Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Blood ; 127(24): 3004-14, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26966091

ABSTRACT

The spectrum of somatic alterations in hematologic malignancies includes substitutions, insertions/deletions (indels), copy number alterations (CNAs), and a wide range of gene fusions; no current clinically available single assay captures the different types of alterations. We developed a novel next-generation sequencing-based assay to identify all classes of genomic alterations using archived formalin-fixed paraffin-embedded blood and bone marrow samples with high accuracy in a clinically relevant time frame, which is performed in our Clinical Laboratory Improvement Amendments-certified College of American Pathologists-accredited laboratory. Targeted capture of DNA/RNA and next-generation sequencing reliably identifies substitutions, indels, CNAs, and gene fusions, with similar accuracy to lower-throughput assays that focus on specific genes and types of genomic alterations. Profiling of 3696 samples identified recurrent somatic alterations that impact diagnosis, prognosis, and therapy selection. This comprehensive genomic profiling approach has proved effective in detecting all types of genomic alterations, including fusion transcripts, which increases the ability to identify clinically relevant genomic alterations with therapeutic relevance.


Subject(s)
DNA Fingerprinting/methods , Gene Expression Profiling/methods , Genomics/methods , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Chromosome Aberrations , Clinical Laboratory Techniques/methods , DNA Mutational Analysis/methods , DNA, Neoplasm/analysis , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Polymorphism, Genetic , RNA, Neoplasm/analysis , Sensitivity and Specificity , Systems Integration
2.
Int J Cancer ; 138(4): 881-90, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26314551

ABSTRACT

Although the BRAF V600E base substitution is an approved target for the BRAF inhibitors in melanoma, BRAF gene fusions have not been investigated as anticancer drug targets. In our study, a wide variety of tumors underwent comprehensive genomic profiling for hundreds of known cancer genes using the FoundationOne™ or FoundationOne Heme™ comprehensive genomic profiling assays. BRAF fusions involving the intact in-frame BRAF kinase domain were observed in 55 (0.3%) of 20,573 tumors, across 12 distinct tumor types, including 20 novel BRAF fusions. These comprised 29 unique 5' fusion partners, of which 31% (9) were known and 69% (20) were novel. BRAF fusions included 3% (14/531) of melanomas; 2% (15/701) of gliomas; 1.0% (3/294) of thyroid cancers; 0.3% (3/1,062) pancreatic carcinomas; 0.2% (8/4,013) nonsmall-cell lung cancers and 0.2% (4/2,154) of colorectal cancers, and were enriched in pilocytic (30%) vs. nonpilocytic gliomas (1%; p < 0.0001), Spitzoid (75%) vs. nonSpitzoid melanomas (1%; p = 0.0001), acinar (67%) vs. nonacinar pancreatic cancers (<1%; p < 0.0001) and papillary (3%) vs. nonpapillary thyroid cancers (0%; p < 0.03). Clinical responses to trametinib and sorafenib are presented. In conclusion, BRAF fusions are rare driver alterations in a wide variety of malignant neoplasms, but enriched in Spitzoid melanoma, pilocytic astrocytomas, pancreatic acinar and papillary thyroid cancers.


Subject(s)
Molecular Targeted Therapy/methods , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Infant , Male , Middle Aged , Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Oncogene Proteins, Fusion/genetics , Phenylurea Compounds/therapeutic use , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Sorafenib , Transcriptome , Treatment Outcome , Young Adult
3.
J Mol Diagn ; 20(5): 686-702, 2018 09.
Article in English | MEDLINE | ID: mdl-29936259

ABSTRACT

Genomic profiling of circulating tumor DNA derived from cell-free DNA (cfDNA) in blood can provide a noninvasive method for detecting genomic biomarkers to guide clinical decision making for cancer patients. We developed a hybrid capture-based next-generation sequencing assay for genomic profiling of circulating tumor DNA from blood (FoundationACT). High-sequencing coverage and molecular barcode-based error detection enabled accurate detection of genomic alterations, including short variants (base substitutions, short insertions/deletions) and genomic re-arrangements at low allele frequencies (AFs), and copy number amplifications. Analytical validation was performed on 2666 reference alterations. The assay achieved >99% overall sensitivity (95% CI, 99.1%-99.4%) for short variants at AF >0.5%, >95% sensitivity (95% CI, 94.2%-95.7%) for AF 0.25% to 0.5%, and 70% sensitivity (95% CI, 68.2%-71.5%) for AF 0.125% to 0.25%. No false positives were detected in 62 samples from healthy volunteers. Genomic alterations detected by FoundationACT demonstrated high concordance with orthogonal assays run on the same clinical cfDNA samples. In 860 routine clinical FoundationACT cases, genomic alterations were detected in cfDNA at comparable frequencies to tissue; for the subset of cases with temporally matched tissue and blood samples, 75% of genomic alterations and 83% of short variant mutations detected in tissue were also detected in cfDNA. On the basis of analytical validation results, FoundationACT has been approved for use in our Clinical Laboratory Improvement Amendments-certified/College of American Pathologists-accredited/New York State-approved laboratory.


Subject(s)
Circulating Tumor DNA/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Circulating Tumor DNA/blood , Gene Amplification , Gene Dosage , Gene Rearrangement , Humans , INDEL Mutation/genetics
4.
Genome Med ; 9(1): 34, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28420421

ABSTRACT

BACKGROUND: High tumor mutational burden (TMB) is an emerging biomarker of sensitivity to immune checkpoint inhibitors and has been shown to be more significantly associated with response to PD-1 and PD-L1 blockade immunotherapy than PD-1 or PD-L1 expression, as measured by immunohistochemistry (IHC). The distribution of TMB and the subset of patients with high TMB has not been well characterized in the majority of cancer types. METHODS: In this study, we compare TMB measured by a targeted comprehensive genomic profiling (CGP) assay to TMB measured by exome sequencing and simulate the expected variance in TMB when sequencing less than the whole exome. We then describe the distribution of TMB across a diverse cohort of 100,000 cancer cases and test for association between somatic alterations and TMB in over 100 tumor types. RESULTS: We demonstrate that measurements of TMB from comprehensive genomic profiling are strongly reflective of measurements from whole exome sequencing and model that below 0.5 Mb the variance in measurement increases significantly. We find that a subset of patients exhibits high TMB across almost all types of cancer, including many rare tumor types, and characterize the relationship between high TMB and microsatellite instability status. We find that TMB increases significantly with age, showing a 2.4-fold difference between age 10 and age 90 years. Finally, we investigate the molecular basis of TMB and identify genes and mutations associated with TMB level. We identify a cluster of somatic mutations in the promoter of the gene PMS2, which occur in 10% of skin cancers and are highly associated with increased TMB. CONCLUSIONS: These results show that a CGP assay targeting ~1.1 Mb of coding genome can accurately assess TMB compared with sequencing the whole exome. Using this method, we find that many disease types have a substantial portion of patients with high TMB who might benefit from immunotherapy. Finally, we identify novel, recurrent promoter mutations in PMS2, which may be another example of regulatory mutations contributing to tumorigenesis.


Subject(s)
DNA Mutational Analysis , Genome, Human , Mutation , Neoplasms/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Child , DNA, Neoplasm , Exome , Humans , Middle Aged , Mismatch Repair Endonuclease PMS2 , Neoplasms/epidemiology , Neoplasms/metabolism , Neoplasms/pathology , Young Adult
5.
Cancer Discov ; 5(8): 850-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25971938

ABSTRACT

UNLABELLED: Focal amplification and activating point mutation of the MET gene are well-characterized oncogenic drivers that confer susceptibility to targeted MET inhibitors. Recurrent somatic splice site alterations at MET exon 14 (METex14) that result in exon skipping and MET activation have been characterized, but their full diversity and prevalence across tumor types are unknown. Here, we report analysis of tumor genomic profiles from 38,028 patients to identify 221 cases with METex14 mutations (0.6%), including 126 distinct sequence variants. METex14 mutations are detected most frequently in lung adenocarcinoma (3%), but also frequently in other lung neoplasms (2.3%), brain glioma (0.4%), and tumors of unknown primary origin (0.4%). Further in vitro studies demonstrate sensitivity to MET inhibitors in cells harboring METex14 alterations. We also report three new patient cases with METex14 alterations in lung or histiocytic sarcoma tumors that showed durable response to two different MET-targeted therapies. The diversity of METex14 mutations indicates that diagnostic testing via comprehensive genomic profiling is necessary for detection in a clinical setting. SIGNIFICANCE: Here we report the identification of diverse exon 14 splice site alterations in MET that result in constitutive activity of this receptor and oncogenic transformation in vitro. Patients whose tumors harbored these alterations derived meaningful clinical benefit from MET inhibitors. Collectively, these data support the role of METex14 alterations as drivers of tumorigenesis, and identify a unique subset of patients likely to derive benefit from MET inhibitors.


Subject(s)
Alternative Splicing , Antineoplastic Agents/therapeutic use , Exons , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cluster Analysis , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Genomics/methods , Humans , Immunohistochemistry , Male , Mutation , Neoplasm Staging , Neoplasms/diagnosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Tomography, X-Ray Computed
6.
Blood ; 109(8): 3177-88, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17185464

ABSTRACT

The aims of this study were to assess the feasibility of prospective pharmacogenomics research in multicenter international clinical trials of bortezomib in multiple myeloma and to develop predictive classifiers of response and survival with bortezomib. Patients with relapsed myeloma enrolled in phase 2 and phase 3 clinical trials of bortezomib and consented to genomic analyses of pretreatment tumor samples. Bone marrow aspirates were subject to a negative-selection procedure to enrich for tumor cells, and these samples were used for gene expression profiling using DNA microarrays. Data quality and correlations with trial outcomes were assessed by multiple groups. Gene expression in this dataset was consistent with data published from a single-center study of newly diagnosed multiple myeloma. Response and survival classifiers were developed and shown to be significantly associated with outcome via testing on independent data. The survival classifier improved on the risk stratification provided by the International Staging System. Predictive models and biologic correlates of response show some specificity for bortezomib rather than dexamethasone. Informative gene expression data and genomic classifiers that predict clinical outcome can be derived from prospective clinical trials of new anticancer agents.


Subject(s)
Boronic Acids/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Models, Biological , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protease Inhibitors/administration & dosage , Pyrazines/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Bortezomib , Dexamethasone/administration & dosage , Female , Gene Expression Profiling , Humans , Male , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Oligonucleotide Array Sequence Analysis , Pharmacogenetics , Predictive Value of Tests , Prospective Studies , Proteasome Inhibitors , Recurrence , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL