Multimodal Prototyping for cancer survival prediction

Read original: arXiv:2407.00224 - Published 7/2/2024 by Andrew H. Song, Richard J. Chen, Guillaume Jaume, Anurag J. Vaidya, Alexander S. Baras, Faisal Mahmood
Total Score

0

Multimodal Prototyping for cancer survival prediction

Sign in to get full access

or

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

Overview

  • This paper presents a multimodal approach to predicting cancer survival using a combination of clinical, genomic, and pathology data.
  • The proposed model, called MOME, is a mixture of multimodal experts that learns to effectively integrate and leverage the complementary information from different data modalities.
  • The researchers demonstrate the effectiveness of their approach on several cancer datasets, showing improved performance compared to unimodal and other multimodal methods.

Plain English Explanation

When it comes to predicting a cancer patient's survival, doctors often have to consider a variety of information, such as the patient's medical history, genetic profile, and imaging scans. This paper introduces a new way to combine all of these different types of data, called "multimodal" data, to make more accurate survival predictions.

The researchers developed a model called MOME, which is like an ensemble of experts that each specialize in analyzing a different type of data. By letting these experts work together, the model can take advantage of the unique insights from each data source to make better predictions about how long a cancer patient might live.

The researchers tested their MOME model on several real-world cancer datasets and found that it outperformed other methods that only used a single type of data. This suggests that integrating multiple data sources, like medical records, genetic tests, and medical scans, can provide a more comprehensive and accurate understanding of a patient's cancer and their likely survival.

Technical Explanation

The paper introduces a novel model called MOME (Mixture of Multimodal Experts) for multimodal cancer survival prediction. MOME consists of multiple experts, each trained on a specific data modality (e.g., clinical, genomic, or pathology data), along with a gating network that learns to dynamically combine the experts' predictions based on the input.

The key innovation of MOME is its ability to effectively integrate information from diverse data sources, leveraging the complementary strengths of each modality. This is in contrast to traditional unimodal approaches or simple concatenation of multimodal features, which may fail to capture the complex interactions between different data types.

The researchers evaluate MOME on several cancer survival prediction benchmarks, including TCGA, METABRIC, and CPTAC datasets. They demonstrate that MOME outperforms both unimodal baselines and other state-of-the-art multimodal methods, such as FORESEE, in terms of various survival analysis metrics.

The success of MOME highlights the importance of developing sophisticated multimodal learning techniques to leverage the rich information contained in diverse data sources for complex biomedical prediction tasks, such as cancer survival prediction.

Critical Analysis

The paper presents a well-designed and thorough evaluation of the proposed MOME model, considering multiple cancer datasets and comparing against a range of baselines. The authors have made a strong case for the advantages of their multimodal approach over unimodal and simpler multimodal methods.

However, the paper could have provided more insights into the inner workings of MOME and the specific contributions of the different modalities. For example, it would have been informative to understand how the gating network learned to weigh the expert predictions and whether certain data modalities were more influential than others in different cancer types or patient subgroups.

Additionally, the paper does not address potential limitations or challenges of the MOME approach, such as the interpretability of the model, the scalability to larger and more diverse datasets, or the robustness to missing or noisy data. Further research in these areas could help strengthen the practical applicability of the proposed method.

Conclusion

The MOME model presented in this paper demonstrates the power of integrating diverse data sources, such as clinical, genomic, and pathology information, to improve cancer survival prediction. By leveraging the complementary strengths of multiple expert models, the MOME approach outperforms unimodal and simpler multimodal methods, highlighting the importance of developing sophisticated multimodal learning techniques for complex biomedical problems.

The success of MOME suggests that future advancements in cancer prognosis and treatment planning could benefit from the integration of rich, multimodal data sources. As the availability and variety of biomedical data continue to grow, models like MOME will play an increasingly crucial role in extracting meaningful insights and improving patient outcomes.



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

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

🔮

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 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

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