1
|
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
|
2
|
Najar Najafi N, Karbassian R, Hajihassani H, Azimzadeh Irani M. Unveiling the influence of fastest nobel prize winner discovery: alphafold's algorithmic intelligence in medical sciences. J Mol Model 2025; 31:163. [PMID: 40387957 DOI: 10.1007/s00894-025-06392-x] [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: 12/01/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
CONTEXT AlphaFold's advanced AI technology has transformed protein structure interpretation. By predicting three-dimensional protein structures from amino acid sequences, AlphaFold has solved the complex protein-folding problem, previously challenging for experimental methods due to numerous possible conformations. Since its inception, AlphaFold has introduced several versions, including AlphaFold2, AlphaFold DB, AlphaFold Multimer, Alpha Missense, and AlphaFold3, each further enhancing protein structure prediction. Remarkably, AlphaFold is recognized as the fastest Nobel Prize winner in science history. This technology has extensive applications, potentially transforming treatment and diagnosis in medical sciences by reducing drug design costs and time, while elucidating structural pathways of human body systems. Numerous studies have demonstrated how AlphaFold aids in understanding health conditions by providing critical information about protein mutations, abnormal protein-protein interactions, and changes in protein dynamics. Researchers have also developed new technologies and pipelines using different versions of AlphaFold to amplify its potential. However, addressing existing limitations is crucial to maximizing AlphaFold's capacity to redefine medical research. This article reviews AlphaFold's impact on five key aspects of medical sciences: protein mutation, protein-protein interaction, molecular dynamics, drug design, and immunotherapy. METHODS This review examines the contributions of various AlphaFold versions AlphaFold2, AlphaFold DB, AlphaFold Multimer, Alpha Missense, and AlphaFold3 to protein structure prediction. The methods include an extensive analysis of computational techniques and software used in interpreting and predicting protein structures, emphasizing advances in AI technology and its applications in medical research.
Collapse
Affiliation(s)
- Niki Najar Najafi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Reyhaneh Karbassian
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Helia Hajihassani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
3
|
Yuan R, Zhang J, Zhou J, Cong Q. Recent progress and future challenges in structure-based protein-protein interaction prediction. Mol Ther 2025; 33:2252-2268. [PMID: 40195117 DOI: 10.1016/j.ymthe.2025.04.003] [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: 02/07/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
Protein-protein interactions (PPIs) play a fundamental role in cellular processes, and understanding these interactions is crucial for advances in both basic biological science and biomedical applications. This review presents an overview of recent progress in computational methods for modeling protein complexes and predicting PPIs based on 3D structures, focusing on the transformative role of artificial intelligence-based approaches. We further discuss the expanding biomedical applications of PPI research, including the elucidation of disease mechanisms, drug discovery, and therapeutic design. Despite these advances, significant challenges remain in predicting host-pathogen interactions, interactions between intrinsically disordered regions, and interactions related to immune responses. These challenges are worthwhile for future explorations and represent the frontier of research in this field.
Collapse
Affiliation(s)
- Rongqing Yuan
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jing Zhang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Ye F, Chen M, Huang Y, Zhang R, Li X, Wang X, Han S, Ma L, Liu X. LightCTL: lightweight contrastive TCR-pMHC specificity learning with context-aware prompt. Brief Bioinform 2025; 26:bbaf246. [PMID: 40439672 PMCID: PMC12121355 DOI: 10.1093/bib/bbaf246] [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: 08/29/2024] [Revised: 03/15/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Identification of T cell receptor (TCR) specificities for antigens from large-scale single-cell or bulk TCR repertoire data plays a vital role in disease diagnosis and immunotherapy. In silico prediction models have emerged in recent years. However, the generalizability and transferability of current computational models remain significant hurdles in accurately predicting TCR-pMHC binding specificity, primarily due to the limited availability of experimental data and the vast diversity of TCR sequences. In this paper, we propose a lightweight contrastive TCR-pMHC learning with context-aware prompts, named LightCTL, to infer TCR-pMHC binding specificity. For each TCR and peptide-MHC sequence, we utilize a TCR encoding module and a pMHC encoding module to transform them into latent representations. Specifically, we introduce a contrastive TCR-pMHC learning paradigm to enhance the generalization ability of TCR-pMHC binding specificity prediction by learning the matching relationship between TCR-pMHC and MHC-peptide. We fuse the TCR and pMHC latent representations and employ a novel context-aware prompt module to consider the varying importance of different feature maps. Compared with existing methods, LightCTL substantially improves the accuracy of predicting TCR-pMHC binding specificity. Moreover, comparative experiments across eight independent datasets demonstrate the generalization ability of LightCTL, showing superior performance for predicting unknown TCR-pMHC pairs. Finally, we assess LightCTL's efficacy across different TCR sequence lengths and distinct unseen epitopes, as well as estimate cytomegalovirus-specific TCR diversity and clone frequency from peripheral TCR repertoire data. Overall, our findings highlight LightCTL as a versatile analytical method for advancing novel T-cell therapies and identifying novel biomarkers for disease diagnosis.
Collapse
Affiliation(s)
- Fei Ye
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Mao Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Yixuan Huang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Ruihao Zhang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Xuqi Li
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Xiuyuan Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Lan Ma
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| | - Xiao Liu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua ShenZhen International Graduate School, Tsinghua University, Lishui Road, Nanshan District, Shenzhen, Guangdong Province 518055, China
| |
Collapse
|
5
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
6
|
Gasperini G, Baylor N, Black S, Bloom DE, Cramer J, de Lannoy G, Denoel P, Feinberg M, Helleputte T, Kang G, Schief WR, Stuart L, Weller C, Zwierzyna M, Rappuoli R. Vaccinology in the artificial intelligence era. Sci Transl Med 2025; 17:eadu3791. [PMID: 40238919 DOI: 10.1126/scitranslmed.adu3791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Artificial intelligence (AI) has already transformed vaccine antigen design and could transform the entire vaccinology pipeline, including immune responses and emerging infectious disease prediction, manufacturing and regulatory processes, clinical trial design and implementation, and vaccine access and equity. However, realizing the promise of AI for vaccinology requires more high-quality data.
Collapse
Affiliation(s)
| | | | - Steve Black
- Global Vaccine Data Network, Auckland, New Zealand
| | - David E Bloom
- Harvard T.H. Chan School of Public Health, Boston MA, USA
| | - Jakob Cramer
- Coalition for Epidemic Preparedness Innovations (CEPI), London, UK
| | | | | | | | | | - Gagandeep Kang
- Bill and Melinda Gates Foundation (BMGF), Seattle, WA, USA
| | - William R Schief
- Moderna Inc., Boston, MA, USA
- Department of Immunology and Microbial Science, Scripps Research Institute, La Jolla, CA, USA
- IAVI Neutralizing Antibody Center, Scripps Research Institute, La Jolla, CA, USA
- Center for HIV/AIDS Vaccine Development, Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
7
|
Visani GM, Pun MN, Minervina AA, Bradley P, Thomas P, Nourmohammad A. T-cell receptor specificity landscape revealed through de novo peptide design. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640903. [PMID: 40093114 PMCID: PMC11908224 DOI: 10.1101/2025.02.28.640903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
T-cells play a key role in adaptive immunity by mounting specific responses against diverse pathogens. An effective binding between T-cell receptors (TCRs) and pathogen-derived peptides presented on Major Histocompatibility Complexes (MHCs) mediate an immune response. However, predicting these interactions remains challenging due to limited functional data on T-cell reactivities. Here, we introduce a computational approach to predict TCR interactions with peptides presented on MHC class I alleles, and to design novel immunogenic peptides for specified TCR-MHC complexes. Our method leverages HERMES, a structure-based, physics-guided machine learning model trained on the protein universe to predict amino acid preferences based on local structural environments. Despite no direct training on TCR-pMHC data, the implicit physical reasoning in HERMES enables us to make accurate predictions of both TCR-pMHC binding affinities and T-cell activities across diverse viral epitopes and cancer neoantigens, achieving up to 72% correlation with experimental data. Leveraging our TCR recognition model, we develop a computational protocol for de novo design of immunogenic peptides. Through experimental validation in three TCR-MHC systems targeting viral and cancer peptides, we demonstrate that our designs-with up to five substitutions from the native sequence-activate T-cells at success rates of up to 50%. Lastly, we use our generative framework to quantify the diversity of the peptide recognition landscape for various TCR-MHC complexes, offering key insights into T-cell specificity in both humans and mice. Our approach provides a platform for immunogenic peptide and neoantigen design, opening new computational paths for T-cell vaccine development against viruses and cancer.
Collapse
Affiliation(s)
- Gian Marco Visani
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 85 E Stevens Way NE, Seattle, WA 98195, USA
| | - Michael N Pun
- Department of Physics, University of Washington, 3910 15th Avenue Northeast, Seattle, WA 98195, USA
| | | | - Philip Bradley
- Fred Hutchinson Cancer Center, 1241 Eastlake Ave E, Seattle, WA 98102, USA
| | - Paul Thomas
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Armita Nourmohammad
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 85 E Stevens Way NE, Seattle, WA 98195, USA
- Department of Physics, University of Washington, 3910 15th Avenue Northeast, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, 1241 Eastlake Ave E, Seattle, WA 98102, USA
- Department of Applied Mathematics, University of Washington, 4182 W Stevens Way NE, Seattle, WA 98105, USA
| |
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
|
Elyaman W, Stern LJ, Jiang N, Dressman D, Bradley P, Klatzmann D, Bradshaw EM, Farber DL, Kent SC, Chizari S, Funk K, Devanand D, Thakur KT, Raj T, Dalahmah OA, Sarkis RA, Weiner HL, Shneider NA, Przedborski S. Exploring the role of T cells in Alzheimer's and other neurodegenerative diseases: Emerging therapeutic insights from the T Cells in the Brain symposium. Alzheimers Dement 2025; 21:e14548. [PMID: 39868844 PMCID: PMC11851166 DOI: 10.1002/alz.14548] [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: 09/12/2024] [Revised: 12/02/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025]
Abstract
This proceedings article summarizes the inaugural "T Cells in the Brain" symposium held at Columbia University. Experts gathered to explore the role of T cells in neurodegenerative diseases. Key topics included characterization of antigen-specific immune responses, T cell receptor (TCR) repertoire, microbial etiology in Alzheimer's disease (AD), and microglia-T cell crosstalk, with a focus on how T cells affect neuroinflammation and AD biomarkers like amyloid beta and tau. The symposium also examined immunotherapies for AD, including the Valacyclovir Treatment of Alzheimer's Disease (VALAD) trial, and two clinical trials leveraging regulatory T cell approaches for multiple sclerosis and amyotrophic lateral sclerosis therapy. Additionally, single-cell RNA/TCR sequencing of T cells and other immune cells provided insights into immune dynamics in neurodegenerative diseases. This article highlights key findings from the symposium and outlines future research directions to further understand the role of T cells in neurodegeneration, offering innovative therapeutic approaches for AD and other neurodegenerative diseases. HIGHLIGHTS: Researchers gathered to discuss approaches to study T cells in brain disorders. New technologies allow high-throughput screening of antigen-specific T cells. Microbial infections can precede several serious and chronic neurological diseases. Central and peripheral T cell responses shape neurological disease pathology. Immunotherapy can induce regulatory T cell responses in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Wassim Elyaman
- Division of Translational NeurobiologyDepartment of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Medical CenterNew YorkNew YorkUSA
- Center for Motor Neuron Biology and DiseaseColumbia University Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Lawrence J. Stern
- Department of Pathology, and Immunology and Microbiology ProgramUMass Chan Medical SchoolWorcesterMassachusettsUSA
| | - Ning Jiang
- Department of BioengineeringInstitute for Immunology and Immune HealthCenter for Cellular ImmunotherapiesAbramson Cancer CenterInstitute for RNA InnovationCenter for Precision Engineering for HealthUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Dallin Dressman
- Division of Translational NeurobiologyDepartment of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Medical CenterNew YorkNew YorkUSA
- Center for Motor Neuron Biology and DiseaseColumbia University Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Philip Bradley
- Computational Biology ProgramPublic Health Sciences DivisionFred Hutchinson Cancer CenterSeattleWashingtonUSA
| | - David Klatzmann
- INSERM UMRS 959Immunology‐Immunopathology‐Immunotherapy (i3)Sorbonne UniversitéParisFrance
| | - Elizabeth M. Bradshaw
- Division of Translational NeurobiologyDepartment of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Medical CenterNew YorkNew YorkUSA
- Center for Motor Neuron Biology and DiseaseColumbia University Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
- Carol and Gene Ludwig Center for Research on NeurodegenerationDepartment of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Donna L. Farber
- Department of Microbiology and Immunology, and Department of SurgeryColumbia UniversityNew YorkNew YorkUSA
| | - Sally C. Kent
- Diabetes Center of ExcellenceDepartment of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Shahab Chizari
- Department of BioengineeringInstitute for Immunology and Immune HealthCenter for Cellular ImmunotherapiesAbramson Cancer CenterInstitute for RNA InnovationCenter for Precision Engineering for HealthUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kristen Funk
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| | - Davangere Devanand
- Department of PsychiatryColumbia University Medical CenterNew YorkNew YorkUSA
| | - Kiran T. Thakur
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| | - Towfique Raj
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic TechnologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Osama Al Dalahmah
- Department of Pathology and Cell BiologyVagelos College of Physicians and SurgeonsColumbia University Irving Medical Center and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Rani A. Sarkis
- Epilepsy DivisionDepartment of NeurologyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Howard L. Weiner
- Ann Romney Center for Neurologic DiseasesBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Neil A. Shneider
- Center for Motor Neuron Biology and DiseaseColumbia University Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
- Eleanor and Lou Gehrig ALS CenterColumbia UniversityNew YorkNew YorkUSA
| | - Serge Przedborski
- Center for Motor Neuron Biology and DiseaseColumbia University Medical CenterNew YorkNew YorkUSA
- Department of NeurologyColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
10
|
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
|
11
|
Lin V, Cheung M, Gowthaman R, Eisenberg M, Baker B, Pierce B. TCR3d 2.0: expanding the T cell receptor structure database with new structures, tools and interactions. Nucleic Acids Res 2025; 53:D604-D608. [PMID: 39329260 PMCID: PMC11701517 DOI: 10.1093/nar/gkae840] [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: 08/14/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Recognition of antigens by T cell receptors (TCRs) is a key component of adaptive immunity. Understanding the structures of these TCR interactions provides major insights into immune protection and diseases, and enables design of therapeutics, vaccines and predictive modeling algorithms. Previously, we released TCR3d, a database and resource for structures of TCRs and their recognition. Due to the growth of available structures and categories of complexes, the content of TCR3d has expanded substantially in the past 5 years. This expansion includes new tables dedicated to TCR mimic antibody complex structures, TCR-CD3 complexes and annotated Class I and II peptide-MHC complexes. Additionally, tools are available for users to calculate docking geometries for input TCR and TCR mimic complex structures. The core tables of TCR-peptide-MHC complexes have grown by 50%, and include binding affinity data for experimentally determined structures. These major content and feature updates enhance TCR3d as a resource for immunology, therapeutics and structural biology research, and enable advanced approaches for predictive TCR modeling and design. TCR3d is available at: https://tcr3d.ibbr.umd.edu.
Collapse
Affiliation(s)
- Valerie Lin
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Melyssa Cheung
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Ragul Gowthaman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Maya Eisenberg
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Brian G Pierce
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Ma X, Zhang J, Jiang Q, Li YX, Yang G. Human microbiome-derived peptide affects the development of experimental autoimmune encephalomyelitis via molecular mimicry. EBioMedicine 2025; 111:105516. [PMID: 39724786 PMCID: PMC11732510 DOI: 10.1016/j.ebiom.2024.105516] [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: 07/31/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Gut commensal microbiota has been identified as a potential environmental risk factor for multiple sclerosis (MS), and numerous studies have linked the commensal microorganism with the onset of MS. However, little is known about the mechanisms underlying the gut microbiome and host-immune system interaction. METHODS We employed bioinformatics methodologies to identify human microbial-derived peptides by analyzing their similarity to the MHC II-TCR binding patterns of self-antigens. Subsequently, we conducted a range of in vitro and in vivo assays to assess the encephalitogenic potential of these microbial-derived peptides. FINDINGS We analyzed 304,246 human microbiome genomes and 103 metagenomes collected from the MS cohort and identified 731 nonredundant analogs of myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). Of note, half of these analogs could bind to MHC II and interact with TCR through structural modeling of the interaction using fine-tuned AlphaFold. Among the 8 selected peptides, the peptide (P3) shows the ability to activate MOG35-55-specific CD4+ T cells in vitro. Furthermore, P3 shows encephalitogenic capacity and has the potential to induce EAE in some animals. Notably, mice immunized with a combination of P3 and MOG35-55 develop severe EAE. Additionally, dendritic cells could process and present P3 to MOG35-55-specific CD4+ T cells and activate these cells. INTERPRETATION Our data suggests the potential involvement of a MOG35-55-mimic peptide derived from the gut microbiota as a molecular trigger of EAE pathogenesis. Our findings offer direct evidence of how microbes can initiate the development of EAE, suggesting a potential explanation for the correlation between certain gut microorganisms and MS prevalence. FUNDING National Natural Science Foundation of China (82371350 to GY).
Collapse
MESH Headings
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Animals
- Molecular Mimicry
- Mice
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/chemistry
- Gastrointestinal Microbiome
- Peptides/chemistry
- Peptides/immunology
- Peptide Fragments/immunology
- Peptide Fragments/chemistry
- Disease Models, Animal
- Receptors, Antigen, T-Cell/metabolism
- Computational Biology/methods
- Histocompatibility Antigens Class II/metabolism
- Protein Binding
- Microbiota
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Multiple Sclerosis
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
Collapse
Affiliation(s)
- Xin Ma
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Jian Zhang
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Qianling Jiang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yong-Xin Li
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
13
|
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
|
14
|
O'Donnell TJ, Kanduri C, Isacchini G, Limenitakis JP, Brachman RA, Alvarez RA, Haff IH, Sandve GK, Greiff V. Reading the repertoire: Progress in adaptive immune receptor analysis using machine learning. Cell Syst 2024; 15:1168-1189. [PMID: 39701034 DOI: 10.1016/j.cels.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/16/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024]
Abstract
The adaptive immune system holds invaluable information on past and present immune responses in the form of B and T cell receptor sequences, but we are limited in our ability to decode this information. Machine learning approaches are under active investigation for a range of tasks relevant to understanding and manipulating the adaptive immune receptor repertoire, including matching receptors to the antigens they bind, generating antibodies or T cell receptors for use as therapeutics, and diagnosing disease based on patient repertoires. Progress on these tasks has the potential to substantially improve the development of vaccines, therapeutics, and diagnostics, as well as advance our understanding of fundamental immunological principles. We outline key challenges for the field, highlighting the need for software benchmarking, targeted large-scale data generation, and coordinated research efforts.
Collapse
Affiliation(s)
| | - Chakravarthi Kanduri
- Department of Informatics, University of Oslo, Oslo, Norway; UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
| | | | | | - Rebecca A Brachman
- Imprint Labs, LLC, New York, NY, USA; Cornell Tech, Cornell University, New York, NY, USA
| | | | - Ingrid H Haff
- Department of Mathematics, University of Oslo, 0371 Oslo, Norway
| | - Geir K Sandve
- Department of Informatics, University of Oslo, Oslo, Norway; UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
| | - Victor Greiff
- Imprint Labs, LLC, New York, NY, USA; Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
15
|
Pressley KR, Schwegman L, De Oca Arena MM, Huizar CC, Zamvil SS, Forsthuber TG. HLA-transgenic mouse models to study autoimmune central nervous system diseases. Autoimmunity 2024; 57:2387414. [PMID: 39167553 PMCID: PMC11470778 DOI: 10.1080/08916934.2024.2387414] [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: 03/31/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/23/2024]
Abstract
It is known that certain human leukocyte antigen (HLA) genes are associated with autoimmune central nervous system (CNS) diseases, such as multiple sclerosis (MS), but their exact role in disease susceptibility and etiopathogenesis remains unclear. The best studied HLA-associated autoimmune CNS disease is MS, and thus will be the primary focus of this review. Other HLA-associated autoimmune CNS diseases, such as autoimmune encephalitis and neuromyelitis optica will be discussed. The lack of animal models to accurately capture the complex human autoimmune response remains a major challenge. HLA transgenic (tg) mice provide researchers with powerful tools to investigate the underlying mechanisms promoting susceptibility and progression of HLA-associated autoimmune CNS diseases, as well as for elucidating the myelin epitopes potentially targeted by T cells in autoimmune disease patients. We will discuss the potential role(s) of autoimmune disease-associated HLA alleles in autoimmune CNS diseases and highlight information provided by studies using HLA tg mice to investigate the underlying pathological mechanisms and opportunities to use these models for development of novel therapies.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | | | - Carol Chase Huizar
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Scott S. Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
16
|
Le HN, de Freitas MV, Antunes DA. Strengths and limitations of web servers for the modeling of TCRpMHC complexes. Comput Struct Biotechnol J 2024; 23:2938-2948. [PMID: 39104710 PMCID: PMC11298609 DOI: 10.1016/j.csbj.2024.06.028] [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: 03/31/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 08/07/2024] Open
Abstract
Cellular immunity relies on the ability of a T-cell receptor (TCR) to recognize a peptide (p) presented by a class I major histocompatibility complex (MHC) receptor on the surface of a cell. The TCR-peptide-MHC (TCRpMHC) interaction is a crucial step in activating T-cells, and the structural characteristics of these molecules play a significant role in determining the specificity and affinity of this interaction. Hence, obtaining 3D structures of TCRpMHC complexes offers valuable insights into various aspects of cellular immunity and can facilitate the development of T-cell-based immunotherapies. Here, we aimed to compare three popular web servers for modeling the structures of TCRpMHC complexes, namely ImmuneScape (IS), TCRpMHCmodels, and TCRmodel2, to examine their strengths and limitations. Each method employs a different modeling strategy, including docking, homology modeling, and deep learning. The accuracy of each method was evaluated by reproducing the 3D structures of a dataset of 87 TCRpMHC complexes with experimentally determined crystal structures available on the Protein Data Bank (PDB). All selected structures were limited to human MHC alleles, presenting a diverse set of peptide ligands. A detailed analysis of produced models was conducted using multiple metrics, including Root Mean Square Deviation (RMSD) and standardized assessments from CAPRI and DockQ. Special attention was given to the complementarity-determining region (CDR) loops of the TCRs and to the peptide ligands, which define most of the unique features and specificity of a given TCRpMHC interaction. Our study provides an optimistic view of the current state-of-the-art for TCRpMHC modeling but highlights some remaining challenges that must be addressed in order to support the future application of these tools for TCR engineering and computer-aided design of TCR-based immunotherapies.
Collapse
Affiliation(s)
- Hoa Nhu Le
- University of Houston, Departments of Biology and Biochemistry, Houston, 77204, TX, USA
| | | | - Dinler Amaral Antunes
- University of Houston, Departments of Biology and Biochemistry, Houston, 77204, TX, USA
| |
Collapse
|
17
|
Wu D, Yin R, Chen G, Ribeiro-Filho HV, Cheung M, Robbins PF, Mariuzza RA, Pierce BG. Structural characterization and AlphaFold modeling of human T cell receptor recognition of NRAS cancer neoantigens. SCIENCE ADVANCES 2024; 10:eadq6150. [PMID: 39576860 PMCID: PMC11584006 DOI: 10.1126/sciadv.adq6150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024]
Abstract
T cell receptors (TCRs) that recognize cancer neoantigens are important for anticancer immune responses and immunotherapy. Understanding the structural basis of TCR recognition of neoantigens provides insights into their exquisite specificity and can enable design of optimized TCRs. We determined crystal structures of a human TCR in complex with NRAS Q61K and Q61R neoantigen peptides and HLA-A1 major histocompatibility complex (MHC), revealing the molecular underpinnings for dual recognition and specificity versus wild-type NRAS peptide. We then used multiple versions of AlphaFold to model the corresponding complex structures, given the challenge of immune recognition for such methods. One implementation of AlphaFold2 (TCRmodel2) with additional sampling was able to generate accurate models of the complexes, while AlphaFold3 also showed strong performance, although success was lower for other complexes. This study provides insights into TCR recognition of a shared cancer neoantigen as well as the utility and practical considerations for using AlphaFold to model TCR-peptide-MHC complexes.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/metabolism
- Membrane Proteins/chemistry
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Models, Molecular
- GTP Phosphohydrolases/metabolism
- GTP Phosphohydrolases/chemistry
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/immunology
- Protein Binding
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/metabolism
- Crystallography, X-Ray
- Protein Conformation
- Peptides/chemistry
- Peptides/immunology
- Peptides/metabolism
Collapse
Affiliation(s)
- Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Rui Yin
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Helder V. Ribeiro-Filho
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas 13083-100, Brazil
| | - Melyssa Cheung
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Paul F. Robbins
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Roy A. Mariuzza
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Brian G. Pierce
- W. M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
18
|
Wei Y, Qiu T, Ai Y, Zhang Y, Xie J, Zhang D, Luo X, Sun X, Wang X, Qiu J. Advances of computational methods enhance the development of multi-epitope vaccines. Brief Bioinform 2024; 26:bbaf055. [PMID: 39951549 PMCID: PMC11827616 DOI: 10.1093/bib/bbaf055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Vaccine development is one of the most promising fields, and multi-epitope vaccine, which does not need laborious culture processes, is an attractive alternative to classical vaccines with the advantage of safety, and efficiency. The rapid development of algorithms and the accumulation of immune data have facilitated the advancement of computer-aided vaccine design. Here we systemically reviewed the in silico data and algorithms resource, for different steps of computational vaccine design, including immunogen selection, epitope prediction, vaccine construction, optimization, and evaluation. The performance of different available tools on epitope prediction and immunogenicity evaluation was tested and compared on benchmark datasets. Finally, we discuss the future research direction for the construction of a multiepitope vaccine.
Collapse
Affiliation(s)
- Yiwen Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital; Intelligent Medicine Institute; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, No. 180, Fenglin Road, Xuhui Destrict, Shanghai 200032, China
| | - Yisi Ai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Yuxi Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Junting Xie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dong Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiaochuan Luo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214122, China
| | - Xin Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| |
Collapse
|
19
|
Postovskaya A, Vercauteren K, Meysman P, Laukens K. tcrBLOSUM: an amino acid substitution matrix for sensitive alignment of distant epitope-specific TCRs. Brief Bioinform 2024; 26:bbae602. [PMID: 39576224 PMCID: PMC11583439 DOI: 10.1093/bib/bbae602] [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: 05/22/2024] [Revised: 10/07/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
Deciphering the specificity of T-cell receptor (TCR) repertoires is crucial for monitoring adaptive immune responses and developing targeted immunotherapies and vaccines. To elucidate the specificity of previously unseen TCRs, many methods employ the BLOSUM62 matrix to find TCRs with similar amino acid (AA) sequences. However, while BLOSUM62 reflects the AA substitutions within conserved regions of proteins with similar functions, the remarkable diversity of TCRs means that both TCRs with similar and dissimilar sequences can bind the same epitope. Therefore, reliance on BLOSUM62 may bias detection towards epitope-specific TCRs with similar biochemical properties, overlooking those with more diverse AA compositions. In this study, we introduce tcrBLOSUMa and tcrBLOSUMb, specialized AA substitution matrices for CDR3 alpha and CDR3 beta TCR chains, respectively. The matrices reflect AA frequencies and variations occurring within TCRs that bind the same epitope, revealing that both CDR3 alpha and CDR3 beta display tolerance to a wide range of AA substitutions and differ noticeably from the standard BLOSUM62. By accurately aligning distant TCRs employing tcrBLOSUMb, we were able to improve clustering performance and capture a large number of epitope-specific TCRs with diverse AA compositions and physicochemical profiles overlooked by BLOSUM62. Utilizing both the general BLOSUM62 and specialized tcrBLOSUM matrices in existing computational tools will broaden the range of TCRs that can be associated with their cognate epitopes, thereby enhancing TCR repertoire analysis.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/chemistry
- Amino Acid Substitution
- Humans
- Amino Acid Sequence
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Sequence Alignment
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Complementarity Determining Regions/chemistry
- Computational Biology/methods
- Epitopes/immunology
- Epitopes/chemistry
- Algorithms
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
Collapse
Affiliation(s)
- Anna Postovskaya
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Koen Vercauteren
- Clinical Virology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 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), University of Antwerp, 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), University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (BIOMINA), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
20
|
Velez-Arce A, Li MM, Gao W, Lin X, Huang K, Fu T, Pentelute BL, Kellis M, Zitnik M. Signals in the Cells: Multimodal and Contextualized Machine Learning Foundations for Therapeutics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598655. [PMID: 38948789 PMCID: PMC11212894 DOI: 10.1101/2024.06.12.598655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drug discovery AI datasets and benchmarks have not traditionally included single-cell analysis biomarkers. While benchmarking efforts in single-cell analysis have recently released collections of single-cell tasks, they have yet to comprehensively release datasets, models, and benchmarks that integrate a broad range of therapeutic discovery tasks with cell-type-specific biomarkers. Therapeutics Commons (TDC-2) presents datasets, tools, models, and benchmarks integrating cell-type-specific contextual features with ML tasks across therapeutics. We present four tasks for contextual learning at single-cell resolution: drug-target nomination, genetic perturbation response prediction, chemical perturbation response prediction, and protein-peptide interaction prediction. We introduce datasets, models, and benchmarks for these four tasks. Finally, we detail the advancements and challenges in machine learning and biology that drove the implementation of TDC-2 and how they are reflected in its architecture, datasets and benchmarks, and foundation model tooling.
Collapse
Affiliation(s)
| | - Michelle M. Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115
| | - Wenhao Gao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Xiang Lin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115
| | - Kexin Huang
- Department of Computer Science, Stanford School of Engineering, Stanford, CA 94305
| | - Tianfan Fu
- Department of Computational Science, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Computer Science and Artificial Intelligence Laboratory, MIT, Electrical Engineering and Computer Science Department, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Marinka Zitnik
- Broad Institute of MIT and Harvard, Harvard Data Science Initiative, Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
21
|
Jagota M, Hsu C, Mazumder T, Sung K, DeWitt WS, Listgarten J, Matsen FA, Ye CJ, Song YS. Learning antibody sequence constraints from allelic inclusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619760. [PMID: 39484623 PMCID: PMC11526943 DOI: 10.1101/2024.10.22.619760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Antibodies and B-cell receptors (BCRs) are produced by B cells, and are built of a heavy chain and a light chain. Although each B cell could express two different heavy chains and four different light chains, usually only a unique pair of heavy chain and light chain is expressed-a phenomenon known as allelic exclusion. However, a small fraction of naive-B cells violate allelic exclusion by expressing two productive light chains, one of which has impaired function; this has been called allelic inclusion. We demonstrate that these B cells can be used to learn constraints on antibody sequence. Using large-scale single-cell sequencing data from humans, we find examples of light chain allelic inclusion in thousands of naive-B cells, which is an order of magnitude larger than existing datasets. We train machine learning models to identify the abnormal sequences in these cells. The resulting models correlate with antibody properties that they were not trained on, including polyreactivity, surface expression, and mutation usage in affinity maturation. These correlations are larger than what is achieved by existing antibody modeling approaches, indicating that allelic inclusion data contains useful new information. We also investigate the impact of similar selection forces on the heavy chain in mouse, and observe that pairing with the surrogate light chain significantly restricts heavy chain diversity.
Collapse
Affiliation(s)
- Milind Jagota
- Computer Science Division, UC Berkeley, Berkeley, CA USA
| | - Chloe Hsu
- Computer Science Division, UC Berkeley, Berkeley, CA USA
| | - Thomas Mazumder
- Division of Rheumatology, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - Frederick A. Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, UCSF, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, UCSF, San Francisco, CA, USA
- Institute for Human Genetics, UCSF, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, USA
| | - Yun S. Song
- Computer Science Division, UC Berkeley, Berkeley, CA USA
- Department of Statistics, UC Berkeley, Berkeley, CA, USA October 23, 2024
| |
Collapse
|
22
|
Henderson J, Nagano Y, Milighetti M, Tiffeau-Mayer A. Limits on inferring T cell specificity from partial information. Proc Natl Acad Sci U S A 2024; 121:e2408696121. [PMID: 39374400 PMCID: PMC11494314 DOI: 10.1073/pnas.2408696121] [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: 05/01/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
A key challenge in molecular biology is to decipher the mapping of protein sequence to function. To perform this mapping requires the identification of sequence features most informative about function. Here, we quantify the amount of information (in bits) that T cell receptor (TCR) sequence features provide about antigen specificity. We identify informative features by their degree of conservation among antigen-specific receptors relative to null expectations. We find that TCR specificity synergistically depends on the hypervariable regions of both receptor chains, with a degree of synergy that strongly depends on the ligand. Using a coincidence-based approach to measuring information enables us to directly bound the accuracy with which TCR specificity can be predicted from partial matches to reference sequences. We anticipate that our statistical framework will be of use for developing machine learning models for TCR specificity prediction and for optimizing TCRs for cell therapies. The proposed coincidence-based information measures might find further applications in bounding the performance of pairwise classifiers in other fields.
Collapse
Affiliation(s)
- James Henderson
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, LondonWC1E 6BT, United Kingdom
| | - Yuta Nagano
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Division of Medicine, University College London, LondonWC1E 6BT, United Kingdom
| | - Martina Milighetti
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Cancer Institute, University College London, LondonWC1E 6DD, United Kingdom
| | - Andreas Tiffeau-Mayer
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, LondonWC1E 6BT, United Kingdom
| |
Collapse
|
23
|
Lee JW, Chen EY, Hu T, Perret R, Chaffee ME, Martinov T, Mureli S, McCurdy CL, Jones LA, Gafken PR, Chanana P, Su Y, Chapuis AG, Bradley P, Schmitt TM, Greenberg PD. Overcoming immune evasion from post-translational modification of a mutant KRAS epitope to achieve TCR-T cell-mediated antitumor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.612965. [PMID: 39345486 PMCID: PMC11429761 DOI: 10.1101/2024.09.18.612965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
T cell receptor (TCR)-T cell immunotherapy, in which T cells are engineered to express a TCR targeting a tumor epitope, is a form of adoptive cell therapy (ACT) that has exhibited promise against various tumor types. Mutants of oncoprotein KRAS, particularly at glycine-12 (G12), are frequent drivers of tumorigenicity, making them attractive targets for TCR-T cell therapy. However, class I-restricted TCRs specifically targeting G12-mutant KRAS epitopes in the context of tumors expressing HLA-A2, the most common human HLA-A allele, have remained elusive despite evidence an epitope encompassing such mutations can bind HLA-A2 and induce T cell responses. We report post-translational modifications (PTMs) on this epitope may allow tumor cells to evade immunologic pressure from TCR-T cells. A lysine side chain-methylated KRAS G12V peptide, rather than the unmodified epitope, may be presented in HLA-A2 by tumor cells and impact TCR recognition. Using a novel computationally guided approach, we developed by mutagenesis TCRs that recognize this methylated peptide, enhancing tumor recognition and destruction. Additionally, we identified TCRs with similar functional activity in normal repertoires from primary T cells by stimulation with modified peptide, clonal expansion, and selection. Mechanistically, a gene knockout screen to identify mechanism(s) by which tumor cells methylate/demethylate this epitope unveiled SPT6 as a demethylating protein that could be targeted to improve effectiveness of these new TCRs. Our findings highlight the role of PTMs in immune evasion and suggest identifying and targeting such modifications should make effective ACTs available for a substantially greater range of tumors than the current therapeutic landscape. One-sentence summary Tumor cell methylation of KRAS G12V epitope in HLA-A2 permits immune evasion, and new TCRs were generated to overcome this with engineered cell therapy.
Collapse
|
24
|
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
|
25
|
Yang A, Poholek AC. Systems immunology approaches to study T cells in health and disease. NPJ Syst Biol Appl 2024; 10:117. [PMID: 39384819 PMCID: PMC11464710 DOI: 10.1038/s41540-024-00446-1] [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: 04/08/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
T cells are dynamically regulated immune cells that are implicated in a variety of diseases ranging from infection, cancer and autoimmunity. Recent advancements in sequencing methods have provided valuable insights in the transcriptional and epigenetic regulation of T cells in various disease settings. In this review, we identify the key sequencing-based methods that have been applied to understand the transcriptomic and epigenomic regulation of T cells in diseases.
Collapse
Affiliation(s)
- Aaron Yang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amanda C Poholek
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Perez MAS, Chiffelle J, Bobisse S, Mayol‐Rullan F, Bugnon M, Bragina ME, Arnaud M, Sauvage C, Barras D, Laniti DD, Huber F, Bassani‐Sternberg M, Coukos G, Harari A, Zoete V. Predicting Antigen-Specificities of Orphan T Cell Receptors from Cancer Patients with TCRpcDist. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405949. [PMID: 39159239 PMCID: PMC11516110 DOI: 10.1002/advs.202405949] [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: 05/30/2024] [Revised: 07/19/2024] [Indexed: 08/21/2024]
Abstract
Approaches to analyze and cluster T-cell receptor (TCR) repertoires to reflect antigen specificity are critical for the diagnosis and prognosis of immune-related diseases and the development of personalized therapies. Sequence-based approaches showed success but remain restrictive, especially when the amount of experimental data used for the training is scarce. Structure-based approaches which represent powerful alternatives, notably to optimize TCRs affinity toward specific epitopes, show limitations for large-scale predictions. To handle these challenges, TCRpcDist is presented, a 3D-based approach that calculates similarities between TCRs using a metric related to the physico-chemical properties of the loop residues predicted to interact with the epitope. By exploiting private and public datasets and comparing TCRpcDist with competing approaches, it is demonstrated that TCRpcDist can accurately identify groups of TCRs that are likely to bind the same epitopes. Importantly, the ability of TCRpcDist is experimentally validated to determine antigen specificities (neoantigens and tumor-associated antigens) of orphan tumor-infiltrating lymphocytes (TILs) in cancer patients. TCRpcDist is thus a promising approach to support TCR repertoire analysis and TCR deorphanization for individualized treatments including cancer immunotherapies.
Collapse
Affiliation(s)
- Marta A. S. Perez
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Molecular Modeling GroupSIB Swiss Institute of BioinformaticsUniversity of LausanneQuartier UNIL‐Sorge, Bâtiment AmphipoleLausanneCH‐1015Switzerland
| | - Johanna Chiffelle
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Sara Bobisse
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Francesca Mayol‐Rullan
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Molecular Modeling GroupSIB Swiss Institute of BioinformaticsUniversity of LausanneQuartier UNIL‐Sorge, Bâtiment AmphipoleLausanneCH‐1015Switzerland
| | - Marine Bugnon
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Molecular Modeling GroupSIB Swiss Institute of BioinformaticsUniversity of LausanneQuartier UNIL‐Sorge, Bâtiment AmphipoleLausanneCH‐1015Switzerland
| | - Maiia E. Bragina
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Molecular Modeling GroupSIB Swiss Institute of BioinformaticsUniversity of LausanneQuartier UNIL‐Sorge, Bâtiment AmphipoleLausanneCH‐1015Switzerland
| | - Marion Arnaud
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Christophe Sauvage
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - David Barras
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Denarda Dangaj Laniti
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Florian Huber
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Michal Bassani‐Sternberg
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - George Coukos
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
- Department of OncologyImmuno‐Oncology ServiceLausanne University HospitalLausanneCH‐1011Switzerland
| | - Alexandre Harari
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Center for Cell TherapyCHUV‐Ludwig InstituteLausanneCH‐1011Switzerland
| | - Vincent Zoete
- Department of OncologyLudwig Institute for Cancer ResearchLausanne BranchLausanne University Hospital (CHUV) and University of Lausanne (UNIL)Agora Cancer Research CenterLausanneCH‐1005Switzerland
- Molecular Modeling GroupSIB Swiss Institute of BioinformaticsUniversity of LausanneQuartier UNIL‐Sorge, Bâtiment AmphipoleLausanneCH‐1015Switzerland
| |
Collapse
|
27
|
Yao Z, Zeng Y, Liu C, Jin H, Wang H, Zhang Y, Ding C, Chen G, Wu D. Focusing on CD8 + T-cell phenotypes: improving solid tumor therapy. J Exp Clin Cancer Res 2024; 43:266. [PMID: 39342365 PMCID: PMC11437975 DOI: 10.1186/s13046-024-03195-5] [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: 07/31/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Vigorous CD8+ T cells play a crucial role in recognizing tumor cells and combating solid tumors. How T cells efficiently recognize and target tumor antigens, and how they maintain the activity in the "rejection" of solid tumor microenvironment, are major concerns. Recent advances in understanding of the immunological trajectory and lifespan of CD8+ T cells have provided guidance for the design of more optimal anti-tumor immunotherapy regimens. Here, we review the newly discovered methods to enhance the function of CD8+ T cells against solid tumors, focusing on optimizing T cell receptor (TCR) expression, improving antigen recognition by engineered T cells, enhancing signal transduction of the TCR-CD3 complex, inducing the homing of polyclonal functional T cells to tumors, reversing T cell exhaustion under chronic antigen stimulation, and reprogramming the energy and metabolic pathways of T cells. We also discuss how to participate in the epigenetic changes of CD8+ T cells to regulate two key indicators of anti-tumor responses, namely effectiveness and persistence.
Collapse
Affiliation(s)
- Zhouchi Yao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yayun Zeng
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Cheng Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huimin Jin
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hong Wang
- Department of Scientific Research, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yue Zhang
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Chengming Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
28
|
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
|
29
|
Glukhov E, Kalitin D, Stepanenko D, Zhu Y, Nguyen T, Jones G, Patsahan T, Simmerling C, Mitchell JC, Vajda S, Dill KA, Padhorny D, Kozakov D. MHC-Fine: Fine-tuned AlphaFold for precise MHC-peptide complex prediction. Biophys J 2024; 123:2902-2909. [PMID: 38751115 PMCID: PMC11393670 DOI: 10.1016/j.bpj.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/13/2024] [Accepted: 05/10/2024] [Indexed: 05/28/2024] Open
Abstract
The precise prediction of major histocompatibility complex (MHC)-peptide complex structures is pivotal for understanding cellular immune responses and advancing vaccine design. In this study, we enhanced AlphaFold's capabilities by fine-tuning it with a specialized dataset consisting of exclusively high-resolution class I MHC-peptide crystal structures. This tailored approach aimed to address the generalist nature of AlphaFold's original training, which, while broad-ranging, lacked the granularity necessary for the high-precision demands of class I MHC-peptide interaction prediction. A comparative analysis was conducted against the homology-modeling-based method Pandora as well as the AlphaFold multimer model. Our results demonstrate that our fine-tuned model outperforms others in terms of root-mean-square deviation (median value for Cα atoms for peptides is 0.66 Å) and also provides enhanced predicted local distance difference test scores, offering a more reliable assessment of the predicted structures. These advances have substantial implications for computational immunology, potentially accelerating the development of novel therapeutics and vaccines by providing a more precise computational lens through which to view MHC-peptide interactions.
Collapse
Affiliation(s)
- Ernest Glukhov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Dmytro Kalitin
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York; Faculty of Applied Science, Ukrainian Catholic University, Lviv, Ukraine
| | - Darya Stepanenko
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Yimin Zhu
- Department of Computer Science, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Thu Nguyen
- Department of Computer Science, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - George Jones
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Taras Patsahan
- Institute for Condensed Matter Physics of the National Academy of Sciences of Ukraine, Lviv, Ukraine; Institute of Applied Mathematics and Fundamental Sciences, Lviv Polytechnic National University, Lviv, Ukraine
| | - Carlos Simmerling
- Department of Chemistry, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Julie C Mitchell
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Ken A Dill
- Department of Chemistry, Stony Brook University, Stony Brook, New York; Department of Physics and Astronomy, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Dzmitry Padhorny
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York.
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
30
|
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
|
31
|
Gaglione SA, Rosales TJ, Schmidt-Hong L, Baker BM, Birnbaum ME. SARS-CoV-2 spike does not interact with the T cell receptor or directly activate T cells. Proc Natl Acad Sci U S A 2024; 121:e2406615121. [PMID: 39042676 PMCID: PMC11295079 DOI: 10.1073/pnas.2406615121] [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: 04/02/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Suggested edit: SARS-CoV-2infection can induce multisystem inflammatory syndrome in children, which resembles superantigen-induced toxic shock syndrome. Recent work has suggested that the SARS-CoV-2 spike (S) protein could act as a superantigen by binding T cell receptors (TCRs) and inducing broad antigen-independent T cell responses. Structure-based computational modeling identified potential TCR-binding sites near the S receptor-binding domain, in addition to a site with homology to known neurotoxins. We experimentally examined the mechanism underpinning this theory-the direct interaction between the TCR and S protein. Surface plasmon resonance of recombinantly expressed S protein and TCR revealed no detectable binding. Orthogonally, we pseudotyped lentiviruses with SARS-CoV-2 S in both wild-type and prefusion-stabilized forms, demonstrated their functionality in a cell line assay, and observed no transduction, activation, or stimulation of proliferation of CD8+ T cells. We conclude that it is unlikely that the SARS-CoV-2 spike protein engages nonspecifically with TCRs or has superantigenic character.
Collapse
Affiliation(s)
- Stephanie A. Gaglione
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Tatiana J. Rosales
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN46556
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN46556
| | - Laura Schmidt-Hong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Brian M. Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN46556
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN46556
| | - Michael E. Birnbaum
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA02139
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
Wu D, Yin R, Chen G, Ribeiro-Filho HV, Cheung M, Robbins PF, Mariuzza RA, Pierce BG. Structural characterization and AlphaFold modeling of human T cell receptor recognition of NRAS cancer neoantigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595215. [PMID: 38826362 PMCID: PMC11142219 DOI: 10.1101/2024.05.21.595215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
T cell receptors (TCRs) that recognize cancer neoantigens are important for anti-cancer immune responses and immunotherapy. Understanding the structural basis of TCR recognition of neoantigens provides insights into their exquisite specificity and can enable design of optimized TCRs. We determined crystal structures of a human TCR in complex with NRAS Q61K and Q61R neoantigen peptides and HLA-A1 MHC, revealing the molecular underpinnings for dual recognition and specificity versus wild-type NRAS peptide. We then used multiple versions of AlphaFold to model the corresponding complex structures, given the challenge of immune recognition for such methods. Interestingly, one implementation of AlphaFold2 (TCRmodel2) was able to generate accurate models of the complexes, while AlphaFold3 also showed strong performance, although success was lower for other complexes. This study provides insights into TCR recognition of a shared cancer neoantigen, as well as the utility and practical considerations for using AlphaFold to model TCR-peptide-MHC complexes.
Collapse
Affiliation(s)
- Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Rui Yin
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Helder V. Ribeiro-Filho
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas 13083-100, Brazil
| | - Melyssa Cheung
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Paul F. Robbins
- Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Roy A. Mariuzza
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Brian G. Pierce
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
37
|
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
|
38
|
McMaster B, Thorpe C, Ogg G, Deane CM, Koohy H. Can AlphaFold's breakthrough in protein structure help decode the fundamental principles of adaptive cellular immunity? Nat Methods 2024; 21:766-776. [PMID: 38654083 DOI: 10.1038/s41592-024-02240-7] [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: 08/23/2023] [Accepted: 03/08/2024] [Indexed: 04/25/2024]
Abstract
T cells are essential immune cells responsible for identifying and eliminating pathogens. Through interactions between their T-cell antigen receptors (TCRs) and antigens presented by major histocompatibility complex molecules (MHCs) or MHC-like molecules, T cells discriminate foreign and self peptides. Determining the fundamental principles that govern these interactions has important implications in numerous medical contexts. However, reconstructing a map between T cells and their antagonist antigens remains an open challenge for the field of immunology, and success of in silico reconstructions of this relationship has remained incremental. In this Perspective, we discuss the role that new state-of-the-art deep-learning models for predicting protein structure may play in resolving some of the unanswered questions the field faces linking TCR and peptide-MHC properties to T-cell specificity. We provide a comprehensive overview of structural databases and the evolution of predictive models, and highlight the breakthrough AlphaFold provided the field.
Collapse
Affiliation(s)
- Benjamin McMaster
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Christopher Thorpe
- Open Targets, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
| | - Graham Ogg
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | | | - Hashem Koohy
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Alan Turning Fellow in Health and Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
39
|
de Miranda NFCC, Scheeren FA. Immunogenetic Diversity and Cancer Immunotherapy Disparities. Cancer Discov 2024; 14:585-588. [PMID: 38571423 DOI: 10.1158/2159-8290.cd-23-1536] [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: 04/05/2024]
Abstract
SUMMARY The success of checkpoint blockade cancer immunotherapies has unequivocally confirmed the critical role of T cells in cancer immunity and boosted the development of immunotherapeutic strategies targeting specific antigens on cancer cells. The vast immunogenetic diversity of human leukocyte antigen (HLA) class I alleles across populations is a key factor influencing the advancement of HLA class I-restricted therapies and related research and diagnostic tools.
Collapse
Affiliation(s)
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
40
|
Shao W, Yao Y, Yang L, Li X, Ge T, Zheng Y, Zhu Q, Ge S, Gu X, Jia R, Song X, Zhuang A. Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy. Exp Hematol Oncol 2024; 13:37. [PMID: 38570883 PMCID: PMC10988985 DOI: 10.1186/s40164-024-00504-8] [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: 12/08/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Weihuan Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiaoran Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Qiuyi Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
41
|
Zhang M, Wang Z, Cui J, Ma X, Qiao Z, Kong X, Xu R, Li S, Du J, Zhao C. Development and characterization of 40 InDel markers from the major histocompatibility complex genes of largemouth bass (Micropterus salmoides). Mol Biol Rep 2024; 51:470. [PMID: 38551799 DOI: 10.1007/s11033-024-09429-1] [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: 01/19/2024] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The genetic improvement in growth and food habit domestication of largemouth bass (Micropterus salmoides) have made breakthroughs in past decades, while the relevant work on disease resistance were rarely carried out. Major histocompatibility complex (MHC) genes, which are well known as their numbers and high polymorphisms, have been used as candidate genes to mine disease-resistant-related molecular markers in many species. METHODS AND RESULTS In present study, we developed and characterized 40 polymorphic and biallelic InDel markers from the major histocompatibility complex genes of largemouth bass. The minor allele frequency, observed heterozygosity, expected heterozygosity and polymorphic information content of these markers ranged from 0.0556 to 0.5000, 0.1111 to 0.6389, 0.1064 to 0.5070, and 0.0994 to 0.3750, respectively. Three loci deviated significantly from Hardy-Weinberg equilibrium, while linkage disequilibrium existed at none of these loci. CONCLUSION These InDel markers might provide references for the further correlation analysis and molecular assisted selection of disease resistance in largemouth bass.
Collapse
Affiliation(s)
- Meng Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China.
- Hangzhou Xiaoshan Donghai Aquaculture Co., Ltd, Hangzhou, 310012, China.
- Hebei Key Laboratory of Marine Biological Resources and Environment, Hebei Ocean and Fisheries Science Research Institute, Qinhuangdao, 066200, China.
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China.
| | - Zerui Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Jiao Cui
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Xiao Ma
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zhigang Qiao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Xianghui Kong
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Ruwei Xu
- Hangzhou Xiaoshan Donghai Aquaculture Co., Ltd, Hangzhou, 310012, China
| | - Shengjie Li
- Pearl River Fisheries Research Institute, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Chinese Academy of Fisheries Sciences, China Ministry of Agriculture, Guangzhou, 510380, China
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Jinxing Du
- Pearl River Fisheries Research Institute, Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Chinese Academy of Fisheries Sciences, China Ministry of Agriculture, Guangzhou, 510380, China
| | - Chunlong Zhao
- Hebei Key Laboratory of Marine Biological Resources and Environment, Hebei Ocean and Fisheries Science Research Institute, Qinhuangdao, 066200, China.
| |
Collapse
|
42
|
Ji H, Wang XX, Zhang Q, Zhang C, Zhang HM. Predicting TCR sequences for unseen antigen epitopes using structural and sequence features. Brief Bioinform 2024; 25:bbae210. [PMID: 38711371 PMCID: PMC11074592 DOI: 10.1093/bib/bbae210] [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: 02/19/2024] [Revised: 04/04/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
T-cell receptor (TCR) recognition of antigens is fundamental to the adaptive immune response. With the expansion of experimental techniques, a substantial database of matched TCR-antigen pairs has emerged, presenting opportunities for computational prediction models. However, accurately forecasting the binding affinities of unseen antigen-TCR pairs remains a major challenge. Here, we present convolutional-self-attention TCR (CATCR), a novel framework tailored to enhance the prediction of epitope and TCR interactions. Our approach utilizes convolutional neural networks to extract peptide features from residue contact matrices, as generated by OpenFold, and a transformer to encode segment-based coded sequences. We introduce CATCR-D, a discriminator that can assess binding by analyzing the structural and sequence features of epitopes and CDR3-β regions. Additionally, the framework comprises CATCR-G, a generative module designed for CDR3-β sequences, which applies the pretrained encoder to deduce epitope characteristics and a transformer decoder for predicting matching CDR3-β sequences. CATCR-D achieved an AUROC of 0.89 on previously unseen epitope-TCR pairs and outperformed four benchmark models by a margin of 17.4%. CATCR-G has demonstrated high precision, recall and F1 scores, surpassing 95% in bidirectional encoder representations from transformers score assessments. Our results indicate that CATCR is an effective tool for predicting unseen epitope-TCR interactions. Incorporating structural insights enhances our understanding of the general rules governing TCR-epitope recognition significantly. The ability to predict TCRs for novel epitopes using structural and sequence information is promising, and broadening the repository of experimental TCR-epitope data could further improve the precision of epitope-TCR binding predictions.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Humans
- Epitopes/chemistry
- Epitopes/immunology
- Computational Biology/methods
- Neural Networks, Computer
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Antigens/chemistry
- Antigens/immunology
- Amino Acid Sequence
Collapse
Affiliation(s)
- Hongchen Ji
- Department of Oncology of Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xiang-Xu Wang
- Department of Oncology of Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Qiong Zhang
- Department of Oncology of Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Chengkai Zhang
- Department of Oncology of Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Hong-Mei Zhang
- Department of Oncology of Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
43
|
Mikhaylov V, Brambley CA, Keller GLJ, Arbuiso AG, Weiss LI, Baker BM, Levine AJ. Accurate modeling of peptide-MHC structures with AlphaFold. Structure 2024; 32:228-241.e4. [PMID: 38113889 PMCID: PMC10872456 DOI: 10.1016/j.str.2023.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/17/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023]
Abstract
Major histocompatibility complex (MHC) proteins present peptides on the cell surface for T cell surveillance. Reliable in silico prediction of which peptides would be presented and which T cell receptors would recognize them is an important problem in structural immunology. Here, we introduce an AlphaFold-based pipeline for predicting the three-dimensional structures of peptide-MHC complexes for class I and class II MHC molecules. Our method demonstrates high accuracy, outperforming existing tools in class I modeling accuracy and class II peptide register prediction. We validate its performance and utility with new experimental data on a recently described cancer neoantigen/wild-type peptide pair and explore applications toward improving peptide-MHC binding prediction.
Collapse
Affiliation(s)
- Victor Mikhaylov
- The Simons Center for Systems Biology, Institute for Advanced Study, 1 Einstein Drive, Princeton, NJ 08540, USA.
| | - Chad A Brambley
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Grant L J Keller
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Alyssa G Arbuiso
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Laura I Weiss
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Arnold J Levine
- The Simons Center for Systems Biology, Institute for Advanced Study, 1 Einstein Drive, Princeton, NJ 08540, USA
| |
Collapse
|
44
|
Bravi B. Development and use of machine learning algorithms in vaccine target selection. NPJ Vaccines 2024; 9:15. [PMID: 38242890 PMCID: PMC10798987 DOI: 10.1038/s41541-023-00795-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/07/2023] [Indexed: 01/21/2024] Open
Abstract
Computer-aided discovery of vaccine targets has become a cornerstone of rational vaccine design. In this article, I discuss how Machine Learning (ML) can inform and guide key computational steps in rational vaccine design concerned with the identification of B and T cell epitopes and correlates of protection. I provide examples of ML models, as well as types of data and predictions for which they are built. I argue that interpretable ML has the potential to improve the identification of immunogens also as a tool for scientific discovery, by helping elucidate the molecular processes underlying vaccine-induced immune responses. I outline the limitations and challenges in terms of data availability and method development that need to be addressed to bridge the gap between advances in ML predictions and their translational application to vaccine design.
Collapse
Affiliation(s)
- Barbara Bravi
- Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
45
|
Ricker CA, Meli K, Van Allen EM. Historical perspective and future directions: computational science in immuno-oncology. J Immunother Cancer 2024; 12:e008306. [PMID: 38191244 PMCID: PMC10826578 DOI: 10.1136/jitc-2023-008306] [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] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Immuno-oncology holds promise for transforming patient care having achieved durable clinical response rates across a variety of advanced and metastatic cancers. Despite these achievements, only a minority of patients respond to immunotherapy, underscoring the importance of elucidating molecular mechanisms responsible for response and resistance to inform the development and selection of treatments. Breakthroughs in molecular sequencing technologies have led to the generation of an immense amount of genomic and transcriptomic sequencing data that can be mined to uncover complex tumor-immune interactions using computational tools. In this review, we discuss existing and emerging computational methods that contextualize the composition and functional state of the tumor microenvironment, infer the reactivity and clonal dynamics from reconstructed immune cell receptor repertoires, and predict the antigenic landscape for immune cell recognition. We further describe the advantage of multi-omics analyses for capturing multidimensional relationships and artificial intelligence techniques for integrating omics data with histopathological and radiological images to encapsulate patterns of treatment response and tumor-immune biology. Finally, we discuss key challenges impeding their widespread use and clinical application and conclude with future perspectives. We are hopeful that this review will both serve as a guide for prospective researchers seeking to use existing tools for scientific discoveries and inspire the optimization or development of novel tools to enhance precision, ultimately expediting advancements in immunotherapy that improve patient survival and quality of life.
Collapse
Affiliation(s)
- Cora A Ricker
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kevin Meli
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
46
|
Chang L, Mondal A, Singh B, Martínez-Noa Y, Perez A. Revolutionizing Peptide-Based Drug Discovery: Advances in the Post-AlphaFold Era. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL MOLECULAR SCIENCE 2024; 14:e1693. [PMID: 38680429 PMCID: PMC11052547 DOI: 10.1002/wcms.1693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 05/01/2024]
Abstract
Peptide-based drugs offer high specificity, potency, and selectivity. However, their inherent flexibility and differences in conformational preferences between their free and bound states create unique challenges that have hindered progress in effective drug discovery pipelines. The emergence of AlphaFold (AF) and Artificial Intelligence (AI) presents new opportunities for enhancing peptide-based drug discovery. We explore recent advancements that facilitate a successful peptide drug discovery pipeline, considering peptides' attractive therapeutic properties and strategies to enhance their stability and bioavailability. AF enables efficient and accurate prediction of peptide-protein structures, addressing a critical requirement in computational drug discovery pipelines. In the post-AF era, we are witnessing rapid progress with the potential to revolutionize peptide-based drug discovery such as the ability to rank peptide binders or classify them as binders/non-binders and the ability to design novel peptide sequences. However, AI-based methods are struggling due to the lack of well-curated datasets, for example to accommodate modified amino acids or unconventional cyclization. Thus, physics-based methods, such as docking or molecular dynamics simulations, continue to hold a complementary role in peptide drug discovery pipelines. Moreover, MD-based tools offer valuable insights into binding mechanisms, as well as the thermodynamic and kinetic properties of complexes. As we navigate this evolving landscape, a synergistic integration of AI and physics-based methods holds the promise of reshaping the landscape of peptide-based drug discovery.
Collapse
Affiliation(s)
- Liwei Chang
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Arup Mondal
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Bhumika Singh
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | | | - Alberto Perez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32611
| |
Collapse
|
47
|
Kleiman DE, Nadeem H, Shukla D. Adaptive Sampling Methods for Molecular Dynamics in the Era of Machine Learning. J Phys Chem B 2023; 127:10669-10681. [PMID: 38081185 DOI: 10.1021/acs.jpcb.3c04843] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Molecular dynamics (MD) simulations are fundamental computational tools for the study of proteins and their free energy landscapes. However, sampling protein conformational changes through MD simulations is challenging due to the relatively long time scales of these processes. Many enhanced sampling approaches have emerged to tackle this problem, including biased sampling and path-sampling methods. In this Perspective, we focus on adaptive sampling algorithms. These techniques differ from other approaches because the thermodynamic ensemble is preserved and the sampling is enhanced solely by restarting MD trajectories at particularly chosen seeds rather than introducing biasing forces. We begin our treatment with an overview of theoretically transparent methods, where we discuss principles and guidelines for adaptive sampling. Then, we present a brief summary of select methods that have been applied to realistic systems in the past. Finally, we discuss recent advances in adaptive sampling methodology powered by deep learning techniques, as well as their shortcomings.
Collapse
Affiliation(s)
- Diego E Kleiman
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hassan Nadeem
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Diwakar Shukla
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
48
|
Glukhov E, Kalitin D, Stepanenko D, Zhu Y, Nguyen T, Jones G, Simmerling C, Mitchell JC, Vajda S, Dill KA, Padhorny D, Kozakov D. MHC-Fine: Fine-tuned AlphaFold for Precise MHC-Peptide Complex Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569310. [PMID: 38077000 PMCID: PMC10705405 DOI: 10.1101/2023.11.29.569310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
The precise prediction of Major Histocompatibility Complex (MHC)-peptide complex structures is pivotal for understanding cellular immune responses and advancing vaccine design. In this study, we enhanced AlphaFold's capabilities by fine-tuning it with a specialized dataset comprised by exclusively high-resolution MHC-peptide crystal structures. This tailored approach aimed to address the generalist nature of AlphaFold's original training, which, while broad-ranging, lacked the granularity necessary for the high-precision demands of MHC-peptide interaction prediction. A comparative analysis was conducted against the homology-modeling-based method Pandora [13], as well as the AlphaFold multimer model [8]. Our results demonstrate that our fine-tuned model outperforms both in terms of RMSD (median value is 0.65 Å) but also provides enhanced predicted lDDT scores, offering a more reliable assessment of the predicted structures. These advances have substantial implications for computational immunology, potentially accelerating the development of novel therapeutics and vaccines by providing a more precise computational lens through which to view MHC-peptide interactions.
Collapse
Affiliation(s)
- Ernest Glukhov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Dmytro Kalitin
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Darya Stepanenko
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Yimin Zhu
- Department of Computer Science, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Thu Nguyen
- Department of Computer Science, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - George Jones
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Carlos Simmerling
- Department of Chemistry, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Julie C. Mitchell
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, 02215, MA, USA
| | - Ken A. Dill
- Department of Chemistry, Stony Brook University, Stony Brook, 11794, NY, USA
- Department of Physics and Astronomy, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Dzmitry Padhorny
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, 11794, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, 11794, NY, USA
| |
Collapse
|
49
|
Mariuzza RA, Wu D, Pierce BG. Structural basis for T cell recognition of cancer neoantigens and implications for predicting neoepitope immunogenicity. Front Immunol 2023; 14:1303304. [PMID: 38045695 PMCID: PMC10693334 DOI: 10.3389/fimmu.2023.1303304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023] Open
Abstract
Adoptive cell therapy (ACT) with tumor-specific T cells has been shown to mediate durable cancer regression. Tumor-specific T cells are also the basis of other therapies, notably cancer vaccines. The main target of tumor-specific T cells are neoantigens resulting from mutations in self-antigens over the course of malignant transformation. The detection of neoantigens presents a major challenge to T cells because of their high structural similarity to self-antigens, and the need to avoid autoimmunity. How different a neoantigen must be from its wild-type parent for it to induce a T cell response is poorly understood. Here we review recent structural and biophysical studies of T cell receptor (TCR) recognition of shared cancer neoantigens derived from oncogenes, including p53R175H, KRASG12D, KRASG12V, HHATp8F, and PIK3CAH1047L. These studies have revealed that, in some cases, the oncogenic mutation improves antigen presentation by strengthening peptide-MHC binding. In other cases, the mutation is detected by direct interactions with TCR, or by energetically driven or other indirect strategies not requiring direct TCR contacts with the mutation. We also review antibodies designed to recognize peptide-MHC on cell surfaces (TCR-mimic antibodies) as an alternative to TCRs for targeting cancer neoantigens. Finally, we review recent computational advances in this area, including efforts to predict neoepitope immunogenicity and how these efforts may be advanced by structural information on peptide-MHC binding and peptide-MHC recognition by TCRs.
Collapse
Affiliation(s)
- Roy A. Mariuzza
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, United States
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Brian G. Pierce
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD, United States
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| |
Collapse
|
50
|
Gupta S, Nerli S, Kutti Kandy S, Mersky GL, Sgourakis NG. HLA3DB: comprehensive annotation of peptide/HLA complexes enables blind structure prediction of T cell epitopes. Nat Commun 2023; 14:6349. [PMID: 37816745 PMCID: PMC10564892 DOI: 10.1038/s41467-023-42163-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
The class I proteins of the major histocompatibility complex (MHC-I) display epitopic peptides derived from endogenous proteins on the cell surface for immune surveillance. Accurate modeling of peptides bound to the human MHC, HLA, has been mired by conformational diversity of the central peptide residues, which are critical for recognition by T cell receptors. Here, analysis of X-ray crystal structures within our curated database (HLA3DB) shows that pHLA complexes encompassing multiple HLA allotypes present a discrete set of peptide backbone conformations. Leveraging these backbones, we employ a regression model trained on terms of a physically relevant energy function to develop a comparative modeling approach for nonamer pHLA structures named RepPred. Our method outperforms the top pHLA modeling approach by up to 19% in structural accuracy, and consistently predicts blind targets not included in our training set. Insights from our work may be applied towards predicting antigen immunogenicity, and receptor cross-reactivity.
Collapse
Affiliation(s)
- Sagar Gupta
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Santrupti Nerli
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sreeja Kutti Kandy
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Glenn L Mersky
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nikolaos G Sgourakis
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|