Balancing sensitivity and specificity in distinguishing TCR groups by CDR sequence similarity

BackgroundRepertoire sequencing is enabling deep explorations into the cellular immune response, including the characterization of commonalities and differences among T cell receptor (TCR) repertoires from different individuals, pathologies, and antigen specificities. In seeking to understand the generality of patterns observed in different groups of TCRs, it is necessary to balance how well each pattern represents the diversity among TCRs from one group (sensitivity) vs. how many TCRs from other groups it also represents (specificity). The variable complementarity determining regions (CDRs), particularly the third CDRs (CDR3s) interact with major histocompatibility complex (MHC)-presented epitopes from putative antigens, and thus encode the determinants of recognition.ResultsWe here systematically characterize the predictive power that can be obtained from CDR3 sequences, using representative, readily interpretable methods for evaluating CDR sequence similarity and then clustering and classifying sequences based on similarity. An initial analysis of CDR3s of known structure, clustered by structural similarity, helps calibrate the limits of sequence diversity among CDRs that might have a common mode of interaction with presented epitopes. Subsequent analyses demonstrate that this same range of sequence similarity strikes a favorable specificity/sensitivity balance in distinguishing twins from non-twins based on overall CDR3 repertoires, classifying CDR3 repertoires by antigen specificity, and distinguishing general pathologies.ConclusionWe conclude that within a fairly broad range of sequence similarity, matching CDR3 sequences are likely to share specificities.

[1]  Regina S. Salvat,et al.  Structure-based redesign of lysostaphin yields potent antistaphylococcal enzymes that evade immune cell surveillance , 2015, Molecular therapy. Methods & clinical development.

[2]  Abigail Wacher,et al.  Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. , 2009, Blood.

[3]  Sandeep Kumar Dhanda,et al.  A Review on T Cell Epitopes Identified Using Prediction and Cell-Mediated Immune Models for Mycobacterium tuberculosis and Bordetella pertussis , 2018, Front. Immunol..

[4]  Charlotte M. Deane,et al.  STCRDab: the structural T-cell receptor database , 2017, Nucleic Acids Res..

[5]  O. Lund,et al.  Definition of supertypes for HLA molecules using clustering of specificity matrices , 2004, Immunogenetics.

[6]  Mark M. Davis,et al.  Deconstructing the Peptide-MHC Specificity of T Cell Recognition , 2014, Cell.

[7]  Bjoern Peters,et al.  Molecular Determinants of T Cell Epitope Recognition to the Common Timothy Grass Allergen , 2010, The Journal of Immunology.

[8]  C. Bailey-Kellogg,et al.  Design and engineering of deimmunized biotherapeutics. , 2016, Current opinion in structural biology.

[9]  D. Price,et al.  A Single Autoimmune T Cell Receptor Recognizes More Than a Million Different Peptides* , 2011, The Journal of Biological Chemistry.

[10]  Mikhail Shugay,et al.  Distinctive properties of identical twins' TCR repertoires revealed by high-throughput sequencing , 2014, Proceedings of the National Academy of Sciences.

[11]  Olivier Michielin,et al.  TCRep 3D: An Automated In Silico Approach to Study the Structural Properties of TCR Repertoires , 2011, PloS one.

[12]  Chris Bailey-Kellogg,et al.  Computationally optimized deimmunization libraries yield highly mutated enzymes with low immunogenicity and enhanced activity , 2017, Proceedings of the National Academy of Sciences.

[13]  Sai T Reddy,et al.  Advanced Methodologies in High-Throughput Sequencing of Immune Repertoires. , 2017, Trends in biotechnology.

[14]  G. S. Pandey,et al.  Post hoc assessment of the immunogenicity of bioengineered factor VIIa demonstrates the use of preclinical tools , 2017, Science Translational Medicine.

[15]  W. A. Beyer,et al.  Some Biological Sequence Metrics , 1976 .

[16]  M. Hilleman Strategies and mechanisms for host and pathogen survival in acute and persistent viral infections , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[17]  Alessandro Sette,et al.  Identifying specificity groups in the T cell receptor repertoire , 2017, Nature.

[18]  Chris Bailey-Kellogg,et al.  Mapping the Pareto Optimal Design Space for a Functionally Deimmunized Biotherapeutic Candidate , 2015, PLoS Comput. Biol..

[19]  R. Houghten,et al.  Predictable TCR antigen recognition based on peptide scans leads to the identification of agonist ligands with no sequence homology. , 1998, Journal of immunology.

[20]  Andrew Sewell,et al.  Structural and kinetic basis for heightened immunogenicity of T cell vaccines , 2005, The Journal of experimental medicine.

[21]  P. Bradley,et al.  Quantifiable predictive features define epitope-specific T cell receptor repertoires , 2017, Nature.

[22]  Mark M Davis,et al.  Linking T-cell receptor sequence to functional phenotype at the single-cell level , 2014, Nature Biotechnology.

[23]  Amelie Stein,et al.  Improvements to Robotics-Inspired Conformational Sampling in Rosetta , 2013, PloS one.

[24]  Rob J. De Boer,et al.  Degenerate T-cell Recognition of Peptides on MHC Molecules Creates Large Holes in the T-cell Repertoire , 2012, PLoS Comput. Biol..

[25]  Jun S. Liu,et al.  Comprehensive analyses of tumor immunity: implications for cancer immunotherapy , 2016, Genome Biology.

[26]  Mark M. Davis,et al.  T-cell antigen receptor genes and T-cell recognition , 1988, Nature.

[27]  Richard A. Olshen,et al.  Diversity and clonal selection in the human T-cell repertoire , 2014, Proceedings of the National Academy of Sciences.

[28]  Chris Bailey-Kellogg,et al.  Depletion of T cell epitopes in lysostaphin mitigates anti-drug antibody response and enhances antibacterial efficacy in vivo. , 2015, Chemistry & biology.

[29]  Eric Jones,et al.  SciPy: Open Source Scientific Tools for Python , 2001 .

[30]  J. Sidney,et al.  Nine major HLA class I supertypes account for the vast preponderance of HLA-A and -B polymorphism , 1999, Immunogenetics.

[31]  Richard A. Moore,et al.  Exhaustive T-cell repertoire sequencing of human peripheral blood samples reveals signatures of antigen selection and a directly measured repertoire size of at least 1 million clonotypes. , 2011, Genome research.

[32]  Ravi V. Kolla,et al.  T Cell Responses to Known Allergen Proteins Are Differently Polarized and Account for a Variable Fraction of Total Response to Allergen Extracts , 2012, The Journal of Immunology.

[33]  Andreas Suhrbier,et al.  Phase I Trial of a CD8+ T-Cell Peptide Epitope-Based Vaccine for Infectious Mononucleosis , 2007, Journal of Virology.

[34]  Ragul Gowthaman,et al.  TCRmodel: high resolution modeling of T cell receptors from sequence , 2018, Nucleic Acids Res..

[35]  R. Holt,et al.  Profiling the T-cell receptor beta-chain repertoire by massively parallel sequencing. , 2009, Genome research.

[36]  W. Delano The PyMOL Molecular Graphics System , 2002 .

[37]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[38]  J. Shawe-Taylor,et al.  Specificity, Privacy, and Degeneracy in the CD4 T Cell Receptor Repertoire Following Immunization , 2017, Front. Immunol..

[39]  A. Steere,et al.  Identification of LFA-1 as a candidate autoantigen in treatment-resistant Lyme arthritis. , 1998, Science.

[40]  C. Bailey-Kellogg,et al.  Hit-and-run, hit-and-stay, and commensal bacteria present different peptide content when viewed from the perspective of the T cell. , 2015, Vaccine.

[41]  Jared Becksfort,et al.  Paired analysis of TCRα and TCRβ chains at the single-cell level in mice. , 2011, The Journal of clinical investigation.

[42]  Sergey Lukyanov,et al.  Next generation sequencing for TCR repertoire profiling: Platform‐specific features and correction algorithms , 2012, European journal of immunology.

[43]  S. Henikoff,et al.  Amino acid substitution matrices from protein blocks. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[44]  C. Bailey-Kellogg,et al.  HCV epitope, homologous to multiple human protein sequences, induces a regulatory T cell response in infected patients. , 2015, Journal of hepatology.

[45]  Yuxin Sun,et al.  Feature selection using a one dimensional naïve Bayes’ classifier increases the accuracy of support vector machine classification of CDR3 repertoires , 2017, Bioinform..

[46]  Jaime Prilusky,et al.  McPAS‐TCR: a manually curated catalogue of pathology‐associated T cell receptor sequences , 2017, Bioinform..

[47]  T. D. Schneider,et al.  Sequence logos: a new way to display consensus sequences. , 1990, Nucleic acids research.

[48]  George Georgiou,et al.  Therapeutic enzyme deimmunization by combinatorial T-cell epitope removal using neutral drift , 2011, Proceedings of the National Academy of Sciences.

[49]  David Baker,et al.  Removing T-cell epitopes with computational protein design , 2014, Proceedings of the National Academy of Sciences.

[50]  A. Casrouge,et al.  A direct estimate of the human alphabeta T cell receptor diversity. , 1999, Science.

[51]  Robyn L Stanfield,et al.  How TCRs bind MHCs, peptides, and coreceptors. , 2006, Annual review of immunology.

[52]  Irini Doytchinova,et al.  T-cell epitope vaccine design by immunoinformatics , 2013, Open Biology.

[53]  J. Calis,et al.  Characterizing immune repertoires by high throughput sequencing: strategies and applications. , 2014, Trends in immunology.

[54]  Charles H. Yoon,et al.  An immunogenic personal neoantigen vaccine for patients with melanoma , 2017, Nature.

[55]  A. Casrouge,et al.  A Direct Estimate of the Human αβ T Cell Receptor Diversity , 1999 .

[56]  James Theiler,et al.  Graph‐based optimization of epitope coverage for vaccine antigen design , 2017, Statistics in medicine.

[57]  Cédric R. Weber,et al.  Computational Strategies for Dissecting the High-Dimensional Complexity of Adaptive Immune Repertoires , 2017, Front. Immunol..

[58]  Jimmy Lin,et al.  Mining Exomic Sequencing Data to Identify Mutated Antigens Recognized by Adoptively Transferred Tumor-reactive T cells , 2013, Nature Medicine.

[59]  Bartek Wilczynski,et al.  Biopython: freely available Python tools for computational molecular biology and bioinformatics , 2009, Bioinform..

[60]  D. Wiley,et al.  T Cell Receptor–MHC Interactions up Close , 2001, Cell.

[61]  Chris Bailey-Kellogg,et al.  Integrated assessment of predicted MHC binding and cross-conservation with self reveals patterns of viral camouflage , 2014, BMC Bioinformatics.

[62]  Y. Louzoun,et al.  Rep‐Seq: uncovering the immunological repertoire through next‐generation sequencing , 2012, Immunology.

[63]  Chris Bailey-Kellogg,et al.  Protein deimmunization via structure‐based design enables efficient epitope deletion at high mutational loads , 2015, Biotechnology and bioengineering.

[64]  G. Crooks,et al.  WebLogo: a sequence logo generator. , 2004, Genome research.

[65]  James Theiler,et al.  Polyvalent vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants , 2007, Nature Medicine.

[66]  William S. DeWitt,et al.  Human T cell receptor occurrence patterns encode immune history, genetic background, and receptor specificity , 2018, bioRxiv.

[67]  Cédric R. Weber,et al.  Learning the High-Dimensional Immunogenomic Features That Predict Public and Private Antibody Repertoires , 2017, The Journal of Immunology.

[68]  Bette Korber,et al.  Mosaic HIV-1 Vaccines Expand the Breadth and Depth of Cellular Immune Responses in Rhesus Monkeys , 2010, Nature Medicine.