Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Clin Oncol ; 40(18): 2036-2047, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35263119

ABSTRACT

PURPOSE: Tubo-ovarian cancer (TOC) is a sentinel cancer for BRCA1 and BRCA2 pathogenic variants (PVs). Identification of a PV in the first member of a family at increased genetic risk (the proband) provides opportunities for cancer prevention in other at-risk family members. Although Australian testing rates are now high, PVs in patients with TOC whose diagnosis predated revised testing guidelines might have been missed. We assessed the feasibility of detecting PVs in this population to enable genetic risk reduction in relatives. PATIENTS AND METHODS: In this pilot study, deceased probands were ascertained from research cohort studies, identification by a relative, and gynecologic oncology clinics. DNA was extracted from archival tissue or stored blood for panel sequencing of 10 risk-associated genes. Testing of deceased probands ascertained through clinic records was performed with a consent waiver. RESULTS: We identified 85 PVs in 84 of 787 (11%) probands. Familial contacts of 39 of 60 (65%) deceased probands with an identified recipient (60 of 84; 71%) have received a written notification of results, with follow-up verbal contact made in 85% (33 of 39). A minority of families (n = 4) were already aware of the PV. For many (29 of 33; 88%), the genetic result provided new information and referral to a genetic service was accepted in most cases (66%; 19 of 29). Those who declined referral (4 of 29) were all male next of kin whose family member had died more than 10 years before. CONCLUSION: We overcame ethical and logistic challenges to demonstrate that retrospective genetic testing to identify PVs in previously untested deceased probands with TOC is feasible. Understanding reasons for a family member's decision to accept or decline a referral will be important for guiding future TRACEBACK projects.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Australia , Breast Neoplasms/genetics , Carcinoma, Ovarian Epithelial/genetics , Family , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Male , Ovarian Neoplasms/genetics , Ovarian Neoplasms/prevention & control , Pilot Projects , Retrospective Studies
2.
Cell Death Dis ; 12(11): 959, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663790

ABSTRACT

Anal cancer is a rare disease that has doubled in incidence over the last four decades. Current treatment and survival of patients with this disease has not changed substantially over this period of time, due, in part, to a paucity of preclinical models to assess new therapeutic options. To address this hiatus, we set-out to establish, validate and characterise a panel of human anal squamous cell carcinoma (ASCC) cell lines by employing an explant technique using fresh human ASCC tumour tissue. The panel of five human ASCC cell lines were validated to confirm their origin, squamous features and tumourigenicity, followed by molecular and genomic (whole-exome sequencing) characterisation. This panel recapitulates the genetic and molecular characteristics previously described in ASCC including phosphoinositide-3-kinase (PI3K) mutations in three of the human papillomavirus (HPV) positive lines and TP53 mutations in the HPV negative line. The cell lines demonstrate the ability to form tumouroids and retain their tumourigenic potential upon xenotransplantation, with varied inducible expression of major histocompatibility complex class I (MHC class I) and Programmed cell death ligand 1 (PD-L1). We observed differential responses to standard chemotherapy, radiotherapy and a PI3K specific molecular targeted agent in vitro, which correlated with the clinical response of the patient tumours from which they were derived. We anticipate this novel panel of human ASCC cell lines will form a valuable resource for future studies into the biology and therapeutics of this rare disease.


Subject(s)
Anus Neoplasms/genetics , Anus Neoplasms/pathology , Genomics , Animals , Anus Neoplasms/therapy , Anus Neoplasms/ultrastructure , B7-H1 Antigen/metabolism , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/ultrastructure , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Copy Number Variations/genetics , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Gene Dosage , Histocompatibility Antigens Class I/metabolism , Humans , Male , Mice, Nude , Middle Aged , Mitomycin/pharmacology , Mitomycin/therapeutic use , Mutation/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
3.
BMC Res Notes ; 13(1): 349, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32698852

ABSTRACT

OBJECTIVE: Ovarian fibromas and adenofibromas are rare ovarian tumours. They are benign tumours composed of spindle-like stromal cells (pure fibroma) or a mixture of fibroblast and epithelial components (adenofibroma). We have previously shown that 40% of benign serous ovarian tumours are likely primary fibromas due to the neoplastic alterations being restricted to the stromal compartment of these tumours. We further explore this finding by comparing benign serous tumours to pure fibromas. RESULTS: Performing copy number aberration (CNA) analysis on the stromal component of 45 benign serous tumours and 8 pure fibromas, we have again shown that trisomy of chromosome 12 is the most common aberration in ovarian fibromas. CNAs were more frequent in the pure fibromas than the benign serous tumours (88% vs 33%), however pure fibromas more frequently harboured more than one CNA event compared with benign serous tumours. As these extra CNA events observed in the pure fibromas were unique to this subset our data indicates a unique tumour evolution. Gene expression analysis on the two cohorts was unable to show gene expression changes that differed based on tumour subtype. Exome analysis did not reveal any recurrently mutated genes.


Subject(s)
Fibroma , Ovarian Neoplasms , DNA Copy Number Variations , Exome , Female , Fibroma/genetics , Humans , Ovarian Neoplasms/genetics , Trisomy
5.
Gynecol Oncol ; 156(3): 552-560, 2020 03.
Article in English | MEDLINE | ID: mdl-31902686

ABSTRACT

OBJECTIVE: Mucinous ovarian carcinoma (MOC) is an uncommon ovarian cancer histotype that responds poorly to conventional chemotherapy regimens. Although long overall survival outcomes can occur with early detection and optimal surgical resection, recurrent and advanced disease are associated with extremely poor survival. There are no current guidelines specifically for the systemic management of recurrent MOC. We analyzed data from a large cohort of women with MOC to evaluate the potential for clinical utility from a range of systemic agents. METHODS: We analyzed gene copy number (n = 191) and DNA sequencing data (n = 184) from primary MOC to evaluate signatures of mismatch repair deficiency and homologous recombination deficiency, and other genetic events. Immunohistochemistry data were collated for ER, CK7, CK20, CDX2, HER2, PAX8 and p16 (n = 117-166). RESULTS: Molecular aberrations noted in MOC that suggest a match with current targeted therapies include amplification of ERBB2 (26.7%) and BRAF mutation (9%). Observed genetic events that suggest potential efficacy for agents currently in clinical trials include: KRAS/NRAS mutations (66%), TP53 missense mutation (49%), RNF43 mutation (11%), ARID1A mutation (10%), and PIK3CA/PTEN mutation (9%). Therapies exploiting homologous recombination deficiency (HRD) may not be effective in MOC, as only 1/191 had a high HRD score. Mismatch repair deficiency was similarly rare (1/184). CONCLUSIONS: Although genetically diverse, MOC has several potential therapeutic targets. Importantly, the lack of response to platinum-based therapy observed clinically corresponds to the lack of a genomic signature associated with HRD, and MOC are thus also unlikely to respond to PARP inhibition.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Aged , Cohort Studies , DNA Mismatch Repair , Female , Homologous Recombination , Humans , Immunohistochemistry , Mutation , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics
6.
Nat Commun ; 10(1): 3935, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477716

ABSTRACT

Mucinous ovarian carcinoma (MOC) is a unique subtype of ovarian cancer with an uncertain etiology, including whether it genuinely arises at the ovary or is metastatic disease from other organs. In addition, the molecular drivers of invasive progression, high-grade and metastatic disease are poorly defined. We perform genetic analysis of MOC across all histological grades, including benign and borderline mucinous ovarian tumors, and compare these to tumors from other potential extra-ovarian sites of origin. Here we show that MOC is distinct from tumors from other sites and supports a progressive model of evolution from borderline precursors to high-grade invasive MOC. Key drivers of progression identified are TP53 mutation and copy number aberrations, including a notable amplicon on 9p13. High copy number aberration burden is associated with worse prognosis in MOC. Our data conclusively demonstrate that MOC arise from benign and borderline precursors at the ovary and are not extra-ovarian metastases.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Ovarian Epithelial/genetics , Gene Expression Profiling/methods , Ovarian Neoplasms/genetics , Adenocarcinoma, Mucinous/classification , Adenocarcinoma, Mucinous/metabolism , Carcinoma, Ovarian Epithelial/classification , Carcinoma, Ovarian Epithelial/metabolism , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Ovarian Neoplasms/classification , Ovarian Neoplasms/metabolism , Sequence Analysis, DNA/methods , Survival Analysis
7.
Sci Rep ; 9(1): 6426, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015508

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 . A server running CNspector loaded with the figures from this paper can be accessed at https://shiny.wehi.edu.au/jmarkham/CNspector/index.html .


Subject(s)
Basal Cell Nevus Syndrome/diagnosis , Carcinoma, Basal Cell/diagnosis , DNA Copy Number Variations , High-Throughput Nucleotide Sequencing/methods , Web Browser , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Chromosome Deletion , Chromosome Duplication , Exons , Genome, Human , Humans , Internet , Sequence Analysis, DNA
9.
Breast Cancer Res ; 20(1): 3, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29316957

ABSTRACT

BACKGROUND: Genome-wide association studies (GWASs) have identified numerous single-nucleotide polymorphisms (SNPs) associated with small increases in breast cancer risk. Studies to date suggest that some SNPs alter the expression of the associated genes, which potentially mediates risk modification. On this basis, we hypothesised that some of these genes may be enriched for rare coding variants associated with a higher breast cancer risk. METHODS: The coding regions and exon-intron boundaries of 56 genes that have either been proposed by GWASs to be the regulatory targets of the SNPs and/or located < 500 kb from the risk SNPs were sequenced in index cases from 1043 familial breast cancer families that previously had negative test results for BRCA1 and BRCA2 mutations and 944 population-matched cancer-free control participants from an Australian population. Rare (minor allele frequency ≤ 0.001 in the Exome Aggregation Consortium and Exome Variant Server databases) loss-of-function (LoF) and missense variants were studied. RESULTS: LoF variants were rare in both the cases and control participants across all the candidate genes, with only 38 different LoF variants observed in a total of 39 carriers. For the majority of genes (n = 36), no LoF variants were detected in either the case or control cohorts. No individual gene showed a significant excess of LoF or missense variants in the cases compared with control participants. Among all candidate genes as a group, the total number of carriers with LoF variants was higher in the cases than in the control participants (26 cases and 13 control participants), as was the total number of carriers with missense variants (406 versus 353), but neither reached statistical significance (p = 0.077 and p = 0.512, respectively). The genes contributing most of the excess of LoF variants in the cases included TET2, NRIP1, RAD51B and SNX32 (12 cases versus 2 control participants), whereas ZNF283 and CASP8 contributed largely to the excess of missense variants (25 cases versus 8 control participants). CONCLUSIONS: Our data suggest that rare LoF and missense variants in genes associated with low-penetrance breast cancer risk SNPs may contribute some additional risk, but as a group these genes are unlikely to be major contributors to breast cancer heritability.


Subject(s)
Breast Neoplasms/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Loss of Function Mutation/genetics , Adult , Aged , Aged, 80 and over , Australia , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/pathology , Caspase 8/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Gene Frequency , Heterozygote , Humans , Middle Aged , Mutation, Missense , Nuclear Receptor Interacting Protein 1/genetics , Penetrance , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins/genetics
10.
Endocr Relat Cancer ; 25(1): 1-9, 2018 01.
Article in English | MEDLINE | ID: mdl-28974544

ABSTRACT

Pheochromocytomas (PC) and paragangliomas (PGL) are endocrine tumors for which the genetic and clinicopathological features of metastatic progression remain incompletely understood. As a result, the risk of metastasis from a primary tumor cannot be predicted. Early diagnosis of individuals at high risk of developing metastases is clinically important and the identification of new biomarkers that are predictive of metastatic potential is of high value. Activation of TERT has been associated with a number of malignant tumors, including PC/PGL. However, the mechanism of TERT activation in the majority of PC/PGL remains unclear. As TERT promoter mutations occur rarely in PC/PGL, we hypothesized that other mechanisms - such as structural variations - may underlie TERT activation in these tumors. From 35 PC and four PGL, we identified three primary PCs that developed metastases with elevated TERT expression, each of which lacked TERT promoter mutations and promoter DNA methylation. Using whole genome sequencing, we identified somatic structural alterations proximal to the TERT locus in two of these tumors. In both tumors, the genomic rearrangements led to the positioning of super-enhancers proximal to the TERT promoter, that are likely responsible for the activation of the normally tightly repressed TERT expression in chromaffin cells.


Subject(s)
Adrenal Gland Neoplasms/genetics , Biomarkers, Tumor/genetics , Mutation , Paraganglioma/genetics , Pheochromocytoma/genetics , Promoter Regions, Genetic , Telomerase/genetics , Adrenal Gland Neoplasms/secondary , DNA Methylation , Genetic Predisposition to Disease , Humans , Paraganglioma/pathology , Pheochromocytoma/pathology , Prognosis , Whole Genome Sequencing
11.
Nat Commun ; 8: 14581, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262675

ABSTRACT

The Eµ-Myc mouse is an extensively used model of MYC driven malignancy; however to date there has only been partial characterization of MYC co-operative mutations leading to spontaneous lymphomagenesis. Here we sequence spontaneously arising Eµ-Myc lymphomas to define transgene architecture, somatic mutations, and structural alterations. We identify frequent disruptive mutations in the PRC1-like component and BCL6-corepressor gene Bcor. Moreover, we find unexpected concomitant multigenic lesions involving Cdkn2a loss and other cancer genes including Nras, Kras and Bcor. These findings challenge the assumed two-hit model of Eµ-Myc lymphoma and demonstrate a functional in vivo role for Bcor in suppressing tumorigenesis.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Mutation , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/genetics , Alleles , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CRISPR-Cas Systems , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , Disease Models, Animal , Gene Editing , Gene Frequency , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Repressor Proteins/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Whole Genome Sequencing
12.
Urol Oncol ; 34(3): 120.e9-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585945

ABSTRACT

OBJECTIVE: A family history of prostate cancer (PC) is a well-recognized high-risk factor for the development of clinically significant PC. To date, traditional linkage and association studies have identified only a limited number of genes and specific gene variants that account for only a small proportion of PC risk. To identify novel PC predisposition genes we performed whole-exome sequencing of PC-affected men from families with a significant history of PC. METHODS AND MATERIALS: Exome sequencing was performed on 5 PC-affected men from 3 families where there were multiple cases of PCs and where diagnostic testing returned a negative result for BRCA1 and BRCA2 mutations. Genotyping was performed for all potentially predisposing variants detected within each family on the affected and unaffected male participants. RESULTS: Essential splice site, missense, and stop-lost variants were filtered against a recently published candidate gene list. A total of 19 truncating variants and 17 missense variants were identified for genotyping in all prostate-affected and unaffected male participants. In all, 3 missense variants, PCTP, MCRS1, and ATRIP, demonstrated complete segregation and 1 missense variant, PARP2, demonstrated partial segregation with PC. In addition, 3 truncating variants, CYP3A43, DOK3, and PLEKHH3, demonstrated complete segregation and 3 truncation mutations, HEATR5B, GPR124, and HKR1, demonstrated partial segregation with PC. No segregating variants between the 3 families were shared. CONCLUSIONS: In all, 10 truncating or missense variants showed either complete or partial segregation with PC in the relevant families. CYP3A43 and PARP2 variants have been shown to occur in other familial PCs and our findings add to the contribution that these variants potentially have in the risk and development of PC in BRCAX cases.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Biomarkers, Tumor/genetics , Exome/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/genetics , Aged , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Pedigree , Prognosis , Prostatic Neoplasms/pathology
13.
Cancer Res ; 75(24): 5228-34, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26627015

ABSTRACT

Merkel cell carcinoma (MCC) is an uncommon, but highly malignant, cutaneous tumor. Merkel cell polyoma virus (MCV) has been implicated in a majority of MCC tumors; however, viral-negative tumors have been reported to be more prevalent in some geographic regions subject to high sun exposure. While the impact of MCV and viral T-antigens on MCC development has been extensively investigated, little is known about the etiology of viral-negative tumors. We performed targeted capture and massively parallel DNA sequencing of 619 cancer genes to compare the gene mutations and copy number alterations in MCV-positive (n = 13) and -negative (n = 21) MCC tumors and cell lines. We found that MCV-positive tumors displayed very low mutation rates, but MCV-negative tumors exhibited a high mutation burden associated with a UV-induced DNA damage signature. All viral-negative tumors harbored mutations in RB1, TP53, and a high frequency of mutations in NOTCH1 and FAT1. Additional mutated or amplified cancer genes of potential clinical importance included PI3K (PIK3CA, AKT1, PIK3CG) and MAPK (HRAS, NF1) pathway members and the receptor tyrosine kinase FGFR2. Furthermore, looking ahead to potential therapeutic strategies encompassing immune checkpoint inhibitors such as anti-PD-L1, we also assessed the status of T-cell-infiltrating lymphocytes (TIL) and PD-L1 in MCC tumors. A subset of viral-negative tumors exhibited high TILs and PD-L1 expression, corresponding with the higher mutation load within these cancers. Taken together, this study provides new insights into the underlying biology of viral-negative MCC and paves the road for further investigation into new treatment opportunities.


Subject(s)
Carcinoma, Merkel Cell/genetics , DNA Damage/radiation effects , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Blotting, Western , Cells, Cultured , DNA Mutational Analysis , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Merkel cell polyomavirus , Mutation , Polymerase Chain Reaction , Transcriptome
14.
Breast Cancer Res ; 17: 111, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26283626

ABSTRACT

INTRODUCTION: PALB2 is emerging as a high-penetrance breast cancer predisposition gene in the order of BRCA1 and BRCA2. However, large studies that have evaluated the full gene rather than just the most common variants in both cases and controls are required before all truncating variants can be included in familial breast cancer variant testing. METHODS: In this study we analyse almost 2000 breast cancer cases sourced from individuals referred to familial cancer clinics, thus representing typical cases presenting in clinical practice. These cases were compared to a similar number of population-based cancer-free controls. RESULTS: We identified a significant excess of truncating variants in cases (1.3 %) versus controls (0.2 %), including six novel variants (p = 0.0001; odds ratio (OR) 6.58, 95 % confidence interval (CI) 2.3-18.9). Three of the four control individuals carrying truncating variants had at least one relative with breast cancer. There was no excess of missense variants in cases overall, but the common c.1676A > G variant (rs152451) was significantly enriched in cases and may represent a low-penetrance polymorphism (p = 0.002; OR 1.24 (95 % CI 1.09-1.47). CONCLUSIONS: Our findings support truncating variants in PALB2 as high-penetrance breast cancer susceptibility alleles, and suggest that a common missense variant may also lead to a low level of increased breast cancer risk.


Subject(s)
Mutation/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Alleles , Australia , Breast Neoplasms/genetics , Case-Control Studies , Fanconi Anemia Complementation Group N Protein , Genetic Predisposition to Disease/genetics , Humans , Middle Aged , Prevalence , Risk , Young Adult
15.
Bioinformatics ; 28(10): 1307-13, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22474122

ABSTRACT

MOTIVATION: In light of the increasing adoption of targeted resequencing (TR) as a cost-effective strategy to identify disease-causing variants, a robust method for copy number variation (CNV) analysis is needed to maximize the value of this promising technology. RESULTS: We present a method for CNV detection for TR data, including whole-exome capture data. Our method calls copy number gains and losses for each target region based on normalized depth of coverage. Our key strategies include the use of base-level log-ratios to remove GC-content bias, correction for an imbalanced library size effect on log-ratios, and the estimation of log-ratio variations via binning and interpolation. Our methods are made available via CONTRA (COpy Number Targeted Resequencing Analysis), a software package that takes standard alignment formats (BAM/SAM) and outputs in variant call format (VCF4.0), for easy integration with other next-generation sequencing analysis packages. We assessed our methods using samples from seven different target enrichment assays, and evaluated our results using simulated data and real germline data with known CNV genotypes.


Subject(s)
DNA Copy Number Variations , Exome , Sequence Analysis, DNA , Animals , Computer Simulation , HapMap Project , Humans , Mice , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...