Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters











Publication year range
2.
Nat Commun ; 7: 11589, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27199251

ABSTRACT

Resistance to the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has been attributed solely to mutations in BTK and related pathway molecules. Using whole-exome and deep-targeted sequencing, we dissect evolution of ibrutinib resistance in serial samples from five chronic lymphocytic leukaemia patients. In two patients, we detect BTK-C481S mutation or multiple PLCG2 mutations. The other three patients exhibit an expansion of clones harbouring del(8p) with additional driver mutations (EP300, MLL2 and EIF2A), with one patient developing trans-differentiation into CD19-negative histiocytic sarcoma. Using droplet-microfluidic technology and growth kinetic analyses, we demonstrate the presence of ibrutinib-resistant subclones and estimate subclone size before treatment initiation. Haploinsufficiency of TRAIL-R, a consequence of del(8p), results in TRAIL insensitivity, which may contribute to ibrutinib resistance. These findings demonstrate that the ibrutinib therapy favours selection and expansion of rare subclones already present before ibrutinib treatment, and provide insight into the heterogeneity of genetic changes associated with ibrutinib resistance.


Subject(s)
Clonal Evolution , Drug Resistance, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Neoplasm Recurrence, Local/genetics , Protein-Tyrosine Kinases/genetics , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Adult , Agammaglobulinaemia Tyrosine Kinase , Aged, 80 and over , Apoptosis , Cell Transdifferentiation , Female , Histiocytic Sarcoma/etiology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Male , Middle Aged , Mutation , Piperidines , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Selection, Genetic
3.
Nature ; 526(7574): 525-30, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26466571

ABSTRACT

Which genetic alterations drive tumorigenesis and how they evolve over the course of disease and therapy are central questions in cancer biology. Here we identify 44 recurrently mutated genes and 11 recurrent somatic copy number variations through whole-exome sequencing of 538 chronic lymphocytic leukaemia (CLL) and matched germline DNA samples, 278 of which were collected in a prospective clinical trial. These include previously unrecognized putative cancer drivers (RPS15, IKZF3), and collectively identify RNA processing and export, MYC activity, and MAPK signalling as central pathways involved in CLL. Clonality analysis of this large data set further enabled reconstruction of temporal relationships between driver events. Direct comparison between matched pre-treatment and relapse samples from 59 patients demonstrated highly frequent clonal evolution. Thus, large sequencing data sets of clinically informative samples enable the discovery of novel genes associated with cancer, the network of relationships between the driver events, and their impact on disease relapse and clinical outcome.


Subject(s)
Disease Progression , Evolution, Molecular , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Cell Transformation, Neoplastic/genetics , Clone Cells/metabolism , Clone Cells/pathology , DNA Copy Number Variations/genetics , Exome/genetics , Genes, myc/genetics , Humans , Ikaros Transcription Factor/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , MAP Kinase Signaling System/genetics , Prognosis , RNA Processing, Post-Transcriptional/genetics , RNA Transport/genetics , Ribosomal Proteins/genetics , Treatment Outcome
4.
Cancer Discov ; 5(11): 1164-1177, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26410082

ABSTRACT

UNLABELLED: Brain metastases are associated with a dismal prognosis. Whether brain metastases harbor distinct genetic alterations beyond those observed in primary tumors is unknown. We performed whole-exome sequencing of 86 matched brain metastases, primary tumors, and normal tissue. In all clonally related cancer samples, we observed branched evolution, where all metastatic and primary sites shared a common ancestor yet continued to evolve independently. In 53% of cases, we found potentially clinically informative alterations in the brain metastases not detected in the matched primary-tumor sample. In contrast, spatially and temporally separated brain metastasis sites were genetically homogenous. Distal extracranial and regional lymph node metastases were highly divergent from brain metastases. We detected alterations associated with sensitivity to PI3K/AKT/mTOR, CDK, and HER2/EGFR inhibitors in the brain metastases. Genomic analysis of brain metastases provides an opportunity to identify potentially clinically informative alterations not detected in clinically sampled primary tumors, regional lymph nodes, or extracranial metastases. SIGNIFICANCE: Decisions for individualized therapies in patients with brain metastasis are often made from primary-tumor biopsies. We demonstrate that clinically actionable alterations present in brain metastases are frequently not detected in primary biopsies, suggesting that sequencing of primary biopsies alone may miss a substantial number of opportunities for targeted therapy.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Neoplasms/genetics , Neoplasms/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Cluster Analysis , Disease Progression , Drug Resistance, Neoplasm/genetics , Exome , Genetic Heterogeneity , Genetic Variation , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Molecular Targeted Therapy , Mutation , Neoplasms/metabolism , Signal Transduction/drug effects
5.
Nat Biotechnol ; 33(11): 1152-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26372948

ABSTRACT

Detection of somatic mutations in human leukocyte antigen (HLA) genes using whole-exome sequencing (WES) is hampered by the high polymorphism of the HLA loci, which prevents alignment of sequencing reads to the human reference genome. We describe a computational pipeline that enables accurate inference of germline alleles of class I HLA-A, B and C genes and subsequent detection of mutations in these genes using the inferred alleles as a reference. Analysis of WES data from 7,930 pairs of tumor and healthy tissue from the same patient revealed 298 nonsilent HLA mutations in tumors from 266 patients. These 298 mutations are enriched for likely functional mutations, including putative loss-of-function events. Recurrence of mutations suggested that these 'hotspot' sites were positively selected. Cancers with recurrent somatic HLA mutations were associated with upregulation of signatures of cytolytic activity characteristic of tumor infiltration by effector lymphocytes, supporting immune evasion by altered HLA function as a contributory mechanism in cancer.


Subject(s)
Histocompatibility Antigens Class I/genetics , Mutation/genetics , Neoplasms/genetics , Computational Biology , DNA Mutational Analysis , Databases, Genetic , Humans , Software
6.
Mol Cell ; 59(3): 478-90, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26253028

ABSTRACT

Repair of DNA interstrand crosslinks requires action of multiple DNA repair pathways, including homologous recombination. Here, we report a de novo heterozygous T131P mutation in RAD51/FANCR, the key recombinase essential for homologous recombination, in a patient with Fanconi anemia-like phenotype. In vitro, RAD51-T131P displays DNA-independent ATPase activity, no DNA pairing capacity, and a co-dominant-negative effect on RAD51 recombinase function. However, the patient cells are homologous recombination proficient due to the low ratio of mutant to wild-type RAD51 in cells. Instead, patient cells are sensitive to crosslinking agents and display hyperphosphorylation of Replication Protein A due to increased activity of DNA2 and WRN at the DNA interstrand crosslinks. Thus, proper RAD51 function is important during DNA interstrand crosslink repair outside of homologous recombination. Our study provides a molecular basis for how RAD51 and its associated factors may operate in a homologous recombination-independent manner to maintain genomic integrity.


Subject(s)
DNA Repair , DNA/metabolism , Fanconi Anemia/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Replication Protein A/metabolism , Cell Survival , Cross-Linking Reagents , DNA Helicases/metabolism , Exodeoxyribonucleases/metabolism , Fanconi Anemia/metabolism , Female , Genomic Instability , HEK293 Cells , Heterozygote , Humans , Infant , Mutation , RecQ Helicases/metabolism , Werner Syndrome Helicase
7.
JAMA Oncol ; 1(2): 238-44, 2015 May.
Article in English | MEDLINE | ID: mdl-26181029

ABSTRACT

IMPORTANCE: Randomized clinical trials demonstrate no benefit for epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in unselected patients with head and neck squamous cell carcinoma (HNSCC). However, a patient with stage IVA HNSCC received 13 days of neoadjuvant erlotinib and experienced a near-complete histologic response. OBJECTIVE: To determine a mechanism of exceptional response to erlotinib therapy in HNSCC. DESIGN, SETTING, AND PARTICIPANTS: Single patient with locally advanced HNSCC who received erlotinib monotherapy in a window-of-opportunity clinical trial (patients scheduled to undergo primary cancer surgery are treated briefly with an investigational agent). Whole-exome sequencing of pretreatment tumor and germline patient samples was performed at a quaternary care academic medical center, and a candidate somatic variant was experimentally investigated for mediating erlotinib response. INTERVENTION: A brief course of erlotinib monotherapy followed by surgical resection. MAIN OUTCOMES AND MEASURES: Identification of pretreatment tumor somatic alterations that may contribute to the exceptional response to erlotinib. Hypotheses were formulated regarding enhanced erlotinib response in preclinical models harboring the patient tumor somatic variant MAPK1 E322K following the identification of tumor somatic variants. RESULTS: No EGFR alterations were observed in the pretreatment tumor DNA. Paradoxically, the tumor harbored an activating MAPK1 E322K mutation (allelic fraction 0.13), which predicts ERK activation and erlotinib resistance in EGFR-mutant lung cancer. The HNSCC cells with MAPK1 E322K exhibited enhanced EGFR phosphorylation and erlotinib sensitivity compared with wild-type MAPK1 cells. CONCLUSIONS AND RELEVANCE: Selective erlotinib use in HNSCC may be informed by precision oncology approaches.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Erlotinib Hydrochloride/administration & dosage , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Mitogen-Activated Protein Kinase 1/genetics , Protein Kinase Inhibitors/administration & dosage , Tongue Neoplasms/drug therapy , Tongue Neoplasms/genetics , Adult , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Chemotherapy, Adjuvant , DNA Mutational Analysis , Drug Administration Schedule , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Genetic Predisposition to Disease , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Targeted Therapy , Mutation , Neoadjuvant Therapy , Neoplasm Staging , Phenotype , Phosphorylation , Predictive Value of Tests , Randomized Controlled Trials as Topic , Remission Induction , Squamous Cell Carcinoma of Head and Neck , Time Factors , Tongue Neoplasms/enzymology , Tongue Neoplasms/pathology , Treatment Outcome
8.
N Engl J Med ; 372(26): 2481-98, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26061751

ABSTRACT

BACKGROUND: Diffuse low-grade and intermediate-grade gliomas (which together make up the lower-grade gliomas, World Health Organization grades II and III) have highly variable clinical behavior that is not adequately predicted on the basis of histologic class. Some are indolent; others quickly progress to glioblastoma. The uncertainty is compounded by interobserver variability in histologic diagnosis. Mutations in IDH, TP53, and ATRX and codeletion of chromosome arms 1p and 19q (1p/19q codeletion) have been implicated as clinically relevant markers of lower-grade gliomas. METHODS: We performed genomewide analyses of 293 lower-grade gliomas from adults, incorporating exome sequence, DNA copy number, DNA methylation, messenger RNA expression, microRNA expression, and targeted protein expression. These data were integrated and tested for correlation with clinical outcomes. RESULTS: Unsupervised clustering of mutations and data from RNA, DNA-copy-number, and DNA-methylation platforms uncovered concordant classification of three robust, nonoverlapping, prognostically significant subtypes of lower-grade glioma that were captured more accurately by IDH, 1p/19q, and TP53 status than by histologic class. Patients who had lower-grade gliomas with an IDH mutation and 1p/19q codeletion had the most favorable clinical outcomes. Their gliomas harbored mutations in CIC, FUBP1, NOTCH1, and the TERT promoter. Nearly all lower-grade gliomas with IDH mutations and no 1p/19q codeletion had mutations in TP53 (94%) and ATRX inactivation (86%). The large majority of lower-grade gliomas without an IDH mutation had genomic aberrations and clinical behavior strikingly similar to those found in primary glioblastoma. CONCLUSIONS: The integration of genomewide data from multiple platforms delineated three molecular classes of lower-grade gliomas that were more concordant with IDH, 1p/19q, and TP53 status than with histologic class. Lower-grade gliomas with an IDH mutation either had 1p/19q codeletion or carried a TP53 mutation. Most lower-grade gliomas without an IDH mutation were molecularly and clinically similar to glioblastoma. (Funded by the National Institutes of Health.).


Subject(s)
DNA, Neoplasm/analysis , Genes, p53 , Glioma/genetics , Mutation , Adolescent , Adult , Aged , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Cluster Analysis , Female , Glioblastoma/genetics , Glioma/metabolism , Glioma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Proportional Hazards Models , Sequence Analysis, DNA , Signal Transduction
9.
Cell Rep ; 12(1): 35-41, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26119737

ABSTRACT

Fanconi anemia (FA) is a rare bone marrow failure and cancer predisposition syndrome resulting from pathogenic mutations in genes encoding proteins participating in the repair of DNA interstrand crosslinks (ICLs). Mutations in 17 genes (FANCA-FANCS) have been identified in FA patients, defining 17 complementation groups. Here, we describe an individual presenting with typical FA features who is deficient for the ubiquitin-conjugating enzyme (E2), UBE2T. UBE2T is known to interact with FANCL, the E3 ubiquitin-ligase component of the multiprotein FA core complex, and is necessary for the monoubiquitination of FANCD2 and FANCI. Proband fibroblasts do not display FANCD2 and FANCI monoubiquitination, do not form FANCD2 foci following treatment with mitomycin C, and are hypersensitive to crosslinking agents. These cellular defects are complemented by expression of wild-type UBE2T, demonstrating that deficiency of the protein UBE2T can lead to Fanconi anemia. UBE2T gene gains an alias of FANCT.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group L Protein/genetics , Fibroblasts/metabolism , Gene Deletion , HEK293 Cells , Humans , Protein Binding , Ubiquitin-Conjugating Enzymes/deficiency , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitination
10.
Cell ; 161(5): 1215-1228, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000489

ABSTRACT

Toward development of a precision medicine framework for metastatic, castration-resistant prostate cancer (mCRPC), we established a multi-institutional clinical sequencing infrastructure to conduct prospective whole-exome and transcriptome sequencing of bone or soft tissue tumor biopsies from a cohort of 150 mCRPC affected individuals. Aberrations of AR, ETS genes, TP53, and PTEN were frequent (40%-60% of cases), with TP53 and AR alterations enriched in mCRPC compared to primary prostate cancer. We identified new genomic alterations in PIK3CA/B, R-spondin, BRAF/RAF1, APC, ß-catenin, and ZBTB16/PLZF. Moreover, aberrations of BRCA2, BRCA1, and ATM were observed at substantially higher frequencies (19.3% overall) compared to those in primary prostate cancers. 89% of affected individuals harbored a clinically actionable aberration, including 62.7% with aberrations in AR, 65% in other cancer-related genes, and 8% with actionable pathogenic germline alterations. This cohort study provides clinically actionable information that could impact treatment decisions for these affected individuals.


Subject(s)
Gene Expression Profiling/methods , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Cohort Studies , Humans , Male , Mutation , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy
11.
Genome Res ; 25(3): 316-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25650244

ABSTRACT

Glioblastoma (GBM) is a prototypical heterogeneous brain tumor refractory to conventional therapy. A small residual population of cells escapes surgery and chemoradiation, resulting in a typically fatal tumor recurrence ∼ 7 mo after diagnosis. Understanding the molecular architecture of this residual population is critical for the development of successful therapies. We used whole-genome sequencing and whole-exome sequencing of multiple sectors from primary and paired recurrent GBM tumors to reconstruct the genomic profile of residual, therapy resistant tumor initiating cells. We found that genetic alteration of the p53 pathway is a primary molecular event predictive of a high number of subclonal mutations in glioblastoma. The genomic road leading to recurrence is highly idiosyncratic but can be broadly classified into linear recurrences that share extensive genetic similarity with the primary tumor and can be directly traced to one of its specific sectors, and divergent recurrences that share few genetic alterations with the primary tumor and originate from cells that branched off early during tumorigenesis. Our study provides mechanistic insights into how genetic alterations in primary tumors impact the ensuing evolution of tumor cells and the emergence of subclonal heterogeneity.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Exome , Genome, Human , Glioblastoma/genetics , Glioblastoma/pathology , High-Throughput Nucleotide Sequencing , Adult , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Clonal Evolution/genetics , DNA Copy Number Variations , DNA Methylation , Genomics/methods , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Middle Aged , Mutation , Mutation Rate , Neoplasm Grading , Neoplasm Recurrence, Local , Polymorphism, Single Nucleotide , Signal Transduction , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Young Adult
13.
Proc Natl Acad Sci U S A ; 111(51): E5564-73, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25512523

ABSTRACT

Osteosarcoma is the most common primary bone tumor, yet there have been no substantial advances in treatment or survival in three decades. We examined 59 tumor/normal pairs by whole-exome, whole-genome, and RNA-sequencing. Only the TP53 gene was mutated at significant frequency across all samples. The mean nonsilent somatic mutation rate was 1.2 mutations per megabase, and there was a median of 230 somatic rearrangements per tumor. Complex chains of rearrangements and localized hypermutation were detected in almost all cases. Given the intertumor heterogeneity, the extent of genomic instability, and the difficulty in acquiring a large sample size in a rare tumor, we used several methods to identify genomic events contributing to osteosarcoma survival. Pathway analysis, a heuristic analytic algorithm, a comparative oncology approach, and an shRNA screen converged on the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway as a central vulnerability for therapeutic exploitation in osteosarcoma. Osteosarcoma cell lines are responsive to pharmacologic and genetic inhibition of the PI3K/mTOR pathway both in vitro and in vivo.


Subject(s)
Bone Neoplasms/metabolism , Genome, Human , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Genetic Heterogeneity , Germ-Line Mutation , Humans , Osteosarcoma/genetics , Osteosarcoma/pathology , Tumor Suppressor Protein p53/genetics
14.
Cancer Cell ; 26(6): 813-825, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490447

ABSTRACT

Intratumoral heterogeneity plays a critical role in tumor evolution. To define the contribution of DNA methylation to heterogeneity within tumors, we performed genome-scale bisulfite sequencing of 104 primary chronic lymphocytic leukemias (CLLs). Compared with 26 normal B cell samples, CLLs consistently displayed higher intrasample variability of DNA methylation patterns across the genome, which appears to arise from stochastically disordered methylation in malignant cells. Transcriptome analysis of bulk and single CLL cells revealed that methylation disorder was linked to low-level expression. Disordered methylation was further associated with adverse clinical outcome. We therefore propose that disordered methylation plays a similar role to that of genetic instability, enhancing the ability of cancer cells to search for superior evolutionary trajectories.


Subject(s)
B-Lymphocytes/metabolism , DNA Methylation , Epigenesis, Genetic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , CpG Islands , Gene Expression Regulation, Leukemic , Genetic Variation , Genome, Human , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Molecular Sequence Data , Sequence Analysis, DNA , Sulfites/chemistry
15.
Mol Cancer ; 13: 141, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24894453

ABSTRACT

BACKGROUND: Inhibition of the activated epidermal growth factor receptor (EGFR) with either enzymatic kinase inhibitors or anti-EGFR antibodies such as cetuximab, is an effective modality of treatment for multiple human cancers. Enzymatic EGFR inhibitors are effective for lung adenocarcinomas with somatic kinase domain EGFR mutations while, paradoxically, anti-EGFR antibodies are more effective in colon and head and neck cancers where EGFR mutations occur less frequently. In colorectal cancer, anti-EGFR antibodies are routinely used as second-line therapy of KRAS wild-type tumors. However, detailed mechanisms and genomic predictors for pharmacological response to these antibodies in colon cancer remain unclear. FINDINGS: We describe a case of colorectal adenocarcinoma, which was found to harbor a kinase domain mutation, G724S, in EGFR through whole genome sequencing. We show that G724S mutant EGFR is oncogenic and that it differs from classic lung cancer derived EGFR mutants in that it is cetuximab responsive in vitro, yet relatively insensitive to small molecule kinase inhibitors. Through biochemical and cellular pharmacologic studies, we have determined that cells harboring the colon cancer-derived G719S and G724S mutants are responsive to cetuximab therapy in vitro and found that the requirement for asymmetric dimerization of these mutant EGFR to promote cellular transformation may explain their greater inhibition by cetuximab than small-molecule kinase inhibitors. CONCLUSION: The colon-cancer derived G719S and G724S mutants are oncogenic and sensitive in vitro to cetuximab. These data suggest that patients with these mutations may benefit from the use of anti-EGFR antibodies as part of the first-line therapy.


Subject(s)
Adenocarcinoma/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Colorectal Neoplasms/genetics , ErbB Receptors/genetics , Mutation , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Protein Kinase Inhibitors/therapeutic use , Protein Multimerization , Protein Structure, Tertiary
16.
Blood ; 124(3): 453-62, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24891321

ABSTRACT

Genome sequencing has revealed a large number of shared and personal somatic mutations across human cancers. In principle, any genetic alteration affecting a protein-coding region has the potential to generate mutated peptides that are presented by surface HLA class I proteins that might be recognized by cytotoxic T cells. To test this possibility, we implemented a streamlined approach for the prediction and validation of such neoantigens derived from individual tumors and presented by patient-specific HLA alleles. We applied our computational pipeline to 91 chronic lymphocytic leukemias (CLLs) that underwent whole-exome sequencing (WES). We predicted ∼22 mutated HLA-binding peptides per leukemia (derived from ∼16 missense mutations) and experimentally confirmed HLA binding for ∼55% of such peptides. Two CLL patients that achieved long-term remission following allogeneic hematopoietic stem cell transplantation were monitored for CD8(+) T-cell responses against predicted or confirmed HLA-binding peptides. Long-lived cytotoxic T-cell responses were detected against peptides generated from personal tumor mutations in ALMS1, C6ORF89, and FNDC3B presented on tumor cells. Finally, we applied our computational pipeline to WES data (N = 2488 samples) across 13 different cancer types and estimated dozens to thousands of predicted neoantigens per individual tumor, suggesting that neoantigens are frequent in most tumors.


Subject(s)
Antigens, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mutation , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Exome , Female , HLA Antigens/metabolism , Humans , Immunologic Memory , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Precision Medicine , Protein Binding , T-Lymphocytes, Cytotoxic/immunology
17.
Genome Res ; 24(7): 1053-63, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24823667

ABSTRACT

Retrotransposons constitute a major source of genetic variation, and somatic retrotransposon insertions have been reported in cancer. Here, we applied TranspoSeq, a computational framework that identifies retrotransposon insertions from sequencing data, to whole genomes from 200 tumor/normal pairs across 11 tumor types as part of The Cancer Genome Atlas (TCGA) Pan-Cancer Project. In addition to novel germline polymorphisms, we find 810 somatic retrotransposon insertions primarily in lung squamous, head and neck, colorectal, and endometrial carcinomas. Many somatic retrotransposon insertions occur in known cancer genes. We find that high somatic retrotransposition rates in tumors are associated with high rates of genomic rearrangement and somatic mutation. Finally, we developed TranspoSeq-Exome to interrogate an additional 767 tumor samples with hybrid-capture exome data and discovered 35 novel somatic retrotransposon insertions into exonic regions, including an insertion into an exon of the PTEN tumor suppressor gene. The results of this large-scale, comprehensive analysis of retrotransposon movement across tumor types suggest that somatic retrotransposon insertions may represent an important class of structural variation in cancer.


Subject(s)
Exome , Genome, Human , Neoplasms/genetics , Retroelements/genetics , Base Sequence , Computational Biology/methods , DNA Transposable Elements , Databases, Nucleic Acid , High-Throughput Nucleotide Sequencing , Humans , Mutagenesis, Insertional , Sequence Analysis, DNA
18.
Blood ; 124(7): 1089-98, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24778153

ABSTRACT

One major goal of cancer genome sequencing is to identify key genes and pathways that drive tumor pathogenesis. Although many studies have identified candidate driver genes based on recurrence of mutations in individual genes, subsets of genes with nonrecurrent mutations may also be defined as putative drivers if they affect a single biological pathway. In this fashion, we previously identified Wnt signaling as significantly mutated through large-scale massively parallel DNA sequencing of chronic lymphocytic leukemia (CLL). Here, we use a novel method of biomolecule delivery, vertical silicon nanowires, to efficiently introduce small interfering RNAs into CLL cells, and interrogate the effects of 8 of 15 mutated Wnt pathway members identified across 91 CLLs. In HEK293T cells, mutations in 2 genes did not generate functional changes, 3 led to dysregulated pathway activation, and 3 led to further activation or loss of repression of pathway activation. Silencing 4 of 8 mutated genes in CLL samples harboring the mutated alleles resulted in reduced viability compared with leukemia samples with wild-type alleles. We demonstrate that somatic mutations in CLL can generate dependence on this pathway for survival. These findings support the notion that nonrecurrent mutations at different nodes of the Wnt pathway can contribute to leukemogenesis.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Signal Transduction/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Adult , Cell Line, Tumor , Cell Survival/genetics , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
19.
Nat Biotechnol ; 32(5): 479-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24752078

ABSTRACT

Comprehensive analyses of cancer genomes promise to inform prognoses and precise cancer treatments. A major barrier, however, is inaccessibility of metastatic tissue. A potential solution is to characterize circulating tumor cells (CTCs), but this requires overcoming the challenges of isolating rare cells and sequencing low-input material. Here we report an integrated process to isolate, qualify and sequence whole exomes of CTCs with high fidelity using a census-based sequencing strategy. Power calculations suggest that mapping of >99.995% of the standard exome is possible in CTCs. We validated our process in two patients with prostate cancer, including one for whom we sequenced CTCs, a lymph node metastasis and nine cores of the primary tumor. Fifty-one of 73 CTC mutations (70%) were present in matched tissue. Moreover, we identified 10 early trunk and 56 metastatic trunk mutations in the non-CTC tumor samples and found 90% and 73% of these mutations, respectively, in CTC exomes. This study establishes a foundation for CTC genomics in the clinic.


Subject(s)
Exome/genetics , Neoplastic Cells, Circulating , Prostatic Neoplasms/genetics , Humans , Male , Mutation/genetics
20.
Cell ; 156(6): 1298-1311, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24630729

ABSTRACT

Small cell lung carcinoma (SCLC) is a highly lethal, smoking-associated cancer with few known targetable genetic alterations. Using genome sequencing, we characterized the somatic evolution of a genetically engineered mouse model (GEMM) of SCLC initiated by loss of Trp53 and Rb1. We identified alterations in DNA copy number and complex genomic rearrangements and demonstrated a low somatic point mutation frequency in the absence of tobacco mutagens. Alterations targeting the tumor suppressor Pten occurred in the majority of murine SCLC studied, and engineered Pten deletion accelerated murine SCLC and abrogated loss of Chr19 in Trp53; Rb1; Pten compound mutant tumors. Finally, we found evidence for polyclonal and sequential metastatic spread of murine SCLC by comparative sequencing of families of related primary tumors and metastases. We propose a temporal model of SCLC tumorigenesis with implications for human SCLC therapeutics and the nature of cancer-genome evolution in GEMMs.


Subject(s)
Carcinogenesis , Disease Models, Animal , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Animals , Humans , Liver Neoplasms/secondary , Lymphatic Metastasis , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Small Cell Lung Carcinoma/secondary
SELECTION OF CITATIONS
SEARCH DETAIL