101
|
CNspector: a web-based tool for visualisation and clinical diagnosis of copy number variation from next generation sequencing. Sci Rep 2019; 9:6426. [PMID: 31015508 PMCID: PMC6478945 DOI: 10.1038/s41598-019-42858-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/08/2019] [Indexed: 11/23/2022] Open
Abstract
Next Generation Sequencing is now routinely used in the practice of diagnostic pathology to detect clinically relevant somatic and germline sequence variations in patient samples. However, clinical assessment of copy number variations (CNVs) and large-scale structural variations (SVs) is still challenging. While tools exist to estimate both, their results are typically presented separately in tables or static plots which can be difficult to read and are unable to show the context needed for clinical interpretation and reporting. We have addressed this problem with CNspector, a multi-scale interactive browser that shows CNVs in the context of other relevant genomic features to enable fast and effective clinical reporting. We illustrate the utility of CNspector at different genomic scales across a variety of sample types in a range of case studies. We show how CNspector can be used for diagnosis and reporting of exon-level deletions, focal gene-level amplifications, chromosome and chromosome arm level amplifications/deletions and in complex genomic rearrangements. CNspector is a web-based clinical variant browser tailored to the clinical application of next generation sequencing for CNV assessment. We have demonstrated the utility of this interactive software in typical applications across a range of tissue types and disease contexts encountered in the context of diagnostic pathology. CNspector is written in R and the source code is available for download under the GPL3 Licence from https://github.com/PapenfussLab/CNspector.
Collapse
|
102
|
Kader T, Hill P, Zethoven M, Goode DL, Elder K, Thio N, Doyle M, Semple T, Sufyan W, Byrne DJ, Pang JMB, Murugasu A, Miligy IM, Green AR, Rakha EA, Fox SB, Mann GB, Campbell IG, Gorringe KL. Atypical ductal hyperplasia is a multipotent precursor of breast carcinoma. J Pathol 2019; 248:326-338. [PMID: 30843206 DOI: 10.1002/path.5262] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022]
Abstract
The current model for breast cancer progression proposes independent 'low grade (LG)-like' and 'high grade (HG)-like' pathways but lacks a known precursor to HG cancer. We applied low-coverage whole-genome sequencing to atypical ductal hyperplasia (ADH) with and without carcinoma to shed light on breast cancer progression. Fourteen out of twenty isolated ADH cases harboured at least one copy number alteration (CNA), but had fewer aberrations than LG or HG ductal carcinoma in situ (DCIS). ADH carried more HG-like CNA than LG DCIS (e.g. 8q gain). Correspondingly, 64% (7/11) of ADH cases with synchronous HG carcinoma were clonally related, similar to LG carcinoma (67%, 6/9). This study represents a significant shift in our understanding of breast cancer progression, with ADH as a common precursor lesion to the independent 'low grade-like' and 'high grade-like' pathways. These data suggest that ADH can be a precursor of HG breast cancer and that LG and HG carcinomas can evolve from a similar ancestor lesion. We propose that although LG DCIS may be committed to a LG molecular pathway, ADH may remain multipotent, progressing to either LG or HG carcinoma. This multipotent nature suggests that some ADH cases could be more clinically significant than LG DCIS, requiring biomarkers for personalising management. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tanjina Kader
- Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Prue Hill
- Department of Anatomical Pathology, St Vincent's Hospital, Fitzroy, Australia
| | | | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Kenneth Elder
- The Breast Service, The Royal Women's Hospital, Melbourne, Australia
| | - Niko Thio
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Maria Doyle
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Wajiha Sufyan
- Territory Pathology, Royal Darwin Hospital, Darwin, Australia
| | | | | | - Anand Murugasu
- The Breast Service, The Royal Women's Hospital, Melbourne, Australia
| | - Islam M Miligy
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Histopathology, Nottingham University Hospitals NHS Trust, City Hospital, Nottingham, UK
| | - Andrew R Green
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Histopathology, Nottingham University Hospitals NHS Trust, City Hospital, Nottingham, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Histopathology, Nottingham University Hospitals NHS Trust, City Hospital, Nottingham, UK
| | | | - G Bruce Mann
- The Breast Service, The Royal Women's Hospital, Melbourne, Australia
| | - Ian G Campbell
- Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Australia
| | - Kylie L Gorringe
- Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
103
|
Mack SC, Singh I, Wang X, Hirsch R, Wu Q, Villagomez R, Bernatchez JA, Zhu Z, Gimple RC, Kim LJY, Morton A, Lai S, Qiu Z, Prager BC, Bertrand KC, Mah C, Zhou W, Lee C, Barnett GH, Vogelbaum MA, Sloan AE, Chavez L, Bao S, Scacheri PC, Siqueira-Neto JL, Lin CY, Rich JN. Chromatin landscapes reveal developmentally encoded transcriptional states that define human glioblastoma. J Exp Med 2019; 216:1071-1090. [PMID: 30948495 PMCID: PMC6504206 DOI: 10.1084/jem.20190196] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022] Open
Abstract
Mack et al. defined active chromatin landscapes of glioblastoma stem cells (GSCs) and primary tumor specimens, revealing novel transcriptional regulatory circuits and therapeutic targets. Super-enhancers identified essential transcription factors that underlie GSC identity and intertumoral diversity, potentially informing precision medicine. Glioblastoma is an incurable brain cancer characterized by high genetic and pathological heterogeneity. Here, we mapped active chromatin landscapes with gene expression, whole exomes, copy number profiles, and DNA methylomes across 44 patient-derived glioblastoma stem cells (GSCs), 50 primary tumors, and 10 neural stem cells (NSCs) to identify essential super-enhancer (SE)–associated genes and the core transcription factors that establish SEs and maintain GSC identity. GSCs segregate into two groups dominated by distinct enhancer profiles and unique developmental core transcription factor regulatory programs. Group-specific transcription factors enforce GSC identity; they exhibit higher activity in glioblastomas versus NSCs, are associated with poor clinical outcomes, and are required for glioblastoma growth in vivo. Although transcription factors are commonly considered undruggable, group-specific enhancer regulation of the MAPK/ERK pathway predicts sensitivity to MEK inhibition. These data demonstrate that transcriptional identity can be leveraged to identify novel dependencies and therapeutic approaches.
Collapse
Affiliation(s)
- Stephen C Mack
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Baylor College of Medicine, Houston, TX
| | - Irtisha Singh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Xiuxing Wang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Rachel Hirsch
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Quilian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Rosie Villagomez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Jean A Bernatchez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA.,Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, La Jolla, CA
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA.,Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA.,Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Andrew Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| | - Sisi Lai
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| | - Zhixin Qiu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA.,Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Kelsey C Bertrand
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Baylor College of Medicine, Houston, TX
| | - Clarence Mah
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Wenchao Zhou
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Christine Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Gene H Barnett
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH
| | - Michael A Vogelbaum
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH
| | - Andrew E Sloan
- Department of Neurological Surgery, Seidman Cancer Center & University Hospitals - Cleveland Medical Center, Cleveland, OH
| | - Lukas Chavez
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Peter C Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA.,Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, La Jolla, CA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX .,Therapeutic Innovation Center, Verna and Marrs McClean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA
| |
Collapse
|
104
|
Beyond sequence variation: assessment of copy number variation in adult glioblastoma through targeted tumor somatic profiling. Hum Pathol 2019; 86:170-181. [DOI: 10.1016/j.humpath.2018.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/06/2018] [Accepted: 12/14/2018] [Indexed: 01/07/2023]
|
105
|
Krimpenfort P, Snoek M, Lambooij JP, Song JY, van der Weide R, Bhaskaran R, Teunissen H, Adams DJ, de Wit E, Berns A. A natural WNT signaling variant potently synergizes with Cdkn2ab loss in skin carcinogenesis. Nat Commun 2019; 10:1425. [PMID: 30926782 PMCID: PMC6441055 DOI: 10.1038/s41467-019-09321-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
Cdkn2ab knockout mice, generated from 129P2 ES cells develop skin carcinomas. Here we show that the incidence of these carcinomas drops gradually in the course of backcrossing to the FVB/N background. Microsatellite analyses indicate that this cancer phenotype is linked to a 20 Mb region of 129P2 chromosome 15 harboring the Wnt7b gene, which is preferentially expressed from the 129P2 allele in skin carcinomas and derived cell lines. ChIPseq analysis shows enrichment of H3K27-Ac, a mark for active enhancers, in the 5' region of the Wnt7b 129P2 gene. The Wnt7b 129P2 allele appears sufficient to cause in vitro transformation of Cdkn2ab-deficient cell lines primarily through CDK6 activation. These results point to a critical role of the Cdkn2ab locus in keeping the oncogenic potential of physiological levels of WNT signaling in check and illustrate that GWAS-based searches for cancer predisposing allelic variants can be enhanced by including defined somatically acquired lesions as an additional input.
Collapse
Affiliation(s)
- Paul Krimpenfort
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Margriet Snoek
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Jan-Paul Lambooij
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Robin van der Weide
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Rajith Bhaskaran
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Hans Teunissen
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - David J Adams
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Elzo de Wit
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Anton Berns
- Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
106
|
de Matos MR, Posa I, Carvalho FS, Morais VA, Grosso AR, de Almeida SF. A Systematic Pan-Cancer Analysis of Genetic Heterogeneity Reveals Associations with Epigenetic Modifiers. Cancers (Basel) 2019; 11:E391. [PMID: 30897760 PMCID: PMC6468518 DOI: 10.3390/cancers11030391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/09/2019] [Accepted: 03/17/2019] [Indexed: 12/30/2022] Open
Abstract
Intratumor genetic heterogeneity (ITH) is the main obstacle to effective cancer treatment and a major mechanism of drug resistance. It results from the continuous evolution of different clones of a tumor over time. However, the molecular features underlying the emergence of genetically-distinct subclonal cell populations remain elusive. Here, we conducted an exhaustive characterization of ITH across 2807 tumor samples from 16 cancer types. Integration of ITH scores and somatic variants detected in each tumor sample revealed that mutations in epigenetic modifier genes are associated with higher ITH levels. In particular, genes that regulate genome-wide histone and DNA methylation emerged as being determinant of high ITH. Indeed, the knockout of histone methyltransferase SETD2 or DNA methyltransferase DNMT3A using the CRISPR/Cas9 system on cancer cells led to significant expansion of genetically-distinct clones and culminated in highly heterogeneous cell populations. The ITH scores observed in knockout cells recapitulated the heterogeneity levels observed in patient tumor samples and correlated with a better mitochondrial bioenergetic performance under stress conditions. Our work provides new insights into tumor development, and discloses new drivers of ITH, which may be useful as either predictive biomarkers or therapeutic targets to improve cancer treatment.
Collapse
Affiliation(s)
- Mafalda Ramos de Matos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Ioana Posa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Filipa Sofia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Vanessa Alexandra Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana Rita Grosso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.
| | - Sérgio Fernandes de Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
107
|
Integrated analysis of relapsed B-cell precursor Acute Lymphoblastic Leukemia identifies subtype-specific cytokine and metabolic signatures. Sci Rep 2019; 9:4188. [PMID: 30862934 PMCID: PMC6414622 DOI: 10.1038/s41598-019-40786-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/22/2019] [Indexed: 12/20/2022] Open
Abstract
Recent efforts reclassified B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) into more refined subtypes. Nevertheless, outcomes of relapsed BCP-ALL remain unsatisfactory, particularly in adult patients where the molecular basis of relapse is still poorly understood. To elucidate the evolution of relapse in BCP-ALL, we established a comprehensive multi-omics dataset including DNA-sequencing, RNA-sequencing, DNA methylation array and proteome MASS-spec data from matched diagnosis and relapse samples of BCP-ALL patients (n = 50) including the subtypes DUX4, Ph-like and two aneuploid subtypes. Relapse-specific alterations were enriched for chromatin modifiers, nucleotide and steroid metabolism including the novel candidates FPGS, AGBL and ZNF483. The proteome expression analysis unraveled deregulation of metabolic pathways at relapse including the key proteins G6PD, TKT, GPI and PGD. Moreover, we identified a novel relapse-specific gene signature specific for DUX4 BCP-ALL patients highlighting chemotaxis and cytokine environment as a possible driver event at relapse. This study presents novel insights at distinct molecular levels of relapsed BCP-ALL based on a comprehensive multi-omics integrated data set including a valuable proteomics data set. The relapse specific aberrations reveal metabolic signatures on genomic and proteomic levels in BCP-ALL relapse. Furthermore, the chemokine expression signature in DUX4 relapse underscores the distinct status of DUX4-fusion BCP-ALL.
Collapse
|
108
|
Salinas EA, Miller MD, Newtson AM, Sharma D, McDonald ME, Keeney ME, Smith BJ, Bender DP, Goodheart MJ, Thiel KW, Devor EJ, Leslie KK, Gonzalez Bosquet J. A Prediction Model for Preoperative Risk Assessment in Endometrial Cancer Utilizing Clinical and Molecular Variables. Int J Mol Sci 2019; 20:ijms20051205. [PMID: 30857319 PMCID: PMC6429416 DOI: 10.3390/ijms20051205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 01/27/2023] Open
Abstract
The utility of comprehensive surgical staging in patients with low risk disease has been questioned. Thus, a reliable means of determining risk would be quite useful. The aim of our study was to create the best performing prediction model to classify endometrioid endometrial cancer (EEC) patients into low or high risk using a combination of molecular and clinical-pathological variables. We then validated these models with publicly available datasets. Analyses between low and high risk EEC were performed using clinical and pathological data, gene and miRNA expression data, gene copy number variation and somatic mutation data. Variables were selected to be included in the prediction model of risk using cross-validation analysis; prediction models were then constructed using these variables. Model performance was assessed by area under the curve (AUC). Prediction models were validated using appropriate datasets in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A prediction model with only clinical variables performed at 88%. Integrating clinical and molecular data improved prediction performance up to 97%. The best prediction models included clinical, miRNA expression and/or somatic mutation data, and stratified pre-operative risk in EEC patients. Integrating molecular and clinical data improved the performance of prediction models to over 95%, resulting in potentially useful clinical tests.
Collapse
Affiliation(s)
| | - Marina D Miller
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Andreea M Newtson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Deepti Sharma
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 52242, USA.
| | - Megan E McDonald
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Matthew E Keeney
- Winfield Pathology Consultants, Central DuPage Hospital, Winfield, IL 60190, USA.
| | - Brian J Smith
- Department of Biostatistics, University of Iowa College of Public Health, Iowa City, IA 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - David P Bender
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Michael J Goodheart
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Kristina W Thiel
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| | - Jesus Gonzalez Bosquet
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| |
Collapse
|
109
|
Cheasley D, Li N, Rowley SM, Elder K, Mann GB, Loi S, Savas P, Goode DL, Kader T, Zethoven M, Semple T, Fox SB, Pang JM, Byrne D, Devereux L, Nickson C, Procopio P, Lee G, Hughes S, Saunders H, Fujihara KM, Kuykhoven K, Connaughton J, James PA, Gorringe KL, Campbell IG. Molecular comparison of interval and screen-detected breast cancers. J Pathol 2019; 248:243-252. [PMID: 30746706 DOI: 10.1002/path.5251] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 01/22/2023]
Abstract
Breast cancer (BC) diagnosed after a negative mammogram but prior to the next screening episode is termed an 'interval BC' (IBC). Understanding the molecular differences between IBC and screen-detected BCs (SDBC) could improve mammographic screening and management options. Therefore, we assessed both germline and somatic genomic aberrations in a prospective cohort. Utilising the Lifepool cohort of >54 000 women attending mammographic screening programs, 930 BC cases with screening status were identified (726 SDBC and 204 IBC). Clinico-pathological and family history information were recorded. Germline and tumour DNA were collected where available and sequenced for BC predisposition and driver gene mutations. Compared to SDBC, IBCs were significantly associated with a younger age at diagnosis and tumour characteristics associated with worse prognosis. Germline DNA assessment of BC cases that developed post-enrolment (276 SDBCs and 77 IBCs) for pathogenic mutations in 12 hereditary BC predisposition genes identified 8 carriers (2.27%). The germline mutation frequency was higher in IBC versus SDBC, although not statistically significant (3.90% versus 1.81%, p = 0.174). Comparing somatic genetic features of IBC and SDBC matched for grade, histological subtype and hormone receptor revealed no significant differences, with the exception of higher homologous recombination deficiency scores in IBC, and copy number changes on chromosome Xq in triple negative SDBCs. Our data demonstrates that while IBCs are clinically more aggressive than SDBC, when matched for confounding clinico-pathological features they do not represent a unique molecular class of invasive BC, but could be a consequence of timing of tumour initiation and mammographic screening. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kenneth Elder
- Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.,The Royal Melbourne and Royal Women's Hospitals, Parkville, Victoria, Australia.,The Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - G Bruce Mann
- Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia.,The Royal Melbourne and Royal Women's Hospitals, Parkville, Victoria, Australia
| | - Sherene Loi
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Savas
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David L Goode
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Tanjina Kader
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Magnus Zethoven
- Bioinformatics Consulting Core, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Tim Semple
- Genomics Core, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - Jia-Min Pang
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - David Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, Victoria, Australia
| | - Lisa Devereux
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Lifepool, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Carolyn Nickson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Pietro Procopio
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Grant Lee
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Siobhan Hughes
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Hugo Saunders
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kenji M Fujihara
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Keilly Kuykhoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jacquie Connaughton
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Cancer Genetics and Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
110
|
Kim B, Lee H, Jang J, Kim SJ, Lee ST, Cheong JW, Lyu CJ, Min YH, Choi JR. Targeted next generation sequencing can serve as an alternative to conventional tests in myeloid neoplasms. PLoS One 2019; 14:e0212228. [PMID: 30840646 PMCID: PMC6402635 DOI: 10.1371/journal.pone.0212228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/29/2019] [Indexed: 12/30/2022] Open
Abstract
The 2016 World Health Organization classification introduced a number of genes with somatic mutations and a category for germline predisposition syndromes in myeloid neoplasms. We have designed a comprehensive next-generation sequencing assay to detect somatic mutations, translocations, and germline mutations in a single assay and have evaluated its clinical utility in patients with myeloid neoplasms. Extensive and specified bioinformatics analyses were undertaken to detect single nucleotide variations, FLT3 internal tandem duplication, genic copy number variations, and chromosomal copy number variations. This enabled us to maximize the clinical utility of the assay, and we concluded that, as a single assay, it can be a good supplement for many conventional tests, including Sanger sequencing, RT-PCR, and cytogenetics. Of note, we found that 8.4-11.6% of patients with acute myeloid leukemia and 12.9% of patients with myeloproliferative neoplasms had germline mutations, and most were heterozygous carriers for autosomal recessive marrow failure syndromes. These patients often did not respond to standard chemotherapy, suggesting that germline predisposition may have distinct and significant clinical implications.
Collapse
Affiliation(s)
- Borahm Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyeonah Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Jieun Jang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Jeong Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (STL); (JRC)
| | - June-Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chuhl Joo Lyu
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Yoo Hong Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (STL); (JRC)
| |
Collapse
|
111
|
PAX5 biallelic genomic alterations define a novel subgroup of B-cell precursor acute lymphoblastic leukemia. Leukemia 2019; 33:1895-1909. [PMID: 30842609 DOI: 10.1038/s41375-019-0430-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 11/08/2022]
Abstract
Chromosomal rearrangements and specific aneuploidy patterns are initiating events and define subgroups in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Here we analyzed 250 BCP-ALL cases and identified a novel subgroup ('PAX5-plus', n = 19) by distinct DNA methylation and gene expression profiles. All patients in this subgroup harbored mutations in the B-lineage transcription factor PAX5, with p.P80R as hotspot. Mutations either affected two independent codons, consistent with compound heterozygosity, or suffered LOH predominantly through chromosome 9p aberrations. These biallelic events resulted in disruption of PAX5 transcriptional programs regulating B-cell differentiation and tumor suppressor functions. Homozygous CDKN2A/B deletions and RAS-activating hotspot mutations were highly enriched as cooperating events in the genomic profile of PAX5-plus ALL. Together, this defined a specific pattern of triple alterations, exclusive to the novel subgroup. PAX5-plus ALL was observed in pediatric and adult patients. Although restricted by the limited sample size, a tendency for more favorable clinical outcome was observed, with 10 of 12 adult PAX5-plus patients achieving long-term survival. PAX5-plus represents the first BCP-ALL subgroup defined by sequence alterations in contrast to gross chromosomal events and exemplifies how deregulated differentiation (PAX5), impaired cell cycle control (CDKN2A/B) and sustained proliferative signaling (RAS) cooperatively drive leukemogenesis.
Collapse
|
112
|
Park H, Chun SM, Shim J, Oh JH, Cho EJ, Hwang HS, Lee JY, Kim D, Jang SJ, Nam SJ, Hwang C, Sohn I, Sung CO. Detection of chromosome structural variation by targeted next-generation sequencing and a deep learning application. Sci Rep 2019; 9:3644. [PMID: 30842562 PMCID: PMC6403216 DOI: 10.1038/s41598-019-40364-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Molecular testing is increasingly important in cancer diagnosis. Targeted next generation sequencing (NGS) is widely accepted method but structural variation (SV) detection by targeted NGS remains challenging. In the brain tumor, identification of molecular alterations, including 1p/19q co-deletion, is essential for accurate glial tumor classification. Hence, we used targeted NGS to detect 1p/19q co-deletion using a newly developed deep learning (DL) model in 61 tumors, including 19 oligodendroglial tumors. An ensemble 1-dimentional convolution neural network was developed and used to detect the 1p/19q co-deletion. External validation was performed using 427 low-grade glial tumors from The Cancer Genome Atlas (TCGA). Manual review of the copy number plot from the targeted NGS identified the 1p/19q co-deletion in all 19 oligodendroglial tumors. Our DL model also perfectly detected the 1p/19q co-deletion (area under the curve, AUC = 1) in the testing set, and yielded reproducible results (AUC = 0.9652) in the validation set (n = 427), although the validation data were generated on a completely different platform (SNP Array 6.0 platform). In conclusion, targeted NGS using a cancer gene panel is a promising approach for classifying glial tumors, and DL can be successfully integrated for the SV detection in NGS data.
Collapse
Affiliation(s)
- Hosub Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sung-Min Chun
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jooyong Shim
- Institute of Statistical Information, Department of Statistics, Inje University, Gyeongsangnam-do, Korea
| | - Ji-Hye Oh
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Eun Jeong Cho
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Hee Sang Hwang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ji-Young Lee
- Center for Cancer Genome Discovery, Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Deokhoon Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Se Jin Jang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Changha Hwang
- Department of Applied Statistics, Dankook University, Gyeonggido, Korea.
| | - Insuk Sohn
- Biostatistics and Clinical Epidemiology Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.
| | - Chang Ohk Sung
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea. .,Center for Cancer Genome Discovery, Asan Institute for Life Science, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea. .,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
113
|
Genetic drivers of oncogenic pathways in molecular subgroups of peripheral T-cell lymphoma. Blood 2019; 133:1664-1676. [PMID: 30782609 DOI: 10.1182/blood-2018-09-872549] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/10/2019] [Indexed: 02/07/2023] Open
Abstract
Peripheral T-cell lymphoma (PTCL) is a group of complex clinicopathological entities, often associated with an aggressive clinical course. Angioimmunoblastic T-cell lymphoma (AITL) and PTCL-not otherwise specified (PTCL-NOS) are the 2 most frequent categories, accounting for >50% of PTCLs. Gene expression profiling (GEP) defined molecular signatures for AITL and delineated biological and prognostic subgroups within PTCL-NOS (PTCL-GATA3 and PTCL-TBX21). Genomic copy number (CN) analysis and targeted sequencing of these molecular subgroups revealed unique CN abnormalities (CNAs) and oncogenic pathways, indicating distinct oncogenic evolution. PTCL-GATA3 exhibited greater genomic complexity that was characterized by frequent loss or mutation of tumor suppressor genes targeting the CDKN2A /B-TP53 axis and PTEN-PI3K pathways. Co-occurring gains/amplifications of STAT3 and MYC occurred in PTCL-GATA3. Several CNAs, in particular loss of CDKN2A, exhibited prognostic significance in PTCL-NOS as a single entity and in the PTCL-GATA3 subgroup. The PTCL-TBX21 subgroup had fewer CNAs, primarily targeting cytotoxic effector genes, and was enriched in mutations of genes regulating DNA methylation. CNAs affecting metabolic processes regulating RNA/protein degradation and T-cell receptor signaling were common in both subgroups. AITL showed lower genomic complexity compared with other PTCL entities, with frequent co-occurring gains of chromosome 5 (chr5) and chr21 that were significantly associated with IDH2 R172 mutation. CN losses were enriched in genes regulating PI3K-AKT-mTOR signaling in cases without IDH2 mutation. Overall, we demonstrated that novel GEP-defined PTCL subgroups likely evolve by distinct genetic pathways and provided biological rationale for therapies that may be investigated in future clinical trials.
Collapse
|
114
|
Fu JM, Leslie EJ, Scott AF, Murray JC, Marazita ML, Beaty TH, Scharpf RB, Ruczinski I. Detection of de novo copy number deletions from targeted sequencing of trios. Bioinformatics 2019; 35:571-578. [PMID: 30084993 PMCID: PMC6378941 DOI: 10.1093/bioinformatics/bty677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 11/25/2022] Open
Abstract
MOTIVATION De novo copy number deletions have been implicated in many diseases, but there is no formal method to date that identifies de novo deletions in parent-offspring trios from capture-based sequencing platforms. RESULTS We developed Minimum Distance for Targeted Sequencing (MDTS) to fill this void. MDTS has similar sensitivity (recall), but a much lower false positive rate compared to less specific CNV callers, resulting in a much higher positive predictive value (precision). MDTS also exhibited much better scalability. AVAILABILITY AND IMPLEMENTATION MDTS is freely available as open source software from the Bioconductor repository. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jack M Fu
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Alan F Scott
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeffrey C Murray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mary L Marazita
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Robert B Scharpf
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
115
|
Analysis of point mutations and copy number variation in Grade II and III meningioma. Exp Mol Pathol 2018; 105:328-333. [DOI: 10.1016/j.yexmp.2018.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/05/2018] [Accepted: 10/10/2018] [Indexed: 01/23/2023]
|
116
|
Integrative epigenetic taxonomy of primary prostate cancer. Nat Commun 2018; 9:4900. [PMID: 30464211 PMCID: PMC6249266 DOI: 10.1038/s41467-018-07270-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/23/2018] [Indexed: 12/31/2022] Open
Abstract
The Androgen Receptor (AR) is the key-driving transcription factor in prostate cancer, tightly controlled by epigenetic regulation. To date, most epigenetic profiling has been performed in cell lines or limited tissue samples. Here, to comprehensively study the epigenetic landscape, we perform RNA-seq with ChIP-seq for AR and histone modification marks (H3K27ac, H3K4me3, H3K27me3) in 100 primary prostate carcinomas. Integrative molecular subtyping of the five data streams revealed three major subtypes of which two were clearly TMPRSS2-ERG dictated. Importantly, we identify a third subtype with low chromatin binding and activity of AR, but with high activity of FGF and WNT signaling. While positive for neuroendocrine-hallmark genes, these tumors were copy number-neutral with low mutational burden, significantly depleted for genes characteristic of poor-outcome associated luminal B-subtype. We present a unique resource on transcriptional and epigenetic control in prostate cancer, revealing tight control of gene regulation differentially dictated by AR over three subtypes. The Androgen Receptor (AR) is the main driver of prostate cancer and functions in conjunction with chromatin modifications. Here, the authors comprehensively profile 100 primary prostate carcinomas by sequencing RNA transcripts in combination with ChIP-sequencing for AR and the active histone marks H3K27ac, H3K4me3 and repressive mark H3K27me3.
Collapse
|
117
|
Li Z, Chen G, Cai Z, Dong X, Qiu L, Xu H, Zeng Y, Liu X, Liu J. Genomic and transcriptional Profiling of tumor infiltrated CD8 + T cells revealed functional heterogeneity of antitumor immunity in hepatocellular carcinoma. Oncoimmunology 2018; 8:e1538436. [PMID: 30713796 DOI: 10.1080/2162402x.2018.1538436] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023] Open
Abstract
As key players in HCC antitumor response, the functions of tumor infiltrated CD8+ T cells are significantly affected by surrounding microenvironment. A detailed profiling of their genomic and transcriptional changes could provide valuable insights for both future immunotherapy development and prognosis evaluation. We performed whole exome and transcriptome sequencing on tumor infiltrated CD8+ T cells and CD8+ T cells isolated from other tissue origins (peritumor tissues and corresponding PBMCs) in eight treatment-naive HCC patients. The results demonstrated that transcriptional changes, rather than genomic alterations were the main contributors to the functional alterations of CD8+ T cells in the process of tumor progression. The origins of CD8+ T cells defined their transcriptional landscape, while the tumor infiltrated CD8+ T cells shared more similarity with peritumor-derived CD8+ T cells compared with those CD8+ T cells in blood. In addition, tumor infiltrated CD8+ T cells also showed larger transcriptional heterogeneity among individuals, which was modulated by clinical features such as HBV levels, preoperative anti-viral treatment and the degree of T cell infiltration. We also identified multiple inter-connected pathways involved in the activation and exhaustion of tumor infiltrated CD8+ T cells, among which IL-12 mediated pathway could dynamically reflect the functional status of CD8+ TILs and activation of this pathway indicated a better prognosis. Our results presented an overview picture of CD8+ TILs' genomic and transcriptional landscape and features, as well as how the functional status of CD8+ TILs correlated with patients' clinical course.
Collapse
Affiliation(s)
- Zhenli Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Liman Qiu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| |
Collapse
|
118
|
Clinical Targeted Next-Generation Sequencing Shows Increased Mutational Load in Endometrioid-type Endometrial Adenocarcinoma With Deficient DNA Mismatch Repair. Int J Gynecol Pathol 2018; 37:581-589. [DOI: 10.1097/pgp.0000000000000459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
119
|
Kang Y, Nam SH, Park KS, Kim Y, Kim JW, Lee E, Ko JM, Lee KA, Park I. DeviCNV: detection and visualization of exon-level copy number variants in targeted next-generation sequencing data. BMC Bioinformatics 2018; 19:381. [PMID: 30326846 PMCID: PMC6192323 DOI: 10.1186/s12859-018-2409-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/04/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Targeted next-generation sequencing (NGS) is increasingly being adopted in clinical laboratories for genomic diagnostic tests. RESULTS We developed a new computational method, DeviCNV, intended for the detection of exon-level copy number variants (CNVs) in targeted NGS data. DeviCNV builds linear regression models with bootstrapping for every probe to capture the relationship between read depth of an individual probe and the median of read depth values of all probes in the sample. From the regression models, it estimates the read depth ratio of the observed and predicted read depth with confidence interval for each probe which is applied to a circular binary segmentation (CBS) algorithm to obtain CNV candidates. Then, it assigns confidence scores to those candidates based on the reliability and strength of the CNV signals inferred from the read depth ratios of the probes within them. Finally, it also provides gene-centric plots with confidence levels of CNV candidates for visual inspection. We applied DeviCNV to targeted NGS data generated for newborn screening and demonstrated its ability to detect novel pathogenic CNVs from clinical samples. CONCLUSIONS We propose a new pragmatic method for detecting CNVs in targeted NGS data with an intuitive visualization and a systematic method to assign confidence scores for candidate CNVs. Since DeviCNV was developed for use in clinical diagnosis, sensitivity is increased by the detection of exon-level CNVs.
Collapse
Affiliation(s)
- Yeeok Kang
- SD Genomics Co., Ltd., 11F, Seoul Gangnam Post Office, 619 Gaepo-ro, Gangnam-gu, Seoul, 06336, Republic of Korea.,Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Seong-Hyeuk Nam
- SD Genomics Co., Ltd., 11F, Seoul Gangnam Post Office, 619 Gaepo-ro, Gangnam-gu, Seoul, 06336, Republic of Korea
| | - Kyung Sun Park
- SD Genomics Co., Ltd., 11F, Seoul Gangnam Post Office, 619 Gaepo-ro, Gangnam-gu, Seoul, 06336, Republic of Korea
| | - Yoonjung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Jong-Won Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eunjung Lee
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, USA
| | - Jung Min Ko
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-A Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| | - Inho Park
- SD Genomics Co., Ltd., 11F, Seoul Gangnam Post Office, 619 Gaepo-ro, Gangnam-gu, Seoul, 06336, Republic of Korea.
| |
Collapse
|
120
|
The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med 2018; 24:1611-1624. [PMID: 30150718 DOI: 10.1038/s41591-018-0156-x] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Glioblastoma is characterized by widespread genetic and transcriptional heterogeneity, yet little is known about the role of the epigenome in glioblastoma disease progression. Here, we present genome-scale maps of DNA methylation in matched primary and recurring glioblastoma tumors, using data from a highly annotated clinical cohort that was selected through a national patient registry. We demonstrate the feasibility of DNA methylation mapping in a large set of routinely collected FFPE samples, and we validate bisulfite sequencing as a multipurpose assay that allowed us to infer a range of different genetic, epigenetic, and transcriptional characteristics of the profiled tumor samples. On the basis of these data, we identified subtle differences between primary and recurring tumors, links between DNA methylation and the tumor microenvironment, and an association of epigenetic tumor heterogeneity with patient survival. In summary, this study establishes an open resource for dissecting DNA methylation heterogeneity in a genetically diverse and heterogeneous cancer, and it demonstrates the feasibility of integrating epigenomics, radiology, and digital pathology for a national cohort, thereby leveraging existing samples and data collected as part of routine clinical practice.
Collapse
|
121
|
Human glioblastoma arises from subventricular zone cells with low-level driver mutations. Nature 2018; 560:243-247. [PMID: 30069053 DOI: 10.1038/s41586-018-0389-3] [Citation(s) in RCA: 404] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/11/2018] [Indexed: 12/17/2022]
Abstract
Glioblastoma (GBM) is a devastating and incurable brain tumour, with a median overall survival of fifteen months1,2. Identifying the cell of origin that harbours mutations that drive GBM could provide a fundamental basis for understanding disease progression and developing new treatments. Given that the accumulation of somatic mutations has been implicated in gliomagenesis, studies have suggested that neural stem cells (NSCs), with their self-renewal and proliferative capacities, in the subventricular zone (SVZ) of the adult human brain may be the cells from which GBM originates3-5. However, there is a lack of direct genetic evidence from human patients with GBM4,6-10. Here we describe direct molecular genetic evidence from patient brain tissue and genome-edited mouse models that show astrocyte-like NSCs in the SVZ to be the cell of origin that contains the driver mutations of human GBM. First, we performed deep sequencing of triple-matched tissues, consisting of (i) normal SVZ tissue away from the tumour mass, (ii) tumour tissue, and (iii) normal cortical tissue (or blood), from 28 patients with isocitrate dehydrogenase (IDH) wild-type GBM or other types of brain tumour. We found that normal SVZ tissue away from the tumour in 56.3% of patients with wild-type IDH GBM contained low-level GBM driver mutations (down to approximately 1% of the mutational burden) that were observed at high levels in their matching tumours. Moreover, by single-cell sequencing and laser microdissection analysis of patient brain tissue and genome editing of a mouse model, we found that astrocyte-like NSCs that carry driver mutations migrate from the SVZ and lead to the development of high-grade malignant gliomas in distant brain regions. Together, our results show that NSCs in human SVZ tissue are the cells of origin that contain the driver mutations of GBM.
Collapse
|
122
|
Cioni B, Nevedomskaya E, Melis MHM, van Burgsteden J, Stelloo S, Hodel E, Spinozzi D, de Jong J, van der Poel H, de Boer JP, Wessels LFA, Zwart W, Bergman AM. Loss of androgen receptor signaling in prostate cancer-associated fibroblasts (CAFs) promotes CCL2- and CXCL8-mediated cancer cell migration. Mol Oncol 2018; 12:1308-1323. [PMID: 29808619 PMCID: PMC6068356 DOI: 10.1002/1878-0261.12327] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblasts are abundantly present in the prostate tumor microenvironment (TME), including cancer‐associated fibroblasts (CAFs) which play a key role in cancer development. Androgen receptor (AR) signaling is the main driver of prostate cancer (PCa) progression, and stromal cells in the TME also express AR. High‐grade tumor and poor clinical outcome are associated with low AR expression in the TME, which suggests a protective role of AR signaling in the stroma against PCa development. However, the mechanism of this relation is not clear. In this study, we isolated AR‐expressing CAF‐like cells. Testosterone (R1881) exposure did not affect CAF‐like cell morphology, proliferation, or motility. PCa cell growth was not affected by culturing in medium from R1881‐exposed CAF‐like cells; however, migration of PCa cells was inhibited. AR chromatin immune precipitation sequencing (ChIP‐seq) was performed and motif search suggested that AR in CAF‐like cells bound the chromatin through AP‐1‐elements upon R1881 exposure, inducing enhancer‐mediated AR chromatin interactions. The vast majority of chromatin binding sites in CAF‐like cells were unique and not shared with AR sites observed in PCa cell lines or tumors. AR signaling in CAF‐like cells decreased expression of multiple cytokines; most notably CCL2 and CXCL8 and both cytokines increased migration of PCa cells. These results suggest direct paracrine regulation of PCa cell migration by CAFs through AR signaling.
Collapse
Affiliation(s)
- Bianca Cioni
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Ekaterina Nevedomskaya
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Division of Molecular Carcinogenesis, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Oncode Institute, The Netherlands
| | - Monique H M Melis
- Division of Molecular Genetics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Johan van Burgsteden
- Division of Molecular Genetics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Suzan Stelloo
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Faculty of EEMCS, Delft University of Technology, Delft, The Netherlands
| | - Emma Hodel
- Division of Molecular Genetics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Daniele Spinozzi
- Division of Molecular Genetics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Jeroen de Jong
- Division of Pathology, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Henk van der Poel
- Division of Urology, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Jan Paul de Boer
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Division of Medical Oncology, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Oncode Institute, The Netherlands.,Faculty of EEMCS, Delft University of Technology, Delft, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Oncode Institute, The Netherlands
| | - Andries M Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.,Division of Medical Oncology, The Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| |
Collapse
|
123
|
van Dijk E, Biesma HD, Cordes M, Smeets D, Neerincx M, Das S, Eijk PP, Murphy V, Barat A, Bacon O, Prehn JHM, Betge J, Gaiser T, Fender B, Meijer GA, McNamara DA, Klinger R, Koopman M, Ebert MPA, Kay EW, Hennessey BT, Verheul HMW, Gallagher WM, O'Connor DP, Punt CJA, Loupakis F, Lambrechts D, Byrne AT, van Grieken NCT, Ylstra B. Loss of Chromosome 18q11.2-q12.1 Is Predictive for Survival in Patients With Metastatic Colorectal Cancer Treated With Bevacizumab. J Clin Oncol 2018; 36:2052-2060. [PMID: 29792754 DOI: 10.1200/jco.2017.77.1782] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Patients with metastatic colorectal cancer (mCRC) have limited benefit from the addition of bevacizumab to standard chemotherapy. However, a subset probably benefits substantially, highlighting an unmet clinical need for a biomarker of response to bevacizumab. Previously, we demonstrated that losses of chromosomes 5q34, 17q12, and 18q11.2-q12.1 had a significant correlation with progression-free survival (PFS) in patients with mCRC treated with bevacizumab in the CAIRO2 clinical trial but not in patients who did not receive bevacizumab in the CAIRO trial. This study was designed to validate these findings. Materials and Methods Primary mCRC samples were analyzed from two cohorts of patients who received bevacizumab as first-line treatment; 96 samples from the European multicenter study Angiopredict (APD) and 81 samples from the Italian multicenter study, MOMA. A third cohort of 90 samples from patients with mCRC who did not receive bevacizumab was analyzed. Copy number aberrations of tumor biopsy specimens were measured by shallow whole-genome sequencing and were correlated with PFS, overall survival (OS), and response. Results Loss of chromosome 18q11.2-q12.1 was associated with prolonged PFS most significantly in both the cohorts that received bevacizumab (APD: hazard ratio, 0.54; P = .01; PFS difference, 65 days; MOMA: hazard ratio, 0.55; P = .019; PFS difference, 49 days). A similar association was found for OS and overall response rate in these two cohorts, which became significant when combined with the CAIRO2 cohort. Median PFS in the cohort of patients with mCRC who did not receive bevacizumab and in the CAIRO cohort was similar to that of the APD, MOMA, and CAIRO2 patients without an 18q11.2-q12.1 loss. Conclusion We conclude that the loss of chromosome 18q11.2-q12.1 is consistently predictive for prolonged PFS in patients receiving bevacizumab. The predictive value of this loss is substantiated by a significant gain in OS and overall response rate.
Collapse
Affiliation(s)
- Erik van Dijk
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Hedde D Biesma
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Martijn Cordes
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Dominiek Smeets
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Maarten Neerincx
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Sudipto Das
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Paul P Eijk
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Verena Murphy
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Anna Barat
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Orna Bacon
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Jochen H M Prehn
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Johannes Betge
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Timo Gaiser
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bozena Fender
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Gerrit A Meijer
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Deborah A McNamara
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Rut Klinger
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Miriam Koopman
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Matthias P A Ebert
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Elaine W Kay
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bryan T Hennessey
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Henk M W Verheul
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - William M Gallagher
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Darran P O'Connor
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Cornelis J A Punt
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Fotios Loupakis
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Diether Lambrechts
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Annette T Byrne
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Nicole C T van Grieken
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bauke Ylstra
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| |
Collapse
|
124
|
Assessment of concordance between fresh-frozen and formalin-fixed paraffin embedded tumor DNA methylation using a targeted sequencing approach. Oncotarget 2018; 8:48126-48137. [PMID: 28611295 PMCID: PMC5564631 DOI: 10.18632/oncotarget.18296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 04/03/2017] [Indexed: 12/28/2022] Open
Abstract
DNA methylation is altered in many types of disease, including metastatic colorectal cancer. However, the methylome has not yet been fully described in archival formalin-fixed paraffin embedded (FFPE) samples in the context of matched fresh-frozen (FF) tumor material at base-pair resolution using a targeted approach. Using next-generation sequencing, we investigated three pairs of matched FFPE and FF samples to determine the extent of their similarity. We identified a ‘bowing’ pattern specific to FFPE samples categorized by a lower CG proportion at the start of sequence reads. We have found no evidence that this affected methylation calling, nor concordance of results. We also found no significant increase in deamination, measured by C>T transitions, previously considered a result of crosslinking DNA by formalin fixation and a barrier to the use of FFPE in methylation studies. The methods used in this study have shown sensitivity of between 60-70% based on positions also methylated in colorectal cancer cell lines. We demonstrate that FFPE material is a useful source of tumor material for methylation studies using targeted sequencing.
Collapse
|
125
|
Semeraro R, Orlandini V, Magi A. Xome-Blender: A novel cancer genome simulator. PLoS One 2018; 13:e0194472. [PMID: 29621252 PMCID: PMC5886411 DOI: 10.1371/journal.pone.0194472] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 02/05/2018] [Indexed: 11/18/2022] Open
Abstract
The adoption of next generation sequencing based methods in cancer research allowed for the investigation of the complex genetic structure of tumor samples. In the last few years, considerable importance was given to the research of somatic variants and several computational approaches were developed for this purpose. Despite continuous improvements to these programs, the validation of their results it’s a hard challenge due to multiple sources of error. To overcome this drawback different simulation approaches are used to generate synthetic samples but they are often based on the addition of artificial mutations that mimic the complexity of genomic variations. For these reasons, we developed a novel software, Xome-Blender, that generates synthetic cancer genomes with user defined features such as the number of subclones, the number of somatic variants and the presence of copy number alterations (CNAs), without the addition of any synthetic element. The singularity of our method is the “morphological approach” used to generate mutation events. To demonstrate the power of our tool we used it to address the hard challenge of evaluating the performance of nine state-of-the-art somatic variant calling methods for small and large variants (VarScan2, MuTect, Shimmer, BCFtools, Strelka, EXCAVATOR2, Control-FREEC and CopywriteR). Through these analyses we observed that by using Xome-Blender data it is possible to appraise small differences between their performance and we have designated VarScan2 and EXCAVATOR2 as best tool for this kind of applications. Xome-Blender is unix-based, licensed under the GPLv3 and freely available at https://github.com/rsemeraro/XomeBlender.
Collapse
Affiliation(s)
- Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- * E-mail:
| | - Valerio Orlandini
- Medical Genetics Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
126
|
Lee JEA, Li N, Rowley SM, Cheasley D, Zethoven M, McInerny S, Gorringe KL, James PA, Campbell IG. Molecular analysis of PALB2-associated breast cancers. J Pathol 2018; 245:53-60. [PMID: 29431189 DOI: 10.1002/path.5055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 12/15/2022]
Abstract
PALB2 is established as the most clinically important moderate to high penetrance breast cancer predisposition gene after BRCA1 and BRCA2. Mutations in classical familial cancer predisposition genes are presumed to be recessive at the cellular level and therefore a second inactivating somatic mutation is required in the tumour tissue. However, from the limited data that exist, PALB2 may be an example of a cancer predisposition gene that does not conform to Knudson's 'two hit' paradigm. We conducted genome-wide copy number analysis and targeted sequencing of PALB2 and other breast cancer driver genes in 15 invasive breast cancers from individuals carrying pathogenic germline mutations in PALB2. The majority of cancers showed clear evidence of bi-allelic inactivation of PALB2 (10/15) either as loss of heterozygosity involving the wild-type allele (six tumours) or as somatic point mutations (four tumours). All PALB2-null cancers had high homologous recombination deficiency (HRD) scores consistent with a homologous recombination repair deficiency. Interestingly, all but one of the PALB2 heterozygous cancers also had high HRD scores, suggesting that alternative mechanisms of PALB2 functional loss might be operating in these cancers. Our findings demonstrate that PALB2 does undergo bi-allelic inactivation in the majority of breast cancers from PALB2 germline mutation carriers. This feature has implications for the discovery of new moderate to high penetrance breast cancer predisposition genes as it supports using the existence of a 'second hit' and mutation signatures as important search criteria. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jue Er Amanda Lee
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Cancer Biology Medical Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Magnus Zethoven
- Bioinformatics Consulting Core, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Simone McInerny
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
127
|
Douville C, Springer S, Kinde I, Cohen JD, Hruban RH, Lennon AM, Papadopoulos N, Kinzler KW, Vogelstein B, Karchin R. Detection of aneuploidy in patients with cancer through amplification of long interspersed nucleotide elements (LINEs). Proc Natl Acad Sci U S A 2018; 115:1871-1876. [PMID: 29432176 PMCID: PMC5828610 DOI: 10.1073/pnas.1717846115] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aneuploidy is a feature of most cancer cells, and a myriad of approaches have been developed to detect it in clinical samples. We previously described primers that could be used to amplify ∼38,000 unique long interspersed nucleotide elements (LINEs) from throughout the genome. Here we have developed an approach to evaluate the sequencing data obtained from these amplicons. This approach, called Within-Sample AneupLoidy DetectiOn (WALDO), employs supervised machine learning to detect the small changes in multiple chromosome arms that are often present in cancers. We used WALDO to search for chromosome arm gains and losses in 1,677 tumors and in 1,522 liquid biopsies of blood from cancer patients or normal individuals. Aneuploidy was detected in 95% of cancer biopsies and in 22% of liquid biopsies. Using single-nucleotide polymorphisms within the amplified LINEs, WALDO concomitantly assesses allelic imbalances, microsatellite instability, and sample identification. WALDO can be used on samples containing only a few nanograms of DNA and as little as 1% neoplastic content and has a variety of applications in cancer diagnostics and forensic science.
Collapse
Affiliation(s)
- Christopher Douville
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21287
| | - Simeon Springer
- Ludwig Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Howard Hughes Medical Institute, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | | | - Joshua D Cohen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287
- Ludwig Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Howard Hughes Medical Institute, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Ralph H Hruban
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Anne Marie Lennon
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Department of Surgery, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Nickolas Papadopoulos
- Ludwig Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Kenneth W Kinzler
- Ludwig Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Bert Vogelstein
- Ludwig Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287;
- Howard Hughes Medical Institute, Johns Hopkins Medical Institutions, Baltimore, MD 21287
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| | - Rachel Karchin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287;
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21287
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD 21287
| |
Collapse
|
128
|
Isobe T, Seki M, Yoshida K, Sekiguchi M, Shiozawa Y, Shiraishi Y, Kimura S, Yoshida M, Inoue Y, Yokoyama A, Kakiuchi N, Suzuki H, Kataoka K, Sato Y, Kawai T, Chiba K, Tanaka H, Shimamura T, Kato M, Iguchi A, Hama A, Taguchi T, Akiyama M, Fujimura J, Inoue A, Ito T, Deguchi T, Kiyotani C, Iehara T, Hosoi H, Oka A, Sanada M, Tanaka Y, Hata K, Miyano S, Ogawa S, Takita J. Integrated Molecular Characterization of the Lethal Pediatric Cancer Pancreatoblastoma. Cancer Res 2018; 78:865-876. [PMID: 29233928 DOI: 10.1158/0008-5472.can-17-2581] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/24/2017] [Accepted: 12/07/2017] [Indexed: 11/16/2022]
Abstract
Pancreatoblastoma is a rare pediatric pancreatic malignancy for which the molecular pathogenesis is not understood. In this study, we report the findings of an integrated multiomics study of whole-exome and RNA sequencing as well as genome-wide copy number and methylation analyses of ten pancreatoblastoma cases. The pancreatoblastoma genome was characterized by a high frequency of aberrant activation of the Wnt signaling pathway, either via somatic mutations of CTNNB1 (90%) and copy-neutral loss of heterozygosity (CN-LOH) of APC (10%). In addition, imprinting dysregulation of IGF2 as a consequence of CN-LOH (80%), gain of paternal allele (10%), and gain of methylation (10%) was universally detected. At the transcriptome level, pancreatoblastoma exhibited an expression profile characteristic of early pancreas progenitor-like cells along with upregulation of the R-spondin/LGR5/RNF43 module. Our results offer a comprehensive description of the molecular basis for pancreatoblastoma and highlight rational therapeutic targets for its treatment.Significance: Molecular genetic analysis of a rare untreatable pediatric tumor reveals Wnt/IGF2 aberrations and features of early pancreas progenitor-like cells, suggesting cellular origins and rational strategies for therapeutic targeting. Cancer Res; 78(4); 865-76. ©2017 AACR.
Collapse
Affiliation(s)
- Tomoya Isobe
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masafumi Seki
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Sekiguchi
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Shiozawa
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichi Shiraishi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Kimura
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pediatrics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Misa Yoshida
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yoshikage Inoue
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Yokoyama
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromichi Suzuki
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keisuke Kataoka
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Sato
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenichi Chiba
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Tanaka
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Teppei Shimamura
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Motohiro Kato
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akihiro Iguchi
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Asahito Hama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Masaharu Akiyama
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | - Junya Fujimura
- Department of Pediatrics, Juntendo University School of Medicine, Tokyo, Japan
| | - Akiko Inoue
- Department of Pediatrics, Osaka Medical College, Osaka, Japan
| | - Tsuyoshi Ito
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Takao Deguchi
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Chikako Kiyotani
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tomoko Iehara
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Akira Oka
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Sanada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yukichi Tanaka
- Department of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
129
|
Characterizing steroid hormone receptor chromatin binding landscapes in male and female breast cancer. Nat Commun 2018; 9:482. [PMID: 29396493 PMCID: PMC5797120 DOI: 10.1038/s41467-018-02856-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/04/2018] [Indexed: 11/09/2022] Open
Abstract
Male breast cancer (MBC) is rare and poorly characterized. Like the female counterpart, most MBCs are hormonally driven, but relapse after hormonal treatment is also noted. The pan-hormonal action of steroid hormonal receptors, including estrogen receptor alpha (ERα), androgen receptor (AR), progesterone receptor (PR), and glucocorticoid receptor (GR) in this understudied tumor type remains wholly unexamined. This study reveals genomic cross-talk of steroid hormone receptor action and interplay in human tumors, here in the context of MBC, in relation to the female disease and patient outcome. Here we report the characterization of human breast tumors of both genders for cistromic make-up of hormonal regulation in human tumors, revealing genome-wide chromatin binding landscapes of ERα, AR, PR, GR, FOXA1, and GATA3 and enhancer-enriched histone mark H3K4me1. We integrate these data with transcriptomics to reveal gender-selective and genomic location-specific hormone receptor actions, which associate with survival in MBC patients.
Collapse
|
130
|
Severson TM, Nevedomskaya E, Peeters J, Kuilman T, Krijgsman O, van Rossum A, Droog M, Kim Y, Koornstra R, Beumer I, Glas AM, Peeper D, Wesseling J, Simon IM, Wessels L, Linn SC, Zwart W. Neoadjuvant tamoxifen synchronizes ERα binding and gene expression profiles related to outcome and proliferation. Oncotarget 2017; 7:33901-18. [PMID: 27129152 PMCID: PMC5085127 DOI: 10.18632/oncotarget.8983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/13/2016] [Indexed: 12/17/2022] Open
Abstract
Estrogen receptor alpha (ERα)-positive breast cancers are frequently treated with tamoxifen, but resistance is common. It remains elusive how tamoxifen resistance occurs and predictive biomarkers for treatment outcome are needed. Because most biomarker discovery studies are performed using pre-treatment surgical resections, the effects of tamoxifen therapy directly on the tumor cell in vivo remain unexamined. In this study, we assessed DNA copy number, gene expression profiles and ERα/chromatin binding landscapes on breast tumor specimens, both before and after neoadjuvant tamoxifen treatment. We observed neoadjuvant tamoxifen treatment synchronized ERα/chromatin interactions and downstream gene expression, indicating that hormonal therapy reduces inter-tumor molecular variability. ERα-synchronized sites are associated with dynamic FOXA1 action at these sites, which is under control of growth factor signaling. Genes associated with tamoxifen-synchronized sites are capable of differentiating patients for tamoxifen benefit. Due to the direct effects of therapeutics on ERα behavior and transcriptional output, our study highlights the added value of biomarker discovery studies after neoadjuvant drug exposure.
Collapse
Affiliation(s)
- Tesa M Severson
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Ekaterina Nevedomskaya
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | | | - Thomas Kuilman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Oscar Krijgsman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Annelot van Rossum
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Marjolein Droog
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Yongsoo Kim
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Rutger Koornstra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, GA, The Netherlands
| | | | | | - Daniel Peeper
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | | | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Department of Pathology, University Medical Center Utrecht, CX, The Netherlands
| | - Wilbert Zwart
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| |
Collapse
|
131
|
BRCA-deficient mouse mammary tumor organoids to study cancer-drug resistance. Nat Methods 2017; 15:134-140. [PMID: 29256493 DOI: 10.1038/nmeth.4535] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023]
Abstract
Poly(ADP-ribose) polymerase inhibition (PARPi) is a promising new therapeutic approach for the treatment of cancers that show homologous recombination deficiency (HRD). Despite the success of PARPi in targeting HRD in tumors that lack the tumor suppressor function of BRCA1 or BRCA2, drug resistance poses a major obstacle. We developed three-dimensional cancer organoids derived from genetically engineered mouse models (GEMMs) for BRCA1- and BRCA2-deficient cancers. Unlike conventional cell lines or mammospheres, organoid cultures can be efficiently derived and rapidly expanded in vitro. Orthotopically transplanted organoids give rise to mammary tumors that recapitulate the epithelial morphology and preserve the drug response of the original tumor. Notably, GEMM-tumor-derived organoids can be easily genetically modified, making them a powerful tool for genetic studies of tumor biology and drug resistance.
Collapse
|
132
|
Rim JH, Lee ST, Gee HY, Lee BJ, Choi JR, Park HW, Han SH, Han J. Accuracy of Next-Generation Sequencing for Molecular Diagnosis in Patients With Infantile Nystagmus Syndrome. JAMA Ophthalmol 2017; 135:1376-1385. [PMID: 29145603 PMCID: PMC6583550 DOI: 10.1001/jamaophthalmol.2017.4859] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/23/2017] [Indexed: 01/10/2023]
Abstract
Importance Infantile nystagmus syndrome (INS) is a group of disorders presenting with genetic and clinical heterogeneities that have challenged the genetic and clinical diagnoses of INS. Precise molecular diagnosis in early infancy may result in more accurate genetic counseling and improved patient management. Objective To assess the accuracy of genomic data from next-generation sequencing (NGS) and phenotypic data to enhance the definitive diagnosis of INS. Design, Setting, and Participants A single-center retrospective case series was conducted in 48 unrelated, consecutive patients with INS, with or without associated ocular or systemic conditions, who underwent genetic testing between June 1, 2015, and January 31, 2017. Next-generation sequencing analysis was performed using a target panel that included 113 genes associated with INS (n = 47) or a TruSight One sequencing panel that included 4813 genes associated with known human phenotypes (n = 1). Variants were filtered and prioritized by in-depth clinical review, and finally classified according to the American College of Medical Genetics and Genomics guidelines. Patients underwent a detailed ophthalmic examination, including electroretinography and optical coherence tomography, if feasible. Main Outcomes and Measures Diagnostic yield of targeted NGS testing. Results Among the 48 patients (21 female and 27 male; mean [SD] age at genetic testing, 9.2 [10.3] years), 8 had a family history of nystagmus and 40 were simplex. All patients were of a single ethnicity (Korean). Genetic variants that were highly likely to be causative were identified in 28 of the 48 patients, corresponding to a molecular diagnostic yield of 58.3% (95% CI, 44.4%-72.2%). FRMD7, GPR143, and PAX6 mutations appeared to be the major genetic causes of familial INS. A total of 10 patients (21%) were reclassified to a different diagnosis based on results of NGS testing, enabling accurate clinical management. Conclusions and Relevance These findings suggest that NGS is an accurate diagnostic tool to differentiate causes of INS because diagnostic tests, such as electroretinography and optical coherence tomography, are not easily applicable in young infants. Accurate application of NGS using a standardized, stepwise, team-based approach in early childhood not only facilitated early molecular diagnosis but also led to improved personalized management in patients with INS.
Collapse
Affiliation(s)
- John Hoon Rim
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Medicine, Yonsei University Graduate School of Medicine, Seoul, Korea
- Department of Pharmacology, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Heon Yung Gee
- Department of Medicine, Yonsei University Graduate School of Medicine, Seoul, Korea
- Department of Pharmacology, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Byung Joo Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Won Park
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Sueng-Han Han
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jinu Han
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
- Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
133
|
Villela D, Costa SS, Vianna-Morgante AM, Krepischi AC, Rosenberg C. Efficient detection of chromosome imbalances and single nucleotide variants using targeted sequencing in the clinical setting. Eur J Med Genet 2017; 60:667-674. [DOI: 10.1016/j.ejmg.2017.08.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 11/29/2022]
|
134
|
Carter JH, Cottrell CE, McNulty SN, Vigh-Conrad KA, Lamp S, Heusel JW, Duncavage EJ. FGFR2 amplification in colorectal adenocarcinoma. Cold Spring Harb Mol Case Stud 2017; 3:mcs.a001495. [PMID: 28835367 PMCID: PMC5701301 DOI: 10.1101/mcs.a001495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 05/30/2017] [Indexed: 01/13/2023] Open
Abstract
FGFR2 is recurrently amplified in 5% of gastric cancers and 1%–4% of breast cancers; however, this molecular alteration has never been reported in a primary colorectal cancer specimen. Preclinical studies indicate that several FGFR tyrosine-kinase inhibitors (TKIs), such as AZD4547, have in vitro activity against the FGFR2-amplified colorectal cell line, NCI-H716. The efficacy of these inhibitors is currently under investigation in clinical trials for breast and gastric cancer. Thus, better characterizing colorectal tumors for FGFR2 amplification could identify a subset of patients who may benefit from FGFR TKI therapies. Here, we describe a novel FGFR2 amplification identified by clinical next-generation sequencing in a primary colorectal cancer. Further characterization of the tumor by immunohistochemistry showed neuroendocrine differentiation, similar to the reported properties of the NCI-H716 cell line. These findings demonstrate that the spectrum of potentially clinically actionable mutations detected by targeted clinical sequencing panels is not limited to only single-nucleotide polymorphisms and insertions/deletions but also to copy-number alterations.
Collapse
Affiliation(s)
- Jamal H Carter
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Catherine E Cottrell
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri 63130, USA.,Department of Genetics, Washington University in St. Louis, Missouri 63130, USA
| | - Samantha N McNulty
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri 63130, USA
| | | | - Stephen Lamp
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri 63130, USA
| | - Jonathan W Heusel
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri 63130, USA.,Department of Genetics, Washington University in St. Louis, Missouri 63130, USA
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri 63130, USA
| |
Collapse
|
135
|
Jiménez-Sánchez A, Memon D, Pourpe S, Veeraraghavan H, Li Y, Vargas HA, Gill MB, Park KJ, Zivanovic O, Konner J, Ricca J, Zamarin D, Walther T, Aghajanian C, Wolchok JD, Sala E, Merghoub T, Snyder A, Miller ML. Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient. Cell 2017; 170:927-938.e20. [PMID: 28841418 PMCID: PMC5589211 DOI: 10.1016/j.cell.2017.07.025] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/06/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022]
Abstract
We present an exceptional case of a patient with high-grade serous ovarian cancer, treated with multiple chemotherapy regimens, who exhibited regression of some metastatic lesions with concomitant progression of other lesions during a treatment-free period. Using immunogenomic approaches, we found that progressing metastases were characterized by immune cell exclusion, whereas regressing and stable metastases were infiltrated by CD8+ and CD4+ T cells and exhibited oligoclonal expansion of specific T cell subsets. We also detected CD8+ T cell reactivity against predicted neoepitopes after isolation of cells from a blood sample taken almost 3 years after the tumors were resected. These findings suggest that multiple distinct tumor immune microenvironments co-exist within a single individual and may explain in part the heterogeneous fates of metastatic lesions often observed in the clinic post-therapy. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Alejandro Jiménez-Sánchez
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Danish Memon
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Stephane Pourpe
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Yanyun Li
- Ludwig Collaborative/Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Michael B Gill
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Kay J Park
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Oliver Zivanovic
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jason Konner
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jacob Ricca
- Ludwig Collaborative/Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Dmitriy Zamarin
- Ludwig Collaborative/Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Tyler Walther
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jedd D Wolchok
- Ludwig Collaborative/Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Immunology and Microbial Pathogenesis Programs, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Taha Merghoub
- Ludwig Collaborative/Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Martin L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| |
Collapse
|
136
|
Wein L, Savas P, Van Geelen C, Caramia F, Moodie K, Joshi S, Loi S. FGFR2 amplification in metastatic hormone-positive breast cancer and response to an mTOR inhibitor. Ann Oncol 2017; 28:2025-2027. [DOI: 10.1093/annonc/mdx194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
137
|
Breast ductal carcinoma in situ carry mutational driver events representative of invasive breast cancer. Mod Pathol 2017; 30:952-963. [PMID: 28338653 DOI: 10.1038/modpathol.2017.21] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/11/2017] [Accepted: 02/19/2017] [Indexed: 12/31/2022]
Abstract
The spectrum of genomic alterations in ductal carcinoma in situ (DCIS) is relatively unexplored, but is likely to provide useful insights into its biology, its progression to invasive carcinoma and the risk of recurrence. DCIS (n=20) with a range of phenotypes was assessed by massively parallel sequencing for mutations and copy number alterations and variants validated by Sanger sequencing. PIK3CA mutations were identified in 11/20 (55%), TP53 mutations in 6/20 (30%), and GATA3 mutations in 9/20 (45%). Screening an additional 91 cases for GATA3 mutations identified a final frequency of 27% (30/111), with a high proportion of missense variants (8/30). TP53 mutations were exclusive to high grade DCIS and more frequent in PR-negative tumors compared with PR-positive tumors (P=0.037). TP53 mutant tumors also had a significantly higher fraction of the genome altered by copy number than wild-type tumors (P=0.005), including a significant positive association with amplification or gain of ERBB2 (P<0.05). The association between TP53 mutation and ERBB2 amplification was confirmed in a wider DCIS cohort using p53 immunohistochemistry as a surrogate marker for TP53 mutations (P=0.03). RUNX1 mutations and MAP2K4 copy number loss were novel findings in DCIS. Frequent copy number alterations included gains on 1q, 8q, 17q, and 20q and losses on 8p, 11q, 16q, and 17p. Patterns of genomic alterations observed in DCIS were similar to those previously reported for invasive breast cancers, with all DCIS having at least one bona fide breast cancer driver event. However, an increase in GATA3 mutations and fewer copy number changes were noted in DCIS compared with invasive carcinomas. The role of such alterations as prognostic and predictive biomarkers in DCIS is an avenue for further investigation.
Collapse
|
138
|
The AURORA pilot study for molecular screening of patients with advanced breast cancer-a study of the breast international group. NPJ Breast Cancer 2017; 3:23. [PMID: 28685159 PMCID: PMC5491498 DOI: 10.1038/s41523-017-0026-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/04/2017] [Accepted: 06/01/2017] [Indexed: 12/24/2022] Open
Abstract
Several studies have demonstrated the feasibility of molecular screening of tumour samples for matching patients with cancer to targeted therapies. However, most of them have been carried out at institutional or national level. Herein, we report on the pilot phase of AURORA (NCT02102165), a European multinational collaborative molecular screening initiative for advanced breast cancer patients. Forty-one patients were prospectively enroled at four participating centres across Europe. Metastatic tumours were biopsied and profiled using an Ion Torrent sequencing platform at a central facility. Sequencing results were obtained for 63% of the patients in real-time with variable turnaround time stemming from delays between patient consent and biopsy. At least one clinically actionable mutation was identified in 73% of patients. We used the Illumina sequencing technology for orthogonal validation and achieved an average of 66% concordance of substitution calls per patient. Additionally, copy number aberrations inferred from the Ion Torrent sequencing were compared to single nucleotide polymorphism arrays and found to be 59% concordant on average. Although this study demonstrates that powerful next generation genomic techniques are logistically ready for international molecular screening programs in routine clinical settings, technical challenges remain to be addressed in order to ensure the accuracy and clinical utility of the genomic data. A pilot study demonstrated that a large-scale, international screening programme for women with metastatic breast cancer is feasible. The study, coordinated by the Institut Jules Bordet and the Breast International Group, aimed to determine whether biopsies and blood could be collected from women with metastatic breast cancer across Europe and sent to a central laboratory for targeted gene sequencing. Genetic information was successfully obtained for 26 of the 41 participants, 19 of whom had mutations that could be targeted with a known drug, potentially influencing treatment decision-making. They concluded that genomic testing is logistically ready for international molecular screening in routine clinical settings laying the groundwork for the parent European AURORA molecular screening programme which aims at recruiting 1300 metastatic breast cancer patients. However, technical challenges remain to be addressed to ensure the accuracy and robustness across different sequencing platforms.
Collapse
|
139
|
Bernhardt N, Brassac J, Kilian B, Blattner FR. Dated tribe-wide whole chloroplast genome phylogeny indicates recurrent hybridizations within Triticeae. BMC Evol Biol 2017; 17:141. [PMID: 28622761 PMCID: PMC5474006 DOI: 10.1186/s12862-017-0989-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/03/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Triticeae, the tribe of wheat grasses, harbours the cereals barley, rye and wheat and their wild relatives. Although economically important, relationships within the tribe are still not understood. We analysed the phylogeny of chloroplast lineages among nearly all monogenomic Triticeae taxa and polyploid wheat species aiming at a deeper understanding of the tribe's evolution. We used on- and off-target reads of a target-enrichment experiment followed by Illumina sequencing. RESULTS The read data was used to assemble the plastid locus ndhF for 194 individuals and the whole chloroplast genome for 183 individuals, representing 53 Triticeae species and 15 genera. We conducted Bayesian and multispecies coalescent analyses to infer relationships and estimate divergence times of the taxa. We present the most comprehensive dated Triticeae chloroplast phylogeny and review previous hypotheses in the framework of our results. Monophyly of Triticeae chloroplasts could not be confirmed, as either Bromus or Psathyrostachys captured a chloroplast from a lineage closely related to a Bromus-Triticeae ancestor. The most recent common ancestor of Triticeae occurred approximately between ten and 19 million years ago. CONCLUSIONS The comparison of the chloroplast phylogeny with available nuclear data in several cases revealed incongruences indicating past hybridizations. Recent events of chloroplast capture were detected as individuals grouped apart from con-specific accessions in otherwise monopyhletic groups.
Collapse
Affiliation(s)
- Nadine Bernhardt
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany.
| | - Jonathan Brassac
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Benjamin Kilian
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
- Present address: Crop Trust, Bonn, Germany
| | - Frank R Blattner
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
140
|
De Sousa SMC, McCabe MJ, Wu K, Roscioli T, Gayevskiy V, Brook K, Rawlings L, Scott HS, Thompson TJ, Earls P, Gill AJ, Cowley MJ, Dinger ME, McCormack AI. Germline variants in familial pituitary tumour syndrome genes are common in young patients and families with additional endocrine tumours. Eur J Endocrinol 2017; 176:635-644. [PMID: 28220018 DOI: 10.1530/eje-16-0944] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/08/2017] [Accepted: 02/17/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Familial pituitary tumour syndromes (FPTS) account for 5% of pituitary adenomas. Multi-gene analysis via next-generation sequencing (NGS) may unveil greater prevalence and inform clinical care. We aimed to identify germline variants in selected patients with pituitary adenomas using a targeted NGS panel. DESIGN We undertook a nationwide cross-sectional study of patients with pituitary adenomas with onset ≤40 years of age and/or other personal/family history of endocrine neoplasia. A custom NGS panel was performed on germline DNA to interrogate eight FPTS genes. Genome data were analysed via a custom bioinformatic pipeline, and validation was performed by Sanger sequencing. Multiplex ligation-dependent probe amplification (MLPA) was performed in cases with heightened suspicion for MEN1, CDKN1B and AIP mutations. The main outcomes were frequency and pathogenicity of rare variants in AIP, CDKN1B, MEN1, PRKAR1A, SDHA, SDHB, SDHC and SDHD. RESULTS Forty-four patients with pituitary tumours, 14 of whom had a personal history of other endocrine tumours and/or a family history of pituitary or other endocrine tumours, were referred from endocrine tertiary-referral centres across Australia. Eleven patients (25%) had a rare variant across the eight FPTS genes tested: AIP (p.A299V, p.R106C, p.F269F, p.R304X, p.K156K, p.R271W), MEN1 (p.R176Q), SDHB (p.A2V, p.S8S), SDHC (p.E110Q) and SDHD (p.G12S), with two patients harbouring dual variants. Variants were classified as pathogenic or of uncertain significance in 9/44 patients (20%). No deletions/duplications were identified in MEN1, CDKN1B or AIP. CONCLUSIONS A high yield of rare variants in genes implicated in FPTS can be found in selected patients using an NGS panel. It may also identify individuals harbouring more than one rare variant.
Collapse
Affiliation(s)
- Sunita M C De Sousa
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- Endocrine and Metabolic UnitRoyal Adelaide Hospital, Adelaide, Australia
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
- School of MedicineUniversity of Adelaide, Adelaide, Australia
| | - Mark J McCabe
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Kathy Wu
- Familial Cancer ServiceWestmead Hospital, Westmead, Australia
- School of MedicineUniversity of Sydney, Sydney, Australia
| | - Tony Roscioli
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
- Department of Medical GeneticsSydney Children's Hospital, Sydney, Australia
| | - Velimir Gayevskiy
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
| | - Katelyn Brook
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
| | - Lesley Rawlings
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
| | - Hamish S Scott
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
- School of MedicineUniversity of Adelaide, Adelaide, Australia
- ACRF Cancer Genomics FacilityCentre for Cancer Biology, SA Pathology, Adelaide, Australia
- School of Biological SciencesUniversity of Adelaide, Adelaide, Australia
- School of Pharmacy and Medical SciencesUniversity of South Australia, Adelaide, Australia
| | - Tanya J Thompson
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
| | - Peter Earls
- School of Pharmacy and Medical SciencesUniversity of South Australia, Adelaide, Australia
| | - Anthony J Gill
- School of MedicineUniversity of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology GroupKolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
- Sydney Vital Translational Cancer Research CentreRoyal North Shore Hospital and University of Sydney, Sydney, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Ann I McCormack
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
- Department of EndocrinologySt Vincent's Hospital, Sydney, Australia
| |
Collapse
|
141
|
Silva GO, Siegel MB, Mose LE, Parker JS, Sun W, Perou CM, Chen M. SynthEx: a synthetic-normal-based DNA sequencing tool for copy number alteration detection and tumor heterogeneity profiling. Genome Biol 2017; 18:66. [PMID: 28390427 PMCID: PMC5385048 DOI: 10.1186/s13059-017-1193-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/16/2017] [Indexed: 01/22/2023] Open
Abstract
Changes in the quantity of genetic material, known as somatic copy number alterations (CNAs), can drive tumorigenesis. Many methods exist for assessing CNAs using microarrays, but considerable technical issues limit current CNA calling based upon DNA sequencing. We present SynthEx, a novel tool for detecting CNAs from whole exome and genome sequencing. SynthEx utilizes a “synthetic-normal” strategy to overcome technical and financial issues. In terms of accuracy and precision, SynthEx is highly comparable to array-based methods and outperforms sequencing-based CNA detection tools. SynthEx robustly identifies CNAs using sequencing data without the additional costs associated with matched normal specimens.
Collapse
Affiliation(s)
- Grace O Silva
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Marni B Siegel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lisle E Mose
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Joel S Parker
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wei Sun
- Public Health Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mengjie Chen
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, 900 East 57th Street, KCBD 3220A, Chicago, IL, 60637, USA.
| |
Collapse
|
142
|
Somatic Mutations in TSC1 and TSC2 Cause Focal Cortical Dysplasia. Am J Hum Genet 2017; 100:454-472. [PMID: 28215400 DOI: 10.1016/j.ajhg.2017.01.030] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/23/2017] [Indexed: 12/18/2022] Open
Abstract
Focal cortical dysplasia (FCD) is a major cause of the sporadic form of intractable focal epilepsies that require surgical treatment. It has recently been reported that brain somatic mutations in MTOR account for 15%-25% of FCD type II (FCDII), characterized by cortical dyslamination and dysmorphic neurons. However, the genetic etiologies of FCDII-affected individuals who lack the MTOR mutation remain unclear. Here, we performed deep hybrid capture and amplicon sequencing (read depth of 100×-20,012×) of five important mTOR pathway genes-PIK3CA, PIK3R2, AKT3, TSC1, and TSC2-by using paired brain and saliva samples from 40 FCDII individuals negative for MTOR mutations. We found that 5 of 40 individuals (12.5%) had brain somatic mutations in TSC1 (c.64C>T [p.Arg22Trp] and c.610C>T [p.Arg204Cys]) and TSC2 (c.4639G>A [p.Val1547Ile]), and these results were reproducible on two different sequencing platforms. All identified mutations induced hyperactivation of the mTOR pathway by disrupting the formation or function of the TSC1-TSC2 complex. Furthermore, in utero CRISPR-Cas9-mediated genome editing of Tsc1 or Tsc2 induced the development of spontaneous behavioral seizures, as well as cytomegalic neurons and cortical dyslamination. These results show that brain somatic mutations in TSC1 and TSC2 cause FCD and that in utero application of the CRISPR-Cas9 system is useful for generating neurodevelopmental disease models of somatic mutations in the brain.
Collapse
|
143
|
Carter JH, McNulty SN, Cimino PJ, Cottrell CE, Heusel JW, Vigh-Conrad KA, Duncavage EJ. Targeted Next-Generation Sequencing in Molecular Subtyping of Lower-Grade Diffuse Gliomas. J Mol Diagn 2017; 19:328-337. [DOI: 10.1016/j.jmoldx.2016.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/16/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022] Open
|
144
|
Estrogen receptor α wields treatment-specific enhancers between morphologically similar endometrial tumors. Proc Natl Acad Sci U S A 2017; 114:E1316-E1325. [PMID: 28167798 DOI: 10.1073/pnas.1615233114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The DNA-binding sites of estrogen receptor α (ERα) show great plasticity under the control of hormones and endocrine therapy. Tamoxifen is a widely applied therapy in breast cancer that affects ERα interactions with coregulators and shifts the DNA-binding signature of ERα upon prolonged exposure in breast cancer. Although tamoxifen inhibits the progression of breast cancer, it increases the risk of endometrial cancer in postmenopausal women. We therefore asked whether the DNA-binding signature of ERα differs between endometrial tumors that arise in the presence or absence of tamoxifen, indicating divergent enhancer activity for tumors that develop in different endocrine milieus. Using ChIP sequencing (ChIP-seq), we compared the ERα profiles of 10 endometrial tumors from tamoxifen users with those of six endometrial tumors from nonusers and integrated these results with the transcriptomic data of 47 endometrial tumors from tamoxifen users and 64 endometrial tumors from nonusers. The ERα-binding sites in tamoxifen-associated endometrial tumors differed from those in the tumors from nonusers and had distinct underlying DNA sequences and divergent enhancer activity as marked by histone 3 containing the acetylated lysine 27 (H3K27ac). Because tamoxifen acts as an agonist in the postmenopausal endometrium, similar to estrogen in the breast, we compared ERα sites in tamoxifen-associated endometrial cancers with publicly available ERα ChIP-seq data in breast tumors and found a striking resemblance in the ERα patterns of the two tissue types. Our study highlights the divergence between endometrial tumors that arise in different hormonal conditions and shows that ERα enhancer use in human cancer differs in the presence of nonphysiological endocrine stimuli.
Collapse
|
145
|
CNARA: reliability assessment for genomic copy number profiles. BMC Genomics 2016; 17:799. [PMID: 27733115 PMCID: PMC5062840 DOI: 10.1186/s12864-016-3074-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/07/2016] [Indexed: 01/22/2023] Open
Abstract
Background DNA copy number profiles from microarray and sequencing experiments sometimes contain wave artefacts which may be introduced during sample preparation and cannot be removed completely by existing preprocessing methods. Besides, large derivative log ratio spread (DLRS) of the probes correlating with poor DNA quality is sometimes observed in genome screening experiments and may lead to unreliable copy number profiles. Depending on the extent of these artefacts and the resulting misidentification of copy number alterations/variations (CNA/CNV), it may be desirable to exclude such samples from analyses or to adapt the downstream data analysis strategy accordingly. Results Here, we propose a method to distinguish reliable genomic copy number profiles from those containing heavy wave artefacts and/or large DLRS. We define four features that adequately summarize the copy number profiles for reliability assessment, and train a classifier on a dataset of 1522 copy number profiles from various microarray platforms. The method can be applied to predict the reliability of copy number profiles irrespective of the underlying microarray platform and may be adapted for those sequencing platforms from which copy number estimates could be computed as a piecewise constant signal. Further details can be found at https://github.com/baudisgroup/CNARA. Conclusions We have developed a method for the assessment of genomic copy number profiling data, and suggest to apply the method in addition to and after other state-of-the-art noise correction and quality control procedures. CNARA could be instrumental in improving the assessment of data used for genomic data mining experiments and support the reliable functional attribution of copy number aberrations especially in cancer research. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3074-7) contains supplementary material, which is available to authorized users.
Collapse
|
146
|
Bruna A, Rueda OM, Greenwood W, Batra AS, Callari M, Batra RN, Pogrebniak K, Sandoval J, Cassidy JW, Tufegdzic-Vidakovic A, Sammut SJ, Jones L, Provenzano E, Baird R, Eirew P, Hadfield J, Eldridge M, McLaren-Douglas A, Barthorpe A, Lightfoot H, O'Connor MJ, Gray J, Cortes J, Baselga J, Marangoni E, Welm AL, Aparicio S, Serra V, Garnett MJ, Caldas C. A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds. Cell 2016; 167:260-274.e22. [PMID: 27641504 PMCID: PMC5037319 DOI: 10.1016/j.cell.2016.08.041] [Citation(s) in RCA: 306] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/21/2016] [Accepted: 08/18/2016] [Indexed: 12/17/2022]
Abstract
The inter- and intra-tumor heterogeneity of breast cancer needs to be adequately captured in pre-clinical models. We have created a large collection of breast cancer patient-derived tumor xenografts (PDTXs), in which the morphological and molecular characteristics of the originating tumor are preserved through passaging in the mouse. An integrated platform combining in vivo maintenance of these PDTXs along with short-term cultures of PDTX-derived tumor cells (PDTCs) was optimized. Remarkably, the intra-tumor genomic clonal architecture present in the originating breast cancers was mostly preserved upon serial passaging in xenografts and in short-term cultured PDTCs. We assessed drug responses in PDTCs on a high-throughput platform and validated several ex vivo responses in vivo. The biobank represents a powerful resource for pre-clinical breast cancer pharmacogenomic studies (http://caldaslab.cruk.cam.ac.uk/bcape), including identification of biomarkers of response or resistance.
Collapse
Affiliation(s)
- Alejandra Bruna
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Oscar M Rueda
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Wendy Greenwood
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Ankita Sati Batra
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Maurizio Callari
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Rajbir Nath Batra
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Katherine Pogrebniak
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Jose Sandoval
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - John W Cassidy
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Ana Tufegdzic-Vidakovic
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Stephen-John Sammut
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Linda Jones
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK; Cambridge Breast Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre at Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 2QQ, UK
| | - Elena Provenzano
- Cambridge Breast Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre at Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 2QQ, UK
| | - Richard Baird
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK; Cambridge Breast Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre at Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 2QQ, UK
| | - Peter Eirew
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - James Hadfield
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Matthew Eldridge
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK
| | - Anne McLaren-Douglas
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Andrew Barthorpe
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Howard Lightfoot
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Mark J O'Connor
- DNA Damage Response Biology Area, Oncology IMED, AstraZeneca, Alderley Park, Macclesfield SK10 4TG, UK
| | - Joe Gray
- OHSU Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Javier Cortes
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Jose Baselga
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, NY 10065, USA
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | - Alana L Welm
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Violeta Serra
- Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Mathew J Garnett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Carlos Caldas
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, UK; Cambridge Breast Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre at Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 2QQ, UK.
| |
Collapse
|
147
|
Macintyre G, Ylstra B, Brenton JD. Sequencing Structural Variants in Cancer for Precision Therapeutics. Trends Genet 2016; 32:530-542. [PMID: 27478068 DOI: 10.1016/j.tig.2016.07.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022]
Abstract
The identification of mutations that guide therapy selection for patients with cancer is now routine in many clinical centres. The majority of assays used for solid tumour profiling use DNA sequencing to interrogate somatic point mutations because they are relatively easy to identify and interpret. Many cancers, however, including high-grade serous ovarian, oesophageal, and small-cell lung cancer, are driven by somatic structural variants that are not measured by these assays. Therefore, there is currently an unmet need for clinical assays that can cheaply and rapidly profile structural variants in solid tumours. In this review we survey the landscape of 'actionable' structural variants in cancer and identify promising detection strategies based on massively-parallel sequencing.
Collapse
Affiliation(s)
- Geoff Macintyre
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, UK.
| |
Collapse
|
148
|
Yu Z, Li A, Wang M. CloneCNA: detecting subclonal somatic copy number alterations in heterogeneous tumor samples from whole-exome sequencing data. BMC Bioinformatics 2016; 17:310. [PMID: 27538789 PMCID: PMC4990858 DOI: 10.1186/s12859-016-1174-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022] Open
Abstract
Background Copy number alteration is a main genetic structural variation that plays an important role in tumor initialization and progression. Accurate detection of copy number alterations is necessary for discovering cancer-causing genes. Whole-exome sequencing has become a widely used technology in the last decade for detecting various types of genomic aberrations in cancer genomes. However, there are several major issues encountered in these detection problems, including normal cell contamination, tumor aneuploidy, and intra-tumor heterogeneity. Especially, deciphering the intra-tumor heterogeneity is imperative for identifying clonal and subclonal copy number alterations. Results We introduce CloneCNA, a novel bioinformatics tool for efficiently addressing these issues and automatically detecting clonal and subclonal somatic copy number alterations from heterogeneous tumor samples. CloneCNA fully explores the log ratio of read counts between paired tumor-normal samples and tumor B allele frequency of germline heterozygous SNP positions, further employs efficient statistical models to quantitatively represent copy number status of tumor sample containing multiple clones. We examine CloneCNA on simulated heterogeneous and real tumor samples, and the results demonstrate that CloneCNA has higher power to detect copy number alterations than existing methods. Conclusions CloneCNA, a novel algorithm is developed to efficiently and accurately identify somatic copy number alterations from heterogeneous tumor samples. We demonstrate the statistical framework of CloneCNA represents a remarkable advance for tumor whole-exome sequencing data. We expect that CloneCNA will promote cancer-focused studies for investigating the role of clonal evolution and elucidating critical events benefiting tumor tumourigenesis and progression. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1174-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenhua Yu
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China. .,Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, AH230027, China.
| | - Minghui Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei, AH230027, China.,Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, AH230027, China
| |
Collapse
|
149
|
D'Aurizio R, Pippucci T, Tattini L, Giusti B, Pellegrini M, Magi A. Enhanced copy number variants detection from whole-exome sequencing data using EXCAVATOR2. Nucleic Acids Res 2016; 44:e154. [PMID: 27507884 PMCID: PMC5175347 DOI: 10.1093/nar/gkw695] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Copy Number Variants (CNVs) are structural rearrangements contributing to phenotypic variation that have been proved to be associated with many disease states. Over the last years, the identification of CNVs from whole-exome sequencing (WES) data has become a common practice for research and clinical purpose and, consequently, the demand for more and more efficient and accurate methods has increased. In this paper, we demonstrate that more than 30% of WES data map outside the targeted regions and that these reads, usually discarded, can be exploited to enhance the identification of CNVs from WES experiments. Here, we present EXCAVATOR2, the first read count based tool that exploits all the reads produced by WES experiments to detect CNVs with a genome-wide resolution. To evaluate the performance of our novel tool we use it for analysing two WES data sets, a population data set sequenced by the 1000 Genomes Project and a tumor data set made of bladder cancer samples. The results obtained from these analyses demonstrate that EXCAVATOR2 outperforms other four state-of-the-art methods and that our combined approach enlarge the spectrum of detectable CNVs from WES data with an unprecedented resolution. EXCAVATOR2 is freely available at http://sourceforge.net/projects/excavator2tool/.
Collapse
Affiliation(s)
- Romina D'Aurizio
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics and Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Tommaso Pippucci
- Medical Genetics Unit, Sant'Orsola Malpighi Polyclinic, Bologna, Italy
| | - Lorenzo Tattini
- Department of Computer Science, University of Pisa, Pisa, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence
| | - Marco Pellegrini
- Laboratory of Integrative Systems Medicine (LISM), Institute of Informatics and Telematics and Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence
| |
Collapse
|
150
|
Semenova EA, Kwon MC, Monkhorst K, Song JY, Bhaskaran R, Krijgsman O, Kuilman T, Peters D, Buikhuisen WA, Smit EF, Pritchard C, Cozijnsen M, van der Vliet J, Zevenhoven J, Lambooij JP, Proost N, van Montfort E, Velds A, Huijbers IJ, Berns A. Transcription Factor NFIB Is a Driver of Small Cell Lung Cancer Progression in Mice and Marks Metastatic Disease in Patients. Cell Rep 2016; 16:631-43. [PMID: 27373156 PMCID: PMC4956617 DOI: 10.1016/j.celrep.2016.06.020] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 12/01/2022] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor, and no effective treatment is available to date. Mouse models of SCLC based on the inactivation of Rb1 and Trp53 show frequent amplifications of the Nfib and Mycl genes. Here, we report that, although overexpression of either transcription factor accelerates tumor growth, NFIB specifically promotes metastatic spread. High NFIB levels are associated with expansive growth of a poorly differentiated and almost exclusively E-cadherin (CDH1)-negative invasive tumor cell population. Consistent with the mouse data, we find that NFIB is overexpressed in almost all tested human metastatic high-grade neuroendocrine lung tumors, warranting further assessment of NFIB as a tumor progression marker in a clinical setting. NFIB drives tumor initiation and progression in mouse models of SCLC NFIB enhances metastasis and changes the metastatic profile NFIB promotes dedifferentiation and invasion in SCLC NFIB marks stage III/IV high-grade neuroendocrine carcinomas in patients
Collapse
Affiliation(s)
- Ekaterina A Semenova
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Min-Chul Kwon
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Kim Monkhorst
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Rajith Bhaskaran
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Oscar Krijgsman
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Thomas Kuilman
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Dennis Peters
- Core Facility for Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Wieneke A Buikhuisen
- Division of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Egbert F Smit
- Division of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Colin Pritchard
- Mouse Clinic for Cancer and Aging research Transgenic Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Miranda Cozijnsen
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Jan van der Vliet
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - John Zevenhoven
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Jan-Paul Lambooij
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Natalie Proost
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Erwin van Montfort
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Arno Velds
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Ivo J Huijbers
- Mouse Clinic for Cancer and Aging research Transgenic Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands.
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands; Skolkovo Institute of Science and Technology, Moscow 143026, Russia.
| |
Collapse
|