RGDA-DDI: Residual graph attention network and dual-attention based framework for drug-drug interaction prediction

Read original: arXiv:2408.15310 - Published 8/29/2024 by Changjian Zhou, Xin Zhang, Jiafeng Li, Jia Song, Wensheng Xiang
Total Score

0

RGDA-DDI: Residual graph attention network and dual-attention based framework for drug-drug interaction prediction

Sign in to get full access

or

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

Overview

  • Presents a novel framework called RGDA-DDI for predicting drug-drug interactions (DDIs)
  • Combines a residual graph attention network (RGAT) and a dual-attention mechanism to capture multi-scale features
  • Achieves state-of-the-art performance on benchmark DDI datasets

Plain English Explanation

The paper introduces a new machine learning framework called RGDA-DDI for predicting drug-drug interactions (DDIs). DDIs occur when one drug affects the way another drug works, which can be harmful to patients. Accurately predicting DDIs is important for ensuring drug safety.

RGDA-DDI uses a residual graph attention network (RGAT) to learn representations of the drugs and their interactions. This allows the model to capture important features at multiple scales, from the individual drug properties to the complex relationships between drugs. The model also employs a dual-attention mechanism to further refine these representations and make more accurate predictions.

Overall, the RGDA-DDI framework demonstrates state-of-the-art performance on standard DDI datasets, suggesting it is a promising approach for this important problem in drug safety and pharmacology.

Technical Explanation

The key innovations of RGDA-DDI are the residual graph attention network (RGAT) and the dual-attention mechanism. The RGAT builds on the standard graph attention network (GAT) by adding residual connections, which help the model learn more effective representations of the drug-drug interaction graph.

The dual-attention mechanism consists of two attention layers that operate at different scales. The first attention layer focuses on learning representations of individual drugs, while the second layer learns how the drugs interact with each other. By combining these multi-scale features, the model can make more accurate predictions of DDIs.

The authors evaluate RGDA-DDI on several benchmark DDI datasets and show that it outperforms previous state-of-the-art methods. They also conduct ablation studies to understand the contributions of the different components of their framework.

Critical Analysis

The authors acknowledge several limitations of their work. First, the model is trained and evaluated on existing DDI datasets, which may not fully capture the complexity of real-world drug interactions. Additionally, the model relies on the availability of drug feature information, which may not always be easily accessible.

Furthermore, while the RGDA-DDI framework demonstrates strong performance, the authors do not provide a comprehensive analysis of its computational complexity or inference time. These practical considerations are important for deploying such models in real-world drug discovery and development pipelines.

Finally, the authors do not discuss potential ethical implications or societal impacts of their work. As DDI prediction models become more advanced, it will be important to consider how they can be responsibly developed and deployed to ensure patient safety and equitable access to healthcare.

Conclusion

The RGDA-DDI framework presented in this paper is a significant advancement in the field of drug-drug interaction prediction. By combining a residual graph attention network with a dual-attention mechanism, the model is able to capture multi-scale features of the drug-drug interaction graph and achieve state-of-the-art performance on benchmark datasets.

While the authors acknowledge several limitations, the RGDA-DDI framework represents an important step forward in using machine learning to improve drug safety and patient outcomes. As the field of computational pharmacology continues to evolve, this work highlights the potential for advanced neural network architectures to tackle complex problems in drug discovery and development.



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

RGDA-DDI: Residual graph attention network and dual-attention based framework for drug-drug interaction prediction
Total Score

0

RGDA-DDI: Residual graph attention network and dual-attention based framework for drug-drug interaction prediction

Changjian Zhou, Xin Zhang, Jiafeng Li, Jia Song, Wensheng Xiang

Recent studies suggest that drug-drug interaction (DDI) prediction via computational approaches has significant importance for understanding the functions and co-prescriptions of multiple drugs. However, the existing silico DDI prediction methods either ignore the potential interactions among drug-drug pairs (DDPs), or fail to explicitly model and fuse the multi-scale drug feature representations for better prediction. In this study, we propose RGDA-DDI, a residual graph attention network (residual-GAT) and dual-attention based framework for drug-drug interaction prediction. A residual-GAT module is introduced to simultaneously learn multi-scale feature representations from drugs and DDPs. In addition, a dual-attention based feature fusion block is constructed to learn local joint interaction representations. A series of evaluation metrics demonstrate that the RGDA-DDI significantly improved DDI prediction performance on two public benchmark datasets, which provides a new insight into drug development.

Read more

8/29/2024

🔮

Total Score

0

HiGraphDTI: Hierarchical Graph Representation Learning for Drug-Target Interaction Prediction

Bin Liu, Siqi Wu, Jin Wang, Xin Deng, Ao Zhou

The discovery of drug-target interactions (DTIs) plays a crucial role in pharmaceutical development. The deep learning model achieves more accurate results in DTI prediction due to its ability to extract robust and expressive features from drug and target chemical structures. However, existing deep learning methods typically generate drug features via aggregating molecular atom representations, ignoring the chemical properties carried by motifs, i.e., substructures of the molecular graph. The atom-drug double-level molecular representation learning can not fully exploit structure information and fails to interpret the DTI mechanism from the motif perspective. In addition, sequential model-based target feature extraction either fuses limited contextual information or requires expensive computational resources. To tackle the above issues, we propose a hierarchical graph representation learning-based DTI prediction method (HiGraphDTI). Specifically, HiGraphDTI learns hierarchical drug representations from triple-level molecular graphs to thoroughly exploit chemical information embedded in atoms, motifs, and molecules. Then, an attentional feature fusion module incorporates information from different receptive fields to extract expressive target features.Last, the hierarchical attention mechanism identifies crucial molecular segments, which offers complementary views for interpreting interaction mechanisms. The experiment results not only demonstrate the superiority of HiGraphDTI to the state-of-the-art methods, but also confirm the practical ability of our model in interaction interpretation and new DTI discovery.

Read more

4/17/2024

🌐

Total Score

0

MKDTI: Predicting drug-target interactions via multiple kernel fusion on graph attention network

Yuhuan Zhou, Yulin Wu, Weiwei Yuan, Xuan Wang, Junyi Li

Drug-target relationships may now be predicted computationally using bioinformatics data, which is a valuable tool for understanding pharmacological effects, enhancing drug development efficiency, and advancing related research. A number of structure-based, ligand-based and network-based approaches have now emerged. Furthermore, the integration of graph attention networks with intricate drug target studies is an application area of growing interest. In our work, we formulate a model called MKDTI by extracting kernel information from various layer embeddings of a graph attention network. This combination improves the prediction ability with respect to novel drug-target relationships. We first build a drug-target heterogeneous network using heterogeneous data of drugs and targets, and then use a self-enhanced multi-head graph attention network to extract potential features in each layer. Next, we utilize embeddings of each layer to computationally extract kernel matrices and fuse multiple kernel matrices. Finally, we use a Dual Laplacian Regularized Least Squares framework to forecast novel drug-target entity connections. This prediction can be facilitated by integrating the kernel matrix associated with the drug-target. We measured our model's efficacy using AUPR and AUC. Compared to the benchmark algorithms, our model outperforms them in the prediction outcomes. In addition, we conducted an experiment on kernel selection. The results show that the multi-kernel fusion approach combined with the kernel matrix generated by the graph attention network provides complementary insights into the model. The fusion of this information helps to enhance the accuracy of the predictions.

Read more

7/16/2024

🔮

Total Score

0

DDIPrompt: Drug-Drug Interaction Event Prediction based on Graph Prompt Learning

Yingying Wang, Yun Xiong, Xixi Wu, Xiangguo Sun, Jiawei Zhang

Predicting drug-drug interaction adverse events, so-called DDI events, is increasingly valuable as it facilitates the study of mechanisms underlying drug use or adverse reactions. Existing models often neglect the distinctive characteristics of individual event classes when integrating multi-source features, which contributes to systematic unfairness when dealing with highly imbalanced event samples. Moreover, the limited capacity of these models to abstract the unique attributes of each event subclass considerably hampers their application in predicting rare drug-drug interaction events with a limited sample size. Reducing dataset bias and abstracting event subclass characteristics are two unresolved challenges. Recently, prompt tuning with frozen pre-trained graph models, namely pre-train, prompt, fine-tune strategy, has demonstrated impressive performance in few-shot tasks. Motivated by this, we propose an advanced method as a solution to address these aforementioned challenges. Specifically, our proposed approach entails a hierarchical pre-training task that aims to capture crucial aspects of drug molecular structure and intermolecular interactions while effectively mitigating implicit dataset bias within the node embeddings. Furthermore, we construct a prototypical graph by strategically sampling data from distinct event types and design subgraph prompts utilizing pre-trained node features. Through comprehensive benchmark experiments, we validate the efficacy of our subgraph prompts in accurately representing event classes and achieve exemplary results in both overall and subclass prediction tasks.

Read more

5/24/2024