1
|
Shuaibi A, Chitra U, Raphael BJ. A latent variable model for evaluating mutual exclusivity and co-occurrence between driver mutations in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590995. [PMID: 38712136 PMCID: PMC11071465 DOI: 10.1101/2024.04.24.590995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
A key challenge in cancer genomics is understanding the functional relationships and dependencies between combinations of somatic mutations that drive cancer development. Such driver mutations frequently exhibit patterns of mutual exclusivity or co-occurrence across tumors, and many methods have been developed to identify such dependency patterns from bulk DNA sequencing data of a cohort of patients. However, while mutual exclusivity and co-occurrence are described as properties of driver mutations, existing methods do not explicitly disentangle functional, driver mutations from neutral, passenger mutations. In particular, nearly all existing methods evaluate mutual exclusivity or co-occurrence at the gene level, marking a gene as mutated if any mutation - driver or passenger - is present. Since some genes have a large number of passenger mutations, existing methods either restrict their analyses to a small subset of suspected driver genes - limiting their ability to identify novel dependencies - or make spurious inferences of mutual exclusivity and co-occurrence involving genes with many passenger mutations. We introduce DIALECT, an algorithm to identify dependencies between pairs of driver mutations from somatic mutation counts. We derive a latent variable mixture model for drivers and passengers that combines existing probabilistic models of passenger mutation rates with a latent variable describing the unknown status of a mutation as a driver or passenger. We use an expectation maximization (EM) algorithm to estimate the parameters of our model, including the rates of mutually exclusivity and co-occurrence between drivers. We demonstrate that DIALECT more accurately infers mutual exclusivity and co-occurrence between driver mutations compared to existing methods on both simulated mutation data and somatic mutation data from 5 cancer types in The Cancer Genome Atlas (TCGA).
Collapse
Affiliation(s)
- Ahmed Shuaibi
- Department of Computer Science, Princeton University
- Lewis-Sigler Institute for Integrative Genomics, Princeton University
| | - Uthsav Chitra
- Department of Computer Science, Princeton University
| | | |
Collapse
|
2
|
Wang X, Kostrzewa C, Reiner A, Shen R, Begg C. Adaptation of a mutual exclusivity framework to identify driver mutations within oncogenic pathways. Am J Hum Genet 2024; 111:227-241. [PMID: 38232729 PMCID: PMC10870134 DOI: 10.1016/j.ajhg.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
Distinguishing genomic alterations in cancer-associated genes that have functional impact on tumor growth and disease progression from the ones that are passengers and confer no fitness advantage have important clinical implications. Evidence-based methods for nominating drivers are limited by existing knowledge on the oncogenic effects and therapeutic benefits of specific variants from clinical trials or experimental settings. As clinical sequencing becomes a mainstay of patient care, applying computational methods to mine the rapidly growing clinical genomic data holds promise in uncovering functional candidates beyond the existing knowledge base and expanding the patient population that could potentially benefit from genetically targeted therapies. We propose a statistical and computational method (MAGPIE) that builds on a likelihood approach leveraging the mutual exclusivity pattern within an oncogenic pathway for identifying probabilistically both the specific genes within a pathway and the individual mutations within such genes that are truly the drivers. Alterations in a cancer-associated gene are assumed to be a mixture of driver and passenger mutations with the passenger rates modeled in relationship to tumor mutational burden. We use simulations to study the operating characteristics of the method and assess false-positive and false-negative rates in driver nomination. When applied to a large study of primary melanomas, the method accurately identifies the known driver genes within the RTK-RAS pathway and nominates several rare variants as prime candidates for functional validation. A comprehensive evaluation of MAGPIE against existing tools has also been conducted leveraging the Cancer Genome Atlas data.
Collapse
Affiliation(s)
- Xinjun Wang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Caroline Kostrzewa
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allison Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Colin Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
3
|
Wang X, Kostrzewa C, Reiner A, Shen R, Begg C. Adaptation of a Mutual Exclusivity Framework to Identify Driver Mutations within Biological Pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558469. [PMID: 37786694 PMCID: PMC10541562 DOI: 10.1101/2023.09.19.558469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Distinguishing genomic alterations in cancer genes that have functional impact on tumor growth and disease progression from the ones that are passengers and confer no fitness advantage has important clinical implications. Evidence-based methods for nominating drivers are limited by existing knowledge on the oncogenic effects and therapeutic benefits of specific variants from clinical trials or experimental settings. As clinical sequencing becomes a mainstay of patient care, applying computational methods to mine the rapidly growing clinical genomic data holds promise in uncovering novel functional candidates beyond the existing knowledge-base and expanding the patient population that could potentially benefit from genetically targeted therapies. We propose a statistical and computational method (MAGPIE) that builds on a likelihood approach leveraging the mutual exclusivity pattern within an oncogenic pathway for identifying probabilistically both the specific genes within a pathway and the individual mutations within such genes that are truly the drivers. Alterations in a cancer gene are assumed to be a mixture of driver and passenger mutations with the passenger rates modeled in relationship to tumor mutational burden. A limited memory BFGS algorithm is used to facilitate large scale optimization. We use simulations to study the operating characteristics of the method and assess false positive and false negative rates in driver nomination. When applied to a large study of primary melanomas the method accurately identified the known driver genes within the RTK-RAS pathway and nominated a number of rare variants with previously unknown biological and clinical relevance as prime candidates for functional validation.
Collapse
|
4
|
Berber I, Erten C, Kazan H. Predator: Predicting the Impact of Cancer Somatic Mutations on Protein-Protein Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3163-3172. [PMID: 37030791 DOI: 10.1109/tcbb.2023.3262119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Since many biological processes are governed by protein-protein interactions, understanding which mutations lead to a disruption in these interactions is profoundly important for cancer research. Most of the existing methods focus on the stability of the protein without considering the specific effects of a mutation on its interactions with other proteins. Here, we focus on somatic mutations that appear on the interface regions of the protein and predict the interactions that would be affected by a mutation of interest. We build an ensemble model, Predator, that classifies the interface mutations as disruptive or nondisruptive based on the predicted effects of mutations on specific protein-protein interactions. We show that Predator outperforms existing approaches in literature in terms of prediction accuracy. We then apply Predator on various TCGA cancer cohorts and perform comprehensive analysis at cohort level, patient level, and gene level in determining the genes whose interface mutations tend to yield a disruption in its interactions. The predictions obtained by Predator shed light on interesting patterns on several genes for each cohort regarding their potential as cancer drivers. Our analyses further reveal that the identified genes and their frequently disrupted partners exhibit patterns of mutually exclusivity across cancer cohorts under study.
Collapse
|
5
|
Mina M, Iyer A, Ciriello G. Epistasis and evolutionary dependencies in human cancers. Curr Opin Genet Dev 2022; 77:101989. [PMID: 36182742 DOI: 10.1016/j.gde.2022.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 01/27/2023]
Abstract
Cancer evolution is driven by the concerted action of multiple molecular alterations, which emerge and are selected during tumor progression. An alteration is selected when it provides an advantage to the tumor cell. However, the advantage provided by a specific alteration depends on the tumor lineage, cell epigenetic state, and presence of additional alterations. In this case, we say that an evolutionary dependency exists between an alteration and what influences its selection. Epistatic interactions between altered genes lead to evolutionary dependencies (EDs), by favoring or vetoing specific combinations of events. Large-scale cancer genomics studies have discovered examples of such dependencies, and showed that they influence tumor progression, disease phenotypes, and therapeutic response. In the past decade, several algorithmic approaches have been proposed to infer EDs from large-scale genomics datasets. These methods adopt diverse strategies to address common challenges and shed new light on cancer evolutionary trajectories. Here, we review these efforts starting from a simple conceptualization of the problem, presenting the tackled and still unmet needs in the field, and discussing the implications of EDs in cancer biology and precision oncology.
Collapse
Affiliation(s)
- Marco Mina
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Arvind Iyer
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
6
|
Giannuzzi D, Marconato L, Fanelli A, Licenziato L, De Maria R, Rinaldi A, Rotta L, Rouquet N, Birolo G, Fariselli P, Mensah AA, Bertoni F, Aresu L. The genomic landscape of canine diffuse large B-cell lymphoma identifies distinct subtypes with clinical and therapeutic implications. Lab Anim (NY) 2022; 51:191-202. [PMID: 35726023 DOI: 10.1038/s41684-022-00998-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/13/2022] [Indexed: 12/13/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid neoplasm in dogs and in humans. It is characterized by a remarkable degree of clinical heterogeneity that is not completely elucidated by molecular data. This poses a major barrier to understanding the disease and its response to therapy, or when treating dogs with DLBCL within clinical trials. We performed an integrated analysis of exome (n = 77) and RNA sequencing (n = 43) data in a cohort of canine DLBCL to define the genetic landscape of this tumor. A wide range of signaling pathways and cellular processes were found in common with human DLBCL, but the frequencies of the most recurrently mutated genes (TRAF3, SETD2, POT1, TP53, MYC, FBXW7, DDX3X and TBL1XR1) differed. We developed a prognostic model integrating exonic variants and clinical and transcriptomic features to predict the outcome in dogs with DLBCL. These results comprehensively define the genetic drivers of canine DLBCL and can be prospectively utilized to identify new therapeutic opportunities.
Collapse
Affiliation(s)
- Diana Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padua, Padua, Italy
| | - Laura Marconato
- Department of Veterinary Medical Science, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | - Antonella Fanelli
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Luca Licenziato
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Andrea Rinaldi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Luca Rotta
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Giovanni Birolo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Afua A Mensah
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland. .,Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy.
| |
Collapse
|
7
|
Vokes NI, Chambers E, Nguyen T, Coolidge A, Lydon CA, Le X, Sholl L, Heymach JV, Nishino M, Van Allen EM, Jänne PA. Concurrent TP53 Mutations Facilitate Resistance Evolution in EGFR-Mutant Lung Adenocarcinoma. J Thorac Oncol 2022; 17:779-792. [PMID: 35331964 PMCID: PMC10478031 DOI: 10.1016/j.jtho.2022.02.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Patients with EGFR-mutant NSCLC experience variable duration of benefit on EGFR tyrosine kinase inhibitors. The effect of concurrent genomic alterations on outcome has been incompletely described. METHODS In this retrospective study, targeted next-generation sequencing data were collected from patients with EGFR-mutant lung cancer treated at the Dana-Farber Cancer Institute. Clinical data were collected and correlated with somatic mutation data. Associations between TP53 mutation status, genomic features, and mutational processes were analyzed. RESULTS A total of 269 patients were identified for inclusion in the cohort. Among 185 response-assessable patients with pretreatment specimens, TP53 alterations were the most common event associated with decreased first-line progression-free survival and decreased overall survival, along with DNMT3A, KEAP1, and ASXL1 alterations. Reduced progression-free survival on later-line osimertinib in 33 patients was associated with MET, APC, and ERBB4 alterations. Further investigation of the effect of TP53 alterations revealed an association with worse outcomes even in patients with good initial radiographic response, and faster acquisition of T790M and other resistance mechanisms. TP53-mutated tumors had higher mutational burdens and increased mutagenesis with exposure to therapy and tobacco. Cell cycle alterations were not independently predictive, but portended worse OS in conjunction with TP53 alterations. CONCLUSIONS TP53 alterations associate with faster resistance evolution independent of mechanism in EGFR-mutant NSCLC and may cooperate with other genomic events to mediate acquisition of resistance mutations to EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Natalie I Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emily Chambers
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alexis Coolidge
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Christine A Lydon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiuning Le
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts; Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
8
|
Germline mutations in mitochondrial complex I reveal genetic and targetable vulnerability in IDH1-mutant acute myeloid leukaemia. Nat Commun 2022; 13:2614. [PMID: 35551192 PMCID: PMC9098909 DOI: 10.1038/s41467-022-30223-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
The interaction of germline variation and somatic cancer driver mutations is under-investigated. Here we describe the genomic mitochondrial landscape in adult acute myeloid leukaemia (AML) and show that rare variants affecting the nuclear- and mitochondrially-encoded complex I genes show near-mutual exclusivity with somatic driver mutations affecting isocitrate dehydrogenase 1 (IDH1), but not IDH2 suggesting a unique epistatic relationship. Whereas AML cells with rare complex I variants or mutations in IDH1 or IDH2 all display attenuated mitochondrial respiration, heightened sensitivity to complex I inhibitors including the clinical-grade inhibitor, IACS-010759, is observed only for IDH1-mutant AML. Furthermore, IDH1 mutant blasts that are resistant to the IDH1-mutant inhibitor, ivosidenib, retain sensitivity to complex I inhibition. We propose that the IDH1 mutation limits the flexibility for citrate utilization in the presence of impaired complex I activity to a degree that is not apparent in IDH2 mutant cells, exposing a mutation-specific metabolic vulnerability. This reduced metabolic plasticity explains the epistatic relationship between the germline complex I variants and oncogenic IDH1 mutation underscoring the utility of genomic data in revealing metabolic vulnerabilities with implications for therapy. Mitochondrial metabolism has been associated with tumourigenesis in acute myeloid leukaemia (AML) and currently considered as a potential therapeutic target. Here, the authors show, in patients with AML, that germline mutations in mitochondrial complex I are mutually exclusive with somatic mutations in the metabolic enzyme IDH1, and find IDH1 mutant cells have increased sensitivity to complex I inhibitors.
Collapse
|
9
|
Sangster AG, Gooding RJ, Garven A, Ghaedi H, Berman DM, Davey SK. Mutually exclusive mutation profiles define functionally related genes in muscle invasive bladder cancer. PLoS One 2022; 17:e0259992. [PMID: 35073341 PMCID: PMC8786205 DOI: 10.1371/journal.pone.0259992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/31/2021] [Indexed: 11/18/2022] Open
Abstract
Muscle Invasive bladder cancer is known to have an abundance of mutations, particularly in DNA damage response and chromatin modification genes. The role of these mutations in the development and progression of the disease is not well understood. However, a mutually exclusive mutation pattern between gene pairs could suggest gene mutations of significance. For example, a mutually exclusive mutation pattern could suggest an epistatic relationship where the outcome of a mutation in one gene would have the same outcome as a mutation in a different gene. The significance of a mutually exclusive relationship was determined by establishing a normal distribution of the conditional probabilities for having a mutation in one gene and not the other as well as the reverse relationship for each gene pairing. Then these distributions were used to determine the sigma–magnitude of standard deviation by which the observed value differed from the expected, a value that can also be interpreted as the ‘p-value’. This approach led to the identification of mutually exclusive mutation patterns in KDM6A and KMT2D as well as KDM6A and RB1 that suggested the observed mutation pattern did not happen by chance. Upon further investigation of these genes and their interactions, a potential similar outcome was identified that supports the concept of epistasis. Knowledge of these mutational interactions provides a better understanding of the mechanisms underlying muscle invasive bladder cancer development, and may direct therapeutic development exploiting genotoxic chemotherapy and synthetic lethality in these pathways.
Collapse
Affiliation(s)
- Ami G. Sangster
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen’s University Cancer Research Institute, Kingston, Ontario, Canada
| | - Robert J. Gooding
- Department of Physics, Queen’s University, Kingston, Ontario, Canada
| | - Andrew Garven
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen’s University Cancer Research Institute, Kingston, Ontario, Canada
| | - Hamid Ghaedi
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen’s University Cancer Research Institute, Kingston, Ontario, Canada
| | - David M. Berman
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen’s University Cancer Research Institute, Kingston, Ontario, Canada
| | - Scott K. Davey
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen’s University Cancer Research Institute, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
10
|
Ahmed R, Erten C, Houdjedj A, Kazan H, Yalcin C. A Network-Centric Framework for the Evaluation of Mutual Exclusivity Tests on Cancer Drivers. Front Genet 2021; 12:746495. [PMID: 34899838 PMCID: PMC8664367 DOI: 10.3389/fgene.2021.746495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/27/2021] [Indexed: 12/03/2022] Open
Abstract
One of the key concepts employed in cancer driver gene identification is that of mutual exclusivity (ME); a driver mutation is less likely to occur in case of an earlier mutation that has common functionality in the same molecular pathway. Several ME tests have been proposed recently, however the current protocols to evaluate ME tests have two main limitations. Firstly the evaluations are mostly with respect to simulated data and secondly the evaluation metrics lack a network-centric view. The latter is especially crucial as the notion of common functionality can be achieved through searching for interaction patterns in relevant networks. We propose a network-centric framework to evaluate the pairwise significances found by statistical ME tests. It has three main components. The first component consists of metrics employed in the network-centric ME evaluations. Such metrics are designed so that network knowledge and the reference set of known cancer genes are incorporated in ME evaluations under a careful definition of proper control groups. The other two components are designed as further mechanisms to avoid confounders inherent in ME detection on top of the network-centric view. To this end, our second objective is to dissect the side effects caused by mutation load artifacts where mutations driving tumor subtypes with low mutation load might be incorrectly diagnosed as mutually exclusive. Finally, as part of the third main component, the confounding issue stemming from the use of nonspecific interaction networks generated as combinations of interactions from different tissues is resolved through the creation and use of tissue-specific networks in the proposed framework. The data, the source code and useful scripts are available at: https://github.com/abu-compbio/NetCentric.
Collapse
Affiliation(s)
- Rafsan Ahmed
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Antalya, Turkey
| | - Cesim Erten
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Aissa Houdjedj
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Cansu Yalcin
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| |
Collapse
|
11
|
Cutigi JF, Evangelista AF, Reis RM, Simao A. A computational approach for the discovery of significant cancer genes by weighted mutation and asymmetric spreading strength in networks. Sci Rep 2021; 11:23551. [PMID: 34876593 PMCID: PMC8651746 DOI: 10.1038/s41598-021-02671-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022] Open
Abstract
Identifying significantly mutated genes in cancer is essential for understanding the mechanisms of tumor initiation and progression. This task is a key challenge since large-scale genomic studies have reported an endless number of genes mutated at a shallow frequency. Towards uncovering infrequently mutated genes, gene interaction networks combined with mutation data have been explored. This work proposes Discovering Significant Cancer Genes (DiSCaGe), a computational method for discovering significant genes for cancer. DiSCaGe computes a mutation score for the genes based on the type of mutations they have. The influence received for their neighbors in the network is also considered and obtained through an asymmetric spreading strength applied to a consensus gene network. DiSCaGe produces a ranking of prioritized possible cancer genes. An experimental evaluation with six types of cancer revealed the potential of DiSCaGe for discovering known and possible novel significant cancer genes.
Collapse
Affiliation(s)
- Jorge Francisco Cutigi
- Federal Institute of Sao Paulo, Sao Carlos, SP, Brazil.
- University of Sao Paulo, Sao Carlos, SP, Brazil.
| | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | | |
Collapse
|
12
|
Fedrizzi T, Ciani Y, Lorenzin F, Cantore T, Gasperini P, Demichelis F. Fast mutual exclusivity algorithm nominates potential synthetic lethal gene pairs through brute force matrix product computations. Comput Struct Biotechnol J 2021; 19:4394-4403. [PMID: 34429855 PMCID: PMC8369001 DOI: 10.1016/j.csbj.2021.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Mutual Exclusivity analysis of genomic aberrations contributes to the exploration of potential synthetic lethal (SL) relationships thus guiding the nomination of specific cancer cells vulnerabilities. When multiple classes of genomic aberrations and large cohorts of patients are interrogated, exhaustive genome-wide analyses are not computationally feasible with commonly used approaches. Here we present Fast Mutual Exclusivity (FaME), an algorithm based on matrix multiplication that employs a logarithm-based implementation of the Fisher's exact test to achieve fast computation of genome-wide mutual exclusivity tests; we show that brute force testing for mutual exclusivity of hundreds of millions of aberrations combinations can be performed in few minutes. We applied FaME to allele-specific data from whole exome experiments of 27 TCGA studies cohorts, detecting both mutual exclusivity of point mutations, as well as allele-specific copy number signals that span sets of contiguous cytobands. We next focused on a case study involving the loss of tumor suppressors and druggable genes while exploiting an integrated analysis of both public cell lines loss of function screens data and patients' transcriptomic profiles. FaME algorithm implementation as well as allele-specific analysis output are publicly available at https://github.com/demichelislab/FaME.
Collapse
Affiliation(s)
- Tarcisio Fedrizzi
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Francesca Lorenzin
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Thomas Cantore
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Paola Gasperini
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Al-Saud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
13
|
Liu S, Liu J, Xie Y, Zhai T, Hinderer EW, Stromberg AJ, Vanderford NL, Kolesar JM, Moseley HNB, Chen L, Liu C, Wang C. MEScan: a powerful statistical framework for genome-scale mutual exclusivity analysis of cancer mutations. Bioinformatics 2021; 37:1189-1197. [PMID: 33165532 PMCID: PMC8189684 DOI: 10.1093/bioinformatics/btaa957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 03/20/2020] [Accepted: 10/31/2020] [Indexed: 12/17/2022] Open
Abstract
MOTIVATION Cancer somatic driver mutations associated with genes within a pathway often show a mutually exclusive pattern across a cohort of patients. This mutually exclusive mutational signal has been frequently used to distinguish driver from passenger mutations and to investigate relationships among driver mutations. Current methods for de novo discovery of mutually exclusive mutational patterns are limited because the heterogeneity in background mutation rate can confound mutational patterns, and the presence of highly mutated genes can lead to spurious patterns. In addition, most methods only focus on a limited number of pre-selected genes and are unable to perform genome-wide analysis due to computational inefficiency. RESULTS We introduce a statistical framework, MEScan, for accurate and efficient mutual exclusivity analysis at the genomic scale. Our framework contains a fast and powerful statistical test for mutual exclusivity with adjustment of the background mutation rate and impact of highly mutated genes, and a multi-step procedure for genome-wide screening with the control of false discovery rate. We demonstrate that MEScan more accurately identifies mutually exclusive gene sets than existing methods and is at least two orders of magnitude faster than most methods. By applying MEScan to data from four different cancer types and pan-cancer, we have identified several biologically meaningful mutually exclusive gene sets. AVAILABILITY AND IMPLEMENTATION MEScan is available as an R package at https://github.com/MarkeyBBSRF/MEScan. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yanqi Xie
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Tingting Zhai
- Department of Statistics, University of Kentucky, Lexington, KY 40536, USA
| | - Eugene W Hinderer
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Arnold J Stromberg
- Department of Statistics, University of Kentucky, Lexington, KY 40536, USA
| | - Nathan L Vanderford
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jill M Kolesar
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Department of Pharmacy Practice and Science, University of Kentucky, Lexington, KY 40536, USA
| | - Hunter N B Moseley
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Li Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Chunming Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.,Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
14
|
Liang L, Zhu K, Tao J, Lu S. ORN: Inferring patient-specific dysregulation status of pathway modules in cancer with OR-gate Network. PLoS Comput Biol 2021; 17:e1008792. [PMID: 33819263 PMCID: PMC8049496 DOI: 10.1371/journal.pcbi.1008792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 04/15/2021] [Accepted: 02/15/2021] [Indexed: 01/26/2023] Open
Abstract
Pathway level understanding of cancer plays a key role in precision oncology. However, the current amount of high-throughput data cannot support the elucidation of full pathway topology. In this study, instead of directly learning the pathway network, we adapted the probabilistic OR gate to model the modular structure of pathways and regulon. The resulting model, OR-gate Network (ORN), can simultaneously infer pathway modules of somatic alterations, patient-specific pathway dysregulation status, and downstream regulon. In a trained ORN, the differentially expressed genes (DEGs) in each tumour can be explained by somatic mutations perturbing a pathway module. Furthermore, the ORN handles one of the most important properties of pathway perturbation in tumours, the mutual exclusivity. We have applied the ORN to lower-grade glioma (LGG) samples and liver hepatocellular carcinoma (LIHC) samples in TCGA and breast cancer samples from METABRIC. Both datasets have shown abnormal pathway activities related to immune response and cell cycles. In LGG samples, ORN identified pathway modules closely related to glioma development and revealed two pathways closely related to patient survival. We had similar results with LIHC samples. Additional results from the METABRIC datasets showed that ORN could characterize critical mechanisms of cancer and connect them to less studied somatic mutations (e.g., BAP1, MIR604, MICAL3, and telomere activities), which may generate novel hypothesis for targeted therapy.
Collapse
Affiliation(s)
- Lifan Liang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kunju Zhu
- Clinical Medicine Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
15
|
Cook JH, Melloni GEM, Gulhan DC, Park PJ, Haigis KM. The origins and genetic interactions of KRAS mutations are allele- and tissue-specific. Nat Commun 2021; 12:1808. [PMID: 33753749 PMCID: PMC7985210 DOI: 10.1038/s41467-021-22125-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Mutational activation of KRAS promotes the initiation and progression of cancers, especially in the colorectum, pancreas, lung, and blood plasma, with varying prevalence of specific activating missense mutations. Although epidemiological studies connect specific alleles to clinical outcomes, the mechanisms underlying the distinct clinical characteristics of mutant KRAS alleles are unclear. Here, we analyze 13,492 samples from these four tumor types to examine allele- and tissue-specific genetic properties associated with oncogenic KRAS mutations. The prevalence of known mutagenic mechanisms partially explains the observed spectrum of KRAS activating mutations. However, there are substantial differences between the observed and predicted frequencies for many alleles, suggesting that biological selection underlies the tissue-specific frequencies of mutant alleles. Consistent with experimental studies that have identified distinct signaling properties associated with each mutant form of KRAS, our genetic analysis reveals that each KRAS allele is associated with a distinct tissue-specific comutation network. Moreover, we identify tissue-specific genetic dependencies associated with specific mutant KRAS alleles. Overall, this analysis demonstrates that the genetic interactions of oncogenic KRAS mutations are allele- and tissue-specific, underscoring the complexity that drives their clinical consequences.
Collapse
Affiliation(s)
- Joshua H Cook
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Giorgio E M Melloni
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Doga C Gulhan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- Harvard Digestive Disease Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Cai Y, Xu G, Wu F, Michelini F, Chan C, Qu X, Selenica P, Ladewig E, Castel P, Cheng Y, Zhao A, Jhaveri K, Toska E, Jimenez M, Jacquet A, Tran-Dien A, Andre F, Chandarlapaty S, Reis-Filho JS, Razavi P, Scaltriti M. Genomic Alterations in PIK3CA-Mutated Breast Cancer Result in mTORC1 Activation and Limit the Sensitivity to PI3Kα Inhibitors. Cancer Res 2021; 81:2470-2480. [PMID: 33685991 DOI: 10.1158/0008-5472.can-20-3232] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/02/2021] [Accepted: 03/02/2021] [Indexed: 11/16/2022]
Abstract
PI3Kα inhibitors have shown clinical activity in PIK3CA-mutated estrogen receptor-positive (ER+) patients with breast cancer. Using whole genome CRISPR/Cas9 sgRNA knockout screens, we identified and validated several negative regulators of mTORC1 whose loss confers resistance to PI3Kα inhibition. Among the top candidates were TSC1, TSC2, TBC1D7, AKT1S1, STK11, MARK2, PDE7A, DEPDC5, NPRL2, NPRL3, C12orf66, SZT2, and ITFG2. Loss of these genes invariably results in sustained mTOR signaling under pharmacologic inhibition of the PI3K-AKT pathway. Moreover, resistance could be prevented or overcome by mTOR inhibition, confirming the causative role of sustained mTOR activity in limiting the sensitivity to PI3Kα inhibition. Cumulatively, genomic alterations affecting these genes are identified in about 15% of PIK3CA-mutated breast tumors and appear to be mutually exclusive. This study improves our understanding of the role of mTOR signaling restoration in leading to resistance to PI3Kα inhibition and proposes therapeutic strategies to prevent or revert this resistance. SIGNIFICANCE: These findings show that genetic lesions of multiple negative regulators of mTORC1 could limit the efficacy of PI3Kα inhibitors in breast cancer, which may guide patient selection strategies for future clinical trials.
Collapse
Affiliation(s)
- Yanyan Cai
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Guotai Xu
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,National Institute of Biological Sciences (NIBS), Beijing, China
| | - Fan Wu
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Flavia Michelini
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carmen Chan
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xuan Qu
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Erik Ladewig
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pau Castel
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Yuanming Cheng
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alison Zhao
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Komal Jhaveri
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eneda Toska
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, Maryland
| | | | | | - Alicia Tran-Dien
- INSERM UMR981 and Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Fabrice Andre
- INSERM UMR981 and Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Le Kremlin-Bicetre, France.,Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Sarat Chandarlapaty
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pedram Razavi
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maurizio Scaltriti
- Human Oncology & Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
17
|
Fang H, Zhang Z, Zhou Y, Jin L, Yang Y. A greedy approach for mutual exclusivity analysis in cancer study. Biostatistics 2021; 23:910-925. [PMID: 33634822 DOI: 10.1093/biostatistics/kxab004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/26/2020] [Accepted: 01/13/2021] [Indexed: 11/14/2022] Open
Abstract
The main challenge in cancer genomics is to distinguish the driver genes from passenger or neutral genes. Cancer genomes exhibit extensive mutational heterogeneity that no two genomes contain exactly the same somatic mutations. Such mutual exclusivity (ME) of mutations has been observed in cancer data and is associated with functional pathways. Analysis of ME patterns may provide useful clues to driver genes or pathways and may suggest novel understandings of cancer progression. In this article, we consider a probabilistic, generative model of ME, and propose a powerful and greedy algorithm to select the mutual exclusivity gene sets. The greedy method includes a pre-selection procedure and a stepwise forward algorithm which can significantly reduce computation time. Power calculations suggest that the new method is efficient and powerful for one ME set or multiple ME sets with overlapping genes. We illustrate this approach by analysis of the whole-exome sequencing data of cancer types from TCGA.
Collapse
Affiliation(s)
- Hongyan Fang
- School of Mathematical Sciences, Anhui University, Hefei, Anhui, China
| | - Zeyu Zhang
- Department of Statistics and Finance, University of Science and Technology of China, Hefei, Anhui, China
| | - Yinsheng Zhou
- Department of Statistics and Finance, University of Science and Technology of China, Hefei, Anhui, China
| | - Lishuai Jin
- School of Mathematical Sciences, Anhui University, Hefei, Anhui, China
| | - Yaning Yang
- Department of Statistics and Finance, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
18
|
A forward selection algorithm to identify mutually exclusive alterations in cancer studies. J Hum Genet 2020; 66:509-518. [PMID: 33177701 DOI: 10.1038/s10038-020-00870-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/11/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Mutual exclusivity analyses provide an effective tool to identify driver genes from passenger genes for cancer studies. Various algorithms have been developed for the detection of mutual exclusivity, but controlling false positive and improving accuracy remain challenging. We propose a forward selection algorithm for identification of mutually exclusive gene sets (FSME) in this paper. The method includes an initial search of seed pair of mutually exclusive (ME) genes and subsequently including more genes into the current ME set. Simulations demonstrated that, compared to recently published approaches (i.e., CoMEt, WExT, and MEGSA), FSME could provide higher precision or recall rate to identify ME gene sets, and had superior control of false positive rates. With application to TCGA real data sets for AML, BRCA, and GBM, we confirmed that FSME can be utilized to discover cancer driver genes.
Collapse
|
19
|
Völkel G, Laban S, Fürstberger A, Kühlwein SD, Ikonomi N, Hoffmann TK, Brunner C, Neuberg DS, Gaidzik V, Döhner H, Kraus JM, Kestler HA. Analysis, identification and visualization of subgroups in genomics. Brief Bioinform 2020; 22:5909009. [PMID: 32954413 PMCID: PMC8138884 DOI: 10.1093/bib/bbaa217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Motivation Cancer is a complex and heterogeneous disease involving multiple somatic mutations that accumulate during its progression. In the past years, the wide availability of genomic data from patients’ samples opened new perspectives in the analysis of gene mutations and alterations. Hence, visualizing and further identifying genes mutated in massive sets of patients are nowadays a critical task that sheds light on more personalized intervention approaches. Results Here, we extensively review existing tools for visualization and analysis of alteration data. We compare different approaches to study mutual exclusivity and sample coverage in large-scale omics data. We complement our review with the standalone software AVAtar (‘analysis and visualization of alteration data’) that integrates diverse aspects known from different tools into a comprehensive platform. AVAtar supplements customizable alteration plots by a multi-objective evolutionary algorithm for subset identification and provides an innovative and user-friendly interface for the evaluation of concurrent solutions. A use case from personalized medicine demonstrates its unique features showing an application on vaccination target selection. Availability AVAtar is available at: https://github.com/sysbio-bioinf/avatar Contact hans.kestler@uni-ulm.de, phone: +49 (0) 731 500 24 500, fax: +49 (0) 731 500 24 502
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas K Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Germany
| | - Donna S Neuberg
- Department of Biostatistics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Verena Gaidzik
- Department of Internal Medicine III, Ulm University Medical Center, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Medical Center, Germany
| | | | | |
Collapse
|
20
|
Kim JY, Choi JK, Jung H. Genome-wide methylation patterns predict clinical benefit of immunotherapy in lung cancer. Clin Epigenetics 2020; 12:119. [PMID: 32762727 PMCID: PMC7410160 DOI: 10.1186/s13148-020-00907-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background It is crucial to unravel molecular determinants of responses to immune checkpoint blockade (ICB) therapy because only a small subset of advanced non-small cell lung cancer (NSCLC) patients responds to ICB therapy. Previous studies were concentrated on genomic and transcriptomic markers (e.g., mutation burden and immune gene expression). However, these markers are not sufficient to accurately predict a response to ICB therapy. Results Here, we analyzed DNA methylomes of 141 advanced NSCLC samples subjected to ICB therapy (i.e., anti-programmed death-1) from two independent cohorts (60 and 81 patients from our and IDIBELL cohorts). Integrative analysis of patients with matched transcriptome data in our cohort (n = 28) at pathway level revealed significant overlaps between promoter hypermethylation and transcriptional repression in nonresponders relative to responders. Fifteen immune-related pathways, including interferon signaling, were identified to be enriched for both hypermethylation and repression. We built a reliable prognostic risk model based on eight genes using LASSO model and successfully validated the model in independent cohorts. Furthermore, we found 30 survival-associated molecular interaction networks, in which two or three hypermethylated genes showed significant mutual exclusion across nonresponders. Conclusions Our study demonstrates that methylation patterns can provide insight into molecular determinants underlying the clinical benefit of ICB therapy.
Collapse
Affiliation(s)
- Jeong Yeon Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Penta Medix Co., Ltd., Seongnam-si, Gyeongi-do, 13449, Republic of Korea.
| | - Hyunchul Jung
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Cancer Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Cambridge, UK.
| |
Collapse
|
21
|
Cutigi JF, Evangelista AF, Simao A. Approaches for the identification of driver mutations in cancer: A tutorial from a computational perspective. J Bioinform Comput Biol 2020; 18:2050016. [PMID: 32698724 DOI: 10.1142/s021972002050016x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer is a complex disease caused by the accumulation of genetic alterations during the individual's life. Such alterations are called genetic mutations and can be divided into two groups: (1) Passenger mutations, which are not responsible for cancer and (2) Driver mutations, which are significant for cancer and responsible for its initiation and progression. Cancer cells undergo a large number of mutations, of which most are passengers, and few are drivers. The identification of driver mutations is a key point and one of the biggest challenges in Cancer Genomics. Many computational methods for such a purpose have been developed in Cancer Bioinformatics. Such computational methods are complex and are usually described in a high level of abstraction. This tutorial details some classical computational methods, from a computational perspective, with the transcription in an algorithmic format towards an easy access by researchers.
Collapse
Affiliation(s)
- Jorge Francisco Cutigi
- Federal Institute of São Paulo (IFSP), São Carlos, SP, Brazil.,University of São Paulo (USP), São Carlos, SP, Brazil
| | | | | |
Collapse
|
22
|
van de Haar J, Canisius S, Yu MK, Voest EE, Wessels LFA, Ideker T. Identifying Epistasis in Cancer Genomes: A Delicate Affair. Cell 2020; 177:1375-1383. [PMID: 31150618 DOI: 10.1016/j.cell.2019.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/04/2019] [Accepted: 04/30/2019] [Indexed: 12/30/2022]
Abstract
Recent studies of the tumor genome seek to identify cancer pathways as groups of genes in which mutations are epistatic with one another or, specifically, "mutually exclusive." Here, we show that most mutations are mutually exclusive not due to pathway structure but to interactions with disease subtype and tumor mutation load. In particular, many cancer driver genes are mutated preferentially in tumors with few mutations overall, causing mutations in these cancer genes to appear mutually exclusive with numerous others. Researchers should view current epistasis maps with caution until we better understand the multiple cause-and-effect relationships among factors such as tumor subtype, positive selection for mutations, and gross tumor characteristics including mutational signatures and load.
Collapse
Affiliation(s)
- Joris van de Haar
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands; Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sander Canisius
- Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael K Yu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Program in Bioinformatics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emile E Voest
- Division of Molecular Oncology & Immunology, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, the Netherlands Cancer Institute, Amsterdam, 1066 CX, the Netherlands; Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Faculty of EEMCS, Delft University of Technology, Delft, 2628 CD, the Netherlands.
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Program in Bioinformatics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Cancer Cell Map Initiative, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Al Hajri Q, Dash S, Feng WC, Garner HR, Anandakrishnan R. Identifying multi-hit carcinogenic gene combinations: Scaling up a weighted set cover algorithm using compressed binary matrix representation on a GPU. Sci Rep 2020; 10:2022. [PMID: 32029803 PMCID: PMC7005272 DOI: 10.1038/s41598-020-58785-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/20/2020] [Indexed: 01/16/2023] Open
Abstract
Despite decades of research, effective treatments for most cancers remain elusive. One reason is that different instances of cancer result from different combinations of multiple genetic mutations (hits). Therefore, treatments that may be effective in some cases are not effective in others. We previously developed an algorithm for identifying combinations of carcinogenic genes with mutations (multi-hit combinations), which could suggest a likely cause for individual instances of cancer. Most cancers are estimated to require three or more hits. However, the computational complexity of the algorithm scales exponentially with the number of hits, making it impractical for identifying combinations of more than two hits. To identify combinations of greater than two hits, we used a compressed binary matrix representation, and optimized the algorithm for parallel execution on an NVIDIA V100 graphics processing unit (GPU). With these enhancements, the optimized GPU implementation was on average an estimated 12,144 times faster than the original integer matrix based CPU implementation, for the 3-hit algorithm, allowing us to identify 3-hit combinations. The 3-hit combinations identified using a training set were able to differentiate between tumor and normal samples in a separate test set with 90% overall sensitivity and 93% overall specificity. We illustrate how the distribution of mutations in tumor and normal samples in the multi-hit gene combinations can suggest potential driver mutations for further investigation. With experimental validation, these combinations may provide insight into the etiology of cancer and a rational basis for targeted combination therapy.
Collapse
Affiliation(s)
- Qais Al Hajri
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Sajal Dash
- Department of Computer Science, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Wu-Chun Feng
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, 24060, USA
- Department of Computer Science, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Harold R Garner
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, 24060, USA
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, 29303, USA
| | - Ramu Anandakrishnan
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, 24060, USA.
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, 29303, USA.
| |
Collapse
|
24
|
Calabrese C, Davidson NR, Demircioğlu D, Fonseca NA, He Y, Kahles A, Lehmann KV, Liu F, Shiraishi Y, Soulette CM, Urban L, Greger L, Li S, Liu D, Perry MD, Xiang Q, Zhang F, Zhang J, Bailey P, Erkek S, Hoadley KA, Hou Y, Huska MR, Kilpinen H, Korbel JO, Marin MG, Markowski J, Nandi T, Pan-Hammarström Q, Pedamallu CS, Siebert R, Stark SG, Su H, Tan P, Waszak SM, Yung C, Zhu S, Awadalla P, Creighton CJ, Meyerson M, Ouellette BFF, Wu K, Yang H, Brazma A, Brooks AN, Göke J, Rätsch G, Schwarz RF, Stegle O, Zhang Z. Genomic basis for RNA alterations in cancer. Nature 2020; 578:129-136. [PMID: 32025019 PMCID: PMC7054216 DOI: 10.1038/s41586-020-1970-0] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 12/11/2019] [Indexed: 01/27/2023]
Abstract
Transcript alterations often result from somatic changes in cancer genomes1. Various forms of RNA alterations have been described in cancer, including overexpression2, altered splicing3 and gene fusions4; however, it is difficult to attribute these to underlying genomic changes owing to heterogeneity among patients and tumour types, and the relatively small cohorts of patients for whom samples have been analysed by both transcriptome and whole-genome sequencing. Here we present, to our knowledge, the most comprehensive catalogue of cancer-associated gene alterations to date, obtained by characterizing tumour transcriptomes from 1,188 donors of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA)5. Using matched whole-genome sequencing data, we associated several categories of RNA alterations with germline and somatic DNA alterations, and identified probable genetic mechanisms. Somatic copy-number alterations were the major drivers of variations in total gene and allele-specific expression. We identified 649 associations of somatic single-nucleotide variants with gene expression in cis, of which 68.4% involved associations with flanking non-coding regions of the gene. We found 1,900 splicing alterations associated with somatic mutations, including the formation of exons within introns in proximity to Alu elements. In addition, 82% of gene fusions were associated with structural variants, including 75 of a new class, termed 'bridged' fusions, in which a third genomic location bridges two genes. We observed transcriptomic alteration signatures that differ between cancer types and have associations with variations in DNA mutational signatures. This compendium of RNA alterations in the genomic context provides a rich resource for identifying genes and mechanisms that are functionally implicated in cancer.
Collapse
Affiliation(s)
| | - Claudia Calabrese
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Natalie R. Davidson
- 0000 0001 2156 2780grid.5801.cETH Zurich, Zurich, Switzerland ,0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY USA ,000000041936877Xgrid.5386.8Weill Cornell Medical College, New York, NY USA ,0000 0001 2223 3006grid.419765.8SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland ,0000 0004 0478 9977grid.412004.3University Hospital Zurich, Zurich, Switzerland
| | - Deniz Demircioğlu
- 0000 0001 2180 6431grid.4280.eNational University of Singapore, Singapore, Singapore ,0000 0004 0620 715Xgrid.418377.eGenome Institute of Singapore, Singapore, Singapore
| | - Nuno A. Fonseca
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Yao He
- 0000 0001 2256 9319grid.11135.37Peking University, Beijing, China
| | - André Kahles
- 0000 0001 2156 2780grid.5801.cETH Zurich, Zurich, Switzerland ,0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY USA ,0000 0001 2223 3006grid.419765.8SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland ,0000 0004 0478 9977grid.412004.3University Hospital Zurich, Zurich, Switzerland
| | - Kjong-Van Lehmann
- 0000 0001 2156 2780grid.5801.cETH Zurich, Zurich, Switzerland ,0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY USA ,0000 0001 2223 3006grid.419765.8SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland ,0000 0004 0478 9977grid.412004.3University Hospital Zurich, Zurich, Switzerland
| | - Fenglin Liu
- 0000 0001 2256 9319grid.11135.37Peking University, Beijing, China
| | - Yuichi Shiraishi
- 0000 0001 2151 536Xgrid.26999.3dThe University of Tokyo, Minato-ku, Japan
| | - Cameron M. Soulette
- 0000 0001 0740 6917grid.205975.cUniversity of California, Santa Cruz, Santa Cruz, CA USA
| | - Lara Urban
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Liliana Greger
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Siliang Li
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Dongbing Liu
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Marc D. Perry
- 0000 0004 0626 690Xgrid.419890.dOntario Institute for Cancer Research, Toronto, Ontario, Canada ,0000 0001 2297 6811grid.266102.1University of California, San Francisco, San Francisco, CA USA
| | - Qian Xiang
- 0000 0004 0626 690Xgrid.419890.dOntario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Fan Zhang
- 0000 0001 2256 9319grid.11135.37Peking University, Beijing, China
| | - Junjun Zhang
- 0000 0004 0626 690Xgrid.419890.dOntario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Peter Bailey
- 0000 0001 2193 314Xgrid.8756.cUniversity of Glasgow, Glasgow, UK
| | - Serap Erkek
- 0000 0004 0495 846Xgrid.4709.aEuropean Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Katherine A. Hoadley
- 0000000122483208grid.10698.36The University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Yong Hou
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Matthew R. Huska
- 0000 0001 1014 0849grid.419491.0Berlin Institute for Medical Systems Biology, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Helena Kilpinen
- 0000000121901201grid.83440.3bUniversity College London, London, UK
| | - Jan O. Korbel
- 0000 0004 0495 846Xgrid.4709.aEuropean Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Maximillian G. Marin
- 0000 0001 0740 6917grid.205975.cUniversity of California, Santa Cruz, Santa Cruz, CA USA
| | - Julia Markowski
- 0000 0001 1014 0849grid.419491.0Berlin Institute for Medical Systems Biology, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Tannistha Nandi
- 0000 0004 0620 715Xgrid.418377.eGenome Institute of Singapore, Singapore, Singapore
| | - Qiang Pan-Hammarström
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,0000 0004 1937 0626grid.4714.6Karolinska Institutet, Stockholm, Sweden
| | - Chandra Sekhar Pedamallu
- grid.66859.34Broad Institute, Cambridge, MA USA ,0000 0001 2106 9910grid.65499.37Dana-Farber Cancer Institute, Boston, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - Reiner Siebert
- grid.410712.1Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Stefan G. Stark
- 0000 0001 2156 2780grid.5801.cETH Zurich, Zurich, Switzerland ,0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY USA ,0000 0001 2223 3006grid.419765.8SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland ,0000 0004 0478 9977grid.412004.3University Hospital Zurich, Zurich, Switzerland
| | - Hong Su
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Patrick Tan
- 0000 0004 0620 715Xgrid.418377.eGenome Institute of Singapore, Singapore, Singapore ,0000 0004 0385 0924grid.428397.3Duke-NUS Medical School, Singapore, Singapore
| | - Sebastian M. Waszak
- 0000 0004 0495 846Xgrid.4709.aEuropean Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Christina Yung
- 0000 0004 0626 690Xgrid.419890.dOntario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Shida Zhu
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Philip Awadalla
- 0000 0004 0626 690Xgrid.419890.dOntario Institute for Cancer Research, Toronto, Ontario, Canada ,0000 0001 2157 2938grid.17063.33University of Toronto, Toronto, Ontario Canada
| | - Chad J. Creighton
- 0000 0001 2160 926Xgrid.39382.33Baylor College of Medicine, Houston, TX USA
| | - Matthew Meyerson
- grid.66859.34Broad Institute, Cambridge, MA USA ,0000 0001 2106 9910grid.65499.37Dana-Farber Cancer Institute, Boston, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | | | - Kui Wu
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China ,China National GeneBank-Shenzhen, Shenzhen, China
| | - Huanming Yang
- 0000 0001 2034 1839grid.21155.32BGI-Shenzhen, Shenzhen, China
| | | | - Alvis Brazma
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Angela N. Brooks
- 0000 0001 0740 6917grid.205975.cUniversity of California, Santa Cruz, Santa Cruz, CA USA ,grid.66859.34Broad Institute, Cambridge, MA USA ,0000 0001 2106 9910grid.65499.37Dana-Farber Cancer Institute, Boston, MA USA
| | - Jonathan Göke
- 0000 0004 0620 715Xgrid.418377.eGenome Institute of Singapore, Singapore, Singapore ,0000 0004 0620 9745grid.410724.4National Cancer Centre Singapore, Singapore, Singapore
| | - Gunnar Rätsch
- 0000 0001 2156 2780grid.5801.cETH Zurich, Zurich, Switzerland ,0000 0001 2171 9952grid.51462.34Memorial Sloan Kettering Cancer Center, New York, NY USA ,000000041936877Xgrid.5386.8Weill Cornell Medical College, New York, NY USA ,0000 0001 2223 3006grid.419765.8SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland ,0000 0004 0478 9977grid.412004.3University Hospital Zurich, Zurich, Switzerland
| | - Roland F. Schwarz
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK ,0000 0001 1014 0849grid.419491.0Berlin Institute for Medical Systems Biology, Max Delbruck Center for Molecular Medicine, Berlin, Germany ,0000 0004 0492 0584grid.7497.dGerman Cancer Consortium (DKTK), partner site Berlin, Germany ,0000 0004 0492 0584grid.7497.dGerman Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Stegle
- 0000 0000 9709 7726grid.225360.0European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK ,0000 0004 0495 846Xgrid.4709.aEuropean Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany ,0000 0004 0492 0584grid.7497.dGerman Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zemin Zhang
- 0000 0001 2256 9319grid.11135.37Peking University, Beijing, China
| | | |
Collapse
|
25
|
Wang M, Yu T, Liu J, Chen L, Stromberg AJ, Villano JL, Arnold SM, Liu C, Wang C. A probabilistic method for leveraging functional annotations to enhance estimation of the temporal order of pathway mutations during carcinogenesis. BMC Bioinformatics 2019; 20:620. [PMID: 31791231 PMCID: PMC6889196 DOI: 10.1186/s12859-019-3218-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 11/12/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Cancer arises through accumulation of somatically acquired genetic mutations. An important question is to delineate the temporal order of somatic mutations during carcinogenesis, which contributes to better understanding of cancer biology and facilitates identification of new therapeutic targets. Although a number of statistical and computational methods have been proposed to estimate the temporal order of mutations, they do not account for the differences in the functional impacts of mutations and thus are likely to be obscured by the presence of passenger mutations that do not contribute to cancer progression. In addition, many methods infer the order of mutations at the gene level, which have limited power due to the low mutation rate in most genes. RESULTS In this paper, we develop a Probabilistic Approach for estimating the Temporal Order of Pathway mutations by leveraging functional Annotations of mutations (PATOPA). PATOPA infers the order of mutations at the pathway level, wherein it uses a probabilistic method to characterize the likelihood of mutational events from different pathways occurring in a certain order. The functional impact of each mutation is incorporated to weigh more on a mutation that is more integral to tumor development. A maximum likelihood method is used to estimate parameters and infer the probability of one pathway being mutated prior to another. Simulation studies and analysis of whole exome sequencing data from The Cancer Genome Atlas (TCGA) demonstrate that PATOPA is able to accurately estimate the temporal order of pathway mutations and provides new biological insights on carcinogenesis of colorectal and lung cancers. CONCLUSIONS PATOPA provides a useful tool to estimate temporal order of mutations at the pathway level while leveraging functional annotations of mutations.
Collapse
Affiliation(s)
- Menghan Wang
- Department of Statistics, University of Kentucky, Lexington, USA
| | - Tianxin Yu
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, USA
| | - Li Chen
- Markey Cancer Center, University of Kentucky, Lexington, USA
- Department of Biostatistics, University of Kentucky, Lexington, USA
| | | | - John L. Villano
- Markey Cancer Center, University of Kentucky, Lexington, USA
- Department of Internal Medicine, University of Kentucky, Lexington, USA
| | - Susanne M. Arnold
- Markey Cancer Center, University of Kentucky, Lexington, USA
- Department of Internal Medicine, University of Kentucky, Lexington, USA
| | - Chunming Liu
- Markey Cancer Center, University of Kentucky, Lexington, USA
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, USA
- Department of Biostatistics, University of Kentucky, Lexington, USA
| |
Collapse
|
26
|
Reyna MA, Leiserson MDM, Raphael BJ. Hierarchical HotNet: identifying hierarchies of altered subnetworks. Bioinformatics 2019; 34:i972-i980. [PMID: 30423088 PMCID: PMC6129270 DOI: 10.1093/bioinformatics/bty613] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Motivation The analysis of high-dimensional ‘omics data is often informed by the use of biological interaction networks. For example, protein–protein interaction networks have been used to analyze gene expression data, to prioritize germline variants, and to identify somatic driver mutations in cancer. In these and other applications, the underlying computational problem is to identify altered subnetworks containing genes that are both highly altered in an ‘omics dataset and are topologically close (e.g. connected) on an interaction network. Results We introduce Hierarchical HotNet, an algorithm that finds a hierarchy of altered subnetworks. Hierarchical HotNet assesses the statistical significance of the resulting subnetworks over a range of biological scales and explicitly controls for ascertainment bias in the network. We evaluate the performance of Hierarchical HotNet and several other algorithms that identify altered subnetworks on the problem of predicting cancer genes and significantly mutated subnetworks. On somatic mutation data from The Cancer Genome Atlas, Hierarchical HotNet outperforms other methods and identifies significantly mutated subnetworks containing both well-known cancer genes and candidate cancer genes that are rarely mutated in the cohort. Hierarchical HotNet is a robust algorithm for identifying altered subnetworks across different ‘omics datasets. Availability and implementation http://github.com/raphael-group/hierarchical-hotnet. Supplementary information Supplementary material are available at Bioinformatics online.
Collapse
Affiliation(s)
- Matthew A Reyna
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Mark D M Leiserson
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| |
Collapse
|
27
|
Deng Y, Luo S, Deng C, Luo T, Yin W, Zhang H, Zhang Y, Zhang X, Lan Y, Ping Y, Xiao Y, Li X. Identifying mutual exclusivity across cancer genomes: computational approaches to discover genetic interaction and reveal tumor vulnerability. Brief Bioinform 2019; 20:254-266. [PMID: 28968730 DOI: 10.1093/bib/bbx109] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Indexed: 02/06/2023] Open
Abstract
Systematic sequencing of cancer genomes has revealed prevalent heterogeneity, with patients harboring various combinatorial patterns of genetic alteration. In particular, a phenomenon that a group of genes exhibits mutually exclusive patterns has been widespread across cancers, covering a broad spectrum of crucial cancer pathways. Recently, there is considerable evidence showing that, mutual exclusivity reflects alternative functions in tumor initiation and progression, or suggests adverse effects of their concurrence. Given its importance, numerous computational approaches have been proposed to study mutual exclusivity using genomic profiles alone, or by integrating networks and phenotypes. Some of them have been routinely used to explore genetic associations, which lead to a deeper understanding of carcinogenic mechanisms and reveals unexpected tumor vulnerabilities. Here, we present an overview of mutual exclusivity from the perspective of cancer genome. We describe the common hypothesis underlying mutual exclusivity, summarize the strategies for the identification of significant mutually exclusive patterns, compare the performance of representative algorithms from simulated data sets and discuss their common confounders.
Collapse
Affiliation(s)
- Yulan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shangyi Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chunyu Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tao Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenkang Yin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyi Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
28
|
Gao B, Zhao Y, Li Y, Liu J, Wang L, Li G, Su Z. Prediction of Driver Modules via Balancing Exclusive Coverages of Mutations in Cancer Samples. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801384. [PMID: 30828525 PMCID: PMC6382311 DOI: 10.1002/advs.201801384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/04/2018] [Indexed: 05/07/2023]
Abstract
Mutual exclusivity of cancer driving mutations is a frequently observed phenomenon in the mutational landscape of cancer. The long tail of rare mutations complicates the discovery of mutually exclusive driver modules. The existing methods usually suffer from the problem that only few genes in some identified modules cover most of the cancer samples. To overcome this hurdle, an efficient method UniCovEx is presented via identifying mutually exclusive driver modules of balanced exclusive coverages. UniCovEx first searches for candidate driver modules with a strong topological relationship in signaling networks using a greedy strategy. It then evaluates the candidate modules by considering their coverage, exclusivity, and balance of coverage, using a novel metric termed exclusive entropy of modules, which measures how balanced the modules are. Finally, UniCovEx predicts sample-specific driver modules by solving a minimum set cover problem using a greedy strategy. When tested on 12 The Cancer Genome Atlas datasets of different cancer types, UniCovEx shows a significant superiority over the previous methods. The software is available at: https://sourceforge.net/projects/cancer-pathway/files/.
Collapse
Affiliation(s)
- Bo Gao
- School of MathematicsShandong UniversityJinan250100China
- State Key Laboratory of Microbial TechnologyShandong UniversityJinan250100China
| | - Yue Zhao
- IAMMADISNCMISAcademy of Mathematics and Systems ScienceChinese Academy of SciencesBeijing100190China
- School of Mathematical SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yang Li
- School of MathematicsShandong UniversityJinan250100China
- State Key Laboratory of Microbial TechnologyShandong UniversityJinan250100China
| | - Juntao Liu
- School of MathematicsShandong UniversityJinan250100China
| | - Lushan Wang
- State Key Laboratory of Microbial TechnologyShandong UniversityJinan250100China
| | - Guojun Li
- School of MathematicsShandong UniversityJinan250100China
- State Key Laboratory of Microbial TechnologyShandong UniversityJinan250100China
| | - Zhengchang Su
- Department of Bioinformatics and GenomicsCollege of Computing and InformaticsThe University of North Carolina at Charlotte9201 University City BlvdCharlotteNC28223USA
| |
Collapse
|
29
|
Ruffalo M, Thomas R, Chen J, Lee AV, Oesterreich S, Bar-Joseph Z. Network-guided prediction of aromatase inhibitor response in breast cancer. PLoS Comput Biol 2019; 15:e1006730. [PMID: 30742607 PMCID: PMC6386390 DOI: 10.1371/journal.pcbi.1006730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 02/22/2019] [Accepted: 12/19/2018] [Indexed: 01/07/2023] Open
Abstract
Prediction of response to specific cancer treatments is complicated by significant heterogeneity between tumors in terms of mutational profiles, gene expression, and clinical measures. Here we focus on the response of Estrogen Receptor (ER)+ post-menopausal breast cancer tumors to aromatase inhibitors (AI). We use a network smoothing algorithm to learn novel features that integrate several types of high throughput data and new cell line experiments. These features greatly improve the ability to predict response to AI when compared to prior methods. For a subset of the patients, for which we obtained more detailed clinical information, we can further predict response to a specific AI drug. Breast cancer is the second most common type of cancer in women, with an incidence rate of over 250,000 cases per year, and breast cancer cases show significant heterogeneity in clinical and omic measures. Estrogen receptor positive (ER+) tumors typically grow in response to estrogen, and in post menopausal women, estrogen is only produced in peripheral tissues via the aromatase enzyme. Inhibition of aromatase is often an effective treatment for ER+ tumors, but aromatase inhibitor therapy is not effective for all tumors, and causes of this heterogeneity in response are largely not known. In this work, we present a feature construction and classification method to predict response to aromatase inhibitor therapy. We use network smoothing techniques to combine tumor omic data into predictive features, which we use as input to standard machine learning algorithms. We train predictive models using clinical data, including high-quality clinical data from UPMC patients, and show that our method outperforms previous approaches in predicting response to aromatase inhibitor therapy.
Collapse
Affiliation(s)
- Matthew Ruffalo
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Roby Thomas
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Jian Chen
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Adrian V. Lee
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Steffi Oesterreich
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
Dash S, Kinney NA, Varghese RT, Garner HR, Feng WC, Anandakrishnan R. Differentiating between cancer and normal tissue samples using multi-hit combinations of genetic mutations. Sci Rep 2019; 9:1005. [PMID: 30700767 PMCID: PMC6353925 DOI: 10.1038/s41598-018-37835-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/14/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer is known to result from a combination of a small number of genetic defects. However, the specific combinations of mutations responsible for the vast majority of cancers have not been identified. Current computational approaches focus on identifying driver genes and mutations. Although individually these mutations can increase the risk of cancer they do not result in cancer without additional mutations. We present a fundamentally different approach for identifying the cause of individual instances of cancer: we search for combinations of genes with carcinogenic mutations (multi-hit combinations) instead of individual driver genes or mutations. We developed an algorithm that identified a set of multi-hit combinations that differentiate between tumor and normal tissue samples with 91% sensitivity (95% Confidence Interval (CI) = 89-92%) and 93% specificity (95% CI = 91-94%) on average for seventeen cancer types. We then present an approach based on mutational profile that can be used to distinguish between driver and passenger mutations within these genes. These combinations, with experimental validation, can aid in better diagnosis, provide insights into the etiology of cancer, and provide a rational basis for designing targeted combination therapies.
Collapse
Affiliation(s)
- Sajal Dash
- Department of Computer Science, Virginia Tech, Blacksburg, VA, USA
| | - Nicholas A Kinney
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - Robin T Varghese
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - Harold R Garner
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - Wu-Chun Feng
- Department of Computer Science, Virginia Tech, Blacksburg, VA, USA
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Ramu Anandakrishnan
- Biomedical Sciences, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.
- Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA.
| |
Collapse
|
31
|
Alon M, Arafeh R, Lee JS, Madan S, Kalaora S, Nagler A, Abgarian T, Greenberg P, Ruppin E, Samuels Y. CAPN1 is a novel binding partner and regulator of the tumor suppressor NF1 in melanoma. Oncotarget 2018; 9:31264-31277. [PMID: 30131853 PMCID: PMC6101293 DOI: 10.18632/oncotarget.25805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/05/2018] [Indexed: 11/25/2022] Open
Abstract
Neurofibromin 1 (NF1), a tumor suppressor that negatively regulates RAS through its GTPase activity, is highly mutated in various types of sporadic human cancers, including melanoma. However, the binding partners of NF1 and the pathways in which it is involved in melanoma have not been characterized in an in depth manner. Utilizing a mass spectrometry analysis of NF1 binding partners, we revealed Calpain1 (CAPN1), a calcium-dependent neutral cysteine protease, as a novel NF1 binding partner that regulates NF1 degradation in melanoma cells. ShRNA-mediated knockdown of CAPN1 or treatment with a CAPN1 inhibitor stabilizes NF1 protein levels, downregulates AKT signaling and melanoma cell growth. Combination treatment of Calpain inhibitor I with MEKi Trametinib in different melanoma cells is more effective in reducing melanoma cell growth compared to treatment with Trametinib alone, suggesting that this combination may have a therapeutic potential in melanoma. This novel mechanism for regulating NF1 in melanoma provides a molecular basis for targeting CAPN1 in order to stabilize NF1 levels and, in doing so, suppressing Ras activation; this mechanism can be exploited therapeutically in melanoma and other cancers.
Collapse
Affiliation(s)
- Michal Alon
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Rand Arafeh
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Joo Sang Lee
- Center for Bioinformatics and Computational Biology, The University of Maryland, College Park, Maryland, USA
- Cancer Data Science Lab, National Cancer Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Sanna Madan
- Center for Bioinformatics and Computational Biology, The University of Maryland, College Park, Maryland, USA
- Cancer Data Science Lab, National Cancer Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Shelly Kalaora
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Nagler
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Tereza Abgarian
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Polina Greenberg
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eytan Ruppin
- Center for Bioinformatics and Computational Biology, The University of Maryland, College Park, Maryland, USA
- Cancer Data Science Lab, National Cancer Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Yardena Samuels
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
32
|
Abstract
Motivation Current technologies for single-cell DNA sequencing require whole-genome amplification (WGA), as a single cell contains too little DNA for direct sequencing. Unfortunately, WGA introduces biases in the resulting sequencing data, including non-uniformity in genome coverage and high rates of allele dropout. These biases complicate many downstream analyses, including the detection of genomic variants. Results We show that amplification biases have a potential upside: long-range correlations in rates of allele dropout provide a signal for phasing haplotypes at the lengths of amplicons from WGA, lengths which are generally longer than than individual sequence reads. We describe a statistical test to measure concurrent allele dropout between single-nucleotide polymorphisms (SNPs) across multiple sequenced single cells. We use results of this test to perform haplotype assembly across a collection of single cells. We demonstrate that the algorithm predicts phasing between pairs of SNPs with higher accuracy than phasing from reads alone. Using whole-genome sequencing data from only seven neural cells, we obtain haplotype blocks that are orders of magnitude longer than with sequence reads alone (median length 10.2 kb versus 312 bp), with error rates <2%. We demonstrate similar advantages on whole-exome data from 16 cells, where we obtain haplotype blocks with median length 9.2 kb-comparable to typical gene lengths-compared with median lengths of 41 bp with sequence reads alone, with error rates <4%. Our algorithm will be useful for haplotyping of rare alleles and studies of allele-specific somatic aberrations. Availability and implementation Source code is available at https://www.github.com/raphael-group. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gryte Satas
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Department of Computer Science, Brown University, Providence, RI, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| |
Collapse
|
33
|
Choi J, Lee K, Ingvarsdottir K, Bonasio R, Saraf A, Florens L, Washburn MP, Tadros S, Green MR, Busino L. Loss of KLHL6 promotes diffuse large B-cell lymphoma growth and survival by stabilizing the mRNA decay factor roquin2. Nat Cell Biol 2018; 20:586-596. [PMID: 29695787 PMCID: PMC5926793 DOI: 10.1038/s41556-018-0084-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/13/2018] [Indexed: 12/30/2022]
Abstract
Kelch-like protein 6 (KLHL6) is an uncharacterized gene mutated in diffuse large B-cell lymphoma (DLBCL). We report that KLHL6 assembles with CULLIN3 to form a functional CULLIN-Ring ubiquitin ligase. Mutations of KLHL6 inhibit its ligase activity by disrupting the interaction with CULLIN3. Loss of KLHL6 favors DLBCL growth and survival both in vitro and in xenograft models. We further established the mRNA decay factor Roquin2 as a substrate of KLHL6. Degradation of Roquin2 is dependent on B-cell receptor activation, and requires the integrity of the tyrosine 691 in Roquin2 that is essential for its interaction with KLHL6. A non-degradable Roquin2 (Y691F) mutant requires its RNA binding ability to phenocopy the effect of KLHL6 loss. Stabilization of Roquin2 promotes mRNA decay of the tumor suppressor and NF-κB pathway inhibitor, tumor necrosis factor-α-inducible gene 3 (TNFAIP3). Collectively, our findings uncover the tumor suppressing mechanism of KLHL6.
Collapse
Affiliation(s)
- Jaewoo Choi
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyutae Lee
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristin Ingvarsdottir
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Roberto Bonasio
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Anita Saraf
- The Stowers Institute of Medical Research, Kansas City, MO, USA
| | | | - Michael P Washburn
- The Stowers Institute of Medical Research, Kansas City, MO, USA.,Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Saber Tadros
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R Green
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luca Busino
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, USA. .,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Huang EW, Wang S, Lee DJL, Zhang R, Liu B, Zhou X, Zhai C. Framing Electronic Medical Records as Polylingual Documents in Query Expansion. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2018; 2017:940-949. [PMID: 29854161 PMCID: PMC5977599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We present a study of electronic medical record (EMR) retrieval that emulates situations in which a doctor treats a new patient. Given a query consisting of a new patient's symptoms, the retrieval system returns the set of most relevant records of previously treated patients. However, due to semantic, functional, and treatment synonyms in medical terminology, queries are often incomplete and thus require enhancement. In this paper, we present a topic model that frames symptoms and treatments as separate languages. Our experimental results show that this method improves retrieval performance over several baselines with statistical significance. These baselines include methods used in prior studies as well as state-of-the-art embedding techniques. Finally, we show that our proposed topic model discovers all three types of synonyms to improve medical record retrieval.
Collapse
Affiliation(s)
- Edward W Huang
- University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sheng Wang
- University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | | | - Baoyan Liu
- National Data Center of Traditional Chinese Medicine, Beijing, China
| | | | - ChengXiang Zhai
- University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
35
|
Ding L, Bailey MH, Porta-Pardo E, Thorsson V, Colaprico A, Bertrand D, Gibbs DL, Weerasinghe A, Huang KL, Tokheim C, Cortés-Ciriano I, Jayasinghe R, Chen F, Yu L, Sun S, Olsen C, Kim J, Taylor AM, Cherniack AD, Akbani R, Suphavilai C, Nagarajan N, Stuart JM, Mills GB, Wyczalkowski MA, Vincent BG, Hutter CM, Zenklusen JC, Hoadley KA, Wendl MC, Shmulevich L, Lazar AJ, Wheeler DA, Getz G. Perspective on Oncogenic Processes at the End of the Beginning of Cancer Genomics. Cell 2018; 173:305-320.e10. [PMID: 29625049 PMCID: PMC5916814 DOI: 10.1016/j.cell.2018.03.033] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/20/2018] [Accepted: 03/13/2018] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Matthew H Bailey
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Eduard Porta-Pardo
- Barcelona Supercomputing Centre, 08034 Barcelona, Spain; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Antonio Colaprico
- Machine Learning Group (MLG), Département d'Informatique, Université Libre de Bruxelles, 1050 Brussels, Belgium; Department of Human Genetics, University of Miami, Miami, FL 33136, USA
| | - Denis Bertrand
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 13862
| | - David L Gibbs
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Amila Weerasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Kuan-Lin Huang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Collin Tokheim
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Isidro Cortés-Ciriano
- Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA 02115, USA; Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Reyka Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Lihua Yu
- H3 Biomedicine Inc., Cambridge, MA 02139, USA
| | - Sam Sun
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Catharina Olsen
- Machine Learning Group (MLG), Département d'Informatique, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Jaegil Kim
- Broad Institute, Cambridge, MA 02142, USA
| | - Alison M Taylor
- Broad Institute, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Andrew D Cherniack
- Broad Institute, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77498, USA
| | - Chayaporn Suphavilai
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 13862
| | - Niranjan Nagarajan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 13862
| | - Joshua M Stuart
- Baskin School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77498, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Carolyn M Hutter
- National Human Genome Research Institute, Bethesda, MD 20892, USA
| | | | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael C Wendl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77498, USA
| | - David A Wheeler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Gad Getz
- Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02142, USA; Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
36
|
Gao B, Li G, Liu J, Li Y, Huang X. Identification of driver modules in pan-cancer via coordinating coverage and exclusivity. Oncotarget 2018; 8:36115-36126. [PMID: 28415609 PMCID: PMC5482642 DOI: 10.18632/oncotarget.16433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/13/2017] [Indexed: 12/30/2022] Open
Abstract
It is widely accepted that cancer is driven by accumulated somatic mutations during the lifetime of an individual. Cancer mutations may target relatively small number of cell functional modules. The heterogeneity in different cancer patients makes it difficult to identify driver mutations or functional modules related to cancer. It is biologically desired to be capable of identifying cancer pathway modules through coordination between coverage and exclusivity. There have been a few approaches developed for this purpose, but they all have limitations in practice due to their computational complexity and prediction accuracy. We present a network based approach, CovEx, to predict the specific patient oriented modules by 1) discovering candidate modules for each considered gene, 2) extracting significant candidates by harmonizing coverage and exclusivity and, 3) further selecting the patient oriented modules based on a set cover model. Applying CovEx to pan-cancer datasets spanning 12 cancer types collecting from public database TCGA, it demonstrates significant superiority over the current leading competitors in performance. It is published under GNU GENERAL PUBLIC LICENSE and the source code is available at:https://sourceforge.net/projects/cancer-pathway/files/
Collapse
Affiliation(s)
- Bo Gao
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China.,Department of Computer Science, Arkansas State University, Jonesboro, Arkansas, 72401, USA
| | - Guojun Li
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China.,Department of Computer Science, Arkansas State University, Jonesboro, Arkansas, 72401, USA
| | - Juntao Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Yang Li
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Xiuzhen Huang
- Department of Computer Science, Arkansas State University, Jonesboro, Arkansas, 72401, USA.,Molecular Biosciences Program, Arkansas State University, Jonesboro, Arkansas, 72401, USA
| |
Collapse
|
37
|
Reddy A, Zhang J, Davis NS, Moffitt AB, Love CL, Waldrop A, Leppa S, Pasanen A, Meriranta L, Karjalainen-Lindsberg ML, Nørgaard P, Pedersen M, Gang AO, Høgdall E, Heavican TB, Lone W, Iqbal J, Qin Q, Li G, Kim SY, Healy J, Richards KL, Fedoriw Y, Bernal-Mizrachi L, Koff JL, Staton AD, Flowers CR, Paltiel O, Goldschmidt N, Calaminici M, Clear A, Gribben J, Nguyen E, Czader MB, Ondrejka SL, Collie A, Hsi ED, Tse E, Au-Yeung RKH, Kwong YL, Srivastava G, Choi WWL, Evens AM, Pilichowska M, Sengar M, Reddy N, Li S, Chadburn A, Gordon LI, Jaffe ES, Levy S, Rempel R, Tzeng T, Happ LE, Dave T, Rajagopalan D, Datta J, Dunson DB, Dave SS. Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell 2017; 171:481-494.e15. [PMID: 28985567 DOI: 10.1016/j.cell.2017.09.027] [Citation(s) in RCA: 791] [Impact Index Per Article: 98.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common form of blood cancer and is characterized by a striking degree of genetic and clinical heterogeneity. This heterogeneity poses a major barrier to understanding the genetic basis of the disease and its response to therapy. Here, we performed an integrative analysis of whole-exome sequencing and transcriptome sequencing in a cohort of 1,001 DLBCL patients to comprehensively define the landscape of 150 genetic drivers of the disease. We characterized the functional impact of these genes using an unbiased CRISPR screen of DLBCL cell lines to define oncogenes that promote cell growth. A prognostic model comprising these genetic alterations outperformed current established methods: cell of origin, the International Prognostic Index comprising clinical variables, and dual MYC and BCL2 expression. These results comprehensively define the genetic drivers and their functional roles in DLBCL to identify new therapeutic opportunities in the disease.
Collapse
Affiliation(s)
- Anupama Reddy
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jenny Zhang
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Nicholas S Davis
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Andrea B Moffitt
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Cassandra L Love
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Alexander Waldrop
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Sirpa Leppa
- Helsinki University Hospital Cancer Center and University of Helsinki, Helsinki, Finland
| | - Annika Pasanen
- Helsinki University Hospital Cancer Center and University of Helsinki, Helsinki, Finland
| | - Leo Meriranta
- Helsinki University Hospital Cancer Center and University of Helsinki, Helsinki, Finland
| | | | - Peter Nørgaard
- Herlev and Gentofte Hospital, Copenhagen University, Herlev, Denmark
| | - Mette Pedersen
- Herlev and Gentofte Hospital, Copenhagen University, Herlev, Denmark
| | - Anne O Gang
- Herlev and Gentofte Hospital, Copenhagen University, Herlev, Denmark
| | - Estrid Høgdall
- Herlev and Gentofte Hospital, Copenhagen University, Herlev, Denmark
| | - Tayla B Heavican
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Waseem Lone
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Qiu Qin
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Guojie Li
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - So Young Kim
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Jane Healy
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Kristy L Richards
- Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yuri Fedoriw
- Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jean L Koff
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ashley D Staton
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Ora Paltiel
- Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Maria Calaminici
- Barts Cancer Institute of Queen Mary University of London, London, UK
| | - Andrew Clear
- Barts Cancer Institute of Queen Mary University of London, London, UK
| | - John Gribben
- Barts Cancer Institute of Queen Mary University of London, London, UK
| | - Evelyn Nguyen
- Pathology, Indiana University, Indianapolis, IN, USA
| | | | - Sarah L Ondrejka
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Angela Collie
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric D Hsi
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric Tse
- Queen Mary Hospital, University of Hong Kong, Hong Kong
| | | | - Yok-Lam Kwong
- Queen Mary Hospital, University of Hong Kong, Hong Kong
| | | | | | | | | | | | - Nishitha Reddy
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Amy Chadburn
- Columbia-Presbyterian Hospital, New York, NY, USA
| | - Leo I Gordon
- Northwestern University Medical School, Chicago, IL, USA
| | | | - Shawn Levy
- Hudson Alpha Institute for Biotechnology, Huntsville, AL, USA
| | - Rachel Rempel
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Tiffany Tzeng
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Lanie E Happ
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Tushar Dave
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Deepthi Rajagopalan
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Jyotishka Datta
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - David B Dunson
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - Sandeep S Dave
- Duke Cancer Institute and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
38
|
Dao P, Kim YA, Wojtowicz D, Madan S, Sharan R, Przytycka TM. BeWith: A Between-Within method to discover relationships between cancer modules via integrated analysis of mutual exclusivity, co-occurrence and functional interactions. PLoS Comput Biol 2017; 13:e1005695. [PMID: 29023534 PMCID: PMC5638227 DOI: 10.1371/journal.pcbi.1005695] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022] Open
Abstract
The analysis of the mutational landscape of cancer, including mutual exclusivity and co-occurrence of mutations, has been instrumental in studying the disease. We hypothesized that exploring the interplay between co-occurrence, mutual exclusivity, and functional interactions between genes will further improve our understanding of the disease and help to uncover new relations between cancer driving genes and pathways. To this end, we designed a general framework, BeWith, for identifying modules with different combinations of mutation and interaction patterns. We focused on three different settings of the BeWith schema: (i) BeME-WithFun, in which the relations between modules are enriched with mutual exclusivity, while genes within each module are functionally related; (ii) BeME-WithCo, which combines mutual exclusivity between modules with co-occurrence within modules; and (iii) BeCo-WithMEFun, which ensures co-occurrence between modules, while the within module relations combine mutual exclusivity and functional interactions. We formulated the BeWith framework using Integer Linear Programming (ILP), enabling us to find optimally scoring sets of modules. Our results demonstrate the utility of BeWith in providing novel information about mutational patterns, driver genes, and pathways. In particular, BeME-WithFun helped identify functionally coherent modules that might be relevant for cancer progression. In addition to finding previously well-known drivers, the identified modules pointed to other novel findings such as the interaction between NCOR2 and NCOA3 in breast cancer. Additionally, an application of the BeME-WithCo setting revealed that gene groups differ with respect to their vulnerability to different mutagenic processes, and helped us to uncover pairs of genes with potentially synergistic effects, including a potential synergy between mutations in TP53 and the metastasis related DCC gene. Overall, BeWith not only helped us uncover relations between potential driver genes and pathways, but also provided additional insights on patterns of the mutational landscape, going beyond cancer driving mutations. Implementation is available at https://www.ncbi.nlm.nih.gov/CBBresearch/Przytycka/software/bewith.html The genomic landscape of cancer obtained from large scale studies revealed mutational patterns such as mutual exclusivity and co-occurrences. Despite significant efforts, the understanding of the patterns harbored by specific genes and their interplay with functional interactions are still limited. Both mutual exclusivity and co-occurrence can arise due to several reasons. For example, two functionally interacting genes may dysregulate the same cancer related pathway when either of them is mutated, leading to mutually exclusive mutations. Alternatively, mutual exclusivity might reflect mutations specific to two different cancer types. Methods for joint analysis of co-occurrence, mutual exclusivity, and functional interaction relationships can lead to a better understanding of the causes and impacts of mutations in cancer. We report a new computational approach, BeWith, which identifies groups of genes (or gene modules) with coherent patterns within modules, but distinct properties among genes in different modules. The general formulation of our method allows us to investigate various aspects of the cancer mutational landscape, leading to uncovering relationships between mutated gene modules, cancer subtypes, and mutational signatures.
Collapse
Affiliation(s)
- Phuong Dao
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, United States of America
| | - Yoo-Ah Kim
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, United States of America
| | - Damian Wojtowicz
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, United States of America
| | - Sanna Madan
- Department of Computer Science, University of Maryland, College Park, MD, United States of America
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (RS); (TMP)
| | - Teresa M. Przytycka
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, United States of America
- * E-mail: (RS); (TMP)
| |
Collapse
|
39
|
Riaz N, Blecua P, Lim RS, Shen R, Higginson DS, Weinhold N, Norton L, Weigelt B, Powell SN, Reis-Filho JS. Pan-cancer analysis of bi-allelic alterations in homologous recombination DNA repair genes. Nat Commun 2017; 8:857. [PMID: 29021619 PMCID: PMC5636842 DOI: 10.1038/s41467-017-00921-w] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/04/2017] [Indexed: 12/22/2022] Open
Abstract
BRCA1 and BRCA2 are involved in homologous recombination (HR) DNA repair and are germ-line cancer pre-disposition genes that result in a syndrome of hereditary breast and ovarian cancer (HBOC). Whether germ-line or somatic alterations in these genes or other members of the HR pathway and if mono- or bi-allelic alterations of HR-related genes have a phenotypic impact on other cancers remains to be fully elucidated. Here, we perform a pan-cancer analysis of The Cancer Genome Atlas (TCGA) data set and observe that bi-allelic pathogenic alterations in homologous recombination (HR) DNA repair-related genes are prevalent across many malignancies. These bi-allelic alterations often associate with genomic features of HR deficiency. Further, in ovarian, breast and prostate cancers, bi-allelic alterations are mutually exclusive of each other. The combination of these two properties facilitates reclassification of variants of unknown significance affecting DNA repair genes, and may help personalize HR directed therapies in the clinic.Germline mutations in homologous recombination (HR) DNA repair genes are linked to breast and ovarian cancer. Here, the authors show that mutually exclusive bi-allelic inactivation of HR genes are present in other cancer types and associated with genomic features of HR deficiency, expanding the potential use of HR-directed therapies.
Collapse
Affiliation(s)
- Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Pedro Blecua
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Raymond S Lim
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Daniel S Higginson
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Nils Weinhold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
40
|
Senft D, Leiserson MDM, Ruppin E, Ronai ZA. Precision Oncology: The Road Ahead. Trends Mol Med 2017; 23:874-898. [PMID: 28887051 PMCID: PMC5718207 DOI: 10.1016/j.molmed.2017.08.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Current efforts in precision oncology largely focus on the benefit of genomics-guided therapy. Yet, advances in sequencing techniques provide an unprecedented view of the complex genetic and nongenetic heterogeneity within individual tumors. Herein, we outline the benefits of integrating genomic and transcriptomic analyses for advanced precision oncology. We summarize relevant computational approaches to detect novel drivers and genetic vulnerabilities, suitable for therapeutic exploration. Clinically relevant platforms to functionally test predicted drugs/drug combinations for individual patients are reviewed. Finally, we highlight the technological advances in single cell analysis of tumor specimens. These may ultimately lead to the development of next-generation cancer drugs, capable of tackling the hurdles imposed by genetic and phenotypic heterogeneity on current anticancer therapies.
Collapse
Affiliation(s)
- Daniela Senft
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mark D M Leiserson
- Microsoft Research New England, Cambridge, MA 02142, USA; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Eytan Ruppin
- School of Computer Sciences and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Technion Integrated Cancer Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
41
|
Moffitt AB, Ondrejka SL, McKinney M, Rempel RE, Goodlad JR, Teh CH, Leppa S, Mannisto S, Kovanen PE, Tse E, Au-Yeung RKH, Kwong YL, Srivastava G, Iqbal J, Yu J, Naresh K, Villa D, Gascoyne RD, Said J, Czader MB, Chadburn A, Richards KL, Rajagopalan D, Davis NS, Smith EC, Palus BC, Tzeng TJ, Healy JA, Lugar PL, Datta J, Love C, Levy S, Dunson DB, Zhuang Y, Hsi ED, Dave SS. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. J Exp Med 2017; 214:1371-1386. [PMID: 28424246 PMCID: PMC5413324 DOI: 10.1084/jem.20160894] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
Enteropathy-associated T cell lymphoma (EATL) is the most common oncologic complication of celiac disease. Moffitt and colleagues identify novel EATL-defining mutations in SETD2, as well as clinically relevant mutations in the JAK-STAT pathway. Enteropathy-associated T cell lymphoma (EATL) is a lethal, and the most common, neoplastic complication of celiac disease. Here, we defined the genetic landscape of EATL through whole-exome sequencing of 69 EATL tumors. SETD2 was the most frequently silenced gene in EATL (32% of cases). The JAK-STAT pathway was the most frequently mutated pathway, with frequent mutations in STAT5B as well as JAK1, JAK3, STAT3, and SOCS1. We also identified mutations in KRAS, TP53, and TERT. Type I EATL and type II EATL (monomorphic epitheliotropic intestinal T cell lymphoma) had highly overlapping genetic alterations indicating shared mechanisms underlying their pathogenesis. We modeled the effects of SETD2 loss in vivo by developing a T cell–specific knockout mouse. These mice manifested an expansion of γδ T cells, indicating novel roles for SETD2 in T cell development and lymphomagenesis. Our data render the most comprehensive genetic portrait yet of this uncommon but lethal disease and may inform future classification schemes.
Collapse
Affiliation(s)
- Andrea B Moffitt
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Sarah L Ondrejka
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Matthew McKinney
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Rachel E Rempel
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - John R Goodlad
- Haematological Malignancy Diagnostic Service, St. James's University Hospital, Leeds LS9 7TF, England, UK
| | - Chun Huat Teh
- Haematology Department, Western General Hospital, Edinburgh EH14 1TY, Scotland, UK
| | - Sirpa Leppa
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Susanna Mannisto
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Panu E Kovanen
- HUSLAB and Medicum, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Eric Tse
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Yok-Lam Kwong
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Javeed Iqbal
- University of Nebraska Medical Center, Omaha, NE 68198
| | - Jiayu Yu
- University of Nebraska Medical Center, Omaha, NE 68198
| | | | - Diego Villa
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Randy D Gascoyne
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Jonathan Said
- University of California, Los Angeles, Los Angeles, CA 90095
| | | | - Amy Chadburn
- Presbyterian Hospital, Pathology and Cell Biology, Cornell University, New York, NY 10065
| | | | | | - Nicholas S Davis
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Eileen C Smith
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Brooke C Palus
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Tiffany J Tzeng
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Jane A Healy
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Patricia L Lugar
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - Jyotishka Datta
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Cassandra Love
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Shawn Levy
- Hudson Alpha Institute for Biotechnology, Huntsville, AL 35806
| | - David B Dunson
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Yuan Zhuang
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710
| | - Eric D Hsi
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sandeep S Dave
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| |
Collapse
|
42
|
Graim K, Liu TT, Achrol AS, Paull EO, Newton Y, Chang SD, Harsh GR, Cordero SP, Rubin DL, Stuart JM. Revealing cancer subtypes with higher-order correlations applied to imaging and omics data. BMC Med Genomics 2017; 10:20. [PMID: 28359308 PMCID: PMC5374737 DOI: 10.1186/s12920-017-0256-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/15/2017] [Indexed: 12/14/2022] Open
Abstract
Background Patient stratification to identify subtypes with different disease manifestations, severity, and expected survival time is a critical task in cancer diagnosis and treatment. While stratification approaches using various biomarkers (including high-throughput gene expression measurements) for patient-to-patient comparisons have been successful in elucidating previously unseen subtypes, there remains an untapped potential of incorporating various genotypic and phenotypic data to discover novel or improved groupings. Methods Here, we present HOCUS, a unified analytical framework for patient stratification that uses a community detection technique to extract subtypes out of sparse patient measurements. HOCUS constructs a patient-to-patient network from similarities in the data and iteratively groups and reconstructs the network into higher order clusters. We investigate the merits of using higher-order correlations to cluster samples of cancer patients in terms of their associations with survival outcomes. Results In an initial test of the method, the approach identifies cancer subtypes in mutation data of glioblastoma, ovarian, breast, prostate, and bladder cancers. In several cases, HOCUS provides an improvement over using the molecular features directly to compare samples. Application of HOCUS to glioblastoma images reveals a size and location classification of tumors that improves over human expert-based stratification. Conclusions Subtypes based on higher order features can reveal comparable or distinct groupings. The distinct solutions can provide biologically- and treatment-relevant solutions that are just as significant as solutions based on the original data. Electronic supplementary material The online version of this article (doi:10.1186/s12920-017-0256-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kiley Graim
- Biomedical Engineering, University of California, Santa Cruz, USA.,UC Santa Cruz Genomics Institute, University of California, Santa Cruz, USA
| | - Tiffany Ting Liu
- Stanford Center for Biomedical Informatics Research and Biomedical Informatics Training Program, Stanford University School of Medicine, Stanford, USA.,Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, Stanford, USA
| | - Achal S Achrol
- Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, Stanford, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA.,Departments of Neurosurgery, Stanford University School of Medicine, Stanford, USA
| | - Evan O Paull
- Biomedical Engineering, University of California, Santa Cruz, USA.,UC Santa Cruz Genomics Institute, University of California, Santa Cruz, USA
| | - Yulia Newton
- Biomedical Engineering, University of California, Santa Cruz, USA.,UC Santa Cruz Genomics Institute, University of California, Santa Cruz, USA
| | - Steven D Chang
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, USA
| | - Griffith R Harsh
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, USA
| | - Sergio P Cordero
- Biomedical Engineering, University of California, Santa Cruz, USA.,UC Santa Cruz Genomics Institute, University of California, Santa Cruz, USA
| | - Daniel L Rubin
- Stanford Center for Biomedical Informatics Research and Biomedical Informatics Training Program, Stanford University School of Medicine, Stanford, USA.,Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, Stanford, USA
| | - Joshua M Stuart
- Biomedical Engineering, University of California, Santa Cruz, USA. .,UC Santa Cruz Genomics Institute, University of California, Santa Cruz, USA.
| |
Collapse
|