-
CovDocker: Benchmarking Covalent Drug Design with Tasks, Datasets, and Solutions
Authors:
Yangzhe Peng,
Kaiyuan Gao,
Liang He,
Yuheng Cong,
Haiguang Liu,
Kun He,
Lijun Wu
Abstract:
Molecular docking plays a crucial role in predicting the binding mode of ligands to target proteins, and covalent interactions, which involve the formation of a covalent bond between the ligand and the target, are particularly valuable due to their strong, enduring binding nature. However, most existing docking methods and deep learning approaches hardly account for the formation of covalent bonds…
▽ More
Molecular docking plays a crucial role in predicting the binding mode of ligands to target proteins, and covalent interactions, which involve the formation of a covalent bond between the ligand and the target, are particularly valuable due to their strong, enduring binding nature. However, most existing docking methods and deep learning approaches hardly account for the formation of covalent bonds and the associated structural changes. To address this gap, we introduce a comprehensive benchmark for covalent docking, CovDocker, which is designed to better capture the complexities of covalent binding. We decompose the covalent docking process into three main tasks: reactive location prediction, covalent reaction prediction, and covalent docking. By adapting state-of-the-art models, such as Uni-Mol and Chemformer, we establish baseline performances and demonstrate the effectiveness of the benchmark in accurately predicting interaction sites and modeling the molecular transformations involved in covalent binding. These results confirm the role of the benchmark as a rigorous framework for advancing research in covalent drug design. It underscores the potential of data-driven approaches to accelerate the discovery of selective covalent inhibitors and addresses critical challenges in therapeutic development.
△ Less
Submitted 26 June, 2025;
originally announced June 2025.
-
GTR-CoT: Graph Traversal as Visual Chain of Thought for Molecular Structure Recognition
Authors:
Jingchao Wang,
Haote Yang,
Jiang Wu,
Yifan He,
Xingjian Wei,
Yinfan Wang,
Chengjin Liu,
Lingli Ge,
Lijun Wu,
Bin Wang,
Dahua Lin,
Conghui He
Abstract:
Optical Chemical Structure Recognition (OCSR) is crucial for digitizing chemical knowledge by converting molecular images into machine-readable formats. While recent vision-language models (VLMs) have shown potential in this task, their image-captioning approach often struggles with complex molecular structures and inconsistent annotations. To overcome these challenges, we introduce GTR-Mol-VLM, a…
▽ More
Optical Chemical Structure Recognition (OCSR) is crucial for digitizing chemical knowledge by converting molecular images into machine-readable formats. While recent vision-language models (VLMs) have shown potential in this task, their image-captioning approach often struggles with complex molecular structures and inconsistent annotations. To overcome these challenges, we introduce GTR-Mol-VLM, a novel framework featuring two key innovations: (1) the Graph Traversal as Visual Chain of Thought mechanism that emulates human reasoning by incrementally parsing molecular graphs through sequential atom-bond predictions, and (2) the data-centric principle of Faithfully Recognize What You've Seen, which addresses the mismatch between abbreviated structures in images and their expanded annotations. To support model development, we constructed GTR-CoT-1.3M, a large-scale instruction-tuning dataset with meticulously corrected annotations, and introduced MolRec-Bench, the first benchmark designed for a fine-grained evaluation of graph-parsing accuracy in OCSR. Comprehensive experiments demonstrate that GTR-Mol-VLM achieves superior results compared to specialist models, chemistry-domain VLMs, and commercial general-purpose VLMs. Notably, in scenarios involving molecular images with functional group abbreviations, GTR-Mol-VLM outperforms the second-best baseline by approximately 14 percentage points, both in SMILES-based and graph-based metrics. We hope that this work will drive OCSR technology to more effectively meet real-world needs, thereby advancing the fields of cheminformatics and AI for Science. We will release GTR-CoT at https://github.com/opendatalab/GTR-CoT.
△ Less
Submitted 9 June, 2025; v1 submitted 9 June, 2025;
originally announced June 2025.
-
Tokenizing Electron Cloud in Protein-Ligand Interaction Learning
Authors:
Haitao Lin,
Odin Zhang,
Jia Xu,
Yunfan Liu,
Zheng Cheng,
Lirong Wu,
Yufei Huang,
Zhifeng Gao,
Stan Z. Li
Abstract:
The affinity and specificity of protein-molecule binding directly impact functional outcomes, uncovering the mechanisms underlying biological regulation and signal transduction. Most deep-learning-based prediction approaches focus on structures of atoms or fragments. However, quantum chemical properties, such as electronic structures, are the key to unveiling interaction patterns but remain largel…
▽ More
The affinity and specificity of protein-molecule binding directly impact functional outcomes, uncovering the mechanisms underlying biological regulation and signal transduction. Most deep-learning-based prediction approaches focus on structures of atoms or fragments. However, quantum chemical properties, such as electronic structures, are the key to unveiling interaction patterns but remain largely underexplored. To bridge this gap, we propose ECBind, a method for tokenizing electron cloud signals into quantized embeddings, enabling their integration into downstream tasks such as binding affinity prediction. By incorporating electron densities, ECBind helps uncover binding modes that cannot be fully represented by atom-level models. Specifically, to remove the redundancy inherent in electron cloud signals, a structure-aware transformer and hierarchical codebooks encode 3D binding sites enriched with electron structures into tokens. These tokenized codes are then used for specific tasks with labels. To extend its applicability to a wider range of scenarios, we utilize knowledge distillation to develop an electron-cloud-agnostic prediction model. Experimentally, ECBind demonstrates state-of-the-art performance across multiple tasks, achieving improvements of 6.42\% and 15.58\% in per-structure Pearson and Spearman correlation coefficients, respectively.
△ Less
Submitted 31 May, 2025; v1 submitted 25 May, 2025;
originally announced May 2025.
-
Analysis of the MICCAI Brain Tumor Segmentation -- Metastases (BraTS-METS) 2025 Lighthouse Challenge: Brain Metastasis Segmentation on Pre- and Post-treatment MRI
Authors:
Nazanin Maleki,
Raisa Amiruddin,
Ahmed W. Moawad,
Nikolay Yordanov,
Athanasios Gkampenis,
Pascal Fehringer,
Fabian Umeh,
Crystal Chukwurah,
Fatima Memon,
Bojan Petrovic,
Justin Cramer,
Mark Krycia,
Elizabeth B. Shrickel,
Ichiro Ikuta,
Gerard Thompson,
Lorenna Vidal,
Vilma Kosovic,
Adam E. Goldman-Yassen,
Virginia Hill,
Tiffany So,
Sedra Mhana,
Albara Alotaibi,
Nathan Page,
Prisha Bhatia,
Melisa S. Guelen
, et al. (219 additional authors not shown)
Abstract:
Despite continuous advancements in cancer treatment, brain metastatic disease remains a significant complication of primary cancer and is associated with an unfavorable prognosis. One approach for improving diagnosis, management, and outcomes is to implement algorithms based on artificial intelligence for the automated segmentation of both pre- and post-treatment MRI brain images. Such algorithms…
▽ More
Despite continuous advancements in cancer treatment, brain metastatic disease remains a significant complication of primary cancer and is associated with an unfavorable prognosis. One approach for improving diagnosis, management, and outcomes is to implement algorithms based on artificial intelligence for the automated segmentation of both pre- and post-treatment MRI brain images. Such algorithms rely on volumetric criteria for lesion identification and treatment response assessment, which are still not available in clinical practice. Therefore, it is critical to establish tools for rapid volumetric segmentations methods that can be translated to clinical practice and that are trained on high quality annotated data. The BraTS-METS 2025 Lighthouse Challenge aims to address this critical need by establishing inter-rater and intra-rater variability in dataset annotation by generating high quality annotated datasets from four individual instances of segmentation by neuroradiologists while being recorded on video (two instances doing "from scratch" and two instances after AI pre-segmentation). This high-quality annotated dataset will be used for testing phase in 2025 Lighthouse challenge and will be publicly released at the completion of the challenge. The 2025 Lighthouse challenge will also release the 2023 and 2024 segmented datasets that were annotated using an established pipeline of pre-segmentation, student annotation, two neuroradiologists checking, and one neuroradiologist finalizing the process. It builds upon its previous edition by including post-treatment cases in the dataset. Using these high-quality annotated datasets, the 2025 Lighthouse challenge plans to test benchmark algorithms for automated segmentation of pre-and post-treatment brain metastases (BM), trained on diverse and multi-institutional datasets of MRI images obtained from patients with brain metastases.
△ Less
Submitted 10 July, 2025; v1 submitted 16 April, 2025;
originally announced April 2025.
-
dyAb: Flow Matching for Flexible Antibody Design with AlphaFold-driven Pre-binding Antigen
Authors:
Cheng Tan,
Yijie Zhang,
Zhangyang Gao,
Yufei Huang,
Haitao Lin,
Lirong Wu,
Fandi Wu,
Mathieu Blanchette,
Stan. Z. Li
Abstract:
The development of therapeutic antibodies heavily relies on accurate predictions of how antigens will interact with antibodies. Existing computational methods in antibody design often overlook crucial conformational changes that antigens undergo during the binding process, significantly impacting the reliability of the resulting antibodies. To bridge this gap, we introduce dyAb, a flexible framewo…
▽ More
The development of therapeutic antibodies heavily relies on accurate predictions of how antigens will interact with antibodies. Existing computational methods in antibody design often overlook crucial conformational changes that antigens undergo during the binding process, significantly impacting the reliability of the resulting antibodies. To bridge this gap, we introduce dyAb, a flexible framework that incorporates AlphaFold2-driven predictions to model pre-binding antigen structures and specifically addresses the dynamic nature of antigen conformation changes. Our dyAb model leverages a unique combination of coarse-grained interface alignment and fine-grained flow matching techniques to simulate the interaction dynamics and structural evolution of the antigen-antibody complex, providing a realistic representation of the binding process. Extensive experiments show that dyAb significantly outperforms existing models in antibody design involving changing antigen conformations. These results highlight dyAb's potential to streamline the design process for therapeutic antibodies, promising more efficient development cycles and improved outcomes in clinical applications.
△ Less
Submitted 28 February, 2025;
originally announced March 2025.
-
Fast and Accurate Blind Flexible Docking
Authors:
Zizhuo Zhang,
Lijun Wu,
Kaiyuan Gao,
Jiangchao Yao,
Tao Qin,
Bo Han
Abstract:
Molecular docking that predicts the bound structures of small molecules (ligands) to their protein targets, plays a vital role in drug discovery. However, existing docking methods often face limitations: they either overlook crucial structural changes by assuming protein rigidity or suffer from low computational efficiency due to their reliance on generative models for structure sampling. To addre…
▽ More
Molecular docking that predicts the bound structures of small molecules (ligands) to their protein targets, plays a vital role in drug discovery. However, existing docking methods often face limitations: they either overlook crucial structural changes by assuming protein rigidity or suffer from low computational efficiency due to their reliance on generative models for structure sampling. To address these challenges, we propose FABFlex, a fast and accurate regression-based multi-task learning model designed for realistic blind flexible docking scenarios, where proteins exhibit flexibility and binding pocket sites are unknown (blind). Specifically, FABFlex's architecture comprises three specialized modules working in concert: (1) A pocket prediction module that identifies potential binding sites, addressing the challenges inherent in blind docking scenarios. (2) A ligand docking module that predicts the bound (holo) structures of ligands from their unbound (apo) states. (3) A pocket docking module that forecasts the holo structures of protein pockets from their apo conformations. Notably, FABFlex incorporates an iterative update mechanism that serves as a conduit between the ligand and pocket docking modules, enabling continuous structural refinements. This approach effectively integrates the three subtasks of blind flexible docking-pocket identification, ligand conformation prediction, and protein flexibility modeling-into a unified, coherent framework. Extensive experiments on public benchmark datasets demonstrate that FABFlex not only achieves superior effectiveness in predicting accurate binding modes but also exhibits a significant speed advantage (208 $\times$) compared to existing state-of-the-art methods. Our code is released at https://github.com/tmlr-group/FABFlex.
△ Less
Submitted 20 February, 2025;
originally announced February 2025.
-
A Simple yet Effective DDG Predictor is An Unsupervised Antibody Optimizer and Explainer
Authors:
Lirong Wu,
Yunfan Liu,
Haitao Lin,
Yufei Huang,
Guojiang Zhao,
Zhifeng Gao,
Stan Z. Li
Abstract:
The proteins that exist today have been optimized over billions of years of natural evolution, during which nature creates random mutations and selects them. The discovery of functionally promising mutations is challenged by the limited evolutionary accessible regions, i.e., only a small region on the fitness landscape is beneficial. There have been numerous priors used to constrain protein evolut…
▽ More
The proteins that exist today have been optimized over billions of years of natural evolution, during which nature creates random mutations and selects them. The discovery of functionally promising mutations is challenged by the limited evolutionary accessible regions, i.e., only a small region on the fitness landscape is beneficial. There have been numerous priors used to constrain protein evolution to regions of landscapes with high-fitness variants, among which the change in binding free energy (DDG) of protein complexes upon mutations is one of the most commonly used priors. However, the huge mutation space poses two challenges: (1) how to improve the efficiency of DDG prediction for fast mutation screening; and (2) how to explain mutation preferences and efficiently explore accessible evolutionary regions. To address these challenges, we propose a lightweight DDG predictor (Light-DDG), which adopts a structure-aware Transformer as the backbone and enhances it by knowledge distilled from existing powerful but computationally heavy DDG predictors. Additionally, we augmented, annotated, and released a large-scale dataset containing millions of mutation data for pre-training Light-DDG. We find that such a simple yet effective Light-DDG can serve as a good unsupervised antibody optimizer and explainer. For the target antibody, we propose a novel Mutation Explainer to learn mutation preferences, which accounts for the marginal benefit of each mutation per residue. To further explore accessible evolutionary regions, we conduct preference-guided antibody optimization and evaluate antibody candidates quickly using Light-DDG to identify desirable mutations.
△ Less
Submitted 13 February, 2025; v1 submitted 10 February, 2025;
originally announced February 2025.
-
Relation-Aware Equivariant Graph Networks for Epitope-Unknown Antibody Design and Specificity Optimization
Authors:
Lirong Wu,
Haitao Lin,
Yufei Huang,
Zhangyang Gao,
Cheng Tan,
Yunfan Liu,
Tailin Wu,
Stan Z. Li
Abstract:
Antibodies are Y-shaped proteins that protect the host by binding to specific antigens, and their binding is mainly determined by the Complementary Determining Regions (CDRs) in the antibody. Despite the great progress made in CDR design, existing computational methods still encounter several challenges: 1) poor capability of modeling complex CDRs with long sequences due to insufficient contextual…
▽ More
Antibodies are Y-shaped proteins that protect the host by binding to specific antigens, and their binding is mainly determined by the Complementary Determining Regions (CDRs) in the antibody. Despite the great progress made in CDR design, existing computational methods still encounter several challenges: 1) poor capability of modeling complex CDRs with long sequences due to insufficient contextual information; 2) conditioned on pre-given antigenic epitopes and their static interaction with the target antibody; 3) neglect of specificity during antibody optimization leads to non-specific antibodies. In this paper, we take into account a variety of node features, edge features, and edge relations to include more contextual and geometric information. We propose a novel Relation-Aware Antibody Design (RAAD) framework, which dynamically models antigen-antibody interactions for co-designing the sequences and structures of antigen-specific CDRs. Furthermore, we propose a new evaluation metric to better measure antibody specificity and develop a contrasting specificity-enhancing constraint to optimize the specificity of antibodies. Extensive experiments have demonstrated the superior capability of RAAD in terms of antibody modeling, generation, and optimization across different CDR types, sequence lengths, pre-training strategies, and input contexts.
△ Less
Submitted 13 December, 2024;
originally announced January 2025.
-
Tokenizing 3D Molecule Structure with Quantized Spherical Coordinates
Authors:
Kaiyuan Gao,
Yusong Wang,
Haoxiang Guan,
Zun Wang,
Qizhi Pei,
John E. Hopcroft,
Kun He,
Lijun Wu
Abstract:
The application of language models (LMs) to molecular structure generation using line notations such as SMILES and SELFIES has been well-established in the field of cheminformatics. However, extending these models to generate 3D molecular structures presents significant challenges. Two primary obstacles emerge: (1) the difficulty in designing a 3D line notation that ensures SE(3)-invariant atomic…
▽ More
The application of language models (LMs) to molecular structure generation using line notations such as SMILES and SELFIES has been well-established in the field of cheminformatics. However, extending these models to generate 3D molecular structures presents significant challenges. Two primary obstacles emerge: (1) the difficulty in designing a 3D line notation that ensures SE(3)-invariant atomic coordinates, and (2) the non-trivial task of tokenizing continuous coordinates for use in LMs, which inherently require discrete inputs. To address these challenges, we propose Mol-StrucTok, a novel method for tokenizing 3D molecular structures. Our approach comprises two key innovations: (1) We design a line notation for 3D molecules by extracting local atomic coordinates in a spherical coordinate system. This notation builds upon existing 2D line notations and remains agnostic to their specific forms, ensuring compatibility with various molecular representation schemes. (2) We employ a Vector Quantized Variational Autoencoder (VQ-VAE) to tokenize these coordinates, treating them as generation descriptors. To further enhance the representation, we incorporate neighborhood bond lengths and bond angles as understanding descriptors. Leveraging this tokenization framework, we train a GPT-2 style model for 3D molecular generation tasks. Results demonstrate strong performance with significantly faster generation speeds and competitive chemical stability compared to previous methods. Further, by integrating our learned discrete representations into Graphormer model for property prediction on QM9 dataset, Mol-StrucTok reveals consistent improvements across various molecular properties, underscoring the versatility and robustness of our approach.
△ Less
Submitted 2 December, 2024;
originally announced December 2024.
-
MeToken: Uniform Micro-environment Token Boosts Post-Translational Modification Prediction
Authors:
Cheng Tan,
Zhenxiao Cao,
Zhangyang Gao,
Lirong Wu,
Siyuan Li,
Yufei Huang,
Jun Xia,
Bozhen Hu,
Stan Z. Li
Abstract:
Post-translational modifications (PTMs) profoundly expand the complexity and functionality of the proteome, regulating protein attributes and interactions that are crucial for biological processes. Accurately predicting PTM sites and their specific types is therefore essential for elucidating protein function and understanding disease mechanisms. Existing computational approaches predominantly foc…
▽ More
Post-translational modifications (PTMs) profoundly expand the complexity and functionality of the proteome, regulating protein attributes and interactions that are crucial for biological processes. Accurately predicting PTM sites and their specific types is therefore essential for elucidating protein function and understanding disease mechanisms. Existing computational approaches predominantly focus on protein sequences to predict PTM sites, driven by the recognition of sequence-dependent motifs. However, these approaches often overlook protein structural contexts. In this work, we first compile a large-scale sequence-structure PTM dataset, which serves as the foundation for fair comparison. We introduce the MeToken model, which tokenizes the micro-environment of each amino acid, integrating both sequence and structural information into unified discrete tokens. This model not only captures the typical sequence motifs associated with PTMs but also leverages the spatial arrangements dictated by protein tertiary structures, thus providing a holistic view of the factors influencing PTM sites. Designed to address the long-tail distribution of PTM types, MeToken employs uniform sub-codebooks that ensure even the rarest PTMs are adequately represented and distinguished. We validate the effectiveness and generalizability of MeToken across multiple datasets, demonstrating its superior performance in accurately identifying PTM types. The results underscore the importance of incorporating structural data and highlight MeToken's potential in facilitating accurate and comprehensive PTM predictions, which could significantly impact proteomics research. The code and datasets are available at https://github.com/A4Bio/MeToken.
△ Less
Submitted 4 November, 2024;
originally announced November 2024.
-
SFM-Protein: Integrative Co-evolutionary Pre-training for Advanced Protein Sequence Representation
Authors:
Liang He,
Peiran Jin,
Yaosen Min,
Shufang Xie,
Lijun Wu,
Tao Qin,
Xiaozhuan Liang,
Kaiyuan Gao,
Yuliang Jiang,
Tie-Yan Liu
Abstract:
Proteins, essential to biological systems, perform functions intricately linked to their three-dimensional structures. Understanding the relationship between protein structures and their amino acid sequences remains a core challenge in protein modeling. While traditional protein foundation models benefit from pre-training on vast unlabeled datasets, they often struggle to capture critical co-evolu…
▽ More
Proteins, essential to biological systems, perform functions intricately linked to their three-dimensional structures. Understanding the relationship between protein structures and their amino acid sequences remains a core challenge in protein modeling. While traditional protein foundation models benefit from pre-training on vast unlabeled datasets, they often struggle to capture critical co-evolutionary information, which evolutionary-based methods excel at. In this study, we introduce a novel pre-training strategy for protein foundation models that emphasizes the interactions among amino acid residues to enhance the extraction of both short-range and long-range co-evolutionary features from sequence data. Trained on a large-scale protein sequence dataset, our model demonstrates superior generalization ability, outperforming established baselines of similar size, including the ESM model, across diverse downstream tasks. Experimental results confirm the model's effectiveness in integrating co-evolutionary information, marking a significant step forward in protein sequence-based modeling.
△ Less
Submitted 31 October, 2024;
originally announced October 2024.
-
Exploiting Pre-trained Models for Drug Target Affinity Prediction with Nearest Neighbors
Authors:
Qizhi Pei,
Lijun Wu,
Zhenyu He,
Jinhua Zhu,
Yingce Xia,
Shufang Xie,
Rui Yan
Abstract:
Drug-Target binding Affinity (DTA) prediction is essential for drug discovery. Despite the application of deep learning methods to DTA prediction, the achieved accuracy remain suboptimal. In this work, inspired by the recent success of retrieval methods, we propose $k$NN-DTA, a non-parametric embedding-based retrieval method adopted on a pre-trained DTA prediction model, which can extend the power…
▽ More
Drug-Target binding Affinity (DTA) prediction is essential for drug discovery. Despite the application of deep learning methods to DTA prediction, the achieved accuracy remain suboptimal. In this work, inspired by the recent success of retrieval methods, we propose $k$NN-DTA, a non-parametric embedding-based retrieval method adopted on a pre-trained DTA prediction model, which can extend the power of the DTA model with no or negligible cost. Different from existing methods, we introduce two neighbor aggregation ways from both embedding space and label space that are integrated into a unified framework. Specifically, we propose a \emph{label aggregation} with \emph{pair-wise retrieval} and a \emph{representation aggregation} with \emph{point-wise retrieval} of the nearest neighbors. This method executes in the inference phase and can efficiently boost the DTA prediction performance with no training cost. In addition, we propose an extension, Ada-$k$NN-DTA, an instance-wise and adaptive aggregation with lightweight learning. Results on four benchmark datasets show that $k$NN-DTA brings significant improvements, outperforming previous state-of-the-art (SOTA) results, e.g, on BindingDB IC$_{50}$ and $K_i$ testbeds, $k$NN-DTA obtains new records of RMSE $\bf{0.684}$ and $\bf{0.750}$. The extended Ada-$k$NN-DTA further improves the performance to be $\bf{0.675}$ and $\bf{0.735}$ RMSE. These results strongly prove the effectiveness of our method. Results in other settings and comprehensive studies/analyses also show the great potential of our $k$NN-DTA approach.
△ Less
Submitted 21 July, 2024;
originally announced July 2024.
-
Diffusion-driven self-assembly of emerin nanodomains at the nuclear envelope
Authors:
Carlos D. Alas,
Liying Wu,
Fabien Pinaud,
Christoph A. Haselwandter
Abstract:
Emerin, a nuclear membrane protein with important biological roles in mechanotransduction and nuclear shape adaptation, self-assembles into nanometer-size domains at the inner nuclear membrane. The size and emerin occupancy of these nanodomains change with applied mechanical stress as well as under emerin mutations associated with Emery-Dreifuss muscular dystrophy (EDMD). Through a combination of…
▽ More
Emerin, a nuclear membrane protein with important biological roles in mechanotransduction and nuclear shape adaptation, self-assembles into nanometer-size domains at the inner nuclear membrane. The size and emerin occupancy of these nanodomains change with applied mechanical stress as well as under emerin mutations associated with Emery-Dreifuss muscular dystrophy (EDMD). Through a combination of theory and experiment we show here that a simple reaction-diffusion model explains the self-assembly of emerin nanodomains. Our model yields quantitative agreement with experimental observations on the size and occupancy of emerin nanodomains for wild-type emerin and EDMD-associated mutations of emerin, with and without applied forces, and allows successful prediction of emerin diffusion coefficients from observations on the overall properties of emerin nanodomains. Our results provide a physical understanding of EDMD-associated defects in emerin organization in terms of changes in key reaction and diffusion properties of emerin and its nuclear binding partners.
△ Less
Submitted 16 July, 2024;
originally announced July 2024.
-
CBGBench: Fill in the Blank of Protein-Molecule Complex Binding Graph
Authors:
Haitao Lin,
Guojiang Zhao,
Odin Zhang,
Yufei Huang,
Lirong Wu,
Zicheng Liu,
Siyuan Li,
Cheng Tan,
Zhifeng Gao,
Stan Z. Li
Abstract:
Structure-based drug design (SBDD) aims to generate potential drugs that can bind to a target protein and is greatly expedited by the aid of AI techniques in generative models. However, a lack of systematic understanding persists due to the diverse settings, complex implementation, difficult reproducibility, and task singularity. Firstly, the absence of standardization can lead to unfair compariso…
▽ More
Structure-based drug design (SBDD) aims to generate potential drugs that can bind to a target protein and is greatly expedited by the aid of AI techniques in generative models. However, a lack of systematic understanding persists due to the diverse settings, complex implementation, difficult reproducibility, and task singularity. Firstly, the absence of standardization can lead to unfair comparisons and inconclusive insights. To address this dilemma, we propose CBGBench, a comprehensive benchmark for SBDD, that unifies the task as a generative heterogeneous graph completion, analogous to fill-in-the-blank of the 3D complex binding graph. By categorizing existing methods based on their attributes, CBGBench facilitates a modular and extensible framework that implements various cutting-edge methods. Secondly, a single task on \textit{de novo} molecule generation can hardly reflect their capabilities. To broaden the scope, we have adapted these models to a range of tasks essential in drug design, which are considered sub-tasks within the graph fill-in-the-blank tasks. These tasks include the generative designation of \textit{de novo} molecules, linkers, fragments, scaffolds, and sidechains, all conditioned on the structures of protein pockets. Our evaluations are conducted with fairness, encompassing comprehensive perspectives on interaction, chemical properties, geometry authenticity, and substructure validity. We further provide the pre-trained versions of the state-of-the-art models and deep insights with analysis from empirical studies. The codebase for CBGBench is publicly accessible at \url{https://github.com/Edapinenut/CBGBench}.
△ Less
Submitted 10 October, 2024; v1 submitted 16 June, 2024;
originally announced June 2024.
-
3D-MolT5: Leveraging Discrete Structural Information for Molecule-Text Modeling
Authors:
Qizhi Pei,
Rui Yan,
Kaiyuan Gao,
Jinhua Zhu,
Lijun Wu
Abstract:
The integration of molecular and natural language representations has emerged as a focal point in molecular science, with recent advancements in Language Models (LMs) demonstrating significant potential for comprehensive modeling of both domains. However, existing approaches face notable limitations, particularly in their neglect of three-dimensional (3D) information, which is crucial for understa…
▽ More
The integration of molecular and natural language representations has emerged as a focal point in molecular science, with recent advancements in Language Models (LMs) demonstrating significant potential for comprehensive modeling of both domains. However, existing approaches face notable limitations, particularly in their neglect of three-dimensional (3D) information, which is crucial for understanding molecular structures and functions. While some efforts have been made to incorporate 3D molecular information into LMs using external structure encoding modules, significant difficulties remain, such as insufficient interaction across modalities in pre-training and challenges in modality alignment. To address the limitations, we propose \textbf{3D-MolT5}, a unified framework designed to model molecule in both sequence and 3D structure spaces. The key innovation of our approach lies in mapping fine-grained 3D substructure representations into a specialized 3D token vocabulary. This methodology facilitates the seamless integration of sequence and structure representations in a tokenized format, enabling 3D-MolT5 to encode molecular sequences, molecular structures, and text sequences within a unified architecture. Leveraging this tokenized input strategy, we build a foundation model that unifies the sequence and structure data formats. We then conduct joint pre-training with multi-task objectives to enhance the model's comprehension of these diverse modalities within a shared representation space. Thus, our approach significantly improves cross-modal interaction and alignment, addressing key challenges in previous work. Further instruction tuning demonstrated that our 3D-MolT5 has strong generalization ability and surpasses existing methods with superior performance in multiple downstream tasks. Our code is available at https://github.com/QizhiPei/3D-MolT5.
△ Less
Submitted 18 March, 2025; v1 submitted 9 June, 2024;
originally announced June 2024.
-
UniIF: Unified Molecule Inverse Folding
Authors:
Zhangyang Gao,
Jue Wang,
Cheng Tan,
Lirong Wu,
Yufei Huang,
Siyuan Li,
Zhirui Ye,
Stan Z. Li
Abstract:
Molecule inverse folding has been a long-standing challenge in chemistry and biology, with the potential to revolutionize drug discovery and material science. Despite specified models have been proposed for different small- or macro-molecules, few have attempted to unify the learning process, resulting in redundant efforts. Complementary to recent advancements in molecular structure prediction, su…
▽ More
Molecule inverse folding has been a long-standing challenge in chemistry and biology, with the potential to revolutionize drug discovery and material science. Despite specified models have been proposed for different small- or macro-molecules, few have attempted to unify the learning process, resulting in redundant efforts. Complementary to recent advancements in molecular structure prediction, such as RoseTTAFold All-Atom and AlphaFold3, we propose the unified model UniIF for the inverse folding of all molecules. We do such unification in two levels: 1) Data-Level: We propose a unified block graph data form for all molecules, including the local frame building and geometric feature initialization. 2) Model-Level: We introduce a geometric block attention network, comprising a geometric interaction, interactive attention and virtual long-term dependency modules, to capture the 3D interactions of all molecules. Through comprehensive evaluations across various tasks such as protein design, RNA design, and material design, we demonstrate that our proposed method surpasses state-of-the-art methods on all tasks. UniIF offers a versatile and effective solution for general molecule inverse folding.
△ Less
Submitted 29 May, 2024;
originally announced May 2024.
-
Learning to Predict Mutation Effects of Protein-Protein Interactions by Microenvironment-aware Hierarchical Prompt Learning
Authors:
Lirong Wu,
Yijun Tian,
Haitao Lin,
Yufei Huang,
Siyuan Li,
Nitesh V Chawla,
Stan Z. Li
Abstract:
Protein-protein bindings play a key role in a variety of fundamental biological processes, and thus predicting the effects of amino acid mutations on protein-protein binding is crucial. To tackle the scarcity of annotated mutation data, pre-training with massive unlabeled data has emerged as a promising solution. However, this process faces a series of challenges: (1) complex higher-order dependen…
▽ More
Protein-protein bindings play a key role in a variety of fundamental biological processes, and thus predicting the effects of amino acid mutations on protein-protein binding is crucial. To tackle the scarcity of annotated mutation data, pre-training with massive unlabeled data has emerged as a promising solution. However, this process faces a series of challenges: (1) complex higher-order dependencies among multiple (more than paired) structural scales have not yet been fully captured; (2) it is rarely explored how mutations alter the local conformation of the surrounding microenvironment; (3) pre-training is costly, both in data size and computational burden. In this paper, we first construct a hierarchical prompt codebook to record common microenvironmental patterns at different structural scales independently. Then, we develop a novel codebook pre-training task, namely masked microenvironment modeling, to model the joint distribution of each mutation with their residue types, angular statistics, and local conformational changes in the microenvironment. With the constructed prompt codebook, we encode the microenvironment around each mutation into multiple hierarchical prompts and combine them to flexibly provide information to wild-type and mutated protein complexes about their microenvironmental differences. Such a hierarchical prompt learning framework has demonstrated superior performance and training efficiency over state-of-the-art pre-training-based methods in mutation effect prediction and a case study of optimizing human antibodies against SARS-CoV-2.
△ Less
Submitted 15 May, 2024;
originally announced May 2024.
-
PPFlow: Target-aware Peptide Design with Torsional Flow Matching
Authors:
Haitao Lin,
Odin Zhang,
Huifeng Zhao,
Dejun Jiang,
Lirong Wu,
Zicheng Liu,
Yufei Huang,
Stan Z. Li
Abstract:
Therapeutic peptides have proven to have great pharmaceutical value and potential in recent decades. However, methods of AI-assisted peptide drug discovery are not fully explored. To fill the gap, we propose a target-aware peptide design method called \textsc{PPFlow}, based on conditional flow matching on torus manifolds, to model the internal geometries of torsion angles for the peptide structure…
▽ More
Therapeutic peptides have proven to have great pharmaceutical value and potential in recent decades. However, methods of AI-assisted peptide drug discovery are not fully explored. To fill the gap, we propose a target-aware peptide design method called \textsc{PPFlow}, based on conditional flow matching on torus manifolds, to model the internal geometries of torsion angles for the peptide structure design. Besides, we establish a protein-peptide binding dataset named PPBench2024 to fill the void of massive data for the task of structure-based peptide drug design and to allow the training of deep learning methods. Extensive experiments show that PPFlow reaches state-of-the-art performance in tasks of peptide drug generation and optimization in comparison with baseline models, and can be generalized to other tasks including docking and side-chain packing.
△ Less
Submitted 9 December, 2024; v1 submitted 5 March, 2024;
originally announced May 2024.
-
FABind+: Enhancing Molecular Docking through Improved Pocket Prediction and Pose Generation
Authors:
Kaiyuan Gao,
Qizhi Pei,
Gongbo Zhang,
Jinhua Zhu,
Kun He,
Lijun Wu
Abstract:
Molecular docking is a pivotal process in drug discovery. While traditional techniques rely on extensive sampling and simulation governed by physical principles, these methods are often slow and costly. The advent of deep learning-based approaches has shown significant promise, offering increases in both accuracy and efficiency. Building upon the foundational work of FABind, a model designed with…
▽ More
Molecular docking is a pivotal process in drug discovery. While traditional techniques rely on extensive sampling and simulation governed by physical principles, these methods are often slow and costly. The advent of deep learning-based approaches has shown significant promise, offering increases in both accuracy and efficiency. Building upon the foundational work of FABind, a model designed with a focus on speed and accuracy, we present FABind+, an enhanced iteration that largely boosts the performance of its predecessor. We identify pocket prediction as a critical bottleneck in molecular docking and propose a novel methodology that significantly refines pocket prediction, thereby streamlining the docking process. Furthermore, we introduce modifications to the docking module to enhance its pose generation capabilities. In an effort to bridge the gap with conventional sampling/generative methods, we incorporate a simple yet effective sampling technique coupled with a confidence model, requiring only minor adjustments to the regression framework of FABind. Experimental results and analysis reveal that FABind+ remarkably outperforms the original FABind, achieves competitive state-of-the-art performance, and delivers insightful modeling strategies. This demonstrates FABind+ represents a substantial step forward in molecular docking and drug discovery. Our code is in https://github.com/QizhiPei/FABind.
△ Less
Submitted 24 February, 2025; v1 submitted 29 March, 2024;
originally announced March 2024.
-
FoldToken: Learning Protein Language via Vector Quantization and Beyond
Authors:
Zhangyang Gao,
Cheng Tan,
Jue Wang,
Yufei Huang,
Lirong Wu,
Stan Z. Li
Abstract:
Is there a foreign language describing protein sequences and structures simultaneously? Protein structures, represented by continuous 3D points, have long posed a challenge due to the contrasting modeling paradigms of discrete sequences. We introduce \textbf{FoldTokenizer} to represent protein sequence-structure as discrete symbols. This innovative approach involves projecting residue types and st…
▽ More
Is there a foreign language describing protein sequences and structures simultaneously? Protein structures, represented by continuous 3D points, have long posed a challenge due to the contrasting modeling paradigms of discrete sequences. We introduce \textbf{FoldTokenizer} to represent protein sequence-structure as discrete symbols. This innovative approach involves projecting residue types and structures into a discrete space, guided by a reconstruction loss for information preservation. We refer to the learned discrete symbols as \textbf{FoldToken}, and the sequence of FoldTokens serves as a new protein language, transforming the protein sequence-structure into a unified modality. We apply the created protein language on general backbone inpainting and antibody design tasks, building the first GPT-style model (\textbf{FoldGPT}) for sequence-structure co-generation with promising results. Key to our success is the substantial enhancement of the vector quantization module, Soft Conditional Vector Quantization (\textbf{SoftCVQ}).
△ Less
Submitted 19 March, 2024; v1 submitted 4 February, 2024;
originally announced March 2024.
-
Advances of Deep Learning in Protein Science: A Comprehensive Survey
Authors:
Bozhen Hu,
Cheng Tan,
Lirong Wu,
Jiangbin Zheng,
Jun Xia,
Zhangyang Gao,
Zicheng Liu,
Fandi Wu,
Guijun Zhang,
Stan Z. Li
Abstract:
Protein representation learning plays a crucial role in understanding the structure and function of proteins, which are essential biomolecules involved in various biological processes. In recent years, deep learning has emerged as a powerful tool for protein modeling due to its ability to learn complex patterns and representations from large-scale protein data. This comprehensive survey aims to pr…
▽ More
Protein representation learning plays a crucial role in understanding the structure and function of proteins, which are essential biomolecules involved in various biological processes. In recent years, deep learning has emerged as a powerful tool for protein modeling due to its ability to learn complex patterns and representations from large-scale protein data. This comprehensive survey aims to provide an overview of the recent advances in deep learning techniques applied to protein science. The survey begins by introducing the developments of deep learning based protein models and emphasizes the importance of protein representation learning in drug discovery, protein engineering, and function annotation. It then delves into the fundamentals of deep learning, including convolutional neural networks, recurrent neural networks, attention models, and graph neural networks in modeling protein sequences, structures, and functions, and explores how these techniques can be used to extract meaningful features and capture intricate relationships within protein data. Next, the survey presents various applications of deep learning in the field of proteins, including protein structure prediction, protein-protein interaction prediction, protein function prediction, etc. Furthermore, it highlights the challenges and limitations of these deep learning techniques and also discusses potential solutions and future directions for overcoming these challenges. This comprehensive survey provides a valuable resource for researchers and practitioners in the field of proteins who are interested in harnessing the power of deep learning techniques. By consolidating the latest advancements and discussing potential avenues for improvement, this review contributes to the ongoing progress in protein research and paves the way for future breakthroughs in the field.
△ Less
Submitted 8 March, 2024;
originally announced March 2024.
-
Renal function changes in chronic hepatitis B patients
Authors:
Jinhua Zhao,
Lili Wu,
Xiaoan Yang,
Zhilaing Gao,
Hong Deng
Abstract:
The best way to treat chronic hepatitis B is with pegylated interferon alone or with oral antiviral drugs. There is limited research comparing the renal safety of entecavir and tenofovir when used with pegylated interferon. This study will compare changes in renal function in chronic hepatitis B patients treated with pegylated interferon and either entecavir or tenofovir. The study included a coho…
▽ More
The best way to treat chronic hepatitis B is with pegylated interferon alone or with oral antiviral drugs. There is limited research comparing the renal safety of entecavir and tenofovir when used with pegylated interferon. This study will compare changes in renal function in chronic hepatitis B patients treated with pegylated interferon and either entecavir or tenofovir. The study included a cohort of 836 patients with chronic hepatitis B (CHB) who received treatment with pegylated interferon (IFN) either alone or in combination with entecavir (ETV) and tenofovir (TDF) between the years 2018 and 2021. Of these patients, 713 were included in a matched analysis comparing outcomes between those who were cured and those who were uncured, while 123 patients received IFN alone as a control group for comparison with the ETV and TDF treatment groups. The primary outcome measured was the change in renal function, specifically estimated glomerular filtration rate (eGFR), cystatin C (CysC), and inorganic phosphorus (IPHOS). Patients were categorized into stage 1 or stage 2 based on a baseline eGFR of less than 90 ml/min/m^2 Results: 125 CHB patients were matched 1:1 in both the combined treatment and cured groups. Baseline eGFR, CysC, and IPHOS levels were similar between the groups. Renal function in stage 1 and stage 2 groups showed a decreasing trend at 48 weeks after an initial increase.Correlation analysis showed significant relationships between changes in ALT and eGFR values at 12 weeks in both non-cured and cured groups. Conclusions: Over the 48-week duration of combined treatment in patients with chronic hepatitis B (CHB), it was found that both Tenofovir Disoproxil Fumarate (TDF) and Entecavir (ETV) did not lead to an increase in renal injury.
△ Less
Submitted 4 March, 2024;
originally announced March 2024.
-
Leveraging Biomolecule and Natural Language through Multi-Modal Learning: A Survey
Authors:
Qizhi Pei,
Lijun Wu,
Kaiyuan Gao,
Jinhua Zhu,
Yue Wang,
Zun Wang,
Tao Qin,
Rui Yan
Abstract:
The integration of biomolecular modeling with natural language (BL) has emerged as a promising interdisciplinary area at the intersection of artificial intelligence, chemistry and biology. This approach leverages the rich, multifaceted descriptions of biomolecules contained within textual data sources to enhance our fundamental understanding and enable downstream computational tasks such as biomol…
▽ More
The integration of biomolecular modeling with natural language (BL) has emerged as a promising interdisciplinary area at the intersection of artificial intelligence, chemistry and biology. This approach leverages the rich, multifaceted descriptions of biomolecules contained within textual data sources to enhance our fundamental understanding and enable downstream computational tasks such as biomolecule property prediction. The fusion of the nuanced narratives expressed through natural language with the structural and functional specifics of biomolecules described via various molecular modeling techniques opens new avenues for comprehensively representing and analyzing biomolecules. By incorporating the contextual language data that surrounds biomolecules into their modeling, BL aims to capture a holistic view encompassing both the symbolic qualities conveyed through language as well as quantitative structural characteristics. In this review, we provide an extensive analysis of recent advancements achieved through cross modeling of biomolecules and natural language. (1) We begin by outlining the technical representations of biomolecules employed, including sequences, 2D graphs, and 3D structures. (2) We then examine in depth the rationale and key objectives underlying effective multi-modal integration of language and molecular data sources. (3) We subsequently survey the practical applications enabled to date in this developing research area. (4) We also compile and summarize the available resources and datasets to facilitate future work. (5) Looking ahead, we identify several promising research directions worthy of further exploration and investment to continue advancing the field. The related resources and contents are updating in \url{https://github.com/QizhiPei/Awesome-Biomolecule-Language-Cross-Modeling}.
△ Less
Submitted 5 March, 2024; v1 submitted 3 March, 2024;
originally announced March 2024.
-
Enhancing Protein Predictive Models via Proteins Data Augmentation: A Benchmark and New Directions
Authors:
Rui Sun,
Lirong Wu,
Haitao Lin,
Yufei Huang,
Stan Z. Li
Abstract:
Augmentation is an effective alternative to utilize the small amount of labeled protein data. However, most of the existing work focuses on design-ing new architectures or pre-training tasks, and relatively little work has studied data augmentation for proteins. This paper extends data augmentation techniques previously used for images and texts to proteins and then benchmarks these techniques on…
▽ More
Augmentation is an effective alternative to utilize the small amount of labeled protein data. However, most of the existing work focuses on design-ing new architectures or pre-training tasks, and relatively little work has studied data augmentation for proteins. This paper extends data augmentation techniques previously used for images and texts to proteins and then benchmarks these techniques on a variety of protein-related tasks, providing the first comprehensive evaluation of protein augmentation. Furthermore, we propose two novel semantic-level protein augmentation methods, namely Integrated Gradients Substitution and Back Translation Substitution, which enable protein semantic-aware augmentation through saliency detection and biological knowledge. Finally, we integrate extended and proposed augmentations into an augmentation pool and propose a simple but effective framework, namely Automated Protein Augmentation (APA), which can adaptively select the most suitable augmentation combinations for different tasks. Extensive experiments have shown that APA enhances the performance of five protein related tasks by an average of 10.55% across three architectures compared to vanilla implementations without augmentation, highlighting its potential to make a great impact on the field.
△ Less
Submitted 1 March, 2024;
originally announced March 2024.
-
BioT5+: Towards Generalized Biological Understanding with IUPAC Integration and Multi-task Tuning
Authors:
Qizhi Pei,
Lijun Wu,
Kaiyuan Gao,
Xiaozhuan Liang,
Yin Fang,
Jinhua Zhu,
Shufang Xie,
Tao Qin,
Rui Yan
Abstract:
Recent research trends in computational biology have increasingly focused on integrating text and bio-entity modeling, especially in the context of molecules and proteins. However, previous efforts like BioT5 faced challenges in generalizing across diverse tasks and lacked a nuanced understanding of molecular structures, particularly in their textual representations (e.g., IUPAC). This paper intro…
▽ More
Recent research trends in computational biology have increasingly focused on integrating text and bio-entity modeling, especially in the context of molecules and proteins. However, previous efforts like BioT5 faced challenges in generalizing across diverse tasks and lacked a nuanced understanding of molecular structures, particularly in their textual representations (e.g., IUPAC). This paper introduces BioT5+, an extension of the BioT5 framework, tailored to enhance biological research and drug discovery. BioT5+ incorporates several novel features: integration of IUPAC names for molecular understanding, inclusion of extensive bio-text and molecule data from sources like bioRxiv and PubChem, the multi-task instruction tuning for generality across tasks, and a numerical tokenization technique for improved processing of numerical data. These enhancements allow BioT5+ to bridge the gap between molecular representations and their textual descriptions, providing a more holistic understanding of biological entities, and largely improving the grounded reasoning of bio-text and bio-sequences. The model is pre-trained and fine-tuned with a large number of experiments, including \emph{3 types of problems (classification, regression, generation), 15 kinds of tasks, and 21 total benchmark datasets}, demonstrating the remarkable performance and state-of-the-art results in most cases. BioT5+ stands out for its ability to capture intricate relationships in biological data, thereby contributing significantly to bioinformatics and computational biology. Our code is available at \url{https://github.com/QizhiPei/BioT5}.
△ Less
Submitted 31 May, 2024; v1 submitted 27 February, 2024;
originally announced February 2024.
-
MAPE-PPI: Towards Effective and Efficient Protein-Protein Interaction Prediction via Microenvironment-Aware Protein Embedding
Authors:
Lirong Wu,
Yijun Tian,
Yufei Huang,
Siyuan Li,
Haitao Lin,
Nitesh V Chawla,
Stan Z. Li
Abstract:
Protein-Protein Interactions (PPIs) are fundamental in various biological processes and play a key role in life activities. The growing demand and cost of experimental PPI assays require computational methods for efficient PPI prediction. While existing methods rely heavily on protein sequence for PPI prediction, it is the protein structure that is the key to determine the interactions. To take bo…
▽ More
Protein-Protein Interactions (PPIs) are fundamental in various biological processes and play a key role in life activities. The growing demand and cost of experimental PPI assays require computational methods for efficient PPI prediction. While existing methods rely heavily on protein sequence for PPI prediction, it is the protein structure that is the key to determine the interactions. To take both protein modalities into account, we define the microenvironment of an amino acid residue by its sequence and structural contexts, which describe the surrounding chemical properties and geometric features. In addition, microenvironments defined in previous work are largely based on experimentally assayed physicochemical properties, for which the "vocabulary" is usually extremely small. This makes it difficult to cover the diversity and complexity of microenvironments. In this paper, we propose Microenvironment-Aware Protein Embedding for PPI prediction (MPAE-PPI), which encodes microenvironments into chemically meaningful discrete codes via a sufficiently large microenvironment "vocabulary" (i.e., codebook). Moreover, we propose a novel pre-training strategy, namely Masked Codebook Modeling (MCM), to capture the dependencies between different microenvironments by randomly masking the codebook and reconstructing the input. With the learned microenvironment codebook, we can reuse it as an off-the-shelf tool to efficiently and effectively encode proteins of different sizes and functions for large-scale PPI prediction. Extensive experiments show that MAPE-PPI can scale to PPI prediction with millions of PPIs with superior trade-offs between effectiveness and computational efficiency than the state-of-the-art competitors.
△ Less
Submitted 22 February, 2024;
originally announced February 2024.
-
Re-Dock: Towards Flexible and Realistic Molecular Docking with Diffusion Bridge
Authors:
Yufei Huang,
Odin Zhang,
Lirong Wu,
Cheng Tan,
Haitao Lin,
Zhangyang Gao,
Siyuan Li,
Stan. Z. Li
Abstract:
Accurate prediction of protein-ligand binding structures, a task known as molecular docking is crucial for drug design but remains challenging. While deep learning has shown promise, existing methods often depend on holo-protein structures (docked, and not accessible in realistic tasks) or neglect pocket sidechain conformations, leading to limited practical utility and unrealistic conformation pre…
▽ More
Accurate prediction of protein-ligand binding structures, a task known as molecular docking is crucial for drug design but remains challenging. While deep learning has shown promise, existing methods often depend on holo-protein structures (docked, and not accessible in realistic tasks) or neglect pocket sidechain conformations, leading to limited practical utility and unrealistic conformation predictions. To fill these gaps, we introduce an under-explored task, named flexible docking to predict poses of ligand and pocket sidechains simultaneously and introduce Re-Dock, a novel diffusion bridge generative model extended to geometric manifolds. Specifically, we propose energy-to-geometry mapping inspired by the Newton-Euler equation to co-model the binding energy and conformations for reflecting the energy-constrained docking generative process. Comprehensive experiments on designed benchmark datasets including apo-dock and cross-dock demonstrate our model's superior effectiveness and efficiency over current methods.
△ Less
Submitted 21 February, 2024; v1 submitted 18 February, 2024;
originally announced February 2024.
-
PSC-CPI: Multi-Scale Protein Sequence-Structure Contrasting for Efficient and Generalizable Compound-Protein Interaction Prediction
Authors:
Lirong Wu,
Yufei Huang,
Cheng Tan,
Zhangyang Gao,
Bozhen Hu,
Haitao Lin,
Zicheng Liu,
Stan Z. Li
Abstract:
Compound-Protein Interaction (CPI) prediction aims to predict the pattern and strength of compound-protein interactions for rational drug discovery. Existing deep learning-based methods utilize only the single modality of protein sequences or structures and lack the co-modeling of the joint distribution of the two modalities, which may lead to significant performance drops in complex real-world sc…
▽ More
Compound-Protein Interaction (CPI) prediction aims to predict the pattern and strength of compound-protein interactions for rational drug discovery. Existing deep learning-based methods utilize only the single modality of protein sequences or structures and lack the co-modeling of the joint distribution of the two modalities, which may lead to significant performance drops in complex real-world scenarios due to various factors, e.g., modality missing and domain shifting. More importantly, these methods only model protein sequences and structures at a single fixed scale, neglecting more fine-grained multi-scale information, such as those embedded in key protein fragments. In this paper, we propose a novel multi-scale Protein Sequence-structure Contrasting framework for CPI prediction (PSC-CPI), which captures the dependencies between protein sequences and structures through both intra-modality and cross-modality contrasting. We further apply length-variable protein augmentation to allow contrasting to be performed at different scales, from the amino acid level to the sequence level. Finally, in order to more fairly evaluate the model generalizability, we split the test data into four settings based on whether compounds and proteins have been observed during the training stage. Extensive experiments have shown that PSC-CPI generalizes well in all four settings, particularly in the more challenging ``Unseen-Both" setting, where neither compounds nor proteins have been observed during training. Furthermore, even when encountering a situation of modality missing, i.e., inference with only single-modality protein data, PSC-CPI still exhibits comparable or even better performance than previous approaches.
△ Less
Submitted 12 February, 2024;
originally announced February 2024.
-
De novo protein design using geometric vector field networks
Authors:
Weian Mao,
Muzhi Zhu,
Zheng Sun,
Shuaike Shen,
Lin Yuanbo Wu,
Hao Chen,
Chunhua Shen
Abstract:
Innovations like protein diffusion have enabled significant progress in de novo protein design, which is a vital topic in life science. These methods typically depend on protein structure encoders to model residue backbone frames, where atoms do not exist. Most prior encoders rely on atom-wise features, such as angles and distances between atoms, which are not available in this context. Thus far,…
▽ More
Innovations like protein diffusion have enabled significant progress in de novo protein design, which is a vital topic in life science. These methods typically depend on protein structure encoders to model residue backbone frames, where atoms do not exist. Most prior encoders rely on atom-wise features, such as angles and distances between atoms, which are not available in this context. Thus far, only several simple encoders, such as IPA, have been proposed for this scenario, exposing the frame modeling as a bottleneck. In this work, we proffer the Vector Field Network (VFN), which enables network layers to perform learnable vector computations between coordinates of frame-anchored virtual atoms, thus achieving a higher capability for modeling frames. The vector computation operates in a manner similar to a linear layer, with each input channel receiving 3D virtual atom coordinates instead of scalar values. The multiple feature vectors output by the vector computation are then used to update the residue representations and virtual atom coordinates via attention aggregation. Remarkably, VFN also excels in modeling both frames and atoms, as the real atoms can be treated as the virtual atoms for modeling, positioning VFN as a potential universal encoder. In protein diffusion (frame modeling), VFN exhibits an impressive performance advantage over IPA, excelling in terms of both designability (67.04% vs. 53.58%) and diversity (66.54% vs. 51.98%). In inverse folding (frame and atom modeling), VFN outperforms the previous SoTA model, PiFold (54.7% vs. 51.66%), on sequence recovery rate. We also propose a method of equipping VFN with the ESM model, which significantly surpasses the previous ESM-based SoTA (62.67% vs. 55.65%), LM-Design, by a substantial margin.
△ Less
Submitted 18 October, 2023;
originally announced October 2023.
-
Protein 3D Graph Structure Learning for Robust Structure-based Protein Property Prediction
Authors:
Yufei Huang,
Siyuan Li,
Jin Su,
Lirong Wu,
Odin Zhang,
Haitao Lin,
Jingqi Qi,
Zihan Liu,
Zhangyang Gao,
Yuyang Liu,
Jiangbin Zheng,
Stan. ZQ. Li
Abstract:
Protein structure-based property prediction has emerged as a promising approach for various biological tasks, such as protein function prediction and sub-cellular location estimation. The existing methods highly rely on experimental protein structure data and fail in scenarios where these data are unavailable. Predicted protein structures from AI tools (e.g., AlphaFold2) were utilized as alternati…
▽ More
Protein structure-based property prediction has emerged as a promising approach for various biological tasks, such as protein function prediction and sub-cellular location estimation. The existing methods highly rely on experimental protein structure data and fail in scenarios where these data are unavailable. Predicted protein structures from AI tools (e.g., AlphaFold2) were utilized as alternatives. However, we observed that current practices, which simply employ accurately predicted structures during inference, suffer from notable degradation in prediction accuracy. While similar phenomena have been extensively studied in general fields (e.g., Computer Vision) as model robustness, their impact on protein property prediction remains unexplored. In this paper, we first investigate the reason behind the performance decrease when utilizing predicted structures, attributing it to the structure embedding bias from the perspective of structure representation learning. To study this problem, we identify a Protein 3D Graph Structure Learning Problem for Robust Protein Property Prediction (PGSL-RP3), collect benchmark datasets, and present a protein Structure embedding Alignment Optimization framework (SAO) to mitigate the problem of structure embedding bias between the predicted and experimental protein structures. Extensive experiments have shown that our framework is model-agnostic and effective in improving the property prediction of both predicted structures and experimental structures. The benchmark datasets and codes will be released to benefit the community.
△ Less
Submitted 19 October, 2023; v1 submitted 14 October, 2023;
originally announced October 2023.
-
BioT5: Enriching Cross-modal Integration in Biology with Chemical Knowledge and Natural Language Associations
Authors:
Qizhi Pei,
Wei Zhang,
Jinhua Zhu,
Kehan Wu,
Kaiyuan Gao,
Lijun Wu,
Yingce Xia,
Rui Yan
Abstract:
Recent advancements in biological research leverage the integration of molecules, proteins, and natural language to enhance drug discovery. However, current models exhibit several limitations, such as the generation of invalid molecular SMILES, underutilization of contextual information, and equal treatment of structured and unstructured knowledge. To address these issues, we propose…
▽ More
Recent advancements in biological research leverage the integration of molecules, proteins, and natural language to enhance drug discovery. However, current models exhibit several limitations, such as the generation of invalid molecular SMILES, underutilization of contextual information, and equal treatment of structured and unstructured knowledge. To address these issues, we propose $\mathbf{BioT5}$, a comprehensive pre-training framework that enriches cross-modal integration in biology with chemical knowledge and natural language associations. $\mathbf{BioT5}$ utilizes SELFIES for $100%$ robust molecular representations and extracts knowledge from the surrounding context of bio-entities in unstructured biological literature. Furthermore, $\mathbf{BioT5}$ distinguishes between structured and unstructured knowledge, leading to more effective utilization of information. After fine-tuning, BioT5 shows superior performance across a wide range of tasks, demonstrating its strong capability of capturing underlying relations and properties of bio-entities. Our code is available at $\href{https://github.com/QizhiPei/BioT5}{Github}$.
△ Less
Submitted 28 January, 2024; v1 submitted 11 October, 2023;
originally announced October 2023.
-
FABind: Fast and Accurate Protein-Ligand Binding
Authors:
Qizhi Pei,
Kaiyuan Gao,
Lijun Wu,
Jinhua Zhu,
Yingce Xia,
Shufang Xie,
Tao Qin,
Kun He,
Tie-Yan Liu,
Rui Yan
Abstract:
Modeling the interaction between proteins and ligands and accurately predicting their binding structures is a critical yet challenging task in drug discovery. Recent advancements in deep learning have shown promise in addressing this challenge, with sampling-based and regression-based methods emerging as two prominent approaches. However, these methods have notable limitations. Sampling-based meth…
▽ More
Modeling the interaction between proteins and ligands and accurately predicting their binding structures is a critical yet challenging task in drug discovery. Recent advancements in deep learning have shown promise in addressing this challenge, with sampling-based and regression-based methods emerging as two prominent approaches. However, these methods have notable limitations. Sampling-based methods often suffer from low efficiency due to the need for generating multiple candidate structures for selection. On the other hand, regression-based methods offer fast predictions but may experience decreased accuracy. Additionally, the variation in protein sizes often requires external modules for selecting suitable binding pockets, further impacting efficiency. In this work, we propose $\mathbf{FABind}$, an end-to-end model that combines pocket prediction and docking to achieve accurate and fast protein-ligand binding. $\mathbf{FABind}$ incorporates a unique ligand-informed pocket prediction module, which is also leveraged for docking pose estimation. The model further enhances the docking process by incrementally integrating the predicted pocket to optimize protein-ligand binding, reducing discrepancies between training and inference. Through extensive experiments on benchmark datasets, our proposed $\mathbf{FABind}$ demonstrates strong advantages in terms of effectiveness and efficiency compared to existing methods. Our code is available at https://github.com/QizhiPei/FABind
△ Less
Submitted 8 January, 2024; v1 submitted 10 October, 2023;
originally announced October 2023.
-
Practical and Asymptotically Exact Conditional Sampling in Diffusion Models
Authors:
Luhuan Wu,
Brian L. Trippe,
Christian A. Naesseth,
David M. Blei,
John P. Cunningham
Abstract:
Diffusion models have been successful on a range of conditional generation tasks including molecular design and text-to-image generation. However, these achievements have primarily depended on task-specific conditional training or error-prone heuristic approximations. Ideally, a conditional generation method should provide exact samples for a broad range of conditional distributions without requir…
▽ More
Diffusion models have been successful on a range of conditional generation tasks including molecular design and text-to-image generation. However, these achievements have primarily depended on task-specific conditional training or error-prone heuristic approximations. Ideally, a conditional generation method should provide exact samples for a broad range of conditional distributions without requiring task-specific training. To this end, we introduce the Twisted Diffusion Sampler, or TDS. TDS is a sequential Monte Carlo (SMC) algorithm that targets the conditional distributions of diffusion models through simulating a set of weighted particles. The main idea is to use twisting, an SMC technique that enjoys good computational efficiency, to incorporate heuristic approximations without compromising asymptotic exactness. We first find in simulation and in conditional image generation tasks that TDS provides a computational statistical trade-off, yielding more accurate approximations with many particles but with empirical improvements over heuristics with as few as two particles. We then turn to motif-scaffolding, a core task in protein design, using a TDS extension to Riemannian diffusion models. On benchmark test cases, TDS allows flexible conditioning criteria and often outperforms the state of the art.
△ Less
Submitted 22 November, 2024; v1 submitted 30 June, 2023;
originally announced June 2023.
-
Functional-Group-Based Diffusion for Pocket-Specific Molecule Generation and Elaboration
Authors:
Haitao Lin,
Yufei Huang,
Odin Zhang,
Lirong Wu,
Siyuan Li,
Zhiyuan Chen,
Stan Z. Li
Abstract:
In recent years, AI-assisted drug design methods have been proposed to generate molecules given the pockets' structures of target proteins. Most of them are atom-level-based methods, which consider atoms as basic components and generate atom positions and types. In this way, however, it is hard to generate realistic fragments with complicated structures. To solve this, we propose D3FG, a functiona…
▽ More
In recent years, AI-assisted drug design methods have been proposed to generate molecules given the pockets' structures of target proteins. Most of them are atom-level-based methods, which consider atoms as basic components and generate atom positions and types. In this way, however, it is hard to generate realistic fragments with complicated structures. To solve this, we propose D3FG, a functional-group-based diffusion model for pocket-specific molecule generation and elaboration. D3FG decomposes molecules into two categories of components: functional groups defined as rigid bodies and linkers as mass points. And the two kinds of components can together form complicated fragments that enhance ligand-protein interactions.
To be specific, in the diffusion process, D3FG diffuses the data distribution of the positions, orientations, and types of the components into a prior distribution; In the generative process, the noise is gradually removed from the three variables by denoisers parameterized with designed equivariant graph neural networks. In the experiments, our method can generate molecules with more realistic 3D structures, competitive affinities toward the protein targets, and better drug properties. Besides, D3FG as a solution to a new task of molecule elaboration, could generate molecules with high affinities based on existing ligands and the hotspots of target proteins.
△ Less
Submitted 18 March, 2024; v1 submitted 30 May, 2023;
originally announced June 2023.
-
The Brain Tumor Segmentation (BraTS-METS) Challenge 2023: Brain Metastasis Segmentation on Pre-treatment MRI
Authors:
Ahmed W. Moawad,
Anastasia Janas,
Ujjwal Baid,
Divya Ramakrishnan,
Rachit Saluja,
Nader Ashraf,
Nazanin Maleki,
Leon Jekel,
Nikolay Yordanov,
Pascal Fehringer,
Athanasios Gkampenis,
Raisa Amiruddin,
Amirreza Manteghinejad,
Maruf Adewole,
Jake Albrecht,
Udunna Anazodo,
Sanjay Aneja,
Syed Muhammad Anwar,
Timothy Bergquist,
Veronica Chiang,
Verena Chung,
Gian Marco Conte,
Farouk Dako,
James Eddy,
Ivan Ezhov
, et al. (207 additional authors not shown)
Abstract:
The translation of AI-generated brain metastases (BM) segmentation into clinical practice relies heavily on diverse, high-quality annotated medical imaging datasets. The BraTS-METS 2023 challenge has gained momentum for testing and benchmarking algorithms using rigorously annotated internationally compiled real-world datasets. This study presents the results of the segmentation challenge and chara…
▽ More
The translation of AI-generated brain metastases (BM) segmentation into clinical practice relies heavily on diverse, high-quality annotated medical imaging datasets. The BraTS-METS 2023 challenge has gained momentum for testing and benchmarking algorithms using rigorously annotated internationally compiled real-world datasets. This study presents the results of the segmentation challenge and characterizes the challenging cases that impacted the performance of the winning algorithms. Untreated brain metastases on standard anatomic MRI sequences (T1, T2, FLAIR, T1PG) from eight contributed international datasets were annotated in stepwise method: published UNET algorithms, student, neuroradiologist, final approver neuroradiologist. Segmentations were ranked based on lesion-wise Dice and Hausdorff distance (HD95) scores. False positives (FP) and false negatives (FN) were rigorously penalized, receiving a score of 0 for Dice and a fixed penalty of 374 for HD95. Eight datasets comprising 1303 studies were annotated, with 402 studies (3076 lesions) released on Synapse as publicly available datasets to challenge competitors. Additionally, 31 studies (139 lesions) were held out for validation, and 59 studies (218 lesions) were used for testing. Segmentation accuracy was measured as rank across subjects, with the winning team achieving a LesionWise mean score of 7.9. Common errors among the leading teams included false negatives for small lesions and misregistration of masks in space.The BraTS-METS 2023 challenge successfully curated well-annotated, diverse datasets and identified common errors, facilitating the translation of BM segmentation across varied clinical environments and providing personalized volumetric reports to patients undergoing BM treatment.
△ Less
Submitted 8 December, 2024; v1 submitted 1 June, 2023;
originally announced June 2023.
-
Cross-Gate MLP with Protein Complex Invariant Embedding is A One-Shot Antibody Designer
Authors:
Cheng Tan,
Zhangyang Gao,
Lirong Wu,
Jun Xia,
Jiangbin Zheng,
Xihong Yang,
Yue Liu,
Bozhen Hu,
Stan Z. Li
Abstract:
Antibodies are crucial proteins produced by the immune system in response to foreign substances or antigens. The specificity of an antibody is determined by its complementarity-determining regions (CDRs), which are located in the variable domains of the antibody chains and form the antigen-binding site. Previous studies have utilized complex techniques to generate CDRs, but they suffer from inadeq…
▽ More
Antibodies are crucial proteins produced by the immune system in response to foreign substances or antigens. The specificity of an antibody is determined by its complementarity-determining regions (CDRs), which are located in the variable domains of the antibody chains and form the antigen-binding site. Previous studies have utilized complex techniques to generate CDRs, but they suffer from inadequate geometric modeling. Moreover, the common iterative refinement strategies lead to an inefficient inference. In this paper, we propose a \textit{simple yet effective} model that can co-design 1D sequences and 3D structures of CDRs in a one-shot manner. To achieve this, we decouple the antibody CDR design problem into two stages: (i) geometric modeling of protein complex structures and (ii) sequence-structure co-learning. We develop a novel macromolecular structure invariant embedding, typically for protein complexes, that captures both intra- and inter-component interactions among the backbone atoms, including C$α$, N, C, and O atoms, to achieve comprehensive geometric modeling. Then, we introduce a simple cross-gate MLP for sequence-structure co-learning, allowing sequence and structure representations to implicitly refine each other. This enables our model to design desired sequences and structures in a one-shot manner. Extensive experiments are conducted to evaluate our results at both the sequence and structure levels, which demonstrate that our model achieves superior performance compared to the state-of-the-art antibody CDR design methods.
△ Less
Submitted 10 January, 2024; v1 submitted 21 April, 2023;
originally announced May 2023.
-
Retrieved Sequence Augmentation for Protein Representation Learning
Authors:
Chang Ma,
Haiteng Zhao,
Lin Zheng,
Jiayi Xin,
Qintong Li,
Lijun Wu,
Zhihong Deng,
Yang Lu,
Qi Liu,
Lingpeng Kong
Abstract:
Protein language models have excelled in a variety of tasks, ranging from structure prediction to protein engineering. However, proteins are highly diverse in functions and structures, and current state-of-the-art models including the latest version of AlphaFold rely on Multiple Sequence Alignments (MSA) to feed in the evolutionary knowledge. Despite their success, heavy computational overheads, a…
▽ More
Protein language models have excelled in a variety of tasks, ranging from structure prediction to protein engineering. However, proteins are highly diverse in functions and structures, and current state-of-the-art models including the latest version of AlphaFold rely on Multiple Sequence Alignments (MSA) to feed in the evolutionary knowledge. Despite their success, heavy computational overheads, as well as the de novo and orphan proteins remain great challenges in protein representation learning. In this work, we show that MSAaugmented models inherently belong to retrievalaugmented methods. Motivated by this finding, we introduce Retrieved Sequence Augmentation(RSA) for protein representation learning without additional alignment or pre-processing. RSA links query protein sequences to a set of sequences with similar structures or properties in the database and combines these sequences for downstream prediction. We show that protein language models benefit from the retrieval enhancement on both structure prediction and property prediction tasks, with a 5% improvement on MSA Transformer on average while being 373 times faster. In addition, we show that our model can transfer to new protein domains better and outperforms MSA Transformer on de novo protein prediction. Our study fills a much-encountered gap in protein prediction and brings us a step closer to demystifying the domain knowledge needed to understand protein sequences. Code is available on https://github.com/HKUNLP/RSA.
△ Less
Submitted 24 February, 2023;
originally announced February 2023.
-
Data-Efficient Protein 3D Geometric Pretraining via Refinement of Diffused Protein Structure Decoy
Authors:
Yufei Huang,
Lirong Wu,
Haitao Lin,
Jiangbin Zheng,
Ge Wang,
Stan Z. Li
Abstract:
Learning meaningful protein representation is important for a variety of biological downstream tasks such as structure-based drug design. Having witnessed the success of protein sequence pretraining, pretraining for structural data which is more informative has become a promising research topic. However, there are three major challenges facing protein structure pretraining: insufficient sample div…
▽ More
Learning meaningful protein representation is important for a variety of biological downstream tasks such as structure-based drug design. Having witnessed the success of protein sequence pretraining, pretraining for structural data which is more informative has become a promising research topic. However, there are three major challenges facing protein structure pretraining: insufficient sample diversity, physically unrealistic modeling, and the lack of protein-specific pretext tasks. To try to address these challenges, we present the 3D Geometric Pretraining. In this paper, we propose a unified framework for protein pretraining and a 3D geometric-based, data-efficient, and protein-specific pretext task: RefineDiff (Refine the Diffused Protein Structure Decoy). After pretraining our geometric-aware model with this task on limited data(less than 1% of SOTA models), we obtained informative protein representations that can achieve comparable performance for various downstream tasks.
△ Less
Submitted 5 February, 2023;
originally announced February 2023.
-
Deciphering RNA Secondary Structure Prediction: A Probabilistic K-Rook Matching Perspective
Authors:
Cheng Tan,
Zhangyang Gao,
Hanqun Cao,
Xingran Chen,
Ge Wang,
Lirong Wu,
Jun Xia,
Jiangbin Zheng,
Stan Z. Li
Abstract:
The secondary structure of ribonucleic acid (RNA) is more stable and accessible in the cell than its tertiary structure, making it essential for functional prediction. Although deep learning has shown promising results in this field, current methods suffer from poor generalization and high complexity. In this work, we reformulate the RNA secondary structure prediction as a K-Rook problem, thereby…
▽ More
The secondary structure of ribonucleic acid (RNA) is more stable and accessible in the cell than its tertiary structure, making it essential for functional prediction. Although deep learning has shown promising results in this field, current methods suffer from poor generalization and high complexity. In this work, we reformulate the RNA secondary structure prediction as a K-Rook problem, thereby simplifying the prediction process into probabilistic matching within a finite solution space. Building on this innovative perspective, we introduce RFold, a simple yet effective method that learns to predict the most matching K-Rook solution from the given sequence. RFold employs a bi-dimensional optimization strategy that decomposes the probabilistic matching problem into row-wise and column-wise components to reduce the matching complexity, simplifying the solving process while guaranteeing the validity of the output. Extensive experiments demonstrate that RFold achieves competitive performance and about eight times faster inference efficiency than the state-of-the-art approaches. The code and Colab demo are available in (http://github.com/A4Bio/RFold).
△ Less
Submitted 19 June, 2024; v1 submitted 2 December, 2022;
originally announced December 2022.
-
Integration of Pre-trained Protein Language Models into Geometric Deep Learning Networks
Authors:
Fang Wu,
Lirong Wu,
Dragomir Radev,
Jinbo Xu,
Stan Z. Li
Abstract:
Geometric deep learning has recently achieved great success in non-Euclidean domains, and learning on 3D structures of large biomolecules is emerging as a distinct research area. However, its efficacy is largely constrained due to the limited quantity of structural data. Meanwhile, protein language models trained on substantial 1D sequences have shown burgeoning capabilities with scale in a broad…
▽ More
Geometric deep learning has recently achieved great success in non-Euclidean domains, and learning on 3D structures of large biomolecules is emerging as a distinct research area. However, its efficacy is largely constrained due to the limited quantity of structural data. Meanwhile, protein language models trained on substantial 1D sequences have shown burgeoning capabilities with scale in a broad range of applications. Several previous studies consider combining these different protein modalities to promote the representation power of geometric neural networks, but fail to present a comprehensive understanding of their benefits. In this work, we integrate the knowledge learned by well-trained protein language models into several state-of-the-art geometric networks and evaluate a variety of protein representation learning benchmarks, including protein-protein interface prediction, model quality assessment, protein-protein rigid-body docking, and binding affinity prediction. Our findings show an overall improvement of 20% over baselines. Strong evidence indicates that the incorporation of protein language models' knowledge enhances geometric networks' capacity by a significant margin and can be generalized to complex tasks.
△ Less
Submitted 29 October, 2023; v1 submitted 6 December, 2022;
originally announced December 2022.
-
DiffBP: Generative Diffusion of 3D Molecules for Target Protein Binding
Authors:
Haitao Lin,
Yufei Huang,
Odin Zhang,
Siqi Ma,
Meng Liu,
Xuanjing Li,
Lirong Wu,
Jishui Wang,
Tingjun Hou,
Stan Z. Li
Abstract:
Generating molecules that bind to specific proteins is an important but challenging task in drug discovery. Previous works usually generate atoms in an auto-regressive way, where element types and 3D coordinates of atoms are generated one by one. However, in real-world molecular systems, the interactions among atoms in an entire molecule are global, leading to the energy function pair-coupled amon…
▽ More
Generating molecules that bind to specific proteins is an important but challenging task in drug discovery. Previous works usually generate atoms in an auto-regressive way, where element types and 3D coordinates of atoms are generated one by one. However, in real-world molecular systems, the interactions among atoms in an entire molecule are global, leading to the energy function pair-coupled among atoms. With such energy-based consideration, the modeling of probability should be based on joint distributions, rather than sequentially conditional ones. Thus, the unnatural sequentially auto-regressive modeling of molecule generation is likely to violate the physical rules, thus resulting in poor properties of the generated molecules. In this work, a generative diffusion model for molecular 3D structures based on target proteins as contextual constraints is established, at a full-atom level in a non-autoregressive way. Given a designated 3D protein binding site, our model learns the generative process that denoises both element types and 3D coordinates of an entire molecule, with an equivariant network. Experimentally, the proposed method shows competitive performance compared with prevailing works in terms of high affinity with proteins and appropriate molecule sizes as well as other drug properties such as drug-likeness of the generated molecules.
△ Less
Submitted 14 July, 2024; v1 submitted 21 November, 2022;
originally announced November 2022.
-
Incorporating Pre-training Paradigm for Antibody Sequence-Structure Co-design
Authors:
Kaiyuan Gao,
Lijun Wu,
Jinhua Zhu,
Tianbo Peng,
Yingce Xia,
Liang He,
Shufang Xie,
Tao Qin,
Haiguang Liu,
Kun He,
Tie-Yan Liu
Abstract:
Antibodies are versatile proteins that can bind to pathogens and provide effective protection for human body. Recently, deep learning-based computational antibody design has attracted popular attention since it automatically mines the antibody patterns from data that could be complementary to human experiences. However, the computational methods heavily rely on high-quality antibody structure data…
▽ More
Antibodies are versatile proteins that can bind to pathogens and provide effective protection for human body. Recently, deep learning-based computational antibody design has attracted popular attention since it automatically mines the antibody patterns from data that could be complementary to human experiences. However, the computational methods heavily rely on high-quality antibody structure data, which is quite limited. Besides, the complementarity-determining region (CDR), which is the key component of an antibody that determines the specificity and binding affinity, is highly variable and hard to predict. Therefore, the data limitation issue further raises the difficulty of CDR generation for antibodies. Fortunately, there exists a large amount of sequence data of antibodies that can help model the CDR and alleviate the reliance on structure data. By witnessing the success of pre-training models for protein modeling, in this paper, we develop the antibody pre-training language model and incorporate it into the (antigen-specific) antibody design model in a systemic way. Specifically, we first pre-train an antibody language model based on the sequence data, then propose a one-shot way for sequence and structure generation of CDR to avoid the heavy cost and error propagation from an autoregressive manner, and finally leverage the pre-trained antibody model for the antigen-specific antibody generation model with some carefully designed modules. Through various experiments, we show that our method achieves superior performances over previous baselines on different tasks, such as sequence and structure generation and antigen-binding CDR-H3 design.
△ Less
Submitted 17 November, 2022; v1 submitted 26 October, 2022;
originally announced November 2022.
-
Multi-view information fusion using multi-view variational autoencoders to predict proximal femoral strength
Authors:
Chen Zhao,
Joyce H Keyak,
Xuewei Cao,
Qiuying Sha,
Li Wu,
Zhe Luo,
Lanjuan Zhao,
Qing Tian,
Chuan Qiu,
Ray Su,
Hui Shen,
Hong-Wen Deng,
Weihua Zhou
Abstract:
The aim of this paper is to design a deep learning-based model to predict proximal femoral strength using multi-view information fusion. Method: We developed new models using multi-view variational autoencoder (MVAE) for feature representation learning and a product of expert (PoE) model for multi-view information fusion. We applied the proposed models to an in-house Louisiana Osteoporosis Study (…
▽ More
The aim of this paper is to design a deep learning-based model to predict proximal femoral strength using multi-view information fusion. Method: We developed new models using multi-view variational autoencoder (MVAE) for feature representation learning and a product of expert (PoE) model for multi-view information fusion. We applied the proposed models to an in-house Louisiana Osteoporosis Study (LOS) cohort with 931 male subjects, including 345 African Americans and 586 Caucasians. With an analytical solution of the product of Gaussian distribution, we adopted variational inference to train the designed MVAE-PoE model to perform common latent feature extraction. We performed genome-wide association studies (GWAS) to select 256 genetic variants with the lowest p-values for each proximal femoral strength and integrated whole genome sequence (WGS) features and DXA-derived imaging features to predict proximal femoral strength. Results: The best prediction model for fall fracture load was acquired by integrating WGS features and DXA-derived imaging features. The designed models achieved the mean absolute percentage error of 18.04%, 6.84% and 7.95% for predicting proximal femoral fracture loads using linear models of fall loading, nonlinear models of fall loading, and nonlinear models of stance loading, respectively. Compared to existing multi-view information fusion methods, the proposed MVAE-PoE achieved the best performance. Conclusion: The proposed models are capable of predicting proximal femoral strength using WGS features and DXA-derived imaging features. Though this tool is not a substitute for FEA using QCT images, it would make improved assessment of hip fracture risk more widely available while avoiding the increased radiation dosage and clinical costs from QCT.
△ Less
Submitted 27 March, 2023; v1 submitted 2 October, 2022;
originally announced October 2022.
-
Tailoring Molecules for Protein Pockets: a Transformer-based Generative Solution for Structured-based Drug Design
Authors:
Kehan Wu,
Yingce Xia,
Yang Fan,
Pan Deng,
Haiguang Liu,
Lijun Wu,
Shufang Xie,
Tong Wang,
Tao Qin,
Tie-Yan Liu
Abstract:
Structure-based drug design is drawing growing attentions in computer-aided drug discovery. Compared with the virtual screening approach where a pre-defined library of compounds are computationally screened, de novo drug design based on the structure of a target protein can provide novel drug candidates. In this paper, we present a generative solution named TamGent (Target-aware molecule generator…
▽ More
Structure-based drug design is drawing growing attentions in computer-aided drug discovery. Compared with the virtual screening approach where a pre-defined library of compounds are computationally screened, de novo drug design based on the structure of a target protein can provide novel drug candidates. In this paper, we present a generative solution named TamGent (Target-aware molecule generator with Transformer) that can directly generate candidate drugs from scratch for a given target, overcoming the limits imposed by existing compound libraries. Following the Transformer framework (a state-of-the-art framework in deep learning), we design a variant of Transformer encoder to process 3D geometric information of targets and pre-train the Transformer decoder on 10 million compounds from PubChem for candidate drug generation. Systematical evaluation on candidate compounds generated for targets from DrugBank shows that both binding affinity and drugability are largely improved. TamGent outperforms previous baselines in terms of both effectiveness and efficiency. The method is further verified by generating candidate compounds for the SARS-CoV-2 main protease and the oncogenic mutant KRAS G12C. The results show that our method not only re-discovers previously verified drug molecules , but also generates novel molecules with better docking scores, expanding the compound pool and potentially leading to the discovery of novel drugs.
△ Less
Submitted 30 August, 2022;
originally announced September 2022.
-
Self-supervised multimodal neuroimaging yields predictive representations for a spectrum of Alzheimer's phenotypes
Authors:
Alex Fedorov,
Eloy Geenjaar,
Lei Wu,
Tristan Sylvain,
Thomas P. DeRamus,
Margaux Luck,
Maria Misiura,
R Devon Hjelm,
Sergey M. Plis,
Vince D. Calhoun
Abstract:
Recent neuroimaging studies that focus on predicting brain disorders via modern machine learning approaches commonly include a single modality and rely on supervised over-parameterized models.However, a single modality provides only a limited view of the highly complex brain. Critically, supervised models in clinical settings lack accurate diagnostic labels for training. Coarse labels do not captu…
▽ More
Recent neuroimaging studies that focus on predicting brain disorders via modern machine learning approaches commonly include a single modality and rely on supervised over-parameterized models.However, a single modality provides only a limited view of the highly complex brain. Critically, supervised models in clinical settings lack accurate diagnostic labels for training. Coarse labels do not capture the long-tailed spectrum of brain disorder phenotypes, which leads to a loss of generalizability of the model that makes them less useful in diagnostic settings. This work presents a novel multi-scale coordinated framework for learning multiple representations from multimodal neuroimaging data. We propose a general taxonomy of informative inductive biases to capture unique and joint information in multimodal self-supervised fusion. The taxonomy forms a family of decoder-free models with reduced computational complexity and a propensity to capture multi-scale relationships between local and global representations of the multimodal inputs. We conduct a comprehensive evaluation of the taxonomy using functional and structural magnetic resonance imaging (MRI) data across a spectrum of Alzheimer's disease phenotypes and show that self-supervised models reveal disorder-relevant brain regions and multimodal links without access to the labels during pre-training. The proposed multimodal self-supervised learning yields representations with improved classification performance for both modalities. The concomitant rich and flexible unsupervised deep learning framework captures complex multimodal relationships and provides predictive performance that meets or exceeds that of a more narrow supervised classification analysis. We present elaborate quantitative evidence of how this framework can significantly advance our search for missing links in complex brain disorders.
△ Less
Submitted 6 September, 2022;
originally announced September 2022.
-
SSM-DTA: Breaking the Barriers of Data Scarcity in Drug-Target Affinity Prediction
Authors:
Qizhi Pei,
Lijun Wu,
Jinhua Zhu,
Yingce Xia,
Shufang Xie,
Tao Qin,
Haiguang Liu,
Tie-Yan Liu,
Rui Yan
Abstract:
Accurate prediction of Drug-Target Affinity (DTA) is of vital importance in early-stage drug discovery, facilitating the identification of drugs that can effectively interact with specific targets and regulate their activities. While wet experiments remain the most reliable method, they are time-consuming and resource-intensive, resulting in limited data availability that poses challenges for deep…
▽ More
Accurate prediction of Drug-Target Affinity (DTA) is of vital importance in early-stage drug discovery, facilitating the identification of drugs that can effectively interact with specific targets and regulate their activities. While wet experiments remain the most reliable method, they are time-consuming and resource-intensive, resulting in limited data availability that poses challenges for deep learning approaches. Existing methods have primarily focused on developing techniques based on the available DTA data, without adequately addressing the data scarcity issue. To overcome this challenge, we present the SSM-DTA framework, which incorporates three simple yet highly effective strategies: (1) A multi-task training approach that combines DTA prediction with masked language modeling (MLM) using paired drug-target data. (2) A semi-supervised training method that leverages large-scale unpaired molecules and proteins to enhance drug and target representations. This approach differs from previous methods that only employed molecules or proteins in pre-training. (3) The integration of a lightweight cross-attention module to improve the interaction between drugs and targets, further enhancing prediction accuracy. Through extensive experiments on benchmark datasets such as BindingDB, DAVIS, and KIBA, we demonstrate the superior performance of our framework. Additionally, we conduct case studies on specific drug-target binding activities, virtual screening experiments, drug feature visualizations, and real-world applications, all of which showcase the significant potential of our work. In conclusion, our proposed SSM-DTA framework addresses the data limitation challenge in DTA prediction and yields promising results, paving the way for more efficient and accurate drug discovery processes. Our code is available at $\href{https://github.com/QizhiPei/SSM-DTA}{Github}$.
△ Less
Submitted 17 October, 2023; v1 submitted 20 June, 2022;
originally announced June 2022.
-
Understanding the network formation pattern for better link prediction
Authors:
Jiating Yu,
Ling-Yun Wu
Abstract:
As a classical problem in the field of complex networks, link prediction has attracted much attention from researchers, which is of great significance to help us understand the evolution and dynamic development mechanisms of networks. Although various network type-specific algorithms have been proposed to tackle the link prediction problem, most of them suppose that the network structure is domina…
▽ More
As a classical problem in the field of complex networks, link prediction has attracted much attention from researchers, which is of great significance to help us understand the evolution and dynamic development mechanisms of networks. Although various network type-specific algorithms have been proposed to tackle the link prediction problem, most of them suppose that the network structure is dominated by the Triadic Closure Principle. We still lack an adaptive and comprehensive understanding of network formation patterns for predicting potential links. In addition, it is valuable to investigate how network local information can be better utilized. To this end, we proposed a novel method named Link prediction using Multiple Order Local Information (MOLI) that exploits the local information from the neighbors of different distances, with parameters that can be a prior-driven based on prior knowledge, or data-driven by solving an optimization problem on observed networks. MOLI defined a local network diffusion process via random walks on the graph, resulting in better use of network information. We show that MOLI outperforms the other 11 widely used link prediction algorithms on 11 different types of simulated and real-world networks. We also conclude that there are different patterns of local information utilization for different networks, including social networks, communication networks, biological networks, etc. In particular, the classical common neighbor-based algorithm is not as adaptable to all social networks as it is perceived to be; instead, some of the social networks obey the Quadrilateral Closure Principle which preferentially connects paths of length three.
△ Less
Submitted 17 October, 2021;
originally announced October 2021.
-
Network Refinement: A unified framework for enhancing signal or removing noise of networks
Authors:
Jiating Yu,
Jiacheng Leng,
Ling-Yun Wu
Abstract:
Networks are widely used in many fields for their powerful ability to provide vivid representations of relationships between variables. However, many of them may be corrupted by experimental noise or inappropriate network inference methods that inherently hamper the efficacy of network-based downstream analysis. Consequently, it's necessary to develop systematic methods for denoising networks, nam…
▽ More
Networks are widely used in many fields for their powerful ability to provide vivid representations of relationships between variables. However, many of them may be corrupted by experimental noise or inappropriate network inference methods that inherently hamper the efficacy of network-based downstream analysis. Consequently, it's necessary to develop systematic methods for denoising networks, namely, improve the Signal-to-Noise Ratio (SNR) of noisy networks. In this paper, we have explored the properties of network signal and noise and proposed a novel network denoising framework called Network Refinement (NR) that adjusts the edge weights by applying a nonlinear graph operator based on a diffusion process defined by random walk on the graph. Specifically, this unified framework consists of two closely linked approaches named NR-F and NR-B, which improve the SNR of noisy input networks from two different perspectives: NR-F aims at enhancing signal strength, while NR-B aims at weakening noise strength. Users can choose from which angle to improve the SNR of the network according to the characteristics of the network itself. We show that NR can significantly refine the quality of many networks by several applications on simulated networks and typical real-world biological and social networks.
△ Less
Submitted 19 September, 2021;
originally announced September 2021.
-
ChemiRise: a data-driven retrosynthesis engine
Authors:
Xiangyan Sun,
Ke Liu,
Yuquan Lin,
Lingjie Wu,
Haoming Xing,
Minghong Gao,
Ji Liu,
Suocheng Tan,
Zekun Ni,
Qi Han,
Junqiu Wu,
Jie Fan
Abstract:
We have developed an end-to-end, retrosynthesis system, named ChemiRise, that can propose complete retrosynthesis routes for organic compounds rapidly and reliably. The system was trained on a processed patent database of over 3 million organic reactions. Experimental reactions were atom-mapped, clustered, and extracted into reaction templates. We then trained a graph convolutional neural network-…
▽ More
We have developed an end-to-end, retrosynthesis system, named ChemiRise, that can propose complete retrosynthesis routes for organic compounds rapidly and reliably. The system was trained on a processed patent database of over 3 million organic reactions. Experimental reactions were atom-mapped, clustered, and extracted into reaction templates. We then trained a graph convolutional neural network-based one-step reaction proposer using template embeddings and developed a guiding algorithm on the directed acyclic graph (DAG) of chemical compounds to find the best candidate to explore. The atom-mapping algorithm and the one-step reaction proposer were benchmarked against previous studies and showed better results. The final product was demonstrated by retrosynthesis routes reviewed and rated by human experts, showing satisfying functionality and a potential productivity boost in real-life use cases.
△ Less
Submitted 9 August, 2021;
originally announced August 2021.
-
Evolution of cooperation driven by active information spreading
Authors:
Bin Wu,
Hye Jin Park,
Lingshan Wu,
Da Zhou
Abstract:
Cooperators forgo their interest to benefit others. Thus cooperation should not be favored by natural selection. It challenges the evolutionists, since cooperation is widespread. As one of the resolutions, information spreading has been revealed to play a key role in the emergence of cooperation. Individuals, however, are typically assumed to be passive in the information spreading. Here we assume…
▽ More
Cooperators forgo their interest to benefit others. Thus cooperation should not be favored by natural selection. It challenges the evolutionists, since cooperation is widespread. As one of the resolutions, information spreading has been revealed to play a key role in the emergence of cooperation. Individuals, however, are typically assumed to be passive in the information spreading. Here we assume that individuals are active to spread the information via self-recommendation. Individuals with higher intensities of self-recommendation are likely to have more neighbors. We find that i) eloquent cooperators are necessary to promote cooperation; ii) individuals need to be open to the self-recommendation to enhance cooperation level; iii) the cost-to-benefit ratio should be smaller than one minus the ratio between self-recommendation intensities of defector and cooperator, which qualitatively measures the viscosity of the population. Our results highlight the importance of active information spreading on cooperation.
△ Less
Submitted 3 July, 2019;
originally announced July 2019.