Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 323, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238294

ABSTRACT

The unexpected contamination of normal samples with tumour cells reduces variant detection sensitivity, compromising downstream analyses in canonical tumour-normal analyses. Leveraging whole-genome sequencing data available at Genomics England, we develop a tool for normal sample contamination assessment, which we validate in silico and against minimal residual disease testing. From a systematic review of [Formula: see text] patients with haematological malignancies and sarcomas, we find contamination across a range of cancer clinical indications and DNA sources, with highest prevalence in saliva samples from acute myeloid leukaemia patients, and sorted CD3+ T-cells from myeloproliferative neoplasms. Further exploration reveals 108 hotspot mutations in genes associated with haematological cancers at risk of being subtracted by standard variant calling pipelines. Our work highlights the importance of contamination assessment for accurate somatic variants detection in research and clinical settings, especially with large-scale sequencing projects being utilised to deliver accurate data from which to make clinical decisions for patient care.


Subject(s)
Neoplasms , Whole Genome Sequencing , Humans , Genomics , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/genetics , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/pathology
2.
Genome Biol ; 25(1): 38, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38297376

ABSTRACT

Copy number alterations (CNAs) are among the most important genetic events in cancer, but their detection from sequencing data is challenging because of unknown sample purity, tumor ploidy, and general intra-tumor heterogeneity. Here, we present CNAqc, an evolution-inspired method to perform the computational validation of clonal and subclonal CNAs detected from bulk DNA sequencing. CNAqc is validated using single-cell data and simulations, is applied to over 4000 TCGA and PCAWG samples, and is incorporated into the validation process for the clinically accredited bioinformatics pipeline at Genomics England. CNAqc is designed to support automated quality control procedures for tumor somatic data validation.


Subject(s)
DNA Copy Number Variations , Neoplasms , Humans , Algorithms , Polymorphism, Single Nucleotide , Neoplasms/genetics , Neoplasms/pathology , Genomics/methods , Computational Biology/methods
3.
PLoS Comput Biol ; 19(11): e1011557, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37917660

ABSTRACT

Single-cell RNA and ATAC sequencing technologies enable the examination of gene expression and chromatin accessibility in individual cells, providing insights into cellular phenotypes. In cancer research, it is important to consistently analyze these states within an evolutionary context on genetic clones. Here we present CONGAS+, a Bayesian model to map single-cell RNA and ATAC profiles onto the latent space of copy number clones. CONGAS+ clusters cells into tumour subclones with similar ploidy, rendering straightforward to compare their expression and chromatin profiles. The framework, implemented on GPU and tested on real and simulated data, scales to analyse seamlessly thousands of cells, demonstrating better performance than single-molecule models, and supporting new multi-omics assays. In prostate cancer, lymphoma and basal cell carcinoma, CONGAS+ successfully identifies complex subclonal architectures while providing a coherent mapping between ATAC and RNA, facilitating the study of genotype-phenotype maps and their connection to genomic instability.


Subject(s)
DNA Copy Number Variations , RNA , RNA/genetics , Bayes Theorem , DNA Copy Number Variations/genetics , Clone Cells , High-Throughput Nucleotide Sequencing/methods , Chromatin
4.
Nat Genet ; 55(3): 451-460, 2023 03.
Article in English | MEDLINE | ID: mdl-36894710

ABSTRACT

In cancer, evolutionary forces select for clones that evade the immune system. Here we analyzed >10,000 primary tumors and 356 immune-checkpoint-treated metastases using immune dN/dS, the ratio of nonsynonymous to synonymous mutations in the immunopeptidome, to measure immune selection in cohorts and individuals. We classified tumors as immune edited when antigenic mutations were removed by negative selection and immune escaped when antigenicity was covered up by aberrant immune modulation. Only in immune-edited tumors was immune predation linked to CD8 T cell infiltration. Immune-escaped metastases experienced the best response to immunotherapy, whereas immune-edited patients did not benefit, suggesting a preexisting resistance mechanism. Similarly, in a longitudinal cohort, nivolumab treatment removes neoantigens exclusively in the immunopeptidome of nonimmune-edited patients, the group with the best overall survival response. Our work uses dN/dS to differentiate between immune-edited and immune-escaped tumors, measuring potential antigenicity and ultimately helping predict response to treatment.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Nivolumab , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes , Mutation
5.
Bioinformatics ; 38(9): 2512-2518, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35298589

ABSTRACT

MOTIVATION: Cancers are composed by several heterogeneous subpopulations, each one harbouring different genetic and epigenetic somatic alterations that contribute to disease onset and therapy response. In recent years, copy number alterations (CNAs) leading to tumour aneuploidy have been identified as potential key drivers of such populations, but the definition of the precise makeup of cancer subclones from sequencing assays remains challenging. In the end, little is known about the mapping between complex CNAs and their effect on cancer phenotypes. RESULTS: We introduce CONGAS, a Bayesian probabilistic method to phase bulk DNA and single-cell RNA measurements from independent assays. CONGAS jointly identifies clusters of single cells with subclonal CNAs, and differences in RNA expression. The model builds statistical priors leveraging bulk DNA sequencing data, does not require a normal reference and scales fast thanks to a GPU backend and variational inference. We test CONGAS on both simulated and real data, and find that it can determine the tumour subclonal composition at the single-cell level together with clone-specific RNA phenotypes in tumour data generated from both 10× and Smart-Seq assays. AVAILABILITY AND IMPLEMENTATION: CONGAS is available as 2 packages: CONGAS (https://github.com/caravagnalab/congas), which implements the model in Python, and RCONGAS (https://caravagnalab.github.io/rcongas/), which provides R functions to process inputs, outputs and run CONGAS fits. The analysis of real data and scripts to generate figures of this paper are available via RCONGAS; code associated to simulations is available at https://github.com/caravagnalab/rcongas_test. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
DNA Copy Number Variations , Neoplasms , Humans , Bayes Theorem , Software , Sequence Analysis, RNA , RNA , Neoplasms/genetics , Single-Cell Analysis
6.
Cells ; 8(12)2019 12 01.
Article in English | MEDLINE | ID: mdl-31805750

ABSTRACT

Stage I epithelial ovarian cancer (EOC) represents about 10% of all EOCs. It is characterized by a complex histopathological and molecular heterogeneity, and it is composed of five main histological subtypes (mucinous, endometrioid, clear cell and high, and low grade serous), which have peculiar genetic, molecular, and clinical characteristics. As it occurs less frequently than advanced-stage EOC, its molecular features have not been thoroughly investigated. In this study, using in silico approaches and gene expression data, on a multicentric cohort composed of 208 snap-frozen tumor biopsies, we explored the subtype-specific molecular alterations that regulate tumor aggressiveness in stage I EOC. We found that single genes rather than pathways are responsible for histotype specificities and that a cAMP-PKA-CREB1 signaling axis seems to play a central role in histotype differentiation. Moreover, our results indicate that immune response seems to be, at least in part, involved in histotype differences, as a higher immune-reactive behavior of serous and mucinous samples was observed with respect to other histotypes.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transcription, Genetic , Biomarkers, Tumor , Carcinoma, Ovarian Epithelial/immunology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Metabolic Networks and Pathways , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...