ayer, Donald E, Hurlin, Peter J, Cherniack, Andrew D, Eisenman, Robert N, Bernard, Brady, Grandori, Carla, Network, The Cancer Genome Atlas, 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, Chambwe, Nyasha, Knijnenburg, Theo, Kramer, Roger, Leinonen, Kalle, 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, Weinstein, John N, 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, and Schultz, Nikolaus
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Cancer Genomics ,Human Genome ,Cancer ,Genetics ,Biotechnology ,2.1 Biological and endogenous factors ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Basic Helix-Loop-Helix Transcription Factors ,Biomarkers ,Tumor ,Carcinogenesis ,Chromatin ,Computational Biology ,Genes ,myc ,Genomics ,Humans ,Neoplasms ,Oncogenes ,Proteomics ,Proto-Oncogene Proteins c-myc ,Repressor Proteins ,Signal Transduction ,Transcription Factors ,Cancer Genome Atlas Network ,MAX ,MNT ,MYC genomic alterations ,TCGA ,The Cancer Genome Atlas ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
Although the MYC oncogene has been implicated in cancer, a systematic assessment of alterations of MYC, related transcription factors, and co-regulatory proteins, forming the proximal MYC network (PMN), across human cancers is lacking. Using computational approaches, we define genomic and proteomic features associated with MYC and the PMN across the 33 cancers of The Cancer Genome Atlas. Pan-cancer, 28% of all samples had at least one of the MYC paralogs amplified. In contrast, the MYC antagonists MGA and MNT were the most frequently mutated or deleted members, proposing a role as tumor suppressors. MYC alterations were mutually exclusive with PIK3CA, PTEN, APC, or BRAF alterations, suggesting that MYC is a distinct oncogenic driver. Expression analysis revealed MYC-associated pathways in tumor subtypes, such as immune response and growth factor signaling; chromatin, translation, and DNA replication/repair were conserved pan-cancer. This analysis reveals insights into MYC biology and is a reference for biomarkers and therapeutics for cancers with alterations of MYC or the PMN.
- Published
- 2018
94. The Integrated Genomic Landscape of Thymic Epithelial Tumors
- Author
-
Radovich, Milan, Pickering, Curtis R, Felau, Ina, Ha, Gavin, Zhang, Hailei, Jo, Heejoon, Hoadley, Katherine A, Anur, Pavana, Zhang, Jiexin, McLellan, Mike, Bowlby, Reanne, Matthew, Thomas, Danilova, Ludmila, Hegde, Apurva M, Kim, Jaegil, Leiserson, Mark DM, Sethi, Geetika, Lu, Charles, Ryan, Michael, Su, Xiaoping, Cherniack, Andrew D, Robertson, Gordon, Akbani, Rehan, Spellman, Paul, Weinstein, John N, Hayes, D Neil, Raphael, Ben, Lichtenberg, Tara, Leraas, Kristen, Zenklusen, Jean Claude, Network, The Cancer Genome Atlas, Ally, Adrian, Appelbaum, Elizabeth L, Auman, J Todd, Balasundaram, Miruna, Balu, Saianand, Behera, Madhusmita, Beroukhim, Rameen, Berrios, Mario, Blandino, Giovanni, Bodenheimer, Tom, Bootwalla, Moiz S, Bowen, Jay, Brooks, Denise, Carcano, Flavio M, Carlsen, Rebecca, Carvalho, Andre L, Castro, Patricia, Chalabreysse, Lara, Chin, Lynda, Cho, Juok, Choe, Gina, Chuah, Eric, Chudamani, Sudha, Cibulskis, Carrie, Cope, Leslie, Cordes, Matthew G, Crain, Daniel, Curley, Erin, Defreitas, Timothy, Demchok, John A, Detterbeck, Frank, Dhalla, Noreen, Dienemann, Hendrik, Edenfield, W Jeff, Facciolo, Francesco, Ferguson, Martin L, Frazer, Scott, Fronick, Catrina C, Fulton, Lucinda A, Fulton, Robert S, Gabriel, Stacey B, Gardner, Johanna, Gastier-Foster, Julie M, Gehlenborg, Nils, Gerken, Mark, Getz, Gad, Heiman, David I, Hobensack, Shital, Holbrook, Andrea, Holt, Robert A, Hoyle, Alan P, Hutter, Carolyn M, Ittmann, Michael, Jefferys, Stuart R, Jones, Corbin D, Jones, Steven JM, Kasaian, Katayoon, Kimes, Patrick K, Lai, Phillip H, Laird, Peter W, Lawrence, Michael S, Lin, Pei, Liu, Jia, Lolla, Laxmi, Lu, Yiling, Ma, Yussanne, Maglinte, Dennis T, and Mallery, David
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Human Genome ,Autoimmune Disease ,Cancer ,Rare Diseases ,Biotechnology ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Female ,Genomics ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasms ,Glandular and Epithelial ,Thymoma ,Thymus Neoplasms ,Transcription Factors ,TFII ,Young Adult ,Cancer Genome Atlas Network ,TCGA ,autoimmunity ,genomics ,myasthenia gravis ,proteomics ,thymic carcinoma ,thymic epithelial tumors ,thymoma ,transcriptomics ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Thymic epithelial tumors (TETs) are one of the rarest adult malignancies. Among TETs, thymoma is the most predominant, characterized by a unique association with autoimmune diseases, followed by thymic carcinoma, which is less common but more clinically aggressive. Using multi-platform omics analyses on 117 TETs, we define four subtypes of these tumors defined by genomic hallmarks and an association with survival and World Health Organization histological subtype. We further demonstrate a marked prevalence of a thymoma-specific mutated oncogene, GTF2I, and explore its biological effects on multi-platform analysis. We further observe enrichment of mutations in HRAS, NRAS, and TP53. Last, we identify a molecular link between thymoma and the autoimmune disease myasthenia gravis, characterized by tumoral overexpression of muscle autoantigens, and increased aneuploidy.
- Published
- 2018
95. Supplementary Figure S9 from Acquired Cross-Resistance in Small Cell Lung Cancer due to Extrachromosomal DNA Amplification of MYC Paralogs
- Author
-
Pal Choudhuri, Shreoshi, primary, Girard, Luc, primary, Lim, Jun Yi Stanley, primary, Wise, Jillian F., primary, Freitas, Braeden, primary, Yang, Di, primary, Wong, Edmond, primary, Hamilton, Seth, primary, Chien, Victor D., primary, Kim, Yoon Jung, primary, Gilbreath, Collin, primary, Zhong, Jun, primary, Phat, Sarah, primary, Myers, David T., primary, Christensen, Camilla L., primary, Mazloom-Farsibaf, Hanieh, primary, Stanzione, Marcello, primary, Wong, Kwok-Kin, primary, Hung, Yin P., primary, Farago, Anna F., primary, Meador, Catherine B., primary, Dyson, Nicholas J., primary, Lawrence, Michael S., primary, Wu, Sihan, primary, and Drapkin, Benjamin J., primary
- Published
- 2024
- Full Text
- View/download PDF
96. Supplementary Tables from Acquired Cross-Resistance in Small Cell Lung Cancer due to Extrachromosomal DNA Amplification of MYC Paralogs
- Author
-
Pal Choudhuri, Shreoshi, primary, Girard, Luc, primary, Lim, Jun Yi Stanley, primary, Wise, Jillian F., primary, Freitas, Braeden, primary, Yang, Di, primary, Wong, Edmond, primary, Hamilton, Seth, primary, Chien, Victor D., primary, Kim, Yoon Jung, primary, Gilbreath, Collin, primary, Zhong, Jun, primary, Phat, Sarah, primary, Myers, David T., primary, Christensen, Camilla L., primary, Mazloom-Farsibaf, Hanieh, primary, Stanzione, Marcello, primary, Wong, Kwok-Kin, primary, Hung, Yin P., primary, Farago, Anna F., primary, Meador, Catherine B., primary, Dyson, Nicholas J., primary, Lawrence, Michael S., primary, Wu, Sihan, primary, and Drapkin, Benjamin J., primary
- Published
- 2024
- Full Text
- View/download PDF
97. Predictors of Response to an Upper Extremity Telerehabilitation-Home Practice Program after Stroke: An Interim Analysis
- Author
-
Rishe, Kelly, primary, Goedeken, Susan, additional, DiCarlo, Julie, additional, McKiernan, Sydney, additional, Hamilton, Taya, additional, Lawrence, Michael, additional, Friesen, Christopher, additional, Ingram, Tony, additional, and Lin, David, additional
- Published
- 2024
- Full Text
- View/download PDF
98. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product
- Author
-
Mishra, Avanish, primary, Huang, Shih-Bo, additional, Dubash, Taronish, additional, Burr, Risa, additional, Edd, Jon F., additional, Wittner, Ben S., additional, Cunneely, Quinn E., additional, Putaturo, Victor R., additional, Deshpande, Akansha, additional, Antmen, Ezgi, additional, Gopinathan, Kaustav A., additional, Otani, Keisuke, additional, Miyazawa, Yoshiyuki, additional, Kwak, Ji Eun, additional, Guay, Sara Y., additional, Kelly, Justin, additional, Walsh, John, additional, Nieman, Linda, additional, Galler, Isabella, additional, Chan, PuiYee, additional, Lawrence, Michael S., additional, Sullivan, Ryan J., additional, Bardia, Aditya, additional, Micalizzi, Douglas S., additional, Sequist, Lecia V., additional, Lee, Richard J., additional, Franses, Joseph W., additional, Ting, David T., additional, Brunker, Patricia A. R., additional, Maheswaran, Shyamala, additional, Miyamoto, David T., additional, Haber, Daniel A., additional, and Toner, Mehmet, additional
- Published
- 2024
- Full Text
- View/download PDF
99. Acquired Cross-Resistance in Small Cell Lung Cancer due to Extrachromosomal DNA Amplification of MYC Paralogs
- Author
-
Pal Choudhuri, Shreoshi, primary, Girard, Luc, additional, Lim, Jun Yi Stanley, additional, Wise, Jillian F., additional, Freitas, Braeden, additional, Yang, Di, additional, Wong, Edmond, additional, Hamilton, Seth, additional, Chien, Victor D., additional, Kim, Yoon Jung, additional, Gilbreath, Collin, additional, Zhong, Jun, additional, Phat, Sarah, additional, Myers, David T., additional, Christensen, Camilla L., additional, Mazloom-Farsibaf, Hanieh, additional, Stanzione, Marcello, additional, Wong, Kwok-Kin, additional, Hung, Yin P., additional, Farago, Anna F., additional, Meador, Catherine B., additional, Dyson, Nicholas J., additional, Lawrence, Michael S., additional, Wu, Sihan, additional, and Drapkin, Benjamin J., additional
- Published
- 2024
- Full Text
- View/download PDF
100. Physico-chemical Characterization and Development of Hemp Aggregates for Highly Insulating Construction Building Materials
- Author
-
Jiang, Yunhong, Hussain, Atif, Heidari, Davoud M., Lawrence, Michael, Ansell, Martin, Lichtfouse, Eric, Series Editor, Ranjan, Shivendu, Advisory Editor, Dasgupta, Nandita, Advisory Editor, and Crini, Grégorio, editor
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.