Engineering immune cells to target cancer is a rapidly advancing technology. The first commercial products, chimeric-antigen receptor (CAR) T cells, are now approved for hematologic malignancies. However, solid tumors pose a greater challenge for cellular therapy, in part because suitable cancer-specific antigens are more difficult to identify and surrounding healthy tissues are harder to avoid. In addition, impaired trafficking of immune cells to solid tumors, the harsh immune-inhibitory microenvironment, and variable antigen density and presentation help tumors evade immune cells targeting cancer-specific antigens. To overcome these obstacles, T cells are being engineered to express defined T-cell receptors (TCR). Given that TCRs target intracellular peptides expressed on tumor MHC molecules, this provides an expanded pool of potential targetable tumor-specific antigens relative to the cell-surface antigens that are targeted by CAR T cells. The affinity of TCR T cells can be tuned to allow for better tumor recognition, even with varying levels of antigen presentation on the tumor and surrounding healthy tissue. Further enhancements to TCR T cells include improved platforms that enable more robust cell expansion and persistence; coadministration of small molecules that enhance tumor recognition and immune activation; and coexpression of cytokine-producing moieties, activating coreceptors, or mediators that relieve checkpoint blockade. Early-phase clinical trials pose logistical challenges involving production, large-scale manufacturing, and more. The challenges and obstacles to successful TCR T-cell therapy, and ways to overcome these and improve anticancer activity and efficacy, are discussed herein.

Adoptive cell therapy is a rapidly advancing field of medicine that is now focused mostly on cancer immunotherapy but already extending to immune manipulation against infections and autoimmune diseases. One of the first FDA-approved cellular therapy products was sipuleucel-T, which consists of autologous antigen-presenting cells (APC) cultured ex vivo and administered to patients with prostate cancer. Newer classes of cellular therapy include tumor-infiltrating lymphocytes (TIL), which recognize tumor-associated antigens (TAA), but are often suppressed in the tumor microenvironment (TME), precluding efficient killing of cancer cells. Nevertheless, TILs, which are isolated from tumor samples, show clinical efficacy after expansion in culture, especially if the patient was not previously exposed to checkpoint inhibitors (1). TIL therapy was recently combined with checkpoint inhibition or vaccination with tumor lysate–loaded dendritic cells (DC), and this elicited prolonged antitumor responses in some patients (2). Indeed, TILs have shown promising preliminary results against various tumor types, mainly melanoma, and research is ongoing to develop ways to more carefully select and expand TILs aimed at specific TAAs, and to enhance TIL antitumor efficacy.

Advances in genetic engineering have also enabled production of cells with receptors directed at antigens presented on cancer cells. The chimeric-antigen receptor (CAR) T-cell therapies tisagenlecleucel, axicabtagene ciloleucel, idecabtagene vicleucel, and lisocabtagene maraleucel became the first commercially approved engineered cellular products after showing remarkable efficacy against hematologic malignancies. Unfortunately, CAR T cells have been less effective against solid tumors (3). CAR T cells can only detect surface TAAs, yet suitable antigens are challenging to identify in many tumors, especially since most aberrant cancer proteins are intracellular and are only expressed in the context of MHC complexes after proteasomal degradation.

Constructs encoding a T-cell receptor (TCR) that recognizes MHC–antigen complexes can be generated and introduced into T cells (known as TCR T cells), and the ensuing TCR–peptide-MHC interaction can be exploited to trigger an immune response (Fig. 1). Importantly, TCR T cells elicit lower maximal cytokine levels at high antigen density, whereas some CAR T cells can prompt significant immune toxicities that may even be life threatening (4). In this review, we summarize preliminary TCR T-cell clinical trials, and survey preclinical studies seeking to enhance the safety and efficacy of this modality through better construct design, combination with other treatments, better TAA identification, and improved platform selection.

Figure 1.

Structure of an engineered TCR. The TCR on CD8+ T cells interacts with MHC class I molecules presenting peptides derived from intracellular proteins. The TCR, as shown here, comprises an α chain and a β chain. The CD3 complex comprises several motifs. The δ, γ, and ϵ subunits all contain one immunoreceptor tyrosine–motif (ITAM); the ζ subunit has three ITAM motifs. When the CD3 complex is activated, it induces T-cell proliferation, activation, and survival. The CD8αβ coreceptor stabilizes the TCR–peptide–MHC complex and participates in triggering the intracellular T-cell activation cascade.

Figure 1.

Structure of an engineered TCR. The TCR on CD8+ T cells interacts with MHC class I molecules presenting peptides derived from intracellular proteins. The TCR, as shown here, comprises an α chain and a β chain. The CD3 complex comprises several motifs. The δ, γ, and ϵ subunits all contain one immunoreceptor tyrosine–motif (ITAM); the ζ subunit has three ITAM motifs. When the CD3 complex is activated, it induces T-cell proliferation, activation, and survival. The CD8αβ coreceptor stabilizes the TCR–peptide–MHC complex and participates in triggering the intracellular T-cell activation cascade.

Close modal

MHC class I molecules are expressed on all nucleated cells, and present peptides derived from intracellular proteins to CD8+ T cells (Fig. 1). Conversely, MHC class II molecules are expressed predominantly on APCs, and they usually present extracellular peptides, internalized through endocytosis, to CD4+ T cells. In both cases, the TCR–MHC interaction is HLA subtype–restricted, which necessitates a specific match between the TCR T cellular therapy product and the patient's MHC haplotype. Furthermore, TCR–MHC interactions can only take place in the presence of an MHC-bound peptide, and each TCR is specific to an MHC–peptide complex, although some cross-reactivity with other MHC–peptide complexes can occur. In addition, T-cell activation requires adjacent molecules to interact with target cells. For example, the CD8αβ coreceptor stabilizes the TCR–peptide–MHC complex, and triggers the intracellular T-cell activation cascade (5). The CD3 complex participates in this process as well, as it comprises several motifs that when activated lead to T-cell proliferation, activation, and survival.

Several TAAs presented on MHC molecules have been identified, enabling the engineering of TCR T cells that recognize such TAAs (Table 1). Many trials of these cells are in progress or have reported preliminary results. One of the first trials to report results evaluated engineered TCR T cells that recognize a cancer-testis antigen, NY-ESO-1, normally expressed in spermatogonia and placenta, but lost during spermatid differentiation and not expressed on healthy tissues. It is overexpressed on epithelial tumors, as well as in synovial cell sarcoma and melanoma. In a trial using TCR T cells targeting NY-ESO-1, 4 of 6 patients with synovial cell sarcoma and 5 of 11 patients with melanoma had objective clinical responses (6). The responses seen in patients with synovial sarcoma may be at least partially attributable to the conditioning regimen administered prior to the TCR T cells, as alkylating agents demonstrated responses in this etiology (7). However, 2 of the patients with melanoma who had a response demonstrated complete responses (CR) lasting over a year. In fact, NY-ESO-1 has been a preferred TAA for TCR T-cell trials due to its relatively abundant expression on various tumors and lack of on-target, off-tumor effects (8–10). On the other hand, a dose-escalation study of TCR T cells targeting MAGE-A10, a TAA presented on cancers like melanoma, head and neck, urothelial, and non–small cell lung cancers (NSCLC), showed TCR T-cell persistence in peripheral blood, but no antitumor effects (11). Remarkably, 1 patient receiving TCR T cells targeting MAGE-A3 in another dose-escalation trial achieved CR lasting 29 months, and 3 of 9 patients receiving the full dose of the TCR T cells achieved partial response (PR), one of which lasted more than 19 months (12). Researchers are focusing on ways to detect TAAs presented on MHC molecules, as well as on ways to enhance the ability of TCR T cells to recognize, bind to, and attack cancer cells, and to recruit other immune components that can reinforce the immune response. These techniques, and clinical trials using the resultant enhanced TCR T cells, are discussed in the following sections.

Table 1.

Key TCR T-cell clinical trials.

TargetDiagnosisConstructNaToxicityResponseLong-term responsesStudy
NY-ESO-1 Melanoma, synovial sarcoma  17 No cell-related toxicity Clinical responses in 4/6 sarcoma, 5/11 melanoma CR >1 y in 2/11 melanoma, PR >18 m in 1 sarcoma Robbins et al. (6) 
NY-ESO-1b, LAGE-1 Multiple myeloma Affinity-enhanced 20 No cell-related toxicity Clinical responses in 16/20 pts Median PFS 19.1 m Rapoport et al. (8) 
NY-ESO-1 Synovial sarcoma Affinity-enhanced, siRNA to silence endogenous TCR 3 pts with CRS PR in 3 pts N/A Ishihara et al. (10)c 
NY-ESO-1 Melanoma, synovial sarcoma siRNA to silence endogenous TCR 5 pts with CRS, 2 with tumor pain 2 PR, 5 SD N/A Butler et al. (46)c 
NY-ESO-1 Synovial sarcoma Affinity-enhanced SPEAR T cells 42 Not reported 1 CR, 14 PR, 24 SD Median duration of response 30.9 weeks Ramachandran et al. (41) 
NY-ESO-1 Sarcoma, melanoma  10 No cell-related toxicity 6 with tumor regression 1 pt with ongoing response at 4 y Nowicki et al. (51) 
MAGE-A3 Urothelial, esophageal, osteosarcoma  17 2 transaminase elevation 3/9 PR at highest dose 1 PR ≥19 m Lu et al. (12) 
MAGE-A3 Melanoma, synovial sarcoma, esophageal  3 neurotoxicity, 2 fatal 1 CR, 4 PR 1 CR >15 m, 1 PR >12 m Morgan et al. (22) 
MAGE-A4 Tumors expressing MAGE-A4 Affinity-enhanced SPEAR T cells 34 No cell-related toxicity 7/28 PR, 11/28 SD N/A Hong et al. (39)c 
        
MAGE-A4 Esophageal cancer  10 No cell-related toxicity 3 SD (with minimal tumor burden at transfusion) 3 with SD had no disease progression for up to 26 m Kageyama et al. (30) 
MAGE A4 Tumors expressing MAGE-A4 Affinity-enhanced SPEAR T cells, CD8α coreceptor No cell-related toxicity 2 PR, 3 SD  Hong et al. (65) 
MAGE-A10 Urothelial, melanoma, H + N, NSCLC Affinity-enhanced 1 CRS, 1 amylase elevation No antitumor effects N/A Lam et al. (11)c 
HPV16 E6 Metastatic HPV16+ tumors  12 No cell-related toxicity 2 objective responses 1 free of disease at 3 y Doran et al. (13) 
HPV-16 E7 HPV16+ epithelial tumors  12 N/A 6 objective responses Response duration 3–9 m Norberg et al. (14) 
MART-1 Melanoma  36 Vitiligo, uveitis, hearing loss, 6/20 objective responses for human construct, 3/16 with mouse construct N/A Johnson et al. (36) 
MART-1 Melanoma  14 Skin rash, ARDS 9/13 tumor regression No long-term responses Chodon et al. (43) 
WT1 AML in remission post-alloSCT EBV-specific, donor-derived 12 No excess chronic GVHD All pts in remission at cell transfusion Relapse free-survival 100% at a median of 44 m Chapuis et al. (27) 
WT1 AML, MDS siRNA to silence endogenous TCR No cell-related toxicity 2 pts with transient responses N/A Tawara et al. (31) 
Multiple targets Tumors expressing prespecified targets ACTolog T cells 14 Grade 1–2 CRS 11/14 with SD 6/16 with SD at 12 weeks Tsimberidou et al. (19) 
TargetDiagnosisConstructNaToxicityResponseLong-term responsesStudy
NY-ESO-1 Melanoma, synovial sarcoma  17 No cell-related toxicity Clinical responses in 4/6 sarcoma, 5/11 melanoma CR >1 y in 2/11 melanoma, PR >18 m in 1 sarcoma Robbins et al. (6) 
NY-ESO-1b, LAGE-1 Multiple myeloma Affinity-enhanced 20 No cell-related toxicity Clinical responses in 16/20 pts Median PFS 19.1 m Rapoport et al. (8) 
NY-ESO-1 Synovial sarcoma Affinity-enhanced, siRNA to silence endogenous TCR 3 pts with CRS PR in 3 pts N/A Ishihara et al. (10)c 
NY-ESO-1 Melanoma, synovial sarcoma siRNA to silence endogenous TCR 5 pts with CRS, 2 with tumor pain 2 PR, 5 SD N/A Butler et al. (46)c 
NY-ESO-1 Synovial sarcoma Affinity-enhanced SPEAR T cells 42 Not reported 1 CR, 14 PR, 24 SD Median duration of response 30.9 weeks Ramachandran et al. (41) 
NY-ESO-1 Sarcoma, melanoma  10 No cell-related toxicity 6 with tumor regression 1 pt with ongoing response at 4 y Nowicki et al. (51) 
MAGE-A3 Urothelial, esophageal, osteosarcoma  17 2 transaminase elevation 3/9 PR at highest dose 1 PR ≥19 m Lu et al. (12) 
MAGE-A3 Melanoma, synovial sarcoma, esophageal  3 neurotoxicity, 2 fatal 1 CR, 4 PR 1 CR >15 m, 1 PR >12 m Morgan et al. (22) 
MAGE-A4 Tumors expressing MAGE-A4 Affinity-enhanced SPEAR T cells 34 No cell-related toxicity 7/28 PR, 11/28 SD N/A Hong et al. (39)c 
        
MAGE-A4 Esophageal cancer  10 No cell-related toxicity 3 SD (with minimal tumor burden at transfusion) 3 with SD had no disease progression for up to 26 m Kageyama et al. (30) 
MAGE A4 Tumors expressing MAGE-A4 Affinity-enhanced SPEAR T cells, CD8α coreceptor No cell-related toxicity 2 PR, 3 SD  Hong et al. (65) 
MAGE-A10 Urothelial, melanoma, H + N, NSCLC Affinity-enhanced 1 CRS, 1 amylase elevation No antitumor effects N/A Lam et al. (11)c 
HPV16 E6 Metastatic HPV16+ tumors  12 No cell-related toxicity 2 objective responses 1 free of disease at 3 y Doran et al. (13) 
HPV-16 E7 HPV16+ epithelial tumors  12 N/A 6 objective responses Response duration 3–9 m Norberg et al. (14) 
MART-1 Melanoma  36 Vitiligo, uveitis, hearing loss, 6/20 objective responses for human construct, 3/16 with mouse construct N/A Johnson et al. (36) 
MART-1 Melanoma  14 Skin rash, ARDS 9/13 tumor regression No long-term responses Chodon et al. (43) 
WT1 AML in remission post-alloSCT EBV-specific, donor-derived 12 No excess chronic GVHD All pts in remission at cell transfusion Relapse free-survival 100% at a median of 44 m Chapuis et al. (27) 
WT1 AML, MDS siRNA to silence endogenous TCR No cell-related toxicity 2 pts with transient responses N/A Tawara et al. (31) 
Multiple targets Tumors expressing prespecified targets ACTolog T cells 14 Grade 1–2 CRS 11/14 with SD 6/16 with SD at 12 weeks Tsimberidou et al. (19) 

Abbreviations: alloSCT, allogeneic stem cell transplant; AML, acute myeloid leukemia; ARDS, acute respiratory distress syndrome; CR, complete response; CRS, cytokine release syndrome; EBV, Epstein–Barr virus; GVHD, graft-versus-host disease; H + N, head and neck; m, month(s); MDS, myelodysplastic syndrome; NSCLC, non–small cell lung cancer; PFS, progression free survival; PR, partial response; pt(s), patient(s); SD, stable disease; SPEAR, specific peptide-enhanced affinity receptor; y, year(s).

aOnly studies with published results for ≥5 patients are listed.

bTarget peptide shared by both antigens.

cPublished only as abstract.

For effective and safe administration of cellular therapy, an ideal target antigen must be uniformly expressed on tumor cells but absent from healthy tissue. Such antigens include tumor-specific neo-antigens produced as a consequence of cancer-associated mutations, e.g., KRAS mutations. The proteins encoded by viral oncogenes can also be targets for TCR T cells, e.g., E6 and E7 in human papillomavirus (HPV)–associated tumors. One trial of TCR T cells targeting HPV16 E6 elicited objective tumor responses at higher doses in 2 of 12 patients (13). No off-tumor toxicity was observed. Similarly, a trial targeting HPV16 E7 elicited objective clinical responses in 6 of 12 patients (14). TAAs can also consist of proteins produced by abnormal transcription of silenced genes, such as some members of the MAGE antigen family, α-fetoprotein (AFP), and carcinoembryonic antigen, as well as cancer-testis antigens such as PRAME and synovial sarcoma X breakpoint 2 (SSX2), neither of which are expressed on healthy tissue except testis and placenta (15, 16). Other possible TAAs are antigens overexpressed on tumors, such as WT1 and mesothelin, which are observed on healthy tissues at concentrations low enough not to trigger immune activation.

One of the main challenges in target selection for TCR T cells is cancer-cell plasticity. Due to the high division rate, often with chromosomal instability, cancer cells mutate at a high rate, and at times, different areas of a tumor display different antigens, or different densities of a given antigen (17). Furthermore, cells have been shown to downregulate cell surface proteins, as well as downregulate TAA presentation on MHC molecules (18). Alternative splicing and proteasome degradation can also cause the presentation of different peptides on MHC molecules, even without actual genetic mutations. These genetic and proteomic mechanisms can explain tumor resistance to TCR T cells, as well as loss of response or relapse of previously responding cancers. Developing TCR T cells that target multiple domains is a possible strategy to mitigate differential antigen presentation (19), and to improve cell expansion and persistence to prevent mutational escape over time.

TCR T-cell therapy carries an inherent risk that an engineered TCR will recognize healthy tissues that express the same antigen, triggering immune attack, and that TCR T cells will cross-react with similar antigens. Although extensive cell line and animal testing is always performed prior to patient administration, it is sometimes hard to predict the toxicities that may occur. For instance, marked reduction in circulating carcinoembryonic antigen was achieved in a Phase I trial of TCR T cells targeting this TAA, yet inflammatory colitis developed in all 3 patients in whom this reduction was observed (20). In another study of TCR T cells expressing an affinity-enhanced TCR against MAGE-A3, the first 2 patients to receive therapy developed severe cardiotoxicity, leading to cardiogenic shock and death within a few days of product administration (21), although no MAGE-A3 expression was found on myocardial tissue on autopsy, despite evident myocardial damage. Subsequent studies demonstrated that the engineered TCR T cells also targeted titin, a protein presented on HLA molecules on cardiac myocytes that is unrelated to MAGE-A3. Titin was not detected in preclinical studies, as it is only presented on beating cardiomyocytes. In a different trial of TCR T cells targeting MAGE A3, 3 of 9 patients had severe neurotoxicity that led to death in 2 (22). This was caused by cross-reactivity of TCR T cells with MAGE-A12 in brain tissue.

These dramatic off-tumor effects highlight the need for careful target selection and safety testing that takes into account not only TCR affinity and specificity toward the TAA, but also TAA expression on malignant and healthy tissues (Fig. 2). Complex TCR interactions may preclude complete assurance of product safety prior to clinical trials since both the target and the TCR must be carefully tuned for efficacy and safety. Some adoptive cell therapies have used a “kill switch” as a conditional way to stop T-cell activity through activation of an apoptosis pathway in case of severe toxicities. However, as with other introduced transgenes, this strategy has been shown to induce immune-mediated elimination of the engineered cells (23). Less immunogenic transgenes are being developed (24, 25), and this important way to overcome unexpected toxicities is now being explored preclinically (15).

Figure 2.

Interactions of TCR affinity, specificity, and antigen expression on tumor and healthy tissues. Enhanced affinity and avidity can improve tumor targeting but may render the TCR T cells less specific, causing off-tumor toxicity. Thus, these must be optimized based on antigen expression on tumor and healthy cells to maximize efficacy and limit toxicity.

Figure 2.

Interactions of TCR affinity, specificity, and antigen expression on tumor and healthy tissues. Enhanced affinity and avidity can improve tumor targeting but may render the TCR T cells less specific, causing off-tumor toxicity. Thus, these must be optimized based on antigen expression on tumor and healthy cells to maximize efficacy and limit toxicity.

Close modal

One suggested method for generating safe and effective TCR T cells is to use T cells from patients responding to treatment with vaccines designed to induce cancer-specific T-cell responses (26). T cells from such patients have been generated by expanding peripheral blood mononuclear cells (PBMC) using a panel of overlapping peptides that collectively encompass the full NY-ESO-1 sequence, with selected cells further expanded into clonal cell lines. This method has also been used for T-cell selection from healthy donors (27), who typically have low-affinity TCRs against WT1, which is expressed at very low levels in normal adult tissues. However, some healthy individuals express higher-affinity TCRs that may be more active against WT1-presenting tumor cells but are unreactive against normal tissues. Accordingly, one higher-affinity, optimal TCR was identified after testing of more than 1,000 T-cell clones from more than 50 healthy subjects with the HLA-A2+ haplotype. This TCR was engineered into T cells derived from donors of allogeneic stem cells to patients with acute myeloid leukemia, and the cells transfused to the latter following the initial stem-cell transplant. Twelve patients at high risk for relapse received these TCR T cells, and at a median follow-up of 44 months, all patients had no evidence of disease relapse and did not require any further treatment. When compared with a group of patients with similar disease characteristics who did not receive the TCR T cells, the study cohort had significantly higher overall survival and relapse-free survival.

In another trial (28), tumor-specific neoantigens were found by whole-exome sequencing of cells from tumor samples, and used to identify and expand T cells, the TCR of which was then cloned and engineered into TCR T cells from healthy donors with the same HLA subtype. These cells reacted to cells bearing the neoantigenic peptides. As above, the risk of off-tumor effects was minimal, as these TCRs were found in patient T cells. In the future, this technique may enable the design of patient-specific TCR T cells based on the mutational profile of the patient's own tumor. Similarly, patient DCs pulsed with cancer-related peptides such as Hormad1, a cancer-testis antigen found in lung, esophageal, and other head and neck tumors, allowed for selection and cloning of TCR T cells for therapeutic use in the same patients (29). Others have tried to augment TCR T-cell activity with direct injection of their target protein, e.g., MAGE-A or WT1, but no clinically significant benefit has been observed (30, 31).

Cellular therapy products are extensively screened for activity against healthy tissue. However, some of this potential activity may not be apparent in vitro, as described above and as observed with the HLA-A*01–restricted MAGE-A3 TCR T cells that elicited cardiogenic shock following cell infusion due to cross-reactivation with a protein expressed in striated muscle, titin (21). To enhance screening for possible off-tumor effects, some investigators have tested each possible mutation in tumor-associated proteins for T-cell activation against primary cell lines. This systematic screening is possible due to the small number of possible mutations in small proteins, e.g., in a cell line targeting MAGE-A10, all 172 possible amino-acid substitutions were tested (32). Moving forward, computational methods may also be used to analyze immune-cell repertoires for use as possible targets for adoptive cell therapy (33).

The avidity and affinity of the TCR–MHC peptide complex defines the strength of the interaction between a single TCR and the peptide–MHC complex (Fig. 2). Avidity relates to a broader phenomenon: the interaction of many TCR molecules, as well as their coreceptors, with target cells (34). The specific affinity of the TCR for the MHC–peptide complex, as part of the general avidity between interacting cells, can significantly affect cytotoxic activity. In a trial of TCR T cells targeting MART1 in patients with MART1-positive melanoma, only 2 of 15 patients had objective responses (35). A higher-affinity TCR boosted the response rate to 30%, although nonmalignant melanocytes were also affected, resulting in uveitis, hearing loss, vitiligo, and other off-tumor effects (36). These trials show that affinity and avidity are crucial in achieving efficacy while controlling off-tumor effects, which may worsen with high-affinity TCRs.

The delicate balance of affinity and avidity sufficient to induce an effective antitumor response, sparing healthy tissue presenting the same TAAs at a lower density, can be achieved by tuning TCR affinity to target only cells abundantly expressing the TAA, while sparing healthy cells with low TAA expression. AFP, a known TAA also used for monitoring hepatocellular carcinoma, is expressed at low concentrations on nonmalignant liver cells, and usually only during inflammation and stress conditions. Some researchers are developing specific peptide-enhanced affinity receptor (SPEAR) T cells in which the TCRs of a patient's T cells are engineered in silico and ex vivo to target cells with higher antigen levels, such as malignant liver cells expressing AFP, while sparing nonmalignant cells (37). These are now being used in preliminary clinical trials, and initial safety data show no on-target, off-tumor toxicity from AFP-targeted SPEAR T cells in hepatocellular carcinoma (38), MAGE-A4–targeted SPEAR T cells in sarcoma (39, 40), and MAGE-A10–targeted SPEAR T cells in melanoma, head and neck, urothelial, and NSCLCs (11). Another report of SPEAR T cells targeting NY-ESO-1 showed promising results in 42 patients with synovial sarcoma, with 14 experiencing PR and one experiencing CR (41). The response was associated with peak TCR T-cell levels, which in turn was associated with lymphodepletion doses. It is important to note that higher-affinity constructs may lose specificity, and thus may have reduced efficacy (42). A fine balance between affinity and specificity is therefore necessary when designing and selecting TCR constructs (Fig. 2).

Many of the first TCR T-cell clinical trials did not demonstrate clinically significant efficacy (10, 11, 31, 43). Although the use of MHC–bound peptides as targets for engineered TCR T cells exploits T-cell cytotoxicity to induce tumor-cell killing, T cells are under a complex system of checks and balances to control cytotoxic activity and direct it only at specific targets, preventing autoimmune phenomena and damage to healthy tissue due to uncontrolled immune responses. This intricate regulatory network is important at steady state yet may pose substantial limitations on immune reactions against cancer cells, which may be identified as bearing self-antigens, or even deploy heightened immune-suppressing mechanisms that provide tumors with survival advantage. Thus, researchers are studying ways to overcome these obstacles and thereby improve TCR T-cell efficacy.

Some investigators have used a vector that encodes both the therapeutic TCR and siRNAs that silence the T cell's endogenous TCR (31, 44) to improve efficacy and to avoid “combined” TCRs that may trigger autoimmune toxicity (45). In a study of 9 patients with endometrial cancer (n = 1), ovarian cancer (n = 1), melanoma (n = 3), and synovial sarcoma (n = 4), who received cells engineered to express a TCR targeting NY-ESO-1 and siRNA to silence the native TCR, 2 patients achieved PR and 5 achieved stable disease (SD; ref. 46). Other investigators have used a novel CRISPR/CAS9 mechanism that does not require transduction with a viral vector to knock out native TCRs and replace them with engineered cancer-specific TCR (47). In another study that used a viral vector following CRISPR/CAS9 mediated TCR knockout, the transgene was expressed at a higher level due to the lack of competitive native TCR expression (48). A different group generated engineered cells with the native TCR knocked out and demonstrated that these cells yielded a stronger response to cancer cell lines, with higher antigen sensitivity, than transduced cells that did not undergo native TCR deletion (49). In a subsequent Phase I study of such cells targeting NY-ESO-1, all 3 treated patients had durable cell engraftment, but NY-ESO-1 expression on tumor cells declined over time, signifying tumor evasion (18).

Cell persistence and expansion has been a concern in adoptive cell therapy. This is especially true for TCR T cells, which bear a structure similar to the native TCR, and thus may require high antigen exposure to activate and persist. Accordingly, many clinical protocols incorporate cytokine injection, mainly IL2 administration for up to 14 days, to enhance cell expansion, engraftment, and cytotoxicity (50). To overcome low antigen presentation by some tumors, DCs pulsed with viral peptides have been given to patients with melanoma together with MART1-directed TCR T cells. In this manner, transgenic T cells are exposed to more presented antigens, and, importantly, to antigens presented by professional APCs. A clinical trial using this method had promising results, with 9 of 13 (69%) patients experiencing tumor regression (43). Another study of TCR T cells against NY-ESO-1 included, in one arm, three subcutaneous shots of DCs pulsed with NY-ESO-1 peptide, once every 2 weeks, following TCR T-cell infusion (51). Another arm in that trial included the anti-CTLA4 immune checkpoint inhibitor ipilimumab in addition to DCs to bypass checkpoint blockade. Of the first 10 patients with sarcoma or melanoma treated, all with progressive stage 3 or 4 disease, 4 of 6 patients treated with DCs showed evidence of tumor regression and 2 of 4 patients treated with DCs and ipilimumab showed similar outcomes. Interestingly, 1 patient has an ongoing CR lasting more than 4 years without additional therapy.

The use of checkpoint inhibition is also being explored in a clinical trial, now recruiting, that will compare two cohorts of patients with multiple myeloma receiving TCR T cells targeting NY-ESO-1/LAGE-1a with or without the anti-PD1 immune checkpoint inhibitor pembrolizumab (52). Other small molecules have been examined as adjuncts to TCR T-cell therapy, either to enhance the immune response or overcome intrinsic tumor-related mechanisms that impede it.

The TME, especially in solid tumors, is inhibitory to immune cells. This is both due to harsh conditions, including hypoxia, low pH, and electrolyte disturbances as a result of accelerated and uncontrolled tumor growth (53), and to mitochondrial dysfunction and upregulation of coinhibitory molecules that prevent sustained T-cell antitumor activity and lead to T-cell exhaustion (54). Likewise, tumor size and decreased blood supply relative to healthy tissue can impair T-cell migration and trafficking. For example, DCs lacking CD103 in the TME impede immune-cell infiltration, thereby promoting tumor immune escape (55). Immune inhibitory pathways enhanced by tumor cells and causing immune evasion may also impair T-cell cytotoxicity, as is the case with the TGF-β pathway and its activation of T regulatory cells (Treg) and several other suppressive cell types (56, 57).

To overcome these inhibitory pathways, novel TCR T and CAR T cells are being developed. Coexpression of a dominant-negative TGF-β receptor and a TCR T cell has been shown in a mouse model to reduce the number of Tregs in the TME (58). Other costimulatory receptors have been coexpressed to mitigate the immune-suppressive TME. For example, a receptor for IL4, an immune inhibitory cytokine, was fused with the intracellular domain of the IL7 receptor, an activating cytokine, to overcome IL4-mediated immune suppression in pancreatic cancer (59). Cytokine-producing motifs are also being incorporated to improve cell persistence via autocrine signaling (60, 61). For example, TCR T cells producing IL18, in comparison with the same cells producing IL12, shifted the immune milieu in the TME toward CD8+ cytotoxic T cells, and resulted in reduction of melanoma tumor burden and prolonged survival in mice (62).

Several groups are working on constructs incorporating the advantages of the native TCR and its cell-activating machinery with a CAR-like antibody-derived binding domain (Fig. 3). For example, an antibody-based CD19-binding domain fused to a TCR and termed a "T-cell receptor fusion construct" (TRuC) showed better in vivo activity when compared with CAR T cells (63). One advantage of this novel receptor is that binding is mediated by a single-chain variable fragment, and it is therefore not HLA-dependent like “standard” TCR T cells, paving the way for future allogeneic applications. Another study found that transgenic coexpression of both CD8αβ and a tumor antigen–specific TCR enhanced tumor-cell killing through upregulation of transcriptional pathways that led to antigen-dependent cytotoxicity with less T-cell exhaustion (5). Furthermore, transgenic CD8αβ expression can maintain previous cell specificity to other antigens, such as viral peptides, and still allow for activity against tumor cells via the engineered TCR (64). The SURPASS trial, based on an affinity-enhanced MAGE A4-directed TCR and a CD8α coreceptor, showed 2 of 5 patients had PR, and the 3 others SD (65). Interestingly, CD4+ T cells engineered with these receptors exhibited direct cytotoxic activity in vitro. These improvements may prove instrumental in enhancing TCR T-cell antitumor activity through better target recognition, cell persistence, and decreased exhaustion.

Figure 3.

Methods to enhance engineered TCRs for improved efficacy. A, Engineered TCRs comprise a modified antigen recognition domain targeting a specific tumor associated antigen. B, Intracellular domains can be added that induce the production of cytokines aimed at the TCR T cell (autocrine signaling) or at other immune components (paracrine signaling) for improved expansion, persistence, and immune activation. C, The TCR tumor recognition domain can be fused to an antibody-based domain to form a “TRuC.”

Figure 3.

Methods to enhance engineered TCRs for improved efficacy. A, Engineered TCRs comprise a modified antigen recognition domain targeting a specific tumor associated antigen. B, Intracellular domains can be added that induce the production of cytokines aimed at the TCR T cell (autocrine signaling) or at other immune components (paracrine signaling) for improved expansion, persistence, and immune activation. C, The TCR tumor recognition domain can be fused to an antibody-based domain to form a “TRuC.”

Close modal

DNA methylation is known to suppress expression of various tumor-related antigens, and the addition of an epigenetic modulator to NY-ESO-1–targeted therapy in a mouse model of breast cancer elicited expression of NY-ESO-1 in 5 of 7 cell lines that did not previously express it (58). This, in turn, caused significantly enhanced tumor infiltration and control by TCR T cells in comparison with administration of TCR T cells alone. Interestingly, TCR transgene expression may also be controlled through epigenetic mechanisms. Although insertion into DNA occurs during TCR T-cell manufacturing, leading to transgene persistence, TCR transgene expression, as can be measured by transgene RNA levels and TCR cell-surface expression, can fluctuate over time. Several studies have shown rapidly declining TCR T-cell persistence over time (43). Furthermore, retroviral or lentiviral promoter regions can be silenced through epigenetic methylation or histone modification (66, 67), hampering transgene expression (68). In a follow-up study of the aforementioned MART-1– and NY-ESO-1– TCR T cells, comparison of samples collected from 16 patients on day of and day 70 after infusion showed large differences in RNA and cell-surface expression between patients that were not due to loss of transgenic TCR DNA in circulating cells (69). Patients with reduced TCR surface expression had increased levels of methylation at the transgenic viral promoter region at day 70. Another study showed that exhausted CAR T cells could regain functionality with drug-induced “rest periods” (i.e., intermittent deactivation) through epigenetic and other mechanisms (70). Epigenetic control of transgene expression as well as other cellular stemness states are subjects of ongoing research and may provide further avenues toward enhancing TCR T-cell therapy.

Another important consideration for successful cellular therapy is the choice of platform to be used for adoptive cell therapy in general, and for TCR T cells specifically. T cells have different maturation and differentiations subtypes, and although most T cells actively fighting infection are cytotoxic, these cells are short lived. T cells with memory phenotypes are usually home to the lymph nodes and may be better candidates than cytotoxic T cells for adoptive cell therapy due to persistence and ability to mount an immune response upon rechallenge with the cognate antigen (71–73). Some CD8+ T cells, termed “veto cells,” can cause apoptosis of T cells targeting their HLA (74). Thus, veto T cells have been proposed as a platform for off-the-shelf allogeneic CAR or TCR cell therapy, with prolonged persistence due to suppression of host rejection (75). Researchers have also proposed inhibition of the AKT pathway as a method of inducing T-cell expansion without terminal differentiation and exhaustion (76). SPEAR T cells produced with AKT inhibition during the expansion phase showed enhanced responses, better proliferation even in the presence of prolonged antigen exposure, and improved cytotoxic activity after restimulation (77). Other immune cells have been used as adoptive cell platforms, and natural killer cells bearing CARs have demonstrated clinical responses in CD19 malignancies (78). Hematopoietic stem cells derived from umbilical cord blood and induced pluripotent stem cells from healthy donors have also been used as sources for cellular therapy platforms in preliminary clinical trials and may provide an “off-the-shelf” alternative to autologous cells (79–82). For example, CD8αβ T cells derived from induced pluripotent stem cells were transduced with a TCR targeting GPC3 and tested in NOD/SCID gamma (NSG) mice against cell lines expressing GPC3, eliciting a marked reduction in tumor growth (83).

Preliminary trials using TCR T-cell therapy usually recruited small numbers of patients (Table 1). These were mostly Phase I or limited Phase II trials, focusing mostly on safety or initial signals of efficacy. As TCR T-cell therapy design advances and more promising results are demonstrated, the idea of using off-the-shelf products is enticing, and this is being advanced as a way to improve availability and widespread implementation of the treatment. Options being explored include mass-produced allogeneic cells that evade immune rejection (78) and soluble TCR-like molecules named "immune mobilizing monoclonal TCRs against cancer" (ImmTAC) that can bind to native T cells and to the TAAs they are targeting (84, 85).

Terminal differentiation or exhaustion is an important factor influencing both ex vivo production and in vivo persistence of T cells. It often arises when cells are exposed to their cognate antigen over an extended duration, as in the setting of high tumor burden or lengthy production. To this end, researchers are looking into different T-cell subtypes as substrates of transgenic T-cell production, and initial signals hint at memory stem-cell or central memory phenotypes as potential ways to enhance persistence of the transgenic clone without reaching phenotypic exhaustion of a large proportion of the cells (86).

Initial clinical trials, especially in cancer therapies with low therapeutic index, have usually been designed with small subcohorts, reflecting the need for careful examination of the balance between efficacy and toxicity. However, the pharmacokinetics of cellular therapies are very different from those of other drugs, as they proliferate in vivo and the initial dose does not always reflect the toxicity that manifests after cell expansion. Dose–response curves are often nonlinear, necessitating a different early-phase trial design that can assess toxicity more closely, also considering other factors such as lymphodepletion, patient and disease status, and product dosing. This is especially true since lymphodepletion regimens vary greatly between trials, and while higher doses of chemotherapy may enable more robust expansion and persistence of TCR T cells (87), organ toxicity, and bone marrow suppression that ensue may outweigh benefits. For instance, prolonged cytopenia in a patient resulted in an amendment to a study to lower the fludarabine dose (43), and a death was attributed to a plastic marrow following lymphodepletion in another trial (39). Furthermore, when direct local or regional administration of cells is employed to improve trafficking of infused cells to the tumor, special interventional expertise is needed. Since cellular therapy has novel toxicities, namely cytokine release syndrome, neurotoxicity, and other immune manifestations, only specialized centers with experience in immune toxicity management can participate in clinical trials, especially as there is no way of predicting which patients will have these adverse effects. To address these issues, the FDA is developing a separate regulatory process for cellular therapy than the usual approval pathway for pharmacologic agents.

Great strides are being made in adoptive cell therapy, with commercial CAR T-cell therapies now available, and many more in various stages of preclinical and clinical development. TCR T cells are a promising approach to immune-mediated cancer treatment, as well as to immune modulation and therapy for nonmalignant diseases such as infections and autoimmune diseases. However, use of TCR T cells necessitates fine-tuning of the delicate balance between affinity and specificity to the cognate antigen and the ability of the T cells to expand, persist, and mount an immune response.

Novel strategies may enhance the usability of TCR T cells, and adjuncts such as small molecules, immune modulators, and epigenetic modulators may further improve efficacy. Further exploration of cellular therapy platforms, including off-the-shelf cellular therapies, may increase availability and accessibility, and with further improvements in efficacy may significantly widen the spectrum of diseases treatable with this promising therapeutic modality.

C.L. Haymaker reports personal fees from Briacell Therapeutics; other support from Mesothelioma Applied Research Foundation; and personal fees from Nanobiotix outside the submitted work. D.S. Hong reports grants from AbbVie, Adaptimmune, Aldi-Norte, Amgen, AstraZeneca, Bayer, Bristol Myer Squibb, Daiichi Sankyo, Deciphera, Eisai, Erasca, Fate Therapeutics, Genentech, Genmab, Infinity, Kite, Kyowa, Lilly, LOXO, Merck, MedImmune, Mirati, Mologen, Navier, National Cancer Institute-Cancer Therapy Evaluation Program (NCI-CTEP), Novartis, Numab, Pfizer, Pyramid Bio, SeraGen, Takeda, Turning Point Therapeutics, Vernstam, and VM Oncology during the conduct of the study; other support from Bayer, Genmab, American Association for Cancer Research (AACR), Society for Immunotherapy of Cancer (SITC), and Telperian and personal fees from Adaptimmune, Alpha Insights, Acuta, Alkermes, Amgen, Aumbiosciences, Antheneum, Axiom, Barclays, Bayer, Baxter, Boxer Capital, BridgeBio, CDR-Life AG, COR2ed, COG, ECOR1, Genentech, Gilead, GLG, Group H, Guidepoint, HCW Precision, Immunogen, Infinity, Janssen, Liberium, Medscape, Numab, Oncologia Brasil, Pfizer, Pharma Intelligence, POET Congress, Prime Oncology, Seattle Genetics, ST Cube, Takeda, Tavistock, Trieza Therapeutics, Turning Point, WebMD, and ZioPharma outside the submitted work; and other ownership interests in OncoResponse (founder) and Telperian (advisor). No disclosures were reported by the other authors.

U. Greenbaum is a recipient of a fellowship grant from the American Physicians' Fellowship for Medicine in Israel (APF). Some elements of the graphics are adapted from Servier Medical Art, licensed under a Creative Commons Attribution 3.0 Unported License.

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.
Forget
MA
,
Haymaker
C
,
Hess
KR
,
Meng
YJ
,
Creasy
C
,
Karpinets
T
, et al
Prospective analysis of adoptive TIL therapy in patients with metastatic melanoma: response, impact of Anti-CTLA4, and biomarkers to predict clinical outcome
.
Clin Cancer Res
2018
;
24
:
4416
28
.
2.
Lövgren
T
,
Wolodarski
M
,
Wickström
S
,
Edbäck
U
,
Wallin
M
,
Martell
E
, et al
Complete and long-lasting clinical responses in immune checkpoint inhibitor-resistant, metastasized melanoma treated with adoptive T cell transfer combined with DC vaccination
.
Oncoimmunology
2020
;
9
:
1792058
.
3.
Greenbaum
U
,
Yalniz
FF
,
Srour
SA
,
Rezvani
K
,
Singh
H
,
Olson
A
, et al
Chimeric antigen receptor therapy: how are we driving in solid tumors?
Biol Blood Marrow Transplant
2020
;
26
:
1759
69
.
4.
Harris
DT
,
Hager
MV
,
Smith
SN
,
Cai
Q
,
Stone
JD
,
Kruger
P
, et al
Comparison of T cell activities mediated by human TCRs and CARs that use the same recognition domains
.
J Immunol
2018
;
200
:
1088
100
.
5.
Rath
JA
,
Bajwa
G
,
Carreres
B
,
Hoyer
E
,
Gruber
I
,
Martínez-Paniagua
MA
, et al
Single-cell transcriptomics identifies multiple pathways underlying antitumor function of TCR-and CD8αβ-engineered human CD4+ T cells
.
Sci Adv
2020
;
6
:
eaaz7809
.
6.
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
.
7.
Vlenterie
M
,
Litière
S
,
Rizzo
E
,
Marréaud
S
,
Judson
I
,
Gelderblom
H
, et al
Outcome of chemotherapy in advanced synovial sarcoma patients: review of 15 clinical trials from the European Organisation for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group; setting a new landmark for studies in this entity
.
Eur J Cancer
2016
;
58
:
62
72
.
8.
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
.
9.
D'Angelo
SP
,
Melchiori
L
,
Merchant
MS
,
Bernstein
D
,
Glod
J
,
Kaplan
R
, et al
Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 c259T cells in synovial sarcoma
.
Cancer Discov
2018
;
8
:
944
57
.
10.
Ishihara
M
,
Kitano
S
,
Hattori
H
,
Miyahara
Y
,
Kato
H
,
Mishima
H
, et al
Tumor responses and early onset cytokine release syndrome in synovial sarcoma patients treated with a novel affinity-enhanced NY-ESO-1-targeting TCR-redirected T cell transfer
.
J Clin Oncol
37
, 
2019
(
suppl; abstr 2530
).
11.
Lam
VK
,
Hong
DS
,
Heymach
J
,
Blumenschein
GR
,
Creelan
BC
,
Bradbury
PA
, et al
Initial safety assessment of MAGE-A10c796TCR T-cells in two clinical trials
.
J Clin Oncol
36
, 
2018
(
suppl; abstr 3056
).
12.
Lu
YC
,
Parker
LL
,
Lu
T
,
Zheng
Z
,
Toomey
MA
,
White
DE
, 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
.
J Clin Oncol
2017
;
35
:
3322
9
.
13.
Doran
SL
,
Stevanović
S
,
Adhikary
S
,
Gartner
JJ
,
Jia
L
,
Kwong
MLM
, et al
T-Cell receptor gene therapy for human papillomavirus-associated epithelial cancers: a first-in-human, phase I/II study
.
J Clin Oncol
2019
;
37
:
2759
68
.
14.
Norberg
S
,
Nagarsheth
N
,
Sinkoe
A
,
Adhikary
S
,
Meyer
T
,
Lack
J
, et al
Safety and clinical activity of gene-engineered T-cell therapy targeting HPV-16 E7 for epithelial cancers
.
J Clin Oncol
38
: 
2020
(
suppl; abstr 101
).
15.
Orlando
D
,
Miele
E
,
De Angelis
B
,
Guercio
M
,
Boffa
I
,
Sinibaldi
M
, et al
Adoptive immunotherapy using PRAME-specific T cells in medulloblastoma
.
Cancer Res
2018
;
78
:
3337
49
.
16.
Abate-Daga
D
,
Speiser
DE
,
Chinnasamy
N
,
Zheng
Z
,
Xu
H
,
Feldman
SA
, et al
Development of a T cell receptor targeting an HLA-A*0201 restricted epitope from the cancer-testis antigen SSX2 for adoptive immunotherapy of cancer
.
PLoS One
2014
;
9
:
e93321
.
17.
van den Bent
MJ
,
Gao
Y
,
Kerkhof
M
,
Kros
JM
,
Gorlia
T
,
van Zwieten
K
, et al
Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas
.
Neuro Oncol
2015
;
17
:
935
41
.
18.
Stadtmauer
EA
,
Fraietta
JA
,
Davis
MM
,
Cohen
AD
,
Weber
KL
,
Lancaster
E
, et al
CRISPR-engineered T cells in patients with refractory cancer
.
Science
2020
;
367
:
eaba7365
.
19.
Tsimberidou
A
,
Guenther
K
,
Alpert
A
,
Andersson
B
,
Coughlin
Z
,
Fritsche
J
, et al
Results of the first-in-human clinical trial with personalized multi-target adoptive cell therapy (ACTolog IMA101)
.
J Immunother Cancer
2020
;
8
(
Suppl 3
):
293
.
Available from
: .
20.
Parkhurst
MR
,
Yang
JC
,
Langan
RC
,
Dudley
ME
,
Nathan
DAN
,
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
.
21.
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
.
22.
Morgan
RA
,
Chinnasamy
N
,
Abate-Daga
DD
,
Gros
A
,
Robbins
PF
,
Zheng
Z
, et al
Cancer regression and neurologic toxicity following anti-MAGE-A3 TCR gene therapy
.
J Immunother
2013
;
36
:
133
.
23.
Berger
C
,
Flowers
ME
,
Warren
EH
,
Riddell
SR
. 
Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation
.
Blood
2006
;
107
:
2294
302
.
24.
Giordano-Attianese
G
,
Gainza
P
,
Gray-Gaillard
E
,
Cribioli
E
,
Shui
S
,
Kim
S
, et al
A computationally designed chimeric antigen receptor provides a small-molecule safety switch for T-cell therapy
.
Nat Biotechnol
2020
;
38
:
426
32
.
25.
Warda
W
,
Da Rocha
MN
,
Trad
R
,
Haderbache
R
,
Salma
Y
,
Bouquet
L
, et al
Overcoming target epitope masking resistance that can occur on low-antigen-expresser AML blasts after IL-1RAP chimeric antigen receptor T cell therapy using the inducible caspase 9 suicide gene safety switch
.
Cancer Gene Ther
2021
Jan 7 [Epub ahead of print]
.
26.
Inderberg
EM
,
Wälchli
S
. 
Long-term surviving cancer patients as a source of therapeutic TCR
.
Cancer Immunol Immunother
2020
;
69
:
859
65
.
27.
Chapuis
AG
,
Egan
DN
,
Bar
M
,
Schmitt
TM
,
McAfee
MS
,
Paulson
KG
, et al
T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant
.
Nat Med
2019
;
25
:
1064
72
.
28.
Ren
L
,
Leisegang
M
,
Deng
B
,
Matsuda
T
,
Kiyotani
K
,
Kato
T
, et al
Identification of neoantigen-specific T cells and their targets: implications for immunotherapy of head and neck squamous cell carcinoma
.
OncoImmunology
2019
;
8
:
e1568813
.
29.
Pan
K
,
Chiu
Y
,
Hasan
F
,
Miroballi
N
,
Whitehurst
A
,
Yee
C
. 
Identification of HLA-A0201 restricted epitope of cancer/testis antigen (CTA) Hormad1 and generation of antigen-specific T-cell receptor-engineered T cells (TCR-T) for treatment of solid tumor malignancies [abstract]
. In:
Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17–20; Boston, MA
.
Philadelphia (PA)
:
AACR
;
Cancer Immunol Res 2020;8(3 Suppl):Abstract nr A69
.
30.
Kageyama
S
,
Ikeda
H
,
Miyahara
Y
,
Imai
N
,
Ishihara
M
,
Saito
K
, et al
Adoptive transfer of MAGE-A4 T-cell receptor gene-transduced lymphocytes in patients with recurrent esophageal cancer
.
Clin Cancer Res
2015
;
21
:
2268
.
31.
Tawara
I
,
Kageyama
S
,
Miyahara
Y
,
Fujiwara
H
,
Nishida
T
,
Akatsuka
Y
, et al
Safety and persistence of WT1-specific T-cell receptor gene−transduced lymphocytes in patients with AML and MDS
.
Blood
2017
;
130
:
1985
94
.
32.
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
2019
;
8
:
e1532759
.
33.
Holland
CJ
,
MacLachlan
BJ
,
Bianchi
V
,
Hesketh
SJ
,
Morgan
R
,
Vickery
O
, et al
In silico and structural analyses demonstrate that intrinsic protein motions guide T cell receptor complementarity determining region loop flexibility
.
Front Immunol
2018
;
9
:
674
.
34.
Campillo-Davo
D
,
Flumens
D
,
Lion
E
. 
The quest for the best: how TCR affinity, avidity, and functional avidity affect TCR-engineered T-cell antitumor responses
.
Cells
2020
;
9
:
1720
.
35.
Morgan
RA
,
Dudley
ME
,
Wunderlich
JR
,
Hughes
MS
,
Yang
JC
,
Sherry
RM
, et al
Cancer regression in patients after transfer of genetically engineered lymphocytes
.
Science
2006
;
314
:
126
9
.
36.
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
.
37.
Docta
RY
,
Ferronha
T
,
Sanderson
JP
,
Weissensteiner
T
,
Pope
GR
,
Bennett
AD
, et al
Tuning T-cell receptor affinity to optimize clinical risk-benefit when targeting alpha-fetoprotein–positive liver cancer
.
Hepatology
2019
;
69
:
2061
75
.
38.
Goyal
L
,
Frigault
M
,
Meyer
T
,
Feun
LG
,
Bruix
J
,
El-Khoueiry
A
, et al
Initial safety of AFP SPEAR T-cells in patients with advanced hepatocellular carcinoma. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29–Apr 3
;
Atlanta, GA. Philadelphia (PA)
:
AACR
;
Cancer Res
2019
;
79
(
13 Suppl
):
Abstract nr 3183
.
39.
Hong
DS
,
Van Tine
BA
,
Olszanski
AJ
,
Johnson
ML
,
Liebner
DA
,
Trivedi
T
, et al
Phase I dose escalation and expansion trial to assess the safety and efficacy of ADP-A2M4 SPEAR T cells in advanced solid tumors
.
J Clin Oncol
38
: 
2020
(
suppl; abstr 102
).
40.
Araujo
DM
,
Druta
M
,
Agulnik
M
,
D'Angelo
SP
,
Blay
J-Y
,
Strauss
SJ
, et al
SPEARHEAD-1: a phase II trial of ADP-A2M4 SPEAR T cells in patients with advanced synovial sarcoma or myxoid/round cell liposarcoma
.
J Clin Oncol
38
: 
2020
(
suppl; abstr TPS11569
).
41.
Ramachandran
I
,
Lowther
DE
,
Dryer-Minnerly
R
,
Wang
R
,
Fayngerts
S
,
Nunez
D
, et al
Systemic and local immunity following adoptive transfer of NY-ESO-1 SPEAR T cells in synovial sarcoma
.
J Immunother Cancer
2019
;
7
:
276
.
42.
Oren
R
,
Hod-Marco
M
,
Haus-Cohen
M
,
Thomas
S
,
Blat
D
,
Duvshani
N
, et al
Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody-based chimeric antigen receptors indicates affinity/avidity thresholds
.
J Immunol
2014
;
193
:
5733
43
.
43.
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
.
44.
Sun
Q
,
Zhang
X
,
Wang
L
,
Gao
X
,
Xiong
Y
,
Liu
L
, et al
T-cell receptor gene therapy targeting melanoma-associated antigen-A4 by silencing of endogenous TCR inhibits tumor growth in mice and human
.
Cell Death Dis
2019
;
10
:
475
.
45.
Bunse
M
,
Bendle
GM
,
Linnemann
C
,
Bies
L
,
Schulz
S
,
Schumacher
TN
, et al
RNAi-mediated TCR knockdown prevents autoimmunity in mice caused by mixed TCR dimers following TCR gene transfer
.
Mol Ther
2014
;
22
:
1983
91
.
46.
Butler
MO
,
Saibil
S
,
Bonilla
L
,
Franke
N
,
Hakgor
S
,
Boross-Harmer
S
, et al
Effect of minimal lymphodepletion prior to ACT with TBI-1301, NY-ESO-1 specific gene-engineered TCR-T cells, on clinical responses and CRS
.
J Clin Oncol
37
, 
2019
(
suppl; abstr 2537
).
47.
Roth
TL
,
Puig-Saus
C
,
Yu
R
,
Shifrut
E
,
Carnevale
J
,
Li
PJ
, et al
Reprogramming human T cell function and specificity with non-viral genome targeting
.
Nature
2018
;
559
:
405
9
.
48.
Morton
LT
,
Reijmers
RM
,
Wouters
AK
,
Kweekel
C
,
Remst
DFG
,
Pothast
CR
, et al
Simultaneous deletion of endogenous TCRαβ for TCR gene therapy creates an improved and safe cellular therapeutic
.
Mol Ther
2020
;
28
:
64
74
.
49.
Legut
M
,
Dolton
G
,
Mian
AA
,
Ottmann
OG
,
Sewell
AK
. 
CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells
.
Blood
2018
;
131
:
311
22
.
50.
Spolski
R
,
Li
P
,
Leonard
WJ
. 
Biology and regulation of IL-2: from molecular mechanisms to human therapy
.
Nat Rev Immunol
2018
;
18
:
648
59
.
51.
Nowicki
TS
,
Berent-Maoz
B
,
Cheung-Lau
G
,
Huang
RR
,
Wang
X
,
Tsoi
J
, et al
A pilot trial of the combination of transgenic NY-ESO-1–reactive adoptive cellular therapy with dendritic cell vaccination with or without ipilimumab
.
Clin Cancer Res
2019
;
25
:
2096
108
.
52.
Rapoport
A
,
Hoffman
JE
,
Kaufman
JL
,
Blouch
K
,
Butler
E
,
Deyoung
MP
, et al
Open-label pilot study of genetically engineered NY-ESO-1–specific T cells (GSK3377794) alone or in combination with pembrolizumab in relapsed/refractory multiple myeloma
.
J Clin Oncol
38
: 
2020
(
suppl; abstr TPS8555
).
53.
Schurich
A
,
Magalhaes
I
,
Mattsson
J
. 
Metabolic regulation of CAR T cell function by the hypoxic microenvironment in solid tumors
.
Immunotherapy
2019
;
11
:
335
45
.
54.
Scharping
NE
,
Delgoffe
GM
. 
Tumor microenvironment metabolism: a new checkpoint for anti-tumor immunity
.
Vaccines
2016
;
4
:
46
.
55.
Spranger
S
,
Dai
D
,
Horton
B
,
Gajewski
TF
. 
Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy
.
Cancer Cell
2017
;
31
:
711
23
.
56.
Beatty
GL
,
Moon
EK
. 
Chimeric antigen receptor T cells are vulnerable to immunosuppressive mechanisms present within the tumor microenvironment
.
Oncoimmunology
2014
;
3
:
e970027
.
57.
Budhu
S
,
Schaer
DA
,
Li
Y
,
Toledo-Crow
R
,
Panageas
K
,
Yang
X
, et al
Blockade of surface-bound TGF-β on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment
.
Sci Signal
2017
;
10
:
eaak9702
.
58.
Matsueda
S
,
Odunsi
K
,
Koya
RC
. 
Abstract B36: TGFβ blockade and epigenetic modulation for cancer treatment: efficient breast cancer targeted therapy with TCR-T cell transfer [abstract]
.
In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27–30
;
Miami Beach, FL. Philadelphia (PA)
:
AACR
;
Cancer Immunol Res
2020
;
8
(
4 Suppl
):
Abstract nr B36
.
59.
Mohammed
S
,
Sukumaran
S
,
Bajgain
P
,
Watanabe
N
,
Heslop
HE
,
Rooney
CM
, et al
Improving chimeric antigen receptor-modified T cell function by reversing the immunosuppressive tumor microenvironment of pancreatic cancer
.
Mol Ther
2017
;
25
:
249
58
.
60.
Yeku
OO
,
Purdon
T
,
Spriggs
DR
,
Brentjens
RJ
. 
Interleukin-12 armored chimeric antigen receptor (CAR) T cells for heterogeneous antigen-expressing ovarian cancer
.
J Clin Oncol
36
, 
2018
(
suppl 5S; abstr 12
).
61.
Steffin
DHM
,
Batra
SA
,
Rathi
P
,
Guo
L
,
Li
W
,
Courtney
AN
, et al
A phase I clinical trial using armored GPC3 CAR T cells for children with relapsed/refractory liver tumors
.
J Clin Oncol
37
, 
2019
(
suppl; abstr TPS2647
).
62.
Kunert
A
,
Chmielewski
M
,
Wijers
R
,
Berrevoets
C
,
Abken
H
,
Debets
R
. 
Intra-tumoral production of IL18, but not IL12, by TCR-engineered T cells is non-toxic and counteracts immune evasion of solid tumors
.
Oncoimmunology
2018
;
7
:
e1378842
.
63.
Baeuerle
PA
,
Ding
J
,
Patel
E
,
Thorausch
N
,
Horton
H
,
Gierut
J
, et al
Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response
.
Nat Commun
2019
;
10
:
2087
.
64.
Bajwa
G
,
Lanz
I
,
Cardenas
M
,
Brenner
MK
,
Arber
C
. 
Transgenic CD8αβ co-receptor rescues endogenous TCR function in TCR-transgenic virus-specific T cells
.
J Immunother Cancer
2020
;
8
:
e001487
.
65.
Hong
D
,
Clarke
J
,
Johanns
T
,
Kebriaei
P
,
Heymach
J
,
Galal
A
, et al
Initial safety, efficacy, and product attributes from the SURPASS trial with ADP-A2M4CD8, a SPEAR T-cell therapy incorporating an affinity optimized TCR targeting MAGE-A4 and a CD8α co-receptor
.
J Immunother Cancer
2020
;
8
(
Suppl 3
):
379
.
Available from
: .
66.
Yao
S
,
Sukonnik
T
,
Kean
T
,
Bharadwaj
RR
,
Pasceri
P
,
Ellis
J
. 
Retrovirus silencing, variegation, extinction, and memory are controlled by a dynamic interplay of multiple epigenetic modifications
.
Mol Ther
2004
;
10
:
27
36
.
67.
Ngai
SC
,
Rosli
R
,
Al Abbar
A
,
Abdullah
S
. 
DNA methylation and histone modifications are the molecular lock in lentivirally transduced hematopoietic progenitor cells
.
Biomed Res Int
2015
;
2015
:
346134
.
68.
Hoffmann
D
,
Schott
JW
,
Geis
FK
,
Lange
L
,
Müller
FJ
,
Lenz
D
, et al
Detailed comparison of retroviral vectors and promoter configurations for stable and high transgene expression in human induced pluripotent stem cells
.
Gene Ther
2017
;
24
:
298
307
.
69.
Nowicki
TS
,
Farrell
C
,
Morselli
M
,
Rubbi
L
,
Campbell
KM
,
Macabali
MH
, et al
Epigenetic suppression of transgenic T-cell receptor expression via gamma-retroviral vector methylation in adoptive cell transfer therapy
.
Cancer Discov
2020
;
10
:
1645
53
.
70.
Weber
EW
,
Parker
KR
,
Sotillo
E
,
Lynn
RC
,
Anbunathan
H
,
Lattin
J
, et al
Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling
.
Science
2021
;
372
:
eaba1786
.
71.
Kotani
H
,
Li
G
,
Yao
J
,
Mesa
TE
,
Chen
J
,
Boucher
JC
, et al
Aged CAR T cells exhibit enhanced cytotoxicity and effector function but shorter persistence and less memory-like phenotypes
.
Blood
2018
;
132
:
2047
.
72.
Wang
X
,
Popplewell
LL
,
Wagner
JR
,
Naranjo
A
,
Blanchard
MS
,
Mott
MR
, et al
Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL
.
Blood
2016
;
127
:
2980
90
.
73.
McLellan
AD
,
Ali Hosseini Rad
SM
. 
Chimeric antigen receptor T cell persistence and memory cell formation
.
Immunol Cell Biol
2019
;
97
:
664
74
.
74.
Ophir
E
,
Eidelstein
Y
,
Afik
R
,
Bachar-Lustig
E
,
Reisner
Y
. 
Induction of tolerance to bone marrow allografts by donor-derived host nonreactive ex vivo–induced central memory CD8 T cells
.
Blood
2010
;
115
:
2095
104
.
75.
Or-Geva
N
,
Gidron-Budovsky
R
,
Sidlik-Muskatel
R
,
Singh
AK
,
Reisner
Y
. 
Next-generation CD8 memory veto T cells directed against memory antigens
.
Leukemia
2019
;
33
:
2737
41
.
76.
Klebanoff
CA
,
Crompton
JG
,
Leonardi
AJ
,
Yamamoto
TN
,
Chandran
SS
,
Eil
RL
, et al
Inhibition of AKT signaling uncouples T cell differentiation from expansion for receptor-engineered adoptive immunotherapy
.
JCI Insight
2017
;
2
:
e95103
.
77.
Mardilovich
K
,
Wang
L
,
Kenneil
R
,
Betts
G
,
Bath
N
,
Spinner
W
, et al
Inhibition of AKT signaling during expansion of TCR-engineered T-cells from patient leukocyte material generates SPEAR T-cells with enhanced functional potential in vitro
.
J Immunother Cancer
2020
;
8
(
Suppl 3
):
95
.
Available from
: .
78.
Liu
E
,
Marin
D
,
Banerjee
P
,
Macapinlac
HA
,
Thompson
P
,
Basar
R
, et al
Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors
.
N Engl J Med
2020
;
382
:
545
53
.
79.
Zhu
H
,
Blum
RH
,
Bjordahl
R
,
Gaidarova
S
,
Rogers
P
,
Lee
TT
, et al
Pluripotent stem cell–derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity
.
Blood
2020
;
135
:
399
410
.
80.
Yu
M
,
Mansour
AG
,
Teng
KY
,
Sun
G
,
Shi
Y
,
Caligiuri
MA
. 
iPSC-derived natural killer cells expressing EGFR-CAR against glioblastoma [abstract]
.
In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27–28 and Jun 22–24
.
Philadelphia (PA)
:
AACR
;
Cancer Res
2020
;
80
(
16 Suppl
):
Abstract nr 3313
.
81.
Li
Q
,
Wang
Y
,
Lin
M
,
Xia
L
,
Bao
Y
,
Sun
X
, et al
Phase I clinical trial with PD-1/MUC1 CAR-pNK92 immunotherapy [abstract]
.
In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30–Oct 3, 2018
;
New York, NY. Philadelphia (PA)
:
AACR
;
Cancer Immunol Res
2019
;
7
(
2 Suppl
):
Abstract nr A014
.
82.
Zhang
L
,
Tian
L
,
Dai
X
,
Yu
H
,
Wang
J
,
Lei
A
, et al
Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions
.
J Hematol Oncol
2020
;
13
:
1
5
.
83.
Minagawa
A
,
Yoshikawa
T
,
Yasukawa
M
,
Hotta
A
,
Kunitomo
M
,
Iriguchi
S
, et al
Enhancing T cell receptor stability in rejuvenated iPSC-derived T cells improves their use in cancer immunotherapy
.
Cell Stem Cell
2018
;
23
:
850
8
.
84.
Lowe
KL
,
Cole
D
,
Kenefeck
R
,
Okelly
I
,
Lepore
M
,
Jakobsen
BK
. 
Novel TCR-based biologics: mobilising T cells to warm ‘cold’ tumours
.
Cancer Treat Rev
2019
;
77
:
35
43
.
85.
Sacco
JJ
,
Carvajal
R
,
Butler
MO
,
Shoushtari
AN
,
Hassel
JC
,
Ikeguchi
A
, et al
A phase (ph) II, multi-center study of the safety and efficacy of tebentafusp (tebe) (IMCgp100) in patients (pts) with metastatic uveal melanoma (mUM)
.
Ann Oncol
2020
;
31
:
S1442
S3
.
Abstract nr 64MO
.
86.
Mastaglio
S
,
Genovese
P
,
Magnani
Z
,
Ruggiero
E
,
Landoni
E
,
Camisa
B
, et al
NY-ESO-1 TCR single edited stem and central memory T cells to treat multiple myeloma without graft-versus-host disease
.
Blood
2017
;
130
:
606
18
.
87.
D'Angelo
S
,
Demetri
G
,
Tine
BV
,
Druta
M
,
Glod
J
,
Chow
W
, et al
298 Final analysis of the phase 1 trial of NY-ESO-1–specific T-cell receptor (TCR) T-cell therapy (letetresgene autoleucel; GSK3377794) in patients with advanced synovial sarcoma (SS)
.
J Immunother Cancer
2020
;
8
:
A182
A3
.