Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
multi-omics data: the input data of the analysis, including miRNA, gene expression data, DNA methylation data, and survival outcome data. All the data were downloaded from TCGA.code: 1. data preprocessing. 2. clustering patients in each omics layer and performing Kaplan-Meier survival analysis to determine the association between patient clusters and survival outcomes. 3. differential expression analysis to identify features that are associated with patients with consistent survival outcomes.
https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/
At the time of our study, 108 cases with breast MRI data were available in the The Cancer Genome Atlas Breast Invasive Carcinoma Collection (TCGA-BRCA) collection. In order to minimize variations in image quality across the multi-institutional cases we included only breast MRI studies acquired on GE 1.5 Tesla magnet strength scanners (GE Medical Systems, Milwaukee, Wisconsin, USA) scanners, yielding a total of 93 cases. We then excluded cases that had missing images in the dynamic sequence (1 patient), or at the time did not have gene expression analysis available in the TCGA Data Portal (8 patients). After these criteria, a dataset of 84 breast cancer patients resulted, with MRIs from four institutions: Memorial Sloan Kettering Cancer Center, the Mayo Clinic, the University of Pittsburgh Medical Center, and the Roswell Park Cancer Institute. The resulting cases contributed by each institution were 9 (date range 1999-2002), 5 (1999-2003), 46 (1999-2004), and 24 (1999-2002), respectively. The dataset of biopsy proven invasive breast cancers included 74 (88%) ductal, 8 (10%) lobular, and 2 (2%) mixed. Of these, 73 (87%) were ER+, 67 (80%) were PR+, and 19 (23%) were HER2+. Various types of analyses were conducted using the combined imaging, genomic, and clinical data. Those analyses are described within several manuscripts created by the group (cited below). Additional information about the methodology for how the Radiologist Annotations file can be found on the TCGA Breast Image Feature Scoring Project page.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
TCGA RNA-seq V2 Level3 data were downloaded from TCGA Genomic Data Commons Data Portal (https://gdc-portal.nci.nih.gov), consisting of 11,303 samples in 34 cancer projects (33 cancer types). Nine cancer types that do not have corresponding non-tumour samples were filtered out, and the analysis was focused on tumour versus non-tumour comparison. 24 cancer types were used in this meta-analysis: BLCA, BRCA, CESC, CHOL, COAD, ESCA, GBM, HNSC, KICH, KIRC, KIRP, LIHC, LUAD, LUSC, PAAD, PCPG, PRAD, READ, SARC, SKCM, STAD, THCA, THYM, UCEC (https://gdc-portal.nci.nih.gov). The nine filtered cancer types were ACC, DLBC, LAML, LGG, MESO, OV, TGCT, UCS and UVM. To extract expression values from TCGA RNA-seq data, we used genomic coordinates to retrieve UCSC Transcript IDs that correspond to the identifiers in TCGA RNA-seq V2 Level3 data (isoform level). The GAF (General Annotation Format) file was used to map the coordinate to UCSC Transcript ID, and it was downloaded form https://tcga-data.nci.nih.gov/docs/GAF/GAF.hg19.June2011.bundle/outputs/TCGA.hg19.June2011.gaf. This file contains genomic annotations shared by all TCGA projects. More details of the GAF file format can be found at https://tcga-data.nci.nih.gov/docs/GAF/GAF3.0/GAF_v3_file_description.docx. We filtered out any coding exons overlapping UCSC Transcript IDs to eliminate expression value of coding genes and evaluate lncRNA expression.We could find the expression values of 443 pcRNAs and 203 tapRNAs in TCGA data, as many of non-coding regions are not yet fully annotated in the TCGA RNA-seq V2 Level3 data. The expression value of pcRNAs and tapRNAs were extracted and clustered by un-supervised Pearson correlation method (Supplementary Figure 18A). The expression values of tapRNA-associated coding genes were also extracted and used to generate the heat-map (Supplementary Figure 18B), which shows the similar pattern of expression with tapRNAs across the cancer types.To show that tapRNAs and associated coding genes have similar expression profiles in cancers we generated a Spearman's Rank-Order Correlation heatmap (Figure 6A) between tapRNAs and their associated coding genes based on the TCGA RNA-seq data. We used the MatLab function corr to calculate the Spearman's rho. This function takes two matrices X (197-by-8,850 expression profiling matrix of tapRNA) and Y (197-by-8,850 expression profiling matrix of tapRNA-assocated coding gene) and returns an 8,850-by-8,850 matrix containing the pairwise correlation coefficient between each pair of 8,850 columns (TCGA cancer samples in Supplementary Figure 18A and B). Thus, the rank-order correlation matrix that we computed from the matrices of expression profiling data (Supplementary Figure S18A and B) allowed us to compare the correlation between two column vectors i.e. cancer samples. This function also returns a matrix of p-values for testing the hypothesis of no correlation against the alternative that there is a nonzero correlation. Each element of a matrix of p-values is the p value for the corresponding element of Spearman's rho. The p-values for Spearman's rho are calculated using large-sample approximations. To check significance level of correlation between tapRNA and its associated coding gene, the diagonal of the p-value matrix was extracted and used. The median is 1.31x10-11 and the mean is 1.03x10-4 with standard deviation 0.0029.To identify cancer-specific tapRNAs, we considered not only the global expression pattern of a given tapRNA in each cancer type, but also expression pattern of specific sub-group that is significantly distinct, to take into account cancer sample heterogeneity. Thus, two conditions were applied: (1) average expression level of a tapRNA in a given cancer type is in top 10% or bottom 10% and (2) a tapRNA has at least 10% of samples in a given cancer type that are significantly up-regulated (Z-score > 2) or down-regulated (Z-score < -2).
This dataset contains summary data visualizations and clinical data from a broad sampling of 442 samples of skin cutaneous melanomas from 488 patients. The data was gathered as part of the PanCancer Atlas initiative, which aims to answer big, overarching questions about cancer by examining the full set of tumors characterized in the robust TCGA dataset. The clinical data includes mutation count, information about mutated genes, patient demographics, disease status, tumor typing, and chromosomal gain or loss. The data set also includes copy-number segment data downloadable as .seg files and viewable via the Integrative Genomics Viewer.
https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/
The Cancer Genome Atlas Ovarian Cancer (TCGA-OV) data collection is part of a larger effort to build a research community focused on connecting cancer phenotypes to genotypes by providing clinical images matched to subjects from The Cancer Genome Atlas (TCGA). Clinical, genetic, and pathological data resides in the Genomic Data Commons (GDC) Data Portal while the radiological data is stored on The Cancer Imaging Archive (TCIA).
Matched TCGA patient identifiers allow researchers to explore the TCGA/TCIA databases for correlations between tissue genotype, radiological phenotype and patient outcomes. Tissues for TCGA were collected from many sites all over the world in order to reach their accrual targets, usually around 500 specimens per cancer type. For this reason the image data sets are also extremely heterogeneous in terms of scanner modalities, manufacturers and acquisition protocols. In most cases the images were acquired as part of routine care and not as part of a controlled research study or clinical trial.
Imaging Source Site (ISS) Groups are being populated and governed by participants from institutions that have provided imaging data to the archive for a given cancer type. Modeled after TCGA analysis groups, ISS groups are given the opportunity to publish a marker paper for a given cancer type per the guidelines in the table above. This opportunity will generate increased participation in building these multi-institutional data sets as they become an open community resource. Learn more about the TCGA Ovarian Phenotype Research Group.
CC0 1.0 Universal Public Domain Dedicationhttps://creativecommons.org/publicdomain/zero/1.0/
License information was derived automatically
The following datasets were created for Project Cognoma:expression-matrix.tsv.bz2
is a sample × gene matrix indicating a gene's expression level for a given sample. This dataset will be the feature/x/predictor information for Project Cognoma.expression-genes.tsv
provides information and summary statistics for every gene in expression-matrix.tsv.bz2
.mutation-matrix.tsv.bz2
is a sample × gene matrix indicating whether a gene is mutated for a given sample. Select columns (or unions of several columns) in this dataset will be the status/y/outcome for Project Cognoma.mutation-genes.tsv
provides information and summary statistics for every gene in mutation-matrix.tsv.bz2
.samples.tsv
is a sample × attribute matrix providing sample information and clinical measures for each sample.covariates.tsv
is a sample × attribute matrix for modeling that encodes categorical variables in samples.tsv using dummies.All datasets contain the same samples as rows (in the same order). No two samples correspond to the same patient.The data was retrieved from the UCSC Xena Browser.These datasets were created by the GitHub repository commit below. See the download directory of the cancer-data repository for metadata files with the version info for the Xena downloads this release is based on.See the data/subset directory of the cancer-data repository on GitHub to browse small subsets of the expression and mutation datasets.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
This dataset contains mitosis detections, mitotic network structures, and social network analysis (SNA) measures derived from 11,161 diagnostic slides in The Cancer Genome Atlas (TCGA). Mitoses were automatically identified using the MDFS algorithm [1], and each detected mitosis was converted into a node within a mitotic network. The resulting graphs are provided in JSON format, with each file representing a single diagnostic slide.
Each JSON file contains four primary fields:
edge_index
Two parallel lists representing edges between nodes. The ii-th element in the first list corresponds to the source node index, and the ii-th element in the second list is the target node index.
coordinates
A list of [x, y] positions for each node (mitosis). The (x,y) coordinates can be used for spatial visualization or further spatial analyses.
feats
A list of feature vectors, with each row corresponding to a node. These features include:
feat_names
The names of the features in feats
. The order matches the columns in each node’s feature vector.
{
"edge_index": [[1, 2, 6, 10], [2, 4, 8, 11]],
"coordinates": [[27689.0, 12005.0], [24517.0, 17809.0], ...],
"feats": [[1.0, 0.0, 0.0, 0.0], [1.0, 1.0, 0.0, 0.115], ...],
"feat_names": ["type", "Node_Degree", "Clustering_Coeff", "Harmonic_Cen"]
}
Below is a sample Python snippet to load one JSON file, extract node coordinates and the type
feature, and combine them into a single NumPy array:
import json
import numpy as np
# Path to your JSON file
json_file_path = "example_graph.json"
with open(json_file_path, 'r') as f:
data = json.load(f)
# Convert coordinates to NumPy
coordinates = np.array(data["coordinates"])
# Identify the "type" column
feat_names = data["feat_names"]
type_index = feat_names.index("type")
# Extract features and isolate the "type" column
feats = np.array(data["feats"])
node_types = feats[:, type_index].reshape(-1, 1)
# Combine x, y, and type into a single array (N x 3)
combined_data = np.hstack([coordinates, node_types])
print(combined_data)
To visualize or analyze the network structure, you can construct a NetworkX graph as follows:
import json
import networkx as nx
import matplotlib.pyplot as plt
json_file_path = "example_graph.json"
with open(json_file_path, "r") as f:
data = json.load(f)
# Create a NetworkX Graph
G = nx.Graph()
# Add each node with position attributes
for i, (x, y) in enumerate(data["coordinates"]):
G.add_node(i, pos=(x, y))
# Add edges using the parallel lists in edge_index
# (Adjust for 1-based indexing if necessary)
for src, dst in zip(data["edge_index"][0], data["edge_index"][1]):
G.add_edge(src, dst)
Having TIAToolbox installed, one can easily visualize the mitotic network on their respective whole slide images using the following command:
tiatoolbox visualize --slides path/to/slides --overlays path/to/overlays
The only thing to consider is that slides and overlays (provided graph json files) should have the same name. For more information, please refer to Visualization Interface Usage - TIA Toolbox 1.5.1 Documentation.
In case of using this dataset, please cite the following publication:
@article{jahanifar2024mitosis, title={Mitosis detection, fast and slow: robust and efficient detection of mitotic figures}, author={Jahanifar, Mostafa and Shephard, Adam and Zamanitajeddin, Neda and Graham, Simon and Raza, Shan E Ahmed and Minhas, Fayyaz and Rajpoot, Nasir}, journal={Medical Image Analysis}, volume={94}, pages={103132}, year={2024}, publisher={Elsevier} }
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Abstract:
The Cancer Genome Atlas (TCGA) was a large-scale collaborative project initiated by the National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI). It aimed to comprehensively characterize the genomic and molecular landscape of various cancer types. This dataset includes curated survival data from the Pan-cancer Atlas paper titled "An Integrated TCGA Pan-Cancer Clinical Data Resource (TCGA-CDR) to drive high quality survival outcome analytics". The paper highlights four types of carefully curated survival endpoints, and recommends the use of the endpoints of OS, PFI, DFI, and DSS for each TCGA cancer type. The dataset also includes phenotypic information about GBM. The Sample IDs are unique identifiers, which can be paired with the gene expression dataset.
Inspiration:
This dataset was uploaded to UBRITE for GTKB project.
Instruction:
The survival and phenotype data were merged into one file. Empty columns were removed. Columns with the same value for every sample were also removed.
Acknowledgments:
Goldman, M.J., Craft, B., Hastie, M. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol (2020). https://doi.org/10.1038/s41587-020-0546-8
Liu, Jianfang, Caesar-Johnson, Samantha J. et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell, Volume 173, Issue 2, 400 - 416.e11. https://doi.org/10.1016/j.cell.2018.02.052
The Cancer Genome Atlas Research Network., Weinstein, J., Collisson, E. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113–1120 (2013). https://doi.org/10.1038/ng.2764
U-BRITE last update: 07/13/2023
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Figure S1 Functional analysis based on the DEGs between the two-risk groups in the TCGA-SKCM cohort. (A) The vocalno plot shows the differential expression genes between high risk and low-risk groups. Red, upregulated in high-risk group; blue, upregulated in low-risk group; Grey, no significant change. Bubble graph for GO enrichment (B) and KEGG pathways (C) (the bigger bubble means the more genes enriched, and the increasing depth of red means the differences were more obvious; q-value: the adjusted p-value). (D) Barplot shows the differences in enrichment in the cancer hallmark pathways between high-risk and low-risk groups in TCGA SKCM dataset. TCGA: The Cancer Genome Atlas; SKCM: Skin cutaneous melanoma; DEGs: Differentially expressed genes; KEGG: Kyoto Encyclopedia of Genes and Genomes
https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/https://www.cancerimagingarchive.net/data-usage-policies-and-restrictions/
The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) data collection is part of a larger effort to build a research community focused on connecting cancer phenotypes to genotypes by providing clinical images matched to subjects from The Cancer Genome Atlas (TCGA). Clinical, genetic, and pathological data resides in the Genomic Data Commons (GDC) Data Portal while the radiological data is stored on The Cancer Imaging Archive (TCIA).
Matched TCGA patient identifiers allow researchers to explore the TCGA/TCIA databases for correlations between tissue genotype, radiological phenotype and patient outcomes. Tissues for TCGA were collected from many sites all over the world in order to reach their accrual targets, usually around 500 specimens per cancer type. For this reason the image data sets are also extremely heterogeneous in terms of scanner modalities, manufacturers and acquisition protocols. In most cases the images were acquired as part of routine care and not as part of a controlled research study or clinical trial.
Imaging Source Site (ISS) Groups are being populated and governed by participants from institutions that have provided imaging data to the archive for a given cancer type. Modeled after TCGA analysis groups, ISS groups are given the opportunity to publish a marker paper for a given cancer type per the guidelines in the table above. This opportunity will generate increased participation in building these multi-institutional data sets as they become an open community resource. Learn more about the TCGA Breast Phenotype Research Group.
https://entrepot.recherche.data.gouv.fr/api/datasets/:persistentId/versions/3.0/customlicense?persistentId=doi:10.15454/YNMQUYhttps://entrepot.recherche.data.gouv.fr/api/datasets/:persistentId/versions/3.0/customlicense?persistentId=doi:10.15454/YNMQUY
This dataset is issued from the public repository TCGA (https://portal.gdc.cancer.gov/) and contain several files, each corresponding to a given omic on the same individuals with breast cancer. Raw data have been obtained from the mixOmics case study described in http://mixomics.org/mixdiablo/case-study-tcga/ [link accessed on August 18, 2021] and were made available by the package authors at http://mixomics.org/wp-content/uploads/2016/08/TCGA.normalised.mixDIABLO.RData_.zip (R data format). Data in the zip file had been normalised for technical biases by the package authors. Data from the train and test sets were exported as TXT/CSV files and completed with miRNA expression on the smae individuals and toy datasets to handle missing value cases and alike. They serve as a basis for the illustration of the web data analysis tool ASTERICS (Project 20008788 funded by Région Occitanie).
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Abstract:
The Cancer Genome Atlas (TCGA) was a large-scale collaborative project initiated by the National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI). It aimed to comprehensively characterize the genomic and molecular landscape of various cancer types. This dataset includes curated survival data from the Pan-cancer Atlas paper titled "An Integrated TCGA Pan-Cancer Clinical Data Resource (TCGA-CDR) to drive high quality survival outcome analytics". The paper highlights four types of carefully curated survival endpoints, and recommends the use of the endpoints of OS, PFI, DFI, and DSS for each TCGA cancer type. The dataset also includes phenotypic information about KIRC. The Sample IDs are unique identifiers, which can be paired with the gene expression dataset.
Inspiration:
This dataset was uploaded to UBRITE for GTKB project.
Instruction:
The survival and phenotype data were merged into one file. Empty columns were removed. Columns with the same value for every sample were also removed.
Acknowledgments:
Goldman, M.J., Craft, B., Hastie, M. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol (2020). https://doi.org/10.1038/s41587-020-0546-8
Liu, Jianfang, Caesar-Johnson, Samantha J. et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell, Volume 173, Issue 2, 400 - 416.e11. https://doi.org/10.1016/j.cell.2018.02.052
The Cancer Genome Atlas Research Network., Weinstein, J., Collisson, E. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113–1120 (2013). https://doi.org/10.1038/ng.2764
U-BRITE last update: 07/13/2023
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Background: While the acquired risk factors of liver cancer in Asia are relatively well understood, the underlying genetic background of liver cancer in Asians has not been well established or correlated with clinical outcomes. Objective: To identify gene mutations linked with worse outcomes in Asian patients with hepatocellular carcinoma (HCC). Methods: A total of 347 Asian and Non-Asian patients with HCC were analyzed in this study. TCGA patient mutation and clinical data were downloaded through TCGAbiolinksGUI and analyzed using the Python NumPy, Matplotlib, seaborn, and SciPy libraries. Statistical significance was determined by Welch’s t-test (unequal variances t-test), with P-values < 0.05 considered to be statistically significant. Results: Mutations in five genes (TP53, TTN, OBSCN, MUC5B, CSMD1) were statistically linked with increased mortality in Asians compared to non-Asians, four of which (TTN, OBSCN, MUC5B, CSMD1) were also more prevalent in the Asian population. Within the Asian cohort, two gene mutations (TTN, HMCN1) were statistically linked with worse outcomes. The TP53 mutation predicts worse outcomes within the non-Asian cohort, but not within the Asian cohort. Conclusions: This study identified multiple genetic biomarkers that can aid in the recognition, surveillance, prognosis, and gene therapy of HCC.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
The subgroup data (BPS-LumA and WPS-LumA) of the 415 TCGA luminal-A breast cancer samples.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Information about the dataset files:
1) pancan_rnaseq_freeze.tsv.gz: Publicly available gene expression data for the TCGA Pan-cancer dataset. File: PanCanAtlas EBPlusPlusAdjustPANCAN_IlluminaHiSeq_RNASeqV2.geneExp.tsv was processed using script process_sample_freeze.py by Gregory Way et al as described in https://github.com/greenelab/pancancer/ data processing and initialization steps. [http://api.gdc.cancer.gov/data/3586c0da-64d0-4b74-a449-5ff4d9136611] [https://doi.org/10.1016/j.celrep.2018.03.046]
2) pancan_mutation_freeze.tsv.gz: Publicly available Mutational information for TCGA Pan-cancer dataset. File: mc3.v0.2.8.PUBLIC.maf.gz was processed using script process_sample_freeze.py by Gregory Way et al as described in https://github.com/greenelab/pancancer/ data processing and initialization steps. [http://api.gdc.cancer.gov/data/1c8cfe5f-e52d-41ba-94da-f15ea1337efc] [https://doi.org/10.1016/j.celrep.2018.03.046]
3) pancan_GISTIC_threshold.tsv.gz: Publicly available Gene- level copy number information of the TCGA Pan-cancer dataset. This file is processed using script process_copynumber.py by Gregory Way et al as described in https://github.com/greenelab/pancancer/ data processing and initialization steps. The files copy_number_loss_status.tsv.gz and copy_number_gain_status.tsv.gz generated from this data are used as inputs in our Galaxy pipeline. [https://xenabrowser.net/datapages/?cohort=TCGA%20Pan-Cancer%20(PANCAN)&removeHub=https%3A%2F%2Fxena.treehouse.gi.ucsc.edu%3A443] [https://doi.org/10.1016/j.celrep.2018.03.046]
4) mutation_burden_freeze.tsv.gz: Publicly available Mutational information for TCGA Pan-cancer dataset mc3.v0.2.8.PUBLIC.maf.gz was processed using script process_sample_freeze.py by Gregory Way et al as described in https://github.com/greenelab/pancancer/ data processing and initialization steps. [https://github.com/greenelab/pancancer/][http://api.gdc.cancer.gov/data/1c8cfe5f-e52d-41ba-94da-f15ea1337efc] [https://doi.org/10.1016/j.celrep.2018.03.046]
5) sample_freeze.tsv or sample_freeze_version4_modify.tsv: The file lists the frozen samples as determined by TCGA PanCancer Atlas consortium along with raw RNAseq and mutation data. These were previously determined and included for all downstream analysis All other datasets were processed and subset according to the frozen samples.[https://github.com/greenelab/pancancer/]
6) vogelstein_cancergenes.tsv: compendium of OG and TSG used for the analysis. [https://github.com/greenelab/pancancer/]
7) CCLE_DepMap_18Q1_maf_20180207.txt.gz Publicly available Mutational data for CCLE cell lines from Broad Institute Cancer Cell Line Encyclopedia (CCLE) / DepMap Portal. [https://depmap.org/portal/download/api/download/external?file_name=ccle%2FCCLE_DepMap_18Q1_maf_20180207.txt]
8) ccle_rnaseq_genes_rpkm_20180929.gct.gz: Publicly available Expression data for 1019 cell lines (RPKM) from Broad Institute Cancer Cell Line Encyclopedia (CCLE) / DepMap Portal. [https://depmap.org/portal/download/api/download/external?file_name=ccle%2Fccle_2019%2FCCLE_RNAseq_genes_rpkm_20180929.gct.gz]
9) CCLE_MUT_CNA_AMP_DEL_binary_Revealer.gct: Publicly available merged Mutational and copy number alterations that include gene amplifications and deletions for the CCLE cell lines. This data is represented in the binary format and provided by the Broad Institute Cancer Cell Line Encyclopedia (CCLE) / DepMap Portal. [https://data.broadinstitute.org/ccle_legacy_data/binary_calls_for_copy_number_and_mutation_data/CCLE_MUT_CNA_AMP_DEL_binary_Revealer.gct]
10) GDSC_cell_lines_EXP_CCLE_names.csv.gz Publicly available RMA normalized expression data for Genomics of Drug Sensitivity in Cancer(GDSC) cell-lines. File gdsc_cell_line_RMA_proc_basalExp.csv was downloaded. This data was subsetted to 389 cell lines that are common among CCLE and GDSC. All the GDSC cell line names were replaced with CCLE cell line names for further processing. [https://www.cancerrxgene.org/gdsc1000/GDSC1000_WebResources//Data/preprocessed/Cell_line_RMA_proc_basalExp.txt.zip]
11) GDSC_CCLE_common_mut_cnv_binary.csv.gz: A subset of merged Mutational and copy number alterations that include gene amplifications and deletions for common cell lines between GDSC and CCLE. This file is generated using CCLE_MUT_CNA_AMP_DEL_binary_Revealer.gct and a list of common cell lines.
12) gdsc1_ccle_pharm_fitted_dose_data.txt.gz: Pharmacological data for GDSC1 cell lines. [ftp://ftp.sanger.ac.uk/pub/project/cancerrxgene/releases/current_release/GDSC1_fitted_dose_response_15Oct19.xlsx]
13) gdsc2_ccle_pharm_fitted_dose_data.txt.gz: Pharmacological data for GDSC2 cell lines. [ftp://ftp.sanger.ac.uk/pub/project/cancerrxgene/releases/current_release/GDSC2_fitted_dose_response_15Oct19.xlsx]
14) compounds.csv: list of pharmacological compounds tested for our analysis
15) tcga_dictonary.tsv: list of cancer types used in the analysis.
16) seg_based_scores.tsv: Measurement of total copy number burden, Percent of genome altered by copy number alterations. This file was used as part of the Pancancer analysis by Gregory Way et al as described in https://github.com/greenelab/pancancer/ data processing and initialization steps. [https://github.com/greenelab/pancancer/]
Abstract: The Cancer Genome Atlas (TCGA) was a large-scale collaborative project initiated by the National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI). It aimed to comprehensively characterize the genomic and molecular landscape of various cancer types. This dataset includes curated survival data from the Pan-cancer Atlas paper titled "An Integrated TCGA Pan-Cancer Clinical Data Resource (TCGA-CDR) to drive high quality survival outcome analytics". The paper highlights four types of carefully curated survival endpoints, and recommends the use of the endpoints of OS, PFI, DFI, and DSS for each TCGA cancer type. The dataset also includes phenotypic information about HNSC. The Sample IDs are unique identifiers, which can be paired with the gene expression dataset. Inspiration: This dataset was uploaded to UBRITE for GTKB project. Instruction: The survival and phenotype data were merged into one file. Empty columns were removed. Columns with the same value for every sample were also removed. Acknowledgments: Goldman, M.J., Craft, B., Hastie, M. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol (2020). https://doi.org/10.1038/s41587-020-0546-8 Liu, Jianfang, Caesar-Johnson, Samantha J. et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell, Volume 173, Issue 2, 400 - 416.e11. https://doi.org/10.1016/j.cell.2018.02.052 The Cancer Genome Atlas Research Network., Weinstein, J., Collisson, E. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113–1120 (2013). https://doi.org/10.1038/ng.2764 U-BRITE last update: 07/13/2023 {"references": ["Goldman, M.J., Craft, B., Hastie, M. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol (2020). https://doi.org/10.1038/s41587-020-0546-8", "Liu, Jianfang, Caesar-Johnson, Samantha J. et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell, Volume 173, Issue 2, 400 - 416.e11.\u00a0https://doi.org/10.1016/j.cell.2018.02.052", "The Cancer Genome Atlas Research Network., Weinstein, J., Collisson, E. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113\u20131120 (2013). https://doi.org/10.1038/ng.2764"]} UBRITE location: /data/project/ubrite/gtkb/TCGA/Clinical
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
Interactions of the extracellular matrix (ECM) and cellular receptors constitute one of the crucial pathways involved in colorectal cancer progression and metastasis. With the use of bioinformatics analysis, we comprehensively evaluated the prognostic information concentrated in the genes from this pathway. First, we constructed a ECM–receptor regulatory network by integrating the transcription factor (TF) and 5’-isomiR interaction databases with mRNA/miRNA-seq data from The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD). Notably, one-third of interactions mediated by 5’-isomiRs was represented by noncanonical isomiRs (isomiRs, whose 5’-end sequence did not match with the canonical miRBase version). Then, exhaustive search-based feature selection was used to fit prognostic signatures composed of nodes from the network for overall survival prediction. Two reliable prognostic signatures were identified and validated on the independent The Cancer Genome Atlas Rectum Adenocarcinoma (TCGA-READ) cohort. The first signature was made up by six genes, directly involved in ECM–receptor interaction: AGRN, DAG1, FN1, ITGA5, THBS3, and TNC (concordance index 0.61, logrank test p = 0.0164, 3-years ROC AUC = 0.68). The second hybrid signature was composed of three regulators: hsa-miR-32-5p, NR1H2, and SNAI1 (concordance index 0.64, logrank test p = 0.0229, 3-years ROC AUC = 0.71). While hsa-miR-32-5p exclusively regulated ECM-related genes (COL1A2 and ITGA5), NR1H2 and SNAI1 also targeted other pathways (adhesion, cell cycle, and cell division). Concordant distributions of the respective risk scores across four stages of colorectal cancer and adjacent normal mucosa additionally confirmed reliability of the models.
TCGA Glioblastoma Multiforme. Source data from GDAC Firehose. Previously known as TCGA Provisional. This dataset contains summary data visualizations and clinical data from a broad sampling of 619 glioblastoma multiformes from 606 patients. The data was gathered as part of the Broad Institute of MIT and Harvard Firehose initiative, a cancer analysis pipeline. The clinical data includes mutation count, information about mutated genes, patient demographics, disease status, tumor typing, chromosomal gain or loss, Adjuvant Postoperative Pharmaceutical Therapy Administered Indicator, Days to Sample Collection, if the patient start adjuvant postoperative radiotherapy, Disease Free (Months), Disease Free Status, and First Pathologic Diagnosis Biospecimen Acquisition Method Type. The dataset includes Next-Generation Clustered Heat Maps (NG-CHM) viewable via an embedded NG-CHM Heat Map Viewer, provided my MD Anderson Cancer Center, which provides a graphical environment for exploration of clustered or non-clustered heat map data. The data set also includes copy-number segment data downloadable as .seg files and viewable via the Integrative Genomics Viewer.
This dataset contains summary data visualizations and clinical data from a broad sampling of over 200 acute myeloid leukemias from 200 patients. The data was gathered as part of the PanCancer Atlas initiative, which aims to answer big, overarching questions about cancer by examining the full set of tumors characterized in the robust TCGA dataset. The clinical data includes mutation count, information about mutated genes, patient demographics, disease status, tumor typing, and chromosomal gain or loss. The data set also includes copy-number segment data downloadable as .seg files and viewable via the Integrative Genomics Viewer.
TCGA Acute Myeloid Leukemia. Source data from GDAC Firehose. Previously known as TCGA Provisional. This dataset contains summary data visualizations and clinical data from a broad sampling of 200 carcinomas from 200 patients. The data was gathered as part of the Broad Institute of MIT and Harvard Firehose initiative, a cancer analysis pipeline. The clinical data includes mutation count, information about mutated genes, patient demographics, sample type, disease code, Abnormal Lymphocyte Percent, Atra Exposure, Basophils Cell Count, Blast Count, Cytogenetic abnormality type, and FAB. The dataset includes Next-Generation Clustered Heat Maps (NG-CHM) viewable via an embedded NG-CHM Heat Map Viewer, provided my MD Anderson Cancer Center, which provides a graphical environment for exploration of clustered or non-clustered heat map data. The data set also includes copy-number segment data downloadable as .seg files and viewable via the Integrative Genomics Viewer.
Attribution 4.0 (CC BY 4.0)https://creativecommons.org/licenses/by/4.0/
License information was derived automatically
multi-omics data: the input data of the analysis, including miRNA, gene expression data, DNA methylation data, and survival outcome data. All the data were downloaded from TCGA.code: 1. data preprocessing. 2. clustering patients in each omics layer and performing Kaplan-Meier survival analysis to determine the association between patient clusters and survival outcomes. 3. differential expression analysis to identify features that are associated with patients with consistent survival outcomes.