Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Genome Biol ; 20(1): 168, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31416464

ABSTRACT

BACKGROUND: The classical genetic model of colorectal cancer presents APC mutations as the earliest genomic alterations, followed by KRAS and TP53 mutations. However, the timing and relative order of clonal expansion and other types of genomic alterations, such as genomic rearrangements, are still unclear. RESULTS: Here, we perform comprehensive bioinformatic analysis to dissect the relative timing of somatic genetic alterations in 63 colorectal cancers with whole-genome sequencing data. Utilizing allele fractions of somatic single nucleotide variants as molecular clocks while accounting for the presence of copy number changes and structural alterations, we identify key events in the evolution of colorectal tumors. We find that driver point mutations, gene fusions, and arm-level copy losses typically arise early in tumorigenesis; different mechanisms act on distinct genomic regions to drive DNA copy changes; and chromothripsis-clustered rearrangements previously thought to occur as a single catastrophic event-is frequent and may occur multiple times independently in the same tumor through different mechanisms. Furthermore, our computational approach reveals that, in contrast to recent studies, selection is often present on subclones and that multiple evolutionary models can operate in a single tumor at different stages. CONCLUSION: Combining these results, we present a refined tumor progression model which significantly expands our understanding of the tumorigenic process of human colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Models, Genetic , Chromothripsis , Cohort Studies , DNA Copy Number Variations/genetics , Gene Dosage , Humans , Mutation/genetics
2.
Cell Rep ; 23(1): 239-254.e6, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617664

ABSTRACT

DNA damage repair (DDR) pathways modulate cancer risk, progression, and therapeutic response. We systematically analyzed somatic alterations to provide a comprehensive view of DDR deficiency across 33 cancer types. Mutations with accompanying loss of heterozygosity were observed in over 1/3 of DDR genes, including TP53 and BRCA1/2. Other prevalent alterations included epigenetic silencing of the direct repair genes EXO5, MGMT, and ALKBH3 in ∼20% of samples. Homologous recombination deficiency (HRD) was present at varying frequency in many cancer types, most notably ovarian cancer. However, in contrast to ovarian cancer, HRD was associated with worse outcomes in several other cancers. Protein structure-based analyses allowed us to predict functional consequences of rare, recurrent DDR mutations. A new machine-learning-based classifier developed from gene expression data allowed us to identify alterations that phenocopy deleterious TP53 mutations. These frequent DDR gene alterations in many human cancers have functional consequences that may determine cancer progression and guide therapy.


Subject(s)
Genome, Human , Neoplasms/genetics , Recombinational DNA Repair , Cell Line, Tumor , DNA Damage , Gene Silencing , Humans , Loss of Heterozygosity , Machine Learning , Mutation , Neoplasms/classification , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Cancer Discov ; 8(6): 730-749, 2018 06.
Article in English | MEDLINE | ID: mdl-29510987

ABSTRACT

To understand the genetic drivers of immune recognition and evasion in colorectal cancer, we analyzed 1,211 colorectal cancer primary tumor samples, including 179 classified as microsatellite instability-high (MSI-high). This set includes The Cancer Genome Atlas colorectal cancer cohort of 592 samples, completed and analyzed here. MSI-high, a hypermutated, immunogenic subtype of colorectal cancer, had a high rate of significantly mutated genes in important immune-modulating pathways and in the antigen presentation machinery, including biallelic losses of B2M and HLA genes due to copy-number alterations and copy-neutral loss of heterozygosity. WNT/ß-catenin signaling genes were significantly mutated in all colorectal cancer subtypes, and activated WNT/ß-catenin signaling was correlated with the absence of T-cell infiltration. This large-scale genomic analysis of colorectal cancer demonstrates that MSI-high cases frequently undergo an immunoediting process that provides them with genetic events allowing immune escape despite high mutational load and frequent lymphocytic infiltration and, furthermore, that colorectal cancer tumors have genetic and methylation events associated with activated WNT signaling and T-cell exclusion.Significance: This multi-omic analysis of 1,211 colorectal cancer primary tumors reveals that it should be possible to better monitor resistance in the 15% of cases that respond to immune blockade therapy and also to use WNT signaling inhibitors to reverse immune exclusion in the 85% of cases that currently do not. Cancer Discov; 8(6); 730-49. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 663.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Tumor Escape , DNA Copy Number Variations , DNA Methylation , Germ-Line Mutation , HLA Antigens/genetics , Humans , Loss of Heterozygosity , Microsatellite Instability , Wnt Signaling Pathway , beta 2-Microglobulin/genetics
5.
Cell Rep ; 18(11): 2780-2794, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297679

ABSTRACT

Cholangiocarcinoma (CCA) is an aggressive malignancy of the bile ducts, with poor prognosis and limited treatment options. Here, we describe the integrated analysis of somatic mutations, RNA expression, copy number, and DNA methylation by The Cancer Genome Atlas of a set of predominantly intrahepatic CCA cases and propose a molecular classification scheme. We identified an IDH mutant-enriched subtype with distinct molecular features including low expression of chromatin modifiers, elevated expression of mitochondrial genes, and increased mitochondrial DNA copy number. Leveraging the multi-platform data, we observed that ARID1A exhibited DNA hypermethylation and decreased expression in the IDH mutant subtype. More broadly, we found that IDH mutations are associated with an expanded histological spectrum of liver tumors with molecular features that stratify with CCA. Our studies reveal insights into the molecular pathogenesis and heterogeneity of cholangiocarcinoma and provide classification information of potential therapeutic significance.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Genomics/methods , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/enzymology , Cholangiocarcinoma/enzymology , Chromatin/metabolism , DNA Methylation/genetics , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Mitochondria/metabolism , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics
6.
Cell Rep ; 14(10): 2476-89, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947078

ABSTRACT

On the basis of multidimensional and comprehensive molecular characterization (including DNA methalylation and copy number, RNA, and protein expression), we classified 894 renal cell carcinomas (RCCs) of various histologic types into nine major genomic subtypes. Site of origin within the nephron was one major determinant in the classification, reflecting differences among clear cell, chromophobe, and papillary RCC. Widespread molecular changes associated with TFE3 gene fusion or chromatin modifier genes were present within a specific subtype and spanned multiple subtypes. Differences in patient survival and in alteration of specific pathways (including hypoxia, metabolism, MAP kinase, NRF2-ARE, Hippo, immune checkpoint, and PI3K/AKT/mTOR) could further distinguish the subtypes. Immune checkpoint markers and molecular signatures of T cell infiltrates were both highest in the subtype associated with aggressive clear cell RCC. Differences between the genomic subtypes suggest that therapeutic strategies could be tailored to each RCC disease subset.


Subject(s)
Carcinoma, Renal Cell/pathology , Genomics , Kidney Neoplasms/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Chromatin/metabolism , Gene Expression Profiling , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , MicroRNAs/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction/genetics , Survival Rate , TOR Serine-Threonine Kinases/metabolism
7.
Cell Rep ; 14(4): 907-919, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26804919

ABSTRACT

The ampulla of Vater is a complex cellular environment from which adenocarcinomas arise to form a group of histopathologically heterogenous tumors. To evaluate the molecular features of these tumors, 98 ampullary adenocarcinomas were evaluated and compared to 44 distal bile duct and 18 duodenal adenocarcinomas. Genomic analyses revealed mutations in the WNT signaling pathway among half of the patients and in all three adenocarcinomas irrespective of their origin and histological morphology. These tumors were characterized by a high frequency of inactivating mutations of ELF3, a high rate of microsatellite instability, and common focal deletions and amplifications, suggesting common attributes in the molecular pathogenesis are at play in these tumors. The high frequency of WNT pathway activating mutation, coupled with small-molecule inhibitors of ß-catenin in clinical trials, suggests future treatment decisions for these patients may be guided by genomic analysis.


Subject(s)
Adenocarcinoma/genetics , DNA-Binding Proteins/genetics , Duodenal Neoplasms/genetics , Mutation , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Wnt Signaling Pathway , Adenocarcinoma/metabolism , Ampulla of Vater/pathology , Base Sequence , Duodenal Neoplasms/metabolism , Genomic Instability , Humans , Microsatellite Repeats , Molecular Sequence Data , Pancreatic Neoplasms/metabolism
8.
Cell ; 164(3): 538-49, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26806129

ABSTRACT

Mutational processes constantly shape the somatic genome, leading to immunity, aging, cancer, and other diseases. When cancer is the outcome, we are afforded a glimpse into these processes by the clonal expansion of the malignant cell. Here, we characterize a less explored layer of the mutational landscape of cancer: mutational asymmetries between the two DNA strands. Analyzing whole-genome sequences of 590 tumors from 14 different cancer types, we reveal widespread asymmetries across mutagenic processes, with transcriptional ("T-class") asymmetry dominating UV-, smoking-, and liver-cancer-associated mutations and replicative ("R-class") asymmetry dominating POLE-, APOBEC-, and MSI-associated mutations. We report a striking phenomenon of transcription-coupled damage (TCD) on the non-transcribed DNA strand and provide evidence that APOBEC mutagenesis occurs on the lagging-strand template during DNA replication. As more genomes are sequenced, studying and classifying their asymmetries will illuminate the underlying biological mechanisms of DNA damage and repair.


Subject(s)
DNA Damage , DNA Mutational Analysis , DNA Repair , Neoplasms/genetics , DNA Replication , Genome, Human , Genome-Wide Association Study , Humans , Mutation , Neoplasms/pathology , Transcription, Genetic
9.
N Engl J Med ; 374(2): 135-45, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26536169

ABSTRACT

BACKGROUND: Papillary renal-cell carcinoma, which accounts for 15 to 20% of renal-cell carcinomas, is a heterogeneous disease that consists of various types of renal cancer, including tumors with indolent, multifocal presentation and solitary tumors with an aggressive, highly lethal phenotype. Little is known about the genetic basis of sporadic papillary renal-cell carcinoma, and no effective forms of therapy for advanced disease exist. METHODS: We performed comprehensive molecular characterization of 161 primary papillary renal-cell carcinomas, using whole-exome sequencing, copy-number analysis, messenger RNA and microRNA sequencing, DNA-methylation analysis, and proteomic analysis. RESULTS: Type 1 and type 2 papillary renal-cell carcinomas were shown to be different types of renal cancer characterized by specific genetic alterations, with type 2 further classified into three individual subgroups on the basis of molecular differences associated with patient survival. Type 1 tumors were associated with MET alterations, whereas type 2 tumors were characterized by CDKN2A silencing, SETD2 mutations, TFE3 fusions, and increased expression of the NRF2-antioxidant response element (ARE) pathway. A CpG island methylator phenotype (CIMP) was observed in a distinct subgroup of type 2 papillary renal-cell carcinomas that was characterized by poor survival and mutation of the gene encoding fumarate hydratase (FH). CONCLUSIONS: Type 1 and type 2 papillary renal-cell carcinomas were shown to be clinically and biologically distinct. Alterations in the MET pathway were associated with type 1, and activation of the NRF2-ARE pathway was associated with type 2; CDKN2A loss and CIMP in type 2 conveyed a poor prognosis. Furthermore, type 2 papillary renal-cell carcinoma consisted of at least three subtypes based on molecular and phenotypic features. (Funded by the National Institutes of Health.).


Subject(s)
Carcinoma, Papillary/metabolism , Kidney Neoplasms/metabolism , Mutation , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-met/metabolism , Carcinoma, Papillary/genetics , CpG Islands/physiology , DNA Methylation , Humans , Kidney Neoplasms/genetics , MicroRNAs/chemistry , NF-E2-Related Factor 2/genetics , Phenotype , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/chemistry , RNA, Neoplasm/chemistry , Sequence Analysis, RNA , Signal Transduction/physiology
10.
Nat Genet ; 46(12): 1267-73, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25362482

ABSTRACT

Diverse epidemiological factors are associated with hepatocellular carcinoma (HCC) prevalence in different populations. However, the global landscape of the genetic changes in HCC genomes underpinning different epidemiological and ancestral backgrounds still remains uncharted. Here a collection of data from 503 liver cancer genomes from different populations uncovered 30 candidate driver genes and 11 core pathway modules. Furthermore, a collaboration of two large-scale cancer genome projects comparatively analyzed the trans-ancestry substitution signatures in 608 liver cancer cases and identified unique mutational signatures that predominantly contribute to Asian cases. This work elucidates previously unexplored ancestry-associated mutational processes in HCC development. A combination of hotspot TERT promoter mutation, TERT focal amplification and viral genome integration occurs in more than 68% of cases, implicating TERT as a central and ancestry-independent node of hepatocarcinogenesis. Newly identified alterations in genes encoding metabolic enzymes, chromatin remodelers and a high proportion of mTOR pathway activations offer potential therapeutic and diagnostic opportunities.


Subject(s)
Carcinoma, Hepatocellular/ethnology , Carcinoma, Hepatocellular/genetics , Genome, Human , Liver Neoplasms/ethnology , Liver Neoplasms/genetics , Mutation , Algorithms , Asian People , Carcinoma, Hepatocellular/epidemiology , CpG Islands , DNA Mutational Analysis , Exome , Gene Expression Regulation, Neoplastic , Genome, Viral , Hepacivirus/genetics , Hepatitis B virus/genetics , Humans , Japan , Liver Neoplasms/epidemiology , Models, Statistical , Principal Component Analysis , TOR Serine-Threonine Kinases/genetics , Telomerase/genetics , United States , White People
11.
Clin Cancer Res ; 20(24): 6582-92, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25303977

ABSTRACT

PURPOSE: Aggressive cutaneous squamous cell carcinoma (cSCC) is often a disfiguring and lethal disease. Very little is currently known about the mutations that drive aggressive cSCC. EXPERIMENTAL DESIGN: Whole-exome sequencing was performed on 39 cases of aggressive cSCC to identify driver genes and novel therapeutic targets. Significantly, mutated genes were identified with MutSig or complementary methods developed to specifically identify candidate tumor suppressors based upon their inactivating mutation bias. RESULTS: Despite the very high-mutational background caused by UV exposure, 23 candidate drivers were identified, including the well-known cancer-associated genes TP53, CDKN2A, NOTCH1, AJUBA, HRAS, CASP8, FAT1, and KMT2C (MLL3). Three novel candidate tumor suppressors with putative links to cancer or differentiation, NOTCH2, PARD3, and RASA1, were also identified as possible drivers in cSCC. KMT2C mutations were associated with poor outcome and increased bone invasion. CONCLUSIONS: The mutational spectrum of cSCC is similar to that of head and neck squamous cell carcinoma and dominated by tumor-suppressor genes. These results improve the foundation for understanding this disease and should aid in identifying and treating aggressive cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Mutation , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/mortality , Cluster Analysis , Computational Biology , DNA Copy Number Variations , Disease Progression , Exome , Genomics , High-Throughput Nucleotide Sequencing , Humans , Prognosis
12.
Genome Res ; 24(11): 1740-50, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25228659

ABSTRACT

Tumors with somatic mutations in the proofreading exonuclease domain of DNA polymerase epsilon (POLE-exo*) exhibit a novel mutator phenotype, with markedly elevated TCT→TAT and TCG→TTG mutations and overall mutation frequencies often exceeding 100 mutations/Mb. Here, we identify POLE-exo* tumors in numerous cancers and classify them into two groups, A and B, according to their mutational properties. Group A mutants are found only in POLE, whereas Group B mutants are found in POLE and POLD1 and appear to be nonfunctional. In Group A, cell-free polymerase assays confirm that mutations in the exonuclease domain result in high mutation frequencies with a preference for C→A mutation. We describe the patterns of amino acid substitutions caused by POLE-exo* and compare them to other tumor types. The nucleotide preference of POLE-exo* leads to increased frequencies of recurrent nonsense mutations in key tumor suppressors such as TP53, ATM, and PIK3R1. We further demonstrate that strand-specific mutation patterns arise from some of these POLE-exo* mutants during genome duplication. This is the first direct proof of leading strand-specific replication by human POLE, which has only been demonstrated in yeast so far. Taken together, the extremely high mutation frequency and strand specificity of mutations provide a unique identifier of eukaryotic origins of replication.


Subject(s)
DNA Polymerase II/genetics , DNA Replication , Exonucleases/genetics , Mutation, Missense , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Class Ia Phosphatidylinositol 3-Kinase , Codon, Nonsense , DNA Mutational Analysis , DNA Polymerase II/chemistry , DNA Polymerase II/metabolism , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Databases, Genetic , Exonucleases/chemistry , Exonucleases/metabolism , Genome-Wide Association Study , Humans , Microsatellite Instability , Models, Molecular , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Replication Origin/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Pathol ; 232(5): 522-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374933

ABSTRACT

Mutations in the TP53 tumour suppressor gene occur in half of all human cancers, indicating its critical importance in inhibiting cancer development. Despite extensive studies, the mechanisms by which mutant p53 enhances tumour progression remain only partially understood. Here, using data from the Cancer Genome Atlas (TCGA), genomic and transcriptomic analyses were performed on 2256 tumours from 10 human cancer types. We show that tumours with TP53 mutations have altered gene expression profiles compared to tumours retaining two wild-type TP53 alleles. Among 113 known p53-up-regulated target genes identified from cell culture assays, 10 were consistently up-regulated in at least eight of 10 cancer types that retain both copies of wild-type TP53. RPS27L, CDKN1A (p21(CIP1)) and ZMAT3 were significantly up-regulated in all 10 cancer types retaining wild-type TP53. Using this p53-based expression analysis as a discovery tool, we used cell-based assays to identify five novel p53 target genes from genes consistently up-regulated in wild-type p53 cancers. Global gene expression analyses revealed that cell cycle regulatory genes and transcription factors E2F1, MYBL2 and FOXM1 were disproportionately up-regulated in many TP53 mutant cancer types. Finally, > 93% of tumours with a TP53 mutation exhibited greatly reduced wild-type p53 messenger expression, due to loss of heterozygosity or copy neutral loss of heterozygosity, supporting the concept of p53 as a recessive tumour suppressor. The data indicate that tumours with wild-type TP53 retain some aspects of p53-mediated growth inhibitory signalling through activation of p53 target genes and suppression of cell cycle regulatory genes.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Mutation , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gene Expression Profiling/methods , Humans , Loss of Heterozygosity , Neoplasms/metabolism , RNA, Messenger/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
14.
J Pathol ; 229(1): 99-110, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22899370

ABSTRACT

Approximately 15% of colorectal carcinomas (CRCs) exhibit a hypermutated genotype accompanied by high levels of microsatellite instability (MSI-H) and defects in DNA mismatch repair. These tumours, unlike the majority of colorectal carcinomas, are often diploid, exhibit frequent epigenetic silencing of the MLH1 DNA mismatch repair gene, and have a better clinical prognosis. As an adjunct study to The Cancer Genome Atlas consortium that recently analysed 224 colorectal cancers by whole exome sequencing, we compared the 35 CRCs (15.6%) with a hypermutated genotype to those with a non-hypermutated genotype. We found that 22 (63%) of the hypermutated CRCs exhibited transcriptional silencing of the MLH1 gene, a high frequency of BRAF V600E gene mutations, and infrequent APC and KRAS mutations, a mutational pattern significantly different from their non-hypermutated counterparts. However, the remaining 13 (37%) hypermutated CRCs lacked MLH1 silencing, contained tumours with the highest mutation rates ('ultramutated' CRCs), and exhibited higher incidences of APC and KRAS mutations, but infrequent BRAF mutations. These patterns were confirmed in an independent validation set of 250 exome-sequenced CRCs. Analysis of mRNA and microRNA expression signatures revealed that hypermutated CRCs with MLH1 silencing had greatly reduced levels of WNT signalling and increased BRAF signalling relative to non-hypermutated CRCs. Our findings suggest that hypermutated CRCs include one subgroup with fundamentally different pathways to malignancy than the majority of CRCs. Examination of MLH1 expression status and frequencies of APC, KRAS, and BRAF mutation in CRC may provide a useful diagnostic tool that could supplement the standard microsatellite instability assays and influence therapeutic decisions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , Gene Silencing , Mutation , Nuclear Proteins/genetics , Adenomatous Polyposis Coli Protein/genetics , Carcinoma/enzymology , Carcinoma/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Frequency , Genetic Predisposition to Disease , Humans , MicroRNAs/metabolism , Microsatellite Instability , MutL Protein Homolog 1 , Oligonucleotide Array Sequence Analysis , Phenotype , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , RNA, Messenger/metabolism , Wnt Signaling Pathway/genetics , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...