1
|
Liu Z, Park T. DMOIT: denoised multi-omics integration approach based on transformer multi-head self-attention mechanism. Front Genet 2024; 15:1488683. [PMID: 39720180 PMCID: PMC11666520 DOI: 10.3389/fgene.2024.1488683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Multi-omics data integration has become increasingly crucial for a deeper understanding of the complexity of biological systems. However, effectively integrating and analyzing multi-omics data remains challenging due to their heterogeneity and high dimensionality. Existing methods often struggle with noise, redundant features, and the complex interactions between different omics layers, leading to suboptimal performance. Additionally, they face difficulties in adequately capturing intra-omics interactions due to simplistic concatenation techiniques, and they risk losing critical inter-omics interaction information when using hierarchical attention layers. To address these challenges, we propose a novel Denoised Multi-Omics Integration approach that leverages the Transformer multi-head self-attention mechanism (DMOIT). DMOIT consists of three key modules: a generative adversarial imputation network for handling missing values, a sampling-based robust feature selection module to reduce noise and redundant features, and a multi-head self-attention (MHSA) based feature extractor with a noval architecture that enchance the intra-omics interaction capture. We validated model porformance using cancer datasets from the Cancer Genome Atlas (TCGA), conducting two tasks: survival time classification across different cancer types and estrogen receptor status classification for breast cancer. Our results show that DMOIT outperforms traditional machine learning methods and the state-of-the-art integration method MoGCN in terms of accuracy and weighted F1 score. Furthermore, we compared DMOIT with various alternative MHSA-based architectures to further validate our approach. Our results show that DMOIT consistently outperforms these models across various cancer types and different omics combinations. The strong performance and robustness of DMOIT demonstrate its potential as a valuable tool for integrating multi-omics data across various applications.
Collapse
Affiliation(s)
- Zhe Liu
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Kin K, Bhogale S, Zhu L, Thomas D, Bertol J, Zheng WJ, Sinha S, Fakhouri WD. Sequence-to-expression approach to identify etiological non-coding DNA variations in P53 and cMYC-driven diseases. Hum Mol Genet 2024; 33:1697-1710. [PMID: 39017605 PMCID: PMC11413647 DOI: 10.1093/hmg/ddae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/08/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
Disease risk prediction based on genomic sequence and transcriptional profile can improve disease screening and prevention. Despite identifying many disease-associated DNA variants, distinguishing deleterious non-coding DNA variations remains poor for most common diseases. In this study, we designed in vitro experiments to uncover the significance of occupancy and competitive binding between P53 and cMYC on common target genes. Analyzing publicly available ChIP-seq data for P53 and cMYC in embryonic stem cells showed that ~344-366 regions are co-occupied, and on average, two cis-overlapping motifs (CisOMs) per region were identified, suggesting that co-occupancy is evolutionarily conserved. Using U2OS and Raji cells untreated and treated with doxorubicin to increase P53 protein level while potentially reducing cMYC level, ChIP-seq analysis illustrated that around 16 to 922 genomic regions were co-occupied by P53 and cMYC, and substitutions of cMYC signals by P53 were detected post doxorubicin treatment. Around 187 expressed genes near co-occupied regions were altered at mRNA level according to RNA-seq data analysis. We utilized a computational motif-matching approach to illustrate that changes in predicted P53 binding affinity in CisOMs of co-occupied elements significantly correlate with alterations in reporter gene expression. We performed a similar analysis using SNPs mapped in CisOMs for P53 and cMYC from ChIP-seq data, and expression of target genes from GTEx portal. We found significant correlation between change in cMYC-motif binding affinity in CisOMs and altered expression. Our study brings us closer to developing a generally applicable approach to filter etiological non-coding variations associated with common diseases.
Collapse
Affiliation(s)
- Katherine Kin
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - Shounak Bhogale
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana–Champaign, 600 S Mathews Ave, Urbana, IL 61801, United States
| | - Lisha Zhu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX 77030, United States
| | - Derrick Thomas
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - Jessica Bertol
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St #600, Houston, TX 77030, United States
| | - Saurabh Sinha
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana–Champaign, 600 S Mathews Ave, Urbana, IL 61801, United States
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, North Avenue Atlanta, GA 30332, United States
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston, 7500 Cambridge St, Houston, TX 77054, United States
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, United States
| |
Collapse
|
3
|
Samuels E, Parks J, Chu J, McDonald T, Spinelli J, Murphy RA, Bhatti P. Metabolites Associated with Polygenic Risk of Breast Cancer. Metabolites 2024; 14:295. [PMID: 38921430 PMCID: PMC11205321 DOI: 10.3390/metabo14060295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
While hundreds of germline genetic variants have been associated with breast cancer risk, the mechanisms underlying the impacts of most of these variants on breast cancer remain uncertain. Metabolomics may offer valuable insights into the mechanisms underlying genetic risks of breast cancer. Among 143 cancer-free female participants, we used linear regression analyses to explore associations between the genetic risk of breast cancer, as determined by a previously developed polygenic risk score (PRS) that included 266 single-nucleotide polymorphisms (SNPs), and 223 measures of metabolites obtained from blood samples using nuclear magnetic resonance (NMR). A false discovery rate of 10% was applied to account for multiple comparisons. PRS was statistically significantly associated with 45 metabolite measures. These were primarily measures of very low-density lipoproteins (VLDLs) and high-density lipoproteins (HDLs), including triglycerides, cholesterol, and phospholipids. For example, the strongest effect was observed with the percent ratio of medium VLDL triglycerides to total lipids (0.53 unit increase in mean-standardized ln-transformed percent ratio per unit increase in PRS; q = 0.1). While larger-scale studies are needed to confirm these results, this exploratory study presents biologically plausible findings that are consistent with previously reported associations between lipids and breast cancer risk. If confirmed, these lipids could be targeted for lifestyle and pharmaceutical interventions among women at increased genetic risk of breast cancer.
Collapse
Affiliation(s)
- Elizabeth Samuels
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jaclyn Parks
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Jessica Chu
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Treena McDonald
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - John Spinelli
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Rachel A. Murphy
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
4
|
Salmerón-Bárcenas EG, Zacapala-Gómez AE, Torres-Rojas FI, Antonio-Véjar V, Ávila-López PA, Baños-Hernández CJ, Núñez-Martínez HN, Dircio-Maldonado R, Martínez-Carrillo DN, Ortiz-Ortiz J, Jiménez-Wences H. TET Enzymes and 5hmC Levels in Carcinogenesis and Progression of Breast Cancer: Potential Therapeutic Targets. Int J Mol Sci 2023; 25:272. [PMID: 38203443 PMCID: PMC10779134 DOI: 10.3390/ijms25010272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Breast Cancer (BC) was the most common female cancer in incidence and mortality worldwide in 2020. Similarly, BC was the top female cancer in the USA in 2022. Risk factors include earlier age at menarche, oral contraceptive use, hormone replacement therapy, high body mass index, and mutations in BRCA1/2 genes, among others. BC is classified into Luminal A, Luminal B, HER2-like, and Basal-like subtypes. These BC subtypes present differences in gene expression signatures, which can impact clinical behavior, treatment response, aggressiveness, metastasis, and survival of patients. Therefore, it is necessary to understand the epigenetic molecular mechanism of transcriptional regulation in BC, such as DNA demethylation. Ten-Eleven Translocation (TET) enzymes catalyze the oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) on DNA, which in turn inhibits or promotes the gene expression. Interestingly, the expression of TET enzymes as well as the levels of the 5hmC epigenetic mark are altered in several types of human cancers, including BC. Several studies have demonstrated that TET enzymes and 5hmC play a key role in the regulation of gene expression in BC, directly (dependent or independent of DNA de-methylation) or indirectly (via interaction with other proteins such as transcription factors). In this review, we describe our recent understanding of the regulatory and physiological function of the TET enzymes, as well as their potential role as biomarkers in BC biology.
Collapse
Affiliation(s)
- Eric Genaro Salmerón-Bárcenas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México C.P. 07360, Mexico; (E.G.S.-B.); (P.A.Á.-L.)
| | - Ana Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (A.E.Z.-G.); (F.I.T.-R.); (V.A.-V.); (J.O.-O.)
| | - Francisco Israel Torres-Rojas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (A.E.Z.-G.); (F.I.T.-R.); (V.A.-V.); (J.O.-O.)
| | - Verónica Antonio-Véjar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (A.E.Z.-G.); (F.I.T.-R.); (V.A.-V.); (J.O.-O.)
| | - Pedro Antonio Ávila-López
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México C.P. 07360, Mexico; (E.G.S.-B.); (P.A.Á.-L.)
| | - Christian Johana Baños-Hernández
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C. P. 44340, Jalisco, Mexico;
| | - Hober Nelson Núñez-Martínez
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México C. P. 04510, Mexico;
| | - Roberto Dircio-Maldonado
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (R.D.-M.); (D.N.M.-C.)
| | - Dinorah Nashely Martínez-Carrillo
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (R.D.-M.); (D.N.M.-C.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (A.E.Z.-G.); (F.I.T.-R.); (V.A.-V.); (J.O.-O.)
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico; (R.D.-M.); (D.N.M.-C.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo C. P. 39090, Guerrero, Mexico
| |
Collapse
|
5
|
Letsou W, Wang F, Moon W, Im C, Sapkota Y, Robison LL, Yasui Y. Refining the genetic risk of breast cancer with rare haplotypes and pattern mining. Life Sci Alliance 2023; 6:e202302183. [PMID: 37541849 PMCID: PMC10403637 DOI: 10.26508/lsa.202302183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
Hundreds of common variants have been found to confer small but significant differences in breast cancer risk, supporting the widely accepted polygenic model of inherited predisposition. Using a novel closed-pattern mining algorithm, we provide evidence that rare haplotypes may refine the association of breast cancer risk with common germline alleles. Our method, called Chromosome Overlap, consists in iteratively pairing chromosomes from affected individuals and looking for noncontiguous patterns of shared alleles. We applied Chromosome Overlap to haplotypes of genotyped SNPs from female breast cancer cases from the UK Biobank at four loci containing common breast cancer-risk SNPs. We found two rare (frequency <0.1%) haplotypes bearing a GWAS hit at 11q13 (hazard ratio = 4.21 and 16.7) which replicated in an independent, European ancestry population at P < 0.05, and another at 22q12 (frequency <0.2%, hazard ratio = 2.58) which expanded the risk pool to noncarriers of a GWAS hit. These results suggest that rare haplotypes (or mutations) may underlie the "synthetic association" of breast cancer risk with at least some common variants.
Collapse
Affiliation(s)
- William Letsou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Fan Wang
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wonjong Moon
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cindy Im
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yutaka Yasui
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
- School of Public Health, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Kin K, Bhogale S, Zhu L, Thomas D, Bertol J, Zheng WJ, Sinha S, Fakhouri WD. Sequence-to-expression approach to identify etiological non-coding DNA variations in P53 and cMYC-driven diseases. RESEARCH SQUARE 2023:rs.3.rs-3037310. [PMID: 37503250 PMCID: PMC10371153 DOI: 10.21203/rs.3.rs-3037310/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background and methods Disease risk prediction based on DNA sequence and transcriptional profile can improve disease screening, prevention, and potential therapeutic approaches by revealing contributing genetic factors and altered regulatory networks. Despite identifying many disease-associated DNA variants through genome-wide association studies, distinguishing deleterious non-coding DNA variations remains poor for most common diseases. We previously reported that non-coding variations disrupting cis-overlapping motifs (CisOMs) of opposing transcription factors significantly affect enhancer activity. We designed in vitro experiments to uncover the significance of the co-occupancy and competitive binding and inhibition between P53 and cMYC on common target gene expression. Results Analyzing publicly available ChIP-seq data for P53 and cMYC in human embryonic stem cells and mouse embryonic cells showed that ~ 344-366 genomic regions are co-occupied by P53 and cMYC. We identified, on average, two CisOMs per region, suggesting that co-occupancy is evolutionarily conserved in vertebrates. Our data showed that treating U2OS cells with doxorubicin increased P53 protein level while reducing cMYC level. In contrast, no change in protein levels was observed in Raji cells. ChIP-seq analysis illustrated that 16-922 genomic regions were co-occupied by P53 and cMYC before and after treatment, and substitutions of cMYC signals by P53 were detected after doxorubicin treatment in U2OS. Around 187 expressed genes near co-occupied regions were altered at mRNA level according to RNA-seq data. We utilized a computational motif-matching approach to determine that changes in predicted P53 binding affinity by DNA variations in CisOMs of co-occupied elements significantly correlate with alterations in reporter gene expression. We performed a similar analysis using SNPs mapped in CisOMs for P53 and cMYC from ChIP-seq data in U2OS and Raji, and expression of target genes from the GTEx portal. Conclusions We found a significant correlation between change in motif-predicted cMYC binding affinity by SNPs in CisOMs and altered gene expression. Our study brings us closer to developing a generally applicable approach to filter etiological non-coding variations associated with P53 and cMYC-dependent diseases.
Collapse
Affiliation(s)
- Katherine Kin
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | | | - Lisha Zhu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston
| | - Derrick Thomas
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | - Jessica Bertol
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston
| | - Saurabh Sinha
- The Wallace H. Coulter Department of Biomedical Engineering
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of Dentistry, University of Texas Health Science Center at Houston
| |
Collapse
|
7
|
Li N, Zethoven M, McInerny S, Healey E, DeSilva D, Devereux L, Scott RJ, James PA, Campbell IG. Contribution of large genomic rearrangements in PALB2 to familial breast cancer: implications for genetic testing. J Med Genet 2023; 60:112-118. [PMID: 35396271 DOI: 10.1136/jmedgenet-2021-108399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/08/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND PALB2 is the most important contributor to familial breast cancer after BRCA1 and BRCA2. Large genomic rearrangements (LGRs) in BRCA1 and BRCA2 are routinely assessed in clinical testing and are a significant contributor to the yield of actionable findings. In contrast, the contribution of LGRs in PALB2 has not been systematically studied. METHODS We performed targeted sequencing and real-time qPCR validation to identify LGRs in PALB2 in 5770 unrelated patients with familial breast cancer and 5741 cancer-free control women from the same Australian population. RESULTS Seven large deletions ranging in size from 0.96 kbp to 18.07 kbp involving PALB2 were identified in seven cases, while no LGRs were identified in any of the controls. Six LGRs were considered pathogenic as they included one or more exons of PALB2 and disrupted the WD40 domain at the C terminal end of the PALB2 protein while one LGR only involved a partial region of intron 10 and was considered a variant of unknown significance. Altogether, pathogenic LGRs identified in this study accounted for 10.3% (6 of 58) of the pathogenic PALB2 variants detected among the 5770 families with familial breast cancer. CONCLUSIONS Our data show that a clinically important proportion of PALB2 pathogenic mutations in Australian patients with familial breast cancer are LGRs. Such observations have provided strong support for inclusion of PALB2 LGRs in routine clinical genetic testing.
Collapse
Affiliation(s)
- Na Li
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Magnus Zethoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Simone McInerny
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Eliza Healey
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Dilanka DeSilva
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Lisa Devereux
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Lifepool, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Division of Molecular Medicine, Pathology North, Newcastle, New South Wales, Australia.,Discipline of Medical Genetics, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Paul A James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, 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
|
8
|
Liu C, Zhou X, Jin J, Zhu Q, Li L, Yin Q, Xu T, Gu W, Ma F, Yang R. The Association Between Breast Cancer and Blood-Based Methylation of CD160, ISYNA1 and RAD51B in the Chinese Population. Front Genet 2022; 13:927519. [PMID: 35812748 PMCID: PMC9261985 DOI: 10.3389/fgene.2022.927519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/23/2022] [Indexed: 12/25/2022] Open
Abstract
Recent studies have identified DNA methylation signatures in the white blood cells as potential biomarkers for breast cancer (BC) in the European population. Here, we investigated the association between BC and blood-based methylation of cluster of differentiation 160 (CD160), inositol-3-phosphate synthase 1 (ISYNA1) and RAD51 paralog B (RAD51B) genes in the Chinese population. Peripheral blood samples were collected from two independent case-control studies with a total of 272 sporadic early-stage BC cases (76.5% at stage I&II) and 272 cancer-free female controls. Mass spectrometry was applied to quantitatively measure the levels of DNA methylation. The logistic regression and non-parametric tests were used for the statistical analyses. In contrast to the protective effects reported in European women, we reported the blood-based hypomethylation in CD160, ISYNA1 and RAD51B as risk factors for BC in the Chinese population (CD160_CpG_3, CD160_CpG_4/cg20975414, ISYNA1_CpG_2, RAD51B_CpG_3 and RAD51B_CpG_4; odds ratios (ORs) per -10% methylation ranging from 1.08 to 1.67, p < 0.05 for all). Moreover, hypomethylation of CD160, ISYNA1 and RAD51B was significantly correlated with age, BC subtypes including estrogen receptor (ER)-negative BC tumors, triple negative tumors, BC cases with larger size, advanced stages and more lymph node involvement. Our results supported the report in European women that BC is associated with altered methylation of CD160, ISYNA1 and RAD51B in the peripheral blood, although the effects are opposite in the Chinese population. The difference between the two populations may be due to variant genetic background or life styles, implicating that the validations of epigenetic biomarkers in variant ethnic groups are warranted.
Collapse
Affiliation(s)
- Chunlan Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiajie Zhou
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jialie Jin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiang Zhu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lixi Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qiming Yin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tian Xu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rongxi Yang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Rongxi Yang,
| |
Collapse
|
9
|
Xu B, Wang H, Tan L. Dysregulated TET Family Genes and Aberrant 5mC Oxidation in Breast Cancer: Causes and Consequences. Cancers (Basel) 2021; 13:cancers13236039. [PMID: 34885145 PMCID: PMC8657367 DOI: 10.3390/cancers13236039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Both genetic and epigenetic mechanisms contribute to the pathogenesis of breast cancer. Since Tahiliani et al. identified TET1 as the first methyl-cytosine dioxygenase in 2009, accumulating evidence has shown that aberrant 5mC oxidation and dysregulated TET family genes are associated with diseases, including breast cancer. In this review we provide an overview on the diagnosis and prognosis values of aberrant 5mC oxidation in breast cancer and emphasize the causes and consequences of such epigenetic alterations. Abstract DNA methylation (5-methylcytosine, 5mC) was once viewed as a stable epigenetic modification until Rao and colleagues identified Ten-eleven translocation 1 (TET1) as the first 5mC dioxygenase in 2009. TET family genes (including TET1, TET2, and TET3) encode proteins that can catalyze 5mC oxidation and consequently modulate DNA methylation, not only regulating embryonic development and cellular differentiation, but also playing critical roles in various physiological and pathophysiological processes. Soon after the discovery of TET family 5mC dioxygenases, aberrant 5mC oxidation and dysregulation of TET family genes have been reported in breast cancer as well as other malignancies. The impacts of aberrant 5mC oxidation and dysregulated TET family genes on the different aspects (so-called cancer hallmarks) of breast cancer have also been extensively investigated in the past decade. In this review, we summarize current understanding of the causes and consequences of aberrant 5mC oxidation in the pathogenesis of breast cancer. The challenges and future perspectives of this field are also discussed.
Collapse
Affiliation(s)
- Bo Xu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China;
| | - Hao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Correspondence: (H.W.); (L.T.); Tel.: +86-21-54237876 (L.T.)
| | - Li Tan
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China;
- Correspondence: (H.W.); (L.T.); Tel.: +86-21-54237876 (L.T.)
| |
Collapse
|
10
|
Identification of Novel CircRNA-miRNA-mRNA Regulatory Network and Its Prognostic Prediction in Breast Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2916398. [PMID: 34745276 PMCID: PMC8570857 DOI: 10.1155/2021/2916398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022]
Abstract
Aim This study aimed to investigate the expression profiles of circRNAs and candidate circRNA-miRNA-mRNA network in BC. Methods Differentially expressed circRNAs, miRNAs, and mRNAs (DEcircRNAs, DEmiRNAs, and DEmRNAs) between BC and normal breast tissue samples were screened by analyzing raw data of the RNA sequencing profile. The expression levels of hub genes in 48 pairs of cancerous and tumor-free breast tissues surgically resected from BC patients were determined by RT-qPCR analysis. Results A total of 145 DEcircRNAs, 140 DEmiRNAs, and 2451 DEmRNAs between BC and normal breast tissue samples were screened out. There were 5 pairs of upcircRNA-downmiRNA-upmRNA network and 20 pairs of downcircRNA-upmiRNA-downmRNA network. EIF4EBP1, DUSP1, EGR2, EZH1, and CBX7 were found to be correlated with overall survival of the patients with BC. The expression level of EIF4EBP1 was increased and the expression levels of DUSP1, EGR2, EZH1, and CBX7 were decreased in cancerous breast tissues compared to tumor-free breast tissues (p < 0.0001). The RT-qPCR results from 48 BC patients were consistent with the bioinformatics results. Conclusion This study provides a novel perspective to study circRNA-miRNA-mRNA network in BC and assists in the identification of new potential biomarkers to be used for diagnostic and prognostic purposes.
Collapse
|
11
|
Fonfria M, de Juan Jiménez I, Tena I, Chirivella I, Richart-Aznar P, Segura A, Sánchez-Heras AB, Martinez-Dueñas E. Prevalence and Clinicopathological Characteristics of Moderate and High-Penetrance Genes in Non-BRCA1/2 Breast Cancer High-Risk Spanish Families. J Pers Med 2021; 11:548. [PMID: 34204722 PMCID: PMC8231620 DOI: 10.3390/jpm11060548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 01/10/2023] Open
Abstract
(1) Background: Over the last decade, genetic counseling clinics have moved from single-gene sequencing to multigene panel sequencing. Multiple genes related to a moderate risk of breast cancer (BC) have emerged, although many questions remain regarding the risks and clinical features associated with these genes. (2) Methods: Ninety-six BC index cases (ICs) with high-risk features for hereditary breast and ovarian cancer (HBOC) and with a previous uninformative result for BRCA1/2 were tested with a panel of 41 genes associated with BC risk. The frequency of pathogenic variants (PVs) was related to the clinical characteristics of BC. (3) Results: We detected a PV rate of 13.5% (excluding two cases each of BRCA1 and MUTYH). Among the 95 assessed cases, 17 PVs were identified in 16 ICs, as follows: BRCA1 (n = 2), CHEK2 (n = 3), ATM (n = 5), MUTYH (n = 2), TP53 (n = 2), BRIP1 (n = 1), CASP8 (n = 1), and MSH2 (n = 1). We also identified a novel loss-of-function variant in CASP8, a candidate gene for increased BC risk. There was no evidence that the clinical characteristics of BC might be related to a higher chance of identifying a PV. (4) Conclusions: In our cohort, which was enriched with families with a high number of BC cases, a high proportion of mutations in ATM and CHEK2 were identified. The clinical characteristics of BC associated with moderate-risk genes were different from those related to BRCA1/2 genes.
Collapse
Affiliation(s)
- Maria Fonfria
- Cancer Genetic Counseling Unit, Medical Oncology Department, Castellon Provincial Hospital, 12002 Castellon, Spain; (M.F.); (I.T.); (E.M.-D.)
| | | | - Isabel Tena
- Cancer Genetic Counseling Unit, Medical Oncology Department, Castellon Provincial Hospital, 12002 Castellon, Spain; (M.F.); (I.T.); (E.M.-D.)
| | - Isabel Chirivella
- Medical Oncology Department, INCLIVA Biomedical Research Institute, University of Valencia, 46001 Valencia, Spain;
| | - Paula Richart-Aznar
- Cancer Genetic Counseling Unit, Medical Oncology Department, La Fe University Hospital, 46026 Valencia, Spain; (P.R.-A.); (A.S.)
| | - Angel Segura
- Cancer Genetic Counseling Unit, Medical Oncology Department, La Fe University Hospital, 46026 Valencia, Spain; (P.R.-A.); (A.S.)
| | - Ana Beatriz Sánchez-Heras
- Cancer Genetic Counseling Unit, Medical Oncology Department, Elche University Hospital, 03203 Elche, Spain;
| | - Eduardo Martinez-Dueñas
- Cancer Genetic Counseling Unit, Medical Oncology Department, Castellon Provincial Hospital, 12002 Castellon, Spain; (M.F.); (I.T.); (E.M.-D.)
| |
Collapse
|
12
|
Li N, Zethoven M, McInerny S, Devereux L, Huang YK, Thio N, Cheasley D, Gutiérrez-Enríquez S, Moles-Fernández A, Diez O, Nguyen-Dumont T, Southey MC, Hopper JL, Simard J, Dumont M, Soucy P, Meindl A, Schmutzler R, Schmidt MK, Adank MA, Andrulis IL, Hahnen E, Engel C, Lesueur F, Girard E, Neuhausen SL, Ziv E, Allen J, Easton DF, Scott RJ, Gorringe KL, James PA, Campbell IG. Evaluation of the association of heterozygous germline variants in NTHL1 with breast cancer predisposition: an international multi-center study of 47,180 subjects. NPJ Breast Cancer 2021; 7:52. [PMID: 33980861 PMCID: PMC8115524 DOI: 10.1038/s41523-021-00255-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Bi-allelic loss-of-function (LoF) variants in the base excision repair (BER) gene NTHL1 cause a high-risk hereditary multi-tumor syndrome that includes breast cancer, but the contribution of heterozygous variants to hereditary breast cancer is unknown. An analysis of 4985 women with breast cancer, enriched for familial features, and 4786 cancer-free women revealed significant enrichment for NTHL1 LoF variants. Immunohistochemistry confirmed reduced NTHL1 expression in tumors from heterozygous carriers but the NTHL1 bi-allelic loss characteristic mutational signature (SBS 30) was not present. The analysis was extended to 27,421 breast cancer cases and 19,759 controls from 10 international studies revealing 138 cases and 93 controls with a heterozygous LoF variant (OR 1.06, 95% CI: 0.82-1.39) and 316 cases and 179 controls with a missense variant (OR 1.31, 95% CI: 1.09-1.57). Missense variants selected for deleterious features by a number of in silico bioinformatic prediction tools or located within the endonuclease III functional domain showed a stronger association with breast cancer. Somatic sequencing of breast cancers from carriers indicated that the risk associated with NTHL1 appears to operate through haploinsufficiency, consistent with other described low-penetrance breast cancer genes. Data from this very large international multicenter study suggests that heterozygous pathogenic germline coding variants in NTHL1 may be associated with low- to moderate- increased risk of breast cancer.
Collapse
Affiliation(s)
- Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Magnus Zethoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Simone McInerny
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Lisa Devereux
- Lifepool, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Yu-Kuan Huang
- Upper Gastrointestinal Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Vic, Australia
| | - Niko Thio
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
| | - Sara Gutiérrez-Enríquez
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Alejandro Moles-Fernández
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Orland Diez
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO); Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Area of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Tu Nguyen-Dumont
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Martine Dumont
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Penny Soucy
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec, Canada
| | - Alfons Meindl
- University of Munich, Campus Großhadern, Department of Gynecology and Obstetrics, Munich, Germany
| | - Rita Schmutzler
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Familial Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Integrated Oncology (CIO), Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Muriel A Adank
- Family Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eric Hahnen
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Familial Breast and Ovarian Cancer, Cologne, Germany
| | - Christoph Engel
- Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Fabienne Lesueur
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Elodie Girard
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Jamie Allen
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Discipline of Medical Genetics, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Division of Molecular Medicine, Pathology North, Newcastle, NSW, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Vic, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Vic, Australia.
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
13
|
Cheasley D, Nigam A, Zethoven M, Hunter S, Etemadmoghadam D, Semple T, Allan P, Carey MS, Fernandez ML, Dawson A, Köbel M, Huntsman DG, Le Page C, Mes-Masson AM, Provencher D, Hacker N, Gao Y, Bowtell D, deFazio A, Gorringe KL, Campbell IG. Genomic analysis of low-grade serous ovarian carcinoma to identify key drivers and therapeutic vulnerabilities. J Pathol 2020; 253:41-54. [PMID: 32901952 DOI: 10.1002/path.5545] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 12/22/2022]
Abstract
Low-grade serous ovarian carcinoma (LGSOC) is associated with a poor response to existing chemotherapy, highlighting the need to perform comprehensive genomic analysis and identify new therapeutic vulnerabilities. The data presented here represent the largest genetic study of LGSOCs to date (n = 71), analysing 127 candidate genes derived from whole exome sequencing cohorts to generate mutation and copy-number variation data. Additionally, immunohistochemistry was performed on our LGSOC cohort assessing oestrogen receptor, progesterone receptor, TP53, and CDKN2A status. Targeted sequencing identified 47% of cases with mutations in key RAS/RAF pathway genes (KRAS, BRAF, and NRAS), as well as mutations in putative novel driver genes including USP9X (27%), MACF1 (11%), ARID1A (9%), NF2 (4%), DOT1L (6%), and ASH1L (4%). Immunohistochemistry evaluation revealed frequent oestrogen/progesterone receptor positivity (85%), along with CDKN2A protein loss (10%) and CDKN2A protein overexpression (6%), which were linked to shorter disease outcomes. Indeed, 90% of LGSOC samples harboured at least one potentially actionable alteration, which in 19/71 (27%) cases were predictive of clinical benefit from a standard treatment, either in another cancer's indication or in LGSOC specifically. In addition, we validated ubiquitin-specific protease 9X (USP9X), which is a chromosome X-linked substrate-specific deubiquitinase and tumour suppressor, as a relevant therapeutic target for LGSOC. Our comprehensive genomic study highlighted that there is an addiction to a limited number of unique 'driver' aberrations that could be translated into improved therapeutic paths. © 2020 The 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, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Abhimanyu Nigam
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Magnus Zethoven
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Bioinformatics Consulting Core, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sally Hunter
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Dariush Etemadmoghadam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Timothy Semple
- Molecular Genomics Core, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Prue Allan
- Department of Clinical Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, VIC, Australia
| | - Mark S Carey
- Department of Obstetrics & Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marta L Fernandez
- Department of Obstetrics & Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Amy Dawson
- Department of Obstetrics & Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Cécile Le Page
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Neville Hacker
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Yunkai Gao
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - David Bowtell
- Cancer Genetics and Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Anna deFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney and the Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Kylie L Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Cheasley D, Devereux L, Hughes S, Nickson C, Procopio P, Lee G, Li N, Pridmore V, Elder K, Bruce Mann G, Kader T, Rowley SM, Fox SB, Byrne D, Saunders H, Fujihara KM, Lim B, Gorringe KL, Campbell IG. The TP53 mutation rate differs in breast cancers that arise in women with high or low mammographic density. NPJ Breast Cancer 2020; 6:34. [PMID: 32802943 PMCID: PMC7414106 DOI: 10.1038/s41523-020-00176-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 07/13/2020] [Indexed: 01/01/2023] Open
Abstract
Mammographic density (MD) influences breast cancer risk, but how this is mediated is unknown. Molecular differences between breast cancers arising in the context of the lowest and highest quintiles of mammographic density may identify the mechanism through which MD drives breast cancer development. Women diagnosed with invasive or in situ breast cancer where MD measurement was also available (n = 842) were identified from the Lifepool cohort of >54,000 women participating in population-based mammographic screening. This group included 142 carcinomas in the lowest quintile of MD and 119 carcinomas in the highest quintile. Clinico-pathological and family history information were recorded. Tumor DNA was collected where available (n = 56) and sequenced for breast cancer predisposition and driver gene mutations, including copy number alterations. Compared to carcinomas from low-MD breasts, those from high-MD breasts were significantly associated with a younger age at diagnosis and features associated with poor prognosis. Low- and high-MD carcinomas matched for grade, histological subtype, and hormone receptor status were compared for somatic genetic features. Low-MD carcinomas had a significantly increased frequency of TP53 mutations, higher homologous recombination deficiency, higher fraction of the genome altered, and more copy number gains on chromosome 1q and losses on 17p. While high-MD carcinomas showed enrichment of tumor-infiltrating lymphocytes in the stroma. The data demonstrate that when tumors were matched for confounding clinico-pathological features, a proportion in the lowest quintile of MD appear biologically distinct, reflective of microenvironment differences between the lowest and highest quintiles of MD.
Collapse
Affiliation(s)
- Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Lisa Devereux
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Lifepool, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Siobhan Hughes
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Carolyn Nickson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
- Cancer Research Division, Cancer Council NSW, Sydney, NSW Australia
- Sydney School of Public Health, University of Sydney, Sydney, NSW Australia
| | - Pietro Procopio
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
- Cancer Research Division, Cancer Council NSW, Sydney, NSW Australia
- Sydney School of Public Health, University of Sydney, Sydney, NSW Australia
| | - Grant Lee
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | | | - Kenneth Elder
- Department of Surgery, University of Melbourne, Melbourne, VIC Australia
- The Royal Melbourne and Royal Women’s Hospitals, Parkville, VIC Australia
- The Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - G. Bruce Mann
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC Australia
- The Royal Melbourne and Royal Women’s Hospitals, Parkville, VIC Australia
| | - Tanjina Kader
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Simone M. Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Stephen B. Fox
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, VIC Australia
| | - David Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, VIC Australia
| | - Hugo Saunders
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Kenji M. Fujihara
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Belle Lim
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kylie L. Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Cancer Genetics and Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Ian G. Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
15
|
Li N, McInerny S, Zethoven M, Cheasley D, Lim BWX, Rowley SM, Devereux L, Grewal N, Ahmadloo S, Byrne D, Lee JEA, Li J, Fox SB, John T, Antill Y, Gorringe KL, James PA, Campbell IG. Combined Tumor Sequencing and Case-Control Analyses of RAD51C in Breast Cancer. J Natl Cancer Inst 2020; 111:1332-1338. [PMID: 30949688 DOI: 10.1093/jnci/djz045] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/07/2019] [Accepted: 04/03/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Loss-of-function variants in RAD51C are associated with familial ovarian cancer, but its role in hereditary breast cancer remains unclear. The aim of this study was to couple breast tumor sequencing with case-control data to clarify the contribution of RAD51C to hereditary breast cancer. METHODS RAD51C was sequenced in 3080 breast cancer index cases that were negative in BRCA1/2 clinical tests and 4840 population-matched cancer-free controls. Pedigree and pathology data were analyzed. Nine breast cancers and one ovarian cancer from RAD51C variant carriers were sequenced to identify biallelic inactivation of RAD51C, copy number variation, mutational signatures, and the spectrum of somatic mutations in breast cancer driver genes. The promoter of RAD51C was analyzed for DNA methylation. RESULTS A statistically significant excess of loss-of-function variants was identified in 3080 cases (0.4%) compared with 2 among 4840 controls (0.04%; odds ratio = 8.67, 95% confidence interval = 1.89 to 80.52, P< .001), with more than half of the carriers having no personal or family history of ovarian cancer. In addition, the association was highly statistically significant among cases with estrogen-negative (P <. 001) or triple-negative cancer (P < .001), but not in estrogen-positive cases. Tumor sequencing from carriers confirmed bi-allelic inactivation in all the triple-negative cases and was associated with high homologous recombination deficiency scores and mutational signature 3 indicating homologous recombination repair deficiency. CONCLUSIONS This study provides evidence that germline loss-of-function variants of RAD51C are associated with hereditary breast cancer, particularly triple-negative type. RAD51C-null breast cancers possess similar genomic and clinical features to BRCA1-null cancers and may also be vulnerable to DNA double-strand break inducing chemotherapies and poly ADP-ribose polymerase inhibitors.
Collapse
|
16
|
Yanes T, Young MA, Meiser B, James PA. Clinical applications of polygenic breast cancer risk: a critical review and perspectives of an emerging field. Breast Cancer Res 2020; 22:21. [PMID: 32066492 PMCID: PMC7026946 DOI: 10.1186/s13058-020-01260-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/07/2020] [Indexed: 01/04/2023] Open
Abstract
Polygenic factors are estimated to account for an additional 18% of the familial relative risk of breast cancer, with those at the highest level of polygenic risk distribution having a least a twofold increased risk of the disease. Polygenic testing promises to revolutionize health services by providing personalized risk assessments to women at high-risk of breast cancer and within population breast screening programs. However, implementation of polygenic testing needs to be considered in light of its current limitations, such as limited risk prediction for women of non-European ancestry. This article aims to provide a comprehensive review of the evidence for polygenic breast cancer risk, including the discovery of variants associated with breast cancer at the genome-wide level of significance and the use of polygenic risk scores to estimate breast cancer risk. We also review the different applications of this technology including testing of women from high-risk breast cancer families with uninformative genetic testing results, as a moderator of monogenic risk, and for population screening programs. Finally, a potential framework for introducing testing for polygenic risk in familial cancer clinics and the potential challenges with implementing this technology in clinical practice are discussed.
Collapse
Affiliation(s)
- Tatiane Yanes
- Psychosocial Research Group, Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia. .,The University of Queensland Diamantina Institute, Dermatology Research Centre, University of Queensland, Brisbane, QLD, 4102, Australia.
| | - Mary-Anne Young
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Bettina Meiser
- Psychosocial Research Group, Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Paul A James
- Parkville Integrated Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| |
Collapse
|
17
|
Chen J, Wu JS, Mize T, Moreno M, Hamid M, Servin F, Bashy B, Zhao Z, Jia P, Tsuang MT, Kendler KS, Xiong M, Chen X. A Frameshift Variant in the CHST9 Gene Identified by Family-Based Whole Genome Sequencing Is Associated with Schizophrenia in Chinese Population. Sci Rep 2019; 9:12717. [PMID: 31481703 PMCID: PMC6722128 DOI: 10.1038/s41598-019-49052-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/19/2019] [Indexed: 12/30/2022] Open
Abstract
Recent studies imply that rare variants contribute to the risk of schizophrenia, however, the exact variants or genes responsible for this condition are largely unknown. In this study, we conducted whole genome sequencing (WGS) of 20 Chinese families. Each family consisted of at least two affected siblings diagnosed with schizophrenia and at least one unaffected sibling. We examined functional variants that were found in affected sibling(s) but not in unaffected sibling(s) within a family. Matching this criterion, a frameshift heterozygous deletion of CA (–/CA) at chromosome 18:24722722, also referred to as rs752084147, in the Carbohydrate Sulfotransferase 9 (CHST9) gene, was detected in two families. This deletion was confirmed by PCR-based Sanger sequencing. With the observed frequency of 0.00076 in Han Chinese population, we performed both case-control and family-based analyses to evaluate its association with schizophrenia. In the case-control analyses, Chi-square test P-value was 6.80e-12 and the P-value was 0.0008 after one million simulations. In family-based segregation analyses, segregation P-value was 7.72e-7 and simulated P-value was 5.70e-6. For both the case-control and family-based analyses, the CA deletion was significantly associated with schizophrenia in the Chinese population. Further investigation of this gene is warranted in the development of schizophrenia by utilizing larger and more ethnically diverse samples.
Collapse
Affiliation(s)
- Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA.
| | - Jain-Shing Wu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Travis Mize
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA.,Department of Psychology, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Marvi Moreno
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Mahtab Hamid
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Francisco Servin
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Bita Bashy
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S, Maryland Parkway, Las Vegas, NV, 89154-4009, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Ming T Tsuang
- Department of Psychiatry, University of California at San Diego, San Diego, CA, 92093, USA
| | - Kenneth S Kendler
- Virginia Institute of Psychiatric and Behavioral Genetics, Medical College of Virginia and Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Momiao Xiong
- Department of Biostatistics and Data Science, Human Genetics Center, University of Texas School of Public Health, Houston, TX, 77030, USA
| | | |
Collapse
|
18
|
Danková Z, Žúbor P, Grendár M, Zelinová K, Jagelková M, Stastny I, Kapinová A, Vargová D, Kasajová P, Dvorská D, Kalman M, Danko J, Lasabová Z. Predictive accuracy of the breast cancer genetic risk model based on eight common genetic variants: The BACkSIDE study. J Biotechnol 2019; 299:1-7. [DOI: 10.1016/j.jbiotec.2019.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 12/24/2022]
|
19
|
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: 15] [Impact Index Per Article: 2.5] [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
|