Pre-Training on Large-Scale Generated Docking Conformations with HelixDock to Unlock the Potential of Protein-ligand Structure Prediction Models

Read original: arXiv:2310.13913 - Published 5/24/2024 by Lihang Liu, Shanzhuo Zhang, Donglong He, Xianbin Ye, Jingbo Zhou, Xiaonan Zhang, Yaoyao Jiang, Weiming Diao, Hang Yin, Hua Chai and 5 others
Total Score

0

🔮

Sign in to get full access

or

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

Overview

  • This paper presents a new approach to protein-ligand structure prediction, a crucial task in drug discovery.
  • Protein-ligand structure prediction involves predicting how small drug-like molecules (ligands) will bind to target proteins (receptors).
  • The researchers used a combination of traditional physics-based docking tools and deep learning techniques to develop a model called HelixDock that can accurately predict protein-ligand binding conformations.
  • HelixDock was pre-trained on a large dataset of docking conformations generated by physics-based tools, then fine-tuned on a smaller set of experimentally validated receptor-ligand complexes.
  • The model demonstrates exceptional performance on benchmark tasks and shows promise for practical applications in drug discovery.

Plain English Explanation

When designing new drugs, researchers need to understand how potential drug molecules will bind to and interact with their target proteins in the body. This is known as protein-ligand structure prediction. Traditionally, this has been done using physics-based computer models that simulate the interactions between molecules.

However, these physics-based methods can be computationally expensive and have limitations in their accuracy. In recent years, researchers have started using deep learning techniques to try to improve protein-ligand structure prediction. But these deep learning models often rely on a relatively small amount of experimental data, which can limit their generalizability.

The researchers in this paper developed a new approach called HelixDock that combines the strengths of physics-based and deep learning methods. First, they used physics-based docking tools to generate a massive dataset of over 100 million potential protein-ligand binding conformations. They then used this large dataset to pre-train a deep learning model, allowing it to learn the underlying physics of molecular interactions.

Next, they fine-tuned this pre-trained model using a smaller set of experimentally validated protein-ligand complexes. This allowed HelixDock to learn from both the large-scale physics-based dataset and the high-quality experimental data, resulting in a model that can accurately predict protein-ligand binding conformations.

The researchers thoroughly tested HelixDock and found that it outperforms both physics-based and other deep learning-based methods on a range of benchmarks. They also applied HelixDock to several drug discovery-related tasks, demonstrating its practical utility for real-world applications.

Overall, this work shows how combining the strengths of physics-based and deep learning approaches can lead to significant improvements in protein-ligand structure prediction, which is a crucial step in the drug discovery process.

Technical Explanation

The researchers in this paper developed a new deep learning-based model called HelixDock for predicting the binding conformations of protein-ligand complexes. To address the limitations of previous deep learning approaches, which often suffer from a lack of diverse training data, the researchers employed a two-stage training process.

First, they generated a massive dataset of over 100 million docking conformations using traditional physics-based docking tools. This large-scale dataset allowed HelixDock to learn the underlying physical principles governing protein-ligand interactions during the pre-training phase.

Next, the pre-trained HelixDock model was fine-tuned on a smaller set of experimentally validated protein-ligand complexes. This allowed the model to refine its predictions and capture the nuances of real-world protein-ligand binding interactions.

The researchers thoroughly benchmarked HelixDock against both physics-based and deep learning-based methods, demonstrating its exceptional performance in predicting binding conformations. They also investigated the scaling laws governing pre-trained protein-ligand structure prediction models, finding that increasing the model size and pre-training dataset size consistently improves performance.

Furthermore, the researchers applied HelixDock to several drug discovery-related tasks, such as cross-docking and structure-based virtual screening, and found that it outperformed existing approaches.

Critical Analysis

The researchers in this paper have taken an innovative approach to addressing the limitations of previous deep learning-based protein-ligand structure prediction methods. By leveraging a large-scale dataset of docking conformations generated by physics-based tools, they were able to pre-train their model to learn the underlying physical principles governing protein-ligand interactions.

However, it is worth noting that the generation of this 100 million-sample dataset was an extremely computationally intensive process, requiring roughly 1 million CPU core days. This raises questions about the scalability and accessibility of the approach, as not all research groups may have access to such significant computational resources.

Additionally, while the researchers extensively benchmarked HelixDock against existing methods, the true test of its practical utility will be its performance in real-world drug discovery pipelines. Further validation on a wider range of datasets and use cases would help to strengthen the case for the model's broader applicability.

Another potential concern is the reliance on experimental data for fine-tuning the pre-trained model. The availability and quality of such data can vary, and the researchers acknowledge that this may limit the generalizability of their approach. Exploring ways to further improve the model's performance without extensive fine-tuning could be an area for future research.

Despite these potential limitations, the researchers have made a significant contribution to the field of protein-ligand structure prediction. The HelixDock model demonstrates impressive performance and the potential to enhance drug discovery efforts. Continued advancements in this area, coupled with a critical examination of the underlying assumptions and limitations, will be crucial for realizing the full impact of these techniques.

Conclusion

This paper presents a novel approach to protein-ligand structure prediction that combines the strengths of physics-based and deep learning techniques. By pre-training on a large-scale dataset of docking conformations and then fine-tuning on experimentally validated data, the researchers developed the HelixDock model, which demonstrates exceptional performance on a range of benchmarks.

The key innovation of this work is the leveraging of physics-based docking tools to generate a massive training dataset, allowing the deep learning model to learn the underlying principles of protein-ligand interactions. This approach addresses the limitations of previous deep learning methods that often suffer from a lack of diverse training data.

The researchers' findings indicate that pre-trained protein-ligand structure prediction models can be further enhanced through increases in model size and pre-training dataset volume, suggesting promising avenues for continued progress in this field. Additionally, the successful application of HelixDock to drug discovery-related tasks underscores the practical utility of this approach for real-world drug development pipelines.

Overall, this work represents a significant advancement in the field of protein-ligand structure prediction and highlights the potential of combined physics-based and deep learning techniques to drive innovation in drug discovery and other areas of computational biology.



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

Pre-Training on Large-Scale Generated Docking Conformations with HelixDock to Unlock the Potential of Protein-ligand Structure Prediction Models

Lihang Liu, Shanzhuo Zhang, Donglong He, Xianbin Ye, Jingbo Zhou, Xiaonan Zhang, Yaoyao Jiang, Weiming Diao, Hang Yin, Hua Chai, Fan Wang, Jingzhou He, Liang Zheng, Yonghui Li, Xiaomin Fang

Protein-ligand structure prediction is an essential task in drug discovery, predicting the binding interactions between small molecules (ligands) and target proteins (receptors). Recent advances have incorporated deep learning techniques to improve the accuracy of protein-ligand structure prediction. Nevertheless, the experimental validation of docking conformations remains costly, it raises concerns regarding the generalizability of these deep learning-based methods due to the limited training data. In this work, we show that by pre-training on a large-scale docking conformation generated by traditional physics-based docking tools and then fine-tuning with a limited set of experimentally validated receptor-ligand complexes, we can obtain a protein-ligand structure prediction model with outstanding performance. Specifically, this process involved the generation of 100 million docking conformations for protein-ligand pairings, an endeavor consuming roughly 1 million CPU core days. The proposed model, HelixDock, aims to acquire the physical knowledge encapsulated by the physics-based docking tools during the pre-training phase. HelixDock has been rigorously benchmarked against both physics-based and deep learning-based baselines, demonstrating its exceptional precision and robust transferability in predicting binding confirmation. In addition, our investigation reveals the scaling laws governing pre-trained protein-ligand structure prediction models, indicating a consistent enhancement in performance with increases in model parameters and the volume of pre-training data. Moreover, we applied HelixDock to several drug discovery-related tasks to validate its practical utility. HelixDock demonstrates outstanding capabilities on both cross-docking and structure-based virtual screening benchmarks.

Read more

5/24/2024

One-step Structure Prediction and Screening for Protein-Ligand Complexes using Multi-Task Geometric Deep Learning
Total Score

0

One-step Structure Prediction and Screening for Protein-Ligand Complexes using Multi-Task Geometric Deep Learning

Kelei He, Tiejun Dong, Jinhui Wu, Junfeng Zhang

Understanding the structure of the protein-ligand complex is crucial to drug development. Existing virtual structure measurement and screening methods are dominated by docking and its derived methods combined with deep learning. However, the sampling and scoring methodology have largely restricted the accuracy and efficiency. Here, we show that these two fundamental tasks can be accurately tackled with a single model, namely LigPose, based on multi-task geometric deep learning. By representing the ligand and the protein pair as a graph, LigPose directly optimizes the three-dimensional structure of the complex, with the learning of binding strength and atomic interactions as auxiliary tasks, enabling its one-step prediction ability without docking tools. Extensive experiments show LigPose achieved state-of-the-art performance on major tasks in drug research. Its considerable improvements indicate a promising paradigm of AI-based pipeline for drug development.

Read more

8/22/2024

🤿

Total Score

0

Deep Learning for Protein-Ligand Docking: Are We There Yet?

Alex Morehead, Nabin Giri, Jian Liu, Jianlin Cheng

The effects of ligand binding on protein structures and their in vivo functions carry numerous implications for modern biomedical research and biotechnology development efforts such as drug discovery. Although several deep learning (DL) methods and benchmarks designed for protein-ligand docking have recently been introduced, to date no prior works have systematically studied the behavior of docking methods within the practical context of (1) using predicted (apo) protein structures for docking (e.g., for broad applicability); (2) docking multiple ligands concurrently to a given target protein (e.g., for enzyme design); and (3) having no prior knowledge of binding pockets (e.g., for pocket generalization). To enable a deeper understanding of docking methods' real-world utility, we introduce PoseBench, the first comprehensive benchmark for practical protein-ligand docking. PoseBench enables researchers to rigorously and systematically evaluate DL docking methods for apo-to-holo protein-ligand docking and protein-ligand structure generation using both single and multi-ligand benchmark datasets, the latter of which we introduce for the first time to the DL community. Empirically, using PoseBench, we find that all recent DL docking methods but one fail to generalize to multi-ligand protein targets and also that template-based docking algorithms perform equally well or better for multi-ligand docking as recent single-ligand DL docking methods, suggesting areas of improvement for future work. Code, data, tutorials, and benchmark results are available at https://github.com/BioinfoMachineLearning/PoseBench.

Read more

7/9/2024

🔮

Total Score

0

Improved prediction of ligand-protein binding affinities by meta-modeling

Ho-Joon Lee, Prashant S. Emani, Mark B. Gerstein

The accurate screening of candidate drug ligands against target proteins through computational approaches is of prime interest to drug development efforts. Such virtual screening depends in part on methods to predict the binding affinity between ligands and proteins. Many computational models for binding affinity prediction have been developed, but with varying results across targets. Given that ensembling or meta-modeling methods have shown great promise in reducing model-specific biases, we develop a framework to integrate published force-field-based empirical docking and sequence-based deep learning models. In building this framework, we evaluate many combinations of individual base models, training databases, and several meta-modeling approaches. We show that many of our meta-models significantly improve affinity predictions over base models. Our best meta-models achieve comparable performance to state-of-the-art deep learning tools exclusively based on structures, while allowing for improved database scalability and flexibility through the explicit inclusion of features such as physicochemical properties or molecular descriptors. Overall, we demonstrate that diverse modeling approaches can be ensembled together to gain improvement in binding affinity prediction.

Read more

5/21/2024