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

Read original: arXiv:2402.08703 - Published 6/27/2024 by Xiangru Tang, Howard Dai, Elizabeth Knight, Fang Wu, Yunyang Li, Tianxiao Li, Mark Gerstein
Total Score

0

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

Sign in to get full access

or

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

Overview

  • This paper provides a comprehensive survey of the latest advancements in generative AI models for de novo drug design, exploring the frontiers of molecule and protein generation.
  • It examines the use of cutting-edge techniques like deep lead optimization, fragment-based molecule generation, molecular optimization driven by machine learning, and diffusion models for drug design.
  • The paper also discusses the potential of synthetic data from diffusion models to accelerate drug discovery.

Plain English Explanation

This paper explores how the latest advancements in artificial intelligence, specifically generative models, are being used to help design new drug molecules from scratch. Traditionally, developing new drugs has been a slow and expensive process, often relying on trial-and-error methods. However, these AI-powered techniques are showing promise in streamlining and accelerating the drug discovery pipeline.

The paper examines several cutting-edge approaches, such as using deep learning models to optimize the molecular structures of lead compounds, generating novel drug candidates by combining molecular fragments, and applying diffusion models to explore the vast space of possible drug molecules. These methods aim to harness the power of machine learning to navigate the complex chemical landscape more efficiently and identify promising drug candidates that could have valuable therapeutic properties.

Additionally, the paper discusses how synthetic data generated by these AI models can be used to further enhance the drug discovery process, providing researchers with large, diverse datasets to train and validate their models.

Technical Explanation

The paper begins by providing an overview of the key generative AI models used in the context of de novo drug design, including variational autoencoders (VAEs), generative adversarial networks (GANs), and diffusion models. It explains how these models can be applied to generate novel molecular structures and optimize existing compounds.

The survey then delves into several specific approaches. For deep lead optimization, the paper examines how deep learning models can leverage the structural information of lead compounds to guide the optimization process, leading to more potent and selective drug candidates.

The fragment-based molecule generation section explores techniques that build new molecules by combining smaller molecular building blocks, allowing for the exploration of a vast chemical space. The paper also discusses molecular optimization driven by machine learning, where AI models are trained to navigate the optimization landscape and suggest modifications to improve desired properties.

Regarding diffusion models for drug design, the paper explains how these generative models can be used to generate diverse and novel molecular structures, potentially leading to the discovery of unexpected drug candidates.

Finally, the paper examines the potential of synthetic data from diffusion models to augment the training of drug discovery models, addressing the common challenge of limited experimental data.

Critical Analysis

The paper provides a comprehensive overview of the state-of-the-art in generative AI for de novo drug design, highlighting the significant progress made in this field. However, the authors also acknowledge several caveats and limitations to the current approaches.

One key challenge is the need for better integration of the various generative models with downstream drug discovery workflows, ensuring seamless translation from molecule generation to experimental validation and lead optimization. The paper suggests that further research is required to develop more holistic and end-to-end solutions.

Additionally, the authors note that while the synthetic data generated by diffusion models can be valuable, ensuring the chemical and biological relevance of these molecules remains an important consideration. Careful evaluation and validation of the generated compounds are necessary before they can be reliably used in drug discovery pipelines.

Another potential limitation is the inherent complexity of the drug discovery process, which involves not only molecule generation but also considerations of pharmacokinetics, toxicity, and target engagement. The paper suggests that future research should explore ways to integrate these additional factors into the generative AI frameworks.

Conclusion

This comprehensive survey paper showcases the remarkable advancements in generative AI for de novo drug design, demonstrating the potential of these techniques to revolutionize the drug discovery process. By leveraging cutting-edge models like VAEs, GANs, and diffusion models, researchers can now explore the vast chemical space more efficiently, generate novel and promising drug candidates, and even leverage synthetic data to accelerate the overall discovery pipeline.

While challenges remain, the progress highlighted in this paper suggests that generative AI is poised to play an increasingly crucial role in the future of drug discovery, paving the way for the development of novel and more effective therapeutic solutions.



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

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

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

👀

Total Score

0

Evaluation Framework for AI-driven Molecular Design of Multi-target Drugs: Brain Diseases as a Case Study

Arthur Cerveira, Frederico Kremer, Darling de Andrade Lourenc{c}o, Ulisses B Corr^ea

The widespread application of Artificial Intelligence (AI) techniques has significantly influenced the development of new therapeutic agents. These computational methods can be used to design and predict the properties of generated molecules. Multi-target Drug Discovery (MTDD) is an emerging paradigm for discovering drugs against complex disorders that do not respond well to more traditional target-specific treatments, such as central nervous system, immune system, and cardiovascular diseases. Still, there is yet to be an established benchmark suite for assessing the effectiveness of AI tools for designing multi-target compounds. Standardized benchmarks allow for comparing existing techniques and promote rapid research progress. Hence, this work proposes an evaluation framework for molecule generation techniques in MTDD scenarios, considering brain diseases as a case study. Our methodology involves using large language models to select the appropriate molecular targets, gathering and preprocessing the bioassay datasets, training quantitative structure-activity relationship models to predict target modulation, and assessing other essential drug-likeness properties for implementing the benchmarks. Additionally, this work will assess the performance of four deep generative models and evolutionary algorithms over our benchmark suite. In our findings, both evolutionary algorithms and generative models can achieve competitive results across the proposed benchmarks.

Read more

8/21/2024

🛸

Total Score

0

A Review on Fragment-based De Novo 2D Molecule Generation

Sergei Voloboev

In the field of computational molecule generation, an essential task in the discovery of new chemical compounds, fragment-based deep generative models are a leading approach, consistently achieving state-of-the-art results in molecular design benchmarks as of 2023. We present a detailed comparative assessment of their architectures, highlighting their unique approaches to molecular fragmentation and generative modeling. This review also includes comparisons of output quality, generation speed, and the current limitations of specific models. We also highlight promising avenues for future research that could bridge fragment-based models to real-world applications.

Read more

5/10/2024