Adoptive therapy with T-cell receptor (TCR)–engineered T cells has shown promising results in the treatment of patients with tumors, and the number of TCRs amenable for clinical testing is expanding rapidly. Notably, adoptive therapy with T cells is challenged by treatment-related side effects, which calls for cautious selection of target antigens and TCRs that goes beyond their mere ability to induce high T-cell reactivity. Here, we propose a sequence of in vitro assays to improve selection of TCRs and exemplify risk assessments of on-target as well as off-target toxicities using TCRs directed against cancer germline antigens. The proposed panel of assays covers parameters considered key to safety, such as expression of target antigen in healthy tissues, determination of a TCR's recognition motif toward its cognate peptide, and a TCR's cross-reactivity toward noncognate peptides. Clin Cancer Res; 23(20); 6012–20. ©2017 AACR.

Therapeutic use of antitumor T cells has proven feasible in a multitude of trials over the past decade. Alongside the demonstration of clinical benefit and enthusiasm about the therapeutic efficacy, the occurrence of toxicities has stimulated awareness of safety pitfalls. Although initial studies demonstrated recognition, and sometimes destruction of healthy tissues (1–3), later studies demonstrated lethal adverse effects in individual patients (4, 5). These studies highlight the two main challenges facing the safety of T-cell receptor (TCR) gene therapy: on- and off-target toxicities. With a quickly expanding panel of TCRs that have generally been selected for their ability to provide T cells with high avidities toward tumor antigens, there is an urgent need for streamlining the safety assessment of these TCRs prior to clinical usage.

An ideal target antigen for adoptive T-cell therapy (AT) displays two important features: It is immunogenic, and it shows selective and homogenous expression in tumor tissue. Immunogenicity is best explained as an antigen's ability to be recognized by and sufficiently activate T cells, a feature that is generally well addressed when selecting a target antigen and its corresponding TCR (reviewed in ref. 6). For example, characteristics related to the immunogenicity of two MAGE-C2 (MC2) antigen epitopes are summarized in Table 1. Selective expression in tumor tissue, and hence its absence in healthy tissues, would reduce the risk for on-target toxicities. Differentiation, overexpressed, or oncofetal antigens are not absent from healthy tissues and the targeting of these antigens, for example, MART-1 or carcinoembryonic antigen, by TCR-transduced T cells has resulted in severe destruction of melanocytes in the eyes, ears, and skin (1) or inflammation of the colon (2, 3). Candidate antigens that are exclusively expressed by tumor tissues include neoantigens and oncoviral antigens. Neoantigens are derived from somatic DNA alterations, and their identification requires analyses of mutations, gene expression, and algorithms that predict antigen processing and presentation by MHC (7–9). AT studies with tumor-infiltrating lymphocytes (TIL) in patients with melanoma and cholangiocarcinoma showed that clinical benefit was associated with T-cell responses against neoantigens (9–11). Current exploitation of neoantigens in AT, however, is challenged by the uncertainty of current algorithms accurately predicting immunogenicity and the fact that neoantigens are usually specific per patient (12–14). Viral antigens are present in more than 10% of human cancers and are often the result of viral insertion into the genome and subsequent reactivation in tumors. AT studies using TILs reactive against either human papillomavirus (HPV) or Epstein–Barr virus (EBV) have shown clinical successes in patients with cervical cancer or nasopharyngeal carcinoma, respectively (15, 16). Similar to neoantigens and oncoviral antigens, certain cancer germline antigens (CGA) also demonstrate tumor-selective expression. In fact, CGAs are expressed in gonadal tissues and some in thymus (17), and certain CGAs are considered to be selectively derepressed in tumor tissuess (for detailed reviews, see refs. 6, 18, 19). MAGE-A3 and NY-ESO1 are examples of CGAs that have already been targeted by TCR-engineered T cells in patients with metastatic melanoma, metastatic synovial sarcoma, or multiple myeloma (4, 20). Although off-target toxicities were observed with the targeting of the former antigen (most likely an issue of the TCRs; see next section), the safe use of selected CGAs was suggested by the targeting of NY-ESO1 demonstrating clinical benefit without toxicities (20, 21).

Table 1.

Properties of cognate MAGE-C2 epitopes and corresponding TCRs that relate to T-cell reactivity

EpitopesLLFGLALIEV (MAGE-C2191-200)ALKDVEERV (MAGE-C2336-344)
HLA-A2 binding Predicted affinitya: 9.9 nmol/L Predicted affinitya: 342.2 nmol/L 
 Measured affinityb: 2.5 μmol/L Measured affinityb: 20.0 μmol/L 
TCR TCR6 TCR16 
TCR genesc α-chain: Vα12-2*01/Jα23*01/Cα α-chain: Vα3*01/Jα3*01/Cα 
 β-chain: Vβ15*02/Jβ2-3*01/Cβ2 β-chain: Vβ28*01/Jβ2-5*01/Cβ2 
pMHC bindingd EC50 CD8+ T cells: 348 nmol/L EC50 CD8+ T cells: 663 nmol/L 
 EC50 CD4+ T cells: 501 nmol/L EC50 CD4+ T cells: n.d. 
Functional T-cell aviditye EC50: 3.19 nmol/L EC50: 73.8 nmol/L 
Tumor cell recognitionf EB81-MEL 1,090 pg/mL EB81-MEL 1,738 pg/mL 
 518-A2 605 pg/mL 518-A2 461 pg/mL 
 607-B 835 pg/mL 607-B 626 pg/mL 
 SCC-9 311 pg/mL SCC-9 207 pg/mL 
 93-VU-120 292 pg/mL 93-VU-120 26 pg/mL 
 SUM-195-PT 217 pg/mL SUM-195-PT 466 pg/mL 
EpitopesLLFGLALIEV (MAGE-C2191-200)ALKDVEERV (MAGE-C2336-344)
HLA-A2 binding Predicted affinitya: 9.9 nmol/L Predicted affinitya: 342.2 nmol/L 
 Measured affinityb: 2.5 μmol/L Measured affinityb: 20.0 μmol/L 
TCR TCR6 TCR16 
TCR genesc α-chain: Vα12-2*01/Jα23*01/Cα α-chain: Vα3*01/Jα3*01/Cα 
 β-chain: Vβ15*02/Jβ2-3*01/Cβ2 β-chain: Vβ28*01/Jβ2-5*01/Cβ2 
pMHC bindingd EC50 CD8+ T cells: 348 nmol/L EC50 CD8+ T cells: 663 nmol/L 
 EC50 CD4+ T cells: 501 nmol/L EC50 CD4+ T cells: n.d. 
Functional T-cell aviditye EC50: 3.19 nmol/L EC50: 73.8 nmol/L 
Tumor cell recognitionf EB81-MEL 1,090 pg/mL EB81-MEL 1,738 pg/mL 
 518-A2 605 pg/mL 518-A2 461 pg/mL 
 607-B 835 pg/mL 607-B 626 pg/mL 
 SCC-9 311 pg/mL SCC-9 207 pg/mL 
 93-VU-120 292 pg/mL 93-VU-120 26 pg/mL 
 SUM-195-PT 217 pg/mL SUM-195-PT 466 pg/mL 

Abbreviation: n.d., not determined.

bT2 cells were pulsed with titrated amounts of ALK or LLF peptide, and formation of pMHC class I complexes on the cell surface was quantified via flow cytometry using PE-labeled HLA-A2 mAb (23).

ccDNAs derived from patient-derived T-cell clones were PCR amplified using either a set of TCR-Vα or Vβ sense primers and a corresponding TCR-Cα or Cβ antisense primer or 5′RACE (rapid amplification of cDNA ends). Following nested PCRs and cloning, TCRα and β sequences were identified using www.imgt.org and classified according to the Lefranc nomenclature (see ref. 23 for details).

dTCR T cells were pulsed with titrated amounts of PE-labeled pMHC multimer, and binding was quantified via flow cytometry.

eTCR T cells were antigen-presenting cells that were pulsed with titrated amounts of cognate peptide, and functional T-cell avidity was quantified via ELISA measurements of IFNγ production.

fTCR-transduced T cells were cocultured with three melanoma, two head and neck carcinoma, and one triple-negative breast cancer cell lines all positive for HLA-A2 and MC2 (determined by qPCR) at an E:T ratio of 3:1 for 24 hours. Cell lines were treated with epigenetic drugs azacytidine and valproate as well as IFNγ prior to coculture (see ref. 23 for details). IFNγ levels in 24-hour culture supernatants were measured by ELISA; displayed values are means of five experiments. Note that epigenetic drugs induce enhanced and tumor-selective expression of MC2 in vitro (23) as well as enhanced tumor immunogenicity in vivo (18).

We recommend for any antigen, with the exception of neoantigens, to test the antigen's absence from a large panel of healthy organs. Online databases, such as the protein atlas (http://www.proteinatlas.org/) or the CGA database (www.cta.lncc.br), combine extensive data from transcriptomic analyses and antibody stainings from numerous normal, noncancerous cell lines and tissues. When applying these tools to assess the expression of MC2, we observed that mRNA expression is restricted to cells from cancers and testes—the latter considered to possess an immune-privileged status (no MHC expression; thus no detection by T cells). The use of commercially available cDNA libraries of a large series of healthy tissues enables researchers to extend online analyses and quantify antigen expression with a laboratory assay. When performing qPCR using such a cDNA library, we demonstrated absence of MC2 mRNA in healthy tissues, as illustrated in Fig. 1A. In case specific antibodies are available, we would recommend to follow up qPCR with IHC. Using an MC2-specific antibody, we confirmed the presence of MC2 protein in testis and melanoma as well as its absence in multiple healthy tissues, such as brain, heart, intestine, and lung (see Fig. 1B). However, both qPCR and IHC cannot formally exclude the presence of rare antigen-positive cells within a tissue, for example, stem cells. For example, mRNA of certain CGAs has been detected only in medullary thymic epithelial cells, but not total thymus (17). As an additional means to exclude target antigen expression, sophisticated in vitro cell cultures have been developed to closely mimic complete tissues or organs (commented on in the next section; refs. 5, 22).

Figure 1.

Target antigen is absent or shows restricted expression in healthy tissues. A, MC2, MC1, and MB4 qPCRs were performed on a cDNA library of 48 healthy human tissue samples (OriGene Technologies). Patient-derived melanoma cell line EB81-MEL and patient-derived esophageal cancer cell line OEC-19 served as positive controls. Spiking experiments were conducted separately to determine optimal PCR conditions for maximum sensitivity (detection limit: 1:1,000–10,000 MC2+ cells: MC2 cells; data not shown). Relative mRNA levels are corrected for GAPDH and expressed as fold increase compared with antigen expression in the testis. Part of the displayed data is derived from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc. B, Immune stainings of melanoma and healthy tissues were conducted using a mAb specific for MC2 (42) on tissue microarrays (TMA). EnVision System (Agilent Technologies) was used for signal visualization; TMAs were scanned via NanoZoomer (Hamatsu) and manually scored using Distiller (SlidePath). Exemplary tissue sections of patient-derived melanoma tissue as well as various healthy tissues obtained from the Erasmus MC Pathology Department are displayed with ×20 magnification. MC2 positivity is scored as brown-colored nuclear staining, with three smaller TMAs exemplifying different staining intensities.

Figure 1.

Target antigen is absent or shows restricted expression in healthy tissues. A, MC2, MC1, and MB4 qPCRs were performed on a cDNA library of 48 healthy human tissue samples (OriGene Technologies). Patient-derived melanoma cell line EB81-MEL and patient-derived esophageal cancer cell line OEC-19 served as positive controls. Spiking experiments were conducted separately to determine optimal PCR conditions for maximum sensitivity (detection limit: 1:1,000–10,000 MC2+ cells: MC2 cells; data not shown). Relative mRNA levels are corrected for GAPDH and expressed as fold increase compared with antigen expression in the testis. Part of the displayed data is derived from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc. B, Immune stainings of melanoma and healthy tissues were conducted using a mAb specific for MC2 (42) on tissue microarrays (TMA). EnVision System (Agilent Technologies) was used for signal visualization; TMAs were scanned via NanoZoomer (Hamatsu) and manually scored using Distiller (SlidePath). Exemplary tissue sections of patient-derived melanoma tissue as well as various healthy tissues obtained from the Erasmus MC Pathology Department are displayed with ×20 magnification. MC2 positivity is scored as brown-colored nuclear staining, with three smaller TMAs exemplifying different staining intensities.

Close modal

Once the safety of the target antigen has been assessed, one can start selecting TCRs. Procedures to obtain tumor-reactive T cells and hence TCRs can generally be divided into those that rely on tolerant and those that rely on nontolerant repertoires of T cells. Tolerant repertoires, where deletion of T cells with an avidity outside the thymic selection window has occurred, have been used to obtain T-cell clones from patients following successful TIL therapy, following peptide vaccination, or using in vitro pulsing of autologous dendritic cells (23, 24). Notably, the thymic selection of T cells is most likely a trade-off between producing a self-tolerant yet sufficiently diverse and responsive TCR repertoire, and escape of self-reactive TCRs cannot be negated. Indeed, TCRs specific for overexpressed antigens and obtained from the native repertoire have been shown to initiate autoimmune side effects (1). The use of nontolerant repertoires, with the rationale of allowing the generation of high-avidity T cells, has been applied in allogeneic in vitro as well as in vivo systems. For example, HLA-mismatched antigen-presenting cells (25) or artificial antigen-presenting cells pulsed with peptides of interest facilitate the in vitro generation of tumor-reactive T cells (26). In vivo, mice transgenic for human HLA as well as mice transgenic for human TCR and HLA-A2 genes (27) have been immunized and used as a source of TCRs. After having obtained antigen-reactive T cells, sequences of the TCRα and β chains can be determined by molecular techniques such as 5′ RACE (23), enhanced PCR methods, capturing and indexing of genomic DNA-encoding TCR chains (28), or sequencing and pairing of TCR chains based on combinatorial algorithms (29).

It is important to note that TCRs derived from the aforementioned repertoires, in particular the nontolerant repertoir, have not been selected in the presence of all patient MHC alleles and in the case of mice neither against human peptides present in the thymus, and may show allo- and noncognate reactivity. Moreover, TCRs, even though obtained from highly tumor-reactive T cells, have an inherent degeneracy for peptide recognition and are able to recognize more than a single peptide. This dynamic flexibility in antigen recognition is in part credited not only to the bending ability of the TCR–CDR domains (30, 31) but also to the dominant interaction of the TCR with a restricted number and order of amino acids present in the MHC-presented peptide. To minimize the risk of selecting a TCR that recognizes noncognate self-peptides, we recommend a series of assays that assess the risk of this so-called off-target toxicity. These assays are illustrated with two patient-derived, MC2-specific TCRs, 6 and 16, with details and evaluation of antitumor T-cell responses mediated by TCR6 and TCR16 summarized in Table 1.

Recognition of random epitopes

Initial assessment of a TCR's self-reactivity can be done by testing the responsiveness of TCR-transduced T cells toward random peptides known to be presented by the respective HLA restriction allele. Mathematical projections indicate that among a pool of approximately 1012 peptides, a single TCR may react with >106 peptides, supporting the notion of TCR degeneracy for peptide recognition, which potentially contributes to a more diverse TCR repertoire (32). We cocultured TCR6 and TCR16 T cells with antigen-presenting cells loaded with saturating concentrations of >100 common, HLA-A2–eluted self-peptides (4). As depicted in Fig. 2, both T-cell populations mediated a T-cell response to their respective cognate peptides but to none of the other peptides. Importantly, these data hint at a lack of cross-reactivity of these two TCRs, but we cannot fully exclude recognition to random peptides. Another assessment of self-reactivity can be conducted by testing TCR-transduced T cells toward allogenic HLA molecules. To exclude activation of T cells upon recognition of foreign HLA molecules, panels of lymphoblastoid B cell lines with various HLA allotypes have proven valuable (33, 34).

Figure 2.

TCRs do not recognize random peptides eluted from the restricting MHC allele. TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of 114 different HLA-A2–eluted peptides (4) for 24 hours. Cognate MC2 peptides (TCR6: LLFGLALIEV; TCR16: ALKDVEERV) served as positive controls. IFNγ levels in 24-hour culture supernatants were measured by ELISA and are displayed as mean ± SEM (n = 3). Left-hand panel is adapted from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc.

Figure 2.

TCRs do not recognize random peptides eluted from the restricting MHC allele. TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of 114 different HLA-A2–eluted peptides (4) for 24 hours. Cognate MC2 peptides (TCR6: LLFGLALIEV; TCR16: ALKDVEERV) served as positive controls. IFNγ levels in 24-hour culture supernatants were measured by ELISA and are displayed as mean ± SEM (n = 3). Left-hand panel is adapted from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc.

Close modal

Recognition of cognate epitope via critical amino acids

Further assessment of a TCR's self-reactivity, and a key assay in this communication, is the testing of a TCR's intrinsic capacity to recognize peptides highly homologous to its cognate epitope (35, 36). To this end, one can determine the recognition motif, that is, the position and sequence of amino acids within the cognate epitopes that are crucial for binding to the TCR. This motif is unique per TCR and can be considered a surrogate measure for the extent of cross-reactivity of TCRs (22, 33). The importance to assess such motifs became apparent from two recent clinical trials using AT with TCR-engineered T cells. The first trial targeting MAGE-A3 and A9 (MA3/9) in the context of HLA-A2 utilized the TCR 9W11 and reported neurologic toxicities in two patients with metastatic melanoma (4). The second trial targeting MA3 in the context of HLA-A1 utilized the TCR a3a and reported cardiac toxicities in one patient with metastatic melanoma and one patient with multiple myeloma (5). Both TCRs were affinity enhanced in vitro and mediated toxicity by recognizing peptides highly similar to the cognate peptide, namely peptides derived from MAGE-A12 and Titin present in brain and heart tissue, respectively. These studies clearly underline the need to assess recognition motifs and search for T-cell reactivities against homologous self-peptides prior to clinical application. The importance of recognition motifs is timely, and its assessment has only occurred for a limited number of TCRs to date, which is summarized in Table 2.

Table 2.

Recognition motifs of TCRs utilized in clinical and preclinical research

TCR nameTarget antigenHLA restrictionCognate epitopeRecognition motifaNumber of antigens with motifbNumber of epitopes with high MHC affinity (<10 μmol/L)cReference
a3a MAGE-A3 A1 EVDPIGHLY ExDPIxxxY 22 
9W11 MAGE-A3 A2 KVAELVHFL -xxE-xH–d — — 43 
T1367 MAGE-A1 A2 KVLEYVIKV xxxEYxIKx 62 33 
s24-TCR survivin A2 ELTLGEFLKL xLTxGEFLKx 44 
gp100 wt gp100 A2 YLEPGPVTA xLEPGPxxA C. Govers; submitted for publication 
fl-MPD gp100 A2 YLEPGPVTA YxEPxxxxx >1,000 >500 45 
fl-296 gp100 A2 YLEPGPVTA YxExxxxxx >1,000 >500 45 
TCR 4 MAGE-C2 A2 LLFGLALIEV xxFGLxLxxx 260 122 23 
TCR 6 MAGE-C2 A2 LLFGLALIEV LxFxLxLxEx 28 23 
TCR 11 MAGE-C2 A2 LLFGLALIEV xxFGLxLxEx 21 17 23 
TCR 16 MAGE-C2 A2 ALKDVEERV xLKDVEERx 23 
TCR nameTarget antigenHLA restrictionCognate epitopeRecognition motifaNumber of antigens with motifbNumber of epitopes with high MHC affinity (<10 μmol/L)cReference
a3a MAGE-A3 A1 EVDPIGHLY ExDPIxxxY 22 
9W11 MAGE-A3 A2 KVAELVHFL -xxE-xH–d — — 43 
T1367 MAGE-A1 A2 KVLEYVIKV xxxEYxIKx 62 33 
s24-TCR survivin A2 ELTLGEFLKL xLTxGEFLKx 44 
gp100 wt gp100 A2 YLEPGPVTA xLEPGPxxA C. Govers; submitted for publication 
fl-MPD gp100 A2 YLEPGPVTA YxEPxxxxx >1,000 >500 45 
fl-296 gp100 A2 YLEPGPVTA YxExxxxxx >1,000 >500 45 
TCR 4 MAGE-C2 A2 LLFGLALIEV xxFGLxLxxx 260 122 23 
TCR 6 MAGE-C2 A2 LLFGLALIEV LxFxLxLxEx 28 23 
TCR 11 MAGE-C2 A2 LLFGLALIEV xxFGLxLxEx 21 17 23 
TCR 16 MAGE-C2 A2 ALKDVEERV xLKDVEERx 23 

aRecogniton motifs are defined through T-cell IFNγ production in response to alanine-scanned cognate epitopes.

bNumber of human proteins containing matching recognition motif according to ScanProSite (http://prosite.expasy.org/scanprosite/).

cNumber of proteins containing matching recognition motif (according to b) and a predicted affinity value <10 μmol/L for binding of the peptide to its respective MHC; affinity calculations according to NetMHCpan (http://www.cbs.dtu.dk/services/NetMHCpan/).

dRecognition motif incomplete; amino acids on positions 2–4 and 6–7 are based on stimulation assays conducted by Chinnasamy and colleagues with a panel of MAGE peptides with highly similar sequences (43); “—” indicates amino acids with unknown relevance to the recognition motif.

Using a set of altered peptide ligands (APL), peptides containing individual alanine replacements at every single position in the cognate peptide (in case of an endogenous alanine→glycine), we conducted stimulation assays with TCR6 and 16T cells. Critical amino acids are defined as those that, when testing the respective APLs, result in a drop of the T-cell response (generally using IFNγ production as a readout) of >50% when compared with the response toward the cognate peptide. Following the determination of these motifs, as exemplified in Fig. 3A, we used ScanProSite (http://prosite.expasy.org/scanprosite/; ref. 37) and identified target antigens, listed in Fig. 3B, that harbor the recognition motif and represent potential cross-reactive targets for TCRs 6 and 16. Subsequently, these self-peptides were tested for their ability to induce T-cell responses toward HLA-A2–postive antigen-presenting cells loaded with saturating concentrations (1 μmol/L) of peptide. These experiments yielded a short list of self-peptides that are actually recognized by the TCRs under study, defined as those that resulted in at least a T-cell IFNγ response >2.5% of the response to cognate peptide (Fig. 3B: underlined antigens).

Figure 3.

TCRs recognize cognate peptide via unique or restricted motif. A, TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of cognate peptides or peptides with a single alanine replacement (APL; in case of alanine in original peptide→glycine). IFNγ levels in 24-hour culture supernatants were measured by ELISA. IFNγ response to APLs is displayed as mean % relative to response to cognate peptides ± SEM (n = 4). Responses <50% (dashed line) were indicative of amino acids critical for TCR recognition (recognition motif: underlined amino acids). B, Homologous motifs from A were queried against a human protein database using ScanProSite. This yielded 27 and one noncognate matches for TCR6 and TCR16, respectively. Subsequently, TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of these 27 and one peptide(s), and IFNγ levels were measured in 24-hour culture supernatants by ELISA (n = 4). Underlined peptides induced a T-cell IFNγ response >2.5% of the response to cognate peptide. Homology to cognate peptide (diverging amino acids underlined) as well as peptide–MHC affinity (IC50 calculations according to NetMHCpan; http://www.cbs.dtu.dk/services/NetMHCpan/) are indicated for those peptides with a detectable IFNγ response. Major facilitator superf, domain-contain. Prot. 1, Major facilitator superfamily domain containing protein 1; μM, μmol/L.

Figure 3.

TCRs recognize cognate peptide via unique or restricted motif. A, TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of cognate peptides or peptides with a single alanine replacement (APL; in case of alanine in original peptide→glycine). IFNγ levels in 24-hour culture supernatants were measured by ELISA. IFNγ response to APLs is displayed as mean % relative to response to cognate peptides ± SEM (n = 4). Responses <50% (dashed line) were indicative of amino acids critical for TCR recognition (recognition motif: underlined amino acids). B, Homologous motifs from A were queried against a human protein database using ScanProSite. This yielded 27 and one noncognate matches for TCR6 and TCR16, respectively. Subsequently, TCR6 or TCR16 T cells were cocultured with T2 cells loaded with 1 μmol/L of these 27 and one peptide(s), and IFNγ levels were measured in 24-hour culture supernatants by ELISA (n = 4). Underlined peptides induced a T-cell IFNγ response >2.5% of the response to cognate peptide. Homology to cognate peptide (diverging amino acids underlined) as well as peptide–MHC affinity (IC50 calculations according to NetMHCpan; http://www.cbs.dtu.dk/services/NetMHCpan/) are indicated for those peptides with a detectable IFNγ response. Major facilitator superf, domain-contain. Prot. 1, Major facilitator superfamily domain containing protein 1; μM, μmol/L.

Close modal

Recognition of noncognate epitopes that contain recognition motif

Once self-peptides that can be recognized by TCR T cells have been identified, we recommend executing two additional tests to more stringently assess the risk for self-reactivity. These tests aim to provide measures for T-cell avidity as well as efficiency of cellular processing and presentation. Toward the first test, one can titrate amounts of noncognate peptides and determine the concentration of these peptides that elicits 50% of the maximal T-cell response (EC50). For both TCR6 and 16 T cells, we found that only a single self-peptide, namely a peptide from the antigen MAGE-B4 (MB4) or MAGE-C1 (MC1; see Fig. 3B for homology and predicted peptide–HLA binding), respectively, revealed detectable EC50 values that were only 2- to 5-fold lower than those of the cognate peptides (Fig. 4). Extent of homology and predicted peptide–MHC binding of peptides that induce T-cell IFNγ are listed in Fig. 3B, which shows that loss of homology and peptide–MHC affinity was least affected for MB4 and MC1 peptides. All other self-peptides revealed no T-cell reactivities at titrated doses, or at the very best 5 log scales lower compared with cognate peptide. These were considered not recognizable by the TCRs under study and excluded from further assays. Toward the second test, one can predict whether self-peptides are the result of antigen processing and presentation to enable T-cell recognition in vitro. NetCTLpan (http://www.cbs.dtu.dk/services/NetCTLpan/; ref. 38) takes proteasomal C terminal cleavage, TAP transport efficiency, and peptide MHC class I binding of peptides into account and can be employed to obtain an initial score for antigen processing and presentation. Analysis of the MB4 peptide, but not MC1 peptide, yielded a high score according to this Web-based tool. Such predictions may not be fully accurate and should be verified using cells known to express the antigen or antigen-presenting cells (e.g., dentritic cells) transfected with antigen-encoded RNA followed by cocultivation with TCR-transduced T cells. To this end, we stimulated TCR T cells with the esophageal cancer cells line OEC-19, which natively expresses the MB4 and MC1 antigens but is devoid of the MC2 antigen, and observed that both TCRs failed to initiate T-cell activation against either MB4 or MC1 (see Fig. 4B). When using cell lines that natively express the MC2 antigen, as a control for the processing and presentation of MAGE antigens, we observed that both TCRs did initiate T-cell activation. It is noteworthy that standard tissue culture systems may not always accurately reflect antigen processing and presentation. This was evidenced by the recognition of Titin by the TCR a3a that could only be observed in more elaborate tissue culture systems, such as 3D cultures of beating cardiomyocytes derived from induced pluripotent stem cells (22). In case the aforementioned two assays do not exclude self-reactivity of TCR T cells, one could pursue assessment of the tumor-selective expression of such new antigens. In case the expression of new antigens is not selective for tumors, the corresponding TCR should be excluded. Using online tools (39) as well as qPCR (Fig. 1A), MB4 showed expression within epididymis and vagina, whereas MC1 showed no expression in any of the healthy non-gonadal tissues. These data highlight the stringent safety profile of TCR 16, the TCR selected for a clinical trial to treat melanoma and head and neck carcinoma, currently prepared at Erasmus MC (Rotterdam, the Netherlands).

Figure 4.

TCRs mediate negligible T-cell avidity and do not recognize natively presented noncognate peptides that harbor recognition motif. A, Noncognate peptides underlined in Fig. 3B were titrated from 10 μmol/L (10−5 mol/L) to 1 pmol/L (10−12 mol/L) and tested for T-cell IFNγ response as described in previous legends and displayed as mean ± SEM (n = 4). EC50 values for cognate and selected noncognate peptides were calculated in GraphPad, using nonlinear regression; n.d., not detectable. B, TCR6 or TCR16 T cells were cocultured with tumor cell lines or T2 cells (as a negative control). Expression status of MC2, MC1, MB4, and HLA-A2 for these cells was assessed via qPCR and indicated below plots with plus or minus symbols. IFNγ levels in 24-hour culture supernatants were measured by ELISA and are displayed as mean ± SEM (n = 3). Left-hand plots are adapted from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc. nM, nmol/L.

Figure 4.

TCRs mediate negligible T-cell avidity and do not recognize natively presented noncognate peptides that harbor recognition motif. A, Noncognate peptides underlined in Fig. 3B were titrated from 10 μmol/L (10−5 mol/L) to 1 pmol/L (10−12 mol/L) and tested for T-cell IFNγ response as described in previous legends and displayed as mean ± SEM (n = 4). EC50 values for cognate and selected noncognate peptides were calculated in GraphPad, using nonlinear regression; n.d., not detectable. B, TCR6 or TCR16 T cells were cocultured with tumor cell lines or T2 cells (as a negative control). Expression status of MC2, MC1, MB4, and HLA-A2 for these cells was assessed via qPCR and indicated below plots with plus or minus symbols. IFNγ levels in 24-hour culture supernatants were measured by ELISA and are displayed as mean ± SEM (n = 3). Left-hand plots are adapted from ref. 23; Copyright © 2016 The American Association of Immunologists, Inc. nM, nmol/L.

Close modal

The proposed collection and sequence of in vitro assays to assess risks for toxicities are presented in Fig. 5. We advocate this testing for TCRs with clinical intent, in particular those TCRs reactive against a self-peptide and derived from a nontolerant repertoire and/or following gene enhancement. In extension to gene enhancement, introduction of TCR–CDR mutations has been a commonly used tool to generate high-affinity TCRs (2, 4, 5, 20). Although such gene-enhanced TCRs recognize target peptides at increased affinities when compared with the corresponding wild-type TCRs, consequently, such TCRs are also at risk to recognize noncognate peptides. To test whether affinity enhancement led to an increase in degeneracy for peptide recognition, we made use of a panel of eight TCRs specific for the same cognate peptide gp100181-188:HLA-A2) but harboring two to three mutated amino acids in either their CDR2β, CDR3α, or CDR3β domains (C. Govers; submitted for publication). Upon assessment of the recognition motifs and search for motif-harboring self-peptides, it became apparent that enhanced affinity was accompanied by a drastic increase in the TCR's ability to recognize self-peptides (Table 3). These data extend earlier findings regarding a correlation between affinity enhancement and loss of TCR specificity (40) and warrant caution when trying to change the TCR–CDR structure, as it compromises the stringet recognition of cognate peptide (31, 41).

Figure 5.

Platform of in vitro assays to select antigens and TCRs with limited risk for in vivo toxicity. This flowchart proposes a series and sequence of in vitro techniques, exemplified in Figs. 1 to 4 and explained in detail in text. Such a sequence of assays would facilitate selections of potentially safe target antigens and TCRs prior to their use in clinical trials.

Figure 5.

Platform of in vitro assays to select antigens and TCRs with limited risk for in vivo toxicity. This flowchart proposes a series and sequence of in vitro techniques, exemplified in Figs. 1 to 4 and explained in detail in text. Such a sequence of assays would facilitate selections of potentially safe target antigens and TCRs prior to their use in clinical trials.

Close modal
Table 3.

TCR affinity enhancement and its effect on off-target recognitiona

TCR nameKD (μmol/L)Target antigenCognate epitopeRecognition motifaNumber of antigens with motifbNumber of epitopes with high MHC affinity (<10 μmol/L)c
gp100 wt 18.5 gp100/HLA-A2 YLEPGPVTA xLEPGPxxA 
gp100 TCR 1 7.9 gp100/HLA-A2 YLEPGPVTA xLExGPxxA 23 13 
gp100 TCR 2 4.0 gp100/HLA-A2 YLEPGPVTA xLExGPxxx 240 97 
gp100 TCR 5 1.1 gp100/HLA-A2 YLEPGPVTA xLExGPxxx 240 97 
gp100 TCR 8 0.026 gp100/HLA-A2 YLEPGPVTA xLxxxxxxx >10,000 >10,000 
TCR nameKD (μmol/L)Target antigenCognate epitopeRecognition motifaNumber of antigens with motifbNumber of epitopes with high MHC affinity (<10 μmol/L)c
gp100 wt 18.5 gp100/HLA-A2 YLEPGPVTA xLEPGPxxA 
gp100 TCR 1 7.9 gp100/HLA-A2 YLEPGPVTA xLExGPxxA 23 13 
gp100 TCR 2 4.0 gp100/HLA-A2 YLEPGPVTA xLExGPxxx 240 97 
gp100 TCR 5 1.1 gp100/HLA-A2 YLEPGPVTA xLExGPxxx 240 97 
gp100 TCR 8 0.026 gp100/HLA-A2 YLEPGPVTA xLxxxxxxx >10,000 >10,000 

aC. Govers; submitted for publication.

bNumber of human proteins containing matching recognition motif according to ScanProSite (http://prosite.expasy.org/scanprosite/).

cNumber of human proteins containing matching recognition motif (according to b) and a predicted affinity value of <10 μmol/L for binding of the peptide to its respective MHC; affinity calculations according to NetMHCpan (http://www.cbs.dtu.dk/services/NetMHCpan/).

Taken together, here, we propose a platform of in vitro assays that in combination with available online databases and tools allows for optimal toxicity risk assessment for target antigens and TCRs currently under consideration for clinical trials.

No potential conflicts of interest were disclosed.

The authors would like to thank Mandy van Brakel and Mieke Timmermans for technical support toward Table 1 and Fig. 1B, respectively.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Johnson
LA
,
Morgan
RA
,
Dudley
ME
,
Cassard
L
,
Yang
JC
,
Hughes
MS
, et al
Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen
.
Blood
2009
;
114
:
535
46
.
2.
Parkhurst
MR
,
Yang
JC
,
Langan
RC
,
Dudley
ME
,
Nathan
DA
,
Feldman
SA
, et al
T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis
.
Mol Ther
2011
;
19
:
620
6
.
3.
Chodon
T
,
Comin-Anduix
B
,
Chmielowski
B
,
Koya
RC
,
Wu
Z
,
Auerbach
M
, et al
Adoptive transfer of MART-1 T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with metastatic melanoma
.
Clin Cancer Res
2014
;
20
:
2457
65
.
4.
Morgan
RA
,
Chinnasamy
N
,
Abate-Daga
D
,
Gros
A
,
Robbins
PF
,
Zheng
Z
, et al
Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy
.
J Immunother
2013
;
36
:
133
51
.
5.
Linette
GP
,
Stadtmauer
EA
,
Maus
MV
,
Rapoport
AP
,
Levine
BL
,
Emery
L
, et al
Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma
.
Blood
2013
;
122
:
863
71
.
6.
Kunert
A
,
Straetemans
T
,
Govers
C
,
Lamers
C
,
Mathijssen
R
,
Sleijfer
S
, et al
TCR-engineered T cells meet new challenges to treat solid tumors: choice of antigen, T cell fitness, and sensitization of tumor milieu
.
Front Immunol
2013
;
4
:
363
.
7.
Brown
SD
,
Warren
RL
,
Gibb
EA
,
Martin
SD
,
Spinelli
JJ
,
Nelson
BH
, et al
Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival
.
Genome Res
2014
;
24
:
743
50
.
8.
van Rooij
N
,
van Buuren
MM
,
Philips
D
,
Velds
A
,
Toebes
M
,
Heemskerk
B
, et al
Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma
.
J Clin Oncol
2013
;
31
:
e439
42
.
9.
Lu
YC
,
Yao
X
,
Crystal
JS
,
Li
YF
,
El-Gamil
M
,
Gross
C
, et al
Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions
.
Clin Cancer Res
2014
;
20
:
3401
10
.
10.
Tran
E
,
Ahmadzadeh
M
,
Lu
YC
,
Gros
A
,
Turcotte
S
,
Robbins
PF
, et al
Immunogenicity of somatic mutations in human gastrointestinal cancers
.
Science
2015
;
350
:
1387
90
.
11.
Verdegaal
EM
,
de Miranda
NF
,
Visser
M
,
Harryvan
T
,
van Buuren
MM
,
Andersen
RS
, et al
Neoantigen landscape dynamics during human melanoma-T cell interactions
.
Nature
2016
;
536
:
91
5
.
12.
Angelova
M
,
Charoentong
P
,
Hackl
H
,
Fischer
ML
,
Snajder
R
,
Krogsdam
AM
, et al
Characterization of the immunophenotypes and antigenomes of colorectal cancers reveals distinct tumor escape mechanisms and novel targets for immunotherapy
.
Genome Biol
2015
;
16
:
64
.
13.
Blankenstein
T
,
Leisegang
M
,
Uckert
W
,
Schreiber
H
. 
Targeting cancer-specific mutations by T cell receptor gene therapy
.
Curr Opin Immunol
2015
;
33
:
112
9
.
14.
The problem with neoantigen prediction
.
Nat Biotechnol
2017
;
35
:
97
.
15.
Li
J
,
Chen
QY
,
He
J
,
Li
ZL
,
Tang
XF
,
Chen
SP
, et al
Phase I trial of adoptively transferred tumor-infiltrating lymphocyte immunotherapy following concurrent chemoradiotherapy in patients with locoregionally advanced nasopharyngeal carcinoma
.
Oncoimmunology
2015
;
4
:
e976507
.
16.
Stevanovic
S
,
Draper
LM
,
Langhan
MM
,
Campbell
TE
,
Kwong
ML
,
Wunderlich
JR
, et al
Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells
.
J Clin Oncol
2015
;
33
:
1543
50
.
17.
Gotter
J
,
Brors
B
,
Hergenhahn
M
,
Kyewski
B
. 
Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters
.
J Exp Med
2004
;
199
:
155
66
.
18.
Debets
R
,
Donnadieu
E
,
Chouaib
S
,
Coukos
G
. 
TCR-engineered T cells to treat tumors: Seeing but not touching?
Semin Immunol
2016
;
28
:
10
21
.
19.
Hong
JA
,
Kang
Y
,
Abdullaev
Z
,
Flanagan
PT
,
Pack
SD
,
Fischette
MR
, et al
Reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter coincides with derepression of this cancer-testis gene in lung cancer cells
.
Cancer Res
2005
;
65
:
7763
74
.
20.
Robbins
PF
,
Morgan
RA
,
Feldman
SA
,
Yang
JC
,
Sherry
RM
,
Dudley
ME
, et al
Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1
.
J Clin Oncol
2011
;
29
:
917
24
.
21.
Rapoport
AP
,
Stadtmauer
EA
,
Binder-Scholl
GK
,
Goloubeva
O
,
Vogl
DT
,
Lacey
SF
, et al
NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma
.
Nat Med
2015
;
21
:
914
21
.
22.
Cameron
BJ
,
Gerry
AB
,
Dukes
J
,
Harper
JV
,
Kannan
V
,
Bianchi
FC
, et al
Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells
.
Sci Transl Med
2013
;
5
:
197ra03
.
23.
Kunert
A
,
van Brakel
M
,
van Steenbergen-Langeveld
S
,
da Silva
M
,
Coulie
PG
,
Lamers
C
, et al
MAGE-C2-Specific TCRs combined with epigenetic drug-enhanced antigenicity yield robust and tumor-selective T cell responses
.
J Immunol
2016
;
197
:
2541
52
.
24.
Lurquin
C
,
Lethe
B
,
De Plaen
E
,
Corbiere
V
,
Theate
I
,
van Baren
N
, et al
Contrasting frequencies of antitumor and anti-vaccine T cells in metastases of a melanoma patient vaccinated with a MAGE tumor antigen
.
J Exp Med
2005
;
201
:
249
57
.
25.
Sadovnikova
E
,
Jopling
LA
,
Soo
KS
,
Stauss
HJ
. 
Generation of human tumor-reactive cytotoxic T cells against peptides presented by non-self HLA class I molecules
.
Eur J Immunol
1998
;
28
:
193
200
.
26.
Butler
MO
,
Friedlander
P
,
Milstein
MI
,
Mooney
MM
,
Metzler
G
,
Murray
AP
, et al
Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells
.
Sci Transl Med
2011
;
3
:
80ra34
.
27.
Li
LP
,
Lampert
JC
,
Chen
X
,
Leitao
C
,
Popovic
J
,
Muller
W
, et al
Transgenic mice with a diverse human T cell antigen receptor repertoire
.
Nat Med
2010
;
16
:
1029
34
.
28.
Linnemann
C
,
van Buuren
MM
,
Bies
L
,
Verdegaal
EM
,
Schotte
R
,
Calis
JJ
, et al
High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma
.
Nat Med
2015
;
21
:
81
5
.
29.
Howie
B
,
Sherwood
AM
,
Berkebile
AD
,
Berka
J
,
Emerson
RO
,
Williamson
DW
, et al
High-throughput pairing of T cell receptor alpha and beta sequences
.
Sci Transl Med
2015
;
7
:
301ra131
.
30.
Mason
D
. 
A very high level of crossreactivity is an essential feature of the T-cell receptor
.
Immunol Today
1998
;
19
:
395
404
.
31.
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
2011
;
414
:
385
400
.
32.
Wooldridge
L
,
Ekeruche-Makinde
J
,
van den Berg
HA
,
Skowera
A
,
Miles
JJ
,
Tan
MP
, et al
A single autoimmune T cell receptor recognizes more than a million different peptides
.
J Biol Chem
2012
;
287
:
1168
77
.
33.
Obenaus
M
,
Leitao
C
,
Leisegang
M
,
Chen
X
,
Gavvovidis
I
,
van der Bruggen
P
, et al
Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice
.
Nat Biotechnol
2015
;
33
:
402
7
.
34.
Heemskerk
MH
,
de Paus
RA
,
Lurvink
EG
,
Koning
F
,
Mulder
A
,
Willemze
R
, et al
Dual HLA class I and class II restricted recognition of alloreactive T lymphocytes mediated by a single T cell receptor complex
.
Proc Natl Acad Sci U S A
2001
;
98
:
6806
11
.
35.
Wilson
DB
,
Wilson
DH
,
Schroder
K
,
Pinilla
C
,
Blondelle
S
,
Houghten
RA
, et al
Specificity and degeneracy of T cells
.
Mol Immunol
2004
;
40
:
1047
55
.
36.
Birnbaum
ME
,
Mendoza
JL
,
Sethi
DK
,
Dong
S
,
Glanville
J
,
Dobbins
J
, et al
Deconstructing the peptide-MHC specificity of T cell recognition
.
Cell
2014
;
157
:
1073
87
.
37.
de Castro
E
,
Sigrist
CJ
,
Gattiker
A
,
Bulliard
V
,
Langendijk-Genevaux
PS
,
Gasteiger
E
, et al
ScanProsite: detection of PROSITE signature matches and ProRule-associated functional and structural residues in proteins
.
Nucleic Acids Res
2006
;
34
:
W362
5
.
38.
Stranzl
T
,
Larsen
MV
,
Lundegaard
C
,
Nielsen
M
. 
NetCTLpan: pan-specific MHC class I pathway epitope predictions
.
Immunogenetics
2010
;
62
:
357
68
.
39.
Hofmann
O
,
Caballero
OL
,
Stevenson
BJ
,
Chen
YT
,
Cohen
T
,
Chua
R
, et al
Genome-wide analysis of cancer/testis gene expression
.
Proc Natl Acad Sci U S A
2008
;
105
:
20422
7
.
40.
Tan
MP
,
Gerry
AB
,
Brewer
JE
,
Melchiori
L
,
Bridgeman
JS
,
Bennett
AD
, et al
T cell receptor binding affinity governs the functional profile of cancer-specific CD8+ T cells
.
Clin Exp Immunol
2015
;
180
:
255
70
.
41.
Burrows
SR
,
Chen
Z
,
Archbold
JK
,
Tynan
FE
,
Beddoe
T
,
Kjer-Nielsen
L
, et al
Hard wiring of T cell receptor specificity for the major histocompatibility complex is underpinned by TCR adaptability
.
Proc Natl Acad Sci U S A
2010
;
107
:
10608
13
.
42.
Zhuang
R
,
Zhu
Y
,
Fang
L
,
Liu
XS
,
Tian
Y
,
Chen
LH
, et al
Generation of monoclonal antibodies to cancer/testis (CT) antigen CT10/MAGE-C2
.
Cancer Immun
2006
;
6
:
7
.
43.
Chinnasamy
N
,
Wargo
JA
,
Yu
Z
,
Rao
M
,
Frankel
TL
,
Riley
JP
, 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
.
J Immunol
2011
;
186
:
685
96
.
44.
Arber
C
,
Feng
X
,
Abhyankar
H
,
Romero
E
,
Wu
MF
,
Heslop
HE
, et al
Survivin-specific T cell receptor targets tumor but not T cells
.
J Clin Invest
2015
;
125
:
157
68
.
45.
Schaft
N
,
Willemsen
RA
,
de Vries
J
,
Lankiewicz
B
,
Essers
BW
,
Gratama
JW
, et al
Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCR alpha beta genes into primary human T lymphocytes
.
J Immunol
2003
;
170
:
2186
94
.