Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cell ; 155(1): 27-38, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074859

ABSTRACT

Genomics is a relatively new scientific discipline, having DNA sequencing as its core technology. As technology has improved the cost and scale of genome characterization over sequencing's 40-year history, the scope of inquiry has commensurately broadened. Massively parallel sequencing has proven revolutionary, shifting the paradigm of genomics to address biological questions at a genome-wide scale. Sequencing now empowers clinical diagnostics and other aspects of medical care, including disease risk, therapeutic identification, and prenatal testing. This Review explores the current state of genomics in the massively parallel sequencing era.


Subject(s)
Genomics , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Animals , Disease/genetics , Genetic Variation , High-Throughput Nucleotide Sequencing/history , History, 20th Century , History, 21st Century , Humans , Mutation , Sequence Analysis, DNA/history
2.
Cell ; 150(2): 264-78, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22817890

ABSTRACT

Most mutations in cancer genomes are thought to be acquired after the initiating event, which may cause genomic instability and drive clonal evolution. However, for acute myeloid leukemia (AML), normal karyotypes are common, and genomic instability is unusual. To better understand clonal evolution in AML, we sequenced the genomes of M3-AML samples with a known initiating event (PML-RARA) versus the genomes of normal karyotype M1-AML samples and the exomes of hematopoietic stem/progenitor cells (HSPCs) from healthy people. Collectively, the data suggest that most of the mutations found in AML genomes are actually random events that occurred in HSPCs before they acquired the initiating mutation; the mutational history of that cell is "captured" as the clone expands. In many cases, only one or two additional, cooperating mutations are needed to generate the malignant founding clone. Cells from the founding clone can acquire additional cooperating mutations, yielding subclones that can contribute to disease progression and/or relapse.


Subject(s)
Clonal Evolution , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , DNA Mutational Analysis , Disease Progression , Female , Genome-Wide Association Study , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/physiopathology , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Recurrence , Skin/metabolism , Young Adult
3.
Nature ; 583(7814): 83-89, 2020 07.
Article in English | MEDLINE | ID: mdl-32460305

ABSTRACT

A key goal of whole-genome sequencing for studies of human genetics is to interrogate all forms of variation, including single-nucleotide variants, small insertion or deletion (indel) variants and structural variants. However, tools and resources for the study of structural variants have lagged behind those for smaller variants. Here we used a scalable pipeline1 to map and characterize structural variants in 17,795 deeply sequenced human genomes. We publicly release site-frequency data to create the largest, to our knowledge, whole-genome-sequencing-based structural variant resource so far. On average, individuals carry 2.9 rare structural variants that alter coding regions; these variants affect the dosage or structure of 4.2 genes and account for 4.0-11.2% of rare high-impact coding alleles. Using a computational model, we estimate that structural variants account for 17.2% of rare alleles genome-wide, with predicted deleterious effects that are equivalent to loss-of-function coding alleles; approximately 90% of such structural variants are noncoding deletions (mean 19.1 per genome). We report 158,991 ultra-rare structural variants and show that 2% of individuals carry ultra-rare megabase-scale structural variants, nearly half of which are balanced or complex rearrangements. Finally, we infer the dosage sensitivity of genes and noncoding elements, and reveal trends that relate to element class and conservation. This work will help to guide the analysis and interpretation of structural variants in the era of whole-genome sequencing.


Subject(s)
Genetic Variation , Genome, Human/genetics , Whole Genome Sequencing , Alleles , Case-Control Studies , Epigenesis, Genetic , Female , Gene Dosage/genetics , Genetics, Population , High-Throughput Nucleotide Sequencing , Humans , Male , Molecular Sequence Annotation , Quantitative Trait Loci , Racial Groups/genetics , Software
5.
Nature ; 572(7769): 323-328, 2019 08.
Article in English | MEDLINE | ID: mdl-31367044

ABSTRACT

Exome-sequencing studies have generally been underpowered to identify deleterious alleles with a large effect on complex traits as such alleles are mostly rare. Because the population of northern and eastern Finland has expanded considerably and in isolation following a series of bottlenecks, individuals of these populations have numerous deleterious alleles at a relatively high frequency. Here, using exome sequencing of nearly 20,000 individuals from these regions, we investigate the role of rare coding variants in clinically relevant quantitative cardiometabolic traits. Exome-wide association studies for 64 quantitative traits identified 26 newly associated deleterious alleles. Of these 26 alleles, 19 are either unique to or more than 20 times more frequent in Finnish individuals than in other Europeans and show geographical clustering comparable to Mendelian disease mutations that are characteristic of the Finnish population. We estimate that sequencing studies of populations without this unique history would require hundreds of thousands to millions of participants to achieve comparable association power.


Subject(s)
Exome Sequencing , Genetic Association Studies/methods , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Quantitative Trait Loci/genetics , Alleles , Cholesterol, HDL/genetics , Cluster Analysis , Endpoint Determination , Finland , Geographic Mapping , Humans , Multifactorial Inheritance/genetics , Reproducibility of Results
6.
Am J Hum Genet ; 108(4): 583-596, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33798444

ABSTRACT

The contribution of genome structural variation (SV) to quantitative traits associated with cardiometabolic diseases remains largely unknown. Here, we present the results of a study examining genetic association between SVs and cardiometabolic traits in the Finnish population. We used sensitive methods to identify and genotype 129,166 high-confidence SVs from deep whole-genome sequencing (WGS) data of 4,848 individuals. We tested the 64,572 common and low-frequency SVs for association with 116 quantitative traits and tested candidate associations using exome sequencing and array genotype data from an additional 15,205 individuals. We discovered 31 genome-wide significant associations at 15 loci, including 2 loci at which SVs have strong phenotypic effects: (1) a deletion of the ALB promoter that is greatly enriched in the Finnish population and causes decreased serum albumin level in carriers (p = 1.47 × 10-54) and is also associated with increased levels of total cholesterol (p = 1.22 × 10-28) and 14 additional cholesterol-related traits, and (2) a multi-allelic copy number variant (CNV) at PDPR that is strongly associated with pyruvate (p = 4.81 × 10-21) and alanine (p = 6.14 × 10-12) levels and resides within a structurally complex genomic region that has accumulated many rearrangements over evolutionary time. We also confirmed six previously reported associations, including five led by stronger signals in single nucleotide variants (SNVs) and one linking recurrent HP gene deletion and cholesterol levels (p = 6.24 × 10-10), which was also found to be strongly associated with increased glycoprotein level (p = 3.53 × 10-35). Our study confirms that integrating SVs in trait-mapping studies will expand our knowledge of genetic factors underlying disease risk.


Subject(s)
Cardiovascular Diseases/genetics , Genomic Structural Variation/genetics , Alleles , Cholesterol/blood , DNA Copy Number Variations/genetics , Female , Finland , Genome, Human/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Male , Mitochondrial Proteins/genetics , Promoter Regions, Genetic/genetics , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics , Pyruvic Acid/metabolism , Serum Albumin, Human/genetics
7.
Bioinformatics ; 35(22): 4782-4787, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31218349

ABSTRACT

SUMMARY: Large-scale human genetics studies are now employing whole genome sequencing with the goal of conducting comprehensive trait mapping analyses of all forms of genome variation. However, methods for structural variation (SV) analysis have lagged far behind those for smaller scale variants, and there is an urgent need to develop more efficient tools that scale to the size of human populations. Here, we present a fast and highly scalable software toolkit (svtools) and cloud-based pipeline for assembling high quality SV maps-including deletions, duplications, mobile element insertions, inversions and other rearrangements-in many thousands of human genomes. We show that this pipeline achieves similar variant detection performance to established per-sample methods (e.g. LUMPY), while providing fast and affordable joint analysis at the scale of ≥100 000 genomes. These tools will help enable the next generation of human genetics studies. AVAILABILITY AND IMPLEMENTATION: svtools is implemented in Python and freely available (MIT) from https://github.com/hall-lab/svtools. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome, Human , Software , Humans , Sequence Deletion , Whole Genome Sequencing
8.
Am J Hum Genet ; 96(3): 397-411, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25704602

ABSTRACT

Although genome-wide association studies (GWASs) for nonsyndromic orofacial clefts have identified multiple strongly associated regions, the causal variants are unknown. To address this, we selected 13 regions from GWASs and other studies, performed targeted sequencing in 1,409 Asian and European trios, and carried out a series of statistical and functional analyses. Within a cluster of strongly associated common variants near NOG, we found that one, rs227727, disrupts enhancer activity. We furthermore identified significant clusters of non-coding rare variants near NTN1 and NOG and found several rare coding variants likely to affect protein function, including four nonsense variants in ARHGAP29. We confirmed 48 de novo mutations and, based on best biological evidence available, chose two of these for functional assays. One mutation in PAX7 disrupted the DNA binding of the encoded transcription factor in an in vitro assay. The second, a non-coding mutation, disrupted the activity of a neural crest enhancer downstream of FGFR2 both in vitro and in vivo. This targeted sequencing study provides strong functional evidence implicating several specific variants as primary contributory risk alleles for nonsyndromic clefting in humans.


Subject(s)
Brain/abnormalities , Carrier Proteins/genetics , Cleft Lip/genetics , Cleft Palate/genetics , PAX7 Transcription Factor/genetics , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 2/genetics , Alleles , Amino Acid Sequence , Animals , Asian People/genetics , Carrier Proteins/metabolism , Cell Line , Epithelial Cells/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation, Missense , PAX7 Transcription Factor/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , White People/genetics , Zebrafish/genetics
9.
Nature ; 481(7382): 506-10, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22237025

ABSTRACT

Most patients with acute myeloid leukaemia (AML) die from progressive disease after relapse, which is associated with clonal evolution at the cytogenetic level. To determine the mutational spectrum associated with relapse, we sequenced the primary tumour and relapse genomes from eight AML patients, and validated hundreds of somatic mutations using deep sequencing; this allowed us to define clonality and clonal evolution patterns precisely at relapse. In addition to discovering novel, recurrently mutated genes (for example, WAC, SMC3, DIS3, DDX41 and DAXX) in AML, we also found two major clonal evolution patterns during AML relapse: (1) the founding clone in the primary tumour gained mutations and evolved into the relapse clone, or (2) a subclone of the founding clone survived initial therapy, gained additional mutations and expanded at relapse. In all cases, chemotherapy failed to eradicate the founding clone. The comparison of relapse-specific versus primary tumour mutations in all eight cases revealed an increase in transversions, probably due to DNA damage caused by cytotoxic chemotherapy. These data demonstrate that AML relapse is associated with the addition of new mutations and clonal evolution, which is shaped, in part, by the chemotherapy that the patients receive to establish and maintain remissions.


Subject(s)
Clonal Evolution/genetics , Genome, Human/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Damage/drug effects , DNA Mutational Analysis , Genes, Neoplasm/genetics , Genome, Human/drug effects , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Mutagenesis/drug effects , Mutagenesis/genetics , Recurrence , Reproducibility of Results
10.
Am J Hum Genet ; 94(3): 373-84, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560519

ABSTRACT

Exome sequencing in families affected by rare genetic disorders has the potential to rapidly identify new disease genes (genes in which mutations cause disease), but the identification of a single causal mutation among thousands of variants remains a significant challenge. We developed a scoring algorithm to prioritize potential causal variants within a family according to segregation with the phenotype, population frequency, predicted effect, and gene expression in the tissue(s) of interest. To narrow the search space in families with multiple affected individuals, we also developed two complementary approaches to exome-based mapping of autosomal-dominant disorders. One approach identifies segments of maximum identity by descent among affected individuals; the other nominates regions on the basis of shared rare variants and the absence of homozygous differences between affected individuals. We showcase our methods by using exome sequence data from families affected by autosomal-dominant retinitis pigmentosa (adRP), a rare disorder characterized by night blindness and progressive vision loss. We performed exome capture and sequencing on 91 samples representing 24 families affected by probable adRP but lacking common disease-causing mutations. Eight of 24 families (33%) were revealed to harbor high-scoring, most likely pathogenic (by clinical assessment) mutations affecting known RP genes. Analysis of the remaining 17 families identified candidate variants in a number of interesting genes, some of which have withstood further segregation testing in extended pedigrees. To empower the search for Mendelian-disease genes in family-based sequencing studies, we implemented them in a cross-platform-compatible software package, MendelScan, which is freely available to the research community.


Subject(s)
Chromosome Mapping/methods , Exome , Retinitis Pigmentosa/genetics , Cohort Studies , Computational Biology/methods , Databases, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation , Genes, Dominant , Genetic Linkage , Genetic Markers , Genetic Variation , Genotype , Heterozygote , Humans , Male , Mutation , Pedigree , Phenotype , Retina/metabolism , Software
11.
PLoS Genet ; 10(1): e1004147, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24497850

ABSTRACT

Genome-wide association studies (GWAS) have identified >500 common variants associated with quantitative metabolic traits, but in aggregate such variants explain at most 20-30% of the heritable component of population variation in these traits. To further investigate the impact of genotypic variation on metabolic traits, we conducted re-sequencing studies in >6,000 members of a Finnish population cohort (The Northern Finland Birth Cohort of 1966 [NFBC]) and a type 2 diabetes case-control sample (The Finland-United States Investigation of NIDDM Genetics [FUSION] study). By sequencing the coding sequence and 5' and 3' untranslated regions of 78 genes at 17 GWAS loci associated with one or more of six metabolic traits (serum levels of fasting HDL-C, LDL-C, total cholesterol, triglycerides, plasma glucose, and insulin), and conducting both single-variant and gene-level association tests, we obtained a more complete understanding of phenotype-genotype associations at eight of these loci. At all eight of these loci, the identification of new associations provides significant evidence for multiple genetic signals to one or more phenotypes, and at two loci, in the genes ABCA1 and CETP, we found significant gene-level evidence of association to non-synonymous variants with MAF<1%. Additionally, two potentially deleterious variants that demonstrated significant associations (rs138726309, a missense variant in G6PC2, and rs28933094, a missense variant in LIPC) were considerably more common in these Finnish samples than in European reference populations, supporting our prior hypothesis that deleterious variants could attain high frequencies in this isolated population, likely due to the effects of population bottlenecks. Our results highlight the value of large, well-phenotyped samples for rare-variant association analysis, and the challenge of evaluating the phenotypic impact of such variants.


Subject(s)
Cholesterol, HDL/genetics , Cholesterol/genetics , Genome-Wide Association Study , Quantitative Trait Loci , Cholesterol/metabolism , Cholesterol, HDL/metabolism , Finland , Genotype , High-Throughput Nucleotide Sequencing , Humans , Linkage Disequilibrium , Phenotype , Population Groups , White People
12.
N Engl J Med ; 368(22): 2059-74, 2013 05 30.
Article in English | MEDLINE | ID: mdl-23634996

ABSTRACT

BACKGROUND: Many mutations that contribute to the pathogenesis of acute myeloid leukemia (AML) are undefined. The relationships between patterns of mutations and epigenetic phenotypes are not yet clear. METHODS: We analyzed the genomes of 200 clinically annotated adult cases of de novo AML, using either whole-genome sequencing (50 cases) or whole-exome sequencing (150 cases), along with RNA and microRNA sequencing and DNA-methylation analysis. RESULTS: AML genomes have fewer mutations than most other adult cancers, with an average of only 13 mutations found in genes. Of these, an average of 5 are in genes that are recurrently mutated in AML. A total of 23 genes were significantly mutated, and another 237 were mutated in two or more samples. Nearly all samples had at least 1 nonsynonymous mutation in one of nine categories of genes that are almost certainly relevant for pathogenesis, including transcription-factor fusions (18% of cases), the gene encoding nucleophosmin (NPM1) (27%), tumor-suppressor genes (16%), DNA-methylation-related genes (44%), signaling genes (59%), chromatin-modifying genes (30%), myeloid transcription-factor genes (22%), cohesin-complex genes (13%), and spliceosome-complex genes (14%). Patterns of cooperation and mutual exclusivity suggested strong biologic relationships among several of the genes and categories. CONCLUSIONS: We identified at least one potential driver mutation in nearly all AML samples and found that a complex interplay of genetic events contributes to AML pathogenesis in individual patients. The databases from this study are widely available to serve as a foundation for further investigations of AML pathogenesis, classification, and risk stratification. (Funded by the National Institutes of Health.).


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Adult , CpG Islands , DNA Methylation , Epigenomics , Female , Gene Expression , Gene Fusion , Genome, Human , Humans , Leukemia, Myeloid, Acute/classification , Male , MicroRNAs/genetics , Middle Aged , Nucleophosmin , Sequence Analysis, DNA/methods
13.
Nat Methods ; 10(12): 1209-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24122041

ABSTRACT

The Drug-Gene Interaction database (DGIdb) mines existing resources that generate hypotheses about how mutated genes might be targeted therapeutically or prioritized for drug development. It provides an interface for searching lists of genes against a compendium of drug-gene interactions and potentially 'druggable' genes. DGIdb can be accessed at http://dgidb.org/.


Subject(s)
Data Mining/methods , Databases, Genetic , Drug Discovery/methods , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Computational Biology/methods , Drug Interactions , Gene Expression Regulation/drug effects , Genetic Variation , Genome , Genomics/methods , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Software , Technology, Pharmaceutical/methods
14.
Nature ; 464(7291): 999-1005, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20393555

ABSTRACT

Massively parallel DNA sequencing technologies provide an unprecedented ability to screen entire genomes for genetic changes associated with tumour progression. Here we describe the genomic analyses of four DNA samples from an African-American patient with basal-like breast cancer: peripheral blood, the primary tumour, a brain metastasis and a xenograft derived from the primary tumour. The metastasis contained two de novo mutations and a large deletion not present in the primary tumour, and was significantly enriched for 20 shared mutations. The xenograft retained all primary tumour mutations and displayed a mutation enrichment pattern that resembled the metastasis. Two overlapping large deletions, encompassing CTNNA1, were present in all three tumour samples. The differential mutation frequencies and structural variation patterns in metastasis and xenograft compared with the primary tumour indicate that secondary tumours may arise from a minority of cells within the primary tumour.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Genome, Human/genetics , Mutation/genetics , Neoplasm Transplantation , Adult , Breast Neoplasms/pathology , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Disease Progression , Female , Gene Frequency/genetics , Genomics , Humans , Translocation, Genetic/genetics , Transplantation, Heterologous , alpha Catenin/genetics
15.
N Engl J Med ; 366(12): 1090-8, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22417201

ABSTRACT

BACKGROUND: The myelodysplastic syndromes are a group of hematologic disorders that often evolve into secondary acute myeloid leukemia (AML). The genetic changes that underlie progression from the myelodysplastic syndromes to secondary AML are not well understood. METHODS: We performed whole-genome sequencing of seven paired samples of skin and bone marrow in seven subjects with secondary AML to identify somatic mutations specific to secondary AML. We then genotyped a bone marrow sample obtained during the antecedent myelodysplastic-syndrome stage from each subject to determine the presence or absence of the specific somatic mutations. We identified recurrent mutations in coding genes and defined the clonal architecture of each pair of samples from the myelodysplastic-syndrome stage and the secondary-AML stage, using the allele burden of hundreds of mutations. RESULTS: Approximately 85% of bone marrow cells were clonal in the myelodysplastic-syndrome and secondary-AML samples, regardless of the myeloblast count. The secondary-AML samples contained mutations in 11 recurrently mutated genes, including 4 genes that have not been previously implicated in the myelodysplastic syndromes or AML. In every case, progression to acute leukemia was defined by the persistence of an antecedent founding clone containing 182 to 660 somatic mutations and the outgrowth or emergence of at least one subclone, harboring dozens to hundreds of new mutations. All founding clones and subclones contained at least one mutation in a coding gene. CONCLUSIONS: Nearly all the bone marrow cells in patients with myelodysplastic syndromes and secondary AML are clonally derived. Genetic evolution of secondary AML is a dynamic process shaped by multiple cycles of mutation acquisition and clonal selection. Recurrent gene mutations are found in both founding clones and daughter subclones. (Funded by the National Institutes of Health and others.).


Subject(s)
Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics , Adolescent , Adult , Clone Cells , Genome, Human , Humans , Leukemia, Myeloid, Acute/etiology , Middle Aged , Myelodysplastic Syndromes/complications , Oligonucleotide Array Sequence Analysis , Skin , Young Adult
16.
Genome Res ; 22(3): 568-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22300766

ABSTRACT

Cancer is a disease driven by genetic variation and mutation. Exome sequencing can be utilized for discovering these variants and mutations across hundreds of tumors. Here we present an analysis tool, VarScan 2, for the detection of somatic mutations and copy number alterations (CNAs) in exome data from tumor-normal pairs. Unlike most current approaches, our algorithm reads data from both samples simultaneously; a heuristic and statistical algorithm detects sequence variants and classifies them by somatic status (germline, somatic, or LOH); while a comparison of normalized read depth delineates relative copy number changes. We apply these methods to the analysis of exome sequence data from 151 high-grade ovarian tumors characterized as part of the Cancer Genome Atlas (TCGA). We validated some 7790 somatic coding mutations, achieving 93% sensitivity and 85% precision for single nucleotide variant (SNV) detection. Exome-based CNA analysis identified 29 large-scale alterations and 619 focal events per tumor on average. As in our previous analysis of these data, we observed frequent amplification of oncogenes (e.g., CCNE1, MYC) and deletion of tumor suppressors (NF1, PTEN, and CDKN2A). We searched for additional recurrent focal CNAs using the correlation matrix diagonal segmentation (CMDS) algorithm, which identified 424 significant events affecting 582 genes. Taken together, our results demonstrate the robust performance of VarScan 2 for somatic mutation and CNA detection and shed new light on the landscape of genetic alterations in ovarian cancer.


Subject(s)
DNA Copy Number Variations , Exome , Mutation , Neoplasms/genetics , Software , Algorithms , Computational Biology , Female , Genomics/methods , Genotype , Germ-Line Mutation , Humans , Internet , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Reproducibility of Results , Sensitivity and Specificity
17.
JAMA ; 314(8): 811-22, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26305651

ABSTRACT

IMPORTANCE: Tests that predict outcomes for patients with acute myeloid leukemia (AML) are imprecise, especially for those with intermediate risk AML. OBJECTIVES: To determine whether genomic approaches can provide novel prognostic information for adult patients with de novo AML. DESIGN, SETTING, AND PARTICIPANTS: Whole-genome or exome sequencing was performed on samples obtained at disease presentation from 71 patients with AML (mean age, 50.8 years) treated with standard induction chemotherapy at a single site starting in March 2002, with follow-up through January 2015. In addition, deep digital sequencing was performed on paired diagnosis and remission samples from 50 patients (including 32 with intermediate-risk AML), approximately 30 days after successful induction therapy. Twenty-five of the 50 were from the cohort of 71 patients, and 25 were new, additional cases. EXPOSURES: Whole-genome or exome sequencing and targeted deep sequencing. Risk of identification based on genetic data. MAIN OUTCOMES AND MEASURES: Mutation patterns (including clearance of leukemia-associated variants after chemotherapy) and their association with event-free survival and overall survival. RESULTS: Analysis of comprehensive genomic data from the 71 patients did not improve outcome assessment over current standard-of-care metrics. In an analysis of 50 patients with both presentation and documented remission samples, 24 (48%) had persistent leukemia-associated mutations in at least 5% of bone marrow cells at remission. The 24 with persistent mutations had significantly reduced event-free and overall survival vs the 26 who cleared all mutations. Patients with intermediate cytogenetic risk profiles had similar findings. [table: see text]. CONCLUSIONS AND RELEVANCE: The detection of persistent leukemia-associated mutations in at least 5% of bone marrow cells in day 30 remission samples was associated with a significantly increased risk of relapse, and reduced overall survival. These data suggest that this genomic approach may improve risk stratification for patients with AML.


Subject(s)
Induction Chemotherapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Disease-Free Survival , Female , Genome, Human , Humans , Idarubicin/administration & dosage , Leukemia, Myeloid, Acute/mortality , Male , MicroRNAs/analysis , Middle Aged , Outcome Assessment, Health Care , Polymorphism, Genetic , Prognosis , RNA, Messenger/analysis , Recurrence , Sequence Analysis, RNA/methods
18.
Nature ; 456(7218): 66-72, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18987736

ABSTRACT

Acute myeloid leukaemia is a highly malignant haematopoietic tumour that affects about 13,000 adults in the United States each year. The treatment of this disease has changed little in the past two decades, because most of the genetic events that initiate the disease remain undiscovered. Whole-genome sequencing is now possible at a reasonable cost and timeframe to use this approach for the unbiased discovery of tumour-specific somatic mutations that alter the protein-coding genes. Here we present the results obtained from sequencing a typical acute myeloid leukaemia genome, and its matched normal counterpart obtained from the same patient's skin. We discovered ten genes with acquired mutations; two were previously described mutations that are thought to contribute to tumour progression, and eight were new mutations present in virtually all tumour cells at presentation and relapse, the function of which is not yet known. Our study establishes whole-genome sequencing as an unbiased method for discovering cancer-initiating mutations in previously unidentified genes that may respond to targeted therapies.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Leukemia, Myeloid, Acute/genetics , Case-Control Studies , Disease Progression , Gene Expression Profiling , Genomics , Humans , Mutagenesis, Insertional , Mutation , Polymorphism, Single Nucleotide , Recurrence , Sequence Analysis, DNA , Sequence Deletion , Skin/metabolism
19.
Nature ; 455(7216): 1069-75, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18948947

ABSTRACT

Determining the genetic basis of cancer requires comprehensive analyses of large collections of histopathologically well-classified primary tumours. Here we report the results of a collaborative study to discover somatic mutations in 188 human lung adenocarcinomas. DNA sequencing of 623 genes with known or potential relationships to cancer revealed more than 1,000 somatic mutations across the samples. Our analysis identified 26 genes that are mutated at significantly high frequencies and thus are probably involved in carcinogenesis. The frequently mutated genes include tyrosine kinases, among them the EGFR homologue ERBB4; multiple ephrin receptor genes, notably EPHA3; vascular endothelial growth factor receptor KDR; and NTRK genes. These data provide evidence of somatic mutations in primary lung adenocarcinoma for several tumour suppressor genes involved in other cancers--including NF1, APC, RB1 and ATM--and for sequence changes in PTPRD as well as the frequently deleted gene LRP1B. The observed mutational profiles correlate with clinical features, smoking status and DNA repair defects. These results are reinforced by data integration including single nucleotide polymorphism array and gene expression array. Our findings shed further light on several important signalling pathways involved in lung adenocarcinoma, and suggest new molecular targets for treatment.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/genetics , Lung Neoplasms/genetics , Mutation/genetics , Female , Gene Dosage , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Male , Proto-Oncogenes/genetics
20.
N Engl J Med ; 363(25): 2424-33, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21067377

ABSTRACT

BACKGROUND: The genetic alterations responsible for an adverse outcome in most patients with acute myeloid leukemia (AML) are unknown. METHODS: Using massively parallel DNA sequencing, we identified a somatic mutation in DNMT3A, encoding a DNA methyltransferase, in the genome of cells from a patient with AML with a normal karyotype. We sequenced the exons of DNMT3A in 280 additional patients with de novo AML to define recurring mutations. RESULTS: A total of 62 of 281 patients (22.1%) had mutations in DNMT3A that were predicted to affect translation. We identified 18 different missense mutations, the most common of which was predicted to affect amino acid R882 (in 37 patients). We also identified six frameshift, six nonsense, and three splice-site mutations and a 1.5-Mbp deletion encompassing DNMT3A. These mutations were highly enriched in the group of patients with an intermediate-risk cytogenetic profile (56 of 166 patients, or 33.7%) but were absent in all 79 patients with a favorable-risk cytogenetic profile (P<0.001 for both comparisons). The median overall survival among patients with DNMT3A mutations was significantly shorter than that among patients without such mutations (12.3 months vs. 41.1 months, P<0.001). DNMT3A mutations were associated with adverse outcomes among patients with an intermediate-risk cytogenetic profile or FLT3 mutations, regardless of age, and were independently associated with a poor outcome in Cox proportional-hazards analysis. CONCLUSIONS: DNMT3A mutations are highly recurrent in patients with de novo AML with an intermediate-risk cytogenetic profile and are independently associated with a poor outcome. (Funded by the National Institutes of Health and others.).


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , DNA Methylation , DNA Methyltransferase 3A , DNA Mutational Analysis/methods , Female , Frameshift Mutation , Gene Expression , Humans , Karyotyping , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Nucleic Acid Amplification Techniques , Prognosis , Proportional Hazards Models , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL