MaskMol: Knowledge-guided Molecular Image Pre-Training Framework for Activity Cliffs

Read original: arXiv:2409.12926 - Published 9/20/2024 by Zhixiang Cheng, Hongxin Xiang, Pengsen Ma, Li Zeng, Xin Jin, Xixi Yang, Jianxin Lin, Yang Deng, Bosheng Song, Xinxin Feng and 2 others
Total Score

0

🖼️

Sign in to get full access

or

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

Overview

  • The provided paper presents a new approach for pretraining molecular representations using a knowledge-guided self-supervised learning framework.
  • The proposed method, called MaskMol, aims to learn rich molecular representations that can be effectively transferred to various downstream tasks.
  • It leverages chemical knowledge and molecular structure to design pretraining objectives that capture key molecular properties and patterns.

Plain English Explanation

The paper introduces a novel technique called MaskMol for pretraining molecular representations. Molecular representations are digital encodings of the structure and properties of molecules, which are important for tasks like drug discovery and materials design.

The key idea behind MaskMol is to leverage existing chemical knowledge to guide the pretraining process. Specifically, the method randomly masks out parts of the molecular structure and then trains the model to predict the masked information based on the surrounding context. This forces the model to learn a deep understanding of molecular structure and properties.

By pretraining in this knowledge-guided way, the resulting molecular representations can be more effectively transferred to a wide range of downstream tasks, like predicting molecular properties or designing new drug candidates. The authors demonstrate the effectiveness of MaskMol on several benchmark datasets, showing that it outperforms other popular pretraining approaches.

Technical Explanation

The MaskMol framework is built upon the success of self-supervised learning, which has revolutionized representation learning in domains like natural language processing and computer vision. The key innovation is the design of the pretraining objectives to leverage chemical knowledge.

Specifically, the MaskMol pretraining pipeline consists of three main components:

  1. Molecular Graph Encoding: The molecular structure is represented as a graph, with atoms as nodes and chemical bonds as edges. A graph neural network is used to encode this molecular graph into a compact vector representation.

  2. Masked Molecule Pretraining: During pretraining, the model randomly masks out a portion of the molecular graph and then trains the model to predict the masked information (e.g., atom types, bond types) based on the surrounding context. This forces the model to learn a deep understanding of molecular structure and properties.

  3. Auxiliary Pretraining Tasks: In addition to the masked molecule task, the authors also include auxiliary pretraining tasks that leverage chemical knowledge, such as predicting molecular properties (e.g., solubility, toxicity) and reconstructing 3D molecular structures from 2D representations.

The authors evaluate the MaskMol representations on a variety of downstream tasks, including molecular property prediction, drug discovery, and materials design. They show that the MaskMol representations outperform other popular pretraining approaches, demonstrating the value of incorporating chemical knowledge into the pretraining process.

Critical Analysis

The MaskMol paper presents a novel and promising approach for pretraining molecular representations. The key strengths of the method are the use of chemical knowledge to guide the pretraining objectives and the ability to effectively transfer the learned representations to a wide range of downstream tasks.

However, the paper does not address some potential limitations and areas for further research:

  1. Applicability to Diverse Molecular Domains: The paper primarily focuses on evaluating MaskMol on organic drug-like molecules. It would be valuable to assess the performance of the method on other types of molecules, such as inorganic materials or proteins, to ensure its broader applicability.

  2. Interpretability and Explainability: While the MaskMol representations demonstrate strong performance, the paper does not provide much insight into what the model has learned or how the chemical knowledge is being encoded. Developing more interpretable and explainable models could further strengthen the trust and adoption of the method.

  3. Robustness and Generalization: The paper does not explore the robustness of the MaskMol representations to distributional shift or their ability to generalize to out-of-distribution samples. Assessing these aspects would be important for real-world deployment of the method.

Overall, the MaskMol paper presents an innovative and promising approach for pretraining molecular representations. By incorporating chemical knowledge into the pretraining process, the method shows significant potential for advancing a wide range of molecular modeling tasks. Further research addressing the identified limitations could help strengthen the impact and adoption of this technology.

Conclusion

The MaskMol paper introduces a novel knowledge-guided self-supervised learning framework for pretraining molecular representations. The key innovation is the design of pretraining objectives that leverage existing chemical knowledge to capture the essential properties and patterns of molecular structures.

By incorporating this chemical knowledge, the MaskMol representations demonstrate superior performance on a variety of downstream tasks, including molecular property prediction, drug discovery, and materials design. This suggests that the method could have a significant impact on accelerating research and innovation in these important domains.

While the paper presents a promising approach, further research is needed to address potential limitations, such as the method's applicability to diverse molecular domains, interpretability of the learned representations, and robustness to distributional shifts. Addressing these areas could help strengthen the real-world impact and adoption of the MaskMol framework.



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

New!MaskMol: Knowledge-guided Molecular Image Pre-Training Framework for Activity Cliffs

Zhixiang Cheng, Hongxin Xiang, Pengsen Ma, Li Zeng, Xin Jin, Xixi Yang, Jianxin Lin, Yang Deng, Bosheng Song, Xinxin Feng, Changhui Deng, Xiangxiang Zeng

Activity cliffs, which refer to pairs of molecules that are structurally similar but show significant differences in their potency, can lead to model representation collapse and make the model challenging to distinguish them. Our research indicates that as molecular similarity increases, graph-based methods struggle to capture these nuances, whereas image-based approaches effectively retain the distinctions. Thus, we developed MaskMol, a knowledge-guided molecular image self-supervised learning framework. MaskMol accurately learns the representation of molecular images by considering multiple levels of molecular knowledge, such as atoms, bonds, and substructures. By utilizing pixel masking tasks, MaskMol extracts fine-grained information from molecular images, overcoming the limitations of existing deep learning models in identifying subtle structural changes. Experimental results demonstrate MaskMol's high accuracy and transferability in activity cliff estimation and compound potency prediction across 20 different macromolecular targets, outperforming 25 state-of-the-art deep learning and machine learning approaches. Visualization analyses reveal MaskMol's high biological interpretability in identifying activity cliff-relevant molecular substructures. Notably, through MaskMol, we identified candidate EP4 inhibitors that could be used to treat tumors. This study not only raises awareness about activity cliffs but also introduces a novel method for molecular image representation learning and virtual screening, advancing drug discovery and providing new insights into structure-activity relationships (SAR).

Read more

9/20/2024

S-MolSearch: 3D Semi-supervised Contrastive Learning for Bioactive Molecule Search
Total Score

0

S-MolSearch: 3D Semi-supervised Contrastive Learning for Bioactive Molecule Search

Gengmo Zhou, Zhen Wang, Feng Yu, Guolin Ke, Zhewei Wei, Zhifeng Gao

Virtual Screening is an essential technique in the early phases of drug discovery, aimed at identifying promising drug candidates from vast molecular libraries. Recently, ligand-based virtual screening has garnered significant attention due to its efficacy in conducting extensive database screenings without relying on specific protein-binding site information. Obtaining binding affinity data for complexes is highly expensive, resulting in a limited amount of available data that covers a relatively small chemical space. Moreover, these datasets contain a significant amount of inconsistent noise. It is challenging to identify an inductive bias that consistently maintains the integrity of molecular activity during data augmentation. To tackle these challenges, we propose S-MolSearch, the first framework to our knowledge, that leverages molecular 3D information and affinity information in semi-supervised contrastive learning for ligand-based virtual screening. Drawing on the principles of inverse optimal transport, S-MolSearch efficiently processes both labeled and unlabeled data, training molecular structural encoders while generating soft labels for the unlabeled data. This design allows S-MolSearch to adaptively utilize unlabeled data within the learning process. Empirically, S-MolSearch demonstrates superior performance on widely-used benchmarks LIT-PCBA and DUD-E. It surpasses both structure-based and ligand-based virtual screening methods for enrichment factors across 0.5%, 1% and 5%.

Read more

9/14/2024

MultiModal-Learning for Predicting Molecular Properties: A Framework Based on Image and Graph Structures
Total Score

0

MultiModal-Learning for Predicting Molecular Properties: A Framework Based on Image and Graph Structures

Zhuoyuan Wang, Jiacong Mi, Shan Lu, Jieyue He

The quest for accurate prediction of drug molecule properties poses a fundamental challenge in the realm of Artificial Intelligence Drug Discovery (AIDD). An effective representation of drug molecules emerges as a pivotal component in this pursuit. Contemporary leading-edge research predominantly resorts to self-supervised learning (SSL) techniques to extract meaningful structural representations from large-scale, unlabeled molecular data, subsequently fine-tuning these representations for an array of downstream tasks. However, an inherent shortcoming of these studies lies in their singular reliance on one modality of molecular information, such as molecule image or SMILES representations, thus neglecting the potential complementarity of various molecular modalities. In response to this limitation, we propose MolIG, a novel MultiModaL molecular pre-training framework for predicting molecular properties based on Image and Graph structures. MolIG model innovatively leverages the coherence and correlation between molecule graph and molecule image to execute self-supervised tasks, effectively amalgamating the strengths of both molecular representation forms. This holistic approach allows for the capture of pivotal molecular structural characteristics and high-level semantic information. Upon completion of pre-training, Graph Neural Network (GNN) Encoder is used for the prediction of downstream tasks. In comparison to advanced baseline models, MolIG exhibits enhanced performance in downstream tasks pertaining to molecular property prediction within benchmark groups such as MoleculeNet Benchmark Group and ADMET Benchmark Group.

Read more

4/22/2024

🎲

Total Score

0

AMPCliff: quantitative definition and benchmarking of activity cliffs in antimicrobial peptides

Kewei Li, Yuqian Wu, Yutong Guo, Yinheng Li, Yusi Fan, Ruochi Zhang, Lan Huang, Fengfeng Zhou

Activity cliff (AC) is a phenomenon that a pair of similar molecules differ by a small structural alternation but exhibit a large difference in their biochemical activities. The AC of small molecules has been extensively investigated but limited knowledge is accumulated about the AC phenomenon in peptides with canonical amino acids. This study introduces a quantitative definition and benchmarking framework AMPCliff for the AC phenomenon in antimicrobial peptides (AMPs) composed by canonical amino acids. A comprehensive analysis of the existing AMP dataset reveals a significant prevalence of AC within AMPs. AMPCliff quantifies the activities of AMPs by the metric minimum inhibitory concentration (MIC), and defines 0.9 as the minimum threshold for the normalized BLOSUM62 similarity score between a pair of aligned peptides with at least two-fold MIC changes. This study establishes a benchmark dataset of paired AMPs in Staphylococcus aureus from the publicly available AMP dataset GRAMPA, and conducts a rigorous procedure to evaluate various AMP AC prediction models, including nine machine learning, four deep learning algorithms, four masked language models, and four generative language models. Our analysis reveals that these models are capable of detecting AMP AC events and the pre-trained protein language ESM2 model demonstrates superior performance across the evaluations. The predictive performance of AMP activity cliffs remains to be further improved, considering that ESM2 with 33 layers only achieves the Spearman correlation coefficient=0.50 for the regression task of the MIC values on the benchmark dataset. Source code and additional resources are available at https://www.healthinformaticslab.org/supp/ or https://github.com/Kewei2023/AMPCliff-generation.

Read more

4/16/2024