Abstract
The T cell receptor (TCR) complex is a naturally occurring antigen sensor that detects, amplifies, and coordinates cellular immune responses to epitopes derived from membrane-associated and intracellular proteins. Thus, TCRs enable the targeting of proteins selectively expressed by cancer cells, including neoantigens, cancer-germline antigens, and viral oncoproteins. As such, they have become an emerging class of oncology therapeutics. Herein, we review the current cancer treatment landscape using TCRs and TCR-like molecules. This includes adoptive cell transfer of T cells expressing endogenous or engineered TCRs, TCR bi-specific engagers, and antibodies specific for human leukocyte antigen (HLA)-bound peptides (TCR mimics). We discuss the unique complexities associated with TCR clinical development such as HLA restriction, TCR retrieval, potency assessment, and the potential for cross-reactivity. In addition, we highlight emerging clinical data that establishes the antitumour potential of TCR-based therapies, including tumour infiltrating lymphocytes, for the treatment of diverse human malignancies. Finally, we explore the future of TCR therapeutics, including emerging genome editing methods to safely enhance potency and strategies to streamline patient identification.
Introduction.
The T cell receptor (TCR) is a lineage-defining, membrane-anchored, clonotypic immune receptor that plays a central role in the ligand-dependent activation of T lymphocytes1,2. Nearly all FDA approved cancer immunotherapies promote the activation and expansion of T cells expressing TCRs that confer recognition to tumour antigens3–5. Unlike antibodies, which physiologically bind solely to cell surface and soluble epitopes, TCRs can respond to antigens derived from the entirety of the cancer cell proteome6. This includes the ~88% of proteins that reside exclusively within intracellular compartments, such as the cytoplasm, nucleus, and mitochondria7. The unique capacity of TCRs to engage intracellular antigens, a property that dramatically expands the landscape of actionable immunologic targets, stems from the fact that they do not bind to intact proteins. Rather, they recognize proteolytically degraded polypeptides bound to human leukocyte antigen (HLA) [G] molecules that have been trafficked to the surface of human cells for extracellular presentation8.
TCRs display high sensitivity for ligands. The number of specific peptide/HLA (p/HLA) complexes on the surface of tumour cells is typically within the range of 1–100 molecules9,10. This value is orders of magnitude smaller than the number of molecules required by therapeutic antibodies11,12. Nevertheless, only one to three p/HLA complexes are sufficient to trigger T cell effector functions13,14. Simultaneously, TCRs are capable of remarkable specificity. They can discriminate between peptides that differ by a single amino acid, such as those resulting from a somatic point mutation15 or germline polymorphism16, as well as different stereoisomers of the same amino acid17.
Despite these virtues, the first TCR therapeutic (tebentafusp) only entered the standard of oncologic care in 202118. By contrast, antibodies and their derivatives, including antibody-drug conjugates, bispecific proteins, and chimeric antigen receptor (CAR)-modified T cells, have been a mainstay of cancer treatments since the mid-1990s19. In this Review, we provide a comprehensive overview of the opportunities and challenges associated with developing TCR-based therapies for the treatment of human cancers. We describe the architecture of naturally occurring TCRs and discuss strategies to rationally modify different structural domains of the TCR to create receptors with distinct biochemical, functional, and pharmacologic properties. Next, we summarize how TCRs can be retrieved and compared with respect to potency and off-target toxicity risk. Finally, we conclude by assessing recent clinical developments using the TCR as a drug and provide a vision for how TCRs will be further integrated into clinical care. On the subjects of HLA-restricted tumour antigens [G] and the molecular mechanisms of resistance to T cell-based therapies, we refer the reader to several recent reviews6,20,21.
Architecture of TCR-based therapeutics
Endogenous TCR.
The TCR is not a single molecule; rather, it is a complex of proteins in which the functions of antigen recognition and signal transduction are divided among distinct subunits. The functional unit of a TCR is an octameric complex composed of six proteins: the clonotypic TCRα/TCRβ hemichains and the invariant CD3γ, δ, ε, and ζ chains (Fig. 1a)22. These proteins assemble with a 1:1:1:1 stoichiometry comprised of the dimeric subunits TCRαβ:CD3δε:CD3γε:CD3ζζ. In lieu of TCRαβ, a minority of circulating T cells express TCRγδ [G] 23. Neither TCR hemichain possess a signal transduction domain. Rather, the receptor depends on non-covalent interactions with the CD3 molecules to initiate intracellular signaling, T cell activation, and cell fate decisions. The CD3γ, CD3δ, and CD3ε subunits are genetically related and contain a single immune receptor tyrosine-based activation motif (ITAM) [G], whereas the CD3ζ subunit is genetically unrelated and contains three ITAMs.
Fig. 1: The molecular architecture of TCR-based therapeutics.

(a-d) Comparison of the structural features of TCR and TCR-like molecules that bind specific peptide/human leukocyte antigen (p/HLA) complexes. (a) The endogenous TCR is comprised of an octameric complex composed of six proteins: the clonotypic TCRα (red) / TCRβ (blue) membrane-anchored heterodimer and the invariant CD3γ, δ, ε, and ζ chains. These proteins assemble with a 1:1:1:1 stoichiometry comprised of the dimeric subunits TCRαβ:CD3δε:CD3γε:CD3ζζ. Each TCR hemichain is composed of an antigen-binding variable (V) domain, a constant (C) domain, a transmembrane domain, and a short non-signaling cytoplasmic tail. The endogenous TCRα/TCRβ hemichains are covalently linked though a single interchain disulfide bond (grey spheres). Non-covalent interactions with the CD3 molecules facilitates intracellular signaling. HLA class I-restricted TCRs bind to a pHLA complex comprised of three alpha subunits (blue), beta-2-microglobulin (light grey) and a short polypeptide sequence typically 8–10 amino acids in length (red). (b) T-cell specificity can be genetically redirected to recognize p/HLA complexes displayed by tumour cells through expression of an exogenous TCRα (light grey) and TCRβ (dark grey) hemichain. Mispairing with the endogenous TCR hemichains can be minimized by introduction of a second interchain disulfide bond. (c) Soluble TCRs are recombinant bispecific proteins that contain a TCR’s α/β variable domains linked in a single-chain format on one end and an antibody-derived antigen binding variable heavy (VH, rouge) and variable light (VL, pink) chains specific for CD3ε on the other. (d) TCR-mimics are an alternative class of recombinant bispecific proteins that use an antibody’s VH/VL domains (yellow and purple) in place of a TCRα/TCRβ to engage a specific p/HLA complex. Shown is a diabody format TCR-mimic. (e-g) Comparison of the structural features of next-generation antigen receptors that repurpose one or several components of the TCR’s CD3 signaling complex. (e) The T cell antigen coupler (TAC) is a bispecific transmembrane protein expressed as a transgene in polyclonal T cells. One domain of a TAC uses an antibody-derived variable sequence to engage a membrane-associated tumor antigen (dark grey) while the other binds CD3ε. (f) The T cell receptor fusion construct (TRuC) is a transgene expressed in polyclonal T cells that covalently links an antibody variable sequence with specificity for a tumour antigen to an exogenous CD3ε molecule. (g) The synthetic TCR antigen receptor (STAR)/HLA-independent TCR (HIT) is a non-HLA restricted receptor that replaces the TCR variable domains with the tumor antigen-binding variable domains of an antibody. By retaining the TCR constant domains, the STAR/HIT receptor can recruit the full complex of CD3 signaling molecules upon ligand binding. V = TCR variable domain, C = TCR constant domain, VH = immunoglobulin variable heavy chain, VL = immunoglobulin variable light chain.
Structurally, each TCR hemichain is composed of a variable (V) domain, a constant (C) domain, a transmembrane domain, and a short cytoplasmic tail24. TCR diversity, and in turn specificity, is generated through combinatorial and junctional diversity involving the Vα/Vβ domains. Like an immunoglobulin’s variable heavy chain (VH), combinatorial diversity of the TCR Vβ domain results from the somatic recombination [G] of germline encoded variable (V), diversity (D), and junctional (J) gene segments1,2. The Vα domain, like an immunoglobulin’s variable light chain (VL), forms through the recombination of chain-specific V and J gene sequences. The recombined Vα and Vβ sequences are in turn linked to a Cα domain (encoded by the TRAC locus) and one of two Cβ domains (encoded by TRBC1 and TRBC2). Additional combinatorial diversity results from pairing of recombined TCRα and TCRβ hemichains. The two TCR hemichains become covalently linked through a single disulfide bond formed by conserved cysteine residues located in the Cα and Cβ domains24.
Each TCR heterodimer contains six regions of sequence hypervariability, termed complementarity-determining regions (CDRs). The CDRs are looped structures that project from a TCR’s variable domains to form the principal sites of contact with a p/HLA complex22. Between the CDRs, each variable chain possesses three framework (FR) regions that facilitate interchain packing of the Vα/Vβ domains and intrachain interfaces of the Vα/Cα and Vβ/Cβ domains25. The CDR1 and CDR2 loops are peripherally located in the solvent-exposed terminus of a TCR and are germline encoded. By contrast, the CDR3 loops are centrally located and generated through combinatorial and junctional diversification to create the most polymorphic sequences of each TCR. Beyond the juxtaposition of different V(D)J gene segments, additional diversity in the CDR3 loops is created through the deletion and addition of non-template encoded nucleotides26.
Most TCRs dock diagonally over the p/HLA complex, placing the Vα and Vβ domains over the N- and C-terminus of an HLA-bound peptide, respectively8. This configuration establishes a broad interface between the TCR and p/HLA complex. Further, it positions the regions of greatest TCR sequence diversity, the somatically rearranged CDR3 loops, over a peptide’s central core where they contribute to the receptor’s fine specificity. The germline-encoded CDR1 and CDR2 loops, by contrast, primarily (although not exclusively) contact the two α-helices that define the binding groove of an HLA molecule. These interactions ensure that a TCR recognizes the p/HLA complex in a peptide-dependent manner. Most naturally occurring TCRs possess comparatively weak binding affinities with a typical disassociation constant (Kd) measured in a micromolar range27–29. By comparison, the affinities for mature antibodies are generally in a nanomolar to picomolar range30.
Exogenous TCRs and genome editing.
The specificity of primary human T cells can be genetically redirected to recognize p/HLA complexes displayed by tumour cells through expression of an exogenous TCRαβ gene sequence (Fig. 1b)31,32. To date, integrating retroviral33–41 and lentiviral42–45 vectors have been the most common means of introducing exogenous TCRs for clinical applications. However, non-viral genome integration technologies, including the Sleeping Beauty transposon/transposase system [G] and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas946,47, can also be used. Despite an extensive safety track record48, integrating viral vectors possess certain limitations. Viruses integrate semi-randomly with variable copy numbers resulting in heterogenous transgene expression49,50. Additionally, viruses are expensive to manufacture51, can cause insertional mutagenesis52,53, and possess a relatively limited cargo capacity. Finally, integrating viral vectors leave the endogenous TCR hemichains intact. This can result in mispaired (α’/β, α/β’) heterodimers in addition to the properly paired endogenous (α/β) and exogenous (α’/β’) TCRs. Expression of endogenous and mispaired TCRs may compromise therapeutic potency by competing for a limited pool of signaling molecules54–56. Moreover, because mispaired TCRs have not undergone thymic selection, they can possess novel reactivities, including to self-antigens57. T cells expressing mispaired TCRs trigger lethal graft versus host disease (GVHD) in syngeneic mice58. Fortunately, GVHD resulting from TCR mispairing has not been observed in human clinical trials59.
To reduce TCR mispairing, nuclease-based genome editing techniques, including zinc finger nucleases60, transcription activator-like effector endonucleases61, and CRISPR/Cas949,62,63 can be used to disrupt the endogenous TRAC and/or TRBC1/2 loci. However, these strategies induce double-strand DNA breaks that risk off-target genomic effects, including insertions/deletions and chromosomal translocations64. Inhibitory RNAs can silence expression of the native TCR without inducing double-strand DNA breaks, but this approach cannot completely mitigate the risk of mispairing as knock-down is often incomplete65. DNA base editors have recently been developed that disrupt protein expression without causing double-strand DNA breaks through splice site inactivation or introduction of a premature stop codon66. Manipulation of the TRAC67 and TRBC1/268,69 loci using DNA base editors might provide a safer alternative for genetic removal of the endogenous TCR.
Beyond disrupting the endogenous TCR, CRISPR/Cas9 may also facilitate the targeted genomic integration of exogenous TCRαβ genes at the TRAC locus using template-guided homology directed repair (HDR)47,62,63. Targeted integration simultaneously provides for enhanced functionality and a more predictable safety profile compared with TCRs inserted semi-randomly into the genome70. Targeted integration enables physiologic antigen receptor regulation using the gene’s endogenous promoter, a feature that may prevent immunologic exhaustion49,70. Importantly, targeted knock-in using CRISPR/Cas9 can be performed in a non-viral manner through co-electroporation of Cas9 ribonucleoprotein (RNP) and a DNA HDR template47,62. This innovation dramatically reduces the time and cost required to generate TCR constructs for clinical use. Non-viral TCR integration has historically been less efficient than viral approaches. Process improvements, including single-stranded DNA donor templates62,71, nanoplasmids72, intranuclear template shuttling73, Cas9 RNP stabilization73, and small-molecule cocktails71 may increase editing rates.
Domain engineering.
TCR domain engineering is an alternative means of enhancing exogenous TCR potency and safety. Alterations to the constant and transmembrane domains are especially versatile because they are applicable across therapeutic candidates. For example, placement of additional cysteine residues along the Cα/Cβ interface facilitates the creation of a second interchain disulfide bond, enhancing proper hemichain pairing74,75. Murinization of all54 or selected76,77 amino acid residues in place of the human Cα/Cβ sequences can also minimize mispairing. Although this approach introduces potentially immunogenic foreign sequences, analysis of patients who received TCRs with murine constant regions has failed to find a correlation between immunogenicity, cellular persistence, and clinical outcomes41,78. If no xenogeneic sequences are desired in a cell product, inversion of the human Cα/Cβ domains (domain swapping) can be used as an alternative method to minimize mispairing79. The constant chains contain residues that undergo post-translational glycosylation80. Removal of glycosylation sites through site-directed mutagenesis enhances the functional avidity of exogenous TCRs, possibly by improving TCR clustering within the immunologic synapse. Finally, strategic replacement of non-ionizable amino acids with aliphatic residues in TCRα’s transmembrane domain can increase TCR surface expression81.
Modifications to the TCR variable domains, including the framework and CDR regions, can also improve TCR function. Unlike constant and transmembrane domain alterations, these modifications require empiric testing and optimization. Exogenously expressed TCRs with identical constant regions are not equally expressed at the cell surface25,55, a finding that suggests the variable domains contribute to TCR assembly and stability. Comparison of recurrent amino acid sequences in the framework regions of highly versus weakly expressed TCRs revealed three optimal residues at the interface of the variable and constant domains25. For TCRs containing suboptimal residues, substitution with optimal amino acids enhances surface expression and in vivo antitumour efficacy. Importantly, because framework regions do not participate in antigen binding, changes to these sites will not alter a TCR’s cross-reactivity profile.
Mutagenesis of selected CDR residues may enhance a TCR’s binding affinity. This can be accomplished through empiric testing82 or directed evolution techniques83–85. Single or combinatorial substitutions to the CDR1, CDR2, or CDR3 loops can result in as much as a million-fold increase in affinity compared with the native sequence83. Beyond modulating binding affinity, alterations to the CDR sequences may also alter TCR specificity. Two clinical trials using affinity-enhanced receptors have resulted in lethal off-tumoru/off-target toxicities attributable to changes in the TCR’s cross-reactivity profile36,86. Thus, a detailed assessment of the cross-reactivity profile for an affinity-enhanced TCR is requisite before clinical development. Notwithstanding early setbacks, many TCR therapeutics currently in advanced clinical development have undergone affinity enhancement9,82,87. A recent strategy that draws from fields in which specificity concerns are paramount (such as DNA binding and enzyme catalysis) is to engineer TCRs with improved specificity88. Rather than altering affinity, this structure-guided approach seeks to redistribute attractive interactions across the TCR:p/HLA interface to reduce off-target peptide recognition89.
Finally, proof-of-concept studies have illustrated how catch bond [G] engineering can increase TCR potency without altering binding affinity90. Catch bonds are created by the transient formation of hydrogen bonds and salt bridges under shear force conditions immediately prior to receptor disengagement91. This results in extended bond lifetimes leading to augmented TCR signaling and T cell activation. Catch bond engineering can be achieved through introduction of polar and charged amino acids into CDR loops located in proximity to but not directly in contact with the p/HLA surface. The goal of this approach is to facilitate the creation of favourable interactions during TCR disengagement from the p/HLA complex akin to a fishhook engaging its prey. Because the affinity of catch bond engineered TCRs generally remains within a physiologic range, the cross-reactivity profile of these receptors should be unaltered as residues that directly contact the ground state p/HLA complex remain fixed.
Soluble bispecific TCRs.
TCRαβ heterodimers can be expressed as a soluble, single-chain, recombinant protein, enabling the development of off-the-shelf reagents that do not require genetic engineering. Soluble TCRs can be fused to a single-chain variable fragment (scFv) derived from an agonistic anti-CD3ε antibody to create bispecific proteins termed ‘immune-mobilizing monoclonal TCR against cancer’ (ImmTAC) and ‘T cell engaging receptor’ (TCER) (Fig. 1c)9,85,92. ImmTACs and TCERs create a synthetic immunologic synapse between polyclonal T cells and target cells that express a specific p/HLA complex. However, to bind a p/HLA complex in a stable manner as a monomer requires CDR mutagenesis to enhance the TCR’s binding affinity into a picomolar range, a value ~106 times higher than naturally occurring TCRs. Altering a TCR’s binding affinity to this degree may lead to a loss in antigen specificity93–95. In the extreme, this can result in a highly promiscuous receptor that binds to a particular HLA molecule independent of the sequence of the bound peptide. Careful assessment of the specificity of affinity-enhanced TCRs, including the use of large panels of antigen and HLA mismatched target cells, is therefore obligatory. T cells redirected using a bispecific TCR exhibit antigen-specific cytolysis of target cells expressing as few as 10 p/HLA molecules9. ImmTACs have a smaller molecular weight compared with monoclonal antibodies (75 kDa versus 150 kDa); therefore, they require re-iterative infusions to maintain potency. Serum half-life extension, such as fusion to a modified IgG Fc molecule, might overcome this limitation96.
TCR-mimics.
An alternative class of bispecific T cell engaging proteins, termed TCR-mimics, use two antibody-derived scFvs covalently linked through a peptide linker10,97. One scFv binds an epitope presented as a p/HLA molecule on cancer cells while the other binds a member of the CD3 complex (Fig. 1d) on T cells. An advantage to using an scFv as the antigen binding domain is that they can be generated de novo using high-throughput techniques such as phage98 and yeast display libraries98,99, a feature that presently cannot be done for TCRs. This attribute streamlines the time required to develop candidate molecules. Like soluble TCRs, TCR-mimics are small (~55k kDA), enabling close interactions between target cells and T cells. Functional, structural, and molecular dynamic comparisons of TCRs and TCR-mimics has revealed that the two antigen receptors engage their targets in distinct ways100,101. These differences are attributable in part to the different orientation of the variable domains of antibodies versus TCRs102. TCRs typically bind a broad region of the p/HLA complex, centered along the peptide’s core, using energetically balanced interactions involving peptide side chains and invariant regions of the HLA molecule8. By contrast, many TCR-mimics dock with a bias towards the extreme terminus of the HLA-bound peptide or to one of the HLA helices using a limited number of highly focused ‘hot spot’ interactions100. A similar binding mode has been observed in conventional TCRs associated with a high degree of cross-reactivity103,104. Therefore, TCR-mimics seem to dock with p/HLA in manner that is permissive of a greater degree of peptide sequence variability compared with most TCRs, a property that can result in increased cross-reactivity. This limitation might be overcome by screening for ultra-rare scFvs which bind in a peptide-centric manner101,105 or using existing TCR-mimic antibodies that dock with a TCR-like topology as a template for mutagenesis99. In addition to bispecific engaging proteins, TCR-mimics can be incorporated into various CAR designs99,101,105.
Designs based on TCR signal transduction
Beyond strategies that use the TCR as a tumour antigen-binding domain, several novel therapies repurpose the physiologic signal transduction machinery of the TCR complex. These alternative TCR-based approaches can enhance receptor sensitivity while potentially reducing cytokine-related toxicities and T cell exhaustion associated with conventional CAR designs.
T cell antigen coupler (TAC).
TAC is a modular, bispecific transmembrane protein expressed as a transgene within T cells that incorporates two binding domains106. One binding domain is used for tumour antigen recognition and the second is used for recruitment of signaling components from the endogenous TCR complex (Fig. 1e). Unlike conventional CARs that integrate domains for T cell activation, co-stimulation, and antigen binding into a single molecule, a TAC lacks the intrinsic capacity to signal. Functionally, TAC-modified T cells exhibit antigen-specific cytokine production and cytolytic activity against target cells in the absence of tonic-signaling, a feature of some CAR designs that results in accelerated T cell exhaustion107. In vivo, TAC-modified T cells demonstrate comparable antitumour efficacy to conventional CAR-modified T cells but without cytokine-related toxicities.
TCR fusion construct (TRuC).
TRuC is an alternative antigen receptor design that, like a TAC, also engages the endogenous TCR signaling complex108,109. However, unlike a TAC, the TRuC covalently links an antigen-binding scFv to the extracellular region of the CD3ε molecule using a non-immunogenic (Gly4Ser)3 linker (Fig. 1f). Biochemical studies demonstrate that up to two TRuC molecules are incorporated into the TCR complex, a finding that is consistent with the natural stoichiometry of CD3ε within the TCR complex22. Like TAC-modified T cells, TRuC-modified T cells do not exhibit tonic-signaling and release significantly lower levels of cytokines compared with CARs while retaining antigen-specific cytolytic potency.
Synthetic TCR antigen receptor (STAR)/HLA-independent TCR (HIT).
STAR/HIT is a non-HLA restricted receptor that replaces the variable domains of the TCR with the VH/VL domains of an antibody (Fig. 1g)12,110. Because the TCR variable regions have a similar size and tertiary fold as VH/VL, these domains can be replaced interchangeably. By retaining the TCR constant domains, the STAR/HIT receptor recruits the full complex of CD3 signaling molecules. Like exogenous TCRs, the STAR/HIT receptor is a heterodimer capable of mispairing with endogenous TCR hemichains. Mispairing can be minimized using the same strategies as exogenous TCRs. Following antigen-stimulation, the STAR/HIT receptor induces TCR-like signaling, enabling enhanced responsiveness to target cells with lower antigen site densities.
Discovery of therapeutic TCR candidates.
From tumour infiltrating lymphocytes (TILs).
Cancers are frequently infiltrated by T cells, a subset of which are reactive against various classes of tumour antigens. These include tissue differentiation antigens111, cancer germline antigens112,113, antigens associated with transforming oncoviruses112, and mutation-derived neoantigens111,112,114–116. Consequently, TILs can serve as a source for TCR gene sequences that confer antitumour immunity (Fig. 2a). Despite being enriched in tumour-reactive T cells, TCR frequency alone is generally insufficient to predict tumour-reactivity117,118. This is because most TILs are passive bystanders with specificity for viruses or other pathogens111,116,119. Strategies to identify and isolate tumour-specific TIL have therefore been developed which fall into three categories: phenotypic, functional, and transcriptomic.
Fig. 2: Discovery of TCR therapeutic candidates.

(a) Fully human TCR gene sequences that confer recognition to tumour-derived peptide/HLA (p/HLA) complexes can be retrieved from healthy donors and cancer patients. Healthy donors have a broad circulating TCR repertoire that has not been subjected to the negative influence of immune-depleting cancer treatments, thymic involution, and peripheral tolerance (left). However, because the frequency of tumour-reactive TCR clonotypes is exceedingly rare within the naïve repertoire, healthy donor T cells must undergo in vitro stimulation (IVS) to enable detection. Tumour antigen-reactive T cells can be detected within the peripheral blood and tumour infiltrating lymphocytes (TILs) of patients with cancer (right). Although TCR diversity (represented as a grey bar) is typically lower in patients compared with healthy donors, the TCR repertoire often is enriched in tumour-reactive T cells that have undergone in vivo clonal expansion (represented as a purple bar). This feature may enable the retrieval of a larger number of tumour-reactive TCR clonotypes than is possible using a comparable sample volume obtained from healthy donors. (b) Tumour-reactive T cells can be generated through antigen-specific vaccination of HLA transgenic mice. HLA transgenic mice possess a diverse TCR repertoire that has not been subjected to central thymic tolerance against human proteins that differ in sequence from their murine counterparts. However, TCRs retrieved from HLA transgenic mice possess immunogenic murine variable sequences capable of triggering host-versus-graft rejection when infused into humans. To overcome this limitation, “humanized” mice have been generated in which the genetic sequences encoding the human TCR variable chains have been knocked into the genetic loci encoding the murine TCR chains (light blue, right). TCRs generated in these mice therefore possess fully human TCR variable sequences. α1/α2 = highly polymorphic domains of HLA class I; α3 = HLA class I constant domain; β2M = beta-2 microglobulin (HLA light chain); V = TCR variable domain; C = TCR constant domain; Mhc = genetic locus encoding the major histocompatibility complex proteins (the murine ortholog of HLA).
Phenotypic markers are constitutively expressed membrane-associated proteins, making them straightforward to measure and convenient to use in large-scale isolations. Detection of phenotypic markers is accomplished with fluorescently-labeled monoclonal antibodies using FACS analysis or molecularly barcoded antibodies followed by cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) [G]120,121. Expression of the ATPase CD39119,120,122–124, the tissue-resident marker CD103120,122, or the immune checkpoint receptors PD-1118,125–127, lymphocyte activation gene 3 protein (LAG-3)125, and T-cell immunoglobulin mucin receptor 3 (TIM-3, also known as Hepatitis A virus cellular receptor 2 (HAVCR2)) 125 identify tumour-reactive TIL populations. In the case of CD4+ TIL, cells with a regulatory T (Treg) cell phenotype may serve as an additional source of TCR sequences that confer reactivity to HLA class II-restricted tumour antigens121,128.
Functional markers either directly assess the antigen-specificity of TIL or measure TCR ligation-induced changes in surface marker expression. Although technically more challenging to assess than phenotypic markers, functional markers enable enrichment of tumour-reactive TIL with greater specificity. TCRs specific for cancer antigens can be identified through binding to synthetic HLA multimers loaded with tumour-associated peptides. Fluorochrome-conjugated peptide (p)/HLA multimers facilitate the sorting of antigen-specific TIL using FACS or magnetic bead isolation129. However, this approach requires a large starting population and has a limited capacity to simultaneously screen multiple antigen specificities. CITE-seq using DNA-barcoded p/HLA multimers combined with single-cell sequencing enables high-throughput screening and retrieval of antigen-linked TCR sequences using smaller sample sizes130. Alternatively, the specificity of TCR clonotypes from TILs of unknown specificity can be de-orphaned using yeast display or antigen-presenting cells (APCs) displaying highly diverse p/HLA libraries131–135. Despite the utility of each of these approaches, practical limitations restrict the total number of HLA alleles, epitopes, and peptide lengths that can be screened at a time. Two members of the tumour necrosis factor receptor superfamily, 4–1BB (also known as TNFRSF9 and CD137) and OX40 (also known as TNFRSF4 and CD134), are not expressed on resting T cells but are upregulated after antigen stimulation. Unlike cytokine expression, expression of 4–1BB and OX40 is dependent on TCR-ligation and occurs independently of T cell differentiation. TIL selection based on expression of these two markers can therefore serve as a functional strategy to retrieve tumour-reactive TCRs without a priori knowledge of the epitopes they recognize136,137.
Finally, single-cell transcriptomic signatures can identify tumour-reactive TCR clonotypes directly ex vivo without the need for expansion or functional testing. Expression of the chemokine CXCL13120,127,138,139 and multiple exhaustion-related genes, including ENTPD1 (encoding CD39), PDCD1 (encoding PD-1), HAVCR2, and TIGIT are higher in cancer-specific T cells compared with bystander T cells111,116,120,121,127,139. Bystander T cells, by contrast, often express high levels of memory-associated genes such as IL7R and TCF7111,116.
From circulating T cells in cancer patients.
TCR clonotypes expressed by TILs can be found among circulating T cells, albeit at significantly lower frequencies111,116,140. Thus, peripheral blood can serve as a minimally invasive source of tumour-reactive T cells (Fig. 2a). In select cases, T cells that are specific for cancer antigens and have undergone clonal expansion are detectable directly ex vivo using p/HLA multimers129. More commonly, strategies that enrich for rare TCR clonotypes are required. The co-inhibitory molecule PD1112,140 and T-cell memory marker CD45RO141 identify circulating populations containing antitumour T cells. However, because tumour-reactive T cells comprise a minor fraction of these pools, ex vivo T cell expansion is required for reactivity screening and TCR sequencing. This can be accomplished by in vitro sensitization (IVS) using monocyte-derived dendritic cells pulsed with tumour-associated peptides or electroporated with RNA encoding tumour antigens.
If the peptide sequence and restricting HLA allele for a tumour antigen are known, antigen-specific T cells can be isolated directly using synthetic p/HLA complexes. Conjugation of the p/HLA complex to a fluorescent dye129, Strep-tagII sequence [G]142, or paramagnetic artificial APC [G]143 permits enrichment and expansion of rare T cell populations. Alternatively, tumour-reactive T cells can be captured directly ex vivo and their corresponding TCR gene sequences retrieved in a single step using DNA-barcoded magnetic nanoparticles displaying p/HLA multimers47.
From healthy donors.
A significant proportion of HLA-presented peptides fail to elicit T cell responses in patients with cancer144,145. This may occur because of ineffective priming or because prior lines of cancer treatment negatively influence tumour-reactive T cell fitness146. To overcome these limitations, TCR discovery can be ‘outsourced’ using the naïve T (TN) cell repertoire of healthy donors (Fig. 2a). In this approach, TN cells are isolated to maximize TCR repertoire diversity prior to IVS using APCs. APCs can be pulsed with peptide63,147 or transfected with mRNA encoding a tumour antigen to enforce more physiologic antigen presentation requiring proteolytic degradation and endogenous HLA loading144,148,149. Alternatively, antigen-specific TN cells may be isolated directly without prior IVS using p/HLA multimer enrichment followed by single-cell sorting150. Screening multiple healthy donors increases the likelihood of detecting a T cell response and the diversity of T cell clonotypes generated63,144,148. In the case of tumour-associated antigens, in which expression is shared by cancer cells and normal tissues, HLA mismatched donors may be used as a strategy to retrieve high-affinity TCR sequences32,149,151. This is because the T cell repertoire reactive against allogeneic HLA molecules is not subject to negative thymic selection. However, because TCRs retrieved from mismatched donors may exhibit promiscuity for generic allogeneic HLA molecules152, systematic screening for potential off-target reactivities is required prior to clinical testing153.
From HLA transgenic mice.
Transgenic (Tg) mice that express HLA molecules and have been immunized with human tumour antigens can be used as a source of TCR candidates (Fig. 2b). There are several advantages to this approach. First, it exploits differences in the protein sequences of mice and humans as a strategy to overcome the negative influence of thymic selection on the pool of TCRs targeting self-antigens154. Second, because rodents are relatively easy to immunize, HLA Tg mice are a time and cost-efficient way of generating diverse TCRs. Variable sequences of T cell clones retrieved from immunized HLA Tg mice have been used as the source of TCRs for several clinical trials34–36. Evidence of on-target antitumour immunity in patients who received TCRs obtained from HLA Tg mice validates the therapeutic potential of this approach. However, a major limitation of TCRs sourced in this manner is that the receptor’s murine variable sequences can contain immunogenic epitopes78.
From humanized mice.
To overcome the limitations of using TCRs with murine variable sequences, Tg mouse strains that express the entire human TCRαβ genomic loci but are deficient in murine TCR sequences have been generated155,156. Conceptually, this approach builds on prior work demonstrating that Tg mice engineered with the human immunoglobulin VH/VL gene loci can serve as a source of fully human antibody sequences157. Crossing TCR humanized mice with HLA Tg mice enables the sampling of a diverse human TCR repertoire that is HLA-restricted. Immunization of TCR humanized mice with cancer antigens generates TCR candidates with fully human variable sequences (Fig. 2b). In some cases, TCRs cloned from humanized mice demonstrate higher functional avidity compared with human-derived TCRs158,159.
Vetting TCRs for safety and potency.
Assessing on-target/off-tumour toxicity risk.
On-target/off-tumour toxicities are expected when tumour cells and normal tissues share expression of the same p/HLA complexes160. Consequently, toxicity risk mitigation for therapeutic TCR candidates begins with rigorously assessing antigen expression by healthy cells (Fig. 3a). In the case of epitopes resulting from somatic mutations and transforming oncoviruses, off-tumor toxicity risk is minimized because normal tissues do not express these proteins. For other antigen classes, an initial assessment of expression can be made using publicly available transcriptomic and proteomic databases. RNA-sequencing (seq) has high sensitivity, high specificity, and a large dynamic range161. RNA-seq databases, such as the Genotype-Tissue Expression (GTEx) project162, provide high-quality sequencing results from multiple normal tissues. Human single-cell RNA expression atlases complement bulk RNA-seq data by facilitating the identification of rare cell populations163. Because the correlation between gene and protein expression is imperfect164, detectable RNA transcripts within critical tissues is alone insufficient to conclude a candidate antigen represents an unsafe target. Analysis of protein level expression is therefore advisable as a confirmatory step. Finally, it is important to consider that select tissues, such as the testes, do not express HLA molecules and therefore are impervious to direct T cell recognition165.
Fig. 3: Strategies to resolve the safety profile and potency and of TCR therapeutic candidates.

(a-c) Methods to quantify the safety profile of a TCR candidate. (a) Normal tissue expression assesses the presence of a target antigen in healthy tissues to assess the risk for on-target/off-tumour toxicities from a TCR candidate. Ideally, the risk profile for a novel antigen will be determined using multiple assays, including bulk tissue RNA sequencing, single-cell RNA sequencing, and measurement of protein level expression. (b) Allogeneic (allo) reactivity measures the capacity of a TCR to respond to a mismatched HLA molecule irrespective of the bound peptide sequence. Allo-reactivity is assessed by co-culture of T cells expressing a candidate TCR with a panel of target cells, such as EBV-transformed B-lymphoblastic cell lines (B-LCLs), that express diverse HLA alleles. (c) TCR degeneracy is the capacity of a single TCR to respond to unrelated peptide sequences but restricted by the same HLA molecule. The degeneracy potential of a TCR can be measured using sequential amino acid scanning mutagenesis (eg. X-Scan) or large combinatorial libraries. In the example shown, the TCR is capable of recognizing both the cognate peptide sequence (highlighted boxes) and unrelated peptide sequences with similar or even enhanced potency. In the heatmap, dark blue represents an amino acid that results in enhanced TCR-mediated cytokine release or binding relative to other amino acids. In all instances where potential cross-reactive peptide sequences are identified, confirmatory studies are required to establish physiologic significance. (d-i) Methods to quantify TCR potency. (d) Functional avidity measures the capacity of T cells expressing multiple copies of a membrane-associated TCR to functionally respond to progressively lower concentrations of a specific p/HLA complex. Functional avidity results from the summation of all binding interactions between a T cell and target cell, including contributions from the TCR, the CD8 or CD4 co-receptors, and intercellular adhesion molecules. (e) Structural avidity measures the TCR-p/HLA dissociation rate (koff) using fluorophore-conjugated (star) p/HLA multimers that dissociate into monomers following addition of an inert chemical (grey diamond). Unlike functional avidity, measurement of structural avidity is not affected by the differentiation state of T cells. (f) Co-receptor dependency measures the capacity of T cells expressing a TCR candidate to respond to target cells in the absence of the avidity and signaling contributions facilitated by the CD8α/β or CD4 co-receptors. The ability of a TCR to function in a coreceptor-independent manner suggests the TCR has a relatively high binding affinity. (g) TCR affinity is the strength of interaction between a single TCR molecule and a single p/HLA complex. Most commonly, affinity is measured using surface plasmon resonance by flowing a recombinant, soluble, single-chain TCR over a metal surface containing immobilized p/HLA complexes. Under equilibrium conditions, a TCR’s binding affinity is inversely proportional to its dissociation constant (Kd) which in turn is defined by the ratio of the rates of dissociation and association (koff/kon). Plot illustrates time-dependent changes in binding (measured as relative response units) of a single-chain TCR flowed over a sensor containing immobilized p/HLA complexes at different concentrations. (h) In vitro tumour recognition measures the capacity of a TCR to trigger T cell responses to physiologic levels of an endogenously processed peptide displayed in the context of a specific HLA allele by tumour cells. Tumour recognition can be quantified by measuring T cell-mediated cytolysis or cytokine production. Solid red and grey lines represent the time-dependent cytolytic activity of two therapeutic TCR candidates while the grey dashed lines represents the cytolytic activity of a control TCR. (i) In vivo tumour regression assesses the ability of a TCR candidate (solid red line) to penetrate an established tumour mass and cause a sustained antitumour response over time. Grey solid and dashed lines represent a control TCR and no treatment control, respectively.
Assessing cross-reactivity potential.
TCRs can also mediate off-target toxicities resulting from the degeneracy potential of any individual TCR (Box 1). The safety profile of a TCR is not driven by the absolute number of peptide sequences it can potentially bind; rather, it is determined by the capacity of a TCR to engage peptides resulting from the endogenous processing and presentation of off-target human proteins in the context of an HLA molecule. Unacceptable toxicities will result only in cases in which an off-target protein is expressed by healthy tissues that perform critical functions. Differences in protein sequences between laboratory animals and humans combined with the absence of HLA expression limit the utility of in vivo toxicology studies for assessing on- and off-target TCR toxicity risks. In vitro strategies are therefore generally required to quantify the degeneracy potential and safety profile of TCR candidates. It remains an outstanding question in the field which assays should be performed and under what circumstances to assess for cross reactivity. Below, we present an overview of several techniques which have been used to support Investigational New Drug applications [G] for TCRs as well as evolving technologies which may be useful for de-risking candidates.
Box #1: TCR diversity, degeneracy, and cross-reactivity.
The processes of combinatorial and junctional diversity can theoretically create as many as 1×1015-1020 unique TCR sequences24. However, the human TCR repertoire is far more constrained and is typically measured within the range of 2×107-1×108 unique sequences301,302. In part, the discordance between the theoretical and measured size of the TCR repertoire reflects the influence of negative and positive thymic selection303. To enable a limited T cell repertoire to respond to the largest possible universe of potential epitopes, many TCRs are capable of cross-reacting to multiple unrelated peptides, a parameter termed TCR degeneracy. The biologic requirement for TCR cross-reactivity has been established using both a theoretical framework304,305 and experimental data306. The degeneracy potential of any individual TCR is highly variable. Some TCRs bind only to a limited number of structurally related peptides whereas others are capable of binding to more than 1×106 unique peptide sequences306. Adding additional complexity, the peptides recognized by a TCR can share little to no sequence or structural homology131,169. Thus, TCR cross-reactivity is an expected rule rather than an exception.
Three distinct mechanisms contribute to TCR degeneracy. First, some TCRs and p/HLA complexes display flexibility in how they engage one another. Flexibility can occur as a result of plasticity in the conformations of CDR loops, enabling a single receptor to accommodate different p/HLA landscapes307. Alternatively, flexibility can occur through the orientation of a TCR’s variable domains over different p/HLA complexes252,308 or rearrangements in a peptide and presenting HLA protein to accommodate a TCR169,309. Second, some TCRs display highly focused interactions involving a minimal ‘hot spot’ motif displayed by an HLA-bound peptide103. Such TCRs retain specificity for structurally and chemically similar amino acid residues contained within this motif but otherwise tolerate multiple substitutions outside this region (a process termed molecular mimicry). Finally, because most HLA alleles can accommodate related amino acids at primary anchor positions310, multiple peptide sequences can function as agonists for the same TCR.
Alloreactivity occurs when a TCR displays cross-reactivity to HLA molecules that were not encountered during thymic development166. Using a panel of HLA mismatched cell lines that express prevalent HLA alleles but lack expression of the target antigen can establish whether a TCR possesses alloreactivity (Fig. 3b)87. If a TCR does not display alloreactivity, additional methods should be used to assess the specificity of a TCR for targets that express the restricting HLA allele but are bound by alternative self-peptides. This can be accomplished by screening candidate TCRs against a panel of normal cells obtained from HLA-matched healthy donors87,92,153. However, even large panels may fail to include highly specialized or rare cell types. One strategy to enhance representation of cellular subsets from vital organs, such as the heart or kidney, is the use of induced pluripotent stem cells or normal tissue organoids86,167.
A complementary technique to define a TCR’s cross-reactivity potential, termed amino acid positional scanning, begins by establishing which peptide residues form critical contacts with the receptor (Fig. 3c)86,168. In this approach, each amino acid in the cognate peptide is sequentially replaced with alanine, the smallest chiral amino acid. An alternative compact amino acid, such as glycine, can be used in cases in which the native residue is alanine. The recognition motif of a TCR is defined by peptide positions in which an amino acid substitution results in significant loss of function (typically >50%) compared with the native amino acid. The clinical utility of amino acid scanning was demonstrated when it identified an off-target peptide derived from titin, a cardiac structural protein, as the cause of lethal cardiac toxicities resulting from an affinity-enhanced TCR targeting an epitope in the melanoma-associated antigen (MAGE)-A3 protein86. Positional scanning can be extended to include all 20 standard amino acids to measure a TCR’s permissiveness for peptides containing chemically similar residues94. Once a TCR’s recognition motif has been determined, these data are used to perform in silico searches to establish whether homologous motifs exist elsewhere in the human proteome.
Although positional scanning provides useful information on which peptide positions contribute to TCR recognition, the technique can miss important cross-reactivity. For example, some peptide sequences function as potent agonists yet display minimal homology with the native peptide recognized by a TCR169. This can occur as a result of intrapeptide coupling whereby modification to residues outside a peptide’s central core facilitates the generation of new TCR contacts103,170. Combinatorial peptide libraries (CPLs) are highly diverse (often 108 to >1011 variants) and contain peptides in which one amino acid position is sequentially fixed while the remaining positions are substituted with all 20 amino acids. CPLs have shown promise in identifying peptide targets153,171, especially when combined with bioinformatic database screening172. Alternatively, yeast display170, phage display92, and molecularly-barcoded artificial APCs132–135 can enhance the speed and sensitivity of cross reactivity screening. In all instances where potential cross-reactive peptides are identified, additional studies are required to establish physiologic significance.
Measuring potency.
A minimal measure of potency for TCRs being considered for clinical development is the capacity to recognize polypeptides resulting from endogenous processing and presentation of a protein (Fig. 3d–i). For HLA-I-restricted TCRs, this should ideally be complemented with assays that establish whether physiologic expression levels of the full-length protein and restriction element are sufficient for recognition. In addition to cytokine secretion, it is important to also compare the capacity of different TCR candidates to cause tumour cell lysis90. Public databases can identify commercially available tumour cell lines that co-express a target antigen and the restricting HLA allele173. However, it is not always possible to identify established lines that express a p/HLA pair of interest. One solution to overcome this limitation is the creation of an ‘avatar’ that represents a tumour’s HLA-restricted antigenic landscape. This can be accomplished using tandem minigenes (TMGs) or synthetic long peptides containing somatic mutations, gene fusions, insertions/deletions, integrating viruses, or cancer-germline antigens114,174. Transfection of TMGs or pulsing of synthetic long peptides onto autologous APCs enforces antigen processing and presentation by a patient’s complement of HLA molecules. More recently, patient-derived tumour organoids [G] have been used to sample the repertoire of naturally processed peptides displayed on a cancer cell’s surface175,176. Relative to established cancer cell lines, cancer organoids better approximate the genomic heterogeneity, phenotype, and three-dimensional characteristics of human tumours in vivo177. Further, unlike patient-derived xenografts, normal tissue organoids may be established in parallel, providing a complementary approach for assessing off-target effects. Notwithstanding these virtues, organoids are time consuming and expensive to generate. Characterization of TCRs retrieved from CD4+ T cells poses additional challenges because many solid cancers lack steady-state expression of the genes required for HLA class II presentation178. This barrier can be overcome by exogenous expression of class II major histocompatibility complex transactivator (CIITA)179, a master transcriptional regulator for HLA II expression.
Functional avidity, the capacity of a T cell to respond to progressively lower concentrations of cognate peptide, is a commonly used and technically straightforward method to compare different TCRs. In general, functional avidity correlates with the magnitude of tumour cell recognition180–182. However, functional avidity measurements are influenced by multiple factors independent of the intrinsic properties of a TCR, including T cell differentiation183 and activation history184. Moreover, distinct T cell functions (such as cytokine production and cellular proliferation) are trigged by different antigen thresholds185. Alternative assays that assess TCR potency which are quantitative, reproducible, and independent of T cell state are therefore highly desirable.
Binding affinity is the strength with which a single TCR molecule interacts with a single p/HLA complex and is typically represented by the disassociation equilibrium constant (Kd). The binding affinity of naturally occurring TCRs has correlated with antitumour efficacy in preclinical models180,186 and human clinical trials33,34,40,41. Under equilibrium conditions, Kd is defined by the ratio of the rates of dissociation and association (koff/kon). Most thymically-selected TCRs have Kd values in a 1–200 μM range27–29; however, directed evolution techniques can generate TCRs with picomolar binding affinities83,84. TCRs that bind self-antigens often possess Kd values at a higher (weaker) end of the physiologic range, likely because of negative thymic selection. By contrast, TCRs that target mutation-derived neoantigens tend to have lower (stronger) Kd values in a range that overlaps with pathogen-associated receptors148,187–189. Kd values are commonly measured using surface plasmon resonance (SPR), a three-dimensional technique in which a recombinant single-chain soluble TCR is flowed over a sensor chip containing an immobilized p/HLA complex (or vice versa)190. Binding affinity correlates to varying degrees with TCR functional responses90,191. However, this correlation is not universally true. In the extreme, two TCRs can bind the same p/HLA complex with near equivalent Kd values yet display disparate signaling responses (for example, one receptor may trigger T cell activation while the other does not)91. The uncoupling of a TCR’s three dimensional binding affinity from its signaling capacity has in some cases been attributed to the inability of SPR to account for force-dependent interactions, such as catch bond formation90,91. Two dimensional methods directly measure TCR affinity and/or binding kinetics on living T cells, bypassing the need for recombinant TCR expression and purification192. Although two dimensional affinities correlate more closely with biological outcomes192,193, this technique remains relatively low throughput and requires specialized instrumentation.
To enable more rapid throughput, flow cytometry-based approaches have been developed that either infer TCR affinity or directly measure the dissociation kinetics of monomeric TCR-p/HLA complexes on living T cells194–196. The degree to which a T cell depends on the CD8 co-receptor for activation is inversely correlated to a TCR’s affinity and its disassociation half-life from a p/HLA molecule197. Thus, a TCR that binds p/HLA multimers182,197 or triggers antigen-specific effector functions in a co-receptor independent manner148,182 qualitatively implies the receptor is relatively high-affinity. A quantitative flow cytometry-based measurement is structural avidity, the capacity of an individual membrane-associated TCR to bind monomeric p/HLA molecules. Unlike functional avidity, structural avidity measurements are agnostic of T cell differentiation state and therefore demonstrate a high degree of concordance between T cell clones and TCR transduced T cells198. Methodologically, structural avidity measures the TCR-p/HLA dissociation rate (koff) using p/HLA multimers that dissociate into monomers following addition of an inert chemical compound. Adoptive cell transfer (ACT) of T cells expressing TCRs with slower koff-rates provide superior antitumour efficacy compared with TCRs with faster koff-rates198–200.
Clinical efficacy of TCR-based cancer immunotherapies.
TIL.
Because TILs are frequently enriched in tumour-reactive TCR clonotypes, they can be used as a source of therapeutic T cells for non-receptor engineered ACT. TIL therapies are generated through the surgical resection of a metastatic tumour followed by ex vivo T cell expansion to achieve treatment numbers (up to 1011 cells). T cells can either be expanded in bulk or following selection based on evidence of tumour reactivity. TILs are typically administered following chemotherapy pre-conditioning to deplete bystander lymphocytes that function as ‘sinks’ for homeostatic cytokines, such as IL-15, and remodel the immunosuppressive tumor microenvironment201–203. TIL infusion is often followed by a multi-day course of the common gamma chain cytokine [G] IL-2 to promote T cell engraftment204. Melanoma111,121,205,206 and cancers of the bladder207, breast208, cervix112,209, gastrointestinal (GI) tract115, head and neck209, kidney210, lung113,116,211, and ovary136,206 contain TILs with HLA-dependent reactivity to cancer antigens. For many of these cancers, TIL ACT caused objective tumour regression in early phase clinical trials113,174,205,208,209,212, a registration enabling single-arm phase II trial213, and a randomized phase III trial214. These findings establish proof of principle that TCR-based therapies can mediate cancer regression in a broad range of human malignancies.
The largest clinical experience with TIL comes from patients with metastatic cutaneous melanoma215. In patients who have not received prior treatment with anti-PD-1/PD-L1 immune checkpoint inhibitors (ICIs), both the overall and complete response rates (ORR and CR, respectively) are relatively high (ORR: 40–62%; CR: 7–24%)212,214,216. Importantly, ~96% of patients with a CR to TIL therapy do not have disease recurrence217. Response to TILs in melanoma can occur following progression on other therapies, including ICIs 213,214,217. However, whereas prior anti-CTLA-4 does not appear to impact TIL efficacy217,218, response rates often are lower following progression on antibodies targeting the PD1/PD-L1 axis213,217,219. This suggests that TIL and anti-PD1/PD-L1 therapies have partially overlapping mechanisms of response and resistance.
Consistent with this hypothesis, meta-analyses of >1000 patients treated with ICIs established a significant association between tumour mutational burden (TMB) and the likelihood of ICI response138,220. In parallel, TIL studies have similarly discovered correlations between response rate and TMB219,221 or response and the frequency of neoantigen-reactive T cells in the infusion product222. These data suggest that the success of TIL and ICI therapies is likely dependent on the abundance of neoantigen-reactive T cells. Consequently, next-generation TIL approaches in which neoantigen-reactive T cells are selectively expanded and/or enriched might allow for more consistent tumour control.
Beyond cutaneous melanoma, TIL therapies have clinical activity in uveal melanoma [G] 205 and several common epithelial malignancies174,208,209. Unlike cutaneous melanoma, uveal melanomas are modestly mutated and largely refractory to ICIs223. In a phase II clinical trial, ACT of uveal melanoma TILs caused cancer regression in seven out of 20 patients, including one CR (ORR: 35%; CR: 5%)205. Among responding patients, three had previously progressed on ICIs. In a post hoc analysis, responding patients received significantly higher numbers of tumour-reactive T cells compared with non-responders. In a second study involving 16 patients with non-small cell lung cancer who received TIL after tumour progression on an anti-PD1 antibody, three had evidence of cancer regression including two durable CRs (ORR: 19%; CR: 13%) ongoing 1.5 years later113. Responders in this study were significantly more likely to have received TIL containing neoantigen and cancer germline-reactive T cells compared with non-responders. Similar results were observed in a third trial that tested TIL therapy in human papillomavirus (HPV)-associated cancers. Among 18 patients with HPV+ cervical cancer who received TILs, five had objective responses including two durable CRs (ORR: 28%; CR: 11%) ongoing 4 years later209. Immune-monitoring studies revealed that the infusion products of the two patients with CRs contained T cells specific for neoantigens, cancer germline antigens, and peptides derived from the HPV E6/E7 oncoproteins112. In a cohort of patients with HPV+ oropharyngeal, anal, and vaginal cancers, two out of 11 subjects had a partial response (ORR: 18%; CR: 0%). Across both cohorts, the frequency of HPV E6/E7-reactive T cells in the infusion product and the persistence of tumour-reactive T cells in the peripheral blood correlated with the likelihood of response.
Finally, case reports have documented responses to TIL ACT in modestly mutated epithelial malignancies. For example, ACT of neoantigen-selected TILs resulted in tumour regression in two patients with gastrointestinal malignancies associated with DNA mismatch repair proficiency [G]174,224. One patient with cholangiocarcinoma, an aggressive bile duct cancer, had a prolonged PR following infusion of a near clonal CD4+ TIL population that recognized a patient-specific private neoantigen174. Of note, this patient failed to respond to an unselected TIL population before receiving >10-fold higher dose of neoantigen-selected T cells. A second patient with metastatic colorectal cancer also had a partial response following infusion of neoantigen-selected CD8+ T cells224. In this case, ~75% of transferred T cells recognized a shared, or public225, neopeptide derived from a recurrent KRASG12D hotspot mutation. All metastases regressed in this patient except for a single lung lesion. Analysis of this escape lesion revealed loss of heterozygosity (LOH) for the HLA allele that presents the KRASG12D-derived peptide, establishing a mechanism of targeted immune escape. Another patient in this study received TILs with reactivity to the same KRASG12D public neoantigen with no response. In this case, only 0.002% of the infusion product was neopeptide-specific. Finally, three patients with metastatic breast cancer experienced objective tumour regression following neoantigen-selected TIL therapy208. In one patient who achieved a durable CR, ~23% of T cells in the infusion product targeted neoepitopes resulting from four somatically mutated genes that were restricted by HLA-I and HLA-II alleles. The other two patients had PRs lasting 6 and 10 months, respectively. Together, these examples provide further evidence that the infusion of neoantigen-reactive T cells can trigger tumour regression in humans.
T cell clones.
Although TILs are relatively enriched in tumour-reactive T cells, they typically contain large numbers of bystander T cells that do not contribute to antitumour immunity111,116,117,119. One strategy to enrich for a homogenous cell product is ACT of expanded T cell clones of a single, well-defined specificity. T cell clones have been generated from TILs and the circulating repertoire of cancer patients using IVS alone226, IVS followed by p/HLA sorting227, or IVS followed by limited dilution cloning228–232. Initial clinical experience of ACT with T cell clones focused on the targeting of non-mutated tissue differentiation antigens. Most early T cell clone clinical trials targeted epitopes derived from the shared melanocyte/melanoma differentiation antigens MART-1 [G], gp100, and tyrosinase228,232,233. Overall, patients experienced limited treatment-related toxicities in these studies, with most side effects attributable to expected on-target/off-tumour destruction of normal melanocytes. However, clinical activity has generally been modest (ORR <10%).
Another important antigen class are the cancer-germline antigens (CGAs), a family of >100 immunogenic intracellular proteins whose normal tissue expression is typically, although not universally, restricted to germ cells and fetal tissues234. Because germ cells lack HLA expression165, they are immune privileged and impervious to T cell-mediated attack. Consequently, targeting CGAs can afford a wider therapeutic index compared with tissue differentiation antigens. Many CGAs are epigenetically silenced in somatic tissues through promoter methylation235. Epigenetic dysregulation during tumourigenesis can lead to de-repression of CGA genetic loci, resulting in expression of a cancer-selective target. In a case study, a patient with refractory metastatic melanoma who received an HLA-II restricted CD4+ T cell clone targeting the CGA New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1) experienced long-term and complete tumour regression230. Similar to findings using TILs, a correlation exists between the in vivo persistence of transferred T cell clones and the likelihood of clinical response236. The limited clinical efficacy of T cell clones observed across most trials is likely attributable to the extended clone manufacturing process resulting in enhanced cellular differentiation and poor in vivo persistence237. Strategies that enhance cellular fitness by altering cytokines during T cell priming227, selection of clones with memory-like attributes238,239, and optimize co-stimulation during expansion226 may improve outcomes.
TCR-engineered T cells.
TCR gene transfer streamlines many practical challenges associated with TIL and T cell clone therapies. These benefits include: 1) a minimally invasive procedure to procure autologous T cells (such as leukapheresis [G]); 2) a high probability of developing potent cell products in a relatively short time; 3) the opportunity to pre-select TCRs with optimal potency and off-target profiles; 4) the capacity to introduce TCRs into minimally-differentiated T cell populations with superior engraftment and proliferative potential240–242; and 5) the opportunity to concurrently introduce genetic manipulations that enhance T cell function through augmented T cell survival243–245, resistance to inhibitory ligands45,245,246, or enhanced antigen-driven signaling through the TCR complex247–250. As summarized below, TCR clinical trials have targeted diverse classes of antigens and, in many cases, distinct epitopes derived from the same antigen (Table 1; Supplementary Table 1).
Table 1:
Summary of selected TCR gene therapy clinical trials.
| Antigen class | HLA | Co-receptor independent | CDR modification | Tumour | ORR* | Reference |
|---|---|---|---|---|---|---|
| Tissue differentiation | ||||||
| MART-1 | A*02:01 | No | No | MEL | 13% | 33 |
| MART-1 | A*02:01 | Yes | No | MEL | 30% | 34 |
| gp100 | A*02:01 | Yes | No | MEL | 19% | 34 |
| Tyrosinase | A*02:01 | Yes | No | MEL | 33% | 299 |
| CEA | A*02:01 | Yes | Yes | CRC | 33% | 35 |
| Cancer germline | ||||||
| NY-ESO-1 | A*02:01 | Yes | Yes | MEL | 55% | 258 |
| NY-ESO-1 | A*02:01 | Yes | Yes | SS | 61% | 258 |
| NY-ESO-1 | A*02:01 | Yes | Yes | SS | 50% | 43 |
| NY-ESO-1 | A*02:01 | Yes | Yes | MRCLS | 40% | 259 |
| MAGE-A3/9/12 | A*02:01 | Yes | Yes | various | 56% | 36 |
| MAGE-A3/6 | DPB1*04:01 | Yes | No | various | 24% | 38 |
| MAGE-A4 | A*24:01 | No | No | ESCA | 0% | 37 |
| MAGE-A4 | A*02:01 | Yes | Yes | SS and MRCLS | 36% | 262 |
| MAGE-A4 | A*02:01 | Yes | Yes | various | 24% | 263 |
| MAGE-A4 + CD8α | A*02:01 | Yes | Yes | various | 36% | 264 |
| MAGE-A10 | A*02:01 | ND | Yes | NSCLC | 11% | 266 |
| PRAME | A*02:01 | No | No | various | 50% | 268 |
| Overexpressed | ||||||
| WT1 | A*24:02 | No | No | MDS/AML | 0% | 39 |
| WT1 | A*02:01 | Yes | No | AML | NED | 44 |
| Viral | ||||||
| HPV16 E6 | A*02:01 | Yes | No | HPV16+ | 17% | 40 |
| HPV16 E7 | A*02:01 | Yes | No | HPV16+ | 50% | 41 |
| Neoantigen | ||||||
| Private | various | various | No | various | 0% | 47 |
| TP53 (R175H) | A*02:01 | Yes | No | BRCA | 1/1 | 275 |
| KRAS (G12D) | C*08:02 | Yes | No | PDAC | 1/2 | 274 |
| KRAS (G12D) | A*11:01 | Yes | No | NSCLC | 1/1 | 300 |
Abbreviations: AML, acute myeloid leukemia; BRCA, breast cancer; CRC, colorectal cancer; ESCA, oesophageal carcinoma; HPV, human papilloma virus; MDS, myelodysplastic syndrome; Mel, melanoma; MRCLS, myxoid/round cell liposarcoma; ND, not defined; NED, no evaluable disease; NSCLC, non-small cell lung cancer; ORR, overall response rate; PDAC, pancreatic ductal adenocarcinoma; SS, synovial cell sarcoma.
ORR reported if ≥3 patients treated.
Tissue differentiation antigens.
As in studies using T cell clones, initial TCR gene therapy studies targeted non-mutated tissue differentiation antigens. The first published human TCR clinical trial tested an HLA-A*02:01-restricted receptor specific for a MART-1 peptide that was cloned from melanoma TILs and transduced into an allogeneic T cell line251. The first two TCR clinical trials using autologous T cells sequentially tested a pair of receptors (DMF4 and DMF5) specific for the same MART-1 peptide but cloned from the TILs of a separate patient. In the first autologous trial, out of 31 patients with melanoma who received T cells retrovirally transduced with the CD8 co-receptor dependent DMF4 TCR, four achieved a PR (ORR: 13%)33,34. No patients developed toxicities related to TCR-modified cells. In a second trial, patients received T cells modified with the DMF5 receptor, a CD8 co-receptor independent TCR with an affinity ~5-fold higher than DMF4252. Among 20 patients who received the DMF5 TCR, six had an objective tumour response (ORR: 30%)34. However, a significant proportion developed on-target toxicities related to destruction of MART-1 expressing melanocytes present in the skin, eye, and inner ear. Antitumour activity and on-target toxicities were observed in a third trial testing a CD8 co-receptor independent TCR targeting an HLA-A*02:01-restricted epitope derived from the melanocyte-associated protein gp10034. Unlike DMF4 and DMF5, this receptor was generated in an HLA-A*02:01 Tg mouse. Among 16 treated patients, three had responses (ORR: 19%; CR: 6%). Significant on-target toxicities related to melanocyte targeting was also observed in this study. The liability of targeting tissue differentiation antigens was established outside the context of melanoma in a fourth trial targeting a peptide derived from the gastrointestinal lineage marker carcinoembryonic antigen (CEA)35. Three patients with colorectal cancer received T cells engineered with an affinity-enhanced TCR generated in an HLA Tg mouse. Following T cell infusion, all patients had significant reductions in serum CEA, indicating on-target engagement, and one had a PR (ORR: 33%). However, all patients developed severe inflammatory colitis resulting from T cell mediated destruction of CEA+ colonic epithelial cells. Collectively, these early clinical trials established that TCR gene therapy can trigger cancer regression but simultaneously highlighted the critical importance of target selection and receptor affinity.
Overexpressed, non-mutated antigens.
Non-mutated epitopes derived from transcriptional regulators, including Wilms’ tumour 1 (WT1) and P53, are frequently overexpressed by haematologic and solid malignancies253–255. Although not cancer-specific167, expression of these proteins can differ by as much as a 1000-fold between normal and transformed tissues, providing a potential therapeutic window. Several HLA-I restricted WT1 epitopes have been targeted using TCRs cloned from healthy donors with no evidence of normal tissue toxicity39,44. In one study which tested a CD8 co-receptor dependent TCR in patients with leukaemia39, no objective antitumour responses were observed. Two CD8 co-receptor independent WT1 TCRs44,63 have entered therapeutic trials for liquid and solid cancers. In one study, the receptor is integrated into the TRAC locus followed by disruption of the TRBC1/2 loci using CRISPR editing (NCT05066165)63. Efficacy results for these studies are forthcoming. A CD8 co-receptor independent TCR targeting a wild-type P53 epitope generated in an HLA Tg mouse256 was tested in an early phase clinical trial. Although allogeneic T cells transduced with this TCR demonstrated reactivity to a diverse range of cancers in vitro, autologous T cells expressing this receptor could not be efficiently expanded257. This finding correlates with increased wild-type P53 expression by activated T cells and TCR-dependent fratricide. These data indicate that differences in wild type P53 expression by normal cells compared with cancer cells is likely insufficient to permit safe immunologic targeting.
Cancer germline antigens.
The first CGA TCR trials targeted an HLA-A*02:01-restricted epitope derived from NY-ESO-1 using an affinity-enhanced, CD8 co-receptor independent receptor cloned from a patient with melanoma82. An initial trial in patients with melanoma reported a >50% overall response rate (ORR: 55%; CR: 20%) without off-tumour toxicities258. This response rate is comparable to that observed in a contemporaneous group of patients with melanoma treated with TILs212, suggesting that single epitope targeting may be comparable to approaches targeting multiple antigens. Antitumour efficacy using this TCR has also been observed in synovial cell sarcoma and myxoid/round cell liposarcoma (ORR: 40–61%; CR: 0–8%)43,258,259, two mesenchymal cancers that respond poorly to other immunotherapies.
The MAGE-A family of CGAs is comprised of 12 genes260, several of which have been targeted in TCR trials. The first two MAGE TCR trials targeted the A3 isoform using HLA-I restricted receptors. One trial used an affinity-enhanced, CD8 co-receptor independent TCR generated in an HLA-A*02:01 Tg mouse261. Objective responses occurred in 5 out of 9 patients (ORR: 56%; CR: 11%) treated as part of a dose-escalation study36. However, 3 out of 5 patients who received the highest T cell dose developed severe neurologic toxicities resulting in two treatment-related deaths. These toxicities resulted from the off-tumour destruction of a previously unappreciated neuronal population expressing MAGE-A12, a protein with >95% homology to MAGE-A3. At the same time, a second trial targeting MAGE-A3 reported lethal toxicities using an affinity-enhanced TCR restricted by HLA-A*01:0142,86. Two patients in this study developed cardiogenic shock following T cell infusion resulting from the off-tumour/off-target recognition of an epitope derived from titin. In this case, affinity-enhancement altered the native receptor’s specificity in a manner that was initially not detected in preclinical studies using a diverse panel of normal cells that express the restricting HLA allele. A third trial also targeted a MAGE-A3 epitope38. Unlike the other studies, the patient-derived TCR used in this trial was restricted by HLA-II and did not undergo affinity-enhancement. As part of the manufacturing process, CD4+ T cells were isolated to test the safety and antitumour efficacy of an HLA-II-restricted TCR in a physiologic context. Among 17 treated patients, none developed off-tumour toxicities and four had objective tumour responses (ORR: 24%; CR: 6%). Interestingly, many patients experienced prolonged fevers following treatment, a finding that may be attributable to enhanced cytokine secretion by CD4+ T cells.
Both MAGE-A4 and MAGE-A10 have also been targeted in TCR gene therapy trials. An initial trial targeting MAGE-A4 using an HLA-A*24:02-restricted, CD8 co-receptor dependent, non-affinity enhanced receptor failed to demonstrate antitumour activity37. By contrast, an affinity-enhanced TCR targeting this CGA showed efficacy in soft tissue sarcomas262 but limited activity in other solid cancers263. A multi-cistronic vector that co-expresses CD8α and the TCR seems to improve responses in common epithelial malignancies (ORR: 36%; CR: 5%), including ovarian, head and neck, and gastroesophageal cancers264. Several trials have targeted MAGE-A10 with an affinity-enhanced HLA-A*02:01-restricted TCR94. To date, there has been no evidence of off-tumour toxicities with this receptor; however, there has been minimal evidence of clinical activity265,266.
Despite its name, preferentially expressed antigen in melanoma (PRAME) is a CGA that is frequently expressed by both melanoma and non-melanoma cancers267. A dose-escalation trial using a naturally occurring TCR targeting an HLA-A*02:01-restricted PRAME epitope recently reported responses in multiple solid cancers (ORR: 50%; CR: 0%) without evidence of off-target toxicities268. A TCR targeting an alternative HLA-A*02:01-restricted PRAME epitope has entered clinical studies in patients with haematologic cancers (NCT03503968).
Viral oncoproteins.
Viral proteins are immunologically foreign, not expressed by normal tissues, and in certain cases directly contribute to malignant transformation269. Therefore, they represent an attractive source of shared cancer-specific epitopes. Several clinical trials have tested the safety and antitumour activity of TCRs targeting virally encoded oncoproteins.
An association between TCR avidity and clinical activity was highlighted in two clinical trials targeting HLA-A*02:01 restricted epitopes derived from the human papillomavirus (HPV)-16 E6 and E7 oncoproteins. In one phase I/II study, patients with HPV-16-associated cancers received T cells transduced with a TCR that binds an epitope derived from the E6 oncoprotein40. The patient-derived TCR used in this study functions in a CD8 co-receptor independent manner and contains no modifications to its variable domains. Among the twelve patients treated, none had evidence of off-target toxicities and two had cancer shrinkage (ORR: 17%). Analysis of post-treatment tumour samples from non-responding patients identified one with HLA-A*02:01 LOH and a second with a frameshift mutation in IFGNR1, a critical gene involved in antigen presentation. By contrast, no mutations in antigen processing and presentation genes were observed in a responding patient.
In a second study, patients received T cells transduced with a patient-derived TCR targeting an epitope derived from the HPV-16 E7 oncoprotein41. Like the E6 TCR, the E7 TCR functions in a CD8 co-receptor independent manner and contains unaltered variable domains. However, the E7 TCR has a significantly slower Koff rate compared with the E6 TCR199, indicating higher structural avidity. Among 12 patients who received E7 TCR-modified T cells, six had PRs (ORR: 50%), including four who had progressed on prior anti-PD1 therapies. No patients experienced off-target toxicities. Four patients had tumours with loss of function mutations in interferon-signaling, HLA-I presentation, or HLA-A*02:01 expression. When considered together with results from the E6 TCR trial, these data indicate that immune-editing occurs frequently in HPV-associated cancers.
Merkel cell carcinoma is a rare and aggressive form of skin cancer that in ~80% of cases is caused by the transforming Merkel cell polyomavirus (MCPyV)270. Multiple epitopes derived from MCPyV-encoded oncoproteins drive T cell responses in patients271, including an HLA-A*02:01 restricted epitope272. A patient-derived, CD8 co-receptor independent TCR targeting the HLA-A*02:01 epitope is now being tested in a phase I/II clinical trial273.
Private neoantigens.
A recent clinical trial tested a highly personalized cell therapy approach that introduced private neoantigen-reactive TCRαβ sequences into polyclonal T cells using non-viral CRISPR/Cas9 genome editing47. In this study, tumours from each patient underwent whole exome sequencing and RNA-seq to define its unique expressed mutational landscape. Then, a curated list of in silico predicted HLA-I neoantigens was selected for incorporation into a custom p/HLA capture library. Gene sequences for TCRs that bound to these p/HLA complexes were retrieved from circulating T cells and the functionality of these TCR was tested for the capacity to trigger antigen-specific cytokine release. Patients received autologous T cells modified with up to three unique TCRs. The exogenous TCRs were inserted into the TRAC locus by HDR following CRISPR editing of TRAC/TRBC to remove the endogenous TCR. Immune monitoring revealed that TCR edited cells engrafted in the peripheral blood and trafficked to metastatic tumour sites. Among 16 treated patients, five demonstrated stable disease and 11 had progressive disease. Although no objective responses were observed, this study established the feasibility and safety of targeting multiple neoantigens simultaneously using a fully non-viral TCR genome editing approach.
Public neoantigens.
Two proof of principle clinical trials have recently established the therapeutic potential of targeting public neoantigens resulting from recurrently mutated driver genes using TCR gene therapy. In one trial, patients were co-infused with T cells individually transduced with two HLA-C*08:02-restricted TCRs specific for a 9mer or 10mer peptide resulting from the KRASG12D hotspot mutation274. Both TCRs were cloned from a patient with KRASG12D colorectal cancer who had experienced an objective response following neoantigen-selected TIL therapy224. One patient with pancreatic cancer achieved a durable PR lasting >6 months following infusion of TCR engineered T cells in the absence of toxicities attributable to the cell product. A second patient with pancreatic cancer who also received TCR engineered T cells developed cytokine release syndrome (CRS) and did not have tumour regression. In a second trial, a patient with treatment-refractory breast cancer received T cells transduced with an HLA-A*02:01-restricted TCR specific for an epitope resulting from a hotspot mutation (R175H) in the tumour suppressor gene TP53275. The TCR, which functions in a CD8 co-receptor independent manner, was cloned from an HLA-A*02:01+ patient with metastatic colorectal cancer who received neoantigen-selected TIL276. Immediately following T cell infusion, the patient developed CRS; however, these symptoms promptly resolved with administration of intravenous steroids. The patient then achieved an objective PR that lasted six months. Genomic sequencing of a new metastatic tumour site revealed LOH for HLA-A*02:01 as a likely resistance mechanism.
Soluble bispecific TCRs and TCR mimics.
Multiple ImmTACs and TCR-mimics have entered human clinical trials (Table 2). Tebentafusp, an ImmTAC that targets an HLA-A*02:01-restricted gp100 epitope, recently received FDA approval for patients with unresectable or metastatic uveal melanoma18. ImmTACs targeting additional HLA-A*02:01-restricted epitopes resulting from NY-ESO-1, MAGE-A4, MAGE-A8, and PRAME are now in early clinical development. In phase I-III trials, the overall response rate following tebentafusp administration has been modest (4.7–9.1%)18,277,278. Nevertheless, in a randomized phase III trial, patients who received tebentafusp had a significant overall survival benefit compared with patients in the control arm18. The fact that conventional response criteria did not strongly correlate with overall survival is reminiscent of other immunotherapies279,280 and suggests that tebentafusp might alter tumour growth kinetics. If the uncoupling of radiographic responses from survival benefit is a class effect, the clinical development of soluble bispecific TCRs may be relatively prolonged and expensive. Unlike T cell-based therapies that have received FDA approval in the relapsed/refractory setting on the basis of high overall response rates in single-arm phase II trials281–287, a soluble bispecific TCR registration trial would require a randomized design. Overall, the toxicity profile of ImmTACs seems similar to other bispecific T cell engaging proteins. This includes a reversible and generally mild CRS that abates after the first few doses18,278. Expected on-target/off-tumour cutaneous toxicities, such as rash and vitiligo, have been observed with tebentafusp. Importantly, no significant neurotoxicity has been observed in patients receiving ImmTACs thus far.
Table 2:
Summary of soluble bispecific TCR and TCR-mimic clinical trials.
| Antigen | HLA | TCR Vs. TCR-mimic | Disease | ORR | Reference or Trial identifier |
|---|---|---|---|---|---|
| Tissue differentiation | |||||
| gp100 | A*02:01 | TCR | MEL | 8.7% | 277 |
| gp100 | A*02:01 | TCR | Uveal MEL | 4.7% | 278 |
| gp100 | A*02:01 | TCR | Uveal MEL | 9.1% | 18 |
| Cancer germline | |||||
| NY-ESO-1 | A*02:01 | TCR | Solid cancers | n.d. | NCT03515551 |
| PRAME | A*02:01 | TCR | Solid cancers | n.d. | NCT04262466 |
| MAGE-A4 | A*02:01 | TCR | Solid cancers | n.d. | NCT03973333 |
| MAGEA4/8 | A*02:01 | TCR | Solid cancers | n.d. | NCT05359445 |
| MAGE-A4 | A*02:01 | TCR-mimic | Solid cancers | n.d. | NCT05129280 |
| Overexpressed | |||||
| WT-1 | A*02:01 | TCR-mimic | AML | n.d. | NCT04580121 |
Abbreviations: AML = acute myeloid leukemia; Mel, melanoma; n.d., no data.
Conclusions and future perspective.
TCR-based therapies represent a new class of precision oncology treatments. Through a unique mechanism of action, TCRs enable the intracellular proteome to become a source of actionable cancer-selective and cancer-specific immunologic targets. Simultaneously, TCRs pose new challenges compared with conventional molecularly targeted therapies because they require co-expression of two biomarkers: a target protein and a specific HLA molecule. Because the HLA locus is the most polymorphic region of the human genome288, identifying patients who express both biomarkers can be inefficient when performed in an unguided fashion. Prospective clinical next-generation sequencing (NGS) has revolutionized the ability to match approved and investigational treatments to specific molecular abnormalities identified in a patient’s tumour289. Many widely used clinical NGS platforms capture sequencing reads that permit inference of a patient’s HLA haplotype290,291. However, these data are currently not systematically reported in a patient’s medical record, partly because of a perception that they do not represent actionable information. The FDA’s recent approval of tebentafusp, which requires confirmation of HLA-A*02:0118, challenges this notion and provides a rationale to begin routinely ascertaining every cancer patient’s HLA haplotype.
Several drugs targeting specific genomic alterations have recently gained approval regardless of the site of disease origin. This new paradigm contrasts with the historical norm of developing cancer therapies in a tissue-specific but genome agnostic manner. Examples of tissue-agnostic therapies include the NTRK gene fusion inhibitors292,293 and ICIs for malignancies associated with a high TMB294,295. Single-arm studies that lack a conventional control arm have recently been used in support of tissue-agnostic small molecule and antibody drug approvals. This study design is defensible in light of high response rates and acceptable toxicities and has dramatically accelerated the timelines for developing new treatments. A similar approach might be applied to TCR-based therapies, particularly those that target public neoantigens or viral oncoproteins, in which target antigen expression can be determined through NGS. With one notable exception47, nearly all genetically engineered TCR, bispecific TCR, and TCR-mimic clinical trials have targeted only a single p/HLA complex. Concurrent targeting of multiple epitopes restricted by distinct HLA alleles might be a future strategy to minimize therapeutic resistance resulting from antigen heterogeneity or HLA LOH.
Thus far, most TCR-engineered cell therapies have used autologous αβ T cells. Although autologous T cells have a track record of antitumour efficacy and avoid the risk of GVHD, their use poses challenges to scalability and cost efficiency296. Third party sources of T cells, including allogeneic viral-specific T cells44, allogeneic TRAC-edited naturally occurring or induced pluripotent T cells297, γδ T cells23, and CD3 complex engineered-NK cells298 can each generate ‘off-the-shelf’ products that support TCR signaling. Whether allogeneic TCR-modified lymphocytes will match the potency of autologous T cells, particularly in patients with solid malignancies, remains unknown and will be an important area of ongoing clinical research.
Supplementary Material
Acknowledgements:
This study was supported, in part, by National Institutes of Health (NIH) grants R37 CA259177 (C.A.K.), R01 CA269733 (C.A.K.), R01 CA286507 (C.A.K.), R35 GM118166 (B.M.B.), R35 CA197633 (A.R.), P01 CA168585 (A.R.), P30 CA008748 (C.A.K.), and the NIH SPORE in Soft Tissue Sarcoma P50 CA217694 (C.A.K.); the Parker Institute for Cancer Immunotherapy (C.A.K. and A.R.); the Cancer Research Institute CRI3176 (C.A.K.); The Sarcoma Center at MSKCC (C.A.K.); the Mr. William H. Goodwin and Mrs. Alice Goodwin and the Commonwealth Foundation for Cancer Research (C.A.K and S.S.C.); and The Center for Experimental Therapeutics at Memorial Sloan Kettering Cancer Center (C.A.K and S.S.C.). S.A.Q. is funded by a CRUK Senior Cancer Research Fellowship (C36463/A22246) and a CRUK Biotherapeutic Programme Grant (C36463/A20764).
Glossary:
- Human leukocyte antigen (HLA)
A family of highly polymorphic, germline encoded transmembrane proteins that bind proteolytically degraded polypeptides. In vertebrate species, related proteins are referred to as the major histocompatibility complex (MHC)
- HLA-restricted tumour antigens
Cancer-specific and cancer-associated polypeptides resulting from proteasomal, endosomal, or lysosomal protein degradation. These polypeptides are bound non-covalently within the binding groove of an HLA class I or class II molecule and facilitate the activation of antigen-specific T cells.
- γδ T cells
~1–5% of circulating T cells express a somatically recombined γδ TCR that associates with the CD3 subunits and mediates antigen-specific cellular immunity. γδ T cells recognize a limited number of ligands presented in the context of non-polymorphic antigen presentation molecules.
- Immune receptor tyrosine-based activation motif (ITAM)
A conserved four amino acid sequence (YxxL/I) contained in the cytoplasmic tails of non-catalytic tyrosine-phosphorylated receptors found in immune cells.
- Somatic recombination
The genes for the V, D, J, and C segments of the TCRα and TCRβ hemichains do not encode functional proteins in their germline configuration. Rather, each segment undergoes site-specific recombination with the aid of recombination activation genes to assemble a single functional frame.
- Sleeping Beauty transposon/transposase system
A gene therapy method that uses co-transfer of two DNA plasmids to achieve stable transgene genomic integration and expression. One plasmid transiently expresses a transposase enzyme that digests the second plasmid, the Sleeping Beauty transposon, at inverted/direct repeats and ligates the transposon cassette containing a gene of interest into TA dinucleotide repeats within the genome.
- Catch bond
A property of many low-affinity cell surface adhesion systems, including selectins, integrins, adhesins, and TCRs in which a bond’s likelihood of separating is reduced as tensile force is applied. This property contrasts with the more common slip bond in which a bond disassociates rapidly following application of sheer force.
- Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq)
A sequencing-based method that enables the simultaneous detection and quantification of cell surface proteins, immune receptor binding specificity, and transcriptomic data at single-cell resolution.
- DNA-barcoded p/HLA multimers
A synthetic p/HLA molecule conjugated to a unique oligonucleotide sequence that is detected and quantified using next-generation sequencing methods. This reagent enables the parallel detection of >1,000 T-cell specificities in a single sample.
- Strep-tagII sequence
A short polypeptide that binds with intermediate affinity to the biotin binding site of a mutated form of streptavidin. In the presence of excess d-biotin, Strep-tagII multimers dissociate into monomers. When p/HLA molecules are conjugated to Strep-tagII, this reagent facilitates the capture, enrichment, and release of antigen-specific T cells.
- Paramagnetic artificial APC
An iron-dextran nanoparticle conjugated to a synthetic p/HLA complex and an anti-CD28 co-stimulatory antibody. In the presence of a magnetic column, this reagent simultaneously enriches for rare antigen-specific T cells and induces T-cell proliferation.
- Investigational New Drug application (IND)
A request from a clinical study sponsor to obtain authorization from the FDA to administer an investigational drug or biological product to humans. An IND application is comprised of pre-clinical data establishing whether the product is reasonably safe and can be manufactured consistently alongside a protocol for the study’s conduct that ensures patients are not exposed to unnecessary risks.
- Tumour organoids
Multicellular in vitro structures that preserve the genetic diversity, phenotype, and structural features of a tumor in vivo. In vitro responses of tumour organoids to different treatments, including immunotherapies, often correlates with patient responses.
- Common gamma chain cytokines
a family of six cytokines that share the common gamma chain (γc, CD132) as part of a receptor complex. Members of this cytokine family include IL-2, IL-7, IL-9, IL-15, and IL-21.
- Uveal melanoma
a rare malignancy that arises from melanocytes within the uveal tract of the eye, which includes the iris, ciliary body, and choroid. Unlike cutaneous melanoma, uveal melanomas are modestly mutated and generally respond poorly to immune checkpoint blockade.
- DNA mismatch repair proficiency (pMMR)
The vast majority (~95%) of colorectal cancers and other GI malignancies are proficient in DNA mismatch repair enzymatic function. These cancers are associated with a modest tumour mutational burden and are largely unresponsive to immune checkpoint inhibitors.
- Leukapheresis
An outpatient procedure to obtain large numbers of circulating T cells, B cells, and monocytes for downstream clinical applications, including genetic engineering and in vitro stimulation. In this procedure, mononuclear cells are separated from red blood cells, platelets, and plasma through differential centrifugation.
- Melanoma antigen recognized by T cells 1 (MART-1; Melan-A)
A transmembrane protein associated with normal melanocytes and the majority of melanomas.
Footnotes
Competing interests:
C.A.K. and S.S.C. are inventors on patents related to TCR discovery and public neoantigen-specific TCRs and are recipients of licensing revenue shared according to MSKCC institutional policies. C.A.K. has consulted for or is on the scientific advisory boards for Achilles Therapeutics, Affini-T Therapeutics, Aleta BioTherapeutics, Bellicum Pharmaceuticals, Bristol Myers Squibb, Catamaran Bio, Cell Design Labs, Decheng Capital, G1 Therapeutics, Klus Pharma, Obsidian Therapeutics, PACT Pharma, Roche/Genentech and T-knife. C.A.K. is a scientific co-founder and equity holder in Affini-T Therapeutics. B.M.B. is an inventor on patents related to TCR engineering and neoantigen discovery, has consulted for Eureka Therapeutics and EnaraBio, and is on the scientific advisory board of T-cure Bioscience. S.A.Q is an inventor in patents related to the use of T cell therapies targeting tumour clonal neoantigens in cancer. S.A.Q. is also a founder, CSO, and equity holder of Achilles Therapeutics. A.R. reports personal fees from Amgen, Chugai, Genentech, Merck, Novartis, Roche, Sanofi, Vedanta, 4C Biomed, Appia, Apricity, Arcus, Highlight, Compugen, ImaginAb, Kalthera/ImmPACT Bio, MapKure, Merus, Rgenix, Lutris, Nextech, PACT Pharma, Synthekine, Tango, Advaxis, CytomX, Five Prime, RAPT, Isoplexis, and Kite/Gilead and has received grants from Agilent and Bristol Myers Squibb outside the submitted work.
Reference list:
- 1. Hedrick SM, Cohen DI, Nielsen EA & Davis MM Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature 308, 149–153 (1984). Together with reference 2, these papers report the genetic sequence for the TCRβ chain in mice and humans for the first time.
- 2.Yanagi Y, et al. A human T cell-specific cDNA clone encodes a protein having extensive homology to immunoglobulin chains. Nature 308, 145–149 (1984). [DOI] [PubMed] [Google Scholar]
- 3.Rosenberg SA IL-2: the first effective immunotherapy for human cancer. Journal of immunology 192, 5451–5458 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ribas A & Wolchok JD Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Guedan S, Ruella M & June CH Emerging Cellular Therapies for Cancer. Annu Rev Immunol 37, 145–171 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chandran SS & Klebanoff CA T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance. Immunol Rev 290, 127–147 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Thul PJ, et al. A subcellular map of the human proteome. Science 356(2017). [DOI] [PubMed] [Google Scholar]
- 8.Rossjohn J, et al. T cell antigen receptor recognition of antigen-presenting molecules. Annu Rev Immunol 33, 169–200 (2015). [DOI] [PubMed] [Google Scholar]
- 9.Liddy N, et al. Monoclonal TCR-redirected tumor cell killing. Nature medicine 18, 980–987 (2012). [DOI] [PubMed] [Google Scholar]
- 10.Hsiue EH, et al. Targeting a neoantigen derived from a common TP53 mutation. Science 371(2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Walker AJ, et al. Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase. Molecular therapy : the journal of the American Society of Gene Therapy 25, 2189–2201 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mansilla-Soto J, et al. HLA-independent T cell receptors for targeting tumors with low antigen density. Nature medicine (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Huang J, et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4(+) T cells. Immunity 39, 846–857 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Purbhoo MA, Irvine DJ, Huppa JB & Davis MM T cell killing does not require the formation of a stable mature immunological synapse. Nat Immunol 5, 524–530 (2004). [DOI] [PubMed] [Google Scholar]
- 15.Sibille C, et al. Structure of the gene of tum- transplantation antigen P198: a point mutation generates a new antigenic peptide. The Journal of experimental medicine 172, 35–45 (1990). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.den Haan JM, et al. The minor histocompatibility antigen HA-1: a diallelic gene with a single amino acid polymorphism. Science 279, 1054–1057 (1998). [DOI] [PubMed] [Google Scholar]
- 17.Lamb JR, Feldmann M, Green N & Lerner RA Influence of antigen structure on the activation and induction of unresponsiveness in cloned human T lymphocytes. Immunology 57, 331–335 (1986). [PMC free article] [PubMed] [Google Scholar]
- 18. Nathan P, et al. Overall Survival Benefit with Tebentafusp in Metastatic Uveal Melanoma. The New England journal of medicine 385, 1196–1206 (2021). This paper reports on a randomized clinical trial in patients with metatstatic uveal melanoma which led to the first FDA approved TCR therapeutic for the treatment of cancer.
- 19.Carter PJ & Lazar GA Next generation antibody drugs: pursuit of the ‘high-hanging fruit’. Nat Rev Drug Discov 17, 197–223 (2018). [DOI] [PubMed] [Google Scholar]
- 20.Kalbasi A & Ribas A Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 20, 25–39 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Peri A, et al. The landscape of T cell antigens for cancer immunotherapy. Nat Cancer 4, 937–954 (2023). [DOI] [PubMed] [Google Scholar]
- 22.Susac L, et al. Structure of a fully assembled tumor-specific T cell receptor ligated by pMHC. Cell 185, 3201–3213 e3219 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sebestyen Z, Prinz I, Dechanet-Merville J, Silva-Santos B & Kuball J Translating gammadelta (gammadelta) T cells and their receptors into cancer cell therapies. Nat Rev Drug Discov 19, 169–184 (2020). [DOI] [PubMed] [Google Scholar]
- 24.Davis MM & Bjorkman PJ T-cell antigen receptor genes and T-cell recognition. Nature 334, 395–402 (1988). [DOI] [PubMed] [Google Scholar]
- 25.Thomas S, et al. Framework engineering to produce dominant T cell receptors with enhanced antigen-specific function. Nature communications 10, 4451 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cabaniols JP, Fazilleau N, Casrouge A, Kourilsky P & Kanellopoulos JM Most alpha/beta T cell receptor diversity is due to terminal deoxynucleotidyl transferase. The Journal of experimental medicine 194, 1385–1390 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Borrman T, et al. ATLAS: A database linking binding affinities with structures for wild-type and mutant TCR-pMHC complexes. Proteins 85, 908–916 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Leem J, de Oliveira SHP, Krawczyk K & Deane CM STCRDab: the structural T-cell receptor database. Nucleic Acids Res 46, D406–D412 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Aleksic M, et al. Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur J Immunol 42, 3174–3179 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Foote J & Eisen HN Kinetic and affinity limits on antibodies produced during immune responses. Proc Natl Acad Sci U S A 92, 1254–1256 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Clay TM, et al. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. Journal of immunology 163, 507–513 (1999). This paper provides the first demonstration that genetically modifying primary human T cells to express an exogenous TCR can confer tumor recognition.
- 32.Stanislawski T, et al. Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol 2, 962–970 (2001). [DOI] [PubMed] [Google Scholar]
- 33. Morgan RA, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314, 126–129 (2006). This paper reports the first demonstration that adoptive transfer of TCR engineered T cells can lead to cancer regression in humans.
- 34.Johnson LA, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114, 535–546 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Parkhurst MR, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Molecular therapy : the journal of the American Society of Gene Therapy 19, 620–626 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Morgan RA, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. Journal of immunotherapy 36, 133–151 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kageyama S, et al. Adoptive Transfer of MAGE-A4 T-cell Receptor Gene-Transduced Lymphocytes in Patients with Recurrent Esophageal Cancer. Clin Cancer Res 21, 2268–2277 (2015). [DOI] [PubMed] [Google Scholar]
- 38. Lu YC, et al. Treatment of Patients With Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3. Journal of clinical oncology : official journal of the American Society of Clinical Oncology, JCO2017745463 (2017). This paper provides the first demonstration that adoptive transfer of CD4+T cells genetically modified with an HLA class II-restricted TCR can cause cancer regression in patients with diverse solid tumors.
- 39.Tawara I, et al. Safety and persistence of WT1-specific T-cell receptor gene-transduced lymphocytes in patients with AML and MDS. Blood 130, 1985–1994 (2017). [DOI] [PubMed] [Google Scholar]
- 40. Doran SL, et al. T-Cell Receptor Gene Therapy for Human Papillomavirus-Associated Epithelial Cancers: A First-in-Human, Phase I/II Study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 37, 2759–2768 (2019). This clinical report, together with references 41 and 224, highlight diverse mechanisms of acquired immune resistance to adoptively transferred CD8+ T cells expressing TCRs specific for HLA class I-restricted epitopes.
- 41.Nagarsheth NB, et al. TCR-engineered T cells targeting E7 for patients with metastatic HPV-associated epithelial cancers. Nature medicine (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Linette GP, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 122, 863–871 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.D’Angelo SP, et al. Antitumor Activity Associated with Prolonged Persistence of Adoptively Transferred NY-ESO-1 (c259)T Cells in Synovial Sarcoma. Cancer Discov 8, 944–957 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chapuis AG, et al. T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant. Nature medicine 25, 1064–1072 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Stadtmauer EA, et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Zhang J, et al. Non-viral, specifically targeted CAR-T cells achieve high safety and efficacy in B-NHL. Nature 609, 369–374 (2022). This paper provides the first clinical report demonstrating the feasibility of simultaneously targeting multiple neoantigens using a nonviral TCR integration approach.
- 47.Foy SP, et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Marcucci KT, et al. Retroviral and Lentiviral Safety Analysis of Gene-Modified T Cell Products and Infused HIV and Oncology Patients. Molecular therapy : the journal of the American Society of Gene Therapy 26, 269–279 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Eyquem J, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Muller TR, et al. Targeted T cell receptor gene editing provides predictable T cell product function for immunotherapy. Cell Rep Med 2, 100374 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kaiser AD, et al. Towards a commercial process for the manufacture of genetically modified T cells for therapy. Cancer Gene Ther 22, 72–78 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fraietta JA, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shah NN, et al. Clonal expansion of CAR T cells harboring lentivector integration in the CBL gene following anti-CD22 CAR T-cell therapy. Blood Adv 3, 2317–2322 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Cohen CJ, Zhao Y, Zheng Z, Rosenberg SA & Morgan RA Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res 66, 8878–8886 (2006). This paper, together with references 74 and 75, describe commonly used modifications to the TCR constant chains which enhances both the potency and safety of exogenously expressed TCRs.
- 55.Heemskerk MH, et al. Efficiency of T-cell receptor expression in dual-specific T cells is controlled by the intrinsic qualities of the TCR chains within the TCR-CD3 complex. Blood 109, 235–243 (2007). [DOI] [PubMed] [Google Scholar]
- 56.Ahmadi M, et al. CD3 limits the efficacy of TCR gene therapy in vivo. Blood 118, 3528–3537 (2011). [DOI] [PubMed] [Google Scholar]
- 57.van Loenen MM, et al. Mixed T cell receptor dimers harbor potentially harmful neoreactivity. Proc Natl Acad Sci U S A 107, 10972–10977 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bendle GM, et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nature medicine 16, 565–570, 561p following 570 (2010). [DOI] [PubMed] [Google Scholar]
- 59.Rosenberg SA Of mice, not men: no evidence for graft-versus-host disease in humans receiving T-cell receptor-transduced autologous T cells. Molecular therapy : the journal of the American Society of Gene Therapy 18, 1744–1745 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Provasi E, et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nature medicine 18, 807–815 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Berdien B, Mock U, Atanackovic D & Fehse B TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther 21, 539–548 (2014). [DOI] [PubMed] [Google Scholar]
- 62.Roth TL, et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ruggiero E, et al. CRISPR-based gene disruption and integration of high-avidity, WT1-specific T cell receptors improve antitumor T cell function. Science translational medicine 14, eabg8027 (2022). [DOI] [PubMed] [Google Scholar]
- 64.Nahmad AD, et al. Frequent aneuploidy in primary human T cells after CRISPR-Cas9 cleavage. Nat Biotechnol (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ishihara M, et al. NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome. J Immunother Cancer 10(2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rees HA & Liu DR Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet 19, 770–788 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Webber BR, et al. Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors. Nature communications 10, 5222 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Preece R, et al. CRISPR-Mediated Base Conversion Allows Discriminatory Depletion of Endogenous T Cell Receptors for Enhanced Synthetic Immunity. Mol Ther Methods Clin Dev 19, 149–161 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Chiesa R, et al. Base-Edited CAR7 T Cells for Relapsed T-Cell Acute Lymphoblastic Leukemia. The New England journal of medicine (2023). [DOI] [PubMed] [Google Scholar]
- 70.Schober K, et al. Orthotopic replacement of T-cell receptor alpha- and beta-chains with preservation of near-physiological T-cell function. Nat Biomed Eng 3, 974–984 (2019). [DOI] [PubMed] [Google Scholar]
- 71.Shy BR, et al. High-yield genome engineering in primary cells using a hybrid ssDNA repair template and small-molecule cocktails. Nat Biotechnol (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Oh SA, et al. High-efficiency nonviral CRISPR/Cas9-mediated gene editing of human T cells using plasmid donor DNA. The Journal of experimental medicine 219(2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Nguyen DN, et al. Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat Biotechnol 38, 44–49 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cohen CJ, et al. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res 67, 3898–3903 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kuball J, et al. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 109, 2331–2338 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bialer G, Horovitz-Fried M, Ya’acobi S, Morgan RA & Cohen CJ Selected murine residues endow human TCR with enhanced tumor recognition. Journal of immunology 184, 6232–6241 (2010). [DOI] [PubMed] [Google Scholar]
- 77.Sommermeyer D & Uckert W Minimal amino acid exchange in human TCR constant regions fosters improved function of TCR gene-modified T cells. Journal of immunology 184, 6223–6231 (2010). [DOI] [PubMed] [Google Scholar]
- 78.Davis JL, et al. Development of human anti-murine T-cell receptor antibodies in both responding and nonresponding patients enrolled in TCR gene therapy trials. Clin Cancer Res 16, 5852–5861 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bethune MT, et al. Domain-swapped T cell receptors improve the safety of TCR gene therapy. Elife 5(2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kuball J, et al. Increasing functional avidity of TCR-redirected T cells by removing defined N-glycosylation sites in the TCR constant domain. The Journal of experimental medicine 206, 463–475 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Haga-Friedman A, Horovitz-Fried M & Cohen CJ Incorporation of transmembrane hydrophobic mutations in the TCR enhance its surface expression and T cell functional avidity. Journal of immunology 188, 5538–5546 (2012). [DOI] [PubMed] [Google Scholar]
- 82.Robbins PF, et al. Single and dual amino acid substitutions in TCR CDRs can enhance antigen-specific T cell functions. Journal of immunology 180, 6116–6131 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Li Y, et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat Biotechnol 23, 349–354 (2005). This paper reports on the use of phage-display libraries for the high-throughput evolution and selection of affinity-enhanced TCRs.
- 84.Holler PD, et al. In vitro evolution of a T cell receptor with high affinity for peptide/MHC. Proc Natl Acad Sci U S A 97, 5387–5392 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Dilchert J, Hofmann M, Unverdorben F, Kontermann R & Bunk S Mammalian Display Platform for the Maturation of Bispecific TCR-Based Molecules. Antibodies (Basel) 11(2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Cameron BJ, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Science translational medicine 5, 197ra103 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sanderson JP, et al. Preclinical evaluation of an affinity-enhanced MAGE-A4-specific T-cell receptor for adoptive T-cell therapy. Oncoimmunology 9, 1682381 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hellman LM, et al. Improving T Cell Receptor On-Target Specificity via Structure-Guided Design. Molecular therapy : the journal of the American Society of Gene Therapy 27, 300–313 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rosenberg AM & Baker BM Engineering the T cell receptor for fun and profit: Uncovering complex biology, interrogating the immune system, and targeting disease. Curr Opin Struct Biol 74, 102358 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zhao X, et al. Tuning T cell receptor sensitivity through catch bond engineering. Science 376, eabl5282 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sibener LV, et al. Isolation of a Structural Mechanism for Uncoupling T Cell Receptor Signaling from Peptide-MHC Binding. Cell 174, 672–687 e627 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Poole A, et al. Therapeutic high affinity T cell receptor targeting a KRAS(G12D) cancer neoantigen. Nature communications 13, 5333 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zhao Y, et al. High-affinity TCRs generated by phage display provide CD4+ T cells with the ability to recognize and kill tumor cell lines. Journal of immunology 179, 5845–5854 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Border EC, Sanderson JP, Weissensteiner T, Gerry AB & Pumphrey NJ Affinity-enhanced T-cell receptors for adoptive T-cell therapy targeting MAGE-A10: strategy for selection of an optimal candidate. Oncoimmunology 8, e1532759 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Docta RY, et al. Tuning T-Cell Receptor Affinity to Optimize Clinical Risk-Benefit When Targeting Alpha-Fetoprotein-Positive Liver Cancer. Hepatology 69, 2061–2075 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Huang C Receptor-Fc fusion therapeutics, traps, and MIMETIBODY technology. Curr Opin Biotechnol 20, 692–699 (2009). [DOI] [PubMed] [Google Scholar]
- 97.Dao T, et al. Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1. Nat Biotechnol 33, 1079–1086 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hoogenboom HR, et al. Antibody phage display technology and its applications. Immunotechnology 4, 1–20 (1998). [DOI] [PubMed] [Google Scholar]
- 99.Yang X, et al. Facile repurposing of peptide-MHC-restricted antibodies for cancer immunotherapy. Nat Biotechnol (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Holland CJ, et al. Specificity of bispecific T cell receptors and antibodies targeting peptide-HLA. J Clin Invest 130, 2673–2688 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Yarmarkovich M, et al. Cross-HLA targeting of intracellular oncoproteins with peptide-centric CARs. Nature 599, 477–484 (2021). This paper describes the use of peptide-centric antibody binders that recognize the same epitope presented in the context of different HLA alleles as a strtaegy to enhance the scalability of TCR therapeutics.
- 102.Dunbar J, Knapp B, Fuchs A, Shi J & Deane CM Examining variable domain orientations in antigen receptors gives insight into TCR-like antibody design. PLoS Comput Biol 10, e1003852 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Cole DK, et al. Hotspot autoimmune T cell receptor binding underlies pathogen and insulin peptide cross-reactivity. J Clin Invest 126, 3626 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Adams JJ, et al. Structural interplay between germline interactions and adaptive recognition determines the bandwidth of TCR-peptide-MHC cross-reactivity. Nat Immunol 17, 87–94 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Liu C, et al. Validation and promise of a TCR mimic antibody for cancer immunotherapy of hepatocellular carcinoma. Sci Rep 12, 12068 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Helsen CW, et al. The chimeric TAC receptor co-opts the T cell receptor yielding robust anti-tumor activity without toxicity. Nature communications 9, 3049 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Long AH, et al. 4–1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nature medicine 21, 581–590 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Baeuerle PA, et al. Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nature communications 10, 2087 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hassan R, et al. Mesothelin-targeting T cell receptor fusion construct cell therapy in refractory solid tumors: phase 1/2 trial interim results. Nature medicine (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Liu Y, et al. Chimeric STAR receptors using TCR machinery mediate robust responses against solid tumors. Science translational medicine 13(2021). [DOI] [PubMed] [Google Scholar]
- 111.Oliveira G, et al. Phenotype, specificity and avidity of antitumour CD8(+) T cells in melanoma. Nature 596, 119–125 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Stevanovic S, et al. Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer. Science 356, 200–205 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Creelan BC, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nature medicine 27, 1410–1418 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nature medicine 21, 81–85 (2015). [DOI] [PubMed] [Google Scholar]
- 115.Parkhurst MR, et al. Unique Neoantigens Arise from Somatic Mutations in Patients with Gastrointestinal Cancers. Cancer Discov 9, 1022–1035 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Caushi JX, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Scheper W, et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nature medicine 25, 89–94 (2019). [DOI] [PubMed] [Google Scholar]
- 118.Pasetto A, et al. Tumor- and Neoantigen-Reactive T-cell Receptors Can Be Identified Based on Their Frequency in Fresh Tumor. Cancer immunology research 4, 734–743 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Simoni Y, et al. Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018). [DOI] [PubMed] [Google Scholar]
- 120.Hanada KI, et al. A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell 40, 479–493 e476 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Oliveira G, et al. Landscape of helper and regulatory antitumour CD4(+) T cells in melanoma. Nature 605, 532–538 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Duhen T, et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nature communications 9, 2724 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Krishna S, et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chow A, et al. The ectonucleotidase CD39 identifies tumor-reactive CD8(+) T cells predictive of immune checkpoint blockade efficacy in human lung cancer. Immunity 56, 93–106 e106 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Gros A, et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J Clin Invest 124, 2246–2259 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Eberhardt CS, et al. Functional HPV-specific PD-1(+) stem-like CD8 T cells in head and neck cancer. Nature 597, 279–284 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Veatch JR, et al. Neoantigen-specific CD4(+) T cells in human melanoma have diverse differentiation states and correlate with CD8(+) T cell, macrophage, and B cell function. Cancer Cell 40, 393–409 e399 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ahmadzadeh M, et al. Tumor-infiltrating human CD4(+) regulatory T cells display a distinct TCR repertoire and exhibit tumor and neoantigen reactivity. Sci Immunol 4(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Cohen CJ, et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J Clin Invest 125, 3981–3991 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhang SQ, et al. High-throughput determination of the antigen specificities of T cell receptors in single cells. Nat Biotechnol (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Gee MH, et al. Antigen Identification for Orphan T Cell Receptors Expressed on Tumor-Infiltrating Lymphocytes. Cell 172, 549–563 e516 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Li G, et al. T cell antigen discovery via trogocytosis. Nat Methods 16, 183–190 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kula T, et al. T-Scan: A Genome-wide Method for the Systematic Discovery of T Cell Epitopes. Cell 178, 1016–1028 e1013 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gejman RS, et al. Identification of the Targets of T-cell Receptor Therapeutic Agents and Cells by Use of a High-Throughput Genetic Platform. Cancer immunology research 8, 672–684 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dobson CS, et al. Antigen identification and high-throughput interaction mapping by reprogramming viral entry. Nat Methods 19, 449–460 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ye Q, et al. CD137 accurately identifies and enriches for naturally occurring tumor-reactive T cells in tumor. Clin Cancer Res 20, 44–55 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Parkhurst M, et al. Isolation of T-Cell Receptors Specifically Reactive with Mutated Tumor-Associated Antigens from Tumor-Infiltrating Lymphocytes Based on CD137 Expression. Clin Cancer Res 23, 2491–2505 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Litchfield K, et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614 e514 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lowery FJ, et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science, eabl5447 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Gros A, et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nature medicine 22, 433–438 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Cafri G, et al. Memory T cells targeting oncogenic mutations detected in peripheral blood of epithelial cancer patients. Nature communications 10, 449 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Neudorfer J, et al. Reversible HLA multimers (Streptamers) for the isolation of human cytotoxic T lymphocytes functionally active against tumor- and virus-derived antigens. J Immunol Methods 320, 119–131 (2007). [DOI] [PubMed] [Google Scholar]
- 143.Perica K, et al. Enrichment and Expansion with Nanoscale Artificial Antigen Presenting Cells for Adoptive Immunotherapy. ACS Nano 9, 6861–6871 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Stronen E, et al. Targeting of cancer neoantigens with donor-derived T cell receptor repertoires. Science 352, 1337–1341 (2016). [DOI] [PubMed] [Google Scholar]
- 145.Bassani-Sternberg M, et al. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nature communications 7, 13404 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Pai CS, et al. Clonal Deletion of Tumor-Specific T Cells by Interferon-gamma Confers Therapeutic Resistance to Combination Immune Checkpoint Blockade. Immunity 50, 477–492 e478 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wolfl M & Greenberg PD Antigen-specific activation and cytokine-facilitated expansion of naive, human CD8+ T cells. Nat Protoc 9, 950–966 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Chandran SS, et al. Immunogenicity and therapeutic targeting of a public neoantigen derived from mutated PIK3CA. Nature medicine 28, 946–957 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Ali M, et al. T cells targeted to TdT kill leukemic lymphoblasts while sparing normal lymphocytes. Nat Biotechnol 40, 488–498 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.van der Lee DI, et al. Mutated nucleophosmin 1 as immunotherapy target in acute myeloid leukemia. J Clin Invest 129, 774–785 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Jahn L, et al. TCR-based therapy for multiple myeloma and other B-cell malignancies targeting intracellular transcription factor BOB1. Blood 129, 1284–1295 (2017). [DOI] [PubMed] [Google Scholar]
- 152.Falkenburg WJ, et al. Allogeneic HLA-A*02-restricted WT1-specific T cells from mismatched donors are highly reactive but show off-target promiscuity. Journal of immunology 187, 2824–2833 (2011). [DOI] [PubMed] [Google Scholar]
- 153.Bijen HM, et al. Preclinical Strategies to Identify Off-Target Toxicity of High-Affinity TCRs. Molecular therapy : the journal of the American Society of Gene Therapy 26, 1206–1214 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Theobald M, et al. Tolerance to p53 by A2.1-restricted cytotoxic T lymphocytes. The Journal of experimental medicine 185, 833–841 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Li LP, et al. Transgenic mice with a diverse human T cell antigen receptor repertoire. Nature medicine 16, 1029–1034 (2010). [DOI] [PubMed] [Google Scholar]
- 156.Moore MJ, et al. Humanization of T cell-mediated immunity in mice. Sci Immunol 6, eabj4026 (2021). [DOI] [PubMed] [Google Scholar]
- 157.Lonberg N Human antibodies from transgenic animals. Nat Biotechnol 23, 1117–1125 (2005). [DOI] [PubMed] [Google Scholar]
- 158.Obenaus M, et al. Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice. Nat Biotechnol 33, 402–407 (2015). [DOI] [PubMed] [Google Scholar]
- 159.Poncette L, Chen X, Lorenz FK & Blankenstein T Effective NY-ESO-1-specific MHC II-restricted T cell receptors from antigen-negative hosts enhance tumor regression. J Clin Invest 129, 324–335 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Klebanoff CA, Rosenberg SA & Restifo NP Prospects for gene-engineered T cell immunotherapy for solid cancers. Nature medicine 22, 26–36 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Mortazavi A, Williams BA, McCue K, Schaeffer L & Wold B Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5, 621–628 (2008). [DOI] [PubMed] [Google Scholar]
- 162.Consortium GT The Genotype-Tissue Expression (GTEx) project. Nat Genet 45, 580–585 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Parker KR, et al. Single-Cell Analyses Identify Brain Mural Cells Expressing CD19 as Potential Off-Tumor Targets for CAR-T Immunotherapies. Cell 183, 126–142 e117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Wu L, et al. Variation and genetic control of protein abundance in humans. Nature 499, 79–82 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Haas GG Jr., D’Cruz OJ & De Bault LE Distribution of human leukocyte antigen-ABC and -D/DR antigens in the unfixed human testis. Am J Reprod Immunol Microbiol 18, 47–51 (1988). [DOI] [PubMed] [Google Scholar]
- 166.Felix NJ & Allen PM Specificity of T-cell alloreactivity. Nat Rev Immunol 7, 942–953 (2007). [DOI] [PubMed] [Google Scholar]
- 167.van Amerongen RA, et al. Human iPSC-derived preclinical models to identify toxicity of tumor-specific T cells with clinical potential. Mol Ther Methods Clin Dev 28, 249–261 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kunert A, Obenaus M, Lamers CHJ, Blankenstein T & Debets R T-cell Receptors for Clinical Therapy: In Vitro Assessment of Toxicity Risk. Clin Cancer Res 23, 6012–6020 (2017). [DOI] [PubMed] [Google Scholar]
- 169.Riley TP, et al. T cell receptor cross-reactivity expanded by dramatic peptide-MHC adaptability. Nat Chem Biol 14, 934–942 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Birnbaum ME, et al. Deconstructing the peptide-MHC specificity of T cell recognition. Cell 157, 1073–1087 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Wilson DB, et al. Specificity and degeneracy of T cells. Mol Immunol 40, 1047–1055 (2004). [DOI] [PubMed] [Google Scholar]
- 172.Whalley T, et al. GPU-Accelerated Discovery of Pathogen-Derived Molecular Mimics of a T-Cell Insulin Epitope. Front Immunol 11, 296 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Scholtalbers J, et al. TCLP: an online cancer cell line catalogue integrating HLA type, predicted neo-epitopes, virus and gene expression. Genome Med 7, 118 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Tran E, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344, 641–645 (2014). This clinical case report provides the first evidence that HLA class II-resricted CD4+ T cells targeting a private neoantigen can mediate durable tumor regression in a human.
- 175.Dijkstra KK, et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell 174, 1586–1598 e1512 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Demmers LC, et al. Single-cell derived tumor organoids display diversity in HLA class I peptide presentation. Nature communications 11, 5338 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Tuveson D & Clevers H Cancer modeling meets human organoid technology. Science 364, 952–955 (2019). [DOI] [PubMed] [Google Scholar]
- 178.Axelrod ML, Cook RS, Johnson DB & Balko JM Biological Consequences of MHC-II Expression by Tumor Cells in Cancer. Clin Cancer Res 25, 2392–2402 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Hos BJ, et al. Cancer-specific T helper shared and neo-epitopes uncovered by expression of the MHC class II master regulator CIITA. Cell Rep 41, 111485 (2022). [DOI] [PubMed] [Google Scholar]
- 180.Zeh HJ 3rd, Perry-Lalley D, Dudley ME, Rosenberg SA & Yang JC High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. Journal of immunology 162, 989–994 (1999). [PubMed] [Google Scholar]
- 181.Dutoit V, et al. Heterogeneous T-cell response to MAGE-A10(254–262): high avidity-specific cytolytic T lymphocytes show superior antitumor activity. Cancer Res 61, 5850–5856 (2001). [PubMed] [Google Scholar]
- 182.Johnson LA, et al. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes. Journal of immunology 177, 6548–6559 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Slifka MK & Whitton JL Functional avidity maturation of CD8(+) T cells without selection of higher affinity TCR. Nat Immunol 2, 711–717 (2001). [DOI] [PubMed] [Google Scholar]
- 184.Gallegos AM, et al. Control of T cell antigen reactivity via programmed TCR downregulation. Nat Immunol 17, 379–386 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Guy CS, et al. Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. Nat Immunol 14, 262–270 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhong S, et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci U S A 110, 6973–6978 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sim MJW, et al. High-affinity oligoclonal TCRs define effective adoptive T cell therapy targeting mutant KRAS-G12D. Proc Natl Acad Sci U S A 117, 12826–12835 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Wu D, Gallagher DT, Gowthaman R, Pierce BG & Mariuzza RA Structural basis for oligoclonal T cell recognition of a shared p53 cancer neoantigen. Nature communications 11, 2908 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Devlin JR, et al. Structural dissimilarity from self drives neoepitope escape from immune tolerance. Nat Chem Biol 16, 1269–1276 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Margulies DH, Plaksin D, Khilko SN & Jelonek MT Studying interactions involving the T-cell antigen receptor by surface plasmon resonance. Curr Opin Immunol 8, 262–270 (1996). [DOI] [PubMed] [Google Scholar]
- 191.Tian S, Maile R, Collins EJ & Frelinger JA CD8+ T cell activation is governed by TCR-peptide/MHC affinity, not dissociation rate. Journal of immunology 179, 2952–2960 (2007). [DOI] [PubMed] [Google Scholar]
- 192.Huang J, et al. The kinetics of two-dimensional TCR and pMHC interactions determine T-cell responsiveness. Nature 464, 932–936 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Liu B, et al. 2D TCR-pMHC-CD8 kinetics determines T-cell responses in a self-antigen-specific TCR system. Eur J Immunol 44, 239–250 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Gannon PO, et al. Quantitative TCR:pMHC Dissociation Rate Assessment by NTAmers Reveals Antimelanoma T Cell Repertoires Enriched for High Functional Competence. Journal of immunology 195, 356–366 (2015). [DOI] [PubMed] [Google Scholar]
- 195.Hebeisen M, et al. Identification of Rare High-Avidity, Tumor-Reactive CD8+ T Cells by Monomeric TCR-Ligand Off-Rates Measurements on Living Cells. Cancer Res 75, 1983–1991 (2015). [DOI] [PubMed] [Google Scholar]
- 196.Nauerth M, et al. Flow cytometry-based TCR-ligand Koff -rate assay for fast avidity screening of even very small antigen-specific T cell populations ex vivo. Cytometry A 89, 816–825 (2016). [DOI] [PubMed] [Google Scholar]
- 197.Laugel B, et al. Different T cell receptor affinity thresholds and CD8 coreceptor dependence govern cytotoxic T lymphocyte activation and tetramer binding properties. J Biol Chem 282, 23799–23810 (2007). [DOI] [PubMed] [Google Scholar]
- 198.Schmidt J, et al. Neoantigen-specific CD8 T cells with high structural avidity preferentially reside in and eliminate tumors. Nature communications 14, 3188 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Jin BY, et al. Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model. JCI Insight 3(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Allard M, et al. TCR-ligand dissociation rate is a robust and stable biomarker of CD8+ T cell potency. JCI Insight 2(2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Gattinoni L, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. The Journal of experimental medicine 202, 907–912 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Klebanoff CA, Khong HT, Antony PA, Palmer DC & Restifo NP Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 26, 111–117 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Paulos CM, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest 117, 2197–2204 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Klebanoff CA, et al. Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clin Cancer Res 17, 5343–5352 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Chandran SS, et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol 18, 792–802 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Arnaud M, et al. Sensitive identification of neoantigens and cognate TCRs in human solid tumors. Nat Biotechnol (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Leko V, et al. Identification of Neoantigen-Reactive Tumor-Infiltrating Lymphocytes in Primary Bladder Cancer. Journal of immunology 202, 3458–3467 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Zacharakis N, et al. Breast Cancers Are Immunogenic: Immunologic Analyses and a Phase II Pilot Clinical Trial Using Mutation-Reactive Autologous Lymphocytes. Journal of clinical oncology : official journal of the American Society of Clinical Oncology, JCO2102170 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Stevanovic S, et al. A Phase II Study of Tumor-infiltrating Lymphocyte Therapy for Human Papillomavirus-associated Epithelial Cancers. Clin Cancer Res 25, 1486–1493 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Hanada K, Perry-Lalley DM, Ohnmacht GA, Bettinotti MP & Yang JC Identification of fibroblast growth factor-5 as an overexpressed antigen in multiple human adenocarcinomas. Cancer Res 61, 5511–5516 (2001). [PubMed] [Google Scholar]
- 211.McGranahan N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Goff SL, et al. Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients With Metastatic Melanoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34, 2389–2397 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Sarnaik AA, et al. Lifileucel, a Tumor-Infiltrating Lymphocyte Therapy, in Metastatic Melanoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 39, 2656–2666 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Haanen JBAG, et al. LBA3 Treatment with tumor-infiltrating lymphocytes (TIL) versus ipilimumab for advanced melanoma: Results from a multicenter, randomized phase III trial. Annals of Oncology 33, S1406 (2022). This randomized phase III clinical trial demonstrated that adoptive transfer of TILs results in significiantly longer progression free survival compared with anti-CTLA-4 as a salvage therapy following anti-PD1 treatment failure.
- 215.Dafni U, et al. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol 30, 1902–1913 (2019). [DOI] [PubMed] [Google Scholar]
- 216.Radvanyi LG, et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 18, 6758–6770 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Seitter SJ, et al. Impact of Prior Treatment on the Efficacy of Adoptive Transfer of Tumor-Infiltrating Lymphocytes in Patients with Metastatic Melanoma. Clin Cancer Res (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Besser MJ, et al. Adoptive transfer of tumor-infiltrating lymphocytes in patients with metastatic melanoma: intent-to-treat analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res 19, 4792–4800 (2013). [DOI] [PubMed] [Google Scholar]
- 219.Levi ST, et al. Neoantigen Identification and Response to Adoptive Cell Transfer in anti PD-1 Naive and Experienced Patients with Metastatic Melanoma. Clin Cancer Res (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Samstein RM, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 51, 202–206 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Lauss M, et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nature communications 8, 1738 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kristensen NP, et al. Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive transfer with tumor-infiltrating lymphocytes in melanoma. J Clin Invest (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Algazi AP, et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer 122, 3344–3353 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Tran E, et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. The New England journal of medicine 375, 2255–2262 (2016). This report, together with reference 275, demonstrate that adoptive transfer of T cells targeting an epitope derived from the recurrent KRAS G12D hotspot mutation can mediate tumor regression in patients with common solid cancers.
- 225.Klebanoff CA & Wolchok JD Shared cancer neoantigens: Making private matters public. The Journal of experimental medicine 215, 5–7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Butler MO, et al. Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells. Science translational medicine 3, 80ra34 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Chapuis AG, et al. T-Cell Therapy Using Interleukin-21-Primed Cytotoxic T-Cell Lymphocytes Combined With Cytotoxic T-Cell Lymphocyte Antigen-4 Blockade Results in Long-Term Cell Persistence and Durable Tumor Regression. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34, 3787–3795 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Yee C, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A 99, 16168–16173 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Khammari A, et al. Treatment of metastatic melanoma with autologous Melan-A/MART-1-specific cytotoxic T lymphocyte clones. J Invest Dermatol 129, 2835–2842 (2009). [DOI] [PubMed] [Google Scholar]
- 230.Hunder NN, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. The New England journal of medicine 358, 2698–2703 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Chandran SS, et al. Persistence of CTL clones targeting melanocyte differentiation antigens was insufficient to mediate significant melanoma regression in humans. Clin Cancer Res 21, 534–543 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Dudley ME, et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. Journal of immunotherapy 24, 363–373 (2001). [DOI] [PubMed] [Google Scholar]
- 233.Chapuis AG, et al. Transferred melanoma-specific CD8+ T cells persist, mediate tumor regression, and acquire central memory phenotype. Proc Natl Acad Sci U S A 109, 4592–4597 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Caballero OL & Chen YT Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci 100, 2014–2021 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.De Smet C, Lurquin C, Lethe B, Martelange V & Boon T DNA methylation is the primary silencing mechanism for a set of germ line- and tumor-specific genes with a CpG-rich promoter. Mol Cell Biol 19, 7327–7335 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Chapuis AG, et al. Tracking the Fate and Origin of Clinically Relevant Adoptively Transferred CD8(+) T Cells In Vivo. Sci Immunol 2(2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Klebanoff CA, Gattinoni L & Restifo NP Sorting through subsets: which T-cell populations mediate highly effective adoptive immunotherapy? Journal of immunotherapy 35, 651–660 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Chandran SS, et al. Tumor-Specific Effector CD8+ T Cells That Can Establish Immunological Memory in Humans after Adoptive Transfer Are Marked by Expression of IL7 Receptor and c-myc. Cancer Res 75, 3216–3226 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Wang A, et al. The stoichiometric production of IL-2 and IFN-gamma mRNA defines memory T cells that can self-renew after adoptive transfer in humans. Science translational medicine 4, 149ra120 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Klebanoff CA, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A 102, 9571–9576 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Gattinoni L, et al. A human memory T cell subset with stem cell-like properties. Nature medicine 17, 1290–1297 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Graef P, et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells. Immunity 41, 116–126 (2014). [DOI] [PubMed] [Google Scholar]
- 243.Yamamoto TN, et al. T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J Clin Invest 129, 1551–1565 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Oda SK, et al. A Fas-4–1BB fusion protein converts a death to a pro-survival signal and enhances T cell therapy. The Journal of experimental medicine 217(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Roth TL, et al. Pooled Knockin Targeting for Genome Engineering of Cellular Immunotherapies. Cell 181, 728–744 e721 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Silk JD, et al. Engineering Cancer Antigen-Specific T Cells to Overcome the Immunosuppressive Effects of TGF-beta. Journal of immunology 208, 169–180 (2022). [DOI] [PubMed] [Google Scholar]
- 247.Stromnes IM, et al. Abrogating Cbl-b in effector CD8(+) T cells improves the efficacy of adoptive therapy of leukemia in mice. J Clin Invest 120, 3722–3734 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Palmer DC, et al. Cish actively silences TCR signaling in CD8+ T cells to maintain tumor tolerance. The Journal of experimental medicine 212, 2095–2113 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Shifrut E, et al. Genome-wide CRISPR Screens in Primary Human T Cells Reveal Key Regulators of Immune Function. Cell 175, 1958–1971 e1915 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Carnevale J, et al. RASA2 ablation in T cells boosts antigen sensitivity and long-term function. Nature (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Duval L, et al. Adoptive transfer of allogeneic cytotoxic T lymphocytes equipped with a HLA-A2 restricted MART-1 T-cell receptor: a phase I trial in metastatic melanoma. Clin Cancer Res 12, 1229–1236 (2006). [DOI] [PubMed] [Google Scholar]
- 252.Borbulevych OY, Santhanagopolan SM, Hossain M & Baker BM TCRs used in cancer gene therapy cross-react with MART-1/Melan-A tumor antigens via distinct mechanisms. Journal of immunology 187, 2453–2463 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Sugiyama H WT1 (Wilms’ tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol 40, 377–387 (2010). [DOI] [PubMed] [Google Scholar]
- 254.Levine AJ, Momand J & Finlay CA The p53 tumour suppressor gene. Nature 351, 453–456 (1991). [DOI] [PubMed] [Google Scholar]
- 255.Theobald M, Biggs J, Dittmer D, Levine AJ & Sherman LA Targeting p53 as a general tumor antigen. Proc Natl Acad Sci U S A 92, 11993–11997 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Cohen CJ, et al. Recognition of fresh human tumor by human peripheral blood lymphocytes transduced with a bicistronic retroviral vector encoding a murine anti-p53 TCR. Journal of immunology 175, 5799–5808 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Theoret MR, et al. Relationship of p53 overexpression on cancers and recognition by anti-p53 T cell receptor-transduced T cells. Hum Gene Ther 19, 1219–1232 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 21, 1019–1027 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.D’Angelo SP, et al. Primary efficacy and safety of letetresgene autoleucel (lete-cel; GSK3377794) pilot study in patients with advanced and metastatic myxoid/round cell liposarcoma (MRCLS). Journal of Clinical Oncology 40, 11500–11500 (2022). [Google Scholar]
- 260.Kerkar SP, et al. MAGE-A is More Highly Expressed Than NY-ESO-1 in a Systematic Immunohistochemical Analysis of 3668 Cases. Journal of immunotherapy 39, 181–187 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Chinnasamy N, et al. A TCR targeting the HLA-A*0201-restricted epitope of MAGE-A3 recognizes multiple epitopes of the MAGE-A antigen superfamily in several types of cancer. Journal of immunology 186, 685–696 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.D’Angelo SP, et al. Identification of response stratification factors from pooled efficacy analyses of afamitresgene autoleucel (“Afami-cel” [Formerly ADP-A2M4]) in metastatic synovial sarcoma and myxoid/round cell liposarcoma phase 1 and phase 2 trials. Journal of Clinical Oncology 40, 11562–11562 (2022). [Google Scholar]
- 263.Hong DS, et al. Autologous T cell therapy for MAGE-A4(+) solid cancers in HLA-A*02(+) patients: a phase 1 trial. Nature medicine 29, 104–114 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Hong DS, et al. 540P Safety and efficacy from the SURPASS trial with ADP-A2M4CD8, a SPEAR T-cell therapy incorporating a CD8α co-receptor and an affinity optimized TCR targeting MAGE-A4. Annals of Oncology 32, S604–S605 (2021). [Google Scholar]
- 265.Hong DS, et al. Phase 1 Clinical Trial Evaluating the Safety and Anti-Tumor Activity of ADP-A2M10 SPEAR T-Cells in Patients With MAGE-A10+ Head and Neck, Melanoma, or Urothelial Tumors. Front Oncol 12, 818679 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Blumenschein GR, et al. Phase I clinical trial evaluating the safety and efficacy of ADP-A2M10 SPEAR T cells in patients with MAGE-A10(+) advanced non-small cell lung cancer. J Immunother Cancer 10(2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Xu Y, Zou R, Wang J, Wang ZW & Zhu X The role of the cancer testis antigen PRAME in tumorigenesis and immunotherapy in human cancer. Cell Prolif 53, e12770 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Wermke M, et al. 959 Safety and anti-tumor activity of TCR-engineered autologous, PRAME-directed T cells across multiple advanced solid cancers at low doses – clinical update on the ACTengine® IMA203 trial. Journal for ImmunoTherapy of Cancer 9, A1009–A1009 (2021). [Google Scholar]
- 269.Schiller JT & Lowy DR Vaccines to prevent infections by oncoviruses. Annu Rev Microbiol 64, 23–41 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Feng HC, Shuda M, Chang Y & Moore PS Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 319, 1096–1100 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Jing LC, et al. Prevalent and Diverse Intratumoral Oncoprotein-Specific CD8(+) T Cells within Polyomavirus-Driven Merkel Cell Carcinomas. Cancer immunology research 8, 648–659 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Miller NJ, et al. Tumor-Infiltrating Merkel Cell Polyomavirus-Specific T Cells Are Diverse and Associated with Improved Patient Survival. Cancer immunology research 5, 137–147 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Veatch J, et al. Merkel polyoma virus specific T-cell receptor transgenic T-cell therapy in PD-1 inhibitor refractory Merkel cell carcinoma. Journal of Clinical Oncology 40, 9549–9549 (2022). [Google Scholar]
- 274.Leidner R, et al. Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. The New England journal of medicine 386, 2112–2119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Kim SP, et al. Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor-Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors. Cancer immunology research, OF1–OF15 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Lo W, et al. Immunologic recognition of a shared p53 mutated neoantigen in a patient with metastatic colorectal cancer. Cancer immunology research (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Middleton MR, et al. Tebentafusp, A TCR/Anti-CD3 Bispecific Fusion Protein Targeting gp100, Potently Activated Antitumor Immune Responses in Patients with Metastatic Melanoma. Clin Cancer Res 26, 5869–5878 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Carvajal RD, et al. Clinical and molecular response to tebentafusp in previously treated patients with metastatic uveal melanoma: a phase 2 trial. Nature medicine (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. The New England journal of medicine 363, 411–422 (2010). [DOI] [PubMed] [Google Scholar]
- 280.Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine 363, 711–723 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Neelapu SS, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. The New England journal of medicine 377, 2531–2544 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Maude SL, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. The New England journal of medicine 378, 439–448 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Schuster SJ, et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. The New England journal of medicine 380, 45–56 (2019). [DOI] [PubMed] [Google Scholar]
- 284.Wang M, et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. The New England journal of medicine 382, 1331–1342 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Abramson JS, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet 396, 839–852 (2020). [DOI] [PubMed] [Google Scholar]
- 286.Shah BD, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet 398, 491–502 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Munshi NC, et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. The New England journal of medicine 384, 705–716 (2021). [DOI] [PubMed] [Google Scholar]
- 288.Mungall AJ, et al. The DNA sequence and analysis of human chromosome 6. Nature 425, 805–811 (2003). [DOI] [PubMed] [Google Scholar]
- 289.Donoghue MTA, Schram AM, Hyman DM & Taylor BS Discovery through clinical sequencing in oncology. Nat Cancer 1, 774–783 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Rosenbaum E, et al. HLA Genotyping in Synovial Sarcoma: Identifying HLA-A*02 and Its Association with Clinical Outcome. Clin Cancer Res 26, 5448–5455 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Montesion M, et al. Somatic HLA Class I Loss Is a Widespread Mechanism of Immune Evasion Which Refines the Use of Tumor Mutational Burden as a Biomarker of Checkpoint Inhibitor Response. Cancer Discov 11, 282–292 (2021). [DOI] [PubMed] [Google Scholar]
- 292.Doebele RC, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol 21, 271–282 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Hong DS, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 21, 531–540 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Le DT, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017). This paper provides the first comprehensive demonstration that a genomic biomarker (MSI-high) can be used to enrich for patients with diverse common solid cancers who are likely to respond to a single-agent cancer immunotherapy (anti-PD1).
- 295.Marabelle A, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 21, 1353–1365 (2020). [DOI] [PubMed] [Google Scholar]
- 296.Lipsitz YY, et al. A roadmap for cost-of-goods planning to guide economic production of cell therapy products. Cytotherapy 19, 1383–1391 (2017). [DOI] [PubMed] [Google Scholar]
- 297.Iriguchi S, et al. A clinically applicable and scalable method to regenerate T-cells from iPSCs for off-the-shelf T-cell immunotherapy. Nature communications 12, 430 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Morton LT, et al. T cell receptor engineering of primary NK cells to therapeutically target tumors and tumor immune evasion. J Immunother Cancer 10(2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Moore T, et al. Clinical and immunologic evaluation of three metastatic melanoma patients treated with autologous melanoma-reactive TCR-transduced T cells. Cancer Immunol Immunother 67, 311–325 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Negrao Marcelo V., M.P.M., Collinson-Pautz Matthew R., Demars Nathan, Adeyemi Frances, Carpio Victor H., Jazaeri Amir A., Johnson Benny, Pant Shubham, Partow Kebriaei, Deniger Drew C., Heymach John V., Kopetz Scott. Objective clinical response by KRAS mutationspecific TCR-T cell therapy in previously treated advanced non-small cell lung cancer. in CRI-ENCI-AACR (CICON22) (New York, N.Y., 2022). [Google Scholar]
- 301.Arstila TP, et al. A direct estimate of the human alphabeta T cell receptor diversity. Science 286, 958–961 (1999). [DOI] [PubMed] [Google Scholar]
- 302.Qi Q, et al. Diversity and clonal selection in the human T-cell repertoire. Proc Natl Acad Sci U S A 111, 13139–13144 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Merkenschlager M, et al. How many thymocytes audition for selection? The Journal of experimental medicine 186, 1149–1158 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Mason D A very high level of crossreactivity is an essential feature of the T-cell receptor. Immunol Today 19, 395–404 (1998). [DOI] [PubMed] [Google Scholar]
- 305.Sewell AK Why must T cells be cross-reactive? Nat Rev Immunol 12, 669–677 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Wooldridge L, et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem 287, 1168–1177 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Scott DR, Borbulevych OY, Piepenbrink KH, Corcelli SA & Baker BM Disparate degrees of hypervariable loop flexibility control T-cell receptor cross-reactivity, specificity, and binding mechanism. J Mol Biol 414, 385–400 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Colf LA, et al. How a single T cell receptor recognizes both self and foreign MHC. Cell 129, 135–146 (2007). [DOI] [PubMed] [Google Scholar]
- 309.Borbulevych OY, Piepenbrink KH & Baker BM Conformational melding permits a conserved binding geometry in TCR recognition of foreign and self molecular mimics. Journal of immunology 186, 2950–2958 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Sarkizova S, et al. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat Biotechnol 38, 199–209 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
