--- title: "Moonlight: an approach to identify multiple role of biomarkers as oncogene or tumorsuppressor in different tumor types and stages." author: "Antonio Colaprico^+^ , Catharina Olsen^+^, Claudia Cava, Thilde Terkelsen, Laura Cantini, Gloria Bertoli, Andre Olsen, Andrei Zinovyev, Emmanuel Barillot, Isabella Castiglioni, Elena Papaleo, Gianluca Bontempi" subtitle: ^+^ These authors contributed equally to the paper as first authors. date: "`r Sys.Date()`" output: BiocStyle::html_document: toc: true number_sections: false toc_depth: 2 highlight: haddock references: - id: ref1 title: Orchestrating high-throughput genomic analysis with Bioconductor author: - family: Huber, Wolfgang and Carey, Vincent J and Gentleman, Robert and Anders, Simon and Carlson, Marc and Carvalho, Benilton S and Bravo, Hector Corrada and Davis, Sean and Gatto, Laurent and Girke, Thomas and others given: journal: Nature methods volume: 12 number: 2 pages: 115-121 issued: year: 2015 - id: ref2 title: GC-content normalization for RNA-Seq data author: - family: Risso, Davide and Schwartz, Katja and Sherlock, Gavin and Dudoit, Sandrine given: journal: BMC bioinformatics volume: 12 number: 1 pages: 480 issued: year: 2011 - id: ref3 title: Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments author: - family: Bullard, James H and Purdom, Elizabeth and Hansen, Kasper D and Dudoit, Sandrine given: journal: BMC bioinformatics volume: 11 number: 1 pages: 94 issued: year: 2010 - id: ref4 title: Inferring regulatory element landscapes and transcription factor networks from cancer methylomes author: - family: Yao, L., Shen, H., Laird, P. W., Farnham, P. J., & Berman, B. P. given: journal: Genome biology volume: 16 number: 1 pages: 105 issued: year: 2015 - id: ref5 title: Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments author: - family: James H Bullard, Elizabeth Purdom, Kasper D Hansen and Sandrine Dudoit given: journal: BMC Bioinformatics volume: 11 number: 1 pages: 94 issued: year: 2010 - id: ref6 title: GC-content normalization for RNA-Seq data author: - family: Risso, D., Schwartz, K., Sherlock, G., & Dudoit, S. given: journal: BMC Bioinformatics volume: 12 number: 1 pages: 480 issued: year: 2011 - id: ref7 title: Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma author: - family: Noushmehr, H., Weisenberger, D.J., Diefes, K., Phillips, H.S., Pujara, K., Berman, B.P., Pan, F., Pelloski, C.E., Sulman, E.P., Bhat, K.P. et al. given: journal: Cancer cell volume: 17 number: 5 pages: 510-522 issued: year: 2010 - id: ref8 title: Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma author: - family: Ceccarelli, Michele and Barthel, Floris P and Malta, Tathiane M and Sabedot, Thais S and Salama, Sofie R and Murray, Bradley A and Morozova, Olena and Newton, Yulia and Radenbaugh, Amie and Pagnotta, Stefano M and others given: journal: Cell URL: "http://doi.org/10.1016/j.cell.2015.12.028" DOI: "10.1016/j.cell.2015.12.028" volume: 164 number: 3 pages: 550-563 issued: year: 2016 - id: ref9 title: Comprehensive molecular profiling of lung adenocarcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature13385" DOI: "10.1038/nature13385" volume: 511 number: 7511 pages: 543-550 issued: year: 2014 - id: ref10 title: Comprehensive molecular characterization of gastric adenocarcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature13480" DOI: "10.1038/nature13480" issued: year: 2014 - id: ref11 title: Comprehensive molecular portraits of human breast tumours author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature11412" DOI: "10.1038/nature11412" volume: 490 number: 7418 pages: 61-70 issued: year: 2012 - id: ref12 title: Comprehensive molecular characterization of human colon and rectal cancer author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature11252" DOI: "10.1038/nature11252" volume: 487 number: 7407 pages: 330-337 issued: year: 2012 - id: ref13 title: Genomic classification of cutaneous melanoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Cell URL: "http://doi.org/10.1016/j.cell.2015.05.044" DOI: "10.1016/j.cell.2015.05.044" volume: 161 number: 7 pages: 1681-1696 issued: year: 2015 - id: ref14 title: Comprehensive genomic characterization of head and neck squamous cell carcinomas author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature14129" DOI: "10.1038/nature14129" volume: 517 number: 7536 pages: 576-582 issued: year: 2015 - id: ref15 title: The somatic genomic landscape of chromophobe renal cell carcinoma author: - family: Davis, Caleb F and Ricketts, Christopher J and Wang, Min and Yang, Lixing and Cherniack, Andrew D and Shen, Hui and Buhay, Christian and Kang, Hyojin and Kim, Sang Cheol and Fahey, Catherine C and others given: journal: Cancer Cell URL: "http://doi.org/10.1016/j.ccr.2014.07.014" DOI: "10.1016/j.ccr.2014.07.014" volume: 26 number: 3 pages: 319-330 issued: year: 2014 - id: ref16 title: Comprehensive genomic characterization of squamous cell lung cancers author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature11404" DOI: "10.1038/nature11404" volume: 489 number: 7417 pages: 519-525 issued: year: 2012 - id: ref17 title: Integrated genomic characterization of endometrial carcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature12113" DOI: "10.1038/nature12113" volume: 497 number: 7447 pages: 67-73 issued: year: 2013 - id: ref18 title: Integrated genomic characterization of papillary thyroid carcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Cell URL: "http://doi.org/10.1016/j.cell.2014.09.050" DOI: "10.1016/j.cell.2014.09.050" volume: 159 number: 3 pages: 676-690 issued: year: 2014 - id: ref19 title: The molecular taxonomy of primary prostate cancer author: - family: Cancer Genome Atlas Research Network and others given: journal: Cell URL: "http://doi.org/10.1016/j.cell.2015.10.025" DOI: "10.1016/j.cell.2015.10.025" volume: 163 number: 4 pages: 1011-1025 issued: year: 2015 - id: ref20 title: Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma author: - family: Linehan, W Marston and Spellman, Paul T and Ricketts, Christopher J and Creighton, Chad J and Fei, Suzanne S and Davis, Caleb and Wheeler, David A and Murray, Bradley A and Schmidt, Laura and Vocke, Cathy D and others given: journal: NEW ENGLAND JOURNAL OF MEDICINE URL: "http://doi.org/10.1056/NEJMoa1505917" DOI: "10.1056/NEJMoa1505917" volume: 374 number: 2 pages: 135-145 issued: year: 2016 - id: ref21 title: Comprehensive molecular characterization of clear cell renal cell carcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Nature URL: "http://doi.org/10.1038/nature12222" DOI: "10.1038/nature12222" volume: 499 number: 7456 pages: 43-49 issued: year: 2013 - id: ref22 title: Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma author: - family: Cancer Genome Atlas Research Network and others given: journal: Cancer Cell URL: "http://dx.doi.org/10.1016/j.ccell.2016.04.002" DOI: "10.1016/j.ccell.2016.04.002" volume: 29 pages: 43-49 issued: year: 2016 - id: ref23 title: Complex heatmaps reveal patterns and correlations in multidimensional genomic data author: - family: Gu, Zuguang and Eils, Roland and Schlesner, Matthias given: journal: Bioinformatics URL: "http://dx.doi.org/10.1016/j.ccell.2016.04.002" DOI: "10.1016/j.ccell.2016.04.002" pages: "btw313" issued: year: 2016 - id: ref24 title: "TCGA Workflow: Analyze cancer genomics and epigenomics data using Bioconductor packages" author: - family: Silva, TC and Colaprico, A and Olsen, C and D'Angelo, F and Bontempi, G and Ceccarelli, M and Noushmehr, H given: journal: F1000Research URL: "http://dx.doi.org/10.12688/f1000research.8923.1" DOI: "10.12688/f1000research.8923.1" volume: 5 number: 1542 issued: year: 2016 - id: ref25 title: "TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data" author: - family: Colaprico, Antonio and Silva, Tiago C. and Olsen, Catharina and Garofano, Luciano and Cava, Claudia and Garolini, Davide and Sabedot, Thais S. and Malta, Tathiane M. and Pagnotta, Stefano M. and Castiglioni, Isabella and Ceccarelli, Michele and Bontempi, Gianluca and Noushmehr, Houtan given: journal: Nucleic Acids Research URL: "http://dx.doi.org/10.1093/nar/gkv1507" DOI: "10.1093/nar/gkv1507" volume: 44 number: 8 pages: e71 issued: year: 2016 vignette: > %\VignetteIndexEntry{Vignette Title} %\VignetteEngine{knitr::rmarkdown} \usepackage[utf8]{inputenc} --- ```{r setup, include=FALSE} knitr::opts_chunk$set(dpi = 300) knitr::opts_chunk$set(cache=FALSE) ``` ```{r, echo = FALSE,hide=TRUE, message=FALSE,warning=FALSE} devtools::load_all(".") ``` # Abstract In order to make light of cancer development, it is crucial to understand which genes play a role in the mechanisms linked to this disease and moreover which role that is. Commonly biological processes such as proliferation and apoptosis have been linked to cancer progression. Based on expression data we perform functional enrichment analysis, infer gene regulatory networks and upstream regulator analysis to score the importance of well-known biological processes with respect to the studied cancer. We then use these scores to predict two specific roles: genes that act as tumor suppressor genes (TSGs) and genes that act as oncogenes (OCGs). This methodology not only allows us to identify genes with dual role (TSG in one cancer type and OCG in another) but also to elucidate the underlying biological processes. # Introduction Cancer development is influenced by mutations in two distinctly different categories of genes, known as tumor suppressor genes (TSG) and oncogenes (OCG). The occurrence of mutations in genes of the first category leads to faster cell proliferation while mutations in genes of second category increases or changes their function. We propose `MoonlightR` a new approach to define TSGs and OCGs based on functional enrichment analysis, infer gene regulatory networks and upstream regulator analysis to score the importance of well-known biological processes with respect to the studied cancer. # Moonlight's pipeline Moonlight's pipeline is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} library(png) library(grid) img <- readPNG("Moonlight_Pipeline.png") grid.raster(img) ``` # Moonlight's proposed workflow The proposed pipeline consists of following eight steps: 1. **getDataTCGA** \& **getDataGEO** for Data collection: expression levels of genes in all samples obtained with IlluminaHiSeq RNASeqV2 in 18 normal tissues (NT) and 18 cancer tissues (CT) according to TCGA criteria, and GEO data set matched to one of the 18 given TCGA cancer types as described in following Table TCGA / GEO. 2. **DPA** Differential Phenotype Analysis (DEA) to identify genes or probes that are different significantly with two phenotypes such as normal and tumor, or normal and stageI, normal and molecular subtype. 3. **FEA** Functional Enrichment Analysis (EA), using Fisher's test, to identify gene sets (with biological functions linked to cancer1) significantly enriched by RG. 4. **GRN** Gene regulatory network inferred between each single DEG (sDEG) and all genes by means of mutual information, obtaining for each DEG a list of regulated genes (RG). 5. **URA** Upstream Regulator Analysis for DEGs in each enriched gene set, we applied z-score being the ratio between the sum of all predicted effects for all the gene involved in the specific function and the square-root of the number of all genes. 6. **PRA** Pattern recognition analysis identifies candidate TCGs (down) and OCGs (up). We either use user defined biological processes or random forests. 7. We applied the above procedure to multiple cancer types to obtain cancer-specific lists of TCGs and OCGs. We compared the lists for each cancer: if a sDEG was TSG in a cancer and OCG in another we defined it as dual-role TSG-OCG. Otherwise if we found a sDEG defined as OCG or TSG only in one tissue we defined it tissue specific biomarker. 8. We use the COSMIC database to define a list of gold standard TSG and OCGs to assess the accuracy of the proposed method. 1 For the devel version of MoonlightR we use a short extract of ten biological functions from QIAGEN'S Ingenuity Pathway Analysis (IPA). We are still working to integrate the\Biocpkg{ReactomePA} package. # Installation To install use the code below. ```{r, eval = FALSE} if (!requireNamespace("BiocManager", quietly=TRUE)) install.packages("BiocManager") BiocManager::install("MoonlightR") ``` ## Citation Please cite TCGAbiolinks package: * "TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data." Nucleic acids research (2015): [gkv1507](http://dx.doi.org/doi:10.1093/nar/gkv1507). [@ref25] Related publications to this package: * "TCGA Workflow: Analyze cancer genomics and epigenomics data using Bioconductor packages". F1000Research [10.12688/f1000research.8923.1](http://dx.doi.org/doi:10.12688/f1000research.8923.1) [@ref24] # `Download`: Get TCGA data You can search TCGA data using the `getDataTCGA` function. ## `getDataTCGA`: Search by cancer type and data type [Gene Expression] The user can query and download the cancer types supported by TCGA, using the function `getDataTCGA`: In this example we used LUAD gene expression data with only 4 samples to reduce time downloading. ```{r, eval = FALSE} dataFilt <- getDataTCGA(cancerType = "LUAD", dataType = "Gene expression", directory = "data", nSample = 4) ``` ## `getDataTCGA`: Search by cancer type and data type [Methylation] The user can also query and download methylation data using the function `getDataTCGA`: ```{r, eval = FALSE} dataFilt <- getDataTCGA(cancerType = "BRCA", dataType = "Methylation", directory = "data",nSample = 4) ``` # `Download`: Get GEO data You can search GEO data using the `getDataGEO` function. GEO_TCGAtab: a 18x12 matrix that provides the GEO data set we matched to one of the 18 given TCGA cancer types ```{r, eval = TRUE, echo = TRUE} knitr::kable(GEO_TCGAtab, digits = 2, caption = "Table with GEO data set matched to one of the 18 given TCGA cancer types ", row.names = TRUE) ``` ## `getDataGEO`: Search by cancer type and data type [Gene Expression] The user can query and download the cancer types supported by GEO, using the function `getDataGEO`: ```{r, eval = FALSE , echo = TRUE, results='hide', warning = FALSE, message = FALSE} dataFilt <- getDataGEO(GEOobject = "GSE20347",platform = "GPL571") ``` ```{r, eval = FALSE, echo = TRUE, results='hide', warning = FALSE, message = FALSE} dataFilt <- getDataGEO(TCGAtumor = "ESCA") ``` # `Analysis`: To analyze TCGA data ## `DPA`: Differential Phenotype Analysis Differential phenotype analysis is able to identify genes or probes that are significantly different between two phenotypes such as normal vs. tumor, or normal vs. stageI, normal vs. molecular subtype. For gene expression data, DPA is running a differential expression analysis (DEA) to identify differentially expressed genes (DEGs) using the `TCGAanalyze_DEA` function from \Biocpkg{TCGAbiolinks}. For methylation data, DPA is running a differentially methylated regions analysis (DMR) to identify differentially methylated CpG sites using the `TCGAanalyze_DMR` the `TCGAanalyze_DMR` function from \Biocpkg{TCGAbiolinks}. ```{r, eval = FALSE, message=FALSE, results='hide', warning=FALSE} dataDEGs <- DPA(dataFilt = dataFilt, dataType = "Gene expression") ``` For gene expression data, DPA dealing with GEO data is running a differential expression analysis (DEA) to identify differentially expressed genes (DEGs) using to the `eBayes` and `topTable` functions from \Biocpkg{limma}. ```{r, eval = FALSE, echo = TRUE, hide=TRUE, results='hide', warning = FALSE, message = FALSE} data(GEO_TCGAtab) DataAnalysisGEO<- "../GEO_dataset/" i<-5 cancer <- GEO_TCGAtab$Cancer[i] cancerGEO <- GEO_TCGAtab$Dataset[i] cancerPLT <-GEO_TCGAtab$Platform[i] fileCancerGEO <- paste0(cancer,"_GEO_",cancerGEO,"_",cancerPLT, ".RData") dataFilt <- getDataGEO(TCGAtumor = cancer) xContrast <- c("G1-G0") GEOdegs <- DPA(dataConsortium = "GEO", gset = dataFilt , colDescription = "title", samplesType = c(GEO_TCGAtab$GEO_Normal[i], GEO_TCGAtab$GEO_Tumor[i]), fdr.cut = 0.01, logFC.cut = 1, gsetFile = paste0(DataAnalysisGEO,fileCancerGEO)) ``` We can visualize those differentially expressed genes (DEGs) with a volcano plot using the `TCGAVisualize_volcano` function from \Biocpkg{TCGAbiolinks.}. ```{r, eval = TRUE, echo = TRUE} library(TCGAbiolinks) TCGAVisualize_volcano(DEGsmatrix$logFC, DEGsmatrix$FDR, filename = "DEGs_volcano.png", x.cut = 1, y.cut = 0.05, names = rownames(DEGsmatrix), color = c("black","red","dodgerblue3"), names.size = 2, show.names = "highlighted", highlight = c("gene1","gene2"), xlab = " Gene expression fold change (Log2)", legend = "State", title = "Volcano plot (Normal NT vs Tumor TP)", width = 10) ``` The figure generated by the code above is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} img <- readPNG("DEGs_volcano.png") grid.raster(img) ``` ## `FEA`: Functional Enrichment Analysis The user can perform a functional enrichment analysis using the function `FEAcomplete`. For each DEG in the gene set a z-score is calculated. This score indicates how the genes act in the gene set. ```{r, eval = TRUE, echo = TRUE, results='hide'} data(DEGsmatrix) dataFEA <- FEA(DEGsmatrix = DEGsmatrix) ``` The output can be visualized with a FEA plot. ## `FEAplot`: Functional Enrichment Analysis Plot The user can plot the result of a functional enrichment analysis using the function `plotFEA`. A negative z-score indicates that the process' activity is decreased. A positive z-score indicates that the process' activity is increased. ```{r, eval = TRUE, echo = TRUE, message=FALSE, results='hide', warning=FALSE} plotFEA(dataFEA = dataFEA, additionalFilename = "_exampleVignette", height = 20, width = 10) ``` The figure generated by the above code is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} img <- readPNG("FEAplot.png") grid.raster(img) ``` ## `GRN`: Gene Regulatory Network The user can perform a gene regulatory network analysis using the function `GRN` which infers the network using the parmigene package. ```{r, eval = TRUE} dataGRN <- GRN(TFs = rownames(DEGsmatrix)[1:100], normCounts = dataFilt, nGenesPerm = 10,kNearest = 3,nBoot = 10) ``` ## `URA`: Upstream Regulator Analysis The user can perform upstream regulator analysis using the function `URA`. This function is applied to each DEG in the enriched gene set and its neighbors in the GRN. ```{r, eval = FALSE, echo = TRUE, results='hide'} data(dataGRN) data(DEGsmatrix) dataFEA <- FEA(DEGsmatrix = DEGsmatrix) BPselected <- dataFEA$Diseases.or.Functions.Annotation[1:5] dataURA <- URA(dataGRN = dataGRN, DEGsmatrix = DEGsmatrix, BPname = BPselected, nCores=1) ``` ## `PRA`: Pattern Regognition Analysis The user can retrieve TSG/OCG candidates using either selected biological processes or a random forest classifier trained on known COSMIC OCGs/TSGs. ```{r, eval = TRUE} data(dataURA) dataDual <- PRA(dataURA = dataURA, BPname = c("apoptosis","proliferation of cells"), thres.role = 0) ``` ## `plotNetworkHive`: GRN hive visualization taking into account Cosmic cancer genes In the following plot the nodes are separated into three groups: known tumor suppressor genes (yellow), known oncogenes (green) and the rest (gray). ```{r, eval = TRUE, echo = TRUE, results='hide', warning = FALSE, message = FALSE} data(knownDriverGenes) data(dataGRN) plotNetworkHive(dataGRN, knownDriverGenes, 0.55) ``` # TCGA Downstream Analysis: Case Studies ### Introduction This vignette shows a complete workflow of the 'MoonlightR' package except for the data download. The code is divided into three case study: * 1. Downstream analysis LUAD using RNA expression data * 2. Expression pipeline Pan Cancer with five cancer types * 3. Expression pipeline with stages I II III IV (BRCA) ## Case study n. 1: Downstream analysis LUAD ```{r,eval = FALSE,echo=TRUE,message=FALSE,warning=FALSE, results='hide'} dataDEGs <- DPA(dataFilt = dataFilt, dataType = "Gene expression") dataFEA <- FEA(DEGsmatrix = dataDEGs) dataGRN <- GRN(TFs = rownames(dataDEGs)[1:100], DEGsmatrix = dataDEGs, DiffGenes = TRUE, normCounts = dataFilt) dataURA <- URA(dataGRN = dataGRN, DEGsmatrix = dataDEGs, BPname = c("apoptosis", "proliferation of cells")) dataDual <- PRA(dataURA = dataURA, BPname = c("apoptosis", "proliferation of cells"), thres.role = 0) CancerGenes <- list("TSG"=names(dataDual$TSG), "OCG"=names(dataDual$OCG)) ``` ## `plotURA`: Upstream regulatory analysis plot The user can plot the result of the upstream regulatory analysis using the function `plotURA`. ```{r, eval = TRUE,message=FALSE,warning=FALSE, results='hide'} plotURA(dataURA = dataURA[c(names(dataDual$TSG), names(dataDual$OCG)),, drop = FALSE], additionalFilename = "_exampleVignette") ``` The figure resulted from the code above is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} img <- readPNG("URAplot.png") grid.raster(img) ``` ## Case study n. 2: Expression pipeline Pan Cancer 5 cancer types ```{r,eval = FALSE,echo=TRUE,message=FALSE,warning=FALSE} cancerList <- c("BLCA","COAD","ESCA","HNSC","STAD") listMoonlight <- moonlight(cancerType = cancerList, dataType = "Gene expression", directory = "data", nSample = 10, nTF = 100, DiffGenes = TRUE, BPname = c("apoptosis","proliferation of cells")) save(listMoonlight, file = paste0("listMoonlight_ncancer4.Rdata")) ``` ## `plotCircos`: Moonlight Circos Plot The results of the moonlight pipeline can be visualized with a circos plot. Outer ring: color by cancer type, Inner ring: OCGs and TSGs, Inner connections: green: common OCGs yellow: common TSGs red: possible dual role ```{r, eval = TRUE, echo = TRUE, results='hide', warning = FALSE, message = FALSE} plotCircos(listMoonlight = listMoonlight, additionalFilename = "_ncancer5") ``` The figure generated by the code above is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} img <- readPNG("circos_ocg_tsg_ncancer5.png") grid.raster(img) ``` ## Case study n. 3: Downstream analysis BRCA with stages ```{r,eval = FALSE,echo=TRUE,message=FALSE,warning=FALSE} listMoonlight <- NULL for (i in 1:4){ dataDual <- moonlight(cancerType = "BRCA", dataType = "Gene expression", directory = "data", nSample = 10, nTF = 5, DiffGenes = TRUE, BPname = c("apoptosis","proliferation of cells"), stage = i) listMoonlight <- c(listMoonlight, list(dataDual)) save(dataDual, file = paste0("dataDual_stage",as.roman(i), ".Rdata")) } names(listMoonlight) <- c("stage1", "stage2", "stage3", "stage4") # Prepare mutation data for stages mutation <- GDCquery_Maf(tumor = "BRCA") res.mutation <- NULL for(stage in 1:4){ curStage <- paste0("Stage ", as.roman(stage)) dataClin$tumor_stage <- toupper(dataClin$tumor_stage) dataClin$tumor_stage <- gsub("[ABCDEFGH]","",dataClin$tumor_stage) dataClin$tumor_stage <- gsub("ST","Stage",dataClin$tumor_stage) dataStg <- dataClin[dataClin$tumor_stage %in% curStage,] message(paste(curStage, "with", nrow(dataStg), "samples")) dataSmTP <- mutation$Tumor_Sample_Barcode dataStgC <- dataSmTP[substr(dataSmTP,1,12) %in% dataStg$bcr_patient_barcode] dataSmTP <- dataStgC info.mutation <- mutation[mutation$Tumor_Sample_Barcode %in% dataSmTP,] ind <- which(info.mutation[,"Consequence"]=="inframe_deletion") ind2 <- which(info.mutation[,"Consequence"]=="inframe_insertion") ind3 <- which(info.mutation[,"Consequence"]=="missense_variant") res.mutation <- c(res.mutation, list(info.mutation[c(ind, ind2, ind3),c(1,51)])) } names(res.mutation) <- c("stage1", "stage2", "stage3", "stage4") tmp <- NULL tmp <- c(tmp, list(listMoonlight[[1]][[1]])) tmp <- c(tmp, list(listMoonlight[[2]][[1]])) tmp <- c(tmp, list(listMoonlight[[3]][[1]])) tmp <- c(tmp, list(listMoonlight[[4]][[1]])) names(tmp) <- names(listMoonlight) mutation <- GDCquery_Maf(tumor = "BRCA") plotCircos(listMoonlight=listMoonlight,listMutation=res.mutation, additionalFilename="proc2_wmutation", intensityColDual=0.2,fontSize = 2) ``` The results of the moonlight pipeline can be visualized with a circos plot. Outer ring: color by cancer type, Inner ring: OCGs and TSGs, Inner connections: green: common OCGs yellow: common TSGs red: possible dual role The figure generated by the code above is shown below: ```{r, fig.width=6, fig.height=4, echo = FALSE, fig.align="center",hide=TRUE, message=FALSE,warning=FALSE} img <- readPNG("circos_ocg_tsg_stages.png") grid.raster(img) ``` ****** Session Information ****** ```{r sessionInfo} sessionInfo() ``` # References