Predicting T-Cell Receptor Specificity

Read original: arXiv:2407.19349 - Published 7/30/2024 by Tengyao Tu, Wei Zeng, Kun Zhao, Zhenyu Zhang
Total Score

0

↗️

Sign in to get full access

or

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

Overview

  • Researchers developed a framework to efficiently screen out T-cell receptors (TCRs) and target antigens, and achieve TCR specificity prediction.
  • The framework consists of an antigen selector and a TCR classifier based on the Random Forest algorithm.
  • The researchers used k-fold validation to compare the performance of their model with ordinary deep learning methods.

Plain English Explanation

The human body's immune system uses T-cells to recognize and fight off threats like cancer cells. Each T-cell has a unique receptor (TCR) that can bind to specific targets, called antigens. Understanding the specificity of TCRs is important for developing immunotherapy, a type of cancer treatment that harnesses the power of the immune system.

The researchers created a framework to help identify which TCRs can recognize which antigens. This involves two main components: an "antigen selector" that screens potential antigens, and a "TCR classifier" that predicts whether a given TCR can bind to a particular antigen. The TCR classifier uses a machine learning algorithm called Random Forest, which is effective at this kind of prediction task.

The researchers tested their framework by comparing its performance to ordinary deep learning methods. They used a technique called k-fold validation, which involves repeatedly training and testing the model on different subsets of data. This helps ensure the results are reliable and not just due to chance.

Overall, the researchers found that adding the Random Forest classifier to their model significantly improved its ability to accurately predict TCR-antigen binding. Their framework outperformed the regular deep learning approaches. The researchers also discussed ways to further optimize their model to address any remaining challenges.

Technical Explanation

The researchers established a TCR generative specificity detection framework consisting of two key components:

  1. Antigen Selector: This component screens potential target antigens to identify the most relevant ones for a given TCR.

  2. TCR Classifier: This component uses a Random Forest algorithm to classify whether a TCR can bind to a particular antigen.

To evaluate their framework, the researchers used the k-fold validation method. This involves repeatedly splitting the data into training and testing sets, training the model on the training set, and evaluating its performance on the testing set. Averaging the results across multiple folds helps ensure the model's performance is not due to chance.

The researchers compared the performance of their TCR specificity detection framework to ordinary deep learning methods. Their results showed that adding the Random Forest-based TCR classifier significantly improved the model's ability to accurately predict TCR-antigen binding compared to the deep learning-only approaches.

Critical Analysis

The researchers acknowledged that their framework has some limitations and challenges that require further optimization:

  • Antigen Screening Accuracy: The performance of the overall framework is dependent on the accuracy of the antigen selector component. Improving the antigen screening process could lead to better TCR-antigen binding predictions.

  • TCR Classifier Robustness: While the Random Forest-based TCR classifier outperformed the deep learning-only models, there may be opportunities to further improve its robustness and generalization capabilities.

  • Computational Efficiency: The researchers noted that the current implementation of their framework may not be computationally efficient enough for real-world deployment. Optimizing the computational complexity could make the system more practical.

Additionally, it would be valuable to see the framework tested on a wider range of datasets and scenarios to better understand its broader applicability and limitations. Validating the model's performance on clinical data would also be an important next step.

Conclusion

This research presents a promising framework for efficiently screening TCRs and target antigens to achieve TCR specificity prediction. By incorporating a Random Forest-based TCR classifier, the researchers were able to outperform ordinary deep learning methods in predicting TCR-antigen binding.

The insights gained from this work can contribute to the development of more effective cancer immunotherapies by helping identify the most relevant TCR-antigen interactions. Further optimization and validation of the framework could lead to even more accurate cancer classification and personalized treatment strategies.



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

Predicting T-Cell Receptor Specificity

Tengyao Tu, Wei Zeng, Kun Zhao, Zhenyu Zhang

Researching the specificity of TCR contributes to the development of immunotherapy and provides new opportunities and strategies for personalized cancer immunotherapy. Therefore, we established a TCR generative specificity detection framework consisting of an antigen selector and a TCR classifier based on the Random Forest algorithm, aiming to efficiently screen out TCRs and target antigens and achieve TCR specificity prediction. Furthermore, we used the k-fold validation method to compare the performance of our model with ordinary deep learning methods. The result proves that adding a classifier to the model based on the random forest algorithm is very effective, and our model generally outperforms ordinary deep learning methods. Moreover, we put forward feasible optimization suggestions for the shortcomings and challenges of our model found during model implementation.

Read more

7/30/2024

Contrastive learning of T cell receptor representations
Total Score

0

Contrastive learning of T cell receptor representations

Yuta Nagano, Andrew Pyo, Martina Milighetti, James Henderson, John Shawe-Taylor, Benny Chain, Andreas Tiffeau-Mayer

Computational prediction of the interaction of T cell receptors (TCRs) and their ligands is a grand challenge in immunology. Despite advances in high-throughput assays, specificity-labelled TCR data remains sparse. In other domains, the pre-training of language models on unlabelled data has been successfully used to address data bottlenecks. However, it is unclear how to best pre-train protein language models for TCR specificity prediction. Here we introduce a TCR language model called SCEPTR (Simple Contrastive Embedding of the Primary sequence of T cell Receptors), capable of data-efficient transfer learning. Through our model, we introduce a novel pre-training strategy combining autocontrastive learning and masked-language modelling, which enables SCEPTR to achieve its state-of-the-art performance. In contrast, existing protein language models and a variant of SCEPTR pre-trained without autocontrastive learning are outperformed by sequence alignment-based methods. We anticipate that contrastive learning will be a useful paradigm to decode the rules of TCR specificity.

Read more

6/11/2024

TCR-GPT: Integrating Autoregressive Model and Reinforcement Learning for T-Cell Receptor Repertoires Generation
Total Score

0

TCR-GPT: Integrating Autoregressive Model and Reinforcement Learning for T-Cell Receptor Repertoires Generation

Yicheng Lin, Dandan Zhang, Yun Liu

T-cell receptors (TCRs) play a crucial role in the immune system by recognizing and binding to specific antigens presented by infected or cancerous cells. Understanding the sequence patterns of TCRs is essential for developing targeted immune therapies and designing effective vaccines. Language models, such as auto-regressive transformers, offer a powerful solution to this problem by learning the probability distributions of TCR repertoires, enabling the generation of new TCR sequences that inherit the underlying patterns of the repertoire. We introduce TCR-GPT, a probabilistic model built on a decoder-only transformer architecture, designed to uncover and replicate sequence patterns in TCR repertoires. TCR-GPT demonstrates an accuracy of 0.953 in inferring sequence probability distributions measured by Pearson correlation coefficient. Furthermore, by leveraging Reinforcement Learning(RL), we adapted the distribution of TCR sequences to generate TCRs capable of recognizing specific peptides, offering significant potential for advancing targeted immune therapies and vaccine development. With the efficacy of RL, fine-tuned pretrained TCR-GPT models demonstrated the ability to produce TCR repertoires likely to bind specific peptides, illustrating RL's efficiency in enhancing the model's adaptability to the probability distributions of biologically relevant TCR sequences.

Read more

8/6/2024

📈

Total Score

0

A unified cross-attention model for predicting antigen binding specificity to both HLA and TCR molecules

Chenpeng Yu, Xing Fang, Hui Liu

The immune checkpoint inhibitors have demonstrated promising clinical efficacy across various tumor types, yet the percentage of patients who benefit from them remains low. The binding affinity between antigens and HLA-I/TCR molecules plays a critical role in antigen presentation and T-cell activation. Some computational methods have been developed to predict antigen-HLA or antigen-TCR binding specificity, but they focus solely on one task at a time. In this paper, we propose UnifyImmun, a unified cross-attention transformer model designed to simultaneously predicts the binding of antigens to both HLA and TCR molecules, thereby providing more comprehensive evaluation of antigen immunogenicity. We devise a two-phase progressive training strategy that enables these two tasks to mutually reinforce each other, by compelling the encoders to extract more expressive features. To further enhance the model generalizability, we incorporate virtual adversarial training. Compared to over ten existing methods for predicting antigen-HLA and antigen-TCR binding, our method demonstrates better performance in both tasks. Notably, on a large-scale COVID-19 antigen-TCR binding test set, our method improves performance by at least 9% compared to the current state-of-the-art methods. The validation experiments on three clinical cohorts confirm that our approach effectively predicts immunotherapy response and clinical outcomes. Furthermore, the cross-attention scores reveal the amino acids sites critical for antigen binding to receptors. In essence, our approach marks a significant step towards comprehensive evaluation of antigen immunogenicity.

Read more

5/14/2024