Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
PLoS Comput Biol ; 19(10): e1011379, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37871126

ABSTRACT

Most computational methods that infer somatic copy number alterations (SCNAs) from bulk sequencing of DNA analyse tumour samples individually. However, the sequencing of multiple tumour samples from a patient's disease is an increasingly common practice. We introduce Refphase, an algorithm that leverages this multi-sampling approach to infer haplotype-specific copy numbers through multi-sample phasing. We demonstrate Refphase's ability to infer haplotype-specific SCNAs and characterise their intra-tumour heterogeneity, to uncover previously undetected allelic imbalance in low purity samples, and to identify parallel evolution in the context of whole genome doubling in a pan-cancer cohort of 336 samples from 99 tumours.


Subject(s)
DNA Copy Number Variations , Neoplasms , Humans , DNA Copy Number Variations/genetics , Haplotypes/genetics , Neoplasms/genetics , Neoplasms/pathology , Algorithms
2.
Nature ; 616(7957): 553-562, 2023 04.
Article in English | MEDLINE | ID: mdl-37055640

ABSTRACT

Circulating tumour DNA (ctDNA) can be used to detect and profile residual tumour cells persisting after curative intent therapy1. The study of large patient cohorts incorporating longitudinal plasma sampling and extended follow-up is required to determine the role of ctDNA as a phylogenetic biomarker of relapse in early-stage non-small-cell lung cancer (NSCLC). Here we developed ctDNA methods tracking a median of 200 mutations identified in resected NSCLC tissue across 1,069 plasma samples collected from 197 patients enrolled in the TRACERx study2. A lack of preoperative ctDNA detection distinguished biologically indolent lung adenocarcinoma with good clinical outcome. Postoperative plasma analyses were interpreted within the context of standard-of-care radiological surveillance and administration of cytotoxic adjuvant therapy. Landmark analyses of plasma samples collected within 120 days after surgery revealed ctDNA detection in 25% of patients, including 49% of all patients who experienced clinical relapse; 3 to 6 monthly ctDNA surveillance identified impending disease relapse in an additional 20% of landmark-negative patients. We developed a bioinformatic tool (ECLIPSE) for non-invasive tracking of subclonal architecture at low ctDNA levels. ECLIPSE identified patients with polyclonal metastatic dissemination, which was associated with a poor clinical outcome. By measuring subclone cancer cell fractions in preoperative plasma, we found that subclones seeding future metastases were significantly more expanded compared with non-metastatic subclones. Our findings will support (neo)adjuvant trial advances and provide insights into the process of metastatic dissemination using low-ctDNA-level liquid biopsy.


Subject(s)
Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung , Circulating Tumor DNA , Lung Neoplasms , Mutation , Neoplasm Metastasis , Small Cell Lung Carcinoma , Humans , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Cohort Studies , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Phylogeny , Small Cell Lung Carcinoma/pathology , Liquid Biopsy
3.
Nat Med ; 29(4): 833-845, 2023 04.
Article in English | MEDLINE | ID: mdl-37045996

ABSTRACT

Lung adenocarcinomas (LUADs) display a broad histological spectrum from low-grade lepidic tumors through to mid-grade acinar and papillary and high-grade solid, cribriform and micropapillary tumors. How morphology reflects tumor evolution and disease progression is poorly understood. Whole-exome sequencing data generated from 805 primary tumor regions and 121 paired metastatic samples across 248 LUADs from the TRACERx 421 cohort, together with RNA-sequencing data from 463 primary tumor regions, were integrated with detailed whole-tumor and regional histopathological analysis. Tumors with predominantly high-grade patterns showed increased chromosomal complexity, with higher burden of loss of heterozygosity and subclonal somatic copy number alterations. Individual regions in predominantly high-grade pattern tumors exhibited higher proliferation and lower clonal diversity, potentially reflecting large recent subclonal expansions. Co-occurrence of truncal loss of chromosomes 3p and 3q was enriched in predominantly low-/mid-grade tumors, while purely undifferentiated solid-pattern tumors had a higher frequency of truncal arm or focal 3q gains and SMARCA4 gene alterations compared with mixed-pattern tumors with a solid component, suggesting distinct evolutionary trajectories. Clonal evolution analysis revealed that tumors tend to evolve toward higher-grade patterns. The presence of micropapillary pattern and 'tumor spread through air spaces' were associated with intrathoracic recurrence, in contrast to the presence of solid/cribriform patterns, necrosis and preoperative circulating tumor DNA detection, which were associated with extra-thoracic recurrence. These data provide insights into the relationship between LUAD morphology, the underlying evolutionary genomic landscape, and clinical and anatomical relapse risk.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Neoplasm Recurrence, Local/pathology , Adenocarcinoma of Lung/genetics , Disease Progression , DNA Helicases , Nuclear Proteins , Transcription Factors
4.
Nature ; 616(7957): 543-552, 2023 04.
Article in English | MEDLINE | ID: mdl-37046093

ABSTRACT

Intratumour heterogeneity (ITH) fuels lung cancer evolution, which leads to immune evasion and resistance to therapy1. Here, using paired whole-exome and RNA sequencing data, we investigate intratumour transcriptomic diversity in 354 non-small cell lung cancer tumours from 347 out of the first 421 patients prospectively recruited into the TRACERx study2,3. Analyses of 947 tumour regions, representing both primary and metastatic disease, alongside 96 tumour-adjacent normal tissue samples implicate the transcriptome as a major source of phenotypic variation. Gene expression levels and ITH relate to patterns of positive and negative selection during tumour evolution. We observe frequent copy number-independent allele-specific expression that is linked to epigenomic dysfunction. Allele-specific expression can also result in genomic-transcriptomic parallel evolution, which converges on cancer gene disruption. We extract signatures of RNA single-base substitutions and link their aetiology to the activity of the RNA-editing enzymes ADAR and APOBEC3A, thereby revealing otherwise undetected ongoing APOBEC activity in tumours. Characterizing the transcriptomes of primary-metastatic tumour pairs, we combine multiple machine-learning approaches that leverage genomic and transcriptomic variables to link metastasis-seeding potential to the evolutionary context of mutations and increased proliferation within primary tumour regions. These results highlight the interplay between the genome and transcriptome in influencing ITH, lung cancer evolution and metastasis.


Subject(s)
Evolution, Molecular , Genome, Human , Lung Neoplasms , Neoplasm Metastasis , Transcriptome , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Genomics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Neoplasm Metastasis/genetics , Transcriptome/genetics , Alleles , Machine Learning , Genome, Human/genetics
5.
Nature ; 616(7957): 534-542, 2023 04.
Article in English | MEDLINE | ID: mdl-37046095

ABSTRACT

Metastatic disease is responsible for the majority of cancer-related deaths1. We report the longitudinal evolutionary analysis of 126 non-small cell lung cancer (NSCLC) tumours from 421 prospectively recruited patients in TRACERx who developed metastatic disease, compared with a control cohort of 144 non-metastatic tumours. In 25% of cases, metastases diverged early, before the last clonal sweep in the primary tumour, and early divergence was enriched for patients who were smokers at the time of initial diagnosis. Simulations suggested that early metastatic divergence more frequently occurred at smaller tumour diameters (less than 8 mm). Single-region primary tumour sampling resulted in 83% of late divergence cases being misclassified as early, highlighting the importance of extensive primary tumour sampling. Polyclonal dissemination, which was associated with extrathoracic disease recurrence, was found in 32% of cases. Primary lymph node disease contributed to metastatic relapse in less than 20% of cases, representing a hallmark of metastatic potential rather than a route to subsequent recurrences/disease progression. Metastasis-seeding subclones exhibited subclonal expansions within primary tumours, probably reflecting positive selection. Our findings highlight the importance of selection in metastatic clone evolution within untreated primary tumours, the distinction between monoclonal versus polyclonal seeding in dictating site of recurrence, the limitations of current radiological screening approaches for early diverging tumours and the need to develop strategies to target metastasis-seeding subclones before relapse.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Clonal Evolution , Clone Cells , Evolution, Molecular , Lung Neoplasms , Neoplasm Metastasis , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Clone Cells/pathology , Cohort Studies , Disease Progression , Lung Neoplasms/pathology , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local
6.
Nature ; 616(7957): 525-533, 2023 04.
Article in English | MEDLINE | ID: mdl-37046096

ABSTRACT

Lung cancer is the leading cause of cancer-associated mortality worldwide1. Here we analysed 1,644 tumour regions sampled at surgery or during follow-up from the first 421 patients with non-small cell lung cancer prospectively enrolled into the TRACERx study. This project aims to decipher lung cancer evolution and address the primary study endpoint: determining the relationship between intratumour heterogeneity and clinical outcome. In lung adenocarcinoma, mutations in 22 out of 40 common cancer genes were under significant subclonal selection, including classical tumour initiators such as TP53 and KRAS. We defined evolutionary dependencies between drivers, mutational processes and whole genome doubling (WGD) events. Despite patients having a history of smoking, 8% of lung adenocarcinomas lacked evidence of tobacco-induced mutagenesis. These tumours also had similar detection rates for EGFR mutations and for RET, ROS1, ALK and MET oncogenic isoforms compared with tumours in never-smokers, which suggests that they have a similar aetiology and pathogenesis. Large subclonal expansions were associated with positive subclonal selection. Patients with tumours harbouring recent subclonal expansions, on the terminus of a phylogenetic branch, had significantly shorter disease-free survival. Subclonal WGD was detected in 19% of tumours, and 10% of tumours harboured multiple subclonal WGDs in parallel. Subclonal, but not truncal, WGD was associated with shorter disease-free survival. Copy number heterogeneity was associated with extrathoracic relapse within 1 year after surgery. These data demonstrate the importance of clonal expansion, WGD and copy number instability in determining the timing and patterns of relapse in non-small cell lung cancer and provide a comprehensive clinical cancer evolutionary data resource.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Adenocarcinoma of Lung/etiology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Neoplasm Recurrence, Local/genetics , Phylogeny , Treatment Outcome , Smoking/genetics , Smoking/physiopathology , Mutagenesis , DNA Copy Number Variations
7.
Nature ; 616(7957): 563-573, 2023 04.
Article in English | MEDLINE | ID: mdl-37046094

ABSTRACT

B cells are frequently found in the margins of solid tumours as organized follicles in ectopic lymphoid organs called tertiary lymphoid structures (TLS)1,2. Although TLS have been found to correlate with improved patient survival and response to immune checkpoint blockade (ICB), the underlying mechanisms of this association remain elusive1,2. Here we investigate lung-resident B cell responses in patients from the TRACERx 421 (Tracking Non-Small-Cell Lung Cancer Evolution Through Therapy) and other lung cancer cohorts, and in a recently established immunogenic mouse model for lung adenocarcinoma3. We find that both human and mouse lung adenocarcinomas elicit local germinal centre responses and tumour-binding antibodies, and further identify endogenous retrovirus (ERV) envelope glycoproteins as a dominant anti-tumour antibody target. ERV-targeting B cell responses are amplified by ICB in both humans and mice, and by targeted inhibition of KRAS(G12C) in the mouse model. ERV-reactive antibodies exert anti-tumour activity that extends survival in the mouse model, and ERV expression predicts the outcome of ICB in human lung adenocarcinoma. Finally, we find that effective immunotherapy in the mouse model requires CXCL13-dependent TLS formation. Conversely, therapeutic CXCL13 treatment potentiates anti-tumour immunity and synergizes with ICB. Our findings provide a possible mechanistic basis for the association of TLS with immunotherapy response.


Subject(s)
Endogenous Retroviruses , Immunotherapy , Lung Neoplasms , Animals , Humans , Mice , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/therapy , Adenocarcinoma of Lung/virology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/virology , Disease Models, Animal , Endogenous Retroviruses/immunology , Immunotherapy/methods , Lung/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/virology , Tumor Microenvironment , B-Lymphocytes/immunology , Cohort Studies , Antibodies/immunology , Antibodies/therapeutic use
8.
Cell ; 186(8): 1541-1563, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059064

ABSTRACT

Recent identification of oncogenic cells within healthy tissues and the prevalence of indolent cancers found incidentally at autopsies reveal a greater complexity in tumor initiation than previously appreciated. The human body contains roughly 40 trillion cells of 200 different types that are organized within a complex three-dimensional matrix, necessitating exquisite mechanisms to restrain aberrant outgrowth of malignant cells that have the capacity to kill the host. Understanding how this defense is overcome to trigger tumorigenesis and why cancer is so extraordinarily rare at the cellular level is vital to future prevention therapies. In this review, we discuss how early initiated cells are protected from further tumorigenesis and the non-mutagenic pathways by which cancer risk factors promote tumor growth. By nature, the absence of permanent genomic alterations potentially renders these tumor-promoting mechanisms clinically targetable. Finally, we consider existing strategies for early cancer interception with perspectives on the next steps for molecular cancer prevention.


Subject(s)
Neoplasms , Humans , Carcinogenesis , Cell Transformation, Neoplastic , Genomics/methods , Neoplasms/epidemiology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Risk Factors
9.
Nature ; 616(7955): 159-167, 2023 04.
Article in English | MEDLINE | ID: mdl-37020004

ABSTRACT

A complete understanding of how exposure to environmental substances promotes cancer formation is lacking. More than 70 years ago, tumorigenesis was proposed to occur in a two-step process: an initiating step that induces mutations in healthy cells, followed by a promoter step that triggers cancer development1. Here we propose that environmental particulate matter measuring ≤2.5 µm (PM2.5), known to be associated with lung cancer risk, promotes lung cancer by acting on cells that harbour pre-existing oncogenic mutations in healthy lung tissue. Focusing on EGFR-driven lung cancer, which is more common in never-smokers or light smokers, we found a significant association between PM2.5 levels and the incidence of lung cancer for 32,957 EGFR-driven lung cancer cases in four within-country cohorts. Functional mouse models revealed that air pollutants cause an influx of macrophages into the lung and release of interleukin-1ß. This process results in a progenitor-like cell state within EGFR mutant lung alveolar type II epithelial cells that fuels tumorigenesis. Ultradeep mutational profiling of histologically normal lung tissue from 295 individuals across 3 clinical cohorts revealed oncogenic EGFR and KRAS driver mutations in 18% and 53% of healthy tissue samples, respectively. These findings collectively support a tumour-promoting role for  PM2.5 air pollutants  and provide impetus for public health policy initiatives to address air pollution to reduce disease burden.


Subject(s)
Adenocarcinoma of Lung , Air Pollutants , Air Pollution , Cell Transformation, Neoplastic , Lung Neoplasms , Animals , Mice , Adenocarcinoma of Lung/chemically induced , Adenocarcinoma of Lung/genetics , Air Pollutants/adverse effects , Air Pollutants/analysis , Air Pollution/adverse effects , Air Pollution/analysis , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Environmental Exposure , ErbB Receptors/genetics , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Particulate Matter/adverse effects , Particulate Matter/analysis , Particle Size , Cohort Studies , Macrophages, Alveolar/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology
10.
Nature ; 597(7877): 555-560, 2021 09.
Article in English | MEDLINE | ID: mdl-34497419

ABSTRACT

The immune microenvironment influences tumour evolution and can be both prognostic and predict response to immunotherapy1,2. However, measurements of tumour infiltrating lymphocytes (TILs) are limited by a shortage of appropriate data. Whole-exome sequencing (WES) of DNA is frequently performed to calculate tumour mutational burden and identify actionable mutations. Here we develop T cell exome TREC tool (T cell ExTRECT), a method for estimation of T cell fraction from WES samples using a signal from T cell receptor excision circle (TREC) loss during V(D)J recombination of the T cell receptor-α gene (TCRA (also known as TRA)). TCRA T cell fraction correlates with orthogonal TIL estimates and is agnostic to sample type. Blood TCRA T cell fraction is higher in females than in males and correlates with both tumour immune infiltrate and presence of bacterial sequencing reads. Tumour TCRA T cell fraction is prognostic in lung adenocarcinoma. Using a meta-analysis of tumours treated with immunotherapy, we show that tumour TCRA T cell fraction predicts immunotherapy response, providing value beyond measuring tumour mutational burden. Applying T cell ExTRECT to a multi-sample pan-cancer cohort reveals a high diversity of the degree of immune infiltration within tumours. Subclonal loss of 12q24.31-32, encompassing SPPL3, is associated with reduced TCRA T cell fraction. T cell ExTRECT provides a cost-effective technique to characterize immune infiltrate alongside somatic changes.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/therapy , Aspartic Acid Endopeptidases/genetics , Cohort Studies , Exome/genetics , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Prognosis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Exome Sequencing/economics
11.
Cancer Discov ; 11(10): 2456-2473, 2021 10.
Article in English | MEDLINE | ID: mdl-33947663

ABSTRACT

APOBEC3 enzymes are cytosine deaminases implicated in cancer. Precisely when APOBEC3 expression is induced during cancer development remains to be defined. Here we show that specific APOBEC3 genes are upregulated in breast ductal carcinoma in situ, and in preinvasive lung cancer lesions coincident with cellular proliferation. We observe evidence of APOBEC3-mediated subclonal mutagenesis propagated from TRACERx preinvasive to invasive non-small cell lung cancer (NSCLC) lesions. We find that APOBEC3B exacerbates DNA replication stress and chromosomal instability through incomplete replication of genomic DNA, manifested by accumulation of mitotic ultrafine bridges and 53BP1 nuclear bodies in the G1 phase of the cell cycle. Analysis of TRACERx NSCLC clinical samples and mouse lung cancer models revealed APOBEC3B expression driving replication stress and chromosome missegregation. We propose that APOBEC3 is functionally implicated in the onset of chromosomal instability and somatic mutational heterogeneity in preinvasive disease, providing fuel for selection early in cancer evolution. SIGNIFICANCE: This study reveals the dynamics and drivers of APOBEC3 gene expression in preinvasive disease and the exacerbation of cellular diversity by APOBEC3B through DNA replication stress to promote chromosomal instability early in cancer evolution.This article is highlighted in the In This Issue feature, p. 2355.


Subject(s)
APOBEC Deaminases/genetics , Breast Neoplasms/genetics , Carcinoma, Ductal/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosomal Instability , DNA Replication , Female , Humans , Mice
12.
Cell ; 184(3): 596-614.e14, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508232

ABSTRACT

Checkpoint inhibitors (CPIs) augment adaptive immunity. Systematic pan-tumor analyses may reveal the relative importance of tumor-cell-intrinsic and microenvironmental features underpinning CPI sensitization. Here, we collated whole-exome and transcriptomic data for >1,000 CPI-treated patients across seven tumor types, utilizing standardized bioinformatics workflows and clinical outcome criteria to validate multivariable predictors of CPI sensitization. Clonal tumor mutation burden (TMB) was the strongest predictor of CPI response, followed by total TMB and CXCL9 expression. Subclonal TMB, somatic copy alteration burden, and histocompatibility leukocyte antigen (HLA) evolutionary divergence failed to attain pan-cancer significance. Dinucleotide variants were identified as a source of immunogenic epitopes associated with radical amino acid substitutions and enhanced peptide hydrophobicity/immunogenicity. Copy-number analysis revealed two additional determinants of CPI outcome supported by prior functional evidence: 9q34 (TRAF2) loss associated with response and CCND1 amplification associated with resistance. Finally, single-cell RNA sequencing (RNA-seq) of clonal neoantigen-reactive CD8 tumor-infiltrating lymphocytes (TILs), combined with bulk RNA-seq analysis of CPI-responding tumors, identified CCR5 and CXCL13 as T-cell-intrinsic markers of CPI sensitivity.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Neoplasms/immunology , T-Lymphocytes/immunology , Biomarkers, Tumor/metabolism , CD8 Antigens/metabolism , Chemokine CXCL13/metabolism , Chromosomes, Human, Pair 9/genetics , Cohort Studies , Cyclin D1/genetics , DNA Copy Number Variations/genetics , Exome/genetics , Gene Amplification , Humans , Immune Evasion/drug effects , Multivariate Analysis , Mutation/genetics , Neoplasms/pathology , Polymorphism, Single Nucleotide/genetics , Receptors, CCR5/metabolism , T-Lymphocytes/drug effects , Tumor Burden/genetics
14.
Nature ; 587(7832): 126-132, 2020 11.
Article in English | MEDLINE | ID: mdl-32879494

ABSTRACT

Chromosomal instability in cancer consists of dynamic changes to the number and structure of chromosomes1,2. The resulting diversity in somatic copy number alterations (SCNAs) may provide the variation necessary for tumour evolution1,3,4. Here we use multi-sample phasing and SCNA analysis of 1,421 samples from 394 tumours across 22 tumour types to show that continuous chromosomal instability results in pervasive SCNA heterogeneity. Parallel evolutionary events, which cause disruption in the same genes (such as BCL9, MCL1, ARNT (also known as HIF1B), TERT and MYC) within separate subclones, were present in 37% of tumours. Most recurrent losses probably occurred before whole-genome doubling, that was found as a clonal event in 49% of tumours. However, loss of heterozygosity at the human leukocyte antigen (HLA) locus and loss of chromosome 8p to a single haploid copy recurred at substantial subclonal frequencies, even in tumours with whole-genome doubling, indicating ongoing karyotype remodelling. Focal amplifications that affected chromosomes 1q21 (which encompasses BCL9, MCL1 and ARNT), 5p15.33 (TERT), 11q13.3 (CCND1), 19q12 (CCNE1) and 8q24.1 (MYC) were frequently subclonal yet appeared to be clonal within single samples. Analysis of an independent series of 1,024 metastatic samples revealed that 13 focal SCNAs were enriched in metastatic samples, including gains in chromosome 8q24.1 (encompassing MYC) in clear cell renal cell carcinoma and chromosome 11q13.3 (encompassing CCND1) in HER2+ breast cancer. Chromosomal instability may enable the continuous selection of SCNAs, which are established as ordered events that often occur in parallel, throughout tumour evolution.


Subject(s)
Chromosomal Instability/genetics , Evolution, Molecular , Karyotype , Neoplasm Metastasis/genetics , Neoplasms/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 8/genetics , Clone Cells/metabolism , Clone Cells/pathology , Cyclin E/genetics , DNA Copy Number Variations/genetics , Female , Humans , Loss of Heterozygosity/genetics , Male , Mutagenesis , Neoplasm Metastasis/pathology , Neoplasms/pathology , Oncogene Proteins/genetics
15.
Nat Commun ; 11(1): 3800, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32733040

ABSTRACT

Frameshift insertion/deletions (fs-indels) are an infrequent but highly immunogenic mutation subtype. Although fs-indels are degraded through the nonsense-mediated decay (NMD) pathway, we hypothesise that some fs-indels escape degradation and elicit anti-tumor immune responses. Using allele-specific expression analysis, expressed fs-indels are enriched in genomic positions predicted to escape NMD, and associated with higher protein expression, consistent with degradation escape (NMD-escape). Across four independent melanoma cohorts, NMD-escape mutations are significantly associated with clinical-benefit to checkpoint inhibitor (CPI) therapy (Pmeta = 0.0039). NMD-escape mutations are additionally found to associate with clinical-benefit in the low-TMB setting. Furthermore, in an adoptive cell therapy treated melanoma cohort, NMD-escape mutation count is the most significant biomarker associated with clinical-benefit. Analysis of functional T cell reactivity screens from personalized vaccine studies shows direct evidence of fs-indel derived neoantigens eliciting immune response, particularly those with highly elongated neo open reading frames. NMD-escape fs-indels represent an attractive target for biomarker optimisation and immunotherapy design.


Subject(s)
Melanoma/genetics , Melanoma/immunology , Nonsense Mediated mRNA Decay/genetics , T-Lymphocytes/immunology , Tumor Escape/genetics , Adoptive Transfer , Antigens, Neoplasm/immunology , Biomarkers, Tumor/genetics , Frameshift Mutation/genetics , Humans , INDEL Mutation/genetics , Immunotherapy, Adoptive , T-Lymphocytes/transplantation , Exome Sequencing
16.
Nature ; 583(7818): 807-812, 2020 07.
Article in English | MEDLINE | ID: mdl-32669708

ABSTRACT

The majority of targeted therapies for non-small-cell lung cancer (NSCLC) are directed against oncogenic drivers that are more prevalent in patients with light exposure to tobacco smoke1-3. As this group represents around 20% of all patients with lung cancer, the discovery of stratified medicine options for tobacco-associated NSCLC is a high priority. Umbrella trials seek to streamline the investigation of genotype-based treatments by screening tumours for multiple genomic alterations and triaging patients to one of several genotype-matched therapeutic agents. Here we report the current outcomes of 19 drug-biomarker cohorts from the ongoing National Lung Matrix Trial, the largest umbrella trial in NSCLC. We use next-generation sequencing to match patients to appropriate targeted therapies on the basis of their tumour genotype. The Bayesian trial design enables outcome data from open cohorts that are still recruiting to be reported alongside data from closed cohorts. Of the 5,467 patients that were screened, 2,007 were molecularly eligible for entry into the trial, and 302 entered the trial to receive genotype-matched therapy-including 14 that re-registered to the trial for a sequential trial drug. Despite pre-clinical data supporting the drug-biomarker combinations, current evidence shows that a limited number of combinations demonstrate clinically relevant benefits, which remain concentrated in patients with lung cancers that are associated with minimal exposure to tobacco smoke.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Markers , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Molecular Targeted Therapy , Precision Medicine , Smoking/genetics , Bayes Theorem , Carcinoma, Non-Small-Cell Lung/etiology , Clinical Protocols , Clinical Trials as Topic , Cohort Studies , Genotype , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/etiology , Oncogenes/genetics , Patient Selection , Smoke/adverse effects , Triage
18.
Cell Rep ; 31(5): 107550, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32375028

ABSTRACT

Although thousands of solid tumors have been sequenced to date, a fundamental under-sampling bias is inherent in current methodologies. This is caused by a tissue sample input of fixed dimensions (e.g., 6 mm biopsy), which becomes grossly under-powered as tumor volume scales. Here, we demonstrate representative sequencing (Rep-Seq) as a new method to achieve unbiased tumor tissue sampling. Rep-Seq uses fixed residual tumor material, which is homogenized and subjected to next-generation sequencing. Analysis of intratumor tumor mutation burden (TMB) variability shows a high level of misclassification using current single-biopsy methods, with 20% of lung and 52% of bladder tumors having at least one biopsy with high TMB but low clonal TMB overall. Misclassification rates by contrast are reduced to 2% (lung) and 4% (bladder) when a more representative sampling methodology is used. Rep-Seq offers an improved sampling protocol for tumor profiling, with significant potential for improved clinical utility and more accurate deconvolution of clonal structure.


Subject(s)
Biomarkers, Tumor/genetics , High-Throughput Nucleotide Sequencing , Lung Neoplasms/genetics , Tumor Burden/genetics , Urinary Bladder Neoplasms/genetics , Biopsy/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Lung Neoplasms/pathology , Mutation/genetics , Urinary Bladder Neoplasms/pathology
19.
Nat Genet ; 52(3): 283-293, 2020 03.
Article in English | MEDLINE | ID: mdl-32139907

ABSTRACT

Whole-genome doubling (WGD) is a prevalent event in cancer, involving a doubling of the entire chromosome complement. However, despite its prevalence and prognostic relevance, the evolutionary selection pressures for WGD in cancer have not been investigated. Here, we combine evolutionary simulations with an analysis of cancer sequencing data to explore WGD during cancer evolution. Simulations suggest that WGD can be selected to mitigate the irreversible, ratchet-like, accumulation of deleterious somatic alterations, provided that they occur at a sufficiently high rate. Consistent with this, we observe an enrichment for WGD in tumor types with extensive loss of heterozygosity, including lung squamous cell carcinoma and triple-negative breast cancers, and we find evidence for negative selection against homozygous loss of essential genes before, but not after, WGD. Finally, we demonstrate that loss of heterozygosity and temporal dissection of mutations can be exploited to identify novel tumor suppressor genes and to obtain a deeper characterization of known cancer genes.


Subject(s)
Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Gene Duplication , Genome, Human/genetics , Lung Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Cohort Studies , Computer Simulation , DNA Copy Number Variations , Evolution, Molecular , Humans , Longitudinal Studies , Loss of Heterozygosity , Mutation , Prospective Studies
20.
Nat Med ; 25(10): 1540-1548, 2019 10.
Article in English | MEDLINE | ID: mdl-31591602

ABSTRACT

An aim of molecular biomarkers is to stratify patients with cancer into disease subtypes predictive of outcome, improving diagnostic precision beyond clinical descriptors such as tumor stage1. Transcriptomic intratumor heterogeneity (RNA-ITH) has been shown to confound existing expression-based biomarkers across multiple cancer types2-6. Here, we analyze multi-region whole-exome and RNA sequencing data for 156 tumor regions from 48 patients enrolled in the TRACERx study to explore and control for RNA-ITH in non-small cell lung cancer. We find that chromosomal instability is a major driver of RNA-ITH, and existing prognostic gene expression signatures are vulnerable to tumor sampling bias. To address this, we identify genes expressed homogeneously within individual tumors that encode expression modules of cancer cell proliferation and are often driven by DNA copy-number gains selected early in tumor evolution. Clonal transcriptomic biomarkers overcome tumor sampling bias, associate with survival independent of clinicopathological risk factors, and may provide a general strategy to refine biomarker design across cancer types.


Subject(s)
Clonal Evolution/genetics , Lung Neoplasms/genetics , Prognosis , Transcriptome/genetics , Aged , Biomarkers, Tumor/genetics , DNA Copy Number Variations/genetics , Disease-Free Survival , Exome/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Risk Factors , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...