Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Leukemia ; 32(12): 2604-2616, 2018 12.
Article in English | MEDLINE | ID: mdl-29789651

ABSTRACT

In multiple myeloma, next-generation sequencing (NGS) has expanded our knowledge of genomic lesions, and highlighted a dynamic and heterogeneous composition of the tumor. Here we used NGS to characterize the genomic landscape of 418 multiple myeloma cases at diagnosis and correlate this with prognosis and classification. Translocations and copy number abnormalities (CNAs) had a preponderant contribution over gene mutations in defining the genotype and prognosis of each case. Known and novel independent prognostic markers were identified in our cohort of proteasome inhibitor and immunomodulatory drug-treated patients with long follow-up, including events with context-specific prognostic value, such as deletions of the PRDM1 gene. Taking advantage of the comprehensive genomic annotation of each case, we used innovative statistical approaches to identify potential novel myeloma subgroups. We observed clusters of patients stratified based on the overall number of mutations and number/type of CNAs, with distinct effects on survival, suggesting that extended genotype of multiple myeloma at diagnosis may lead to improved disease classification and prognostication.


Subject(s)
Biomarkers, Tumor/genetics , Multiple Myeloma/genetics , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Multiple Myeloma/pathology , Mutation/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Prognosis , Translocation, Genetic/genetics
2.
Cancer Cell ; 32(2): 169-184.e7, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28810143

ABSTRACT

Patterns of genomic evolution between primary and metastatic breast cancer have not been studied in large numbers, despite patients with metastatic breast cancer having dismal survival. We sequenced whole genomes or a panel of 365 genes on 299 samples from 170 patients with locally relapsed or metastatic breast cancer. Several lines of analysis indicate that clones seeding metastasis or relapse disseminate late from primary tumors, but continue to acquire mutations, mostly accessing the same mutational processes active in the primary tumor. Most distant metastases acquired driver mutations not seen in the primary tumor, drawing from a wider repertoire of cancer genes than early drivers. These include a number of clinically actionable alterations and mutations inactivating SWI-SNF and JAK2-STAT3 pathways.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Evolution, Molecular , Mutation , Neoplasm Recurrence, Local/genetics , Adult , Aged , Aged, 80 and over , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Female , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Male , Middle Aged , Neoplasm Metastasis/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
3.
N Engl J Med ; 374(23): 2209-2221, 2016 Jun 09.
Article in English | MEDLINE | ID: mdl-27276561

ABSTRACT

BACKGROUND: Recent studies have provided a detailed census of genes that are mutated in acute myeloid leukemia (AML). Our next challenge is to understand how this genetic diversity defines the pathophysiology of AML and informs clinical practice. METHODS: We enrolled a total of 1540 patients in three prospective trials of intensive therapy. Combining driver mutations in 111 cancer genes with cytogenetic and clinical data, we defined AML genomic subgroups and their relevance to clinical outcomes. RESULTS: We identified 5234 driver mutations across 76 genes or genomic regions, with 2 or more drivers identified in 86% of the patients. Patterns of co-mutation compartmentalized the cohort into 11 classes, each with distinct diagnostic features and clinical outcomes. In addition to currently defined AML subgroups, three heterogeneous genomic categories emerged: AML with mutations in genes encoding chromatin, RNA-splicing regulators, or both (in 18% of patients); AML with TP53 mutations, chromosomal aneuploidies, or both (in 13%); and, provisionally, AML with IDH2(R172) mutations (in 1%). Patients with chromatin-spliceosome and TP53-aneuploidy AML had poor outcomes, with the various class-defining mutations contributing independently and additively to the outcome. In addition to class-defining lesions, other co-occurring driver mutations also had a substantial effect on overall survival. The prognostic effects of individual mutations were often significantly altered by the presence or absence of other driver mutations. Such gene-gene interactions were especially pronounced for NPM1-mutated AML, in which patterns of co-mutation identified groups with a favorable or adverse prognosis. These predictions require validation in prospective clinical trials. CONCLUSIONS: The driver landscape in AML reveals distinct molecular subgroups that reflect discrete paths in the evolution of AML, informing disease classification and prognostic stratification. (Funded by the Wellcome Trust and others; ClinicalTrials.gov number, NCT00146120.).


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Adult , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA Mutational Analysis , Epistasis, Genetic , Gene Fusion , Genotype , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Middle Aged , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Proportional Hazards Models , Prospective Studies , RNA Splicing , Survival Analysis
4.
Nat Med ; 21(7): 751-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26099045

ABSTRACT

The sequencing of cancer genomes may enable tailoring of therapeutics to the underlying biological abnormalities driving a particular patient's tumor. However, sequencing-based strategies rely heavily on representative sampling of tumors. To understand the subclonal structure of primary breast cancer, we applied whole-genome and targeted sequencing to multiple samples from each of 50 patients' tumors (303 samples in total). The extent of subclonal diversification varied among cases and followed spatial patterns. No strict temporal order was evident, with point mutations and rearrangements affecting the most common breast cancer genes, including PIK3CA, TP53, PTEN, BRCA2 and MYC, occurring early in some tumors and late in others. In 13 out of 50 cancers, potentially targetable mutations were subclonal. Landmarks of disease progression, such as resistance to chemotherapy and the acquisition of invasive or metastatic potential, arose within detectable subclones of antecedent lesions. These findings highlight the importance of including analyses of subclonal structure and tumor evolution in clinical trials of primary breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Proliferation , Clone Cells , Cohort Studies , DNA Copy Number Variations/genetics , Female , Genomics , Humans , Middle Aged , Mutation/genetics
6.
J Pathol ; 236(4): 457-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25850943

ABSTRACT

Multifocal breast cancer (MFBC), defined as multiple synchronous unilateral lesions of invasive breast cancer, is relatively frequent and has been associated with more aggressive features than unifocal cancer. Here, we aimed to investigate the genomic heterogeneity between MFBC lesions sharing similar histopathological parameters. Characterization of different lesions from 36 patients with ductal MFBC involved the identification of non-silent coding mutations in 360 protein-coding genes (171 tumour and 36 matched normal samples). We selected only patients with lesions presenting the same grade, ER, and HER2 status. Mutations were classified as 'oncogenic' in the case of recurrent substitutions reported in COSMIC or truncating mutations affecting tumour suppressor genes. All mutations identified in a given patient were further interrogated in all samples from that patient through deep resequencing using an orthogonal platform. Whole-genome rearrangement screen was further conducted in 8/36 patients. Twenty-four patients (67%) had substitutions/indels shared by all their lesions, of which 11 carried the same mutations in all lesions, and 13 had lesions with both common and private mutations. Three-quarters of those 24 patients shared oncogenic variants. The remaining 12 patients (33%) did not share any substitution/indels, with inter-lesion heterogeneity observed for oncogenic mutation(s) in genes such as PIK3CA, TP53, GATA3, and PTEN. Genomically heterogeneous lesions tended to be further apart in the mammary gland than homogeneous lesions. Genome-wide analyses of a limited number of patients identified a common somatic background in all studied MFBCs, including those with no mutation in common between the lesions. To conclude, as the number of molecular targeted therapies increases and trials driven by genomic screening are ongoing, our findings highlight the presence of genomic inter-lesion heterogeneity in one-third, despite similar pathological features. This implies that deeper molecular characterization of all MFBC lesions is warranted for the adequate management of those cancers.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Mutation , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Intraductal, Noninfiltrating/chemistry , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Neoplasm Grading , Neoplasms, Multiple Primary/chemistry , Phenotype , Predictive Value of Tests , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Retrospective Studies
7.
Nat Genet ; 47(4): 367-372, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25730763

ABSTRACT

Genome-wide DNA sequencing was used to decrypt the phylogeny of multiple samples from distinct areas of cancer and morphologically normal tissue taken from the prostates of three men. Mutations were present at high levels in morphologically normal tissue distant from the cancer, reflecting clonal expansions, and the underlying mutational processes at work in morphologically normal tissue were also at work in cancer. Our observations demonstrate the existence of ongoing abnormal mutational processes, consistent with field effects, underlying carcinogenesis. This mechanism gives rise to extensive branching evolution and cancer clone mixing, as exemplified by the coexistence of multiple cancer lineages harboring distinct ERG fusions within a single cancer nodule. Subsets of mutations were shared either by morphologically normal and malignant tissues or between different ERG lineages, indicating earlier or separate clonal cell expansions. Our observations inform on the origin of multifocal disease and have implications for prostate cancer therapy in individual cases.


Subject(s)
Clonal Evolution/genetics , DNA Mutational Analysis , Neoplasms, Multiple Primary/genetics , Prostate/cytology , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Case-Control Studies , Cell Lineage/genetics , Clone Cells/pathology , Humans , Male , Mutation , Phylogeny
8.
Science ; 345(6196): 1251343, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25082706

ABSTRACT

Long interspersed nuclear element-1 (L1) retrotransposons are mobile repetitive elements that are abundant in the human genome. L1 elements propagate through RNA intermediates. In the germ line, neighboring, nonrepetitive sequences are occasionally mobilized by the L1 machinery, a process called 3' transduction. Because 3' transductions are potentially mutagenic, we explored the extent to which they occur somatically during tumorigenesis. Studying cancer genomes from 244 patients, we found that tumors from 53% of the patients had somatic retrotranspositions, of which 24% were 3' transductions. Fingerprinting of donor L1s revealed that a handful of source L1 elements in a tumor can spawn from tens to hundreds of 3' transductions, which can themselves seed further retrotranspositions. The activity of individual L1 elements fluctuated during tumor evolution and correlated with L1 promoter hypomethylation. The 3' transductions disseminated genes, exons, and regulatory elements to new locations, most often to heterochromatic regions of the genome.


Subject(s)
DNA Transposable Elements , Long Interspersed Nucleotide Elements , Neoplasms/genetics , Transduction, Genetic , Carcinogenesis/genetics , Chromatin/chemistry , Exons , Genome, Human , Humans , Mutagenesis, Insertional , Translocation, Genetic
9.
Nature ; 513(7518): 422-425, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25043003

ABSTRACT

The somatic mutations present in the genome of a cell accumulate over the lifetime of a multicellular organism. These mutations can provide insights into the developmental lineage tree, the number of divisions that each cell has undergone and the mutational processes that have been operative. Here we describe whole genomes of clonal lines derived from multiple tissues of healthy mice. Using somatic base substitutions, we reconstructed the early cell divisions of each animal, demonstrating the contributions of embryonic cells to adult tissues. Differences were observed between tissues in the numbers and types of mutations accumulated by each cell, which likely reflect differences in the number of cell divisions they have undergone and varying contributions of different mutational processes. If somatic mutation rates are similar to those in mice, the results indicate that precise insights into development and mutagenesis of normal human cells will be possible.


Subject(s)
Cell Lineage/genetics , Clone Cells/cytology , Clone Cells/metabolism , Genome/genetics , Mutagenesis/genetics , Mutation/genetics , Animals , Biological Clocks/genetics , Cell Division , Cells, Cultured , Embryo, Mammalian/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mutation Rate , Organoids/cytology , Organoids/metabolism , Phylogeny , Sequence Analysis, DNA , Tail/cytology
10.
Nat Commun ; 5: 3644, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24714652

ABSTRACT

Cancer evolves by mutation, with somatic reactivation of retrotransposons being one such mutational process. Germline retrotransposition can cause processed pseudogenes, but whether this occurs somatically has not been evaluated. Here we screen sequencing data from 660 cancer samples for somatically acquired pseudogenes. We find 42 events in 17 samples, especially non-small cell lung cancer (5/27) and colorectal cancer (2/11). Genomic features mirror those of germline LINE element retrotranspositions, with frequent target-site duplications (67%), consensus TTTTAA sites at insertion points, inverted rearrangements (21%), 5' truncation (74%) and polyA tails (88%). Transcriptional consequences include expression of pseudogenes from UTRs or introns of target genes. In addition, a somatic pseudogene that integrated into the promoter and first exon of the tumour suppressor gene, MGA, abrogated expression from that allele. Thus, formation of processed pseudogenes represents a new class of mutation occurring during cancer development, with potentially diverse functional consequences depending on genomic context.


Subject(s)
Neoplasms/genetics , Pseudogenes/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Pseudogenes/physiology
11.
Nat Genet ; 46(4): 376-379, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24633157

ABSTRACT

Angiosarcoma is an aggressive malignancy that arises spontaneously or secondarily to ionizing radiation or chronic lymphoedema. Previous work has identified aberrant angiogenesis, including occasional somatic mutations in angiogenesis signaling genes, as a key driver of angiosarcoma. Here we employed whole-genome, whole-exome and targeted sequencing to study the somatic changes underpinning primary and secondary angiosarcoma. We identified recurrent mutations in two genes, PTPRB and PLCG1, which are intimately linked to angiogenesis. The endothelial phosphatase PTPRB, a negative regulator of vascular growth factor tyrosine kinases, harbored predominantly truncating mutations in 10 of 39 tumors (26%). PLCG1, a signal transducer of tyrosine kinases, encoded a recurrent, likely activating p.Arg707Gln missense variant in 3 of 34 cases (9%). Overall, 15 of 39 tumors (38%) harbored at least one driver mutation in angiogenesis signaling genes. Our findings inform and reinforce current therapeutic efforts to target angiogenesis signaling in angiosarcoma.


Subject(s)
Hemangiosarcoma/drug therapy , Hemangiosarcoma/genetics , Neovascularization, Pathologic/drug therapy , Phospholipase C gamma/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Analysis of Variance , Base Sequence , Exome/genetics , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , Mutation/genetics , Neovascularization, Pathologic/genetics , RNA Interference , Sequence Analysis, RNA , Vascular Endothelial Growth Factor A/antagonists & inhibitors
12.
Nat Commun ; 5: 2997, 2014.
Article in English | MEDLINE | ID: mdl-24429703

ABSTRACT

Multiple myeloma is an incurable plasma cell malignancy with a complex and incompletely understood molecular pathogenesis. Here we use whole-exome sequencing, copy-number profiling and cytogenetics to analyse 84 myeloma samples. Most cases have a complex subclonal structure and show clusters of subclonal variants, including subclonal driver mutations. Serial sampling reveals diverse patterns of clonal evolution, including linear evolution, differential clonal response and branching evolution. Diverse processes contribute to the mutational repertoire, including kataegis and somatic hypermutation, and their relative contribution changes over time. We find heterogeneity of mutational spectrum across samples, with few recurrent genes. We identify new candidate genes, including truncations of SP140, LTB, ROBO1 and clustered missense mutations in EGR1. The myeloma genome is heterogeneous across the cohort, and exhibits diversity in clonal admixture and in dynamics of evolution, which may impact prognostic stratification, therapeutic approaches and assessment of disease response to treatment.


Subject(s)
Exome , Multiple Myeloma/genetics , Adult , Aged , Antigens, Nuclear , Cohort Studies , DNA Copy Number Variations , Early Growth Response Protein 1 , Evolution, Molecular , GTP Phosphohydrolases , Genetic Heterogeneity , Humans , Lymphotoxin-beta , Membrane Proteins , Middle Aged , Mutation , Mutation, Missense , Nerve Tissue Proteins , Proto-Oncogene Proteins , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins p21(ras) , Receptors, Immunologic , Sequence Analysis, DNA , Transcription Factors , Tumor Suppressor Protein p53 , ras Proteins , Roundabout Proteins
13.
Nat Genet ; 46(2): 116-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24413735

ABSTRACT

The ETV6-RUNX1 fusion gene, found in 25% of childhood acute lymphoblastic leukemia (ALL) cases, is acquired in utero but requires additional somatic mutations for overt leukemia. We used exome and low-coverage whole-genome sequencing to characterize secondary events associated with leukemic transformation. RAG-mediated deletions emerge as the dominant mutational process, characterized by recombination signal sequence motifs near breakpoints, incorporation of non-templated sequence at junctions, ∼30-fold enrichment at promoters and enhancers of genes actively transcribed in B cell development and an unexpectedly high ratio of recurrent to non-recurrent structural variants. Single-cell tracking shows that this mechanism is active throughout leukemic evolution, with evidence of localized clustering and reiterated deletions. Integration of data on point mutations and rearrangements identifies ATF7IP and MGA as two new tumor-suppressor genes in ALL. Thus, a remarkably parsimonious mutational process transforms ETV6-RUNX1-positive lymphoblasts, targeting the promoters, enhancers and first exons of genes that normally regulate B cell differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Rearrangement/genetics , Genetic Variation , Homeodomain Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombination, Genetic/genetics , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Copy Number Variations/genetics , Gene Library , Genes, Tumor Suppressor , Humans , Molecular Sequence Data , Repressor Proteins , Sequence Analysis, DNA , Sequence Deletion/genetics , Transcription Factors/genetics , V(D)J Recombination/genetics
14.
Genome Biol ; 14(10): R113, 2013.
Article in English | MEDLINE | ID: mdl-24148783

ABSTRACT

BACKGROUND: Melanoma is the most deadly form of skin cancer. Expression of oncogenic BRAF or NRAS, which are frequently mutated in human melanomas, promote the formation of nevi but are not sufficient for tumorigenesis. Even with germline mutated p53, these engineered melanomas present with variable onset and pathology, implicating additional somatic mutations in a multi-hit tumorigenic process. RESULTS: To decipher the genetics of these melanomas, we sequence the protein coding exons of 53 primary melanomas generated from several BRAF(V600E) or NRAS(Q61K) driven transgenic zebrafish lines. We find that engineered zebrafish melanomas show an overall low mutation burden, which has a strong, inverse association with the number of initiating germline drivers. Although tumors reveal distinct mutation spectrums, they show mostly C > T transitions without UV light exposure, and enrichment of mutations in melanogenesis, p53 and MAPK signaling. Importantly, a recurrent amplification occurring with pre-configured drivers BRAF(V600E) and p53-/- suggests a novel path of BRAF cooperativity through the protein kinase A pathway. CONCLUSION: This is the first analysis of a melanoma mutational landscape in the absence of UV light, where tumors manifest with remarkably low mutation burden and high heterogeneity. Genotype specific amplification of protein kinase A in cooperation with BRAF and p53 mutation suggests the involvement of melanogenesis in these tumors. This work is important for defining the spectrum of events in BRAF or NRAS driven melanoma in the absence of UV light, and for informed exploitation of models such as transgenic zebrafish to better understand mechanisms leading to human melanoma formation.


Subject(s)
Genetic Heterogeneity , Melanoma/genetics , Mutation , Zebrafish/genetics , Animals , Animals, Genetically Modified , DNA Copy Number Variations , Disease Models, Animal , Gene Amplification , Gene Knockout Techniques , Homozygote , INDEL Mutation , Melanoma/pathology , Mutation/radiation effects , Polymorphism, Single Nucleotide , Risk Factors , Sequence Deletion , Ultraviolet Rays
15.
Blood ; 122(22): 3616-27; quiz 3699, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24030381

ABSTRACT

Myelodysplastic syndromes (MDS) are a heterogeneous group of chronic hematological malignancies characterized by dysplasia, ineffective hematopoiesis and a variable risk of progression to acute myeloid leukemia. Sequencing of MDS genomes has identified mutations in genes implicated in RNA splicing, DNA modification, chromatin regulation, and cell signaling. We sequenced 111 genes across 738 patients with MDS or closely related neoplasms (including chronic myelomonocytic leukemia and MDS-myeloproliferative neoplasms) to explore the role of acquired mutations in MDS biology and clinical phenotype. Seventy-eight percent of patients had 1 or more oncogenic mutations. We identify complex patterns of pairwise association between genes, indicative of epistatic interactions involving components of the spliceosome machinery and epigenetic modifiers. Coupled with inferences on subclonal mutations, these data suggest a hypothesis of genetic "predestination," in which early driver mutations, typically affecting genes involved in RNA splicing, dictate future trajectories of disease evolution with distinct clinical phenotypes. Driver mutations had equivalent prognostic significance, whether clonal or subclonal, and leukemia-free survival deteriorated steadily as numbers of driver mutations increased. Thus, analysis of oncogenic mutations in large, well-characterized cohorts of patients illustrates the interconnections between the cancer genome and disease biology, with considerable potential for clinical application.


Subject(s)
Mutation , Myelodysplastic Syndromes/genetics , Aged , Aged, 80 and over , Cohort Studies , Disease Progression , Epistasis, Genetic , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Male , Middle Aged , Myelodysplastic-Myeloproliferative Diseases/genetics , Oncogenes , Prognosis , RNA Splicing/genetics , Spliceosomes/genetics
16.
J Clin Invest ; 123(7): 2965-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778141

ABSTRACT

Adenoid cystic carcinoma (ACC) is a rare malignancy that can occur in multiple organ sites and is primarily found in the salivary gland. While the identification of recurrent fusions of the MYB-NFIB genes have begun to shed light on the molecular underpinnings, little else is known about the molecular genetics of this frequently fatal cancer. We have undertaken exome sequencing in a series of 24 ACC to further delineate the genetics of the disease. We identified multiple mutated genes that, combined, implicate chromatin deregulation in half of cases. Further, mutations were identified in known cancer genes, including PIK3CA, ATM, CDKN2A, SF3B1, SUFU, TSC1, and CYLD. Mutations in NOTCH1/2 were identified in 3 cases, and we identify the negative NOTCH signaling regulator, SPEN, as a new cancer gene in ACC with mutations in 5 cases. Finally, the identification of 3 likely activating mutations in the tyrosine kinase receptor FGFR2, analogous to those reported in ovarian and endometrial carcinoma, point to potential therapeutic avenues for a subset of cases.


Subject(s)
Carcinoma, Adenoid Cystic/genetics , Exome , Salivary Gland Neoplasms/genetics , DNA Mutational Analysis , Genes, Neoplasm , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Mutation , Polymorphism, Single Nucleotide
17.
Nat Genet ; 45(8): 923-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23770606

ABSTRACT

Chondrosarcoma is a heterogeneous collection of malignant bone tumors and is the second most common primary malignancy of bone after osteosarcoma. Recent work has identified frequent, recurrent mutations in IDH1 or IDH2 in nearly half of central chondrosarcomas. However, there has been little systematic genomic analysis of this tumor type, and, thus, the contribution of other genes is unclear. Here we report comprehensive genomic analyses of 49 individuals with chondrosarcoma (cases). We identified hypermutability of the major cartilage collagen gene COL2A1, with insertions, deletions and rearrangements identified in 37% of cases. The patterns of mutation were consistent with selection for variants likely to impair normal collagen biosynthesis. In addition, we identified mutations in IDH1 or IDH2 (59%), TP53 (20%), the RB1 pathway (33%) and Hedgehog signaling (18%).


Subject(s)
Bone Neoplasms/genetics , Chondrosarcoma/genetics , Collagen Type II/genetics , Mutation , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Collagen Type II/metabolism , Computational Biology , DNA Copy Number Variations , Databases, Genetic , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Neoplasm Grading , Polymorphism, Single Nucleotide , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction
18.
Nature ; 486(7403): 400-4, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22722201

ABSTRACT

All cancers carry somatic mutations in their genomes. A subset, known as driver mutations, confer clonal selective advantage on cancer cells and are causally implicated in oncogenesis, and the remainder are passenger mutations. The driver mutations and mutational processes operative in breast cancer have not yet been comprehensively explored. Here we examine the genomes of 100 tumours for somatic copy number changes and mutations in the coding exons of protein-coding genes. The number of somatic mutations varied markedly between individual tumours. We found strong correlations between mutation number, age at which cancer was diagnosed and cancer histological grade, and observed multiple mutational signatures, including one present in about ten per cent of tumours characterized by numerous mutations of cytosine at TpC dinucleotides. Driver mutations were identified in several new cancer genes including AKT2, ARID1B, CASP8, CDKN1B, MAP3K1, MAP3K13, NCOR1, SMARCD1 and TBX3. Among the 100 tumours, we found driver mutations in at least 40 cancer genes and 73 different combinations of mutated cancer genes. The results highlight the substantial genetic diversity underlying this common disease.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Mutagenesis/genetics , Mutation/genetics , Oncogenes/genetics , Age Factors , Breast Neoplasms/classification , Breast Neoplasms/pathology , Cytosine/metabolism , DNA Mutational Analysis , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasm Grading , Reproducibility of Results , Signal Transduction/genetics
19.
Cell ; 149(5): 994-1007, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608083

ABSTRACT

Cancer evolves dynamically as clonal expansions supersede one another driven by shifting selective pressures, mutational processes, and disrupted cancer genes. These processes mark the genome, such that a cancer's life history is encrypted in the somatic mutations present. We developed algorithms to decipher this narrative and applied them to 21 breast cancers. Mutational processes evolve across a cancer's lifespan, with many emerging late but contributing extensive genetic variation. Subclonal diversification is prominent, and most mutations are found in just a fraction of tumor cells. Every tumor has a dominant subclonal lineage, representing more than 50% of tumor cells. Minimal expansion of these subclones occurs until many hundreds to thousands of mutations have accumulated, implying the existence of long-lived, quiescent cell lineages capable of substantial proliferation upon acquisition of enabling genomic changes. Expansion of the dominant subclone to an appreciable mass may therefore represent the final rate-limiting step in a breast cancer's development, triggering diagnosis.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Clonal Evolution , Mutation , Algorithms , Chromosome Aberrations , Female , Humans , Point Mutation
20.
Cell ; 149(5): 979-93, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608084

ABSTRACT

All cancers carry somatic mutations. The patterns of mutation in cancer genomes reflect the DNA damage and repair processes to which cancer cells and their precursors have been exposed. To explore these mechanisms further, we generated catalogs of somatic mutation from 21 breast cancers and applied mathematical methods to extract mutational signatures of the underlying processes. Multiple distinct single- and double-nucleotide substitution signatures were discernible. Cancers with BRCA1 or BRCA2 mutations exhibited a characteristic combination of substitution mutation signatures and a distinctive profile of deletions. Complex relationships between somatic mutation prevalence and transcription were detected. A remarkable phenomenon of localized hypermutation, termed "kataegis," was observed. Regions of kataegis differed between cancers but usually colocalized with somatic rearrangements. Base substitutions in these regions were almost exclusively of cytosine at TpC dinucleotides. The mechanisms underlying most of these mutational signatures are unknown. However, a role for the APOBEC family of cytidine deaminases is proposed.


Subject(s)
Breast Neoplasms/genetics , DNA Mutational Analysis , Genome-Wide Association Study , Mutation , APOBEC-1 Deaminase , BRCA2 Protein/genetics , Cytidine Deaminase/metabolism , Female , Genes, BRCA1 , High-Throughput Nucleotide Sequencing , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...