309 research outputs found

    Cold and heterogeneous T cell repertoire is associated with copy number aberrations and loss of immune genes in small-cell lung cancer

    Get PDF
    Small-cell lung cancer (SCLC) is speculated to harbor complex genomic intratumor heterogeneity (ITH) associated with high recurrence rate and suboptimal response to immunotherapy. Here, using multi-region whole exome/T cell receptor (TCR) sequencing as well as immunohistochemistry, we reveal a rather homogeneous mutational landscape but extremely cold and heterogeneous TCR repertoire in limited-stage SCLC tumors (LS-SCLCs). Compared to localized non-small cell lung cancers, LS-SCLCs have similar predicted neoantigen burden and genomic ITH, but significantly colder and more heterogeneous TCR repertoire associated with higher chromosomal copy number aberration (CNA) burden. Furthermore, copy number loss of IFN-γ pathway genes is frequently observed and positively correlates with CNA burden. Higher mutational burden, higher T cell infiltration and positive PD-L1 expression are associated with longer overall survival (OS), while higher CNA burden is associated with shorter OS in patients with LS-SCLC

    GENOMIC AND TRANSCRIPTOMIC LANDSCAPE OF COLORECTAL PREMALIGNANCY

    Get PDF
    Colorectal cancer (CRC) is the third most commonly diagnosed cancer among men and women in the United States, with 3 to 5 percent of the cases diagnosed in the background of a hereditary form of the disease. Biologically, CRC is divided into two groups: microsatellite instable (MSI) and chromosomally unstable (CIN). Genomic and transcriptomic characterization of CRC has emerged from large-scale studies in recent years due to the advancement of next-generation sequencing technologies. These studies have identified key genes and pathways altered in CRC and provided insights to the discovery of therapeutic targets. Despite the wealth of knowledge acquired in the carcinoma stage, there have been insufficient efforts to systematically characterize premalignant lesions at the molecular level, which could lead to a better understanding of neoplastic initiation, risk prediction, and the development of targeted chemoprevention strategies. The challenge in characterizing premalignancy has always been the limited availability of sample material. This challenge is tackled by getting more samples, integrating public datasets, deploying better technology that use less amount of nucleic acids and in-silico tools to extract multi-layer information from the same experiment. My genomic study consisted of whole exome sequencing (WES) and high-depth targeted sequencing on 80 premalignant lesions bulk tissue and crypts to assess clonality and mutational heterogeneity. WES results showed the presence of multiple clone in premalignancy based on clustering somatic mutation allele frequency. In addition, I determined that multiple clones originate from independent crypts harboring distinct APC and KRAS alterations. In my second study, I performed immune expression profiling and assessment of mutation and neoantigen rate of 28 premalignant lesions with DNA mismatch repair (MMR) deficient and proficient background using RNAseq. My results showed an activated immune profile despite low mutational and neoantigen rate, which challenges the canonical view in MMR-deficient carcinoma stage that immune activation is largely due to high mutation and neoantigen rate. In the last study, I performed transcriptomic sub-classifications of 398 premalignant lesions that associate them with different carcinomas subtypes, and clinical and histopathological features. My results revealed two major findings: prominent immune activation and WNT and MYC activation in premalignancy. In summary, my large-scale genomic and transcriptomic analyses of colorectal adenomas have identified key molecular characteristics in early colorectal tumorigenesis and provide a foundation for discovering novel preventive strategies

    Unraveling the clonal hierarchy of somatic genomic aberrations

    Get PDF
    Defining the chronology of molecular alterations may identify milestones in carcinogenesis. To unravel the temporal evolution of aberrations from clinical tumors, we developed CLONET, which upon estimation of tumor admixture and ploidy infers the clonal hierarchy of genomic aberrations. Comparative analysis across 100 sequenced genomes from prostate, melanoma, and lung cancers established diverse evolutionary hierarchies, demonstrating the early disruption of tumor-specific pathways. The analyses highlight the diversity of clonal evolution within and across tumor types that might be informative for risk stratification and patient selection for targeted therapies. CLONET addresses heterogeneous clinical samples seen in the setting of precision medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13059-014-0439-6) contains supplementary material, which is available to authorized users

    SuperFreq: Integrated mutation detection and clonal tracking in cancer.

    Get PDF
    Analysing multiple cancer samples from an individual patient can provide insight into the way the disease evolves. Monitoring the expansion and contraction of distinct clones helps to reveal the mutations that initiate the disease and those that drive progression. Existing approaches for clonal tracking from sequencing data typically require the user to combine multiple tools that are not purpose-built for this task. Furthermore, most methods require a matched normal (non-tumour) sample, which limits the scope of application. We developed SuperFreq, a cancer exome sequencing analysis pipeline that integrates identification of somatic single nucleotide variants (SNVs) and copy number alterations (CNAs) and clonal tracking for both. SuperFreq does not require a matched normal and instead relies on unrelated controls. When analysing multiple samples from a single patient, SuperFreq cross checks variant calls to improve clonal tracking, which helps to separate somatic from germline variants, and to resolve overlapping CNA calls. To demonstrate our software we analysed 304 cancer-normal exome samples across 33 cancer types in The Cancer Genome Atlas (TCGA) and evaluated the quality of the SNV and CNA calls. We simulated clonal evolution through in silico mixing of cancer and normal samples in known proportion. We found that SuperFreq identified 93% of clones with a cellular fraction of at least 50% and mutations were assigned to the correct clone with high recall and precision. In addition, SuperFreq maintained a similar level of performance for most aspects of the analysis when run without a matched normal. SuperFreq is highly versatile and can be applied in many different experimental settings for the analysis of exomes and other capture libraries. We demonstrate an application of SuperFreq to leukaemia patients with diagnosis and relapse samples

    Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab.

    Get PDF
    The mechanisms by which immune checkpoint blockade modulates tumor evolution during therapy are unclear. We assessed genomic changes in tumors from 68 patients with advanced melanoma, who progressed on ipilimumab or were ipilimumab-naive, before and after nivolumab initiation (CA209-038 study). Tumors were analyzed by whole-exome, transcriptome, and/or T cell receptor (TCR) sequencing. In responding patients, mutation and neoantigen load were reduced from baseline, and analysis of intratumoral heterogeneity during therapy demonstrated differential clonal evolution within tumors and putative selection against neoantigenic mutations on-therapy. Transcriptome analyses before and during nivolumab therapy revealed increases in distinct immune cell subsets, activation of specific transcriptional networks, and upregulation of immune checkpoint genes that were more pronounced in patients with response. Temporal changes in intratumoral TCR repertoire revealed expansion of T cell clones in the setting of neoantigen loss. Comprehensive genomic profiling data in this study provide insight into nivolumab\u27s mechanism of action

    The role of chromosomal instability and parallel evolution in cancer

    Get PDF
    Although chromosomal instability (CIN) is recognised as an initiating process in cancer, the extent and relevance of ongoing somatic copy number alterations (SCNAs) that result from it later in tumour development is unclear. In this thesis I describe a comprehensive analysis, including 1421 tumour samples (394 patients; 22 tumour types), to evaluate ongoing CIN and SCNAs in tumour evolution and show that intratumor heterogeneity mediated through chromosomal instability is associated with an increased risk of recurrence or death in non-small cell lung cancer (NSCLC), a finding that supports the potential value of CIN as a prognostic predictor. I also uncover pervasive SCNA intratumour heterogeneity across cancers, with recurrent clonal and subclonal events identified and found to demonstrate enrichment for cancer genes. I develop novel techniques for obtaining a phasing of heterozygous SNPs from multi-region next generation sequencing data and apply them to observe recurrent parallel evolutionary events converging upon disruption to the same genes in distinct subclones within 146 individual tumours. The most prevalent recurrent parallel loss event involved chromosome 14, including HIF1A and HIF1B. In addition, chromosome 5p, including TERT, was recurrently gained and subject to parallel evolution in 7 tumour types. Tumour type-specific constraints to early tumour development were identified in the form of obligatory clonal LOH, including LOH of 3p in clear cell renal cell carcinoma, lung squamous cell carcinoma (LUSC) and triple-negative breast cancer and LOH of 17p in LUSC, colorectal adenocarcinoma, triple negative and HER2+ breast cancer. Wholegenome doubling (WGD) was generally an early event in tumour evolution, associated with an increased acquisition of both clonal and subclonal SCNAs. For instance, CCNE1 amplifications, which occurred exclusively in WGD tumours, were subclonal in 45% of these cases, suggesting this event may be selected following a WGD event. Mathematical modelling of subclonal SCNA evolution demonstrated that models that incorporate ongoing selection with respect to SCNAs significantly outperform evolutionary neutral models, particularly in the context of WGD. This thesis highlights the importance of ongoing CIN and recurrent subclonal chromosomal alterations in tumour evolution, reveals parallel evolution of SCNAs, and sheds light on the dynamics and order of events that influence metastasis

    Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features

    Get PDF
    The mechanism by which anti-cancer immunity shapes early carcinogenesis of lung adenocarcinoma (ADC) is unknown. In this study, we characterize the immune contexture of invasive lung ADC and its precursors by transcriptomic immune profiling, T cell receptor (TCR) sequencing and multiplex immunofluorescence (mIF). Our results demonstrate that anti-tumor immunity evolved as a continuum from lung preneoplasia, to preinvasive ADC, minimally-invasive ADC and frankly invasive lung ADC with a gradually less effective and more intensively regulated immune response including down-regulation of immune-activation pathways, up-regulation of immunosuppressive pathways, lower infiltration of cytotoxic T cells (CTLs) and anti-tumor helper T cells (Th), higher infiltration of regulatory T cells (Tregs), decreased T cell clonality, and lower frequencies of top T cell clones in later-stages. Driver mutations, chromosomal copy number aberrations (CNAs) and aberrant DNA methylation may collectively impinge host immune responses and facilitate immune evasion, promoting the outgrowth of fit subclones in preneoplasia into dominant clones in invasive ADC

    Molecular Portraits of Cancer Evolution and Ecology

    Get PDF
    Research on the molecular lesions that drive cancers holds the translational promise of unmasking distinct disease subtypes in otherwise pathologically identical patients. Yet clinical adoption is hindered by the reproducibility crisis for cancer biomarkers. In this thesis, a novel metric uncovered transcriptional diversity within individual non-small cell lung cancers, driven by chromosomal instability. Existing prognostic biomarkers were confounded by tumour sampling bias, arising from this diversity, in ~50% of patients assessed. An atlas of consistently expressed genes was derived to address this diagnostic challenge, yielding a clonal biomarker robust to sampling bias. This diagnostic based on cancer evolutionary principles maintained prognostic value in a metaanalysis of >900 patients, and over known risk factors in stage I disease, motivating further development as a clinical assay. Next, in situ RNA profiles of immune, fibroblast and endothelial cell subsets were generated from cancerous and adjacent non-malignant lung tissue. The phenotypic adaptation of stromal cells in the tumour microenvironment undermined the performance of existing molecular signatures for cell-type enumeration. Transcriptome-wide analysis delineated ~10% of genes displaying cell-type-specific expression, paving the way for high-fidelity signatures for the accurate digital dissection of tumour ecology. Lastly, the impact of branching, Darwinian evolution on the detection of epistatic interactions was evaluated in a pan-cancer analysis. The clonal status of driver genes was associated with the proportion of significant epistatic findings in 44-78% of the cancer-types assessed. Integrating the clonal architecture of tumours in future analyses could help decipher evolutionary dependencies. This work provides pragmatic solutions for refining molecular portraits of cancer in the light of their evolutionary and ecological features, moving the needle for precision cancer diagnostics

    Whole-genome analysis of Nigerian patients with breast cancer reveals ethnic-driven somatic evolution and distinct genomic subtypes

    Get PDF
    Black women across the African diaspora experience more aggressive breast cancer with higher mortality rates than white women of European ancestry. Although inter-ethnic germline variation is known, differential somatic evolution has not been investigated in detail. Analysis of deep whole genomes of 97 breast cancers, with RNA-seq in a subset, from women in Nigeria in comparison with The Cancer Genome Atlas (n = 76) reveal a higher rate of genomic instability and increased intra-tumoral heterogeneity as well as a unique genomic subtype defined by early clonal GATA3 mutations with a 10.5-year younger age at diagnosis. We also find non-coding mutations in bona fide drivers (ZNF217 and SYPL1) and a previously unreported INDEL signature strongly associated with African ancestry proportion, underscoring the need to expand inclusion of diverse populations in biomedical research. Finally, we demonstrate that characterizing tumors for homologous recombination deficiency has significant clinical relevance in stratifying patients for potentially life-saving therapies

    Characterization of the genomic and immunologic diversity of malignant brain tumors through multisector analysis

    Get PDF
    Despite some success in secondary brain metastases, targeted or immune-based therapies have shown limited efficacy against primary brain malignancies such as glioblastoma (GBM). Although the intratumoral heterogeneity of GBM is implicated in treatment resistance, it remains unclear whether this diversity is observed within brain metastases and to what extent cancer cell-intrinsic heterogeneity sculpts the local immune microenvironment. Here, we profiled the immunogenomic state of 93 spatially distinct regions from 30 malignant brain tumors through whole-exome, RNA, and T-cell receptor sequencing. Our analyses identified differences between primary and secondary malignancies, with gliomas displaying more spatial heterogeneity at the genomic and neoantigen levels. In addition, this spatial diversity was recapitulated in the distribution of T-cell clones in which some gliomas harbored highly expanded but spatially restricted clonotypes. This study defines the immunogenomic landscape across a cohort of malignant brain tumors and contains implications for the design of targeted and immune-based therapies against intracranial malignancies. SIGNIFICANCE: This study describes the impact of spatial heterogeneity on genomic and immunologic characteristics of gliomas and brain metastases. The results suggest that gliomas harbor significantly greater intratumoral heterogeneity of genomic alterations, neoantigens, and T-cell clones than brain metastases, indicating the importance of multisector analysis for clinical or translational studies
    corecore