دورية أكاديمية

Hydra: A mixture modeling framework for subtyping pediatric cancer cohorts using multimodal gene expression signatures.

التفاصيل البيبلوغرافية
العنوان: Hydra: A mixture modeling framework for subtyping pediatric cancer cohorts using multimodal gene expression signatures.
المؤلفون: Pfeil, Jacob, Sanders, Lauren M., Anastopoulos, Ioannis, Lyle, A. Geoffrey, Weinstein, Alana S., Xue, Yuanqing, Blair, Andrew, Beale, Holly C., Lee, Alex, Leung, Stanley G., Dinh, Phuong T., Shah, Avanthi Tayi, Breese, Marcus R., Devine, W. Patrick, Bjork, Isabel, Salama, Sofie R., Sweet-Cordero, E. Alejandro, Haussler, David, Vaske, Olena Morozova
المصدر: PLoS Computational Biology; 4/10/2020, Vol. 16 Issue 4, p1-25, 25p, 2 Diagrams, 5 Graphs
مصطلحات موضوعية: CHILDHOOD cancer, GENE expression, EWING'S sarcoma, CELL tumors, SYNOVIOMA, PROGRAMMED cell death 1 receptors, TUMOR suppressor genes
مستخلص: Precision oncology has primarily relied on coding mutations as biomarkers of response to therapies. While transcriptome analysis can provide valuable information, incorporation into workflows has been difficult. For example, the relative rather than absolute gene expression level needs to be considered, requiring differential expression analysis across samples. However, expression programs related to the cell-of-origin and tumor microenvironment effects confound the search for cancer-specific expression changes. To address these challenges, we developed an unsupervised clustering approach for discovering differential pathway expression within cancer cohorts using gene expression measurements. The hydra approach uses a Dirichlet process mixture model to automatically detect multimodally distributed genes and expression signatures without the need for matched normal tissue. We demonstrate that the hydra approach is more sensitive than widely-used gene set enrichment approaches for detecting multimodal expression signatures. Application of the hydra analysis framework to small blue round cell tumors (including rhabdomyosarcoma, synovial sarcoma, neuroblastoma, Ewing sarcoma, and osteosarcoma) identified expression signatures associated with changes in the tumor microenvironment. The hydra approach also identified an association between ATRX deletions and elevated immune marker expression in high-risk neuroblastoma. Notably, hydra analysis of all small blue round cell tumors revealed similar subtypes, characterized by changes to infiltrating immune and stromal expression signatures. Author summary: Pediatric cancers generally have few somatic mutations. To increase the number of actionable treatment leads, precision pediatric oncology initiatives also analyze tumor gene expression patterns. However, currently available approaches for gene expression data analysis in the clinical setting often use arbitrary thresholds for assessing overexpression and assume gene expression is normally distributed. These methods also rely on reference distributions of related cancer types or normal samples for assessing expression distributions. Often adequate normal samples are not available, and comparing matched cancer cohorts without accounting for subtype expression overestimates the uncertainty in the analysis. We developed a computational framework to automatically detect multimodal expression distributions within well-defined disease populations. Our analysis of small blue round cell tumors (including rhabdomyosarcoma, synovial sarcoma, neuroblastoma, Ewing sarcoma and osteosarcoma) discovered a significant number of multimodally expressed genes. Multimodally expressed genes were associated with proliferative signaling, extracellular matrix organization, and immune signaling pathways across cancer types. Expression signatures correlated with differences in patient outcomes for MYCN non-amplified neuroblastoma, osteosarcoma, and synovial sarcoma. The low mutation rate in pediatric cancers has led some to suggest that pediatric cancers are less immunogenic. However, our analysis suggests that immune infiltration can be identified across small blue round cell tumors. Thus, further research into modulating immune cells for patient benefit may be warranted. [ABSTRACT FROM AUTHOR]
Copyright of PLoS Computational Biology is the property of Public Library of Science and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
قاعدة البيانات: Complementary Index
الوصف
تدمد:1553734X
DOI:10.1371/journal.pcbi.1007753