Updates

Recently the TCGA data has been moved from the DCC server to The National Cancer Institute (NCI) Genomic Data Commons (GDC) Data Portal In this version of the package, we rewrote all the functions that were acessing the old TCGA server to GDC.

The GDC, which receives, processes, harmonizes, and distributes clinical, biospecimen, and genomic data from multiple cancer research programs, has data from the following programs:

  • The Cancer Genome Atlas (TCGA)
  • Therapeutically Applicable Research to Generate Effective Treatments (TARGET)
  • the Cancer Genome Characterization Initiative (CGCI)

The big change is that the GDC data is harmonized against GRCh38. However, not all data has been harmonized yet. The old TCGA data can be acessed through GDC legacy Archive, in which the majority of data can be found.

More information about the project can be found in GDC FAQS

The functions TCGAquery, TCGAdownload, TCGAPrepare, TCGAquery_maf, TCGAquery_clinical, were replaced by GDCquery, GDCdownload, GDCprepare, GDCquery_maf, GDCquery_clinical.

And it can acess both the GDC and GDC Legacy Archive.

Note: Not all the examples in this vignette were updated.

Introduction

Motivation: The Cancer Genome Atlas (TCGA) provides us with an enormous collection of data sets, not only spanning a large number of cancers but also a large number of experimental platforms. Even though the data can be accessed and downloaded from the database, the possibility to analyse these downloaded data directly in one single R package has not yet been available.

TCGAbiolinks consists of three parts or levels. Firstly, we provide different options to query and download from TCGA relevant data from all currently platforms and their subsequent pre-processing for commonly used bio-informatics (tools) packages in Bioconductor or CRAN. Secondly, the package allows to integrate different data types and it can be used for different types of analyses dealing with all platforms such as diff.expression, network inference or survival analysis, etc, and then it allows to visualize the obtained results. Thirdly we added a social level where a researcher can found a similar intereset in a bioinformatic community, and allows both to find a validation of results in literature in pubmed and also to retrieve questions and answers from site such as support.bioconductor.org, biostars.org, stackoverflow,etc.

This document describes how to search, download and analyze TCGA data using the TCGAbiolinks package.

Installation

To install use the code below.

source("https://bioconductor.org/biocLite.R")
biocLite("TCGAbiolinks")

For a Graphical User Interface, please see TCGAbiolinksGUI. The GUI in under review and will soon be available in Bioconductor repository.

Citation

Please cite TCGAbiolinks package:

  • “TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data.” Nucleic acids research (2015): gkv1507. (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 2016)

Related publications to this package:

  • “TCGA Workflow: Analyze cancer genomics and epigenomics data using Bioconductor packages”. F1000Research 10.12688/f1000research.8923.1 (Silva, TC and Colaprico, A and Olsen, C and D’Angelo, F and Bontempi, G and Ceccarelli, M and Noushmehr, H 2016)

Also, if you have used ELMER analysis please cite:

  • Yao, L., Shen, H., Laird, P. W., Farnham, P. J., & Berman, B. P. “Inferring regulatory element landscapes and transcription factor networks from cancer methylomes.” Genome Biol 16 (2015): 105.
  • Yao, Lijing, Benjamin P. Berman, and Peggy J. Farnham. “Demystifying the secret mission of enhancers: linking distal regulatory elements to target genes.” Critical reviews in biochemistry and molecular biology 50.6 (2015): 550-573.

GDCquery: Searching TCGA open-access data

GDCquery: Searching GDC data for download

You can easily search GDC data using the GDCquery function.

Using a summary of filters as used in the TCGA portal, the function works with the following arguments:

  • project A list of valid project (see table below)
  • data.category A valid project (see list with getProjectSummary(project))
  • data.type A data type to filter the files to download
  • sample.type A sample type to filter the files to download (See table below)
  • workflow.type GDC workflow type
  • barcode A list of barcodes to filter the files to download
  • legacy Search in the legacy repository? Default: FALSE
  • platform Experimental data platform (HumanMethylation450, AgilentG4502A_07 etc). Used only for legacy repository
  • file.type A string to filter files, based on its names. Used only for legacy repository

The next subsections will detail each of the search arguments. Below, we show some search examples:

#---------------------------------------------------------------
#  For available entries and combinations please se table below
#---------------------------------------------------------------

# Gene expression aligned against Hg38
query <- GDCquery(project = "TARGET-AML",
                  data.category = "Transcriptome Profiling",
                  data.type = "Gene Expression Quantification", 
                  workflow.type = "HTSeq - Counts")

# All DNA methylation data for TCGA-GBM and TCGA-GBM
query.met <- GDCquery(project = c("TCGA-GBM","TCGA-LGG"),
                      legacy = TRUE,
                      data.category = "DNA methylation",
                      platform = c("Illumina Human Methylation 450", "Illumina Human Methylation 27"))

# Using sample type to get only Primary solid Tumor samples and Solid Tissue Normal
query.mirna <- GDCquery(project = "TCGA-ACC", 
                        data.category = "Transcriptome Profiling", 
                        data.type = "miRNA Expression Quantification",
                        sample.type = c("Primary solid Tumor","Solid Tissue Normal"))

# Example Using legacy to accessing hg19 and filtering by barcode
query <- GDCquery(project = "TCGA-GBM",
                  data.category = "DNA methylation", 
                  platform = "Illumina Human Methylation 27", 
                  legacy = TRUE,
                  barcode = c("TCGA-02-0047-01A-01D-0186-05","TCGA-06-2559-01A-01D-0788-05"))

# Gene expression aligned against hg19.
query.exp.hg19 <- GDCquery(project = "TCGA-GBM",
                  data.category = "Gene expression",
                  data.type = "Gene expression quantification",
                  platform = "Illumina HiSeq", 
                  file.type  = "normalized_results",
                  experimental.strategy = "RNA-Seq",
                  barcode = c("TCGA-14-0736-02A-01R-2005-01", "TCGA-06-0211-02A-02R-2005-01"),
                  legacy = TRUE)

# Searching idat file for DNA methylation
query <- GDCquery(project = "TCGA-OV",
                  data.category = "Raw microarray data",
                  data.type = "Raw intensities", 
                  experimental.strategy = "Methylation array", 
                  legacy = TRUE,
                  file.type = ".idat",
                  platform = "Illumina Human Methylation 450")

The list of projects is below:

List of projects
disease_type.0 project_id name primary_site.0 released state dbgap_accession_number id tumor
Cholangiocarcinoma TCGA-CHOL Cholangiocarcinoma Bile Duct True legacy NA TCGA-CHOL CHOL
Ovarian Serous Cystadenocarcinoma TCGA-OV Ovarian Serous Cystadenocarcinoma Ovary True legacy NA TCGA-OV OV
Liver Hepatocellular Carcinoma TCGA-LIHC Liver Hepatocellular Carcinoma Liver True legacy NA TCGA-LIHC LIHC
Head and Neck Squamous Cell Carcinoma TCGA-HNSC Head and Neck Squamous Cell Carcinoma Head and Neck True legacy NA TCGA-HNSC HNSC
Colon Adenocarcinoma TCGA-COAD Colon Adenocarcinoma Colorectal True legacy NA TCGA-COAD COAD
Adrenocortical Carcinoma TCGA-ACC Adrenocortical Carcinoma Adrenal Gland True legacy NA TCGA-ACC ACC
Mesothelioma TCGA-MESO Mesothelioma Pleura True legacy NA TCGA-MESO MESO
Acute Myeloid Leukemia TARGET-AML Acute Myeloid Leukemia Blood True legacy phs000465 TARGET-AML AML
Neuroblastoma TARGET-NBL Neuroblastoma Nervous System True legacy phs000467 TARGET-NBL NBL
Brain Lower Grade Glioma TCGA-LGG Brain Lower Grade Glioma Brain True legacy NA TCGA-LGG LGG
Pancreatic Adenocarcinoma TCGA-PAAD Pancreatic Adenocarcinoma Pancreas True legacy NA TCGA-PAAD PAAD
High-Risk Wilms Tumor TARGET-WT High-Risk Wilms Tumor Kidney True legacy phs000471 TARGET-WT WT
Uterine Carcinosarcoma TCGA-UCS Uterine Carcinosarcoma Uterus True legacy NA TCGA-UCS UCS
Uveal Melanoma TCGA-UVM Uveal Melanoma Eye True legacy NA TCGA-UVM UVM
Stomach Adenocarcinoma TCGA-STAD Stomach Adenocarcinoma Stomach True legacy NA TCGA-STAD STAD
Lung Squamous Cell Carcinoma TCGA-LUSC Lung Squamous Cell Carcinoma Lung True legacy NA TCGA-LUSC LUSC
Uterine Corpus Endometrial Carcinoma TCGA-UCEC Uterine Corpus Endometrial Carcinoma Uterus True legacy NA TCGA-UCEC UCEC
Esophageal Carcinoma TCGA-ESCA Esophageal Carcinoma Esophagus True legacy NA TCGA-ESCA ESCA
Rhabdoid Tumor TARGET-RT Rhabdoid Tumor Kidney True legacy phs000470 TARGET-RT RT
Osteosarcoma TARGET-OS Osteosarcoma Bone True legacy phs000468 TARGET-OS OS
Prostate Adenocarcinoma TCGA-PRAD Prostate Adenocarcinoma Prostate True legacy NA TCGA-PRAD PRAD
Kidney Chromophobe TCGA-KICH Kidney Chromophobe Kidney True legacy NA TCGA-KICH KICH
Testicular Germ Cell Tumors TCGA-TGCT Testicular Germ Cell Tumors Testis True legacy NA TCGA-TGCT TGCT
Acute Myeloid Leukemia TCGA-LAML Acute Myeloid Leukemia Bone Marrow True legacy NA TCGA-LAML LAML
Thymoma TCGA-THYM Thymoma Thymus True legacy NA TCGA-THYM THYM
Lymphoid Neoplasm Diffuse Large B-cell Lymphoma TCGA-DLBC Lymphoid Neoplasm Diffuse Large B-cell Lymphoma Lymph Nodes True legacy NA TCGA-DLBC DLBC
Pheochromocytoma and Paraganglioma TCGA-PCPG Pheochromocytoma and Paraganglioma Adrenal Gland True legacy NA TCGA-PCPG PCPG
Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma TCGA-CESC Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma Cervix True legacy NA TCGA-CESC CESC
Lung Adenocarcinoma TCGA-LUAD Lung Adenocarcinoma Lung True legacy NA TCGA-LUAD LUAD
Bladder Urothelial Carcinoma TCGA-BLCA Bladder Urothelial Carcinoma Bladder True legacy NA TCGA-BLCA BLCA
Kidney Renal Papillary Cell Carcinoma TCGA-KIRP Kidney Renal Papillary Cell Carcinoma Kidney True legacy NA TCGA-KIRP KIRP
Clear Cell Sarcoma of the Kidney TARGET-CCSK Clear Cell Sarcoma of the Kidney Kidney True legacy phs000466 TARGET-CCSK CCSK
Sarcoma TCGA-SARC Sarcoma Soft Tissue True legacy NA TCGA-SARC SARC
Thyroid Carcinoma TCGA-THCA Thyroid Carcinoma Thyroid True legacy NA TCGA-THCA THCA
Rectum Adenocarcinoma TCGA-READ Rectum Adenocarcinoma Colorectal True legacy NA TCGA-READ READ
Skin Cutaneous Melanoma TCGA-SKCM Skin Cutaneous Melanoma Skin True legacy NA TCGA-SKCM SKCM
Glioblastoma Multiforme TCGA-GBM Glioblastoma Multiforme Brain True legacy NA TCGA-GBM GBM
Kidney Renal Clear Cell Carcinoma TCGA-KIRC Kidney Renal Clear Cell Carcinoma Kidney True legacy NA TCGA-KIRC KIRC
Breast Invasive Carcinoma TCGA-BRCA Breast Invasive Carcinoma Breast True legacy NA TCGA-BRCA BRCA

The list of sample.type is below:

List sample types
tissue.code shortLetterCode tissue.definition
01 TP Primary solid Tumor
02 TR Recurrent Solid Tumor
03 TB Primary Blood Derived Cancer - Peripheral Blood
04 TRBM Recurrent Blood Derived Cancer - Bone Marrow
05 TAP Additional - New Primary
06 TM Metastatic
07 TAM Additional Metastatic
08 THOC Human Tumor Original Cells
09 TBM Primary Blood Derived Cancer - Bone Marrow
10 NB Blood Derived Normal
11 NT Solid Tissue Normal
12 NBC Buccal Cell Normal
13 NEBV EBV Immortalized Normal
14 NBM Bone Marrow Normal
20 CELLC Control Analyte
40 TRB Recurrent Blood Derived Cancer - Peripheral Blood
50 CELL Cell Lines
60 XP Primary Xenograft Tissue
61 XCL Cell Line Derived Xenograft Tissue

The other fields (data.category, data.type, workflow.type, platform, file.type) can be found below. Please, not that these tables are still incomplete.

Harmonized data

data.category data.type workflow.type platform
Transcriptome Profiling Gene Expression Quantification HTSeq - Counts
HTSeq - FPKM-UQ
HTSeq - FPKM
Isoform Expression Quantification -
miRNA Expression Quantification -
Copy Number Variation Copy Number Segment
Masked Copy Number Segment
Simple Nucleotide Variation
Raw Sequencing Data
Biospecimen
Clinical
DNA Methylation Illumina Human Methylation 450
Illumina Human Methylation 27

Legacy data

data.category data.type platform file.type
Copy number variation - Affymetrix SNP Array 6.0 nocnv_hg18.seg
- Affymetrix SNP Array 6.0 hg18.seg
- Affymetrix SNP Array 6.0 nocnv_hg19.seg
- Affymetrix SNP Array 6.0 hg19.seg
- Illumina HiSeq -
Simple nucleotide variation
Raw sequencing data
Biospecimen
Clinical
Protein expression MDA RPPA Core -
Gene expression Gene expression quantification Illumina HiSeq normalized_results
Illumina HiSeq results
HT_HG-U133A -
AgilentG4502A_07_2 -
AgilentG4502A_07_1 -
HuEx-1_0-st-v2 FIRMA.txt
gene.txt
Isoform expression quantification
miRNA gene quantification hg19.mirna
hg19.mirbase20
mirna
Exon junction quantification
Exon quantification
miRNA isoform quantification hg19.isoform
isoform
DNA methylation Illumina Human Methylation 450 -
Illumina Human Methylation 27 -
Illumina DNA Methylation OMA003 CPI -
Illumina DNA Methylation OMA002 CPI -
Illumina Hi Seq
Raw microarray data Raw intensities Illumina Human Methylation 450 idat
Illumina Human Methylation 27 idat
Other

Working with mutation files.

GDCquery_maf: Mutation Annotation Format (MAF) aligned against hg38

In order to download the Mutation Annotation Format (MAF) aligned against hg38, we provide the user with the GDCquery_maf function. Briefly, it will get the open acess maf files from https://gdc-docs.nci.nih.gov/Data/Release_Notes/Data_Release_Notes/. Four separate variant calling pipelines are implemented for GDC data harmonization which are described here

The arguments that will be used to filter the data are:

  • tumor: A TCGA project without the TCGA- prefix.
  • save.csv: Save the maf as csv file
  • pipelines: Four separate variant calling pipelines are implemented for GDC data harmonization. Options: muse, varscan2, somaticsniper, mutect.
acc.muse.maf <- GDCquery_Maf("ACC", pipelines = "muse")
acc.varscan2.maf <- GDCquery_Maf("ACC", pipelines = "varscan2")
acc.somaticsniper.maf <- GDCquery_Maf("ACC", pipelines = "somaticsniper")
acc.mutect.maf <- GDCquery_Maf("ACC", pipelines = "mutect")

This mutation data can be visualized through a oncoprint from the ComplexHeatmap package (Gu, Zuguang and Eils, Roland and Schlesner, Matthias 2016).

mut <- GDCquery_Maf("ACC", pipelines = "mutect2")
clin <- GDCquery_clinic("TCGA-ACC","clinical")
clin <- clin[,c("bcr_patient_barcode","disease","gender","tumor_stage","race","vital_status")]
TCGAvisualize_oncoprint(mut = mut, genes = mut$Hugo_Symbol[1:20],
                        filename = "oncoprint.pdf",
                        annotation = clin,
                        color=c("background"="#CCCCCC","DEL"="purple","INS"="yellow","SNP"="brown"),
                        rows.font.size=10,
                        width = 10,
                        heatmap.legend.side = "right",
                        dist.col = 0,
                        label.font.size = 10)

Mutation Annotation Format (MAF) aligned against hg19

In order to download the Mutation Annotation Format (MAF) aligned against hg19, the user will need to use GDCquery with legacy = TRUE, GDCdownload and GDCprepare.

query.maf.hg19 <- GDCquery(project = "TCGA-COAD", 
                           data.category = "Simple nucleotide variation", 
                           data.type = "Simple somatic mutation",
                           access = "open", 
                           legacy = TRUE)
# Check maf availables
knitr::kable(getResults(query.maf.hg19)[,c("created_datetime","file_name")]) 
created_datetime file_name
2016-06-13T16:30:24.763822-05:00 gsc_COAD_pairs.aggregated.capture.tcga.uuid.automated.somatic.maf
NA hgsc.bcm.edu_COAD.SOLiD_DNASeq.1.somatic.maf
query.maf.hg19 <- GDCquery(project = "TCGA-COAD", 
                           data.category = "Simple nucleotide variation", 
                           data.type = "Simple somatic mutation",
                           access = "open", 
                           file.type = "gsc_COAD_pairs.aggregated.capture.tcga.uuid.automated.somatic.maf",
                           legacy = TRUE)
GDCdownload(query.maf.hg19)
coad.mutect.maf <- GDCprepare(query.maf.hg19)

GDCquery_clinic and GDCprepare_clinic: Working with clinical data.

In GDC database the clinical data can be retrieved in two sources:

  • indexed clinical (from json)
  • xml files

there are two main differences:

  • XML has more information: radiation, drugs information, follow-ups, biospecimen, etc. So the indexed one is only a subset of the XML files
  • The indexed data contains the updated data with the follow up informaiton. For example: if the patient is alive in the first time clinical data was collect and the in the next follow-up he is dead, the indexed data will show dead. The XML will have two fields, one for the first time saying he is alive (in the clinical part) and the follow-up saying he is dead. You can see this case here: https://gist.github.com/tiagochst/cfa384297ff0950a6489e3f4b296ecd7
clin.query <- GDCquery(project = "TCGA-BLCA", data.category = "Clinical", barcode = "TCGA-FD-A5C0")
 json  <- tryCatch(GDCdownload(clin.query), 
                   error = function(e) GDCdownload(clin.query, method = "client"))
clinical.patient <- GDCprepare_clinic(clin.query, clinical.info = "patient")
clinical.patient.followup <- GDCprepare_clinic(clin.query, clinical.info = "follow_up")
clinical.index <- GDCquery_clinic("TCGA-BLCA")
# Example of the second difference:
clinical.patient[,c("vital_status","days_to_death","days_to_last_followup")]
##   vital_status days_to_death days_to_last_followup
## 1        Alive            NA                   209
clinical.patient.followup[,c("vital_status","days_to_death","days_to_last_followup")]
##   vital_status days_to_death days_to_last_followup
## 1        Alive            NA                   407
## 2         Dead           550                    NA
# indexed data is equivalent to follow ups information
clinical.index[clinical.index$submitter_id=="TCGA-FD-A5C0",
               c("vital_status","days_to_death","days_to_last_follow_up")]
##     vital_status days_to_death days_to_last_follow_up
## 197         dead           550                    407

You can retrieve clinical data using the GDCquery_clinic function. This will get only the indexed GDC clinical data. This is the same as clicking on the download clinical buttun in the data portal. This means this function is not parsing the XML files, see GDCprepare_clinic function.

The arguments of this function are:

  • project: project_id (“TCGA-OV”,“TCGA-BRCA”,“TARGET-WT”, etc)
  • type: (“clinical”,“biospecimen”)
  • save.csv: save the clinical/biospeciment into a csv file. Pattern: project_type.csv

Examples of use:

clin <- GDCquery_clinic("TCGA-ACC", type = "clinical", save.csv = TRUE)
clin <- GDCquery_clinic("TCGA-ACC", type = "biospecimen", save.csv = TRUE)

To parse the TCGA clinical XML files, please use GDCprepare_clinic function. It receives as argument the query object and which clinical information we should retrieve. The possible values for the argument clinical.info are:

  • For clinical XML files:
    • admin
    • drug
    • follow_up
    • patient
    • new_tumor_event
    • radiation
    • stage_event
  • For biospecimen XML files:
    • admin
    • aliquot
    • analyte
    • bio_patient
    • portion
    • protocol
    • sample
    • slide
query <- GDCquery(project = "TCGA-COAD", 
                  data.category = "Clinical", 
                  barcode = c("TCGA-RU-A8FL","TCGA-AA-3972"))
GDCdownload(query)
clinical <- GDCprepare_clinic(query, clinical.info = "patient")
clinical.drug <- GDCprepare_clinic(query, clinical.info = "drug")
clinical.radiation <- GDCprepare_clinic(query, clinical.info = "radiation")
clinical.admin <- GDCprepare_clinic(query, clinical.info = "admin")

query <- GDCquery(project = "TCGA-COAD", 
                  data.category = "Biospecimen", 
                  barcode = c("TCGA-RU-A8FL","TCGA-AA-3972"))
GDCdownload(query)
clinical.admin <- GDCprepare_clinic(query, clinical.info = "admin")
clinical.sample <- GDCprepare_clinic(query, clinical.info = "sample")
clinical.slide <- GDCprepare_clinic(query, clinical.info = "slide")
clinical.portion <- GDCprepare_clinic(query, clinical.info = "portion")

To get all the information for TGCA samples you can use the script below:

# This code will get all clinical indexed data from TCGA
library(TCGAbiolinks)
library(data.table)
clinical <- TCGAbiolinks:::getGDCprojects()$project_id %>% 
            regexPipes::grep("TCGA",value=T) %>% 
            sort %>% 
            plyr::alply(1,GDCquery_clinic, .progress = "text") %>% 
            rbindlist
readr::write_csv(clinical,path = paste0("all_clin_indexed.csv"))

# This code will get all clinical XML data from TCGA
getclinical <- function(proj){
    message(proj)
    while(1){
        result = tryCatch({
            query <- GDCquery(project = proj, data.category = "Clinical")
            GDCdownload(query)
            clinical <- GDCprepare_clinic(query, clinical.info = "patient")
            for(i in c("admin","radiation","follow_up","drug","new_tumor_event")){
                message(i)
                aux <- GDCprepare_clinic(query, clinical.info = i)
                if(is.null(aux)) next
                # add suffix manually if it already exists
                replicated <- which(grep("bcr_patient_barcode",colnames(aux), value = T,invert = T) %in% colnames(clinical))
                colnames(aux)[replicated] <- paste0(colnames(aux)[replicated],".",i)
                if(!is.null(aux)) clinical <- merge(clinical,aux,by = "bcr_patient_barcode", all = TRUE)
            }
            readr::write_csv(clinical,path = paste0(proj,"_clinical_from_XML.csv")) # Save the clinical data into a csv file
            return(clinical)
        }, error = function(e) {
            message(paste0("Error clinical: ", proj))
        })
    }
}
clinical <- TCGAbiolinks:::getGDCprojects()$project_id %>% 
    regexPipes::grep("TCGA",value=T) %>% sort %>% 
    plyr::alply(1,getclinical, .progress = "text") %>% 
    rbindlist(fill = TRUE) %>% setDF %>% subset(!duplicated(clinical))
readr::write_csv(clinical,path = "all_clin_XML.csv")
# result: https://drive.google.com/open?id=0B0-8N2fjttG-WWxSVE5MSGpva1U
# Obs: this table has multiple lines for each patient, as the patient might have several followups, drug treatments,
# new tumor events etc...

Also, some functions to work with clinical data are provided.

For example the function TCGAquery_SampleTypes will filter barcodes based on a type the argument typesample. Some of the typesamples possibilities are: TP (PRIMARY SOLID TUMOR), TR (RECURRENT SOLID TUMOR), NT (Solid Tissue Normal) etc. Please, see ?TCGAquery_SampleTypes for all the possible values.

The function TCGAquery_MatchedCoupledSampleTypes will filter the samples that have all the typesample provided as argument. For example, if TP and TR are set as typesample, the function will return the barcodes of a patient if it has both types. So, if it has a TP, but not a TR, no barcode will be returned. If it has a TP and a TR both barcodes are returned.

An example of the function is below:

bar <- c("TCGA-G9-6378-02A-11R-1789-07", "TCGA-CH-5767-04A-11R-1789-07",  
         "TCGA-G9-6332-60A-11R-1789-07", "TCGA-G9-6336-01A-11R-1789-07",
         "TCGA-G9-6336-11A-11R-1789-07", "TCGA-G9-7336-11A-11R-1789-07",
         "TCGA-G9-7336-04A-11R-1789-07", "TCGA-G9-7336-14A-11R-1789-07",
         "TCGA-G9-7036-04A-11R-1789-07", "TCGA-G9-7036-02A-11R-1789-07",
         "TCGA-G9-7036-11A-11R-1789-07", "TCGA-G9-7036-03A-11R-1789-07",
         "TCGA-G9-7036-10A-11R-1789-07", "TCGA-BH-A1ES-10A-11R-1789-07",
         "TCGA-BH-A1F0-10A-11R-1789-07", "TCGA-BH-A0BZ-02A-11R-1789-07",
         "TCGA-B6-A0WY-04A-11R-1789-07", "TCGA-BH-A1FG-04A-11R-1789-08",
         "TCGA-D8-A1JS-04A-11R-2089-08", "TCGA-AN-A0FN-11A-11R-8789-08",
         "TCGA-AR-A2LQ-12A-11R-8799-08", "TCGA-AR-A2LH-03A-11R-1789-07",
         "TCGA-BH-A1F8-04A-11R-5789-07", "TCGA-AR-A24T-04A-55R-1789-07",
         "TCGA-AO-A0J5-05A-11R-1789-07", "TCGA-BH-A0B4-11A-12R-1789-07",
         "TCGA-B6-A1KN-60A-13R-1789-07", "TCGA-AO-A0J5-01A-11R-1789-07",
         "TCGA-AO-A0J5-01A-11R-1789-07", "TCGA-G9-6336-11A-11R-1789-07",
         "TCGA-G9-6380-11A-11R-1789-07", "TCGA-G9-6380-01A-11R-1789-07",
         "TCGA-G9-6340-01A-11R-1789-07", "TCGA-G9-6340-11A-11R-1789-07")

S <- TCGAquery_SampleTypes(bar,"TP")
S2 <- TCGAquery_SampleTypes(bar,"NB")

# Retrieve multiple tissue types  NOT FROM THE SAME PATIENTS
SS <- TCGAquery_SampleTypes(bar,c("TP","NB"))

# Retrieve multiple tissue types  FROM THE SAME PATIENTS
SSS <- TCGAquery_MatchedCoupledSampleTypes(bar,c("NT","TP"))

TCGAquery_subtype: Working with molecular subtypes data.

The Cancer Genome Atlas (TCGA) Research Network has reported integrated genome-wide studies of various diseases. We have added some of the subtypes defined by these report in our package:

  • ACC(Cancer Genome Atlas Research Network and others 2016)
  • BRCA(Cancer Genome Atlas Research Network and others 2012c)
  • COAD (Cancer Genome Atlas Research Network and others 2012b)
  • GBM (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 2016)
  • HNSC (Cancer Genome Atlas Research Network and others 2015a)
  • KICH (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 2014)
  • KIRC(Cancer Genome Atlas Research Network and others 2013a)
  • KIRP (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 2016)
  • LGG(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 2016)
  • LUAD (Cancer Genome Atlas Research Network and others 2014b)
  • LUSC(Cancer Genome Atlas Research Network and others 2012a)
  • PCPG(Fishbein, Lauren and Leshchiner, Ignaty and Walter, Vonn and Danilova, Ludmila and Robertson, A Gordon and Johnson, Amy R and Lichtenberg, Tara M and Murray, Bradley A and Ghayee, Hans K and Else, Tobias and others 2017)
  • PRAD(Cancer Genome Atlas Research Network and others 2015c)
  • READ (Cancer Genome Atlas Research Network and others 2012b)
  • SKCM (Cancer Genome Atlas Research Network and others 2015b)
  • STAD (Cancer Genome Atlas Research Network and others 2014a)
  • THCA (Cancer Genome Atlas Research Network and others 2014c)
  • UCEC (Cancer Genome Atlas Research Network and others 2013b)

These subtypes will be automatically added in the summarizedExperiment object through TCGAprepare. But you can also use the TCGAquery_subtype function to retrive this information.

# Check with subtypes from TCGAprepare and update examples
GBM_path_subtypes <- TCGAquery_subtype(tumor = "gbm")

LGG_path_subtypes <- TCGAquery_subtype(tumor = "lgg")

A subset of the lgg subytpe is shown below:

GDCdownload: Downloading GDC data

You can easily download data using the GDCdownload function. It uses GDC transfer tool to download gdc data doing a system call. For this reason some times the update will stop to show, which does not means that the download process has stopped. Once the process has finished it will give a signal to R.

The data from query will be save in a folder: project/source/data.category (where source is harmonized or legacy)

The arguments are:

  • query A query for GDCquery function
  • token.file A token file to download protect data (not test yet)
  • method Use API (POST method) or gdc client tool (API is faster, but the data might get corrupted in the download, and it might need to be executed again). Options: “api”, “client”
  • directory Directory/Folder where the data was downloaded. Default: GDCdata
  • chunks.per.download This will make the API method only download n (chunks.per.download) files at a time. This may reduce the download problems when the data size is too large.

GDCdownload: Example of use

query <- GDCquery(project = "TCGA-ACC", data.category = "Copy Number Variation",
                  data.type = "Copy Number Segment",
                  barcode = c( "TCGA-OR-A5KU-01A-11D-A29H-01", "TCGA-OR-A5JK-01A-11D-A29H-01"))
GDCdownload(query)
data <- GDCprepare(query)

#-------------------------------------- 
# Gene expression
#-------------------------------------- 
# Aligned against Hg38
# mRNA pipeline: https://gdc-docs.nci.nih.gov/Data/Bioinformatics_Pipelines/Expression_mRNA_Pipeline/
query.exp.hg38 <- GDCquery(project = "TCGA-GBM", 
                  data.category = "Transcriptome Profiling", 
                  data.type = "Gene Expression Quantification", 
                  workflow.type = "HTSeq - FPKM-UQ",
                  barcode =  c("TCGA-14-0736-02A-01R-2005-01", "TCGA-06-0211-02A-02R-2005-01"))
GDCdownload(query.exp.hg38)
expdat <- GDCprepare(query = query.exp.hg38,
                     save = TRUE, 
                     save.filename = "exp.rda")

# Aligned against Hg19
query.exp.hg19 <- GDCquery(project = "TCGA-GBM",
                  data.category = "Gene expression",
                  data.type = "Gene expression quantification",
                  platform = "Illumina HiSeq", 
                  file.type  = "normalized_results",
                  experimental.strategy = "RNA-Seq",
                  barcode = c("TCGA-14-0736-02A-01R-2005-01", "TCGA-06-0211-02A-02R-2005-01"),
                  legacy = TRUE)
GDCdownload(query.exp.hg19)
data <- GDCprepare(query.exp.hg19)


#-------------------------------------- 
# DNA methylation data
#-------------------------------------- 
# DNA methylation aligned to hg38
query_met.hg38 <- GDCquery(project= "TCGA-LGG", 
                           data.category = "DNA Methylation", 
                           platform = "Illumina Human Methylation 450", 
                           barcode = c("TCGA-HT-8111-01A-11D-2399-05","TCGA-HT-A5R5-01A-11D-A28N-05"))
GDCdownload(query_met.hg38)
data.hg38 <- GDCprepare(query_met.hg38)

# DNA methylation aligned to hg19
query_meth.hg19 <- GDCquery(project= "TCGA-LGG", 
                            data.category = "DNA methylation", 
                            platform = "Illumina Human Methylation 450", 
                            barcode = c("TCGA-HT-8111-01A-11D-2399-05","TCGA-HT-A5R5-01A-11D-A28N-05"), 
                            legacy = TRUE)
GDCdownload(query_meth.hg19)
data.hg19 <- GDCprepare(query_meth.hg19)

# A function to download only 20 samples
legacyPipeline <- function(project, data.category, platform, n = 20){
    query <- GDCquery(project = project,
                      data.category = data.category,
                      platform = platform,
                      legacy = TRUE)
    cases <- getResults(query, rows = 1:n, cols="cases") # Get two samples from the search
    query <- GDCquery(project = project,
                      data.category = data.category,
                      platform = platform,
                      legacy = TRUE,
                      barcode = cases)
    GDCdownload(query,chunks.per.download = 5)
    data <- GDCprepare(query)
    return(data)
}
# DNA methylation
data <- legacyPipeline("TCGA-GBM","DNA methylation","Illumina Human Methylation 27")
data <- legacyPipeline("TCGA-GBM","DNA methylation","Illumina Human Methylation 450")
data <- legacyPipeline("TCGA-GBM","DNA methylation","Illumina DNA Methylation OMA003 CPI")
data <- legacyPipeline("TCGA-GBM","DNA methylation","Illumina DNA Methylation OMA002 CPI")

#-------------------------------------------------------
# Example to  idat files from TCGA projects
#-------------------------------------------------------
projects <- TCGAbiolinks:::getGDCprojects()$project_id
projects <- projects[grepl('^TCGA',projects,perl=T)]
match.file.cases.all <- NULL
for(proj in projects){
    print(proj)
    query <- GDCquery(project = proj,
                      data.category = "Raw microarray data",
                      data.type = "Raw intensities", 
                      experimental.strategy = "Methylation array", 
                      legacy = TRUE,
                      file.type = ".idat",
                      platform = "Illumina Human Methylation 450")
    match.file.cases <- getResults(query,cols=c("cases","file_name"))
    match.file.cases$project <- proj
    match.file.cases.all <- rbind(match.file.cases.all,match.file.cases)
    tryCatch(GDCdownload(query, method = "api",chunks.per.download = 20),
             error = function(e) GDCdownload(query, method = "client"))
}
# This will create a map between idat file name, cases (barcode) and project
readr::write_tsv(match.file.cases.all, path =  "idat_filename_case.txt")
# code to move all files to local folder
for(file in dir(".",pattern = ".idat", recursive = T)){
    TCGAbiolinks::move(file,basename(file))
}

GDCprepare: Preparing the data

This function is still under development, it is not working for all cases. See the tables below with the status. Examples of query, download, prepare can be found in this gist.

Harmonized data

Data.category Data.type Workflow Type Status
Transcriptome Profiling Gene Expression Quantification HTSeq - Counts Data frame or SE (losing 5% of information when mapping to genomic regions)
HTSeq - FPKM-UQ Returning only a (losing 5% of information when mapping to genomic regions)
HTSeq - FPKM Returning only a (losing 5% of information when mapping to genomic regions)
Isoform Expression Quantification Not needed
miRNA Expression Quantification Not needed Returning only a dataframe for the moment
Copy number variation Copy Number Segment Returning only a dataframe for the moment
Masked Copy Number Segment Returning only a dataframe for the moment
Simple Nucleotide Variation
Raw Sequencing Data
Biospecimen
Clinical

Legacy data

Data.category Data.type Platform file.type Status
Transcriptome Profiling
Copy number variation - Affymetrix SNP Array 6.0 nocnv_hg18.seg Working
- Affymetrix SNP Array 6.0 hg18.seg Working
- Affymetrix SNP Array 6.0 nocnv_hg19.seg Working
- Affymetrix SNP Array 6.0 hg19.seg Working
- Illumina HiSeq Several Working
Simple Nucleotide Variation Simple somatic mutation
Raw Sequencing Data
Biospecimen
Clinical
Protein expression MDA RPPA Core - Working
Gene expression Gene expression quantification Illumina HiSeq normalized_results Working
Illumina HiSeq results Working
HT_HG-U133A - Working
AgilentG4502A_07_2 - Data frame only
AgilentG4502A_07_1 - Data frame only
HuEx-1_0-st-v2 FIRMA.txt Not Preparing
gene.txt Not Preparing
Isoform expression quantification
miRNA gene quantification
Exon junction quantification
Exon quantification
miRNA isoform quantification
DNA methylation Illumina Human Methylation 450 Not used Working
Illumina Human Methylation 27 Not used Working
Illumina DNA Methylation OMA003 CPI Not used Working
Illumina DNA Methylation OMA002 CPI Not used Working
Illumina Hi Seq Not working
Raw Microarray Data
Structural Rearrangement
Other

You can easily read the downloaded data using the GDCprepare function. This function will prepare the data into a SummarizedExperiment (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 2015) object for downstream analysis. For the moment this function works only with data level 3.

The arguments are:

  • query Data frame as the one returned from TCGAquery
  • save Save a rda object with the prepared object? Default: FALSE
  • save.filename Name of the rda object that will be saved if save is TRUE
  • summarizedExperiment Should the output be a SummarizedExperiment object? Default: TRUE
  • directory Directory/Folder where the data was downloaded. Default: GDCdata
  • remove.files.prepared Remove the files read? Default: FALSE. This argument will be considered only if save argument is set to true
  • add.gistic2.mut If a list of genes (gene symbol) is given columns with gistic2 results from GDAC firehose (hg19) and a column indicating if there is or not mutation in that gene (hg38) (TRUE or FALSE - use the maf file for more information) will be added to the sample matrix in the summarized Experiment object
library(TCGAbiolinks)
# Downloading and prepare 
query <- GDCquery(project = "TARGET-AML", 
                  data.category = "Transcriptome Profiling", 
                  data.type = "Gene Expression Quantification", 
                  workflow.type = "HTSeq - FPKM-UQ")
GDCdownload(query)
data <- GDCprepare(query)

# Downloading and prepare using legacy
query <- GDCquery(project = "TCGA-GBM",
                  data.category = "Protein expression",
                  legacy = TRUE, 
                  barcode = c("TCGA-OX-A56R-01A-21-A44T-20","TCGA-08-0357-01A-21-1898-20"))
GDCdownload(query)
data <- GDCprepare(query, save = TRUE, 
                   save.filename = "gbmProteinExpression.rda",
                   remove.files.prepared = TRUE)

# Downloading and prepare using legacy
query <- GDCquery(project = "TCGA-GBM",
                  data.category = "DNA methylation", 
                  platform = "Illumina Human Methylation 27",legacy = TRUE,
                  barcode = c("TCGA-02-0047-01A-01D-0186-05","TCGA-06-2559-01A-01D-0788-05"))
GDCdownload(query)
data <- GDCprepare(query, add.gistic2.mut = c("PTEN","FOXJ1"))

# To view gistic and mutation information please access the samples information matrix in the summarized Experiment object
library(SummarizedExperiment)
samples.information <- colData(data)

TCGAanalyze: Analyze data from TCGA.

You can easily analyze data using following functions:

TCGAanalyze_Preprocessing: Preprocessing of Gene Expression data (IlluminaHiSeq_RNASeqV2)

You can easily search TCGA samples, download and prepare a matrix of gene expression.

# You can define a list of samples to query and download providing relative TCGA barcodes.
listSamples <- c("TCGA-E9-A1NG-11A-52R-A14M-07","TCGA-BH-A1FC-11A-32R-A13Q-07",
                 "TCGA-A7-A13G-11A-51R-A13Q-07","TCGA-BH-A0DK-11A-13R-A089-07",
                 "TCGA-E9-A1RH-11A-34R-A169-07","TCGA-BH-A0AU-01A-11R-A12P-07",
                 "TCGA-C8-A1HJ-01A-11R-A13Q-07","TCGA-A7-A13D-01A-13R-A12P-07",
                 "TCGA-A2-A0CV-01A-31R-A115-07","TCGA-AQ-A0Y5-01A-11R-A14M-07")

# Query platform Illumina HiSeq with a list of barcode 
query <- GDCquery(project = "TCGA-BRCA", 
                  data.category = "Gene expression",
                  data.type = "Gene expression quantification",
                  experimental.strategy = "RNA-Seq",
                  platform = "Illumina HiSeq",
                  file.type = "results",
                  barcode = listSamples, 
                  legacy = TRUE)

# Download a list of barcodes with platform IlluminaHiSeq_RNASeqV2
GDCdownload(query)

# Prepare expression matrix with geneID in the rows and samples (barcode) in the columns
# rsem.genes.results as values
BRCARnaseqSE <- GDCprepare(query)

BRCAMatrix <- assay(BRCARnaseqSE,"raw_count") # or BRCAMatrix <- assay(BRCARnaseqSE,"raw_count")

# For gene expression if you need to see a boxplot correlation and AAIC plot to define outliers you can run
BRCARnaseq_CorOutliers <- TCGAanalyze_Preprocessing(BRCARnaseqSE)

The result is shown below:

Example of a matrix of gene expression (10 genes in rows and 2 samples in columns)
TCGA-A7-A13G-11A-51R-A13Q-07 TCGA-C8-A1HJ-01A-11R-A13Q-07
KCNJ16|3773 42.0 1064
SEPHS2|22928 2575.0 10767
LOC100268168|100268168 14.0 22
LOC100271722|100271722 170.0 114
DUSP26|78986 0.0 2
ITIH4|3700 68.4 31
IL17C|27189 0.0 1
METRN|79006 83.0 45
ACTL7B|10880 0.0 0
STT3B|201595 4611.0 3130

The result from TCGAanalyze_Preprocessing is shown below:

TCGAanalyze_DEA & TCGAanalyze_LevelTab: Differential expression analysis (DEA)

Perform DEA (Differential expression analysis) to identify differentially expressed genes (DEGs) using the TCGAanalyze_DEA function.

TCGAanalyze_DEA performs DEA using following functions from R :

  1. edgeR::DGEList converts the count matrix into an edgeR object.
  2. edgeR::estimateCommonDisp each gene gets assigned the same dispersion estimate.
  3. edgeR::exactTest performs pair-wise tests for differential expression between two groups.
  4. edgeR::topTags takes the output from exactTest(), adjusts the raw p-values using the False Discovery Rate (FDR) correction, and returns the top differentially expressed genes.

This function receives as arguments:

  • mat1 The matrix of the first group (in the example, group 1 is the normal samples),
  • mat2 The matrix of the second group (in the example, group 2 is tumor samples)
  • Cond1type Label for group 1
  • Cond1type Label for group 2

Next, we filter the output of dataDEGs by abs(LogFC) >=1, and uses the TCGAanalyze_LevelTab function to create a table with DEGs (differentially expressed genes), log Fold Change (FC), false discovery rate (FDR), the gene expression level for samples in Cond1type, and Cond2type, and Delta value (the difference of gene expression between the two conditions multiplied logFC).

# Downstream analysis using gene expression data  
# TCGA samples from IlluminaHiSeq_RNASeqV2 with type rsem.genes.results
# save(dataBRCA, geneInfo , file = "dataGeneExpression.rda")
library(TCGAbiolinks)

# normalization of genes
dataNorm <- TCGAanalyze_Normalization(tabDF = dataBRCA, geneInfo =  geneInfo)

# quantile filter of genes
dataFilt <- TCGAanalyze_Filtering(tabDF = dataNorm,
                                  method = "quantile", 
                                  qnt.cut =  0.25)

# selection of normal samples "NT"
samplesNT <- TCGAquery_SampleTypes(barcode = colnames(dataFilt),
                                   typesample = c("NT"))

# selection of tumor samples "TP"
samplesTP <- TCGAquery_SampleTypes(barcode = colnames(dataFilt), 
                                   typesample = c("TP"))

# Diff.expr.analysis (DEA)
dataDEGs <- TCGAanalyze_DEA(mat1 = dataFilt[,samplesNT],
                            mat2 = dataFilt[,samplesTP],
                            Cond1type = "Normal",
                            Cond2type = "Tumor",
                            fdr.cut = 0.01 ,
                            logFC.cut = 1,
                            method = "glmLRT")

# DEGs table with expression values in normal and tumor samples
dataDEGsFiltLevel <- TCGAanalyze_LevelTab(dataDEGs,"Tumor","Normal",
                                          dataFilt[,samplesTP],dataFilt[,samplesNT])

The result is shown below:

Table of DEGs after DEA
mRNA logFC FDR Tumor Normal Delta
FN1 2.88 1.296151e-19 347787.48 41234.12 1001017.3
COL1A1 1.77 1.680844e-08 358010.32 89293.72 633086.3
C4orf7 5.20 2.826474e-50 87821.36 2132.76 456425.4
COL1A2 1.40 9.480478e-06 273385.44 91241.32 383242.9
GAPDH 1.32 3.290678e-05 179057.44 63663.00 236255.5
CLEC3A 6.79 7.971002e-74 27257.16 259.60 185158.6
IGFBP5 1.24 1.060717e-04 128186.88 53323.12 158674.6
CPB1 4.27 3.044021e-37 37001.76 2637.72 157968.8
CARTPT 6.72 1.023371e-72 21700.96 215.16 145872.8
DCD 7.26 1.047988e-80 19941.20 84.80 144806.3

TCGAanalyze_EAcomplete & TCGAvisualize_EAbarplot: Enrichment Analysis

Researchers, in order to better understand the underlying biological processes, often want to retrieve a functional profile of a set of genes that might have an important role. This can be done by performing an enrichment analysis.

We will perform an enrichment analysis on gene sets using the TCGAanalyze_EAcomplete function. Given a set of genes that are up-regulated under certain conditions, an enrichment analysis will identify classes of genes or proteins that are over-represented using annotations for that gene set.

To view the results you can use the TCGAvisualize_EAbarplot function as shown below.

library(TCGAbiolinks)
# Enrichment Analysis EA
# Gene Ontology (GO) and Pathway enrichment by DEGs list
Genelist <- rownames(dataDEGsFiltLevel)

system.time(ansEA <- TCGAanalyze_EAcomplete(TFname="DEA genes Normal Vs Tumor",Genelist))

# Enrichment Analysis EA (TCGAVisualize)
# Gene Ontology (GO) and Pathway enrichment barPlot

TCGAvisualize_EAbarplot(tf = rownames(ansEA$ResBP), 
                        GOBPTab = ansEA$ResBP,
                        GOCCTab = ansEA$ResCC,
                        GOMFTab = ansEA$ResMF,
                        PathTab = ansEA$ResPat,
                        nRGTab = Genelist, 
                        nBar = 10)

The result is shown below:

The figure shows canonical pathways significantly overrepresented (enriched) by the DEGs (differentially expressed genes). The most statistically significant canonical pathways identified in DEGs list are listed according to their p value corrected FDR (-Log) (colored bars) and the ratio of list genes found in each pathway over the total number of genes in that pathway (Ratio, red line).

TCGAanalyze_survival: Survival Analysis: Cox Regression and dnet package

When analyzing survival, different problems come up than the ones discussed so far. One question is how do we deal with subjects dropping out of a study. For example, assuming that we test a new cancer drug. While some subjects die, others may believe that the new drug is not effective, and decide to drop out of the study before the study is finished. A similar problem would be faced when we investigate how long a machine lasts before it breaks down.

Using the clinical data, it is possible to create a survival plot with the function TCGAanalyze_survival as follows:

clin.gbm <- GDCquery_clinic("TCGA-GBM", "clinical")
TCGAanalyze_survival(clin.gbm,
                     "gender",
                     main = "TCGA Set\n GBM",height = 10, width=10)

The arguments of TCGAanalyze_survival are:

  • clinical_patient TCGA Clinical patient with the information days_to_death
  • clusterCol Column with groups to plot. This is a mandatory field, the caption will be based in this column
  • legend Legend title of the figure
  • xlim xlim x axis limits e.g. xlim = c(0, 1000). Present narrower X axis, but not affect survival estimates.
  • main main title of the plot
  • ylab y-axis text of the plot
  • xlab x-axis text of the plot
  • filename The name of the pdf file
  • color Define the colors of the lines.
  • pvalue Show pvalue in the plot.
  • risk.table Show or not the risk table
  • conf.int Show confidence intervals for point estimaes of survival curves.

The result is shown below:

library(TCGAbiolinks)
# Survival Analysis SA

clinical_patient_Cancer <- GDCquery_clinic("TCGA-BRCA","clinical")
dataBRCAcomplete <- log2(BRCA_rnaseqv2)

tokenStop<- 1

tabSurvKMcomplete <- NULL

for( i in 1: round(nrow(dataBRCAcomplete)/100)){
    message( paste( i, "of ", round(nrow(dataBRCAcomplete)/100)))
    tokenStart <- tokenStop
    tokenStop <-100*i
    tabSurvKM<-TCGAanalyze_SurvivalKM(clinical_patient_Cancer,
                                      dataBRCAcomplete,
                                      Genelist = rownames(dataBRCAcomplete)[tokenStart:tokenStop],
                                      Survresult = F,
                                      ThreshTop=0.67,
                                      ThreshDown=0.33)
    
    tabSurvKMcomplete <- rbind(tabSurvKMcomplete,tabSurvKM)
}

tabSurvKMcomplete <- tabSurvKMcomplete[tabSurvKMcomplete$pvalue < 0.01,]
tabSurvKMcomplete <- tabSurvKMcomplete[order(tabSurvKMcomplete$pvalue, decreasing=F),]

tabSurvKMcompleteDEGs <- tabSurvKMcomplete[
    rownames(tabSurvKMcomplete) %in% dataDEGsFiltLevel$mRNA,
    ]

The result is shown below:

Table KM-survival genes after SA
pvalue Cancer Deaths Cancer Deaths with Top Cancer Deaths with Down
DCTPP1 6.204170e-08 66 46 20
APOO 9.390193e-06 65 49 16
LOC387646 1.039097e-05 69 48 21
PGK1 1.198577e-05 71 49 22
CCNE2 2.100348e-05 65 48 17
Mean Tumor Top Mean Tumor Down Mean Normal
DCTPP1 13.31 12.01 11.74
APOO 11.40 10.17 10.01
LOC387646 7.92 4.64 5.90
PGK1 15.66 14.18 14.28
CCNE2 11.07 8.23 7.03

TCGAanalyze_DMR: Differentially methylated regions Analysis

We will search for differentially methylated CpG sites using the TCGAanalyze_DMR function. In order to find these regions we use the beta-values (methylation values ranging from 0.0 to 1.0) to compare two groups.

First, it calculates the difference between the mean DNA methylation of each group for each probe.

Second, it test for differential expression between two groups using the wilcoxon test adjusting by the Benjamini-Hochberg method. The default arguments was set to require a minimum absolute beta-values difference of 0.2 and am adjusted p-value of < 0.01.

After these tests, we save a volcano plot (x-axis:diff mean methylation, y-axis: statistical significance) that will help the user identify the differentially methylated CpG sites, then the results are saved in a csv file (DMR_results.groupCol.group1.group2.csv) and finally the object is returned with the calculus in the rowRanges.

The arguments of TCGAanalyze_DMR are:

  • data SummarizedExperiment obtained from the GDCprepare
  • groupCol Columns with the groups inside the SummarizedExperiment object. (This will be obtained by the function colData(data)
  • group1 In case our object has more than 2 groups, you should set the name of the group
  • group2 In case our object has more than 2 groups, you should set the name of the group
  • plot.filename pdf filename. Default: volcano.pdf
  • legend Legend title
  • color vector of colors to be used in graph
  • title main title. If not specified it will be “Volcano plot (group1 vs group2)
  • ylab y axis text
  • xlab x axis text
  • xlim x limits to cut image
  • ylim y limits to cut image
  • label vector of labels to be used in the figure. Example: c(“Not Significant”, “Hypermethylated in group1”, “Hypomethylated in group1”))
  • p.cut p values threshold. Default: 0.01
  • diffmean.cut diffmean threshold. Default: 0.2
  • adj.method Adjusted method for the p-value calculation
  • paired Wilcoxon paired parameter. Default: FALSE
  • overwrite Overwrite the pvalues and diffmean values if already in the object for both groups? Default: FALSE
  • save save the object with the results?
  • cores use multiple cores for non-parametric test
  • filename Name of the rda file to save the object
data <- TCGAanalyze_DMR(data, groupCol = "methylation_subtype",
                        group1 = "CIMP.H",
                        group2="CIMP.L",
                        p.cut = 10^-5,
                        diffmean.cut = 0.25,
                        legend = "State",
                        plot.filename = "coad_CIMPHvsCIMPL_metvolcano.png")

The output will be a plot such as the figure below. The green dots are the probes that are hypomethylated in group 2 compared to group 1, while the red dots are the hypermethylated probes in group 2 compared to group 1

Also, the TCGAanalyze_DMR function will save the plot as pdf and return the same SummarizedExperiment that was given as input with the values of p-value, p-value adjusted, diffmean and the group it belongs in the graph (non significant, hypomethylated, hypermethylated) in the rowRanges. The collumns will be (where group1 and group2 are the names of the groups):

  • diffmean.group1.group2 (mean.group2 - mean.group1)
  • diffmean.group2.group1 (mean.group1 - mean.group2)
  • p.value.group1.group2
  • p.value.adj.group1.group2
  • status.group1.group2 (Status of probes in group2 in relation to group1)
  • status.group2.group1 (Status of probes in group1 in relation to group2)

This values can be view/acessed using the rowRanges acessesor (rowRanges(data)).

Observation: Calling the same function again, with the same arguments will only plot the results, as it was already calculated. If you want to have them recalculated, please set overwrite to TRUE or remove the calculated collumns.

TCGAvisualize: Visualize results from analysis functions with TCGA’s data.

You can easily visualize results from some following functions:

TCGAvisualize_Heatmap: Create heatmaps with cluster bars

In order to have a better view of clusters, we normally use heatmaps. TCGAvisualize_Heatmap will plot a heatmap and add to each sample bars representing different features. This function is a wrapper to the package ComplexHeatmap package,

The arguments of this function are:

  • data The object with the heatmap data (expression, methylation)
  • col.metadata Metadata for the columns (patients). It should have the column bcr_patient_barcode or patient or ID with the patients barcodes.
  • row.metadata Metadata for the rows genes (expression) or probes (methylation)
  • col.colors A list of names colors
  • row.colors A list of named colors
  • show_column_names Show column names names? Dafault: FALSE
  • show_row_names Show row names? Dafault: FALSE
  • cluster_rows Cluster rows ? Dafault: FALSE
  • cluster_columns Cluster columns ? Dafault: FALSE
  • sortCol Name of the column to be used to sort the columns
  • title Title of the plot
  • type Select the colors of the heatmap values. Possible values are “expression” (default), “methylation”
  • scale Use z-score to make the heamat? If we want to show differences between genes, it is good to make Z-score by samples (force each sample to have zero mean and standard deviation=1). If we want to show differences between samples, it is good to make Z-score by genes (force each gene to have zero mean and standard deviation=1). Possibilities: “row”, “col. Default”none"

For more information please take a look on case study #2.

The result is shown below:

TCGAvisualize_Volcano: Create volcano plot

Creates a volcano plot for DNA methylation or expression

The arguments of this function are:

  • x x-axis data
  • y y-axis data
  • y.cut p-values threshold. Default: 0.01
  • x.cut x-axis threshold. Default: 0.0
  • filename Filename. Default: volcano.pdf, volcano.svg, volcano.png
  • legend Legend title
  • color vector of colors to be used in graph
  • title main title. If not specified it will be Volcano plot (group1 vs group2)
  • ylab y axis text
  • xlab x axis text
  • xlim x limits to cut image
  • ylim y limits to cut image
  • height Figure height
  • width Figure width
  • names Names to be ploted if significant. Should be the same size of x and y
  • names.fill Names should be filled in a color box? Default: TRUE
  • names.size Size of the names text
  • dpi Figure dpi
  • label vector of labels to be used in the figure. Example: c(“Not Significant”,“Hypermethylated in group1”, “Hypomethylated in group1”))
  • highlight List of genes/probes to be highlighted. It should be in the names argument.
  • highlight.color Color of the points highlighted
  • show.names What names will be showd? Possibilities: “both”, “significant”, “highlighted”

For more information please take a look on case study #3.

TCGAvisualize_oncoprint

The visualize the MAF files we created a function called TCGAvisualize_oncoprint that uses the ComplexHeatmap to create an oncoprint.

  • mut Mutation file (see TCGAquery_maf from TCGAbiolinks)
  • genes Gene list
  • filename name of the pdf
  • color named vector for the plot
  • height pdf height
  • rm.empty.columns If there is no alteration in that sample, whether remove it on the oncoprint
  • show.row.barplot Show barplot annotation on rows?
  • show.column.names Show column names? Default: FALSE
  • rows.font.size Size of the fonts
  • labels.font.size Size of the fonts
  • annotation Matrix or data frame with the annotation. Should have a column bcr_patient_barcode with the same ID of the mutation object
  • annotation.position Position of the annotation “bottom” or “top”
  • label.title Title of the label
mut <- GDCquery_Maf(tumor = "ACC", pipelines = "muse")
clin <- GDCquery_clinic("TCGA-ACC","clinical")
clin <- clin[,c("bcr_patient_barcode","disease","gender","tumor_stage","race","vital_status")]
TCGAvisualize_oncoprint(mut = mut, genes = mut$Hugo_Symbol[1:20],
                        filename = "oncoprint.pdf",
                        annotation = clin,
                        color=c("background"="#CCCCCC","DEL"="purple","INS"="yellow","SNP"="brown"),
                        rows.font.size=10,
                        heatmap.legend.side = "right",
                        dist.col = 0, 
                        width = 5,
                        label.font.size = 10)

The columns are the samples and the rows are genes. The upper histograms shows the number of mutation of each samples, the right histogram shows the number of patients that has that mutation (which is also represented by the percentage numbers in the left side of the plot).

TCGAvisualize_PCA: Principal Component Analysis plot for differentially expressed genes

In order to better understand our genes, we can perform a PCA to reduce the number of dimensions of our gene set. The function TCGAvisualize_PCA will plot the PCA for different groups.

The arguments of this function are:

  • dataFilt The expression matrix after normalization and quantile filter
  • dataDEGsFiltLevel The TCGAanalyze_LevelTab output
  • ntopgenes number of DEGs genes to plot in PCA
  • ** group1 a string containing the barcode list of the samples in control group
  • ** group2 a string containing the barcode list of the samples in disease group
# normalization of genes
dataNorm <- TCGAbiolinks::TCGAanalyze_Normalization(dataBRCA, geneInfo)

# quantile filter of genes
dataFilt <- TCGAanalyze_Filtering(tabDF = dataNorm,
                                  method = "quantile", 
                                  qnt.cut =  0.25)

# selection of normal samples "NT" 
group1 <- TCGAquery_SampleTypes(colnames(dataFilt), typesample = c("NT"))
# selection of normal samples "TP" 
group2 <- TCGAquery_SampleTypes(colnames(dataFilt), typesample = c("TP"))

# Principal Component Analysis plot for ntop selected DEGs
 pca <- TCGAvisualize_PCA(dataFilt,dataDEGsFiltLevel, ntopgenes = 200, group1, group2)

The result is shown below:

TCGAvisualize_meanMethylation: Mean DNA Methylation Analysis

Using the data and calculating the mean DNA methylation per group, it is possible to create a mean DNA methylation boxplot with the function TCGAvisualize_meanMethylation as follows:

query <- GDCquery(project = "TCGA-GBM",
                  data.category = "DNA methylation",
                  platform = "Illumina Human Methylation 27",
                  legacy = TRUE, 
                  barcode = c("TCGA-02-0058-01A-01D-0186-05", "TCGA-12-1597-01B-01D-0915-05",
                              "TCGA-12-0829-01A-01D-0392-05", "TCGA-06-0155-01B-01D-0521-05",
                              "TCGA-02-0099-01A-01D-0199-05", "TCGA-19-4068-01A-01D-1228-05",
                              "TCGA-19-1788-01A-01D-0595-05", "TCGA-16-0848-01A-01D-0392-05"))
GDCdownload(query, method = "api")
data <- GDCprepare(query)
# "shortLetterCode" is a column in the SummarizedExperiment::colData(data) matrix
TCGAvisualize_meanMethylation(data, groupCol = "shortLetterCode",filename = NULL)
##   groups      Mean    Median       Max       Min
## 1     NT 0.5014940 0.5009496 0.5359795 0.4732053
## 2     TP 0.5014747 0.4958995 0.5298975 0.4656788
## 3     TR 0.5030028 0.4986486 0.5415967 0.4673249
##           NT        TP        TR
## NT        NA 0.9987060 0.9004057
## TP 0.9987060        NA 0.9031248
## TR 0.9004057 0.9031248        NA

The arguments of TCGAvisualize_meanMethylation are:

  • data SummarizedExperiment object obtained from GDCprepare
  • groupCol Columns in colData(data) that defines the groups. If no columns defined a columns called “Patients” will be used
  • subgroupCol Columns in colData(data) that defines the subgroups.
  • shapes Shape vector of the subgroups. It must have the size of the levels of the subgroups. Example: shapes = c(21,23) if for two levels
  • filename The name of the pdf that will be saved
  • subgroup.legend Name of the subgroup legend. DEFAULT: subgroupCol
  • group.legend Name of the group legend. DEFAULT: groupCol
  • color vector of colors to be used in graph
  • title main title in the plot
  • ylab y axis text in the plot
  • print.pvalue Print p-value for two groups in the plot
  • xlab x axis text in the plot
  • labels Labels of the groups
  • y.limits Change lower/upper y-axis limit

Other examples using the parameters:

# setting y limits: lower 0, upper 1
TCGAvisualize_meanMethylation(data,groupCol = "shortLetterCode", 
                              filename = NULL, y.limits = c(0,1))
##   groups      Mean    Median       Max       Min
## 1     NT 0.5014940 0.5009496 0.5359795 0.4732053
## 2     TP 0.5014747 0.4958995 0.5298975 0.4656788
## 3     TR 0.5030028 0.4986486 0.5415967 0.4673249
##           NT        TP        TR
## NT        NA 0.9987060 0.9004057
## TP 0.9987060        NA 0.9031248
## TR 0.9004057 0.9031248        NA

# setting y limits: lower 0
TCGAvisualize_meanMethylation(data,groupCol = "shortLetterCode", 
                              filename = NULL, y.limits = 0)
##   groups      Mean    Median       Max       Min
## 1     NT 0.5014940 0.5009496 0.5359795 0.4732053
## 2     TP 0.5014747 0.4958995 0.5298975 0.4656788
## 3     TR 0.5030028 0.4986486 0.5415967 0.4673249
##           NT        TP        TR
## NT        NA 0.9987060 0.9004057
## TP 0.9987060        NA 0.9031248
## TR 0.9004057 0.9031248        NA

# Changing shapes of jitters to show subgroups
TCGAvisualize_meanMethylation(data,
                              groupCol = "subtype_Pan.Glioma.DNA.Methylation.Cluster", 
                              subgroupCol ="vital_status", filename = NULL)
##   groups      Mean    Median       Max       Min
## 1 group1 0.5047761 0.4999907 0.5359795 0.4869987
## 2 group2 0.4913455 0.4921337 0.5188015 0.4656788
## 3 group3 0.5098497 0.5103386 0.5415967 0.4732053
##           group1    group2    group3
## group1        NA 0.2144950 0.6538003
## group2 0.2144950        NA 0.1437124
## group3 0.6538003 0.1437124        NA

# Sorting bars by descending mean methylation
TCGAvisualize_meanMethylation(data,
                              groupCol = "subtype_Pan.Glioma.DNA.Methylation.Cluster",
                              sort="mean.desc",
                              filename=NULL)
##   groups      Mean    Median       Max       Min
## 1 group1 0.5047761 0.4999907 0.5359795 0.4869987
## 2 group2 0.4913455 0.4921337 0.5188015 0.4656788
## 3 group3 0.5098497 0.5103386 0.5415967 0.4732053
##           group1    group2    group3
## group1        NA 0.2144950 0.6538003
## group2 0.2144950        NA 0.1437124
## group3 0.6538003 0.1437124        NA

# Sorting bars by asc mean methylation
TCGAvisualize_meanMethylation(data,
                              groupCol = "subtype_Pan.Glioma.DNA.Methylation.Cluster",
                              sort = "mean.asc",
                              filename=NULL)
##   groups      Mean    Median       Max       Min
## 1 group1 0.5047761 0.4999907 0.5359795 0.4869987
## 2 group2 0.4913455 0.4921337 0.5188015 0.4656788
## 3 group3 0.5098497 0.5103386 0.5415967 0.4732053
##           group1    group2    group3
## group1        NA 0.2144950 0.6538003
## group2 0.2144950        NA 0.1437124
## group3 0.6538003 0.1437124        NA

TCGAvisualize_meanMethylation(data,
                              groupCol = "vital_status",
                              sort = "mean.asc",
                              filename=NULL)
##   groups      Mean    Median       Max       Min
## 1  ALIVE 0.5014747 0.4958995 0.5298975 0.4656788
## 2   DEAD 0.5022484 0.4993197 0.5415967 0.4673249
##           ALIVE      DEAD
## ALIVE        NA 0.9413677
## DEAD  0.9413677        NA

TCGAvisualize_starburst: Integration of gene expression and DNA methylation data

The starburst plot is proposed to combine information from two volcano plots, and is applied for a study of DNA methylation and gene expression. It first introduced in 2010 (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. 2010). In order to reproduce this plot, we will use the TCGAvisualize_starburst function.

The function creates Starburst plot for comparison of DNA methylation and gene expression. The log10 (FDR-corrected P value) for DNA methylation is plotted in the x axis, and for gene expression in the y axis, for each gene. The black dashed line shows the FDR-adjusted P value of 0.01.

The arguments of this function are:

  • met SummarizedExperiment with methylation data obtained from the GDCprepare or Data frame from DMR_results file. Expected colData columns: diffmean, p.value.adj and p.value Execute volcanoPlot function in order to obtain these values for the object.
  • exp Matrix with expression data obtained from the TCGAanalyze_DEA function. Expected colData columns: logFC, FDR
  • names Add the names of the significant genes? Default: FALSE
  • names.fill Names should be filled in a color box? Default: TRUE
  • circle Circle pair gene/probe that respects diffmean.cut and logFC.cut
  • filename pdf filename
  • legend legend title
  • color vector of colors to be used in graph
  • label vector of labels to be used in graph
  • title main title
  • ylab y axis text
  • xlab x axis text
  • xlim x limits to cut image
  • ylim y limits to cut image
  • p.cut p value cut-off
  • group1 The name of the group 1 Obs: Column p.value.adj.group1.group2 should exist
  • group2 The name of the group 2. Obs: Column p.value.adj.group1.group2 should exist
  • exp.p.cut expression p value cut-off
  • met.p.cut methylation p value cut-off
  • diffmean.cut If set, the probes with diffmean higher than methylation cut-off will be highlighted in the plot. And the data frame return will be subseted.
  • logFC.cut If set, the probes with expression fold change higher than methylation cut-off will be highlighted in the plot. And the data frame return will be subseted.
  • height Figure height
  • width Figure width
  • dpi Figure dpi
starburst <- TCGAvisualize_starburst(coad.SummarizeExperiment, 
                                     different.experssion.analysis.data,
                                     group1 = "CIMP.H",
                                     group2 = "CIMP.L",
                                     met.p.cut = 10^-5, 
                                     exp.p.cut=10^-5,
                                     names = TRUE)

As a result, the function will a plot the figure below and return a matrix with the Gene_symbol and it status in relation to expression (up regulated/down regulated) and to methylation (Hyper/Hypo methylated). The case study 3, shows the complete pipeline for creating this figure.

GDC Downstream Pipeline

HTSeq data: Downstream analysis BRCA

CancerProject <- "TCGA-BRCA"
DataDirectory <- paste0("../GDC/",gsub("-","_",CancerProject))
FileNameData <- paste0(DataDirectory, "_","HTSeq_Counts",".rda")

query <- GDCquery(project = CancerProject,
                  data.category = "Transcriptome Profiling",
                  data.type = "Gene Expression Quantification", 
                  workflow.type = "HTSeq - Counts")

samplesDown <- getResults(query,cols=c("cases"))

dataSmTP <- TCGAquery_SampleTypes(barcode = samplesDown,
                                  typesample = "TP")

dataSmNT <- TCGAquery_SampleTypes(barcode = samplesDown,
                                  typesample = "NT")
dataSmTP_short <- dataSmTP[1:10]
dataSmNT_short <- dataSmNT[1:10]

queryDown <- GDCquery(project = CancerProject, 
                      data.category = "Transcriptome Profiling",
                      data.type = "Gene Expression Quantification", 
                      workflow.type = "HTSeq - Counts", 
                      barcode = c(dataSmTP_short, dataSmNT_short))
                    
GDCdownload(query = queryDown,
            directory = DataDirectory)

dataPrep <- GDCprepare(query = queryDown, 
                       save = TRUE, 
                       directory =  DataDirectory,
                       save.filename = FileNameData)

dataPrep <- TCGAanalyze_Preprocessing(object = dataPrep, 
                                      cor.cut = 0.6,
                                      datatype = "HTSeq - Counts")                      

dataNorm <- TCGAanalyze_Normalization(tabDF = dataPrep,
                                      geneInfo = geneInfoHT,
                                      method = "gcContent") 

boxplot(dataPrep, outline = FALSE)

boxplot(dataNorm, outline = FALSE)

dataFilt <- TCGAanalyze_Filtering(tabDF = dataNorm,
                                  method = "quantile", 
                                  qnt.cut =  0.25)   

dataDEGs <- TCGAanalyze_DEA(mat1 = dataFilt[,dataSmTP_short],
                            mat2 = dataFilt[,dataSmNT_short],
                            Cond1type = "Normal",
                            Cond2type = "Tumor",
                            fdr.cut = 0.01 ,
                            logFC.cut = 1,
                            method = "glmLRT")  

miRNA expression data: Downstream analysis BRCA

require(TCGAbiolinks)

CancerProject <- "TCGA-BRCA"
DataDirectory <- paste0("../GDC/",gsub("-","_",CancerProject))
FileNameData <- paste0(DataDirectory, "_","miRNA_gene_quantification",".rda")

query.miR <- GDCquery(project = CancerProject, 
                  data.category = "Gene expression",
                  data.type = "miRNA gene quantification",
                  file.type = "hg19.mirna",
                  legacy = TRUE)

samplesDown.miR <- getResults(query.miR,cols=c("cases"))

dataSmTP.miR <- TCGAquery_SampleTypes(barcode = samplesDown.miR,
                                  typesample = "TP")

dataSmNT.miR <- TCGAquery_SampleTypes(barcode = samplesDown.miR,
                                  typesample = "NT")
dataSmTP_short.miR <- dataSmTP.miR[1:10]
dataSmNT_short.miR <- dataSmNT.miR[1:10]

queryDown.miR <- GDCquery(project = CancerProject, 
                      data.category = "Gene expression",
                      data.type = "miRNA gene quantification",
                      file.type = "hg19.mirna",
                      legacy = TRUE,
                      barcode = c(dataSmTP_short.miR, dataSmNT_short.miR))

GDCdownload(query = queryDown.miR,
            directory = DataDirectory)

dataAssy.miR <- GDCprepare(query = queryDown.miR, 
                           save = TRUE, 
                           save.filename = FileNameData, 
                           summarizedExperiment = TRUE,
                           directory =DataDirectory )
rownames(dataAssy.miR) <- dataAssy.miR$miRNA_ID

# using read_count's data 
read_countData <-  colnames(dataAssy.miR)[grep("count", colnames(dataAssy.miR))]
dataAssy.miR <- dataAssy.miR[,read_countData]
colnames(dataAssy.miR) <- gsub("read_count_","", colnames(dataAssy.miR))

dataFilt <- TCGAanalyze_Filtering(tabDF = dataAssy.miR,
                                  method = "quantile", 
                                  qnt.cut =  0.25)   

dataDEGs <- TCGAanalyze_DEA(mat1 = dataFilt[,dataSmNT_short.miR],
                            mat2 = dataFilt[,dataSmTP_short.miR],
                            Cond1type = "Normal",
                            Cond2type = "Tumor",
                            fdr.cut = 0.01 ,
                            logFC.cut = 1,
                            method = "glmLRT")  

TCGA Downstream Analysis: Case Studies

Introduction

This vignette shows a complete workflow of the TCGAbiolinks package. The code is divided in 4 case study:

    1. Expression pipeline (BRCA)
    1. Expression pipeline (GBM)
    1. Integration of DNA methylation and RNA expression pipeline (COAD)
    1. Elmer pipeline (KIRC)

Case study n. 1: Pan Cancer downstream analysis BRCA

library(SummarizedExperiment)
library(TCGAbiolinks)
query.exp <- GDCquery(project = "TCGA-BRCA", 
                      legacy = TRUE,
                      data.category = "Gene expression",
                      data.type = "Gene expression quantification",
                      platform = "Illumina HiSeq", 
                      file.type = "results",
                      experimental.strategy = "RNA-Seq",
                      sample.type = c("Primary solid Tumor","Solid Tissue Normal"))
GDCdownload(query.exp)
brca.exp <- GDCprepare(query = query.exp, save = TRUE, save.filename = "brcaExp.rda")

# get subtype information
dataSubt <- TCGAquery_subtype(tumor = "BRCA")

# get clinical data
dataClin <- GDCquery_clinic(project = "TCGA-BRCA","clinical") 

# Which samples are primary solid tumor
dataSmTP <- TCGAquery_SampleTypes(getResults(query.exp,cols="cases"),"TP") 
# which samples are solid tissue normal
dataSmNT <- TCGAquery_SampleTypes(getResults(query.exp,cols="cases"),"NT")

Using TCGAnalyze_DEA, we identified 3,390 differentially expression genes (DEG) (log fold change >=1 and FDR < 1%) between 114 normal and 1097 BRCA samples. In order to understand the underlying biological process from DEGs we performed an enrichment analysis using TCGAnalyze_EA_complete function.

dataPrep <- TCGAanalyze_Preprocessing(object = brca.exp, cor.cut = 0.6)                      

dataNorm <- TCGAanalyze_Normalization(tabDF = dataPrep,
                                      geneInfo = geneInfo,
                                      method = "gcContent")                

dataFilt <- TCGAanalyze_Filtering(tabDF = dataNorm,
                                  method = "quantile", 
                                  qnt.cut =  0.25)   

dataDEGs <- TCGAanalyze_DEA(mat1 = dataFilt[,dataSmNT],
                            mat2 = dataFilt[,dataSmTP],
                            Cond1type = "Normal",
                            Cond2type = "Tumor",
                            fdr.cut = 0.01 ,
                            logFC.cut = 1,
                            method = "glmLRT")  

TCGAbiolinks outputs bar chart with the number of genes for the main categories of three ontologies (GO:biological process, GO:cellular component, and GO:molecular function, respectively).

ansEA <- TCGAanalyze_EAcomplete(TFname="DEA genes Normal Vs Tumor",
                                RegulonList = rownames(dataDEGs))  

TCGAvisualize_EAbarplot(tf = rownames(ansEA$ResBP),
                        GOBPTab = ansEA$ResBP,
                        GOCCTab = ansEA$ResCC,
                        GOMFTab = ansEA$ResMF,
                        PathTab = ansEA$ResPat,
                        nRGTab = rownames(dataDEGs),
                        nBar = 20)

The figure resulted from the code above is shown below.

The Kaplan-Meier analysis was used to compute survival univariate curves, and
log-Ratio test was computed to assess the statistical significance by using TCGAanalyze_SurvivalKM function; starting with 3,390 DEGs genes we found 555 significantly genes with p.value <0.05.

group1 <- TCGAquery_SampleTypes(colnames(dataFilt), typesample = c("NT"))
group2 <- TCGAquery_SampleTypes(colnames(dataFilt), typesample = c("TP"))

dataSurv <- TCGAanalyze_SurvivalKM(clinical_patient = dataClin,
                                   dataGE = dataFilt,
                                   Genelist = rownames(dataDEGs),
                                   Survresult = FALSE,
                                   ThreshTop = 0.67,
                                   ThreshDown = 0.33,
                                   p.cut = 0.05, group1, group2)

Cox-regression analysis was used to compute survival multivariate curves, and cox p-value was computed to assess the statistical significance by using TCGAnalyze_SurvivalCoxNET function. Survival multivariate analysis found 160 significantly genes according to the cox p-value FDR 5.00e-02. From DEGs that we found to correlate significantly with survival by both univariate and multivariate analyses we analyzed the following network.

The interactome network graph was generated using STRING.,org.Hs.string version 10 (Human functional protein association network). The network graph was resized by dnet package considering only multivariate survival genes, with strong interaction (threshold = 700) we obtained a subgraphsub graph of 24 nodes and 31 edges.

require(dnet)  # to change
org.Hs.string <- dRDataLoader(RData = "org.Hs.string")

TabCoxNet <- TCGAvisualize_SurvivalCoxNET(dataClin,
                                          dataFilt, 
                                          Genelist = rownames(dataSurv),
                                          scoreConfidence = 700,
                                          org.Hs.string = org.Hs.string,
                                          titlePlot = "Case Study n.1 dnet")

The figure resulted from the code above is shown below.

Case study n. 2: Pan Cancer downstream analysis LGG

We focused on the analysis of LGG samples. In particular, we used TCGAbiolinks to download 293 samples with molecular subtypes. Link the complete complete code. .

library(TCGAbiolinks)
library(SummarizedExperiment)

query.exp <- GDCquery(project = "TCGA-LGG", 
                      legacy = TRUE,
                      data.category = "Gene expression",
                      data.type = "Gene expression quantification",
                      platform = "Illumina HiSeq", 
                      file.type = "results",
                      experimental.strategy = "RNA-Seq",
                      sample.type = "Primary solid Tumor")
GDCdownload(query.exp)
lgg.exp <- GDCprepare(query = query.exp, save = TRUE, save.filename = "lggExp.rda")

# get subtype information
dataSubt <- TCGAquery_subtype(tumor = "LGG")

# get indexed clinical data
dataClin <- GDCquery_clinic(project = "TCGA-LGG", "Clinical")

First, we searched for possible outliers using the TCGAanalyze_Preprocessing function, which performs an Array Array Intensity correlation AAIC. We used all samples in expression data which contain molecular subtypes, filtering out samples without molecular information, and using only IDHmut-codel (n=85), IDHmut-non-codel (n=141) and IDHwt (n=56), NA (11), to define a square symetric matrix of pearson correlation among all samples (n=293). According to this matrix we found no samples with low correlation (cor.cut = 0.6) that can be identified as possible outliers, so we continued our analysis with 70 samples.

Second, using the TCGAanalyze_Normalization function we normalized mRNA transcripts and miRNA, using EDASeq package. This function does use Within-lane normalization procedures to adjust for GC-content effect (or other gene-level effects) on read counts: loess robust local regression, global-scaling, and full-quantile normalization (Risso, D., Schwartz, K., Sherlock, G., & Dudoit, S. 2011) and between-lane normalization procedures to adjust for distributional differences between lanes (e.g., sequencing depth): global-scaling and full-quantile normalization (James H Bullard, Elizabeth Purdom, Kasper D Hansen and Sandrine Dudoit 2010).

Third, using the TCGAanalyze_Filtering function we applied 3 filters removing features / mRNAs with low signal across samples obtaining 4578, 4284, 1187 mRNAs respectively.

Then we applied two Hierarchical cluster analysis on 1187 mRNAs after the three filters described above, the first cluster using as method ward.D2, and the second with ConsensusClusterPlus.

After the two clustering analysis, with cut.tree =4 we obtained n=4 espression clusters (EC).

# expression data with molecular subtypes
lgg.exp <- subset(lgg.exp, select = colData(lgg.exp)$patient %in% dataSubt$patient)

dataPrep <- TCGAanalyze_Preprocessing(object = lgg.exp,cor.cut = 0.6)
dataNorm <- TCGAanalyze_Normalization(tabDF = dataPrep,
                                      geneInfo = geneInfo,
                                      method = "gcContent")

datFilt1 <- TCGAanalyze_Filtering(tabDF = dataNorm,method = "varFilter")
datFilt2 <- TCGAanalyze_Filtering(tabDF = datFilt1,method = "filter1")
datFilt <- TCGAanalyze_Filtering(tabDF = datFilt2,method = "filter2")

data_Hc1 <- TCGAanalyze_Clustering(tabDF = datFilt,
                                   method = "hclust",
                                   methodHC = "ward.D2")
data_Hc2 <- TCGAanalyze_Clustering(tabDF = datFilt,
                                   method = "consensus",
                                   methodHC = "ward.D2") 

For the next steps, we will merge the clinical data with with the the cluster information and we will add the subtype information.

#------  Add cluster information
cluster <- data.frame("groupsHC" = data_Hc2[[4]]$consensusClass)
cluster$groupsHC <- paste0("EC",cluster$groupsHC)
cluster$patient <-  substr(colData(lgg.exp)$patient,1,12)

# Add information about gropus from consensus Cluster in clinical data
dataClin <- merge(dataClin,cluster, by.x="bcr_patient_barcode", by.y="patient")

# Merge subtype and clinical data
clin_subt <- merge(dataClin,dataSubt, by.x="bcr_patient_barcode", by.y="patient")
clin_subt_all <- merge(dataClin,dataSubt, 
                       by.x="bcr_patient_barcode", by.y="patient", all.x = TRUE)

The next steps will be to visualize the data. First, we created the survival plot.

#----------- VISUALIZE --------------------
# plotting survival for groups EC1, EC2, EC3, EC4
TCGAanalyze_survival(data = clin_subt_all,
                     clusterCol = "groupsHC",
                     main = "TCGA kaplan meier survival plot from consensus cluster",
                     legend = "RNA Group",
                     color = c("black","red","blue","green3"),
                     filename = "case2_surv.png")

The result is showed below:

We will also, create a heatmap of the expression.

TCGAvisualize_Heatmap(t(datFilt),
                      col.metadata =  clin_subt[,c("bcr_patient_barcode",
                                                   "groupsHC",
                                                   "Histology",
                                                   "IDH.codel.subtype")],
                      col.colors =  list(
                          groupsHC = c("EC1"="black",
                                       "EC2"="red",
                                       "EC3"="blue",
                                       "EC4"="green3"),
                          Histology=c("astrocytoma"="navy",
                                      "oligoastrocytoma"="green3",
                                      "oligodendroglioma"="red"),
                          IDH.codel.subtype = c("IDHmut-codel"="tomato",
                                                "IDHmut-non-codel"="navy",
                                                "IDHwt"="gold","NA"="white")),
                      sortCol = "groupsHC",
                      type = "expression", # sets default color
                      scale = "row", # use z-scores for better visualization
                      title = "Heatmap from concensus cluster", 
                      filename = "case2_Heatmap.pdf",
                      cluster_rows = TRUE)

The result is shown below:

Finally, we will take a look in the mutation genes. We will first download the maf file with TCGAquery_maf. In this example we will investigate the gene “ATRX”.

LGGmut <- GDCquery_Maf(tumor = "LGG", pipelines = "muse")
# Selecting gene
mRNAsel <- "ATRX"
LGGselected <- LGGmut[LGGmut$Hugo_Symbol == mRNAsel,]

dataMut <- LGGselected[!duplicated(LGGselected$Tumor_Sample_Barcode),]
dataMut$Tumor_Sample_Barcode <- substr(dataMut$Tumor_Sample_Barcode,1,12)

# Adding the Expression Cluster classification found before
dataMut <- merge(dataMut, cluster, by.y="patient", by.x="Tumor_Sample_Barcode")
dataMut <- dataMut[dataMut$Variant_Classification!=0,]

Case study n. 3: Integration of methylation and expression for ACC

In recent years, it has been described the relationship between DNA methylation and gene expression and the study of this relationship is often difficult to accomplish.

This case study will show the steps to investigate the relationship between the two types of data.

First, we downloaded ACC DNA methylation data for HumanMethylation450k platforms, and ACC RNA expression data for Illumina HiSeq platform.

TCGAbiolinks adds by default the subtypes classification already published by researchers.

We will use this classification to do our examples. So, selected the groups CIMP-low and CIMP-high to do RNA expression and DNA methylation comparison.

library(TCGAbiolinks)
library(SummarizedExperiment)
dir.create("case3")
setwd("case3")
#-----------------------------------
# STEP 1: Search, download, prepare |
#-----------------------------------
# 1.1 - DNA methylation
# ----------------------------------
query.met <- GDCquery(project = "TCGA-ACC", 
                      legacy = TRUE,
                      data.category = "DNA methylation",
                      platform = "Illumina Human Methylation 450")
GDCdownload(query.met)

acc.met <- GDCprepare(query = query.met,
                      save = TRUE, 
                      save.filename = "accDNAmet.rda",
                      summarizedExperiment = TRUE)

#-----------------------------------
# 1.2 - RNA expression
# ----------------------------------
query.exp <- GDCquery(project = "TCGA-ACC", 
                      legacy = TRUE,
                      data.category = "Gene expression",
                      data.type = "Gene expression quantification",
                      platform = "Illumina HiSeq", 
                      file.type = "results")
GDCdownload(query.exp)
acc.exp <- GDCprepare(query = query.exp, save = TRUE, save.filename = "accExp.rda")

For DNA methylation, we perform a DMR (different methylated region) analysis, which will give the difference of DNA methylation for the probes of the groups and their significance value. The output can be seen in a volcano plot. Note: Depending on the number of samples this function can be very slow due to the wilcoxon test, taking from hours to days.

# na.omit
acc.met <- subset(acc.met,subset = (rowSums(is.na(assay(acc.met))) == 0))

# Volcano plot
acc.met <- TCGAanalyze_DMR(acc.met, groupCol = "subtype_MethyLevel",
                           group1 = "CIMP-high",
                           group2="CIMP-low",
                           p.cut = 10^-5,
                           diffmean.cut = 0.25,
                           legend = "State",
                           plot.filename = "CIMP-highvsCIMP-low_metvolcano.png")

The figure resulted from the code above is shown below:

For the expression analysis, we do a DEA (differential expression analysis) which will give the fold change of gene expression and their significance value.

#-------------------------------------------------
# 2.3 - DEA - Expression analysis - volcano plot
# ------------------------------------------------
acc.exp.aux <- subset(acc.exp, 
                      select = colData(acc.exp)$subtype_MethyLevel %in% c("CIMP-high","CIMP-low"))

idx <- colData(acc.exp.aux)$subtype_MethyLevel %in% c("CIMP-high")
idx2 <- colData(acc.exp.aux)$subtype_MethyLevel %in% c("CIMP-low")

dataPrep <- TCGAanalyze_Preprocessing(object = acc.exp.aux, cor.cut = 0.6)

dataNorm <- TCGAanalyze_Normalization(tabDF = dataPrep,
                                      geneInfo = geneInfo,
                                      method = "gcContent")

dataFilt <- TCGAanalyze_Filtering(tabDF = dataNorm,
                                  qnt.cut = 0.25,
                                  method='quantile')

dataDEGs <- TCGAanalyze_DEA(mat1 = dataFilt[,idx],
                            mat2 = dataFilt[,idx2],
                            Cond1type = "CIMP-high",
                            Cond2type = "CIMP-low",
                            method = "glmLRT")

TCGAVisualize_volcano(dataDEGs$logFC,dataDEGs$FDR,
                      filename = "Case3_volcanoexp.png",
                      x.cut = 3,
                      y.cut = 10^-5,
                      names = rownames(dataDEGs),
                      color = c("black","red","darkgreen"),
                      names.size = 2,
                      xlab = " Gene expression fold change (Log2)",
                      legend = "State",
                      title = "Volcano plot (CIMP-high vs CIMP-low)",
                      width = 10)

The figure resulted from the code above is shown below:

Finally, using both previous analysis we do a starburst plot to select the genes that are Candidate Biologically Significant.

Observation: over the time, the number of samples has increased and the clinical data updated. We used only the samples that had a classification in the examples.

#------------------------------------------
# 2.4 - Starburst plot
# -----------------------------------------
# If true the argument names of the geenes in circle 
# (biologically significant genes, has a change in gene
# expression and DNA methylation and respects all the thresholds)
# will be shown
# these genes are returned by the function see starburst object after the function is executed
starburst <- TCGAvisualize_starburst(acc.met, dataDEGs,"CIMP-high","CIMP-low",
                                     filename = "starburst.png",
                                     met.p.cut = 10^-5,
                                     exp.p.cut = 10^-5,
                                     diffmean.cut = 0.25,
                                     logFC.cut = 3,
                                     names = FALSE, 
                                     height=10,
                                     width=15,
                                     dpi=300)

The figure resulted from the code above is shown below:

Case study n. 4: Elmer pipeline - KIRC

One of the biggest problems related to anlysis of data is the preparation phase, which often consists of successive steps in order to prepare it to a format acceptable by certain algorithms and software.

With the objective of assisting users in this arduous step, TCGAbiolinks will offer to prepare the data in order to obtain the correct format for different packages.

An example of package to perform DNA methylation and RNA expression analysis is ELMER (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 2015). ELMER, which is designed to combine DNA methylation and gene expression data from human tissues to infer multi-level cis-regulatory networks. ELMER uses DNA methylation to identify enhancers, and correlates enhancer state with expression of nearby genes to identify one or more transcriptional targets. Transcription factor (TF) binding site analysis of enhancers is coupled with expression analysis of all TFs to infer upstream regulators. This package can be easily applied to TCGA public available cancer data sets and custom DNA methylation and gene expression data sets.

ELMER analyses have 5 main steps:

  1. Identify distal enhancer probes on HM450K.
  2. Identify distal enhancer probes with significantly different DNA methyaltion level in control group and experiment group.
  3. Identify putative target genes for differentially methylated distal enhancer probes.
  4. Identify enriched motifs for the distal enhancer probes which are significantly differentially methylated and linked to putative target gene.
  5. Identify regulatory TFs whose expression associate with DNA methylation at motifs.

We will present this the study KIRC by TCGAbiolinks and ELMER integration. For more information, please consult the ELMER package.

For the DNA methylation data we will search the platform HumanMethylation450 and level 3 data with TCGAquery. After, we will download the data with TCGAdownload and the data will be prepared into a data.frame object with TCGAprepare. TCGAprepare_elmer will be used to prepare the data frame to ELMER.

library(TCGAbiolinks)
library(SummarizedExperiment)
library(ELMER)
library(parallel)
dir.create("case4")
setwd("case4")
#-----------------------------------
# STEP 1: Search, download, prepare |
#-----------------------------------
# 1.1 - DNA methylation
# ----------------------------------
query.met <- GDCquery(project = "TCGA-KIRC", 
                      legacy = TRUE,
                      data.category = "DNA methylation",
                      platform = "Illumina Human Methylation 450")
GDCdownload(query.met)
kirc.met <- GDCprepare(query = query.met,
                       save = TRUE, 
                       save.filename = "kircDNAmet.rda",
                       summarizedExperiment = TRUE)


kirc.met <- TCGAprepare_elmer(kirc.met, 
                              platform = "HumanMethylation450",
                              save = TRUE,
                              met.na.cut = 0.2)

For the gene expression data we will search the platform IlluminaHiSeq_RNASeqV2 and level 3 data with TCGAquery. Next, we will download the normalized_results data with TCGAdownload and prepare it into a data.frame object with TCGAprepare. TCGAprepare_elmer will be used to prepare the data frame to ELMER.

# Step 1.2 download expression data
#-----------------------------------
# 1.2 - RNA expression
# ----------------------------------
query.exp <- GDCquery(project = "TCGA-KIRC", 
                      legacy = TRUE,
                      data.category = "Gene expression",
                      data.type = "Gene expression quantification",
                      platform = "Illumina HiSeq", 
                      file.type = "normalized_results")
GDCdownload(query.exp)
kirc.exp <- GDCprepare(query = query.exp, save = TRUE, save.filename = "kirkExp.rda")

kirc.exp <- TCGAprepare_elmer(kirc.exp,
                              save = TRUE,
                              platform = "IlluminaHiSeq_RNASeqV2") 

The ELMER input is a mee object that contains the methylation matrix, the expression matrix, the probe information, the gene information and a table with a summary of the data.

#-----------------------------------
# STEP 2: ELMER integration         |
#-----------------------------------
# Step 2.1 prepare mee object       |
# -----------------------------------
library(ELMER)
library(parallel)

geneAnnot <- txs()
geneAnnot$GENEID <- paste0("ID",geneAnnot$GENEID)
geneInfo <- promoters(geneAnnot,upstream = 0, downstream = 0)
probe <- get.feature.probe()
mee <- fetch.mee(meth = kirc.met, exp = kirc.exp, TCGA = TRUE,
                 probeInfo = probe, geneInfo = geneInfo)

The code below excute steps 2-5 showed above:

direction <- c("hyper","hypo")

for (j in direction){
    print(j)
    dir.out <- paste0("kirc/",j)
    dir.create(dir.out, recursive = TRUE)
    #--------------------------------------
    # STEP 3: Analysis                     |
    #--------------------------------------
    # Step 3.1: Get diff methylated probes |
    #--------------------------------------
    Sig.probes <- get.diff.meth(mee, cores=detectCores(),
                                dir.out =dir.out,
                                diff.dir=j,
                                pvalue = 0.01)
    
    #-------------------------------------------------------------
    # Step 3.2: Identify significant probe-gene pairs            |
    #-------------------------------------------------------------
    # Collect nearby 20 genes for Sig.probes
    nearGenes <- GetNearGenes(TRange=getProbeInfo(mee, probe=Sig.probes$probe),
                              cores=detectCores(),
                              geneAnnot=getGeneInfo(mee))
    
    pair <- get.pair(mee=mee,
                     probes=Sig.probes$probe,
                     nearGenes=nearGenes,
                     permu.dir=paste0(dir.out,"/permu"),
                     dir.out=dir.out,
                     cores=detectCores(),
                     label= j,
                     permu.size=10000,
                     Pe = 0.001)
    
    Sig.probes.paired <- fetch.pair(pair=pair,
                                    probeInfo = getProbeInfo(mee),
                                    geneInfo = getGeneInfo(mee))
    Sig.probes.paired <-read.csv(paste0(dir.out,"/getPair.",j,".pairs.significant.csv"),
                                 stringsAsFactors=FALSE)[,1]
    
    
    #-------------------------------------------------------------
    # Step 3.3: Motif enrichment analysis on the selected probes |
    #-------------------------------------------------------------
    if(length(Sig.probes.paired) > 0 ){
        #-------------------------------------------------------------
        # Step 3.3: Motif enrichment analysis on the selected probes |
        #-------------------------------------------------------------
        enriched.motif <- get.enriched.motif(probes=Sig.probes.paired,
                                             dir.out=dir.out, label=j,
                                             background.probes = probe$name)
        if(length(enriched.motif) > 0){
            #-------------------------------------------------------------
            # Step 3.4: Identifying regulatory TFs                        |
            #-------------------------------------------------------------
            print("get.TFs")
            
            TF <- get.TFs(mee = mee,
                          enriched.motif = enriched.motif,
                          dir.out = dir.out,
                          cores = detectCores(), label = j)
            save(TF, enriched.motif, Sig.probes.paired,
                 pair, nearGenes, Sig.probes,
                 file=paste0(dir.out,"/ELMER_results_",j,".rda"))
        }
    }
}

From this analysis it is possible to verify the relationship between nearby 20 gene expression vs DNA methylation at this probe. The result of this is show by the ELMER scatter plot.

scatter.plot(mee,byProbe=list(probe=c("cg00328720"),geneNum=20), category="TN", lm_line = TRUE)

The result is shown below:

Each scatter plot showing the average DNA methylation level of sites with the UA6 motif in all KIRC samples plotted against the expression of the transcription factor ZNF677 and PEG3 respectively.

scatter.plot(mee,byTF = list(TF = c("ZNF677","PEG3"),
                          probe = enriched.motif[["UA6"]]), category = "TN",
                          save = TRUE, lm_line = TRUE, dir.out = "kirc")

The result is shown below:

The schematic plot shows probe colored in blue and the location of nearby 20 genes. The genes significantly linked to the probe were shown in red.

pair <- fetch.pair(pair="./kirc/getPair.hypo.pairs.significant.csv",
                   probeInfo = mee@probeInfo, geneInfo = mee@geneInfo)
schematic.plot(pair=pair, byProbe="cg15862394",save=FALSE)

The result is shown below:

The plot shows the odds ratio (x axis) for the selected motifs with OR above 1.3 and lower boundary of OR above 1.3. The range shows the 95% confidence interval for each Odds Ratio.


Session Information


sessionInfo()
## R version 3.4.1 (2017-06-30)
## Platform: x86_64-pc-linux-gnu (64-bit)
## Running under: Ubuntu 16.04.3 LTS
## 
## Matrix products: default
## BLAS: /home/biocbuild/bbs-3.5-bioc/R/lib/libRblas.so
## LAPACK: /home/biocbuild/bbs-3.5-bioc/R/lib/libRlapack.so
## 
## locale:
##  [1] LC_CTYPE=en_US.UTF-8       LC_NUMERIC=C              
##  [3] LC_TIME=en_US.UTF-8        LC_COLLATE=C              
##  [5] LC_MONETARY=en_US.UTF-8    LC_MESSAGES=en_US.UTF-8   
##  [7] LC_PAPER=en_US.UTF-8       LC_NAME=C                 
##  [9] LC_ADDRESS=C               LC_TELEPHONE=C            
## [11] LC_MEASUREMENT=en_US.UTF-8 LC_IDENTIFICATION=C       
## 
## attached base packages:
##  [1] grid      parallel  stats4    stats     graphics  grDevices utils    
##  [8] datasets  methods   base     
## 
## other attached packages:
##  [1] png_0.1-7                  TCGAbiolinks_2.5.9        
##  [3] bindrcpp_0.2               DT_0.2                    
##  [5] dplyr_0.7.3                SummarizedExperiment_1.6.3
##  [7] DelayedArray_0.2.7         matrixStats_0.52.2        
##  [9] Biobase_2.36.2             GenomicRanges_1.28.5      
## [11] GenomeInfoDb_1.12.2        IRanges_2.10.3            
## [13] S4Vectors_0.14.4           BiocGenerics_0.22.0       
## 
## loaded via a namespace (and not attached):
##   [1] backports_1.1.0             circlize_0.4.1             
##   [3] aroma.light_3.6.0           plyr_1.8.4                 
##   [5] selectr_0.3-1               ConsensusClusterPlus_1.40.0
##   [7] lazyeval_0.2.0              splines_3.4.1              
##   [9] BiocParallel_1.10.1         ggplot2_2.2.1              
##  [11] digest_0.6.12               foreach_1.4.3              
##  [13] htmltools_0.3.6             viridis_0.4.0              
##  [15] magrittr_1.5                memoise_1.1.0              
##  [17] cluster_2.0.6               doParallel_1.0.10          
##  [19] limma_3.32.6                ComplexHeatmap_1.14.0      
##  [21] Biostrings_2.44.2           readr_1.1.1                
##  [23] annotate_1.54.0             R.utils_2.5.0              
##  [25] colorspace_1.3-2            blob_1.1.0                 
##  [27] rvest_0.3.2                 ggrepel_0.6.5              
##  [29] crayon_1.3.2                RCurl_1.95-4.8             
##  [31] jsonlite_1.5                roxygen2_6.0.1             
##  [33] genefilter_1.58.1           bindr_0.1                  
##  [35] zoo_1.8-0                   survival_2.41-3            
##  [37] iterators_1.0.8             glue_1.1.1                 
##  [39] survminer_0.4.0             gtable_0.2.0               
##  [41] zlibbioc_1.22.0             XVector_0.16.0             
##  [43] GetoptLong_0.1.6            kernlab_0.9-25             
##  [45] shape_1.4.3                 prabclus_2.2-6             
##  [47] DEoptimR_1.0-8              scales_0.5.0               
##  [49] DESeq_1.28.0                mvtnorm_1.0-6              
##  [51] DBI_0.7                     edgeR_3.18.1               
##  [53] ggthemes_3.4.0              Rcpp_0.12.12               
##  [55] viridisLite_0.2.0           xtable_1.8-2               
##  [57] cmprsk_2.2-7                foreign_0.8-69             
##  [59] bit_1.1-12                  matlab_1.0.2               
##  [61] mclust_5.3                  km.ci_0.5-2                
##  [63] htmlwidgets_0.9             httr_1.3.1                 
##  [65] RColorBrewer_1.1-2          fpc_2.1-10                 
##  [67] modeltools_0.2-21           pkgconfig_2.0.1            
##  [69] XML_3.98-1.9                R.methodsS3_1.7.1          
##  [71] flexmix_2.3-14              nnet_7.3-12                
##  [73] locfit_1.5-9.1              labeling_0.3               
##  [75] rlang_0.1.2                 reshape2_1.4.2             
##  [77] AnnotationDbi_1.38.2        munsell_0.4.3              
##  [79] tools_3.4.1                 downloader_0.4             
##  [81] RSQLite_2.0                 devtools_1.13.3            
##  [83] broom_0.4.2                 evaluate_0.10.1            
##  [85] stringr_1.2.0               yaml_2.1.14                
##  [87] knitr_1.17                  bit64_0.9-7                
##  [89] robustbase_0.92-7           survMisc_0.5.4             
##  [91] purrr_0.2.3                 dendextend_1.5.2           
##  [93] EDASeq_2.10.0               nlme_3.1-131               
##  [95] whisker_0.3-2               R.oo_1.21.0                
##  [97] xml2_1.1.1                  biomaRt_2.32.1             
##  [99] BiocStyle_2.4.1             compiler_3.4.1             
## [101] curl_2.8.1                  testthat_1.0.2             
## [103] tibble_1.3.4                geneplotter_1.54.0         
## [105] stringi_1.1.5               highr_0.6                  
## [107] GenomicFeatures_1.28.4      lattice_0.20-35            
## [109] trimcluster_0.1-2           Matrix_1.2-11              
## [111] commonmark_1.4              psych_1.7.8                
## [113] KMsurv_0.1-5                GlobalOptions_0.0.12       
## [115] data.table_1.10.4           bitops_1.0-6               
## [117] rtracklayer_1.36.4          R6_2.2.2                   
## [119] latticeExtra_0.6-28         hwriter_1.3.2              
## [121] ShortRead_1.34.1            gridExtra_2.3              
## [123] codetools_0.2-15            MASS_7.3-47                
## [125] assertthat_0.2.0            rprojroot_1.2              
## [127] rjson_0.2.15                withr_2.0.0                
## [129] GenomicAlignments_1.12.2    Rsamtools_1.28.0           
## [131] mnormt_1.5-5                GenomeInfoDbData_0.99.0    
## [133] diptest_0.75-7              hms_0.3                    
## [135] tidyr_0.7.1                 class_7.3-14               
## [137] rmarkdown_1.6               ggpubr_0.1.5

References

Cancer Genome Atlas Research Network and others. 2012a. “Comprehensive Genomic Characterization of Squamous Cell Lung Cancers.” doi:10.1038/nature11404.

———. 2012b. “Comprehensive Molecular Characterization of Human Colon and Rectal Cancer.” doi:10.1038/nature11252.

———. 2012c. “Comprehensive Molecular Portraits of Human Breast Tumours.” doi:10.1038/nature11412.

———. 2013a. “Comprehensive Molecular Characterization of Clear Cell Renal Cell Carcinoma.” doi:10.1038/nature12222.

———. 2013b. “Integrated Genomic Characterization of Endometrial Carcinoma.” doi:10.1038/nature12113.

———. 2014a. “Comprehensive Molecular Characterization of Gastric Adenocarcinoma.” doi:10.1038/nature13480.

———. 2014b. “Comprehensive Molecular Profiling of Lung Adenocarcinoma.” doi:10.1038/nature13385.

———. 2014c. “Integrated Genomic Characterization of Papillary Thyroid Carcinoma.” doi:10.1016/j.cell.2014.09.050.

———. 2015a. “Comprehensive Genomic Characterization of Head and Neck Squamous Cell Carcinomas.” doi:10.1038/nature14129.

———. 2015b. “Genomic Classification of Cutaneous Melanoma.” doi:10.1016/j.cell.2015.05.044.

———. 2015c. “The Molecular Taxonomy of Primary Prostate Cancer.” doi:10.1016/j.cell.2015.10.025.

———. 2016. “Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma.” doi:10.1016/j.ccell.2016.04.002.

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. 2016. “Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma.” doi:10.1016/j.cell.2015.12.028.

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. 2016. “TCGAbiolinks: An R/Bioconductor Package for Integrative Analysis of TCGA Data.” doi:10.1093/nar/gkv1507.

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. 2014. “The Somatic Genomic Landscape of Chromophobe Renal Cell Carcinoma.” doi:10.1016/j.ccr.2014.07.014.

Fishbein, Lauren and Leshchiner, Ignaty and Walter, Vonn and Danilova, Ludmila and Robertson, A Gordon and Johnson, Amy R and Lichtenberg, Tara M and Murray, Bradley A and Ghayee, Hans K and Else, Tobias and others. 2017. “Comprehensive Molecular Characterization of Pheochromocytoma and Paraganglioma.” doi:10.1016/j.ccell.2017.01.001.

Gu, Zuguang and Eils, Roland and Schlesner, Matthias. 2016. “Complex Heatmaps Reveal Patterns and Correlations in Multidimensional Genomic Data.” doi:10.1016/j.ccell.2016.04.002.

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. 2015. “Orchestrating High-Throughput Genomic Analysis with Bioconductor.”

James H Bullard, Elizabeth Purdom, Kasper D Hansen and Sandrine Dudoit. 2010. “Evaluation of Statistical Methods for Normalization and Differential Expression in MRNA-Seq Experiments.”

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. 2016. “Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma.” doi:10.1056/NEJMoa1505917.

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. 2010. “Identification of a CpG Island Methylator Phenotype That Defines a Distinct Subgroup of Glioma.”

Risso, D., Schwartz, K., Sherlock, G., & Dudoit, S. 2011. “GC-Content Normalization for RNA-Seq Data.”

Silva, TC and Colaprico, A and Olsen, C and D’Angelo, F and Bontempi, G and Ceccarelli, M and Noushmehr, H. 2016. “TCGA Workflow: Analyze Cancer Genomics and Epigenomics Data Using Bioconductor Packages.” doi:10.12688/f1000research.8923.1.