Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
2.
N Engl J Med ; 376(22): 2109-2121, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28445112

ABSTRACT

BACKGROUND: Among patients with non-small-cell lung cancer (NSCLC), data on intratumor heterogeneity and cancer genome evolution have been limited to small retrospective cohorts. We wanted to prospectively investigate intratumor heterogeneity in relation to clinical outcome and to determine the clonal nature of driver events and evolutionary processes in early-stage NSCLC. METHODS: In this prospective cohort study, we performed multiregion whole-exome sequencing on 100 early-stage NSCLC tumors that had been resected before systemic therapy. We sequenced and analyzed 327 tumor regions to define evolutionary histories, obtain a census of clonal and subclonal events, and assess the relationship between intratumor heterogeneity and recurrence-free survival. RESULTS: We observed widespread intratumor heterogeneity for both somatic copy-number alterations and mutations. Driver mutations in EGFR, MET, BRAF, and TP53 were almost always clonal. However, heterogeneous driver alterations that occurred later in evolution were found in more than 75% of the tumors and were common in PIK3CA and NF1 and in genes that are involved in chromatin modification and DNA damage response and repair. Genome doubling and ongoing dynamic chromosomal instability were associated with intratumor heterogeneity and resulted in parallel evolution of driver somatic copy-number alterations, including amplifications in CDK4, FOXA1, and BCL11A. Elevated copy-number heterogeneity was associated with an increased risk of recurrence or death (hazard ratio, 4.9; P=4.4×10-4), which remained significant in multivariate analysis. CONCLUSIONS: Intratumor heterogeneity mediated through chromosome instability was associated with an increased risk of recurrence or death, a finding that supports the potential value of chromosome instability as a prognostic predictor. (Funded by Cancer Research UK and others; TRACERx ClinicalTrials.gov number, NCT01888601 .).


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chromosomal Instability , Genetic Heterogeneity , Lung Neoplasms/genetics , Mutation , Neoplasm Recurrence, Local/genetics , Carcinoma, Non-Small-Cell Lung/mortality , DNA Copy Number Variations , Disease-Free Survival , Evolution, Molecular , Exome , Female , Humans , Lung Neoplasms/mortality , Male , Phylogeny , Prognosis , Prospective Studies , Risk Factors , Sequence Analysis, DNA/methods
3.
Nature ; 545(7655): 446-451, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28445469

ABSTRACT

The early detection of relapse following primary surgery for non-small-cell lung cancer and the characterization of emerging subclones, which seed metastatic sites, might offer new therapeutic approaches for limiting tumour recurrence. The ability to track the evolutionary dynamics of early-stage lung cancer non-invasively in circulating tumour DNA (ctDNA) has not yet been demonstrated. Here we use a tumour-specific phylogenetic approach to profile the ctDNA of the first 100 TRACERx (Tracking Non-Small-Cell Lung Cancer Evolution Through Therapy (Rx)) study participants, including one patient who was also recruited to the PEACE (Posthumous Evaluation of Advanced Cancer Environment) post-mortem study. We identify independent predictors of ctDNA release and analyse the tumour-volume detection limit. Through blinded profiling of postoperative plasma, we observe evidence of adjuvant chemotherapy resistance and identify patients who are very likely to experience recurrence of their lung cancer. Finally, we show that phylogenetic ctDNA profiling tracks the subclonal nature of lung cancer relapse and metastasis, providing a new approach for ctDNA-driven therapeutic studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Lineage/genetics , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Evolution, Molecular , Lung Neoplasms/genetics , Neoplasm Metastasis/diagnosis , Neoplasm Recurrence, Local/diagnosis , Biopsy/methods , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cell Tracking , Clone Cells/metabolism , Clone Cells/pathology , DNA Mutational Analysis , Disease Progression , Drug Resistance, Neoplasm/genetics , Early Detection of Cancer/methods , Humans , Limit of Detection , Lung Neoplasms/blood , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Multiplex Polymerase Chain Reaction , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Postoperative Care/methods , Reproducibility of Results , Tumor Burden
4.
Science ; 351(6280): 1463-9, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26940869

ABSTRACT

As tumors grow, they acquire mutations, some of which create neoantigens that influence the response of patients to immune checkpoint inhibitors. We explored the impact of neoantigen intratumor heterogeneity (ITH) on antitumor immunity. Through integrated analysis of ITH and neoantigen burden, we demonstrate a relationship between clonal neoantigen burden and overall survival in primary lung adenocarcinomas. CD8(+)tumor-infiltrating lymphocytes reactive to clonal neoantigens were identified in early-stage non-small cell lung cancer and expressed high levels of PD-1. Sensitivity to PD-1 and CTLA-4 blockade in patients with advanced NSCLC and melanoma was enhanced in tumors enriched for clonal neoantigens. T cells recognizing clonal neoantigens were detectable in patients with durable clinical benefit. Cytotoxic chemotherapy-induced subclonal neoantigens, contributing to an increased mutational load, were enriched in certain poor responders. These data suggest that neoantigen heterogeneity may influence immune surveillance and support therapeutic developments targeting clonal neoantigens.


Subject(s)
Adenocarcinoma/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Surveillance , Lung Neoplasms/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Cycle Checkpoints/immunology , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/immunology , Middle Aged , Mutation , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology
5.
BMC Med Genomics ; 8: 58, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26429708

ABSTRACT

BACKGROUND: A substantial proportion of cancer cases present with a metastatic tumor and require further testing to determine the primary site; many of these are never fully diagnosed and remain cancer of unknown primary origin (CUP). It has been previously demonstrated that the somatic point mutations detected in a tumor can be used to identify its site of origin with limited accuracy. We hypothesized that higher accuracy could be achieved by a classification algorithm based on the following feature sets: 1) the number of nonsynonymous point mutations in a set of 232 specific cancer-associated genes, 2) frequencies of the 96 classes of single-nucleotide substitution determined by the flanking bases, and 3) copy number profiles, if available. METHODS: We used publicly available somatic mutation data from the COSMIC database to train random forest classifiers to distinguish among those tissues of origin for which sufficient data was available. We selected feature sets using cross-validation and then derived two final classifiers (with or without copy number profiles) using 80 % of the available tumors. We evaluated the accuracy using the remaining 20 %. For further validation, we assessed accuracy of the without-copy-number classifier on three independent data sets: 1669 newly available public tumors of various types, a cohort of 91 breast metastases, and a set of 24 specimens from 9 lung cancer patients subjected to multiregion sequencing. RESULTS: The cross-validation accuracy was highest when all three types of information were used. On the left-out COSMIC data not used for training, we achieved a classification accuracy of 85 % across 6 primary sites (with copy numbers), and 69 % across 10 primary sites (without copy numbers). Importantly, a derived confidence score could distinguish tumors that could be identified with 95 % accuracy (32 %/75 % of tumors with/without copy numbers) from those that were less certain. Accuracy in the independent data sets was 46 %, 53 % and 89 % respectively, similar to the accuracy expected from the training data. CONCLUSIONS: Identification of primary site from point mutation and/or copy number data may be accurate enough to aid clinical diagnosis of cancers of unknown primary origin.


Subject(s)
Breast Neoplasms/genetics , Databases, Genetic , Genes, Neoplasm , Lung Neoplasms/genetics , Point Mutation , Polymorphism, Single Nucleotide , Female , Humans , Male , Organ Specificity
6.
Science ; 346(6206): 251-6, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25301630

ABSTRACT

Spatial and temporal dissection of the genomic changes occurring during the evolution of human non-small cell lung cancer (NSCLC) may help elucidate the basis for its dismal prognosis. We sequenced 25 spatially distinct regions from seven operable NSCLCs and found evidence of branched evolution, with driver mutations arising before and after subclonal diversification. There was pronounced intratumor heterogeneity in copy number alterations, translocations, and mutations associated with APOBEC cytidine deaminase activity. Despite maintained carcinogen exposure, tumors from smokers showed a relative decrease in smoking-related mutations over time, accompanied by an increase in APOBEC-associated mutations. In tumors from former smokers, genome-doubling occurred within a smoking-signature context before subclonal diversification, which suggested that a long period of tumor latency had preceded clinical detection. The regionally separated driver mutations, coupled with the relentless and heterogeneous nature of the genome instability processes, are likely to confound treatment success in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Genetic Heterogeneity , Genomic Instability , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , APOBEC-1 Deaminase , Carcinogens/toxicity , Carcinoma, Non-Small-Cell Lung/chemically induced , Cytidine Deaminase/genetics , Evolution, Molecular , Gene Dosage , Humans , Lung Neoplasms/chemically induced , Mutation , Neoplasm Recurrence, Local/genetics , Prognosis , Smoking/adverse effects , Translocation, Genetic , Tumor Cells, Cultured
7.
PLoS Biol ; 12(7): e1001906, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25003521

ABSTRACT

The importance of intratumour genetic and functional heterogeneity is increasingly recognised as a driver of cancer progression and survival outcome. Understanding how tumour clonal heterogeneity impacts upon therapeutic outcome, however, is still an area of unmet clinical and scientific need. TRACERx (TRAcking non-small cell lung Cancer Evolution through therapy [Rx]), a prospective study of patients with primary non-small cell lung cancer (NSCLC), aims to define the evolutionary trajectories of lung cancer in both space and time through multiregion and longitudinal tumour sampling and genetic analysis. By following cancers from diagnosis to relapse, tracking the evolutionary trajectories of tumours in relation to therapeutic interventions, and determining the impact of clonal heterogeneity on clinical outcomes, TRACERx may help to identify novel therapeutic targets for NSCLC and may also serve as a model applicable to other cancer types.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Disease Progression , Lung Neoplasms/genetics , Antigens, Neoplasm , Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm , Humans , Longitudinal Studies , Neoplasm Metastasis , Treatment Outcome
8.
Sci Transl Med ; 3(113): 113ra124, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22133594

ABSTRACT

DNA damage or other physicochemical stresses may increase the expression of major histocompatibility complex class I-related stress antigens, which then activate lymphocytes. This lymphoid stress surveillance (LSS) not only can limit tumor formation but may also promote immunopathology. MICA is a highly polymorphic human stress antigen implicated in tumor surveillance, inflammation, and transplant rejection. However, LSS has not been conclusively demonstrated in humans, and the functional role for MICA polymorphisms remains to be established. We show that MICA coding sequence polymorphisms substantially affected RNA and protein expression. All donors tested showed LSS responses of γδ T and natural killer cells, but unexpectedly, each was individually "tuned." Hence, some responded optimally to highly expressed alleles, whereas others responded better to lower MICA expression, challenging the orthodoxy that higher stress antigen levels promote greater responsiveness. These individual variations in LSS tuning may help explain patient-specific differences in tumor immune surveillance, transplant rejection, and inflammation, as well as provide insight into immune evasion and immunosuppression.


Subject(s)
Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Immunologic Surveillance/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Polymorphism, Genetic , Stress, Physiological/immunology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL