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

Read original: arXiv:2407.00004 - Published 7/2/2024 by Ayush Noori, I~naki Arango, William E. Byrd, Nada Amin
Total Score

0

🤖

Sign in to get full access

or

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

Overview

  • Researchers used multi-objective generative AI to design small molecules that can cross the blood-brain barrier and bind to the dopamine D2 receptor, a key target for antipsychotic drugs.
  • They built predictive models for properties like blood-brain barrier permeability and safety, then used a Monte Carlo Tree Search algorithm to generate a diverse library of 26,581 novel drug-like molecules.
  • Top scoring molecules were validated through molecular docking simulations, showing predicted binding affinity comparable to an existing antipsychotic drug.
  • This computational approach could enable discovery of new treatments for central nervous system disorders.

Plain English Explanation

The blood-brain barrier is a highly selective filter that makes it very difficult for drugs to reach the brain and nervous system. Computational methods to generate BBB permeable drugs in silico may be valuable tools in the CNS drug design pipeline. However, simply crossing the blood-brain barrier is not enough - the drug molecule must also bind to a specific target or receptor in the brain and be safe and non-toxic.

To address this challenge, the researchers used a type of artificial intelligence called multi-objective generative AI to design small drug-like molecules that can cross the blood-brain barrier and bind to the dopamine D2 receptor. The D2 receptor is an important target for many existing antipsychotic medications.

The researchers first built computational models to predict key properties like blood-brain barrier permeability and safety/toxicity profiles. They then adapted a Monte Carlo Tree Search algorithm called SyntheMol, originally developed for antibiotic design, to generate a diverse library of 26,581 novel small molecules that satisfy these constraints.

Top scoring molecules from this library were validated through molecular docking simulations, which showed they could bind to the D2 receptor with predicted affinity comparable to an existing clinically used antipsychotic drug.

Overall, this computational approach could enable the discovery of new treatments for difficult-to-treat central nervous system disorders by systematically designing drug candidates that can effectively reach and engage targets in the brain.

Technical Explanation

The researchers first developed computational models to predict key properties of small molecules, including blood-brain barrier (BBB) permeability and safety/toxicity profiles, using graph neural network-based property predictors.

They then adapted the SyntheMol algorithm, a Monte Carlo Tree Search-based method originally developed for antibiotic design, to perform a multi-objective guided traversal over a space of easily synthesizable molecules. The goal was to computationally synthesize a diverse library of novel drug-like small molecules with high predicted BBB permeability and favorable predicted safety/toxicity, targeting the dopamine D2 receptor.

The resulting library contained 26,581 unique molecules. The researchers validated the top scoring compounds through molecular docking simulations, which demonstrated predicted binding affinity for the D2 receptor comparable to the clinically used antipsychotic drug risperidone.

Critical Analysis

The researchers acknowledge that while their computational approach enables the generation of BBB-permeable small molecules, further experimental validation would be required to confirm biological activity and safety. The paper does not provide details on how the researchers selected the specific target (D2 receptor) or whether this target is considered an optimal or clinically validated therapeutic target for the disorders they aim to address.

Additionally, the authors do not discuss potential issues around the synthetic accessibility or scalability of producing the designed molecules in the wet lab. The use of a Monte Carlo Tree Search algorithm also raises questions about the efficiency and determinism of the molecule generation process compared to other generative AI approaches.

Overall, the research represents an interesting proof-of-concept for using multi-objective generative AI to design drug candidates tailored for central nervous system delivery. However, significant additional work would be needed to translate these computational results into viable clinical candidates.

Conclusion

This research demonstrates the potential of multi-objective generative AI techniques to aid the discovery of novel small molecule drugs capable of penetrating the blood-brain barrier and binding to specific neurological targets. By systematically designing molecules with predicted BBB permeability, target binding, and safety attributes, the researchers were able to generate a diverse library of compounds that could be further validated experimentally.

If successful, this computational approach could accelerate the development of much-needed new treatments for central nervous system disorders that have traditionally been difficult to drug. However, significant work remains to translate these in silico results into real-world therapeutics. Continued progress in areas like predictive modeling, generative algorithms, and experimental validation will be crucial to realizing the full potential of this technology.



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

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

Efficient Training of Transformers for Molecule Property Prediction on Small-scale Datasets
Total Score

0

Efficient Training of Transformers for Molecule Property Prediction on Small-scale Datasets

Shivesh Prakash

The blood-brain barrier (BBB) serves as a protective barrier that separates the brain from the circulatory system, regulating the passage of substances into the central nervous system. Assessing the BBB permeability of potential drugs is crucial for effective drug targeting. However, traditional experimental methods for measuring BBB permeability are challenging and impractical for large-scale screening. Consequently, there is a need to develop computational approaches to predict BBB permeability. This paper proposes a GPS Transformer architecture augmented with Self Attention, designed to perform well in the low-data regime. The proposed approach achieved a state-of-the-art performance on the BBB permeability prediction task using the BBBP dataset, surpassing existing models. With a ROC-AUC of 78.8%, the approach sets a state-of-the-art by 5.5%. We demonstrate that standard Self Attention coupled with GPS transformer performs better than other variants of attention coupled with GPS Transformer.

Read more

9/10/2024

👀

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

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