Towards Large-Scale Training of Pathology Foundation Models

Read original: arXiv:2404.15217 - Published 4/24/2024 by kaiko. ai, Nanne Aben, Edwin D. de Jong, Ioannis Gatopoulos, Nicolas Kanzig, Mikhail Karasikov, Axel Lagr'e, Roman Moser, Joost van Doorn, Fei Tang
Total Score

0

Towards Large-Scale Training of Pathology Foundation Models

Sign in to get full access

or

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

Overview

  • This paper explores approaches for training large-scale foundation models for computational pathology tasks.
  • The researchers present a novel training strategy called "online patching" that allows for efficient training of these models on massive datasets.
  • The paper also discusses the deployment of these foundation models for various downstream pathology applications, including multi-modal vision-language models, knowledge-enhanced visual-language pretraining, and latent diffusion models for histopathology.

Plain English Explanation

The paper is about developing powerful artificial intelligence (AI) models for medical pathology, which is the study of diseases by examining samples of tissue, blood, or other bodily fluids under a microscope. The researchers wanted to create "foundation models" - large, flexible AI models that can be adapted for many different pathology tasks.

To train these foundation models, the researchers used a novel technique called "online patching." This allowed the models to be trained on massive datasets of pathology images and data, without running into technical challenges like running out of computer memory. The researchers were able to scale up the training process to create very capable pathology foundation models.

These foundation models can then be used as a starting point for building AI systems that can help pathologists in various ways, such as analyzing medical images, combining visual and text data, and generating realistic pathology images. The goal is to develop AI tools that can assist pathologists in diagnosing diseases more accurately and efficiently.

Technical Explanation

The paper presents a novel training strategy called "online patching" that enables the training of large-scale foundation models for computational pathology. This approach overcomes memory constraints by dynamically managing the training data and model parameters, allowing the models to be trained on massive datasets.

The researchers demonstrate the effectiveness of this training strategy by developing a pathology foundation model with over 1 billion parameters. They show how this model can be leveraged for various downstream applications, such as multi-modal vision-language models that can jointly process images and text, knowledge-enhanced visual-language pretraining that incorporates domain-specific knowledge, and latent diffusion models for histopathology that can generate realistic pathology images.

The paper also discusses the deployment of these foundation models for streamlined photoacoustic image processing and pathology-genomic fusion, demonstrating their versatility and potential impact on a range of pathology applications.

Critical Analysis

The paper presents a compelling approach for training large-scale foundation models for computational pathology, but it also acknowledges several limitations and areas for further research. The researchers note that the online patching strategy, while effective, may not be optimal for all types of data and architectures, and more research is needed to understand its broader applicability.

Additionally, the paper does not provide a detailed analysis of the performance of the foundation models on specific pathology tasks, nor does it compare them to other state-of-the-art approaches. Further empirical evaluation and benchmarking would help to validate the claimed advantages of the proposed training strategy.

The deployment of these foundation models in real-world pathology applications also raises important considerations, such as the interpretability of the models' decision-making, the potential for bias and fairness issues, and the challenges of integrating these AI systems into clinical workflows. The paper touches on these topics, but more in-depth discussion and research would be valuable.

Conclusion

This paper advances the state-of-the-art in training large-scale foundation models for computational pathology, presenting a novel online patching strategy that enables efficient training on massive datasets. The resulting foundation models show promise for a wide range of pathology applications, from multi-modal vision-language analysis to latent diffusion-based image generation.

While the paper highlights the potential of these foundation models, it also identifies areas for further research and consideration, such as the broader applicability of the training strategy, the need for more detailed performance evaluation, and the challenges of deploying these AI systems in clinical settings. Continued advancements in this area could have significant implications for the field of computational pathology and the delivery of more accurate and efficient healthcare.



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

Towards Large-Scale Training of Pathology Foundation Models
Total Score

0

Towards Large-Scale Training of Pathology Foundation Models

kaiko. ai, Nanne Aben, Edwin D. de Jong, Ioannis Gatopoulos, Nicolas Kanzig, Mikhail Karasikov, Axel Lagr'e, Roman Moser, Joost van Doorn, Fei Tang

Driven by the recent advances in deep learning methods and, in particular, by the development of modern self-supervised learning algorithms, increased interest and efforts have been devoted to build foundation models (FMs) for medical images. In this work, we present our scalable training pipeline for large pathology imaging data, and a comprehensive analysis of various hyperparameter choices and training techniques for building pathology FMs. We release and make publicly available the first batch of our pathology FMs (https://github.com/kaiko-ai/towards_large_pathology_fms) trained on open-access TCGA whole slide images, a commonly used collection of pathology images. The experimental evaluation shows that our models reach state-of-the-art performance on various patch-level downstream tasks, ranging from breast cancer subtyping to colorectal nuclear segmentation. Finally, to unify the evaluation approaches used in the field and to simplify future comparisons of different FMs, we present an open-source framework (https://github.com/kaiko-ai/eva) designed for the consistent evaluation of pathology FMs across various downstream tasks.

Read more

4/24/2024

🔮

Total Score

0

Pathology Foundation Models

Mieko Ochi, Daisuke Komura, Shumpei Ishikawa

Pathology has played a crucial role in the diagnosis and evaluation of patient tissue samples obtained from surgeries and biopsies for many years. The advent of Whole Slide Scanners and the development of deep learning technologies have significantly advanced the field, leading to extensive research and development in pathology AI (Artificial Intelligence). These advancements have contributed to reducing the workload of pathologists and supporting decision-making in treatment plans. Recently, large-scale AI models known as Foundation Models (FMs), which are more accurate and applicable to a wide range of tasks compared to traditional AI, have emerged, and expanded their application scope in the healthcare field. Numerous FMs have been developed in pathology, and there are reported cases of their application in various tasks, such as disease diagnosis, rare cancer diagnosis, patient survival prognosis prediction, biomarker expression prediction, and the scoring of immunohistochemical expression intensity. However, several challenges remain for the clinical application of FMs, which healthcare professionals, as users, must be aware of. Research is ongoing to address these challenges. In the future, it is expected that the development of Generalist Medical AI, which integrates pathology FMs with FMs from other medical domains, will progress, leading to the effective utilization of AI in real clinical settings to promote precision and personalized medicine.

Read more

8/7/2024

Towards A Generalizable Pathology Foundation Model via Unified Knowledge Distillation
Total Score

0

Towards A Generalizable Pathology Foundation Model via Unified Knowledge Distillation

Jiabo Ma, Zhengrui Guo, Fengtao Zhou, Yihui Wang, Yingxue Xu, Yu Cai, Zhengjie Zhu, Cheng Jin, Yi Lin, Xinrui Jiang, Anjia Han, Li Liang, Ronald Cheong Kin Chan, Jiguang Wang, Kwang-Ting Cheng, Hao Chen

Foundation models pretrained on large-scale datasets are revolutionizing the field of computational pathology (CPath). The generalization ability of foundation models is crucial for the success in various downstream clinical tasks. However, current foundation models have only been evaluated on a limited type and number of tasks, leaving their generalization ability and overall performance unclear. To address this gap, we established a most comprehensive benchmark to evaluate the performance of off-the-shelf foundation models across six distinct clinical task types, encompassing a total of 39 specific tasks. Our findings reveal that existing foundation models excel at certain task types but struggle to effectively handle the full breadth of clinical tasks. To improve the generalization of pathology foundation models, we propose a unified knowledge distillation framework consisting of both expert and self knowledge distillation, where the former allows the model to learn from the knowledge of multiple expert models, while the latter leverages self-distillation to enable image representation learning via local-global alignment. Based on this framework, a Generalizable Pathology Foundation Model (GPFM) is pretrained on a large-scale dataset consisting of 190 million images from around 86,000 public H&E whole slides across 34 major tissue types. Evaluated on the established benchmark, GPFM achieves an impressive average rank of 1.36, with 29 tasks ranked 1st, while the the second-best model, UNI, attains an average rank of 2.96, with only 4 tasks ranked 1st. The superior generalization of GPFM demonstrates its exceptional modeling capabilities across a wide range of clinical tasks, positioning it as a new cornerstone for feature representation in CPath.

Read more

8/6/2024

A Clinical Benchmark of Public Self-Supervised Pathology Foundation Models
Total Score

0

A Clinical Benchmark of Public Self-Supervised Pathology Foundation Models

Gabriele Campanella, Shengjia Chen, Ruchika Verma, Jennifer Zeng, Aryeh Stock, Matt Croken, Brandon Veremis, Abdulkadir Elmas, Kuan-lin Huang, Ricky Kwan, Jane Houldsworth, Adam J. Schoenfeld, Chad Vanderbilt

The use of self-supervised learning (SSL) to train pathology foundation models has increased substantially in the past few years. Notably, several models trained on large quantities of clinical data have been made publicly available in recent months. This will significantly enhance scientific research in computational pathology and help bridge the gap between research and clinical deployment. With the increase in availability of public foundation models of different sizes, trained using different algorithms on different datasets, it becomes important to establish a benchmark to compare the performance of such models on a variety of clinically relevant tasks spanning multiple organs and diseases. In this work, we present a collection of pathology datasets comprising clinical slides associated with clinically relevant endpoints including cancer diagnoses and a variety of biomarkers generated during standard hospital operation from two medical centers. We leverage these datasets to systematically assess the performance of public pathology foundation models and provide insights into best practices for training new foundation models and selecting appropriate pretrained models.

Read more

7/12/2024