De novo antibody design with SE(3) diffusion

Read original: arXiv:2405.07622 - Published 5/14/2024 by Daniel Cutting, Fr'ed'eric A. Dreyer, David Errington, Constantin Schneider, Charlotte M. Deane
Total Score

0

👀

Sign in to get full access

or

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

Overview

  • The researchers introduce IgDiff, a new antibody variable domain diffusion model based on a general protein backbone diffusion framework.
  • IgDiff can generate highly designable antibodies with novel binding regions, while maintaining good agreement with the distribution of a reference antibody dataset.
  • The researchers experimentally validate the designed antibodies, finding they all express with high yield.
  • IgDiff outperforms a state-of-the-art generative backbone diffusion model on various antibody design tasks.

Plain English Explanation

The researchers have developed a new computational model called IgDiff that can design novel antibody structures. Antibodies are Y-shaped proteins used by the immune system to recognize and neutralize foreign substances. The variable regions of the antibody are the parts that bind to the target.

IgDiff uses a diffusion model to generate new antibody structures by starting with random shapes and slowly refining them to match the characteristics of natural antibodies. This approach allows IgDiff to efficiently explore the space of possible antibody structures and identify ones that are both designable (i.e., can be manufactured) and contain novel binding regions.

The researchers show that the antibodies generated by IgDiff have backbone structures that align well with those found in natural antibodies. They also experimentally validate the designed antibodies, confirming that they can all be produced with high yields, an important practical consideration.

Compared to a previous state-of-the-art model, IgDiff demonstrates improved performance on a variety of antibody design tasks, such as designing the complementarity-determining regions or pairing a light chain to an existing heavy chain. This suggests IgDiff could be a valuable tool for accelerating the development of new antibody-based therapies.

Technical Explanation

The researchers develop IgDiff, an antibody variable domain diffusion model that extends a general protein backbone diffusion framework to handle multiple chains. The key innovations are:

  1. Modeling the variable domain regions of antibodies, which are responsible for binding to targets, rather than the full antibody structure.
  2. Incorporating a diffusion process to efficiently explore the space of possible antibody structures.
  3. Extending the diffusion framework to support multiple protein chains, as antibodies consist of both heavy and light chains.

To assess the designability and novelty of the generated structures, the researchers analyze the backbone dihedral angles of the sampled antibodies and find good agreement with a reference antibody distribution. They also experimentally validate the designed antibodies, confirming they can all be expressed with high yields.

The researchers compare IgDiff to a state-of-the-art generative backbone diffusion model on several antibody design tasks, such as designing the complementarity-determining regions or pairing a light chain to an existing heavy chain. IgDiff demonstrates improved properties and designability compared to the baseline.

Critical Analysis

The researchers acknowledge some limitations of their work, such as the need to further validate the designed antibodies' binding affinity and specificity. They also note that the model is currently limited to the variable domain regions and does not yet generate full antibody structures.

Additionally, while the experimentally validated antibodies all expressed well, the researchers do not provide details on the specific expression levels or other biophysical characteristics that would be important for real-world therapeutic applications.

The researchers could also have explored the model's ability to generate antibodies targeting specific antigens or diseases, which would better demonstrate its utility for practical antibody discovery and development.

Overall, the IgDiff model represents a promising step forward in using diffusion-based generative methods for antibody design, but further research and validation will be needed to fully assess its capabilities and limitations.

Conclusion

The IgDiff model introduces a novel approach to designing antibody variable domains using a diffusion-based generative framework. By efficiently exploring the space of possible antibody structures, IgDiff can generate highly designable antibodies with novel binding regions while maintaining structural characteristics consistent with natural antibodies.

The experimental validation of the designed antibodies, and the improved performance on various antibody design tasks compared to a state-of-the-art baseline, suggest that IgDiff could be a valuable tool for accelerating the development of new antibody-based therapies. However, additional research is needed to further assess the model's capabilities and limitations, particularly in terms of binding affinity, specificity, and real-world therapeutic applications.



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

De novo antibody design with SE(3) diffusion

Daniel Cutting, Fr'ed'eric A. Dreyer, David Errington, Constantin Schneider, Charlotte M. Deane

We introduce IgDiff, an antibody variable domain diffusion model based on a general protein backbone diffusion framework which was extended to handle multiple chains. Assessing the designability and novelty of the structures generated with our model, we find that IgDiff produces highly designable antibodies that can contain novel binding regions. The backbone dihedral angles of sampled structures show good agreement with a reference antibody distribution. We verify these designed antibodies experimentally and find that all express with high yield. Finally, we compare our model with a state-of-the-art generative backbone diffusion model on a range of antibody design tasks, such as the design of the complementarity determining regions or the pairing of a light chain to an existing heavy chain, and show improved properties and designability.

Read more

5/14/2024

Improving Antibody Design with Force-Guided Sampling in Diffusion Models
Total Score

0

Improving Antibody Design with Force-Guided Sampling in Diffusion Models

Paulina Kulyt.e, Francisco Vargas, Simon Valentin Mathis, Yu Guang Wang, Jos'e Miguel Hern'andez-Lobato, Pietro Li`o

Antibodies, crucial for immune defense, primarily rely on complementarity-determining regions (CDRs) to bind and neutralize antigens, such as viruses. The design of these CDRs determines the antibody's affinity and specificity towards its target. Generative models, particularly denoising diffusion probabilistic models (DDPMs), have shown potential to advance the structure-based design of CDR regions. However, only a limited dataset of bound antibody-antigen structures is available, and generalization to out-of-distribution interfaces remains a challenge. Physics based force-fields, which approximate atomic interactions, offer a coarse but universal source of information to better mold designs to target interfaces. Integrating this foundational information into diffusion models is, therefore, highly desirable. Here, we propose a novel approach to enhance the sampling process of diffusion models by integrating force field energy-based feedback. Our model, DiffForce, employs forces to guide the diffusion sampling process, effectively blending the two distributions. Through extensive experiments, we demonstrate that our method guides the model to sample CDRs with lower energy, enhancing both the structure and sequence of the generated antibodies.

Read more

9/10/2024

Antibody DomainBed: Out-of-Distribution Generalization in Therapeutic Protein Design
Total Score

0

Antibody DomainBed: Out-of-Distribution Generalization in Therapeutic Protein Design

Natav{s}a Tagasovska, Ji Won Park, Matthieu Kirchmeyer, Nathan C. Frey, Andrew Martin Watkins, Aya Abdelsalam Ismail, Arian Rokkum Jamasb, Edith Lee, Tyler Bryson, Stephen Ra, Kyunghyun Cho

Machine learning (ML) has demonstrated significant promise in accelerating drug design. Active ML-guided optimization of therapeutic molecules typically relies on a surrogate model predicting the target property of interest. The model predictions are used to determine which designs to evaluate in the lab, and the model is updated on the new measurements to inform the next cycle of decisions. A key challenge is that the experimental feedback from each cycle inspires changes in the candidate proposal or experimental protocol for the next cycle, which lead to distribution shifts. To promote robustness to these shifts, we must account for them explicitly in the model training. We apply domain generalization (DG) methods to classify the stability of interactions between an antibody and antigen across five domains defined by design cycles. Our results suggest that foundational models and ensembling improve predictive performance on out-of-distribution domains. We publicly release our codebase extending the DG benchmark ``DomainBed,'' and the associated dataset of antibody sequences and structures emulating distribution shifts across design cycles.

Read more

8/1/2024

AntibodyFlow: Normalizing Flow Model for Designing Antibody Complementarity-Determining Regions
Total Score

0

AntibodyFlow: Normalizing Flow Model for Designing Antibody Complementarity-Determining Regions

Bohao Xu, Yanbo Wang, Wenyu Chen, Shimin Shan

Therapeutic antibodies have been extensively studied in drug discovery and development in the past decades. Antibodies are specialized protective proteins that bind to antigens in a lock-to-key manner. The binding strength/affinity between an antibody and a specific antigen is heavily determined by the complementarity-determining regions (CDRs) on the antibodies. Existing machine learning methods cast in silico development of CDRs as either sequence or 3D graph (with a single chain) generation tasks and have achieved initial success. However, with CDR loops having specific geometry shapes, learning the 3D geometric structures of CDRs remains a challenge. To address this issue, we propose AntibodyFlow, a 3D flow model to design antibody CDR loops. Specifically, AntibodyFlow first constructs the distance matrix, then predicts amino acids conditioned on the distance matrix. Also, AntibodyFlow conducts constraint learning and constrained generation to ensure valid 3D structures. Experimental results indicate that AntibodyFlow outperforms the best baseline consistently with up to 16.0% relative improvement in validity rate and 24.3% relative reduction in geometric graph level error (root mean square deviation, RMSD).

Read more

6/21/2024