Modeling Dense Multimodal Interactions Between Biological Pathways and Histology for Survival Prediction

Read original: arXiv:2304.06819 - Published 4/16/2024 by Guillaume Jaume, Anurag Vaidya, Richard Chen, Drew Williamson, Paul Liang, Faisal Mahmood
Total Score

0

🔮

Sign in to get full access

or

If you already have an account, we'll log you in

Overview

  • This research aims to address two key challenges in integrating whole-slide images (WSIs) and bulk transcriptomics data for predicting patient survival:
  • How to tokenize transcriptomics data in a semantically meaningful and interpretable way?
  • How to effectively capture dense multimodal interactions between WSIs and transcriptomics?

Plain English Explanation

The research paper proposes a novel approach, called SURVPATH, to integrate two types of medical data - whole-slide images (WSIs) and bulk transcriptomics - to better predict patient survival. WSIs provide a detailed spatial description of a tumor, while transcriptomics data gives a global view of gene expression levels within the tumor.

The key ideas are:

  • Meaningful Transcriptomics Tokens: The researchers develop a way to extract "pathway tokens" from the transcriptomics data that encode specific cellular functions, making the data more interpretable.
  • Multimodal Fusion: They propose a memory-efficient multimodal Transformer model that can effectively capture the interactions between the histology patch tokens from the WSIs and the pathway tokens from the transcriptomics data.

By combining these two complementary data sources, the SURVPATH model achieves state-of-the-art performance in predicting patient survival across multiple cancer datasets. Importantly, the model's interpretability framework can provide valuable insights into the complex relationship between tumor genotype and phenotype, potentially leading to a better understanding of the underlying biological mechanisms.

Technical Explanation

The research first addresses the challenge of tokenizing transcriptomics data in a semantically meaningful way. Rather than using individual genes as tokens, the authors propose learning "pathway tokens" that encode specific cellular functions. These pathway tokens, combined with histology patch tokens from the WSIs, form the input to a multimodal Transformer model.

The proposed SURVPATH model uses a memory-efficient Transformer architecture to capture the dense interactions between the two modalities. This allows the model to learn complex relationships between the tumor's spatial morphology (from the WSIs) and its underlying molecular processes (from the transcriptomics data).

The researchers evaluate SURVPATH on five cancer datasets from The Cancer Genome Atlas and show that it outperforms both unimodal and multimodal baseline models in predicting patient survival. Furthermore, the interpretability framework of SURVPATH identifies key multimodal prognostic factors, providing insights into the interplay between tumor genotype and phenotype.

Critical Analysis

The research presents a compelling approach to integrating WSIs and transcriptomics data for improved patient prognosis. The use of pathway tokens to represent the transcriptomics data is a novel and potentially more interpretable approach compared to using individual genes.

However, the paper does not provide a detailed discussion of the limitations of the pathway token extraction process. It would be helpful to understand the potential biases or information loss that may occur during this step and how they may impact the overall model performance.

Additionally, the authors mention that the multimodal Transformer architecture is "memory-efficient," but they do not provide a quantitative comparison of the memory requirements or computational complexity of their approach compared to other multimodal fusion methods. This information would be useful for researchers looking to apply similar techniques in their own work.

Conclusion

The SURVPATH model represents a significant advancement in integrating WSIs and transcriptomics data for patient survival prediction. By learning meaningful pathway tokens from the transcriptomics data and fusing them with histology patch tokens using a memory-efficient Transformer, the researchers have developed a powerful and interpretable multimodal framework.

The findings of this research have the potential to provide valuable insights into the complex interplay between tumor genotype and phenotype, ultimately leading to a better understanding of the underlying biological mechanisms driving cancer progression. As the field of computational pathology continues to evolve, approaches like SURVPATH may become increasingly important for improving patient prognosis and guiding personalized treatment strategies.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Follow @aimodelsfyi on 𝕏 →

Related Papers

🔮

Total Score

0

Modeling Dense Multimodal Interactions Between Biological Pathways and Histology for Survival Prediction

Guillaume Jaume, Anurag Vaidya, Richard Chen, Drew Williamson, Paul Liang, Faisal Mahmood

Integrating whole-slide images (WSIs) and bulk transcriptomics for predicting patient survival can improve our understanding of patient prognosis. However, this multimodal task is particularly challenging due to the different nature of these data: WSIs represent a very high-dimensional spatial description of a tumor, while bulk transcriptomics represent a global description of gene expression levels within that tumor. In this context, our work aims to address two key challenges: (1) how can we tokenize transcriptomics in a semantically meaningful and interpretable way?, and (2) how can we capture dense multimodal interactions between these two modalities? Specifically, we propose to learn biological pathway tokens from transcriptomics that can encode specific cellular functions. Together with histology patch tokens that encode the different morphological patterns in the WSI, we argue that they form appropriate reasoning units for downstream interpretability analyses. We propose fusing both modalities using a memory-efficient multimodal Transformer that can model interactions between pathway and histology patch tokens. Our proposed model, SURVPATH, achieves state-of-the-art performance when evaluated against both unimodal and multimodal baselines on five datasets from The Cancer Genome Atlas. Our interpretability framework identifies key multimodal prognostic factors, and, as such, can provide valuable insights into the interaction between genotype and phenotype, enabling a deeper understanding of the underlying biological mechanisms at play. We make our code public at: https://github.com/ajv012/SurvPath.

Read more

4/16/2024

Multimodal Prototyping for cancer survival prediction
Total Score

0

Multimodal Prototyping for cancer survival prediction

Andrew H. Song, Richard J. Chen, Guillaume Jaume, Anurag J. Vaidya, Alexander S. Baras, Faisal Mahmood

Multimodal survival methods combining gigapixel histology whole-slide images (WSIs) and transcriptomic profiles are particularly promising for patient prognostication and stratification. Current approaches involve tokenizing the WSIs into smaller patches (>10,000 patches) and transcriptomics into gene groups, which are then integrated using a Transformer for predicting outcomes. However, this process generates many tokens, which leads to high memory requirements for computing attention and complicates post-hoc interpretability analyses. Instead, we hypothesize that we can: (1) effectively summarize the morphological content of a WSI by condensing its constituting tokens using morphological prototypes, achieving more than 300x compression; and (2) accurately characterize cellular functions by encoding the transcriptomic profile with biological pathway prototypes, all in an unsupervised fashion. The resulting multimodal tokens are then processed by a fusion network, either with a Transformer or an optimal transport cross-alignment, which now operates with a small and fixed number of tokens without approximations. Extensive evaluation on six cancer types shows that our framework outperforms state-of-the-art methods with much less computation while unlocking new interpretability analyses.

Read more

7/2/2024

Pathology-genomic fusion via biologically informed cross-modality graph learning for survival analysis
Total Score

0

Pathology-genomic fusion via biologically informed cross-modality graph learning for survival analysis

Zeyu Zhang, Yuanshen Zhao, Jingxian Duan, Yaou Liu, Hairong Zheng, Dong Liang, Zhenyu Zhang, Zhi-Cheng Li

The diagnosis and prognosis of cancer are typically based on multi-modal clinical data, including histology images and genomic data, due to the complex pathogenesis and high heterogeneity. Despite the advancements in digital pathology and high-throughput genome sequencing, establishing effective multi-modal fusion models for survival prediction and revealing the potential association between histopathology and transcriptomics remains challenging. In this paper, we propose Pathology-Genome Heterogeneous Graph (PGHG) that integrates whole slide images (WSI) and bulk RNA-Seq expression data with heterogeneous graph neural network for cancer survival analysis. The PGHG consists of biological knowledge-guided representation learning network and pathology-genome heterogeneous graph. The representation learning network utilizes the biological prior knowledge of intra-modal and inter-modal data associations to guide the feature extraction. The node features of each modality are updated through attention-based graph learning strategy. Unimodal features and bi-modal fused features are extracted via attention pooling module and then used for survival prediction. We evaluate the model on low-grade gliomas, glioblastoma, and kidney renal papillary cell carcinoma datasets from the Cancer Genome Atlas (TCGA) and the First Affiliated Hospital of Zhengzhou University (FAHZU). Extensive experimental results demonstrate that the proposed method outperforms both unimodal and other multi-modal fusion models. For demonstrating the model interpretability, we also visualize the attention heatmap of pathological images and utilize integrated gradient algorithm to identify important tissue structure, biological pathways and key genes.

Read more

4/15/2024

Multimodal Cross-Task Interaction for Survival Analysis in Whole Slide Pathological Images
Total Score

0

Multimodal Cross-Task Interaction for Survival Analysis in Whole Slide Pathological Images

Songhan Jiang, Zhengyu Gan, Linghan Cai, Yifeng Wang, Yongbing Zhang

Survival prediction, utilizing pathological images and genomic profiles, is increasingly important in cancer analysis and prognosis. Despite significant progress, precise survival analysis still faces two main challenges: (1) The massive pixels contained in whole slide images (WSIs) complicate the process of pathological images, making it difficult to generate an effective representation of the tumor microenvironment (TME). (2) Existing multimodal methods often rely on alignment strategies to integrate complementary information, which may lead to information loss due to the inherent heterogeneity between pathology and genes. In this paper, we propose a Multimodal Cross-Task Interaction (MCTI) framework to explore the intrinsic correlations between subtype classification and survival analysis tasks. Specifically, to capture TME-related features in WSIs, we leverage the subtype classification task to mine tumor regions. Simultaneously, multi-head attention mechanisms are applied in genomic feature extraction, adaptively performing genes grouping to obtain task-related genomic embedding. With the joint representation of pathological images and genomic data, we further introduce a Transport-Guided Attention (TGA) module that uses optimal transport theory to model the correlation between subtype classification and survival analysis tasks, effectively transferring potential information. Extensive experiments demonstrate the superiority of our approaches, with MCTI outperforming state-of-the-art frameworks on three public benchmarks. href{https://github.com/jsh0792/MCTI}{https://github.com/jsh0792/MCTI}.

Read more

6/26/2024