Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 543(7647): 723-727, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28329770

ABSTRACT

Cancer somatic mutations can generate neoantigens that distinguish malignant from normal cells. However, the personalized identification and validation of neoantigens remains a major challenge. Here we discover neoantigens in human mantle-cell lymphomas by using an integrated genomic and proteomic strategy that interrogates tumour antigen peptides presented by major histocompatibility complex (MHC) class I and class II molecules. We applied this approach to systematically characterize MHC ligands from 17 patients. Remarkably, all discovered neoantigenic peptides were exclusively derived from the lymphoma immunoglobulin heavy- or light-chain variable regions. Although we identified MHC presentation of private polymorphic germline alleles, no mutated peptides were recovered from non-immunoglobulin somatically mutated genes. Somatic mutations within the immunoglobulin variable region were almost exclusively presented by MHC class II. We isolated circulating CD4+ T cells specific for immunoglobulin-derived neoantigens and found these cells could mediate killing of autologous lymphoma cells. These results demonstrate that an integrative approach combining MHC isolation, peptide identification, and exome sequencing is an effective platform to uncover tumour neoantigens. Application of this strategy to human lymphoma implicates immunoglobulin neoantigens as targets for lymphoma immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Immunoglobulin Variable Region/immunology , Lymphoma, Mantle-Cell/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , DNA Mutational Analysis , Epitopes, T-Lymphocyte/immunology , Exome/genetics , Genomics , HLA-D Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunotherapy/trends , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/therapy , Mutation , Proteomics
2.
Blood ; 132(23): 2456-2464, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30249784

ABSTRACT

The introduction of novel agents has led to major improvements in clinical outcomes for patients with multiple myeloma. To shorten evaluation times for new treatments, health agencies are currently examining minimal residual disease (MRD) as a surrogate end point in clinical trials. We assessed the prognostic value of MRD, measured during maintenance therapy by next-generation sequencing (NGS). MRD negativity was defined as the absence of tumor plasma cell within 1 000 000 bone marrow cells (<10-6). Data were analyzed from a recent clinical trial that evaluated the role of transplantation in newly diagnosed myeloma patients treated with lenalidomide, bortezomib, and dexamethasone (RVD). MRD negativity was achieved at least once during maintenance in 127 patients (25%). At the start of maintenance therapy, MRD was a strong prognostic factor for both progression-free survival (adjusted hazard ratio, 0.22; 95% confidence interval, 0.15-0.34; P < .001) and overall survival (adjusted hazard ratio, 0.24; 95% confidence interval, 0.11-0.54; P = .001). Patients who were MRD negative had a higher probability of prolonged progression-free survival than patients with detectable residual disease, regardless of treatment group (RVD vs transplant), cytogenetic risk profile, or International Staging System disease stage at diagnosis. These results were similar after completion of maintenance therapy. Our findings confirm the value of MRD status, as determined by NGS, as a prognostic biomarker in multiple myeloma, and suggest that this approach could be used to adapt treatment strategies in future clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , High-Throughput Nucleotide Sequencing , Multiple Myeloma/metabolism , Aged , Bone Marrow/metabolism , Bone Marrow/pathology , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Disease-Free Survival , Female , Follow-Up Studies , Humans , Lenalidomide/administration & dosage , Maintenance Chemotherapy , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Neoplasm, Residual , Plasma Cells/metabolism , Plasma Cells/pathology , Survival Rate
3.
Br J Haematol ; 175(5): 841-850, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27711974

ABSTRACT

Next-generation sequencing (NGS)-based circulating tumour DNA (ctDNA) detection is a promising monitoring tool for lymphoid malignancies. We evaluated whether the presence of ctDNA was associated with outcome after allogeneic haematopoietic stem cell transplantation (HSCT) in lymphoma patients. We studied 88 patients drawn from a phase 3 clinical trial of reduced-intensity conditioning HSCT in lymphoma. Conventional restaging and collection of peripheral blood samples occurred at pre-specified time points before and after HSCT and were assayed for ctDNA by sequencing of the immunoglobulin or T-cell receptor genes. Tumour clonotypes were identified in 87% of patients with adequate tumour samples. Sixteen of 19 (84%) patients with disease progression after HSCT had detectable ctDNA prior to progression at a median of 3·7 months prior to relapse/progression. Patients with detectable ctDNA 3 months after HSCT had inferior progression-free survival (PFS) (2-year PFS 58% vs. 84% in ctDNA-negative patients, P = 0·033). In multivariate models, detectable ctDNA was associated with increased risk of progression/death (Hazard ratio 3·9, P = 0·003) and increased risk of relapse/progression (Hazard ratio 10·8, P = 0·0006). Detectable ctDNA is associated with an increased risk of relapse/progression, but further validation studies are necessary to confirm these findings and determine the clinical utility of NGS-based minimal residual disease monitoring in lymphoma patients after HSCT.


Subject(s)
DNA, Neoplasm/blood , Hematopoietic Stem Cell Transplantation/standards , Lymphoma/diagnosis , Neoplasm, Residual/diagnosis , Adolescent , Adult , Aged , Death , Disease Progression , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunoglobulins/genetics , Lymphoma/genetics , Lymphoma/therapy , Male , Middle Aged , Receptors, Antigen, T-Cell/genetics , Recurrence , Retrospective Studies , Sequence Analysis, DNA , Transplantation, Homologous , Treatment Outcome , Young Adult
4.
Nature ; 466(7308): 869-73, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20668451

ABSTRACT

The systematic characterization of somatic mutations in cancer genomes is essential for understanding the disease and for developing targeted therapeutics. Here we report the identification of 2,576 somatic mutations across approximately 1,800 megabases of DNA representing 1,507 coding genes from 441 tumours comprising breast, lung, ovarian and prostate cancer types and subtypes. We found that mutation rates and the sets of mutated genes varied substantially across tumour types and subtypes. Statistical analysis identified 77 significantly mutated genes including protein kinases, G-protein-coupled receptors such as GRM8, BAI3, AGTRL1 (also called APLNR) and LPHN3, and other druggable targets. Integrated analysis of somatic mutations and copy number alterations identified another 35 significantly altered genes including GNAS, indicating an expanded role for galpha subunits in multiple cancer types. Furthermore, our experimental analyses demonstrate the functional roles of mutant GNAO1 (a Galpha subunit) and mutant MAP2K4 (a member of the JNK signalling pathway) in oncogenesis. Our study provides an overview of the mutational spectra across major human cancers and identifies several potential therapeutic targets.


Subject(s)
Genes, Neoplasm/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Breast Neoplasms/classification , Breast Neoplasms/genetics , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Female , GTP-Binding Protein alpha Subunits/genetics , Humans , Lung Neoplasms/classification , Lung Neoplasms/genetics , MAP Kinase Kinase 4/genetics , Male , Neoplasms/enzymology , Neoplasms/pathology , Ovarian Neoplasms/classification , Ovarian Neoplasms/genetics , Prostatic Neoplasms/classification , Prostatic Neoplasms/genetics , Protein Kinases/genetics , Receptors, G-Protein-Coupled/genetics
5.
Br J Haematol ; 169(5): 689-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818067

ABSTRACT

We applied a highly sensitive next-generation sequencing method to identify lymphoma-specific immunoglobulin gene segments in classical Hodgkin lymphoma (CHL) at initial diagnosis or recurrence, and assessed the ability of detecting such lymphoma-specific sequences in peripheral blood (PB). Seventeen CHL cases were tested and lymphoma-specific sequences were identified in 12 of the primary tumour biopsies. In 11 of these patients whose paired PB samples were available, tumour-specific clonotypes were detected in PB in eight patients. This data demonstrates the feasibility of detecting circulating tumour-specific sequences, creating an unprecedented opportunity to optimize the future treatment and monitoring strategies for patients with CHL.


Subject(s)
Hodgkin Disease/diagnosis , Hodgkin Disease/genetics , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Herpesvirus 4, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Leukocytes, Mononuclear/metabolism , Neoplasm Staging
6.
Biol Blood Marrow Transplant ; 20(9): 1307-13, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24769317

ABSTRACT

Minimal residual disease (MRD) quantification is an important predictor of outcome after treatment for acute lymphoblastic leukemia (ALL). Bone marrow ALL burden ≥ 10(-4) after induction predicts subsequent relapse. Likewise, MRD ≥ 10(-4) in bone marrow before initiation of conditioning for allogeneic (allo) hematopoietic cell transplantation (HCT) predicts transplantation failure. Current methods for MRD quantification in ALL are not sufficiently sensitive for use with peripheral blood specimens and have not been broadly implemented in the management of adults with ALL. Consensus-primed immunoglobulin (Ig), T cell receptor (TCR) amplification and high-throughput sequencing (HTS) permit use of a standardized algorithm for all patients and can detect leukemia at 10(-6) or lower. We applied the LymphoSIGHT HTS platform (Sequenta Inc., South San Francisco, CA) to quantification of MRD in 237 samples from 29 adult B cell ALL patients before and after allo-HCT. Using primers for the IGH-VDJ, IGH-DJ, IGK, TCRB, TCRD, and TCRG loci, MRD could be quantified in 93% of patients. Leukemia-associated clonotypes at these loci were identified in 52%, 28%, 10%, 35%, 28%, and 41% of patients, respectively. MRD ≥ 10(-4) before HCT conditioning predicted post-HCT relapse (hazard ratio [HR], 7.7; 95% confidence interval [CI], 2.0 to 30; P = .003). In post-HCT blood samples, MRD ≥10(-6) had 100% positive predictive value for relapse with median lead time of 89 days (HR, 14; 95% CI, 4.7 to 44, P < .0001). The use of HTS-based MRD quantification in adults with ALL offers a standardized approach with sufficient sensitivity to quantify leukemia MRD in peripheral blood. Use of this approach may identify a window for clinical intervention before overt relapse.


Subject(s)
Genes, T-Cell Receptor/genetics , Hematopoietic Stem Cell Transplantation/methods , Immunoglobulins/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Adolescent , Adult , Aged , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Neoplasm Recurrence, Local , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Retrospective Studies , Survival Analysis , Young Adult
7.
Blood ; 120(26): 5173-80, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23074282

ABSTRACT

The persistence of minimal residual disease (MRD) during therapy is the strongest adverse prognostic factor in acute lymphoblastic leukemia (ALL). We developed a high-throughput sequencing method that universally amplifies antigen-receptor gene segments and identifies all clonal gene rearrangements (ie, leukemia-specific sequences) at diagnosis, allowing monitoring of disease progression and clonal evolution during therapy. In the present study, the assay specifically detected 1 leukemic cell among greater than 1 million leukocytes in spike-in experiments. We compared this method with the gold-standard MRD assays multiparameter flow cytometry and allele-specific oligonucleotide polymerase chain reaction (ASO-PCR) using diagnostic and follow-up samples from 106 patients with ALL. Sequencing detected MRD in all 28 samples shown to be positive by flow cytometry and in 35 of the 36 shown to be positive by ASO-PCR and revealed MRD in 10 and 3 additional samples that were negative by flow cytometry and ASO-PCR, respectively. We conclude that this new method allows monitoring of treatment response in ALL and other lymphoid malignancies with great sensitivity and precision. The www.clinicaltrials.gov identifier number for the Total XV study is NCT00137111.


Subject(s)
High-Throughput Nucleotide Sequencing , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Base Sequence , Child , Clonal Evolution/genetics , Clonal Evolution/physiology , Genes, Immunoglobulin Heavy Chain/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Models, Biological , Molecular Diagnostic Techniques/methods , Molecular Sequence Data , Neoplasm, Residual , Polymerase Chain Reaction/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prognosis , Sensitivity and Specificity
8.
Blood ; 120(22): 4407-17, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22932801

ABSTRACT

The ability to distinguish clonal B-cell populations based on the sequence of their rearranged immunoglobulin heavy chain (IgH) locus is an important tool for diagnosing B-cell neoplasms and monitoring treatment response. Leukemic precursor B cells may continue to undergo recombination of the IgH gene after malignant transformation; however, the magnitude of evolution at the IgH locus is currently unknown. We used next-generation sequencing to characterize the repertoire of IgH sequences in diagnostic samples of 51 children with B precursor acute lymphoblastic leukemia (B-ALL). We identified clonal IgH rearrangements in 43 of 51 (84%) cases and found that the number of evolved IgH sequences per patient ranged dramatically from 0 to 4024. We demonstrate that the evolved IgH sequences are not the result of amplification artifacts and are unique to leukemic precursor B cells. In addition, the evolution often follows an allelic exclusion pattern, where only 1 of 2 rearranged IgH loci exhibit ongoing recombination. Thus, precursor B-cell leukemias maintain evolution at the IgH locus at levels that were previously underappreciated. This finding sheds light on the mechanisms associated with leukemic clonal evolution and may fundamentally change approaches for monitoring minimal residual disease burden.


Subject(s)
Clonal Evolution/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Genetic Loci , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Age Factors , Algorithms , Bone Marrow/pathology , Case-Control Studies , Child , Child, Preschool , Clonal Evolution/physiology , DNA Mutational Analysis , Genetic Loci/genetics , Humans , Infant , Infant, Newborn , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Validation Studies as Topic
10.
Proc Natl Acad Sci U S A ; 107(28): 12587-92, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616066

ABSTRACT

A unique microarray-based method for determining the extent of DNA methylation has been developed. It relies on a selective enrichment of the regions to be assayed by target amplification by capture and ligation (mTACL). The assay is quantitatively accurate, relatively precise, and lends itself to high-throughput determination using nanogram amounts of DNA. The measurements using mTACLs are highly reproducible and in excellent agreement with those obtained by sequencing (r = 0.94). In the present work, the methylation status of >145,000 CpGs from 5,472 promoters in 221 samples was measured. The methylation levels of nearby CpGs are correlated, but the correlation falls off dramatically over several hundred base pairs. In some instances, nearby CpGs have very different levels of methylation. Comparison of normal and tumor samples indicates that in tumors, the promoter regions of genes involved in differentiation and signaling are preferentially hypermethylated, whereas those of housekeeping genes remain hypomethylated. mTACL is a platform for profiling the state of methylation of a large number of CpG in many samples in a cost-effective fashion, and is capable of scaling to much larger numbers of CpGs than those collected here.


Subject(s)
DNA Methylation , Cell Differentiation/genetics , DNA/genetics , Dinucleoside Phosphates , Genome , Humans , Methylation
11.
Nat Genet ; 34(1): 91-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12704386

ABSTRACT

Familial hypercholanemia (FHC) is characterized by elevated serum bile acid concentrations, itching, and fat malabsorption. We show here that FHC in Amish individuals is associated with mutations in tight junction protein 2 (encoded by TJP2, also known as ZO-2) and bile acid Coenzyme A: amino acid N-acyltransferase (encoded by BAAT). The mutation of TJP2, which occurs in the first PDZ domain, reduces domain stability and ligand binding in vitro. We noted a morphological change in hepatic tight junctions. The mutation of BAAT, a bile acid-conjugating enzyme, abrogates enzyme activity; serum of individuals homozygous with respect to this mutation contains only unconjugated bile acids. Mutations in both TJP2 and BAAT may disrupt bile acid transport and circulation. Inheritance seems to be oligogenic, with genotype at BAAT modifying penetrance in individuals homozygous with respect to the mutation in TJP2.


Subject(s)
Acyltransferases/genetics , Bile Acids and Salts/blood , Malabsorption Syndromes/blood , Malabsorption Syndromes/genetics , Membrane Proteins/genetics , Mutation , Case-Control Studies , Ethnicity/genetics , Female , Genotype , Humans , Linkage Disequilibrium , Liver/pathology , Malabsorption Syndromes/pathology , Male , Pedigree , Pennsylvania , Phenotype , Tight Junctions/pathology , Zonula Occludens-2 Protein
12.
J Mol Diagn ; 25(3): 156-167, 2023 03.
Article in English | MEDLINE | ID: mdl-36563937

ABSTRACT

Nearly 14% of disease-causing germline variants result from the disruption of mRNA splicing. Most (67%) DNA variants predicted in silico to disrupt splicing are classified as variants of uncertain significance. An analytic workflow-splice effect event resolver (SPEER)-was developed and validated to use mRNA sequencing to reveal significant deviations in splicing, pinpoint the DNA variants potentially involved, and measure the deleterious effects of the altered splicing on mRNA transcripts, providing evidence for assessing the pathogenicity of the variant. SPEER was used to analyze leukocyte RNA encoding 63 hereditary cancer syndrome-related genes in 20,317 patients. Among 3563 patients (17.5%) with at least one DNA variant predicted to affect splicing, 971 (4.8%) had altered splicing with a deleterious effect on the transcript, and 40 had altered splicing due to a DNA variant located outside of the reportable range of the test. Integrating SPEER results into the interpretation of variants allowed variants of uncertain significance to be reclassified as pathogenic or likely pathogenic in 0.4%, and as benign or likely benign in 5.9%, of the 20,317 patients. SPEER-based evidence was associated with a significantly greater effect on classifications of pathogenic or likely pathogenic and benign or likely benign in nonwhite versus non-Hispanic white patients, illustrating that evidence derived from mRNA splicing analysis may help to reduce ethnic/ancestral disparities in genetic testing.


Subject(s)
Genetic Testing , Neoplastic Syndromes, Hereditary , Humans , Genetic Testing/methods , RNA Splicing , RNA, Messenger/genetics , RNA , Neoplastic Syndromes, Hereditary/genetics
14.
Proc Natl Acad Sci U S A ; 106(16): 6712-7, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19342489

ABSTRACT

Although genomewide association studies have successfully identified associations of many common single-nucleotide polymorphisms (SNPs) with common diseases, the SNPs implicated so far account for only a small proportion of the genetic variability of tested diseases. It has been suggested that common diseases may often be caused by rare alleles missed by genomewide association studies. To identify these rare alleles we need high-throughput, high-accuracy resequencing technologies. Although array-based genotyping has allowed genomewide association studies of common SNPs in tens of thousands of samples, array-based resequencing has been limited for 2 main reasons: the lack of a fully multiplexed pipeline for high-throughput sample processing, and failure to achieve sufficient performance. We have recently solved both of these problems and created a fully multiplexed high-throughput pipeline that results in high-quality data. The pipeline consists of target amplification from genomic DNA, followed by allele enrichment to generate pools of purified variant (or nonvariant) DNA and ends with interrogation of purified DNA on resequencing arrays. We have used this pipeline to resequence approximately 5 Mb of DNA (on 3 arrays) corresponding to the exons of 1,500 genes in >473 samples; in total >2,350 Mb were sequenced. In the context of this large-scale study we obtained a false positive rate of approximately 1 in 500,000 bp and a false negative rate of approximately 10%.


Subject(s)
Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA/methods , Alleles , Automation , Base Pair Mismatch , Genome, Human/genetics , Humans , Mutation/genetics , ROC Curve , Sequence Analysis, DNA/standards
15.
BMC Bioinformatics ; 11: 305, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20525369

ABSTRACT

BACKGROUND: Methylation of CpG islands within the DNA promoter regions is one mechanism that leads to aberrant gene expression in cancer. In particular, the abnormal methylation of CpG islands may silence associated genes. Therefore, using high-throughput microarrays to measure CpG island methylation will lead to better understanding of tumor pathobiology and progression, while revealing potentially new biomarkers. We have examined a recently developed high-throughput technology for measuring genome-wide methylation patterns called mTACL. Here, we propose a computational pipeline for integrating gene expression and CpG island methylation profiles to identify epigenetically regulated genes for a panel of 45 breast cancer cell lines, which is widely used in the Integrative Cancer Biology Program (ICBP). The pipeline (i) reduces the dimensionality of the methylation data, (ii) associates the reduced methylation data with gene expression data, and (iii) ranks methylation-expression associations according to their epigenetic regulation. Dimensionality reduction is performed in two steps: (i) methylation sites are grouped across the genome to identify regions of interest, and (ii) methylation profiles are clustered within each region. Associations between the clustered methylation and the gene expression data sets generate candidate matches within a fixed neighborhood around each gene. Finally, the methylation-expression associations are ranked through a logistic regression, and their significance is quantified through permutation analysis. RESULTS: Our two-step dimensionality reduction compressed 90% of the original data, reducing 137,688 methylation sites to 14,505 clusters. Methylation-expression associations produced 18,312 correspondences, which were used to further analyze epigenetic regulation. Logistic regression was used to identify 58 genes from these correspondences that showed a statistically significant negative correlation between methylation profiles and gene expression in the panel of breast cancer cell lines. Subnetwork enrichment of these genes has identified 35 common regulators with 6 or more predicted markers. In addition to identifying epigenetically regulated genes, we show evidence of differentially expressed methylation patterns between the basal and luminal subtypes. CONCLUSIONS: Our results indicate that the proposed computational protocol is a viable platform for identifying epigenetically regulated genes. Our protocol has generated a list of predictors including COL1A2, TOP2A, TFF1, and VAV3, genes whose key roles in epigenetic regulation is documented in the literature. Subnetwork enrichment of these predicted markers further suggests that epigenetic regulation of individual genes occurs in a coordinated fashion and through common regulators.


Subject(s)
Breast Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Antigens, Neoplasm/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , CpG Islands , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Genes, Tumor Suppressor , Genome-Wide Association Study , Humans , Oligonucleotide Array Sequence Analysis , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-vav/genetics , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics
16.
Hum Mutat ; 29(3): 441-50, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18186519

ABSTRACT

Mismatch repair detection (MRD) was used to screen 93 matched tumor-normal sample pairs and 22 cell lines for somatic mutations in 30 cancer relevant genes. Using a starting amount of only 150 ng of genomic DNA, we screened 102 kb of sequence for somatic mutations in colon and breast cancer. A total of 152 somatic mutations were discovered, encompassing previously reported mutations, such as BRAF V600E and KRAS G12S, G12V, and G13D, as well as novel mutations, including some in genes in which somatic mutations have not previously been reported, such as MAP2K1 and MAP2K2. The distribution of mutations ranged widely within and across tumor types. The functional significance of many of these mutations is not understood, with patterns of selection only evident in KRAS and BRAF in colon cancer. These results present a novel approach to high-throughput mutation screening using small amounts of starting material and reveal a mutation spectrum across 30 genes in a large cohort of breast and colorectal cancers.


Subject(s)
Breast Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair , DNA Mutational Analysis/methods , Mutation , Base Sequence , Cell Line, Tumor , DNA, Neoplasm/genetics , Female , Humans , Male
18.
Clin Cancer Res ; 13(1): 197-205, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17200355

ABSTRACT

PURPOSE: Our aim was to discover possible inherited factors associated with glioblastoma age at diagnosis and survival. Although new genotyping technologies allow greatly expanded exploration of such factors, they pose many challenges. EXPERIMENTAL DESIGN: In this pilot study, we (a) genotyped 112 newly diagnosed glioblastoma patients ascertained through a population-based study (group 1) with the ParAllele assay panel of approximately 10,000 nonsynonymous coding single-nucleotide polymorphisms (SNP), (b) used several statistical and bioinformatic techniques to identify 17 SNPs potentially related to either glioblastoma age at diagnosis or survival, and (c) genotyped 16 of these SNPs using conventional PCR methods in an independent group of 195 glioblastoma patients (group 2). RESULTS: In group 2, only one of the 16 SNPs, rs8057643 (located on 16p13.2), was significantly associated with glioblastoma age at diagnosis (nominal P = 0.0017; Bonferroni corrected P = 0.054). Median ages at diagnosis for those with 0, 1, or 2 T alleles were 66, 57, and 59 years in group 1 and 64, 57, and 55 years in group 2 (combined P = 0.001). Furthermore, Cox regression analyses of time to death with number of T alleles adjusted for gender and patient group yielded a hazard ratio of 0.82 (95% confidence interval, 0.68-0.98; P = 0.03). CONCLUSIONS: Although limited by a relatively small sample size, this pilot study, using well-characterized, unambiguous disease characteristics, illustrates the necessity of independent replication owing to the likelihood of false positives. Several other challenges are discussed, including attempts to incorporate information on the potential functional importance of SNPs in genome-disease association studies.


Subject(s)
Gene Expression Regulation, Neoplastic , Genome , Glioblastoma/genetics , Glioblastoma/pathology , Polymorphism, Single Nucleotide , Age Factors , Age of Onset , Aged , Alleles , Codon , Codon, Terminator , Female , Genotype , Glioblastoma/mortality , Humans , Male , Middle Aged , Pilot Projects , Treatment Outcome
19.
Hum Genomics ; 2(6): 391-402, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16848977

ABSTRACT

Association studies hold great promise for the elucidation of the genetic basis of diseases. Studies based on functional single nucleotide polymorphisms (SNPs) or on linkage disequilibrium (LD) represent two main types of designs. LD-based association studies can be comprehensive for common causative variants, but they perform poorly for rare alleles. Conversely, functional SNP-based studies are efficient because they focus on the SNPs with the highest a priori chance of being associated. Our poor ability to predict the functional effect of SNPs, however, hampers attempts to make these studies comprehensive. Recent progress in comparative genomics, and evidence that functional elements tend to lie in conserved regions, promises to change the landscape, permitting functional SNP association studies to be carried out that comprehensively assess common and rare alleles. SNP genotyping technologies are already sufficient for such studies, but studies will require continued genomic sequencing of multiple species, research on the functional role of conserved sequences and additional SNP discovery and validation efforts (including targeted SNP discovery to identify the rare alleles in functional regions). With these resources, we expect that comprehensive functional SNP association studies will soon be possible.


Subject(s)
Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Genome, Human/genetics , Humans
20.
Oncotarget ; 8(5): 8765-8774, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28060738

ABSTRACT

Recent advances in next-generation sequencing (NGS) have enabled the quantitation of circulating tumour DNA (ctDNA) encoding the clonal rearranged V(D)J immunoglobulin locus. We aimed to evaluate the clonal heterogeneity of follicular lymphoma (FL) in the tumour and the plasma at diagnosis and to assess the prognostic value of the ctDNA level. Plasma samples at diagnosis were available for 34 patients registered in the PRIMA trial (NCT00140582). One tumour clonotype or more could be detected for 29 (85%) and 25 (74%) patients, respectively, in the tumour or plasma samples. In 18 patients, several subclones were detected in the tumour (2 to 71 subclones/cases) and/or in the plasma (2 to 20 subclones/cases). In more than half of the cases, the distribution of subclones differed between the tumour and plasma samples, reflecting high clonal heterogeneity and diversity in lymphoma subclone dissemination. In multivariate analysis, a high level of ctDNA was the only independent factor associated with patients' progression-free survival (HR 4, IC 95 (1.1-37), p=.039). In conclusion, an NGS-based immunosequencing method reveals the marked clonal heterogeneity of follicular lymphoma in patients with FL, and quantification of ctDNA at diagnosis represents a potential powerful prognostic biomarker that needs to be investigated in larger cohorts.


Subject(s)
Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Lymphoma, Follicular/genetics , V(D)J Recombination , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Chi-Square Distribution , Circulating Tumor DNA/blood , Disease-Free Survival , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Lymphoma, Follicular/blood , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/immunology , Male , Middle Aged , Multivariate Analysis , Phenotype , Predictive Value of Tests , Prospective Studies , Risk Factors , Time Factors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL