Deep Lead Optimization: Leveraging Generative AI for Structural Modification

Read original: arXiv:2404.19230 - Published 5/1/2024 by Odin Zhang, Haitao Lin, Hui Zhang, Huifeng Zhao, Yufei Huang, Yuansheng Huang, Dejun Jiang, Chang-yu Hsieh, Peichen Pan, Tingjun Hou
Total Score

0

🤿

Sign in to get full access

or

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

Overview

  • This paper explores using deep learning-based molecular generation to accelerate the discovery of new drug candidates.
  • It focuses on the subfield of lead optimization, which refines existing molecules into better drug candidates, rather than generating entirely new molecules from scratch.
  • The paper conducts a systematic review of traditional computational methods for lead optimization, organizing them into four main subtasks.
  • It also introduces a unified perspective that sees lead optimization as a task of generating molecules conditioned on certain substructures, which can benefit from innovations in de novo design.

Plain English Explanation

Developing new drugs is a complex and lengthy process. One promising approach is to use deep learning algorithms to generate potential drug molecules. This is known as molecular generation.

There are two main strategies in molecular generation: de novo design, which creates entirely new molecules, and lead optimization, which refines existing molecules to make them better drug candidates. Lead optimization is particularly important in real-world drug development, as it can help create "me-better" drugs that are chemically distinct yet more effective than the original.

Despite its importance, lead optimization has been less explored than de novo design, in part because it requires deep knowledge of biology and chemistry. This paper aims to bridge that gap by reviewing traditional computational methods for lead optimization and organizing them into four main subtasks.

The paper also introduces a new perspective that sees lead optimization as a type of constrained subgraph generation. This allows lead optimization to benefit from innovations in de novo design, while also allowing de novo design to incorporate strategies from lead optimization to generate molecules that are easier to synthesize.

Technical Explanation

The paper conducts a systematic review of traditional computational approaches to lead optimization, organizing them into four main subtasks:

  1. Molecular Modifications: Modifying an existing molecule to improve its properties, such as binding affinity or bioavailability.
  2. Bioisosteric Replacements: Replacing parts of a molecule with chemically similar substructures to maintain or improve its function.
  3. Scaffold Hopping: Identifying new molecular scaffolds that can serve as the core structure for a drug candidate.
  4. Analog Search: Identifying structural analogs of a lead compound that may have improved properties.

The paper then introduces a unified perspective that views lead optimization as a task of generating molecules conditioned on certain substructures. This allows lead optimization to benefit from innovations in de novo design, such as the use of deep generative models and reinforcement learning.

Conversely, this perspective also allows de novo design to incorporate strategies from lead optimization, such as multimodal learning to generate molecules that are more synthetically accessible. This can help address the challenge of generating hard-to-synthesize molecules, which is a common issue in de novo design.

Critical Analysis

The paper provides a comprehensive review of traditional computational methods for lead optimization, which is a valuable contribution to the field. However, it is important to note that the paper does not present any new experimental results or novel deep learning architectures.

Additionally, the paper does not delve deeply into the limitations or potential issues with the reviewed computational methods. For example, it does not discuss the challenges of incorporating complex biological and chemical knowledge into these algorithms, or the difficulty of accurately predicting the properties of modified molecules.

Further research is needed to address these limitations and develop more robust and reliable deep learning-based methods for lead optimization, target-conditioned molecule generation, and 3D molecular modeling.

Conclusion

This paper provides a valuable review of traditional computational methods for lead optimization, a critical task in the drug discovery process. By introducing a unified perspective that views lead optimization as a form of constrained subgraph generation, the paper opens up new avenues for cross-pollination between lead optimization and de novo design approaches.

While the paper does not present any novel deep learning architectures, it lays the groundwork for further research in this area. Developing more effective and reliable deep learning-based methods for lead optimization could significantly accelerate the discovery of new drug candidates and improve the efficiency of the overall drug development pipeline.



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

Follow @aimodelsfyi on 𝕏 →

Related Papers

🤿

Total Score

0

Deep Lead Optimization: Leveraging Generative AI for Structural Modification

Odin Zhang, Haitao Lin, Hui Zhang, Huifeng Zhao, Yufei Huang, Yuansheng Huang, Dejun Jiang, Chang-yu Hsieh, Peichen Pan, Tingjun Hou

The idea of using deep-learning-based molecular generation to accelerate discovery of drug candidates has attracted extraordinary attention, and many deep generative models have been developed for automated drug design, termed molecular generation. In general, molecular generation encompasses two main strategies: de novo design, which generates novel molecular structures from scratch, and lead optimization, which refines existing molecules into drug candidates. Among them, lead optimization plays an important role in real-world drug design. For example, it can enable the development of me-better drugs that are chemically distinct yet more effective than the original drugs. It can also facilitate fragment-based drug design, transforming virtual-screened small ligands with low affinity into first-in-class medicines. Despite its importance, automated lead optimization remains underexplored compared to the well-established de novo generative models, due to its reliance on complex biological and chemical knowledge. To bridge this gap, we conduct a systematic review of traditional computational methods for lead optimization, organizing these strategies into four principal sub-tasks with defined inputs and outputs. This review delves into the basic concepts, goals, conventional CADD techniques, and recent advancements in AIDD. Additionally, we introduce a unified perspective based on constrained subgraph generation to harmonize the methodologies of de novo design and lead optimization. Through this lens, de novo design can incorporate strategies from lead optimization to address the challenge of generating hard-to-synthesize molecules; inversely, lead optimization can benefit from the innovations in de novo design by approaching it as a task of generating molecules conditioned on certain substructures.

Read more

5/1/2024

A Survey of Generative AI for de novo Drug Design: New Frontiers in Molecule and Protein Generation
Total Score

0

A Survey of Generative AI for de novo Drug Design: New Frontiers in Molecule and Protein Generation

Xiangru Tang, Howard Dai, Elizabeth Knight, Fang Wu, Yunyang Li, Tianxiao Li, Mark Gerstein

Artificial intelligence (AI)-driven methods can vastly improve the historically costly drug design process, with various generative models already in widespread use. Generative models for de novo drug design, in particular, focus on the creation of novel biological compounds entirely from scratch, representing a promising future direction. Rapid development in the field, combined with the inherent complexity of the drug design process, creates a difficult landscape for new researchers to enter. In this survey, we organize de novo drug design into two overarching themes: small molecule and protein generation. Within each theme, we identify a variety of subtasks and applications, highlighting important datasets, benchmarks, and model architectures and comparing the performance of top models. We take a broad approach to AI-driven drug design, allowing for both micro-level comparisons of various methods within each subtask and macro-level observations across different fields. We discuss parallel challenges and approaches between the two applications and highlight future directions for AI-driven de novo drug design as a whole. An organized repository of all covered sources is available at https://github.com/gersteinlab/GenAI4Drug.

Read more

6/27/2024

🛠️

Total Score

0

Human-level molecular optimization driven by mol-gene evolution

Jiebin Fang (Hainan Institute of Zhejiang University, Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University), Churu Mao (Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University), Yuchen Zhu (College of Pharmaceutical Sciences and Cancer Center, Zhejiang University), Xiaoming Chen (Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University), Chang-Yu Hsieh (College of Pharmaceutical Sciences and Cancer Center, Zhejiang University), Zhongjun Ma (Hainan Institute of Zhejiang University, Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University)

De novo molecule generation allows the search for more drug-like hits across a vast chemical space. However, lead optimization is still required, and the process of optimizing molecular structures faces the challenge of balancing structural novelty with pharmacological properties. This study introduces the Deep Genetic Molecular Modification Algorithm (DGMM), which brings structure modification to the level of medicinal chemists. A discrete variational autoencoder (D-VAE) is used in DGMM to encode molecules as quantization code, mol-gene, which incorporates deep learning into genetic algorithms for flexible structural optimization. The mol-gene allows for the discovery of pharmacologically similar but structurally distinct compounds, and reveals the trade-offs of structural optimization in drug discovery. We demonstrate the effectiveness of the DGMM in several applications.

Read more

6/21/2024

Leveraging Latent Evolutionary Optimization for Targeted Molecule Generation
Total Score

0

Leveraging Latent Evolutionary Optimization for Targeted Molecule Generation

Siddartha Reddy N, Sai Prakash MV, Varun V, Vishal Vaddina, Saisubramaniam Gopalakrishnan

Lead optimization is a pivotal task in the drug design phase within the drug discovery lifecycle. The primary objective is to refine the lead compound to meet specific molecular properties for progression to the subsequent phase of development. In this work, we present an innovative approach, Latent Evolutionary Optimization for Molecule Generation (LEOMol), a generative modeling framework for the efficient generation of optimized molecules. LEOMol leverages Evolutionary Algorithms, such as Genetic Algorithm and Differential Evolution, to search the latent space of a Variational AutoEncoder (VAE). This search facilitates the identification of the target molecule distribution within the latent space. Our approach consistently demonstrates superior performance compared to previous state-of-the-art models across a range of constrained molecule generation tasks, outperforming existing models in all four sub-tasks related to property targeting. Additionally, we suggest the importance of including toxicity in the evaluation of generative models. Furthermore, an ablation study underscores the improvements that our approach provides over gradient-based latent space optimization methods. This underscores the effectiveness and superiority of LEOMol in addressing the inherent challenges in constrained molecule generation while emphasizing its potential to propel advancements in drug discovery.

Read more

7/22/2024