Structure-based drug design by denoising voxel grids

Read original: arXiv:2405.03961 - Published 5/8/2024 by Pedro O. Pinheiro, Arian Jamasb, Omar Mahmood, Vishnu Sresht, Saeed Saremi
Total Score

0

Structure-based drug design by denoising voxel grids

Sign in to get full access

or

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

Overview

  • This paper proposes a structure-based drug design approach using a deep learning model to "denoise" voxel grids representing molecular structures.
  • The model aims to generate high-quality molecular structures that can be used in the drug design process.
  • The approach leverages recent advancements in generative models and diffusion models to generate novel molecular structures.

Plain English Explanation

The paper introduces a new way to design potential drug compounds using a deep learning model. The key idea is to start with a rough or "noisy" 3D representation of a molecule, and then use a neural network to refine and "denoise" that representation, producing a high-quality 3D structure that could be a promising drug candidate.

This builds on recent AI techniques like generative models and diffusion models, which have shown promise in generating realistic molecular structures. The advantage of this approach is that it can explore a wide range of potential drug molecules, rather than being limited to existing compounds.

Overall, the goal is to streamline the drug design process by using AI to rapidly generate and refine promising drug candidates, which can then be further tested and optimized by human experts.

Technical Explanation

The paper proposes a score-based generative model for structure-based drug design, where the model learns to "denoise" voxel grid representations of molecular structures.

The model architecture consists of a 3D convolutional neural network that takes a noisy voxel grid as input and produces a denoised output. This is trained using a diffusion process that gradually adds noise to clean molecular structures and then learns to reverse that process.

Key innovations include the use of Gaussian splatting to represent molecular structures, and the incorporation of domain-specific knowledge about molecular interactions and pharmacological properties into the model training.

The authors demonstrate the effectiveness of their approach through experiments on generating new drug-like molecules and show that the model can produce high-quality structures that are chemically feasible and have favorable drug-like properties.

Critical Analysis

The paper presents a promising approach for leveraging AI to assist in the drug design process. The use of a generative model to "denoise" molecular structures is an interesting idea that could help explore a wider chemical space than traditional methods.

However, the paper does not fully address the challenge of ensuring that the generated structures are not only chemically feasible, but also biologically active and safe. While the authors incorporate some domain knowledge, there may be additional pharmacological considerations that need to be taken into account.

Additionally, the paper focuses on the technical details of the model architecture and training process, but does not provide a deep analysis of the broader implications or potential limitations of this approach. Further research and real-world validation would be needed to fully understand the practical impact and scalability of this technique.

Overall, the paper is a valuable contribution to the field of structure-based drug design, but additional work is needed to address the remaining challenges and fully realize the potential of this AI-driven approach.

Conclusion

This paper presents a novel approach for structure-based drug design using a deep learning model to "denoise" and refine molecular structures. By leveraging recent advancements in generative models and diffusion processes, the authors demonstrate a promising way to explore a wider chemical space and rapidly generate promising drug candidates.

While the technical details of the model are compelling, there are still open questions around ensuring the biological relevance and safety of the generated structures. Further research and real-world validation would be needed to fully understand the practical impact of this approach.

Nevertheless, this work represents an exciting step forward in the application of AI to the drug design process, and could pave the way for more efficient and effective development of new therapeutic compounds.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Follow @aimodelsfyi on 𝕏 →

Related Papers

Structure-based drug design by denoising voxel grids
Total Score

0

Structure-based drug design by denoising voxel grids

Pedro O. Pinheiro, Arian Jamasb, Omar Mahmood, Vishnu Sresht, Saeed Saremi

We present VoxBind, a new score-based generative model for 3D molecules conditioned on protein structures. Our approach represents molecules as 3D atomic density grids and leverages a 3D voxel-denoising network for learning and generation. We extend the neural empirical Bayes formalism (Saremi & Hyvarinen, 2019) to the conditional setting and generate structure-conditioned molecules with a two-step procedure: (i) sample noisy molecules from the Gaussian-smoothed conditional distribution with underdamped Langevin MCMC using the learned score function and (ii) estimate clean molecules from the noisy samples with single-step denoising. Compared to the current state of the art, our model is simpler to train, significantly faster to sample from, and achieves better results on extensive in silico benchmarks -- the generated molecules are more diverse, exhibit fewer steric clashes, and bind with higher affinity to protein pockets.

Read more

5/8/2024

Total Score

0

DiffBP: Generative Diffusion of 3D Molecules for Target Protein Binding

Haitao Lin, Yufei Huang, Odin Zhang, Siqi Ma, Meng Liu, Xuanjing Li, Lirong Wu, Jishui Wang, Tingjun Hou, Stan Z. Li

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.

Read more

7/16/2024

CBGBench: Fill in the Blank of Protein-Molecule Complex Binding Graph
Total Score

0

CBGBench: Fill in the Blank of Protein-Molecule Complex Binding Graph

Haitao Lin, Guojiang Zhao, Odin Zhang, Yufei Huang, Lirong Wu, Zicheng Liu, Siyuan Li, Cheng Tan, Zhifeng Gao, Stan Z. Li

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}.

Read more

7/23/2024

MolCRAFT: Structure-Based Drug Design in Continuous Parameter Space
Total Score

0

MolCRAFT: Structure-Based Drug Design in Continuous Parameter Space

Yanru Qu, Keyue Qiu, Yuxuan Song, Jingjing Gong, Jiawei Han, Mingyue Zheng, Hao Zhou, Wei-Ying Ma

Generative models for structure-based drug design (SBDD) have shown promising results in recent years. Existing works mainly focus on how to generate molecules with higher binding affinity, ignoring the feasibility prerequisites for generated 3D poses and resulting in false positives. We conduct thorough studies on key factors of ill-conformational problems when applying autoregressive methods and diffusion to SBDD, including mode collapse and hybrid continuous-discrete space. In this paper, we introduce MolCRAFT, the first SBDD model that operates in the continuous parameter space, together with a novel noise reduced sampling strategy. Empirical results show that our model consistently achieves superior performance in binding affinity with more stable 3D structure, demonstrating our ability to accurately model interatomic interactions. To our best knowledge, MolCRAFT is the first to achieve reference-level Vina Scores (-6.59 kcal/mol) with comparable molecular size, outperforming other strong baselines by a wide margin (-0.84 kcal/mol). Code is available at https://github.com/AlgoMole/MolCRAFT.

Read more

5/29/2024