1
|
Wan YT, Nielsen M. TCRCluster: a novel approach to T-cell receptor latent featurization and clustering using contrastive learning-guided two-stage variational autoencoders. NAR Genom Bioinform 2025; 7:lqaf065. [PMID: 40432791 PMCID: PMC12107435 DOI: 10.1093/nargab/lqaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
T cells play a vital role in adaptive immunity by targeting pathogen-infected or cancerous cells, but predicting their specificity remains challenging. Encoding T-cell receptor (TCR) sequences into informative feature spaces is therefore crucial for advancing specificity prediction and downstream applications. For this, we developed a variational autoencoder (VAE)-based model trained on paired TCR α-β chain data, incorporating all six complementarity-determining regions. A semi-supervised 'two-stage VAE' framework, integrating cosine triplet loss and a classifier, was found to further refine peptide-specific latent representations, outperforming sequence-based methods in specificity prediction. Clustering analyses leveraging our VAE latent space were evaluated using K-means, agglomerative clustering, and a novel graph-based method. Agglomerative clustering achieved the most biologically relevant results, balancing cluster purity and retention despite noise in TCR specificity annotations. We extended these insights to evaluate TCR repertoire data. Across datasets, VAE-based models outperformed sequence-based methods, particularly in retention metrics, with notable improvements in the SARS-CoV-2 repertoire dataset. Moreover, the cancer repertoire analysis highlighted the generalizability of our approach, where the model displayed high performance despite minimal similarity between the training and test data. Collectively, these results demonstrate the potential of VAE-based latent representations to offer a robust framework for prediction, clustering, and repertoire analysis.
Collapse
Affiliation(s)
- Yat-Tsai Richie Wan
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby DK 28002, Denmark
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby DK 28002, Denmark
| |
Collapse
|
2
|
Castorina LV, Grazioli F, Machart P, Mösch A, Errica F. Assessing the generalization capabilities of TCR binding predictors via peptide distance analysis. PLoS One 2025; 20:e0324011. [PMID: 40392871 PMCID: PMC12091837 DOI: 10.1371/journal.pone.0324011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
Understanding the interaction between T Cell Receptors (TCRs) and peptide-bound Major Histocompatibility Complexes (pMHCs) is crucial for comprehending immune responses and developing targeted immunotherapies. While recent machine learning (ML) models show remarkable success in predicting TCR-pMHC binding within training data, these models often fail to generalize to peptides outside their training distributions, raising concerns about their applicability in therapeutic settings. Understanding and improving the generalization of these models is therefore critical to ensure real-world applications. To address this issue, we evaluate the effect of the distance between training and testing peptide distributions on ML model empirical risk assessments, using sequence-based and 3D structure-based distance metrics. In our analysis we use several state-of-the-art models for TCR-peptide binding prediction: Attentive Variational Information Bottleneck (AVIB), NetTCR-2.0 and -2.2, and ERGO II (pre-trained autoencoder) and ERGO II (LSTM). In this work, we introduce a novel approach for assessing the generalization capabilities of TCR binding predictors: the Distance Split (DS) algorithm. The DS algorithm controls the distance between training and testing peptides based on both sequence and structure, allowing for a more nuanced evaluation of model performance. We show that lower 3D shape similarity between training and test peptides is associated with a harder out-of-distribution task definition, which is more interesting when measuring the ability to generalize to unseen peptides. However, we observe the opposite effect when splitting using sequence-based similarity. These findings highlight the importance of using a distance-based splitting approach to benchmark models. This could then be used to estimate a confidence score on predictions on novel and unseen peptides, based on how different they are from the training ones. Additionally, our results may hint that employing 3D shape to complement sequence information could improve the accuracy of TCR-pMHC binding predictors.
Collapse
Affiliation(s)
- Leonardo V. Castorina
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
- NEC Laboratories Europe, Heidelberg, Germany
| | | | | | - Anja Mösch
- NEC Laboratories Europe, Heidelberg, Germany
| | | |
Collapse
|
3
|
Gao L, Zhang Y, Ge F, Li S, Guo Y, Song J, Yu DJ. Structure-Directed Pan-Specific T-Cell Receptor-Peptide-Major Histocompatibility Complex Interaction Prediction. J Chem Inf Model 2025; 65:4674-4686. [PMID: 40297927 DOI: 10.1021/acs.jcim.5c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
T-cell receptors (TCRs) play a pivotal role in the adaptive immune system, and understanding their antigen recognition mechanism remains a critical area of research. With the increasing availability of binding and interaction data between TCRs and peptide-major histocompatibility complexes (pMHCs), data-driven computational methods are emerging as powerful tools with significant potential for advancement. In this study, we collected and curated comprehensive sequence and structure data sets of TCRs from human CD8+ T-cells and cognate epitopes presented by MHC class I molecules. We developed two innovative computational frameworks: SG-TPMI, a lightweight, extensible, and structure-guided model for predicting TCR-pMHC binding specificity, and Seq/Struct-TCS, a pair of models (sequence-based and structure-based) for predicting contact sites within TCR-pMHC complexes. Notably, we directly integrated MHC-I alpha helices (or pseudosequences) and structural information on the protein complex into the prediction models. Our comprehensive modeling approach enabled quantitative investigations of TCR-pMHC interaction mechanisms, empowering SG-TPMI and Struct-TCS to achieve performances comparable to those of state-of-the-art methods. Furthermore, our results highlight the necessity of CDR1 and CDR2 loops as well as MHC restriction in pan-specific TCR-pMHC interaction prediction, providing new insights into TCR recognition. In summary, we not only propose SG-TPMI as an effective computational method for predicting TCR-pMHC binary interactions but also introduce the Seq/Struct-TCS design for predicting TCR interacting sites with peptide or MHC alpha helices.
Collapse
Affiliation(s)
- Letao Gao
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Yumeng Zhang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Fang Ge
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210003,China
| | - Shanshan Li
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Yuming Guo
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| |
Collapse
|
4
|
Tang X, Deng J, He C, Xu Y, Bai S, Guo Z, Du G, Ouyang D, Sun X. Application of in-silico approaches in subunit vaccines: Overcoming the challenges of antigen and adjuvant development. J Control Release 2025; 381:113629. [PMID: 40086761 DOI: 10.1016/j.jconrel.2025.113629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Subunit vaccines are crucial in preventing modern diseases due to their safety, stability, and ability to elicit targeted immune responses. However, challenges in antigen and adjuvant design hinder their development. Recent advancements in in-silico approaches, including reverse vaccinology, structural vaccinology, and machine learning, have revolutionized vaccine development from empirical practices to rational design approaches. This review summarizes the transformative impact of in-silico approaches on subunit vaccine development. We address the challenges of antigen identification and designation, highlighting how advanced computational techniques are employed to accelerate antigen acquisition. We also examine the challenges in adjuvant discovery and illustrate how machine learning helps overcome these barriers. Finally, we explore potential future directions for subunit vaccines, highlighting the importance of combining computational methods with other technologies to tackle the challenges associated with subunit vaccine development.
Collapse
Affiliation(s)
- Xue Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiayin Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuting Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhaofei Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China; DPM, Faculty of Health Sciences, University of Macau, Macao SAR, China.
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
5
|
Ge J, Wang J, Ye Q, Pan L, Kang Y, Shen C, Deng Y, Hsieh CY, Hou T. TRAP: a contrastive learning-enhanced framework for robust TCR-pMHC binding prediction with improved generalizability. Chem Sci 2025:d4sc08141b. [PMID: 40321182 PMCID: PMC12046420 DOI: 10.1039/d4sc08141b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
The binding of T cell receptors (TCRs) to peptide-MHC I (pMHC) complexes is critical for triggering adaptive immune responses to potential health threats. Developing highly accurate machine learning (ML) models to predict TCR-pMHC binding could significantly accelerate immunotherapy advancements. However, existing ML models for TCR-pMHC binding prediction often underperform with unseen epitopes, severely limiting their applicability. We introduce TRAP, which leverages contrastive learning to enhance model performance by aligning structural and sequence features of pMHC with TCR sequences. TRAP outperforms previous state-of-the-art models in both random and unseen epitope scenarios, achieving an AUPR of 0.84 (a 22% improvement over the second-best model) and an AUC of 0.92 in the random scenario, and an AUC of 0.75 (almost 11% higher than the second-best model) in the unseen epitope scenario. Furthermore, TRAP demonstrates a noteworthy capability to diagnose potential issues of cross-reactivity between TCRs and similar epitopes. This highly robust performance makes it a suitable tool for large-scale predictions in real-world settings. A specific case study confirmed that TRAP can discover hit TCRs with binding free energies comparable to referenced experimental results. These findings highlight TRAP's potential for practical applications and its role as a powerful tool in developing TCR-based immunotherapies.
Collapse
Affiliation(s)
- Jingxuan Ge
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
- CarbonSilicon AI Technology Company, Ltd Hangzhou 310018 Zhejiang China
| | - Jike Wang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
- CarbonSilicon AI Technology Company, Ltd Hangzhou 310018 Zhejiang China
| | - Qing Ye
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Liqiang Pan
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Chao Shen
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Yafeng Deng
- CarbonSilicon AI Technology Company, Ltd Hangzhou 310018 Zhejiang China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University Hangzhou 310058 Zhejiang China
| |
Collapse
|
6
|
Ullanat V, Jing B, Sledzieski S, Berger B. Learning the language of protein-protein interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642188. [PMID: 40166198 PMCID: PMC11956943 DOI: 10.1101/2025.03.09.642188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Protein Language Models (PLMs) trained on large databases of protein sequences have proven effective in modeling protein biology across a wide range of applications. However, while PLMs excel at capturing individual protein properties, they face challenges in natively representing protein-protein interactions (PPIs), which are crucial to understanding cellular processes and disease mechanisms. Here, we introduce MINT, a PLM specifically designed to model sets of interacting proteins in a contextual and scalable manner. Using unsupervised training on a large curated PPI dataset derived from the STRING database, MINT outperforms existing PLMs in diverse tasks relating to protein-protein interactions, including binding affinity prediction and estimation of mutational effects. Beyond these core capabilities, it excels at modeling interactions in complex protein assemblies and surpasses specialized models in antibody-antigen modeling and T cell receptor-epitope binding prediction. MINT's predictions of mutational impacts on oncogenic PPIs align with experimental studies, and it provides reliable estimates for the potential for cross-neutralization of antibodies against SARS-CoV-2 variants of concern. These findings position MINT as a powerful tool for elucidating complex protein interactions, with significant implications for biomedical research and therapeutic discovery.
Collapse
Affiliation(s)
- Varun Ullanat
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Bowen Jing
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Samuel Sledzieski
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Center for Computational Biology, Flatiron Insitute, New York, NY
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Department of Mathematics, Massachusetts Institute of Technology, MA
| |
Collapse
|
7
|
Metoikidou C, Karnaukhov V, Boeckx B, Timperi E, Bonté PE, Wang L, Espenel M, Albaud B, Loirat D, Wang X, Sotiriou C, Aftimos P, Punie K, Wildiers H, Labroska V, Wang MW, Waterfall JJ, Piccart-Gebhart M, Mora T, Walczak A, Lantz O, Buisseret L, Lambrechts D, Amigorena S, Romano E. Continuous replenishment of the dysfunctional CD8 T cell axis is associated with response to chemoimmunotherapy in advanced breast cancer. Cell Rep Med 2025; 6:101973. [PMID: 39983715 PMCID: PMC11970331 DOI: 10.1016/j.xcrm.2025.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 08/18/2024] [Accepted: 01/22/2025] [Indexed: 02/23/2025]
Abstract
Chemotherapy combined with immune checkpoint blockade has shown clinical activity in breast cancer. Response, however, occurs in only a low proportion of patients. How the immune landscape of the tumor determines the immune and clinical responses to chemoimmunotherapy is not well understood. Here, using a combination of single-cell RNA sequencing (scRNA-seq) and single-cell T cell receptor sequencing (scTCR-seq), we profile 40 biopsies from 27 patients with metastatic triple-negative breast cancer (TNBC), receiving chemotherapy and anti-PD-L1 alone or in combination with anti-CD73, in a phase 2 randomized clinical trial. Our results show an enrichment of late-dysfunctional, clonally expanded CD8+ T cells in responder (R) patients. On treatment, R display an influx of newly emerging clonotypes, as well as expansion of the CD8+ precursors. Collectively, our data suggest that baseline clonal expansion could be a potential predictor of response and that both clonal reinvigoration of pre-existing tumor-reactive T cells and clonal replacement on-treatment are important for a protective response to chemoimmunotherapy.
Collapse
Affiliation(s)
- Christina Metoikidou
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France; Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vadim Karnaukhov
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France; Laboratoire de Physique de l'École Normale Supérieure, Paris Sciences & Lettres University, CNRS, Sorbonne Université and Université Paris Cité, 75005 Paris, France
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Eleonora Timperi
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France
| | - Pierre-Emmanuel Bonté
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France
| | - Ling Wang
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Marion Espenel
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie, 75005 Paris, France
| | - Benoit Albaud
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie, 75005 Paris, France
| | - Delphine Loirat
- Department of Medical Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France
| | - Xiaoxiao Wang
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, Leuven, Belgium; University Hospitals Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, Leuven, Belgium; University Hospitals Leuven, Leuven, Belgium
| | - Viktorija Labroska
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming-Wei Wang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joshua J Waterfall
- Translational Research Department, Institut Curie, 75005 Paris, France; INSERM U830, Institut Curie, 75005 Paris, France
| | | | - Thierry Mora
- Laboratoire de Physique de l'École Normale Supérieure, Paris Sciences & Lettres University, CNRS, Sorbonne Université and Université Paris Cité, 75005 Paris, France
| | - Aleksandra Walczak
- Laboratoire de Physique de l'École Normale Supérieure, Paris Sciences & Lettres University, CNRS, Sorbonne Université and Université Paris Cité, 75005 Paris, France
| | - Olivier Lantz
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France; Laboratoire d'immunologie clinique, Institut Curie, 75005 Paris, France; Centre d'investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428), Paris, France
| | | | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Sebastian Amigorena
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France
| | - Emanuela Romano
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, 75005 Paris, France; Department of Medical Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France.
| |
Collapse
|
8
|
Quast NP, Abanades B, Guloglu B, Karuppiah V, Harper S, Raybould MIJ, Deane CM. T-cell receptor structures and predictive models reveal comparable alpha and beta chain structural diversity despite differing genetic complexity. Commun Biol 2025; 8:362. [PMID: 40038394 PMCID: PMC11880327 DOI: 10.1038/s42003-025-07708-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 02/09/2025] [Indexed: 03/06/2025] Open
Abstract
T-cell receptor (TCR) structures are currently under-utilised in early-stage drug discovery and repertoire-scale informatics. Here, we leverage a large dataset of solved TCR structures from Immunocore to evaluate the current state-of-the-art for TCR structure prediction, and identify which regions of the TCR remain challenging to model. Through clustering analyses and the training of a TCR-specific model capable of large-scale structure prediction, we find that the alpha chain VJ-recombined loop (CDR3α) is as structurally diverse and correspondingly difficult to predict as the beta chain VDJ-recombined loop (CDR3β). This differentiates TCR variable domain loops from the genetically analogous antibody loops and supports the conjecture that both TCR alpha and beta chains are deterministic of antigen specificity. We hypothesise that the larger number of alpha chain joining genes compared to beta chain joining genes compensates for the lack of a diversity gene segment. We also provide over 1.5M predicted TCR structures to enable repertoire structural analysis and elucidate strategies towards improving the accuracy of future TCR structure predictors. Our observations reinforce the importance of paired TCR sequence information and capture the current state-of-the-art for TCR structure prediction, while our model and 1.5M structure predictions enable the use of structural TCR information at an unprecedented scale.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Humans
- Models, Molecular
- Genetic Variation
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Protein Conformation
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/chemistry
Collapse
Affiliation(s)
- Nele P Quast
- Department of Statistics, University of Oxford, Oxford, UK
| | | | - Bora Guloglu
- Department of Statistics, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
9
|
Li J, Zhang Y, Hu L, Ye H, Yan X, Li X, Li Y, Ye S, Wu B, Li Z. T-cell Receptor Repertoire Analysis in the Context of Transarterial Chemoembolization Synergy with Systemic Therapy for Hepatocellular Carcinoma. J Clin Transl Hepatol 2025; 13:69-83. [PMID: 39801788 PMCID: PMC11712086 DOI: 10.14218/jcth.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/16/2025] Open
Abstract
T-cell receptor (TCR) sequencing provides a novel platform for insight into and characterization of intricate T-cell profiles, advancing the understanding of tumor immune heterogeneity. Recently, transarterial chemoembolization (TACE) combined with systemic therapy has become the recommended regimen for advanced hepatocellular carcinoma. The regulation of the immune microenvironment after TACE and its impact on tumor progression and recurrence has been a focus of research. By examining and tracking fluctuations in the TCR repertoire following combination treatment, novel perspectives on the modulation of the tumor microenvironment post-TACE and the underlying mechanisms governing tumor progression and recurrence can be gained. Clarifying the distinctive metrics and dynamic alterations of the TCR repertoire within the context of combination therapy is imperative for understanding the mechanisms of anti-tumor immunity, assessing efficacy, exploiting novel treatments, and further advancing precision oncology in the treatment of hepatocellular carcinoma. In this review, we initially summarized the fundamental characteristics of TCR repertoire and depicted immune microenvironment remodeling after TACE. Ultimately, we illustrated the prospective applications of TCR repertoires in TACE combined with systemic therapy.
Collapse
Affiliation(s)
- Jie Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Luqi Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Heqing Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xingli Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yifan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Shuwen Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Bailu Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Wohlwend J, Nathan A, Shalon N, Crain CR, Tano-Menka R, Goldberg B, Richards E, Gaiha GD, Barzilay R. Deep learning enhances the prediction of HLA class I-presented CD8 + T cell epitopes in foreign pathogens. NAT MACH INTELL 2025; 7:232-243. [PMID: 40008296 PMCID: PMC11847706 DOI: 10.1038/s42256-024-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/10/2024] [Indexed: 02/27/2025]
Abstract
Accurate in silico determination of CD8+ T cell epitopes would greatly enhance T cell-based vaccine development, but current prediction models are not reliably successful. Here, motivated by recent successes applying machine learning to complex biology, we curated a dataset of 651,237 unique human leukocyte antigen class I (HLA-I) ligands and developed MUNIS, a deep learning model that identifies peptides presented by HLA-I alleles. MUNIS shows improved performance compared with existing models in predicting peptide presentation and CD8+ T cell epitope immunodominance hierarchies. Moreover, application of MUNIS to proteins from Epstein-Barr virus led to successful identification of both established and novel HLA-I epitopes which were experimentally validated by in vitro HLA-I-peptide stability and T cell immunogenicity assays. MUNIS performs comparably to an experimental stability assay in terms of immunogenicity prediction, suggesting that deep learning can reduce experimental burden and accelerate identification of CD8+ T cell epitopes for rapid T cell vaccine development.
Collapse
Affiliation(s)
- Jeremy Wohlwend
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Anusha Nathan
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA USA
| | - Nitan Shalon
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Charles R. Crain
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | - Rhoda Tano-Menka
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | | | - Emma Richards
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | - Gaurav D. Gaiha
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA USA
| | - Regina Barzilay
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
11
|
Chandora K, Chandora A, Saeed A, Cavalcante L. Adoptive T Cell Therapy Targeting MAGE-A4. Cancers (Basel) 2025; 17:413. [PMID: 39941782 PMCID: PMC11815873 DOI: 10.3390/cancers17030413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
MAGE A4 (Melanoma Antigen Gene A4) is a cancer testis antigen (CTA) that is expressed normally in germline cells (testis/embryonic tissues) but absent in somatic cells. The MAGE A4 CTA is expressed in a variety of tumor types, like synovial sarcoma, ovarian cancer and non-small cell lung cancer. Having its expression profile limited to germline cells has made MAGE A4 a sought-after immunotherapeutic target in certain malignancies. In this review, we focus on MAGE-A4's function and expression, current clinical trials involving targeted immunotherapy approaches, and challenges and opportunities facing MAGE-A4's targeted therapeutics.
Collapse
Affiliation(s)
- Kapil Chandora
- Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA 30310, USA; (K.C.)
| | - Akshay Chandora
- Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA 30310, USA; (K.C.)
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Ludimila Cavalcante
- Division of Hematology and Oncology, University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22903, USA
| |
Collapse
|
12
|
Teimouri H, Ghoreyshi ZS, Kolomeisky AB, George JT. Feature selection enhances peptide binding predictions for TCR-specific interactions. Front Immunol 2025; 15:1510435. [PMID: 39916960 PMCID: PMC11799297 DOI: 10.3389/fimmu.2024.1510435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
Introduction T-cell receptors (TCRs) play a critical role in the immune response by recognizing specific ligand peptides presented by major histocompatibility complex (MHC) molecules. Accurate prediction of peptide binding to TCRs is essential for advancing immunotherapy, vaccine design, and understanding mechanisms of autoimmune disorders. Methods This study presents a theoretical approach that explores the impact of feature selection techniques on enhancing the predictive accuracy of peptide binding models tailored for specific TCRs. To evaluate our approach across different TCR systems, we utilized a dataset that includes peptide libraries tested against three distinct murine TCRs. A broad range of physicochemical properties, including amino acid composition, dipeptide composition, and tripeptide features, were integrated into the machine learning-based feature selection framework to identify key properties contributing to binding affinity. Results Our analysis reveals that leveraging optimized feature subsets not only simplifies the model complexity but also enhances predictive performance, enabling more precise identification of TCR peptide interactions. The results of our feature selection method are consistent with findings from hybrid approaches that utilize both sequence and structural data as input as well as experimental data. Discussion Our theoretical approach highlights the role of feature selection in peptide-TCR interactions, providing a quantitative tool for uncovering the molecular mechanisms of the T-cell response and assisting in the design of more advanced targeted therapeutics.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Zahra S. Ghoreyshi
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Anatoly B. Kolomeisky
- Department of Chemistry, Rice University, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, United States
| | - Jason T. George
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
- Department of Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, Houston, TX, United States
- Department of Translational Medical Sciences, Texas A&M Health Science Center, Houston, TX, United States
| |
Collapse
|
13
|
Nagano Y, Pyo AGT, Milighetti M, Henderson J, Shawe-Taylor J, Chain B, Tiffeau-Mayer A. Contrastive learning of T cell receptor representations. Cell Syst 2025; 16:101165. [PMID: 39778580 DOI: 10.1016/j.cels.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
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-labeled TCR data remain sparse. In other domains, the pre-training of language models on unlabeled 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), which is capable of data-efficient transfer learning. Through our model, we introduce a pre-training strategy combining autocontrastive learning and masked-language modeling, 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. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yuta Nagano
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; Division of Medicine, University College London, London WC1E 6BT, UK
| | - Andrew G T Pyo
- Center for the Physics of Biological Function, Princeton University, Princeton, NJ 08544, USA
| | - Martina Milighetti
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; Cancer Institute, University College London, London WC1E 6DD, UK
| | - James Henderson
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK
| | - John Shawe-Taylor
- Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Benny Chain
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Andreas Tiffeau-Mayer
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, London WC1E 6BT, UK.
| |
Collapse
|
14
|
Luo S, Notaro A, Lin L. ATLAS-seq: a microfluidic single-cell TCR screen for antigen-reactive TCRs. Nat Commun 2025; 16:216. [PMID: 39746936 PMCID: PMC11696065 DOI: 10.1038/s41467-024-54675-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Discovering antigen-reactive T cell receptors (TCRs) is central to developing effective engineered T cell immunotherapies. However, the conventional technologies for isolating antigen-reactive TCRs (i.e., major histocompatibility complex (MHC) multimer staining) focus on high-affinity interactions between the TCR and MHC-antigen complex, and may fail to identify TCRs with high efficacy for activating T cells. Here, we develop a microfluidic single-cell screening method for antigen-reactive T cells named ATLAS-seq (Aptamer-based T Lymphocyte Activity Screening and SEQuencing). This technology isolates and characterizes activated T cells via an aptamer-based fluorescent molecular sensor, which monitors the cytotoxic cytokine IFNγ secretion from single T cells upon antigen stimulation, followed by single-cell RNA and single-cell TCR sequencing. We use ATLAS-seq to screen TCRs reactive to cytomegalovirus (CMV) or prostate specific antigen (PSA) from peripheral blood mononuclear cells (PBMCs). ATLAS-seq identifies distinct TCR clonotype populations with higher T cell activation levels compared to TCRs recovered by MHC multimer staining. Select TCR clonotypes from ATLAS-seq are more efficient in target cell killing than those from MHC multimer staining. Collectively, ATLAS-seq provides an efficient and broadly applicable technology to screen antigen-reactive TCRs for engineered T cell immunotherapy.
Collapse
Affiliation(s)
- Siwei Luo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amber Notaro
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lan Lin
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Ruiz Ortega M, Pogorelyy MV, Minervina AA, Thomas PG, Mora T, Walczak AM. Learning predictive signatures of HLA type from T-cell repertoires. PLoS Comput Biol 2025; 21:e1012724. [PMID: 39761303 PMCID: PMC11737854 DOI: 10.1371/journal.pcbi.1012724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 01/16/2025] [Accepted: 12/16/2024] [Indexed: 01/15/2025] Open
Abstract
T cells recognize a wide range of pathogens using surface receptors that interact directly with peptides presented on major histocompatibility complexes (MHC) encoded by the HLA loci in humans. Understanding the association between T cell receptors (TCR) and HLA alleles is an important step towards predicting TCR-antigen specificity from sequences. Here we analyze the TCR alpha and beta repertoires of large cohorts of HLA-typed donors to systematically infer such associations, by looking for overrepresentation of TCRs in individuals with a common allele.TCRs, associated with a specific HLA allele, exhibit sequence similarities that suggest prior antigen exposure. Immune repertoire sequencing has produced large numbers of datasets, however the HLA type of the corresponding donors is rarely available. Using our TCR-HLA associations, we trained a computational model to predict the HLA type of individuals from their TCR repertoire alone. We propose an iterative procedure to refine this model by using data from large cohorts of untyped individuals, by recursively typing them using the model itself. The resulting model shows good predictive performance, even for relatively rare HLA alleles.
Collapse
Affiliation(s)
- María Ruiz Ortega
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL Université, Sorbonne Université, and Université Paris-Cité, Paris, France
| | - Mikhail V. Pogorelyy
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Anastasia A. Minervina
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Paul G. Thomas
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Thierry Mora
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL Université, Sorbonne Université, and Université Paris-Cité, Paris, France
| | - Aleksandra M. Walczak
- Laboratoire de physique de l’École Normale Supérieure, CNRS, PSL Université, Sorbonne Université, and Université Paris-Cité, Paris, France
| |
Collapse
|
16
|
Gray GI, Chukwuma PC, Eldaly B, Perera WWJG, Brambley CA, Rosales TJ, Baker BM. The Evolving T Cell Receptor Recognition Code: The Rules Are More Like Guidelines. Immunol Rev 2025; 329:e13439. [PMID: 39804137 PMCID: PMC11771984 DOI: 10.1111/imr.13439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025]
Abstract
αβ T cell receptor (TCR) recognition of peptide-MHC complexes lies at the core of adaptive immunity, balancing specificity and cross-reactivity to facilitate effective antigen discrimination. Early structural studies established basic frameworks helpful for understanding and contextualizing TCR recognition and features such as peptide specificity and MHC restriction. However, the growing TCR structural database and studies launched from structural work continue to reveal exceptions to common assumptions and simplifications derived from earlier work. Here we explore our evolving understanding of TCR recognition, illustrating how structural and biophysical investigations regularly uncover complex phenomena that push against paradigms and expand our understanding of how TCRs bind to and discriminate between peptide/MHC complexes. We discuss the implications of these findings for basic, translational, and predictive immunology, including the challenges in accounting for the inherent adaptability, flexibility, and occasional biophysical sloppiness that characterize TCR recognition.
Collapse
MESH Headings
- Humans
- Animals
- Protein Binding
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
- Peptides/immunology
- Peptides/metabolism
- T-Lymphocytes/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Major Histocompatibility Complex
- Protein Conformation
Collapse
Affiliation(s)
- George I. Gray
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - P. Chukwunalu Chukwuma
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Bassant Eldaly
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - W. W. J. Gihan Perera
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Chad A. Brambley
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Tatiana J. Rosales
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Brian M. Baker
- Department of Chemistry and Biochemistry and the Haper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
17
|
Wang M, Fan W, Wu T, Li M. TPepRet: a deep learning model for characterizing T-cell receptors-antigen binding patterns. Bioinformatics 2024; 41:btaf022. [PMID: 39880376 PMCID: PMC11784750 DOI: 10.1093/bioinformatics/btaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025] Open
Abstract
MOTIVATION T-cell receptors (TCRs) elicit and mediate the adaptive immune response by recognizing antigenic peptides, a process pivotal for cancer immunotherapy, vaccine design, and autoimmune disease management. Understanding the intricate binding patterns between TCRs and peptides is critical for advancing these clinical applications. While several computational tools have been developed, they neglect the directional semantics inherent in sequence data, which are essential for accurately characterizing TCR-peptide interactions. RESULTS To address this gap, we develop TPepRet, an innovative model that integrates subsequence mining with semantic integration capabilities. TPepRet combines the strengths of the Bidirectional Gated Recurrent Unit (BiGRU) network for capturing bidirectional sequence dependencies with the Large Language Model framework to analyze subsequences and global sequences comprehensively, which enables TPepRet to accurately decipher the semantic binding relationship between TCRs and peptides. We have evaluated TPepRet to a range of challenging scenarios, including performance benchmarking against other tools using diverse datasets, analysis of peptide binding preferences, characterization of T cells clonal expansion, identification of true binder in complex environments, assessment of key binding sites through alanine scanning, validation against expression rates from large-scale datasets, and ability to screen SARS-CoV-2 TCRs. The comprehensive results suggest that TPepRet outperforms existing tools. We believe TPepRet will become an effective tool for understanding TCR-peptide binding in clinical treatment. AVAILABILITY AND IMPLEMENTATION The source code can be obtained from https://github.com/CSUBioGroup/TPepRet.git.
Collapse
Affiliation(s)
- Meng Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Wei Fan
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX39DU, United Kingdom
| | - Tianrui Wu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
18
|
许 珉, 张 斯, 鲁 曼, 高 媛, 张 梦, 林 勇, 谢 鹭. [Prediction of MHC II antigen peptide-T cell receptors binding based on foundation model]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:1243-1249. [PMID: 40000215 PMCID: PMC11955369 DOI: 10.7507/1001-5515.202405024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/01/2024] [Indexed: 02/27/2025]
Abstract
The specific binding of T cell receptors (TCRs) to antigenic peptides plays a key role in the regulation and mediation of the immune process and provides an essential basis for the development of tumour vaccines. In recent years, studies have mainly focused on TCR prediction of major histocompatibility complex (MHC) class I antigens, but TCR prediction of MHC class II antigens has not been sufficiently investigated and there is still much room for improvement. In this study, the combination of MHC class II antigen peptide and TCR prediction was investigated using the ProtT5 grand model to explore its feature extraction capability. In addition, the model was fine-tuned to retain the underlying features of the model, and a feed-forward neural network structure was constructed for fusion to achieve the prediction model. The experimental results showed that the method proposed in this study performed better than the traditional methods, with a prediction accuracy of 0.96 and an AUC of 0.93, which verifies the effectiveness of the model proposed in this paper.
Collapse
Affiliation(s)
- 珉瑞 许
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
- 上海市生物医药技术研究院 上海市疾病与健康基因组学重点实验室(上海 200237)Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, P. R. China
| | - 斯文 张
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - 曼曼 鲁
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - 媛 高
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - 梦欢 张
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - 勇 林
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - 鹭 谢
- 上海理工大学 健康科学与工程学院(上海 200093)School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
- 上海市生物医药技术研究院 上海市疾病与健康基因组学重点实验室(上海 200237)Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, P. R. China
| |
Collapse
|
19
|
Vegesana K, Thomas PG. Cracking the code of adaptive immunity: The role of computational tools. Cell Syst 2024; 15:1156-1167. [PMID: 39701033 DOI: 10.1016/j.cels.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/14/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024]
Abstract
In recent years, the advances in high-throughput and deep sequencing have generated a diverse amount of adaptive immune repertoire data. This surge in data has seen a proportional increase in computational methods aimed to characterize T cell receptor (TCR) repertoires. In this perspective, we will provide a brief commentary on the various domains of TCR repertoire analysis, their respective computational methods, and the ongoing challenges. Given the breadth of methods and applications of TCR analysis, we will focus our perspective on sequence-based computational methods.
Collapse
Affiliation(s)
- Kasi Vegesana
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
20
|
Yadav S, Vora DS, Sundar D, Dhanjal JK. TCR-ESM: Employing protein language embeddings to predict TCR-peptide-MHC binding. Comput Struct Biotechnol J 2024; 23:165-173. [PMID: 38146434 PMCID: PMC10749252 DOI: 10.1016/j.csbj.2023.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Cognate target identification for T-cell receptors (TCRs) is a significant barrier in T-cell therapy development, which may be overcome by accurately predicting TCR interaction with peptide-bound major histocompatibility complex (pMHC). In this study, we have employed peptide embeddings learned from a large protein language model- Evolutionary Scale Modeling (ESM), to predict TCR-pMHC binding. The TCR-ESM model presented outperforms existing predictors. The complementarity-determining region 3 (CDR3) of the hypervariable TCR is located at the center of the paratope and plays a crucial role in peptide recognition. TCR-ESM trained on paired TCR data with both CDR3α and CDR3β chain information performs significantly better than those trained on data with only CDR3β, suggesting that both TCR chains contribute to specificity, the relative importance however depends on the specific peptide-MHC targeted. The study illuminates the importance of MHC information in TCR-peptide binding which remained inconclusive so far and was thought dependent on the dataset characteristics. TCR-ESM outperforms existing approaches on external datasets, suggesting generalizability. Overall, the potential of deep learning for predicting TCR-pMHC interactions and improving the understanding of factors driving TCR specificity are highlighted. The prediction model is available at http://tcresm.dhanjal-lab.iiitd.edu.in/ as an online tool.
Collapse
Affiliation(s)
- Shashank Yadav
- Department of Biomedical Engineering, University of Arizona, Tucson 85721, AZ, USA
| | - Dhvani Sandip Vora
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Jaspreet Kaur Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| |
Collapse
|
21
|
Hu W, Bian Y, Ji H. TIL Therapy in Lung Cancer: Current Progress and Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409356. [PMID: 39422665 PMCID: PMC11633538 DOI: 10.1002/advs.202409356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Lung cancer remains the most prevalent malignant tumor worldwide and is the leading cause of cancer-related mortality. Although immune checkpoint blockade has revolutionized the treatment of advanced lung cancer, many patients still do not respond well, often due to the lack of functional T cell infiltration. Adoptive cell therapy (ACT) using expanded immune cells has emerged as an important therapeutic modality. Tumor-infiltrating lymphocytes (TIL) therapy is one form of ACT involving the administration of expanded and activated autologous T cells derived from surgically resected cancer tissues and reinfusion into patients and holds great therapeutic potential for lung cancer. In this review, TIL therapy is introduced and its suitability for lung cancer is discussed. Then its historical and clinical developments are summarized, and the methods developed up-to-date to identify tumor-recognizing TILs and optimize TIL composition. Some perspectives toward future TIL therapy for lung cancer are also provided.
Collapse
Affiliation(s)
- Weilei Hu
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yifei Bian
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hongbin Ji
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
- School of Life Science and TechnologyShanghai Tech UniversityShanghai200120China
| |
Collapse
|
22
|
Mahdy AKH, Lokes E, Schöpfel V, Kriukova V, Britanova OV, Steiert TA, Franke A, ElAbd H. Bulk T cell repertoire sequencing (TCR-Seq) is a powerful technology for understanding inflammation-mediated diseases. J Autoimmun 2024; 149:103337. [PMID: 39571301 DOI: 10.1016/j.jaut.2024.103337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/12/2024] [Accepted: 11/09/2024] [Indexed: 12/15/2024]
Abstract
Multiple alterations in the T cell repertoire were identified in many chronic inflammatory diseases such as inflammatory bowel disease, multiple sclerosis, and rheumatoid arthritis, suggesting that T cells might, directly or indirectly, be implicated in these pathologies. This has sparked a deep interest in characterizing disease-associated T cell clonotypes as well as in identifying and quantifying their contribution to the pathophysiology of different autoimmune and inflammation-mediated diseases. Bulk T cell repertoire sequencing (TCR-Seq) has emerged as a powerful method to profile the T cell repertoire of a sample in a high throughput fashion. Given the increasing utilization of TCR-Seq, we aimed here to provide a comprehensive, up-to-date review of the method, its extensions, and its ability to investigate chronic and autoimmune diseases. Specifically, we started by introducing the immunological basis of TCR repertoire generation and features, followed by discussing different experimental approach to perform TCR-Seq, then we describe different methods and frameworks for analyzing the generated datasets. Subsequently, different experimental techniques for investigating the antigenicity of T cell clonotypes are described. Lastly, we discuss recent studies that utilized TCR-Seq to understand different inflammation-mediated diseases, discuss fallbacks of the technology and potential future directions to overcome current limitations.
Collapse
Affiliation(s)
- Aya K H Mahdy
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Evgeniya Lokes
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Valentina Schöpfel
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Valeriia Kriukova
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Olga V Britanova
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Tim A Steiert
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany.
| | - Hesham ElAbd
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany.
| |
Collapse
|
23
|
Mullan KA, Ha M, Valkiers S, de Vrij N, Ogunjimi B, Laukens K, Meysman P. T cell receptor-centric perspective to multimodal single-cell data analysis. SCIENCE ADVANCES 2024; 10:eadr3196. [PMID: 39612336 PMCID: PMC11606500 DOI: 10.1126/sciadv.adr3196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
The T cell receptor (TCR), despite its importance, is underutilized in single-cell analysis, with gene expression features solely driving current strategies. Here, we argue for a TCR-first approach, more suited toward T cell repertoires. To this end, we curated a large T cell atlas from 12 prominent human studies, containing in total 500,000 T cells spanning multiple diseases, including melanoma, head and neck cancer, blood cancer, and lung transplantation. Here, we identified severe limitations in cell-type annotation using unsupervised approaches and propose a more robust standard using a semi-supervised method or the TCR arrangement. We showcase the utility of a TCR-first approach through application of the STEGO.R tool for the identification of treatment-related dynamics and previously unknown public T cell clusters with potential antigen-specific properties. Thus, the paradigm shift to a TCR-first can highlight overlooked key T cell features that have the potential for improvements in immunotherapy and diagnostics.
Collapse
Affiliation(s)
- Kerry A. Mullan
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| | - My Ha
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Valkiers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| | - Nicky de Vrij
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
Pham MDN, Su CTT, Nguyen TN, Nguyen HN, Nguyen DDA, Giang H, Nguyen DT, Phan MD, Nguyen V. epiTCR-KDA: knowledge distillation model on dihedral angles for TCR-peptide prediction. BIOINFORMATICS ADVANCES 2024; 4:vbae190. [PMID: 39678207 PMCID: PMC11646569 DOI: 10.1093/bioadv/vbae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/03/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Motivation The prediction of the T-cell receptor (TCR) and antigen bindings is crucial for advancements in immunotherapy. However, most current TCR-peptide interaction predictors struggle to perform well on unseen data. This limitation may stem from the conventional use of TCR and/or peptide sequences as input, which may not adequately capture their structural characteristics. Therefore, incorporating the structural information of TCRs and peptides into the prediction model is necessary to improve its generalizability. Results We developed epiTCR-KDA (KDA stands for Knowledge Distillation model on Dihedral Angles), a new predictor of TCR-peptide binding that utilizes the dihedral angles between the residues of the peptide and the TCR as a structural descriptor. This structural information was integrated into a knowledge distillation model to enhance its generalizability. epiTCR-KDA demonstrated competitive prediction performance, with an area under the curve (AUC) of 1.00 for seen data and AUC of 0.91 for unseen data. On public datasets, epiTCR-KDA consistently outperformed other predictors, maintaining a median AUC of 0.93. Further analysis of epiTCR-KDA revealed that the cosine similarity of the dihedral angle vectors between the unseen testing data and training data is crucial for its stable performance. In conclusion, our epiTCR-KDA model represents a significant step forward in developing a highly effective pipeline for antigen-based immunotherapy. Availability and implementation epiTCR-KDA is available on GitHub (https://github.com/ddiem-ri-4D/epiTCR-KDA).
Collapse
Affiliation(s)
- My-Diem Nguyen Pham
- Faculty of Information Technology, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | | | | | | | - Dinh Duy An Nguyen
- Department of Genetics and Genomic Sciences School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States
| | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | - Dinh-Thuc Nguyen
- Faculty of Information Technology, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Minh-Duy Phan
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
- NexCalibur Therapeutics, DE, United States
| | - Vy Nguyen
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| |
Collapse
|
25
|
Feng X, Huo M, Li H, Yang Y, Jiang Y, He L, Cheng Li S. A comprehensive benchmarking for evaluating TCR embeddings in modeling TCR-epitope interactions. Brief Bioinform 2024; 26:bbaf030. [PMID: 39883514 PMCID: PMC11781202 DOI: 10.1093/bib/bbaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/17/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
The complexity of T cell receptor (TCR) sequences, particularly within the complementarity-determining region 3 (CDR3), requires efficient embedding methods for applying machine learning to immunology. While various TCR CDR3 embedding strategies have been proposed, the absence of their systematic evaluations created perplexity in the community. Here, we extracted CDR3 embedding models from 19 existing methods and benchmarked these models with four curated datasets by accessing their impact on the performance of TCR downstream tasks, including TCR-epitope binding affinity prediction, epitope-specific TCR identification, TCR clustering, and visualization analysis. We assessed these models utilizing eight downstream classifiers and five downstream clustering methods, with the performance measured by a diverse range of metrics for precision, robustness, and usability. Overall, handcrafted embeddings outperformed data-driven ones in modeling TCR-epitope interactions. To further refine our comparative findings, we developed an all-in-one TCR CDR3 embedding package comprising all evaluated embedding models. This package will assist users in easily selecting suitable embedding models for their data.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Benchmarking
- Humans
- Complementarity Determining Regions/immunology
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Epitopes, T-Lymphocyte/chemistry
- Machine Learning
- Epitopes/immunology
- Computational Biology/methods
Collapse
Affiliation(s)
- Xikang Feng
- School of Software, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an Shaanxi, 710072, China
| | - Miaozhe Huo
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, 999077, China
| | - He Li
- School of Software, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an Shaanxi, 710072, China
| | - Yongze Yang
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, 999077, China
| | - Yuepeng Jiang
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, 999077, China
| | - Liang He
- School of Software, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an Shaanxi, 710072, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, 999077, China
| |
Collapse
|
26
|
Xu L, Yang Q, Dong W, Li X, Wang K, Dong S, Zhang X, Yang T, Luo G, Liao X, Gao X, Wang G. Meta learning for mutant HLA class I epitope immunogenicity prediction to accelerate cancer clinical immunotherapy. Brief Bioinform 2024; 26:bbae625. [PMID: 39656887 DOI: 10.1093/bib/bbae625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Accurate prediction of binding between human leukocyte antigen (HLA) class I molecules and antigenic peptide segments is a challenging task and a key bottleneck in personalized immunotherapy for cancer. Although existing prediction tools have demonstrated significant results using established datasets, most can only predict the binding affinity of antigenic peptides to HLA and do not enable the immunogenic interpretation of new antigenic epitopes. This limitation results from the training data for the computational models relying heavily on a large amount of peptide-HLA (pHLA) eluting ligand data, in which most of the candidate epitopes lack immunogenicity. Here, we propose an adaptive immunogenicity prediction model, named MHLAPre, which is trained on the large-scale MS-derived HLA I eluted ligandome (mostly presented by epitopes) that are immunogenic. Allele-specific and pan-allelic prediction models are also provided for endogenous peptide presentation. Using a meta-learning strategy, MHLAPre rapidly assessed HLA class I peptide affinities across the whole pHLA pairs and accurately identified tumor-associated endogenous antigens. During the process of adaptive immune response of T-cells, pHLA-specific binding in the antigen presentation is only a pre-task for CD8+ T-cell recognition. The key factor in activating the immune response is the interaction between pHLA complexes and T-cell receptors (TCRs). Therefore, we performed transfer learning on the pHLA model using the pHLA-TCR dataset. In pHLA binding task, MHLAPre demonstrated significant improvement in identifying neoepitope immunogenicity compared with five state-of-the-art models, proving its effectiveness and robustness. After transfer learning of the pHLA-TCR data, MHLAPre also exhibited relatively superior performance in revealing the mechanism of immunotherapy. MHLAPre is a powerful tool to identify neoepitopes that can interact with TCR and induce immune responses. We believe that the proposed method will greatly contribute to clinical immunotherapy, such as anti-tumor immunity, tumor-specific T-cell engineering, and personalized tumor vaccine.
Collapse
Affiliation(s)
- Long Xu
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
| | - Qiang Yang
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
- School of Medicine and Health, Harbin Institute of Technology, Yikuang Street, 150000 Harbin, China
| | - Weihe Dong
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, 150040 Harbin, China
| | - Xiaokun Li
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
- School of Computer Science and Technology, Heilongjiang University, Xuefu Road, 150080 Harbin, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Xuefu Road, 150090 Harbin, China
- Shandong Hengxun Technology Co., Ltd., Miaoling Road, 266100 Qingdao, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
| | - Suyu Dong
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, 150040 Harbin, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Haping Road, 150081 Harbin, China
| | - Tiansong Yang
- Department of Rehabilitation, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Xuefu Road, 150040 Harbin, China
| | - Gongning Luo
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, 4700 KAUST Saudi, Arabia
| | - Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University, 710072 Xian, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, 4700 KAUST Saudi, Arabia
| | - Guohua Wang
- School of Computer Science and Technology, Harbin Institute of Technology, West DaZhi Street, 150001 Harbin, China
- College of Computer and Control Engineering, Northeast Forestry University, Hexing Road, 150040 Harbin, China
| |
Collapse
|
27
|
Niu R, Wang J, Li Y, Zhou J, Guo Y, Shang X. Attention-aware differential learning for predicting peptide-MHC class I binding and T cell receptor recognition. Brief Bioinform 2024; 26:bbaf038. [PMID: 39883517 PMCID: PMC11781218 DOI: 10.1093/bib/bbaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
The identification of neoantigens is crucial for advancing vaccines, diagnostics, and immunotherapies. Despite this importance, a fundamental question remains: how to model the presentation of neoantigens by major histocompatibility complex class I molecules and the recognition of the peptide-MHC-I (pMHC-I) complex by T cell receptors (TCRs). Accurate prediction of pMHC-I binding and TCR recognition remains a significant computational challenge in immunology due to intricate binding motifs and the long-tail distribution of known binding pairs in public databases. Here, we propose an attention-aware framework comprising TranspMHC for pMHC-I binding prediction and TransTCR for TCR-pMHC-I recognition prediction. Leveraging the attention mechanism, TranspMHC surpasses existing algorithms on independent datasets at both pan-specific and allele-specific levels. For TCR-pMHC-I recognition, TransTCR incorporates transfer learning and a differential learning strategy, demonstrating superior performance and enhanced generalization on independent datasets compared to existing methods. Furthermore, we identify key amino acids associated with binding motifs of peptides and TCRs that facilitate pMHC-I and TCR-pMHC-I binding, indicating the potential interpretability of our proposed framework.
Collapse
Affiliation(s)
- Rui Niu
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710129 Shaanxi, China
| | - Jingwei Wang
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710129 Shaanxi, China
| | - Yanli Li
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia
| | - Jiren Zhou
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710129 Shaanxi, China
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia
| | - Yang Guo
- School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710129 Shaanxi, China
| |
Collapse
|
28
|
Pham TMQ, Nguyen TN, Tran Nguyen BQ, Diem Tran TP, Diem Pham NM, Phuc Nguyen HT, Cuong Ho TK, Linh Nguyen DV, Nguyen HT, Tran DH, Tran TS, Pham TVN, Le MT, Vy Nguyen TT, Phan MD, Giang H, Nguyen HN, Tran LS. The T cell receptor β chain repertoire of tumor infiltrating lymphocytes improves neoantigen prediction and prioritization. eLife 2024; 13:RP94658. [PMID: 39466298 PMCID: PMC11517254 DOI: 10.7554/elife.94658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
In the realm of cancer immunotherapy, the meticulous selection of neoantigens plays a fundamental role in enhancing personalized treatments. Traditionally, this selection process has heavily relied on predicting the binding of peptides to human leukocyte antigens (pHLA). Nevertheless, this approach often overlooks the dynamic interaction between tumor cells and the immune system. In response to this limitation, we have developed an innovative prediction algorithm rooted in machine learning, integrating T cell receptor β chain (TCRβ) profiling data from colorectal cancer (CRC) patients for a more precise neoantigen prioritization. TCRβ sequencing was conducted to profile the TCR repertoire of tumor-infiltrating lymphocytes (TILs) from 28 CRC patients. The data unveiled both intra-tumor and inter-patient heterogeneity in the TCRβ repertoires of CRC patients, likely resulting from the stochastic utilization of V and J segments in response to neoantigens. Our novel combined model integrates pHLA binding information with pHLA-TCR binding to prioritize neoantigens, resulting in heightened specificity and sensitivity compared to models using individual features alone. The efficacy of our proposed model was corroborated through ELISpot assays on long peptides, performed on four CRC patients. These assays demonstrated that neoantigen candidates prioritized by our combined model outperformed predictions made by the established tool NetMHCpan. This comprehensive assessment underscores the significance of integrating pHLA binding with pHLA-TCR binding analysis for more effective immunotherapeutic strategies.
Collapse
MESH Headings
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/genetics
- Machine Learning
- Algorithms
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Huu Thinh Nguyen
- University Medical Center Ho Chi Minh CityHo Chi Minh CityViet Nam
| | - Duc Huy Tran
- University Medical Center Ho Chi Minh CityHo Chi Minh CityViet Nam
| | - Thanh Sang Tran
- University Medical Center Ho Chi Minh CityHo Chi Minh CityViet Nam
| | | | - Minh Triet Le
- University Medical Center Ho Chi Minh CityHo Chi Minh CityViet Nam
| | | | | | - Hoa Giang
- Medical Genetics InstituteHo Chi Minh CityViet Nam
| | | | - Le Son Tran
- Medical Genetics InstituteHo Chi Minh CityViet Nam
| |
Collapse
|
29
|
Teimouri H, Ghoreyshi ZS, Kolomeisky AB, George JT. Feature Selection Enhances Peptide Binding Predictions for TCR-Specific Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617901. [PMID: 39416168 PMCID: PMC11482946 DOI: 10.1101/2024.10.11.617901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
T-cell receptors (TCRs) play a critical role in the immune response by recognizing specific ligand peptides presented by major histocompatibility complex (MHC) molecules. Accurate prediction of peptide binding to TCRs is essential for advancing immunotherapy, vaccine design, and understanding mechanisms of autoimmune disorders. This study presents a novel theoretical method that explores the impact of feature selection techniques on enhancing the predictive accuracy of peptide binding models tailored for specific TCRs. To evaluate the universality of our approach across different TCR systems, we utilized a dataset that includes peptide libraries tested against three distinct murine TCRs. A broad range of physicochemical properties, including amino acid composition, dipeptide composition, and tripeptide features, were integrated into the machine learning-based feature selection framework to identify key features contributing to binding affinity. Our analysis reveals that leveraging optimized feature subsets not only simplifies the model complexity but also enhances predictive performance, enabling more precise identification of TCR-peptide interactions. The results of our feature selection method are consistent with findings from hybrid approaches that utilize both sequence and structural data as input as well as experimental data. Our theoretical approach highlights the role of feature selection in peptide-TCR interactions, providing a powerful tool for uncovering the molecular mechanisms of the T-cell response and assisting in the design of more advanced targeted therapeutics.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
| | - Zahra S Ghoreyshi
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Anatoly B Kolomeisky
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, 77005, USA
| | - Jason T George
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
30
|
Li F, Qian X, Zhu X, Lai X, Zhang X, Wang J. TCRcost: a deep learning model utilizing TCR 3D structure for enhanced of TCR-peptide binding. Front Genet 2024; 15:1346784. [PMID: 39415981 PMCID: PMC11479912 DOI: 10.3389/fgene.2024.1346784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Predicting TCR-peptide binding is a complex and significant computational problem in systems immunology. During the past decade, a series of computational methods have been developed for better predicting TCR-peptide binding from amino acid sequences. However, the performance of sequence-based methods appears to have hit a bottleneck. Considering the 3D structures of TCR-peptide complexes, which provide much more information, could potentially lead to better prediction outcomes. Methods In this study, we developed TCRcost, a deep learning method, to predict TCR-peptide binding by incorporating 3D structures. TCRcost overcomes two significant challenges: acquiring a sufficient number of high-quality TCR-peptide structures and effectively extracting information from these structures for binding prediction. TCRcost corrects TCR 3D structures generated by protein structure tools, significantly extending the available datasets. The main and side chains of a TCR structure are separately corrected using a long short-term memory (LSTM) model. This approach prevents interference between the chains and accurately extracts interactions among both adjacent and global atoms. A 3D convolutional neural network (CNN) is designed to extract the atomic features relevant to TCR-peptide binding. The spatial features extracted by the 3DCNN are then processed through a fully connected layer to estimate the probability of TCR-peptide binding. Results Test results demonstrated that predicting TCR-peptide binding from 3D TCR structures is both efficient and highly accurate with an average accuracy of 0.974 on precise structures. Furthermore, the average accuracy on corrected structures was 0.762, significantly higher than the average accuracy of 0.375 on uncorrected original structures. Additionally, the average root mean square distance (RMSD) to precise structures was significantly reduced from 12.753 Å for predicted structures to 8.785 Å for corrected structures. Discussion Thus, utilizing structural information of TCR-peptide complexes is a promising approach to improve the accuracy of binding predictions.
Collapse
Affiliation(s)
- Fan Li
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| | - Xinyang Qian
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| | - Xiaoyan Zhu
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| | - Xin Lai
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| | - Xuanping Zhang
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| | - Jiayin Wang
- School of Computer Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Engineering Research Center of Medical and Health Big Data, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
31
|
Scheffer L, Reber EE, Mehta BB, Pavlović M, Chernigovskaya M, Richardson E, Akbar R, Lund-Johansen F, Greiff V, Haff IH, Sandve GK. Predictability of antigen binding based on short motifs in the antibody CDRH3. Brief Bioinform 2024; 25:bbae537. [PMID: 39438077 PMCID: PMC11495870 DOI: 10.1093/bib/bbae537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Adaptive immune receptors, such as antibodies and T-cell receptors, recognize foreign threats with exquisite specificity. A major challenge in adaptive immunology is discovering the rules governing immune receptor-antigen binding in order to predict the antigen binding status of previously unseen immune receptors. Many studies assume that the antigen binding status of an immune receptor may be determined by the presence of a short motif in the complementarity determining region 3 (CDR3), disregarding other amino acids. To test this assumption, we present a method to discover short motifs which show high precision in predicting antigen binding and generalize well to unseen simulated and experimental data. Our analysis of a mutagenesis-based antibody dataset reveals 11 336 position-specific, mostly gapped motifs of 3-5 amino acids that retain high precision on independently generated experimental data. Using a subset of only 178 motifs, a simple classifier was made that on the independently generated dataset outperformed a deep learning model proposed specifically for such datasets. In conclusion, our findings support the notion that for some antibodies, antigen binding may be largely determined by a short CDR3 motif. As more experimental data emerge, our methodology could serve as a foundation for in-depth investigations into antigen binding signals.
Collapse
Affiliation(s)
- Lonneke Scheffer
- Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| | - Eric Emanuel Reber
- Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| | - Brij Bhushan Mehta
- Department of Immunology, University of Oslo, Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Milena Pavlović
- Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| | - Maria Chernigovskaya
- Department of Immunology, University of Oslo, Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Eve Richardson
- La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA, United States
| | - Rahmad Akbar
- Department of Immunology, University of Oslo, Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, University of Oslo, Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Victor Greiff
- Department of Immunology, University of Oslo, Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Ingrid Hobæk Haff
- Department of Mathematics, University of Oslo, Niels Henrik Abels hus, Moltke Moes vei 35, 0851 Oslo, Norway
| | - Geir Kjetil Sandve
- Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| |
Collapse
|
32
|
Kim J, Lee BJ, Moon S, Lee H, Lee J, Kim BS, Jung K, Seo H, Chung Y. Strategies to Overcome Hurdles in Cancer Immunotherapy. Biomater Res 2024; 28:0080. [PMID: 39301248 PMCID: PMC11411167 DOI: 10.34133/bmr.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024] Open
Abstract
Despite marked advancements in cancer immunotherapy over the past few decades, there remains an urgent need to develop more effective treatments in humans. This review explores strategies to overcome hurdles in cancer immunotherapy, leveraging innovative technologies including multi-specific antibodies, chimeric antigen receptor (CAR) T cells, myeloid cells, cancer-associated fibroblasts, artificial intelligence (AI)-predicted neoantigens, autologous vaccines, and mRNA vaccines. These approaches aim to address the diverse facets and interactions of tumors' immune evasion mechanisms. Specifically, multi-specific antibodies and CAR T cells enhance interactions with tumor cells, bolstering immune responses to facilitate tumor infiltration and destruction. Modulation of myeloid cells and cancer-associated fibroblasts targets the tumor's immunosuppressive microenvironment, enhancing immunotherapy efficacy. AI-predicted neoantigens swiftly and accurately identify antigen targets, which can facilitate the development of personalized anticancer vaccines. Additionally, autologous and mRNA vaccines activate individuals' immune systems, fostering sustained immune responses against cancer neoantigens as therapeutic vaccines. Collectively, these strategies are expected to enhance efficacy of cancer immunotherapy, opening new horizons in anticancer treatment.
Collapse
Affiliation(s)
- Jihyun Kim
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, College of Pharmacy,Seoul National University, Seoul 08826, Republic of Korea
| | - Byung Joon Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehoon Moon
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, College of Pharmacy,Seoul National University, Seoul 08826, Republic of Korea
| | - Hojeong Lee
- Department of Anatomy and Cell Biology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Juyong Lee
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, College of Pharmacy,Seoul National University, Seoul 08826, Republic of Korea
- Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
- Arontier Co., Seoul 06735, Republic of Korea
| | - Byung-Soo Kim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, and BioMAX, Seoul National University, Seoul 08826, Republic of Korea
| | - Keehoon Jung
- Department of Anatomy and Cell Biology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyungseok Seo
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, College of Pharmacy,Seoul National University, Seoul 08826, Republic of Korea
| | - Yeonseok Chung
- Research Institute for Pharmaceutical Sciences, College of Pharmacy, College of Pharmacy,Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
33
|
T. RR, Demerdash ONA, Smith JC. TCR-H: explainable machine learning prediction of T-cell receptor epitope binding on unseen datasets. Front Immunol 2024; 15:1426173. [PMID: 39221256 PMCID: PMC11361934 DOI: 10.3389/fimmu.2024.1426173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Artificial-intelligence and machine-learning (AI/ML) approaches to predicting T-cell receptor (TCR)-epitope specificity achieve high performance metrics on test datasets which include sequences that are also part of the training set but fail to generalize to test sets consisting of epitopes and TCRs that are absent from the training set, i.e., are 'unseen' during training of the ML model. We present TCR-H, a supervised classification Support Vector Machines model using physicochemical features trained on the largest dataset available to date using only experimentally validated non-binders as negative datapoints. TCR-H exhibits an area under the curve of the receiver-operator characteristic (AUC of ROC) of 0.87 for epitope 'hard splitting' (i.e., on test sets with all epitopes unseen during ML training), 0.92 for TCR hard splitting and 0.89 for 'strict splitting' in which neither the epitopes nor the TCRs in the test set are seen in the training data. Furthermore, we employ the SHAP (Shapley additive explanations) eXplainable AI (XAI) method for post hoc interrogation to interpret the models trained with different hard splits, shedding light on the key physiochemical features driving model predictions. TCR-H thus represents a significant step towards general applicability and explainability of epitope:TCR specificity prediction.
Collapse
Affiliation(s)
- Rajitha Rajeshwar T.
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, United States
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Omar N. A. Demerdash
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Jeremy C. Smith
- UT/ORNL Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, United States
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| |
Collapse
|
34
|
Kim HY, Kim S, Park WY, Kim D. TSpred: a robust prediction framework for TCR-epitope interactions using paired chain TCR sequence data. Bioinformatics 2024; 40:btae472. [PMID: 39052940 PMCID: PMC11297499 DOI: 10.1093/bioinformatics/btae472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 07/27/2024] Open
Abstract
MOTIVATION Prediction of T-cell receptor (TCR)-epitope interactions is important for many applications in biomedical research, such as cancer immunotherapy and vaccine design. The prediction of TCR-epitope interactions remains challenging especially for novel epitopes, due to the scarcity of available data. RESULTS We propose TSpred, a new deep learning approach for the pan-specific prediction of TCR binding specificity based on paired chain TCR data. We develop a robust model that generalizes well to unseen epitopes by combining the predictive power of CNN and the attention mechanism. In particular, we design a reciprocal attention mechanism which focuses on extracting the patterns underlying TCR-epitope interactions. Upon a comprehensive evaluation of our model, we find that TSpred achieves state-of-the-art performances in both seen and unseen epitope specificity prediction tasks. Also, compared to other predictors, TSpred is more robust to bias related to peptide imbalance in the dataset. In addition, the reciprocal attention component of our model allows for model interpretability by capturing structurally important binding regions. Results indicate that TSpred is a robust and reliable method for the task of TCR-epitope binding prediction. AVAILABILITY AND IMPLEMENTATION Source code is available at https://github.com/ha01994/TSpred.
Collapse
Affiliation(s)
- Ha Young Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | | | - Woong-Yang Park
- GENINUS Inc., Seoul 05836, South Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, South Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| |
Collapse
|
35
|
Ehrlich R, Glynn E, Singh M, Ghersi D. Computational Methods for Predicting Key Interactions in T Cell-Mediated Adaptive Immunity. Annu Rev Biomed Data Sci 2024; 7:295-316. [PMID: 38748864 DOI: 10.1146/annurev-biodatasci-102423-122741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
The adaptive immune system recognizes pathogen- and cancer-specific features and is endowed with memory, enabling it to respond quickly and efficiently to repeated encounters with the same antigens. T cells play a central role in the adaptive immune system by directly targeting intracellular pathogens and helping to activate B cells to secrete antibodies. Several fundamental protein interactions-including those between major histocompatibility complex (MHC) proteins and antigen-derived peptides as well as between T cell receptors and peptide-MHC complexes-underlie the ability of T cells to recognize antigens with great precision. Computational approaches to predict these interactions are increasingly being used for medically relevant applications, including vaccine design and prediction of patient response to cancer immunotherapies. We provide computational researchers with an accessible introduction to the adaptive immune system, review computational approaches to predict the key protein interactions underlying T cell-mediated adaptive immunity, and highlight remaining challenges.
Collapse
Affiliation(s)
- Ryan Ehrlich
- School of Interdisciplinary Informatics, University of Nebraska, Omaha, Nebraska, USA;
| | - Eric Glynn
- Lewis-Sigler Institute, Princeton University, Princeton, New Jersey, USA
| | - Mona Singh
- Department of Computer Science, Princeton University, Princeton, New Jersey, USA;
- Lewis-Sigler Institute, Princeton University, Princeton, New Jersey, USA
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska, Omaha, Nebraska, USA;
| |
Collapse
|
36
|
Pertseva M, Follonier O, Scarcella D, Reddy ST. TCR clustering by contrastive learning on antigen specificity. Brief Bioinform 2024; 25:bbae375. [PMID: 39129361 PMCID: PMC11317525 DOI: 10.1093/bib/bbae375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Effective clustering of T-cell receptor (TCR) sequences could be used to predict their antigen-specificities. TCRs with highly dissimilar sequences can bind to the same antigen, thus making their clustering into a common antigen group a central challenge. Here, we develop TouCAN, a method that relies on contrastive learning and pretrained protein language models to perform TCR sequence clustering and antigen-specificity predictions. Following training, TouCAN demonstrates the ability to cluster highly dissimilar TCRs into common antigen groups. Additionally, TouCAN demonstrates TCR clustering performance and antigen-specificity predictions comparable to other leading methods in the field.
Collapse
Affiliation(s)
- Margarita Pertseva
- Department of Biosystems Science and Engineering, ETH Zurich, Schanzenstrasse 44, 4056 Basel, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Oceane Follonier
- Department of Biosystems Science and Engineering, ETH Zurich, Schanzenstrasse 44, 4056 Basel, Switzerland
| | - Daniele Scarcella
- Department of Biosystems Science and Engineering, ETH Zurich, Schanzenstrasse 44, 4056 Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, Schanzenstrasse 44, 4056 Basel, Switzerland
| |
Collapse
|
37
|
Guasp P, Reiche C, Sethna Z, Balachandran VP. RNA vaccines for cancer: Principles to practice. Cancer Cell 2024; 42:1163-1184. [PMID: 38848720 DOI: 10.1016/j.ccell.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Vaccines are the most impactful medicines to improve health. Though potent against pathogens, vaccines for cancer remain an unfulfilled promise. However, recent advances in RNA technology coupled with scientific and clinical breakthroughs have spurred rapid discovery and potent delivery of tumor antigens at speed and scale, transforming cancer vaccines into a tantalizing prospect. Yet, despite being at a pivotal juncture, with several randomized clinical trials maturing in upcoming years, several critical questions remain: which antigens, tumors, platforms, and hosts can trigger potent immunity with clinical impact? Here, we address these questions with a principled framework of cancer vaccination from antigen detection to delivery. With this framework, we outline features of emergent RNA technology that enable rapid, robust, real-time vaccination with somatic mutation-derived neoantigens-an emerging "ideal" antigen class-and highlight latent features that have sparked the belief that RNA could realize the enduring vision for vaccines against cancer.
Collapse
Affiliation(s)
- Pablo Guasp
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Reiche
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zachary Sethna
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
38
|
Karnaukhov VK, Shcherbinin DS, Chugunov AO, Chudakov DM, Efremov RG, Zvyagin IV, Shugay M. Structure-based prediction of T cell receptor recognition of unseen epitopes using TCRen. NATURE COMPUTATIONAL SCIENCE 2024; 4:510-521. [PMID: 38987378 DOI: 10.1038/s43588-024-00653-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
T cell receptor (TCR) recognition of foreign peptides presented by major histocompatibility complex protein is a major event in triggering the adaptive immune response to pathogens or cancer. The prediction of TCR-peptide interactions has great importance for therapy of cancer as well as infectious and autoimmune diseases but remains a major challenge, particularly for novel (unseen) peptide epitopes. Here we present TCRen, a structure-based method for ranking candidate unseen epitopes for a given TCR. The first stage of the TCRen pipeline is modeling of the TCR-peptide-major histocompatibility complex structure. Then a TCR-peptide residue contact map is extracted from this structure and used to rank all candidate epitopes on the basis of an interaction score with the target TCR. Scoring is performed using an energy potential derived from the statistics of TCR-peptide contact preferences in existing crystal structures. We show that TCRen has high performance in discriminating cognate versus unrelated peptides and can facilitate the identification of cancer neoepitopes recognized by tumor-infiltrating lymphocytes.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Humans
- Peptides/immunology
- Peptides/chemistry
- Epitopes/immunology
- Epitopes/chemistry
- Models, Molecular
- Neoplasms/immunology
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Major Histocompatibility Complex/immunology
- Protein Conformation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
Collapse
Affiliation(s)
- Vadim K Karnaukhov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - Dmitrii S Shcherbinin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anton O Chugunov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Dmitriy M Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.
- Central European Institute of Technology, Brno, Czech Republic.
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Higher School of Economics, Moscow, Russia
| | - Ivan V Zvyagin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mikhail Shugay
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.
| |
Collapse
|
39
|
Hao Q, Long Y, Yang Y, Deng Y, Ding Z, Yang L, Shu Y, Xu H. Development and Clinical Applications of Therapeutic Cancer Vaccines with Individualized and Shared Neoantigens. Vaccines (Basel) 2024; 12:717. [PMID: 39066355 PMCID: PMC11281709 DOI: 10.3390/vaccines12070717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Neoantigens, presented as peptides on the surfaces of cancer cells, have recently been proposed as optimal targets for immunotherapy in clinical practice. The promising outcomes of neoantigen-based cancer vaccines have inspired enthusiasm for their broader clinical applications. However, the individualized tumor-specific antigens (TSA) entail considerable costs and time due to the variable immunogenicity and response rates of these neoantigens-based vaccines, influenced by factors such as neoantigen response, vaccine types, and combination therapy. Given the crucial role of neoantigen efficacy, a number of bioinformatics algorithms and pipelines have been developed to improve the accuracy rate of prediction through considering a series of factors involving in HLA-peptide-TCR complex formation, including peptide presentation, HLA-peptide affinity, and TCR recognition. On the other hand, shared neoantigens, originating from driver mutations at hot mutation spots (e.g., KRASG12D), offer a promising and ideal target for the development of therapeutic cancer vaccines. A series of clinical practices have established the efficacy of these vaccines in patients with distinct HLA haplotypes. Moreover, increasing evidence demonstrated that a combination of tumor associated antigens (TAAs) and neoantigens can also improve the prognosis, thus expand the repertoire of shared neoantigens for cancer vaccines. In this review, we provide an overview of the complex process involved in identifying personalized neoantigens, their clinical applications, advances in vaccine technology, and explore the therapeutic potential of shared neoantigen strategies.
Collapse
Affiliation(s)
- Qing Hao
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
| | - Yuhang Long
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
| | - Yi Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
| | - Yiqi Deng
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhenyu Ding
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; (Q.H.); (Y.L.); (Y.Y.); (Y.D.); (Z.D.); (L.Y.)
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Center of Clinical Laboratory Medicine, Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Meynard-Piganeau B, Feinauer C, Weigt M, Walczak AM, Mora T. TULIP: A transformer-based unsupervised language model for interacting peptides and T cell receptors that generalizes to unseen epitopes. Proc Natl Acad Sci U S A 2024; 121:e2316401121. [PMID: 38838016 PMCID: PMC11181096 DOI: 10.1073/pnas.2316401121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
The accurate prediction of binding between T cell receptors (TCR) and their cognate epitopes is key to understanding the adaptive immune response and developing immunotherapies. Current methods face two significant limitations: the shortage of comprehensive high-quality data and the bias introduced by the selection of the negative training data commonly used in the supervised learning approaches. We propose a method, Transformer-based Unsupervised Language model for Interacting Peptides and T cell receptors (TULIP), that addresses both limitations by leveraging incomplete data and unsupervised learning and using the transformer architecture of language models. Our model is flexible and integrates all possible data sources, regardless of their quality or completeness. We demonstrate the existence of a bias introduced by the sampling procedure used in previous supervised approaches, emphasizing the need for an unsupervised approach. TULIP recognizes the specific TCRs binding an epitope, performing well on unseen epitopes. Our model outperforms state-of-the-art models and offers a promising direction for the development of more accurate TCR epitope recognition models.
Collapse
Affiliation(s)
- Barthelemy Meynard-Piganeau
- Laboratory of Computational and Quantitative Biology, Institut de Biologie Paris Seine, CNRS, Sorbonne Université, Paris75005, France
- Department of Computing Sciences, Bocconi University, Milan20100, Italy
| | | | - Martin Weigt
- Laboratory of Computational and Quantitative Biology, Institut de Biologie Paris Seine, CNRS, Sorbonne Université, Paris75005, France
| | - Aleksandra M. Walczak
- Laboratoire de Physique de l’Ecole Normale Supérieure, Université Paris Sciences et Lettres, CNRS, Sorbonne Université, Université de Paris Cité, Paris75005, France
| | - Thierry Mora
- Laboratoire de Physique de l’Ecole Normale Supérieure, Université Paris Sciences et Lettres, CNRS, Sorbonne Université, Université de Paris Cité, Paris75005, France
| |
Collapse
|
41
|
Yu Z, Jiang M, Lan X. HeteroTCR: A heterogeneous graph neural network-based method for predicting peptide-TCR interaction. Commun Biol 2024; 7:684. [PMID: 38834836 PMCID: PMC11150398 DOI: 10.1038/s42003-024-06380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 05/23/2024] [Indexed: 06/06/2024] Open
Abstract
Identifying interactions between T-cell receptors (TCRs) and immunogenic peptides holds profound implications across diverse research domains and clinical scenarios. Unsupervised clustering models (UCMs) cannot predict peptide-TCR binding directly, while supervised predictive models (SPMs) often face challenges in identifying antigens previously unencountered by the immune system or possessing limited TCR binding repertoires. Therefore, we propose HeteroTCR, an SPM based on Heterogeneous Graph Neural Network (GNN), to accurately predict peptide-TCR binding probabilities. HeteroTCR captures within-type (TCR-TCR or peptide-peptide) similarity information and between-type (peptide-TCR) interaction insights for predictions on unseen peptides and TCRs, surpassing limitations of existing SPMs. Our evaluation shows HeteroTCR outperforms state-of-the-art models on independent datasets. Ablation studies and visual interpretation underscore the Heterogeneous GNN module's critical role in enhancing HeteroTCR's performance by capturing pivotal binding process features. We further demonstrate the robustness and reliability of HeteroTCR through validation using single-cell datasets, aligning with the expectation that pMHC-TCR complexes with higher predicted binding probabilities correspond to increased binding fractions.
Collapse
Affiliation(s)
- Zilan Yu
- School of Medicine, Tsinghua University, 100084, Beijing, China
- Centre for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Mengnan Jiang
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Xun Lan
- School of Medicine, Tsinghua University, 100084, Beijing, China.
- Centre for Life Sciences, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Tsinghua University, Beijing, China.
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
42
|
Machaca V, Goyzueta V, Cruz MG, Sejje E, Pilco LM, López J, Túpac Y. Transformers meets neoantigen detection: a systematic literature review. J Integr Bioinform 2024; 21:jib-2023-0043. [PMID: 38960869 PMCID: PMC11377031 DOI: 10.1515/jib-2023-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/20/2024] [Indexed: 07/05/2024] Open
Abstract
Cancer immunology offers a new alternative to traditional cancer treatments, such as radiotherapy and chemotherapy. One notable alternative is the development of personalized vaccines based on cancer neoantigens. Moreover, Transformers are considered a revolutionary development in artificial intelligence with a significant impact on natural language processing (NLP) tasks and have been utilized in proteomics studies in recent years. In this context, we conducted a systematic literature review to investigate how Transformers are applied in each stage of the neoantigen detection process. Additionally, we mapped current pipelines and examined the results of clinical trials involving cancer vaccines.
Collapse
Affiliation(s)
| | | | | | - Erika Sejje
- Universidad Nacional de San Agustín, Arequipa, Perú
| | | | | | - Yván Túpac
- 187038 Universidad Católica San Pablo , Arequipa, Perú
| |
Collapse
|
43
|
Bulashevska A, Nacsa Z, Lang F, Braun M, Machyna M, Diken M, Childs L, König R. Artificial intelligence and neoantigens: paving the path for precision cancer immunotherapy. Front Immunol 2024; 15:1394003. [PMID: 38868767 PMCID: PMC11167095 DOI: 10.3389/fimmu.2024.1394003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
Cancer immunotherapy has witnessed rapid advancement in recent years, with a particular focus on neoantigens as promising targets for personalized treatments. The convergence of immunogenomics, bioinformatics, and artificial intelligence (AI) has propelled the development of innovative neoantigen discovery tools and pipelines. These tools have revolutionized our ability to identify tumor-specific antigens, providing the foundation for precision cancer immunotherapy. AI-driven algorithms can process extensive amounts of data, identify patterns, and make predictions that were once challenging to achieve. However, the integration of AI comes with its own set of challenges, leaving space for further research. With particular focus on the computational approaches, in this article we have explored the current landscape of neoantigen prediction, the fundamental concepts behind, the challenges and their potential solutions providing a comprehensive overview of this rapidly evolving field.
Collapse
Affiliation(s)
- Alla Bulashevska
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Zsófia Nacsa
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Franziska Lang
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Markus Braun
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Martin Machyna
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Liam Childs
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Renate König
- Host-Pathogen-Interactions, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
44
|
Jiang F, Guo Y, Ma H, Na S, Zhong W, Han Y, Wang T, Huang J. GTE: a graph learning framework for prediction of T-cell receptors and epitopes binding specificity. Brief Bioinform 2024; 25:bbae343. [PMID: 39007599 PMCID: PMC11247411 DOI: 10.1093/bib/bbae343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/15/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
The interaction between T-cell receptors (TCRs) and peptides (epitopes) presented by major histocompatibility complex molecules (MHC) is fundamental to the immune response. Accurate prediction of TCR-epitope interactions is crucial for advancing the understanding of various diseases and their prevention and treatment. Existing methods primarily rely on sequence-based approaches, overlooking the inherent topology structure of TCR-epitope interaction networks. In this study, we present $GTE$, a novel heterogeneous Graph neural network model based on inductive learning to capture the topological structure between TCRs and Epitopes. Furthermore, we address the challenge of constructing negative samples within the graph by proposing a dynamic edge update strategy, enhancing model learning with the nonbinding TCR-epitope pairs. Additionally, to overcome data imbalance, we adapt the Deep AUC Maximization strategy to the graph domain. Extensive experiments are conducted on four public datasets to demonstrate the superiority of exploring underlying topological structures in predicting TCR-epitope interactions, illustrating the benefits of delving into complex molecular networks. The implementation code and data are available at https://github.com/uta-smile/GTE.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| | - Yuzhi Guo
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| | - Hehuan Ma
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| | - Saiyang Na
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| | - Wenliang Zhong
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| | - Yi Han
- Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, United States
| | - Tao Wang
- Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, United States
| | - Junzhou Huang
- Department of Computer Science and Engineering, University of Texas at Arlington, 701 S. Nedderman Drive, TX 76019, United States
| |
Collapse
|
45
|
Wang A, Lin X, Chau KN, Onuchic JN, Levine H, George JT. RACER-m leverages structural features for sparse T cell specificity prediction. SCIENCE ADVANCES 2024; 10:eadl0161. [PMID: 38748791 PMCID: PMC11095454 DOI: 10.1126/sciadv.adl0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reliable prediction of T cell specificity against antigenic signatures is a formidable task, complicated by the immense diversity of T cell receptor and antigen sequence space and the resulting limited availability of training sets for inferential models. Recent modeling efforts have demonstrated the advantage of incorporating structural information to overcome the need for extensive training sequence data, yet disentangling the heterogeneous TCR-antigen interface to accurately predict MHC-allele-restricted TCR-peptide interactions has remained challenging. Here, we present RACER-m, a coarse-grained structural model leveraging key biophysical information from the diversity of publicly available TCR-antigen crystal structures. Explicit inclusion of structural content substantially reduces the required number of training examples and maintains reliable predictions of TCR-recognition specificity and sensitivity across diverse biological contexts. Our model capably identifies biophysically meaningful point-mutant peptides that affect binding affinity, distinguishing its ability in predicting TCR specificity of point-mutants from alternative sequence-based methods. Its application is broadly applicable to studies involving both closely related and structurally diverse TCR-peptide pairs.
Collapse
Affiliation(s)
- Ailun Wang
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Xingcheng Lin
- Department of Physics, North Carolina State University, Raleigh, NC, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - Kevin Ng Chau
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
| | - José N. Onuchic
- Departments of Physics and Astronomy, Chemistry, and Biosciences, Rice University, Houston, TX, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Jason T. George
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- Department of Biomedical Engineering, Texas A&M University, Houston, TX, USA
| |
Collapse
|
46
|
Jiang M, Yu Z, Lan X. VitTCR: A deep learning method for peptide recognition prediction. iScience 2024; 27:109770. [PMID: 38711451 PMCID: PMC11070698 DOI: 10.1016/j.isci.2024.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 01/21/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
This study introduces VitTCR, a predictive model based on the vision transformer (ViT) architecture, aimed at identifying interactions between T cell receptors (TCRs) and peptides, crucial for developing cancer immunotherapies and vaccines. VitTCR converts TCR-peptide interactions into numerical AtchleyMaps using Atchley factors for prediction, achieving AUROC (0.6485) and AUPR (0.6295) values. Benchmark analysis indicates VitTCR's performance is comparable to other models, with further comparative studies suggested to understand its effectiveness in varied contexts. Additionally, integrating a positional bias weight matrix (PBWM), derived from amino acid contact probabilities in structurally resolved pMHC-TCR complexes, slightly improves VitTCR's accuracy. The model's predictions show weak yet statistically significant correlations with immunological factors like T cell clonal expansion and activation percentages, underscoring the biological relevance of VitTCR's predictive capabilities. VitTCR emerges as a valuable computational tool for predicting TCR-peptide interactions, offering insights for immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Mengnan Jiang
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zilan Yu
- School of Medicine, Tsinghua University, Beijing 100084, China
- Centre for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xun Lan
- School of Medicine, Tsinghua University, Beijing 100084, China
- Centre for Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
47
|
Liu Y, Zhou Y, Hu X, Le-Ge W, Wang H, Jiang T, Li J, Hu Y, Wang Y. DIRMC: a database of immunotherapy-related molecular characteristics. Database (Oxford) 2024; 2024:baae032. [PMID: 38713861 PMCID: PMC11184449 DOI: 10.1093/database/baae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/02/2024] [Accepted: 03/29/2024] [Indexed: 05/09/2024]
Abstract
Cancer immunotherapy has brought about a revolutionary breakthrough in the field of cancer treatment. Immunotherapy has changed the treatment landscape for a variety of solid and hematologic malignancies. To assist researchers in efficiently uncovering valuable information related to cancer immunotherapy, we have presented a manually curated comprehensive database called DIRMC, which focuses on molecular features involved in cancer immunotherapy. All the content was collected manually from published literature, authoritative clinical trial data submitted by clinicians, some databases for drug target prediction such as DrugBank, and some experimentally confirmed high-throughput data sets for the characterization of immune-related molecular interactions in cancer, such as a curated database of T-cell receptor sequences with known antigen specificity (VDJdb), a pathology-associated TCR database (McPAS-TCR) et al. By constructing a fully connected functional network, ranging from cancer-related gene mutations to target genes to translated target proteins to protein regions or sites that may specifically affect protein function, we aim to comprehensively characterize molecular features related to cancer immunotherapy. We have developed the scoring criteria to assess the reliability of each MHC-peptide-T-cell receptor (TCR) interaction item to provide a reference for users. The database provides a user-friendly interface to browse and retrieve data by genes, target proteins, diseases and more. DIRMC also provides a download and submission page for researchers to access data of interest for further investigation or submit new interactions related to cancer immunotherapy targets. Furthermore, DIRMC provides a graphical interface to help users predict the binding affinity between their own peptide of interest and MHC or TCR. This database will provide researchers with a one-stop resource to understand cancer immunotherapy-related targets as well as data on MHC-peptide-TCR interactions. It aims to offer reliable molecular characteristics support for both the analysis of the current status of cancer immunotherapy and the development of new immunotherapy. DIRMC is available at http://www.dirmc.tech/. Database URL: http://www.dirmc.tech/.
Collapse
Affiliation(s)
- Yue Liu
- Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yuhuan Zhou
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Xiumei Hu
- Beidahuang Industry Group General Hospital, Harbin 150001, China
| | - Wuri Le-Ge
- Department of Pain, Tongliao City Hospital, Tongliao 028000, China
| | - Haoyan Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Tao Jiang
- Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Junyi Li
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Yang Hu
- Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yadong Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| |
Collapse
|
48
|
Marrer-Berger E, Nicastri A, Augustin A, Kramar V, Liao H, Hanisch LJ, Carpy A, Weinzierl T, Durr E, Schaub N, Nudischer R, Ortiz-Franyuti D, Breous-Nystrom E, Stucki J, Hobi N, Raggi G, Cabon L, Lezan E, Umaña P, Woodhouse I, Bujotzek A, Klein C, Ternette N. The physiological interactome of TCR-like antibody therapeutics in human tissues. Nat Commun 2024; 15:3271. [PMID: 38627373 PMCID: PMC11021511 DOI: 10.1038/s41467-024-47062-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Selective binding of TCR-like antibodies that target a single tumour-specific peptide antigen presented by human leukocyte antigens (HLA) is the absolute prerequisite for their therapeutic suitability and patient safety. To date, selectivity assessment has been limited to peptide library screening and predictive modeling. We developed an experimental platform to de novo identify interactomes of TCR-like antibodies directly in human tissues using mass spectrometry. As proof of concept, we confirm the target epitope of a MAGE-A4-specific TCR-like antibody. We further determine cross-reactive peptide sequences for ESK1, a TCR-like antibody with known off-target activity, in human liver tissue. We confirm off-target-induced T cell activation and ESK1-mediated liver spheroid killing. Off-target sequences feature an amino acid motif that allows a structural groove-coordination mimicking that of the target peptide, therefore allowing the interaction with the engager molecule. We conclude that our strategy offers an accurate, scalable route for evaluating the non-clinical safety profile of TCR-like antibody therapeutics prior to first-in-human clinical application.
Collapse
Affiliation(s)
- Estelle Marrer-Berger
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Annalisa Nicastri
- The Jenner Institute, Old Road Campus Research Building, Oxford, OX37DQ, UK
- Centre for Immuno-Oncology, Old Road Campus Research Building, Oxford, OX37DQ, UK
| | - Angelique Augustin
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Vesna Kramar
- Roche Innovation Center Zürich, 8952, Schlieren, Switzerland
| | - Hanqing Liao
- The Jenner Institute, Old Road Campus Research Building, Oxford, OX37DQ, UK
- Centre for Immuno-Oncology, Old Road Campus Research Building, Oxford, OX37DQ, UK
| | | | - Alejandro Carpy
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, 82377, Penzberg, Germany
| | - Tina Weinzierl
- Roche Innovation Center Zürich, 8952, Schlieren, Switzerland
| | - Evelyne Durr
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Nathalie Schaub
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Ramona Nudischer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Daniela Ortiz-Franyuti
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Ekaterina Breous-Nystrom
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Janick Stucki
- Alveolix AG, Swiss Organs-on-Chip Innovation, 3010, Bern, Switzerland
| | - Nina Hobi
- Alveolix AG, Swiss Organs-on-Chip Innovation, 3010, Bern, Switzerland
| | - Giulia Raggi
- Alveolix AG, Swiss Organs-on-Chip Innovation, 3010, Bern, Switzerland
| | - Lauriane Cabon
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Emmanuelle Lezan
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zürich, 8952, Schlieren, Switzerland
| | - Isaac Woodhouse
- The Jenner Institute, Old Road Campus Research Building, Oxford, OX37DQ, UK
- Centre for Immuno-Oncology, Old Road Campus Research Building, Oxford, OX37DQ, UK
| | - Alexander Bujotzek
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, 82377, Penzberg, Germany
| | - Christian Klein
- Roche Innovation Center Zürich, 8952, Schlieren, Switzerland.
| | - Nicola Ternette
- The Jenner Institute, Old Road Campus Research Building, Oxford, OX37DQ, UK.
- Centre for Immuno-Oncology, Old Road Campus Research Building, Oxford, OX37DQ, UK.
- Department of Pharmaceutical Sciences, University of Utrecht, 3584, CH, Utrecht, The Netherlands.
| |
Collapse
|
49
|
Croce G, Bobisse S, Moreno DL, Schmidt J, Guillame P, Harari A, Gfeller D. Deep learning predictions of TCR-epitope interactions reveal epitope-specific chains in dual alpha T cells. Nat Commun 2024; 15:3211. [PMID: 38615042 PMCID: PMC11016097 DOI: 10.1038/s41467-024-47461-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/03/2024] [Indexed: 04/15/2024] Open
Abstract
T cells have the ability to eliminate infected and cancer cells and play an essential role in cancer immunotherapy. T cell activation is elicited by the binding of the T cell receptor (TCR) to epitopes displayed on MHC molecules, and the TCR specificity is determined by the sequence of its α and β chains. Here, we collect and curate a dataset of 17,715 αβTCRs interacting with dozens of class I and class II epitopes. We use this curated data to develop MixTCRpred, an epitope-specific TCR-epitope interaction predictor. MixTCRpred accurately predicts TCRs recognizing several viral and cancer epitopes. MixTCRpred further provides a useful quality control tool for multiplexed single-cell TCR sequencing assays of epitope-specific T cells and pinpoints a substantial fraction of putative contaminants in public databases. Analysis of epitope-specific dual α T cells demonstrates that MixTCRpred can identify α chains mediating epitope recognition. Applying MixTCRpred to TCR repertoires from COVID-19 patients reveals enrichment of clonotypes predicted to bind an immunodominant SARS-CoV-2 epitope. Overall, MixTCRpred provides a robust tool to predict TCRs interacting with specific epitopes and interpret TCR-sequencing data from both bulk and epitope-specific T cells.
Collapse
Affiliation(s)
- Giancarlo Croce
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Sara Bobisse
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Dana Léa Moreno
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Julien Schmidt
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Philippe Guillame
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Centre, Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University Hospital of Lausanne, Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
- Agora Cancer Research Centre, Lausanne, Switzerland.
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
50
|
Pothuri VS, Hogg GD, Conant L, Borcherding N, James CA, Mudd J, Williams G, Seo YD, Hawkins WG, Pillarisetty VG, DeNardo DG, Fields RC. Intratumoral T-cell receptor repertoire composition predicts overall survival in patients with pancreatic ductal adenocarcinoma. Oncoimmunology 2024; 13:2320411. [PMID: 38504847 PMCID: PMC10950267 DOI: 10.1080/2162402x.2024.2320411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy that is refractory to immune checkpoint inhibitor therapy. However, intratumoral T-cell infiltration correlates with improved overall survival (OS). Herein, we characterized the diversity and antigen specificity of the PDAC T-cell receptor (TCR) repertoire to identify novel immune-relevant biomarkers. Demographic, clinical, and TCR-beta sequencing data were collated from 353 patients across three cohorts that underwent surgical resection for PDAC. TCR diversity was calculated using Shannon Wiener index, Inverse Simpson index, and "True entropy." Patients were clustered by shared repertoire specificity. TCRs predictive of OS were identified and their associated transcriptional states were characterized by single-cell RNAseq. In multivariate Cox regression models controlling for relevant covariates, high intratumoral TCR diversity predicted OS across multiple cohorts. Conversely, in peripheral blood, high abundance of T-cells, but not high diversity, predicted OS. Clustering patients based on TCR specificity revealed a subset of TCRs that predicts OS. Interestingly, these TCR sequences were more likely to encode CD8+ effector memory and CD4+ T-regulatory (Tregs) T-cells, all with the capacity to recognize beta islet-derived autoantigens. As opposed to T-cell abundance, intratumoral TCR diversity was predictive of OS in multiple PDAC cohorts, and a subset of TCRs enriched in high-diversity patients independently correlated with OS. These findings emphasize the importance of evaluating peripheral and intratumoral TCR repertoires as distinct and relevant biomarkers in PDAC.
Collapse
Affiliation(s)
- Vikram S. Pothuri
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Graham D. Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Leah Conant
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Borcherding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - C. Alston James
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Greg Williams
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yongwoo David Seo
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - William G. Hawkins
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| | - Venu G. Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WAUSA
| | - David G. DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| | - Ryan C. Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MOUSA
| |
Collapse
|