Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Bioinformatics ; 39(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-37027230

ABSTRACT

SUMMARY: Identification of duplicate templates is a common preprocessing step in bulk sequence analysis; for large libraries, this can be resource intensive. Here, we present streammd: a fast, memory-efficient, single-pass duplicate marker operating on the principle of a Bloom filter. streammd closely reproduces outputs from Picard MarkDuplicates while being substantially faster, and requires much less memory than SAMBLASTER. AVAILABILITY AND IMPLEMENTATION: streammd is a C++ program available from GitHub https://github.com/delocalizer/streammd under the MIT license.


Subject(s)
Software , Sequence Analysis, DNA
2.
Nature ; 545(7653): 175-180, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467829

ABSTRACT

Melanoma of the skin is a common cancer only in Europeans, whereas it arises in internal body surfaces (mucosal sites) and on the hands and feet (acral sites) in people throughout the world. Here we report analysis of whole-genome sequences from cutaneous, acral and mucosal subtypes of melanoma. The heavily mutated landscape of coding and non-coding mutations in cutaneous melanoma resolved novel signatures of mutagenesis attributable to ultraviolet radiation. However, acral and mucosal melanomas were dominated by structural changes and mutation signatures of unknown aetiology, not previously identified in melanoma. The number of genes affected by recurrent mutations disrupting non-coding sequences was similar to that affected by recurrent mutations to coding sequences. Significantly mutated genes included BRAF, CDKN2A, NRAS and TP53 in cutaneous melanoma, BRAF, NRAS and NF1 in acral melanoma and SF3B1 in mucosal melanoma. Mutations affecting the TERT promoter were the most frequent of all; however, neither they nor ATRX mutations, which correlate with alternative telomere lengthening, were associated with greater telomere length. Most melanomas had potentially actionable mutations, most in components of the mitogen-activated protein kinase and phosphoinositol kinase pathways. The whole-genome mutation landscape of melanoma reveals diverse carcinogenic processes across its subtypes, some unrelated to sun exposure, and extends potential involvement of the non-coding genome in its pathogenesis.


Subject(s)
Genome, Human/genetics , Melanoma/genetics , Mutation/genetics , DNA Helicases/genetics , GTP Phosphohydrolases/genetics , Genes, p16 , Humans , Melanoma/classification , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Neurofibromatosis 1/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA Splicing Factors/genetics , Signal Transduction/drug effects , Telomerase/genetics , Telomere/genetics , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays/adverse effects , X-linked Nuclear Protein
3.
Nature ; 543(7643): 65-71, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28199314

ABSTRACT

The diagnosis of pancreatic neuroendocrine tumours (PanNETs) is increasing owing to more sensitive detection methods, and this increase is creating challenges for clinical management. We performed whole-genome sequencing of 102 primary PanNETs and defined the genomic events that characterize their pathogenesis. Here we describe the mutational signatures they harbour, including a deficiency in G:C > T:A base excision repair due to inactivation of MUTYH, which encodes a DNA glycosylase. Clinically sporadic PanNETs contain a larger-than-expected proportion of germline mutations, including previously unreported mutations in the DNA repair genes MUTYH, CHEK2 and BRCA2. Together with mutations in MEN1 and VHL, these mutations occur in 17% of patients. Somatic mutations, including point mutations and gene fusions, were commonly found in genes involved in four main pathways: chromatin remodelling, DNA damage repair, activation of mTOR signalling (including previously undescribed EWSR1 gene fusions), and telomere maintenance. In addition, our gene expression analyses identified a subgroup of tumours associated with hypoxia and HIF signalling.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Genome, Human/genetics , Genomics , Pancreatic Neoplasms/genetics , Base Sequence , Calmodulin-Binding Proteins/genetics , Chromatin Assembly and Disassembly/genetics , Chromosome Aberrations , DNA Copy Number Variations/genetics , DNA Glycosylases/genetics , DNA Mutational Analysis , DNA Repair/genetics , Female , Germ-Line Mutation/genetics , Humans , Male , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , TOR Serine-Threonine Kinases/metabolism , Telomere/genetics , Telomere/metabolism
5.
Hum Mutat ; 43(12): 2054-2062, 2022 12.
Article in English | MEDLINE | ID: mdl-36095262

ABSTRACT

The clinical classification of variants may change with new information, however, there is limited guidance on how often significant changes in variant classification occur. We used ClinVar to examine how variant classification changes over time. We developed a custom parser and accessed variant data from ClinVar between January 2015 and July 2021. The ClinVar-assigned "aggregate" classification of variants in 121 hereditary cancer genes was harmonized across releases to align to the American College of Medical Genetics and Genomics and the Association for Molecular Pathology terms. Aggregate classification categories were grouped as: benign/likely benign (B/LB); likely pathogenic/pathogenic (LP/P); variant of uncertain significance (VUS); conflicting interpretations of pathogenicity (Conflicting); or Other. We profiled changes in aggregate variant classification between consecutive semi-annual ClinVar releases. The proportion of variants that changed aggregate classification between semi-annual ClinVar releases ranged from 0.6% to 6.4%. The most frequent changes were "VUS to conflicting," "other to LP/P," and "B/LB to Conflicting." A limited number of variants changed aggregate classification from "LP/P to B/LB," or vice versa. Our analysis indicates need for regular reassessment of clinical variant interpretations. The parser developed for this project will facilitate extraction of relevant interpretation data from ClinVar.


Subject(s)
Genetic Testing , Neoplasms , Humans , United States , Genetic Variation , Genetic Predisposition to Disease , Genomics , Software , Neoplasms/diagnosis , Neoplasms/genetics
6.
Nature ; 531(7592): 47-52, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26909576

ABSTRACT

Integrated genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-ß, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. Expression analysis defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) aberrantly differentiated endocrine exocrine (ADEX) that correlate with histopathological characteristics. Squamous tumours are enriched for TP53 and KDM6A mutations, upregulation of the TP63∆N transcriptional network, hypermethylation of pancreatic endodermal cell-fate determining genes and have a poor prognosis. Pancreatic progenitor tumours preferentially express genes involved in early pancreatic development (FOXA2/3, PDX1 and MNX1). ADEX tumours displayed upregulation of genes that regulate networks involved in KRAS activation, exocrine (NR5A2 and RBPJL), and endocrine differentiation (NEUROD1 and NKX2-2). Immunogenic tumours contained upregulated immune networks including pathways involved in acquired immune suppression. These data infer differences in the molecular evolution of pancreatic cancer subtypes and identify opportunities for therapeutic development.


Subject(s)
Genes, Neoplasm/genetics , Genome, Human/genetics , Genomics , Mutation/genetics , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Pancreatic Ductal/classification , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Histone Demethylases/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Survival Analysis , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Zebrafish Proteins
7.
Hum Mutat ; 42(5): 530-536, 2021 05.
Article in English | MEDLINE | ID: mdl-33600021

ABSTRACT

Aggregate population genomics data from large cohorts are vital for assessing germline variant pathogenicity. However, there are no specifications on how sequencing quality metrics should be considered, and whether exome-derived and genome-derived allele frequencies should be considered in isolation. Germline genome sequence data were simulated for nine read-depths to identify a minimum acceptable read-depth for detecting variants. gnomAD exome-derived and genome-derived datasets were assessed for read-depth, for six key cancer genes selected for variant curation by ClinGen expert panels. Non-Finnish European allele frequency (AF) or filter AF of coding variants in these genes, assigned into frequency bins using modified ACMG-AMP criteria, was compared between exome-derived and genome-derived datasets. A 30X read-depth achieved acceptable precision and recall for detection of substitutions, but poor recall for small insertions/deletions. Exome-derived and genome-derived datasets exhibited low read-depth for different gene exons. Individual variants were mostly assigned to non-divergent AF bins (>95%) or filter AF bins (>97%). Two major bin divergences were resolved by applying the minimal acceptable read-depth threshold. These findings show the importance of assessing read-depth separately for population datasets sourced from different short-read sequencing technologies before assigning a frequency-based ACMG-AMP classification code for variant interpretation.


Subject(s)
Genome, Human , Neoplasms , Gene Frequency , Genetic Testing , Genetic Variation , Genomics , Germ Cells , Humans , Neoplasms/genetics
8.
Nature ; 518(7540): 495-501, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25719666

ABSTRACT

Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.


Subject(s)
DNA Mutational Analysis , Genome, Human/genetics , Genomics , Mutation/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , DNA Repair/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Markers/genetics , Genomic Instability/genetics , Genotype , Humans , Mice , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/drug therapy , Platinum/pharmacology , Point Mutation/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Xenograft Model Antitumor Assays
9.
J Pathol ; 247(2): 214-227, 2019 02.
Article in English | MEDLINE | ID: mdl-30350370

ABSTRACT

Metaplastic breast carcinoma (MBC) is relatively rare but accounts for a significant proportion of global breast cancer mortality. This group is extremely heterogeneous and by definition exhibits metaplastic change to squamous and/or mesenchymal elements, including spindle, squamous, chondroid, osseous, and rhabdomyoid features. Clinically, patients are more likely to present with large primary tumours (higher stage), distant metastases, and overall, have shorter 5-year survival compared to invasive carcinomas of no special type. The current World Health Organisation (WHO) diagnostic classification for this cancer type is based purely on morphology - the biological basis and clinical relevance of its seven sub-categories are currently unclear. By establishing the Asia-Pacific MBC (AP-MBC) Consortium, we amassed a large series of MBCs (n = 347) and analysed the mutation profile of a subset, expression of 14 breast cancer biomarkers, and clinicopathological correlates, contextualising our findings within the WHO guidelines. The most significant indicators of poor prognosis were large tumour size (T3; p = 0.004), loss of cytokeratin expression (lack of staining with pan-cytokeratin AE1/3 antibody; p = 0.007), EGFR overexpression (p = 0.01), and for 'mixed' MBC, the presence of more than three distinct morphological entities (p = 0.007). Conversely, fewer morphological components and EGFR negativity were favourable indicators. Exome sequencing of 30 cases confirmed enrichment of TP53 and PTEN mutations, and intriguingly, concurrent mutations of TP53, PTEN, and PIK3CA. Mutations in neurofibromatosis-1 (NF1) were also overrepresented [16.7% MBCs compared to ∼5% of breast cancers overall; enrichment p = 0.028; mutation significance p = 0.006 (OncodriveFM)], consistent with published case reports implicating germline NF1 mutations in MBC risk. Taken together, we propose a practically minor but clinically significant modification to the guidelines: all WHO_1 mixed-type tumours should have the number of morphologies present recorded, as a mechanism for refining prognosis, and that EGFR and pan-cytokeratin expression are important prognostic markers. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Mutation , Neoplasms, Complex and Mixed/genetics , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/classification , Breast Neoplasms/pathology , Cadherins/analysis , Class I Phosphatidylinositol 3-Kinases/genetics , Cross-Sectional Studies , Epithelial-Mesenchymal Transition , ErbB Receptors/analysis , Female , Genetic Predisposition to Disease , Humans , Keratins/analysis , Metaplasia , Middle Aged , Neoplasm Grading , Neoplasms, Complex and Mixed/chemistry , Neoplasms, Complex and Mixed/classification , Neoplasms, Complex and Mixed/pathology , Neurofibromin 1/genetics , PTEN Phosphohydrolase/genetics , Phenotype , Tumor Burden , Tumor Suppressor Protein p53/genetics
10.
Int J Cancer ; 144(5): 1049-1060, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30178487

ABSTRACT

Cutaneous melanoma accounts for at least >10% of all cancers in adolescents and young adults (AYA, 15-30 years of age) in Western countries. To date, little is known about the correlations between germline variants and somatic mutations and mutation signatures in AYA melanoma patients that might explain why they have developed a cancer predominantly affecting those over 65 years of age. We performed genomic analysis of 50 AYA melanoma patients (onset 10-30 years, median 20); 25 underwent whole genome sequencing (WGS) of both tumor and germline DNA, exome data were retrieved from 12 TCGA AYA cases, and targeted DNA sequencing was conducted on 13 cases. The AYA cases were compared with WGS data from 121 adult cutaneous melanomas. Similar to mature adult cutaneous melanomas, AYA melanomas showed a high mutation burden and mutation signatures of ultraviolet radiation (UVR) damage. The frequencies of somatic mutations in BRAF (96%) and PTEN (36%) in the AYA WGS cohort were double the rates observed in adult melanomas (Q < 6.0 × 10-6 and 0.028, respectively). Furthermore, AYA melanomas contained a higher proportion of non-UVR-related mutation signatures than mature adult melanomas as a proportion of total mutation burden (p = 2.0 × 10-4 ). Interestingly, these non-UVR mutation signatures relate to APOBEC or mismatch repair pathways, and germline variants in related genes were observed in some of these cases. We conclude that AYA melanomas harbor some of the same molecular aberrations and mutagenic insults occurring in older adults, but in different proportions. Germline variants that may have conferred disease susceptibility correlated with somatic mutation signatures in a subset of AYA melanomas.


Subject(s)
Genetic Predisposition to Disease/genetics , Germ Cells/physiology , Melanoma/genetics , Mutation/genetics , Adolescent , Adult , Cohort Studies , Female , Humans , Male , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins B-raf/genetics , Whole Genome Sequencing/methods , Young Adult
11.
Gastroenterology ; 152(1): 68-74.e2, 2017 01.
Article in English | MEDLINE | ID: mdl-27856273

ABSTRACT

Pancreatic cancer is molecularly diverse, with few effective therapies. Increased mutation burden and defective DNA repair are associated with response to immune checkpoint inhibitors in several other cancer types. We interrogated 385 pancreatic cancer genomes to define hypermutation and its causes. Mutational signatures inferring defects in DNA repair were enriched in those with the highest mutation burdens. Mismatch repair deficiency was identified in 1% of tumors harboring different mechanisms of somatic inactivation of MLH1 and MSH2. Defining mutation load in individual pancreatic cancers and the optimal assay for patient selection may inform clinical trial design for immunotherapy in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA Mismatch Repair/genetics , Mutation , Pancreatic Neoplasms/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Genome , Humans , Male , Middle Aged , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Proto-Oncogene Proteins p21(ras)/genetics
12.
Nature ; 491(7424): 399-405, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23103869

ABSTRACT

Pancreatic cancer is a highly lethal malignancy with few effective therapies. We performed exome sequencing and copy number analysis to define genomic aberrations in a prospectively accrued clinical cohort (n = 142) of early (stage I and II) sporadic pancreatic ductal adenocarcinoma. Detailed analysis of 99 informative tumours identified substantial heterogeneity with 2,016 non-silent mutations and 1,628 copy-number variations. We define 16 significantly mutated genes, reaffirming known mutations (KRAS, TP53, CDKN2A, SMAD4, MLL3, TGFBR2, ARID1A and SF3B1), and uncover novel mutated genes including additional genes involved in chromatin modification (EPC1 and ARID2), DNA damage repair (ATM) and other mechanisms (ZIM2, MAP2K4, NALCN, SLC16A4 and MAGEA6). Integrative analysis with in vitro functional data and animal models provided supportive evidence for potential roles for these genetic aberrations in carcinogenesis. Pathway-based analysis of recurrently mutated genes recapitulated clustering in core signalling pathways in pancreatic ductal adenocarcinoma, and identified new mutated genes in each pathway. We also identified frequent and diverse somatic aberrations in genes described traditionally as embryonic regulators of axon guidance, particularly SLIT/ROBO signalling, which was also evident in murine Sleeping Beauty transposon-mediated somatic mutagenesis models of pancreatic cancer, providing further supportive evidence for the potential involvement of axon guidance genes in pancreatic carcinogenesis.


Subject(s)
Axons/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Genome/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice , Mutation , Proteins/genetics , Signal Transduction
13.
J Pathol ; 237(3): 363-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26172396

ABSTRACT

Treatment options for patients with brain metastases (BMs) have limited efficacy and the mortality rate is virtually 100%. Targeted therapy is critically under-utilized, and our understanding of mechanisms underpinning metastatic outgrowth in the brain is limited. To address these deficiencies, we investigated the genomic and transcriptomic landscapes of 36 BMs from breast, lung, melanoma and oesophageal cancers, using DNA copy-number analysis and exome- and RNA-sequencing. The key findings were as follows. (a) Identification of novel candidates with possible roles in BM development, including the significantly mutated genes DSC2, ST7, PIK3R1 and SMC5, and the DNA repair, ERBB-HER signalling, axon guidance and protein kinase-A signalling pathways. (b) Mutational signature analysis was applied to successfully identify the primary cancer type for two BMs with unknown origins. (c) Actionable genomic alterations were identified in 31/36 BMs (86%); in one case we retrospectively identified ERBB2 amplification representing apparent HER2 status conversion, then confirmed progressive enrichment for HER2-positivity across four consecutive metastatic deposits by IHC and SISH, resulting in the deployment of HER2-targeted therapy for the patient. (d) In the ERBB/HER pathway, ERBB2 expression correlated with ERBB3 (r(2) = 0.496; p < 0.0001) and HER3 and HER4 were frequently activated in an independent cohort of 167 archival BM from seven primary cancer types: 57.6% and 52.6% of cases were phospho-HER3(Y1222) or phospho-HER4(Y1162) membrane-positive, respectively. The HER3 ligands NRG1/2 were barely detectable by RNAseq, with NRG1 (8p12) genomic loss in 63.6% breast cancer-BMs, suggesting a microenvironmental source of ligand. In summary, this is the first study to characterize the genomic landscapes of BM. The data revealed novel candidates, potential clinical applications for genomic profiling of resectable BMs, and highlighted the possibility of therapeutically targeting HER3, which is broadly over-expressed and activated in BMs, independent of primary site and systemic therapy.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Gene Expression Profiling/methods , Genomics/methods , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , DNA Mutational Analysis , Enzyme Activation , Gene Amplification , Gene Dosage , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Ligands , Molecular Targeted Therapy , Mutation , Phenotype , Phosphorylation , Precision Medicine , Predictive Value of Tests , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Tumor Microenvironment
14.
Int J Cancer ; 135(5): 1110-8, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24500968

ABSTRACT

The importance of epigenetic modifications such as DNA methylation in tumorigenesis is increasingly being appreciated. To define the genome-wide pattern of DNA methylation in pancreatic ductal adenocarcinomas (PDAC), we captured the methylation profiles of 167 untreated resected PDACs and compared them to a panel of 29 adjacent nontransformed pancreata using high-density arrays. A total of 11,634 CpG sites associated with 3,522 genes were significantly differentially methylated (DM) in PDAC and were capable of segregating PDAC from non-malignant pancreas, regardless of tumor cellularity. As expected, PDAC hypermethylation was most prevalent in the 5' region of genes (including the proximal promoter, 5'UTR and CpG islands). Approximately 33% DM genes showed significant inverse correlation with mRNA expression levels. Pathway analysis revealed an enrichment of aberrantly methylated genes involved in key molecular mechanisms important to PDAC: TGF-ß, WNT, integrin signaling, cell adhesion, stellate cell activation and axon guidance. Given the recent discovery that SLIT-ROBO mutations play a clinically important role in PDAC, the role of epigenetic perturbation of axon guidance was pursued in more detail. Bisulfite amplicon deep sequencing and qRT-PCR expression analyses confirmed recurrent perturbation of axon guidance pathway genes SLIT2, SLIT3, ROBO1, ROBO3, ITGA2 and MET and suggests epigenetic suppression of SLIT-ROBO signaling and up-regulation of MET and ITGA2 expression. Hypomethylation of MET and ITGA2 correlated with high gene expression, which was associated with poor survival. These data suggest that aberrant methylation plays an important role in pancreatic carcinogenesis affecting core signaling pathways with potential implications for the disease pathophysiology and therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Cell Adhesion/genetics , Female , Gene Expression Profiling , Humans , Integrin alpha2/genetics , Integrins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Middle Aged , Nerve Tissue Proteins/genetics , Pancreatic Ducts/pathology , Pancreatic Stellate Cells/pathology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/biosynthesis , Receptors, Immunologic/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics , Roundabout Proteins
15.
Cancers (Basel) ; 16(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38398180

ABSTRACT

Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is often the only source of tumor tissue from patients with advanced, inoperable lung cancer. EBUS-TBNA aspirates are used for the diagnosis, staging, and genomic testing to inform therapy options. Here we extracted DNA and RNA from 220 EBUS-TBNA aspirates to evaluate their suitability for whole genome (WGS), whole exome (WES), and comprehensive panel sequencing. For a subset of 40 cases, the same nucleic acid extraction was sequenced using WGS, WES, and the TruSight Oncology 500 assay. Genomic features were compared between sequencing platforms and compared with those reported by clinical testing. A total of 204 aspirates (92.7%) had sufficient DNA (100 ng) for comprehensive panel sequencing, and 109 aspirates (49.5%) had sufficient material for WGS. Comprehensive sequencing platforms detected all seven clinically reported tier 1 actionable mutations, an additional three (7%) tier 1 mutations, six (15%) tier 2-3 mutations, and biomarkers of potential immunotherapy benefit (tumor mutation burden and microsatellite instability). As expected, WGS was more suited for the detection and discovery of emerging novel biomarkers of treatment response. WGS could be performed in half of all EBUS-TBNA aspirates, which points to the enormous potential of EBUS-TBNA as source material for large, well-curated discovery-based studies for novel and more effective predictors of treatment response. Comprehensive panel sequencing is possible in the vast majority of fresh EBUS-TBNA aspirates and enhances the detection of actionable mutations over current clinical testing.

16.
Nat Commun ; 14(1): 3155, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37258531

ABSTRACT

Oesophageal adenocarcinoma is a poor prognosis cancer and the molecular features underpinning response to treatment remain unclear. We investigate whole genome, transcriptomic and methylation data from 115 oesophageal adenocarcinoma patients mostly from the DOCTOR phase II clinical trial (Australian New Zealand Clinical Trials Registry-ACTRN12609000665235), with exploratory analysis pre-specified in the study protocol of the trial. We report genomic features associated with poorer overall survival, such as the APOBEC mutational and RS3-like rearrangement signatures. We also show that positron emission tomography non-responders have more sub-clonal genomic copy number alterations. Transcriptomic analysis categorises patients into four immune clusters correlated with survival. The immune suppressed cluster is associated with worse survival, enriched with myeloid-derived cells, and an epithelial-mesenchymal transition signature. The immune hot cluster is associated with better survival, enriched with lymphocytes, myeloid-derived cells, and an immune signature including CCL5, CD8A, and NKG7. The immune clusters highlight patients who may respond to immunotherapy and thus may guide future clinical trials.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Humans , Neoadjuvant Therapy , Multiomics , Australia , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics
17.
Genome Med ; 15(1): 74, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37723522

ABSTRACT

BACKGROUND: Many families and individuals do not meet criteria for a known hereditary cancer syndrome but display unusual clusters of cancers. These families may carry pathogenic variants in cancer predisposition genes and be at higher risk for developing cancer. METHODS: This multi-centre prospective study recruited 195 cancer-affected participants suspected to have a hereditary cancer syndrome for whom previous clinical targeted genetic testing was either not informative or not available. To identify pathogenic disease-causing variants explaining participant presentation, germline whole-genome sequencing (WGS) and a comprehensive cancer virtual gene panel analysis were undertaken. RESULTS: Pathogenic variants consistent with the presenting cancer(s) were identified in 5.1% (10/195) of participants and pathogenic variants considered secondary findings with potential risk management implications were identified in another 9.7% (19/195) of participants. Health economic analysis estimated the marginal cost per case with an actionable variant was significantly lower for upfront WGS with virtual panel ($8744AUD) compared to standard testing followed by WGS ($24,894AUD). Financial analysis suggests that national adoption of diagnostic WGS testing would require a ninefold increase in government annual expenditure compared to conventional testing. CONCLUSIONS: These findings make a case for replacing conventional testing with WGS to deliver clinically important benefits for cancer patients and families. The uptake of such an approach will depend on the perspectives of different payers on affordability.


Subject(s)
Neoplastic Syndromes, Hereditary , Humans , Prospective Studies , Oncogenes , Genetic Testing , Germ Cells
18.
Bioinform Adv ; 2(1): vbac005, 2022.
Article in English | MEDLINE | ID: mdl-36699384

ABSTRACT

Motivation: Changes in telomere length have been observed in cancer and can be indicative of mechanisms involved in carcinogenesis. Most methods used to estimate telomere length require laboratory analysis of DNA samples. Here, we present qmotif, a fast and easy tool that determines telomeric repeat sequences content as an estimate of telomere length directly from whole-genome sequencing. Results: qmotif shows similar results to quantitative PCR, the standard method for high-throughput clinical telomere length quantification. qmotif output correlates strongly with the output of other tools for determining telomere sequence content, TelSeq and TelomereHunter, but can run in a fraction of the time-usually under a minute. Availability and implementation: qmotif is implemented in Java and source code is available at https://github.com/AdamaJava/adamajava, with instructions on how to build and use the application available from https://adamajava.readthedocs.io/en/latest/. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

19.
Genome Med ; 14(1): 58, 2022 05 30.
Article in English | MEDLINE | ID: mdl-35637530

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) has a poor overall survival with few treatment options. Whole genome sequencing (WGS) combined with the immune features of MPM offers the prospect of identifying changes that could inform future clinical trials. METHODS: We analysed somatic mutations from 229 MPM samples, including previously published data and 58 samples that had undergone WGS within this study. This was combined with RNA-seq analysis to characterize the tumour immune environment. RESULTS: The comprehensive genome analysis identified 12 driver genes, including new candidate genes. Whole genome doubling was a frequent event that correlated with shorter survival. Mutational signature analysis revealed SBS5/40 were dominant in 93% of samples, and defects in homologous recombination repair were infrequent in our cohort. The tumour immune environment contained high M2 macrophage infiltrate linked with MMP2, MMP14, TGFB1 and CCL2 expression, representing an immune suppressive environment. The expression of TGFB1 was associated with overall survival. A small subset of samples (less than 10%) had a higher proportion of CD8 T cells and a high cytolytic score, suggesting a 'hot' immune environment independent of the somatic mutations. CONCLUSIONS: We propose accounting for genomic and immune microenvironment status may influence therapeutic planning in the future.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Genomics , Humans , Lung Neoplasms/genetics , Mesothelioma/genetics , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Tumor Microenvironment/genetics
20.
Cancer Cell ; 40(1): 88-102.e7, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34951955

ABSTRACT

We concurrently examine the whole genome, transcriptome, methylome, and immune cell infiltrates in baseline tumors from 77 patients with advanced cutaneous melanoma treated with anti-PD-1 with or without anti-CTLA-4. We show that high tumor mutation burden (TMB), neoantigen load, expression of IFNγ-related genes, programmed death ligand expression, low PSMB8 methylation (therefore high expression), and T cells in the tumor microenvironment are associated with response to immunotherapy. No specific mutation correlates with therapy response. A multivariable model combining the TMB and IFNγ-related gene expression robustly predicts response (89% sensitivity, 53% specificity, area under the curve [AUC], 0.84); tumors with high TMB and a high IFNγ signature show the best response to immunotherapy. This model validates in an independent cohort (80% sensitivity, 59% specificity, AUC, 0.79). Except for a JAK3 loss-of-function mutation, for patients who did not respond as predicted there is no obvious biological mechanism that clearly explained their outlier status, consistent with intratumor and intertumor heterogeneity in response to immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , B7-H1 Antigen/immunology , Biomarkers, Tumor/genetics , CTLA-4 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Immunotherapy/methods , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Melanoma/immunology , Mutation/genetics , Skin Neoplasms/immunology , Tumor Microenvironment/immunology , Melanoma, Cutaneous Malignant
SELECTION OF CITATIONS
SEARCH DETAIL