Fact-checked by Grok 2 weeks ago

T-cell receptor

The T-cell receptor (TCR) is a complex expressed on the surface of T lymphocytes, which are key effectors of the , enabling the specific recognition of peptide antigens presented by (MHC) molecules on antigen-presenting cells or target cells. Composed primarily of an αβ or γδ heterodimer paired with invariant CD3 signaling chains (including CD3ε, CD3γ, CD3δ, and the ζζ homodimer), the TCR initiates intracellular signaling cascades upon ligand binding, leading to T-cell activation, proliferation, and differentiation into effector subsets that orchestrate immune responses against pathogens, tumors, and aberrant self-antigens. This recognition is MHC-restricted, with αβ TCRs typically interacting with peptide-MHC class I or II complexes to distinguish infected or abnormal cells from healthy ones, while γδ TCRs often target non-peptide antigens like lipids in mucosal and epithelial tissues. The structural architecture of the TCR features variable (V) and constant (C) immunoglobulin-like domains in its extracellular region, with six complementarity-determining regions (CDRs) forming the antigen-binding site; CDR3 loops, generated through somatic V(D)J recombination, confer the majority of specificity and diversity, yielding a potential diversity estimated at 10^15 or more, with a realized repertoire in the order of 10^8 unique receptors in human adults. Transmembrane and cytoplasmic domains facilitate assembly into an octameric complex with CD3, where immunoreceptor tyrosine-based activation motifs (ITAMs) on CD3 chains become phosphorylated by kinases like LCK upon TCR engagement, propagating signals through adaptors such as LAT and SLP-76 to activate pathways including NF-κB, NFAT, and MAPK for cytokine production and cytotoxicity. Co-receptors CD4 (for MHC II) and CD8 (for MHC I) enhance avidity and signaling by recruiting additional kinases. TCR diversity and function are sculpted during T-cell development in the thymus via positive and negative selection, ensuring self-tolerance while maintaining reactivity to foreign antigens; disruptions in this process contribute to autoimmunity, as seen in diseases like rheumatoid arthritis and multiple sclerosis, whereas TCR signaling deficiencies underlie immunodeficiencies such as severe combined immunodeficiency (SCID). In cancer, TCRs drive antitumor immunity but can be evaded by tumor microenvironments; engineered TCR therapies, redirecting T cells against neoantigens, have shown clinical promise, including response rates up to 69% in a 2014 metastatic melanoma trial, and culminated in the first FDA approval of a TCR-T therapy (afami-cel) for synovial sarcoma in August 2024. Overall, the TCR's versatility positions it as a cornerstone of adaptive immunity, balancing vigilance against infection and malignancy with the prevention of self-reactivity.

Historical Development

Discovery and Early Characterization

The specificity of T-cell responses in allograft rejection experiments during the 1970s provided early evidence for the existence of a dedicated T-cell receptor (TCR), analogous to the B-cell immunoglobulin receptor, capable of recognizing distinct antigens. This hypothesis was strengthened by the discovery of major histocompatibility complex (MHC) restriction, demonstrated by Rolf Zinkernagel and Peter Doherty, who showed that T cells recognize foreign antigens only when presented by self-MHC molecules, implying a receptor that interacts with both components. These findings, building on prior observations of T-cell-mediated graft rejection in model systems, established the conceptual foundation for a clonally distributed TCR on T lymphocytes. A major breakthrough occurred in 1984 when Stephen M. Hedrick and Mark M. Davis cloned the genes encoding the β chain of the murine TCR, revealing its structural similarity to immunoglobulins with variable and constant regions. This work, performed using cDNA libraries from T-cell hybridomas, identified rearranged gene segments that generated diversity, confirming the TCR as a heterodimeric protein distinct from B-cell receptors yet sharing evolutionary origins. Shortly thereafter, the α chain was cloned, solidifying the αβ TCR as the predominant form on most T cells. Early functional characterization relied on monoclonal antibodies generated in the mid-1980s that specifically targeted the TCR complex, enabling the demonstration of its role in antigen recognition. For instance, antibodies such as those developed by Ellis L. Reinherz and colleagues recognized the TCR αβ heterodimer (initially termed Ti) on human T cells and effectively blocked antigen-specific cytotoxic and proliferative responses in vitro. Similarly, in murine systems, antibodies like H57-597 against the αβ TCR inhibited T-cell activation upon antigen encounter, providing direct evidence that the TCR serves as the primary antigen-binding molecule. These experiments confirmed the TCR's essential function in initiating T-cell immunity without involvement in broad cellular adhesion. In 1986, a distinct TCR lineage was identified, comprising γ and δ chains, expanding the understanding of T-cell diversity. This γδ TCR was first noted through of the γ chain in 1984 by groups including , but its pairing with a novel δ chain and expression as a separate heterodimer on a minor T-cell subset was fully characterized in 1986. Unlike the MHC-restricted αβ TCR, γδ T cells were observed in epithelial tissues and suggested roles in innate-like responses to non-peptide antigens.

Key Milestones in Structural and Functional Studies

In the 1990s, crystallographic studies provided the first high-resolution views of T-cell receptor (TCR) interactions with -major histocompatibility complex (pMHC), elucidating the molecular basis of . A landmark achievement was the determination of the structure of the human A6 αβ TCR bound to the HLA-A2/ complex, which revealed a diagonal docking geometry where the TCR Vα and Vβ domains contact the α1 and α2 helices of , positioning the complementarity-determining regions (CDRs) to engage both the and MHC surfaces. This structure established that TCR-pMHC binding involves a conserved orientation, with CDR1 and CDR2 primarily interacting with MHC and CDR3 loops focusing on the , thereby explaining specificity and in T-cell responses. During the , functional studies advanced understanding of TCR triggering mechanisms, particularly through the serial engagement model, which posits that a single pMHC can sequentially bind and activate multiple TCRs to amplify signaling despite low-affinity interactions. This model was refined via biophysical and mathematical analyses, such as kinetic simulations demonstrating that one pMHC could engage up to 100 TCRs over relevant timescales, resolving the paradox of T-cell sensitivity to rare antigens. Experimental validation using fluorescence microscopy confirmed rapid TCR following limited pMHC encounters, supporting serial triggering as a key of downstream signaling pathways like MAPK . Post-2010 advances leveraged cryo-electron microscopy (cryo-EM) to resolve the full TCR-CD3 signaling complex, bridging extracellular recognition with intracellular . The 2019 cryo-EM structure of the intact human αβ TCR-CD3 complex at 3.7 Å resolution visualized the octameric assembly, including TCRαβ paired with CD3δε, CD3γε, and CD3ζζ, and highlighted transmembrane interactions stabilizing the complex for signal propagation. Subsequent refinements, such as the 2022 structure of a ligated tumor-specific TCR-CD3 complex, confirmed conformational rigidity in the extracellular domains upon pMHC binding, emphasizing allosteric transmission through CD3 ITAMs. Recent developments from 2023 to 2025, driven by single-molecule imaging techniques, have illuminated dynamic conformational changes in TCR upon binding. () microscopy studies have shown force-dependent catch-bond formation in TCR-pMHC interactions, where dwell times extend under piconewton forces, contributing to mechanosensitive signaling. In 2024, analyses highlighted how forces enhance TCR signaling for self-nonself . In 2025, studies quantified nonlinear catch-slip transitions that fine-tune T-cell of agonists versus self-peptides.

Molecular Structure

Chain Composition and Domains

The T-cell receptor (TCR) is primarily expressed as an αβ heterodimer on approximately 95% of circulating T lymphocytes in humans. Each α chain and β chain features an extracellular region composed of a (V) domain followed by a (C) domain, with the two chains linked by a bond between their respective C domains. The V domains of both chains adopt an immunoglobulin-like β-sandwich fold, characteristic of the , enabling recognition. Within each V domain, three hypervariable loops known as complementarity-determining regions (CDRs)—CDR1, CDR2, and CDR3—form the antigen-binding surface when the Vα and Vβ domains associate. CDR1 and CDR2 are encoded directly by V gene segments and exhibit relatively conserved lengths and sequences, whereas CDR3 is the most diverse, arising from the imprecise joining of V, D (for β), and J segments during T-cell . The C domains, while more conserved, also follow an immunoglobulin fold but lack CDRs and primarily stabilize the heterodimer. In humans, the mature TCRα chain extracellular region spans approximately 250 , with the Vα domain comprising about 110 residues and the Cα domain around 140 residues; the TCRβ chain is structurally analogous, with a similar overall length. A smaller subset of T cells, comprising about 5%, express a γδ TCR heterodimer instead of αβ. The γ and δ chains share a similar domain architecture to their α and β counterparts, each with an N-terminal V domain and a membrane-proximal C domain exhibiting immunoglobulin folds and three CDRs per V domain, though γδ TCRs generally lack the classical observed in αβ TCRs. The extracellular portions of TCRγ and TCRδ chains are comparable in length to those of α and β, around 250 , with Vγ and Vδ domains of roughly 110 residues each. This variability in CDR3, particularly prominent in the δ chain due to additional usage, contributes to the diverse recognition capabilities of γδ T cells. Both αβ and γδ TCRs possess short transmembrane regions of about 20 and minimal cytoplasmic tails, typically 5 residues long, which preclude direct signaling and necessitate association with invariant chains for . These structural features ensure that the TCR's primary role is detection, with intracellular signaling relayed through partnered molecules.

Variable Regions and Antigen-Binding Sites

The variable regions of the T-cell receptor (TCR) are primarily formed by the Vα and Vβ domains in αβ TCRs, which together create a analogous to the antigen-binding site in antibodies. These variable domains provide the structural framework for three complementarity-determining regions (CDRs) per chain, designated CDR1, CDR2, and CDR3, that directly interact with peptide-major complex (pMHC) ligands. CDR1 and CDR2 loops are encoded directly by the germline V gene segments and exhibit relatively fixed sequences within each V family, contributing to conserved interactions with the MHC molecule. In contrast, the CDR3 loop arises from the junctional regions of V-J (for α chain) or V-D-J (for β chain) segment joining, introducing extensive sequence variability through nucleotide additions and deletions during recombination, which enables fine-tuned recognition of diverse peptides. This hypervariability in CDR3 is critical for TCR specificity, as it allows the loop to protrude into the peptide-binding groove of the MHC, contacting the antigenic peptide directly and accommodating variations in peptide side chains. The overall geometry of the TCR-pMHC interface features a conserved diagonal docking mode, where the TCR binds at an approximately 45–75° across the top of the MHC α-helices. In this orientation, the germline-encoded CDR1 and CDR2 loops primarily contact the conserved helices of the MHC, providing a scaffold for , while the hypervariable CDR3 loops from both α and β chains focus on the exposed , enabling discrimination of specific motifs. This partitioned contact pattern ensures both broad MHC compatibility and precise specificity. Recent (cryo-EM) studies have revealed dynamic conformational changes in the TCR variable domains upon binding, including allosteric shifts in the Vα and Vβ regions that propagate from the CDR loops to modulate overall receptor flexibility. For instance, engagement induces subtle rotations and hinge motions in the V domains, enhancing to the constant regions without large-scale rearrangements. In γδ TCRs, the variable regions differ functionally from their αβ counterparts, as γδ TCRs often recognize non-peptide antigens such as phosphoantigens, , or small metabolites presented by non-classical MHC-like molecules like or MR1, with CDR3 loops playing a dominant role in direct ligand binding independent of a peptide-MHC groove. This allows γδ T cells to survey stress-induced or microbial in an MHC-unrestricted manner, contrasting the peptide-focused recognition of αβ TCRs.

Generation of Diversity

V(D)J Recombination Process

V(D)J recombination is a site-specific genetic rearrangement process that assembles the variable regions of T-cell receptor (TCR) genes during T-cell development in the , enabling the generation of a diverse repertoire of receptors. This involves the precise cutting and rejoining of variable (V), diversity (D), and joining (J) gene segments, guided by recombination signal sequences (RSSs) flanking each segment. The process is initiated by the products RAG1 and RAG2, which form a transposase-like complex that recognizes RSSs and introduces double-strand breaks (DSBs) at the junctions between coding segments and their flanking signals. Each consists of a conserved heptamer (CACAGTG or CACAGTG-like), a spacer of either 12 or 23 base pairs, and a nonamer (ACAAAAACC), with the 12/23 rule dictating that recombination occurs only between a 12-RSS and a 23-RSS to ensure proper segment pairing. Following cleavage, the complex holds the broken DNA ends in a post-cleavage synaptic complex, facilitating subsequent repair by (NHEJ) machinery to ligate the coding segments. For TCR β and δ chains, recombination proceeds in two steps: first, a joins to a J segment (D-to-J joining), followed by V-to-DJ joining, reflecting the presence of D segments in these loci. In contrast, TCR α and γ chains undergo direct V-to-J joining without involvement, as their loci lack D genes. The recombination process is temporally ordered during thymocyte maturation, with TCR β locus rearrangement initiating in CD4⁻CD8⁻ double-negative (DN) thymocytes, where successful β chain expression forms a pre-TCR complex that promotes progression to the CD4⁺CD8⁺ double-positive (DP) stage. TCR α locus recombination then occurs specifically in DP thymocytes, allowing assembly of the complete αβ TCR. This sequential activation ensures coordinated receptor assembly and is regulated by stage-specific expression of RAG1 and RAG2. Allelic exclusion maintains monoallelic expression of TCR genes, preventing dual specificities in individual T cells, through a feedback mechanism where a productive rearrangement on one signals inhibition of further recombination on the other . This is achieved via pre-TCR signaling for the β locus and αβ TCR signaling for the α locus, which downregulates expression and accessibility of the second . In humans, the TCR loci are organized on specific chromosomes: the TRA locus (encoding α chains) and nested TRD locus (encoding δ chains) on chromosome 14q11.2, the TRB locus (β chains) on 7q34, and the TRG locus (γ chains) on 7p14. These genomic arrangements support the ordered and locus-specific recombination essential for TCR diversity. For γδ TCRs, recombination follows similar principles but with distinct locus structures. The TRG locus has approximately 14 Vγ segments and 5 Jγ segments across clusters, undergoing direct V-to-J joining. The TRD locus, nested within TRA, features only 3 functional Vδ segments but 3 Dδ and 3 Jδ segments, enabling V-to-Dδ-to-Jδ joining with extensive junctional modifications that generate much of the γδ repertoire diversity, estimated at 10^3 to 10^6 unique receptors, lower than αβ but crucial for innate-like immunity.

Junctional and Combinatorial Diversity

Junctional and combinatorial diversity mechanisms significantly amplify the variability generated during V(D)J recombination, enabling the T-cell receptor (TCR) to recognize a vast array of antigens. Combinatorial diversity arises from the random selection and joining of variable (V) and joining (J) gene segments, with the TCR α chain locus featuring approximately 70 Vα segments and 61 Jα segments, yielding around 4,000 possible α chain combinations in humans. Similarly, the TCR β chain locus includes about 50 Vβ segments and 13 Jβ segments across two clusters, contributing a comparable scale of β chain variability through Vβ-to-Dβ-to-Jβ joining. These combinatorial pairings form the foundational layer of TCR diversity, independent of junctional modifications. Junctional diversity further enhances variability at the boundaries where V, D, and J segments are joined, primarily through nucleotide trimming and addition processes. Exonucleases remove nucleotides from the ends of the recombining segments, introducing deletions that create irregular junctions and increase sequence heterogeneity. Concurrently, (TdT) adds non-templated N-nucleotides at these junctions, typically 0-15 random nucleotides per junction, which is particularly active in adult thymocytes and absent in fetal stages to allow for invariant receptors in early development. These modifications, occurring at both V-J and D-J junctions for β chains, exponentially expand the potential sequences in the 3 (CDR3), the primary antigen-contact site. The combined effects of combinatorial and junctional result in an estimated theoretical TCR repertoire of 10^15 unique αβ pairs in humans, far exceeding the number of T cells in the body and ensuring broad immune coverage. Junctional , particularly in CDR3, accounts for the majority of the overall TCR repertoire variability.

TCR Complex Assembly

Interaction with CD3 and Zeta Chains

The T-cell receptor (TCR) αβ heterodimer non-covalently associates with three invariant signaling dimers—CD3γε, CD3δε, and the ζζ homodimer—to form a hetero-octameric complex essential for antigen recognition and . This assembly ensures that the variable TCR αβ chains, responsible for antigen specificity, are coupled to the invariant CD3 and ζ chains that mediate intracellular signaling. The non-covalent interactions occur primarily through the extracellular and transmembrane domains, stabilizing the complex on the T-cell surface. Each ζ polypeptide, forming a homodimer of two identical polypeptides, contains three immunoreceptor tyrosine-based activation motifs (ITAMs) in the cytoplasmic tail, providing six ITAMs total for the ζζ homodimer; each ITAM consists of the YxxL/I (where x represents any and L/I is or ), separated by 6-8 residues from a second YxxL/I motif. These ITAMs collectively provide multiple docking sites for downstream signaling molecules, amplifying the response to engagement. In contrast, the CD3 chains each contain a single ITAM, contributing to the total of ten ITAMs in the full complex. Assembly of the TCR-CD3-ζ complex begins in the () via a sequential process: initial pairing of TCR αβ with CD3δε and CD3γε heterodimers, followed by incorporation of the ζζ homodimer. This ordered progression is assisted by the ER chaperone , which binds to individual subunits like CD3δ and TCR α to prevent premature aggregation and promote proper folding and association. Incomplete assembly retains subcomplexes in the ER, blocking transport to the Golgi; thus, full complex formation is required for surface expression. Defects in this process, such as complete deficiency of the CD3ζ chain, impair TCR assembly and surface expression, resulting in T⁻B⁺NK⁺ (SCID). Cryo-electron microscopy (cryo-EM) structures from , including the model deposited as PDB: 7PHR, have revealed the detailed ectodomain of the ligated TCR-CD3 complex, showing multivalent interactions across immunoglobulin-like domains, connecting peptides, and transmembrane helices that rigidify the assembly. These insights highlight how the ectodomains of CD3 chains anchor to the TCR constant regions via hydrophobic and ionic contacts, maintaining a tilted orientation of the antigen-binding site relative to the membrane.

Role of Coreceptors CD4 and CD8

The coreceptors and play essential roles in modulating T-cell receptor (TCR) function by facilitating interactions with (MHC) molecules and enhancing signal initiation. specifically associates with molecules on antigen-presenting cells, binding primarily to the β2 domain, while interacts with molecules through the α3 domain. These interactions stabilize the TCR-pMHC complex, increasing its overall and to low-affinity antigens. Unlike the primary TCR-CD3 complex, which directly recognizes peptide-MHC ligands, the coreceptors contribute by bridging the TCR to the MHC and amplifying downstream responses. Both CD4 and CD8 recruit the Src family kinase Lck through a conserved CXCP motif in their cytoplasmic tails, positioning Lck in proximity to the TCR-CD3 complex to initiate phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs). This recruitment is crucial for efficient signal transduction, as Lck phosphorylates CD3 chains upon TCR engagement, thereby amplifying the activation threshold. In CD4+ helper T cells, which recognize antigens presented by MHC class II, coreceptor engagement promotes cytokine production and B-cell collaboration, whereas in CD8+ cytotoxic T cells interacting with MHC class I, it enhances perforin and granzyme release for target cell lysis. These distinct functions underscore the coreceptors' role in specifying T-cell effector fates based on MHC class recognition. During thymic development, and expression on double-positive thymocytes guides lineage commitment through positive selection. Thymocytes expressing receive sustained signals via , leading to downregulation of and commitment to the + helper lineage, while favor retention and cytotoxic differentiation. This process ensures and functional specialization, with coreceptor dynamics modulating signal strength over days to weeks. Recent structural studies have highlighted coreceptor-independent TCR signaling in certain γδ T cells, where the γδ TCR-CD3 complex directly engages non-peptide antigens without or involvement, revealing alternative activation pathways in innate-like T-cell subsets.

Antigen Recognition

MHC Restriction and Peptide Presentation

The concept of MHC restriction was established in 1974 through experiments demonstrating that cytotoxic T cells recognize viral antigens only when presented by (MHC) molecules matching those of the T cell's origin, rather than the antigen alone. This discovery revealed that T cell activation requires co-recognition of both foreign peptides and self-MHC, ensuring immune responses are directed against altered self-cells while avoiding direct reactivity to free pathogens. In αβ T cells, the T cell receptor (TCR) binds to peptide-MHC (pMHC) complexes, where short peptide fragments derived from antigens are loaded into the MHC binding groove. These peptides typically range from 8 to 20 amino acids in length, with MHC class I molecules accommodating 8-10 residues and MHC class II molecules binding longer sequences of 13-25 residues, though the overall span fits the 8-20 aa approximation for TCR engagement. MHC class I presents intracellular antigens, such as those from viruses or tumors, to CD8+ cytotoxic T cells, enabling the elimination of infected or malignant cells. In contrast, MHC class II displays peptides from extracellular sources, like bacterial proteins, to CD4+ helper T cells, which orchestrate broader immune responses including antibody production and macrophage activation. Unlike αβ TCRs, γδ TCRs are often MHC-unrestricted, directly recognizing non-peptidic ligands such as phosphoantigens, lipids, or stress-induced molecules like and MICB on infected or transformed cells, bypassing the need for MHC-mediated . Thermodynamic analyses of αβ TCR-pMHC interactions reveal low-affinity characterized by dissociation constants (Kd) typically in the range of 1-100 μM, reflecting the weak, transient nature of these contacts that allow rapid T cell scanning of diverse pMHC surfaces while enabling specificity through kinetic proofreading. These models highlight enthalpic and entropic contributions to , with variations influenced by sequence and MHC , underscoring the evolutionary adaptation for broad yet precise surveillance.

Mechanisms of Discrimination and Sensitivity

T-cell receptors (TCRs) discriminate between and peptides presented by (MHC) molecules through kinetic mechanisms that differentiate the duration and intensity of signaling events. peptides, which fully activate T cells, induce sustained TCR-pMHC interactions leading to prolonged signaling, whereas peptides trigger short-lived engagements that fail to propagate full signals. This kinetic allows T cells to distinguish subtle differences in , with agonists typically exhibiting slower dissociation rates from the TCR compared to antagonists. The serial triggering model, proposed in the 1990s, explains how T cells achieve high sensitivity to low antigen densities by enabling a single peptide-MHC (pMHC) complex to sequentially engage and trigger multiple TCRs on the T cell surface. In this process, rapid on-off kinetics allow one pMHC to activate up to 200 TCRs before , amplifying the signal from sparse antigens without requiring high-affinity . This model reconciles the of TCR's low individual affinity for pMHC with the system's overall sensitivity, as demonstrated in studies using fluorescently labeled pMHC to track engagement dynamics. Upon binding pMHC, the TCR undergoes conformational changes that rigidify its structure, stabilizing the interaction and facilitating signal propagation. Recent structural analyses, including hydrogen-deuterium experiments, reveal that agonist engagement dampens TCR flexibility, particularly in the variable-constant domain linker, contrasting with the more dynamic conformations induced by weak or ligands. (FRET) studies further support this by showing distinct bond conformations for agonists, where sustained proximity between TCR domains correlates with enhanced discrimination. Thymic positive and negative selection processes refine the TCR to ensure recognition of self-MHC while preventing , thereby tuning discrimination and . During positive selection, thymocytes with TCRs exhibiting low-affinity interactions with self-pMHC survive and mature, establishing a for self-MHC restriction essential for peripheral recognition. Negative selection eliminates thymocytes with high-affinity self-pMHC , shaping a that avoids strong self-reactivity and balances to foreign antigens against autoimmune . Recent studies as of have also highlighted the role of mechanical forces in TCR-pMHC interactions, where force-dependent catch bonds prolong dwell times and enhance discrimination between and antagonists. TCR sensitivity is amplified such that only 1-3 pMHC ligands are typically sufficient to initiate T cell , enabling rapid responses to minimal ; this low threshold is achieved through serial engagement and kinetic , where each pMHC can trigger multiple TCRs to propagate signals like calcium flux and production in naïve T cells. This efficiency underscores the system's design for detecting rare antigens.

Signal Transduction

Receptor Activation and Proximal Events

Upon engagement of the T-cell receptor (TCR) by peptide-major complex (pMHC) ligands, the TCR-CD3 complex undergoes a conformational shift that releases the CD3 cytoplasmic domains from the inner leaflet of the plasma membrane, thereby exposing immunoreceptor tyrosine-based motifs (ITAMs) for subsequent . This ligand-induced change is essential for initiating signaling and has been demonstrated through structural and biochemical studies showing altered accessibility of proline-rich sequences in CD3ε. The TCR complex, comprising the antigen-binding TCRαβ heterodimer non-covalently associated with CD3 subunits (δε, γε, and ζζ), contains a total of 10 ITAMs that amplify the signal upon activation. The Src-family kinase , recruited via coreceptors or , rapidly the tyrosine residues within these exposed ITAMs on the CD3 and ζ chains. Dual phosphorylation of ITAM tyrosines is required to create high-affinity docking sites, with Lck activity being a rate-limiting step in proximal signaling. This event occurs within seconds of binding and sets the stage for downstream recruitment. Phosphorylated ITAMs serve as binding sites for the tandem Src homology 2 (SH2) domains of zeta-chain-associated protein kinase 70 (ZAP-70), leading to its recruitment and activation through transphosphorylation by Lck on tyrosine 493. Activated ZAP-70 then phosphorylates adaptor proteins, including linker for activation of T cells (LAT) and SH2 domain-containing leukocyte protein of 76 kDa (SLP-76), which nucleate multimeric signaling complexes within lipid rafts. LAT, anchored in rafts via palmitoylation, and SLP-76 form scaffolds that recruit phospholipase Cγ1 (PLCγ1), Grb2, and other effectors, organizing the signalosome for efficient propagation. One of the earliest measurable outcomes is a rapid calcium influx, initiated when ZAP-70-phosphorylated PLCγ1 hydrolyzes (PIP2) to produce inositol 1,4,5-trisphosphate (IP3), which binds IP3 receptors (IP3R) on the to release stored Ca²⁺ within 6-7 seconds of TCR engagement. This is followed by store-operated calcium entry from extracellular sources, sustaining the signal and enabling cytoskeletal reorganization and other proximal responses. The TCR-CD3-ζ complex assembly is crucial for these events, as it positions the ITAMs for ordered .

Downstream Pathways and Transcriptional Regulation

Upon TCR engagement, proximal signaling events initiate the activation of phospholipase Cγ1 (PLCγ1), which hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 triggers the release of calcium from intracellular stores, while DAG recruits and activates protein kinase Cθ (PKCθ) and Ras guanine nucleotide release protein 1 (RasGRP1), amplifying downstream signals essential for T-cell activation. The calcium flux induced by IP3 activates calmodulin, which in turn stimulates the phosphatase calcineurin to dephosphorylate nuclear factor of activated T cells (NFAT) proteins. Dephosphorylated NFAT translocates to the nucleus, where it binds to target DNA sequences to drive transcription of genes involved in T-cell proliferation and cytokine production. DAG-mediated activation of PKCθ promotes the assembly of the CARMA1-BCL10-MALT1 (CBM) complex, which recruits the IκB kinase (IKK) complex to activate nuclear factor κB (NF-κB). This leads to the phosphorylation and degradation of IκB, allowing NF-κB dimers to translocate to the nucleus and induce expression of pro-inflammatory and survival genes in T cells. Parallel to these pathways, DAG also activates the Ras-ERK mitogen-activated protein kinase (MAPK) cascade, culminating in the phosphorylation and dimerization of activator protein-1 (AP-1) components, including Fos and Jun family members. Nuclear AP-1 binds to TPA-responsive elements in promoter regions, regulating genes critical for T-cell differentiation and effector functions. These pathways converge at the interleukin-2 (IL-2) promoter, where cooperative binding of NFAT, NF-κB, and AP-1 transcription factors forms an enhanceosome complex that synergistically drives IL-2 expression, a key cytokine for T-cell expansion. Recent studies highlight the role of epigenetic modifiers, such as histone deacetylases and DNA methyltransferases, in fine-tuning these transcriptional responses during T-cell differentiation, with disruptions linked to impaired effector memory formation.

Therapeutic Applications

TCR-Based Immunotherapies

TCR-T cell therapy involves the of a patient's autologous T cells to express a tumor-specific T-cell receptor (TCR), enabling these cells to recognize and eliminate cancer cells presenting specific peptide on (MHC) molecules. This approach leverages the natural antigen recognition mechanism of TCRs to target intracellular proteins, which are processed and presented via MHC pathways. A prominent example is the targeting of NY-ESO-1, a cancer-testis expressed in various solid tumors such as , , and , where TCR-engineered T cells have demonstrated tumor infiltration and cytotoxicity in clinical settings. In August 2024, the U.S. Food and Drug Administration (FDA) granted accelerated approval to afamitresgene autoleucel (Tecelra), the first , for adults with unresectable or metastatic expressing the in the context of . This approval was based on the phase 2 SPEARHEAD-1 trial, which reported an overall response rate of 43.2% (95% : 28.4–59.0; 19 of 44 patients), including 2 complete responses and 17 partial responses, with a median duration of response of 6.0 months (95% : 4.6, not reached). Compared to chimeric antigen receptor (CAR)-T cell therapies, TCR-T therapies offer distinct advantages, including MHC restriction that ensures precise recognition of peptide-MHC complexes and the ability to target intracellular antigens inaccessible to surface-directed CARs. This MHC dependency enhances specificity for tumor cells while potentially reducing off-tumor effects on healthy tissues lacking the specific antigen presentation. As of 2025, dozens of TCR-T clinical trials are ongoing worldwide, primarily targeting solid tumors such as , , and sarcomas, with reported overall response rates ranging from 20% to 50% across multiple studies. For instance, aggregated data from early-phase trials targeting NY-ESO-1 involving 107 patients showed an average response rate of 47%, highlighting efficacy in antigen-positive tumors but variable durability due to tumor heterogeneity. For example, in January 2025, the FDA granted Designation to letetresgene autoleucel for myxoid/round cell , based on a 42% ORR in phase 2 trials. A key safety concern in TCR-T therapies is the risk of arising from TCR , where engineered TCRs may recognize similar on healthy tissues, leading to off-target toxicities such as or organ damage. Preclinical and clinical assessments, including multi-tiered assays for peptide specificity, are essential to mitigate these risks, as evidenced by rare but severe cases of fatal cross-reactivity in early trials.

Engineering Advances and Challenges

Engineering of T-cell receptors (TCRs) has focused on enhancing their for peptide-major histocompatibility complex (pMHC) ligands to improve therapeutic potency in adoptive T-cell therapies. Affinity maturation techniques, such as directed of the complementarity-determining region 3 (CDR3) loops, enable the generation of TCR variants with significantly higher binding affinities. For instance, yeast display systems combined with CDR3α-directed have produced TCR mutants exhibiting up to 100-fold increases in intrinsic binding to pMHC compared to the parental receptor, while preserving specificity. These modifications leverage the natural diversity of the TCR repertoire as a starting point for optimization, allowing iterative selection of variants that maintain physiological docking modes but augment contacts for stronger interactions. To address the limitations of human leukocyte antigen (HLA) restriction, which confines TCR recognition to specific patient genotypes, universal TCR constructs have been developed to enable broader applicability. Bispecific TCR formats, often incorporating TCR domains fused to antibody-derived modules, bypass traditional HLA dependency by directly engaging tumor antigens in an MHC-independent manner, facilitating off-the-shelf therapies. As of 2025, early clinical trials are evaluating such bispecific constructs, including those targeting non-polymorphic HLA-E molecules for universal peptide presentation across diverse patient populations, demonstrating feasibility in preclinical models of chronic infections and cancers. These approaches aim to expand TCR therapy accessibility beyond HLA-matched settings. Safety mechanisms are integral to engineered TCRs to mitigate risks of uncontrolled T-cell expansion or . The inducible (iC9) suicide switch, activated by administration of a small-molecule dimerizer like AP1903, rapidly eliminates transduced T cells by triggering , providing a controllable safeguard . This system has been successfully integrated into TCR-engineered T cells, achieving dose-dependent depletion in preclinical models and clinical settings for , with near-complete elimination observed within hours of activation. Despite these advances, engineering TCRs presents significant challenges, particularly regarding and . On-target/off-tumor arises when high-affinity TCRs cross-react with similar self-peptides presented on healthy tissues, leading to unintended immune attacks, as evidenced by severe adverse in early trials targeting antigens like MAGE-A3. Manufacturing scalability remains a bottleneck, with personalized TCR-T cell hindered by variable efficiencies, cell expansion inconsistencies, and the need for GMP-compliant processes that support large-scale autologous therapies without compromising viability or potency. Recent innovations, such as automated bioreactors, are addressing these issues but require further optimization for widespread adoption. In 2025, (AI) has emerged as a transformative tool for , particularly in predicting and mitigating . Models like NetTCR-struc employ structure-aware to forecast TCR-pMHC interactions, enabling the design of variants with minimized off-target binding while optimizing . These AI-driven approaches analyze vast datasets of TCR sequences and epitopes to simulate binding landscapes, outperforming traditional methods in identifying safe, high-specificity candidates for therapeutic development.

References

  1. [1]
    T cell receptor (TCR) signaling in health and disease - Nature
    Dec 13, 2021 · This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors.
  2. [2]
    The structural basis of T-cell receptor (TCR) activation - NIH
    In this review, we examine the molecular basis for TCR activation in light of the recently determined cryoEM structure of a complete TCR–CD3 complex.Missing: paper | Show results with:paper
  3. [3]
    The T cell antigen receptor: the Swiss army knife of the immune system
    The mammalian T cell receptor (TCR) orchestrates immunity by responding to many billions of different ligands that it has never encountered before.
  4. [4]
    Legacy of the discovery of the T-cell receptor: 40 years of shaping ...
    May 31, 2024 · The discovery of the TCR has provided valuable insights into how the immune system functions and has paved the way for the development of new ...Fig. 1 · Fig. 3 · Mhc Restriction
  5. [5]
  6. [6]
    DNA Origami Tension Sensors (DOTS) for Single-Molecule Force ...
    Aug 25, 2025 · We report unprecedented measurements of TCR–antigen forces at fluid membranes, detecting forces with magnitudes of 8 to 19 pN, and tracking ...
  7. [7]
    Mechanical force matters in early T cell activation - PNAS
    Sep 6, 2024 · We provide a perspective on how mechanical forces between a T cell and an antigen-presenting cell can influence the bond of a single T cell receptor major ...Sign Up For Pnas Alerts · Rebinding Of Tcr To Pmhc... · Effects Of Tcr Rigidity On...
  8. [8]
    αβ and γδ T cell receptors: Similar but different - Morath - 2020
    Jan 29, 2020 · Review compares the αβ and γδ T cell Ag receptor (TCR) complexes, with a particular focus on their molecular signaling mechanisms.
  9. [9]
    STCRDab: the structural T-cell receptor database - Oxford Academic
    Oct 26, 2017 · Each TCR chain is characterised by two immunoglobulin domains: a variable domain (V) and a constant (C). Both variable and constant domains have ...
  10. [10]
    γδ T cells: origin and fate, subsets, diseases and immunotherapy
    Nov 22, 2023 · In 1984, γδ T cells were first reported by Tonegawa et al., with significant contributions from Adrian C. Hayday. Until 1987, important work on ...<|control11|><|separator|>
  11. [11]
    Crystal Structure of the Vα Domain of a T Cell Antigen Receptor
    The crystal structure of the Vα domain of a T cell antigen receptor (TCR) was determined at a resolution of 2.2 angstroms. This structure represents an ...
  12. [12]
    TRAC - T cell receptor alpha chain constant - Homo sapiens (Human)
    Jul 18, 2018 · Alpha-beta T cell receptors are antigen specific receptors which are essential to the immune response and are present on the cell surface of T ...
  13. [13]
    TRGC1 - T cell receptor gamma constant 1 - UniProt
    Constant region of T cell receptor (TR) gamma chain that participates in the antigen recognition (PubMed ... Amino acids. 173 (go to sequence). Protein existence.
  14. [14]
    T cell receptor delta constant - B7Z8K6 - UniProt
    Constant region of T cell receptor (TR) delta chain that participates in the antigen recognition (PubMed ... Homo sapiens (Human). Amino acids. 153 (go to ...
  15. [15]
    The T Cell Antigen Receptor α Transmembrane Domain ...
    Nov 20, 2018 · TCRα and β each contain an extracellular variable (V) and constant (C) domain, a membrane-proximal connecting peptide (CP), a single ...
  16. [16]
    How the T Cell Receptor Sees Antigen—A Structural View
    Aug 12, 2005 · The CDR1 and 2 loop sequences are constant for each type of chain and are therefore referred to as “germline derived,” whereas the CDR3 loops ...
  17. [17]
    Comparative Analysis of the CDR Loops of Antigen Receptors
    We find that TCR and antibody CDRs tend to have different length distributions, and where they have similar lengths, they mostly occupy distinct structural ...
  18. [18]
    T-cell receptor structures and predictive models reveal comparable ...
    Mar 4, 2025 · TCRs function as heterodimers, typically consisting of an alpha and a beta chain whose recombination and sequence diversification process relies ...
  19. [19]
    The Hypervariable Loops of Free TCRs Sample Multiple Distinct ...
    Nov 12, 2018 · These CDR loops are the structural elements that recognize the peptide-MHC surface. Over two decades of crystallographic work have generated a ...
  20. [20]
    The Evolving T Cell Receptor Recognition Code: The Rules Are ...
    Jan 13, 2025 · Thus, the mutation in the neoepitope pre-organizes the peptide into a conformation compatible for TCR recognition. The concept of pre- ...
  21. [21]
    Structural insights into the editing of germ-line–encoded interactions ...
    T-cell receptors (TCRs) bind peptide–MHC (pMHC) via their six complementarity-determining region (CDR) loops, three from the variable alpha (Vα) domain and ...Sign Up For Pnas Alerts · Results · Peptide Recognition By Tcrs...
  22. [22]
    Reconciling views on T cell receptor germline bias for MHC - PMC
    This view shows the roughly diagonal binding orientation with the germline-derived CDRs 1 and 2 primarily engaging the MHC helices, and the TCR CDR3s engaging ...
  23. [23]
    Two Human T Cell Receptors Bind in a Similar Diagonal Mode to ...
    The structure was determined by molecular replacement using the refined model of the A6 TCR/HLA-A2/Tax peptide complex (Garboczi et al. 1996a). The position of ...
  24. [24]
    The resting and ligand-bound states of the membrane-embedded ...
    Sep 20, 2024 · Specifically, it is unclear whether ligand binding and activation involves conformational change in the TCR–CD3 complex. While reconstitution of ...
  25. [25]
    In situ cell-surface conformation of the TCR-CD3 signaling complex
    Nov 7, 2024 · We show that the TCR-CD3 complex undergoes minimal subunit movements or reorientations upon interaction with activating antibodies and pMHC tetramers.
  26. [26]
    Structures of human γδ T cell receptor–CD3 complex - Nature
    Apr 24, 2024 · Here we present two cryo-electron microscopy (cryo-EM) structures of unliganded human Vγ9Vδ2 and Vγ5Vδ1 TCR–CD3 complexes, revealing two ...Missing: Dong | Show results with:Dong
  27. [27]
    Ligand recognition by the γδ TCR and discrimination between ...
    Jul 24, 2020 · In contrast, the γδ TCR does not require MHC-mediated antigen presentation and no general requirement for co-receptor interaction has been ...
  28. [28]
    V(D)J Recombination: Mechanism, Errors, and Fidelity - PMC - NIH
    Nov 1, 2016 · Immunoglobulin light chain genes and TCR alpha and gamma genes rearrange in a single step, involving V-J recombination, as D segments are absent ...
  29. [29]
    Diagnostic role of tests for T cell receptor (TCR) genes - PMC - NIH
    The rearrangement of TCRB and TCRD is a two step process, in which a D gene first recombines with a J gene and then a V gene recombines with the D–J block (fig ...The Tcr Loci · V(d)j Recombination · T Cell Lymphomas And...
  30. [30]
    T Cell Receptor Gene Rearrangement Lineage Analysis ... - NIH
    The diversity of the TCRs derives from the largely random and imprecise somatic recombination of variable (V), diversity (D), and junctional (J) genes, the ...<|control11|><|separator|>
  31. [31]
    The rearrangement of antigen-receptor gene segments ... - NCBI
    During the period of α-chain gene rearrangement, α:β T-cell receptors are first expressed, and selection by peptide:MHC complexes in the thymus can begin. Along ...<|control11|><|separator|>
  32. [32]
    Mechanics of T cell receptor gene rearrangement - PMC - NIH
    Vβ-to-DβJβ recombination is then suppressed in DP thymocytes despite re-expression of Rag-1 and Rag-2. Almost all Vβ gene segments are several hundred kb ...
  33. [33]
    Antigen Receptor Allelic Exclusion: An Update and Reappraisal - PMC
    To enforce allelic exclusion by feedback inhibition, only one allele can initiate V-to-(D)-J recombination during the time window required for feedback signals ...
  34. [34]
    Chromosome choice for initiation of V-(D)-J recombination is not ...
    We found no parental chromosomal bias in the initiation of V-(D)-J recombination in T or B cells, eliminating genomic imprinting as a potential regulator.
  35. [35]
    T cell clonality assessment: past, present and future - PubMed Central
    Oct 21, 2017 · The human TCR genes are organised into four loci: TRA, TRB, TRD and TRG. The TRA locus is found on the long arm of chromosome 14 in band 14q11.
  36. [36]
    IMGT/GeneInfo: T cell receptor gamma TRG and delta TRD genes in ...
    Apr 26, 2006 · The TR genes are located in four loci, designated as TRA, TRB, TRG and TRD which contribute to the synthesis of the alpha, beta, gamma and ...
  37. [37]
    A conserved αβ transmembrane interface forms the core of a ... - PNAS
    Oct 10, 2016 · The TCR heterodimer could therefore sit above the two closely positioned CD3 modules, consistent with the compact, elongated structure observed ...Results · Tcrαβ Tm Association Is... · Tcrαβ Tm Modeling In...
  38. [38]
    Structure of a fully assembled tumor-specific T cell receptor ligated ...
    Aug 18, 2022 · We used cryogenic electron microscopy to determine the structure of a tumor-reactive TCRαβ/CD3δγε 2 ζ 2 complex bound to a melanoma-specific human class I pMHC.
  39. [39]
    ITAM-mediated Signaling by the T-Cell Antigen Receptor - PMC
    Moreover, although all TCR ITAMs share the conserved YxxL/I-X6-8-YXXL/I motif, the amino acid sequence of each ITAM is distinctive (Fig. 1). The question that ...
  40. [40]
    ITAMs versus ITIMs: striking a balance during cell regulation - JCI
    In total, the TCR complex contains ten ITAMs, one from each of the CD3 chains and six from the ζ dimer. By contrast, other ITAM-containing receptors have two to ...<|control11|><|separator|>
  41. [41]
    Phosphorylation of ITAM motifs in CD3 complexes
    ITAMs consist of evolutionarily conserved amino-acid sequence motifs of D/ExYxxLx(6-8)YxxL. ... CD3 chains including the three tandem ITAMs in each zeta chains.
  42. [42]
    Calnexin Associates Exclusively with Individual CD3δ and T Cell ...
    Assembly of the T cell antigen receptor (TCR) complex occurs within the endoplasmic reticulum (ER) and proceeds in a highly ordered manner by: (i) formation ...
  43. [43]
    Retention of unassembled components of integral ... - PubMed
    It is concluded that calnexin interacts with incompletely assembled TCR components and retains them in the ER.
  44. [44]
    Retention of Unassembled Components of Integral Membrane ...
    CD3 ε chains cotransfected with the forms of calnexin that were not retained in the ER exited the ER and colocalized with calnexin. Since engineered calnexin ...
  45. [45]
    T−B+NK+ severe combined immunodeficiency caused by complete ...
    Dec 14, 2006 · These findings provide the first demonstration that complete CD3ζ deficiency in humans can cause SCID by preventing normal TCR assembly and surface expression.
  46. [46]
    7PHR: Structure of a fully assembled T-cell receptor ... - RCSB PDB
    Aug 31, 2022 · The antigen-bound complex comprises 11 subunits stabilized by multivalent interactions across three structural layers, with clustered membrane- ...Missing: 7PZB | Show results with:7PZB
  47. [47]
    Structural and Biophysical Insights into the Role of CD4 ... - Frontiers
    CD4 and CD8 enhance T cell signaling by binding MHC class II (CD4) or MHC class I (CD8) molecules on APCs. The interaction of CD4 with MHC class II greatly ...Abstract · Introduction · Role of the CD8 Stalk Region... · Co-Receptors and TCR...
  48. [48]
    Role of alpha3 domain of class I MHC molecules in the activation of ...
    As a co-receptor, CD8 can bind to the alpha3 domain of the same MHC class I molecules as the TCR to facilitate TCR signaling. To evaluate the role of the MHC ...
  49. [49]
    Cooperative binding of T cell receptor and CD4 to peptide-MHC ...
    Nov 17, 2022 · Here we show, using two-dimensional (2D) mechanical-based assays, that the affinity of CD4–pMHC interaction is 3-4 logs lower than that of cognate TCR–pMHC ...
  50. [50]
    Adjusting to self in the thymus: CD4 versus CD8 lineage ...
    Jul 9, 2024 · This model helps to explain the link between MHC recognition and CD4 versus CD8 lineage commitment and highlights the importance of TCR signal ...
  51. [51]
    The order and logic of CD4 versus CD8 lineage choice and ... - Nature
    Jan 4, 2021 · In the unperturbed thymus, the order of coreceptor gene expression states is Cd4+ Cd8a+ followed by Cd4+ Cd8a− and later by Cd4− Cd8a+, ...
  52. [52]
    Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic ...
    Apr 19, 1974 · R. M. ZINKERNAGEL &; P. C. DOHERTY. Nature volume 248, pages 701–702 (1974)Cite this article. 10k Accesses. 1842 Citations. 43 Altmetric.Missing: MHC | Show results with:MHC
  53. [53]
    Thermodynamics of T cell receptor – peptide/MHC interactions - NIH
    Here we review the use of thermodynamic measurements in the study of TCR-pMHC interactions, with attention to the diversity in binding thermodynamics.
  54. [54]
    Unconventional Peptide Presentation by Classical MHC Class I and ...
    Oct 13, 2020 · The majority of MHC I peptides are 8–10 amino acids long (9-mers are preferred) and longer peptides either bind in a zig-zag orientation [23] ...
  55. [55]
    Improved Prediction of MHC II Antigen Presentation through ...
    Apr 18, 2020 · MHC II has an open binding cleft, allowing it to interact with peptides of a broad length range, most commonly of 13–25 amino acids. (4) The ...
  56. [56]
    Major Histocompatibility Complex (MHC) Class I and MHC Class II ...
    Mar 17, 2017 · Here, we review the recent literature that describe MHC class I and II dynamics from a theoretical and experimental point of view.
  57. [57]
    The ins and outs of MHC class II-mediated antigen processing and ...
    In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide–MHC class II ...
  58. [58]
    Quantitative Analysis of the Contribution of TCR/pepMHC Affinity ...
    Various studies have shown that the affinities of productive TCR:pepMHC interactions are low, with KD values in the 1 to 50 μM range Davis et al.
  59. [59]
    Kinetic discrimination in T-cell activation. - PNAS
    Kinetic discrimination in T-cell activation. J D Rabinowitz, C Beeson, D S ... peptides referred to as T-cell receptor antagonists. Results of recent ...
  60. [60]
    Serial triggering of many T-cell receptors by a few peptide–MHC ...
    May 11, 1995 · Here we show that a small number of peptide–MHC complexes can achieve a high TCR occupancy, because a single complex can serially engage and trigger up to ∼200 ...
  61. [61]
    Serial triggering of many T-cell receptors by a few peptide ... - PubMed
    Here we show that a small number of peptide-MHC complexes can achieve a high TCR occupancy, because a single complex can serially engage and trigger up to ...
  62. [62]
    TCR-pMHC complex formation triggers CD3 dynamics - eLife
    Mar 22, 2023 · Additional support comes from hydrogen-deuterium exchange experiments, which point to a rigidification of the TCR upon TCR-pMHC binding, which ...
  63. [63]
    TCR–pMHC bond conformation controls TCR ligand discrimination
    Sep 17, 2019 · The TCR conformational change model postulates that a conformational change in a TCR occurs upon pMHC binding, but no conformational changes at ...Missing: Kd | Show results with:Kd<|control11|><|separator|>
  64. [64]
    The MHC Reactivity of the T Cell Repertoire Prior to Positive and ...
    It is thought that positive selection favors immature CD4+CD8+ thymocytes that bind weakly or strongly to self-MHC/peptide complexes, whereas negative selection ...
  65. [65]
    Direct single molecule measurement of TCR triggering by agonist ...
    Jul 3, 2013 · We report direct single molecule measurements of TCR triggering by agonist pMHC in hybrid junctions between live primary T cells and supported lipid membranes.Elife Digest · Agonist Pmhc Binding To Tcr... · Tcr And Mcc Agonist Pmhc...
  66. [66]
    An induced rebinding model of antigen discrimination - PMC
    T cells are able to respond rapidly to very low levels of agonist pMHC, indicating that they are sensitive to a small number of productive TCR–pMHC interactions ...
  67. [67]
  68. [68]
  69. [69]
    Receptor signaling in immune cell development and function - PMC
    These proximal signaling events lead to the activation of PLCγ1 [15]. Activated PLCγ1 hydrolyzes phosphatidylinositol 4,5-bisphosphate [PIP2] to generate two ...
  70. [70]
    Dephosphorylation of the nuclear factor of activated T cells (NFAT ...
    ... NFAT proteins are dephosphorylated by the Ca2+/calmodulin-dependent phosphatase calcineurin and translocate to the nucleus to activate target gene expression.
  71. [71]
    Antigen Receptor Signaling to NF-κB via CARMA1, BCL10, and ...
    The transcription factor NF-κB plays a key role in lymphocyte activation and lymphoma formation. Scaffold proteins and the protease MALT1 link antigen receptors ...
  72. [72]
    Assembly mechanism of the CARMA1–BCL10–MALT1–TRAF6 ...
    Jan 30, 2018 · The CARMA1–BCL10–MALT1 (CBM) complex is the central mediator of T cell receptor and B cell receptor-induced NF-κB activation in lymphocytes.
  73. [73]
    Recent insights of T cell receptor-mediated signaling pathways for T ...
    May 21, 2020 · ERK1/2 signaling has an important role in controlling T cell development, differentiation, and TCR-induced signal strength. AP-1 is a basic ...
  74. [74]
    Hierarchy of signaling thresholds downstream of the T cell receptor ...
    Aug 27, 2021 · Pathways downstream of the TCR, critical for robust T cell activation, are highly dependent on the activation of NFAT, NF-κB, and AP-1 ...
  75. [75]
    CD8+ T cell stressors converge on shared metabolic–epigenetic ...
    Sep 22, 2025 · In this review, we explore how these intersecting metabolic and epigenetic pathways create self-reinforcing loops of immune paralysis across ...
  76. [76]
    Epigenetic priming as a driver of memory recall and dysfunction in T ...
    Jul 31, 2025 · This review highlights how memory T cells fight infections and cancer by retaining epigenetic “memories” that enable the rapid reactivation ...
  77. [77]
    TCR-engineered T cell therapy in solid tumors: State of the art ... - NIH
    Feb 15, 2023 · TCR-T and CAR-T cell therapies consist in genetically engineered T cells, modified to express a receptor directed against a tumor antigen. CAR-T ...
  78. [78]
    A Pilot Trial Using Lymphocytes Genetically Engineered with an NY ...
    The CG antigen NY-ESO-1 is expressed in 10% to 50% of metastatic melanomas, lung, breast, prostate, thyroid, and ovarian cancers (6–9) as well as between 70% ...
  79. [79]
    NY-ESO-1-specific redirected T cells with endogenous TCR ...
    Jun 29, 2022 · The TCR-T cell infusion exhibited significant tumor response and early-onset CRS in patients with tumors that express NY-ESO-1 at high levels.
  80. [80]
    FDA grants accelerated approval to afamitresgene autoleucel for ...
    Aug 2, 2024 · On August 2, 2024, the Food and Drug Administration granted accelerated approval to afamitresgene autoleucel (TECELRA, Adaptimmune, LLC).
  81. [81]
    Next-Generation CAR-T and TCR-T Cell Therapies for Solid Tumors
    Jun 11, 2025 · This review provides a comprehensive analysis of the current clinical development strategies for CAR-T and TCR-T cell therapies for solid tumors.
  82. [82]
    Cancer Therapy With TCR-Engineered T Cells - PubMed Central - NIH
    Therefore, TCR T therapies may have an advantage over CAR T therapies in the ability to aggressively target cancer cells while minimizing toxicity. TCR Targeted ...
  83. [83]
    TCR-engineered T cell therapy in solid tumors: State of the art and ...
    Feb 15, 2023 · This increased sensitivity may improve tumor cell detection and killing. Last, the high avidity of TCR-T cells may also improve their efficacy, ...
  84. [84]
    TCR-T cell therapy: current development approaches, preclinical ...
    Oct 4, 2024 · Cross-reactivity of TCRs represents a major safety hazard in TCR gene therapy. T-cell receptors recognize peptides presented by human ...Safety Assessment · Toxicity Assessment · Off-Tumor Toxicity And...
  85. [85]
    Multi-tiered approach to detect autoimmune cross-reactivity of ...
    Jul 26, 2023 · One of the main safety concerns for TCR-engineered T cell therapies using high-affinity TCRs is off-target toxicity (cross-reactivity) (10–12).Results · Single-Cell Tcr Paired... · T Cell Functional Assays
  86. [86]
    In vitro evolution of a T cell receptor with high affinity for peptide/MHC
    These findings show that the yeast system, combined with CDR3α-directed mutagenesis, is capable of selecting mutants with 100-fold higher intrinsic binding ...
  87. [87]
    Increased Peptide Contacts Govern High Affinity Binding ... - Frontiers
    Natural TCRs generally bind to their cognate pMHC molecules with weak affinity and fast kinetics, limiting their use as therapeutic agents.
  88. [88]
    Viral sequence determines HLA-E-restricted T cell recognition of ...
    Nov 22, 2024 · Abstract. The non-polymorphic HLA-E molecule offers opportunities for new universal immunotherapeutic approaches to chronic infectious diseases.
  89. [89]
    JNJ-78306358, a first-in-class bispecific T cell engaging antibody ...
    Mar 21, 2025 · JNJ-78306358 is a bispecific T cell engager that targets CD3 and the oncofetal protein HLA-G. JNJ-78306358 binds a unique epitope on HLA-G and competes with ...
  90. [90]
    Genetically engineered T cells for cancer immunotherapy - Nature
    Sep 20, 2019 · Similar to CAR-T therapy, the TME often inhibits the efficacy of TCR-T cell therapy. ... An inducible caspase 9 safety switch for T-cell therapy.
  91. [91]
    A systematic safety pipeline for selection of T-cell receptors to enter ...
    Aug 22, 2023 · In addition to the potential risk of cross-reactivity of TCRs to unintended peptides, the possibility of TCRs reacting to HLA alleles other ...Results · Discussion · T-Cell Activation And...
  92. [92]
    Advances and challenges in manufacturing engineered T cell ...
    Here we describe the latest technology used to manufacture CAR- and TCR-engineered T cells in the clinic, including cell purification, transduction/transfection ...
  93. [93]
    NetTCR-struc, a structure driven approach for prediction of TCR ...
    Accurate modeling of T cell receptor (TCR)–peptide–major histocompatibility complex (pMHC) interactions is critical for understanding immune recognition.
  94. [94]
    Methods T cell receptor cross-reactivity prediction improved by a ...
    Aug 20, 2025 · Comprehensively mapping all targets of a T cell receptor (TCR) is important for predicting pathogenic escape and off-target effects of TCR ...Methods · Summary · Introduction