T cells are integral components of the adaptive immune system, and their responses are mediated by unique T-cell receptors (TCR) that recognize specific antigens from a variety of biological contexts. As a result, analyzing the T-cell repertoire offers a better understanding of immune responses and of diseases like cancer. Next-generation sequencing technologies have greatly enabled the high-throughput analysis of the TCR repertoire. On the basis of our extensive experience in the field from the past decade, we provide an overview of TCR sequencing, from the initial library preparation steps to sequencing and analysis methods and finally to functional validation techniques. With regards to data analysis, we detail important TCR repertoire metrics and present several computational tools for predicting antigen specificity. Finally, we highlight important applications of TCR sequencing and repertoire analysis to understanding tumor biology and developing cancer immunotherapies.

Our immune system is composed of the innate and adaptive immune systems; the innate immune system protects against general threats and conserved pathogenic sequences while the adaptive immune system targets and retains memory of specific pathogens (1). T cells are critical mediators of adaptive immunity, specifically driving cellular immunity as opposed to B cells that govern humoral immunity. Functionally, T cells kill infected cells or release cytokines to recruit other immune cells after recognizing foreign antigens presented on infected cells (2). Immature T cells migrate from hemopoietic tissues to the thymus where they mature into naïve T cells that express a unique and functional T-cell receptor (TCR) that grants them the ability to recognize specific antigens (3). The thymus is also where T cells differentiate from an initial CD4/CD8 double-positive state into either a CD4+ or CD8+ lineage. CD4+ T cells execute helper functions while CD8+ T cells are cytotoxic (4). Naïve T cells become antigen-experienced T cells after encountering their cognate antigen presented on major histocompatibility complex (MHC) molecules through their TCRs.

TCRs are highly diverse heterodimeric surface receptors that enable T cells to provide protection against a variety of different pathogens. A functional TCR is comprised of two paired protein chains, with either alpha (α) and beta (β) chains or gamma (γ) and delta (δ) chains (Fig. 1A). The majority of T cells express αβ TCRs that recognize antigens presented on MHC proteins (3). Only 1%–5% of T cells express γδ TCRs, which are not MHC restricted and participate in innate immune responses. It is unclear which exact ligands γδ T cells bind (5). For these reasons, αβ TCRs are typically the focus of TCR sequencing and subsequent clinical applications.

Figure 1.

Schematic representation of TCR, MHC, and V(D)J recombination. A, TCR with paired alpha and beta chains at the cell surface. In the variable region of the TCR, variable (V), diversity (D), and joining (J) gene segments are shown in dark blue, purple, and green, respectively. The constant (C) region is shown in light blue. The commonly sequenced CDR3 region of the TCRβ chain is shown in the antigen-binding site. B, V(D)J recombination of the beta chain. Initially, a D segment randomly recombines with a J segment, then the D-J segment recombines with a random V segment. The complete variable region joins with the constant region to encode a functioning beta chain. C, Interaction of TCR and pMHC. MHC class I molecule is represented with alpha chains in yellow and beta chain in orange. The peptide is shown in dark blue.

Figure 1.

Schematic representation of TCR, MHC, and V(D)J recombination. A, TCR with paired alpha and beta chains at the cell surface. In the variable region of the TCR, variable (V), diversity (D), and joining (J) gene segments are shown in dark blue, purple, and green, respectively. The constant (C) region is shown in light blue. The commonly sequenced CDR3 region of the TCRβ chain is shown in the antigen-binding site. B, V(D)J recombination of the beta chain. Initially, a D segment randomly recombines with a J segment, then the D-J segment recombines with a random V segment. The complete variable region joins with the constant region to encode a functioning beta chain. C, Interaction of TCR and pMHC. MHC class I molecule is represented with alpha chains in yellow and beta chain in orange. The peptide is shown in dark blue.

Close modal

Regardless of type, the chains of a TCR contain an N-terminus variable region capable of antigen recognition and a C-terminus constant region. The variable region is assembled from variable (V), diversity (D), and joining (J) gene segments through an ordered process called V(D)J recombination, in which one allele of each gene segment is randomly recombined with other gene segment alleles to form a functional antigen recognition region (Fig. 1B; refs. 6, 7). TCRα and γ chains are comprised of V and J gene segments, while TCRβ and δ chains also include the D gene segment, offering more diversity in structure. The overall combinatorial diversity of the gene segments is accompanied by junctional diversity, which is driven by the random addition or deletion of nucleotides at junctions between gene segments. Both combinational and junctional diversity grant T cells a substantial range of antigen specificities, potentially accounting for 1015 to 1020 possible TCR chains (8). Importantly, immature T cells commit to either an αβ or γδ lineage during V(D)J recombination, where the commitment to αβ is facilitated by the successful recombination of the β-chain and commitment to γδ is facilitated by successful recombination of both the γ and δ chain (9).

The variable domains for each of the TCR chains have three complementarity-determining regions (CDR): CDR1, CDR2, and CDR3. CDR1 and CDR2, which are encoded by the V gene segment, primarily facilitate the interaction between the TCR and MHC through contact with the conserved α-helices of MHC. CDR3 is encoded by the junction of V and J or D and J gene segments, resulting in high variability. This region is responsible for binding peptide antigens presented in the binding groove of MHC (8, 10). Because of its direct interaction with antigens and inherent hypervariability, CDR3 offers a wealth of knowledge about TCR specificity and is consequently a commonly used target region for TCR sequencing.

TCRs are of no consequence in the absence of their critical interaction partners, MHC molecules. MHC class I proteins are expressed by all nucleated cells and are recognized by cytotoxic CD8+ T cells while MHC class II proteins are expressed by professional antigen-presenting cells (APC) such as dendritic cells, macrophages, and B cells for recognition by CD4+ T cells. Antigen presentation by both classes of MHC proteins is an essential process in adaptive immunity (11, 12). These MHC molecules present processed peptides at the cell's surface for recognition by T cells through their specific TCRs (Fig. 1C). Antigen-processing pathways differ between both MHC classes; however, all pathways lead to the expression of a fully assembled peptide-MHC (pMHC) complex at the cell surface for TCR recognition (13). Structurally, the binding surface of both classes of MHC proteins is comprised of two domains; this surface originates from a single α-chain for MHC class I and from an α-chain and β-chain heterodimer for MHC class II (12). Accordingly, this diversity of MHC sequence and structure must be accounted for by any predictive software seeking to identify high-confidence MHC epitopes. Methods for predicting MHC epitopes are further discussed in the Computational Tools for Predicting Antigen Specificity section.

TCR specificity serves as the foundation for using TCR sequencing to understand T-cell biology and dynamics, as well as to study T cells in disease contexts and informing the development of therapeutics. Although identical TCRs may tend to recognize the same antigens, cross-reactivity has been observed, in which a single TCR has the ability to recognize multiple pMHC pairs (14–16). Because cross-reactivity of TCRs has the potential to complicate the characterization and surveillance of T-cell populations, it must not be overlooked in analysis of TCR sequencing and validation of antigen specificity.

The earliest approaches for characterizing the T-cell repertoire relied on Sanger sequencing. Because this approach is only able to capture small alterations in the genome, such as substitutions and short insertions/deletions, it is not robust enough to account for the immense diversity of TCRs (3). The advent of high-throughput sequencing (HTS) platforms has allowed for the rapid and comprehensive sequencing of genomic DNA (gDNA) or RNA, fueling major advances in the understanding of the T-cell repertoire (17). HTS methods, such as next-generation sequencing (NGS; ref. 12), are available for all TCR chains, but most kits target TCRαβ chains, as αβ T cells make up the majority of the T-cell population (8). While there are a multitude of different NGS platforms offered by companies like Illumina, Life Technologies, and Oxford Nanopore Technologies, the general workflow for using HTS in research remains consistent (18–20). The process begins with library preparation, followed by NGS, then computational analysis and deconvolution of the sequencing data, and finally, experimental validation (19). For TCR sequencing, each of these steps involves specialized techniques or programs designed to accommodate the complexity of TCRs.

An important first step of NGS library preparation is targeted enrichment of transcripts to be sequenced (21). Multiplex PCR and rapid amplification of cDNA ends (5′ RACE) are two commonly used methods for targeted amplification during T-cell repertoire library preparation. Both methods commonly aim to amplify the CDR3 region of the TCR, which offers the most information on antigen specificity. Multiplex PCR is widely used and it involves the amplification of either gDNA or RNA of the CDR3 region using primers for J gene segment alleles or the TCR constant region in conjunction with a mixture of primers for known V gene segment alleles. Inherently, multiplex PCR is limited by the set of primers available; because of this, novel V-alleles cannot be accurately characterized. Furthermore, primer bias within multiplex PCR risks uneven amplification of alleles, leading to inaccurate relative TCR sequence frequencies (8). Dual barcoding and unique molecular identifier (UMI) modifications to standard PCR protocols facilitate the removal of artifacts introduced during PCR amplification and support accurate downstream sequence analysis (21, 22).

Alternatively, 5′ RACE utilizes RNA and employs only one primer pair that targets the constant region of a TCR chain and the 5′ mRNA end (23). This eliminates the bias presented by multiplex PCR and allows for the capture of all TCRs present in a given sample. However, 5′ RACE may not be completely efficient in recapitulating entire recombination sequences, as it has been demonstrated to exclude V gene segments in some contexts; this necessitates the need to optimize such techniques further (8, 23). Optimizations to reduce the risk for primer bias during cDNA amplification include template switching modifications which add universal oligonucleotide sequences to 3′ ends of cDNA so that universal 3′ primers can be used during PCR amplification to amplify strands in a sequence-independent manner (24). The addition of unique barcodes to 5′ ends of cDNA further monitors for amplification errors and biases (25, 26).

With regards to sequencing methods, either bulk sequencing or single-cell sequencing can be used to analyze T-cell repertoires. Bulk sequencing, which sequences the aggregation of unpaired TCR chains within a sample, is commonly used to analyze large-scale TCR diversity and compare populations across patient cohorts (8). Historically, bulk analysis methods have focused on sequencing the TCRβ chain (or δ in γδ T cells) due to the presence of the additional D gene segment and greater combinatorial diversity compared with the α-chain. Moreover, T cells participate in “allelic exclusion,” in which only one functional β-chain is present in a T cell whereas multiple α-chains may be expressed (27). However, a major limitation of only sequencing the β-chain is that it does not provide information about αβ-chain pairings or in vivo biological function of the sequenced T cell (3, 8).

Conversely, single-cell sequencing focuses on individual immune cells. Advances in single-cell isolation techniques like fluorescence-activated cell sorting and microfluidic platforms have laid the groundwork for the success of single-cell TCR sequencing (28). While bulk sequencing offers valuable information regarding TCR diversity, it lacks the resolution of full TCRαβ chain pairing of single-cell analysis. Single-cell sequencing offers a more targeted and specific analysis of the T-cell repertoire, providing important information on paired α- and β-chain sequences; however, this method is limited by the number of cells that can be processed from a given sample and may not be capable of detecting rarer clonotypes. Overall, by analyzing gene expression in individual cells, single-cell analysis can provide a deeper understanding of intercellular genetic diversity and in vivo biological function (28).

Several companies offer TCR sequencing platforms, including Adaptive Biotechnologies (ImmunoSEQ), Beijing Genomics Institute (IR-SEQ), iRepertoire, Inc (RepSeq), Clonotech Takara Bio USA, Inc. (SMARTer Human TCR a/b Profiling Kit), CD Genomics (TCR-Seq), and Thermo Fisher Scientific (Oncomine TCR Beta-LR Assay), among others (8). Notably, 10X Genomics offers a single-cell profiling method equipped with feature barcoding technology to provide extensive information regarding gene and protein expression, paired TCR sequences, and antigen specificity (Table 1; ref. 29).

Table 1.

TCR sequencing platforms.

MethodManufacturerSequencing platformSequencing levelLibrary prep kit?MethodologyInputRegions sequencedChainsOrganismUnique features
ImmunoSEQ Adaptive Biotechnologies Illumina HiSeq and MiSeq Bulk (single chain) Yes Multiplex PCR DNA CDR3 portion of V and J genes, complete D gene TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – Extensive metrics generated for TCR repertoire analysis

  • – User-friendly online analyzer with built-in functions to access, view, analyze, and graph data

 
IR-SEQ Beijing Genomics Institute Illumina HiSeq and MiSeq Bulk and single cell (paired chains) No Multiplex PCR (DNA) 5′ RACE (RNA) DNA RNA CDR3 (DNA) CDR1 CDR2 CDR3 (RNA) TCRα TCRβ Human  
RepSeq iRepertoire, Inc. Illumina MiSeq Bulk and single cell Yes Patented multiplex arm-PCR and dam-PCR technologies DNA RNA CDR1 CDR2 CDR3 TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – In-house gDNA and RNA extraction available

  • – Short- and long-read options available

  • – Can sequence all TCR chains in a single reaction

  • – Offers single-cell phenotyping alongside TCR sequencing

 
SMARTer Human TCR a/b Profiling Kit v2 Clonotech Takara Bio USA, Inc. Illumina MiSeq Bulk and single cell Yes Combination of multiplex PCR and 5′ RACE RNA Full chain TCRα TCRβ Human Mouse 
  • – Library preparation kit includes UMIs that facilitate removal of PCR duplicate reads and reads containing sequencing errors

 
Oncomine TCR Beta-LR Assay Thermo Fisher Scientific Ion GeneStudio system Bulk Yes Multiplex PCR DNA RNA CDR1 CDR2 CDR3 TCRβ Human  
Single Cell Immune Profiling 10x genomics Illumina NovaSeq, HiSeq, NextSeq, MiSeq Single cell Yes RT PCR cDNA Full chain TCRα TCRβ Human Mouse 
  • – Offers feature barcoding technology to integrate omics and antigen specificity data with TCR repertoire analysis

 
QIAseq Immune Repertoire RNA Library Kit QIAGEN Illumina Bulk Yes RT PCR RNA Full chain TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – Library preparation kit includes UMIs that facilitate removal of PCR duplicate reads and reads containing sequencing errors

 
AmpliSeq TCRb Panel Illumina Illumina MiniSeq, MiSeq, NextSeq 1000, NextSeq 2000, NextSeq 550 Bulk Yes Multiplex PCR RNA CDR1 CDR2 CDR3 TCRβ Human  
MethodManufacturerSequencing platformSequencing levelLibrary prep kit?MethodologyInputRegions sequencedChainsOrganismUnique features
ImmunoSEQ Adaptive Biotechnologies Illumina HiSeq and MiSeq Bulk (single chain) Yes Multiplex PCR DNA CDR3 portion of V and J genes, complete D gene TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – Extensive metrics generated for TCR repertoire analysis

  • – User-friendly online analyzer with built-in functions to access, view, analyze, and graph data

 
IR-SEQ Beijing Genomics Institute Illumina HiSeq and MiSeq Bulk and single cell (paired chains) No Multiplex PCR (DNA) 5′ RACE (RNA) DNA RNA CDR3 (DNA) CDR1 CDR2 CDR3 (RNA) TCRα TCRβ Human  
RepSeq iRepertoire, Inc. Illumina MiSeq Bulk and single cell Yes Patented multiplex arm-PCR and dam-PCR technologies DNA RNA CDR1 CDR2 CDR3 TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – In-house gDNA and RNA extraction available

  • – Short- and long-read options available

  • – Can sequence all TCR chains in a single reaction

  • – Offers single-cell phenotyping alongside TCR sequencing

 
SMARTer Human TCR a/b Profiling Kit v2 Clonotech Takara Bio USA, Inc. Illumina MiSeq Bulk and single cell Yes Combination of multiplex PCR and 5′ RACE RNA Full chain TCRα TCRβ Human Mouse 
  • – Library preparation kit includes UMIs that facilitate removal of PCR duplicate reads and reads containing sequencing errors

 
Oncomine TCR Beta-LR Assay Thermo Fisher Scientific Ion GeneStudio system Bulk Yes Multiplex PCR DNA RNA CDR1 CDR2 CDR3 TCRβ Human  
Single Cell Immune Profiling 10x genomics Illumina NovaSeq, HiSeq, NextSeq, MiSeq Single cell Yes RT PCR cDNA Full chain TCRα TCRβ Human Mouse 
  • – Offers feature barcoding technology to integrate omics and antigen specificity data with TCR repertoire analysis

 
QIAseq Immune Repertoire RNA Library Kit QIAGEN Illumina Bulk Yes RT PCR RNA Full chain TCRα TCRβ TCRγ TCRδ Human Mouse 
  • – Library preparation kit includes UMIs that facilitate removal of PCR duplicate reads and reads containing sequencing errors

 
AmpliSeq TCRb Panel Illumina Illumina MiniSeq, MiSeq, NextSeq 1000, NextSeq 2000, NextSeq 550 Bulk Yes Multiplex PCR RNA CDR1 CDR2 CDR3 TCRβ Human  

Note: Many companies offer different TCR sequencing platforms. The sequencing level, information about library preparation, input material, regions and chains sequenced, organism type, and unique features are listed.

Moreover, several research groups have developed sequencing methods to address current experimental and technological limitations. For example, Howie and colleagues developed and validated pairSEQ, which assesses the diversity of TCR sequences in a sample and accurately pairs TCRα and TCRβ sequences without the need for single-cell methodologies. This platform is currently available through Adaptive Biotechnologies (30). Also of note, recently developed single-cell RNA sequencing technologies, including TCR functional landscape estimation supervised with scRNA-seq analysis (tessa) and clonotype neighbor graph analysis (CoNGA), can capture both TCR and gene expression data, mapping the functional relevance of specific TCRs (31, 32).

Metrics to characterize TCR repertoires

Computational analyses are performed on TCR sequencing data acquired from NGS to obtain useful information about the properties of the T-cell repertoire. Common metrics used in the analysis of the T-cell repertoire include T-cell density, diversity, and clonality (Fig. 2A). T-cell density refers to the total number of T cells that are found within an area. In the context of cancer, density serves as an estimate of T-cell infiltration into a tumor (33). T-cell diversity accounts for both TCR sequence “richness” and “evenness,” which reflect the number of unique TCR sequences and the distribution of TCR sequences, respectively (33, 34). Specific diversity measures include Hill numbers that refer to the number of effective species in a sample and Rényi entropy values which quantify the randomness of the given system. These measures serve as the basis for diversity-related indices that aim to assess clonal dominance, such as Shannon entropy, the Gini–Simpson index, and Pielou's coefficient (34). Clonality integrates density and diversity metrics and assesses the degree of clonal expansion of different T cells within a sample. Shannon clonality and Simpson clonality are clonality functions that are often included in TCR repertoire analysis, and they are derived from Shannon entropy and Gini–Simpson index diversity metrics, respectively. Recommended by Adaptive Biotechnologies, Simpson clonality is less sensitive to differences in sample size.

Figure 2.

Visualization of metrics to characterize T-cell repertoires. A, Density, diversity, and clonality metrics used to characterize T-cell repertoires. High metrics are contrasted with low metrics, and colors represent different T-cell clonotypes. Density refers to the proportion of T cells in a given area, diversity refers to the number of unique clonotypes in a sample, and clonality refers to expansion of clonotypes in a sample. B, Jaccard index and MOI used to compare different T-cell repertoires. High overlap is contrasted with low overlap, and colors represent different T-cell clonotypes. Jaccard index considers only clonotype, while MOI considers both clonotype and frequency. Colored spheres represent T-cell clonotypes, and gray units in the background represent tumor cells. S1, sample 1; S2, sample 2.

Figure 2.

Visualization of metrics to characterize T-cell repertoires. A, Density, diversity, and clonality metrics used to characterize T-cell repertoires. High metrics are contrasted with low metrics, and colors represent different T-cell clonotypes. Density refers to the proportion of T cells in a given area, diversity refers to the number of unique clonotypes in a sample, and clonality refers to expansion of clonotypes in a sample. B, Jaccard index and MOI used to compare different T-cell repertoires. High overlap is contrasted with low overlap, and colors represent different T-cell clonotypes. Jaccard index considers only clonotype, while MOI considers both clonotype and frequency. Colored spheres represent T-cell clonotypes, and gray units in the background represent tumor cells. S1, sample 1; S2, sample 2.

Close modal

In addition to metrics that report on the properties of a single sample, there are indices that aid in the comparison of T-cell repertoires between different samples. Two similarity indices that are routinely utilized are the Jaccard index and the Morisita overlap index (MOI). The Jaccard index and related indices like the Sorensen index are solely based on the presence or absence of specific TCR sequences across multiple samples. These methods are useful in comparing heterogeneous repertoires but are unable to compare relative frequencies of particular TCR sequences between samples. In contrast, MOI accounts for relative frequencies as well as presence of specific TCR sequences, offering a more comprehensive comparison of T-cell repertoires from different samples (Fig. 2B; ref. 34). TCR metrics and overlap data can be presented in numerous ways for publication (Fig. 3).

Figure 3.

Common representations of TCR-related data. Stacked bar graphs showing proportion of top 10 clonotypes (A), clone tracking graphs (B), Venn diagrams (C), differential abundance plots (D), heatmaps (E), phylogenetic trees (F), alluvial plots (G), and network diagrams (H); all are used to present TCR data. Such graphs offer information about the frequency and evolution of T-cell clonotypes in different patients, in different tissue types, and through different disease or therapeutic conditions.

Figure 3.

Common representations of TCR-related data. Stacked bar graphs showing proportion of top 10 clonotypes (A), clone tracking graphs (B), Venn diagrams (C), differential abundance plots (D), heatmaps (E), phylogenetic trees (F), alluvial plots (G), and network diagrams (H); all are used to present TCR data. Such graphs offer information about the frequency and evolution of T-cell clonotypes in different patients, in different tissue types, and through different disease or therapeutic conditions.

Close modal

Research groups like our own have developed more advanced approaches to characterize the T-cell repertoire. A limitation in the calculations of standard diversity or clonality metrics is that a group of highly similar TCR sequences within a given sample has the same weight as a group of dissimilar TCR sequences. Numeric embedding of TCR sequences allows for the fine-grained capture of TCR similarities, accounting for a continuous scale of TCR similarity when characterizing the T-cell repertoire (31, 35). In addition, computational tools like GLIPH, iSMART, and TCRdist that cluster TCRs based on sequence similarity can help researchers understand the biological relevance of TCR sequences rather than counting total TCR clonotypes (36–39). Additional analysis pipelines, such as Immunarch and Scirpy, facilitate large-scale analysis and visualization of TCR sequencing data (40, 41). Gene usage analysis reports allelic frequency of V, D, and J alleles and can be used to highlight intrinsic allelic bias within samples as well as extrinsic bias in response to disease (42, 43). Clone tracking and differential abundance analyses quickly identify expanded and contracted T-cell populations across longitudinal samples and enable researchers to both monitor and predict immunologic responses to disease and therapy (44–46).

TCR and antigen databases

Because of the expanded mapping of TCR repertoires through HTS, several groups have curated databases to provide a centralized location for TCR-related information (Table 2). ImMunoGeneTics (IMGT) is an information system that was created in 1989 to compile and standardize immunogenetic data. As the first and largest branch of IMGT, IMGT/LIGM-DB offers a collection of known antibody and TCR sequences from humans and other vertebrate species (47, 48). With regards to T-cell antigens, the Immune Epitope Database (IEDB) contains a collection of experimentally isolated antigens from a variety of contexts, including infectious agents, allergens, cancer, and self-antigens (49). The Adaptive Immune Receptor Repertoire (AIRR) Community established the AIRR Data Commons (ADC), an open-source repository for immune receptor sequencing data from individual external databases such as iRepertoire and VDJ Server (50, 51). With over 5.4 billion sequence annotations, the ADC represents the largest database of immune cell receptor sequences and an incredible resource for researchers (51). McPAS-TCR assembles TCR sequences associated with specific pathologies, like infections, cancer, and autoimmune diseases, as well as their corresponding antigens (52). VDJdb is a database that uniquely links TCR sequences with their pMHC ligands, contributing to a more comprehensive characterization of TCR interactions. In contrast to McPAS-TCR which solely focuses on pathologic conditions, VDJdb does not limit the biological contexts in which the collected TCR-pMHC pairs occur (53). In addition, 10X Genomics offers a dataset of over 15,000 distinct paired TCR sequences with specificity for at least one pMHC within the panel used in their single-cell immune profiling platform. Curated by the same group that created VDJdb, TCRdb leverages datasets from 10X Genomics single-cell immune profiling technology and contains more than 270 million TCR sequences from across clinical conditions, tissue, and cell types. The large-scale nature of TCRdb enabled by a new pipeline contributes to its novelty, as previous databases were manually curated and only contained around 100,000 TCR sequences (54). Such databases serve as the foundation for many computational tools that analyze important aspects of the TCR repertoire and immune response.

Table 2.

TCR and antigen databases.

DatabaseInformation typeContextSource organismDatabase sizeReferences
IMGT/LIGM-DB 
  • – Antibody sequences

  • – TCR sequences

 
All Human and other vertebrates 246,976 sequences (48
IEDB 
  • – Antibody sequences

  • – T-cell epitope sequences

  • – MHC alleles and ligand sequences

  • – Epitope analysis and prediction

 
Infectious disease, allergy, cancer, autoimmunity and transplantation Human, non-human primates, and other animal species >6,000,000 sequences (49
AIRR Database Commons 
  • – TCR and antibody/BCR sequences from iRepertoire, VDJServer, and AIRR COVID-19 independent repositories

 
All Human and mouse 5.4 billion sequences (51
McPAS-TCR 
  • – TCR sequences

  • – T-cell epitope sequences

 
Infectious disease, allergy, cancer, autoimmunity, and transplantation Human and mouse >5,000 sequences (52
VDJdb 
  • – TCR sequences with known antigen specificity

 
All Human, non-human primates, and mouse 61,049 sequences (53
10X Genomics Dataset 
  • – paired TCR sequences with known antigen specificity

 
All Human >15,000 sequences (10X Genomics Application Note) 
TCRdb 
  • – TCRβ chain sequences with known antigen specificity

 
Associated with specific tissue, clinical condition, cell type Human >277,000 sequences (54
DatabaseInformation typeContextSource organismDatabase sizeReferences
IMGT/LIGM-DB 
  • – Antibody sequences

  • – TCR sequences

 
All Human and other vertebrates 246,976 sequences (48
IEDB 
  • – Antibody sequences

  • – T-cell epitope sequences

  • – MHC alleles and ligand sequences

  • – Epitope analysis and prediction

 
Infectious disease, allergy, cancer, autoimmunity and transplantation Human, non-human primates, and other animal species >6,000,000 sequences (49
AIRR Database Commons 
  • – TCR and antibody/BCR sequences from iRepertoire, VDJServer, and AIRR COVID-19 independent repositories

 
All Human and mouse 5.4 billion sequences (51
McPAS-TCR 
  • – TCR sequences

  • – T-cell epitope sequences

 
Infectious disease, allergy, cancer, autoimmunity, and transplantation Human and mouse >5,000 sequences (52
VDJdb 
  • – TCR sequences with known antigen specificity

 
All Human, non-human primates, and mouse 61,049 sequences (53
10X Genomics Dataset 
  • – paired TCR sequences with known antigen specificity

 
All Human >15,000 sequences (10X Genomics Application Note) 
TCRdb 
  • – TCRβ chain sequences with known antigen specificity

 
Associated with specific tissue, clinical condition, cell type Human >277,000 sequences (54

Note: Several databases containing different TCR-related information have been curated for research and clinical purposes. The type of TCR-related information, biological context, source organisms, and database size are listed.

Computational tools for predicting antigen specificity

To predict TCR antigen specificity, one must first identify antigens which can be presented to T cells via MHC molecules. As such, numerous tools have been developed to address this critical challenge. Polymorphisms in MHC molecules affect characteristics of the peptide-binding groove and restricts the types of epitopes which can be presented (12). MHC haplotypes are commonly used as inputs into MHC prediction tools, such as NetMHC and the IEDB T-cell epitope prediction tool, to predict the likelihood of known proteins being presented by MHC molecules (55–57). Using the unique amino acid sequence of the peptide-binding groove in MHC molecules and inherent anchor positions important for peptide binding, these prediction algorithms predict the binding affinity for peptides and likelihood of presentation (58, 59). However, such algorithms only predict the presentation of a given peptide sequence and cannot predict what peptides are likely generated by cells through proteasomal degradation (60). Proteasomal processing prediction algorithms, such as Proteasome Cleavage Prediction Server, SpliceMet, and those offered by IEDB, can be used in conjunction with MHC prediction tools to more faithfully predict antigen presentation by MHC molecules and to identify potential sets of personalized tumor antigens using somatic mutations as inputs (61–63). Ultimately, although these tools are unable to predict T-cell reactivity, they do predict the stability of the formed peptide-bound MHC (pMHC), which contributes to its half-life and likelihood of being seen by any given TCR.

In addition to facilitating the characterization of T-cell repertoires by specific metrics, TCR sequencing provides information for the determination of T-cell antigen specificity. Identifying specific antigens targeted by different TCRs holds value in regards to quantifying antigen specificity groups within an individual, predicting disease outcomes, and isolating reactive T cells for therapeutic purposes. There is particular interest in the field of cancer immunology to identify and target neoantigens, peptides that arise from somatic mutations unique to cancer cells (64). With the recent improvements in HTS, many groups including our own have developed machine learning–based and algorithm-based tools aimed at elucidating antigen specificity for a variety of purposes (Table 3).

Table 3.

Computational tools for predicting antigen specificity.

Computational toolInput Output Unique featuresReferences
GLIPH2  CDR3 region of TCRα and β chain  Clustering of TCR sequences into groups of shared specificity 
  • – Efficiently processes and clusters millions of TCR sequences

 
(37
iSMART CDR3 region of TCRβ chain  Clustering of TCR sequences into groups of shared specificity 
  • – Built on pan-cancer multi-omics analysis

  • – Utilizes TRUST algorithm to assemble hypervariable CDR3 regions

 
(38, 65
ERGO-II CDR3 region of TCRα and β chain  TCR-peptide binding prediction 
  • – Includes MHC typing and V and J gene segment alleles in prediction model

 
(67
TCRMatch  CDR3 region of TCRβ chain  TCR-peptide binding prediction  
  • – Can be used in conjunction with other tools

 
(68
NetTCR 2.0  Paired TCRα and β chain  TCR-peptide binding prediction 
  • – Accounts for different TCR sequence lengths

  • – Improved processing of paired TCRα and β chain 

 
(69
pMTnet CDR3 region of TCRβ chain  TCR-peptide binding prediction  
  • – Accurately predicts TCRs, epitopes, and HLAs previously not observed

  • – Utilizes transfer learning for related TCR and pMHC data lacking pairing information 

 
(35
IGoR TCRβ and BCR heavy chains V(D)J recombination and somatic hypermutation prediction 
  • – Performance in identifying the correct V(D)J recombination scenario is 2× better than current methods

  • – Can assess the mutational landscape of BCRs

 
(70
OLGA CDR3 sequence of TCR and BCR V(D)J recombination prediction 
  • – Computes the probabilities of CDR3 sequences and motifs of four chain loci (human and mouse TCRβ, human TCRα and human IGH)

  • – Can identify outlying sequences in repertoire sequencing datasets to study disease immune response

 
(74
SONIA CDR3 sequence of TCRβ chain  V(D)J recombination and selection pressure prediction 
  • – Distinguishes between generation and selection to quantify contribution to the overall variability

 
(79
Computational toolInput Output Unique featuresReferences
GLIPH2  CDR3 region of TCRα and β chain  Clustering of TCR sequences into groups of shared specificity 
  • – Efficiently processes and clusters millions of TCR sequences

 
(37
iSMART CDR3 region of TCRβ chain  Clustering of TCR sequences into groups of shared specificity 
  • – Built on pan-cancer multi-omics analysis

  • – Utilizes TRUST algorithm to assemble hypervariable CDR3 regions

 
(38, 65
ERGO-II CDR3 region of TCRα and β chain  TCR-peptide binding prediction 
  • – Includes MHC typing and V and J gene segment alleles in prediction model

 
(67
TCRMatch  CDR3 region of TCRβ chain  TCR-peptide binding prediction  
  • – Can be used in conjunction with other tools

 
(68
NetTCR 2.0  Paired TCRα and β chain  TCR-peptide binding prediction 
  • – Accounts for different TCR sequence lengths

  • – Improved processing of paired TCRα and β chain 

 
(69
pMTnet CDR3 region of TCRβ chain  TCR-peptide binding prediction  
  • – Accurately predicts TCRs, epitopes, and HLAs previously not observed

  • – Utilizes transfer learning for related TCR and pMHC data lacking pairing information 

 
(35
IGoR TCRβ and BCR heavy chains V(D)J recombination and somatic hypermutation prediction 
  • – Performance in identifying the correct V(D)J recombination scenario is 2× better than current methods

  • – Can assess the mutational landscape of BCRs

 
(70
OLGA CDR3 sequence of TCR and BCR V(D)J recombination prediction 
  • – Computes the probabilities of CDR3 sequences and motifs of four chain loci (human and mouse TCRβ, human TCRα and human IGH)

  • – Can identify outlying sequences in repertoire sequencing datasets to study disease immune response

 
(74
SONIA CDR3 sequence of TCRβ chain  V(D)J recombination and selection pressure prediction 
  • – Distinguishes between generation and selection to quantify contribution to the overall variability

 
(79

Note: Many tools have been developed to improve and streamline the identification of TCR-peptide pairs. Input, output, and unique features are listed.

In 2017, Glanville and colleagues developed GLIPH (grouping of lymphocyte interactions by paratope hotspots) to automatically cluster TCR sequences into different groups of shared specificity. GLIPH integrates global and local TCR sequence similarity of CDR3 sequences to identify sets of TCRs that potentially recognize the same pMHC ligands. Clustering by global similarity refers to grouping α and β CDR3 sequences differing by up to one amino acid and clustering by local similarity refers to grouping sequences with shared enriched CDR3 motifs (36). More recently, the same group introduced GLIPH2, which efficiently processes and clusters millions of TCR sequences; this serves as an improvement to the original algorithm which lost accuracy when greater than 10,000 TCR sequences were inputted (37). Yet, both versions of GLIPH require TCR information from established databases and thus cannot discern new epitopes or TCR-pMHC pairs.

Zhang and colleagues also developed a method in 2020 to cluster TCRs based on antigen specificity, named iSMART (immuno-Similarity Measurement by Aligning Receptors of T cells). The researchers coupled iSMART with their TRUST algorithm (Tcr repertoire utilities for solid tissue) that detects CDR3 hypervariable regions from bulk RNA-sequencing data (65). Within the study, TRUST and iSMART were utilized to profile tumor-associated T cells, using multi-omic data from The Cancer Genome Atlas. Ultimately, iSMART aided in the identification of novel cancer neoantigens (38).

While GLIPH and iSMART indirectly facilitate antigen discovery through clustering TCR sequences based on shared specificity, other algorithms aim to directly predict TCR and antigen pairs. Springer and colleagues developed a deep learning–based method termed ERGO (pEptide tcR matchinG predictiOn) to predict TCR-peptide binding using existing large-scale TCR-peptide datasets as reference. ERGO is based on long short-term memory networks and solely considers the β-chain CDR3 sequence in determining whether a specific TCR will bind to different peptides (66). Because antigen specificity is not solely governed by the β-chain CDR3, ERGO-II was developed to include the α-chain CDR3 sequence, MHC typing, and V and J gene segments (67).

Similar to ERGO, TCRMatch aims to identify the specific antigens targeted by different TCRs. Using the β-chain CDR3 sequence as an input, TCRMatch employs a comprehensive k-mer matching algorithm to both identify TCRs with matches in IEDB and provide the specificity of the matches. The use of IEDB eliminates the need to perform preliminary experiments to train algorithms and streamlines the overall process; however, the range of antigen identification is limited to the dataset availabilities. To offset this limitation, TCRMatch can be used in conjunction with other tools like GLIPH2 (68).

NetTCR, designed by Jurtz and colleagues, predicted the interactions between TCRs and peptides specifically presented by HLA-A*02:01, a common human MHC class I allele. Like TCRMatch and the first version of ERGO, NetTCR used the β-chain CDR3 sequence; however, it was based on convolutional neural networks (CNN), which are uniquely suited to reconcile unaligned peptides and TCR sequences of different lengths (bioRxiv 433706). NetTCR-2.0 retains the advantage of accounting for differing TCR sequence lengths while gaining the improved processing ability for paired TCRαβ sequence data. In this study, the authors demonstrated that models using α-chain CDR3 or β-chain CDR3 data alone showed variability in results and decreased performance (69).

We developed pMHC–TCR binding prediction network (pMTnet) to predict specific TCR binding interactions with MHC class I-bound T-cell neoantigens. pMTnet uses the β-chain CDR3 sequence as an input and relies on a novel branch of deep learning called “transfer learning,” which utilizes related TCR and pMHC data that lack pairing information, as well as a training program that allows for the differentiation between binding versus nonbinding TCRs. pMTnet achieved an AUROC (area under the ROC curve) of greater than 0.83 on multiple cohorts. This accuracy extends to TCRs, epitopes, and HLAs that had not been observed before. Using pMTnet as a knowledge discovery tool, we validated the high immunogenicity of human endogenous retrovirus E (HERV-E) in kidney cancers and showed HERV-E is more immunogenic than most tumor neoantigens. With pMTnet, it is possible to develop prognostic and predictive tools for patients with cancer, using TCR and tumor antigen data (35).

Moreover, several computational tools have been developed to help understand V(D)J recombination and subsequent selection processes. IGoR (Inference and Generation of Repertoires) is a software tool that takes immune sequence reads of cDNA or gDNA as input and then quantifies the statistics of receptor generation (70). IGoR assumes that random choice of germline segments and indels contribute to the recombination of receptor sequences and that there is a constant error probability throughout the sequence when dealing with the TCR. IGoR can be used to process V(D)J recombination–related tasks, such as inferring the recombination statistics, analyzing specific recombination scenarios and their likelihoods, and generating synthetic sequences (70). Recently, IGoR has contributed significantly to T-cell research, facilitating the distinction of T-cell lineages, comparing the efficacy of TCR profiling methods, and characterizing diversity of TCR repertoires (71–73).

OLGA (Optimized Likelihood estimate of immunoGlobulin Amino-acid sequences) is a computational tool developed to estimate the probability of generating TCR and B-cell receptor (BCR) sequences or motifs with dynamic programming (74). OLGA can be directly applied to human and mouse TCRβ loci, as well as human TCRα and IGH loci. Other species and loci can also be analyzed by learning recombination models using IGoR. Studies in many fields have benefited from OLGA, including vaccine development and computational immunology (75–78).

During development, T cells undergo the selection process based on their binding properties; Sethna and colleagues developed a computational method, called SONIA, to quantify the selection pressure of CDR3 amino acid sequence (79). SONIA outputs the selection pressure factors for features of the input TCR CDR3β sequence. These selection factors can be used to indicate how likely a sequence is to pass the selection process. The postselection probability of this sequence can be calculated using this selection factor.

Numerous other methods have also been developed to tackle the issue of TCR antigen-binding prediction, including DeepTCR, TCRGP, and TITAN (Tcr epITope bimodal Attention Networks). DeepTCR uses CNNs to parse through TCR sequencing data; it integrates α-chain CDR3 or β-chain CDR3 sequences with V/D/J gene segment usage to classify and cluster antigen-specific TCRs (80). TCRGP is a computational tool using Gaussian process, which has the ability to generate a robust prediction from small datasets, to predict the binding of TCR and epitopes (81). TITAN is a bimodal neural network method developed to predict the binding of TCR and epitope. It uses k-nearest neighbor algorithm as the baseline and encodes both TCR and epitope sequences for the generalization purpose (82).

Overall, these algorithm-based tools for analyzing TCR sequencing data aim to help researchers better understand antigen specificity, offering a wealth of information regarding the complexity of the immune response. In their respective manners, these tools have all contributed to developing new ways to characterize, monitor, and treat a variety of human diseases, including infectious disease, autoimmune diseases, and cancer.

Because of TCR-antigen promiscuity, functional assays must be performed to validate TCR-pMHC pairing predictions made by algorithm-based TCR sequencing analysis tools (83). Common experimental techniques for such validation include ELISA, enzyme-linked immune absorbent spot (ELIspot), multimer staining, and activation marker expression. Both ELISA and ELIspot assess T-cell activation through cytokine release, which occurs when T cells bind to their cognate pMHC and is routinely used as an indication of activated T cells. ELISA assesses the total concentration of cytokines released, while ELIspot detects individual cytokine-secreting clones (84, 85). On the other hand, multimer sorting is used to assess the binding of a potential TCR-pMHC pair. Multimers, like tetramers and dextramers, can be engineered to present target pMHC for target T cells. Because multimers are labeled with fluorophores, multimer stains allow for the quantification and isolation of T cells by flow cytometry (86).

Beyond these commonly used techniques, the upregulation of both stimulatory and inhibitory proteins during T-cell activation can be monitored for functional validation. 4-1BB, OX40, and CD103 have been found to enhance T-cell expansion and survival while PD-1, TOX, and CD39 have been shown to dampen T-cell responses; all of these markers have been found to be enriched on antigen-specific T cells in different contexts (87–92). Yet, the utility of these markers is limited, as their discovery stems from evaluating only a small minority of antigen-specific T-cell responses and they fail to distinguish between antitumor and bystander T cells. Several novel functional assays were developed to address such shortcomings while also enabling the processing of more TCRs and antigens. Yeast antigen display libraries, which involve the expression of human MHC on the surface of yeast, facilitate the rapid identification of peptide ligands and T-cell recognition events (93). Another assay was developed to track T-cell trogocytosis, a phenomenon where T cells share their lipid membrane and membrane-associated proteins while conjugated with their target APCs (94). Signaling and antigen-presenting bifunctional receptors on APCs have been developed to induce TCR-like signaling after binding of TCR with its cognate antigen (95). Finally, tandem minigenes offer the ability to assess several potential TCR-antigen pairs by designing multiple antigen sequences into one construct (96).

While the previously described assays provide valuable information, they are ultimately either limited in their sensitivity and scope or biased by the focus on a specific TCR. These limitations make assessing the repertoire of functional tumor-associated T cells particularly difficult. As a result, Danilova and colleagues developed MANAFEST (Mutation-Associated NeoAntigen Functional Expansion of Specific T cells), which uses TCR sequencing of peptide-stimulated cultures and a bioinformatics platform to both analyze antigen-specific clonal expansion and characterize the expanded TCR clonotypes in a high-throughput and sensitive manner. This assay can drive the development of personalized cancer immunotherapies, as well as the identification of treatment response biomarkers (97). Importantly, adaptations of MANAFEST can be used to detect T-cell responses to specific viral, bacterial, and self-antigens. The same group developed ViraFEST, which has the ability to characterize T-cell responses against different viral antigens including human immunodeficiency virus epitopes (98).

With major advances in TCR sequencing, T-cell antigen specificity prediction, and functional validation of T-cell antigen specificity, there is motivation to develop strategies to streamline the whole process. In 2015, Klinger and colleagues developed MIRA (Multiplexed Identification of T-cell Receptor Antigen specificity), which combines deep TCR sequencing and several immune assays to simultaneously identify and validate large numbers of antigen-specific T cells (99). Separately, Zhang and colleagues presented tetramer-associated T-cell receptor sequencing (TetTCR-seq) as a means to connect TCRs to their specific antigen, in which functional TCR-pMHC binding is determined using a library of DNA-barcoded antigen tetramers (100). TetTCR-seqHD combines TetTCR-seq with the new ability to characterize gene and protein expression to offer a more informed characterization of T-cell activation. These methods are effective in profiling large amounts of single T cells with precision, while also uniquely identifying T-cell cross-reactivity (101). In addition, Arnaud and colleagues developed NeoScreen as a pipeline for the sensitive identification of rare tumor neoantigens and cognate TCRs expressed by tumor-infiltrating lymphocytes (TIL). This method is based on the early exposure of TILs isolated from tumors to antigens of choice which are loaded onto CD40-activated B cells; TCR and neoantigen predictions are then validated against autologous tumors (102).

Characterization of tumor properties

Advances in TCR sequencing and TCR repertoire analysis have significantly aided in the characterization of tumors (Fig. 4). Notably, TCR sequencing has promoted the description of immune tumor heterogeneity, which also relates to and cooperates with the genomic heterogeneity of tumor cells in tumorigenesis. Performing TCR sequencing for non–small cell lung cancer (NSCLC), our group has demonstrated significant intratumor differences in T-cell density and clonality between distinct tumor regions and correlation between genomic and immune intratumor heterogeneity, indicating that TCR intratumor heterogeneity is potentially driven by specific antigens in different tumor regions (46). More recently, Joshi and colleagues also characterized the heterogeneity of the NSCLC TCR repertoire and provided more evidence that TCR intratumor heterogeneity reflects the mutational landscape across different tumor regions (103). Significant TCR intratumor heterogeneity was also observed in NSCLC metastases, as well as other cancers like melanoma and renal cell carcinoma (104–106). In the context of limited-stage small cell lung cancer (LS-SCLC), whole-exome and TCR sequencing revealed a homogeneous mutational landscape along with a heterogeneous TCR repertoire; this sheds light on the unique nature of LS-SCLC as having high copy-number alteration levels and a particularly immunosuppressive tumor microenvironment (107). In addition to characterizing intratumor heterogeneity, TCR sequencing can be utilized to observe intertumor heterogeneity. Specifically, our group utilized TCR sequencing to observe heterogeneous T-cell profiles across synchronous metastases from lung cancer and melanoma that correspond to heterogeneous mutational and methylation landscapes (104, 105). Through these studies, TCR sequencing of multiple synchronous tumors enables the assessment of differential tumor growth in treatment-naïve patients, in addition to lesion-specific therapeutic responses to targeted therapy and immune checkpoint blockade between metastases within the same patient (105). Overall, TCR intratumor and intertumor heterogeneity have important implications in elucidating mechanisms of cancer immunity and predicting therapeutic responses to immunotherapy.

Figure 4.

Applications of TCR sequencing and T-cell repertoire analysis. TCR sequencing is a valuable tool with fundamental applications to cancer biology and translational implications with cancer immunotherapy. Its utility extends to other contexts as well, including organ transplantation and pathogen immunity.

Figure 4.

Applications of TCR sequencing and T-cell repertoire analysis. TCR sequencing is a valuable tool with fundamental applications to cancer biology and translational implications with cancer immunotherapy. Its utility extends to other contexts as well, including organ transplantation and pathogen immunity.

Close modal

In addition to informing on heterogeneity, TCR sequencing offers a way to track tumor evolution through immune monitoring. Combining gene expression profiling and TCR sequencing, Dejima and colleagues showed that T cell–mediated antitumor immunity decreases as lung cancers progress from preneoplastic lesions to invasive lung adenocarcinomas. Specifically, as lung cancers become more malignant, immune-activation pathways are downregulated while immunosuppressive pathways are upregulated and T-cell clonality and T-cell tumor infiltration decreases (108). Cui and colleagues reported that the diversity of the TCR repertoire in the peripheral blood gradually decreased during tumor progression from cervical intraepithelial neoplasia to established cervical cancer and that specific clonotypes with similar CDR3 motifs were present between patients (109). The identification of immunologic trends during tumor evolution can help in the classification of tumor grades and aid in the optimization of immunotherapies based on cancer stage.

On the basis of recent observations that cancer-associated TCRs share similar biochemical profiles which distinguish them from TCRs in normal tissues, Beshnova and colleagues developed a machine learning method called DeepCAT (Deep CNN Model for Cancer-Associated TCRs) to predict whether cancer-associated TCRs are present in a patient's peripheral blood. DeepCAT demonstrated high prediction accuracy when distinguishing between patients with cancer and healthy individuals, with an AUC greater 0.95 for several types of early-stage cancers, including breast cancer, ovarian cancer, and melanoma. Overall, DeepCAT represents a potential method for noninvasive, early cancer detection (110).

Development and characterization of immunotherapies

TCR sequencing is necessary to improve upon current T cell–based therapies. Adoptive T-cell transfer therapy previously relied on the autologous infusion of expanded tumor-infiltrating T cells; however, many patients do not respond to this therapy due to the low frequencies of neoantigen-specific T cells within the tumor-infiltrating group (111). Advances in T-cell therapies including genetically modified T cells expressing novel TCRs have been developed to elicit tumor-specific immune responses (112). TCR sequencing plays an important role in increasing the efficacy of T-cell therapies through the identification of optimal target antigens and tumor antigen-specific TCRs for these novel T-cell therapies (111, 113). Isolated neoantigen-specific T cells can undergo either bulk or single-cell TCR sequencing to identify dominant TCR clonotypes within a tumor (111).

In addition to driving the specificity of T-cell therapies, TCR sequencing can also be used as a tool to evaluate and track T-cell clonotypes over time after administration of immunotherapies. For example, deep sequencing on the variable region of the TCRβ chain was used to track the in vivo frequency of TCRs reactive against mutant KRAS G12D in a patient with metastatic colorectal cancer who experienced tumor regression after adoptive cellular therapy. Interestingly, the most dominant clonotype upon infusion was not detected after some time while less dominant clonotypes persisted (114). Furthermore, sequencing of the TCRβ chain revealed that clonal diversity of chimeric antigen receptor (CAR)-T cells was highest in the blood of patients with acute lymphoblastic leukemia and non–Hodgkin lymphoma during infusion and declined over time; it was also found that individual T-cell clonotypes exhibited distinct clonal kinetics and had variable impacts on the CAR-T cell pool after infusion (115).

Analysis of the TCR repertoire also allows researchers to elucidate underlying mechanisms of action for immunotherapy, as well as its effects on immune cells. Sipuleucel-T is an autologous cellular therapy and a type of cancer vaccine where APCs are activated against prostatic acid phosphatase antigen, which is widely expressed on prostate cancers (116). Sheik and colleagues employed TCR sequencing to characterize the mechanism of action for sipuleucel-T. They showed that the treatment facilitated the recruitment of T cells into the prostate rather than promoting expansion of T-cell clones, as diversity of circulated T cells decreased while diversity of tumor-infiltrating T cells increased (117). In another study, Wieland and colleagues utilized TCR sequencing to examine the composition of the peripheral blood and tumor-infiltrating CD8+ T cells that were activated upon anti–PD-1 therapy in a patient with melanoma. They found an oligoclonal repertoire of CD8+ T cells in peripheral blood and showed that the majority of dominant peripheral blood clones did not expand following treatment, while expanding clones were found within the resected tumor (118).

Prediction of patient outcomes, treatment response, and toxicities

Aside from informing the development of immunotherapies, TCR sequencing and derived TCR repertoire metrics have utility in predicting patient outcomes. TCR diversity represents a prognostic biomarker in patients with melanoma, in which patients with greater T-cell evenness and richness in their peripheral blood, as well as in lymph node metastases, had longer progression-free and overall survival (119). Increased diversity in the baseline TCR repertoire of tumor-infiltrating T cells was found to be prognostic in various cancers, including breast cancer, melanoma, lung cancer, and kidney cancer (120). In a study focused on high-grade serous ovarian carcinoma, high T-cell clonality combined with either genomic instability signatures, which includes homologous recombination deficiency and copy-number variation, or T-cell infiltration into the tumor resulted in high prognostic value (121). Furthermore, patients with expanding T cells exhibiting higher affinity to truncal neoantigens, which are derived from mutations with higher variant allele frequency, were more likely to demonstrate better prognosis and response to immunotherapies in lung cancer and melanoma (35).

The TCR repertoire can also be used to predict patient responses to different cancer immunotherapies. Both increased richness and evenness of TCR diversity in the peripheral blood of patients with advanced melanoma prior to treatment with ipilimumab, an inhibitor of CTLA-4, corresponded to greater clinical benefit upon treatment (122). From a study on advanced NSCLC, patients who exhibited higher TCR richness in their peripheral blood during treatment had the greatest clinical benefit from anti-PD-1 immunotherapy, as well as prolonged progression-free and overall survival. In the same study, patients with greater Jaccard similarity index values between pretreatment and various treatment timepoints showed improved progression-free survival, highlighting the importance of immune monitoring during treatment (123). Both increased diversity of the TCR repertoire at baseline and increased clonality during treatment related to significant responses to nivolumab, an inhibitor of PD-1, in patients with classical Hodgkin lymphoma, as well as to autologous Epstein–Barr virus (EBV)-expanded CTLs immunotherapy in patients with EBV-positive nasopharyngeal carcinoma (124, 125). In addition, greater TCR clonality of pretreatment tumor-infiltrating T cells was predictive for the efficacy of anti-PD-1 immunotherapy in patients with metastatic melanoma (120).

Although cancer immunotherapies have promoted significant antitumor immune responses, they also have the potential to induce inflammation-related toxicities. In addition to serving as prognostic and predictive biomarkers for patient outcome and immunotherapy responses, TCR repertoire metrics can also offer information on potential immunotherapy-related toxicities. Clonality was assessed in the context of immune-related adverse events (irAE) after ipilimumab anti-CTLA-4 treatment in patients with prostate cancer; the expansion of greater than 55 CD8+ T-cell clones in the peripheral blood preceded the development of severe irAEs (126). A separate study associated CD4+ memory T-cell density and diversity in the peripheral blood of patients with melanoma with the development of severe irAE upon anti-PD-1 monotherapy and anti-PD-1 and anti-CTLA-4 combination therapy (127). TCR repertoire metrics can correlate with both positive and negative clinical outcomes, and it is important to leverage the knowledge gained from TCR sequencing to develop treatments and strategies that promote favorable outcomes while minimizing potential toxicities (Fig. 4).

The utility of TCR sequencing extends beyond applications to cancer. Because T cells and their TCRs play an integral role in the mammalian allogeneic response by recognizing and eliminating incompatible tissues, researchers utilize TCR sequencing to characterize responses to organ transplants (128). Specifically, the TCR repertoire can offer information on the risk of rejection through T cell–mediated rejection and graft-versus-host disease and has been evaluated in the context of kidney, liver, and hematopoietic stem cell transplantations (129–132). In addition to transplants, TCR sequencing has potential use in the biological characterization, disease stratification, and monitoring of autoimmune diseases. For example, from analyzing TCR repertoires, researchers have gained insight into the spatial T-cell dynamics in multiple sclerosis, identified TCR clonotypes specific to celiac disease, and defined both clonotype expansion and TCR chain pairing in type I diabetes (133–135). T cells are also instrumental in the immune response to pathogens. As a result, TCR sequencing is valuable for studying the dynamics of adaptive immunity, surveilling the progress of infection, and developing novel immunotherapies for a number of different viral infections, including SARS-CoV-2 (136, 137). In other health conditions, like ischemic heart disease and acute coronary syndrome, TCR repertoire analysis can be performed for the purpose of risk stratification and diagnosis (138, 139). Overall, TCR sequencing and analysis of the TCR repertoire represents a significant tool in an array of biological contexts beyond the scope of this review (Fig. 4).

Recent technological advances have expanded the scope of possibilities pertaining to analysis of the T-cell repertoire, streamlining the identification and development of novel biomarkers and therapeutics. Single-cell transcriptomics equipped with feature barcoding has been implemented in conjunction with TCR sequencing to obtain paired TCRαβ chain sequences while also providing information about cell surface marker expression and antigen specificity, allowing scientists to effectively link antigen specificity and TCR sequence to phenotype for the first time (29, 140). Importantly, interactions between T cells and their surrounding cells have the ability to influence the function and transcriptome of T cells and require the implementation of spatial transcriptomic technologies like 10X Genomics Visium Spatial Gene Expression platform and Advanced Cell Diagnostic's RNAscope assay, which allows the design of custom probes which could be used to identify specific CDR3β sequences and T-cell clonotypes in tissues (141–143). These methodologies can also be utilized to assess the localization and tumor infiltration of antigen-specific T cells and clonotypes of CAR and TCR engineered T cells. With an increasing number of studies now evaluating the importance of cell distribution, distances between cell types and their impact on clinical outcomes, these advances are poised to contribute to the next generation of discoveries in the TCR sequencing landscape (144, 145). Moreover, use of novel protein folding prediction tools such as Google's DeepMind and AlphaFold are likely to enhance our ability to understand pMHC and TCR interactions going forward and help leverage this knowledge to improve upon immunotherapies and therapeutic responses (146).

TCR sequencing is an invaluable tool for the characterization of a wide range of immune responses, informing on the nature of TCR repertoires and aiding in the identification of TCR-antigen pairs. With specific regards to cancer, TCR sequencing can contribute to the basic characterization of tumor heterogeneity and tumor evolution. From a clinical standpoint, TCR sequencing serves both as the foundation of T cell–based immunotherapies and a tool to assess the efficacy of various types of immunotherapies. Furthermore, TCR repertoire metrics derived from sequencing data analysis can be significant predictors of patient outcomes, anticancer treatment responses, and the risk of immune-related toxicities in many different cancer types. NGS methodologies and TCR data analysis tools are continually improving, exhibiting greater processing capabilities and expanding research and clinical applications.

J.V. Heymach reports other support from AstraZeneca, EMD Serono, Boehringer-Ingelheim, Catalyst, Genetech, GlaxoSmithKline, Hengrui Therapeutics, Eli Lilly, Spectrum, Sanofi, Takeda, Mirati Therapeutics, Bristol Myers Squibb, BrightPath Biotherapeutics, Janssen Global Services, Pneuma Respiratory, RefleXion, and Chugai Pharmaceuticals outside the submitted work. J. Zhang reports personal fees from AstraZeneca, Geneplus, Hengrui, Innovent, Merck, and Roche; grants and personal fees from Johnson and Johnson and Novartis; and grants from OrigMed outside the submitted work. A. Reuben reports personal fees from Adaptive Biotechnologies outside the submitted work. No disclosures were reported by the other authors.

A. Reuben was supported by the Exon 20 Group, Rexanna's Foundation for Fighting Lung Cancer, the Waun Ki Hong Lung Cancer Research Fund, MD Anderson's Lung Cancer Moon Shot, the Petrin Fund, the University Cancer Foundation via the Institutional Research Grant program at the University of Texas MD Anderson Cancer Center, the Happy Lungs Project, RETpositive/LUNGevity (FP00015320), and the Cancer Prevention & Research Institute of Texas (RP210137).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1.
Chaplin
DD
.
Overview of the immune response
.
J Allergy Clin Immunol
2010
;
125
:
S3
23
.
2.
Bonilla
FA
,
Oettgen
HC
.
Adaptive immunity
.
J Allergy Clin Immunol
2010
;
125
:
S33
40
.
3.
De Simone
M
,
Rossetti
G
,
Pagani
M
.
Single cell T cell receptor sequencing: techniques and future challenges
.
Front Immunol
2018
;
9
:
1638
.
4.
Xiong
Y
,
Bosselut
R
.
CD4-CD8 differentiation in the thymus: connecting circuits and building memories
.
Curr Opin Immunol
2012
;
24
:
139
45
.
5.
Lo Presti
E
,
Dieli
F
,
Meraviglia
S
.
Tumor-infiltrating gammadelta T lymphocytes: pathogenic role, clinical significance, and differential programing in the tumor microenvironment
.
Front Immunol
2014
;
5
:
607
.
6.
Schatz
DG
,
Ji
Y
.
Recombination centres and the orchestration of V(D)J recombination
.
Nat Rev Immunol
2011
;
11
:
251
63
.
7.
Roth
DB
.
V(D)J recombination: mechanism, errors, and fidelity
.
Microbiol Spectr
2014
;
2
.
8.
Rosati
E
,
Dowds
CM
,
Liaskou
E
,
Henriksen
EKK
,
Karlsen
TH
,
Franke
A
.
Overview of methodologies for T-cell receptor repertoire analysis
.
BMC Biotechnol
2017
;
17
:
61
.
9.
Krangel
MS
.
Mechanics of T cell receptor gene rearrangement
.
Curr Opin Immunol
2009
;
21
:
133
9
.
10.
Rossjohn
J
,
Gras
S
,
Miles
JJ
,
Turner
SJ
,
Godfrey
DI
,
McCluskey
J
.
T cell antigen receptor recognition of antigen-presenting molecules
.
Annu Rev Immunol
2015
;
33
:
169
200
.
11.
Wieczorek
M
,
Abualrous
ET
,
Sticht
J
,
Alvaro-Benito
M
,
Stolzenberg
S
,
Noe
F
, et al
.
Major histocompatibility complex (MHC) class I and MHC class II proteins: conformational plasticity in antigen presentation
.
Front Immunol
2017
;
8
:
292
.
12.
Neefjes
J
,
Jongsma
ML
,
Paul
P
,
Bakke
O
.
Towards a systems understanding of MHC class I and MHC class II antigen presentation
.
Nat Rev Immunol
2011
;
11
:
823
36
.
13.
Rock
KL
,
Reits
E
,
Neefjes
J
.
Present yourself! By MHC class I and MHC class II molecules
.
Trends Immunol
2016
;
37
:
724
37
.
14.
Matzinger
P
,
Bevan
MJ
.
Hypothesis: why do so many lymphocytes respond to major histocompatibility antigens?
Cell Immunol
1977
;
29
:
1
5
.
15.
Mason
D
.
A very high level of crossreactivity is an essential feature of the T-cell receptor
.
Immunol Today
1998
;
19
:
395
404
.
16.
Wooldridge
L
,
Ekeruche-Makinde
J
,
van den Berg
HA
,
Skowera
A
,
Miles
JJ
,
Tan
MP
, et al
.
A single autoimmune T cell receptor recognizes more than a million different peptides
.
J Biol Chem
2012
;
287
:
1168
77
.
17.
Behjati
S
,
Tarpey
PS
.
What is next generation sequencing?
Arch Dis Child Educ Pract Ed
2013
;
98
:
236
8
.
18.
Reuter
JA
,
Spacek
DV
,
Snyder
MP
.
High-throughput sequencing technologies
.
Mol Cell
2015
;
58
:
586
97
.
19.
Metzker
ML
.
Sequencing technologies - the next generation
.
Nat Rev Genet
2010
;
11
:
31
46
.
20.
Hess
JF
,
Kohl
TA
,
Kotrova
M
,
Ronsch
K
,
Paprotka
T
,
Mohr
V
, et al
.
Library preparation for next generation sequencing: a review of automation strategies
.
Biotechnol Adv
2020
;
41
:
107537
.
21.
Singh
RR
.
Target enrichment approaches for next-generation sequencing applications in oncology
.
Diagnostics
2022
;
12
:
1539
.
22.
Wang
Q
,
Zeng
H
,
Zhu
Y
,
Wang
M
,
Zhang
Y
,
Yang
X
, et al
.
Dual UMIs and dual barcodes with minimal PCR amplification removes artifacts and acquires accurate antibody repertoire
.
Front Immunol
2021
;
12
:
778298
.
23.
Lin
YH
,
Hung
SJ
,
Chen
YL
,
Lin
CH
,
Kung
TF
,
Yeh
YC
, et al
.
Dissecting efficiency of a 5' rapid amplification of cDNA ends (5'-RACE) approach for profiling T-cell receptor beta repertoire
.
PLoS One
2020
;
15
:
e0236366
.
24.
Pinto
FL
,
Lindblad
P
.
A guide for in-house design of template-switch-based 5' rapid amplification of cDNA ends systems
.
Anal Biochem
2010
;
397
:
227
32
.
25.
Mamedov
IZ
,
Britanova
OV
,
Zvyagin
IV
,
Turchaninova
MA
,
Bolotin
DA
,
Putintseva
EV
, et al
.
Preparing unbiased T-cell receptor and antibody cDNA libraries for the deep next generation sequencing profiling
.
Front Immunol
2013
;
4
:
456
.
26.
Adamopoulos
PG
,
Tsiakanikas
P
,
Stolidi
I
,
Scorilas
A
.
A versatile 5' RACE-Seq methodology for the accurate identification of the 5' termini of mRNAs
.
BMC Genomics
2022
;
23
:
163
.
27.
Bergman
Y
.
Allelic exclusion in B and T lymphopoiesis
.
Semin Immunol
1999
;
11
:
319
28
.
28.
Pai
JA
,
Satpathy
AT
.
High-throughput and single-cell T cell receptor sequencing technologies
.
Nat Methods
2021
;
18
:
881
92
.
29.
Zheng
GX
,
Terry
JM
,
Belgrader
P
,
Ryvkin
P
,
Bent
ZW
,
Wilson
R
, et al
.
Massively parallel digital transcriptional profiling of single cells
.
Nat Commun
2017
;
8
:
14049
.
30.
Howie
B
,
Sherwood
AM
,
Berkebile
AD
,
Berka
J
,
Emerson
RO
,
Williamson
DW
, et al
.
High-throughput pairing of T cell receptor alpha and beta sequences
.
Sci Transl Med
2015
;
7
:
301ra131
.
31.
Zhang
Z
,
Xiong
D
,
Wang
X
,
Liu
H
,
Wang
T
.
Mapping the functional landscape of T cell receptor repertoires by single-T cell transcriptomics
.
Nat Methods
2021
;
18
:
92
9
.
32.
Schattgen
SA
,
Guion
K
,
Crawford
JC
,
Souquette
A
,
Barrio
AM
,
Stubbington
MJT
, et al
.
Integrating T cell receptor sequences and transcriptional profiles by clonotype neighbor graph analysis (CoNGA)
.
Nat Biotechnol
2022
;
40
:
54
63
.
33.
Reuben
A
,
Zhang
J
,
Chiou
SH
,
Gittelman
RM
,
Li
J
,
Lee
WC
, et al
.
Comprehensive T cell repertoire characterization of non-small cell lung cancer
.
Nat Commun
2020
;
11
:
603
.
34.
Chiffelle
J
,
Genolet
R
,
Perez
MA
,
Coukos
G
,
Zoete
V
,
Harari
A
.
T-cell repertoire analysis and metrics of diversity and clonality
.
Curr Opin Biotechnol
2020
;
65
:
284
95
.
35.
Lu
T
,
Zhang
Z
,
Zhu
J
,
Wang
Y
,
Jiang
P
,
Xiao
X
, et al
.
Deep learning-based prediction of the T cell receptor–antigen binding specificity
.
Nat Mach Intell
2021
;
3
:
864
75
.
36.
Glanville
J
,
Huang
H
,
Nau
A
,
Hatton
O
,
Wagar
LE
,
Rubelt
F
, et al
.
Identifying specificity groups in the T cell receptor repertoire
.
Nature
2017
;
547
:
94
8
.
37.
Huang
H
,
Wang
C
,
Rubelt
F
,
Scriba
TJ
,
Davis
MM
.
Analyzing the Mycobacterium tuberculosis immune response by T-cell receptor clustering with GLIPH2 and genome-wide antigen screening
.
Nat Biotechnol
2020
;
38
:
1194
202
.
38.
Zhang
H
,
Liu
L
,
Zhang
J
,
Chen
J
,
Ye
J
,
Shukla
S
, et al
.
Investigation of antigen-specific T-cell receptor clusters in human cancers
.
Clin Cancer Res
2020
;
26
:
1359
71
.
39.
Dash
P
,
Fiore-Gartland
AJ
,
Hertz
T
,
Wang
GC
,
Sharma
S
,
Souquette
A
, et al
.
Quantifiable predictive features define epitope-specific T cell receptor repertoires
.
Nature
2017
;
547
:
89
93
.
40.
Nazarov
VI
,
Pogorelyy
MV
,
Komech
EA
,
Zvyagin
IV
,
Bolotin
DA
,
Shugay
M
, et al
.
tcR: an R package for T cell receptor repertoire advanced data analysis
.
BMC Bioinformatics
2015
;
16
:
175
.
41.
Arunkumar
M
,
Zielinski
CE
.
T-cell receptor repertoire analysis with computational tools-an immunologist's perspective
.
Cells
2021
;
10
:
3582
.
42.
Wang
P
,
Jin
X
,
Zhou
W
,
Luo
M
,
Xu
Z
,
Xu
C
, et al
.
Comprehensive analysis of TCR repertoire in COVID-19 using single cell sequencing
.
Genomics
2021
;
113
:
456
62
.
43.
Yang
HQ
,
Wang
YS
,
Zhai
K
,
Tong
ZH
.
Single-cell TCR sequencing reveals the dynamics of T cell repertoire profiling during pneumocystis infection
.
Front Microbiol
2021
;
12
:
637500
.
44.
Gerdemann
U
,
Fleming
RA
,
Kaminski
J
,
McGuckin
C
,
Rui
X
,
Lane
JF
, et al
.
Identification and tracking of alloreactive T cell clones in rhesus macaques through the RM-scTCR-Seq platform
.
Front Immunol
2021
;
12
:
804932
.
45.
Wolf
K
,
Hether
T
,
Gilchuk
P
,
Kumar
A
,
Rajeh
A
,
Schiebout
C
, et al
.
Identifying and tracking low-frequency virus-specific TCR clonotypes using high-throughput sequencing
.
Cell Rep
2018
;
25
:
2369
78
.
46.
Reuben
A
,
Gittelman
R
,
Gao
J
,
Zhang
J
,
Yusko
EC
,
Wu
CJ
, et al
.
TCR repertoire intratumor heterogeneity in localized lung adenocarcinomas: an association with predicted neoantigen heterogeneity and postsurgical recurrence
.
Cancer Discov
2017
;
7
:
1088
97
.
47.
Lefranc
MP
,
Giudicelli
V
,
Ginestoux
C
,
Jabado-Michaloud
J
,
Folch
G
,
Bellahcene
F
, et al
.
IMGT, the international ImMunoGeneTics information system
.
Nucleic Acids Res
2009
;
37
:
D1006
12
.
48.
Giudicelli
V
,
Duroux
P
,
Ginestoux
C
,
Folch
G
,
Jabado-Michaloud
J
,
Chaume
D
, et al
.
IMGT/LIGM-DB, the IMGT comprehensive database of immunoglobulin and T cell receptor nucleotide sequences
.
Nucleic Acids Res
2006
;
34
:
D781
4
.
49.
Fleri
W
,
Paul
S
,
Dhanda
SK
,
Mahajan
S
,
Xu
X
,
Peters
B
, et al
.
The immune epitope database and analysis resource in epitope discovery and synthetic vaccine design
.
Front Immunol
2017
;
8
:
278
.
50.
Rubelt
F
,
Busse
CE
,
Bukhari
SAC
,
Burckert
JP
,
Mariotti-Ferrandiz
E
,
Cowell
LG
, et al
.
Adaptive immune receptor repertoire community recommendations for sharing immune-repertoire sequencing data
.
Nat Immunol
2017
;
18
:
1274
8
.
51.
Christley
S
,
Aguiar
A
,
Blanck
G
,
Breden
F
,
Bukhari
SAC
,
Busse
CE
, et al
.
The ADC API: a web API for the programmatic query of the AIRR data commons
.
Front Big Data
2020
;
3
:
22
.
52.
Tickotsky
N
,
Sagiv
T
,
Prilusky
J
,
Shifrut
E
,
Friedman
N
.
McPAS-TCR: a manually curated catalogue of pathology-associated T cell receptor sequences
.
Bioinformatics
2017
;
33
:
2924
9
.
53.
Shugay
M
,
Bagaev
DV
,
Zvyagin
IV
,
Vroomans
RM
,
Crawford
JC
,
Dolton
G
, et al
.
VDJdb: a curated database of T-cell receptor sequences with known antigen specificity
.
Nucleic Acids Res
2018
;
46
:
D419
D27
.
54.
Chen
SY
,
Yue
T
,
Lei
Q
,
Guo
AY
.
TCRdb: a comprehensive database for T-cell receptor sequences with powerful search function
.
Nucleic Acids Res
2021
;
49
:
D468
D74
.
55.
Kote
S
,
Pirog
A
,
Bedran
G
,
Alfaro
J
,
Dapic
I
.
Mass spectrometry-based identification of MHC-associated peptides
.
Cancers
2020
;
12
:
535
.
56.
Martini
S
,
Nielsen
M
,
Peters
B
,
Sette
A
.
The immune epitope database and analysis resource program 2003–2018: reflections and outlook
.
Immunogenetics
2020
;
72
:
57
76
.
57.
Reynisson
B
,
Alvarez
B
,
Paul
S
,
Peters
B
,
Nielsen
M
.
NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data
.
Nucleic Acids Res
2020
;
48
:
W449
W54
.
58.
Hobohm
U
,
Meyerhans
A
.
A pattern search method for putative anchor residues in T cell epitopes
.
Eur J Immunol
1993
;
23
:
1271
6
.
59.
Hoof
I
,
Peters
B
,
Sidney
J
,
Pedersen
LE
,
Sette
A
,
Lund
O
, et al
.
NetMHCpan, a method for MHC class I binding prediction beyond humans
.
Immunogenetics
2009
;
61
:
1
13
.
60.
Jurtz
V
,
Paul
S
,
Andreatta
M
,
Marcatili
P
,
Peters
B
,
Nielsen
M
.
NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data
.
J Immunol
2017
;
199
:
3360
8
.
61.
Gomez-Perosanz
M
,
Ras-Carmona
A
,
Lafuente
EM
,
Reche
PA
.
Identification of CD8(+) T cell epitopes through proteasome cleavage site predictions
.
BMC Bioinformatics
2020
;
21
:
484
.
62.
Liepe
J
,
Mishto
M
,
Textoris-Taube
K
,
Janek
K
,
Keller
C
,
Henklein
P
, et al
.
The 20S proteasome splicing activity discovered by SpliceMet
.
PLoS Comput Biol
2010
;
6
:
e1000830
.
63.
Kim
Y
,
Ponomarenko
J
,
Zhu
Z
,
Tamang
D
,
Wang
P
,
Greenbaum
J
, et al
.
Immune epitope database analysis resource
.
Nucleic Acids Res
2012
;
40
:
W525
30
.
64.
Schumacher
TN
,
Schreiber
RD
.
Neoantigens in cancer immunotherapy
.
Science
2015
;
348
:
69
74
.
65.
Li
B
,
Li
T
,
Wang
B
,
Dou
R
,
Zhang
J
,
Liu
JS
, et al
.
Ultrasensitive detection of TCR hypervariable-region sequences in solid-tissue RNA-seq data
.
Nat Genet
2017
;
49
:
482
3
.
66.
Springer
I
,
Besser
H
,
Tickotsky-Moskovitz
N
,
Dvorkin
S
,
Louzoun
Y
.
Prediction of specific TCR-peptide binding from large dictionaries of TCR-peptide pairs
.
Front Immunol
2020
;
11
:
1803
.
67.
Springer
I
,
Tickotsky
N
,
Louzoun
Y
.
Contribution of T cell receptor alpha and beta CDR3, MHC typing, V and J genes to peptide binding prediction
.
Front Immunol
2021
;
12
:
664514
.
68.
Chronister
WD
,
Crinklaw
A
,
Mahajan
S
,
Vita
R
,
Kosaloglu-Yalcin
Z
,
Yan
Z
, et al
.
TCRMatch: predicting T-cell receptor specificity based on sequence similarity to previously characterized receptors
.
Front Immunol
2021
;
12
:
640725
.
69.
Montemurro
A
,
Schuster
V
,
Povlsen
HR
,
Bentzen
AK
,
Jurtz
V
,
Chronister
WD
, et al
.
NetTCR-2.0 enables accurate prediction of TCR-peptide binding by using paired TCRalpha and beta sequence data
.
Commun Biol
2021
;
4
:
1060
.
70.
Marcou
Q
,
Mora
T
,
Walczak
AM
.
High-throughput immune repertoire analysis with IGoR
.
Nat Commun
2018
;
9
:
561
.
71.
Liedmann
S
,
Liu
X
,
Guy
CS
,
Crawford
JC
,
Rodriguez
DA
,
Kuzuoglu-Ozturk
D
, et al
.
Localization of a TORC1-eIF4F translation complex during CD8(+) T cell activation drives divergent cell fate
.
Mol Cell
2022
;
82
:
2401
14
.
72.
Barennes
P
,
Quiniou
V
,
Shugay
M
,
Egorov
ES
,
Davydov
AN
,
Chudakov
DM
, et al
.
Benchmarking of T cell receptor repertoire profiling methods reveals large systematic biases
.
Nat Biotechnol
2021
;
39
:
236
45
.
73.
Soto
C
,
Bombardi
RG
,
Kozhevnikov
M
,
Sinkovits
RS
,
Chen
EC
,
Branchizio
A
, et al
.
High frequency of shared clonotypes in human T cell receptor repertoires
.
Cell Rep
2020
;
32
:
107882
.
74.
Sethna
Z
,
Elhanati
Y
,
Callan
CG
,
Walczak
AM
,
Mora
T
.
OLGA: fast computation of generation probabilities of B- and T-cell receptor amino acid sequences and motifs
.
Bioinformatics
2019
;
35
:
2974
81
.
75.
Schultheiss
C
,
Paschold
L
,
Simnica
D
,
Mohme
M
,
Willscher
E
,
von Wenserski
L
, et al
.
Next-generation sequencing of T and B cell receptor repertoires from COVID-19 patients showed signatures associated with severity of disease
.
Immunity
2020
;
53
:
442
55
.
76.
Mukhamedova
M
,
Wrapp
D
,
Shen
CH
,
Gilman
MSA
,
Ruckwardt
TJ
,
Schramm
CA
, et al
.
Vaccination with prefusion-stabilized respiratory syncytial virus fusion protein induces genetically and antigenically diverse antibody responses
.
Immunity
2021
;
54
:
769
80
.
77.
Wahl
I
,
Obraztsova
AS
,
Puchan
J
,
Hundsdorfer
R
,
Chakravarty
S
,
Sim
BKL
, et al
.
Clonal evolution and TCR specificity of the human TFH cell response to Plasmodium falciparum CSP
.
Sci Immunol
2022
;
7
:
eabm9644
.
78.
Rognes
T
,
Scheffer
L
,
Greiff
V
,
Sandve
GK
.
CompAIRR: ultra-fast comparison of adaptive immune receptor repertoires by exact and approximate sequence matching
.
Bioinformatics
2022
;
38
:
4230
2
.
79.
Sethna
Z
,
Isacchini
G
,
Dupic
T
,
Mora
T
,
Walczak
AM
,
Elhanati
Y
.
Population variability in the generation and selection of T-cell repertoires
.
PLoS Comput Biol
2020
;
16
:
e1008394
.
80.
Sidhom
JW
,
Larman
HB
,
Pardoll
DM
,
Baras
AS
.
DeepTCR is a deep learning framework for revealing sequence concepts within T-cell repertoires
.
Nat Commun
2021
;
12
:
1605
.
81.
Jokinen
E
,
Huuhtanen
J
,
Mustjoki
S
,
Heinonen
M
,
Lahdesmaki
H
.
Predicting recognition between T cell receptors and epitopes with TCRGP
.
PLoS Comput Biol
2021
;
17
:
e1008814
.
82.
Weber
A
,
Born
J
,
Rodriguez Martinez
M
.
TITAN: T-cell receptor specificity prediction with bimodal attention networks
.
Bioinformatics
2021
;
37
:
i237
i44
.
83.
Chiou
SH
,
Tseng
D
,
Reuben
A
,
Mallajosyula
V
,
Molina
IS
,
Conley
S
, et al
.
Global analysis of shared T cell specificities in human non-small cell lung cancer enables HLA inference and antigen discovery
.
Immunity
2021
;
54
:
586
602
.
84.
Diaz-Mitoma
F
,
Kumar
A
,
Karimi
S
,
Kryworuchko
M
,
Daftarian
MP
,
Creery
WD
, et al
.
Expression of IL-10, IL-4 and interferon-gamma in unstimulated and mitogen-stimulated peripheral blood lymphocytes from HIV-seropositive patients
.
Clin Exp Immunol
1995
;
102
:
31
9
.
85.
Czerkinsky
CC
,
Nilsson
LA
,
Nygren
H
,
Ouchterlony
O
,
Tarkowski
A
.
A solid-phase enzyme-linked immunospot (ELISPOT) assay for enumeration of specific antibody-secreting cells
.
J Immunol Methods
1983
;
65
:
109
21
.
86.
Altman
JD
,
Moss
PA
,
Goulder
PJ
,
Barouch
DH
,
McHeyzer-Williams
MG
,
Bell
JI
, et al
.
Phenotypic analysis of antigen-specific T lymphocytes
.
Science
1996
;
274
:
94
6
.
87.
Simon
S
,
Labarriere
N
.
PD-1 expression on tumor-specific T cells: Friend or foe for immunotherapy?
Oncoimmunology
2017
;
7
:
e1364828
.
88.
Shuford
WW
,
Klussman
K
,
Tritchler
DD
,
Loo
DT
,
Chalupny
J
,
Siadak
AW
, et al
.
4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses
.
J Exp Med
1997
;
186
:
47
55
.
89.
Sun
G
,
Sun
X
,
Li
W
,
Liu
K
,
Tian
D
,
Dong
Y
, et al
.
Critical role of OX40 in the expansion and survival of CD4 T-cell-derived double-negative T cells
.
Cell Death Dis
2018
;
9
:
616
.
90.
Scott
AC
,
Dundar
F
,
Zumbo
P
,
Chandran
SS
,
Klebanoff
CA
,
Shakiba
M
, et al
.
TOX is a critical regulator of tumour-specific T cell differentiation
.
Nature
2019
;
571
:
270
4
.
91.
Corgnac
S
,
Boutet
M
,
Kfoury
M
,
Naltet
C
,
Mami-Chouaib
F
.
The emerging role of CD8(+) tissue resident memory T (TRM) cells in antitumor immunity: a unique functional contribution of the CD103 integrin
.
Front Immunol
2018
;
9
:
1904
.
92.
Raczkowski
F
,
Rissiek
A
,
Ricklefs
I
,
Heiss
K
,
Schumacher
V
,
Wundenberg
K
, et al
.
CD39 is upregulated during activation of mouse and human T cells and attenuates the immune response to Listeria monocytogenes
.
PLoS One
2018
;
13
:
e0197151
.
93.
Liu
R
,
Jiang
W
,
Mellins
ED
.
Yeast display of MHC-II enables rapid identification of peptide ligands from protein antigens (RIPPA)
.
Cell Mol Immunol
2021
;
18
:
1847
60
.
94.
Li
G
,
Bethune
MT
,
Wong
S
,
Joglekar
AV
,
Leonard
MT
,
Wang
JK
, et al
.
T cell antigen discovery via trogocytosis
.
Nat Methods
2019
;
16
:
183
90
.
95.
Joglekar
AV
,
Leonard
MT
,
Jeppson
JD
,
Swift
M
,
Li
G
,
Wong
S
, et al
.
T cell antigen discovery via signaling and antigen-presenting bifunctional receptors
.
Nat Methods
2019
;
16
:
191
8
.
96.
Tran
E
,
Turcotte
S
,
Gros
A
,
Robbins
PF
,
Lu
YC
,
Dudley
ME
, et al
.
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer
.
Science
2014
;
344
:
641
5
.
97.
Danilova
L
,
Anagnostou
V
,
Caushi
JX
,
Sidhom
JW
,
Guo
H
,
Chan
HY
, et al
.
The mutation-associated neoantigen functional expansion of specific T cells (MANAFEST) assay: a sensitive platform for monitoring antitumor immunity
.
Cancer Immunol Res
2018
;
6
:
888
99
.
98.
Chan
HY
,
Zhang
J
,
Garliss
CC
,
Kwaa
AK
,
Blankson
JN
,
Smith
KN
.
A T cell receptor sequencing-based assay identifies cross-reactive recall CD8(+) T cell clonotypes against autologous HIV-1 epitope variants
.
Front Immunol
2020
;
11
:
591
.
99.
Klinger
M
,
Pepin
F
,
Wilkins
J
,
Asbury
T
,
Wittkop
T
,
Zheng
J
, et al
.
Multiplex identification of antigen-specific T cell receptors using a combination of immune assays and immune receptor sequencing
.
PLoS One
2015
;
10
:
e0141561
.
100.
Zhang
SQ
,
Ma
KY
,
Schonnesen
AA
,
Zhang
M
,
He
C
,
Sun
E
, et al
.
High-throughput determination of the antigen specificities of T cell receptors in single cells
.
Nat Biotechnol
2018
[Online ahead of print].
101.
Ma
KY
,
Schonnesen
AA
,
He
C
,
Xia
AY
,
Sun
E
,
Chen
E
, et al
.
High-throughput and high-dimensional single-cell analysis of antigen-specific CD8(+) T cells
.
Nat Immunol
2021
;
22
:
1590
8
.
102.
Arnaud
M
,
Chiffelle
J
,
Genolet
R
,
Navarro Rodrigo
B
,
Perez
MAS
,
Huber
F
, et al
.
Sensitive identification of neoantigens and cognate TCRs in human solid tumors
.
Nat Biotechnol
2022
;
40
:
656
60
.
103.
Joshi
K
,
de Massy
MR
,
Ismail
M
,
Reading
JL
,
Uddin
I
,
Woolston
A
, et al
.
Spatial heterogeneity of the T cell receptor repertoire reflects the mutational landscape in lung cancer
.
Nat Med
2019
;
25
:
1549
59
.
104.
Chen
R
,
Li
J
,
Fujimoto
J
,
Hong
L
,
Hu
X
,
Quek
K
, et al
.
Immunogenomic intertumor heterogeneity across primary and metastatic sites in a patient with lung adenocarcinoma
.
J Exp Clin Cancer Res
2022
;
41
:
172
.
105.
Reuben
A
,
Spencer
CN
,
Prieto
PA
,
Gopalakrishnan
V
,
Reddy
SM
,
Miller
JP
, et al
.
Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma
.
NPJ Genom Med
2017
;
2
:
10
.
106.
Gerlinger
M
,
Quezada
SA
,
Peggs
KS
,
Furness
AJ
,
Fisher
R
,
Marafioti
T
, et al
.
Ultra-deep T cell receptor sequencing reveals the complexity and intratumour heterogeneity of T cell clones in renal cell carcinomas
.
J Pathol
2013
;
231
:
424
32
.
107.
Chen
M
,
Chen
R
,
Jin
Y
,
Li
J
,
Hu
X
,
Zhang
J
, et al
.
Cold and heterogeneous T cell repertoire is associated with copy number aberrations and loss of immune genes in small-cell lung cancer
.
Nat Commun
2021
;
12
:
6655
.
108.
Dejima
H
,
Hu
X
,
Chen
R
,
Zhang
J
,
Fujimoto
J
,
Parra
ER
, et al
.
Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features
.
Nat Commun
2021
;
12
:
2722
.
109.
Cui
JH
,
Lin
KR
,
Yuan
SH
,
Jin
YB
,
Chen
XP
,
Su
XK
, et al
.
TCR repertoire as a novel indicator for immune monitoring and prognosis assessment of patients with cervical cancer
.
Front Immunol
2018
;
9
:
2729
.
110.
Beshnova
D
,
Ye
J
,
Onabolu
O
,
Moon
B
,
Zheng
W
,
Fu
YX
, et al
.
De novo prediction of cancer-associated T cell receptors for noninvasive cancer detection
.
Sci Transl Med
2020
;
12
:
eaaz3738
.
111.
Arnaud
M
,
Bobisse
S
,
Chiffelle
J
,
Harari
A
.
The promise of personalized TCR-based cellular immunotherapy for cancer patients
.
Front Immunol
2021
;
12
:
701636
.
112.
Rohaan
MW
,
Wilgenhof
S
,
Haanen
J
.
Adoptive cellular therapies: the current landscape
.
Virchows Arch
2019
;
474
:
449
61
.
113.
Hammerl
D
,
Rieder
D
,
Martens
JWM
,
Trajanoski
Z
,
Debets
R
.
Adoptive T cell therapy: new avenues leading to safe targets and powerful allies
.
Trends Immunol
2018
;
39
:
921
36
.
114.
Tran
E
,
Robbins
PF
,
Lu
YC
,
Prickett
TD
,
Gartner
JJ
,
Jia
L
, et al
.
T-cell transfer therapy targeting mutant KRAS in cancer
.
N Engl J Med
2016
;
375
:
2255
62
.
115.
Sheih
A
,
Voillet
V
,
Hanafi
LA
,
DeBerg
HA
,
Yajima
M
,
Hawkins
R
, et al
.
Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19 CAR-T immunotherapy
.
Nat Commun
2020
;
11
:
219
.
116.
Kantoff
PW
,
Higano
CS
,
Shore
ND
,
Berger
ER
,
Small
EJ
,
Penson
DF
, et al
.
Sipuleucel-T immunotherapy for castration-resistant prostate cancer
.
N Engl J Med
2010
;
363
:
411
22
.
117.
Sheikh
N
,
Cham
J
,
Zhang
L
,
DeVries
T
,
Letarte
S
,
Pufnock
J
, et al
.
Clonotypic diversification of intratumoral T cells following sipuleucel-T treatment in prostate cancer subjects
.
Cancer Res
2016
;
76
:
3711
8
.
118.
Wieland
A
,
Kamphorst
AO
,
Adsay
NV
,
Masor
JJ
,
Sarmiento
J
,
Nasti
TH
, et al
.
T cell receptor sequencing of activated CD8 T cells in the blood identifies tumor-infiltrating clones that expand after PD-1 therapy and radiation in a melanoma patient
.
Cancer Immunol Immunother
2018
;
67
:
1767
76
.
119.
Charles
J
,
Mouret
S
,
Challende
I
,
Leccia
MT
,
De Fraipont
F
,
Perez
S
, et al
.
T-cell receptor diversity as a prognostic biomarker in melanoma patients
.
Pigment Cell Melanoma Res
2020
;
33
:
612
24
.
120.
Valpione
S
,
Mundra
PA
,
Galvani
E
,
Campana
LG
,
Lorigan
P
,
De Rosa
F
, et al
.
The T cell receptor repertoire of tumor infiltrating T cells is predictive and prognostic for cancer survival
.
Nat Commun
2021
;
12
:
4098
.
121.
Lecuelle
J
,
Boidot
R
,
Mananet
H
,
Derangere
V
,
Albuisson
J
,
Goussot
V
, et al
.
TCR clonality and genomic instability signatures as prognostic biomarkers in high grade serous ovarian cancer
.
Cancers
2021
;
13
:
4394
.
122.
Postow
MA
,
Manuel
M
,
Wong
P
,
Yuan
J
,
Dong
Z
,
Liu
C
, et al
.
Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma
.
J Immunother Cancer
2015
;
3
:
23
.
123.
Dong
N
,
Moreno-Manuel
A
,
Calabuig-Farinas
S
,
Gallach
S
,
Zhang
F
,
Blasco
A
, et al
.
Characterization of circulating T cell receptor repertoire provides information about clinical outcome after PD-1 blockade in advanced non-small cell lung cancer patients
.
Cancers
2021
;
13
:
2950
.
124.
Wang
G
,
Mudgal
P
,
Wang
L
,
Shuen
TWH
,
Wu
H
,
Alexander
PB
, et al
.
TCR repertoire characteristics predict clinical response to adoptive CTL therapy against nasopharyngeal carcinoma
.
Oncoimmunology
2021
;
10
:
1955545
.
125.
Cader
FZ
,
Hu
X
,
Goh
WL
,
Wienand
K
,
Ouyang
J
,
Mandato
E
, et al
.
A peripheral immune signature of responsiveness to PD-1 blockade in patients with classical Hodgkin lymphoma
.
Nat Med
2020
;
26
:
1468
79
.
126.
Subudhi
SK
,
Aparicio
A
,
Gao
J
,
Zurita
AJ
,
Araujo
JC
,
Logothetis
CJ
, et al
.
Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities
.
Proc Natl Acad Sci U S A
2016
;
113
:
11919
24
.
127.
Lozano
AX
,
Chaudhuri
AA
,
Nene
A
,
Bacchiocchi
A
,
Earland
N
,
Vesely
MD
, et al
.
T cell characteristics associated with toxicity to immune checkpoint blockade in patients with melanoma
.
Nat Med
2022
;
28
:
353
62
.
128.
Lakkis
FG
,
Lechler
RI
.
Origin and biology of the allogeneic response
.
Cold Spring Harb Perspect Med
2013
;
3
:
a014993
.
129.
Alachkar
H
,
Mutonga
M
,
Kato
T
,
Kalluri
S
,
Kakuta
Y
,
Uemura
M
, et al
.
Quantitative characterization of T-cell repertoire and biomarkers in kidney transplant rejection
.
BMC Nephrol
2016
;
17
:
181
.
130.
Aschauer
C
,
Jelencsics
K
,
Hu
K
,
Heinzel
A
,
Gregorich
MG
,
Vetter
J
, et al
.
Prospective tracking of donor-reactive T-cell clones in the circulation and rejecting human kidney allografts
.
Front Immunol
2021
;
12
:
750005
.
131.
Han
FF
,
Fan
H
,
Ren
LL
,
Wang
HG
,
Wang
C
,
Ma
X
, et al
.
Profiling the pattern of human TRB/IGH-CDR3 repertoire in liver transplantation patients via high-throughput sequencing analysis
.
Scand J Immunol
2020
;
92
:
e12912
.
132.
Yew
PY
,
Alachkar
H
,
Yamaguchi
R
,
Kiyotani
K
,
Fang
H
,
Yap
KL
, et al
.
Quantitative characterization of T-cell repertoire in allogeneic hematopoietic stem cell transplant recipients
.
Bone Marrow Transplant
2015
;
50
:
1227
34
.
133.
Amoriello
R
,
Chernigovskaya
M
,
Greiff
V
,
Carnasciali
A
,
Massacesi
L
,
Barilaro
A
, et al
.
TCR repertoire diversity in multiple sclerosis: high-dimensional bioinformatics analysis of sequences from brain, cerebrospinal fluid and peripheral blood
.
EBioMedicine
2021
;
68
:
103429
.
134.
Yohannes
DA
,
Freitag
TL
,
de Kauwe
A
,
Kaukinen
K
,
Kurppa
K
,
Wacklin
P
, et al
.
Deep sequencing of blood and gut T-cell receptor beta-chains reveals gluten-induced immune signatures in celiac disease
.
Sci Rep
2017
;
7
:
17977
.
135.
Fuchs
YF
,
Eugster
A
,
Dietz
S
,
Sebelefsky
C
,
Kuhn
D
,
Wilhelm
C
, et al
.
CD8(+) T cells specific for the islet autoantigen IGRP are restricted in their T cell receptor chain usage
.
Sci Rep
2017
;
7
:
44661
.
136.
Luo
L
,
Liang
W
,
Pang
J
,
Xu
G
,
Chen
Y
,
Guo
X
, et al
.
Dynamics of TCR repertoire and T cell function in COVID-19 convalescent individuals
.
Cell Discov
2021
;
7
:
89
.
137.
Li
Y
,
Hu
J
,
Wang
Y
,
Liu
D
,
Shi
Y
,
Zhang
J
, et al
.
T-cell repertoire characteristics of asymptomatic and re-detectable positive COVID-19 patients
.
Front Immunol
2021
;
12
:
769442
.
138.
Zhong
Z
,
Wu
H
,
Zhang
Q
,
Zhong
W
,
Zhao
P
.
Characteristics of T cell receptor repertoires of patients with acute myocardial infarction through high-throughput sequencing
.
J Transl Med
2019
;
17
:
21
.
139.
Tang
TT
,
Zhu
YC
,
Dong
NG
,
Zhang
S
,
Cai
J
,
Zhang
LX
, et al
.
Pathologic T-cell response in ischaemic failing hearts elucidated by T-cell receptor sequencing and phenotypic characterization
.
Eur Heart J
2019
;
40
:
3924
33
.
140.
Pasetto
A
,
Lu
YC
.
Single-cell TCR and transcriptome analysis: an indispensable tool for studying T-cell biology and cancer immunotherapy
.
Front Immunol
2021
;
12
:
689091
.
141.
Fu
T
,
Dai
LJ
,
Wu
SY
,
Xiao
Y
,
Ma
D
,
Jiang
YZ
, et al
.
Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response
.
J Hematol Oncol
2021
;
14
:
98
.
142.
Hudson
WH
,
Sudmeier
LJ
.
Localization of T cell clonotypes using the Visium spatial transcriptomics platform
.
STAR Protoc
2022
;
3
:
101391
.
143.
Wang
F
,
Flanagan
J
,
Su
N
,
Wang
LC
,
Bui
S
,
Nielson
A
, et al
.
RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues
.
J Mol Diagn
2012
;
14
:
22
9
.
144.
Reddy
SM
,
Reuben
A
,
Barua
S
,
Jiang
H
,
Zhang
S
,
Wang
L
, et al
.
Poor response to neoadjuvant chemotherapy correlates with mast cell infiltration in inflammatory breast cancer
.
Cancer Immunol Res
2019
;
7
:
1025
35
.
145.
Carstens
JL
,
Correa de Sampaio
P
,
Yang
D
,
Barua
S
,
Wang
H
,
Rao
A
, et al
.
Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer
.
Nat Commun
2017
;
8
:
15095
.
146.
Senior
AW
,
Evans
R
,
Jumper
J
,
Kirkpatrick
J
,
Sifre
L
,
Green
T
, et al
.
Improved protein structure prediction using potentials from deep learning
.
Nature
2020
;
577
:
706
10
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.