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

Read original: arXiv:2408.10482 - Published 8/21/2024 by Arthur Cerveira, Frederico Kremer, Darling de Andrade Lourenc{c}o, Ulisses B Corr^ea
Total Score

0

👀

Sign in to get full access

or

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

Overview

  • This paper presents an evaluation framework for AI-driven molecular design of multi-target drugs, focusing on brain diseases as a case study.
  • The framework combines evolutionary algorithms and deep generative models to generate novel drug candidates that can interact with multiple therapeutic targets.
  • The approach aims to address the challenges of developing effective treatments for complex brain disorders by considering the polypharmacological nature of these diseases.

Plain English Explanation

The paper describes a new way to design drugs that can interact with multiple targets in the brain to treat complex disorders. Current drug development often focuses on finding a single drug that targets one specific problem. However, many brain diseases involve complex interactions between different biological systems, so a single-target drug may not be enough.

The researchers' approach uses a combination of two powerful AI techniques - evolutionary algorithms and deep generative models. Evolutionary algorithms mimic the process of natural selection to gradually improve potential drug candidates over many iterations. Deep generative models can then create novel drug-like molecules from scratch, drawing inspiration from successful candidates identified by the evolutionary process.

By using this hybrid AI approach, the researchers hope to generate drug candidates that can address the multiple, interconnected factors involved in brain diseases. This could lead to more effective treatments for conditions like Alzheimer's, Parkinson's, and psychiatric disorders, which often have complex underlying causes.

The paper focuses on brain diseases as an important case study, but the evaluation framework could potentially be applied to developing multi-target drugs for other complex diseases as well.

Technical Explanation

The paper's evaluation framework integrates evolutionary algorithms and deep generative models to tackle the challenge of discovering multi-target drugs for brain diseases. Evolutionary algorithms are used to iteratively optimize and improve candidate drug molecules, while deep generative models are employed to generate novel, drug-like molecular structures.

The evolutionary algorithm component of the framework uses a multi-objective optimization approach to simultaneously optimize a candidate drug's ability to bind to multiple therapeutic targets relevant to brain diseases. The deep generative model, inspired by techniques like conditional VAEs and target-aware molecule generation, is then used to explore the chemical space and propose novel molecular structures that exhibit the desired multi-target binding properties.

The authors test their framework on a case study focused on developing drugs for brain diseases, where the ability to modulate multiple targets is crucial for addressing the complex, multi-factorial nature of these disorders. The results demonstrate the potential of this hybrid AI approach to generate promising multi-target drug candidates that warrant further experimental validation.

Critical Analysis

The paper presents a well-designed evaluation framework that leverages the complementary strengths of evolutionary algorithms and deep generative models for multi-target drug discovery. The authors acknowledge several limitations and avenues for future research, including the need to further validate the generated drug candidates through wet-lab experiments and the potential to expand the framework to consider additional factors, such as drug-like properties and pharmacokinetics.

One potential concern is the reliance on accurate and comprehensive target binding data, which can be challenging to obtain, especially for complex brain diseases. The framework's performance may be sensitive to the quality and completeness of the target binding information used to guide the optimization process.

Additionally, while the case study focuses on brain diseases, the authors could consider discussing the broader applicability of the framework to other complex, multi-factorial disease areas, as well as the potential challenges and adaptations required when scaling the approach to real-world drug discovery pipelines.

Overall, the paper presents a promising AI-driven approach to address the unmet need for effective multi-target drugs, particularly in the context of brain disorders. Further advancements in this direction could have significant implications for the development of more holistic and personalized therapies.

Conclusion

This paper introduces an innovative evaluation framework that combines evolutionary algorithms and deep generative models to tackle the challenge of designing multi-target drugs for complex brain diseases. By leveraging the strengths of these complementary AI techniques, the framework can generate promising drug candidates that can modulate multiple therapeutic targets relevant to brain disorders.

The case study on brain diseases highlights the potential of this approach to address the polypharmacological nature of such conditions, which often involve intricate interactions between different biological systems. While further experimental validation is needed, the presented framework represents a significant step forward in the quest for more effective and personalized treatments for complex neurological and psychiatric disorders.



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

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

Multi-objective generative AI for designing novel brain-targeting small molecules

Ayush Noori, I~naki Arango, William E. Byrd, Nada Amin

The strict selectivity of the blood-brain barrier (BBB) represents one of the most formidable challenges to successful central nervous system (CNS) drug delivery. Computational methods to generate BBB permeable drugs in silico may be valuable tools in the CNS drug design pipeline. However, in real-world applications, BBB penetration alone is insufficient; rather, after transiting the BBB, molecules must bind to a specific target or receptor in the brain and must also be safe and non-toxic. To discover small molecules that concurrently satisfy these constraints, we use multi-objective generative AI to synthesize drug-like BBB-permeable small molecules. Specifically, we computationally synthesize molecules with predicted binding affinity against dopamine receptor D2, the primary target for many clinically effective antipsychotic drugs. After training several graph neural network-based property predictors, we adapt SyntheMol (Swanson et al., 2024), a recently developed Monte Carlo Tree Search-based algorithm for antibiotic design, to perform a multi-objective guided traversal over an easily synthesizable molecular space. We design a library of 26,581 novel and diverse small molecules containing hits with high predicted BBB permeability and favorable predicted safety and toxicity profiles, and that could readily be synthesized for experimental validation in the wet lab. We also validate top scoring molecules with molecular docking simulation against the D2 receptor and demonstrate predicted binding affinity on par with risperidone, a clinically prescribed D2-targeting antipsychotic. In the future, the SyntheMol-based computational approach described here may enable the discovery of novel neurotherapeutics for currently intractable disorders of the CNS.

Read more

7/2/2024

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

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