51
|
Strelka2: fast and accurate calling of germline and somatic variants. Nat Methods 2018; 15:591-594. [PMID: 30013048 DOI: 10.1038/s41592-018-0051-x] [Citation(s) in RCA: 936] [Impact Index Per Article: 133.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
We describe Strelka2 ( https://github.com/Illumina/strelka ), an open-source small-variant-calling method for research and clinical germline and somatic sequencing applications. Strelka2 introduces a novel mixture-model-based estimation of insertion/deletion error parameters from each sample, an efficient tiered haplotype-modeling strategy, and a normal sample contamination model to improve liquid tumor analysis. For both germline and somatic calling, Strelka2 substantially outperformed the current leading tools in terms of both variant-calling accuracy and computing cost.
Collapse
|
52
|
Zhang AW, McPherson A, Milne K, Kroeger DR, Hamilton PT, Miranda A, Funnell T, Little N, de Souza CP, Laan S, LeDoux S, Cochrane DR, Lim JL, Yang W, Roth A, Smith MA, Ho J, Tse K, Zeng T, Shlafman I, Mayo MR, Moore R, Failmezger H, Heindl A, Wang YK, Bashashati A, Grewal DS, Brown SD, Lai D, Wan AN, Nielsen CB, Huebner C, Tessier-Cloutier B, Anglesio MS, Bouchard-Côté A, Yuan Y, Wasserman WW, Gilks CB, Karnezis AN, Aparicio S, McAlpine JN, Huntsman DG, Holt RA, Nelson BH, Shah SP. Interfaces of Malignant and Immunologic Clonal Dynamics in Ovarian Cancer. Cell 2018; 173:1755-1769.e22. [DOI: 10.1016/j.cell.2018.03.073] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/22/2018] [Accepted: 03/27/2018] [Indexed: 02/07/2023]
|
53
|
Espiritu SMG, Liu LY, Rubanova Y, Bhandari V, Holgersen EM, Szyca LM, Fox NS, Chua ML, Yamaguchi TN, Heisler LE, Livingstone J, Wintersinger J, Yousif F, Lalonde E, Rouette A, Salcedo A, Houlahan KE, Li CH, Huang V, Fraser M, van der Kwast T, Morris QD, Bristow RG, Boutros PC. The Evolutionary Landscape of Localized Prostate Cancers Drives Clinical Aggression. Cell 2018; 173:1003-1013.e15. [DOI: 10.1016/j.cell.2018.03.029] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/01/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
|
54
|
Formalin fixation increases deamination mutation signature but should not lead to false positive mutations in clinical practice. PLoS One 2018; 13:e0196434. [PMID: 29698444 PMCID: PMC5919577 DOI: 10.1371/journal.pone.0196434] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/12/2018] [Indexed: 12/28/2022] Open
Abstract
Genomic analysis of cancer tissues is an essential aspect of personalized oncology treatment. Though it has been suggested that formalin fixation of patient tissues may be suboptimal for molecular studies, this tissue processing approach remains the industry standard. Therefore clinical molecular laboratories must be able to work with formalin fixed, paraffin embedded (FFPE) material. This study examines the effects of pre-analytic variables introduced by routine pathology processing on specimens used for clinical reports produced by next-generation sequencing technology. Tissue resected from three colorectal cancer patients was subjected to 2, 15, 24, and 48 hour fixation times in neutral buffered formalin. DNA was extracted from all tissues twice, once with uracil-N-glycosylase (UNG) treatment to counter deamination effects, and once without. Of note, deamination events at methylated cytosine, as found at CpG sites, remains unaffected by UNG. After extraction a two-step PCR targeted sequencing method was performed using the Illumina MiSeq and the data was analyzed via a custom-built bioinformatics pipeline, including filtration of reads with mapping quality <30. A larger baseline group of samples (n = 20) was examined to establish if there was a sample performance difference between the two DNA extraction methods, with/without UNG treatment. There was no statistical difference between sequencing performance of the two extraction methods when comparing read counts (raw, mapped, and filtered) and read quality (% mapped, % filtered). Analyzing mutation type, there was no significant difference between mutation calls until the 48 hour fixation treatment. At 48 hours there is a significant increase in C/G->T/A mutations that is not represented in DNA treated with UNG. This suggests these errors may be due to deamination events triggered by a longer fixation time. However the allelic frequency of these events remained below the limit of detection for reportable mutations in this assay (<2%). We do however recommend that suspected intratumoral heterogeneity events be verified by re-sequencing the same FFPE block.
Collapse
|
55
|
Thibodeau ML, Bonakdar M, Zhao E, Mungall KL, Reisle C, Zhang W, Bye MH, Thiessen N, Bleile D, Mungall AJ, Ma YP, Jones MR, Renouf DJ, Lim HJ, Yip S, Ng T, Ho C, Laskin J, Marra MA, Schrader KA, Jones SJM. Whole genome and whole transcriptome genomic profiling of a metastatic eccrine porocarcinoma. NPJ Precis Oncol 2018; 2:8. [PMID: 29872726 PMCID: PMC5871832 DOI: 10.1038/s41698-018-0050-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/26/2018] [Accepted: 02/01/2018] [Indexed: 12/31/2022] Open
Abstract
Eccrine porocarcinomas (EPs) are rare malignant tumours of the intraepidermic sweat gland duct and most often arise from benign eccrine poromas. Some recurrent somatic genomic events have been identified in these malignancies, but very little is known about the complexity of their molecular pathophysiology. We describe the whole genome and whole transcriptome genomic profiling of a metastatic EP in a 66-year-old male patient with a previous history of localized porocarcinoma of the scalp. Whole genome and whole transcriptome genomic profiling was performed on the metastatic EP. Whole genome sequencing was performed on blood-derived DNA in order to allow a comparison between germline and somatic events. We found somatic copy losses of several tumour suppressor genes including APC, PTEN and CDKN2A, CDKN2B and CDKN1A. We identified a somatic hemizygous CDKN2A pathogenic splice site variant. De novo transcriptome assembly revealed abnormal splicing of CDKN2A p14ARF and p16INK4a. Elevated expression of oncogenes EGFR and NOTCH1 was noted and no somatic mutations were found in these genes. Wnt pathway somatic alterations were also observed. In conclusion, our results suggest that the molecular pathophysiology of malignant EP features high complexity and subtle interactions of multiple key genes. Cell cycle dysregulation and CDKN2A loss of function was found to be a new potential driver in EP tumourigenesis. Moreover, the combination of somatic copy number variants and abnormal gene expression perhaps partly related to epigenetic mechanisms, all likely contribute to the development of this rare malignancy in our patient.
Collapse
Affiliation(s)
- My Linh Thibodeau
- Department of Medical Genetics, University of British Columbia, C201–4500 Oak Street, Vancouver, BC V6H 3N1 Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Melika Bonakdar
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Eric Zhao
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Karen L. Mungall
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Caralyn Reisle
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Wei Zhang
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Morgan H. Bye
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Nina Thiessen
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Dustin Bleile
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Andrew J. Mungall
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Yussanne P. Ma
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Martin R. Jones
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Daniel J. Renouf
- Department of Medical Oncology, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6 Canada
| | - Howard J. Lim
- Department of Medical Oncology, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6 Canada
| | - Stephen Yip
- Department of Pathology & Laboratory Medicine, Vancouver General Hospital, 910 West 10th Avenue, Vancouver, BC V5Z 1M9 Canada
| | - Tony Ng
- Department of Pathology & Laboratory Medicine, Vancouver General Hospital, 910 West 10th Avenue, Vancouver, BC V5Z 1M9 Canada
| | - Cheryl Ho
- Department of Medical Oncology, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6 Canada
| | - Janessa Laskin
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
- Department of Medical Oncology, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6 Canada
| | - Marco A. Marra
- Department of Medical Genetics, University of British Columbia, C201–4500 Oak Street, Vancouver, BC V6H 3N1 Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| | - Kasmintan A. Schrader
- Hereditary Cancer Program, Department of Medical Genetics, British Columbia Cancer Agency, 614–750 West Broadway, Vancouver, BC V5Z 1H5 Canada
| | - Steven J. M. Jones
- Department of Medical Genetics, University of British Columbia, C201–4500 Oak Street, Vancouver, BC V6H 3N1 Canada
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, 100–570 West 7th Avenue, Vancouver, BC V5Z 4S6 Canada
| |
Collapse
|
56
|
Albuquerque MA, Grande BM, Ritch EJ, Pararajalingam P, Jessa S, Krzywinski M, Grewal JK, Shah SP, Boutros PC, Morin RD. Enhancing knowledge discovery from cancer genomics data with Galaxy. Gigascience 2018; 6:1-13. [PMID: 28327945 PMCID: PMC5437943 DOI: 10.1093/gigascience/gix015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/06/2017] [Indexed: 01/15/2023] Open
Abstract
The field of cancer genomics has demonstrated the power of massively parallel sequencing techniques to inform on the genes and specific alterations that drive tumor onset and progression. Although large comprehensive sequence data sets continue to be made increasingly available, data analysis remains an ongoing challenge, particularly for laboratories lacking dedicated resources and bioinformatics expertise. To address this, we have produced a collection of Galaxy tools that represent many popular algorithms for detecting somatic genetic alterations from cancer genome and exome data. We developed new methods for parallelization of these tools within Galaxy to accelerate runtime and have demonstrated their usability and summarized their runtimes on multiple cloud service providers. Some tools represent extensions or refinement of existing toolkits to yield visualizations suited to cohort-wide cancer genomic analysis. For example, we present Oncocircos and Oncoprintplus, which generate data-rich summaries of exome-derived somatic mutation. Workflows that integrate these to achieve data integration and visualizations are demonstrated on a cohort of 96 diffuse large B-cell lymphomas and enabled the discovery of multiple candidate lymphoma-related genes. Our toolkit is available from our GitHub repository as Galaxy tool and dependency definitions and has been deployed using virtualization on multiple platforms including Docker.
Collapse
Affiliation(s)
- Marco A Albuquerque
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Bruno M Grande
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Elie J Ritch
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Prasath Pararajalingam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Selin Jessa
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Martin Krzywinski
- Canada's Michael Smith Genome Sciences Center, BC Cancer Agency, Vancouver, BC, Canada
| | - Jasleen K Grewal
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Sohrab P Shah
- Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Paul C Boutros
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Ryan D Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.,Canada's Michael Smith Genome Sciences Center, BC Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
57
|
Hung SS, Meissner B, Chavez EA, Ben-Neriah S, Ennishi D, Jones MR, Shulha HP, Chan FC, Boyle M, Kridel R, Gascoyne RD, Mungall AJ, Marra MA, Scott DW, Connors JM, Steidl C. Assessment of Capture and Amplicon-Based Approaches for the Development of a Targeted Next-Generation Sequencing Pipeline to Personalize Lymphoma Management. J Mol Diagn 2018; 20:203-214. [PMID: 29429887 DOI: 10.1016/j.jmoldx.2017.11.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/24/2017] [Accepted: 11/03/2017] [Indexed: 01/30/2023] Open
Abstract
Targeted next-generation sequencing panels are increasingly used to assess the value of gene mutations for clinical diagnostic purposes. For assay development, amplicon-based methods have been preferentially used on the basis of short preparation time and small DNA input amounts. However, capture sequencing has emerged as an alternative approach because of high testing accuracy. We compared capture hybridization and amplicon sequencing approaches using fresh-frozen and formalin-fixed, paraffin-embedded tumor samples from eight lymphoma patients. Next, we developed a targeted sequencing pipeline using a 32-gene panel for accurate detection of actionable mutations in formalin-fixed, paraffin-embedded tumor samples of the most common lymphocytic malignancies: chronic lymphocytic leukemia, diffuse large B-cell lymphoma, and follicular lymphoma. We show that hybrid capture is superior to amplicon sequencing by providing deep more uniform coverage and yielding higher sensitivity for variant calling. Sanger sequencing of 588 variants identified specificity limits of thresholds for mutation calling, and orthogonal validation on 66 cases indicated 93% concordance with whole-genome sequencing. The developed pipeline and assay identified at least one actionable mutation in 91% of tumors from 219 lymphoma patients and revealed subtype-specific mutation patterns and frequencies consistent with the literature. This pipeline is an accurate and sensitive method for identifying actionable gene mutations in routinely acquired biopsy materials, suggesting further assessment of capture-based assays in the context of personalized lymphoma management.
Collapse
Affiliation(s)
- Stacy S Hung
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Barbara Meissner
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Elizabeth A Chavez
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Susana Ben-Neriah
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Daisuke Ennishi
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Martin R Jones
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Hennady P Shulha
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Fong Chun Chan
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Merrill Boyle
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Robert Kridel
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Randy D Gascoyne
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - David W Scott
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Joseph M Connors
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer Agency, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
58
|
Xu C. A review of somatic single nucleotide variant calling algorithms for next-generation sequencing data. Comput Struct Biotechnol J 2018; 16:15-24. [PMID: 29552334 PMCID: PMC5852328 DOI: 10.1016/j.csbj.2018.01.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/20/2018] [Accepted: 01/28/2018] [Indexed: 02/06/2023] Open
Abstract
Detection of somatic mutations holds great potential in cancer treatment and has been a very active research field in the past few years, especially since the breakthrough of the next-generation sequencing technology. A collection of variant calling pipelines have been developed with different underlying models, filters, input data requirements, and targeted applications. This review aims to enumerate these unique features of the state-of-the-art variant callers, in the hope to provide a practical guide for selecting the appropriate pipeline for specific applications. We will focus on the detection of somatic single nucleotide variants, ranging from traditional variant callers based on whole genome or exome sequencing of paired tumor-normal samples to recent low-frequency variant callers designed for targeted sequencing protocols with unique molecular identifiers. The variant callers have been extensively benchmarked with inconsistent performances across these studies. We will review the reference materials, datasets, and performance metrics that have been used in the benchmarking studies. In the end, we will discuss emerging trends and future directions of the variant calling algorithms.
Collapse
Affiliation(s)
- Chang Xu
- Life Science Research and Foundation, Qiagen Sciences, Inc., 6951 Executive Way, Frederick, Maryland 21703, USA
| |
Collapse
|
59
|
Detection and genomic characterization of a mammary-like adenocarcinoma. Cold Spring Harb Mol Case Stud 2017; 3:mcs.a002170. [PMID: 28877932 PMCID: PMC5701302 DOI: 10.1101/mcs.a002170] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/02/2017] [Indexed: 12/31/2022] Open
Abstract
Whole-genome and transcriptome sequencing were performed to identify potential therapeutic strategies in the absence of viable treatment options for a patient initially diagnosed with vulvar adenocarcinoma. Genomic events were prioritized by comparison against variant distributions in the TCGA pan-cancer data set and complemented with detailed transcriptome sequencing and copy-number analysis. These findings were considered against published scientific literature in order to evaluate the functional effects of potentially relevant genomic events. Analysis of the transcriptome against a background of 27 TCGA cancer types led to reclassification of the tumor as a primary HER2+ mammary-like adenocarcinoma of the vulva. This revised diagnosis was subsequently confirmed by follow-up immunohistochemistry for a mammary-like adenocarcinoma. The patient was treated with chemotherapy and targeted therapies for HER2+ breast cancer. The detailed pathology and genomic findings of this case are presented herein.
Collapse
|
60
|
Farahani H, de Souza CPE, Billings R, Yap D, Shumansky K, Wan A, Lai D, Mes-Masson AM, Aparicio S, P Shah S. Engineered in-vitro cell line mixtures and robust evaluation of computational methods for clonal decomposition and longitudinal dynamics in cancer. Sci Rep 2017; 7:13467. [PMID: 29044127 PMCID: PMC5647443 DOI: 10.1038/s41598-017-13338-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/19/2017] [Indexed: 11/30/2022] Open
Abstract
Characterization and quantification of tumour clonal populations over time via longitudinal sampling are essential components in understanding and predicting the response to therapeutic interventions. Computational methods for inferring tumour clonal composition from deep-targeted sequencing data are ubiquitous, however due to the lack of a ground truth biological data, evaluating their performance is difficult. In this work, we generate a benchmark data set that simulates tumour longitudinal growth and heterogeneity by in vitro mixing of cancer cell lines with known proportions. We apply four different algorithms to our ground truth data set and assess their performance in inferring clonal composition using different metrics. We also analyse the performance of these algorithms on breast tumour xenograft samples. We conclude that methods that can simultaneously analyse multiple samples while accounting for copy number alterations as a factor in allelic measurements exhibit the most accurate predictions. These results will inform future functional genomics oriented studies of model systems where time series measurements in the context of therapeutic interventions are becoming increasingly common. These studies will need computational models which accurately reflect the multi-factorial nature of allele measurement in cancer including, as we show here, segmental aneuploidies.
Collapse
Affiliation(s)
- Hossein Farahani
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada.,University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, V6T 2B5, Canada
| | - Camila P E de Souza
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada.,University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, V6T 2B5, Canada
| | - Raewyn Billings
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada
| | - Damian Yap
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada
| | - Karey Shumansky
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada
| | - Adrian Wan
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada
| | - Daniel Lai
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l' Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| | - Samuel Aparicio
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada.,University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, V6T 2B5, Canada
| | - Sohrab P Shah
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, V5Z 1L3, Canada. .,University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, V6T 2B5, Canada. .,BC Cancer Agency, Michael Smith Genome Sciences Centre, Vancouver, V5Z 1L3, Canada.
| |
Collapse
|
61
|
Thibodeau ML, Reisle C, Zhao E, Martin LA, Alwelaie Y, Mungall KL, Ch'ng C, Thomas R, Ng T, Yip S, J Lim H, Sun S, Young SS, Karsan A, Zhao Y, Mungall AJ, Moore RA, J Renouf D, Gelmon K, Ma YP, Hayes M, Laskin J, Marra MA, Schrader KA, Jones SJM. Genomic profiling of pelvic genital type leiomyosarcoma in a woman with a germline CHEK2:c.1100delC mutation and a concomitant diagnosis of metastatic invasive ductal breast carcinoma. Cold Spring Harb Mol Case Stud 2017; 3:mcs.a001628. [PMID: 28514723 PMCID: PMC5593158 DOI: 10.1101/mcs.a001628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/14/2017] [Indexed: 12/19/2022] Open
Abstract
We describe a woman with the known pathogenic germline variant CHEK2:c.1100delC and synchronous diagnoses of both pelvic genital type leiomyosarcoma (LMS) and metastatic invasive ductal breast carcinoma. CHEK2 (checkpoint kinase 2) is a tumor-suppressor gene encoding a serine/threonine-protein kinase (CHEK2) involved in double-strand DNA break repair and cell cycle arrest. The CHEK2:c.1100delC variant is a moderate penetrance allele resulting in an approximately twofold increase in breast cancer risk. Whole-genome and whole-transcriptome sequencing were performed on the leiomyosarcoma and matched blood-derived DNA. Despite the presence of several genomic hits within the double-strand DNA damage pathway (CHEK2 germline variant and multiple RAD51B somatic structural variants), tumor profiling did not show an obvious DNA repair deficiency signature. However, even though the LMS displayed clear malignant features, its genomic profiling revealed several characteristics classically associated with leiomyomas including a translocation, t(12;14), with one breakpoint disrupting RAD51B and the other breakpoint upstream of HMGA2 with very high expression of HMGA2 and PLAG1. This is the first report of LMS genomic profiling in a patient with the germline CHEK2:c.1100delC variant and an additional diagnosis of metastatic invasive ductal breast carcinoma. We also describe a possible mechanistic relationship between leiomyoma and LMS based on genomic and transcriptome data. Our findings suggest that RAD51B translocation and HMGA2 overexpression may play an important role in LMS oncogenesis.
Collapse
Affiliation(s)
- My Linh Thibodeau
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - Caralyn Reisle
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Eric Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Lee Ann Martin
- Fraser Valley Cancer Centre, British Columbia Cancer Agency, Surrey, British Columbia V3V 1Z2, Canada
| | - Yazeed Alwelaie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Carolyn Ch'ng
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Ruth Thomas
- Hereditary Cancer Program, British Columbia Cancer Agency-Abbotsford, Abbotsford, British Columbia V2S 0C2, Canada
| | - Tony Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Howard J Lim
- British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Sophie Sun
- British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Sean S Young
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada.,Cancer Genetics Laboratory, Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada.,Cancer Genetics Laboratory, Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Daniel J Renouf
- British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Karen Gelmon
- British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Yussanne P Ma
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Malcolm Hayes
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada.,Cancer Genetics Laboratory, Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Janessa Laskin
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Marco A Marra
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| | - Kasmintan A Schrader
- Hereditary Cancer Program, Department of Medical Genetics, British Columbia Cancer Agency, 614-750 West Broadway, Vancouver British Columbia V5Z 1H5, Canada
| | - Steven J M Jones
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada.,Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, British Columbia V5Z 4S6, Canada
| |
Collapse
|
62
|
Malhotra R, Jha M, Poss M, Acharya R. A random forest classifier for detecting rare variants in NGS data from viral populations. Comput Struct Biotechnol J 2017; 15:388-395. [PMID: 28819548 PMCID: PMC5548337 DOI: 10.1016/j.csbj.2017.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/01/2017] [Accepted: 07/03/2017] [Indexed: 11/28/2022] Open
Abstract
We propose a random forest classifier for detecting rare variants from sequencing errors in Next Generation Sequencing (NGS) data from viral populations. The method utilizes counts of varying length of k-mers from the reads of a viral population to train a Random forest classifier, called MultiRes, that classifies k-mers as erroneous or rare variants. Our algorithm is rooted in concepts from signal processing and uses a frame-based representation of k-mers. Frames are sets of non-orthogonal basis functions that were traditionally used in signal processing for noise removal. We define discrete spatial signals for genomes and sequenced reads, and show that k-mers of a given size constitute a frame. We evaluate MultiRes on simulated and real viral population datasets, which consist of many low frequency variants, and compare it to the error detection methods used in correction tools known in the literature. MultiRes has 4 to 500 times less false positives k-mer predictions compared to other methods, essential for accurate estimation of viral population diversity and their de-novo assembly. It has high recall of the true k-mers, comparable to other error correction methods. MultiRes also has greater than 95% recall for detecting single nucleotide polymorphisms (SNPs) and fewer false positive SNPs, while detecting higher number of rare variants compared to other variant calling methods for viral populations. The software is available freely from the GitHub link https://github.com/raunaq-m/MultiRes.
Collapse
Affiliation(s)
- Raunaq Malhotra
- The School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Manjari Jha
- The School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Mary Poss
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Raj Acharya
- School of Informatics and Computing, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
63
|
Peterson TA, Gauran IIM, Park J, Park D, Kann MG. Oncodomains: A protein domain-centric framework for analyzing rare variants in tumor samples. PLoS Comput Biol 2017; 13:e1005428. [PMID: 28426665 PMCID: PMC5398485 DOI: 10.1371/journal.pcbi.1005428] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/28/2017] [Indexed: 12/28/2022] Open
Abstract
The fight against cancer is hindered by its highly heterogeneous nature. Genome-wide sequencing studies have shown that individual malignancies contain many mutations that range from those commonly found in tumor genomes to rare somatic variants present only in a small fraction of lesions. Such rare somatic variants dominate the landscape of genomic mutations in cancer, yet efforts to correlate somatic mutations found in one or few individuals with functional roles have been largely unsuccessful. Traditional methods for identifying somatic variants that drive cancer are 'gene-centric' in that they consider only somatic variants within a particular gene and make no comparison to other similar genes in the same family that may play a similar role in cancer. In this work, we present oncodomain hotspots, a new 'domain-centric' method for identifying clusters of somatic mutations across entire gene families using protein domain models. Our analysis confirms that our approach creates a framework for leveraging structural and functional information encapsulated by protein domains into the analysis of somatic variants in cancer, enabling the assessment of even rare somatic variants by comparison to similar genes. Our results reveal a vast landscape of somatic variants that act at the level of domain families altering pathways known to be involved with cancer such as protein phosphorylation, signaling, gene regulation, and cell metabolism. Due to oncodomain hotspots' unique ability to assess rare variants, we expect our method to become an important tool for the analysis of sequenced tumor genomes, complementing existing methods.
Collapse
Affiliation(s)
- Thomas A. Peterson
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
- University of California, San Francisco, Institute for Computational Health Science, San Francisco, California, United States of America
| | - Iris Ivy M. Gauran
- Department of Mathematics and Statistics, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Junyong Park
- Department of Mathematics and Statistics, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - DoHwan Park
- Department of Mathematics and Statistics, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Maricel G. Kann
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| |
Collapse
|
64
|
Salehi S, Steif A, Roth A, Aparicio S, Bouchard-Côté A, Shah SP. ddClone: joint statistical inference of clonal populations from single cell and bulk tumour sequencing data. Genome Biol 2017; 18:44. [PMID: 28249593 PMCID: PMC5333399 DOI: 10.1186/s13059-017-1169-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/10/2017] [Indexed: 12/16/2022] Open
Abstract
Next-generation sequencing (NGS) of bulk tumour tissue can identify constituent cell populations in cancers and measure their abundance. This requires computational deconvolution of allelic counts from somatic mutations, which may be incapable of fully resolving the underlying population structure. Single cell sequencing (SCS) is a more direct method, although its replacement of NGS is impeded by technical noise and sampling limitations. We propose ddClone, which analytically integrates NGS and SCS data, leveraging their complementary attributes through joint statistical inference. We show on real and simulated datasets that ddClone produces more accurate results than can be achieved by either method alone.
Collapse
Affiliation(s)
- Sohrab Salehi
- Bioinformatics Graduate Program, University of British Columbia, 570 West 7th Avenue, Vancouver, V5Z 4S6, BC, Canada
| | - Adi Steif
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - Andrew Roth
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z 1L3, BC, Canada
| | - Samuel Aparicio
- Bioinformatics Graduate Program, University of British Columbia, 570 West 7th Avenue, Vancouver, V5Z 4S6, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, BC, Canada
| | - Alexandre Bouchard-Côté
- Department of Statistics, University of British Columbia, 2207 Main Mall, Vancouver, V6T 1Z4, BC, Canada
| | - Sohrab P Shah
- Bioinformatics Graduate Program, University of British Columbia, 570 West 7th Avenue, Vancouver, V5Z 4S6, BC, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 2B5, BC, Canada.
| |
Collapse
|
65
|
Scalable whole-genome single-cell library preparation without preamplification. Nat Methods 2017; 14:167-173. [PMID: 28068316 DOI: 10.1038/nmeth.4140] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 12/02/2016] [Indexed: 12/17/2022]
Abstract
Single-cell genomics is critical for understanding cellular heterogeneity in cancer, but existing library preparation methods are expensive, require sample preamplification and introduce coverage bias. Here we describe direct library preparation (DLP), a robust, scalable, and high-fidelity method that uses nanoliter-volume transposition reactions for single-cell whole-genome library preparation without preamplification. We examined 782 cells from cell lines and triple-negative breast xenograft tumors. Low-depth sequencing, compared with existing methods, revealed greater coverage uniformity and more reliable detection of copy-number alterations. Using phylogenetic analysis, we found minor xenograft subpopulations that were undetectable by bulk sequencing, as well as dynamic clonal expansion and diversification between passages. Merging single-cell genomes in silico, we generated 'bulk-equivalent' genomes with high depth and uniform coverage. Thus, low-depth sequencing of DLP libraries may provide an attractive replacement for conventional bulk sequencing methods, permitting analysis of copy number at the cell level and of other genomic variants at the population level.
Collapse
|
66
|
Hofmann AL, Behr J, Singer J, Kuipers J, Beisel C, Schraml P, Moch H, Beerenwinkel N. Detailed simulation of cancer exome sequencing data reveals differences and common limitations of variant callers. BMC Bioinformatics 2017; 18:8. [PMID: 28049408 PMCID: PMC5209852 DOI: 10.1186/s12859-016-1417-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/10/2016] [Indexed: 12/30/2022] Open
Abstract
Background Next-generation sequencing of matched tumor and normal biopsy pairs has become a technology of paramount importance for precision cancer treatment. Sequencing costs have dropped tremendously, allowing the sequencing of the whole exome of tumors for just a fraction of the total treatment costs. However, clinicians and scientists cannot take full advantage of the generated data because the accuracy of analysis pipelines is limited. This particularly concerns the reliable identification of subclonal mutations in a cancer tissue sample with very low frequencies, which may be clinically relevant. Results Using simulations based on kidney tumor data, we compared the performance of nine state-of-the-art variant callers, namely deepSNV, GATK HaplotypeCaller, GATK UnifiedGenotyper, JointSNVMix2, MuTect, SAMtools, SiNVICT, SomaticSniper, and VarScan2. The comparison was done as a function of variant allele frequencies and coverage. Our analysis revealed that deepSNV and JointSNVMix2 perform very well, especially in the low-frequency range. We attributed false positive and false negative calls of the nine tools to specific error sources and assigned them to processing steps of the pipeline. All of these errors can be expected to occur in real data sets. We found that modifying certain steps of the pipeline or parameters of the tools can lead to substantial improvements in performance. Furthermore, a novel integration strategy that combines the ranks of the variants yielded the best performance. More precisely, the rank-combination of deepSNV, JointSNVMix2, MuTect, SiNVICT and VarScan2 reached a sensitivity of 78% when fixing the precision at 90%, and outperformed all individual tools, where the maximum sensitivity was 71% with the same precision. Conclusions The choice of well-performing tools for alignment and variant calling is crucial for the correct interpretation of exome sequencing data obtained from mixed samples, and common pipelines are suboptimal. We were able to relate observed substantial differences in performance to the underlying statistical models of the tools, and to pinpoint the error sources of false positive and false negative calls. These findings might inspire new software developments that improve exome sequencing pipelines and further the field of precision cancer treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1417-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ariane L Hofmann
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland.,Swiss Institute of Bioinformatics, Mattenstr, Basel, 26, 4058, Switzerland
| | - Jonas Behr
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland.,Swiss Institute of Bioinformatics, Mattenstr, Basel, 26, 4058, Switzerland
| | - Jochen Singer
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland.,Swiss Institute of Bioinformatics, Mattenstr, Basel, 26, 4058, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland.,Swiss Institute of Bioinformatics, Mattenstr, Basel, 26, 4058, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland
| | - Peter Schraml
- Institute for Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Holger Moch
- Institute for Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstr, Basel, 26, 4058, Switzerland. .,Swiss Institute of Bioinformatics, Mattenstr, Basel, 26, 4058, Switzerland.
| |
Collapse
|
67
|
Sheffield BS, Tessier-Cloutier B, Li-Chang H, Shen Y, Pleasance E, Kasaian K, Li Y, Jones SJM, Lim HJ, Renouf DJ, Huntsman DG, Yip S, Laskin J, Marra M, Schaeffer DF. Personalized oncogenomics in the management of gastrointestinal carcinomas-early experiences from a pilot study. ACTA ACUST UNITED AC 2016; 23:e571-e575. [PMID: 28050146 DOI: 10.3747/co.23.3165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Gastrointestinal carcinomas are genomically complex cancers that are lethal in the metastatic setting. Whole-genome and transcriptome sequencing allow for the simultaneous characterization of multiple oncogenic pathways. METHODS We report 3 cases of metastatic gastrointestinal carcinoma in patients enrolled in the Personalized Onco-Genomics program at the BC Cancer Agency. Real-time genomic profiling was combined with clinical expertise to diagnose a carcinoma of unknown primary, to explore treatment response to bevacizumab in a colorectal cancer, and to characterize an appendiceal adenocarcinoma. RESULTS In the first case, genomic profiling revealed an IDH1 somatic mutation, supporting the diagnosis of cholangiocarcinoma in a malignancy of unknown origin, and further guided therapy by identifying epidermal growth factor receptor amplification. In the second case, a BRAF V600E mutation and wild-type KRAS profile justified the use of targeted therapies to treat a colonic adenocarcinoma. The third case was an appendiceal adenocarcinoma defined by a p53 inactivation; Ras/raf/mek, Akt/mtor, Wnt, and notch pathway activation; and overexpression of ret, erbb2 (her2), erbb3, met, and cell cycle regulators. SUMMARY We show that whole-genome and transcriptome sequencing can be achieved within clinically effective timelines, yielding clinically useful and actionable information.
Collapse
Affiliation(s)
- B S Sheffield
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - B Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - H Li-Chang
- Royal Victoria Regional Health Centre, Department of Pathology and Laboratory Medicine, Barrie, ON
| | - Y Shen
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - E Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - K Kasaian
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - Y Li
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - S J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC
| | - H J Lim
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - D J Renouf
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - D G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - S Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| | - J Laskin
- Division of Medical Oncology, BC Cancer Agency, Vancouver, BC
| | - M Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC.; Department of Medical Genetics, University of British Columbia, Vancouver, BC
| | - D F Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
| |
Collapse
|
68
|
McIntyre JB, Rambau PF, Chan A, Yap S, Morris D, Nelson GS, Köbel M. Molecular alterations in indolent, aggressive and recurrent ovarian low-grade serous carcinoma. Histopathology 2016; 70:347-358. [DOI: 10.1111/his.13071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/29/2016] [Indexed: 02/04/2023]
Affiliation(s)
- John B McIntyre
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Peter F Rambau
- Department of Pathology; Catholic University of Health and Allied Sciences-Bugando; Mwanza Tanzania
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| | - Angela Chan
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Sidney Yap
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| | - Don Morris
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Gregg S Nelson
- Department of Gynecological Oncology; Tom Baker Cancer Centre; University of Calgary; Calgary Alberta Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| |
Collapse
|
69
|
Jakaitiene A, Avino M, Guarracino MR. Beta-Binomial Model for the Detection of Rare Mutations in Pooled Next-Generation Sequencing Experiments. J Comput Biol 2016; 24:357-367. [PMID: 27632638 DOI: 10.1089/cmb.2016.0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Against diminishing costs, next-generation sequencing (NGS) still remains expensive for studies with a large number of individuals. As cost saving, sequencing genome of pools containing multiple samples might be used. Currently, there are many software available for the detection of single-nucleotide polymorphisms (SNPs). Sensitivity and specificity depend on the model used and data analyzed, indicating that all software have space for improvement. We use beta-binomial model to detect rare mutations in untagged pooled NGS experiments. We propose a multireference framework for pooled data with ability being specific up to two patients affected by neuromuscular disorders (NMD). We assessed the results comparing with The Genome Analysis Toolkit (GATK), CRISP, SNVer, and FreeBayes. Our results show that the multireference approach applying beta-binomial model is accurate in predicting rare mutations at 0.01 fraction. Finally, we explored the concordance of mutations between the model and software, checking their involvement in any NMD-related gene. We detected seven novel SNPs, for which the functional analysis produced enriched terms related to locomotion and musculature.
Collapse
Affiliation(s)
- Audrone Jakaitiene
- 1 Bioinformatics and Biostatistics Center, Department of Human and Medical Genetics, Faculty of Medicine, Vilnius University , Vilnius, Lithuania
| | - Mariano Avino
- 2 High Performance Computing and Networking Institute , National Research Council, Naples, Italy
| | - Mario Rosario Guarracino
- 2 High Performance Computing and Networking Institute , National Research Council, Naples, Italy
| |
Collapse
|
70
|
Robust high-performance nanoliter-volume single-cell multiple displacement amplification on planar substrates. Proc Natl Acad Sci U S A 2016; 113:8484-9. [PMID: 27412862 DOI: 10.1073/pnas.1520964113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The genomes of large numbers of single cells must be sequenced to further understanding of the biological significance of genomic heterogeneity in complex systems. Whole genome amplification (WGA) of single cells is generally the first step in such studies, but is prone to nonuniformity that can compromise genomic measurement accuracy. Despite recent advances, robust performance in high-throughput single-cell WGA remains elusive. Here, we introduce droplet multiple displacement amplification (MDA), a method that uses commercially available liquid dispensing to perform high-throughput single-cell MDA in nanoliter volumes. The performance of droplet MDA is characterized using a large dataset of 129 normal diploid cells, and is shown to exceed previously reported single-cell WGA methods in amplification uniformity, genome coverage, and/or robustness. We achieve up to 80% coverage of a single-cell genome at 5× sequencing depth, and demonstrate excellent single-nucleotide variant (SNV) detection using targeted sequencing of droplet MDA product to achieve a median allelic dropout of 15%, and using whole genome sequencing to achieve false and true positive rates of 9.66 × 10(-6) and 68.8%, respectively, in a G1-phase cell. We further show that droplet MDA allows for the detection of copy number variants (CNVs) as small as 30 kb in single cells of an ovarian cancer cell line and as small as 9 Mb in two high-grade serous ovarian cancer samples using only 0.02× depth. Droplet MDA provides an accessible and scalable method for performing robust and accurate CNV and SNV measurements on large numbers of single cells.
Collapse
|
71
|
Salama MA, Hassanien AE, Mostafa A. The prediction of virus mutation using neural networks and rough set techniques. EURASIP JOURNAL ON BIOINFORMATICS & SYSTEMS BIOLOGY 2016; 2016:10. [PMID: 27257410 PMCID: PMC4867776 DOI: 10.1186/s13637-016-0042-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 05/03/2016] [Indexed: 11/10/2022]
Abstract
Viral evolution remains to be a main obstacle in the effectiveness of antiviral treatments. The ability to predict this evolution will help in the early detection of drug-resistant strains and will potentially facilitate the design of more efficient antiviral treatments. Various tools has been utilized in genome studies to achieve this goal. One of these tools is machine learning, which facilitates the study of structure-activity relationships, secondary and tertiary structure evolution prediction, and sequence error correction. This work proposes a novel machine learning technique for the prediction of the possible point mutations that appear on alignments of primary RNA sequence structure. It predicts the genotype of each nucleotide in the RNA sequence, and proves that a nucleotide in an RNA sequence changes based on the other nucleotides in the sequence. Neural networks technique is utilized in order to predict new strains, then a rough set theory based algorithm is introduced to extract these point mutation patterns. This algorithm is applied on a number of aligned RNA isolates time-series species of the Newcastle virus. Two different data sets from two sources are used in the validation of these techniques. The results show that the accuracy of this technique in predicting the nucleotides in the new generation is as high as 75 %. The mutation rules are visualized for the analysis of the correlation between different nucleotides in the same RNA sequence.
Collapse
Affiliation(s)
- Mostafa A Salama
- British University in Egypt (BUE), Cairo, Egypt ; Scientific Research Group in Egypt, (SRGE), Cairo, Egypt
| | - Aboul Ella Hassanien
- Cairo University, Cairo, Egypt ; Scientific Research Group in Egypt, (SRGE), Cairo, Egypt
| | | |
Collapse
|
72
|
Parker JDK, Shen Y, Pleasance E, Li Y, Schein JE, Zhao Y, Moore R, Wegrzyn-Woltosz J, Savage KJ, Weng AP, Gascoyne RD, Jones S, Marra M, Laskin J, Karsan A. Molecular etiology of an indolent lymphoproliferative disorder determined by whole-genome sequencing. Cold Spring Harb Mol Case Stud 2016; 2:a000679. [PMID: 27148583 PMCID: PMC4849852 DOI: 10.1101/mcs.a000679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In an attempt to assess potential treatment options, whole-genome and transcriptome sequencing were performed on a patient with an unclassifiable small lymphoproliferative disorder. Variants from genome sequencing were prioritized using a combination of comparative variant distributions in a spectrum of lymphomas, and meta-analyses of gene expression profiling. In this patient, the molecular variants that we believe to be most relevant to the disease presentation most strongly resemble a diffuse large B-cell lymphoma (DLBCL), whereas the gene expression data are most consistent with a low-grade chronic lymphocytic leukemia (CLL). The variant of greatest interest was a predicted NOTCH2-truncating mutation, which has been recently reported in various lymphomas.
Collapse
Affiliation(s)
- Jeremy D K Parker
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Yvonne Li
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Jacqueline E Schein
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Richard Moore
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Joanna Wegrzyn-Woltosz
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Kerry J Savage
- Centre for Lymphoid Cancer and Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Andrew P Weng
- Terry Fox Laboratory and Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Randy D Gascoyne
- Centre for Lymphoid Cancer and Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Steven Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Marco Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| | - Janessa Laskin
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4E6, Canada
| | - Aly Karsan
- Genome Sciences Centre and Department of Pathology, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 1L3, Canada
| |
Collapse
|
73
|
|
74
|
Sakr RA, Schizas M, Carniello JVS, Ng CKY, Piscuoglio S, Giri D, Andrade VP, De Brot M, Lim RS, Towers R, Weigelt B, Reis-Filho JS, King TA. Targeted capture massively parallel sequencing analysis of LCIS and invasive lobular cancer: Repertoire of somatic genetic alterations and clonal relationships. Mol Oncol 2015; 10:360-70. [PMID: 26643573 DOI: 10.1016/j.molonc.2015.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/09/2015] [Accepted: 11/03/2015] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Lobular carcinoma in situ (LCIS) has been proposed as a non-obligate precursor of invasive lobular carcinoma (ILC). Here we sought to define the repertoire of somatic genetic alterations in pure LCIS and in synchronous LCIS and ILC using targeted massively parallel sequencing. METHODS DNA samples extracted from microdissected LCIS, ILC and matched normal breast tissue or peripheral blood from 30 patients were subjected to massively parallel sequencing targeting all exons of 273 genes, including the genes most frequently mutated in breast cancer and DNA repair-related genes. Single nucleotide variants and insertions and deletions were identified using state-of-the-art bioinformatics approaches. RESULTS The constellation of somatic mutations found in LCIS (n = 34) and ILC (n = 21) were similar, with the most frequently mutated genes being CDH1 (56% and 66%, respectively), PIK3CA (41% and 52%, respectively) and CBFB (12% and 19%, respectively). Among 19 LCIS and ILC synchronous pairs, 14 (74%) had at least one identical mutation in common, including identical PIK3CA and CDH1 mutations. Paired analysis of independent foci of LCIS from 3 breasts revealed at least one common mutation in each of the 3 pairs (CDH1, PIK3CA, CBFB and PKHD1L1). CONCLUSION LCIS and ILC have a similar repertoire of somatic mutations, with PIK3CA and CDH1 being the most frequently mutated genes. The presence of identical mutations between LCIS-LCIS and LCIS-ILC pairs demonstrates that LCIS is a clonal neoplastic lesion, and provides additional evidence that at least some LCIS are non-obligate precursors of ILC.
Collapse
Affiliation(s)
- Rita A Sakr
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michail Schizas
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jose V Scarpa Carniello
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dilip Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Victor P Andrade
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Marina De Brot
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Raymond S Lim
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Russell Towers
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| | - Tari A King
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.
| |
Collapse
|
75
|
Szulwach KE, Chen P, Wang X, Wang J, Weaver LS, Gonzales ML, Sun G, Unger MA, Ramakrishnan R. Single-Cell Genetic Analysis Using Automated Microfluidics to Resolve Somatic Mosaicism. PLoS One 2015; 10:e0135007. [PMID: 26302375 PMCID: PMC4547741 DOI: 10.1371/journal.pone.0135007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/16/2015] [Indexed: 12/31/2022] Open
Abstract
Somatic mosaicism occurs throughout normal development and contributes to numerous disease etiologies, including tumorigenesis and neurological disorders. Intratumor genetic heterogeneity is inherent to many cancers, creating challenges for effective treatments. Unfortunately, analysis of bulk DNA masks subclonal phylogenetic architectures created by the acquisition and distribution of somatic mutations amongst cells. As a result, single-cell genetic analysis is becoming recognized as vital for accurately characterizing cancers. Despite this, methods for single-cell genetics are lacking. Here we present an automated microfluidic workflow enabling efficient cell capture, lysis, and whole genome amplification (WGA). We find that ~90% of the genome is accessible in single cells with improved uniformity relative to current single-cell WGA methods. Allelic dropout (ADO) rates were limited to 13.75% and variant false discovery rates (SNV FDR) were 4.11x10(-6), on average. Application to ER-/PR-/HER2+ breast cancer cells and matched normal controls identified novel mutations that arose in a subpopulation of cells and effectively resolved the segregation of known cancer-related mutations with single-cell resolution. Finally, we demonstrate effective cell classification using mutation profiles with 10X average exome coverage depth per cell. Our data demonstrate an efficient automated microfluidic platform for single-cell WGA that enables the resolution of somatic mutation patterns in single cells.
Collapse
Affiliation(s)
- Keith E. Szulwach
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Peilin Chen
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Xiaohui Wang
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Jing Wang
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Lesley S. Weaver
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Michael L. Gonzales
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Gang Sun
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Marc A. Unger
- Fluidigm Corporation, South San Francisco, California, United States of America
| | - Ramesh Ramakrishnan
- Fluidigm Corporation, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
76
|
Di Paolo A, Polillo M, Lastella M, Bocci G, Del Re M, Danesi R. Methods: for studying pharmacogenetic profiles of combination chemotherapeutic drugs. Expert Opin Drug Metab Toxicol 2015; 11:1253-67. [PMID: 26037261 DOI: 10.1517/17425255.2015.1053460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Molecular and genetic analysis of tumors and individuals has led to patient-centered therapies, through the discovery and identification of genetic markers predictive of drug efficacy and tolerability. Present therapies often include a combination of synergic drugs, each of them directed against different targets. Therefore, the pharmacogenetic profiling of tumor masses and patients is becoming a challenge, and several questions may arise when planning a translational study. AREAS COVERED The review presents the different techniques used to stratify oncology patients and to tailor antineoplastic treatments according to individual pharmacogenetic profiling. The advantages of these methodologies are discussed as well as current limits. EXPERT OPINION Facing the rapid technological evolution for genetic analyses, the most pressing issues are the choice of appropriate strategies (i.e., from gene candidate up to next-generation sequencing) and the possibility to replicate study results for their final validation. It is likely that the latter will be the major obstacle in the future. However, the present landscape is opening up new possibilities, overcoming those hurdles that have limited result translation into clinical settings for years.
Collapse
Affiliation(s)
- Antonello Di Paolo
- University of Pisa, Department of Clinical and Experimental Medicine, Via Roma 55, 56126 Pisa , Italy +39 050 2218755 ; +39 050 2218758 ;
| | | | | | | | | | | |
Collapse
|
77
|
Mathelier A, Lefebvre C, Zhang AW, Arenillas DJ, Ding J, Wasserman WW, Shah SP. Cis-regulatory somatic mutations and gene-expression alteration in B-cell lymphomas. Genome Biol 2015; 16:84. [PMID: 25903198 PMCID: PMC4467049 DOI: 10.1186/s13059-015-0648-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/07/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND With the rapid increase of whole-genome sequencing of human cancers, an important opportunity to analyze and characterize somatic mutations lying within cis-regulatory regions has emerged. A focus on protein-coding regions to identify nonsense or missense mutations disruptive to protein structure and/or function has led to important insights; however, the impact on gene expression of mutations lying within cis-regulatory regions remains under-explored. We analyzed somatic mutations from 84 matched tumor-normal whole genomes from B-cell lymphomas with accompanying gene expression measurements to elucidate the extent to which these cancers are disrupted by cis-regulatory mutations. RESULTS We characterize mutations overlapping a high quality set of well-annotated transcription factor binding sites (TFBSs), covering a similar portion of the genome as protein-coding exons. Our results indicate that cis-regulatory mutations overlapping predicted TFBSs are enriched in promoter regions of genes involved in apoptosis or growth/proliferation. By integrating gene expression data with mutation data, our computational approach culminates with identification of cis-regulatory mutations most likely to participate in dysregulation of the gene expression program. The impact can be measured along with protein-coding mutations to highlight key mutations disrupting gene expression and pathways in cancer. CONCLUSIONS Our study yields specific genes with disrupted expression triggered by genomic mutations in either the coding or the regulatory space. It implies that mutated regulatory components of the genome contribute substantially to cancer pathways. Our analyses demonstrate that identifying genomically altered cis-regulatory elements coupled with analysis of gene expression data will augment biological interpretation of mutational landscapes of cancers.
Collapse
Affiliation(s)
- Anthony Mathelier
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, V5Z 4H4, Vancouver, BC, Canada.
| | - Calvin Lefebvre
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, V5Z 1L3, BC, Canada. .,Bioinformatics Graduate Program, University of British Columbia, Vancouver, V5Z 1L3, BC, Canada.
| | - Allen W Zhang
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, V5Z 4H4, Vancouver, BC, Canada. .,Bioinformatics Graduate Program, University of British Columbia, Vancouver, V5Z 1L3, BC, Canada.
| | - David J Arenillas
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, V5Z 4H4, Vancouver, BC, Canada.
| | - Jiarui Ding
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, V5Z 1L3, BC, Canada. .,Department of Computer Science, University of British Columbia, Vancouver, V6T 1Z4, BC, Canada.
| | - Wyeth W Wasserman
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, V5Z 4H4, Vancouver, BC, Canada.
| | - Sohrab P Shah
- Department of Molecular Oncology, British Columbia Cancer Agency, Vancouver, V5Z 1L3, BC, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, G227-2211, BC, Canada.
| |
Collapse
|
78
|
Cancer genomics: why rare is valuable. J Mol Med (Berl) 2015; 93:369-81. [PMID: 25676695 PMCID: PMC4366545 DOI: 10.1007/s00109-015-1260-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/26/2014] [Accepted: 01/29/2015] [Indexed: 02/07/2023]
Abstract
Rare conditions are sometimes ignored in biomedical research because of difficulties in obtaining specimens and limited interest from fund raisers. However, the study of rare diseases such as unusual cancers has again and again led to breakthroughs in our understanding of more common diseases. It is therefore unsurprising that with the development and accessibility of next-generation sequencing, much has been learnt from studying cancers that are rare and in particular those with uniform biological and clinical behavior. Herein, we describe how shotgun sequencing of cancers such as granulosa cell tumor, endometrial stromal sarcoma, epithelioid hemangioendothelioma, ameloblastoma, small-cell carcinoma of the ovary, clear-cell carcinoma of the ovary, nonepithelial ovarian tumors, chondroblastoma, and giant cell tumor of the bone has led to rapidly translatable discoveries in diagnostics and tumor taxonomies, as well as providing insights into cancer biology.
Collapse
|
79
|
Tone AA, McConechy MK, Yang W, Ding J, Yip S, Kong E, Wong KK, Gershenson DM, Mackay H, Shah S, Gilks B, Tinker AV, Clarke B, McAlpine JN, Huntsman D. Intratumoral heterogeneity in a minority of ovarian low-grade serous carcinomas. BMC Cancer 2014; 14:982. [PMID: 25523272 PMCID: PMC4320586 DOI: 10.1186/1471-2407-14-982] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/11/2014] [Indexed: 12/20/2022] Open
Abstract
Background Ovarian low-grade serous carcinoma (LGSC) has fewer mutations than ovarian high-grade serous carcinoma (HGSC) and a less aggressive clinical course. However, an overwhelming majority of LGSC patients do not respond to conventional chemotherapy resulting in a poor long-term prognosis comparable to women diagnosed with HGSC. KRAS and BRAF mutations are common in LGSC, leading to clinical trials targeting the MAPK pathway. We assessed the stability of targetable somatic mutations over space and/or time in LGSC, with a view to inform stratified treatment strategies and clinical trial design. Methods Eleven LGSC cases with primary and recurrent paired samples were identified (stage IIB-IV). Tumor DNA was isolated from 1–4 formalin-fixed paraffin-embedded tumor blocks from both the primary and recurrence (n = 37 tumor and n = 7 normal samples). Mutational analysis was performed using the Ion Torrent AmpliSeqTM Cancer Panel, with targeted validation using Fluidigm-MiSeq, Sanger sequencing and/or Raindance Raindrop digital PCR. Results KRAS (3/11), BRAF (2/11) and/or NRAS (1/11) mutations were identified in five unique cases. A novel, non-synonymous mutation in SMAD4 was observed in one case. No somatic mutations were detected in the remaining six cases. In two cases with a single matched primary and recurrent sample, two KRAS hotspot mutations (G12V, G12R) were both stable over time. In three cases with multiple samplings from both the primary and recurrent surgery some mutations (NRAS Q61R, BRAF V600E, SMAD4 R361G) were stable across all samples, while others (KRAS G12V, BRAF G469V) were unstable. Conclusions Overall, the majority of cases with detectable somatic mutations showed mutational stability over space and time while one of five cases showed both temporal and spatial mutational instability in presumed drivers of disease. Investigation of additional cases is required to confirm whether mutational heterogeneity in a minority of LGSC is a general phenomenon that should be factored into the design of clinical trials and stratified treatment for this patient population. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-982) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - David Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
80
|
Bao R, Huang L, Andrade J, Tan W, Kibbe WA, Jiang H, Feng G. Review of current methods, applications, and data management for the bioinformatics analysis of whole exome sequencing. Cancer Inform 2014; 13:67-82. [PMID: 25288881 PMCID: PMC4179624 DOI: 10.4137/cin.s13779] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/06/2014] [Accepted: 07/07/2014] [Indexed: 12/21/2022] Open
Abstract
The advent of next-generation sequencing technologies has greatly promoted advances in the study of human diseases at the genomic, transcriptomic, and epigenetic levels. Exome sequencing, where the coding region of the genome is captured and sequenced at a deep level, has proven to be a cost-effective method to detect disease-causing variants and discover gene targets. In this review, we outline the general framework of whole exome sequence data analysis. We focus on established bioinformatics tools and applications that support five analytical steps: raw data quality assessment, pre-processing, alignment, post-processing, and variant analysis (detection, annotation, and prioritization). We evaluate the performance of open-source alignment programs and variant calling tools using simulated and benchmark datasets, and highlight the challenges posed by the lack of concordance among variant detection tools. Based on these results, we recommend adopting multiple tools and resources to reduce false positives and increase the sensitivity of variant calling. In addition, we briefly discuss the current status and solutions for big data management, analysis, and summarization in the field of bioinformatics.
Collapse
Affiliation(s)
- Riyue Bao
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Lei Huang
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Wei Tan
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York, USA
| | - Warren A Kibbe
- Biomedical Informatics Center (NUBIC), Clinical and Translational Sciences Institute (NUCATS), Northwestern University, Chicago, IL, USA
| | - Hongmei Jiang
- Department of Statistics, Northwestern University, Evanston, IL, USA
| | - Gang Feng
- Biomedical Informatics Center (NUBIC), Clinical and Translational Sciences Institute (NUCATS), Northwestern University, Chicago, IL, USA
| |
Collapse
|
81
|
Ferraro MB, Savarese M, Di Fruscio G, Nigro V, Guarracino MR. Prediction of rare single-nucleotide causative mutations for muscular diseases in pooled next-generation sequencing experiments. J Comput Biol 2014; 21:665-75. [PMID: 25029289 DOI: 10.1089/cmb.2014.0037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Next-generation sequencing (NGS) is a new approach for biomedical research, useful for the diagnosis of genetic diseases in extremely heterogeneous conditions. In this work, we describe how data generated by high-throughput NGS experiments can be analyzed to find single nucleotide polymorphisms (SNPs) in DNA samples of patients affected by neuromuscular disorders. In particular, we consider untagged pooled NGS data, where DNA samples of different individuals are combined in a single experiment, still providing information with an uncertainty limited to only two patients. At the moment, only few publications address the problem of SNPs detection in pooled experiments, and existing tools are often inaccurate. We propose a computational procedure consisting of two parts. In the first, data are filtered by means of decision rules. The second phase is based on a supervised classification technique. In the present work, we compare different de facto standard supervised and unsupervised procedures to identify and classify variants potentially related to muscular diseases, and we discuss results in terms of statistical and biological validation.
Collapse
|
82
|
Salari R, Saleh SS, Kashef-Haghighi D, Khavari D, Newburger DE, West RB, Sidow A, Batzoglou S. Inference of tumor phylogenies with improved somatic mutation discovery. J Comput Biol 2014; 20:933-44. [PMID: 24195709 DOI: 10.1089/cmb.2013.0106] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Next-generation sequencing technologies provide a powerful tool for studying genome evolution during progression of advanced diseases such as cancer. Although many recent studies have employed new sequencing technologies to detect mutations across multiple, genetically related tumors, current methods do not exploit available phylogenetic information to improve the accuracy of their variant calls. Here, we present a novel algorithm that uses somatic single-nucleotide variations (SNVs) in multiple, related tissue samples as lineage markers for phylogenetic tree reconstruction. Our method then leverages the inferred phylogeny to improve the accuracy of SNV discovery. Experimental analyses demonstrate that our method achieves up to 32% improvement for somatic SNV calling of multiple, related samples over the accuracy of GATK's Unified Genotyper, the state-of-the-art multisample SNV caller.
Collapse
Affiliation(s)
- Raheleh Salari
- 1 Department of Computer Science, Stanford University , Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Kim SY, Jacob L, Speed TP. Combining calls from multiple somatic mutation-callers. BMC Bioinformatics 2014; 15:154. [PMID: 24885750 PMCID: PMC4035752 DOI: 10.1186/1471-2105-15-154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 05/12/2014] [Indexed: 11/29/2022] Open
Abstract
Background Accurate somatic mutation-calling is essential for insightful mutation analyses in cancer studies. Several mutation-callers are publicly available and more are likely to appear. Nonetheless, mutation-calling is still challenging and there is unlikely to be one established caller that systematically outperforms all others. Therefore, fully utilizing multiple callers can be a powerful way to construct a list of final calls for one’s research. Results Using a set of mutations from multiple callers that are impartially validated, we present a statistical approach for building a combined caller, which can be applied to combine calls in a wider dataset generated using a similar protocol. Using the mutation outputs and the validation data from The Cancer Genome Atlas endometrial study (6,746 sites), we demonstrate how to build a statistical model that predicts the probability of each call being a somatic mutation, based on the detection status of multiple callers and a few associated features. Conclusion The approach allows us to build a combined caller across the full range of stringency levels, which outperforms all of the individual callers.
Collapse
Affiliation(s)
- Su Yeon Kim
- Department of Statistics, University of California at Berkeley, Berkeley CA 94720, USA.
| | | | | |
Collapse
|
84
|
Natrajan R, Wilkerson PM, Marchiò C, Piscuoglio S, Ng CKY, Wai P, Lambros MB, Samartzis EP, Dedes KJ, Frankum J, Bajrami I, Kopec A, Mackay A, A'hern R, Fenwick K, Kozarewa I, Hakas J, Mitsopoulos C, Hardisson D, Lord CJ, Kumar-Sinha C, Ashworth A, Weigelt B, Sapino A, Chinnaiyan AM, Maher CA, Reis-Filho JS. Characterization of the genomic features and expressed fusion genes in micropapillary carcinomas of the breast. J Pathol 2014; 232:553-65. [PMID: 24395524 PMCID: PMC4013428 DOI: 10.1002/path.4325] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/04/2013] [Accepted: 12/29/2013] [Indexed: 12/30/2022]
Abstract
Micropapillary carcinoma (MPC) is a rare histological special type of breast cancer, characterized by an aggressive clinical behaviour and a pattern of copy number aberrations (CNAs) distinct from that of grade- and oestrogen receptor (ER)-matched invasive carcinomas of no special type (IC-NSTs). The aims of this study were to determine whether MPCs are underpinned by a recurrent fusion gene(s) or mutations in 273 genes recurrently mutated in breast cancer. Sixteen MPCs were subjected to microarray-based comparative genomic hybridization (aCGH) analysis and Sequenom OncoCarta mutation analysis. Eight and five MPCs were subjected to targeted capture and RNA sequencing, respectively. aCGH analysis confirmed our previous observations about the repertoire of CNAs of MPCs. Sequencing analysis revealed a spectrum of mutations similar to those of luminal B IC-NSTs, and recurrent mutations affecting mitogen-activated protein kinase family genes and NBPF10. RNA-sequencing analysis identified 17 high-confidence fusion genes, eight of which were validated and two of which were in-frame. No recurrent fusions were identified in an independent series of MPCs and IC-NSTs. Forced expression of in-frame fusion genes (SLC2A1-FAF1 and BCAS4-AURKA) resulted in increased viability of breast cancer cells. In addition, genomic disruption of CDK12 caused by out-of-frame rearrangements was found in one MPC and in 13% of HER2-positive breast cancers, identified through a re-analysis of publicly available massively parallel sequencing data. In vitro analyses revealed that CDK12 gene disruption results in sensitivity to PARP inhibition, and forced expression of wild-type CDK12 in a CDK12-null cell line model resulted in relative resistance to PARP inhibition. Our findings demonstrate that MPCs are neither defined by highly recurrent mutations in the 273 genes tested, nor underpinned by a recurrent fusion gene. Although seemingly private genetic events, some of the fusion transcripts found in MPCs may play a role in maintenance of a malignant phenotype and potentially offer therapeutic opportunities.
Collapse
Affiliation(s)
- Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Paul M Wilkerson
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | | | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Charlotte KY Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Patty Wai
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Maryou B Lambros
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | | | | | - Jessica Frankum
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Ilirjana Bajrami
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Alicja Kopec
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Alan Mackay
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Roger A'hern
- Cancer Research UK Clinical Trials Unit, The Institute of Cancer ResearchSutton, UK
| | - Kerry Fenwick
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Iwanka Kozarewa
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Jarle Hakas
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Costas Mitsopoulos
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, Universidad Autonoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ)Madrid, Spain
| | - Christopher J Lord
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology (MCTP), Department of Pathology, University of MichiganAnn Arbor, MI, USA
| | - Alan Ashworth
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer ResearchLondon, UK
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| | - Anna Sapino
- Department of Medical Sciences, University of TurinTurin, Italy
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology (MCTP), Department of Pathology, University of MichiganAnn Arbor, MI, USA
| | - Christopher A Maher
- Washington University Genome Institute, Washington UniversitySt Louis, MO, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
| |
Collapse
|
85
|
Liu Y, Li B, Tan R, Zhu X, Wang Y. A gradient-boosting approach for filtering de novo mutations in parent-offspring trios. ACTA ACUST UNITED AC 2014; 30:1830-6. [PMID: 24618463 DOI: 10.1093/bioinformatics/btu141] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MOTIVATION Whole-genome and -exome sequencing on parent-offspring trios is a powerful approach to identifying disease-associated genes by detecting de novo mutations in patients. Accurate detection of de novo mutations from sequencing data is a critical step in trio-based genetic studies. Existing bioinformatic approaches usually yield high error rates due to sequencing artifacts and alignment issues, which may either miss true de novo mutations or call too many false ones, making downstream validation and analysis difficult. In particular, current approaches have much worse specificity than sensitivity, and developing effective filters to discriminate genuine from spurious de novo mutations remains an unsolved challenge. RESULTS In this article, we curated 59 sequence features in whole genome and exome alignment context which are considered to be relevant to discriminating true de novo mutations from artifacts, and then employed a machine-learning approach to classify candidates as true or false de novo mutations. Specifically, we built a classifier, named De Novo Mutation Filter (DNMFilter), using gradient boosting as the classification algorithm. We built the training set using experimentally validated true and false de novo mutations as well as collected false de novo mutations from an in-house large-scale exome-sequencing project. We evaluated DNMFilter's theoretical performance and investigated relative importance of different sequence features on the classification accuracy. Finally, we applied DNMFilter on our in-house whole exome trios and one CEU trio from the 1000 Genomes Project and found that DNMFilter could be coupled with commonly used de novo mutation detection approaches as an effective filtering approach to significantly reduce false discovery rate without sacrificing sensitivity. AVAILABILITY The software DNMFilter implemented using a combination of Java and R is freely available from the website at http://humangenome.duke.edu/software.
Collapse
Affiliation(s)
- Yongzhuang Liu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USASchool of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Bingshan Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Renjie Tan
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USASchool of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Xiaolin Zhu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China, Center for Human Genome Variation, Duke University, Durham, NC 27708 and Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
86
|
Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma. Nat Genet 2014; 46:329-35. [PMID: 24531327 DOI: 10.1038/ng.2900] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 01/24/2014] [Indexed: 12/20/2022]
Abstract
Classical Hodgkin lymphoma and primary mediastinal B cell lymphoma (PMBCL) are related lymphomas sharing pathological, molecular and clinical characteristics. Here we discovered by whole-genome and whole-transcriptome sequencing recurrent somatic coding-sequence mutations in the PTPN1 gene. Mutations were found in 6 of 30 (20%) Hodgkin lymphoma cases, in 6 of 9 (67%) Hodgkin lymphoma-derived cell lines, in 17 of 77 (22%) PMBCL cases and in 1 of 3 (33%) PMBCL-derived cell lines, consisting of nonsense, missense and frameshift mutations. We demonstrate that PTPN1 mutations lead to reduced phosphatase activity and increased phosphorylation of JAK-STAT pathway members. Moreover, silencing of PTPN1 by RNA interference in Hodgkin lymphoma cell line KM-H2 resulted in hyperphosphorylation and overexpression of downstream oncogenic targets. Our data establish PTPN1 mutations as new drivers in lymphomagenesis.
Collapse
|
87
|
Raphael BJ, Dobson JR, Oesper L, Vandin F. Identifying driver mutations in sequenced cancer genomes: computational approaches to enable precision medicine. Genome Med 2014; 6:5. [PMID: 24479672 PMCID: PMC3978567 DOI: 10.1186/gm524] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
High-throughput DNA sequencing is revolutionizing the study of cancer and enabling the measurement of the somatic mutations that drive cancer development. However, the resulting sequencing datasets are large and complex, obscuring the clinically important mutations in a background of errors, noise, and random mutations. Here, we review computational approaches to identify somatic mutations in cancer genome sequences and to distinguish the driver mutations that are responsible for cancer from random, passenger mutations. First, we describe approaches to detect somatic mutations from high-throughput DNA sequencing data, particularly for tumor samples that comprise heterogeneous populations of cells. Next, we review computational approaches that aim to predict driver mutations according to their frequency of occurrence in a cohort of samples, or according to their predicted functional impact on protein sequence or structure. Finally, we review techniques to identify recurrent combinations of somatic mutations, including approaches that examine mutations in known pathways or protein-interaction networks, as well as de novo approaches that identify combinations of mutations according to statistical patterns of mutual exclusivity. These techniques, coupled with advances in high-throughput DNA sequencing, are enabling precision medicine approaches to the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Benjamin J Raphael
- Department of Computer Science, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
| | - Jason R Dobson
- Department of Computer Science, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Layla Oesper
- Department of Computer Science, Brown University, 115 Waterman Street, Providence, RI 02912, USA
| | - Fabio Vandin
- Department of Computer Science, Brown University, 115 Waterman Street, Providence, RI 02912, USA
- Center for Computational Molecular Biology, Brown University, 115 Waterman Street, Providence, RI 02912, USA
| |
Collapse
|
88
|
McConechy MK, Ding J, Senz J, Yang W, Melnyk N, Tone AA, Prentice LM, Wiegand K, McAlpine JN, Shah SP, Lee CH, Goodfellow PJ, Gilks CB, Huntsman DG. Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles. Mod Pathol 2014; 27:128-34. [PMID: 23765252 PMCID: PMC3915240 DOI: 10.1038/modpathol.2013.107] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/10/2013] [Accepted: 05/11/2013] [Indexed: 12/19/2022]
Abstract
Ovarian endometrioid carcinomas and endometrial endometrioid carcinomas share many histological and molecular alterations. These similarities are likely due to a common endometrial epithelial precursor cell of origin, with most ovarian endometrioid carcinomas arising from endometriosis. To directly compare the mutation profiles of two morphologically similar tumor types, endometrial endometrioid carcinomas (n=307) and ovarian endometrioid carcinomas (n=33), we performed select exon capture sequencing on a panel of genes: ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, TP53. We found that PTEN mutations are more frequent in low-grade endometrial endometrioid carcinomas (67%) compared with low-grade ovarian endometrioid carcinomas (17%) (P<0.0001). By contrast, CTNNB1 mutations are significantly different in low-grade ovarian endometrioid carcinomas (53%) compared with low-grade endometrial endometrioid carcinomas (28%) (P<0.0057). This difference in CTNNB1 mutation frequency may be reflective of the distinct microenvironments; the epithelial cells lining an endometriotic cyst within the ovary are exposed to a highly oxidative environment that promotes tumorigenesis. Understanding the distinct mutation patterns found in the PI3K and Wnt pathways of ovarian and endometrial endometrioid carcinomas may provide future opportunities for stratifying patients for targeted therapeutics.
Collapse
Affiliation(s)
- Melissa K. McConechy
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Jiarui Ding
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada,Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Janine Senz
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Winnie Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Nataliya Melnyk
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Alicia A. Tone
- Division of Gynecology Oncology, Princess Margaret Cancer Centre, Toronto, Ontario
| | - Leah M. Prentice
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Kimberly Wiegand
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada
| | - Jessica N. McAlpine
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sohrab P. Shah
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada,Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Cheng-Han Lee
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada
| | - Paul J. Goodfellow
- Department of Obstetrics and Gynecology, College of Medicine, Ohio State University, Columbus, OH USA
| | - C. Blake Gilks
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada,Corresponding Authors: 1. David G. Huntsman, MD Department of Pathology and Laboratory Medicine, University of British Columbia, British Columbia Cancer Agency, 3427-600 West 10th Ave Vancouver, BC, Canada, V5E 4E6. Phone: 604-877-6000 Fax: 604-877-6089 , 2. C. Blake Gilks, MD, FRCPC, Anatomical Pathology, JP1400, Vancouver General Hospital, 910 West 10th Ave, Vancouver, BC, Canada V5Z 4E3. Phone: 604-875-4901 Fax: 604-877-3888
| | - David G. Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Centre for Translational & Applied Genomics, Vancouver, BC, Canada,Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada,Corresponding Authors: 1. David G. Huntsman, MD Department of Pathology and Laboratory Medicine, University of British Columbia, British Columbia Cancer Agency, 3427-600 West 10th Ave Vancouver, BC, Canada, V5E 4E6. Phone: 604-877-6000 Fax: 604-877-6089 , 2. C. Blake Gilks, MD, FRCPC, Anatomical Pathology, JP1400, Vancouver General Hospital, 910 West 10th Ave, Vancouver, BC, Canada V5Z 4E3. Phone: 604-875-4901 Fax: 604-877-3888
| |
Collapse
|
89
|
|
90
|
Kassahn KS, Holmes O, Nones K, Patch AM, Miller DK, Christ AN, Harliwong I, Bruxner TJ, Xu Q, Anderson M, Wood S, Leonard C, Taylor D, Newell F, Song S, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Steptoe A, Pajic M, Cowley MJ, Pinese M, Chang DK, Gill AJ, Johns AL, Wu J, Wilson PJ, Fink L, Biankin AV, Waddell N, Grimmond SM, Pearson JV. Somatic point mutation calling in low cellularity tumors. PLoS One 2013; 8:e74380. [PMID: 24250782 PMCID: PMC3826759 DOI: 10.1371/journal.pone.0074380] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/31/2013] [Indexed: 12/31/2022] Open
Abstract
Somatic mutation calling from next-generation sequencing data remains a challenge due to the difficulties of distinguishing true somatic events from artifacts arising from PCR, sequencing errors or mis-mapping. Tumor cellularity or purity, sub-clonality and copy number changes also confound the identification of true somatic events against a background of germline variants. We have developed a heuristic strategy and software (http://www.qcmg.org/bioinformatics/qsnp/) for somatic mutation calling in samples with low tumor content and we show the superior sensitivity and precision of our approach using a previously sequenced cell line, a series of tumor/normal admixtures, and 3,253 putative somatic SNVs verified on an orthogonal platform.
Collapse
Affiliation(s)
- Karin S. Kassahn
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Oliver Holmes
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Katia Nones
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ann-Marie Patch
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David K. Miller
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Angelika N. Christ
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ivon Harliwong
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Timothy J. Bruxner
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Qinying Xu
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew Anderson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Scott Wood
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Conrad Leonard
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Darrin Taylor
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Felicity Newell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Sarah Song
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Senel Idrisoglu
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Craig Nourse
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ehsan Nourbakhsh
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Suzanne Manning
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Shivangi Wani
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Anita Steptoe
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Mark J. Cowley
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Mark Pinese
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - David K. Chang
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Surgery, Bankstown Hospital, Sydney, New South Wales, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, New South Wales, Australia
| | - Anthony J. Gill
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Amber L. Johns
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Jianmin Wu
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Peter J. Wilson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Lynn Fink
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Andrew V. Biankin
- The Kinghorn Cancer Centre, and the Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Surgery, Bankstown Hospital, Sydney, New South Wales, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, New South Wales, Australia
| | - Nicola Waddell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Sean M. Grimmond
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - John V. Pearson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
91
|
Wang Q, Jia P, Li F, Chen H, Ji H, Hucks D, Dahlman KB, Pao W, Zhao Z. Detecting somatic point mutations in cancer genome sequencing data: a comparison of mutation callers. Genome Med 2013; 5:91. [PMID: 24112718 PMCID: PMC3971343 DOI: 10.1186/gm495] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/02/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Driven by high throughput next generation sequencing technologies and the pressing need to decipher cancer genomes, computational approaches for detecting somatic single nucleotide variants (sSNVs) have undergone dramatic improvements during the past 2 years. The recently developed tools typically compare a tumor sample directly with a matched normal sample at each variant locus in order to increase the accuracy of sSNV calling. These programs also address the detection of sSNVs at low allele frequencies, allowing for the study of tumor heterogeneity, cancer subclones, and mutation evolution in cancer development. METHODS We used whole genome sequencing (Illumina Genome Analyzer IIx platform) of a melanoma sample and matched blood, whole exome sequencing (Illumina HiSeq 2000 platform) of 18 lung tumor-normal pairs and seven lung cancer cell lines to evaluate six tools for sSNV detection: EBCall, JointSNVMix, MuTect, SomaticSniper, Strelka, and VarScan 2, with a focus on MuTect and VarScan 2, two widely used publicly available software tools. Default/suggested parameters were used to run these tools. The missense sSNVs detected in these samples were validated through PCR and direct sequencing of genomic DNA from the samples. We also simulated 10 tumor-normal pairs to explore the ability of these programs to detect low allelic-frequency sSNVs. RESULTS Out of the 237 sSNVs successfully validated in our cancer samples, VarScan 2 and MuTect detected the most of any tools (that is, 204 and 192, respectively). MuTect identified 11 more low-coverage validated sSNVs than VarScan 2, but missed 11 more sSNVs with alternate alleles in normal samples than VarScan 2. When examining the false calls of each tool using 169 invalidated sSNVs, we observed >63% false calls detected in the lung cancer cell lines had alternate alleles in normal samples. Additionally, from our simulation data, VarScan 2 identified more sSNVs than other tools, while MuTect characterized most low allelic-fraction sSNVs. CONCLUSIONS Our study explored the typical false-positive and false-negative detections that arise from the use of sSNV-calling tools. Our results suggest that despite recent progress, these tools have significant room for improvement, especially in the discrimination of low coverage/allelic-frequency sSNVs and sSNVs with alternate alleles in normal samples.
Collapse
Affiliation(s)
- Qingguo Wang
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Peilin Jia
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA ; Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fei Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China ; Department of Oncology, Shanghai Medical College, Shanghai, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Donald Hucks
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kimberly Brown Dahlman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA ; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - William Pao
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA ; Department of Medicine/Division of Hematology-Oncology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA ; Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA ; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA ; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
92
|
Goode DL, Hunter SM, Doyle MA, Ma T, Rowley SM, Choong D, Ryland GL, Campbell IG. A simple consensus approach improves somatic mutation prediction accuracy. Genome Med 2013; 5:90. [PMID: 24073752 PMCID: PMC3978449 DOI: 10.1186/gm494] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/20/2013] [Indexed: 12/14/2022] Open
Abstract
Differentiating true somatic mutations from artifacts in massively parallel sequencing data is an immense challenge. To develop methods for optimal somatic mutation detection and to identify factors influencing somatic mutation prediction accuracy, we validated predictions from three somatic mutation detection algorithms, MuTect, JointSNVMix2 and SomaticSniper, by Sanger sequencing. Full consensus predictions had a validation rate of >98%, but some partial consensus predictions validated too. In cases of partial consensus, read depth and mapping quality data, along with additional prediction methods, aided in removing inaccurate predictions. Our consensus approach is fast, flexible and provides a high-confidence list of putative somatic mutations.
Collapse
Affiliation(s)
- David L Goode
- Peter MacCallum Cancer Centre, Sarcoma Genetics and Genomics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Sally M Hunter
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia
| | - Maria A Doyle
- Peter MacCallum Cancer Centre, Bioinformatics Core Facility, St. Andrew's Place, East Melbourne, Victoria, Australia
| | - Tao Ma
- Peter MacCallum Cancer Centre, Bioinformatics Core Facility, St. Andrew's Place, East Melbourne, Victoria, Australia ; Bioinformatics Graduate Program, University of Melbourne, Parkville, Victoria, Australia
| | - Simone M Rowley
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia
| | - David Choong
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia
| | - Georgina L Ryland
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia ; Centre for Cancer Research, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Ian G Campbell
- Peter MacCallum Cancer Centre, Cancer Genetics Laboratory, St. Andrew's Place, East Melbourne, Victoria, Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia ; Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
93
|
Kasaian K, Wiseman SM, Thiessen N, Mungall KL, Corbett RD, Qian JQ, Nip KM, He A, Tse K, Chuah E, Varhol RJ, Pandoh P, McDonald H, Zeng T, Tam A, Schein J, Birol I, Mungall AJ, Moore RA, Zhao Y, Hirst M, Marra MA, Walker BA, Jones SJM. Complete genomic landscape of a recurring sporadic parathyroid carcinoma. J Pathol 2013; 230:249-60. [PMID: 23616356 DOI: 10.1002/path.4203] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/14/2013] [Accepted: 04/16/2013] [Indexed: 12/17/2022]
Abstract
Parathyroid carcinoma is a rare endocrine malignancy with an estimated incidence of less than 1 per million population. Excessive secretion of parathyroid hormone, extremely high serum calcium level, and the deleterious effects of hypercalcaemia are the clinical manifestations of the disease. Up to 60% of patients develop multiple disease recurrences and although long-term survival is possible with palliative surgery, permanent remission is rarely achieved. Molecular drivers of sporadic parathyroid carcinoma have remained largely unknown. Previous studies, mostly based on familial cases of the disease, suggested potential roles for the tumour suppressor MEN1 and proto-oncogene RET in benign parathyroid tumourigenesis, while the tumour suppressor HRPT2 and proto-oncogene CCND1 may also act as drivers in parathyroid cancer. Here, we report the complete genomic analysis of a sporadic and recurring parathyroid carcinoma. Mutational landscapes of the primary and recurrent tumour specimens were analysed using high-throughput sequencing technologies. Such molecular profiling allowed for identification of somatic mutations never previously identified in this malignancy. These included single nucleotide point mutations in well-characterized cancer genes such as mTOR, MLL2, CDKN2C, and PIK3CA. Comparison of acquired mutations in patient-matched primary and recurrent tumours revealed loss of PIK3CA activating mutation during the evolution of the tumour from the primary to the recurrence. Structural variations leading to gene fusions and regions of copy loss and gain were identified at a single-base resolution. Loss of the short arm of chromosome 1, along with somatic missense and truncating mutations in CDKN2C and THRAP3, respectively, provides new evidence for the potential role of these genes as tumour suppressors in parathyroid cancer. The key somatic mutations identified in this study can serve as novel diagnostic markers as well as therapeutic targets.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 2013; 122:1256-65. [PMID: 23699601 DOI: 10.1182/blood-2013-02-483727] [Citation(s) in RCA: 325] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous cancer composed of at least 2 molecular subtypes that differ in gene expression and distribution of mutations. Recently, application of genome/exome sequencing and RNA-seq to DLBCL has revealed numerous genes that are recurrent targets of somatic point mutation in this disease. Here we provide a whole-genome-sequencing-based perspective of DLBCL mutational complexity by characterizing 40 de novo DLBCL cases and 13 DLBCL cell lines and combining these data with DNA copy number analysis and RNA-seq from an extended cohort of 96 cases. Our analysis identified widespread genomic rearrangements including evidence for chromothripsis as well as the presence of known and novel fusion transcripts. We uncovered new gene targets of recurrent somatic point mutations and genes that are targeted by focal somatic deletions in this disease. We highlight the recurrence of germinal center B-cell-restricted mutations affecting genes that encode the S1P receptor and 2 small GTPases (GNA13 and GNAI2) that together converge on regulation of B-cell homing. We further analyzed our data to approximate the relative temporal order in which some recurrent mutations were acquired and demonstrate that ongoing acquisition of mutations and intratumoral clonal heterogeneity are common features of DLBCL. This study further improves our understanding of the processes and pathways involved in lymphomagenesis, and some of the pathways mutated here may indicate new avenues for therapeutic intervention.
Collapse
|
95
|
Hansen NF, Gartner JJ, Mei L, Samuels Y, Mullikin JC. Shimmer: detection of genetic alterations in tumors using next-generation sequence data. ACTA ACUST UNITED AC 2013; 29:1498-503. [PMID: 23620360 DOI: 10.1093/bioinformatics/btt183] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
MOTIVATION Extensive DNA sequencing of tumor and matched normal samples using exome and whole-genome sequencing technologies has enabled the discovery of recurrent genetic alterations in cancer cells, but variability in stromal contamination and subclonal heterogeneity still present a severe challenge to available detection algorithms. RESULTS Here, we describe publicly available software, Shimmer, which accurately detects somatic single-nucleotide variants using statistical hypothesis testing with multiple testing correction. This program produces somatic single-nucleotide variant predictions with significantly higher sensitivity and accuracy than other available software when run on highly contaminated or heterogeneous samples, and it gives comparable sensitivity and accuracy when run on samples of high purity. AVAILABILITY http://www.github.com/nhansen/Shimmer
Collapse
Affiliation(s)
- Nancy F Hansen
- Genome Technology Branch, NHGRI/NIH, Bethesda, MD 20892-9400, USA.
| | | | | | | | | |
Collapse
|
96
|
Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW. Cancer genome landscapes. Science 2013. [PMID: 23539594 DOI: 10.1126/science.123512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Over the past decade, comprehensive sequencing efforts have revealed the genomic landscapes of common forms of human cancer. For most cancer types, this landscape consists of a small number of "mountains" (genes altered in a high percentage of tumors) and a much larger number of "hills" (genes altered infrequently). To date, these studies have revealed ~140 genes that, when altered by intragenic mutations, can promote or "drive" tumorigenesis. A typical tumor contains two to eight of these "driver gene" mutations; the remaining mutations are passengers that confer no selective growth advantage. Driver genes can be classified into 12 signaling pathways that regulate three core cellular processes: cell fate, cell survival, and genome maintenance. A better understanding of these pathways is one of the most pressing needs in basic cancer research. Even now, however, our knowledge of cancer genomes is sufficient to guide the development of more effective approaches for reducing cancer morbidity and mortality.
Collapse
Affiliation(s)
- Bert Vogelstein
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
97
|
Abstract
Over the past decade, comprehensive sequencing efforts have revealed the genomic landscapes of common forms of human cancer. For most cancer types, this landscape consists of a small number of "mountains" (genes altered in a high percentage of tumors) and a much larger number of "hills" (genes altered infrequently). To date, these studies have revealed ~140 genes that, when altered by intragenic mutations, can promote or "drive" tumorigenesis. A typical tumor contains two to eight of these "driver gene" mutations; the remaining mutations are passengers that confer no selective growth advantage. Driver genes can be classified into 12 signaling pathways that regulate three core cellular processes: cell fate, cell survival, and genome maintenance. A better understanding of these pathways is one of the most pressing needs in basic cancer research. Even now, however, our knowledge of cancer genomes is sufficient to guide the development of more effective approaches for reducing cancer morbidity and mortality.
Collapse
Affiliation(s)
- Bert Vogelstein
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Nickolas Papadopoulos
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Victor E. Velculescu
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Shibin Zhou
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Luis A. Diaz
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| | - Kenneth W. Kinzler
- The Ludwig Center and The Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA
| |
Collapse
|
98
|
Abstract
Background Structural variations in human genomes, such as deletions, play an important role in cancer development. Next-Generation Sequencing technologies have been central in providing ways to detect such variations. Methods like paired-end mapping allow to simultaneously analyze data from several samples in order to, e.g., distinguish tumor from patient specific variations. However, it has been shown that, especially in this setting, there is a need to explicitly take overlapping deletions into consideration. Existing tools have only minor capabilities to call overlapping deletions, unable to unravel complex signals to obtain consistent predictions. Result We present a first approach specifically designed to cluster short-read paired-end data into possibly overlapping deletion predictions. The method does not make any assumptions on the composition of the data, such as the number of samples, heterogeneity, polyploidy, etc. Taking paired ends mapped to a reference genome as input, it iteratively merges mappings to clusters based on a similarity score that takes both the putative location and size of a deletion into account. Conclusion We demonstrate that agglomerative clustering is suitable to predict deletions. Analyzing real data from three samples of a cancer patient, we found putatively overlapping deletions and observed that, as a side-effect, erroneous mappings are mostly identified as singleton clusters. An evaluation on simulated data shows, compared to other methods which can output overlapping clusters, high accuracy in separating overlapping from single deletions.
Collapse
Affiliation(s)
- Roland Wittler
- Genome Informatics, Faculty of Technology and Institute for Bioinformatics, Center for Biotechnology, Bielefeld University, 33594 Bielefeld, Germany.
| |
Collapse
|
99
|
Paul R, Groza T, Hunter J, Zankl A. Decision support methods for finding phenotype--disorder associations in the bone dysplasia domain. PLoS One 2012; 7:e50614. [PMID: 23226331 PMCID: PMC3511538 DOI: 10.1371/journal.pone.0050614] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 10/26/2012] [Indexed: 11/18/2022] Open
Abstract
A lack of mature domain knowledge and well established guidelines makes the medical diagnosis of skeletal dysplasias (a group of rare genetic disorders) a very complex process. Machine learning techniques can facilitate objective interpretation of medical observations for the purposes of decision support. However, building decision support models using such techniques is highly problematic in the context of rare genetic disorders, because it depends on access to mature domain knowledge. This paper describes an approach for developing a decision support model in medical domains that are underpinned by relatively sparse knowledge bases. We propose a solution that combines association rule mining with the Dempster-Shafer theory (DST) to compute probabilistic associations between sets of clinical features and disorders, which can then serve as support for medical decision making (e.g., diagnosis). We show, via experimental results, that our approach is able to provide meaningful outcomes even on small datasets with sparse distributions, in addition to outperforming other Machine Learning techniques and behaving slightly better than an initial diagnosis by a clinician.
Collapse
Affiliation(s)
- Razan Paul
- School of ITEE, The University of Queensland, St. Lucia, Queensland, Australia
| | - Tudor Groza
- School of ITEE, The University of Queensland, St. Lucia, Queensland, Australia
| | - Jane Hunter
- School of ITEE, The University of Queensland, St. Lucia, Queensland, Australia
| | - Andreas Zankl
- Bone Dysplasia Research Group, UQ Centre for Clinical Research (UQCCR), The University of Queensland, Herston, Queensland, Australia
- Genetic Health Queensland, Royal Brisbane and Women’s Hospital, Herston, Queensland, Australia
| |
Collapse
|
100
|
Mutation discovery in regions of segmental cancer genome amplifications with CoNAn-SNV: a mixture model for next generation sequencing of tumors. PLoS One 2012; 7:e41551. [PMID: 22916110 PMCID: PMC3420914 DOI: 10.1371/journal.pone.0041551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 06/27/2012] [Indexed: 01/08/2023] Open
Abstract
Next generation sequencing has now enabled a cost-effective enumeration of the full mutational complement of a tumor genome—in particular single nucleotide variants (SNVs). Most current computational and statistical models for analyzing next generation sequencing data, however, do not account for cancer-specific biological properties, including somatic segmental copy number alterations (CNAs)—which require special treatment of the data. Here we present CoNAn-SNV (Copy Number Annotated SNV): a novel algorithm for the inference of single nucleotide variants (SNVs) that overlap copy number alterations. The method is based on modelling the notion that genomic regions of segmental duplication and amplification induce an extended genotype space where a subset of genotypes will exhibit heavily skewed allelic distributions in SNVs (and therefore render them undetectable by methods that assume diploidy). We introduce the concept of modelling allelic counts from sequencing data using a panel of Binomial mixture models where the number of mixtures for a given locus in the genome is informed by a discrete copy number state given as input. We applied CoNAn-SNV to a previously published whole genome shotgun data set obtained from a lobular breast cancer and show that it is able to discover 21 experimentally revalidated somatic non-synonymous mutations in a lobular breast cancer genome that were not detected using copy number insensitive SNV detection algorithms. Importantly, ROC analysis shows that the increased sensitivity of CoNAn-SNV does not result in disproportionate loss of specificity. This was also supported by analysis of a recently published lymphoma genome with a relatively quiescent karyotype, where CoNAn-SNV showed similar results to other callers except in regions of copy number gain where increased sensitivity was conferred. Our results indicate that in genomically unstable tumors, copy number annotation for SNV detection will be critical to fully characterize the mutational landscape of cancer genomes.
Collapse
|