Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 173(2): 321-337.e10, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625050

ABSTRACT

Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFß signaling, p53 and ß-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.


Subject(s)
Databases, Genetic , Neoplasms/pathology , Signal Transduction/genetics , Genes, Neoplasm , Humans , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
2.
Bioinformatics ; 33(23): 3799-3801, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28961932

ABSTRACT

SUMMARY: MIRMMR predicts microsatellite instability status in cancer samples using methylation and mutation information, in contrast to existing methods that rely on observed microsatellites. Additionally, MIRMMR highlights those genetic alterations contributing to microsatellite instability. AVAILABILITY AND IMPLEMENTATION: Source code is freely available at https://github.com/ding-lab/MIRMMR under the MIT license, implemented in R and supported on Unix/OS X operating systems. CONTACT: smfoltz@wustl.edu or lding@wustl.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
DNA Methylation , Genomics/methods , Microsatellite Instability , Mutation , Neoplasms/genetics , Software , Humans , Neoplasms/metabolism
3.
Commun Biol ; 6(1): 222, 2023 02 25.
Article in English | MEDLINE | ID: mdl-36841852

ABSTRACT

Large compendia of gene expression data have proven valuable for the discovery of novel biological relationships. Historically, most available RNA assays were run on microarray, while RNA-seq is now the platform of choice for many new experiments. The data structure and distributions between the platforms differ, making it challenging to combine them directly. Here we perform supervised and unsupervised machine learning evaluations to assess which existing normalization methods are best suited for combining microarray and RNA-seq data. We find that quantile and Training Distribution Matching normalization allow for supervised and unsupervised model training on microarray and RNA-seq data simultaneously. Nonparanormal normalization and z-scores are also appropriate for some applications, including pathway analysis with Pathway-Level Information Extractor (PLIER). We demonstrate that it is possible to perform effective cross-platform normalization using existing methods to combine microarray and RNA-seq data for machine learning applications.


Subject(s)
Gene Expression Profiling , Machine Learning , RNA-Seq , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Microarray Analysis
4.
Cancer Cell ; 41(9): 1567-1585.e7, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37582362

ABSTRACT

DNA methylation plays a critical role in establishing and maintaining cellular identity. However, it is frequently dysregulated during tumor development and is closely intertwined with other genetic alterations. Here, we leveraged multi-omic profiling of 687 tumors and matched non-involved adjacent tissues from the kidney, brain, pancreas, lung, head and neck, and endometrium to identify aberrant methylation associated with RNA and protein abundance changes and build a Pan-Cancer catalog. We uncovered lineage-specific epigenetic drivers including hypomethylated FGFR2 in endometrial cancer. We showed that hypermethylated STAT5A is associated with pervasive regulon downregulation and immune cell depletion, suggesting that epigenetic regulation of STAT5A expression constitutes a molecular switch for immunosuppression in squamous tumors. We further demonstrated that methylation subtype-enrichment information can explain cell-of-origin, intra-tumor heterogeneity, and tumor phenotypes. Overall, we identified cis-acting DNA methylation events that drive transcriptional and translational changes, shedding light on the tumor's epigenetic landscape and the role of its cell-of-origin.


Subject(s)
DNA Methylation , Endometrial Neoplasms , Female , Humans , Epigenesis, Genetic , Multiomics , Gene Expression Regulation, Neoplastic , Endometrial Neoplasms/genetics
5.
Cancer Res ; 83(8): 1214-1233, 2023 04 14.
Article in English | MEDLINE | ID: mdl-36779841

ABSTRACT

Multiple myeloma (MM) is a highly refractory hematologic cancer. Targeted immunotherapy has shown promise in MM but remains hindered by the challenge of identifying specific yet broadly representative tumor markers. We analyzed 53 bone marrow (BM) aspirates from 41 MM patients using an unbiased, high-throughput pipeline for therapeutic target discovery via single-cell transcriptomic profiling, yielding 38 MM marker genes encoding cell-surface proteins and 15 encoding intracellular proteins. Of these, 20 candidate genes were highlighted that are not yet under clinical study, 11 of which were previously uncharacterized as therapeutic targets. The findings were cross-validated using bulk RNA sequencing, flow cytometry, and proteomic mass spectrometry of MM cell lines and patient BM, demonstrating high overall concordance across data types. Independent discovery using bulk RNA sequencing reiterated top candidates, further affirming the ability of single-cell transcriptomics to accurately capture marker expression despite limitations in sample size or sequencing depth. Target dynamics and heterogeneity were further examined using both transcriptomic and immuno-imaging methods. In summary, this study presents a robust and broadly applicable strategy for identifying tumor markers to better inform the development of targeted cancer therapy. SIGNIFICANCE: Single-cell transcriptomic profiling and multiomic cross-validation to uncover therapeutic targets identifies 38 myeloma marker genes, including 11 transcribing surface proteins with previously uncharacterized potential for targeted antitumor therapy.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiomics , Proteomics , Biomarkers, Tumor/genetics , Gene Expression Profiling/methods
6.
Cell Genom ; 3(7): 100340, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37492101

ABSTRACT

Pediatric brain and spinal cancers are collectively the leading disease-related cause of death in children; thus, we urgently need curative therapeutic strategies for these tumors. To accelerate such discoveries, the Children's Brain Tumor Network (CBTN) and Pacific Pediatric Neuro-Oncology Consortium (PNOC) created a systematic process for tumor biobanking, model generation, and sequencing with immediate access to harmonized data. We leverage these data to establish OpenPBTA, an open collaborative project with over 40 scalable analysis modules that genomically characterize 1,074 pediatric brain tumors. Transcriptomic classification reveals universal TP53 dysregulation in mismatch repair-deficient hypermutant high-grade gliomas and TP53 loss as a significant marker for poor overall survival in ependymomas and H3 K28-mutant diffuse midline gliomas. Already being actively applied to other pediatric cancers and PNOC molecular tumor board decision-making, OpenPBTA is an invaluable resource to the pediatric oncology community.

7.
Cancer Cell ; 41(8): 1397-1406, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37582339

ABSTRACT

The National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (CPTAC) investigates tumors from a proteogenomic perspective, creating rich multi-omics datasets connecting genomic aberrations to cancer phenotypes. To facilitate pan-cancer investigations, we have generated harmonized genomic, transcriptomic, proteomic, and clinical data for >1000 tumors in 10 cohorts to create a cohesive and powerful dataset for scientific discovery. We outline efforts by the CPTAC pan-cancer working group in data harmonization, data dissemination, and computational resources for aiding biological discoveries. We also discuss challenges for multi-omics data integration and analysis, specifically the unique challenges of working with both nucleotide sequencing and mass spectrometry proteomics data.


Subject(s)
Neoplasms , Proteogenomics , Humans , Proteomics , Genomics , Neoplasms/genetics , Gene Expression Profiling
8.
Nat Commun ; 12(1): 2559, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963182

ABSTRACT

Multiple myeloma (MM) is characterized by the uncontrolled proliferation of plasma cells. Despite recent treatment advances, it is still incurable as disease progression is not fully understood. To investigate MM and its immune environment, we apply single cell RNA and linked-read whole genome sequencing to profile 29 longitudinal samples at different disease stages from 14 patients. Here, we collect 17,267 plasma cells and 57,719 immune cells, discovering patient-specific plasma cell profiles and immune cell expression changes. Patients with the same genetic alterations tend to have both plasma cells and immune cells clustered together. By integrating bulk genomics and single cell mapping, we track plasma cell subpopulations across disease stages and find three patterns: stability (from precancer to diagnosis), and gain or loss (from diagnosis to relapse). In multiple patients, we detect "B cell-featured" plasma cell subpopulations that cluster closely with B cells, implicating their cell of origin. We validate AP-1 complex differential expression (JUN and FOS) in plasma cell subpopulations using CyTOF-based protein assays, and integrated analysis of single-cell RNA and CyTOF data reveals AP-1 downstream targets (IL6 and IL1B) potentially leading to inflammation regulation. Our work represents a longitudinal investigation for tumor and microenvironment during MM progression and paves the way for expanding treatment options.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic/genetics , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Neoplasm Recurrence, Local/genetics , Tumor Microenvironment/immunology , Aged , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Lineage , Clonal Evolution/genetics , Cohort Studies , Disease Progression , Female , Gene Expression Regulation, Neoplastic/immunology , Haplotypes , Humans , Interleukin-1beta/blood , Interleukin-6/blood , Male , Mass Spectrometry , Middle Aged , Multigene Family , Multiple Myeloma/blood , Multiple Myeloma/pathology , Mutation , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/immunology , Proto-Oncogene Proteins c-fos/blood , Proto-Oncogene Proteins c-jun/blood , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology , Single-Cell Analysis
9.
Nat Commun ; 11(1): 2666, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471990

ABSTRACT

Multiple myeloma is a plasma cell blood cancer with frequent chromosomal translocations leading to gene fusions. To determine the clinical relevance of fusion events, we detect gene fusions from a cohort of 742 patients from the Multiple Myeloma Research Foundation CoMMpass Study. Patients with multiple clinic visits enable us to track tumor and fusion evolution, and cases with matching peripheral blood and bone marrow samples allow us to evaluate the concordance of fusion calls in patients with high tumor burden. We examine the joint upregulation of WHSC1 and FGFR3 in samples with t(4;14)-related fusions, and we illustrate a method for detecting fusions from single cell RNA-seq. We report fusions at MYC and a neighboring gene, PVT1, which are related to MYC translocations and associated with divergent progression-free survival patterns. Finally, we find that 4% of patients may be eligible for targeted fusion therapies, including three with an NTRK1 fusion.


Subject(s)
Gene Fusion/genetics , Histone-Lysine N-Methyltransferase/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-myc/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Repressor Proteins/genetics , Adult , Aged , Aged, 80 and over , DNA Copy Number Variations/genetics , Gene Expression Profiling/methods , Histone-Lysine N-Methyltransferase/biosynthesis , Humans , Immunoglobulins/genetics , Middle Aged , Progression-Free Survival , RNA, Long Noncoding/genetics , RNA-Seq/methods , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Receptor, trkA/genetics , Repressor Proteins/biosynthesis
10.
Cell Rep ; 23(1): 227-238.e3, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617662

ABSTRACT

Gene fusions represent an important class of somatic alterations in cancer. We systematically investigated fusions in 9,624 tumors across 33 cancer types using multiple fusion calling tools. We identified a total of 25,664 fusions, with a 63% validation rate. Integration of gene expression, copy number, and fusion annotation data revealed that fusions involving oncogenes tend to exhibit increased expression, whereas fusions involving tumor suppressors have the opposite effect. For fusions involving kinases, we found 1,275 with an intact kinase domain, the proportion of which varied significantly across cancer types. Our study suggests that fusions drive the development of 16.5% of cancer cases and function as the sole driver in more than 1% of them. Finally, we identified druggable fusions involving genes such as TMPRSS2, RET, FGFR3, ALK, and ESR1 in 6.0% of cases, and we predicted immunogenic peptides, suggesting that fusions may provide leads for targeted drug and immune therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Neoplasms/genetics , Oncogene Fusion , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Humans , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/pathology , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
11.
PLoS One ; 10(6): e0131578, 2015.
Article in English | MEDLINE | ID: mdl-26110832

ABSTRACT

CONTEXT: Financial and demographic pressures in US require an understanding of the most efficient distribution of physicians to maximize population-level health benefits. Prior work has assumed a constant negative relationship between physician supply and mortality outcomes throughout the US and has not addressed regional variation. METHODS: In this ecological analysis, geographically weighted regression was used to identify spatially varying relationships between local urologist density and prostate cancer mortality at the county level. Data from 1,492 counties in 30 eastern and southern states from 2006-2010 were analyzed. FINDINGS: The ordinary least squares (OLS) regression found that, on average, increasing urologist density by 1 urologist per 100,000 people resulted in an expected decrease in prostate cancer mortality of -0.499 deaths per 100,000 men (95% CI -0.709 to -0.289, p-value < 0.001), or a 1.5% decrease. Geographic weighted regression demonstrated that the addition of one urologist per 100,000 people in counties in the southern Mississippi River states of Arkansas, Mississippi, and Louisiana, as well as parts of Illinois, Indiana, and Wisconsin is associated with decrease of 0.411 to 0.916 in prostate cancer mortality per 100,000 men (1.6-3.6%). In contrast, the urologist density was not significantly associated with the prostate state mortality in the new England region. CONCLUSIONS: The strength of association between urologist density and prostate cancer mortality varied regionally. Those areas with the highest potential for effects could be targeted for increasing the supply of urologists, as it associated with the largest predicted improvement in prostate cancer mortality.


Subject(s)
Prostatic Neoplasms/mortality , Urology , Adult , Aged , Data Collection , Demography , Geography , Health Services Accessibility , Humans , Least-Squares Analysis , Male , Middle Aged , Multivariate Analysis , Physicians/supply & distribution , Regression Analysis , United States/epidemiology , Workforce
SELECTION OF CITATIONS
SEARCH DETAIL