American Association for Cancer Research
Browse
can-21-0482_supplementary_tables_suppst1-st11.pdf (1.77 MB)

Supplementary Tables from Predicting Molecular Phenotypes from Histopathology Images: A Transcriptome-Wide Expression–Morphology Analysis in Breast Cancer

Download (1.77 MB)
journal contribution
posted on 2023-03-30, 14:20 authored by Yinxi Wang, Kimmo Kartasalo, Philippe Weitz, Balázs Ács, Masi Valkonen, Christer Larsson, Pekka Ruusuvuori, Johan Hartman, Mattias Rantalainen

Online-only supplementary tables: Table S1. Clinicopathological characteristics of patients per cohort. Table S2. ST gene panel. Table S3. The list of features used in the neural network based classification. Table S4. Results of pre-study hyperparameter tunning on 5 genes. Table S5. Prediction performance for the 1011 genes on validation and test sets. Table S6. Prediction performance for transcripts from established biomarker panels. Table S7. Correlation of gene expression with ER status (internal test set). Table S8. Correlation of gene expression with ER status (external test set). Table S9. Enriched pathways for genes cannot be predicted (Reactome gene sets). Table S10. Enriched pathways for genes cannot be predicted (Hallmark gene sets). Table S11. List of proliferation genes.

Funding

Cancer Foundation Finland

CSC - IT Center for Science

Tampere University graduate school

Vetenskapsrådet (VR)

Cancerfonden (Swedish Cancer Society)

Swedish e-science research centre (SeRC)

MedTechLabs

ERA PerMed

Karolinska Institutet (Cancer Research KI; StratCan)

Stockholm Region

Stockholm Cancer Society

Swedish Breast Cancer Association

Academy of Finland (Suomen Akatemia)

Academy of Finland Center of Excellence programme

History

ARTICLE ABSTRACT

Molecular profiling is central in cancer precision medicine but remains costly and is based on tumor average profiles. Morphologic patterns observable in histopathology sections from tumors are determined by the underlying molecular phenotype and therefore have the potential to be exploited for prediction of molecular phenotypes. We report here the first transcriptome-wide expression–morphology (EMO) analysis in breast cancer, where individual deep convolutional neural networks were optimized and validated for prediction of mRNA expression in 17,695 genes from hematoxylin and eosin–stained whole slide images. Predicted expressions in 9,334 (52.75%) genes were significantly associated with RNA sequencing estimates. We also demonstrated successful prediction of an mRNA-based proliferation score with established clinical value. The results were validated in independent internal and external test datasets. Predicted spatial intratumor variabilities in expression were validated through spatial transcriptomics profiling. These results suggest that EMO provides a cost-efficient and scalable approach to predict both tumor average and intratumor spatial expression from histopathology images. Transcriptome-wide expression morphology deep learning analysis enables prediction of mRNA expression and proliferation markers from routine histopathology whole slide images in breast cancer.

Usage metrics

    Cancer Research

    Categories

    Keywords

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC